51
|
Kwak MS, Kim HS, Lee B, Kim YH, Son M, Shin JS. Immunological Significance of HMGB1 Post-Translational Modification and Redox Biology. Front Immunol 2020; 11:1189. [PMID: 32587593 PMCID: PMC7297982 DOI: 10.3389/fimmu.2020.01189] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/13/2020] [Indexed: 12/19/2022] Open
Abstract
Most extracellular proteins are secreted via the classical endoplasmic reticulum (ER)/Golgi-dependent secretion pathway; however, some proteins, including a few danger-associated molecular patterns (DAMPs), are secreted via non-classical ER/Golgi-independent secretion pathways. The evolutionarily conserved high mobility group box1 (HMGB1) is a ubiquitous nuclear protein that can be released by almost all cell types. HMGB1 lacks signal peptide and utilizes diverse non-canonical secretion mechanisms for its extracellular export. Although the post-translational modifications of HMGB1 were demonstrated, the oxidation of HMGB1 and secretion mechanisms are not highlighted yet. We currently investigated that peroxiredoxins I and II (PrxI/II) induce the intramolecular disulfide bond formation of HMGB1 in the nucleus. Disulfide HMGB1 is preferentially transported out of the nucleus by binding to the nuclear exportin chromosome-region maintenance 1 (CRM1). We determined the kinetics of HMGB1 oxidation in bone marrow-derived macrophage as early as a few minutes after lipopolysaccharide treatment, peaking at 4 h while disulfide HMGB1 accumulation was observed within the cells, starting to secrete in the late time point. We have shown that HMGB1 oxidation status, which is known to determine the biological activity in extracellular HMGB1, is crucial for the secretion of HMGB1 from the nucleus. This review summarizes selected aspects of HMGB1 redox biology relevant to the induction and propagation of inflammatory diseases. We implicate the immunological significance and the need for novel HMGB1 inhibitors through mechanism-based studies.
Collapse
Affiliation(s)
- Man Sup Kwak
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| | - Hee Sue Kim
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea
| | - Bin Lee
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea
| | - Young Hun Kim
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| | - Myoungsun Son
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Jeon-Soo Shin
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.,Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, South Korea
| |
Collapse
|
52
|
Xu Y, Zhang S, Rong J, Lin Y, Du L, Wang Y, Zhang Z. Sirt3 is a novel target to treat sepsis induced myocardial dysfunction by acetylated modulation of critical enzymes within cardiac tricarboxylic acid cycle. Pharmacol Res 2020; 159:104887. [PMID: 32526680 DOI: 10.1016/j.phrs.2020.104887] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/29/2020] [Accepted: 05/01/2020] [Indexed: 12/16/2022]
Abstract
Sepsis induced myocardial dysfunction (SIMD) results in high morbidity and mortality. However, the effective therapeutic strategies for SIMD treatment remain limited. Sirt3 is the main mitochondrial Sirtuin member and is a key modulator of mitochondrial metabolism and function. In this study, we aimed to investigate the effect and mechanism of Sirt3 on SIMD. SIMD was induced by 20 mg/kg Lipopolysaccharides (LPS) injection for 6 h in mice. Sepsis could induce the reduction of cardiac Sirt3 expression and global deficiency of Sirt3 exacerbated cardiac function. Quantitative acetyl-proteomics and cardiac metabolomics analysis revealed that loss of Sirt3 led to hyper-acetylation of critical enzymes within cardiac tricarboxylic acid (TCA) cycle and generation of lactate and NADH, subsequently promotion of cardiac dysfunction after sepsis. Additionally, to evaluate whether Emodin could be utilized as a potential Sirt3 modulator to treat SIMD, male wild type mice (WT mice) or global Sirt3 deficient mice (Sirt3-/- mice) were intraperitoneally injected with 40 mg/kg Emodin for 5 days followed by 20 mg/kg LPS administration for another 6 h and observed that exogenous administration of Emodin could attenuate myocardial dysfunction in septic WT mice. However, septic Sirt3-/- mice can not gain benefit on cardiac performance from Emodin infusion. In conclusion, this study presented the protective role of Sirt3 targeting SIMD, which may provide a potential novel approach to maintain normal cardiac performance after sepsis.
Collapse
Affiliation(s)
- Yinchuan Xu
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Shujing Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiabing Rong
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Yao Lin
- Department of Intensive Care Unit, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Linlin Du
- Department of Intensive Care Unit, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Yi Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Zhaocai Zhang
- Department of Intensive Care Unit, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China.
| |
Collapse
|
53
|
Hwang JS, Kim E, Hur J, Yoon TJ, Seo HG. Ring finger protein 219 regulates inflammatory responses by stabilizing sirtuin 1. Br J Pharmacol 2020; 177:4601-4614. [PMID: 32220064 DOI: 10.1111/bph.15060] [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: 06/18/2019] [Revised: 03/11/2020] [Accepted: 03/14/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND AND PURPOSE Ring finger protein 219 (RNF219), a protein containing the C3 HC4 -type RING-HC motif, has been identified as a binding partner of the histone deacetylase sirtuin 1 (SIRT1). To explore the functions of RNF219, we examined its possible roles in the cellular responses to inflammation. EXPERIMENTAL APPROACH Effects of RNF219 on SIRT1 were studied in vitro using RAW264.7 cells and in male BALB/c mice, treated with LPS or IFN-γ. Western blots, RT-PCR, co-immunoprecipitation and ubiquitination assays were used, along with LC-MS/MS analysis. In vivo, survival and serum cytokines and tissue levels of RNF219 and SIRT1 were measured. KEY RESULTS Binding of RNF219 to SIRT1 inhibited degradation of SIRT1 by preventing its ubiquitination, thereby prolonging SIRT1-mediated anti-inflammatory signalling. LPS caused RNF219 deacetylation, leading to instability of RNF219 and preventing its association with SIRT1. Accordingly, the acetylation status of RNF219 is a critical determinant in its interaction with SIRT1, affecting the response to inflammatory stimuli. The deacetylase inhibitor trichostatin A, increased acetylation and stability of RNF219 and survival of mice injected with LPS, through the interaction of RNF219 with SIRT1. CONCLUSION AND IMPLICATIONS RNF219 is involved in a novel mechanism to stabilize SIRT1 protein by protein-protein interaction, leading to the resolution of cellular inflammation. These observations provide new insights into the function of RNF219 in modulation of cellular inflammation, and may aid and encourage the development of new anti-inflammatory drugs.
Collapse
Affiliation(s)
- Jung Seok Hwang
- College of Sang-Huh Life Sciences, Konkuk University, Seoul, Republic of Korea
| | - Eunsu Kim
- College of Sang-Huh Life Sciences, Konkuk University, Seoul, Republic of Korea
| | - Jinwoo Hur
- College of Sang-Huh Life Sciences, Konkuk University, Seoul, Republic of Korea
| | - Taek Joon Yoon
- Department of Food Science and Nutrition, Yuhan University, Bucheon-si, Republic of Korea
| | - Han Geuk Seo
- College of Sang-Huh Life Sciences, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
54
|
Sirtuin-1 and Its Relevance in Vascular Calcification. Int J Mol Sci 2020; 21:ijms21051593. [PMID: 32111067 PMCID: PMC7084838 DOI: 10.3390/ijms21051593] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/23/2020] [Accepted: 02/24/2020] [Indexed: 12/16/2022] Open
Abstract
Vascular calcification (VC) is highly associated with cardiovascular disease and all-cause mortality in patients with chronic kidney disease. Dysregulation of endothelial cells and vascular smooth muscle cells (VSMCs) is related to VC. Sirtuin-1 (Sirt1) deacetylase encompasses a broad range of transcription factors that are linked to an extended lifespan. Sirt1 enhances endothelial NO synthase and upregulates FoxOs to activate its antioxidant properties and delay cell senescence. Sirt1 reverses osteogenic phenotypic transdifferentiation by influencing RUNX2 expression in VSMCs. Low Sirt1 hardly prevents acetylation by p300 and phosphorylation of β-catenin that, following the facilitation of β-catenin translocation, drives osteogenic phenotypic transdifferentiation. Hyperphosphatemia induces VC by osteogenic conversion, apoptosis, and senescence of VSMCs through the Pit-1 cotransporter, which can be retarded by the sirt1 activator resveratrol. Proinflammatory adipocytokines released from dysfunctional perivascular adipose tissue (PVAT) mediate medial calcification and arterial stiffness. Sirt1 ameliorates release of PVAT adipokines and increases adiponectin secretion, which interact with FoxO 1 against oxidative stress and inflammatory arterial insult. Conclusively, Sirt1 decelerates VC by means of influencing endothelial NO bioavailability, senescence of ECs and VSMCs, osteogenic phenotypic transdifferentiation, apoptosis of VSMCs, ECM deposition, and the inflammatory response of PVAT. Factors that aggravate VC include vitamin D deficiency-related macrophage recruitment and further inflammation responses. Supplementation with vitamin D to adequate levels is beneficial in improving PVAT macrophage infiltration and local inflammation, which further prevents VC.
Collapse
|
55
|
Yang C, Yang W, He Z, He H, Yang X, Lu Y, Li H. Kaempferol Improves Lung Ischemia-Reperfusion Injury via Antiinflammation and Antioxidative Stress Regulated by SIRT1/HMGB1/NF-κB Axis. Front Pharmacol 2020; 10:1635. [PMID: 32116668 PMCID: PMC7025570 DOI: 10.3389/fphar.2019.01635] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/16/2019] [Indexed: 12/24/2022] Open
Abstract
Trauma, organ transplantation, and thromboembolism are the main causes of lung ischemia-reperfusion injury (LIRI), and new therapies and drugs are urgent to relieve LIRI. In preliminary experiment, authors found that kaempferol could improve LIRI in rats, and the current study further explored its possible mechanism. The model of rat LIRI was established and appropriate research methods were implemented. Results shown that kaempferol could significantly improve LIRI, inhibit release of inflammatory factors including interleukin (IL) 6 and tumor necrosis factor (TNF) α in bronchoalveolar lavage fluid, and reduce oxidative stress reaction. Western blotting was used to detect protein expression levels and found that kaempferol could up-regulate the protein expressions of phosphorylated (p-) p65 and p65, and down-regulate the protein expression of sirtuin (SIRT) 1. Immunofluorescence was used to localize the expression of high mobility group box (HMGB) 1 and found its higher expression in outside of nucleus. However, the above effects of kaempferol on LIRI markedly attenuated by EX 527, a selective inhibitor of SIRT 1. Taken together, we first reported the protective effect of kaempferol on rat LIRI and confirmed that kaempferol’s antiinflammation and antioxidative stress involving the SIRT1/HMGB1/NF-κB axis.
Collapse
Affiliation(s)
- Chunli Yang
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Wenkai Yang
- Department of Cardiovascular Surgery, Affiliated Central People's Hospital of Zhanjiang of Guangdong Medical University, Zhanjiang, China
| | - Zhaohui He
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Huiwei He
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Xiaogang Yang
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Yuanhua Lu
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Hongbo Li
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| |
Collapse
|
56
|
Khambu B, Yan S, Huda N, Yin XM. Role of High-Mobility Group Box-1 in Liver Pathogenesis. Int J Mol Sci 2019; 20:ijms20215314. [PMID: 31731454 PMCID: PMC6862281 DOI: 10.3390/ijms20215314] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 10/16/2019] [Accepted: 10/23/2019] [Indexed: 12/21/2022] Open
Abstract
High-mobility group box 1 (HMGB1) is a highly abundant DNA-binding protein that can relocate to the cytosol or undergo extracellular release during cellular stress or death. HMGB1 has a functional versatility depending on its cellular location. While intracellular HMGB1 is important for DNA structure maintenance, gene expression, and autophagy induction, extracellular HMGB1 acts as a damage-associated molecular pattern (DAMP) molecule to alert the host of damage by triggering immune responses. The biological function of HMGB1 is mediated by multiple receptors, including the receptor for advanced glycation end products (RAGE) and Toll-like receptors (TLRs), which are expressed in different hepatic cells. Activation of HMGB1 and downstream signaling pathways are contributing factors in the pathogenesis of non-alcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), and drug-induced liver injury (DILI), each of which involves sterile inflammation, liver fibrosis, ductular reaction, and hepatic tumorigenesis. In this review, we will discuss the critical role of HMGB1 in these pathogenic contexts and propose HMGB1 as a bona fide and targetable DAMP in the setting of common liver diseases.
Collapse
Affiliation(s)
- Bilon Khambu
- Correspondence: ; Tel.: +1-317-274-1789; Fax: +1-317-491-6639
| | | | | | | |
Collapse
|
57
|
Xu B, Lang LM, Lian S, Guo JR, Wang JF, Liu J, Yang HM, Li SZ. Neuroinflammation induced by secretion of acetylated HMGB1 from activated microglia in hippocampi of mice following chronic cold exposure. Brain Res 2019; 1726:146495. [PMID: 31586627 DOI: 10.1016/j.brainres.2019.146495] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/12/2019] [Accepted: 10/02/2019] [Indexed: 12/21/2022]
Abstract
Stress is a nonspecific response to adverse circumstances and chronic stress can destroy homeostasis, leading to various primary diseases. Although chronic cold stress is becoming increasingly important for individuals living or working in extreme environments, the risk of associated disorders of the central nervous system remains unstudied. Here, male C57BL/6 mice were exposed to a temperature of 4 °C, for three hours each day for one, two or three weeks. Glial cell activation, neuronal structure, and neuroinflammation were then evaluated by western blotting, immunofluorescence, Nissl staining and co-immunoprecipitation. Microglial activation, accompanied by activation of the NF-κB signaling pathway, release of pro-inflammatory cytokines and loss of Nissl bodies, was observed in mouse hippocampal tissue following cold exposure. We speculate that these phenomena are mediated by the HMGB1/TLR4/NF-κB pathway and closely associated with acetylation of HMGB1 in the hippocampus. These findings provide new insights into the mechanisms of the cold stress response, which should inform the development of new strategies to combat the effects of hypothermia.
Collapse
Affiliation(s)
- Bin Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Li-Min Lang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Shuai Lian
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Jing-Ru Guo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Jian-Fa Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Juxiong Liu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Huan-Min Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China.
| | - Shi-Ze Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China.
| |
Collapse
|
58
|
Mutirangura A. A Hypothesis to Explain How the DNA of Elderly People Is Prone to Damage: Genome-Wide Hypomethylation Drives Genomic Instability in the Elderly by Reducing Youth-Associated Gnome-Stabilizing DNA Gaps. Epigenetics 2019. [DOI: 10.5772/intechopen.83372] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
59
|
Xu S, Zeng Z, Zhao M, Huang Q, Gao Y, Dai X, Lu J, Huang W, Zhao K. Evidence for SIRT1 Mediated HMGB1 Release From Kidney Cells in the Early Stages of Hemorrhagic Shock. Front Physiol 2019; 10:854. [PMID: 31333497 PMCID: PMC6625367 DOI: 10.3389/fphys.2019.00854] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 06/20/2019] [Indexed: 11/13/2022] Open
Abstract
Background This study is to explore the effect of SIRT1 deacetylating inactivation on organ-derived high mobility group box 1 (HMGB1) during the development of severe hemorrhagic shock (HS). Methods Hemorrhagic shock model was reproduced in rats by blood shedding and mimicked in HK-2 cells by hypoxia-reoxygenation (H/R) treatment. The level and acetylation of HMGB1 and the expression and activity of SIRT1 were detected in tissue, serum and cultured cells and supernatant. The deacetylated sites of HMGB1 was determined by Co-IP. Results Serum HMGB1 in HS rats was increased but were reduced in multiple organs, especially in kidney tissue, with enhanced HMGB1 acetylation, and reduced deacetylase SIRT1 activity in isolated RTECs. HMGB1 in suspension of H/R-treated HK-2 cells was increased, accompanying with enhanced HMGB1 acetylation, and nuclear-plasma translocation. SIRT1 down-regulated by siRNA aggravated acetylation of HMGB1 and nucleus-to-cytoplasm translocation and resulted in increased HMGB1 in cultured HK-2 suspension. Immunoprecipitation data suggested that SIRT1-indcuced deacetylated sites of HMGB1 were K90 and K177 following H/R. SIRT1 overexpression inhibited the acetylation of HMGB1 and reduced the content of extracellular HMGB1 in H/R-treated HK-2 cells. Inhibiting mutation of SIRT1 restored the acetylation of HMGB1 and HMGB1 content in extracellular suspension. In HS rat model, the neutralization of HMGB1 with antibody could reduce serum HMGB1 and pro-inflammatory cytokine contents, but had no effect on SIRT1 protein expression and activity. Polydatin (PD), a potential SIRT1 agonist, up-regulated SIRT1 activity and inhibited nucleus-to-cytoplasm translocation of HMGB1 in RTECs. Moreover, PD also attenuated RTEC apoptosis, protected renal function, and prolonged survival in HS rats. These beneficial effect of PD is largely blocked by specific inhibition of SIRT1 with Ex527. Conclusion The acetylation of HMGB1 in K99 and K177 is enhanced during HS due to the downregulation of SIRT1. The nucleus-to-cytoplasm translocation and the release of acetylated HMGB1 from RTECs of kidney might exacerbate the pro-inflammatory effects of HMGB1 during the development of HS.
Collapse
Affiliation(s)
- Siqi Xu
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Department of Pathology, Qingdao Municipal Hospital (Group), Qingdao, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ming Zhao
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qiaobing Huang
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Youguang Gao
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xingui Dai
- Department of Critical Care Medicine, The First People's Hospital of Chenzhou, Institute of Translational Medicine, Chenzhou, China
| | - Jiayin Lu
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Weiqing Huang
- Department of Pathology, Qingdao Municipal Hospital (Group), Qingdao, China
| | - Keseng Zhao
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
60
|
Mohammad G, Abdelaziz GM, Siddiquei MM, Ahmad A, De Hertogh G, Abu El-Asrar AM. Cross-Talk between Sirtuin 1 and the Proinflammatory Mediator High-Mobility Group Box-1 in the Regulation of Blood-Retinal Barrier Breakdown in Diabetic Retinopathy. Curr Eye Res 2019; 44:1133-1143. [PMID: 31136205 DOI: 10.1080/02713683.2019.1625406] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Purpose: High-mobility group box-1 (HMGB1) mediates inflammation and breakdown of blood-retinal barrier (BRB) in diabetic retina. Sirtuin-1 (SIRT1) has protective effects against inflammation and oxidative stress. The aim of this study was to investigate the interaction between HMGB1 and SIRT1 in regulating BRB breakdown in diabetic retina. Methods: BRB breakdown was assessed in vivo with fluorescein isothiocyanate-conjugated dextran. Vitreous samples from 47 proliferative diabetic retinopathy (PDR) and 19 nondiabetic patients, and epiretinal membranes from 13 patients with PDR were studied by enzyme-linked immunosorbent assay and immunohistochemistry. Retinas from 4-week diabetic rats and from normal rats intravitreally injected with HMGB1 were studied by spectrophotometric assay, Western blot analysis, and RT-PCR. We also studied the effect of the HMGB1 inhibitor glycyrrhizin and the SIRT1 activator resveratrol on diabetes-induced biochemical changes in the retina. Results: HMGB1 levels in vitreous samples from PDR patients were significantly higher than in nondiabetic controls, whereas SIRT1 levels were significantly lower in vitreous samples from patients with inactive PDR than those in patients with active PDR and nondiabetic controls. In epiretinal membranes, SIRT1 was expressed in vascular endothelial cells and stromal cells. Diabetes and intravitreal injection of HMGB1 in normal rats downregulated SIRT1expression, whereas glycyrrhizin and resveratrol normalized diabetes-induced downregulation of SIRT1. Resveratrol significantly attenuated diabetes-induced downregulation of occludin and upregulation of HMGB1 and receptor for advanced glycation end products in the retina and breakdown of BRB. Conclusions: Our findings suggest that a functional link between SIRT1 and HMGB1 is involved in regulating of BRB breakdown in diabetic retina.
Collapse
Affiliation(s)
- Ghulam Mohammad
- Department of Ophthalmology, College of Medicine, King Saud University , Riyadh , Saudi Arabia.,Dr. Nasser Al-Rashid Research Chair in Ophthalmology , Riyadh , Saudi Arabia
| | - Ghada Maher Abdelaziz
- Department of Ophthalmology, College of Medicine, King Saud University , Riyadh , Saudi Arabia
| | - Mohammad Mairaj Siddiquei
- Department of Ophthalmology, College of Medicine, King Saud University , Riyadh , Saudi Arabia.,Dr. Nasser Al-Rashid Research Chair in Ophthalmology , Riyadh , Saudi Arabia
| | - Ajmal Ahmad
- Department of Ophthalmology, College of Medicine, King Saud University , Riyadh , Saudi Arabia.,Dr. Nasser Al-Rashid Research Chair in Ophthalmology , Riyadh , Saudi Arabia
| | - Gert De Hertogh
- Laboratory of Histochemistry and Cytochemistry, University of Leuven , KU Leuven , Leuven , Belgium
| | - Ahmed M Abu El-Asrar
- Department of Ophthalmology, College of Medicine, King Saud University , Riyadh , Saudi Arabia.,Dr. Nasser Al-Rashid Research Chair in Ophthalmology , Riyadh , Saudi Arabia
| |
Collapse
|
61
|
Cai X, Biswas I, Panicker SR, Giri H, Rezaie AR. Activated protein C inhibits lipopolysaccharide-mediated acetylation and secretion of high-mobility group box 1 in endothelial cells. J Thromb Haemost 2019; 17:803-817. [PMID: 30865333 PMCID: PMC6494677 DOI: 10.1111/jth.14425] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Indexed: 12/21/2022]
Abstract
Essentials APC elicits cytoprotective responses in endothelial cells via EPCR-dependent cleavage of PAR1. APC inhibits LPS-mediated translocation and extracellular secretion of HMGB1 in endothelial cells. Signaling activity of APC inhibits LPS-mediated acetylation of HMGB1 by epigenetic mechanisms. APC inhibits LPS-mediated HMGB1 expression in CD31-positive endothelial cells in cremaster muscle. SUMMARY: Background Activated protein C (APC) inhibits high-mobility group box 1 (HMGB1) signaling and its lipopolysaccharide (LPS)-mediated release by endothelial protein C receptor (EPCR)-dependent activation of protease-activated receptor 1 (PAR1) in endothelial cells. Post-translational acetylation is known to modulate the subcellular localization of HMGB1, and its hyperacetylated form is translocated to the cytoplasm of innate immune cells before being secreted into the extracellular space. Objective To determine whether APC inhibits LPS-mediated HMGB1 secretion from endothelial cells by modulating its acetylation status. Methods The subcellular localization of HMGB1 in LPS-treated endothelial cells was monitored in the absence and presence of APC by western blot analysis of fractionated cell lysates and confocal immunofluorescence microscopy. Results Both western blot and immunofluorescence data indicated that APC effectively inhibits LPS-mediated translocation of HMGB1 from the nucleus to the cytoplasm by EPCR-dependent and PAR1-dependent mechanisms. When EPCR was ligated by the Gla-domain of protein C/APC, thrombin also inhibited LPS-mediated HMGB1 translocation. Further studies revealed that APC inhibits the translocation of HMGB1 from the nucleus to the cytoplasm by inhibiting LPS-mediated hyperacetylation of HMGB1 by (de)acetylating enzymes. Furthermore, the translocated HMGB1 was found to be associated with lysosome-associated membrane protein 1 in LPS-treated endothelial cells. The in vivo relevance of these findings was investigated in the mouse cremaster muscle, and this demonstrated that both wild-type APC and a signaling-selective mutant of APC inhibit LPS-mediated HMGB1 expression and translocation in CD31-positive endothelial cells. Conclusion These results suggest that APC inhibits LPS-mediated cytoplasmic translocation and secretion of HMGB1 in endothelial cells by epigenetic mechanisms.
Collapse
Affiliation(s)
- Xiaofeng Cai
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation
| | - Indranil Biswas
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation
| | - Sumith R. Panicker
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation
| | - Hemant Giri
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation
| | - Alireza R. Rezaie
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| |
Collapse
|
62
|
Hwang JS, Lee WJ, Hur J, Lee HG, Kim E, Lee GH, Choi MJ, Lim DS, Paek KS, Seo HG. Rosiglitazone-dependent dissociation of HuR from PPAR-γ regulates adiponectin expression at the posttranscriptional level. FASEB J 2019; 33:7707-7720. [PMID: 30897345 DOI: 10.1096/fj.201802643r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Peroxisome proliferator-activated receptor (PPAR)-γ has been implicated as a key player in the regulation of adiponectin levels via both transcriptional and posttranscriptional mechanisms. Herein, we show that PPAR-γ interacts with human antigen R (HuR) and that the PPAR-γ-HuR complex dissociates following activation of PPAR-γ by rosiglitazone, a specific ligand of PPAR-γ. This rosiglitazone-dependent dissociation of HuR from PPAR-γ leads to nucleocytoplasmic shuttling of HuR and its binding to the 3'-UTR of adiponectin mRNA. PPAR-γ with H321A and H447A double mutation (PPAR-γH321/447A), a mutant lacking ligand-binding activity, impaired HuR dissociation from the PPAR-γ-HuR complex, resulting in reduced nucleocytoplasmic shuttling, even in the presence of rosiglitazone. Consequently, rosiglitazone up-regulated adiponectin levels by modulating the stability of adiponectin mRNA, whereas these effects were abolished by HuR ablation or blocked in cells expressing the PPAR-γH321/447A mutant, indicating that the interaction of PPAR-γ and HuR is a critical event during adiponectin expression. Taken together, the findings demonstrate a novel mechanism for regulating adiponectin expression at the posttranscriptional level and suggest that ligand-mediated activation of PPAR-γ to interfere with interaction of HuR could offer a therapeutic strategy for inflammation-associated diseases that involve decreased adiponectin mRNA stability.-Hwang, J. S., Lee, W. J., Hur, J., Lee, H. G., Kim, E., Lee, G. H., Choi, M.-J., Lim, D.-S., Paek, K. S., Seo, H. G. Rosiglitazone-dependent dissociation of HuR from PPAR-γ regulates adiponectin expression at the posttranscriptional level.
Collapse
Affiliation(s)
- Jung Seok Hwang
- Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Won Jin Lee
- Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Jinwoo Hur
- Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Hyuk Gyoon Lee
- Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Eunsu Kim
- Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Gyeong Hee Lee
- Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Mi-Jung Choi
- Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Dae-Seog Lim
- Department of Biotechnology, CHA University, Seongnam, Korea
| | | | - Han Geuk Seo
- Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| |
Collapse
|
63
|
Quan M, Lv Y, Dai Y, Qi B, Fu L, Chen X, Qian Y. Tanshinone IIA protects against lipopolysaccharide-induced lung injury through targeting Sirt1. J Pharm Pharmacol 2019; 71:1142-1151. [PMID: 30868609 DOI: 10.1111/jphp.13087] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/17/2019] [Indexed: 12/11/2022]
Abstract
Abstract
Objectives
This study was designed to investigate the effects and the mechanism of Tanshinone IIA (TIIA) on endotoxic shock-induced lung injury in a mouse model.
Methods
Mice were administered intraperitoneally with TIIA (10 mg/kg) 0.5 h before lipopolysaccharide (LPS) challenge and then received additional injections every 24 h during the 3-day experimental period. The physiological indexes, the survival rate and the parameters for lung injury were examined. The protein levels of Sirt1, and the acetylation and activation of NF-κB p65 were determined. The expression and secretion of pro-inflammatory factors were evaluated, respectively.
Key findings
Treatment with TIIA significantly improved physiological indexes and increased the survival rate of mice in response to LPS challenge. TIIA treatment displayed an obvious up-regulation of Sirt1 protein, in accompany with reduced acetylation and activation of NF-κB p65 following LPS stimulation. In addition, TIIA attenuated LPS-induced lung injury and prevented the expression and secretion of pro-inflammatory factors. However, the protective effects of TIIA were abolished by Sirt1 inhibitor.
Conclusions
Tanshinone IIA prevents LPS-induced secretion of pro-inflammatory cytokines thus exerts protective effects against acute lung injury, probably via modulation of Sirt1/NF-κB signalling pathway.
Collapse
Affiliation(s)
- Minxue Quan
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Yanni Lv
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yang Dai
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Biying Qi
- The Second Clinical Medical School, Nanchang University, Nanchang, China
| | - Longsheng Fu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuanying Chen
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yisong Qian
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| |
Collapse
|
64
|
Hardeland R. Aging, Melatonin, and the Pro- and Anti-Inflammatory Networks. Int J Mol Sci 2019; 20:ijms20051223. [PMID: 30862067 PMCID: PMC6429360 DOI: 10.3390/ijms20051223] [Citation(s) in RCA: 225] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/05/2019] [Accepted: 03/07/2019] [Indexed: 12/17/2022] Open
Abstract
Aging and various age-related diseases are associated with reductions in melatonin secretion, proinflammatory changes in the immune system, a deteriorating circadian system, and reductions in sirtuin-1 (SIRT1) activity. In non-tumor cells, several effects of melatonin are abolished by inhibiting SIRT1, indicating mediation by SIRT1. Melatonin is, in addition to its circadian and antioxidant roles, an immune stimulatory agent. However, it can act as either a pro- or anti-inflammatory regulator in a context-dependent way. Melatonin can stimulate the release of proinflammatory cytokines and other mediators, but also, under different conditions, it can suppress inflammation-promoting processes such as NO release, activation of cyclooxygenase-2, inflammasome NLRP3, gasdermin D, toll-like receptor-4 and mTOR signaling, and cytokine release by SASP (senescence-associated secretory phenotype), and amyloid-β toxicity. It also activates processes in an anti-inflammatory network, in which SIRT1 activation, upregulation of Nrf2 and downregulation of NF-κB, and release of the anti-inflammatory cytokines IL-4 and IL-10 are involved. A perhaps crucial action may be the promotion of macrophage or microglia polarization in favor of the anti-inflammatory phenotype M2. In addition, many factors of the pro- and anti-inflammatory networks are subject to regulation by microRNAs that either target mRNAs of the respective factors or upregulate them by targeting mRNAs of their inhibitor proteins.
Collapse
Affiliation(s)
- Rüdiger Hardeland
- Johann Friedrich Blumenbach Institute of Zoology and Anthropology, University of Göttingen, 37073 Göttingen, Germany.
| |
Collapse
|
65
|
Bhat SM, Massey N, Karriker LA, Singh B, Charavaryamath C. Ethyl pyruvate reduces organic dust-induced airway inflammation by targeting HMGB1-RAGE signaling. Respir Res 2019; 20:27. [PMID: 30728013 PMCID: PMC6364446 DOI: 10.1186/s12931-019-0992-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 01/27/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Animal production workers are persistently exposed to organic dust and can suffer from a variety of respiratory disease symptoms and annual decline in lung function. The role of high mobility group box-1 (HMGB1) in inflammatory airway diseases is emerging. Hence, we tested a hypothesis that organic dust exposure of airway epithelial cells induces nucleocytoplasmic translocation of HMGB1 and blocking this translocation dampens organic dust-induced lung inflammation. METHODS Rats were exposed to either ambient air or swine barn (8 h/day for either 1, 5, or 20 days) and lung tissues were processed for immunohistochemistry. Swine barn dust was collected and organic dust extract (ODE) was prepared and sterilized. Human airway epithelial cell line (BEAS-2B) was exposed to either media or organic dust extract followed by treatment with media or ethyl pyruvate (EP) or anti-HMGB1 antibody. Immunoblotting, ELISA and other assays were performed at 0 (control), 6, 24 and 48 h. Data (as mean ± SEM) was analyzed using one or two-way ANOVA followed by Bonferroni's post hoc comparison test. A p value of less than 0.05 was considered significant. RESULTS Compared to controls, barn exposed rats showed an increase in the expression of HMGB1 in the lungs. Compared to controls, ODE exposed BEAS-2B cells showed nucleocytoplasmic translocation of HMGB1, co-localization of HMGB1 and RAGE, reactive species and pro-inflammatory cytokine production. EP treatment reduced the ODE induced nucleocytoplasmic translocation of HMGB1, HMGB1 expression in the cytoplasmic fraction, GM-CSF and IL-1β production and augmented the production of TGF-β1 and IL-10. Anti-HMGB1 treatment reduced ODE-induced NF-κB p65 expression, IL-6, ROS and RNS but augmented TGF-β1 and IL-10 levels. CONCLUSIONS HMGB1-RAGE signaling is an attractive target to abrogate OD-induced lung inflammation.
Collapse
Affiliation(s)
- Sanjana Mahadev Bhat
- Department of Biomedical Sciences, 2008 Vet Med Building, Iowa State University, Ames, IA USA
| | - Nyzil Massey
- Department of Biomedical Sciences, 2008 Vet Med Building, Iowa State University, Ames, IA USA
| | - Locke A. Karriker
- Department of Veterinary Diagnostic and Production Animal Medicine, 2203 Lloyd Veterinary Medical Center, Iowa State university, Ames, IA USA
| | - Baljit Singh
- Faculty of Veterinary Medicine, 2500 University Dr. NW, University of Calgary, Calgary, T2N 1N4 Canada
| | | |
Collapse
|
66
|
Zhang X, Lu Y, Wu Q, Dai H, Li W, Lv S, Zhou X, Zhang X, Hang C, Wang J. Astaxanthin mitigates subarachnoid hemorrhage injury primarily by increasing sirtuin 1 and inhibiting the Toll-like receptor 4 signaling pathway. FASEB J 2019; 33:722-737. [PMID: 30048156 DOI: 10.1096/fj.201800642rr] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/02/2018] [Indexed: 11/29/2024]
Abstract
Inflammation plays a key role in the progression of subarachnoid hemorrhage (SAH). Here, we examined the effects of astaxanthin (ATX) on the inflammatory response and secondary damage after SAH and the underlying mechanisms of action. In vivo, a prechiasmatic cistern injection model was established in rats and mice. In addition, neuron-microglia cocultures were exposed to oxyhemoglobin to mimic SAH in vitro. Western blotting revealed that protein expression of TLR4 was markedly increased in microglia at 24 h after SAH, with consequent increases in the downstream molecules myeloid differentiation factor 88 and NF-кB. Treatment with ATX significantly inhibited the TLR4 activation, increased sirtuin 1 expression, and inhibited the subsequent inflammatory response both in vivo and in vitro. ATX also significantly decreased high-mobility group box 1 nuclear translocation and secretion in neurons, an effect that was reversed by the sirtuin 1-specific inhibitor sirtinol. ATX administered 4 h after SAH ameliorated cerebral inflammation, brain edema, and neuronal death and improved neurologic function. ATX reduced neuronal death but did not improve neurologic function in TLR4 knockout mice. These results suggest that ATX reduces the proinflammatory response and secondary brain injury after SAH, primarily by increasing sirtuin 1 levels and inhibiting the TLR4 signaling pathway.-Zhang, X., Lu, Y., Wu, Q., Dai, H., Li, W., Lv, S., Zhou, X., Zhang, X., Hang, C., Wang, J. Astaxanthin mitigates subarachnoid hemorrhage injury primarily by increasing sirtuin 1 and inhibiting the Toll-like receptor 4 signaling pathway.
Collapse
Affiliation(s)
- Xiangsheng Zhang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yue Lu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Qi Wu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Haibin Dai
- Department of Neurosurgery, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Shengyin Lv
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Xiaoming Zhou
- Department of Neurosurgery, Changzheng Hospital, School of Medicine, Second Military Medical University, Shanghai, China; and
| | - Xin Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Chunhua Hang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
67
|
Bartoli-Leonard F, Wilkinson FL, Langford-Smith AWW, Alexander MY, Weston R. The Interplay of SIRT1 and Wnt Signaling in Vascular Calcification. Front Cardiovasc Med 2018; 5:183. [PMID: 30619890 PMCID: PMC6305318 DOI: 10.3389/fcvm.2018.00183] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/04/2018] [Indexed: 12/24/2022] Open
Abstract
Vascular calcification is a major health risk and is highly correlated with atherosclerosis, diabetes, and chronic kidney disease. The development of vascular calcification is an active and complex process linked with a multitude of signaling pathways, which regulate promoters and inhibitors of osteogenesis, the balance of which become deregulated in disease conditions. SIRT1, a protein deacetylase, known to be protective in inhibiting oxidative stress and inflammation within the vessel wall, has been shown as a possible key player in modulating the cell-fate determining canonical Wnt signaling pathways. Suppression of SIRT1 has been reported in patients suffering with cardiovascular pathologies, suggesting that the sustained acetylation of osteogenic factors could contribute to their activation and in turn, lead to the progression of calcification. There is clear evidence of the synergy between β-Catenin and elevated Runx2, and with Wnt signaling being β-Catenin dependent, further understanding is needed as to how these molecular pathways converge and interact, in order to provide novel insight into the mechanism by which smooth muscle cells switch to an osteogenic differentiation programme. Therefore, this review will describe the current concepts of pathological soft tissue mineralization, with a focus on the contribution of SIRT1 as a regulator of Wnt signaling and its targets, discussing SIRT1 as a potential target for manipulation and therapy.
Collapse
Affiliation(s)
- Francesca Bartoli-Leonard
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - Fiona L Wilkinson
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - Alex W W Langford-Smith
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - M Y Alexander
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - Ria Weston
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| |
Collapse
|
68
|
Wang A, Zhou F, Li D, Lu JJ, Wang Y, Lin L. γ-Mangostin alleviates liver fibrosis through Sirtuin 3-superoxide-high mobility group box 1 signaling axis. Toxicol Appl Pharmacol 2018; 363:142-153. [PMID: 30502394 DOI: 10.1016/j.taap.2018.11.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 12/29/2022]
Abstract
The activation of hepatic stellate cells (HSCs) plays a critical role in liver fibrosis. In the current study, γ-mangostin (γ-man), one of the major xanthones from mangosteen (Garcinia mangostana), was found to alleviate fibrogenesis in human immortalized HSCs (LX-2 cells) and in liver from chronic carbon tetrachloride (CCl4) injured mice. γ-Man suppressed the expression levels of collagen I and α-smooth muscle actin (α-SMA) in LX-2 cells in both dose and time dependent manners. Furthermore, γ-man inhibited NAD(P)H oxidase activity through induction of sirtuin 3 (SIRT3), resulting in reduced intracellular oxidative stress in LX-2 cells. Moreover, γ-man stimulated the expression of histone deacetylase 1, which in turn decreased the acetylation and cytoplasmic shuttling of high mobility group box 1 (HMGB1), to impair autocrine HMGB1-induced HSC activation. In CCl4-injured mice, γ-man enhanced the expression of SIRT3 and decreased the expression of HMGB1, resulting in decreased accumulation of collagen I and α-SMA in liver. Consequently, γ-man might be a potent candidate to treat oxidative stress induced liver fibrosis.
Collapse
Affiliation(s)
- Anqi Wang
- Guangdong-Macau Traditional Chinese Medicine Technology Industrial Park Development Co., Ltd, Hengqin New Area, Zhuhai, Guangdong 519031, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Fayang Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Dan Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Ligen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
69
|
Li L, Chen Z, Fu W, Cai S, Zeng Z. Emerging Evidence concerning the Role of Sirtuins in Sepsis. Crit Care Res Pract 2018; 2018:5489571. [PMID: 30533222 PMCID: PMC6250024 DOI: 10.1155/2018/5489571] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 10/18/2018] [Indexed: 12/29/2022] Open
Abstract
Sepsis, a dysregulated host response to infection, is a major public health concern. Though experimental and clinical studies relating to sepsis are increasing, the mechanism of sepsis is not completely understood. To date, numerous studies have shown that sirtuins (silent mating type information regulation 2 homolog), which belong to the class III histone deacetylases, may have a varied, or even opposite, effect in the pathogenesis of sepsis. Notably, downstream mechanisms of sirtuins are not fully understood. The sirtuin family consists of sirtuins 1-7; among them, sirtuin 1 (SIRT1) is the most studied one, during the development of sepsis. Furthermore, other sirtuin members are also confirmed to be involved in the regulation of inflammatory or metabolic signaling following sepsis. In addition, sirtuins may interact with each other to form a precise regulatory mechanism in different phases of sepsis. Therefore, in this review, by accumulating data from PubMed, we intend to explain the role of sirtuin in sepsis, which we hope will pave the way for further experimental study and the potential future clinical applications of sirtuins.
Collapse
Affiliation(s)
- Lulan Li
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, Guangdong, China
| | - Zhongqing Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, Guangdong, China
| | - Weijun Fu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, Guangdong, China
| | - Shumin Cai
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, Guangdong, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, Guangdong, China
| |
Collapse
|
70
|
Hardeland R. Melatonin and inflammation-Story of a double-edged blade. J Pineal Res 2018; 65:e12525. [PMID: 30242884 DOI: 10.1111/jpi.12525] [Citation(s) in RCA: 312] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 09/10/2018] [Accepted: 09/14/2018] [Indexed: 12/13/2022]
Abstract
Melatonin is an immune modulator that displays both pro- and anti-inflammatory properties. Proinflammatory actions, which are well documented by many studies in isolated cells or leukocyte-derived cell lines, can be assumed to enhance the resistance against pathogens. However, they can be detrimental in autoimmune diseases. Anti-inflammatory actions are of particular medicinal interest, because they are observed in high-grade inflammation such as sepsis, ischemia/reperfusion, and brain injury, and also in low-grade inflammation during aging and in neurodegenerative diseases. The mechanisms contributing to anti-inflammatory effects are manifold and comprise various pathways of secondary signaling. These include numerous antioxidant effects, downregulation of inducible and inhibition of neuronal NO synthases, downregulation of cyclooxygenase-2, inhibition of high-mobility group box-1 signaling and toll-like receptor-4 activation, prevention of inflammasome NLRP3 activation, inhibition of NF-κB activation and upregulation of nuclear factor erythroid 2-related factor 2 (Nrf2). These effects are also reflected by downregulation of proinflammatory and upregulation of anti-inflammatory cytokines. Proinflammatory actions of amyloid-β peptides are reduced by enhancing α-secretase and inhibition of β- and γ-secretases. A particular role in melatonin's actions seems to be associated with the upregulation of sirtuin-1 (SIRT1), which shares various effects known from melatonin and additionally interferes with the signaling by the mechanistic target of rapamycin (mTOR) and Notch, and reduces the expression of the proinflammatory lncRNA-CCL2. The conclusion on a partial mediation by SIRT1 is supported by repeatedly observed inhibitions of melatonin effects by sirtuin inhibitors or knockdown.
Collapse
Affiliation(s)
- Rüdiger Hardeland
- Johann Friedrich Blumenbach Institute of Zoology and Anthropology, University of Göttingen, Göttingen, Germany
| |
Collapse
|
71
|
Wei S, Gao Y, Dai X, Fu W, Cai S, Fang H, Zeng Z, Chen Z. SIRT1-mediated HMGB1 deacetylation suppresses sepsis-associated acute kidney injury. Am J Physiol Renal Physiol 2018; 316:F20-F31. [PMID: 30379096 DOI: 10.1152/ajprenal.00119.2018] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Sepsis is the leading cause of death in the intensive care unit and continues to lack effective treatment. It is widely accepted that high-mobility group box 1 (HMGB1) is a key inflammatory mediator in the pathogenesis of sepsis. Moreover, some studies indicate that the functions of HMGB1 depend on its molecular localization and posttranslational modifications. Our previous study confirms that sirtuin 1, silent information regulator 2-related enzyme 1 (SIRT1), a type III deacetylase, can ameliorate sepsis-associated acute kidney injury (SA-AKI). We explored the effect and mechanism of SIRT1 on HMGB1 using a mouse model of cecal ligation and puncture-induced sepsis and LPS-treated human kidney (HK-2) cell line. We found that HMGB1 is elevated in the serum but is gradually reduced in kidney cells in the later stages of septic mice. The acetylation modification of HMGB1 is a key process before its nucleus-to-cytoplasm translocation and extracellular secretion in kidney cells, accelerating the development of SA-AKI. Moreover, SIRT1 can physically interact with HMGB1 at the deacetylated lysine sites K28, K29, and K30, subsequently suppressing downstream inflammatory signaling. Thus the SIRT1-HMGB1 signaling pathway is a crucial mechanism in the development of SA-AKI and presents a novel experimental perspective for future SA-AKI research.
Collapse
Affiliation(s)
- Siwei Wei
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Baiyun District, Guangzhou, Guangdong , China.,Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Baiyun District, Guangzhou, Guangdong , China
| | - Youguang Gao
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University , Fuzhou, Fujian , China
| | - Xingui Dai
- The First People's Hospital of Chenzhou, Department of Critical Care Medicine, Institute of Translational Medicine, Chenzhou, Hunan , China
| | - Weijun Fu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Baiyun District, Guangzhou, Guangdong , China
| | - Shumin Cai
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Baiyun District, Guangzhou, Guangdong , China
| | - Haihong Fang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Baiyun District, Guangzhou, Guangdong , China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Baiyun District, Guangzhou, Guangdong , China.,Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Baiyun District, Guangzhou, Guangdong , China
| | - Zhongqing Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Baiyun District, Guangzhou, Guangdong , China.,Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Baiyun District, Guangzhou, Guangdong , China
| |
Collapse
|
72
|
Andersson U, Yang H, Harris H. High-mobility group box 1 protein (HMGB1) operates as an alarmin outside as well as inside cells. Semin Immunol 2018. [DOI: 10.1016/j.smim.2018.02.011] [Citation(s) in RCA: 223] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
73
|
Wang B, Lian YJ, Su WJ, Peng W, Dong X, Liu LL, Gong H, Zhang T, Jiang CL, Wang YX. HMGB1 mediates depressive behavior induced by chronic stress through activating the kynurenine pathway. Brain Behav Immun 2018; 72:51-60. [PMID: 29195782 DOI: 10.1016/j.bbi.2017.11.017] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 11/21/2017] [Accepted: 11/22/2017] [Indexed: 12/20/2022] Open
Abstract
Our previous study has reported that the proactive secretion and role of central high mobility group box 1 (HMGB1) in lipopolysaccharide-induced depressive behavior. Here, the potential mechanism of HMGB1 mediating chronic-stress-induced depression through the kynurenine pathway (KP) was further explored both in vivo and in vitro. Depression model was established with the 4-week chronic unpredictable mild stress (CUMS). Sucrose preference and Barnes maze test were performed to reflect depressive behaviors. The ratio of kynurenine (KYN)/tryptophan (Trp) represented the enzyme activity of indoleamine-2,3-dioxygenase (IDO). Gene transcription and protein expression were assayed by real-time RT-PCR and western-blot or ELISA kit respectively. Along with depressive behaviors, HMGB1 concentrations in the hippocampus and serum substantially increased post 4-week CUMS exposure. Concurrent with the upregulated HMGB1 protein, the regulator of translocation of HMGB1, sirtuin 1 (SIRT1) concentration in the hippocampus remarkably increased. In addition to HMGB1 and SIRT1, IDO, the rate limiting enzyme of KP, was upregulated at the level of mRNA expression and enzyme activity in stressed hippocampi and LPS/HMGB1-treated hippocampal slices. The gene transcription of kynurenine monooxygenase (KMO) and kynureninase (KYNU) in the downstream of KP also increased both in vivo and in vitro. Mice treated with ethyl pyruvate (EP), the inhibitor of HMGB1 releasing, were observed with lower tendency of developing depressive behaviors and reduced activation of enzymes in KP. All of these experiments demonstrate that the role of HMGB1 on the induction of depressive behavior is mediated by KP activation.
Collapse
Affiliation(s)
- Bo Wang
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Yong-Jie Lian
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Wen-Jun Su
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Wei Peng
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China; Department of Psychiatry, The 92nd Hospital of PLA, Nanping 353000, PR China
| | - Xin Dong
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Lin-Lin Liu
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China; Department of Nursing, The 474th Hospital of PLA, Urumqi 830012, PR China
| | - Hong Gong
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Ting Zhang
- Department of Navy Medicine, Second Military Medical University, Shanghai 200433, PR China
| | - Chun-Lei Jiang
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Yun-Xia Wang
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China.
| |
Collapse
|
74
|
Hong G, Zheng D, Zhang L, Ni R, Wang G, Fan GC, Lu Z, Peng T. Administration of nicotinamide riboside prevents oxidative stress and organ injury in sepsis. Free Radic Biol Med 2018; 123:125-137. [PMID: 29803807 PMCID: PMC6236680 DOI: 10.1016/j.freeradbiomed.2018.05.073] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 05/01/2018] [Accepted: 05/19/2018] [Indexed: 01/11/2023]
Abstract
AIMS Sepsis-caused multiple organ failure remains the major cause of morbidity and mortality in intensive care units. Nicotinamide riboside (NR) is a precursor of nicotinamide adenine dinucleotide (NAD+), which is important in regulating oxidative stress. This study investigated whether administration of NR prevented oxidative stress and organ injury in sepsis. METHODS Mouse sepsis models were induced by injection of lipopolysaccharides (LPS) or feces-injection-in-peritoneum. NR was given before sepsis onset. Cultured macrophages and endothelial cells were incubated with various agents. RESULTS Administration of NR elevated the NAD+ levels, and elicited a reduction of oxidative stress, inflammation and caspase-3 activity in lung and heart tissues, which correlated with attenuation of pulmonary microvascular permeability and myocardial dysfunction, leading to less mortality in sepsis models. These protective effects of NR were associated with decreased levels of plasma high mobility group box-1 (HMGB1) in septic mice. Consistently, pre-treatment of macrophages with NR increased NAD+ content and reduced HMGB1 release upon LPS stimulation. NR also prevented reactive oxygen species (ROS) production and apoptosis in endothelial cells induced by a conditioned-medium collected from LPS-treated macrophages. Furthermore, inhibition of SIRT1 by EX527 offset the negative effects of NR on HMGB1 release in macrophages, and ROS and apoptosis in endothelial cells. CONCLUSIONS Administration of NR prevents lung and heart injury, and improves the survival in sepsis, likely by inhibiting HMGB1 release and oxidative stress via the NAD+/SIRT1 signaling. Given NR has been used as a health supplement, it may be a useful agent to prevent organ injury in sepsis.
Collapse
Affiliation(s)
- Guangliang Hong
- Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada N6A 4G5; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada N6A 4G5
| | - Dong Zheng
- Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada N6A 4G5; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada N6A 4G5; Department of Medicine, Western University, London, Ontario, Canada N6A 4G5; Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Lulu Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Rui Ni
- Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada N6A 4G5; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada N6A 4G5
| | - Grace Wang
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Zhongqiu Lu
- Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Tianqing Peng
- Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada N6A 4G5; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada N6A 4G5; Department of Medicine, Western University, London, Ontario, Canada N6A 4G5; Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China.
| |
Collapse
|
75
|
Zhou S, Lu H, Chen R, Tian Y, Jiang Y, Zhang S, Ni D, Su Z, Shao X. Angiotensin II enhances the acetylation and release of HMGB1 in RAW264.7 macrophage. Cell Biol Int 2018; 42:1160-1169. [PMID: 29741224 DOI: 10.1002/cbin.10984] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 05/05/2018] [Indexed: 12/14/2022]
Abstract
The high-mobility group box-1 (HMGB1), as a highly conserved ubiquitous DNA-binding protein, has been widely studied in various diseases, including inflammation and tumor; however, fewer studies were focused on the mechanisms controlling HMGB1 release compared with the function of HMGB1. Previous studies have proven that ANG II can act as a pro-inflammatory cytokine, both of HMGB1 and ANG II were significantly upregulated in autoimmune diseases; however, the exact role of ANG II in regulating HMGB1 release have not been shown. The present study was to define the effects of ANG II on macrophages and the possible mechanisms in controlling HMGB1 release. Our results showed that ANG II can induce M1 macrophage polarization through upregulated the expression of HMGB1 and caused acetylation of HMGB1 and release via its dissociation from SIRT1, which in a positive feedback upregulates ANG II. Subsequently, HMGB1 inhibitors can reduce the ANG II-elicited polarize of macrophage. Meanwhile, we show that JAK/STAT pathways play an essential role in ANG II-induced HMGB1 nuclear translocation, JAK/STAT specific inhibitors can inhibit ANG II-induced HMGB1 expression. Taken together, our results provide a novel evidence that HMGB1 play a critical role in ANG II mediated macrophage polarization, and we suggest that ANG II mediated HMGB1 release via dissociation from SIRT1, induce hyperacetylation of HMGB1, thus for subsequent release, suggesting that the angiotensin II receptor antagonist is a potential drug target for inhibiting HMGB1 release in inflammation diseases.
Collapse
Affiliation(s)
- Shanshan Zhou
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Hongxiang Lu
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Rong Chen
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Yu Tian
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - YuanYuan Jiang
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Shiqing Zhang
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Daobing Ni
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Zhaoliang Su
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Xiaoyi Shao
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China.,Department of Immunology, Medical College, Nantong University, Nantong, Jiangsu, 226001, China
| |
Collapse
|
76
|
Sun Y, Chen H, Dai J, Wan Z, Xiong P, Xu Y, Han Z, Chai W, Gong F, Zheng F. Glycyrrhizin Protects Mice Against Experimental Autoimmune Encephalomyelitis by Inhibiting High-Mobility Group Box 1 (HMGB1) Expression and Neuronal HMGB1 Release. Front Immunol 2018; 9:1518. [PMID: 30013568 PMCID: PMC6036111 DOI: 10.3389/fimmu.2018.01518] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 06/19/2018] [Indexed: 12/12/2022] Open
Abstract
The inflammatory mediator high-mobility group box 1 (HMGB1) plays a critical role in the pathogenesis of human multiple sclerosis (MS) and mouse experimental autoimmune encephalomyelitis (EAE). Glycyrrhizin (GL), a glycoconjugated triterpene extracted from licorice root, has the ability to inhibit the functions of HMGB1; however, GL’s function against EAE has not been thoroughly characterized to date. To determine the benefit of GL as a modulator of neuroinflammation, we used an in vivo study to examine GL’s effect on EAE along with primary cultured cortical neurons to study the GL effect on HMGB1 release. Treatment of EAE mice with GL from onset to the peak stage of disease resulted in marked attenuation of EAE severity, reduced inflammatory cell infiltration and demyelination, decreased tumor necrosis factor-alpha (TNF-α), IFN-γ, IL-17A, IL-6, and transforming growth factor-beta 1, and increased IL-4 both in serum and spinal cord homogenate. Moreover, HMGB1 levels in different body fluids were reduced, accompanied by a decrease in neuronal damage, activated astrocytes and microglia, as well as HMGB1-positive astrocytes and microglia. GL significantly reversed HMGB1 release into the medium induced by TNF-α stimulation in primary cultured cortical neurons. Taken together, the results indicate that GL has a strong neuroprotective effect on EAE mice by reducing HMGB1 expression and release and thus can be used to treat central nervous system inflammatory diseases, such as MS.
Collapse
Affiliation(s)
- Yan Sun
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, China.,Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Neurobiology, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Huoying Chen
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Laboratory Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jiapei Dai
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, China.,Department of Neurobiology, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Zhongjun Wan
- Department of Neurobiology, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Ping Xiong
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Xu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengrong Han
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, China.,Department of Neurobiology, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Weitai Chai
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, China.,Department of Neurobiology, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Feili Gong
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Wuhan, China.,NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
77
|
Mass Spectrometry-based Structural Analysis and Systems Immunoproteomics Strategies for Deciphering the Host Response to Endotoxin. J Mol Biol 2018; 430:2641-2660. [PMID: 29949751 DOI: 10.1016/j.jmb.2018.06.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/23/2018] [Accepted: 06/15/2018] [Indexed: 02/06/2023]
Abstract
One cause of sepsis is systemic maladaptive immune response of the host to bacteria and specifically, to Gram-negative bacterial outer-membrane glycolipid lipopolysaccharide (LPS). On the host myeloid cell surface, proinflammatory LPS activates the innate immune system via Toll-like receptor-4/myeloid differentiation factor-2 complex. Intracellularly, LPS is also sensed by the noncanonical inflammasome through caspase-11 in mice and 4/5 in humans. The minimal functional determinant for innate immune activation is the membrane anchor of LPS called lipid A. Even subtle modifications to the lipid A scaffold can enable, diminish, or abolish immune activation. Bacteria are known to modify their LPS structure during environmental stress and infection of hosts to alter cellular immune phenotypes. In this review, we describe how mass spectrometry-based structural analysis of endotoxin helped uncover major determinations of molecular pathogenesis. Through characterization of LPS modifications, we now better understand resistance to antibiotics and cationic antimicrobial peptides, as well as how the environment impacts overall endotoxin structure. In addition, mass spectrometry-based systems immunoproteomics approaches can assist in elucidating the immune response against LPS. Many regulatory proteins have been characterized through proteomics and global/targeted analysis of protein modifications, enabling the discovery and characterization of novel endotoxin-mediated protein translational modifications.
Collapse
|
78
|
Park EJ, Kim YM, Kim HJ, Chang KC. Degradation of histone deacetylase 4 via the TLR4/JAK/STAT1 signaling pathway promotes the acetylation of high mobility group box 1 (HMGB1) in lipopolysaccharide-activated macrophages. FEBS Open Bio 2018; 8:1119-1126. [PMID: 29988587 PMCID: PMC6026695 DOI: 10.1002/2211-5463.12456] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/16/2018] [Accepted: 05/17/2018] [Indexed: 02/06/2023] Open
Abstract
High mobility group box 1 (HMGB1) has been proposed as crucial in the pathogenesis of many diseases including sepsis. Acetylation of HMGB1 prevents its entry into the nucleus and leads to its secretion from the cell where it can trigger inflammation. We hypothesized that histone deacetylase 4 (HDAC4) controls the acetylation of HMGB1 in lipopolysaccharide (LPS)‐stimulated RAW264.7 cells via the janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway. The results showed that LPS treatment promoted the degradation of HDAC4 in a proteasome‐dependent manner, which led to HMGB1 acetylation. In LPS‐activated RAW264.7 cells, treatment with TAK‐242 (a toll like receptor 4 inhibitor) and pyridone 6 (a JAK inhibitor) significantly inhibited HDAC4 degradation and acetylation of HMGB1, and thus prevented secretion of HMGB1. Decreased phosphorylation of STAT1 was also observed. Interestingly, HDAC4 overexpression significantly prevented the acetylation and secretion of HMGB1 in both RAW264.7 cells and isolated murine peritoneal macrophages. We conclude that HDAC4 might be a useful target for the treatment of sepsis.
Collapse
Affiliation(s)
- Eun J Park
- Department of Pharmacology Institute of Health Sciences College of Medicine Gyeongsang National University Jinju Korea
| | - Young M Kim
- Department of Pharmacology Institute of Health Sciences College of Medicine Gyeongsang National University Jinju Korea
| | - Hye J Kim
- Department of Pharmacology Institute of Health Sciences College of Medicine Gyeongsang National University Jinju Korea
| | - Ki C Chang
- Department of Pharmacology Institute of Health Sciences College of Medicine Gyeongsang National University Jinju Korea
| |
Collapse
|
79
|
Kim YM, Park EJ, Kim HJ, Chang KC. Sirt1 S-nitrosylation induces acetylation of HMGB1 in LPS-activated RAW264.7 cells and endotoxemic mice. Biochem Biophys Res Commun 2018; 501:73-79. [PMID: 29680657 DOI: 10.1016/j.bbrc.2018.04.155] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 04/18/2018] [Indexed: 11/29/2022]
Abstract
Excessive inflammation plays a detrimental role in endotoxemia. A recent study indicated that alarmins such as high mobility group box 1 (HMGB1) have drawn attention as therapeutic targets of sepsis. Post-translational modification (i.e., acetylation of lysine residues) of HMGB1 leads to the release of HMGB1 into the cellular space, operating as a warning signal that induces inflammation. Sirtuin 1 (SIRT1) has been shown to negatively regulate HMGB1 hyperacetylation and its extracellular release in sepsis. Therefore, we hypothesized that the S-nitrosylation (SNO) of SIRT1 may disrupt the ability of SIRT1 to negatively regulate the hyperacetylation of HMGB1. As long as the S-nitrosylation of SIRT1 occurs during septic conditions, it may worsen the situation. We found that the activity of SIRT1 decreased as the SNO-SIRT1 levels increased, resulting in HMGB1 release by LPS in RAW264.7 cells. Both the iNOS inhibitor (1400 W) and silencing iNOS significantly inhibited SNO-SIRT1, allowing increases in SIRT1 activity that decreased the HMGB1 release by LPS. SNAP, a NO donor, significantly increased both SNO-SIRT1 levels and the HMGB1 release that was accompanied by decreased sirt1 activity. However, sirtinol, a Sirt1 inhibitor, by itself decreased Sirt1 activity compared to that of the control, so that it did not affect already increased SNO-SIRT levels by SNAP. Most importantly, in lung tissues of LPS-endotoxic mice, significantly increased levels of SNO-SIRT were found, which was inhibited by 1400 W treatment. Plasma nitrite and HMGB1 levels were significantly higher than those in the sham controls, and the elevated levels were significantly lowered in the presence of 1400 W. We concluded that the S-nitrosylation of Sirt1 under endotoxic conditions may uninhibit the acetylation of HMGB1 and its extracellular release.
Collapse
Affiliation(s)
- Young Min Kim
- Department of Pharmacology, College of Medicine Gyeongsang National University and Institute of Health Sciences, Jinju 660-290, Republic of Korea
| | - Eun Jung Park
- Department of Pharmacology, College of Medicine Gyeongsang National University and Institute of Health Sciences, Jinju 660-290, Republic of Korea
| | - Hye Jung Kim
- Department of Pharmacology, College of Medicine Gyeongsang National University and Institute of Health Sciences, Jinju 660-290, Republic of Korea.
| | - Ki Churl Chang
- Department of Pharmacology, College of Medicine Gyeongsang National University and Institute of Health Sciences, Jinju 660-290, Republic of Korea.
| |
Collapse
|
80
|
Yuan Y, Liu Q, Zhao J, Tang H, Sun J. SIRT1 attenuates murine allergic rhinitis by downregulated HMGB 1/TLR4 pathway. Scand J Immunol 2018; 87:e12667. [PMID: 29701897 DOI: 10.1111/sji.12667] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 04/19/2018] [Indexed: 01/11/2023]
Affiliation(s)
- Y. Yuan
- Department of Otolaryngology; Qingdao Municipal Hospital; Qingdao China
| | - Q. Liu
- Department of Otolaryngology; Qingdao Municipal Hospital; Qingdao China
| | - J. Zhao
- Department of Out-patient; Qingdao Municipal Hospital; Qingdao China
| | - H. Tang
- Department of Respiratory; Qingdao Municipal Hospital (Eastern Campus); Qingdao China
| | - J. Sun
- International Clinic; Qingdao Municipal Hospital (Eastern Campus); Qingdao China
| |
Collapse
|
81
|
Park IA, Heo SH, Song IH, Kim YA, Park HS, Bang WS, Park SY, Jo JH, Lee HJ, Gong G. Endoplasmic reticulum stress induces secretion of high-mobility group proteins and is associated with tumor-infiltrating lymphocytes in triple-negative breast cancer. Oncotarget 2018; 7:59957-59964. [PMID: 27494867 PMCID: PMC5312361 DOI: 10.18632/oncotarget.11010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 07/19/2016] [Indexed: 11/25/2022] Open
Abstract
Background Although the prognostic and predictive significance of tumor-infiltrating lymphocytes (TILs) in triple-negative breast cancer (TNBC) have been shown, the cause of the TIL influx is unclear. Here, we investigated whether extracellular secretion of HMGN1 is associated with TIL influx, as well as increased endoplasmic reticulum stress (ERS), in human TNBC. Methods We reviewed the slides of 767 patients with TNBC and evaluated the TIL levels. We also assessed the expression of HMGs and several ERS-associated molecules using immunohistochemical staining. Western blot analysis of human TNBC cell lines and pharmacological ERS inducers was used to determine if HMGN1 migrates from the nucleus to the extracellular space in response to ERS. Results On immunohistochemical staining, either higher nuclear or cytoplasmic expression of both HMGB1 and HMGN1 was significantly associated with ERS. TILs showed a positive correlation with the cytoplasmic expression of the HMGs. Western blot analysis of TNBC cell lines showed that ERS induction resulted in the secretion of HMG proteins. Conclusions This is the first study to elucidate the associations among ERS, secretion of HMGs, and degree of TILs in TNBCs. Understanding the mechanisms of TIL influx will help in the development of effective immunotherapeutic agents for TNBC.
Collapse
Affiliation(s)
- In Ah Park
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Sun-Hee Heo
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.,Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - In Hye Song
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Young-Ae Kim
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.,Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Hye Seon Park
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.,Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Won Seon Bang
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.,Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Suk Young Park
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.,Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jeong-Hyon Jo
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hee Jin Lee
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Gyungyub Gong
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
82
|
Vogel S, Thein SL. Platelets at the crossroads of thrombosis, inflammation and haemolysis. Br J Haematol 2018; 180:761-767. [PMID: 29383704 DOI: 10.1111/bjh.15117] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Platelets play a critical role at the interphase of thrombosis and inflammation, key features in haemolysis-associated disorders. Exercising this role requires expression of pattern recognition receptors by platelets, including toll-like receptor 4 (TLR4) and nucleotide-binding domain leucine rich repeat containing protein 3 (NLRP3), the latter forming intraplatelet multiprotein inflammasome complexes. Platelets are a potential target of various damage-associated molecular pattern (DAMP) molecules, such as free haem, a degradation by-product of haemoglobin oxidation during haemolysis, and high-mobility group box 1 (HMGB1), a DNA-binding protein released by dying or stressed cells and activated platelets. We have recently identified platelet TLR4, NLRP3, and Bruton tyrosine kinase (BTK) as critical regulators of platelet aggregation and thrombus formation, suggesting that the BTK inhibitor ibrutinib is a potential therapeutic target. Increasing evidence suggests that these and other DAMP-driven signalling mechanisms employed by platelets might be key in mediating inflammation and thrombosis encountered in haemolytic disorders. However, the precise regulatory triggers and their clinical relevance are poorly understood. We provide new insights into these less-well characterised platelet mechanisms, which are potentially targetable in haemolytic disorders.
Collapse
Affiliation(s)
- Sebastian Vogel
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Swee Lay Thein
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
83
|
Hwang JS, Kang ES, Han SG, Lim DS, Paek KS, Lee CH, Seo HG. Formononetin inhibits lipopolysaccharide-induced release of high mobility group box 1 by upregulating SIRT1 in a PPARδ-dependent manner. PeerJ 2018; 6:e4208. [PMID: 29312829 PMCID: PMC5756453 DOI: 10.7717/peerj.4208] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/09/2017] [Indexed: 12/27/2022] Open
Abstract
Background The release of high mobility group box 1 (HMGB1) induced by inflammatory signals acts as a cellular alarmin to trigger a chain of inflammatory responses. Although the inflammatory actions of HMGB1 are well studied, less is known about the therapeutic agents that can impede its release. This study investigated whether the isoflavonoid formononetin can modulate HMGB1 release in cellular inflammatory responses. Methods RAW264.7 murine macrophages were exposed to lipopolysaccharide (LPS) in the presence or absence of formononetin. The levels of HMGB1 release, sirtuin 1 (SIRT1) expression, and HMGB1 acetylation were analyzed by immunoblotting and real-time polymerase chain reaction. The effects of resveratrol and sirtinol, an activator and inhibitor of SIRT1, respectively, on LPS-induced HMGB1 release were also evaluated. Results Formononetin modulated cellular inflammatory responses by suppressing the release of HMGB1 by macrophages exposed to LPS. In RAW264.7 cells, formononetin significantly attenuated LPS-induced release of HMGB1 into the extracellular environment, which was accompanied by a reduction in its translocation from the nucleus to the cytoplasm. In addition, formononetin significantly induced mRNA and protein expression of SIRT1 in a peroxisome proliferator-activated receptor δ (PPARδ)-dependent manner. These effects of formononetin were dramatically attenuated in cells treated with small interfering RNA (siRNA) against PPARδ or with GSK0660, a specific inhibitor of PPARδ, indicating that PPARδ is involved in formononetin-mediated SIRT1 expression. In line with these effects, formononetin-mediated inhibition of HMGB1 release in LPS-treated cells was reversed by treatment with SIRT1-targeting siRNA or sirtinol, a SIRT1 inhibitor. By contrast, resveratrol, a SIRT1 activator, further potentiated the inhibitory effect of formononetin on LPS-induced HMGB1 release, revealing a possible mechanism by which formononetin regulates HMGB1 release through SIRT1. Furthermore, modulation of SIRT1 expression by transfection of SIRT1- or PPARδ-targeting siRNA significantly counteracted the inhibitory effects of formononetin on LPS-induced HMGB1 acetylation, which was responsible for HMGB1 release. Discussion This study shows for the first time that formononetin inhibits HMGB1 release by decreasing HMGB1 acetylation via upregulating SIRT1 in a PPARδ-dependent manner. Formononetin consequently exhibits anti-inflammatory activity. Identification of agents, such as formononetin, which can block HMGB1 release, may help to treat inflammation-related disorders.
Collapse
Affiliation(s)
- Jung Seok Hwang
- Department of Food Science and Biotechnology of Animal Products, Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Eun Sil Kang
- Department of Food Science and Biotechnology of Animal Products, Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Sung Gu Han
- Department of Food Science and Biotechnology of Animal Products, Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Dae-Seog Lim
- Department of Biotechnology, CHA University, Seongnam, Korea
| | | | - Chi-Ho Lee
- Department of Food Science and Biotechnology of Animal Products, Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| | - Han Geuk Seo
- Department of Food Science and Biotechnology of Animal Products, Sanghuh College of Life Sciences, Konkuk University, Seoul, Korea
| |
Collapse
|
84
|
Salidroside Inhibits HMGB1 Acetylation and Release through Upregulation of SirT1 during Inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9821543. [PMID: 29333216 PMCID: PMC5733170 DOI: 10.1155/2017/9821543] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/25/2017] [Accepted: 07/25/2017] [Indexed: 12/24/2022]
Abstract
HMGB1, a highly conserved nonhistone DNA-binding protein, plays an important role in inflammatory diseases. Once released to the extracellular space, HMGB1 acts as a proinflammatory cytokine that triggers inflammatory reaction. Our previous study showed that salidroside exerts anti-inflammatory effect via inhibiting the JAK2-STAT3 signalling pathway. However, whether salidroside inhibits the release of HMGB1 is still unclear. In this study, we aim to study the effects of salidroside on HMGB1 release and then investigate the potential molecular mechanisms. In an experimental rat model of sepsis caused by CLP, salidroside administration significantly attenuated lung injury and reduced the serum HMGB1 level. In RAW264.7 cells, we investigated the effects of salidroside on LPS-induced HMGB1 release and then explored the underlying molecular mechanisms. We found that salidroside significantly inhibited LPS-induced HMGB1 release, and the inhibitory effect was correlated with the HMGB1 acetylation levels. Mechanismly, salidroside inhibits HMGB1 acetylation through the AMPK-SirT1 pathway. In addition, SirT1 overexpression attenuated LPS-induced HMGB1 acetylation and nucleocytoplasmic translocation. Furthermore, in SirT1 shRNA plasmid-transfected cells, salidroside treatment enhanced SirT1 expression and reduced LPS-activated HMGB1 acetylation and nucleocytoplasmic translocation. Collectively, these results demonstrated that salidroside might reduce HMGB1 release through the AMPK-SirT1 signalling pathway and suppress HMGB1 acetylation and nucleocytoplasmic translocation.
Collapse
|
85
|
Lan KC, Chao SC, Wu HY, Chiang CL, Wang CC, Liu SH, Weng TI. Salidroside ameliorates sepsis-induced acute lung injury and mortality via downregulating NF-κB and HMGB1 pathways through the upregulation of SIRT1. Sci Rep 2017; 7:12026. [PMID: 28931916 PMCID: PMC5607272 DOI: 10.1038/s41598-017-12285-8] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 09/06/2017] [Indexed: 12/29/2022] Open
Abstract
Sepsis is a life-threatening medical condition. Salidroside, a substance isolated from Rhodiola rosea, possesses antioxidant and anti-inflammatory properties. The effect and mechanism of salidroside on sepsis-induced acute lung injury still remains to be well clarified. Here, we investigated the effect and mechanism of salidroside on septic mouse models and explored the role of salidroside-upregulated SIRT1. Salidroside inhibited the inflammatory responses and HMGB1 productions in bacterial lipopolysaccharide (LPS)-treated macrophages and mice. Salidroside could also reverse the decreased SIRT1 protein expression in LPS-treated macrophages and mice. Salidroside also alleviated the sepsis-induced lung edema, lipid peroxidation, and histopathological changes and the mortality, and improved the lung PaO2/FiO2 ratio in cecal ligation and puncture (CLP)-induced septic mice. Salidroside significantly decreased the serum TNF-α, IL-6, NO, and HMGB1 productions, pulmonary inducible NO synthase (iNOS) and phosphorylated NF-κB-p65 protein expressions, and pulmonary HMGB1 nuclear translocation in CLP septic mice. Moreover, sepsis decreased the SIRT1 protein expression in the lungs of CLP septic mice. Salidroside significantly upregulated the SIRT1 expression and inhibited the inflammatory responses in CLP septic mouse lungs. These results suggest that salidroside protects against sepsis-induced acute lung injury and mortality, which might be through the SIRT1-mediated repression of NF-κB activation and HMGB1 nucleocytoplasmic translocation.
Collapse
Affiliation(s)
- Kuo-Cheng Lan
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Sung-Chuan Chao
- Department of Surgery, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Hsiao-Yi Wu
- Department of Forensic Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Lien Chiang
- Department of Forensic Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ching-Chia Wang
- Departments of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Shing-Hwa Liu
- Departments of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.,Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Te-I Weng
- Department of Forensic Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan. .,Departments of Emergency Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
86
|
Ahn MY, Hwang JS, Lee SB, Ham SA, Hur J, Kim JT, Seo HG. Curcumin longa extract-loaded nanoemulsion improves the survival of endotoxemic mice by inhibiting nitric oxide-dependent HMGB1 release. PeerJ 2017; 5:e3808. [PMID: 28929026 PMCID: PMC5600948 DOI: 10.7717/peerj.3808] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 08/23/2017] [Indexed: 12/22/2022] Open
Abstract
Background High mobility group box 1 (HMGB1) is a well-known damage-related alarmin that participates in cellular inflammatory responses. However, the mechanisms leading to HMGB1 release in inflammatory conditions and the therapeutic agents that could prevent it remain poorly understood. This study attempted to examine whether the Curcumin longa herb, which is known to have anti-inflammatory property, can modulate cellular inflammatory responses by regulating HMGB1 release. Methods The murine macrophage RAW264.7 cells were treated with lipopolysaccharide (LPS) and/or a C. longa extract-loaded nanoemulsion (CLEN). The levels of released HMGB1, nitric oxide (NO) production, inducible NO synthase (iNOS) expression, and phosphorylation of mitogen-activated protein kinases were analyzed in RAW264.7 macrophages. The effects of CLEN on survival of endotoxemic model mice, circulating HMGB1 levels, and tissue iNOS expression were also evaluated. Results We have shown that a nanoemulsion loaded with an extract from the C. longa rhizome regulates cellular inflammatory responses and LPS-induced systemic inflammation by suppressing the release of HMGB1 by macrophages. First, treatment of RAW264.7 macrophages with the nanoemulsion significantly attenuated their LPS-induced release of HMGB1: this effect was mediated by inhibiting c-Jun N-terminal kinase activation, which in turn suppressed the NO production and iNOS expression of the cells. The nanoemulsion did not affect LPS-induced p38 or extracellular signal-regulated kinase activation. Second, intraperitoneal administration of the nanoemulsion improved the survival rate of LPS-injected endotoxemic mice. This associated with marked reductions in circulating HMGB1 levels and tissue iNOS expression. Discussion The present study shows for the first time the mechanism by which C. longa ameliorates sepsis, namely, by suppressing NO signaling and thereby inhibiting the release of the proinflammatory cytokine HMGB1. These observations suggest that identification of agents, including those in the herb C. longa, that can inhibit HMGB1 production and/or activity may aid the treatment of endotoxemia.
Collapse
Affiliation(s)
- Min Young Ahn
- Department of Food Science and Biotechnology of Animal Products, Konkuk University, Seoul, South Korea
| | - Jung Seok Hwang
- Department of Food Science and Biotechnology of Animal Products, Konkuk University, Seoul, South Korea
| | - Su Bi Lee
- Department of Food Science and Technology, Keimyung University, Daegu, South Korea
| | - Sun Ah Ham
- Department of Food Science and Biotechnology of Animal Products, Konkuk University, Seoul, South Korea
| | - Jinwoo Hur
- Department of Food Science and Biotechnology of Animal Products, Konkuk University, Seoul, South Korea
| | - Jun Tae Kim
- Department of Food Science and Technology, Keimyung University, Daegu, South Korea
| | - Han Geuk Seo
- Department of Food Science and Biotechnology of Animal Products, Konkuk University, Seoul, South Korea
| |
Collapse
|
87
|
Rider P, Voronov E, Dinarello CA, Apte RN, Cohen I. Alarmins: Feel the Stress. THE JOURNAL OF IMMUNOLOGY 2017; 198:1395-1402. [PMID: 28167650 DOI: 10.4049/jimmunol.1601342] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 09/27/2016] [Indexed: 12/20/2022]
Abstract
Over the last decade, danger-associated molecular pattern molecules, or alarmins, have been recognized as signaling mediators of sterile inflammatory responses after trauma and injury. In contrast with the accepted passive release models suggested by the "danger hypothesis," it was recently shown that alarmins can also directly sense and report damage by signaling to the environment when released from live cells undergoing physiological stress, even without loss of subcellular compartmentalization. In this article, we review the involvement of alarmins such as IL-1α, IL-33, IL-16, and high-mobility group box 1 in cellular and physiological stress, and suggest a novel activity of these molecules as central initiators of sterile inflammation in response to nonlethal stress, a function we denote "stressorins." We highlight the role of posttranslational modifications of stressorins as key regulators of their activity and propose that targeted inhibition of stressorins or their modifiers could serve as attractive new anti-inflammatory treatments for a broad range of diseases.
Collapse
Affiliation(s)
- Peleg Rider
- Department of Clinical Biochemistry and Pharmacology, Ben Gurion University of the Negev, 84105 Beer-Sheva, Israel
| | - Elena Voronov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben Gurion University of the Negev, 84105 Beer-Sheva, Israel
| | | | - Ron N Apte
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben Gurion University of the Negev, 84105 Beer-Sheva, Israel
| | - Idan Cohen
- Faculty of Medicine, Galilee Medical Center, Nahariya Hospital, 22100 Nahariya, Israel
| |
Collapse
|
88
|
Bai C, Ren Y, Huang J, Zhang Y, LI L, Du G. High-mobility group Box-1 regulates acute myocardial ischemia-induced injury through the toll-like receptor 4-related pathway. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:8344-8352. [PMID: 31966685 PMCID: PMC6965424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 05/27/2017] [Indexed: 06/10/2023]
Abstract
High-mobility group box-1 (HMGB1) is a nuclear protein released by necrotic cells as a result of its interactions with several receptors, including the receptor for advanced glycation end-products (RAGE) and members of the toll-like receptor family. HMGB1 has been implicated in autoimmune diseases and hepatic and intestinal ischemia/reperfusion (I/R) injury; however, its role in myocardial ischemia-induced injury remains unclear. In this study, isoproterenol (ISO) was used to establish a myocardial ischemia mouse model. Treating mice with recombinant HMGB1 (rHMGB1) worsened myocardial injury, whereas treating mice with antibodies that neutralized HMGB1 significantly reduced tissue damage. Interestingly, myocardial ischemia severity was not affected by rHMGB1 or HMGB1 antibody administration in toll-like receptor 4 (TLR4)-deficient mice (TLR4-/-), which demonstrated significantly reduced ischemia-induced cardiac tissue damage compared with wild-type (WT) mice. HMGB1 plays an important role in myocardial ischemia-induced injury by binding to TLR4, which results in proinflammatory pathway activation and enhanced myocardial injury. Therefore, blocking HMGB1 or TLR4 may represent a novel therapeutic strategy for treating myocardial ischemia-induced injury.
Collapse
Affiliation(s)
- Chaochao Bai
- Department of Cardiology, First Affiliated Hospital, School of Medicine, Shihezi UniversityShihezi, Xinjiang, China
| | - Yun Ren
- The Fifth People’s Hospital of FoshanFoshan, China
| | - Jin Huang
- Department of Cardiology, First Affiliated Hospital, School of Medicine, Shihezi UniversityShihezi, Xinjiang, China
| | - Yuan Zhang
- Department of Cardiology, First Affiliated Hospital, School of Medicine, Shihezi UniversityShihezi, Xinjiang, China
| | - Lingyi LI
- Department of Cardiology, First Affiliated Hospital, School of Medicine, Shihezi UniversityShihezi, Xinjiang, China
| | - Guangsheng Du
- The Fifth People’s Hospital of FoshanFoshan, China
- Department of Cardiology, First Affiliated Hospital, School of Medicine, Shihezi UniversityShihezi, Xinjiang, China
| |
Collapse
|
89
|
Ahn MY, Hwang JS, Ham SA, Hur J, Jo Y, Lee S, Choi MJ, Han SG, Seo HG. Subcritical water-hydrolyzed fish collagen ameliorates survival of endotoxemic mice by inhibiting HMGB1 release in a HO-1-dependent manner. Biomed Pharmacother 2017; 93:923-930. [PMID: 28715873 DOI: 10.1016/j.biopha.2017.07.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/26/2017] [Accepted: 07/09/2017] [Indexed: 02/06/2023] Open
Abstract
To investigate potential mechanisms underlying the bioactivity of hydrolyzed fish collagen, we examined the anti-inflammatory actions of subcritical water-hydrolyzed fish collagen (SWFC) in lipopolysaccharide (LPS)-triggered inflammation and endotoxemia. SWFC markedly inhibited LPS-stimulated release of high mobility group box 1 (HMGB1) in murine RAW264.7 macrophages, along with decreased cytosolic translocation of HMGB1. Both the protein and mRNA levels of heme oxygenase-1 (HO-1) were significantly upregulated in SWFC-treated RAW 264.7 cells in an Nrf2-dependent manner. In line with these effects of SWFC, both HO-1 siRNA and ZnPPIX (zinc protoporphyrin IX) actually attenuated the effects of SWFC on HMGB1 release stimulated by LPS, indicating a possible mechanism by which SWFC modulates HMGB1 release through HO-1 signaling. Notably, administration of SWFC improved the survival rates of LPS-injected endotoxemic mice, in which the serum level of HMGB1 was significantly reduced. Taken together, these results indicate that the anti-inflammatory activities of SWFC are achieved by inhibiting HMGB1 release induced by LPS in a HO-1-sensitive manner.
Collapse
Affiliation(s)
- Min Young Ahn
- Department of Food Science and Biotechnology of Animal Products, Sanghuh College of Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jung Seok Hwang
- Department of Food Science and Biotechnology of Animal Products, Sanghuh College of Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sun Ah Ham
- Department of Food Science and Biotechnology of Animal Products, Sanghuh College of Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jinwoo Hur
- Department of Food Science and Biotechnology of Animal Products, Sanghuh College of Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Yeonji Jo
- Department of Food Science and Biotechnology of Animal Products, Sanghuh College of Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - SangYoon Lee
- Department of Food Science and Biotechnology of Animal Products, Sanghuh College of Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Mi-Jung Choi
- Department of Food Science and Biotechnology of Animal Products, Sanghuh College of Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sung Gu Han
- Department of Food Science and Biotechnology of Animal Products, Sanghuh College of Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Han Geuk Seo
- Department of Food Science and Biotechnology of Animal Products, Sanghuh College of Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
90
|
Zaouali MA, Panisello A, Lopez A, Folch E, Castro-Benítez C, Adam R, Roselló-Catafau J. Cross-Talk Between Sirtuin 1 and High-Mobility Box 1 in Steatotic Liver Graft Preservation. Transplant Proc 2017; 49:765-769. [PMID: 28457391 DOI: 10.1016/j.transproceed.2017.01.071] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Sirtuin 1 (SIRT1) is a nicotinamide adenine dinucleotide +-dependent histone deacetylase that regulates various pathways involved in ischemia-reperfusion injury (IRI). Moreover, high-mobility group box 1 protein (HMGB1) has also been involved in inflammatory processes during IRI. However, the roles of both SIRT1 and HMGB1 in liver preservation is poorly understood. In this communication, we evaluated the potential relationship between SIRT1 and HMGB1 in steatotic and non-steatotic liver grafts preserved in Institute Georges Lopez solution (IGL-1) preservation solution enriched or not enriched with trimetazidine (TMZ). METHODS Steatotic and non-steatotic livers were preserved in IGL-1 preservation solution (24 hours, 4°C), enriched or not enriched with TMZ (10 μmol/L), and then submitted to ex vivo reperfusion (2 hours; 37°C). Liver injury (AST/ALT) and function (bile output, vascular resistance) were evaluated. SIRT1, HMGB1, autophagy parameters (beclin-1, LC3B), PPAR-γ, and heat-shock protein (HO-1, HSP70) expression were determined by means of Western blot. Also, we assessed oxidative stress, mitochondrial damage (glutamate dehydrogenase), and TNF-α levels. RESULTS Elevated SIRT1 and enhanced autophagy were found after reperfusion in steatotic livers preserved in IGL-1+TMZ when compared with IGL-1. However, these changes were not seen in the case of non-steatotic livers. Also, HO-1 increases in the IGL-1 + TMZ group were evident only in the case of steatotic livers, whereas HSP70 and PPAR-γ protein expression were enhanced only in non-steatotic livers. All reported changes were consistent with decreased liver injury diminution, ameliorated hepatic function, and decreased TNF-α and HMGB levels. In addition, the oxidative stress and mitochondrial damage were efficiently prevented by the IGL-1 + TMZ use. CONCLUSIONS SIRT1 is associated with HMGB1 decreases and increased autophagy in steatotic livers, contributing to increased tolerance to cold IRI.
Collapse
Affiliation(s)
- M A Zaouali
- Experimental Hepatic Ischemia-Reperfusion Unit, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC-IDIBAPS, Barcelona, Spain; Research Unit of Biology and Molecular Anthropology Applied to Development and Health (UR12ES11), Faculty of Pharmacy, University of Monastir, Tunisia; High Institut of Biotechnology of Monastir, University of Monastir, Tunisia
| | - A Panisello
- Experimental Hepatic Ischemia-Reperfusion Unit, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC-IDIBAPS, Barcelona, Spain
| | - A Lopez
- Centre Hépato-Biliaire, Hôpital Universitaire Paul Brousse, Villejuif, France
| | - E Folch
- Experimental Hepatic Ischemia-Reperfusion Unit, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC-IDIBAPS, Barcelona, Spain
| | - C Castro-Benítez
- Centre Hépato-Biliaire, Hôpital Universitaire Paul Brousse, Villejuif, France
| | - R Adam
- Centre Hépato-Biliaire, Hôpital Universitaire Paul Brousse, Villejuif, France
| | - J Roselló-Catafau
- Experimental Hepatic Ischemia-Reperfusion Unit, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC-IDIBAPS, Barcelona, Spain.
| |
Collapse
|
91
|
Choi HS, Park JA, Hwang JS, Ham SA, Yoo T, Lee WJ, Paek KS, Shin HC, Lee CH, Seo HG. A Dalbergia odorifera extract improves the survival of endotoxemia model mice by inhibiting HMGB1 release. Altern Ther Health Med 2017; 17:212. [PMID: 28403838 PMCID: PMC5389052 DOI: 10.1186/s12906-017-1725-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 04/05/2017] [Indexed: 01/14/2023]
Abstract
Background Dalbergia odorifera T. Chen (Leguminosae) is an indigenous medicinal herb that is widely used as a popular remedy in northern and eastern Asia. However, the cellular mechanisms underlying the biological activity of D. odorifera are not fully elucidated. Methods Anti-inflammatory effect of D. odorifera extract (DOE) was determined through intraperitoneal injection in a mouse model of endotoxemia induced by lipopolysaccharide (LPS). RAW 264.7 cells, a murine macrophage, were also treated with LPS to generate a cellular model of inflammation, and investigated the anti-inflammatory activity and underlying mechanisms of DOE and its constituent isoliquiritigenin. Results DOE dose-dependently inhibited LPS-induced release of high mobility group box 1 (HMGB1), a late proinflammatory cytokine, and decreased cytosolic translocation of HMGB1 in RAW264.7 cells. This inhibitory effect of DOE on HMGB1 release was observed in cells treated with DOE before or after LPS treatment, suggesting that DOE is effective for both treatment and prevention. In addition, DOE significantly inhibited LPS-induced formation of nitric oxide (NO) and expression of inducible NO synthase (iNOS) in a dose-dependent manner. These effects of DOE were accompanied by suppression of HMGB1 release triggered by LPS, suggesting a possible mechanism by which DOE modulates HMGB1 release through NO signaling. Isoriquiritigenin, a constituent of DOE, also attenuated LPS-triggered NO formation and HMGB1 release in RAW264.7 cells, indicating that isoriquiritigenin is an indexing molecule for the anti-inflammatory properties of DOE. Furthermore, c-Jun N-terminal kinase, but not extracellular signal-regulated kinase and p38, mediated DOE-dependent inhibition of HMGB1 release and NO/iNOS induction in RAW 264.7 cells exposed to LPS. Notably, administration of DOE ameliorated survival rates in a mouse model of endotoxemia induced by LPS, where decreased level of circulating HMGB1 was observed. Conclusion These results suggest that DOE confers resistance to LPS-triggered inflammation through NO-mediated inhibitory effects on HMGB1 release.
Collapse
|
92
|
Yin Y, Feng Y, Zhao H, Zhao Z, Yua H, Xu J, Che H. SIRT1 inhibits releases of HMGB1 and HSP70 from human umbilical vein endothelial cells caused by IL-6 and the serum from a preeclampsia patient and protects the cells from death. Biomed Pharmacother 2017; 88:449-458. [DOI: 10.1016/j.biopha.2017.01.087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/04/2017] [Accepted: 01/14/2017] [Indexed: 12/29/2022] Open
|
93
|
Sun J, Guo E, Yang J, Yang Y, Liu S, Hu J, Jiang X, Dirsch O, Dahmen U, Dong W, Liu A. Carbon monoxide ameliorates hepatic ischemia/reperfusion injury via sirtuin 1-mediated deacetylation of high-mobility group box 1 in rats. Liver Transpl 2017; 23:510-526. [PMID: 28133883 DOI: 10.1002/lt.24733] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 01/09/2017] [Accepted: 01/14/2017] [Indexed: 12/12/2022]
Abstract
Carbon monoxide (CO) exerts protective effects on hepatic ischemia/reperfusion injury (IRI), but the underlying molecular mechanisms are not fully understood. High-mobility group box 1 (HMGB1) is an important mediator of injury and inflammation in hepatic IRI. Here, we investigated whether CO could attenuate hepatic IRI via inhibition of HMGB1 release, particularly through sirtuin 1 (SIRT1). CO was released by treatment with carbon monoxide-releasing molecule (CORM)-2. CORM-2-delivered CO ameliorated hepatic IRI, as indicated by lower serum aminotransferase levels, lower hepatic inflammatory responses, and less severe ischemia/reperfusion-associated histopathologic changes. Treatment with CORM-2 significantly inhibited IRI-induced HMGB1 translocation and release. SIRT1 expression was increased by CORM-2 pretreatment. When CORM-2-induced SIRT1 expression was inhibited using EX527, HMGB1 translocation and release were increased and hepatic IRI was worsened, whereas SIRT1 activation by resveratrol reversed this trend. In vitro, CORM-2 reduced hypoxia/reoxygenation-induced HMGB1 translocation and release, these inhibitions were blocked by SIRT1 inhibition using EX527 or SIRT1 small interfering RNA both in alpha mouse liver 12 cells and RAW264.7 macrophages. Moreover, SIRT1 directly interacted with and deacetylated HMGB1. IRI increased HMGB1 acetylation, which was abolished by CORM-2 treatment via SIRT1. In conclusion, these results suggest that CO may increase SIRT1 expression, which may decrease HMGB1 acetylation and subsequently reduce its translocation and release, thereby protecting against hepatic IRI. Liver Transplantation 23 510-526 2017 AASLD.
Collapse
Affiliation(s)
- Jian Sun
- Department of Biliopancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Enshuang Guo
- Department of Infectious Diseases, Wuhan General Hospital of Guangzhou Military Command, Wuhan, China
| | - Jiankun Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shenpei Liu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jifa Hu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojing Jiang
- Department of Infectious Diseases, Wuhan General Hospital of Guangzhou Military Command, Wuhan, China
| | - Olaf Dirsch
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Friedrich-Schiller-University Jena, Jena, Germany
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Friedrich-Schiller-University Jena, Jena, Germany
| | - Wei Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| | - Anding Liu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
94
|
Kaneko Y, Pappas C, Malapira T, Vale FĹ, Tajiri N, Borlongan CV. Extracellular HMGB1 Modulates Glutamate Metabolism Associated with Kainic Acid-Induced Epilepsy-Like Hyperactivity in Primary Rat Neural Cells. Cell Physiol Biochem 2017; 41:947-959. [PMID: 28222432 DOI: 10.1159/000460513] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 12/21/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Neuroinflammatory processes have been implicated in the pathophysiology of seizure/epilepsy. High mobility group box 1 (HMGB1), a non-histone DNA binding protein, behaves like an inflammatory cytokine in response to epileptogenic insults. Kainic acid (KA) is an excitotoxic reagent commonly used to induce epilepsy in rodents. However, the molecular mechanism by which KA-induced HMGB1 affords the initiation of epilepsy, especially the role of extracellular HMGB1 in neurotransmitter expression, remains to be elucidated. METHODS Experimental early stage of epilepsy-related hyperexcitability was induced in primary rat neural cells (PRNCs) by KA administration. We measured the localization of HMGB1, cell viability, mitochondrial activity, and expression level of glutamate metabolism-associated enzymes. RESULTS KA induced the translocation of HMGB1 from nucleus to cytosol, and its release from the neural cells. The translocation is associated with post-translational modifications. An increase in extracellular HMGB1 decreased PRNC cell viability and mitochondrial activity, downregulated expression of glutamate decarboxylase67 (GAD67) and glutamate dehydrogenase (GLUD1/2), and increased intracellular glutamate concentration and major histocompatibility complex II (MHC II) level. CONCLUSIONS That a surge in extracellular HMGB1 approximated seizure initiation suggests a key pathophysiological contribution of HMGB1 to the onset of epilepsy-related hyperexcitability.
Collapse
|
95
|
Resveratrol treatment reveals a novel role for HMGB1 in regulation of the type 1 interferon response in dengue virus infection. Sci Rep 2017; 7:42998. [PMID: 28216632 PMCID: PMC5316936 DOI: 10.1038/srep42998] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 01/17/2017] [Indexed: 11/09/2022] Open
Abstract
Dengue is one of the most significant mosquito-borne virus diseases worldwide, particularly in tropical and subtropical regions. This study sought to examine the antiviral activity of resveratrol (RESV), a phytoalexin secreted naturally by plants, against dengue virus (DENV) infection. Our data showed that RESV inhibits the translocation of high mobility group box 1 (HMGB1), a DNA binding protein that normally resides in the nucleus, into the cytoplasm and extracellular milieu. HMGB1 migrates out of the nucleus during DENV infection. This migration is inhibited by RESV treatment and is mediated by induction of Sirt1 which leads to the retention of HMGB1 in the nucleus and consequently helps in the increased production of interferon-stimulated genes (ISGs). Nuclear HMGB1 was found to bind to the promoter region of the ISG and positively regulated the expression of ISG. The enhanced transcription of ISGs by nuclear HMGB1 thus contributes to the antiviral activity of RESV against DENV. To the best of our knowledge, this is the first report to demonstrate that RESV antagonizes DENV replication and that nuclear HMGB1 plays a role in regulating ISG production.
Collapse
|
96
|
Hwang JS, Ham SA, Yoo T, Lee WJ, Paek KS, Kim JH, Lee CH, Seo HG. Upregulation of MKP-7 in response to rosiglitazone treatment ameliorates lipopolysaccharide-induced destabilization of SIRT1 by inactivating JNK. Pharmacol Res 2016; 114:47-55. [DOI: 10.1016/j.phrs.2016.10.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/12/2016] [Accepted: 10/17/2016] [Indexed: 01/26/2023]
|
97
|
SIRT1/3 Activation by Resveratrol Attenuates Acute Kidney Injury in a Septic Rat Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:7296092. [PMID: 28003866 PMCID: PMC5149703 DOI: 10.1155/2016/7296092] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 09/07/2016] [Accepted: 10/30/2016] [Indexed: 12/20/2022]
Abstract
Sepsis often results in damage to multiple organ systems, possibly due to severe mitochondrial dysfunction. Two members of the sirtuin family, SIRT1 and SIRT3, have been implicated in the reversal of mitochondrial damage. The aim of this study was to determine the role of SIRT1/3 in acute kidney injury (AKI) following sepsis in a septic rat model. After drug pretreatment and cecal ligation and puncture (CLP) model reproduction in the rats, we performed survival time evaluation and kidney tissue extraction and renal tubular epithelial cell (RTEC) isolation. We observed reduced SIRT1/3 activity, elevated acetylated SOD2 (ac-SOD2) levels and oxidative stress, and damaged mitochondria in RTECs following sepsis. Treatment with resveratrol (RSV), a chemical SIRT1 activator, effectively restored SIRT1/3 activity, reduced acetylated SOD2 levels, ameliorated oxidative stress and mitochondrial function of RTECs, and prolonged survival time. However, the beneficial effects of RSV were greatly abrogated by Ex527, a selective inhibitor of SIRT1. These results suggest a therapeutic role for SIRT1 in the reversal of AKI in septic rat, which may rely on SIRT3-mediated deacetylation of SOD2. SIRT1/3 activation could therefore be a promising therapeutic strategy to treat sepsis-associated AKI.
Collapse
|
98
|
Structural aspects of the interaction of anticancer drug Actinomycin-D to the GC rich region of hmgb1 gene. Int J Biol Macromol 2016; 87:433-42. [DOI: 10.1016/j.ijbiomac.2016.02.060] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 02/22/2016] [Accepted: 02/23/2016] [Indexed: 12/18/2022]
|
99
|
Osinalde N, Mitxelena J, Sánchez-Quiles V, Akimov V, Aloria K, Arizmendi JM, Zubiaga AM, Blagoev B, Kratchmarova I. Nuclear Phosphoproteomic Screen Uncovers ACLY as Mediator of IL-2-induced Proliferation of CD4+ T lymphocytes. Mol Cell Proteomics 2016; 15:2076-92. [PMID: 27067055 DOI: 10.1074/mcp.m115.057158] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Indexed: 02/03/2023] Open
Abstract
Anti-cancer immunotherapies commonly rely on the use of interleukin-2 (IL-2) to promote the expansion of T lymphocytes. IL-2- dependent proliferation is the culmination of a complex network of phosphorylation-driven signaling events that impact on gene transcription through mechanisms that are not clearly understood. To study the role of IL-2 in the regulation of nuclear protein function we have performed an unbiased mass spectrometry-based study of the nuclear phosphoproteome of resting and IL-2-treated CD4(+) T lymphocytes. We detected 8521distinct phosphosites including many that are not yet reported in curated phosphorylation databases. Although most phosphorylation sites remained unaffected upon IL-2 treatment, 391 sites corresponding to 288 gene products showed robust IL-2-dependent regulation. Importantly, we show that ATP-citrate lyase (ACLY) is a key phosphoprotein effector of IL-2-mediated T-cell responses. ACLY becomes phosphorylated on serine 455 in T lymphocytes upon IL-2-driven activation of AKT, and depletion or inactivation of ACLY compromises IL-2-promoted T-cell growth. Mechanistically, we demonstrate that ACLY is required for enhancing histone acetylation levels and inducing the expression of cell cycle regulating genes in response to IL-2. Thus, the metabolic enzyme ACLY emerges as a bridge between cytokine signaling and proliferation of T lymphocytes, and may be an attractive candidate target for the development of more efficient anti-cancer immunotherapies.
Collapse
Affiliation(s)
- Nerea Osinalde
- From the ‡Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Jone Mitxelena
- §Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain
| | - Virginia Sánchez-Quiles
- From the ‡Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Vyacheslav Akimov
- From the ‡Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Kerman Aloria
- ¶Proteomics Core Facility-SGIKER, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain
| | - Jesus M Arizmendi
- ‖Department of Biochemistry and Molecular Biology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain
| | - Ana M Zubiaga
- §Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain
| | - Blagoy Blagoev
- From the ‡Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Irina Kratchmarova
- From the ‡Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark;
| |
Collapse
|