1
|
Hu C, Li M, Chen Y, Cheng W, Wang H, Zhou Y, Teng F, Ling T, Pan J, Xu H, Zheng Y, Ji G, Zhao T, You Q. AIM2 regulates autophagy to mitigate oxidative stress in aged mice with acute liver injury. Cell Death Discov 2024; 10:107. [PMID: 38429284 PMCID: PMC10907373 DOI: 10.1038/s41420-024-01870-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/06/2024] [Accepted: 02/14/2024] [Indexed: 03/03/2024] Open
Abstract
The cytoplasmic pattern recognition receptor, absent in melanoma 2 (AIM2), detects cytosolic DNA, activating the inflammasome and resulting in pro-inflammatory cytokine production and pyroptotic cell death. Recent research has illuminated AIM2's contributions to PANoptosis and host defense. However, the role of AIM2 in acetaminophen (APAP)-induced hepatoxicity remains enigmatic. In this study, we unveil AIM2's novel function as a negative regulator in the pathogenesis of APAP-induced liver damage in aged mice, independently of inflammasome activation. AIM2-deficient aged mice exhibited heightened lipid accumulation and hepatic triglycerides in comparison to their wild-type counterparts. Strikingly, AIM2 knockout mice subjected to APAP overdose demonstrated intensified liver injury, compromised mitochondrial stability, exacerbated glutathione depletion, diminished autophagy, and elevated levels of phosphorylated c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK). Furthermore, our investigation revealed AIM2's mitochondrial localization; its overexpression in mouse hepatocytes amplified autophagy while dampening JNK phosphorylation. Notably, induction of autophagy through rapamycin administration mitigated serum alanine aminotransferase levels and reduced the necrotic liver area in AIM2-deficient aged mice following APAP overdose. Mechanistically, AIM2 deficiency exacerbated APAP-induced acute liver damage and inflammation in aged mice by intensifying oxidative stress and augmenting the phosphorylation of JNK and ERK. Given its regulatory role in autophagy and lipid peroxidation, AIM2 emerges as a promising therapeutic target for age-related acute liver damage treatment.
Collapse
Affiliation(s)
- Chao Hu
- Department of Geriatrics, Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Mengjing Li
- Department of Geriatrics, Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Yongzhen Chen
- Department of general practice, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Wei Cheng
- Department of Geriatrics, Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Haining Wang
- Department of Geriatrics, Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Yiming Zhou
- Department of Geriatrics, Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Fengmeng Teng
- Affilated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Tao Ling
- Department of Geriatrics, Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Jinshun Pan
- Department of Geriatrics, Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Haozhe Xu
- Department of Geriatrics, Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Yanan Zheng
- Department of Geriatrics, Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Guozhong Ji
- Department of general practice, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China.
| | - Ting Zhao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Qiang You
- Department of Geriatrics, Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China.
| |
Collapse
|
2
|
Napodano C, Carnazzo V, Basile V, Pocino K, Stefanile A, Gallucci S, Natali P, Basile U, Marino M. NLRP3 Inflammasome Involvement in Heart, Liver, and Lung Diseases-A Lesson from Cytokine Storm Syndrome. Int J Mol Sci 2023; 24:16556. [PMID: 38068879 PMCID: PMC10706560 DOI: 10.3390/ijms242316556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Inflammation and inflammasomes have been proposed as important regulators of the host-microorganism interaction, playing a key role in morbidity and mortality due to the coronavirus disease 2019 (COVID-19) in subjects with chronic conditions and compromised immune system. The inflammasome consists of a multiprotein complex that finely regulates the activation of caspase-1 and the production and secretion of potent pro-inflammatory cytokines such as IL-1β and IL-18. The pyrin containing NOD (nucleotide-binding oligomerization domain) like receptor (NLRP) is a family of intracellular receptors, sensing patterns associated to pathogens or danger signals and NLRP3 inflammasome is the most deeply analyzed for its involvement in the innate and adaptive immune system as well as its contribution to several autoinflammatory and autoimmune diseases. It is highly expressed in leukocytes and up-regulated in sentinel cells upon inflammatory stimuli. NLRP3 expression has also been reported in B and T lymphocytes, in epithelial cells of oral and genital mucosa, in specific parenchymal cells as cardiomyocytes, and keratinocytes, and chondrocytes. It is well known that a dysregulated activation of the inflammasome is involved in the pathogenesis of different disorders that share the common red line of inflammation in their pathogenetic fingerprint. Here, we review the potential roles of the NLRP3 inflammasome in cardiovascular events, liver damage, pulmonary diseases, and in that wide range of systemic inflammatory syndromes named as a cytokine storm.
Collapse
Affiliation(s)
- Cecilia Napodano
- Department of Laboratory of Medicine and Pathology, S. Agostino Estense Hospital, 41126 Modena, Italy;
| | - Valeria Carnazzo
- Department of Clinical Pathology, Santa Maria Goretti Hospital, AUSL Latina, 04100 Latina, Italy; (V.C.); (U.B.)
| | - Valerio Basile
- Clinical Pathology Unit and Cancer Biobank, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Krizia Pocino
- Unità Operativa Complessa di Patologia Clinica, Ospedale Generale di Zona San Pietro Fatebenefratelli, 00189 Rome, Italy; (K.P.); (A.S.)
| | - Annunziata Stefanile
- Unità Operativa Complessa di Patologia Clinica, Ospedale Generale di Zona San Pietro Fatebenefratelli, 00189 Rome, Italy; (K.P.); (A.S.)
| | - Stefania Gallucci
- Laboratory of Dendritic Cell Biology, Division of Innate Immunity, Department of Medicine, UMass Chan Medical School, Worcester, MA 01655, USA;
| | - Patrizia Natali
- Diagnostic Hematology and Clinical Genomics, Department of Laboratory Medicine and Pathology, AUSL/AOU Modena, 41124 Modena, Italy;
| | - Umberto Basile
- Department of Clinical Pathology, Santa Maria Goretti Hospital, AUSL Latina, 04100 Latina, Italy; (V.C.); (U.B.)
| | - Mariapaola Marino
- Dipartimento di Medicina e Chirurgia Traslazionale, Sezione di Patologia Generale, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
3
|
Tran L, Xie B, Assaf E, Ferrari R, Pipinos II, Casale GP, Alvidrez RIM, Watkins S, Sachdev U. Transcriptomic Profiling Identifies Ferroptosis-Related Gene Signatures in Ischemic Muscle Satellite Cells Affected by Peripheral Artery Disease-Brief Report. Arterioscler Thromb Vasc Biol 2023; 43:2023-2029. [PMID: 37675635 PMCID: PMC10549760 DOI: 10.1161/atvbaha.123.319518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/01/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND We hypothesized that transcriptomic profiling of muscle satellite cells in peripheral artery disease (PAD) would identify damage-related pathways contributing to skeletal muscle myopathy. We identified a potential role for ferroptosis-a form of programmed lytic cell death by iron-mediated lipid peroxidation-as one such pathway. Ferroptosis promotes myopathy in ischemic cardiac muscle but has an unknown role in PAD. METHODS Muscle satellite cells from donors with PAD were obtained during surgery. cDNA libraries were processed for single-cell RNA sequencing using the 10X Genomics platform. Protein expression was confirmed based on pathways inferred by transcriptomic analysis. RESULTS Unsupervised cluster analysis of over 25 000 cells aggregated from 8 donor samples yielded distinct cell populations grouped by a shared unique transcriptional fingerprint. Quiescent cells were diminished in ischemic muscle while myofibroblasts and apoptotic cells were prominent. Differential gene expression demonstrated a surprising increase in genes associated with iron transport and oxidative stress and a decrease in GPX4 (glutathione peroxidase 4) in ischemic PAD-derived cells. Release of the danger signal HMGB1 (high mobility group box-1) correlated with ferroptotic markers including surface transferrin receptor and were higher in ischemia. Furthermore, lipid peroxidation in muscle satellite cells was modulated by ferrostatin, a ferroptosis inhibitor. Histology confirmed iron deposition and lipofuscin, an inducer of ferroptosis in PAD-affected muscle. CONCLUSIONS This report presents a novel finding that genes known to be involved in ferroptosis are differentially expressed in human skeletal muscle affected by PAD. Targeting ferroptosis may be a novel therapeutic strategy to reduce PAD myopathy.
Collapse
Affiliation(s)
- Lillian Tran
- University of Pittsburgh Medical Center Department of Surgery
| | - Bowen Xie
- University of Pittsburgh Medical Center Department of Surgery
| | - Edwyn Assaf
- University of Pittsburgh Medical Center Department of Surgery
| | - Ricardo Ferrari
- University of Pittsburgh Medical Center Department of Surgery
| | - Iraklis I. Pipinos
- University of Nebraska Medical Center Department of Surgery and the VAResearch Service, VA Nebraska-Western Iowa Health Care System
| | - George P. Casale
- University of Nebraska Medical Center Department of Surgery and the VAResearch Service, VA Nebraska-Western Iowa Health Care System
| | | | - Simon Watkins
- University of Pittsburgh Center for Biologic Imaging
| | - Ulka Sachdev
- University of Pittsburgh Medical Center Department of Surgery
| |
Collapse
|
4
|
Tseng YH, Chen IC, Li WC, Hsu JH. Regulatory Cues in Pulmonary Fibrosis-With Emphasis on the AIM2 Inflammasome. Int J Mol Sci 2023; 24:10876. [PMID: 37446052 DOI: 10.3390/ijms241310876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Pulmonary fibrosis (PF) is a chronic lung disorder characterized by the presence of scarred and thickened lung tissues. Although the Food and Drug Administration approved two antifibrotic drugs, pirfenidone, and nintedanib, that are currently utilized for treating idiopathic PF (IPF), the clinical therapeutic efficacy remains unsatisfactory. It is crucial to develop new drugs or treatment schemes that combine pirfenidone or nintedanib to achieve more effective outcomes for PF patients. Understanding the complex mechanisms underlying PF could potentially facilitate drug discovery. Previous studies have found that the activation of inflammasomes, including nucleotide-binding and oligomerization domain (NOD)-like receptor protein (NLRP)1, NLRP3, NOD-like receptor C4, and absent in melanoma (AIM)2, contributes to lung inflammation and fibrosis. This article aims to summarize the cellular and molecular regulatory cues that contribute to PF with a particular emphasis on the role of AIM2 inflammasome in mediating pathophysiologic events during PF development. The insights gained from this research may pave the way for the development of more effective strategies for the prevention and treatment of PF.
Collapse
Affiliation(s)
- Yu-Hsin Tseng
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - I-Chen Chen
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
- Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Wan-Chun Li
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Jong-Hau Hsu
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
- Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
5
|
Wang J, Sun Z, Xie J, Ji W, Cui Y, Ai Z, Liang G. Inflammasome and pyroptosis in autoimmune liver diseases. Front Immunol 2023; 14:1150879. [PMID: 36969233 PMCID: PMC10030845 DOI: 10.3389/fimmu.2023.1150879] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 02/23/2023] [Indexed: 03/11/2023] Open
Abstract
Autoimmune hepatitis (AIH), primary biliary cholangitis (PBC), primary sclerosing cholangitis (PSC), and IgG4-related sclerosing cholangitis (IgG4-SC) are the four main forms of autoimmune liver diseases (AILDs), which are all defined by an aberrant immune system attack on the liver. Most previous studies have shown that apoptosis and necrosis are the two major modes of hepatocyte death in AILDs. Recent studies have reported that inflammasome-mediated pyroptosis is critical for the inflammatory response and severity of liver injury in AILDs. This review summarizes our present understanding of inflammasome activation and function, as well as the connections among inflammasomes, pyroptosis, and AILDs, thus highlighting the shared features across the four disease models and gaps in our knowledge. In addition, we summarize the correlation among NLRP3 inflammasome activation in the liver-gut axis, liver injury, and intestinal barrier disruption in PBC and PSC. We summarize the differences in microbial and metabolic characteristics between PSC and IgG4-SC, and highlight the uniqueness of IgG4-SC. We explore the different roles of NLRP3 in acute and chronic cholestatic liver injury, as well as the complex and controversial crosstalk between various types of cell death in AILDs. We also discuss the most up-to-date developments in inflammasome- and pyroptosis-targeted medicines for autoimmune liver disorders.
Collapse
Affiliation(s)
- Jixuan Wang
- School of First Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhiwen Sun
- Department of Liver, Spleen and Stomach Diseases, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jingri Xie
- Department of Liver, Spleen and Stomach Diseases, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wanli Ji
- School of First Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yang Cui
- School of First Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zongxiong Ai
- School of First Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
- *Correspondence: Guoying Liang, ; Zongxiong Ai,
| | - Guoying Liang
- Department of Liver, Spleen and Stomach Diseases, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
- *Correspondence: Guoying Liang, ; Zongxiong Ai,
| |
Collapse
|
6
|
Kim JS, Chapman WC, Lin Y. Mitochondrial Autophagy in Ischemic Aged Livers. Cells 2022; 11:cells11244083. [PMID: 36552847 PMCID: PMC9816943 DOI: 10.3390/cells11244083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Mitochondrial autophagy (mitophagy) is a central catabolic event for mitochondrial quality control. Defective or insufficient mitophagy, thus, can result in mitochondrial dysfunction, and ultimately cell death. There is a strong causal relationship between ischemia/reperfusion (I/R) injury and mitochondrial dysfunction following liver resection and transplantation. Compared to young patients, elderly patients poorly tolerate I/R injury. Accumulation of abnormal mitochondria after I/R is more prominent in aged livers than in young counterparts. This review highlights how altered autophagy is mechanistically involved in age-dependent hypersensitivity to reperfusion injury.
Collapse
Affiliation(s)
- Jae-Sung Kim
- Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA; (W.C.C.); (Y.L.)
- Department of Cell Biology & Physiology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Correspondence:
| | - William C. Chapman
- Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA; (W.C.C.); (Y.L.)
| | - Yiing Lin
- Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA; (W.C.C.); (Y.L.)
| |
Collapse
|
7
|
Pirnie R, P Gillespie K, Mesaros C, Blair IA. Reappraisal of oxidized HMGB1 as a mediator and biomarker. Future Sci OA 2022; 8:FSO828. [PMID: 36874369 PMCID: PMC9979160 DOI: 10.2144/fsoa-2022-0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 01/12/2023] [Indexed: 02/12/2023] Open
Abstract
HMGB1 is a dual-function protein that acts as a chromatin-binding protein and as a danger-associated molecular pattern (DAMP) when released from activated immune cells or injured tissue. In much of the HMGB1 literature, immunomodulatory effects of extracellular HMGB1 are proposed to depend on its oxidation state. However, many of the foundational studies for this model have been retracted or flagged with expressions of concern. The literature on HMGB1 oxidation reveals a diversity of redox proteoforms of HMGB1 that are inconsistent with current models of redox modulation regulating HMGB1 secretion. A recent study of acetaminophen toxicity has identified previously unrecognized HMGB1 oxidized proteoforms. HMGB1 undergoes oxidative modifications that could serve as pathology-specific biomarkers and drug targets.
Collapse
Affiliation(s)
- Ross Pirnie
- Center of Excellence in Environmental Toxicology & Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kevin P Gillespie
- Center of Excellence in Environmental Toxicology & Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Clementina Mesaros
- Center of Excellence in Environmental Toxicology & Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ian A Blair
- Center of Excellence in Environmental Toxicology & Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
8
|
Pirnie R, Gillespie KP, Weng L, Mesaros C, Blair IA. Characterization and Quantification of Oxidized High Mobility Group Box 1 Proteoforms Secreted from Hepatocytes by Toxic Levels of Acetaminophen. Chem Res Toxicol 2022; 35:1893-1902. [PMID: 35922039 PMCID: PMC9580022 DOI: 10.1021/acs.chemrestox.2c00161] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The high mobility group box 1 (HMGB1), which is released during acute acetaminophen (APAP) overdose, is thought to mediate a subsequent immune response, particularly hepatic infiltration of macrophages. The redox behavior of HMGB1 and the proteoforms of HMGB1 present in oxidative environments has been the subject of a number of confusing and contradictory studies. Therefore, a stable isotope dilution two-dimensional nanoultrahigh-performance liquid chromatography parallel reaction monitoring/high-resolution mass spectrometry method was developed in order to characterize and quantify oxidative modifications to the cysteine (Cys) residues (Cys-23, Cys-45, and Cys-106) that are present in HMGB1. Disulfide linkages were determined using carbamidoethyl derivatization before and after reduction as well as by direct analysis of disulfide cross-linked peptides. A stable isotope labeled form of HMGB1 was used as an internal standard to correct for sample to sample differences in immunoaffinity precipitation, derivatization, and electrospray ionization. Four discrete HMGB1 proteoforms were found to be released from a hepatocarcinoma cell model of APAP overdose after 24 h. Fully reduced HMGB1 with all three Cys-residues in their free thiol state accounted for 18% of the secreted HMGB1. The proteoform with disulfide between Cys-23 and Cys-45 accounted for 24% of the HMGB1. No evidence was obtained for a disulfide cross-link between Cys-106 and the other two Cys-residues. However, 45% of the HMGB1 formed a cross-link with unidentified intracellular proteins via an intermolecular disulfide bond, and 12% was present as the terminally oxidized cysteic acid. Surprisingly, there was no evidence for the formation of HMGB1 disulfides with GSH or other low molecular weight thiols. Secreted plasma HMGB1 Cys-23/Cys45 disulfide proteoform together with the Cys-106/protein disulfide proteoforms could potentially serve as early biomarkers of hepatoxicity after APAP overdose as well as biomarkers of drug-induced liver injury.
Collapse
|
9
|
Dutta A, Das M, Ghosh A, Rana S. Molecular and cellular pathophysiology of circulating cardiomyocyte-specific cell free DNA (cfDNA): Biomarkers of heart failure and potential therapeutic targets. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
|
10
|
Gan C, Cai Q, Tang C, Gao J. Inflammasomes and Pyroptosis of Liver Cells in Liver Fibrosis. Front Immunol 2022; 13:896473. [PMID: 35707547 PMCID: PMC9189314 DOI: 10.3389/fimmu.2022.896473] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/04/2022] [Indexed: 01/18/2023] Open
Abstract
Inflammasomes are multiprotein complexes that can sense danger signals and activate caspase-1 to mediate pro-inflammatory cytokines release and pyroptotic cell death. There are two main canonical and non-canonical signaling pathways that trigger inflammasome activation. Inflammasomes are expressed and assembled in parenchymal and nonparenchymal cells in response to liver injury in the liver. Additionally, the hepatocytes, biliary epithelial cells (cholangiocytes), hepatic stellate cells (HSCs), hepatic macrophages, and liver sinusoidal endothelial cells (LSECs) contribute to liver fibrosis via different mechanisms. However, the underlying mechanism of the inflammasome and pyroptosis in these liver cells in liver fibrosis remains elusive. This review summarizes the activation and function of inflammasome complexes and then discusses the association between inflammasomes, pyroptosis, and liver fibrosis. Unlike other similar reviewers, we will focus on the effect of inflammasome activation and pyroptosis in the various liver cells during the development of liver fibrosis. We will also highlight the latest progress of pharmacological intervention in inflammasome-mediated liver fibrosis.
Collapse
Affiliation(s)
- Can Gan
- Lab of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Qiuyu Cai
- Lab of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Chengwei Tang
- Lab of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Jinhang Gao, ; ; Chengwei Tang,
| | - Jinhang Gao
- Lab of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Jinhang Gao, ; ; Chengwei Tang,
| |
Collapse
|
11
|
Ferrari R, Xie B, Assaf E, Morder K, Scott M, Liao H, Calderon MJ, Ross M, Loughran P, Watkins SC, Pipinos I, Casale G, Tzeng E, McEnaney R, Sachdev U. Inflammatory Caspase Activity Mediates HMGB1 Release and Differentiation in Myoblasts Affected by Peripheral Arterial Disease. Cells 2022; 11:1163. [PMID: 35406727 PMCID: PMC8997414 DOI: 10.3390/cells11071163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 12/10/2022] Open
Abstract
Introduction: We previously showed that caspase-1 and -11, which are activated by inflammasomes, mediate recovery from muscle ischemia in mice. We hypothesized that similar to murine models, inflammatory caspases modulate myogenicity and inflammation in ischemic muscle disease. Methods: Caspase activity was measured in ischemic and perfused human myoblasts in response to the NLRP3 and AIM2 inflammasome agonists (nigericin and poly(dA:dT), respectively) with and without specific caspase-1 or pan-caspase inhibition. mRNA levels of myogenic markers and caspase-1 were assessed, and protein levels of caspases-1, -4, -5, and -3 were measured by Western blot. Results: When compared to perfused cells, ischemic myoblasts demonstrated attenuated MyoD and myogenin and elevated caspase-1 mRNA. Ischemic myoblasts also had significantly higher enzymatic caspase activity with poly(dA:dT) (p < 0.001), but not nigericin stimulation. Inhibition of caspase activity including caspase-4/-5, but not caspase-1, blocked activation effects of poly(dA:dT). Ischemic myoblasts had elevated cleaved caspase-5. Inhibition of caspase activity deterred differentiation in ischemic but not perfused myoblasts and reduced the release of HMGB1 from both groups. Conclusion: Inflammatory caspases can be activated in ischemic myoblasts by AIM2 and influence ischemic myoblast differentiation and release of pro-angiogenic HMGB1. AIM2 inflammasome involvement suggests a role as a DNA damage sensor, and our data suggest that caspase-5 rather than caspase-1 may mediate the downstream mediator of this pathway.
Collapse
Affiliation(s)
- Ricardo Ferrari
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Bowen Xie
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Edwyn Assaf
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Kristin Morder
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Melanie Scott
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Hong Liao
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Michael J. Calderon
- University of Pittsburgh Center for Biologic Imaging, Pittsburgh, PA 15213, USA; (M.J.C.); (M.R.); (P.L.); (S.C.W.)
| | - Mark Ross
- University of Pittsburgh Center for Biologic Imaging, Pittsburgh, PA 15213, USA; (M.J.C.); (M.R.); (P.L.); (S.C.W.)
| | - Patricia Loughran
- University of Pittsburgh Center for Biologic Imaging, Pittsburgh, PA 15213, USA; (M.J.C.); (M.R.); (P.L.); (S.C.W.)
| | - Simon C. Watkins
- University of Pittsburgh Center for Biologic Imaging, Pittsburgh, PA 15213, USA; (M.J.C.); (M.R.); (P.L.); (S.C.W.)
| | - Iraklis Pipinos
- Department of Surgery, University of Nebraska, Omaha, NE 68198, USA; (I.P.); (G.C.)
- Department of Surgery, Veterans Affairs Hospital, Pittsburgh, PA 15240, USA
| | - George Casale
- Department of Surgery, University of Nebraska, Omaha, NE 68198, USA; (I.P.); (G.C.)
| | - Edith Tzeng
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
- Department of Surgery, Veterans Affairs Hospital, Pittsburgh, PA 15240, USA
| | - Ryan McEnaney
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
- Department of Surgery, Veterans Affairs Hospital, Pittsburgh, PA 15240, USA
| | - Ulka Sachdev
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| |
Collapse
|
12
|
Ismail N, Sharma A, Soong L, Walker DH. Review: Protective Immunity and Immunopathology of Ehrlichiosis. ZOONOSES (BURLINGTON, MASS.) 2022; 2:10.15212/zoonoses-2022-0009. [PMID: 35876763 PMCID: PMC9300479 DOI: 10.15212/zoonoses-2022-0009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Human monocytic ehrlichiosis, a tick transmitted infection, ranges in severity from apparently subclinical to a fatal toxic shock-like fatal disease. Models in immunocompetent mice range from an abortive infection to uniformly lethal depending on the infecting Ehrlichia species, dose of inoculum, and route of inoculation. Effective immunity is mediated by CD4+ T lymphocytes and gamma interferon. Lethal infection occurs with early overproduction of proinflammatory cytokines and overproduction of TNF alpha and IL-10 by CD8+ T lymphocytes. Furthermore, fatal ehrlichiosis is associated with signaling via TLR 9/MyD88 with upregulation of several inflammasome complexes and secretion of IL-1 beta, IL-1 alpha, and IL-18 by hepatic mononuclear cells, suggesting activation of canonical and noncanonical inflammasome pathways, a deleterious role for IL-18, and the protective role for caspase 1. Autophagy promotes ehrlichial infection, and MyD88 signaling hinders ehrlichial infection by inhibiting autophagy induction and flux. Activation of caspase 11 during infection of hepatocytes by the lethal ehrlichial species after interferon alpha receptor signaling results in the production of inflammasome-dependent IL-1 beta, extracellular secretion of HMGB1, and pyroptosis. The high level of HMGB1 in lethal ehrlichiosis suggests a role in toxic shock. Studies of primary bone marrow-derived macrophages infected by highly avirulent or mildly avirulent ehrlichiae reveal divergent M1 and M2 macrophage polarization that links with generation of pathogenic CD8 T cells, neutrophils, and excessive inflammation or with strong expansion of protective Th1 and NKT cells, resolution of inflammation and clearance of infection, respectively.
Collapse
Affiliation(s)
- Nahed Ismail
- Clinical Microbiology, Laboratory Medicine, University of Illinois at Chicago-College of Medicine, University of Illinois Hospitals & Health Science System, Chicago, IL
| | - Aditya Sharma
- Clinical Microbiology, Laboratory Medicine, University of Illinois at Chicago-College of Medicine, University of Illinois Hospitals & Health Science System, Chicago, IL
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
- Department of Pathology, Center for Biodefense & Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX
| | - David H. Walker
- Department of Pathology, Center for Biodefense & Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX
| |
Collapse
|
13
|
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: 158] [Impact Index Per Article: 39.5] [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
|
14
|
Shao R, Wang X, Xu T, Xia Y, Cui D. The balance between AIM2-associated inflammation and autophagy: the role of CHMP2A in brain injury after cardiac arrest. J Neuroinflammation 2021; 18:257. [PMID: 34740380 PMCID: PMC8571899 DOI: 10.1186/s12974-021-02307-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/27/2021] [Indexed: 12/30/2022] Open
Abstract
Background Activation of the absent in melanoma 2 (AIM2) inflammasome and impaired autophagosome clearance in neurons contribute significantly to cardiac arrest and return of spontaneous circulation (CA-ROSC) injury, while the mechanism by which the AIM2 inflammasome is regulated and relationship between the processes remain poorly understood. Recently, charged multivesicular body protein 2A (CHMP2A), a subunit of endosomal sorting complex required for transport (ESCRT), was shown to regulate phagophore closure, and its depletion led to the accumulation of autophagosomes and induced cell death. Here, we investigated whether CHMP2A-mediated autophagy was an underlying mechanism of AIM2-associated inflammation after CA-ROSC and explored the potential link between the AIM2 inflammasome and autophagy under ischemic conditions. Methods AIM2 inflammasome activation and autophagic flux in the cortex were assessed in the CA-ROSC rat model. We injected LV-Vector or LV-CHMP2A virus into the motor cortex with stereotaxic coordinates and divided the rats into four groups: Sham, CA, CA+LV-Vector, and CA+LV-CHMP2A. Neurologic deficit scores (NDSs), balance beam tests, histopathological injury of the brain, and expression of the AIM2 inflammasome and proinflammatory cytokines were analyzed. Results AIM2 inflammasome activation and increased interleukin 1 beta (IL-1β) and IL-18 release were concurrent with reduced levels of CHMP2A-induced autophagy in CA-ROSC rat neurons. In addition, silencing CHMP2A resulted in autophagosome accumulation and decreased autophagic degradation of the AIM2 inflammasome. In parallel, a reduction in AIM2 contributed to autophagy activation and mitigated oxygen–glucose deprivation and reperfusion (OGD-Rep)-induced inflammation. Notably, CHMP2A overexpression in the cortex hindered neuroinflammation, protected against ischemic brain damage, and improved neurologic outcomes after CA. Conclusions Our results support a potential link between autophagy and AIM2 signaling, and targeting CHMP2A may provide new insights into neuroinflammation in the early phase during CA-ROSC. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02307-8.
Collapse
Affiliation(s)
- Rongjiao Shao
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, Xuhui District, Shanghai, 200233, China
| | - Xintao Wang
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, Xuhui District, Shanghai, 200233, China
| | - Tianhua Xu
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, Xuhui District, Shanghai, 200233, China
| | - Yiyang Xia
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, Xuhui District, Shanghai, 200233, China
| | - Derong Cui
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, Xuhui District, Shanghai, 200233, China.
| |
Collapse
|
15
|
Li S, Zhu J, Pan L, Wan P, Qin Q, Luo D, Pan W, Wei Y, Xu Y, Shang L, Ye X. Potential protective effect of hesperidin on hypoxia/reoxygenation-induced hepatocyte injury. Exp Ther Med 2021; 22:764. [PMID: 34035861 PMCID: PMC8135133 DOI: 10.3892/etm.2021.10196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
Hesperidin (HDN) has been reported to have hydrogen radical- and hydrogen peroxide-removal activities and to serve an antioxidant role in biological systems. However, whether HDN protects hepatocytes (HCs) against hypoxia/reoxygenation (H/R)-induced injury remains unknown. The present study aimed to explore the role of HDN in H/R-induced injury. HCs were isolated and cultured under H/R conditions with or without HDN treatment. HC damage was markedly induced under H/R, as indicated by cell viability, supernatant lactate dehydrogenase levels and alanine aminotransferase levels; however, HDN treatment significantly reversed HC injury. Oxidative stress markers (malondialdehyde, superoxide dismutase, glutathioneand reactive oxygen species) were increased markedly during H/R in HCs; however, this effect was significantly attenuated after exposure to HDN. Compared with those of the control group, the mRNA expression levels of IL-6 and TNF-α in HCs and the concentrations of IL-6 and TNF-α in the supernatants increased significantly following H/R, and HDN significantly ameliorated these effects. Western blotting demonstrated that microtubule-associated protein 1 light chain 3α (MAP1LC3A, also known as LC3) and Beclin-1 protein expression levels increased, while sequestosome 1 levels decreased during H/R following exposure to HDN. The number of GFP-LC3 puncta in HCs following exposure to HDN was increased compared with that observed in HCs without HDN exposure under the H/R conditions after bafilomycin A1 treatment. In summary, the present study demonstrated that HDN attenuated HC oxidative stress and inflammatory responses while enhancing autophagy during H/R. HDN may have a potential protective effect on HCs during H/R-induced injury.
Collapse
Affiliation(s)
- Shilai Li
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jijin Zhu
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Ling Pan
- Department of Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Peiqi Wan
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Quanlin Qin
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Daqing Luo
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Wenhui Pan
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yuqing Wei
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yansong Xu
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Liming Shang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xinping Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
16
|
Zhang WJ, Chen SJ, Zhou SC, Wu SZ, Wang H. Inflammasomes and Fibrosis. Front Immunol 2021; 12:643149. [PMID: 34177893 PMCID: PMC8226128 DOI: 10.3389/fimmu.2021.643149] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
Fibrosis is the final common pathway of inflammatory diseases in various organs. The inflammasomes play an important role in the progression of fibrosis as innate immune receptors. There are four main members of the inflammasomes, such as NOD-like receptor protein 1 (NLRP1), NOD-like receptor protein 3 (NLRP3), NOD-like receptor C4 (NLRC4), and absent in melanoma 2 (AIM2), among which NLRP3 inflammasome is the most studied. NLRP3 inflammasome is typically composed of NLRP3, ASC and pro-caspase-1. The activation of inflammasome involves both "classical" and "non-classical" pathways and the former pathway is better understood. The "classical" activation pathway of inflammasome is that the backbone protein is activated by endogenous/exogenous stimulation, leading to inflammasome assembly. After the formation of "classic" inflammasome, pro-caspase-1 could self-activate. Caspase-1 cleaves cytokine precursors into mature cytokines, which are secreted extracellularly. At present, the "non-classical" activation pathway of inflammasome has not formed a unified model for activation process. This article reviews the role of NLRP1, NLRP3, NLRC4, AIM2 inflammasome, Caspase-1, IL-1β, IL-18 and IL-33 in the fibrogenesis.
Collapse
Affiliation(s)
- Wen-Juan Zhang
- Department of Immunology, School of Basic Medicine, Gannan Medical University, Ganzhou, China.,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Shu-Juan Chen
- Department of Immunology, School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Shun-Chang Zhou
- Department of Experimental Animals, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Su-Zhen Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Hui Wang
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
17
|
Li S, Qin Q, Luo D, Pan W, Wei Y, Xu Y, Zhu J, Shang L. Hesperidin ameliorates liver ischemia/reperfusion injury via activation of the Akt pathway. Mol Med Rep 2020; 22:4519-4530. [PMID: 33174025 PMCID: PMC7646746 DOI: 10.3892/mmr.2020.11561] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 07/03/2020] [Indexed: 01/07/2023] Open
Abstract
Hesperidin (HDN) is a bioflavonoid that serves a role as an antioxidant in biological systems. However, although HDN has hydrogen radical- and hydrogen peroxide-removal activities, the role of HDN in liver ischemia/reperfusion (I/R) injury remains unknown. This study aimed to determine the role of HDN in liver I/R injury. Male C57BL/6J wild-type (WT) mice were subjected to warm partial liver I/R injury. Liver damage was evaluated by measuring serum alanine aminotransferase (ALT) levels, cytokine production, oxidative stress indicators, tissue hematoxylin-eosin staining and cell death. The Akt signaling pathway was examined to elucidate the underlying mechanisms. HDN had no effect on ALT levels and tissue damage in WT mice without liver I/R injury. However, HDN significantly ameliorated liver I/R injury as measured by serum ALT levels and necrotic tissue areas. HDN decreased malondialdehyde content, but increased the levels of superoxide dismutase, catalase, glutathione peroxidase and glutathione. In addition, HDN significantly attenuated the mRNA expression levels of TNF-α, IL-6 and IL-1β after liver I/R injury. Furthermore, HDN protected the liver against apoptosis in liver I/R injury by increasing the levels of Bcl-2 and decreasing the levels of cleaved-caspase 3. Mechanistically, the levels of phosphorylated Akt were elevated by HDN during liver I/R injury. In addition, HDN could induce Akt activation in hepatocytes in vitro. Most importantly, treatment with the Akt inhibitor LY294002 in WT mice blocked the hepatoprotective effects of HDN in liver I/R injury. In summary, the results of the present study suggested that HDN may protect against liver I/R injury through activating the Akt pathway by ameliorating liver oxidative stress, suppressing inflammation and preventing hepatocyte apoptosis. HDN may be a useful factor for liver injury protection and a potential therapeutic treatment for liver I/R injury in the future.
Collapse
Affiliation(s)
- Shilai Li
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R China
| | - Quanlin Qin
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R China
| | - Daqing Luo
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R China
| | - Wenhui Pan
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R China
| | - Yuqing Wei
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R China
| | - Yansong Xu
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R China
| | - Jijin Zhu
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R China
| | - Liming Shang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R China
| |
Collapse
|
18
|
Sachdev U, Ferrari R, Cui X, Pius A, Sahu A, Reynolds M, Liao H, Sun P, Shinde S, Ambrosio F, Shiva S, Loughran P, Scott M. Caspase1/11 signaling affects muscle regeneration and recovery following ischemia, and can be modulated by chloroquine. Mol Med 2020; 26:69. [PMID: 32641037 PMCID: PMC7341481 DOI: 10.1186/s10020-020-00190-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/19/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND We previously showed that the autophagy inhibitor chloroquine (CQ) increases inflammatory cleaved caspase-1 activity in myocytes, and that caspase-1/11 is protective in sterile liver injury. However, the role of caspase-1/11 in the recovery of muscle from ischemia caused by peripheral arterial disease is unknown. We hypothesized that caspase-1/11 mediates recovery in muscle via effects on autophagy and this is modulated by CQ. METHODS C57Bl/6 J (WT) and caspase-1/11 double-knockout (KO) mice underwent femoral artery ligation (a model of hind-limb ischemia) with or without CQ (50 mg/kg IP every 2nd day). CQ effects on autophagosome formation, microtubule associated protein 1A/1B-light chain 3 (LC3), and caspase-1 expression was measured using electron microscopy and immunofluorescence. Laser Doppler perfusion imaging documented perfusion every 7 days. After 21 days, in situ physiologic testing in tibialis anterior muscle assessed peak force contraction, and myocyte size and fibrosis was also measured. Muscle satellite cell (MuSC) oxygen consumption rate (OCR) and extracellular acidification rate was measured. Caspase-1 and glycolytic enzyme expression was detected by Western blot. RESULTS CQ increased autophagosomes, LC3 consolidation, total caspase-1 expression and cleaved caspase-1 in muscle. Perfusion, fibrosis, myofiber regeneration, muscle contraction, MuSC fusion, OCR, ECAR and glycolytic enzyme expression was variably affected by CQ depending on presence of caspase-1/11. CQ decreased perfusion recovery, fibrosis and myofiber size in WT but not caspase-1/11KO mice. CQ diminished peak force in whole muscle, and myocyte fusion in MuSC and these effects were exacerbated in caspase-1/11KO mice. CQ reductions in maximal respiration and ATP production were reduced in caspase-1/11KO mice. Caspase-1/11KO MuSC had significant increases in protein kinase isoforms and aldolase with decreased ECAR. CONCLUSION Caspase-1/11 signaling affects the response to ischemia in muscle and effects are variably modulated by CQ. This may be critically important for disease treated with CQ and its derivatives, including novel viral diseases (e.g. COVID-19) that are expected to affect patients with comorbidities like cardiovascular disease.
Collapse
Affiliation(s)
- Ulka Sachdev
- Division of Vascular Surgery; Department of Surgery, University of Pittsburgh Medical Center, Magee Women's Hospital, 200 Lothrop Street, Pittsburgh, PA, 15213, USA.
| | - Ricardo Ferrari
- Division of Vascular Surgery; Department of Surgery, University of Pittsburgh Medical Center, Magee Women's Hospital, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
| | - Xiangdong Cui
- Division of Vascular Surgery; Department of Surgery, University of Pittsburgh Medical Center, Magee Women's Hospital, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
| | - Abish Pius
- McGowan Institute for Regenerative Medicine, University of Pittsburgh Medical Center, Bridgeside Point, Pittsburgh, PA, 15213, USA
| | - Amrita Sahu
- McGowan Institute for Regenerative Medicine, University of Pittsburgh Medical Center, Bridgeside Point, Pittsburgh, PA, 15213, USA
| | - Michael Reynolds
- Department of Pharmacology and Chemical Biology, University of Pittsburgh Medical Center, Biomedical Sciences Towe, Pittsburgh, PA, 15213, USA
| | - Hong Liao
- Division of Vascular Surgery; Department of Surgery, University of Pittsburgh Medical Center, Magee Women's Hospital, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
| | - Ping Sun
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Department of Surgery 11/20/2018-11/19/202, Visiting scholar, University of Pittsburgh, Pittsburgh, USA
| | - Sunita Shinde
- McGowan Institute for Regenerative Medicine, University of Pittsburgh Medical Center, Bridgeside Point, Pittsburgh, PA, 15213, USA
| | - Fabrisia Ambrosio
- McGowan Institute for Regenerative Medicine, University of Pittsburgh Medical Center, Bridgeside Point, Pittsburgh, PA, 15213, USA
| | - Sruti Shiva
- Department of Pharmacology and Chemical Biology, University of Pittsburgh Medical Center, Biomedical Sciences Towe, Pittsburgh, PA, 15213, USA
| | - Patricia Loughran
- Center for Biologic Imaging (CBI), University of Pittsburgh Medical Center, Biomedical Sciences Tower, Pittsburgh, PA, 15213, USA
| | - Melanie Scott
- Division of Vascular Surgery; Department of Surgery, University of Pittsburgh Medical Center, Magee Women's Hospital, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
| |
Collapse
|
19
|
Yang H, Wang H, Andersson U. Targeting Inflammation Driven by HMGB1. Front Immunol 2020; 11:484. [PMID: 32265930 PMCID: PMC7099994 DOI: 10.3389/fimmu.2020.00484] [Citation(s) in RCA: 405] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/02/2020] [Indexed: 12/22/2022] Open
Abstract
High mobility group box 1 (HMGB1) is a highly conserved, nuclear protein present in all cell types. It is a multi-facet protein exerting functions both inside and outside of cells. Extracellular HMGB1 has been extensively studied for its prototypical alarmin functions activating innate immunity, after being actively released from cells or passively released upon cell death. TLR4 and RAGE operate as the main HMGB1 receptors. Disulfide HMGB1 activates the TLR4 complex by binding to MD-2. The binding site is separate from that of LPS and it is now feasible to specifically interrupt HMGB1/TLR4 activation without compromising protective LPS/TLR4-dependent functions. Another important therapeutic strategy is established on the administration of HMGB1 antagonists precluding RAGE-mediated endocytosis of HMGB1 and HMGB1-bound molecules capable of activating intracellular cognate receptors. Here we summarize the role of HMGB1 in inflammation, with a focus on recent findings on its mission as a damage-associated molecular pattern molecule and as a therapeutic target in inflammatory diseases. Recently generated HMGB1-specific inhibitors for treatment of inflammatory conditions are discussed.
Collapse
Affiliation(s)
- Huan Yang
- Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Haichao Wang
- Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Ulf Andersson
- Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
20
|
Li S, Yi Z, Deng M, Scott MJ, Yang C, Li W, Lei Z, Santerre NM, Loughran P, Billiar TR. TSLP protects against liver I/R injury via activation of the PI3K/Akt pathway. JCI Insight 2019; 4:129013. [PMID: 31723054 DOI: 10.1172/jci.insight.129013] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 10/10/2019] [Indexed: 01/09/2023] Open
Abstract
Thymic stromal lymphopoietin (TSLP) is a cytokine mainly released by epithelial cells that plays important roles in inflammation, autoimmune disease, and cancer. While TSLP is expressed in the liver at high levels, the role of TSLP in liver ischemia/reperfusion (I/R) injury remains unknown. Experiments were carried out to determine the role of TSLP in liver I/R injury. Wild-type (WT) and TSLP receptor-knockout (TSLPR-/-) mice were subjected to liver partial warm I/R injury. Liver injury was assessed by measuring serum alanine aminotransferase (ALT) level, necrotic areas by liver histology, hepatocyte death, and local hepatic inflammatory responses. Signal pathways were explored in vivo and in vitro to identify possible mechanisms for TSLP in I/R injury. TSLP and TSLPR protein expression increased during liver I/R in vivo and following hepatocyte hypoxia/reoxygenation in vitro. Deletion of TSLPR or neutralization of TSLP with anti-TSLP antibody exacerbated liver injury in terms of serum ALT levels as well as necrotic areas in liver histology. Administration of exogenous recombinant mouse TSLP to WT mice significantly reduced liver damage compared with controls, but failed to prevent I/R injury in TSLPR-/- mice. TSLP induced autophagy in hepatocytes during liver I/R injury. Mechanistically, Akt was activated in WT mice during liver I/R injury. The opposite results were observed in TSLPR-/- mice. In addition, TSLP could directly induce Akt activation in hepatocytes independent of nonparenchymal cells in vitro. Furthermore, the Akt agonist, insulin-like growth factor-1 (IGF-1), prevented I/R injury in TSLPR-/- mice and an Akt inhibitor, LY294002, blocked the protective effects of TSLP in WT mice subjected to I/R. Our data indicate that TSLP protects against liver I/R injury via activation of the PI3K/Akt pathway. Through this pathway, TSLP induces autophagy in hepatocytes. Thus, TSLP is a potent inhibitor of stress-induced hepatocyte necrosis.
Collapse
Affiliation(s)
- Shilai Li
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhongjie Yi
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Hepatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Meihong Deng
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Melanie J Scott
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Chenxuan Yang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,School of Medicine, Tsinghua University, Beijing, China
| | - Wenbo Li
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Plastic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhao Lei
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Hepatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Nicole M Santerre
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Patricia Loughran
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Center for Biologic Imaging, University of Pittsburgh, Pennsylvania, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
21
|
Wang J, Li R, Peng Z, Hu B, Rao X, Li J. HMGB1 participates in LPS‑induced acute lung injury by activating the AIM2 inflammasome in macrophages and inducing polarization of M1 macrophages via TLR2, TLR4, and RAGE/NF‑κB signaling pathways. Int J Mol Med 2019; 45:61-80. [PMID: 31746367 PMCID: PMC6889921 DOI: 10.3892/ijmm.2019.4402] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 09/30/2019] [Indexed: 12/22/2022] Open
Abstract
High mobility group box 1 (HMGB1), a crucial proinflammatory cytokine, was reported to activate the absent in melanoma 2 (AIM2) inflammasome, which are both essential in acute lung injury (ALI). However, their interaction mechanism has remained elusive. Macrophages are known to express the AIM2 inflammasome and the main receptors [receptor for advanced glycation end products (RAGE), Toll‑like receptor 2/4 (TLR‑2/TLR‑4)] of HMGB1 to transmit intracellular signals. The present study aimed to indicate whether HMGB1 participates in the process of lipopolysaccharides (LPS)‑induced ALI through activating the AIM2 inflammasome in macrophages, as well as inducing polarization of M1 macrophages via TLR2, TLR4 and RAGE/ nuclear factor‑κB (NF‑κB) signaling pathways. In an in vivo mouse model of LPS‑induced ALI, anti‑HMGB1, recombinant (r)HMGB1, LPS from Rhodobacter sphaeroides (LPS‑RS, TLR2/4 antagonist) or FPS‑ZM1 (RAGE antagonist) were administrated. In in vitro studies, bone marrow‑derived macrophages from mice primed with LPS were stimulated with or without anti‑HMGB1, rHMGB1, LPS‑RS, or FPS‑ZM1. The findings revealed that anti‑HMGB1, LPS‑RS and FPS‑ZM1 significantly decreased infiltration of inflammatory cells, wet‑to‑dry ratio, myeloperoxidase activity in the lung, the levels of cytokines, as well as macrophages and neutrophil infiltration in the bronchoalveolar lavage fluid. However, rHMGB1 aggravated the inflammatory response in ALI. Mechanistically, anti‑HMGB1, LPS‑RS and FPS‑ZM1 attenuated activation of TLR2, TLR4, and RAGE/NF‑κB signaling pathways and expression of the AIM2 inflammasome in macrophages. However, rHMGB1 enhanced their expression levels and induced polarization of M1 macrophages. These results indicated that HMGB1 could participate in the pathogenesis of ALI by activating the AIM2 inflammasome in macrophages, as well as inducing polarization of M1 macrophages through TLR2, TLR4 and RAGE/NF‑κB signaling pathways.
Collapse
Affiliation(s)
- Jing Wang
- Department of Intensive Care Unit, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Ruiting Li
- Department of Intensive Care Unit, Wuhan Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Zhiyong Peng
- Department of Intensive Care Unit, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Bo Hu
- Department of Intensive Care Unit, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Xin Rao
- Department of Intensive Care Unit, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Jianguo Li
- Department of Intensive Care Unit, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
22
|
Zhang X, Dong P, Xu L, Tian Y, Sun H, Shi H, Duan Z, Chen L, Ren F. The different expression of caspase-1 in HBV-related liver disease and acts as a biomarker for acute-on-chronic liver failure. BMC Gastroenterol 2019; 19:148. [PMID: 31429707 PMCID: PMC6700995 DOI: 10.1186/s12876-019-1064-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
Background Caspase-1 is an evolutionarily conserved enzyme that proteolytically cleaves the precursors of the inflammatory cytokines interleukin 1β and interleukin 18. However, the role of caspase-1 in determining the severity of acute-on-chronic liver failure (ACLF) has yet to be elucidated. We evaluated the expression levels of caspase-1 in HBV-related liver disease and assessed its utility as a biomarker predicting the severity of ACLF. Methods The gene, protein and activity levels of caspase-1 were measured in the liver and/or serum of subjects with HBV-related disease. We also analysed the correlation between the expression levels of caspase-1 and liver injury of ACLF. Results Compared with the values observed in normal subjects, the relative caspase-1 mRNA and protein levels in livers were decreased in patients with CHB, LC, and HCC but increased in those with ACLF; moreover, ACLF patients had the lowest serum level and hepatic activity of caspase-1 among the five groups. The serum caspase-1 levels in ACLF patients showed a negative correlation with total serum bilirubin and a positive correlation with serum total protein and albumin. Importantly, the serum caspase-1 levels in the surviving group with ACLF were higher than those in the non-surviving group and showed different dynamic trends. Analyses of the area under the receiver operating characteristic curve indicated that caspase-1 (AUC = 0.84, AUC of MELD score = 0.72) may be a useful marker for independently predicting ACLF. Conclusion Caspase-1 is a potential non-invasive biomarker of disease progression and prognosis in ACLF.
Collapse
Affiliation(s)
- Xiangying Zhang
- Beijing YouAn Hospital, Capital Medical University, Beijing, 100069, China
| | - Peiling Dong
- Beijing YouAn Hospital, Capital Medical University, Beijing, 100069, China
| | - Lin Xu
- Beijing YouAn Hospital, Capital Medical University, Beijing, 100069, China
| | - Yuan Tian
- Beijing YouAn Hospital, Capital Medical University, Beijing, 100069, China
| | - Huayin Sun
- Beijing YouAn Hospital, Capital Medical University, Beijing, 100069, China
| | - Hongbo Shi
- Beijing YouAn Hospital, Capital Medical University, Beijing, 100069, China
| | - Zhongping Duan
- Beijing YouAn Hospital, Capital Medical University, Beijing, 100069, China
| | - Liyan Chen
- The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| | - Feng Ren
- Beijing YouAn Hospital, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
23
|
Gasdermin D protects against noninfectious liver injury by regulating apoptosis and necroptosis. Cell Death Dis 2019; 10:481. [PMID: 31209224 PMCID: PMC6579760 DOI: 10.1038/s41419-019-1719-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 05/15/2019] [Accepted: 06/04/2019] [Indexed: 12/12/2022]
Abstract
Gasdermin D (GsdmD) was recently identified as the executioner of pyroptotic inflammatory cell death, and is a substrate for caspases-1 and 11. GsdmD is detrimental in lethal endotoxemia but protective in bacterial sepsis. However, little is known about its role during noninfectious/sterile injuries. In this study, we examined the contribution of GsdmD using WT and GsdmD−/− mice in two models of noninfectious liver injury: hemorrhagic shock with resuscitation (HS/R) and acetaminophen (APAP) overdose. GsdmD−/− mice had significantly increased liver damage at 6 h after HS/R or APAP vs WT, shown by significantly elevated ALT level and extended areas of cell death in liver. Caspase-8, a mediator of multiple cell death pathways, was highly elevated in GsdmD−/− mice after injury. Significantly increased cleavage of caspase-8 and subsequent high levels of apoptosis were found in livers of GsdmD−/− mice after HS/R, a relatively mild ROS-induced liver injury. However, during more severe APAP-mediated ROS-induced liver injury, caspase-8 cleavage in GsdmD−/− liver was inhibited compared with WT, resulting in accumulation of pro-caspase-8 and increased levels of necroptosis. Our findings indicate a novel hepatoprotective role for GsdmD in noninfectious inflammation models via regulation of caspase-8 expression and downstream cell death pathways. The effects of GsdmD protection are likely injury specific and may also depend on injury severity and levels of ROS produced. These data suggest modulation of GsdmD/caspase-8 may be a novel therapeutic option in ROS-mediated liver injury.
Collapse
|
24
|
Li XQ, Yu Q, Fang B, Zhang ZL, Ma H. Knockdown of the AIM2 molecule attenuates ischemia-reperfusion-induced spinal neuronal pyroptosis by inhibiting AIM2 inflammasome activation and subsequent release of cleaved caspase-1 and IL-1β. Neuropharmacology 2019; 160:107661. [PMID: 31181224 DOI: 10.1016/j.neuropharm.2019.05.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 05/23/2019] [Accepted: 05/31/2019] [Indexed: 01/05/2023]
Abstract
Ischemia-reperfusion (IR) injury induces activation of several inflammasomes that widely affect neuroinflammation and, subsequently, neuronal viability. The absent in melanoma 2 (AIM2) inflammasome is highly expressed in neurons after traumatic injury. This study was performed to investigate whether the AIM2 molecule acts as an initiator to trigger AIM2 inflammasome activation and regulate neuronal pyroptosis in a mouse IR model. The early motor dysfunction that occurred within the first 8 h post-IR injury was closely associated with a massive increase in dsDNA in serum and cerebrospinal fluid (CSF) at the same observed timepoints. However, the subsequent persistent dysfunction was consistent with the continuously increasing protein levels of apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC), cleaved caspase-1 and IL-1β with time. Upregulated AIM2 immunoreactivity was primarily visualized in neurons. The si-AIM2 treatment in vivo preserved part of motor function, accompanied by decreased protein levels of AIM2, ASC, cleaved caspase-1 and IL-1β. In vitro, the direct interactions between the AIM2 molecule and caspase-1 were demonstrated by immunofluorescence staining and coimmunoprecipitation. In this context, both si-AIM2 and Ac-YVAD-CMK treatments effectively maintained neuronal viability, as demonstrated by the decreased percentage of cells with pyroptosis and release of lactate dehydrogenase (LDH), accompanied by weak immunoreactivity and a decreased number of AIM2-caspase-1 positive neurons. By contrast, poly(dA-dT) treatment exacerbated neuronal pyroptosis by reversing the above-mentioned effects. However, no significant differences were observed after si-Con treatment. These results suggest AIM2 molecule played an important role in initiating AIM2 inflammasome activation through IR-induced release of ectopic dsDNA.
Collapse
Affiliation(s)
- Xiao-Qian Li
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, Shenyang, 110001, Liaoning, China.
| | - Qian Yu
- Department of Thoracic Surgery, Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, Liaoning, China.
| | - Bo Fang
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, Shenyang, 110001, Liaoning, China.
| | - Zai-Li Zhang
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, Shenyang, 110001, Liaoning, China.
| | - Hong Ma
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
25
|
Deng M, Scott MJ, Fan J, Billiar TR. Location is the key to function: HMGB1 in sepsis and trauma-induced inflammation. J Leukoc Biol 2019; 106:161-169. [PMID: 30946496 DOI: 10.1002/jlb.3mir1218-497r] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/18/2019] [Accepted: 03/04/2019] [Indexed: 12/16/2022] Open
Abstract
High mobility group box 1 (HMGB1) is a multifunctional nuclear protein, probably known best as a prototypical alarmin or damage-associated molecular pattern (DAMP) molecule when released from cells. However, HMGB1 has multiple functions that depend on its location in the nucleus, in the cytosol, or extracellularly after either active release from cells, or passive release upon lytic cell death. Movement of HMGB1 between cellular compartments is a dynamic process induced by a variety of cell stresses and disease processes, including sepsis, trauma, and hemorrhagic shock. Location of HMGB1 is intricately linked with its function and is regulated by a series of posttranslational modifications. HMGB1 function is also regulated by the redox status of critical cysteine residues within the protein, and is cell-type dependent. This review highlights some of the mechanisms that contribute to location and functions of HMGB1, and focuses on some recent insights on important intracellular effects of HMGB1 during sepsis and trauma.
Collapse
Affiliation(s)
- Meihong Deng
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Pittsburgh Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Melanie J Scott
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Pittsburgh Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jie Fan
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Pittsburgh Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Pittsburgh Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
26
|
Tang H, Qu EZ, Li K, Ye LS, Zhang Q, Yang Y, Chen GH, Zhang YC. Effect of Hepatic Artery Spasm on a Rat Model of Hepatic Ischemia-Reperfusion Injury. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2019; 38:597-604. [PMID: 30378156 DOI: 10.1002/jum.14726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/07/2018] [Accepted: 05/26/2018] [Indexed: 06/08/2023]
Abstract
OBJECTIVES To investigate hemodynamic changes in the hepatic artery after hepatic ischemia-reperfusion injury (IRI) in rats via ultrasound (US) imaging and to discuss the protective effect of phentolamine (PHT) pretreatment on hepatic IRI. METHODS Fifty rats were randomly divided into 3 groups: a sham operation group (n = 10), a control ischemia-reperfusion group (n = 20), and a PHT pretreatment group (n = 20). Color Doppler flow imaging and contrast-enhanced US examinations were performed in each group at 30 minutes (n = 10) and 90 minutes (n = 10) after reperfusion. Blood samples were obtained to analyze serum alanine aminotransferase and aspartate aminotransferase levels, and liver tissue specimens were collected for pathologic analysis. RESULTS Using US, we found that hepatic artery resistance at 30 minutes after reperfusion in the control group was higher than that in the sham group (mean resistive index [RI] ± SD, 0.65 ± 0.09 versus 0.50 ± 0.09; P < .01), which was higher at 30 than 90 minutes (RI, 0.65 ± 0.09 versus 0.50 ± 0.08; P < .01) after reperfusion in the control group. However, the hepatic artery resistance and liver microcirculation in the PHT group were better than those in the control group at 30 minutes after reperfusion (RI, 0.54 ± 0.09 versus 0.65 ± 0.09; P < .05; time to peak, 31.94 ± 2.02 versus 48.34 ± 4.74 seconds; P < .01). Compared to the control group, the aspartate aminotransferase and alanine aminotransferase levels were significantly lower at 30 minutes after reperfusion in the PHT group (P < .05). A pathologic examination revealed a smaller hepatic artery diameter and a depressed vessel wall in the control group. CONCLUSIONS The hepatic artery can undergo a transient spasm during the hepatic IRI process, which can exacerbate liver damage. Phentolamine treatment can alleviate hepatic artery spasms, improve liver perfusion, and reduce liver injury by ameliorating the hepatic microcirculation.
Collapse
Affiliation(s)
- Hui Tang
- Liver Transplantation Center and Department of Hepatic Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - En-Ze Qu
- Department of Ultrasonography, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kun Li
- Liver Transplantation Center and Department of Hepatic Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lin-Sen Ye
- Liver Transplantation Center and Department of Hepatic Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qi Zhang
- Liver Transplantation Center and Department of Hepatic Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Liver Diseases of Guangdong Province, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yang Yang
- Liver Transplantation Center and Department of Hepatic Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Gui-Hua Chen
- Liver Transplantation Center and Department of Hepatic Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ying-Cai Zhang
- Liver Transplantation Center and Department of Hepatic Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
27
|
van Golen RF, Reiniers MJ, Marsman G, Alles LK, van Rooyen DM, Petri B, Van der Mark VA, van Beek AA, Meijer B, Maas MA, Zeerleder S, Verheij J, Farrell GC, Luken BM, Teoh NC, van Gulik TM, Murphy MP, Heger M. The damage-associated molecular pattern HMGB1 is released early after clinical hepatic ischemia/reperfusion. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1192-1200. [PMID: 30658161 DOI: 10.1016/j.bbadis.2019.01.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 12/21/2018] [Accepted: 01/11/2019] [Indexed: 12/31/2022]
Abstract
OBJECTIVE AND BACKGROUND Activation of sterile inflammation after hepatic ischemia/reperfusion (I/R) culminates in liver injury. The route to liver damage starts with mitochondrial oxidative stress and cell death during early reperfusion. The link between mitochondrial oxidative stress, damage-associate molecular pattern (DAMP) release, and sterile immune signaling is incompletely understood and lacks clinical validation. The aim of the study was to validate this relation in a clinical liver I/R cohort and to limit DAMP release using a mitochondria-targeted antioxidant in I/R-subjected mice. METHODS Plasma levels of the DAMPs high-mobility group box 1 (HMGB1), mitochondrial DNA, and nucleosomes were measured in 39 patients enrolled in an observational study who underwent a major liver resection with (N = 29) or without (N = 13) intraoperative liver ischemia. Circulating cytokine and neutrophil activation markers were also determined. In mice, the mitochondria-targeted antioxidant MitoQ was intravenously infused in an attempt to limit DAMP release, reduce sterile inflammation, and suppress I/R injury. RESULTS In patients, HMGB1 was elevated following liver resection with I/R compared to liver resection without I/R. HMGB1 levels correlated positively with ischemia duration and peak post-operative transaminase (ALT) levels. There were no differences in mitochondrial DNA, nucleosome, or cytokine levels between the two groups. In mice, MitoQ neutralized hepatic oxidative stress and decreased HMGB1 release by ±50%. MitoQ suppressed transaminase release, hepatocellular necrosis, and cytokine production. Reconstituting disulfide HMGB1 during reperfusion reversed these protective effects. CONCLUSION HMGB1 seems the most pertinent DAMP in clinical hepatic I/R injury. Neutralizing mitochondrial oxidative stress may limit DAMP release after hepatic I/R and reduce liver damage.
Collapse
Affiliation(s)
- Rowan F van Golen
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Megan J Reiniers
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Gerben Marsman
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Lindy K Alles
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Derrick M van Rooyen
- Liver Research Group, Australian National University at The Canberra Hospital, Canberra, Australia
| | - Björn Petri
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary AB T2N 1N4, Alberta, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary AB T2N 1N4, Alberta, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada
| | - Vincent A Van der Mark
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Tytgat Institute for Gastrointestinal and Liver Research, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Adriaan A van Beek
- Department of Cell Biology and Immunology, Wageningen University, Wageningen, the Netherlands
| | - Ben Meijer
- Department of Cell Biology and Immunology, Wageningen University, Wageningen, the Netherlands
| | - Martinus A Maas
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Sacha Zeerleder
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Switzerland; Department for BioMedical Research, University of Bern, Switzerland
| | - Joanne Verheij
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Geoffrey C Farrell
- Liver Research Group, Australian National University at The Canberra Hospital, Canberra, Australia
| | - Brenda M Luken
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Narci C Teoh
- Liver Research Group, Australian National University at The Canberra Hospital, Canberra, Australia
| | - Thomas M van Gulik
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Michael P Murphy
- Medical Research Council Mitochondrial Biology Unit, Cambridge, United Kingdom; Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Michal Heger
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, PR China.
| |
Collapse
|
28
|
Luan J, Ju D. Inflammasome: A Double-Edged Sword in Liver Diseases. Front Immunol 2018; 9:2201. [PMID: 30319645 PMCID: PMC6167446 DOI: 10.3389/fimmu.2018.02201] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 09/05/2018] [Indexed: 12/20/2022] Open
Abstract
Inflammasomes have emerged as critical innate sensors of host immune that defense against pathogen infection, metabolism syndrome, cellular stress and cancer metastasis in the liver. The assembly of inflammasome activates caspase-1, which promotes the maturation of interleukin-1β (IL-1β) and interleukin-18 (IL-18), and initiates pyroptotic cell death (pyroptosis). IL-18 exerts pleiotropic effects on hepatic NK cells, priming FasL-mediated cytotoxicity, and interferon-γ (IFN-γ)-dependent responses to prevent the development of liver diseases. However, considerable attention has been attracted to the pathogenic role of inflammasomes in various acute and chronic liver diseases, including viral hepatitis, nanoparticle-induced liver injury, alcoholic and non-alcoholic steatohepatitis. In this review, we summarize the latest advances on the physiological and pathological roles of inflammasomes for further development of inflammasome-based therapeutic strategies for human liver diseases.
Collapse
Affiliation(s)
- Jingyun Luan
- Department of Microbiological and Biochemical Pharmacy & The Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Dianwen Ju
- Department of Microbiological and Biochemical Pharmacy & The Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
29
|
Gaskell H, Ge X, Nieto N. High-Mobility Group Box-1 and Liver Disease. Hepatol Commun 2018; 2:1005-1020. [PMID: 30202816 PMCID: PMC6128227 DOI: 10.1002/hep4.1223] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/03/2018] [Indexed: 12/12/2022] Open
Abstract
High‐mobility group box‐1 (HMGB1) is a ubiquitous protein. While initially thought to be simply an architectural protein due to its DNA‐binding ability, evidence from the last decade suggests that HMGB1 is a key protein participating in the pathogenesis of acute liver injury and chronic liver disease. When it is passively released or actively secreted after injury, HMGB1 acts as a damage‐associated molecular pattern that communicates injury and inflammation to neighboring cells by the receptor for advanced glycation end products or toll‐like receptor 4, among others. In the setting of acute liver injury, HMGB1 participates in ischemia/reperfusion, sepsis, and drug‐induced liver injury. In the context of chronic liver disease, it has been implicated in alcoholic liver disease, liver fibrosis, nonalcoholic steatohepatitis, and hepatocellular carcinoma. Recently, specific posttranslational modifications have been identified that could condition the effects of the protein in the liver. Here, we provide a detailed review of how HMGB1 signaling participates in acute liver injury and chronic liver disease.
Collapse
Affiliation(s)
- Harriet Gaskell
- Department of Pathology University of Illinois at Chicago Chicago IL
| | - Xiaodong Ge
- Department of Pathology University of Illinois at Chicago Chicago IL
| | - Natalia Nieto
- Department of Pathology University of Illinois at Chicago Chicago IL.,Department of Medicine University of Illinois at Chicago Chicago IL
| |
Collapse
|
30
|
Kerr NA, de Rivero Vaccari JP, Abbassi S, Kaur H, Zambrano R, Wu S, Dietrich WD, Keane RW. Traumatic Brain Injury-Induced Acute Lung Injury: Evidence for Activation and Inhibition of a Neural-Respiratory-Inflammasome Axis. J Neurotrauma 2018; 35:2067-2076. [PMID: 29648974 DOI: 10.1089/neu.2017.5430] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Approximately 20-25% of traumatic brain injury (TBI) subjects develop acute lung injury (ALI), but the pathomechanisms of TBI-induced ALI remain poorly defined. Our previous work has shown that the inflammasome plays a critical role in TBI-induced secondary pathophysiology and that inflammasome proteins are released in extracellular vesicles (EV) after TBI. Here we investigated whether EV-mediated inflammasome signaling contributed to the etiology of TBI-induced ALI. C57/BL6 male mice were subjected to controlled cortical impact (CCI), and the brains and lungs were examined for inflammasome activation and ALI at 4 and 24 h after TBI. We show that TBI releases EV containing inflammasome proteins into serum that target the lung to cause ALI, supporting activation of a neural-respiratory-inflammasome axis. Administration of a low-molecular-weight heparin (enoxaparin, a blocker of EV uptake) or treatment with a monoclonal antibody against apoptosis speck-like staining protein containing a caspase recruitment domain (anti-ASC) after adoptive transfer of EV isolated from TBI-injured mice significantly inhibited inflammasome activation in the lungs of recipient mice resulting in improved ALI scores.This axis constitutes an important arm of the innate inflammatory response in lung pathology after TBI and targeting this axis represents a novel therapeutic treatment for TBI-induced ALI.
Collapse
Affiliation(s)
- Nadine A Kerr
- 1 Department of Neurological Surgery, University of Miami Miller School of Medicine , Miami, Florida.,2 Department of Physiology and Biophysics, University of Miami Miller School of Medicine , Miami, Florida
| | - Juan Pablo de Rivero Vaccari
- 1 Department of Neurological Surgery, University of Miami Miller School of Medicine , Miami, Florida.,2 Department of Physiology and Biophysics, University of Miami Miller School of Medicine , Miami, Florida
| | - Sam Abbassi
- 2 Department of Physiology and Biophysics, University of Miami Miller School of Medicine , Miami, Florida
| | - Harmanpreet Kaur
- 1 Department of Neurological Surgery, University of Miami Miller School of Medicine , Miami, Florida
| | - Ronald Zambrano
- 3 Department of Pediatrics, University of Miami Miller School of Medicine , Miami, Florida
| | - Shu Wu
- 3 Department of Pediatrics, University of Miami Miller School of Medicine , Miami, Florida
| | - W Dalton Dietrich
- 1 Department of Neurological Surgery, University of Miami Miller School of Medicine , Miami, Florida
| | - Robert W Keane
- 1 Department of Neurological Surgery, University of Miami Miller School of Medicine , Miami, Florida.,2 Department of Physiology and Biophysics, University of Miami Miller School of Medicine , Miami, Florida
| |
Collapse
|
31
|
Lei Z, Deng M, Yi Z, Sun Q, Shapiro RA, Xu H, Li T, Loughran PA, Griepentrog JE, Huang H, Scott MJ, Huang F, Billiar TR. cGAS-mediated autophagy protects the liver from ischemia-reperfusion injury independently of STING. Am J Physiol Gastrointest Liver Physiol 2018; 314:G655-G667. [PMID: 29446653 PMCID: PMC6032062 DOI: 10.1152/ajpgi.00326.2017] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Liver ischemia-reperfusion (I/R) injury occurs through induction of oxidative stress and release of damage-associated molecular patterns (DAMPs), including cytosolic DNA released from dysfunctional mitochondria or from the nucleus. Cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS) is a cytosolic DNA sensor known to trigger stimulator of interferon genes (STING) and downstream type 1 interferon (IFN-I) pathways, which are pivotal innate immune system responses to pathogen. However, little is known about the role of cGAS/STING in liver I/R injury. We subjected C57BL/6 (WT), cGAS knockout (cGAS-/-), and STING-deficient (STINGgt/gt) mice to warm liver I/R injury and that found cGAS-/- mice had significantly increased liver injury compared with WT or STINGgt/gt mice, suggesting a protective effect of cGAS independent of STING. Liver I/R upregulated cGAS in vivo and also in vitro in hepatocytes subjected to anoxia/reoxygenation (A/R). We confirmed a previously published finding that hepatocytes do not express STING under normoxic conditions or after A/R. Hepatocytes and liver from cGAS-/- mice had increased cell death and reduced induction of autophagy under hypoxic conditions as well as increased apoptosis. Protection could be restored in cGAS-/- hepatocytes by overexpression of cGAS or by pretreatment of mice with autophagy inducer rapamycin. Our findings indicate a novel protective role for cGAS in the regulation of autophagy during liver I/R injury that occurs independently of STING. NEW & NOTEWORTHY Our studies are the first to document the important role of cGAS in the acute setting of sterile injury induced by I/R. Specifically, we provide evidence that cGAS protects liver from I/R injury in a STING-independent manner.
Collapse
Affiliation(s)
- Zhao Lei
- 1Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China,2Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Meihong Deng
- 2Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Zhongjie Yi
- 1Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China,2Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Qian Sun
- 2Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Richard A. Shapiro
- 2Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Hongbo Xu
- 1Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China,2Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Tunliang Li
- 2Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Patricia A. Loughran
- 2Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania,4Center for Biologic Imaging, University of Pittsburgh, Pennsylvania
| | | | - Hai Huang
- 2Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania,3Pittsburgh Liver Research Center, University of Pittsburgh, Pennsylvania
| | - Melanie J. Scott
- 2Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania,3Pittsburgh Liver Research Center, University of Pittsburgh, Pennsylvania
| | - Feizhou Huang
- 1Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Timothy R. Billiar
- 2Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania,3Pittsburgh Liver Research Center, University of Pittsburgh, Pennsylvania
| |
Collapse
|
32
|
Abstract
Traumatic injury as one of the world's most relevant but neglected health concerns results in modulated inflammasome activity, which is closely linked to the development of post-injury complications. Cytokine-producing capacity of cells is important for the appropriate immune response to trauma and requires not only synthesis and transcription of inflammasome components but also their activation. Unfortunately, the precise role of inflammasome in trauma is still largely unknown. However, in the following chapter, we provide an overview on the best described inflammasomes in the various settings of trauma, introducing the recent findings on the up-to-date best described NLRP inflammasomes and underlying cytokines in the inflammatory response to trauma.
Collapse
Affiliation(s)
- Borna Relja
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.
| | | |
Collapse
|
33
|
Vodovotz Y, Simmons RL, Gandhi CR, Barclay D, Jefferson BS, Huang C, Namas R, El-Dehaibi F, Mi Q, Billiar TR, Zamora R. "Thinking" vs. "Talking": Differential Autocrine Inflammatory Networks in Isolated Primary Hepatic Stellate Cells and Hepatocytes under Hypoxic Stress. Front Physiol 2017; 8:1104. [PMID: 29312006 PMCID: PMC5743931 DOI: 10.3389/fphys.2017.01104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 12/14/2017] [Indexed: 12/23/2022] Open
Abstract
We hypothesized that isolated primary mouse hepatic stellate cells (HSC) and hepatocytes (HC) would elaborate different inflammatory responses to hypoxia with or without reoxygenation. We further hypothesized that intracellular information processing (“thinking”) differs from extracellular information transfer (“talking”) in each of these two liver cell types. Finally, we hypothesized that the complexity of these autocrine responses might only be defined in the absence of other non-parenchymal cells or trafficking leukocytes. Accordingly, we assayed 19 inflammatory mediators in the cell culture media (CCM) and whole cell lysates (WCLs) of HSC and HC during hypoxia with and without reoxygenation. We applied a unique set of statistical and data-driven modeling techniques including Two-Way ANOVA, hierarchical clustering, Principal Component Analysis (PCA) and Network Analysis to define the inflammatory responses of these isolated cells to stress. HSC, under hypoxic and reoxygenation stresses, both expressed and secreted larger quantities of nearly all inflammatory mediators as compared to HC. These differential responses allowed for segregation of HSC from HC by hierarchical clustering. PCA suggested, and network analysis supported, the hypothesis that above a certain threshold of cellular stress, the inflammatory response becomes focused on a limited number of functions in both HSC and HC, but with distinct characteristics in each cell type. Network analysis of separate extracellular and intracellular inflammatory responses, as well as analysis of the combined data, also suggested the presence of more complex inflammatory “talking” (but not “thinking”) networks in HSC than in HC. This combined network analysis also suggested an interplay between intracellular and extracellular mediators in HSC under more conditions than that observed in HC, though both cell types exhibited a qualitatively similar phenotype under hypoxia/reoxygenation. Our results thus suggest that a stepwise series of computational and statistical analyses may help decipher how cells respond to environmental stresses, both within the cell and in its secretory products, even in the absence of cooperation from other cells in the liver.
Collapse
Affiliation(s)
- Yoram Vodovotz
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Richard L Simmons
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Derek Barclay
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Chao Huang
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rami Namas
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Fayten El-Dehaibi
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Qi Mi
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ruben Zamora
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
34
|
Sun Q, Fan J, Billiar TR, Scott MJ. Inflammasome and autophagy regulation - a two-way street. Mol Med 2017; 23:188-195. [PMID: 28741645 DOI: 10.2119/molmed.2017.00077] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/20/2017] [Indexed: 01/22/2023] Open
Abstract
Inflammation plays a significant role in protecting hosts against pathogens. Inflammation induced by non-infectious, endogenous agents can be detrimental, and if excessive can result in organ and tissue damage. The inflammasome is a major innate immune pathway that can be activated via both exogenous pathogen-associated molecular patterns (PAMPs) and endogenous damage-associated molecular patterns (DAMPs). Inflammasome activation involves formation and oligomerization of a protein complex including a NOD-like receptor (NLR), an adaptor protein (ASC) and procaspase-1. This then allows cleavage and activation of caspase-1, followed by downstream cleavage and release of proinflammatory cytokines, IL-1β and IL-18, from innate immune cells. Hyperinflammation caused by unrestrained inflammasome activation is linked with multiple inflammatory diseases, including inflammatory bowel disease, Alzheimer's disease and multiple sclerosis. So there is an understandable rush to understand mechanisms that regulate such potent inflammatory pathways. Autophagy has now been identified as a main regulator of inflammasomes. Autophagy is a vital intracellular process involved in cellular homeostasis, recycling and removal of damaged organelles (e.g. mitochondria) and intracellular pathogens. Autophagy is regulated by proteins that are important in endosomal/phagosomal pathways, as well as by specific autophagy proteins coded for by autophagy-related genes. Cytosolic components are surrounded and contained by a double-membraned vesicle, which then fuses with lysosomes to enable degradation of the contents. Autophagic removal of intracellular DAMPs, inflammasome components or cytokines can reduce inflammasome activation. Similarly, inflammasomes can regulate the autophagic process, allowing for a two-way, mutual regulation of inflammation that may hold the key for treatment of multiple diseases.
Collapse
Affiliation(s)
- Qian Sun
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213.,Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Melanie J Scott
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| |
Collapse
|