51
|
Bathish B, Robertson H, Dillon JF, Dinkova-Kostova AT, Hayes JD. Nonalcoholic steatohepatitis and mechanisms by which it is ameliorated by activation of the CNC-bZIP transcription factor Nrf2. Free Radic Biol Med 2022; 188:221-261. [PMID: 35728768 DOI: 10.1016/j.freeradbiomed.2022.06.226] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 12/11/2022]
Abstract
Non-alcoholic steatohepatitis (NASH) represents a global health concern. It is characterised by fatty liver, hepatocyte cell death and inflammation, which are associated with lipotoxicity, endoplasmic reticulum (ER) stress, mitochondrial dysfunction, iron overload and oxidative stress. NF-E2 p45-related factor 2 (Nrf2) is a transcription factor that combats oxidative stress. Remarkably, Nrf2 is downregulated during the development of NASH, which probably accelerates disease, whereas in pre-clinical studies the upregulation of Nrf2 inhibits NASH. We now review the scientific literature that proposes Nrf2 downregulation during NASH involves its increased ubiquitylation and proteasomal degradation, mediated by Kelch-like ECH-associated protein 1 (Keap1) and/or β-transducin repeat-containing protein (β-TrCP) and/or HMG-CoA reductase degradation protein 1 (Hrd1, also called synoviolin (SYVN1)). Additionally, downregulation of Nrf2-mediated transcription during NASH may involve diminished recruitment of coactivators by Nrf2, due to increased levels of activating transcription factor 3 (ATF3) and nuclear factor-kappaB (NF-κB) p65, or competition for promoter binding due to upregulation of BTB and CNC homology 1 (Bach1). Many processes that downregulate Nrf2 are triggered by transforming growth factor-beta (TGF-β), with oxidative stress amplifying its signalling. Oxidative stress may also increase suppression of Nrf2 by β-TrCP through facilitating formation of the DSGIS-containing phosphodegron in Nrf2 by glycogen synthase kinase-3. In animal models, knockout of Nrf2 increases susceptibility to NASH, while pharmacological activation of Nrf2 by inducing agents that target Keap1 inhibits development of NASH. These inducing agents probably counter Nrf2 downregulation affected by β-TrCP, Hrd1/SYVN1, ATF3, NF-κB p65 and Bach1, by suppressing oxidative stress. Activation of Nrf2 is also likely to inhibit NASH by ameliorating lipotoxicity, inflammation, ER stress and iron overload. Crucially, pharmacological activation of Nrf2 in mice in which NASH has already been established supresses liver steatosis and inflammation. There is therefore compelling evidence that pharmacological activation of Nrf2 provides a comprehensive multipronged strategy to treat NASH.
Collapse
Affiliation(s)
- Boushra Bathish
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Holly Robertson
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK; Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - John F Dillon
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - John D Hayes
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK.
| |
Collapse
|
52
|
Hypoxia-induced HMGB1 promotes glioma stem cells self-renewal and tumorigenicity via RAGE. iScience 2022; 25:104872. [PMID: 36034219 PMCID: PMC9399482 DOI: 10.1016/j.isci.2022.104872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/10/2022] [Accepted: 07/29/2022] [Indexed: 11/23/2022] Open
Abstract
Glioma stem cells (GSCs) in the hypoxic niches contribute to tumor initiation, progression, and recurrence in glioblastoma (GBM). Hypoxia induces release of high-mobility group box 1 (HMGB1) from tumor cells, promoting the development of tumor. Here, we report that HMGB1 is overexpressed in human GBM specimens. Hypoxia promotes the expression and secretion of HMGB1 in GSCs. Furthermore, silencing HMGB1 results in the loss of stem cell markers and a reduction in self-renewal ability of GSCs. Additionally, HMGB1 knockdown inhibits the activation of RAGE-dependent ERK1/2 signaling pathway and arrests the cell cycle in GSCs. Consistently, FPS-ZM1, an inhibitor of RAGE, downregulates HMGB1 expression and the phosphorylation of ERK1/2, leading to a reduction in the proliferation of GSCs. In xenograft mice of GBM, HMGB1 knockdown inhibits tumor growth and promotes mouse survival. Collectively, these findings uncover a vital function for HMGB1 in regulating GSC self-renewal potential and tumorigenicity. Glioma stem cells overexpress HMGB1 in human glioblastoma Hypoxia induces the upregulation and release of HMGB1 in glioma stem cells HMGB1 promotes the self-renewal of glioma stem cells via RAGE Targeting HMGB1 inhibits the tumorigenesis of glioma stem cells
Collapse
|
53
|
Li Y, Palmer A, Lupu L, Huber-Lang M. Inflammatory response to the ischaemia-reperfusion insult in the liver after major tissue trauma. Eur J Trauma Emerg Surg 2022; 48:4431-4444. [PMID: 35831749 DOI: 10.1007/s00068-022-02026-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 05/28/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Polytrauma is often accompanied by ischaemia-reperfusion injury to tissues and organs, and the resulting series of immune inflammatory reactions are a major cause of death in patients. The liver is one of the largest organs in the body, a characteristic that makes it the most vulnerable organ after multiple injuries. In addition, the liver is an important digestive organ that secretes a variety of inflammatory mediators involved in local as well as systemic immune inflammatory responses. Therefore, this review considers the main features of post-traumatic liver injury, focusing on the immuno-pathophysiological changes, the interactions between liver organs, and the principles of treatment deduced. METHODS We focus on the local as well as systemic immune response involving the liver after multiple injuries, with emphasis on the pathophysiological mechanisms. RESULTS An overview of the mechanisms underlying the pathophysiology of local as well as systemic immune responses involving the liver after multiple injuries, the latest research findings, and the current mainstream therapeutic approaches. CONCLUSION Cross-reactivity between various organs and cascade amplification effects are among the main causes of systemic immune inflammatory responses after multiple injuries. For the time being, the pathophysiological mechanisms underlying this interaction remain unclear. Future work will continue to focus on identifying potential signalling pathways as well as target genes and intervening at the right time points to prevent more severe immune inflammatory responses and promote better and faster recovery of the patient.
Collapse
Affiliation(s)
- Yang Li
- Institute for Clinical and Experimental Trauma Immunology (ITI), University Hospital Ulm, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Annette Palmer
- Institute for Clinical and Experimental Trauma Immunology (ITI), University Hospital Ulm, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Ludmila Lupu
- Institute for Clinical and Experimental Trauma Immunology (ITI), University Hospital Ulm, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma Immunology (ITI), University Hospital Ulm, Helmholtzstr. 8/1, 89081, Ulm, Germany.
| |
Collapse
|
54
|
Deguchi A, Maru Y. Inflammation-associated premetastatic niche formation. Inflamm Regen 2022; 42:22. [PMID: 35780158 PMCID: PMC9250732 DOI: 10.1186/s41232-022-00208-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 04/04/2022] [Indexed: 11/10/2022] Open
Abstract
Metastasis remains the leading cause of cancer-related death. In 1889, Stephen Paget originally proposed the theory "seed-and-soil." Both cancer cell-intrinsic properties ("seed") and fertile microenvironment ("soil") are essential for metastasis formation. To date, accumulating evidences supported the theory using mouse models. The formation of a premetastatic niche has been widely accepted as an accel for metastasis. Similar to tumor microenvironment, various types of cells, such as immune cells, endothelial cells, and fibroblasts are involved in premetastatic niche formation. We have discovered that primary tumors hijack Toll-like receptor 4 (TLR4) signaling to establish a premetastatic niche in the lung by utilizing the endogenous ligands. In this review, we discuss the mechanisms that underlie inflammation-associated premetastatic niche formation upon metastasis, focusing especially on myeloid cells and macrophages as the cells executing and mediating complicated processes.
Collapse
Affiliation(s)
- Atsuko Deguchi
- Department of Pharmacology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan.
| | - Yoshiro Maru
- Department of Pharmacology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan.
| |
Collapse
|
55
|
Ryan TAJ, O'Neill LAJ. Innate immune signaling and immunothrombosis: New insights and therapeutic opportunities. Eur J Immunol 2022; 52:1024-1034. [PMID: 35569038 PMCID: PMC9543829 DOI: 10.1002/eji.202149410] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 01/10/2023]
Abstract
Activation of the coagulation cascade is a critical, evolutionarily conserved mechanism that maintains hemostasis by rapidly forming blood clots in response to blood-borne infections and damaged blood vessels. Coagulation is a key component of innate immunity since it prevents bacterial dissemination and can provoke inflammation. The term immunothrombosis describes the process by which the innate immune response drives aberrant coagulation, which can result in a lethal condition termed disseminated intravascular coagulation, often seen in sepsis. In this review, we describe the recently uncovered molecular mechanisms underlying inflammasome- and STING-driven immunothrombosis induced by bacterial and viral infections, culminating in tissue factor (TF) activation and release. Current anticoagulant therapeutics, while effective, are associated with a life-threatening bleeding risk, requiring the urgent development of new treatments. Targeting immunothrombosis may provide a safer option. Thus, we highlight preclinical tools which target TF and/or block canonical (NLRP3) or noncanonical (caspase-11) inflammasome activation as well as STING-driven TF release and discuss clinically approved drugs which block key immunothrombotic processes and, therefore, may be redeployed as safer anticoagulants.
Collapse
Affiliation(s)
- Tristram A. J. Ryan
- School of Biochemistry and ImmunologyTrinity Biomedical Sciences InstituteTrinity College DublinDublin 2Ireland
| | - Luke A. J. O'Neill
- School of Biochemistry and ImmunologyTrinity Biomedical Sciences InstituteTrinity College DublinDublin 2Ireland
| |
Collapse
|
56
|
Du D, Lv W, Jing X, Yu C, Wuen J, Hasi S. Camel whey protein alleviates heat stress-induced liver injury by activating the Nrf2/HO-1 signaling pathway and inhibiting HMGB1 release. Cell Stress Chaperones 2022; 27:449-460. [PMID: 35543864 PMCID: PMC9346022 DOI: 10.1007/s12192-022-01277-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 01/03/2023] Open
Abstract
This study aimed to investigate the mechanism by which camel whey protein (CWP) inhibits the release of high-mobility group box 1 (HMGB1) in heat stress (HS)-stimulated rat liver. Administration of CWP by gavage prior to HS inhibited the cytoplasmic translocation of HMGB1 and consequently reduced the inflammatory response in the rat liver, and downregulated the levels of the NLR pyrin domain containing 3 (NLRP3) inflammasome, interleukin (IL)-1β, and tumor necrosis factor (TNF)-α. The use of N-acetyl-L-cysteine (NAC), an inhibitor of reactive oxygen species (ROS) production, indicated that this downregulation effect may be attributed to the antioxidant activity of CWP. We observed that CWP enhanced nuclear factor erythroid 2-related factor (Nrf)2 and heme-oxygenase (HO)-1 expression, which inhibited ROS production, nicotinamide adenine dinucleotide phosphate oxidase (NOX) activity, and malondialdehyde (MDA) levels, and increased superoxide dismutase 1 (SOD1) activity and reduced glutathione (GSH) content in the HS-treated liver, ultimately increasing the total antioxidant capacity (TAC) in the liver. Administration of Nrf2 or HO-1 inhibitors before HS abolished the protective effects of CWP against oxidative damage in the liver of HS-treated rats, accompanied by increased levels of HMGB1 in the cytoplasm and IL-1β and TNF-α in the serum. In conclusion, our study demonstrated that CWP enhanced the TAC of the rat liver after HS by activating Nrf2/HO-1 signaling, which in turn reduced HMGB1 release from hepatocytes and the subsequent inflammatory response and damage. Furthermore, the combination of CWP and NAC abolished the adverse effects of HS in the rat liver. Therefore, dietary CWP could be an effective adjuvant treatment for HS-induced liver damage.
Collapse
Affiliation(s)
- Donghua Du
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease/Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, 010018, China
- Department of Veterinary Medicine, College of Animal Science and Technology, Hebei North University, Zhangjiakou, 075131, Hebei, China
| | - Wenting Lv
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease/Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, 010018, China
- Department of Veterinary Medicine, College of Animal Science and Technology, Hebei North University, Zhangjiakou, 075131, Hebei, China
| | - Xiaoxia Jing
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease/Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Chunwei Yu
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease/Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Jiya Wuen
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease/Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Surong Hasi
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease/Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, 010018, China.
- Inner Mongolia Institute of Camel Research, Alashan, 750300, Inner Mongolia, China.
| |
Collapse
|
57
|
Cao Z, Yang F, Lin Y, Shan J, Cao H, Zhang C, Zhuang Y, Xing C, Hu G. Selenium Antagonizes Cadmium-Induced Inflammation and Oxidative Stress via Suppressing the Interplay between NLRP3 Inflammasome and HMGB1/NF-κB Pathway in Duck Hepatocytes. Int J Mol Sci 2022; 23:ijms23116252. [PMID: 35682929 PMCID: PMC9181349 DOI: 10.3390/ijms23116252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 12/14/2022] Open
Abstract
Cadmium (Cd) is a toxic heavy metal that can accumulate in the liver of animals, damaging liver function. Inflammation and oxidative stress are considered primary causes of Cd-induced liver damage. Selenium (Se) is an antioxidant and can resist the detrimental impacts of Cd on the liver. To elucidate the antagonism of Se on Cd against hepatocyte injury and its mechanism, duck embryo hepatocytes were treated with Cd (4 μM) and/or Se (0.4 μM) for 24 h. Then, the hepatocyte viability, oxidative stress and inflammatory status were assessed. The findings manifested that the accumulation of reactive oxygen species (ROS) and the levels of pro-inflammatory factors were elevated in the Cd group. Simultaneously, immunofluorescence staining revealed that the interaction between NOD-like receptor pyran domain containing 3 (NLRP3) and apoptosis-associated speck-like protein (ASC) was enhanced, the movement of high-mobility group box 1 (HMGB1) from nucleus to cytoplasm was increased and the inflammatory response was further amplified. Nevertheless, the addition of Se relieved the above-mentioned effects, thereby alleviating cellular oxidative stress and inflammation. Collectively, the results suggested that Se could mitigate Cd-stimulated oxidative stress and inflammation in hepatocytes, which might be correlated with the NLRP3 inflammasome and HMGB1/nuclear factor-κB (NF-κB) signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chenghong Xing
- Correspondence: (C.X.); (G.H.); Tel.: +86-18770046182 (C.X.); +86-13807089905 (G.H.)
| | - Guoliang Hu
- Correspondence: (C.X.); (G.H.); Tel.: +86-18770046182 (C.X.); +86-13807089905 (G.H.)
| |
Collapse
|
58
|
Tang SP, Mao XL, Chen YH, Yan LL, Ye LP, Li SW. Reactive Oxygen Species Induce Fatty Liver and Ischemia-Reperfusion Injury by Promoting Inflammation and Cell Death. Front Immunol 2022; 13:870239. [PMID: 35572532 PMCID: PMC9098816 DOI: 10.3389/fimmu.2022.870239] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/07/2022] [Indexed: 01/24/2023] Open
Abstract
Liver transplantation is the ultimate method for treating end-stage liver disease. With the increasing prevalence of obesity, the number of patients with non-alcoholic fatty liver, a common cause of chronic liver disease, is on the rise and may become the main cause of liver transplantation in the future. With the increasing gap between the number of donor livers and patients waiting for liver transplantation and the increasing prevalence of non-alcoholic fatty liver, the proportion of steatosis livers among non-standard donor organs is also increasing. Ischemia-reperfusion injury has historically been the focus of attention in the liver transplantation process, and severe ischemia-reperfusion injury leads to adverse outcomes of liver transplantation. Studies have shown that the production of reactive oxygen species and subsequent oxidative stress play a key role in the pathogenesis of hepatic ischemia and reperfusion injury and non-alcoholic fatty liver. Furthermore, the sensitivity of fatty liver transplantation to ischemia-reperfusion injury has been suggested to be related to the production of reactive oxygen species (ROS) and oxidative stress. In ischemia-reperfusion injury, Kupffer cell and macrophage activation along with mitochondrial damage and the xanthine/xanthine oxidase system promote marked reactive oxygen species production and the inflammatory response and apoptosis, resulting in liver tissue injury. The increased levels of ROS and lipid peroxidation products, vicious circle of ROS and oxidative stress along with mitochondrial dysfunction promoted the progress of non-alcoholic fatty liver. In contrast to the non-fatty liver, a non-alcoholic fatty liver produces more reactive oxygen species and suffers more serious oxidative stress when subjected to ischemia-reperfusion injury. We herein review the effects of reactive oxygen species on ischemia-reperfusion injury and non-alcoholic fatty liver injury as well as highlight several treatment approaches.
Collapse
Affiliation(s)
- Shen-ping Tang
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Xin-li Mao
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Ya-hong Chen
- Health Management Center, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Ling-ling Yan
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, China
| | - Li-ping Ye
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- *Correspondence: Li-ping Ye, ; Shao-wei Li,
| | - Shao-wei Li
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- *Correspondence: Li-ping Ye, ; Shao-wei Li,
| |
Collapse
|
59
|
Shaker ME. The contribution of sterile inflammation to the fatty liver disease and the potential therapies. Biomed Pharmacother 2022; 148:112789. [PMID: 35272137 DOI: 10.1016/j.biopha.2022.112789] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 11/02/2022] Open
Abstract
Hepatic inflammation is prevalent in several metabolic liver diseases. Recent scientific advances about the pathogenesis of metabolic liver diseases showed an emerging role of several damage-associated molecular patterns (DAMPs), including DNA, high-mobility group box 1 (HMGB1), ATP and uric acid. For these DAMPs to induce inflammation, they should stimulate pattern recognition receptors (PRRs), which are located in the hepatic immune cells like resident Kupffer cells, infiltrated neutrophils, monocytes or dendritic cells. As a consequence, proinflammatory cytokines like interleukins (ILs)-1β and 18 alongside tumor necrosis factor (TNF)-α are overproduced and released, leading to pronounced hepatic inflammation and cellular death. This review highlights the contribution of these DAMPs and PRRs in the settings of alcoholic and nonalcoholic steatohepatitis. The review also summarizes the therapeutic usefulness of targeting NLR family pyrin domain containing 3 (NLRP3)-inflammasome, Toll-like receptors (TLRs) 4 and 9, IL-1 receptor (IL-1R), caspase 1, uric acid and GMP-AMP synthase/stimulator of interferon genes (cGAS/STING) in these hepatic inflammatory disorders.
Collapse
Affiliation(s)
- Mohamed E Shaker
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Aljouf, Saudi Arabia.
| |
Collapse
|
60
|
Jung E, Song N, Lee Y, Kwon G, Kwon S, Lee D. H2O2-activatable hybrid prodrug nanoassemblies as a pure nanodrug for hepatic ischemia/reperfusion injury. Biomaterials 2022; 284:121515. [DOI: 10.1016/j.biomaterials.2022.121515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 03/28/2022] [Accepted: 04/04/2022] [Indexed: 11/02/2022]
|
61
|
Deng C, Zhao L, Yang Z, Shang JJ, Wang CY, Shen MZ, Jiang S, Li T, Di WC, Chen Y, Li H, Cheng YD, Yang Y. Targeting HMGB1 for the treatment of sepsis and sepsis-induced organ injury. Acta Pharmacol Sin 2022; 43:520-528. [PMID: 34040166 PMCID: PMC8888646 DOI: 10.1038/s41401-021-00676-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 04/01/2021] [Indexed: 02/05/2023] Open
Abstract
High mobility group box 1 (HMGB1) is a ubiquitous nuclear protein that is present in almost all cells and regulates the activity of innate immune responses in both intracellular and extracellular settings. Current evidence suggests that HMGB1 plays a pivotal role in human pathological and pathophysiological processes such as the inflammatory response, immune reactions, cell migration, aging, and cell death. Sepsis is a systemic inflammatory response syndrome (SIRS) that occurs in hosts in response to microbial infections with a proven or suspected infectious etiology and is the leading cause of death in intensive care units worldwide, particularly in the aging population. Dysregulated systemic inflammation is a classic characteristic of sepsis, and suppression of HMGB1 may ameliorate inflammation and improve patient outcomes. Here, we focus on the latest breakthroughs regarding the roles of HMGB1 in sepsis and sepsis-related organ injury, the ways by which HMGB1 are released, and the signaling pathways and therapeutics associated with HMGB1. This review highlights recent advances related to HMGB1: the regulation of HMBG1 might be helpful for both basic research and drug development for the treatment of sepsis and sepsis-related organ injury.
Collapse
Affiliation(s)
- Chao Deng
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Department of Orthopaedics, Huaian Medical District of Jingling Hospital, Medical School of Nanjing University, Huaian, 213001, China
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lin Zhao
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Zhi Yang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, Xi'an, 710021, China
| | - Jia-Jia Shang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, Xi'an, 710021, China
| | - Chang-Yu Wang
- Department of Cardiology, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
| | - Ming-Zhi Shen
- Hainan Hospital of PLA General Hospital, The Second School of Clinical Medicine, Southern Medical University, Sanya, 572013, China
| | - Shuai Jiang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, Xi'an, 710021, China
| | - Tian Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
| | - Wen-Cheng Di
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518100, China
| | - Ying Chen
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - He Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, Xi'an, 710021, China
| | - Ye-Dong Cheng
- Department of Orthopaedics, Huaian Medical District of Jingling Hospital, Medical School of Nanjing University, Huaian, 213001, China.
| | - Yang Yang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, Xi'an, 710021, China.
| |
Collapse
|
62
|
Zito G, Miceli V, Carcione C, Busà R, Bulati M, Gallo A, Iannolo G, Pagano D, Conaldi PG. Human Amnion-Derived Mesenchymal Stromal/Stem Cells Pre-Conditioning Inhibits Inflammation and Apoptosis of Immune and Parenchymal Cells in an In Vitro Model of Liver Ischemia/Reperfusion. Cells 2022; 11:cells11040709. [PMID: 35203355 PMCID: PMC8870407 DOI: 10.3390/cells11040709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/09/2022] [Accepted: 02/15/2022] [Indexed: 02/07/2023] Open
Abstract
Ischemia/reperfusion injury (IRI) represents one of the leading causes of primary non-function acute liver transplantation failure. IRI, generated by an interruption of organ blood flow and the subsequent restoration upon transplant, i.e., reperfusion, generates the activation of an inflammatory cascade from the resident Kupffer cells, leading first to neutrophils recruitment and second to apoptosis of the parenchyma. Recently, human mesenchymal stromal/stem cells (hMSCs) and derivatives have been implemented for reducing the damage induced by IRI. Interestingly, sparse data in the literature have described the use of human amnion-derived MSCs (hAMSCs) and, more importantly, no evidence regarding hMSCs priming on liver IRI have been described yet. Thus, our study focused on the definition of an in vitro model of liver IRI to test the effect of primed hAMSCs to reduce IRI damage on immune and hepatic cells. We found that the IFNγ pre-treatment and 3D culture of hAMSCs strongly reduced inflammation induced by M1-differentiated macrophages. Furthermore, primed hAMSCs significantly inhibited parenchymal apoptosis at early timepoints of reperfusion by blocking the activation of caspase 3/7. All together, these data demonstrate that hAMSCs priming significantly overcomes IRI effects in vitro by engaging the possibility of defining the molecular pathways involved in this process.
Collapse
Affiliation(s)
- Giovanni Zito
- Research Department, IRCSS ISMETT (Instituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (V.M.); (R.B.); (M.B.); (A.G.); (G.I.); (D.P.); (P.G.C.)
- Correspondence: ; Tel.: +39-091-21-92-649
| | - Vitale Miceli
- Research Department, IRCSS ISMETT (Instituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (V.M.); (R.B.); (M.B.); (A.G.); (G.I.); (D.P.); (P.G.C.)
| | | | - Rosalia Busà
- Research Department, IRCSS ISMETT (Instituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (V.M.); (R.B.); (M.B.); (A.G.); (G.I.); (D.P.); (P.G.C.)
| | - Matteo Bulati
- Research Department, IRCSS ISMETT (Instituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (V.M.); (R.B.); (M.B.); (A.G.); (G.I.); (D.P.); (P.G.C.)
| | - Alessia Gallo
- Research Department, IRCSS ISMETT (Instituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (V.M.); (R.B.); (M.B.); (A.G.); (G.I.); (D.P.); (P.G.C.)
| | - Gioacchin Iannolo
- Research Department, IRCSS ISMETT (Instituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (V.M.); (R.B.); (M.B.); (A.G.); (G.I.); (D.P.); (P.G.C.)
| | - Duilio Pagano
- Research Department, IRCSS ISMETT (Instituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (V.M.); (R.B.); (M.B.); (A.G.); (G.I.); (D.P.); (P.G.C.)
| | - Pier Giulio Conaldi
- Research Department, IRCSS ISMETT (Instituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (V.M.); (R.B.); (M.B.); (A.G.); (G.I.); (D.P.); (P.G.C.)
| |
Collapse
|
63
|
Chen R, Kang R, Tang D. The mechanism of HMGB1 secretion and release. Exp Mol Med 2022; 54:91-102. [PMID: 35217834 PMCID: PMC8894452 DOI: 10.1038/s12276-022-00736-w] [Citation(s) in RCA: 291] [Impact Index Per Article: 145.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/13/2021] [Accepted: 11/04/2021] [Indexed: 02/08/2023] Open
Abstract
High mobility group box 1 (HMGB1) is a nonhistone nuclear protein that has multiple functions according to its subcellular location. In the nucleus, HMGB1 is a DNA chaperone that maintains the structure and function of chromosomes. In the cytoplasm, HMGB1 can promote autophagy by binding to BECN1 protein. After its active secretion or passive release, extracellular HMGB1 usually acts as a damage-associated molecular pattern (DAMP) molecule, regulating inflammation and immune responses through different receptors or direct uptake. The secretion and release of HMGB1 is fine-tuned by a variety of factors, including its posttranslational modification (e.g., acetylation, ADP-ribosylation, phosphorylation, and methylation) and the molecular machinery of cell death (e.g., apoptosis, pyroptosis, necroptosis, alkaliptosis, and ferroptosis). In this minireview, we introduce the basic structure and function of HMGB1 and focus on the regulatory mechanism of HMGB1 secretion and release. Understanding these topics may help us develop new HMGB1-targeted drugs for various conditions, especially inflammatory diseases and tissue damage. A nuclear protein that gets released after cell death or is actively secreted by immune cells offers a promising therapeutic target for treating diseases linked to excessive inflammation. Daolin Tang from the University of Texas Southwestern Medical Center in Dallas, USA, and colleagues review how cellular stresses can trigger the accumulation of HMGB1, a type of alarm signal protein that promotes the recruitment and activation of inflammation-promoting immune cells. The researchers discuss various mechanisms that drive both passive and active release of HMGB1 into the space around cells. These processes, which include enzymatic modifications of the HMGB1 protein, cell–cell interactions and molecular pathways of cell death, could be targeted by drugs to lessen tissue damage and inflammatory disease caused by HMGB1-induced immune responses
Collapse
Affiliation(s)
- Ruochan Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China. .,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
64
|
STING Induces Liver Ischemia-Reperfusion Injury by Promoting Calcium-Dependent Caspase 1-GSDMD Processing in Macrophages. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8123157. [PMID: 35281468 PMCID: PMC8906939 DOI: 10.1155/2022/8123157] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 10/26/2021] [Accepted: 01/15/2022] [Indexed: 12/31/2022]
Abstract
Objectives Although a recent study reported that stimulator of interferon genes (STING) in macrophages has an important regulatory effect on liver ischemia-reperfusion injury (IRI), the underlying mechanism of STING-dependent innate immune activation in liver macrophages (Kupffer cells, KCs) remains unclear. Here, we investigated the effect of STING on liver macrophage pyroptosis and the associated regulatory mechanism of liver IRI. Methods Clodronate liposomes were used to block liver macrophages. AAV-STING-RNAi-F4/80-EGFP, an adenoassociated virus (AAV), was transfected into the portal vein of mice in vivo, and the liver IRI model was established 14 days later. In vitro, liver macrophages were treated with STING-specific siRNA, and a hypoxia-reoxygenation (H/R) model was established. The level of STING was detected via Western blotting (WB), RT-PCR, and immunostaining. Liver tissue and blood samples were collected. Pathological changes in liver tissue were detected by hematoxylin and eosin (H&E) staining. Macrophage pyroptosis was detected by WB, confocal laser scanning microscopy (CLSM), transmission electron microscopy (TEM), and enzyme-linked immunosorbent assay (ELISA). The calcium concentration was measured by immunofluorescence and analyzed with a fluorescence microplate reader. Results The expression of STING increased with liver IRI but decreased significantly after the clodronate liposome blockade of liver macrophages. After knockdown of STING, the activation of caspase 1-GSDMD in macrophages and liver IRI was alleviated. More interestingly, hypoxia/reoxygenation (H/R) increased the calcium concentration in liver macrophages, but the calcium concentration was decreased after STING knockdown. Furthermore, after the inhibition of calcium in H/R-induced liver macrophages by BAPTA-AM, pyroptosis was significantly reduced, but the expression of STING was not significantlydecreased. Conclusions Knockdown of STING reduces calcium-dependent macrophage caspase 1-GSDMD-mediated liver IRI, representing a potential therapeutic approach in the clinic.
Collapse
|
65
|
Kim MJ, Hwang YH, Hwang JW, Alam Z, Lee DY. Heme oxygenase-1 gene delivery for altering high mobility group box-1 protein in pancreatic islet. J Control Release 2022; 343:326-337. [PMID: 35085698 DOI: 10.1016/j.jconrel.2022.01.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 10/19/2022]
Abstract
Pancreatic islet transplantation is a promising strategy for the treatment of type I diabetes. High-mobility group box-1 (HMGB1), highly expressed in islet cells, is a potent immune stimulator in immune rejection. Heme oxygenase-1 (HO1) gene therapy can modulate the release of HMGB1 by altering intracellular molecules for successful cell transplantation. After delivery of the heme oxygenase-1 (HO1) gene to islet cells using an adeno-associated viral vector (AAV), it was evaluated the changes in cytoplasmic Ca2+ ions and calcineurin activity as well as histone acetyltransferase (HAT) and Poly(ADP) ribose polymerase-1 (PARP-1). Inhibition of HMGB1 release was evaluated through altering these intracellular molecules. Then, after transplantation of HO1-transduced islets, the therapeutic effect of them was evaluated through measuring blood glucose level to diabetic mice and through immunohistochemical analysis. The transduced HO1 gene significantly inhibited HMGB1 release in islets that was under the cell damage by hypoxia exposure. It was confirmed that this result was initially due to the decrease in cytoplasmic Ca2+ ion concentration and calcineurin activity. In addition, the delivered HO1 gene simultaneously reduced the activity of HAT and PARP-1, which are involved in the translocation of HMGB1 from the nucleus to the cytoplasm. As a result, when the HO1 gene-transduced islets were transplanted into diabetic mice, the treatment efficiency of diabetes was effectively improved by increasing the survival rate of the islets. Collectively, these results suggest that HO1 gene transfer can be used for successful islet transplantation by altering the activity of intracellular signal molecules and reducing HMGB1 release.
Collapse
Affiliation(s)
- Min Jun Kim
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Yong Hwa Hwang
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Jin Wook Hwang
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Zahid Alam
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea; Institute of Nano Science & Technology (INST), Hanyang University, Seoul 04763, Republic of Korea; Elixir Pharmatech Inc., Seoul 04763, Republic of Korea.
| |
Collapse
|
66
|
Dai Q, Xie D, Zhang C, Zhu L, Xu Y, Li K, Hao W, Yin H. Osthole Blocks HMGB1 Release From the Nucleus and Confers Protective Effects Against Renal Ischemia-Reperfusion Injury. Front Physiol 2022; 12:735425. [PMID: 35002751 PMCID: PMC8727455 DOI: 10.3389/fphys.2021.735425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/20/2021] [Indexed: 11/13/2022] Open
Abstract
Renal ischemia-reperfusion (IR) is one of the main causes of renal injury. In severe cases with serious consequences, IR-related renal damage progresses rapidly and can even lead to acute renal failure. Its clinical treatment is currently difficult. According to various studies at home and abroad, HMGB1 is released from the nucleus into the cytoplasm or extracellular space by damaged parenchymal cells during ischemia and hypoxia, and this plays an important role in the initiation of reperfusion injury as an early inflammatory factor and is closely related to the occurrence and development of renal diseases. In recent years, the protective effect of osthole on IR of tissues and organs has been a key topic among clinical researchers. Osthole can inhibit the inflammatory response, reduce cell apoptosis the progression, and improve the prognosis of IR, thus protecting the kidney. During the development of renal IR, finding a mechanism through which the osthole blocks the release of HMGB1 from the nucleus would be helpful in detecting targets for clinical treatment.
Collapse
Affiliation(s)
- Qing Dai
- Department of Nephrology, The Second People's Hospital of Yibin, Yibin, China.,North Sichuan Medical College, Nanchong, China
| | - Deqiong Xie
- Department of Nephrology, The Second People's Hospital of Yibin, Yibin, China
| | - Chenli Zhang
- Department of Nephrology, The Second People's Hospital of Yibin, Yibin, China
| | - Lei Zhu
- Department of Nephrology, The Second People's Hospital of Yibin, Yibin, China
| | - Ying Xu
- Department of Nephrology, The Second People's Hospital of Yibin, Yibin, China
| | - Kui Li
- Department of Nephrology, The Second People's Hospital of Yibin, Yibin, China
| | - Wen Hao
- Department of Nephrology, The Second People's Hospital of Yibin, Yibin, China
| | - Hefei Yin
- Department of Nephrology, The Second People's Hospital of Yibin, Yibin, China
| |
Collapse
|
67
|
Zhang YY, Ning BT. Signaling pathways and intervention therapies in sepsis. Signal Transduct Target Ther 2021; 6:407. [PMID: 34824200 PMCID: PMC8613465 DOI: 10.1038/s41392-021-00816-9] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by dysregulated host systemic inflammatory and immune response to infection. Over decades, advanced understanding of host-microorganism interaction has gradually unmasked the genuine nature of sepsis, guiding toward new definition and novel therapeutic approaches. Diverse clinical manifestations and outcomes among infectious patients have suggested the heterogeneity of immunopathology, while systemic inflammatory responses and deteriorating organ function observed in critically ill patients imply the extensively hyperactivated cascades by the host defense system. From focusing on microorganism pathogenicity, research interests have turned toward the molecular basis of host responses. Though progress has been made regarding recognition and management of clinical sepsis, incidence and mortality rate remain high. Furthermore, clinical trials of therapeutics have failed to obtain promising results. As far as we know, there was no systematic review addressing sepsis-related molecular signaling pathways and intervention therapy in literature. Increasing studies have succeeded to confirm novel functions of involved signaling pathways and comment on efficacy of intervention therapies amid sepsis. However, few of these studies attempt to elucidate the underlining mechanism in progression of sepsis, while other failed to integrate preliminary findings and describe in a broader view. This review focuses on the important signaling pathways, potential molecular mechanism, and pathway-associated therapy in sepsis. Host-derived molecules interacting with activated cells possess pivotal role for sepsis pathogenesis by dynamic regulation of signaling pathways. Cross-talk and functions of these molecules are also discussed in detail. Lastly, potential novel therapeutic strategies precisely targeting on signaling pathways and molecules are mentioned.
Collapse
Affiliation(s)
- Yun-Yu Zhang
- Department of Pediatric Intensive Care Unit, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Bo-Tao Ning
- Department of Pediatric Intensive Care Unit, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China.
| |
Collapse
|
68
|
Mollah MMI, Choi HW, Yeam I, Lee JM, Kim Y. Salicylic Acid, a Plant Hormone, Suppresses Phytophagous Insect Immune Response by Interrupting HMG-Like DSP1. Front Physiol 2021; 12:744272. [PMID: 34671276 PMCID: PMC8521015 DOI: 10.3389/fphys.2021.744272] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/06/2021] [Indexed: 12/23/2022] Open
Abstract
Salicylic acid is a plant hormone that can mediate various plant physiological processes. Salicylic acid can bind to human high mobility group box 1 (HMGB1) and interrupt its role in mediating immune responses. Dorsal switch protein 1 (DSP1) is an insect homolog of HMGB1. In this study, a DSP1 (Se-DSP1) encoded in Spodoptera exigua, a phytophagous insect, was characterized, and its potential role in immune response was explored. Upon bacterial challenge, Se-DSP1 was localized in the nucleus and released into the hemolymph. The released Se-DSP1 could mediate both cellular and humoral immune responses by activating eicosanoid biosynthesis. Salicylic acid could bind to Se-DSP1 with a high affinity. The immune responses of S. exigua were significantly interrupted by SA feeding. Larvae reared on tomatoes with high endogenous SA levels became more susceptible to entomopathogens. Taken together, these results suggest a tritrophic defensive role of plant SA against phytophagous insects.
Collapse
Affiliation(s)
- Md Mahi Imam Mollah
- Department of Plant Medicals, Andong National University, Andong, South Korea
| | - Hyong Woo Choi
- Department of Plant Medicals, Andong National University, Andong, South Korea
| | - Inhwa Yeam
- Department of Horticulture and Breeding, Andong National University, Andong, South Korea
| | - Je Min Lee
- Department of Horticultural Science, Kyungpook National University, Daegu, South Korea
| | - Yonggyun Kim
- Department of Plant Medicals, Andong National University, Andong, South Korea
| |
Collapse
|
69
|
Ni M, Zhang J, Sosa R, Zhang H, Wang H, Jin D, Crowley K, Naini B, Elaine RF, Busuttil RW, Kupiec-Weglinski JW, Wang X, Zhai Y. T-Cell Immunoglobulin and Mucin Domain-Containing Protein-4 Is Critical for Kupffer Cell Homeostatic Function in the Activation and Resolution of Liver Ischemia Reperfusion Injury. Hepatology 2021; 74:2118-2132. [PMID: 33999437 PMCID: PMC9060306 DOI: 10.1002/hep.31906] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/26/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Liver ischemia reperfusion injury (IRI) remains an unresolved clinical problem. This study dissected roles of liver-resident macrophage Kupffer cells (KCs), with a functional focus on efferocytosis receptor T-cell immunoglobulin and mucin domain-containing protein-4 (TIM-4), in both the activation and resolution of IRI in a murine liver partial warm ischemia model. APPROACH AND RESULTS Fluorescence-activated cell sorting results showed that TIM-4 was expressed exclusively by KCs, but not infiltrating macrophages (iMФs), in IR livers. Anti-TIM-4 antibody depleted TIM-4+ macrophages in vivo, resulting in either alleviation or deterioration of liver IRI, which was determined by the repopulation kinetics of the KC niche with CD11b+ macrophages. To determine the KC-specific function of TIM-4, we reconstituted clodronate-liposome-treated mice with exogenous wild-type or TIM-4-deficient KCs at either 0 hour or 24 hours postreperfusion. TIM-4 deficiency in KCs resulted in not only increases in the severity of liver IRI (at 6 hours postreperfusion), but also impairment of the inflammation resolution (at 7 days postreperfusion). In vitro analysis revealed that TIM-4 promoted KC efferocytosis to regulate their Toll-like receptor response by up-regulating IL-10 and down-regulating TNF-α productions. CONCLUSIONS TIM-4 is critical for KC homeostatic function in both the activation and resolution of liver IRI by efferocytosis.
Collapse
Affiliation(s)
- Ming Ni
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery,Hepatobiliary Center, Key Laboratory of Liver Transplantation of Chinese Academy of Medical Sciences, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jing Zhang
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery
| | - Rebecca Sosa
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA
| | - Hanwen Zhang
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery
| | - Han Wang
- Hepatobiliary Center, Key Laboratory of Liver Transplantation of Chinese Academy of Medical Sciences, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Dan Jin
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery,Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kaitlyn Crowley
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery
| | - Bita Naini
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery
| | - Reed, F. Elaine
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA
| | - Ronald W. Busuttil
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery
| | - Jerzy W. Kupiec-Weglinski
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery
| | - Xuehao Wang
- Hepatobiliary Center, Key Laboratory of Liver Transplantation of Chinese Academy of Medical Sciences, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China,Address for correspondence: Yuan Zhai, MD, PhD. Dumont-UCLA Transplant Center 77-120 CHS, 10833 Le Conte Ave, Los Angeles, CA 90095. Phone: (310) 825-9426; Fax: (310) 267-2367, , Xuehao Wang, MD, Department of Liver Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, P.R.China 210029, Phone: 86-25-68136053; Fax:86-25-84630769,
| | - Yuan Zhai
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery,Address for correspondence: Yuan Zhai, MD, PhD. Dumont-UCLA Transplant Center 77-120 CHS, 10833 Le Conte Ave, Los Angeles, CA 90095. Phone: (310) 825-9426; Fax: (310) 267-2367, , Xuehao Wang, MD, Department of Liver Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, P.R.China 210029, Phone: 86-25-68136053; Fax:86-25-84630769,
| |
Collapse
|
70
|
Berberine-loaded nanostructured lipid carriers mitigate warm hepatic ischemia/reperfusion-induced lesion through modulation of HMGB1/TLR4/NF-κB signaling and autophagy. Biomed Pharmacother 2021; 145:112122. [PMID: 34489150 DOI: 10.1016/j.biopha.2021.112122] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Berberine (BBR) is a known alkaloid that has verified its protective effects against ischemia/reperfusion (I/RN) lesion in multiple organs but its poor oral bioavailability limited its use. Despite the previous works, its possible impact on the warm hepatic I/RN-induced lesion is not clear. Accordingly, a nanostructured lipid carrier of BBR (NLC BBR) was developed for enhancing its efficiency and to inspect its protective mechanistic against warm hepatic I/RN. METHODS NLC BBR formula was evaluated pharmaceutically. Wistar rats were orally pre-treated with either BBR or NLC BBR (100 mg/kg) for 2 weeks followed by hepatic I/RN (30 min/24 h). Biochemical, ELISA, qPCR, western blot, histopathological, and immunohistochemical studies were performed. KEY FINDINGS Optimized NLC BBR was prepared with a particle size of 130 ± 8.3 nm. NLC BBR divulged its aptitude to safeguard the hepatic tissues partly due to anti-inflammatory capacity through downsizing the HMGB1/TLR4/NF-κB trajectory with concomitant rebating of TNF-α, iNOS, COX-2, and MPO content. Furthermore, NLC BBR antiapoptotic trait was confirmed by boosting the prosurvival protein (Bcl-2) and cutting down the pro-apoptotic marker (Bax). Moreover, its antioxidant nature was confirmed by TAC uplifting besides MDA subsiding. On the other hand, NLC BBR action embroiled autophagy flux spiking merit exemplified in Beclin-1 and LC3-II enhancement. Finally, NLC BBR administration ascertained its hepatocyte guarding action by recovering the histopathological ailment and diminishing serum transaminases. CONCLUSION NLC BBR purveyed reasonable shielding mechanisms and subsided incidents contemporaneous to warm hepatic I/RN lesion in part, by moderating HMGB1/TLR4/NF-κB inflammatory signaling, autophagy, and apoptosis.
Collapse
|
71
|
Zhang Y, Li Y, Fu X, Wang P, Wang Q, Meng W, Wang T, Yang J, Chai R. The Detrimental and Beneficial Functions of Macrophages After Cochlear Injury. Front Cell Dev Biol 2021; 9:631904. [PMID: 34458249 PMCID: PMC8385413 DOI: 10.3389/fcell.2021.631904] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 07/14/2021] [Indexed: 12/20/2022] Open
Abstract
Macrophages are the main intrinsic immune cells in the cochlea; they can be activated and play a complicated role after cochlear injury. Many studies have shown that the number of macrophages and their morphological characteristics within the major cochlear partitions undergo significant changes under various pathological conditions including acoustic trauma, ototoxic drug treatment, age-related cochlear degeneration, selective hair cell (HC) and spiral ganglion neuron (SGN) elimination, and surgery. However, the exact role of these macrophages after cochlear injury is still unclear. Regulating the migration and activity of macrophages may be a therapeutic approach to reduce the risk or magnitude of trauma-induced hearing loss, and this review highlights the role of macrophages on the peripheral auditory structures of the cochlea and elucidate the mechanisms of macrophage injury and the strategies to reduce the injury by regulating macrophage.
Collapse
Affiliation(s)
- Yuan Zhang
- MOE Key Laboratory for Developmental Genes and Human Disease, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China.,Department of Otolaryngology Head and Neck, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yiyuan Li
- MOE Key Laboratory for Developmental Genes and Human Disease, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Xiaolong Fu
- MOE Key Laboratory for Developmental Genes and Human Disease, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Pengjun Wang
- Department of Otorhinolaryngology, Affiliated Sixth People's Hospital of Shanghai Jiao Tong University, Shanghai, China
| | - Qin Wang
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Meng
- Department of Otolaryngology Head and Neck, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Tian Wang
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jianming Yang
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Renjie Chai
- MOE Key Laboratory for Developmental Genes and Human Disease, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
72
|
Perrelli A, Retta SF. Polymorphisms in genes related to oxidative stress and inflammation: Emerging links with the pathogenesis and severity of Cerebral Cavernous Malformation disease. Free Radic Biol Med 2021; 172:403-417. [PMID: 34175437 DOI: 10.1016/j.freeradbiomed.2021.06.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/03/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023]
Abstract
Cerebral Cavernous Malformation (CCM) is a cerebrovascular disease of genetic origin affecting 0.5% of the population and characterized by abnormally enlarged and leaky capillaries that predispose to seizures, neurological deficits, and intracerebral hemorrhage (ICH). CCM occurs sporadically or is inherited as dominant condition with incomplete penetrance and highly variable expressivity. Three disease genes have been identified: KRIT1 (CCM1), CCM2 and CCM3. Previous results demonstrated that loss-of-function mutations of CCM genes cause pleiotropic effects, including defective autophagy, altered reactive oxygen species (ROS) homeostasis, and enhanced sensitivity to oxidative stress and inflammatory events, suggesting a novel unifying pathogenetic mechanism, and raising the possibility that CCM disease onset and severity are influenced by the presence of susceptibility and modifier genes. Consistently, genome-wide association studies (GWAS) in large and homogeneous cohorts of patients sharing the familial form of CCM disease and identical mutations in CCM genes have led to the discovery of distinct genetic modifiers of major disease severity phenotypes, such as development of numerous and large CCM lesions, and susceptibility to ICH. This review deals with the identification of genetic modifiers with a significant impact on inter-individual variability in CCM disease onset and severity, including highly polymorphic genes involved in oxidative stress, inflammatory and immune responses, such as cytochrome P450 monooxygenases (CYP), matrix metalloproteinases (MMP), and Toll-like receptors (TLR), pointing to their emerging prognostic value, and opening up new perspectives for risk stratification and personalized medicine strategies.
Collapse
Affiliation(s)
- Andrea Perrelli
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy; CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy.
| | - Saverio Francesco Retta
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy; CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy.
| |
Collapse
|
73
|
Hassan MI, Ali FE, Shalkami AGS. Role of TLR-4/IL-6/TNF-α, COX-II and eNOS/iNOS pathways in the impact of carvedilol against hepatic ischemia reperfusion injury. Hum Exp Toxicol 2021; 40:1362-1373. [PMID: 33655798 DOI: 10.1177/0960327121999442] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
AIM Hepatic ischemia/reperfusion (I/R) injury is a syndrome involved in allograft dysfunction. This work aimed to elucidate carvedilol (CAR) role in hepatic I/R injury. METHODS Male rats were allocated to Sham group, CAR group, I/R group and CAR plus I/R group. Rats subjected to hepatic ischemia for 30 minutes then reperfused for 60 minutes. Oxidative stress markers, inflammatory cytokines and nitric oxide synthases were measured in hepatic tissues. RESULTS Hepatocyte injury following I/R was confirmed by a marked increase in liver enzymes. Also, hepatic I/R increased the contents of malondialdehyde however decreased glutathione contents and activities of antioxidant enzymes. Furthermore, hepatic I/R caused elevation of toll-like receptor-4 (TLR-4) expression and inflammatory mediators levels such as tumor necrosis factor-α, interleukin-6 and cyclooxygenase-II. Hepatic I/R caused down-regulation of endothelial nitric oxide synthase and upregulation of inducible nitric oxide synthase expressions. CAR treatment before hepatic I/R resulted in the restoration of liver enzymes. Administration of CAR caused a significant correction of oxidative stress and inflammation markers as well as modulates the expression of endothelial and inducible nitric oxide synthase. CONCLUSIONS CAR protects liver from I/R injury through reduction of the oxidative stress and inflammation, and modulates endothelial and inducible nitric oxide synthase expressions.
Collapse
Affiliation(s)
- Mohamed Ia Hassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, 195495Al-Azhar University, Assiut, Egypt
| | - Fares Em Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, 195495Al-Azhar University, Assiut, Egypt
| | - Abdel-Gawad S Shalkami
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, 195495Al-Azhar University, Assiut, Egypt
| |
Collapse
|
74
|
Liu H, Man K. New Insights in Mechanisms and Therapeutics for Short- and Long-Term Impacts of Hepatic Ischemia Reperfusion Injury Post Liver Transplantation. Int J Mol Sci 2021; 22:ijms22158210. [PMID: 34360975 PMCID: PMC8348697 DOI: 10.3390/ijms22158210] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 02/07/2023] Open
Abstract
Liver transplantation has been identified as the most effective treatment for patients with end-stage liver diseases. However, hepatic ischemia reperfusion injury (IRI) is associated with poor graft function and poses a risk of adverse clinical outcomes post transplantation. Cell death, including apoptosis, necrosis, ferroptosis and pyroptosis, is induced during the acute phase of liver IRI. The release of danger-associated molecular patterns (DAPMs) and mitochondrial dysfunction resulting from the disturbance of metabolic homeostasis initiates graft inflammation. The inflammation in the short term exacerbates hepatic damage, leading to graft dysfunction and a higher incidence of acute rejection. The subsequent changes in the graft immune environment due to hepatic IRI may result in chronic rejection, cancer recurrence and fibrogenesis in the long term. In this review, we mainly focus on new mechanisms of inflammation initiated by immune activation related to metabolic alteration in the short term during liver IRI. The latest mechanisms of cancer recurrence and fibrogenesis due to the long-term impact of inflammation in hepatic IRI is also discussed. Furthermore, the development of therapeutic strategies, including ischemia preconditioning, pharmacological inhibitors and machine perfusion, for both attenuating acute inflammatory injury and preventing late-phase disease recurrence, will be summarized in the context of clinical, translational and basic research.
Collapse
|
75
|
Activation of the Hippocampal LXRβ Improves Sleep-Deprived Cognitive Impairment by Inhibiting Neuroinflammation. Mol Neurobiol 2021; 58:5272-5288. [PMID: 34278533 DOI: 10.1007/s12035-021-02446-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/06/2021] [Indexed: 10/20/2022]
Abstract
Sleep deprivation (SD) leads to cognitive impairment due to neuroinflammation associated with impaired hippocampal neuronal plasticity and memory processes. Liver X receptors (LXRs), including LXRα and LXRβ isoforms, are crucial for synaptic plasticity and neuroinflammation. However, the potential roles of LXRs in the pathogenesis of cognitive impairment induced by SD remain unclear. We revealed that SD resulted in LXRβ reduction in the hippocampus, which was associated with upregulated expression of high mobility group box 1 (HMGB1)/toll-like receptor 4 (TLR4)/NF-κB p65, and knockdown of hippocampal LXRβ by shRNA (shLXRβ) led to cognitive impairment. GW3965, a dual agonist for both LXRα and LXRβ, ameliorated SD-induced cognitive impairment by inhibiting microglia activation, suppressing HMGB1/TLR4/NF-κB p65 pathway, and ultimately affecting the hippocampal expression of inflammatory cytokines in SD mice. LXRβ knockdown by shLXRβ abrogated the GW3965-mediated inhibition of the HMGB1/TLR4/NF-κB p65 pathway, therefore, abolishing the cognitive improvement. Moreover, inhibition of HMGB1 by glycyrrhizin (GLY) synergistic promoted GW3965-mediated anti-inflammation in activated microglia after lipopolysaccharide (LPS)/ATP stimulation and facilitated the cognitive improvement after GW administration by activating LXRβ. All the data suggested that GW3965 ameliorated impaired cognition in SD mice by suppressing the HMGB1/TLR4/NF-κB p65 pathway followed LXRβ activation. This study correlates a deficit of LXRβ in cognitive dysfunction in SD associated with HMGB1 inflammatory pathway in hippocampus, and LXRs may serve as a potential therapeutic target for cognitive impairment with anti-inflammation.
Collapse
|
76
|
El-Sadek HM, Al-Shorbagy MY, Awny MM, Abdallah DM, El-Abhar HS. Pentoxifylline treatment alleviates kidney ischemia/reperfusion injury: Novel involvement of galectin-3 and ASK-1/JNK & ERK1/2/NF-κB/HMGB-1 trajectories. J Pharmacol Sci 2021; 146:136-148. [PMID: 34030796 DOI: 10.1016/j.jphs.2021.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023] Open
Abstract
Despite the documented renoprotective effect of pentoxifylline (PTX), a non-selective phosphodiesterase-4 inhibitor, the studies appraised only its anti-inflammatory/-oxidant/-apoptotic capacities without assessment of the possible involved trajectories. Here, we evaluated the potential role of galectin-3 and the ASK-1/NF-κB p65 signaling pathway with its upstream/downstream signals in an attempt to unveil part of the cascades involved in the renotherapeutic effect using a renal bilateral ischemia/reperfusion (I/R) model. Rats were randomized into sham-operated, renal I/R (45 min/72 h) and I/R + PTX (100 mg/kg; p.o). Post-treatment with PTX improved renal function and abated serum levels of cystatin C, creatinine, BUN and renal KIM-1 content, effects that were reflected on an improvement of the I/R-induced renal histological changes. On the molecular level, PTX reduced renal contents of galectin-3, ASK-1 with its downstream molecule JNK and ERK1/2, as well as NF-κB p65 and HMGB1. This inhibitory effect extended also to suppress neutrophil infiltration, evidenced by diminishing ICAM-1 and MPO, as well as inflammatory cytokines (TNF-α/IL-18), oxidative stress (MDA/TAC), and caspase-3. The PTX novel renotherapeutic effect involved in part the inhibition of galectin-3 and ASK-1/JNK and ERK1/2/NF-κB/HMGB-1 trajectories to mitigate renal I/R injury and to provide basis for its anti-inflammatory, antioxidant, and anti-apoptotic impacts.
Collapse
Affiliation(s)
- Hagar M El-Sadek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, Giza, 12585, Egypt
| | - Muhammad Y Al-Shorbagy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt; Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman, 4184, United Arab Emirates
| | - Magdy M Awny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, Giza, 12585, Egypt
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Hanan S El-Abhar
- Department of Pharmacology, Toxicology and Biochemistry, Faculty of Pharmaceutical Sciences and Pharmaceutical Industries, Future University in Egypt, Cairo, 84518, Egypt
| |
Collapse
|
77
|
Val‐Blasco A, Gil‐Fernández M, Rueda A, Pereira L, Delgado C, Smani T, Ruiz Hurtado G, Fernández‐Velasco M. Ca 2+ mishandling in heart failure: Potential targets. Acta Physiol (Oxf) 2021; 232:e13691. [PMID: 34022101 DOI: 10.1111/apha.13691] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/14/2022]
Abstract
Ca2+ mishandling is a common feature in several cardiovascular diseases such as heart failure (HF). In many cases, impairment of key players in intracellular Ca2+ homeostasis has been identified as the underlying mechanism of cardiac dysfunction and cardiac arrhythmias associated with HF. In this review, we summarize primary novel findings related to Ca2+ mishandling in HF progression. HF research has increasingly focused on the identification of new targets and the contribution of their role in Ca2+ handling to the progression of the disease. Recent research studies have identified potential targets in three major emerging areas implicated in regulation of Ca2+ handling: the innate immune system, bone metabolism factors and post-translational modification of key proteins involved in regulation of Ca2+ handling. Here, we describe their possible contributions to the progression of HF.
Collapse
Affiliation(s)
| | | | - Angélica Rueda
- Department of Biochemistry Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV‐IPN) México City Mexico
| | - Laetitia Pereira
- INSERM UMR‐S 1180 Laboratory of Ca Signaling and Cardiovascular Physiopathology University Paris‐Saclay Châtenay‐Malabry France
| | - Carmen Delgado
- Instituto de Investigaciones Biomédicas Alberto Sols Madrid Spain
- Department of Metabolism and Cell Signalling Biomedical Research Institute "Alberto Sols" CSIC‐UAM Madrid Spain
| | - Tarik Smani
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV) Madrid Spain
- Department of Medical Physiology and Biophysics University of Seville Seville Spain
- Group of Cardiovascular Pathophysiology Institute of Biomedicine of Seville University Hospital of Virgen del Rocío, University of Seville, CSIC Seville Spain
| | - Gema Ruiz Hurtado
- Cardiorenal Translational Laboratory Institute of Research i+12 University Hospital 12 de Octubre Madrid Spain
- CIBER‐CV University Hospita1 12 de Octubre Madrid Spain
| | - Maria Fernández‐Velasco
- La Paz University Hospital Health Research Institute IdiPAZ Madrid Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV) Madrid Spain
| |
Collapse
|
78
|
Guan Y, Yao W, Yi K, Zheng C, Lv S, Tao Y, Hei Z, Li M. Nanotheranostics for the Management of Hepatic Ischemia-Reperfusion Injury. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2007727. [PMID: 33852769 DOI: 10.1002/smll.202007727] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/21/2021] [Indexed: 06/12/2023]
Abstract
Hepatic ischemia-reperfusion injury (IRI), in which an insufficient oxygen supply followed by reperfusion leads to an inflammatory network and oxidative stress in disease tissue to cause cell death, always occurs after liver transplantations and sections. Although pharmacological treatments favorably prevent or protect the liver against experimental IRI, there have been few successes in clinical applications for patient benefits because of the incomprehension of complicated IRI-induced signaling events as well as short blood circulation time, poor solubility, and severe side reactions of most antioxidants and anti-inflammatory drugs. Nanomaterials can achieve targeted delivery and controllable release of contrast agents and therapeutic drugs in desired hepatic IRI regions for enhanced imaging sensitivity and improved therapeutic effects, emerging as novel alternative approaches for hepatic IRI diagnosis and therapy. In this review, the application of nanotechnology is summarized in the management of hepatic IRI, including nanomaterial-assisted hepatic IRI diagnosis, nanoparticulate systems-mediated remission of reactive oxygen species-induced tissue injury, and nanoparticle-based targeted drug delivery systems for the alleviation of IRI-related inflammation. The current challenges and future perspectives of these nanoenabled strategies for hepatic IRI treatment are also discussed.
Collapse
Affiliation(s)
- Yu Guan
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Weifeng Yao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Chunxiong Zheng
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Shixian Lv
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Ziqing Hei
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| |
Collapse
|
79
|
Shih CP, Kuo CY, Lin YY, Lin YC, Chen HK, Wang H, Chen HC, Wang CH. Inhibition of Cochlear HMGB1 Expression Attenuates Oxidative Stress and Inflammation in an Experimental Murine Model of Noise-Induced Hearing Loss. Cells 2021; 10:810. [PMID: 33916471 PMCID: PMC8066810 DOI: 10.3390/cells10040810] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/22/2021] [Accepted: 04/02/2021] [Indexed: 02/06/2023] Open
Abstract
Noise-induced hearing loss (NIHL) is a common inner ear disease but has complex pathological mechanisms, one of which is increased oxidative stress in the cochlea. The high-mobility group box 1 (HMGB1) protein acts as an inflammatory mediator and shows different activities with redox modifications linked to the generation of reactive oxygen species (ROS). We aimed to investigate whether manipulation of cochlear HMGB1 during noise exposure could prevent noise-induced oxidative stress and hearing loss. Sixty CBA/CaJ mice were divided into two groups. An intraperitoneal injection of anti-HMGB1 antibodies was administered to the experimental group; the control group was injected with saline. Thirty minutes later, all mice were subjected to white noise exposure. Subsequent cochlear damage, including auditory threshold shifts, hair cell loss, expression of cochlear HMGB1, and free radical activity, was then evaluated. The levels of HMGB1 and 4-hydroxynonenal (4-HNE), as respective markers of reactive nitrogen species (RNS) and ROS formation, showed slight increases on post-exposure day 1 and achieved their highest levels on post-exposure day 4. After noise exposure, the antibody-treated mice showed markedly less ROS formation and lower expression of NADPH oxidase 4 (NOX4), nitrotyrosine, inducible nitric oxide synthase (iNOS), and intercellular adhesion molecule-1 (ICAM-1) than the saline-treated control mice. A significant amelioration was also observed in the threshold shifts of the auditory brainstem response and the loss of outer hair cells in the antibody-treated versus the saline-treated mice. Our results suggest that inhibition of HMGB1 by neutralization with anti-HMGB1 antibodies prior to noise exposure effectively attenuated oxidative stress and subsequent inflammation. This procedure could therefore have potential as a therapy for NIHL.
Collapse
Affiliation(s)
- Cheng-Ping Shih
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical Center, Tri-Service General Hospital, Taipei 11490, Taiwan; (C.-P.S.); (C.-Y.K.); (Y.-Y.L.); (H.-K.C.); (H.W.)
| | - Chao-Yin Kuo
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical Center, Tri-Service General Hospital, Taipei 11490, Taiwan; (C.-P.S.); (C.-Y.K.); (Y.-Y.L.); (H.-K.C.); (H.W.)
| | - Yuan-Yung Lin
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical Center, Tri-Service General Hospital, Taipei 11490, Taiwan; (C.-P.S.); (C.-Y.K.); (Y.-Y.L.); (H.-K.C.); (H.W.)
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Yi-Chun Lin
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Hang-Kang Chen
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical Center, Tri-Service General Hospital, Taipei 11490, Taiwan; (C.-P.S.); (C.-Y.K.); (Y.-Y.L.); (H.-K.C.); (H.W.)
- Taichung Armed Forces General Hospital, Taichung 41168, Taiwan
| | - Hao Wang
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical Center, Tri-Service General Hospital, Taipei 11490, Taiwan; (C.-P.S.); (C.-Y.K.); (Y.-Y.L.); (H.-K.C.); (H.W.)
| | - Hsin-Chien Chen
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical Center, Tri-Service General Hospital, Taipei 11490, Taiwan; (C.-P.S.); (C.-Y.K.); (Y.-Y.L.); (H.-K.C.); (H.W.)
| | - Chih-Hung Wang
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical Center, Tri-Service General Hospital, Taipei 11490, Taiwan; (C.-P.S.); (C.-Y.K.); (Y.-Y.L.); (H.-K.C.); (H.W.)
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan;
- Taichung Armed Forces General Hospital, Taichung 41168, Taiwan
- Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei 11490, Taiwan
| |
Collapse
|
80
|
Walsh SA, Hoyt BW, Rowe CJ, Dey D, Davis TA. Alarming Cargo: The Role of Exosomes in Trauma-Induced Inflammation. Biomolecules 2021; 11:biom11040522. [PMID: 33807302 PMCID: PMC8065643 DOI: 10.3390/biom11040522] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
Severe polytraumatic injury initiates a robust immune response. Broad immune dysfunction in patients with such injuries has been well-documented; however, early biomarkers of immune dysfunction post-injury, which are critical for comprehensive intervention and can predict the clinical course of patients, have not been reported. Current circulating markers such as IL-6 and IL-10 are broad, non-specific, and lag behind the clinical course of patients. General blockade of the inflammatory response is detrimental to patients, as a certain degree of regulated inflammation is critical and necessary following trauma. Exosomes, small membrane-bound extracellular vesicles, found in a variety of biofluids, carry within them a complex functional cargo, comprised of coding and non-coding RNAs, proteins, and metabolites. Composition of circulating exosomal cargo is modulated by changes in the intra- and extracellular microenvironment, thereby serving as a homeostasis sensor. With its extensively documented involvement in immune regulation in multiple pathologies, study of exosomal cargo in polytrauma patients can provide critical insights on trauma-specific, temporal immune dysregulation, with tremendous potential to serve as unique biomarkers and therapeutic targets for timely and precise intervention.
Collapse
Affiliation(s)
- Sarah A. Walsh
- USU Walter Reed Surgery, Uniformed Services University, Bethesda, MD 20814, USA; (S.A.W.); (B.W.H.); (C.J.R.); (D.D.)
| | - Benjamin W. Hoyt
- USU Walter Reed Surgery, Uniformed Services University, Bethesda, MD 20814, USA; (S.A.W.); (B.W.H.); (C.J.R.); (D.D.)
| | - Cassie J. Rowe
- USU Walter Reed Surgery, Uniformed Services University, Bethesda, MD 20814, USA; (S.A.W.); (B.W.H.); (C.J.R.); (D.D.)
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Devaveena Dey
- USU Walter Reed Surgery, Uniformed Services University, Bethesda, MD 20814, USA; (S.A.W.); (B.W.H.); (C.J.R.); (D.D.)
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Thomas A. Davis
- USU Walter Reed Surgery, Uniformed Services University, Bethesda, MD 20814, USA; (S.A.W.); (B.W.H.); (C.J.R.); (D.D.)
- Correspondence:
| |
Collapse
|
81
|
Mollah MMI, Ahmed S, Kim Y. Immune mediation of HMG-like DSP1 via Toll-Spätzle pathway and its specific inhibition by salicylic acid analogs. PLoS Pathog 2021; 17:e1009467. [PMID: 33765093 PMCID: PMC8023496 DOI: 10.1371/journal.ppat.1009467] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 04/06/2021] [Accepted: 03/11/2021] [Indexed: 12/22/2022] Open
Abstract
Xenorhabdus hominickii, an entomopathogenic bacterium, inhibits eicosanoid biosynthesis of target insects to suppress their immune responses by inhibiting phospholipase A2 (PLA2) through binding to a damage-associated molecular pattern (DAMP) molecule called dorsal switch protein 1 (DSP1) from Spodoptera exigua, a lepidopteran insect. However, the signalling pathway between DSP1 and PLA2 remains unknown. The objective of this study was to determine whether DSP1 could activate Toll immune signalling pathway to activate PLA2 activation and whether X. hominickii metabolites could inhibit DSP1 to shutdown eicosanoid biosynthesis. Toll-Spätzle (Spz) signalling pathway includes two Spz (SeSpz1 and SeSpz2) and 10 Toll receptors (SeToll1-10) in S. exigua. Loss-of-function approach using RNA interference showed that SeSpz1 and SeToll9 played crucial roles in connecting DSP1 mediation to activate PLA2. Furthermore, a deletion mutant against SeToll9 using CRISPR/Cas9 abolished DSP1 mediation and induced significant immunosuppression. Organic extracts of X. hominickii culture broth could bind to DSP1 at a low micromolar range. Subsequent sequential fractionations along with binding assays led to the identification of seven potent compounds including 3-ethoxy-4-methoxyphenol (EMP). EMP could bind to DSP1 and prevent its translocation to plasma in response to bacterial challenge and suppress the up-regulation of PLA2 activity. These results suggest that X. hominickii inhibits DSP1 and prevents its DAMP role in activating Toll immune signalling pathway including PLA2 activation, leading to significant immunosuppression of target insects. Immune responses of insects are highly effective in defending various entomopathogens. Xenorhabdus hominickii is an entomopathogenic bacterium that uses a pathogenic strategy of suppressing host insect immunity by inhibiting phospholipase A2 (PLA2) which catalyzes the committed step for eicosanoid biosynthesis. Eicosanoids mediate both cellular and humoral immune responses in insects. This study discovers an upstream signalling pathway to activate PLA2 in response to bacterial challenge. Se-DSP1 is an insect homolog of vertebrate HMGB1 that acts as a damage-associated molecular pattern. Upon bacterial infection, Se-DSP1 is released to the circulatory system to activate Spätzle, an insect cytokine that can bind to Toll receptor. Toll immune signalling pathway can activate antimicrobial peptide gene expression and PLA2. A deletion mutant against a Toll gene abolished immune responses mediated by Se-DSP1. Indeed, X. hominickii can produce and secrete secondary metabolites including salicylic acid analogs that can strongly bind to Se-DSP1. These bacterial metabolites prevented the release of Se-DSP1, which impaired the activation of PLA2 and resulted in a significant immunosuppression of target insects against bacterial infection.
Collapse
Affiliation(s)
- Md. Mahi Imam Mollah
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong, Korea
| | - Shabbir Ahmed
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong, Korea
| | - Yonggyun Kim
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong, Korea
- * E-mail:
| |
Collapse
|
82
|
Rosa AC, Corsi D, Cavi N, Bruni N, Dosio F. Superoxide Dismutase Administration: A Review of Proposed Human Uses. Molecules 2021; 26:1844. [PMID: 33805942 PMCID: PMC8037464 DOI: 10.3390/molecules26071844] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Superoxide dismutases (SODs) are metalloenzymes that play a major role in antioxidant defense against oxidative stress in the body. SOD supplementation may therefore trigger the endogenous antioxidant machinery for the neutralization of free-radical excess and be used in a variety of pathological settings. This paper aimed to provide an extensive review of the possible uses of SODs in a range of pathological settings, as well as describe the current pitfalls and the delivery strategies that are in development to solve bioavailability issues. We carried out a PubMed query, using the keywords "SOD", "SOD mimetics", "SOD supplementation", which included papers published in the English language, between 2012 and 2020, on the potential therapeutic applications of SODs, including detoxification strategies. As highlighted in this paper, it can be argued that the generic antioxidant effects of SODs are beneficial under all tested conditions, from ocular and cardiovascular diseases to neurodegenerative disorders and metabolic diseases, including diabetes and its complications and obesity. However, it must be underlined that clinical evidence for its efficacy is limited and consequently, this efficacy is currently far from being demonstrated.
Collapse
Affiliation(s)
- Arianna Carolina Rosa
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via P. Giuria 9, 10125 Turin, Italy; (D.C.); (N.C.); (F.D.)
| | - Daniele Corsi
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via P. Giuria 9, 10125 Turin, Italy; (D.C.); (N.C.); (F.D.)
| | - Niccolò Cavi
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via P. Giuria 9, 10125 Turin, Italy; (D.C.); (N.C.); (F.D.)
| | - Natascia Bruni
- Istituto Farmaceutico Candioli, Strada Comunale di None, 1, 10092 Beinasco, Italy;
| | - Franco Dosio
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via P. Giuria 9, 10125 Turin, Italy; (D.C.); (N.C.); (F.D.)
| |
Collapse
|
83
|
Ren J, He J, Zhang H, Xia Y, Hu Z, Loughran P, Billiar T, Huang H, Tsung A. Platelet TLR4-ERK5 Axis Facilitates NET-Mediated Capturing of Circulating Tumor Cells and Distant Metastasis after Surgical Stress. Cancer Res 2021; 81:2373-2385. [PMID: 33687949 DOI: 10.1158/0008-5472.can-20-3222] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/21/2021] [Accepted: 03/05/2021] [Indexed: 11/16/2022]
Abstract
Surgical removal of malignant tumors is a mainstay in controlling most solid cancers. However, surgical insult also increases the risk of tumor recurrence and metastasis. Tissue trauma activates the innate immune system locally and systemically, mounting an inflammatory response. Platelets and neutrophils are two crucial players in the early innate immune response that heals tissues, but their actions may also contribute to cancer cell dissemination and distant metastasis. Here we report that surgical stress-activated platelets enhance the formation of platelet-tumor cell aggregates, facilitating their entrapment by neutrophil extracellular traps (NET) and subsequent distant metastasis. A murine hepatic ischemia/reperfusion (I/R) injury model of localized surgical stress showed that I/R promotes capturing of aggregated circulating tumor cells (CTC) by NETs and eventual metastasis to the lungs, which are abrogated when platelets are depleted. Hepatic I/R also increased deposition of NETs within the lung microvasculature, but depletion of platelets had no effect. TLR4 was essential for platelet activation and platelet-tumor cell aggregate formation in an ERK5-GPIIb/IIIa integrin-dependent manner. Such aggregation facilitated NET-mediated capture of CTCs in vitro under static and dynamic conditions. Blocking platelet activation or knocking out TLR4 protected mice from hepatic I/R-induced metastasis with no CTC entrapment by NETs. These results uncover a novel mechanism where platelets and neutrophils contribute to metastasis in the setting of acute inflammation. Targeted disruption of the interaction between platelets and NETs holds therapeutic promise to prevent postoperative distant metastasis. SIGNIFICANCE: Targeting platelet activation via TLR4/ERK5/integrin GPIIb/IIIa signaling shows potential for preventing NET-driven distant metastasis in patients post-resection.
Collapse
Affiliation(s)
- Jinghua Ren
- Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio.,Cancer center, Union Hospital, Huazhong University of Science and Technology, Wuhan, P.R. China.,Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jiayi He
- Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio.,Department of Pediatrics, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Hongji Zhang
- Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio.,Department of Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yujia Xia
- Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio.,Department of Gastroenterology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Zhiwei Hu
- Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio
| | - Patricia Loughran
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,Center for Biologic Imaging, Department of Cell Biology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Timothy Billiar
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Hai Huang
- Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio.
| | - Allan Tsung
- Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio.
| |
Collapse
|
84
|
Sosa RA, Terry AQ, Kaldas FM, Jin YP, Rossetti M, Ito T, Li F, Ahn RS, Naini BV, Groysberg VM, Zheng Y, Aziz A, Nevarez-Mejia J, Zarrinpar A, Busuttil RW, Gjertson DW, Kupiec-Weglinski JW, Reed EF. Disulfide High-Mobility Group Box 1 Drives Ischemia-Reperfusion Injury in Human Liver Transplantation. Hepatology 2021; 73:1158-1175. [PMID: 32426849 PMCID: PMC8722704 DOI: 10.1002/hep.31324] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/16/2020] [Accepted: 04/19/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Sterile inflammation is a major clinical concern during ischemia-reperfusion injury (IRI) triggered by traumatic events, including stroke, myocardial infarction, and solid organ transplantation. Despite high-mobility group box 1 (HMGB1) clearly being involved in sterile inflammation, its role is controversial because of a paucity of patient-focused research. APPROACH AND RESULTS Here, we examined the role of HMGB1 oxidation states in human IRI following liver transplantation. Portal blood immediately following allograft reperfusion (liver flush; LF) had increased total HMGB1, but only LF from patients with histopathological IRI had increased disulfide-HMGB1 and induced Toll-like receptor 4-dependent tumor necrosis factor alpha production by macrophages. Disulfide HMGB1 levels increased concomitantly with IRI severity. IRI+ prereperfusion biopsies contained macrophages with hyperacetylated, lysosomal disulfide-HMGB1 that increased postreperfusion at sites of injury, paralleling increased histone acetyltransferase general transcription factor IIIC subunit 4 and decreased histone deacetylase 5 expression. Purified disulfide-HMGB1 or IRI+ blood stimulated further production of disulfide-HMGB1 and increased proinflammatory molecule and cytokine expression in macrophages through a positive feedback loop. CONCLUSIONS These data identify disulfide-HMGB1 as a mechanistic biomarker of, and therapeutic target for, minimizing sterile inflammation during human liver IRI.
Collapse
Affiliation(s)
- Rebecca A. Sosa
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Allyson Q. Terry
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Fady M. Kaldas
- Dumont-UCLA Transplantation Center, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Yi-Ping Jin
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Maura Rossetti
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Takahiro Ito
- Dumont-UCLA Transplantation Center, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Fang Li
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Richard S. Ahn
- Institute of Quantitative and Computational Biosciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Bita V. Naini
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Victoria M. Groysberg
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Ying Zheng
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Antony Aziz
- Dumont-UCLA Transplantation Center, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Jessica Nevarez-Mejia
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Ali Zarrinpar
- Dumont-UCLA Transplantation Center, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Ronald W. Busuttil
- Dumont-UCLA Transplantation Center, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - David W. Gjertson
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Department of Biostatistics, School of Public Health at UCLA, Los Angeles, CA, 90095, USA
| | - Jerzy W. Kupiec-Weglinski
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Dumont-UCLA Transplantation Center, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| |
Collapse
|
85
|
Complement 5 Inhibition Ameliorates Hepatic Ischemia/reperfusion Injury in Mice, Dominantly via the C5a-mediated Cascade. Transplantation 2021; 104:2065-2077. [PMID: 32384381 DOI: 10.1097/tp.0000000000003302] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Hepatic ischemia/reperfusion injury (IRI) is a serious complication in liver surgeries, including transplantation. Complement activation seems to be closely involved in hepatic IRI; however, no complement-targeted intervention has been clinically applied. We investigated the therapeutic potential of Complement 5 (C5)-targeted regulation in hepatic IRI. METHODS C5-knockout (B10D2/oSn) and their corresponding wild-type mice (WT, B10D2/nSn) were exposed to 90-minute partial (70%) hepatic ischemia/reperfusion with either anti-mouse-C5 monoclonal antibody (BB5.1) or corresponding control immunoglobulin administration 30 minutes before ischemia. C5a receptor 1 antagonist was also given to WT to identify which cascade, C5a or C5b-9, is dominant. RESULTS C5-knockout and anti-C5-Ab administration to WT both significantly reduced serum transaminase release and histopathological damages from 2 hours after reperfusion. This improvement was characterized by significantly reduced CD41+ platelet aggregation, maintained F4/80+ cells, and decreased high-mobility group box 1 release. After 6 hours of reperfusion, the infiltration of CD11+ and Ly6-G+ cells, cytokine/chemokine expression, single-stranded DNA+ cells, and cleaved caspase-3 expression were all significantly alleviated by anti-C5-Ab. C5a receptor 1 antagonist was as effective as anti-C5-Ab for reducing transaminases. CONCLUSIONS Anti-C5 antibody significantly ameliorated hepatic IRI, predominantly via the C5a-mediated cascade, not only by inhibiting platelet aggregation during the early phase but also by attenuating the activation of infiltrating macrophages/neutrophils and hepatocyte apoptosis in the late phase of reperfusion. Given its efficacy, clinical availability, and controllability, C5-targeted intervention may provide a novel therapeutic strategy against hepatic IRI.
Collapse
|
86
|
Nakamura K, Kageyama S, Kaldas FM, Hirao H, Ito T, Kadono K, Dery KJ, Kojima H, Gjertson DW, Sosa RA, Kujawski M, Busuttil RW, Reed EF, Kupiec-Weglinski JW. Hepatic CEACAM1 expression indicates donor liver quality and prevents early transplantation injury. J Clin Invest 2021; 130:2689-2704. [PMID: 32027621 DOI: 10.1172/jci133142] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/30/2020] [Indexed: 12/16/2022] Open
Abstract
Although CEACAM1 (CC1) glycoprotein resides at the interface of immune liver injury and metabolic homeostasis, its role in orthotopic liver transplantation (OLT) remains elusive. We aimed to determine whether/how CEACAM1 signaling may affect hepatic ischemia-reperfusion injury (IRI) and OLT outcomes. In the mouse, donor liver CC1 null mutation augmented IRI-OLT (CC1-KO→WT) by enhancing ROS expression and HMGB1 translocation during cold storage, data supported by in vitro studies where hepatic flush from CC1-deficient livers enhanced macrophage activation in bone marrow-derived macrophage cultures. Although hepatic CC1 deficiency augmented cold stress-triggered ASK1/p-p38 upregulation, adjunctive ASK1 inhibition alleviated IRI and improved OLT survival by suppressing p-p38 upregulation, ROS induction, and HMGB1 translocation (CC1-KO→WT), whereas ASK1 silencing (siRNA) promoted cytoprotection in cold-stressed and damage-prone CC1-deficient hepatocyte cultures. Consistent with mouse data, CEACAM1 expression in 60 human donor liver biopsies correlated negatively with activation of the ASK1/p-p38 axis, whereas low CC1 levels associated with increased ROS and HMGB1 translocation, enhanced innate and adaptive immune responses, and inferior early OLT function. Notably, reduced donor liver CEACAM1 expression was identified as one of the independent predictors for early allograft dysfunction (EAD) in human OLT patients. Thus, as a checkpoint regulator of IR stress and sterile inflammation, CEACAM1 may be considered as a denominator of donor hepatic tissue quality, and a target for therapeutic modulation in OLT recipients.
Collapse
Affiliation(s)
- Kojiro Nakamura
- Department of Surgery, Division of Liver and Pancreas Transplantation, Dumont-UCLA Liver Transplant Center
| | - Shoichi Kageyama
- Department of Surgery, Division of Liver and Pancreas Transplantation, Dumont-UCLA Liver Transplant Center
| | - Fady M Kaldas
- Department of Surgery, Division of Liver and Pancreas Transplantation, Dumont-UCLA Liver Transplant Center
| | - Hirofumi Hirao
- Department of Surgery, Division of Liver and Pancreas Transplantation, Dumont-UCLA Liver Transplant Center
| | - Takahiro Ito
- Department of Surgery, Division of Liver and Pancreas Transplantation, Dumont-UCLA Liver Transplant Center
| | - Kentaro Kadono
- Department of Surgery, Division of Liver and Pancreas Transplantation, Dumont-UCLA Liver Transplant Center
| | - Kenneth J Dery
- Department of Surgery, Division of Liver and Pancreas Transplantation, Dumont-UCLA Liver Transplant Center
| | - Hidenobu Kojima
- Department of Surgery, Division of Liver and Pancreas Transplantation, Dumont-UCLA Liver Transplant Center
| | - David W Gjertson
- Department of Biostatistics, UCLA School of Public Health.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Rebecca A Sosa
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Maciej Kujawski
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Ronald W Busuttil
- Department of Surgery, Division of Liver and Pancreas Transplantation, Dumont-UCLA Liver Transplant Center
| | - Elaine F Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jerzy W Kupiec-Weglinski
- Department of Surgery, Division of Liver and Pancreas Transplantation, Dumont-UCLA Liver Transplant Center
| |
Collapse
|
87
|
Shivappagowdar A, Garg S, Srivastava A, Hada RS, Kalia I, Singh AP, Garg LC, Pati S, Singh S. Pathogenic Pore Forming Proteins of Plasmodium Triggers the Necrosis of Endothelial Cells Attributed to Malaria Severity. Toxins (Basel) 2021; 13:toxins13010062. [PMID: 33467515 PMCID: PMC7839052 DOI: 10.3390/toxins13010062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/05/2020] [Accepted: 11/08/2020] [Indexed: 11/17/2022] Open
Abstract
Severe malaria caused by Plasmodium falciparum poses a major global health problem with high morbidity and mortality. P. falciparum harbors a family of pore-forming proteins (PFPs), known as perforin like proteins (PLPs), which are structurally equivalent to prokaryotic PFPs. These PLPs are secreted from the parasites and, they contribute to disease pathogenesis by interacting with host cells. The severe malaria pathogenesis is associated with the dysfunction of various barrier cells, including endothelial cells (EC). Several factors, including PLPs secreted by parasites, contribute to the host cell dysfunction. Herein, we have tested the hypothesis that PLPs mediate dysfunction of barrier cells and might have a role in disease pathogenesis. We analyzed various dysfunctions in barrier cells following rPLP2 exposure and demonstrate that it causes an increase in intracellular Ca2+ levels. Additionally, rPLP2 exposed barrier cells displayed features of cell death, including Annexin/PI positivity, depolarized the mitochondrial membrane potential, and ROS generation. We have further performed the time-lapse video microscopy of barrier cells and found that the treatment of rPLP2 triggers their membrane blebbing. The cytoplasmic localization of HMGB1, a marker of necrosis, further confirmed the necrotic type of cell death. This study highlights the role of parasite factor PLP in endothelial dysfunction and provides a rationale for the design of adjunct therapies against severe malaria.
Collapse
Affiliation(s)
- Abhishek Shivappagowdar
- Department of Life Science, School of Natural Sciences, Shiv Nadar University, Chithera, Gautam Buddha Nagar, Uttar Pradesh 201314, India; (A.S.); (A.S.); (R.S.H.); (S.P.)
| | - Swati Garg
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India;
| | - Akriti Srivastava
- Department of Life Science, School of Natural Sciences, Shiv Nadar University, Chithera, Gautam Buddha Nagar, Uttar Pradesh 201314, India; (A.S.); (A.S.); (R.S.H.); (S.P.)
| | - Rahul S. Hada
- Department of Life Science, School of Natural Sciences, Shiv Nadar University, Chithera, Gautam Buddha Nagar, Uttar Pradesh 201314, India; (A.S.); (A.S.); (R.S.H.); (S.P.)
| | - Inderjeet Kalia
- Infectious Disease Lab, National Institute of Immunology, New Delhi 110067, India; (I.K.); (A.P.S.)
| | - Agam P. Singh
- Infectious Disease Lab, National Institute of Immunology, New Delhi 110067, India; (I.K.); (A.P.S.)
| | - Lalit C. Garg
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi 110067, India;
| | - Soumya Pati
- Department of Life Science, School of Natural Sciences, Shiv Nadar University, Chithera, Gautam Buddha Nagar, Uttar Pradesh 201314, India; (A.S.); (A.S.); (R.S.H.); (S.P.)
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India;
- Correspondence:
| |
Collapse
|
88
|
Functional crosstalk between myeloid Foxo1-β-catenin axis and Hedgehog/Gli1 signaling in oxidative stress response. Cell Death Differ 2020; 28:1705-1719. [PMID: 33288903 PMCID: PMC8167164 DOI: 10.1038/s41418-020-00695-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023] Open
Abstract
Foxo1 transcription factor is an evolutionarily conserved regulator of cell metabolism, oxidative stress, inflammation, and apoptosis. Activation of Hedgehog/Gli signaling is known to regulate cell growth, differentiation, and immune function. However, the molecular mechanisms by which interactive cell signaling networks restrain oxidative stress response and necroptosis are still poorly understood. Here, we report that myeloid-specific Foxo1 knockout (Foxo1M-KO) mice were resistant to oxidative stress-induced hepatocellular damage with reduced macrophage/neutrophil infiltration, and proinflammatory mediators in liver ischemia/reperfusion injury (IRI). Foxo1M-KO enhanced β-catenin-mediated Gli1/Snail activity, and reduced receptor-interacting protein kinase 3 (RIPK3) and NIMA-related kinase 7 (NEK7)/NLRP3 expression in IR-stressed livers. Disruption of Gli1 in Foxo1M-KO livers deteriorated liver function, diminished Snail, and augmented RIPK3 and NEK7/NLRP3. Mechanistically, macrophage Foxo1 and β-catenin colocalized in the nucleus, whereby the Foxo1 competed with T-cell factor (TCF) for interaction with β-catenin under inflammatory conditions. Disruption of the Foxo1–β-catenin axis by Foxo1 deletion enhanced β-catenin/TCF binding, activated Gli1/Snail signaling, leading to inhibited RIPK3 and NEK7/NLRP3. Furthermore, macrophage Gli1 or Snail knockout activated RIPK3 and increased hepatocyte necroptosis, while macrophage RIPK3 ablation diminished NEK7/NLRP3-driven inflammatory response. Our findings underscore a novel molecular mechanism of the myeloid Foxo1–β-catenin axis in regulating Hedgehog/Gli1 function that is key in oxidative stress-induced liver inflammation and necroptosis.
Collapse
|
89
|
Chen R, Huang Y, Quan J, Liu J, Wang H, Billiar TR, Lotze MT, Zeh HJ, Kang R, Tang D. HMGB1 as a potential biomarker and therapeutic target for severe COVID-19. Heliyon 2020; 6:e05672. [PMID: 33313438 PMCID: PMC7720697 DOI: 10.1016/j.heliyon.2020.e05672] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/26/2020] [Accepted: 12/03/2020] [Indexed: 01/08/2023] Open
Abstract
COVID-19 has attracted global attention due to its rapid spread around the world with substantial morbidity and associated mortality. Severe COVID-19 can be complicated by the acute respiratory distress syndrome, sepsis and septic shock leading to death. These complications are thought to result from an overactivation of the immune system, leading to a cytokine storm syndrome associated with multiple organ failure. Here, we report that high mobility group box 1 (HMGB1), a prototypical damage-associated molecular pattern (DAMP) and a central mediator of lethal inflammation, could be a potential target for innovative therapeutic strategies for COVID-19. Serum HMGB1 in severe COVID-19 patients is elevated (189.40 ± 140.88 ng/ml). Exogenous HMGB1 induces the expression of SARS-CoV-2 entry receptor ACE2 in alveolar epithelial cells in an AGER-dependent manner. Importantly, genetic (using AGER siRNA) or pharmacological (using glycyrrhizin, chloroquine, hydroxychloroquine, and FPS-ZM1) inhibition of the HMGB1-AGER pathway blocks ACE2 expression. Thus, HMGB1 inhibitors are likewise promising drug candidates for the treatment of patients suffering from COVID-19.
Collapse
Affiliation(s)
- Ruochan Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yan Huang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jun Quan
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jiao Liu
- The Third Affiliated Hospital, Guangzhou Medical University, Guangdong 510600, China
| | - Haichao Wang
- Laboratory of Emergency Medicine, North Shore University Hospital, Feinstein Institute for Medical Research, Manhasset, New York 11030, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | - Herbert J Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
90
|
Abstract
Bone homeostasis is maintained by a balance in the levels of osteoclast and osteoblast activity. Osteoclasts are bone-resorbing cells and have been shown to act as key players in various osteolytic diseases. Osteoclasts differentiate from monocyte/macrophage lineage cells in the presence of receptor activator of nuclear factor-κB ligand and macrophage colony-stimulating factor. Osteoblasts support osteoclastogenesis by producing several osteoclast differentiation factors. Toll-like receptors (TLRs) are members of the pattern recognition receptor family that are involved in recognizing pathogen-associated molecular patterns and damage-associated molecular patterns in response to pathogen infection. TLRs regulate osteoclastogenesis and bone resorption through either the myeloid differentiation primary response 88 or the Toll/interleukin-1 receptor domain-containing adapter-inducing interferon-β signaling pathways. Since osteoclasts play a central role in the progression of osteolytic diseases, extensive research focusing on TLR downstream signaling in these cells should be conducted to advance the development of effective TLR modulators. In this review, we summarize the currently available information on the role of TLRs in osteoclast differentiation and osteolytic diseases.
Collapse
Affiliation(s)
- Mijung Yim
- College of Pharmacy, Sookmyung Women's University, Seoul, Korea
| |
Collapse
|
91
|
Octreotide and melatonin alleviate inflammasome-induced pyroptosis through inhibition of TLR4-NF-κB-NLRP3 pathway in hepatic ischemia/reperfusion injury. Toxicol Appl Pharmacol 2020; 410:115340. [PMID: 33264646 DOI: 10.1016/j.taap.2020.115340] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 11/07/2020] [Accepted: 11/13/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIM The Toll-like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB)/NLRP3 inflammasome signaling pathway is essential in the pathogenesis of hepatic ischemia/ reperfusion (HIR) injury. Pyroptosis is a proinflammatory programmed cell death that is related to several diseases. Thus, the purpose of this study was to examine whether pretreatment with octreotide (somatostatin analogue, OCT) at different doses or OCT at 75μg/kg combined with melatonin (N-acetyl-5-methoxytryptamine, MLT) can alleviate HIR injury via targeting NLRP3 inflammasome-induced pyroptosis in a TLR4/MyD88/NF-κB dependent manner. METHODS Rats were randomized into sham, HIR, OCT (50, 75, and 100 µg/kg), MLT, and MLT + OCT75 groups. Ischemia was induced via occlusion of the portal triad for 30 min followed by 24 h reperfusion. RESULTS OCT pretreatment at doses (50 or 75 μg/kg), MLT alone, and MLT + OCT75 significantly ameliorated the biochemical with histopathological changes, oxidative stress, inflammation, apoptosis, then augmented anti-oxidant and anti-apoptotic markers through downregulation of HMGB1, TLR4, MyD88, TRAF-6, p-IκBα (S32), p-NF-κBp65 (S536), NLRP3, ASC, caspase-1(p20), and GSDMD-N expressions compared with HIR group. CONCLUSION OCT at doses (50 or 75 µg/kg) showed for the first time a hepatoprotective effect against HIR injury via inhibiting TLR4-NLRP3-mediated pyroptosis in rats. As well, OCT75 was more effective than OCT50 or MLT alone, and its effect was not enhanced after the addition of MLT, through downregulation of TLR4/MyD88/NF-κB/NLRP3 inflammasome pathway.
Collapse
|
92
|
Acetaminophen-Induced Rat Hepatotoxicity Based on M1/M2-Macrophage Polarization, in Possible Relation to Damage-Associated Molecular Patterns and Autophagy. Int J Mol Sci 2020; 21:ijms21238998. [PMID: 33256230 PMCID: PMC7730394 DOI: 10.3390/ijms21238998] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/20/2020] [Accepted: 11/20/2020] [Indexed: 02/07/2023] Open
Abstract
Overdose of acetaminophen (APAP), an antipyretic drug, is an important cause of liver injury. However, the mechanism in the rat model remains undetermined. We analyzed APAP-induced hepatotoxicity using rats based on M1/M2-macrophage functions in relation to damage-associated molecular patterns (DAMPs) and autophagy. Liver samples from six-week-old rats injected with APAP (1000 mg/kg BW, ip, once) after 15 h fasting were collected at hour 10, and on days 1, 2, 3, and 5. Liver lesions consisting of coagulation necrosis and inflammation were seen in the affected centrilobular area on days 1 and 2, and then, recovered with reparative fibrosis by day 5. Liver exudative enzymes increased transiently on day 1. CD68+ M1-macrophages increased significantly on days 1 and 2 with increased mRNAs of M1-related cytokines such as IFN-g and TNF-α, whereas CD163+ M2-macrophages appeared later on days 2 and 3. Macrophages reacting to MHC class II and Iba1 showed M1-type polarization, and CD204+ macrophages tended to be polarized toward M2-type. At hour 10, interestingly, HMGB1 (representative DAMPs) and its related signals, TLR-9 and MyD88, as well as LC3B+ autophagosomes began to increase. Collectively, the pathogenesis of rat APAP hepatotoxicity, which is the first, detailed report for a rat model, might be influenced by macrophage functions of M1 type for tissue injury/inflammation and M2-type for anti-inflammatory/fibrosis; particularly, M1-type may function in relation to DAMPs and autophagy. Understanding the interplayed mechanisms would provide new insight into hepato-pathogenesis and contribute to the possible development of therapeutic strategies.
Collapse
|
93
|
Niu L, Yang W, Duan L, Wang X, Li Y, Xu C, Liu C, Zhang Y, Zhou W, Liu J, Zhao Q, Han Y, Hong L, Fan D. Biological functions and theranostic potential of HMGB family members in human cancers. Ther Adv Med Oncol 2020; 12:1758835920970850. [PMID: 33224279 PMCID: PMC7659026 DOI: 10.1177/1758835920970850] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 10/08/2020] [Indexed: 12/14/2022] Open
Abstract
The high mobility group box (HMGB) protein family consists of four members: HMGB1, 2, 3, and 4. They share similar amino acid sequences and identical functional regions, especially HMGB1, 2, and 3. The homology in structure may lead to similarity in function. In fact, though their targets may be different, they all possess the fundamental function of binding and distorting target DNAs. However, further research confirmed they are distributed differently in tissues and involved in various distinct physiological and pathological cellular processes, including cell proliferation, division, migration, and differentiation. Recently, the roles of HMGB family members in carcinogenesis has been widely investigated; however, systematic discussion on their functions and clinical values in malignant tumors is limited. In this review, we mainly review and summarize recent advances in knowledge of HMGB family members in terms of structure, distribution, biochemical cascades, and specific mechanisms regarding tumor progression. Importantly, the diagnostic, prognostic, and therapeutic value of these proteins in cancers is discussed. Finally, we envisage the orientation and challenges of this field in further studies.
Collapse
Affiliation(s)
- Liaoran Niu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Wanli Yang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Lili Duan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Xiaoqian Wang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yiding Li
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Chengchao Xu
- 94719 Military Hospital, Ji'an, Jiangxi Province, China
| | - Chao Liu
- School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yujie Zhang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Wei Zhou
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Jinqiang Liu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Qingchuan Zhao
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yu Han
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, 710032, China
| | - Liu Hong
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Shaanxi Province, 710032, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
94
|
Arnold K, Liao YE, Liu J. Potential Use of Anti-Inflammatory Synthetic Heparan Sulfate to Attenuate Liver Damage. Biomedicines 2020; 8:E503. [PMID: 33207634 PMCID: PMC7697061 DOI: 10.3390/biomedicines8110503] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/12/2020] [Accepted: 11/14/2020] [Indexed: 12/28/2022] Open
Abstract
Heparan sulfate is a highly sulfated polysaccharide abundant on the surface of hepatocytes and surrounding extracellular matrix. Emerging evidence demonstrates that heparan sulfate plays an important role in neutralizing the activities of proinflammatory damage associate molecular patterns (DAMPs) that are released from hepatocytes under pathological conditions. Unlike proteins and nucleic acids, isolation of homogenous heparan sulfate polysaccharides from biological sources is not possible, adding difficulty to study the functional role of heparan sulfate. Recent advancement in the development of a chemoenzymatic approach allows production of a large number of structurally defined oligosaccharides. These oligosaccharides are used to probe the physiological functions of heparan sulfate in liver damage under different pathological conditions. The findings provide a potential new therapeutic agent to treat liver diseases that are associated with excessive inflammation.
Collapse
Affiliation(s)
| | | | - Jian Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA; (K.A.); (Y.-E.L.)
| |
Collapse
|
95
|
The role of type 1 interferons in coagulation induced by gram-negative bacteria. Blood 2020; 135:1087-1100. [PMID: 32016282 DOI: 10.1182/blood.2019002282] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 01/08/2020] [Indexed: 12/13/2022] Open
Abstract
Bacterial infection not only stimulates innate immune responses but also activates coagulation cascades. Overactivation of the coagulation system in bacterial sepsis leads to disseminated intravascular coagulation (DIC), a life-threatening condition. However, the mechanisms by which bacterial infection activates the coagulation cascade are not fully understood. Here we show that type 1 interferons (IFNs), a widely expressed family of cytokines that orchestrate innate antiviral and antibacterial immunity, mediate bacterial infection-induced DIC by amplifying the release of high-mobility group box 1 (HMGB1) into the bloodstream. Inhibition of the expression of type 1 IFNs and disruption of their receptor IFN-α/βR or downstream effector (eg, HMGB1) uniformly decreased gram-negative bacteria-induced DIC. Mechanistically, extracellular HMGB1 markedly increased the procoagulant activity of tissue factor by promoting the externalization of phosphatidylserine to the outer cell surface, where phosphatidylserine assembles a complex of cofactor-proteases of the coagulation cascades. These findings not only provide novel insights into the link between innate immune responses and coagulation, but they also open a new avenue for developing novel therapeutic strategies to prevent DIC in sepsis.
Collapse
|
96
|
Fagenson AM, Xu K, Saaoud F, Nanayakkara G, Jhala NC, Liu L, Drummer C, Sun Y, Lau KN, Di Carlo A, Jiang X, Wang H, Karhadkar SS, Yang X. Liver Ischemia Reperfusion Injury, Enhanced by Trained Immunity, Is Attenuated in Caspase 1/Caspase 11 Double Gene Knockout Mice. Pathogens 2020; 9:pathogens9110879. [PMID: 33114395 PMCID: PMC7692674 DOI: 10.3390/pathogens9110879] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/17/2020] [Accepted: 10/20/2020] [Indexed: 12/14/2022] Open
Abstract
Ischemia reperfusion injury (IRI) during liver transplantation increases morbidity and contributes to allograft dysfunction. There are no therapeutic strategies to mitigate IRI. We examined a novel hypothesis: caspase 1 and caspase 11 serve as danger-associated molecular pattern (DAMPs) sensors in IRI. By performing microarray analysis and using caspase 1/caspase 11 double-knockout (Casp DKO) mice, we show that the canonical and non-canonical inflammasome regulators are upregulated in mouse liver IRI. Ischemic pre (IPC)- and post-conditioning (IPO) induce upregulation of the canonical and non-canonical inflammasome regulators. Trained immunity (TI) regulators are upregulated in IPC and IPO. Furthermore, caspase 1 is activated during liver IRI, and Casp DKO attenuates liver IRI. Casp DKO maintained normal liver histology via decreased DNA damage. Finally, the decreased TUNEL assay-detected DNA damage is the underlying histopathological and molecular mechanisms of attenuated liver pyroptosis and IRI. In summary, liver IRI induces the upregulation of canonical and non-canonical inflammasomes and TI enzyme pathways. Casp DKO attenuate liver IRI. Development of novel therapeutics targeting caspase 1/caspase 11 and TI may help mitigate injury secondary to IRI. Our findings have provided novel insights on the roles of caspase 1, caspase 11, and inflammasome in sensing IRI derived DAMPs and TI-promoted IRI-induced liver injury.
Collapse
Affiliation(s)
- Alexander M. Fagenson
- Department of Surgery, Division of Abdominal Organ Transplant, Lewis Katz School of Medicine, Temple University, 3401 N. Broad Street, Philadelphia, PA 19140, USA; (K.N.L.); (A.D.C.); (S.S.K.)
- Centers for Cardiovascular Research, Inflammation, Translational and Clinical Lung Research, Metabolic Disease Research, Thrombosis Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (K.X.); (F.S.); (G.N.); (C.D.); (Y.S.); (X.J.)
- Correspondence: (A.M.F.); (X.Y.)
| | - Keman Xu
- Centers for Cardiovascular Research, Inflammation, Translational and Clinical Lung Research, Metabolic Disease Research, Thrombosis Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (K.X.); (F.S.); (G.N.); (C.D.); (Y.S.); (X.J.)
- Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (L.L.); (H.W.)
| | - Fatma Saaoud
- Centers for Cardiovascular Research, Inflammation, Translational and Clinical Lung Research, Metabolic Disease Research, Thrombosis Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (K.X.); (F.S.); (G.N.); (C.D.); (Y.S.); (X.J.)
| | - Gayani Nanayakkara
- Centers for Cardiovascular Research, Inflammation, Translational and Clinical Lung Research, Metabolic Disease Research, Thrombosis Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (K.X.); (F.S.); (G.N.); (C.D.); (Y.S.); (X.J.)
- Eccles Institute of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Nirag C. Jhala
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| | - Lu Liu
- Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (L.L.); (H.W.)
| | - Charles Drummer
- Centers for Cardiovascular Research, Inflammation, Translational and Clinical Lung Research, Metabolic Disease Research, Thrombosis Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (K.X.); (F.S.); (G.N.); (C.D.); (Y.S.); (X.J.)
| | - Yu Sun
- Centers for Cardiovascular Research, Inflammation, Translational and Clinical Lung Research, Metabolic Disease Research, Thrombosis Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (K.X.); (F.S.); (G.N.); (C.D.); (Y.S.); (X.J.)
| | - Kwan N. Lau
- Department of Surgery, Division of Abdominal Organ Transplant, Lewis Katz School of Medicine, Temple University, 3401 N. Broad Street, Philadelphia, PA 19140, USA; (K.N.L.); (A.D.C.); (S.S.K.)
| | - Antonio Di Carlo
- Department of Surgery, Division of Abdominal Organ Transplant, Lewis Katz School of Medicine, Temple University, 3401 N. Broad Street, Philadelphia, PA 19140, USA; (K.N.L.); (A.D.C.); (S.S.K.)
| | - Xiaohua Jiang
- Centers for Cardiovascular Research, Inflammation, Translational and Clinical Lung Research, Metabolic Disease Research, Thrombosis Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (K.X.); (F.S.); (G.N.); (C.D.); (Y.S.); (X.J.)
- Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (L.L.); (H.W.)
| | - Hong Wang
- Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (L.L.); (H.W.)
| | - Sunil S. Karhadkar
- Department of Surgery, Division of Abdominal Organ Transplant, Lewis Katz School of Medicine, Temple University, 3401 N. Broad Street, Philadelphia, PA 19140, USA; (K.N.L.); (A.D.C.); (S.S.K.)
| | - Xiaofeng Yang
- Centers for Cardiovascular Research, Inflammation, Translational and Clinical Lung Research, Metabolic Disease Research, Thrombosis Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (K.X.); (F.S.); (G.N.); (C.D.); (Y.S.); (X.J.)
- Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (L.L.); (H.W.)
- Correspondence: (A.M.F.); (X.Y.)
| |
Collapse
|
97
|
Ghosh SS, Ghosh S. HMGB1 (High-Mobility Group Box-1): A Common Link Determining the Consequences of Tissue Injury, Sterile/Microbial and Low-Grade Chronic Inflammation. Arterioscler Thromb Vasc Biol 2020; 40:2561-2563. [PMID: 33085519 DOI: 10.1161/atvbaha.120.315189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Siddhartha S Ghosh
- Department of Internal Medicine, VCU Medical Center, Richmond, VA (S.S.G., S.G.)
| | - Shobha Ghosh
- Hunter Homes McGuire VA Medical Center, Richmond, VA (S.G.)
| |
Collapse
|
98
|
Arnold K, Xu Y, Liao YE, Cooley BC, Pawlinski R, Liu J. Synthetic anticoagulant heparan sulfate attenuates liver ischemia reperfusion injury. Sci Rep 2020; 10:17187. [PMID: 33057098 PMCID: PMC7566620 DOI: 10.1038/s41598-020-74275-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 09/29/2020] [Indexed: 12/18/2022] Open
Abstract
Heparan sulfate (HS) is a sulfated glycosaminoglycan abundant on the cell surface and in the extracellular matrix and has several biological activities including anticoagulation and anti-inflammation. Liver ischemia reperfusion injury is associated with coagulation and inflammatory responses. Here, we synthesized HS oligosaccharides with defined sulfation patterns and show that synthetic anticoagulant HS oligosaccharides limit liver ischemia reperfusion injury in a mouse model. Using a small targeted HS library, we demonstrate that an oligosaccharide that possesses both anticoagulant activity and binding affinity to HMGB1, the inflammatory target, decreases injury greater than oligosaccharides that only bind to HMGB1 or only have anticoagulant activity. HS oligosaccharides may represent a potential new therapeutic option for decreasing liver damage resulting from ischemia reperfusion injury.
Collapse
Affiliation(s)
- Katelyn Arnold
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Yongmei Xu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Yi-En Liao
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Brian C Cooley
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Rafal Pawlinski
- Division of Hematology/Oncology, Department of Medicine, UNC Blood Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - Jian Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
99
|
Zhang S, Feng Z, Gao W, Duan Y, Fan G, Geng X, Wu B, Li K, Liu K, Peng C. Aucubin Attenuates Liver Ischemia-Reperfusion Injury by Inhibiting the HMGB1/TLR-4/NF-κB Signaling Pathway, Oxidative Stress, and Apoptosis. Front Pharmacol 2020; 11:544124. [PMID: 33013386 PMCID: PMC7506056 DOI: 10.3389/fphar.2020.544124] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/24/2020] [Indexed: 12/16/2022] Open
Abstract
Liver ischemia-reperfusion injury (IRI) is a common clinical event with high morbidity in patients undergoing complex liver surgery or having abdominal trauma. Inflammatory and oxidative stress responses are the main contributing factors in liver IRI. The iridoid glucoside aucubin (AU) has good anti-inflammatory and antioxidative effects; however, there are no relevant reports on the protective effect of glucosides on hepatic IRI. The purpose of this study was to determine whether AU pretreatment could prevent liver IRI and to explore the mechanism. Sprague–Dawley rats were randomly divided into five groups. The sham operation and IRI control groups were given intraperitoneal injections of normal saline, while the AU low-dose (AU-L) group, AU medium-dose (AU-M) group, and AU high-dose (AU-H) group were given intraperitoneal injections of AU at doses of 1, 5, and 10 mg/kg/day, respectively. After 10 d, liver IRI (70% liver ischemia for 1 h, reperfusion for 6 h) was surgically established in all groups except the sham group. Our results confirmed that liver injury was significantly aggravated after hepatic ischemia-reperfusion. AU alleviated the increase of transaminase and pathological changes induced by ischemia-reperfusion and improved liver damage. AU could also ameliorate the inflammatory and oxidative stress responses induced by ischemia-reperfusion and reduced expression of high mobility group protein (HMG)B1, receptor for advanced glycation end-products (RAGE), tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and reactive oxygen species (ROS). Moreover, AU reduced ischemia-reperfusion-induced mitochondrial dysfunction and cells apoptosis, increased peroxisome proliferator-activated receptor γ coactivator (PGC)-1α and uncoupling (UCP)2 protein expression, and reduced caspase-3, cleaved caspase-3, and Cytochrome P450 proteins (CYP) expression. To determine expression levels of the Toll-like receptor (TLR)-4/nuclear factor-κB (NF-κB) pathway-related proteins in vitro and in vivo, we also measured TLR-4, myeloid differentiation factor88 (MyD88), NF-κB P65, p-P65, I-kappa-B-alpha (IκB-α), and p-IκB-α levels. The results showed that AU effectively inhibited activation of the TLR-4/NF-κB signaling pathway. In conclusion, we showed for the first time a hepatoprotective effect for AU in liver IRI, which acted by inhibiting the HMGB1/TLR-4/NF-κB signaling pathway, oxidative stress, and apoptosis. Pretreatment with AU may be a promising strategy for preventing liver IRI.
Collapse
Affiliation(s)
- Shilong Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zanjie Feng
- Department of Biochemistry and Molecular Biology, Zunyi Medical University, Zunyi, China
| | - Weidong Gao
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yuling Duan
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Guoxin Fan
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xin Geng
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Bo Wu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Kai Li
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Kangwei Liu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Cijun Peng
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
100
|
Hirao H, Dery KJ, Kageyama S, Nakamura K, Kupiec-Weglinski JW. Heme Oxygenase-1 in liver transplant ischemia-reperfusion injury: From bench-to-bedside. Free Radic Biol Med 2020; 157:75-82. [PMID: 32084514 PMCID: PMC7434658 DOI: 10.1016/j.freeradbiomed.2020.02.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/02/2020] [Accepted: 02/17/2020] [Indexed: 12/16/2022]
Abstract
Hepatic ischemia-reperfusion injury (IRI), a major risk factor for early allograft dysfunction (EAD) and acute or chronic graft rejection, contributes to donor organ shortage for life-saving orthotopic liver transplantation (OLT). The graft injury caused by local ischemia (warm and/or cold) leads to parenchymal cell death and release of danger-associated molecular patterns (DAMPs), followed by reperfusion-triggered production of reactive oxygen species (ROS), activation of inflammatory cells, hepatocellular damage and ultimate organ failure. Heme oxygenase 1 (HO-1), a heat shock protein-32 induced under IR-stress, is an essential component of the cytoprotective mechanism in stressed livers. HO-1 regulates anti-inflammatory responses and may be crucial in the pathogenesis of chronic diseases, such as arteriosclerosis, hypertension, diabetes and steatosis. An emerging area of study is macrophage-derived HO-1 and its pivotal intrahepatic homeostatic function played in IRI-OLT. Indeed, ectopic hepatic HO-1 overexpression activates intracellular SIRT1/autophagy axis to serve as a key cellular self-defense mechanism in both mouse and human OLT recipients. Recent translational studies in rodents and human liver transplant patients provide novel insights into HO-1 mediated cytoprotection against sterile hepatic inflammation. In this review, we summarize the current bench-to-bedside knowledge on HO-1 molecular signaling and discuss their future therapeutic potential to mitigate IRI in OLT.
Collapse
Affiliation(s)
- Hirofumi Hirao
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Kenneth J Dery
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Shoichi Kageyama
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Kojiro Nakamura
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA; Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Surgery, Nishi-Kobe Medical Center, 5-7-1 Koji-dai, Nishi-ku, Kobe, Hyogo, 651-2273, Japan
| | - Jerzy W Kupiec-Weglinski
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|