1
|
Datta S, Rahman MA, Koka S, Boini KM. High Mobility Group Box 1 (HMGB1): Molecular Signaling and Potential Therapeutic Strategies. Cells 2024; 13:1946. [PMID: 39682695 PMCID: PMC11639863 DOI: 10.3390/cells13231946] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
High Mobility Group Box 1 (HMGB1) is a highly conserved non-histone chromatin-associated protein across species, primarily recognized for its regulatory impact on vital cellular processes, like autophagy, cell survival, and apoptosis. HMGB1 exhibits dual functionality based on its localization: both as a non-histone protein in the nucleus and as an inducer of inflammatory cytokines upon extracellular release. Pathophysiological insights reveal that HMGB1 plays a significant role in the onset and progression of a vast array of diseases, viz., atherosclerosis, kidney damage, cancer, and neurodegeneration. However, a clear mechanistic understanding of HMGB1 release, translocation, and associated signaling cascades in mediating such physiological dysfunctions remains obscure. This review presents a detailed outline of HMGB1 structure-function relationship and its regulatory role in disease onset and progression from a signaling perspective. This review also presents an insight into the status of HMGB1 druggability, potential limitations in understanding HMGB1 pathophysiology, and future perspective of studies that can be undertaken to address the existing scientific gap. Based on existing paradigm of various studies, HMGB1 is a critical regulator of inflammatory cascades and drives the onset and progression of a broad spectrum of dysfunctions. Studies focusing on HMGB1 druggability have enabled the development of biologics with potential clinical benefits. However, deeper understanding of post-translational modifications, redox states, translocation mechanisms, and mitochondrial interactions can potentially enable the development of better courses of therapy against HMGB1-mediated physiological dysfunctions.
Collapse
Affiliation(s)
- Sayantap Datta
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Mohammad Atiqur Rahman
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Saisudha Koka
- Department of Pharmaceutical Sciences, Irma Lerma College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA;
| | - Krishna M. Boini
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
2
|
Yamaga S, Aziz M, Murao A, Brenner M, Wang P. DAMPs and radiation injury. Front Immunol 2024; 15:1353990. [PMID: 38333215 PMCID: PMC10850293 DOI: 10.3389/fimmu.2024.1353990] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
The heightened risk of ionizing radiation exposure, stemming from radiation accidents and potential acts of terrorism, has spurred growing interests in devising effective countermeasures against radiation injury. High-dose ionizing radiation exposure triggers acute radiation syndrome (ARS), manifesting as hematopoietic, gastrointestinal, and neurovascular ARS. Hematopoietic ARS typically presents with neutropenia and thrombocytopenia, while gastrointestinal ARS results in intestinal mucosal injury, often culminating in lethal sepsis and gastrointestinal bleeding. This deleterious impact can be attributed to radiation-induced DNA damage and oxidative stress, leading to various forms of cell death, such as apoptosis, necrosis and ferroptosis. Damage-associated molecular patterns (DAMPs) are intrinsic molecules released by cells undergoing injury or in the process of dying, either through passive or active pathways. These molecules then interact with pattern recognition receptors, triggering inflammatory responses. Such a cascade of events ultimately results in further tissue and organ damage, contributing to the elevated mortality rate. Notably, infection and sepsis often develop in ARS cases, further increasing the release of DAMPs. Given that lethal sepsis stands as a major contributor to the mortality in ARS, DAMPs hold the potential to function as mediators, exacerbating radiation-induced organ injury and consequently worsening overall survival. This review describes the intricate mechanisms underlying radiation-induced release of DAMPs. Furthermore, it discusses the detrimental effects of DAMPs on the immune system and explores potential DAMP-targeting therapeutic strategies to alleviate radiation-induced injury.
Collapse
Affiliation(s)
- Satoshi Yamaga
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Atsushi Murao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Max Brenner
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| |
Collapse
|
3
|
Sheu ML, Pan LY, Yang CN, Sheehan J, Pan LY, You WC, Wang CC, Chen HS, Pan HC. Neuronal Death Caused by HMGB1-Evoked via Inflammasomes from Thrombin-Activated Microglia Cells. Int J Mol Sci 2023; 24:12664. [PMID: 37628850 PMCID: PMC10454604 DOI: 10.3390/ijms241612664] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Microglial cells are a macrophage-like cell type residing within the CNS. These cells evoke pro-inflammatory responses following thrombin-induced brain damage. Inflammasomes, which are large caspase-1-activating protein complexes, play a critical role in mediating the extracellular release of HMGB1 in activated immune cells. The exact role of inflammasomes in microglia activated by thrombin remains unclear, particularly as it relates to the downstream functions of HMGB1. After receiving microinjections of thrombin, Sprague Dawley rats of 200 to 250 gm were studied in terms of behaviors and immunohistochemical staining. Primary culture of microglia cells and BV-2 cells were used for the assessment of signal pathways. In a water maze test and novel object recognition analysis, microinjections of thrombin impaired rats' short-term and long-term memory, and such detrimental effects were alleviated by injecting anti-HMGB-1 antibodies. After thrombin microinjections, the increased oxidative stress of neurons was aggravated by HMGB1 injections but attenuated by anti-HMGB-1 antibodies. Such responses occurred in parallel with the volume of activated microglia cells, as well as their expressions of HMGB-1, IL-1β, IL-18, and caspase-I. In primary microglia cells and BV-2 cell lines, thrombin also induced NO release and mRNA expressions of iNOS, IL-1β, IL-18, and activated caspase-I. HMGB-1 aggravated these responses, which were abolished by anti-HMGB-1 antibodies. In conclusion, thrombin induced microglia activation through triggering inflammasomes to release HMGB1, contributing to neuronal death. Such an action was counteracted by the anti-HMGB-1 antibodies. The refinement of HMGB-1 modulated the neuro-inflammatory response, which was attenuated in thrombin-associated neurodegenerative disorder.
Collapse
Affiliation(s)
- Meei-Ling Sheu
- Institute of Biomedical Sciences, National Chung-Hsing University, Taichung 40227, Taiwan;
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40210, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | - Liang-Yi Pan
- Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Cheng-Ning Yang
- Department of Dentistry, School of Dentistry, College of Medicine, National Taiwan University, Taipei 106319, Taiwan;
| | - Jason Sheehan
- Department of Neurosurgery, University of Virginia, Charlottesville, VA 22904, USA;
| | - Liang-Yu Pan
- Faculty of Medicine, Poznan University of Medical Sciences, 61-701 Poznań, Poland;
| | - Weir-Chiang You
- Department of Radiation Oncology, Taichung Veterans General Hospital, Taichung 40210, Taiwan;
| | - Chien-Chia Wang
- Department of Life Sciences, National Central University, Taoyuan 32001, Taiwan;
| | - Hong-Shiu Chen
- Department of Neurosurgery, Tungs’ Taichung Metro-Harbor Hospital, Taichung 40210, Taiwan;
| | - Hung-Chuan Pan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40210, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung 40210, Taiwan
| |
Collapse
|
4
|
Wu L, Pu L, Zhuang Z. miR-155-5p/FOXO3a promotes pulmonary fibrosis in rats by mediating NLRP3 inflammasome activation. Immunopharmacol Immunotoxicol 2023; 45:257-267. [PMID: 35997271 DOI: 10.1080/08923973.2022.2115923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Pulmonary fibrosis (PF) is regarded as progressive lung disease. miR-155-5p depletion exerts anti-fibrotic effects in silicotic mice. This study explored the effect and possible mechanism of miR-155-5p in PF rats, hoping to find a new target for PF management. METHODS Bleomycin-induced PF rat model was established. Alveolar structure and collagen fiber deposition were observed by HE and Elastica-Masson staining. Alveolitis and PF scores were evaluated using the method of Szapiel. Total collagen content was detected using the Sircol method. PF rats were intraperitoneally injected with NLRP3 inhibitor MCC950 or intravenously injected with miR-155-5p antagomir and si-FOXO3a lentivirus plasmids. Binding sites of miR-155-5p and FOXO3a were predicted using bioinformatics analysis and dual-luciferase reporter assay. The expressions of miR-155-5p, NLRP3, ASC, caspase-1, IL-1β, IL-18, and FOXO3a were detected by RT-qPCR, Western blot, and ELISA. RESULTS MCC950 treatment inhibited NLRP3 inflammasome, alleviated alveolar hemorrhage and alveolitis, and reduced blue collagen fiber deposition, scores of alveolitis and PF, and levels of NLRP3, ASC, caspase-1, IL-1β, and IL-18 in PF rats. miR-155-5p was elevated in lung tissues of PF rats. Inhibition of miR-155-5p downregulated levels of NLRP3, ASC, caspase-1, IL-1β, and IL-18 in lung tissues of PF rats. miR-155-5p targeted FOXO3a. miR-155-5p inhibition and silencing FOXO3a exacerbated alveolitis and PF in rats and increased levels of NLRP3, ASC, caspase-1, IL-1β, and IL-18. CONCLUSIONS miR-155-5p aggravated alveolitis and promoted PF by targeting FOXO3a and prompting the activation of NLRP3 inflammasome and then inducing IL-1β and IL-18 release.
Collapse
Affiliation(s)
- Ling Wu
- Department of Respiratory, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, Changzhou, PR China
| | - Li Pu
- Department of Respiratory, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, Changzhou, PR China
| | - Zhifang Zhuang
- Department of Respiratory, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, Changzhou, PR China
| |
Collapse
|
5
|
Baicalein Attenuates Severe Polymicrobial Sepsis via lleviating Immune Dysfunction of T Lymphocytes and Inflammation. Chin J Integr Med 2022; 28:711-718. [PMID: 35355199 DOI: 10.1007/s11655-022-3510-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To investigate the effect of baicalein on polymicrobial sepsis-induced immune dysfunction and organ injury. METHODS A sepsis model was induced in Sprague-Dawley rats via caecal ligation and puncture (CLP). Specific pathogen free rats were randomly divided into a sham group, CLP group and CLP + baicalein (Bai) group (n=16 each). Rats in the CLP + Bai group were intravenously injected with baicalein (20 mg/kg) at 1 and 10 h after CLP. Survival rate, bacterial load, and organ damage were assessed. Then each group was evaluated at 6, 12, and 24 h to investigate the effect of baicalein on immune cells and inflammatory cytokines in septic rats. RESULTS Baicalein treatment significantly improved the survival of septic rats, decreased the bacterial burden, and moderated tissue damage (spleen, liver, and lung), as observed by haematoxylin and eosin staining. Septic rats treated with baicalein had strikingly increased proportions of CD3+CD4+ T cells and ratios of CD4+/CD8+ T cells in the peripheral blood and spleen (all P<0.05). Moreover, baicalein treatment decreased the apoptotic rate of whole white blood cells and spleen cells at 24 h after surgery (P<0.05). Baicalein significantly reduced the levels of tumor necrosis factor α and interleukin-6 (IL-6) and increased IL-10, and the expression levels of galectin 9 were also raised in the spleen (P<0.01). CONCLUSION Baicalein may be an effective immunomodulator that attenuates overwhelming inflammatory responses in severe abdominal sepsis.
Collapse
|
6
|
Wang Z, Yang X, Wang X, Liang F, Tang Y. Glycyrrhizin attenuates caspase-11-dependent immune responses and coagulopathy by targeting high mobility group box 1. Int Immunopharmacol 2022; 107:108713. [PMID: 35339905 DOI: 10.1016/j.intimp.2022.108713] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/10/2022] [Accepted: 03/15/2022] [Indexed: 11/05/2022]
Abstract
Caspase-11, a cytosolic endotoxin (lipopolysaccharide: LPS) receptor, mediates pyroptosis, coagulopathy and lethality in endoxemia and bacterial sepsis. The activation of caspase-11 requires high mobility group box 1 (HMGB1)-mediated translocation of LPS from the extracellular space to the cytosol. Here we show that HMGB1-dependent cytosolic delivery of LPS was blocked by glycyrrhizin, a medication to treat liver diseases. Glycyrrhizin competitively bound HMGB1 and thereby inhibiting the physical interaction between HMGB1 and LPS. Treatment of glycyrrhizin significantly attenuated caspase-11-dependent immune responses, coagulopathy, organ injury and lethality in endotoxemia and experimental sepsis. Together, our data suggest that pharmacological inhibition of the cytosolic delivery of LPS by glycyrrhizin might be a potential therapeutic strategy to treat sepsis, which is a leading cause of death in hospitals worldwide.
Collapse
Affiliation(s)
- Zhongtai Wang
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha 410000, PR China
| | - Xinyu Yang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Xiangyu Wang
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha 410000, PR China
| | - Fang Liang
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha 410000, PR China.
| | - Yiting Tang
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha 410000, PR China; Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province 410000, PR China.
| |
Collapse
|
7
|
The Effect and Regulatory Mechanism of High Mobility Group Box-1 Protein on Immune Cells in Inflammatory Diseases. Cells 2021; 10:cells10051044. [PMID: 33925132 PMCID: PMC8145631 DOI: 10.3390/cells10051044] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/18/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
High mobility group box-1 protein (HMGB1), a member of the high mobility group protein superfamily, is an abundant and ubiquitously expressed nuclear protein. Intracellular HMGB1 is released by immune and necrotic cells and secreted HMGB1 activates a range of immune cells, contributing to the excessive release of inflammatory cytokines and promoting processes such as cell migration and adhesion. Moreover, HMGB1 is a typical damage-associated molecular pattern molecule that participates in various inflammatory and immune responses. In these ways, it plays a critical role in the pathophysiology of inflammatory diseases. Herein, we review the effects of HMGB1 on various immune cell types and describe the molecular mechanisms by which it contributes to the development of inflammatory disorders. Finally, we address the therapeutic potential of targeting HMGB1.
Collapse
|
8
|
Wang Y, Yu Z, Yuan H, Chen H, Xie N, Wang Z, Sun Q, Zhang W. Structure-based design of glycyrrhetinic acid derivatives as potent anti-sepsis agents targeting high-mobility group box-1. Bioorg Chem 2020; 106:104461. [PMID: 33223202 DOI: 10.1016/j.bioorg.2020.104461] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 10/15/2020] [Accepted: 11/02/2020] [Indexed: 12/15/2022]
Abstract
Novel Glycyrrhetinic Acid (GA) derivatives with fused heterocycles on A ring were structure-based designed and synthesized. Their potential anti-inflammatory effects were investigated by a classical LPS stimulated macrophage model. Surface plasmon resonance (SPR) was used to verify the binding of GA analogues with HMGB1. A preliminary structure-activity relationship was summarized and an analogue GA-60 with ortho-methoxybenzyl pyrozole showed stronger anti-inflammatory effect and higher affinity for HMGB1 with a Kd value of 12.5 μM. In addition, this compound exhibited excellent inhibitory functions on NO (96%), TNF-α (94%), and IL-6 (100%), by interfering with phosphorylation of p38, ERK, JNK MAPKs, as well as that of NF-κB p65 and IKKα/β. Moreover, GA-60 extended the survival of either the classic CLP-induced or LPS-induced sepsis mouse models. Molecular modeling predictions further supported these findings, clearly indicating that inhibiting HMGB1 release, using fused heterocyclic GA derivatives, is a promising strategy for treatment of sepsis.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Interdisciplinary Science Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zongmin Yu
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China; Department of Medical Service Training Center, No. 965 Hospital, Joint Logistics Support Force of PLA, Jilin 132011, China
| | - Hu Yuan
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Jiangxi 341000, China
| | - Hao Chen
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China; State Key Laboratory of Innovative Natural Medicine and TCM Injections, Jiangxi 341000, China
| | - Ning Xie
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Jiangxi 341000, China
| | - Zhibin Wang
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| | - Qingyan Sun
- Shanghai Institute of Pharmaceutical Industry, Shanghai 200040, China.
| | - Weidong Zhang
- Interdisciplinary Science Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Institute of Pharmaceutical Industry, Shanghai 200040, China; School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
9
|
Li J, Bao G, Wang H. Time to Develop Therapeutic Antibodies Against Harmless Proteins Colluding with Sepsis Mediators? Immunotargets Ther 2020; 9:157-166. [PMID: 33117741 PMCID: PMC7547129 DOI: 10.2147/itt.s262605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/22/2020] [Indexed: 12/29/2022] Open
Abstract
Sepsis refers to a systemic inflammatory response syndrome resulting from microbial infections, and is partly attributable to dysregulated inflammation and associated immunosuppression. A ubiquitous nuclear protein, HMGB1, is secreted by activated leukocytes to orchestrate inflammatory responses during early stages of sepsis. When it is released by injured somatic cells at overwhelmingly higher quantities, HMGB1 may induce macrophage pyroptosis and immunosuppression, thereby impairing the host's ability to eradicate microbial infections. A number of endogenous proteins have been shown to bind HMGB1 to modulate its extracellular functions. Here, we discuss an emerging possibility to develop therapeutic antibodies against harmless proteins that collude with pathogenic mediators for the clinical management of human sepsis and other inflammatory diseases.
Collapse
Affiliation(s)
- Jianhua Li
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY11030, USA
| | - Guoqiang Bao
- Department of General Surgery, Tangdu Hospital, Xi’an, Shaanxi710032, People’s Republic of China
| | - Haichao Wang
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY11549, USA
| |
Collapse
|
10
|
Hoffman M, Kyriazis ID, Lucchese AM, de Lucia C, Piedepalumbo M, Bauer M, Schulze PC, Bonios MJ, Koch WJ, Drosatos K. Myocardial Strain and Cardiac Output are Preferable Measurements for Cardiac Dysfunction and Can Predict Mortality in Septic Mice. J Am Heart Assoc 2020; 8:e012260. [PMID: 31112430 PMCID: PMC6585345 DOI: 10.1161/jaha.119.012260] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Sepsis is the overwhelming host response to infection leading to shock and multiple organ dysfunction. Cardiovascular complications greatly increase sepsis‐associated mortality. Although murine models are routinely used for preclinical studies, the benefit of using genetically engineered mice in sepsis is countered by discrepancies between human and mouse sepsis pathophysiology. Therefore, recent guidelines have called for standardization of preclinical methods to document organ dysfunction. We investigated the course of cardiac dysfunction and myocardial load in different mouse models of sepsis to identify the optimal measurements for early systolic and diastolic dysfunction. Methods and Results We performed speckle‐tracking echocardiography and assessed blood pressure, plasma inflammatory cytokines, lactate, B‐type natriuretic peptide, and survival in mouse models of endotoxemia or polymicrobial infection (cecal ligation and puncture, [CLP]) of moderate and high severity. We observed that myocardial strain and cardiac output were consistently impaired early in both sepsis models. Suppression of cardiac output was associated with systolic dysfunction in endotoxemia or combined systolic dysfunction and reduced preload in the CLP model. We found that cardiac output at 2 hours post‐CLP is a negative prognostic indicator with high sensitivity and specificity that predicts mortality at 48 hours. Using a known antibiotic (ertapenem) treatment, we confirmed that this approach can document recovery. Conclusions We propose a non‐invasive approach for assessment of cardiac function in sepsis and myocardial strain and strain rate as preferable measures for monitoring cardiovascular function in sepsis mouse models. We further show that the magnitude of cardiac output suppression 2 hours post‐CLP can be used to predict mortality.
Collapse
Affiliation(s)
- Matthew Hoffman
- 1 Center for Translational Medicine and Department of Pharmacology Lewis Katz School of Medicine Temple University Philadelphia PA
| | - Ioannis D Kyriazis
- 1 Center for Translational Medicine and Department of Pharmacology Lewis Katz School of Medicine Temple University Philadelphia PA
| | - Anna M Lucchese
- 1 Center for Translational Medicine and Department of Pharmacology Lewis Katz School of Medicine Temple University Philadelphia PA
| | - Claudio de Lucia
- 1 Center for Translational Medicine and Department of Pharmacology Lewis Katz School of Medicine Temple University Philadelphia PA
| | - Michela Piedepalumbo
- 1 Center for Translational Medicine and Department of Pharmacology Lewis Katz School of Medicine Temple University Philadelphia PA.,2 Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences University of Campania "Luigi Vanvitelli" Naples Italy
| | - Michael Bauer
- 3 Department for Anesthesiology and Intensive Care Medicine Friedrich-Schiller-University Jena Germany
| | - P Christian Schulze
- 4 Division of Cardiology, Angiology, Intensive Medical Care and Pneumology Department of Internal Medicine I University Hospital Jena Germany
| | - Michael J Bonios
- 5 Heart Failure and Transplant Unit Onassis Cardiac Surgery Center Athens Greece
| | - Walter J Koch
- 1 Center for Translational Medicine and Department of Pharmacology Lewis Katz School of Medicine Temple University Philadelphia PA
| | - Konstantinos Drosatos
- 1 Center for Translational Medicine and Department of Pharmacology Lewis Katz School of Medicine Temple University Philadelphia PA
| |
Collapse
|
11
|
Amada E, Fukuda K, Kumagai K, Kawakubo H, Kitagawa Y. Soluble recombinant human thrombomodulin suppresses inflammation-induced gastrointestinal tumor growth in a murine peritonitis model. Mol Cell Biochem 2020; 475:195-203. [PMID: 32767229 DOI: 10.1007/s11010-020-03872-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 08/01/2020] [Indexed: 12/27/2022]
Abstract
Regulatory T cells (Tregs) and transforming growth factor β (TGF-β) are believed to play key roles in both postoperative pro-inflammatory and anti-inflammatory responses of malignancies. Recombinant human thrombomodulin (rTM) is implied to inhibit the interaction between TGF-β and Tregs. The aim of this study is to evaluate the antitumor effects of rTM against gastrointestinal tumors under systemic inflammation. Mice were subjected to cecal ligation and puncture and percutaneous allogeneic tumor implantation. rTM were introduced by percutaneous injection into the abdominal cavity. The effects of rTM were evaluated by weight of implanted tumor, proportion of Tregs in peripheral blood lymphocytes (PBL) and tumor infiltrating lymphocytes (TIL) and temporal evaluation of serum cytokines. The effect of rTM was also evaluated on the in vitro differentiation of naïve T cells into induced Tregs induced by TGF-β and interleukin (IL) -2. rTM significantly inhibited the proliferation of the implanted tumor cells in an inflammation-dependent manner. rTM also reduced the fractions of regulatory T cells and induced regulatory T cells among both PBL and TIL. Temporal evaluation of serum cytokine levels in the model mice showed that rTM significantly suppressed the increases in the serum levels of IL-2 and TGF-β. An in vitro differentiation assay revealed that rTM inhibited the differentiation of naïve T cells into Tregs triggered by IL-2- and TGF-β. rTM has suppressive effects on inflammation-induced gastrointestinal tumor growth by suggestively affecting differentiation of Tregs.
Collapse
Affiliation(s)
- En Amada
- Department of Surgery, School of Medicine, Keio University, 35, Shinanomachi, Shinjuku, Tokyo, Japan
| | - Kazumasa Fukuda
- Department of Surgery, School of Medicine, Keio University, 35, Shinanomachi, Shinjuku, Tokyo, Japan
| | - Koshi Kumagai
- Department of Gastroenterological Surgery, The Cancer Institute Hospital of JFCR, 3-8-31, Ariake, Koto, Tokyo, Japan.
| | - Hirofumi Kawakubo
- Department of Surgery, School of Medicine, Keio University, 35, Shinanomachi, Shinjuku, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, School of Medicine, Keio University, 35, Shinanomachi, Shinjuku, Tokyo, Japan
| |
Collapse
|
12
|
Infection-induced innate antimicrobial response disorders: from signaling pathways and their modulation to selected biomarkers. Cent Eur J Immunol 2020; 45:104-116. [PMID: 32425688 PMCID: PMC7226557 DOI: 10.5114/ceji.2020.94712] [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: 07/18/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022] Open
Abstract
Severe infections are a major public health problem responsible for about 40-65% of hospitalizations in intensive care units (ICU). The high mortality (30-50%) of persons diagnosed with severe infection is caused by largely unknown mechanisms of sepsis-induced immune system response. Severe infections with dynamic progress are accompanied with SIRS (systemic inflammatory reaction syndrome) and CARS (compensatory anti-inflammatory response syndrome), and require a biological treatment appropriate to the phase of immune response. The mechanisms responsible for severe infection related to immune system response particularly attract extensive interest of non-specific defense mechanisms, including signaling pathways of Toll-like receptors (mainly TLR4 and TLR2) that recognize distinct pathogen-associated molecular patterns (PAMP) and play a critical role in innate immune response. There are attempts of treatment, followed by blocking ligand binding with TLR or modulation of intracellular signaling pathways, to inhibit signal transduction. Moreover, researches regarding new and more efficient diagnostics biomarkers were mostly focused on indicators related to innate response to infection as well as connections of pro-inflammatory response with anti-inflammatory response.According to these studies, in case of ICU septic patients with high-risk of mortality, the solution for the problem will require mainly early immune and genetic diagnostics (e.g. cytokines, microRNA, cluster of differentiation-64 [CD64], triggering receptor expressed on myeloid cells-1 [TREM-1], and high mobility group box 1 protein [HMGB1]).
Collapse
|
13
|
High mobility group box protein 1 neutralization therapy in ovine bacteremia: Lessons learned from an ovine septic shock model incorporating intensive care support. Exp Ther Med 2019; 18:3271-3280. [PMID: 31602200 PMCID: PMC6777221 DOI: 10.3892/etm.2019.7961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 02/19/2019] [Indexed: 12/12/2022] Open
Abstract
Sepsis is a highly complex and often fatal syndrome which varies widely in its clinical manifestations, and therapies that target the underlying uncontrolled immune status in sepsis are needed. The failure of preclinical approaches to provide significant sepsis survival benefit in the clinic is often attributed to inappropriate animal disease models. It has been demonstrated that high mobility group box protein 1 (HMGB1) blockade can reduce inflammation, mortality and morbidity in experimental sepsis without promoting immunosuppression. Within this study, we explored the use of ovine anti-HMGB1 antibodies in a model of ovine septic shock incorporating intensive care supports (OSSICS). Results: Septic sheep exhibited elevated levels of HMGB1 within 12 h after the induction of sepsis. In this study, sepsis was induced in six anaesthetized adult Border Leicester × Merino ewes via intravenous instillation of E. coli and sheep monitored according to intensive care unit standard protocols for 26 h, with the requirement for noradrenaline as the primary endpoint. Septic sheep exhibited a hyperdynamic circulation, renal dysfunction, deranged coagulation profile and severe metabolic acidosis. Sheep were assigned a severity of illness score, which increased over time. While a therapeutic effect of intravenous anti-HMGB1 antibody could not be observed in this model due to limited animal numbers, a reduced bacterial dose induced a septic syndrome of much lower severity. With modifications including a reduced bacterial dose, a longer timeframe and broad spectrum antibiotics, the OSSICS model may become a robust tool for preclinical assessment of sepsis therapeutics.
Collapse
|
14
|
Chen F, Liu Z, Li W, Li D, Yan B. The significance of serum HMGB1 level in humans with acute paraquat poisoning. Sci Rep 2019; 9:7448. [PMID: 31092889 PMCID: PMC6520495 DOI: 10.1038/s41598-019-43877-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 04/28/2019] [Indexed: 01/04/2023] Open
Abstract
High-mobility group box 1 (HMGB1) mediates acute lung injury in a mouse model of paraquat poisoning. However, published reports showing a clinically relevant association between HMGB1 and paraquat exposure are lacking. The objective of the present study was to investigate the potential role of serum HMGB1 level as a prognostic marker of mortality in patients with paraquat poisoning in a clinical setting. This retrospective observational cohort study included a convenience sample of 92 patients with acute paraquat poisoning admitted to the emergency room (ER) of The First Hospital of Jilin University between January 2014 and December 2016. Baseline serum HMGB1 levels and other laboratory parameters were measured on admission. Cumulative incidence of mortality during the first 30 days after admission was 50% (n = 46/92). Serum HMGB1 levels were higher in fatalities than survivors (P = 0.015), 30-day mortality increased with increasing baseline serum HMGB1 level (P < 0.001), and higher serum HMGB1 levels were associated with an increase in 30-day mortality on Kaplan-Meier analysis. Multivariate Cox regression analysis identified baseline serum HMGB1 levels, white blood cell count, and serum lactic acid levels as independent prognostic markers of 30-day mortality. These data suggest that serum HMGB1 levels measured on admission to the ER are an independent predictor of 30-day mortality in patients with acute paraquat poisoning.
Collapse
Affiliation(s)
- Feng Chen
- Dermatology Department, China-Japan Union Hospital of Jilin University, 126Xiantai Street, Changchun, 130033, Jilin Province, People's Republic of China
| | - Zuolong Liu
- Department of Emergency, The First Hospital of Jilin University, 71Xinmin Street, Changchun, 130021, Jilin Province, People's Republic of China
| | - Wei Li
- Department of Emergency, The First Hospital of Jilin University, 71Xinmin Street, Changchun, 130021, Jilin Province, People's Republic of China
| | - Dan Li
- Department of Respiratory Medicine, The First Hospital of Jilin University, 71Xinmin Street, Changchun, 130021, Jilin Province, People's Republic of China.
| | - Bailing Yan
- Department of Emergency, The First Hospital of Jilin University, 71Xinmin Street, Changchun, 130021, Jilin Province, People's Republic of China.
| |
Collapse
|
15
|
Walborn A, Rondina M, Mosier M, Fareed J, Hoppensteadt D. Endothelial Dysfunction Is Associated with Mortality and Severity of Coagulopathy in Patients with Sepsis and Disseminated Intravascular Coagulation. Clin Appl Thromb Hemost 2019; 25:1076029619852163. [PMID: 31140293 PMCID: PMC6714948 DOI: 10.1177/1076029619852163] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 04/25/2019] [Accepted: 04/26/2019] [Indexed: 01/08/2023] Open
Abstract
The role of the endothelium in sepsis-associated disseminated intravascular coagulation (DIC) is multifaceted and may contribute substantially to disease severity and outcome. The purpose of this study was to quantify measures of endothelial function, including markers of activation (endocan, Angiopoietin-2 [Ang-2], and von Willebrand Factor), endogenous anticoagulants (tissue factor pathway inhibitor and protein C), and damage-associated factors (High Mobility Group Box 1 [HMGB-1]) in the plasma of patients with sepsis and DIC, and to determine the relationship of these factors with severity of illness and outcome. Plasma samples were collected from 103 adult patients with sepsis within 48 hours of intensive care unit admission. Biomarker levels were measured using commercially available, standardized methods. Disseminated intravascular coagulation was diagnosed according to the International Society of Thrombosis and Hemostasis scoring algorithm. Twenty-eight-day mortality was used as the primary end point. In this study, endothelial damage and dysfunction were associated with the severity of coagulopathy and mortality in DIC patients. Loss of the endogenous anticoagulant protein C and elevation in the vascular regulator Ang-2 were associated with the development of overt DIC. In addition to Ang-2 and protein C, endocan, a biomarker of endothelial activation, and HMGB-1, a mediator of endothelial damage and activation, were significantly associated with mortality. This underscores the contribution of the endothelium to the pathogenesis of sepsis-associated DIC.
Collapse
Affiliation(s)
- Amanda Walborn
- Departments of Pathology and Pharmacology, Loyola University Medical Center, Maywood, IL, USA
| | - Matthew Rondina
- Department of Internal Medicine and the Molecular Medicine Program, University of Utah and the GRECC, George E. Wahlen VAMC, Salt Lake City, UT, USA
| | - Michael Mosier
- General Surgery, The Oregon Clinic, Surgical and Burn Specialists at Emanuel, Portland, OR, USA
| | - Jawed Fareed
- Departments of Pathology and Pharmacology, Loyola University Medical Center, Maywood, IL, USA
| | - Debra Hoppensteadt
- Departments of Pathology and Pharmacology, Loyola University Medical Center, Maywood, IL, USA
| |
Collapse
|
16
|
Onohara T, Sakamoto Y, Inoue S. Plasma Adsorption Membranes Are Able to Efficiently Remove High Mobility Group Box-1 (HMGB-1). J NIPPON MED SCH 2018; 85:150-156. [PMID: 30135341 DOI: 10.1272/jnms.jnms.2018_85-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND High Mobility Group Box 1 (HMGB-1) is a 30 kDa protein that is a lethal mediator in sepsis and is a recognized therapeutic target. However, no consensus has been reached for acute blood purification therapy as a treatment for sepsis targeting HMGB-1. Previous studies demonstrated that a high anti-HMGB-1 antibody titer and the suppression of HMGB-1 activity improved survival rate in animal sepsis models. The aim of this study was to evaluate whether plasma adsorption therapy is able to decrease the level of HMGB-1, representing a new potential treatment strategy against sepsis. METHODS Plasma adsorption therapy has been known as a treatment for various autoimmune diseases. Three different adsorbent columns, including TR-350 (IM-TR), PH-350 (IM-PH), and BRS-350 (BRS), were used in this study for comparison. We made a 1/350 scale of these three columns. Fetal bovine serum (FBS) contains a significant amount of HMGB-1. After priming with saline, FBS was passed through the columns and the adsorption rates of HMGB-1 at 25 minutes, 50 minutes, and 75 minutes were evaluated. The total adsorbed HMGB-1 level at 75 minutes was also calculated. RESULTS The highest adsorption rate and total adsorbed amount of HMGB-1 were observed in IM-TR, followed by BRS and IM-PH. IM-TR showed a decline in adsorption rate over time. BRS showed a steady adsorption rate at all time points. IM-TR removed HMGB-1 significantly more than IM-PH and BRS. CONCLUSIONS Our findings showed that plasma adsorption therapy efficiently adsorbed HMGB-1 and could be safely applied for the treatment of sepsis.
Collapse
Affiliation(s)
- Takayuki Onohara
- The Department of Emergency and Critical Care, Ureshino Medical Center
| | - Yuichiro Sakamoto
- The Department of Emergency and Critical Care Medicine, Saga University Hospital
| | - Satoshi Inoue
- The Division of Trauma Surgery and Surgical Critical Care, Saga University Hospital
| |
Collapse
|
17
|
Andersson U, Yang H, Harris H. High-mobility group box 1 protein (HMGB1) operates as an alarmin outside as well as inside cells. Semin Immunol 2018. [DOI: 10.1016/j.smim.2018.02.011] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
18
|
He M, Bianchi ME, Coleman TR, Tracey KJ, Al-Abed Y. Exploring the biological functional mechanism of the HMGB1/TLR4/MD-2 complex by surface plasmon resonance. Mol Med 2018; 24:21. [PMID: 30134799 PMCID: PMC6085627 DOI: 10.1186/s10020-018-0023-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 05/01/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND High Mobility Group Box 1 (HMGB1) was first identified as a nonhistone chromatin-binding protein that functions as a pro-inflammatory cytokine and a Damage-Associated Molecular Pattern molecule when released from necrotic cells or activated leukocytes. HMGB1 consists of two structurally similar HMG boxes that comprise the pro-inflammatory (B-box) and the anti-inflammatory (A-box) domains. Paradoxically, the A-box also contains the epitope for the well-characterized anti-HMGB1 monoclonal antibody "2G7", which also potently inhibits HMGB1-mediated inflammation in a wide variety of in vivo models. The molecular mechanisms through which the A-box domain inhibits the inflammatory activity of HMGB1 and 2G7 exerts anti-inflammatory activity after binding the A-box domain have been a mystery. Recently, we demonstrated that: 1) the TLR4/MD-2 receptor is required for HMGB1-mediated cytokine production and 2) the HMGB1-TLR4/MD-2 interaction is controlled by the redox state of HMGB1 isoforms. METHODS We investigated the interactions of HMGB1 isoforms (redox state) or HMGB1 fragments (A- and B-box) with TLR4/MD-2 complex using Surface Plasmon Resonance (SPR) studies. RESULTS Our results demonstrate that: 1) intact HMGB1 binds to TLR4 via the A-box domain with high affinity but an appreciable dissociation rate; 2) intact HMGB1 binds to MD-2 via the B-box domain with low affinity but a very slow dissociation rate; and 3) HMGB1 A-box domain alone binds to TLR4 more stably than the intact protein and thereby antagonizes HMGB1 by blocking HMGB1 from interacting with the TLR4/MD-2 complex. CONCLUSIONS These findings not only suggest a model whereby HMGB1 interacts with TLR4/MD-2 in a two-stage process but also explain how the A-box domain and 2G7 inhibit HMGB1.
Collapse
Affiliation(s)
- Mingzhu He
- Center for Molecular Innovation, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, New York, 11030, USA.
| | - Marco E Bianchi
- Chromatin Dynamics Unit, Division of Genetics and Cell Biology, San Raffaele University and San Raffaele Scientific Institute IRCCS, Via Olgettina 58, 20132, Milan, Italy
| | - Tom R Coleman
- Center for Molecular Innovation, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, New York, 11030, USA
| | - Kevin J Tracey
- Center for Biomedical Science, and Center for Bioelectronic Medicine, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, New York, 11030, USA
| | - Yousef Al-Abed
- Center for Molecular Innovation, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, New York, 11030, USA.
| |
Collapse
|
19
|
Amemiya R, Shinoda M, Yamada M, Ueno Y, Shimada K, Fujieda H, Yagi H, Mizota T, Nishiyama R, Oshima G, Yamada S, Matsubara K, Abe Y, Hibi T, Kitago M, Obara H, Itano O, Kitagawa Y. Hemoadsorption of high-mobility-group box 1 using a porous polymethylmethacrylate fiber in a swine acute liver failure model. Int J Artif Organs 2018. [PMID: 29528759 DOI: 10.1177/0391398817752296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND High-mobility-group box chromosomal protein 1 has been identified as an important mediator of various kinds of acute and chronic inflammation. In this study, we aimed to develop a column that effectively adsorbs high-mobility-group box chromosomal protein 1 by altering the pore size of the fiber. MATERIALS AND METHODS First, we produced three types of porous polymethylmethacrylate fiber by altering the concentration of polymethylmethacrylate dissolved in dimethylsulfoxide. We then selected a fiber based on the results of an in vitro incubation test of high-mobility-group box chromosomal protein 1 adsorption. Using the selected fiber, we constructed a new column and tested its high-mobility-group box chromosomal protein 1 adsorption capacity during 4-h extracorporeal hemoperfusion in a swine acute liver failure model. RESULTS Electron microscope observation showed that the three types of fibers had different pore sizes on the surface and in cross section, which were dependent on the concentration of polymethylmethacrylate. In the in vitro incubation test, fiber with moderate-sized pores demonstrated the highest adsorption capacity. In the in vivo hemoperfusion study, the ratio of the high-mobility-group box chromosomal protein 1 concentration at the outlet versus the inlet of the column was significantly lower with the new column than with the control column during 4-h extracorporeal hemoperfusion. The normalized plasma level of high-mobility-group box chromosomal protein 1 at 12 h after the completion of hemoperfusion was significantly lower with the new column than with the control column. CONCLUSION The newly developed polymethylmethacrylate column adsorbs high-mobility-group box chromosomal protein 1 during hemoperfusion in swine ALF model.
Collapse
Affiliation(s)
- Ryusuke Amemiya
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masahiro Shinoda
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masayuki Yamada
- 2 Medical Devices & Materials Research Unit, Advanced Materials Research Laboratories, Toray Industries, Inc., Shiga, Japan
| | - Yoshiyuki Ueno
- 2 Medical Devices & Materials Research Unit, Advanced Materials Research Laboratories, Toray Industries, Inc., Shiga, Japan
| | - Kaoru Shimada
- 2 Medical Devices & Materials Research Unit, Advanced Materials Research Laboratories, Toray Industries, Inc., Shiga, Japan
| | - Hiroaki Fujieda
- 2 Medical Devices & Materials Research Unit, Advanced Materials Research Laboratories, Toray Industries, Inc., Shiga, Japan
| | - Hiroshi Yagi
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Takamasa Mizota
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Ryo Nishiyama
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Go Oshima
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Shingo Yamada
- 3 Central Institute, Shino-Test Corporation, Kanagawa, Japan
| | - Kentaro Matsubara
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yuta Abe
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Taizo Hibi
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Minoru Kitago
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hideaki Obara
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Osamu Itano
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kitagawa
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
20
|
Zhou Y, Li P, Goodwin AJ, Cook JA, Halushka PV, Chang E, Fan H. Exosomes from Endothelial Progenitor Cells Improve the Outcome of a Murine Model of Sepsis. Mol Ther 2018; 26:1375-1384. [PMID: 29599080 DOI: 10.1016/j.ymthe.2018.02.020] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/15/2018] [Accepted: 02/22/2018] [Indexed: 01/07/2023] Open
Abstract
Microvascular dysfunction leads to multi-organ failure and mortality in sepsis. Our previous studies demonstrated that administration of exogenous endothelial progenitor cells (EPCs) confers protection in sepsis as evidenced by reduced vascular leakage, improved organ function, and increased survival. We hypothesize that EPCs protect the microvasculature through the exosomes-mediated transfer of microRNAs (miRNAs). Mice were rendered septic by cecal ligation and puncture (CLP), and EPC exosomes were administered intravenously at 4 hr after CLP. EPC exosomes treatment improved survival, suppressing lung and renal vascular leakage, and reducing liver and kidney dysfunction in septic mice. EPC exosomes attenuated sepsis-induced increases in plasma levels of cytokines and chemokine. Moreover, we determined miRNA contents of EPC exosomes with next-generation sequencing and found abundant miR-126-3p and 5p. We demonstrated that exosomal miR-126-5p and 3p suppressed LPS-induced high mobility group box 1 (HMGB1) and vascular cell adhesion molecule 1 (VCAM1) levels, respectively, in human microvascular endothelial cells (HMVECs). Inhibition of microRNA-126-5p and 3p through transfection with microRNA-126-5p and 3p inhibitors abrogated the beneficial effect of EPC exosomes. The inhibition of exosomal microRNA-126 failed to block LPS-induced increase in HMGB1 and VCAM1 protein levels in HMVECs and negated the protective effect of exosomes on sepsis survival. Thus, EPC exosomes prevent microvascular dysfunction and improve sepsis outcomes potentially through the delivery of miR-126.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Biopharmaceutics, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Pengfei Li
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Andrew J Goodwin
- Department of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - James A Cook
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Perry V Halushka
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Pharmacology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Eugene Chang
- Department of Obstetrics-Gynecology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Hongkuan Fan
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
21
|
Andersson U, Yang H, Harris H. Extracellular HMGB1 as a therapeutic target in inflammatory diseases. Expert Opin Ther Targets 2018; 22:263-277. [PMID: 29447008 DOI: 10.1080/14728222.2018.1439924] [Citation(s) in RCA: 223] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION High-mobility group box 1 (HMGB1) is a ubiquitous nuclear protein that promotes inflammation when released extracellularly after cellular activation, stress, damage or death. HMGB1 operates as one of the most intriguing molecules in inflammatory disorders via recently elucidated signal and molecular transport mechanisms. Treatments based on antagonists specifically targeting extracellular HMGB1 have generated encouraging results in a wide number of experimental models of infectious and sterile inflammation. Clinical studies are still to come. Areas covered: We here summarize recent advances regarding pathways for extracellular HMGB1 release, receptor usage, and functional consequences of post-translational modifications. The review also addresses results of preclinical HMGB1-targeted therapy studies in multiple inflammatory conditions and outlines the current status of emerging clinical HMGB1-specific antagonists. Expert opinion: Blocking excessive amounts of extracellular HMGB1, particularly the disulfide isoform, offers an attractive clinical opportunity to ameliorate systemic inflammatory diseases. Therapeutic interventions to regulate intracellular HMGB1 biology must still await a deeper understanding of intracellular HMGB1 functions. Future work is needed to create more robust assays to evaluate functional bioactivity of HMGB1 antagonists. Forthcoming clinical studies would also greatly benefit from a development of antibody-based assays to quantify HMGB1 redox isoforms, presently assessed by mass spectrometry methods.
Collapse
Affiliation(s)
- Ulf Andersson
- a Department of Women's and Children's Health, Center for Molecular Medicine (CMM) L8:04, Karolinska Institutet , Karolinska University Hospital , Stockholm , Sweden
| | - Huan Yang
- b Laboratory of Biomedical Science , The Feinstein Institute for Medical Research , Manhasset , NY , USA
| | - Helena Harris
- c Unit of Rheumatology, Department of Medicine, Center for Molecular Medicine (CMM) L, 8:04, Karolinska Institutet , Karolinska University Hospital , Stockholm , Sweden
| |
Collapse
|
22
|
Glycyrrhizin Protects Rats from Sepsis by Blocking HMGB1 Signaling. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9719647. [PMID: 28484719 PMCID: PMC5412259 DOI: 10.1155/2017/9719647] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 02/28/2017] [Accepted: 03/12/2017] [Indexed: 12/28/2022]
Abstract
Background. HMGB1 acts as an important inflammatory mediator and is a potential therapeutic target for sepsis. Glycyrrhizin (GL), a natural triterpene glycoside derived from licorice, has been demonstrated to inhibit HMGB1 activity. The aim of this study is to explore how GL affects the HMGB1 signaling in sepsis. Methods. We used a CLP model of sepsis and in vitro LPS or HMGB1-treated NR8383 cells to examine the effects of GL on expression of HMGB1 and proinflammatory cytokines. Furthermore, we explored the effect of GL on interactions between HMGB1 and RAGE or TLR4 and the activations of NF-κB and MAPKs. Results. GL significantly decreased mortality and reduced serum levels of HMGB1 in vivo. GL also attenuated the release and expression of HMGB1 and proinflammatory cytokines. Direct stimulation by HMGB1 elevated the release of proinflammatory cytokines faster than LPS did and it was also inhibited by GL. Furthermore, GL blocked the interaction of HMGB1 with RAGE and TLR4 and suppressed the downstream MAPKs/NF-κB signaling pathway. Conclusion. GL may protect rats against sepsis by blocking the interaction of HMGB1 with cell surface receptors and HMGB1-mediated inflammatory responses.
Collapse
|
23
|
Paeonol Inhibits Lipopolysaccharide-Induced HMGB1 Translocation from the Nucleus to the Cytoplasm in RAW264.7 Cells. Inflammation 2017; 39:1177-87. [PMID: 27106477 DOI: 10.1007/s10753-016-0353-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Transport of high-mobility group box 1 (HMGB1), a highly conserved non-histone DNA-binding protein, from the nucleus to the cytoplasm is induced by lipopolysaccharide (LPS). Secretion of HMGB1 appears to be a key lethal factor in sepsis, so it is considered to be a therapeutic target. Previous studies have suggested that paeonol (2'-hydroxy-4'-methoxyacetophenone), an active compound of Paeonia lactiflora Pallas, exerts anti-inflammatory effects. However, the effect of paeonol on HMGB1 is unknown. Here, we investigated the effect of paeonol on the expression, location, and secretion of HMGB1 in LPS-induced murine RAW264.7 cells. ELISA revealed HMGB1 supernatant concentrations of 615 ± 30 ng/mL in the LPS group and 600 ± 45, 560 ± 42, and 452 ± 38 ng/mL in cells treated with 0.2, 0.6, or 1 mM paeonol, respectively, suggesting that paeonol inhibits HMGB1 secretion induced by LPS. Immunohistochemistry and Western blotting revealed that paeonol decreased cytoplasmic HMGB1 and increased nuclear HMGB1. Chromatin immunoprecipitation microarrays suggested that HMGB1 relocation to the nucleus induced by paeonol might depress the action of Janus kinase/signal transducers and activators of transcription, chemokine, and mitogen-activated protein kinase pro-inflammatory signaling pathways. Paeonol was also found to inhibit tumor necrosis factor-α promoter activity in a dose-dependent manner. These results indicate that paeonol has the potential to be developed as a novel HMGB1-targeting therapeutic drug for the treatment of inflammatory diseases.
Collapse
|
24
|
Venereau E, De Leo F, Mezzapelle R, Careccia G, Musco G, Bianchi ME. HMGB1 as biomarker and drug target. Pharmacol Res 2016; 111:534-544. [PMID: 27378565 DOI: 10.1016/j.phrs.2016.06.031] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/29/2016] [Accepted: 06/30/2016] [Indexed: 12/27/2022]
Abstract
High Mobility Group Box 1 protein was discovered as a nuclear protein, but it has a "second life" outside the cell where it acts as a damage-associated molecular pattern. HMGB1 is passively released or actively secreted in a number of diseases, including trauma, chronic inflammatory disorders, autoimmune diseases and cancer. Extracellular HMGB1 triggers and sustains the inflammatory response by inducing cytokine release and by recruiting leucocytes. These characteristics make extracellular HMGB1 a key molecular target in multiple diseases. A number of strategies have been used to prevent HMGB1 release or to inhibit its activities. Current pharmacological strategies include antibodies, peptides, decoy receptors and small molecules. Noteworthy, salicylic acid, a metabolite of aspirin, has been recently found to inhibit HMGB1. HMGB1 undergoes extensive post-translational modifications, in particular acetylation and oxidation, which modulate its functions. Notably, high levels of serum HMGB1, in particular of the hyper-acetylated and disulfide isoforms, are sensitive disease biomarkers and are associated with different disease stages. In the future, the development of isoform-specific HMGB1 inhibitors may potentiate and fine-tune the pharmacological control of inflammation. We review here the current therapeutic strategies targeting HMGB1, in particular the emerging and relatively unexplored small molecules-based approach.
Collapse
Affiliation(s)
- Emilie Venereau
- Chromatin Dynamics Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; HMGBiotech s.r.l., Milan, Italy
| | - Federica De Leo
- Biomolecular NMR Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rosanna Mezzapelle
- Chromatin Dynamics Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giorgia Careccia
- Chromatin Dynamics Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; University of Milano Bicocca, Milan, Italy
| | - Giovanna Musco
- Biomolecular NMR Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco E Bianchi
- Chromatin Dynamics Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; San Raffaele University, Milan, Italy.
| |
Collapse
|
25
|
Gao DD, Fu J, Qin B, Huang WX, Yang C, Jia B. Recombinant adenovirus containing hyper-interleukin-6 and hepatocyte growth factor ameliorates acute-on-chronic liver failure in rats. World J Gastroenterol 2016; 22:4136-4148. [PMID: 27122664 PMCID: PMC4837431 DOI: 10.3748/wjg.v22.i16.4136] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/01/2016] [Accepted: 03/02/2016] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the protective efficacy of recombinant adenovirus containing hyper-interleukin-6 (Hyper-IL-6, HIL-6) and hepatocyte growth factor (HGF) (Ad-HGF-HIL-6) compared to that of recombinant adenovirus containing either HIL-6 or HGF (Ad-HIL-6 or Ad-HGF) in rats with acute-on-chronic liver failure (ACLF).
METHODS: The recombinant adenoviruses containing HIL-6 and/or HGF were constructed. We established an ACLF model, and rats were randomly assigned to control, model, Ad-GFP, Ad-HIL-6, Ad-HGF or Ad-HGF-HIL-6 group. We collected serum and liver tissue samples to test pathological changes, biochemical indexes and molecular biological indexes.
RESULTS: Attenuated alanine aminotransferase, prothrombin time, high-mobility group box 1 (HMGB1), endotoxin, tumour necrosis factor (TNF)-α and interferon-γ were observed in the Ad-HGF-, Ad-HIL-6- and Ad-HGF-HIL-6-treated rats with ACLF. Likewise, reduced hepatic damage and apoptotic activity, as well as reduced HMGB1 and Bax proteins, but raised expression of Ki67 and Bcl-2 proteins and Bcl-2/Bax ratio were also observed in the Ad-HGF-, Ad-HIL-6- and Ad-HGF-HIL-6-treated rats with ACLF. More significant changes were observed in the Ad-HGF-HIL-6 treatment group without obvious side effects. Furthermore, caspase-3 at the protein level decreased in the Ad-HIL-6 and Ad-HGF-HIL-6 treatment groups, more predominantly in the latter group.
CONCLUSION: This study identifies that the protective efficacy of Ad-HGF-HIL-6 is more potent than that of Ad-HGF or Ad-HIL-6 in ACLF rats, with no significant side effects.
Collapse
|
26
|
Li K, Yang J, Han X. Ketamine attenuates sepsis-induced acute lung injury via regulation of HMGB1-RAGE pathways. Int Immunopharmacol 2016; 34:114-128. [PMID: 26945830 DOI: 10.1016/j.intimp.2016.01.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 01/05/2016] [Accepted: 01/21/2016] [Indexed: 10/22/2022]
Abstract
High mobility group box protein 1 (HMGB1) and receptor for the advanced glycation end product (RAGE) play important roles in the development of sepsis-induced acute lung injury (ALI). Ketamine is considered to confer protective effects on ALI during sepsis. In this study, we investigated the effects of ketamine on HMGB1-RAGE activation in a rat model of sepsis-induced ALI. ALI was induced in wild type (WT) and RAGE deficient (RAGE(-/-)) rats by cecal ligation and puncture (CLP) or HMGB1 to mimic sepsis-induced ALI. Rats were randomly divided to six groups: sham-operation+normal saline (NS, 10 mL/kg), sham-operation+ketamine (10 mg/kg), CLP/HMGB1+NS (10 mL/kg), CLP/HMGB1+ketamine (5 mg/kg), CLP/HMGB1+ketamine (7.5 mg/kg), and CLP/HMGB1+ketamine (10 mg/kg) groups. NS and ketamine were administered at 3 and 12 h after CLP/HMGB1 via intraperitoneal injection. Pathological changes of lung, inflammatory cell counts, expression of HMGB1 and RAGE, and concentrations of various inflammatory mediators in bronchoalveolar lavage fluids (BALF) and lung tissue were then assessed. Nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinases (MAPK) signaling pathways in the lung were also evaluated. CLP/HMGB1 increased the wet to dry weight ratio and myeloperoxidase activity in lung, the number of total cells, neutrophils, and macrophages in the BALF, and inflammatory mediators in the BALF and lung tissues. Moreover, expression of HMGB1 and RAGE in lung tissues was increased after CLP. Ketamine inhibited all the above effects. It also inhibited the activation of IκB-α, NF-κB p65, and MAPK. Ketamine protects rats against HMGB1-RAGE activation in a rat model of sepsis-induced ALI. These effects may partially result from reductions in NF-κB and MAPK.
Collapse
Affiliation(s)
- Kehan Li
- Department of Anesthesiology, The First Affiliated Hospital of Henan Science and Technology University, Luoyang, Henan, China.
| | - Jianxue Yang
- Department of Neurology, The First Affiliated Hospital of Henan Science and Technology University, Luoyang, Henan, China
| | - Xuechang Han
- Department of Anesthesiology, The First Affiliated Hospital of Henan Science and Technology University, Luoyang, Henan, China
| |
Collapse
|
27
|
Park HS, Kim EN, Kim MY, Lim JH, Kim HW, Park CW, Yang CW, Jin DC, Choi BS. The protective effect of neutralizing high-mobility group box1 against chronic cyclosporine nephrotoxicity in mice. Transpl Immunol 2015; 34:42-9. [PMID: 26603313 DOI: 10.1016/j.trim.2015.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 11/11/2015] [Accepted: 11/17/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND High-mobility group box1 (HMGB1) is known to be involved in innate immune response through interaction with receptor for advanced glycation end products (RAGE) and toll-like receptors (TLRs), besides its proper role within the nucleus. Immunological pathways, including TLR signaling, are also involved in chronic cyclosporine (CsA) nephrotoxicity. This study was designed to determine whether neutralizing HMGB1 prevents chronic CsA nephrotoxicity. METHODS Chronic CsA nephrotoxicity was induced by CsA subcutaneous injection daily for 4weeks under salt-depletion in mice. Anti-HMGB1 neutralizing antibody for HMGB1 blockade (600mcg/mouse) was administered weekly to mice in the anti-HMGB1 treatment group. The effects of HMGB1 neutralization were evaluated in terms of renal function as well as histological and immunopathological examination. RESULTS Anti-HMGB1 administration prevented the increases in serum creatinine and 24h albuminuria and the decrease in creatinine clearance associated with CsA treatment. Increased tubulointerstitial fibrosis and transforming growth factor (TGF)-β immunohistochemical staining associated with CsA treatment were also prevented by anti-HMGB1 administration. Anti-HMGB1 administration prevented the activation of the TLR4 signaling pathway, which resulted in the reduction of nuclear factor kappa B (NF-κB) expression. In cultured tubular cells, anti-HMGB1 pretreatment also prevented the increases in fibronectin and collagen IV levels associated with CsA treatment. CONCLUSIONS Neutralizing HMGB1 with an anti-HMGB1 antibody ameliorated chronic CsA nephrotoxicity via inhibition of the TLR4 signaling pathway. Our study suggests that HMGB1 blockade can be beneficial for increasing allograft survival in renal transplant recipients by protecting against calcineurin inhibitor-induced nephrotoxicity.
Collapse
Affiliation(s)
- Hoon Suk Park
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Nim Kim
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Min Young Kim
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Hee Lim
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyung Wook Kim
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Cheol Whee Park
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chul Woo Yang
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dong Chan Jin
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Bum Soon Choi
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
28
|
CXCR2 expression and postoperative complications affect long-term survival in patients with esophageal cancer. World J Surg Oncol 2015; 13:232. [PMID: 26231560 PMCID: PMC4522106 DOI: 10.1186/s12957-015-0658-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 07/20/2015] [Indexed: 12/25/2022] Open
Abstract
Background Esophagectomy is one of the most invasive surgical treatments for digestive tract cancer, and the blood levels of inflammatory cytokines such as interleukin-1, interleukin-6, and interleukin-8 are increased for several hours after surgery or in patients experiencing postoperative complications. CXCR2, an interleukin-8 receptor, is reportedly expressed in several carcinomas, and interleukin-8 signaling promotes cancer cell proliferation. The impact of postoperative complications following esophagectomy on long-term survival is controversial. In this study, we demonstrate the significance of CXCR2 expression and validate the effects of CXCR2 expression and postoperative complications on long-term prognosis of esophageal squamous cell carcinoma using resected specimens. Methods Eighty-two specimens were sectioned from archived, paraffin-embedded tumor tissues obtained from patients with esophageal squamous cell carcinoma who underwent esophagectomy and extended lymphadenectomy for complete resection of cancer in our institute between 1997 and 2002. Immunohistochemistry was performed using a polyclonal antibody to CXCR2, and the correlation of stainability with clinicopathological factors and long-term survival was examined. Results CXCR2 was expressed in 33 of 82 (40.2 %) specimens. In the CXCR2-positive group, the recurrence-free survival and overall survival rates of patients who developed postoperative complications were both significantly lower than those for patients who did not develop any complications. In contrast, in the CXCR2-negative group, there was no significant difference in long-term prognosis between patients with and without complications. CXCR2 positivity combined with postoperative complications was an independent risk factor for subsequent tumor recurrence, showing the highest hazard ratio. Conclusions Our results suggest that the patients with CXCR2-positive esophageal cancer who develop postoperative complications have a poor prognosis and should be carefully followed. Trial registration This study was approved by Keio University School of Medicine Ethics Committee with a trial registration number of 2011-241.
Collapse
|
29
|
Sodhi CP, Jia H, Yamaguchi Y, Lu P, Good M, Egan C, Ozolek J, Zhu X, Billiar TR, Hackam DJ. Intestinal Epithelial TLR-4 Activation Is Required for the Development of Acute Lung Injury after Trauma/Hemorrhagic Shock via the Release of HMGB1 from the Gut. THE JOURNAL OF IMMUNOLOGY 2015; 194:4931-9. [PMID: 25862813 DOI: 10.4049/jimmunol.1402490] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 03/13/2015] [Indexed: 12/11/2022]
Abstract
The mechanisms that lead to the development of remote lung injury after trauma remain unknown, although a central role for the gut in the induction of lung injury has been postulated. We hypothesized that the development of remote lung injury after trauma/hemorrhagic shock requires activation of TLR4 in the intestinal epithelium, and we sought to determine the mechanisms involved. We show that trauma/hemorrhagic shock caused lung injury in wild-type mice, but not in mice that lack TLR4 in the intestinal epithelium, confirming the importance of intestinal TLR4 activation in the process. Activation of intestinal TLR4 after trauma led to increased endoplasmic reticulum (ER) stress, enterocyte apoptosis, and the release of circulating HMGB1, whereas inhibition of ER stress attenuated apoptosis, reduced circulating HMGB1, and decreased lung injury severity. Neutralization of circulating HMGB1 led to reduced severity of lung injury after trauma, and mice that lack HMGB1 in the intestinal epithelium were protected from the development of lung injury, confirming the importance of the intestine as the source of HMGB1, whose release of HMGB1 induced a rapid protein kinase C ζ-mediated internalization of surface tight junctions in the pulmonary epithelium. Strikingly, the use of a novel small-molecule TLR4 inhibitor reduced intestinal ER stress, decreased circulating HMGB1, and preserved lung architecture after trauma. Thus, intestinal epithelial TLR4 activation leads to HMGB1 release from the gut and the development of lung injury, whereas strategies that block upstream TLR4 signaling may offer pulmonary protective strategies after trauma.
Collapse
Affiliation(s)
- Chhinder P Sodhi
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins University, Baltimore, MD 21287
| | - Hongpeng Jia
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins University, Baltimore, MD 21287
| | - Yukihiro Yamaguchi
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins University, Baltimore, MD 21287
| | - Peng Lu
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins University, Baltimore, MD 21287
| | - Misty Good
- Division of Newborn Medicine, Children's Hospital of Pittsburgh, Pittsburgh, PA 15213; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Charlotte Egan
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins University, Baltimore, MD 21287
| | - John Ozolek
- Division of Pathology, Children's Hospital of Pittsburgh, Pittsburgh, PA 15224; Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Xiaorong Zhu
- Department of Medicine, Gastroenterology, Hepatology and Nutrition, University of Chicago, Chicago, IL 60637; and
| | - Timothy R Billiar
- Division of Trauma and General Surgery, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213
| | - David J Hackam
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins University, Baltimore, MD 21287;
| |
Collapse
|
30
|
Tanaka M, Shinoda M, Takayanagi A, Oshima G, Nishiyama R, Fukuda K, Yagi H, Hayashida T, Masugi Y, Suda K, Yamada S, Miyasho T, Hibi T, Abe Y, Kitago M, Obara H, Itano O, Takeuchi H, Sakamoto M, Tanabe M, Maruyama I, Kitagawa Y. Gene transfer of high-mobility group box 1 box-A domain in a rat acute liver failure model. J Surg Res 2015; 194:571-580. [PMID: 25498512 DOI: 10.1016/j.jss.2014.11.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Revised: 10/13/2014] [Accepted: 11/13/2014] [Indexed: 01/18/2023]
Abstract
BACKGROUND High-mobility group box 1 (HMGB1) has recently been identified as an important mediator of various kinds of acute and chronic inflammation. The protein encoded by the box-A domain of the HMGB1 gene is known to act as a competitive inhibitor of HMGB1. In this study, we investigated whether box-A gene transfer results in box-A protein production in rats and assessed therapeutic efficacy in vivo using an acute liver failure (ALF) model. MATERIALS AND METHODS Three types of adenovirus vectors were constructed-a wild type and two mutants-and a mutant vector was then selected based on the secretion from HeLa cells. The secreted protein was subjected to a tumor necrosis factor (TNF) production inhibition test in vitro. The vector was injected via the portal vein in healthy Wistar rats to confirm box-A protein production in the liver. The vector was then injected via the portal vein in rats with ALF. RESULTS Western blot analysis showed enhanced expression of box-A protein in HeLa cells transfected with one of the mutant vectors. The culture supernatant from HeLa cells transfected with the vector inhibited TNF-α production from macrophages. Expression of box-A protein was confirmed in the transfected liver at 72 h after transfection. Transfected rats showed decreased hepatic enzymes, plasma HMGB1, and hepatic TNF-α messenger RNA levels, and histologic findings and survival were significantly improved. CONCLUSIONS HMGB1 box-A gene transfer results in box-A protein production in the liver and appears to have a beneficial effect on ALF in rats.
Collapse
Affiliation(s)
- Masayuki Tanaka
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Masahiro Shinoda
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan.
| | - Atsushi Takayanagi
- Department of Molecular Biology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Go Oshima
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Ryo Nishiyama
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Kazumasa Fukuda
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Hiroshi Yagi
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Tetsu Hayashida
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Yohei Masugi
- Department of Pathology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Koichi Suda
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Shingo Yamada
- Central Institute, Shino-Test Corporation, Sagamihara, Kanagawa, Japan
| | - Taku Miyasho
- Department of Veterinary Science, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - Taizo Hibi
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Yuta Abe
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Minoru Kitago
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Hideaki Obara
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Osamu Itano
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Hiroya Takeuchi
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Michiie Sakamoto
- Department of Pathology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Minoru Tanabe
- Department of Surgery, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan
| | - Ikuro Maruyama
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medicine and Dental Sciences, Kagoshima, Kagoshima, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| |
Collapse
|
31
|
Wang F, Meng Y, Zhang Y, Zhao G, Zheng X, Xiao Q, Yu Y. RETRACTED: Ketamine reduces lipopolysaccharide-induced high-mobility group box-1 through heme oxygenase-1 and nuclear factor erythroid 2-related factor 2/ p38 mitogen-activated protein kinase. J Surg Res 2015; 194:599-613. [DOI: 10.1016/j.jss.2014.11.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 11/02/2014] [Accepted: 11/21/2014] [Indexed: 01/07/2023]
|
32
|
Foley NM, Wang J, Redmond HP, Wang JH. Current knowledge and future directions of TLR and NOD signaling in sepsis. Mil Med Res 2015; 2:1. [PMID: 25722880 PMCID: PMC4340879 DOI: 10.1186/s40779-014-0029-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 12/17/2014] [Indexed: 01/13/2023] Open
Abstract
The incidence of sepsis is increasing over time, along with an increased risk of dying from the condition. Sepsis care costs billions annually in the United States. Death from sepsis is understood to be a complex process, driven by a lack of normal immune homeostatic functions and excessive production of proinflammatory cytokines, which leads to multi-organ failure. The Toll-like receptor (TLR) family, one of whose members was initially discovered in Drosophila, performs an important role in the recognition of microbial pathogens. These pattern recognition receptors (PRRs), upon sensing invading microorganisms, activate intracellular signal transduction pathways. NOD signaling is also involved in the recognition of bacteria and acts synergistically with the TLR family in initiating an efficient immune response for the eradication of invading microbial pathogens. TLRs and NOD1/NOD2 respond to different pathogen-associated molecular patterns (PAMPs). Modulation of both TLR and NOD signaling is an area of research that has prompted much excitement and debate as a therapeutic strategy in the management of sepsis. Molecules targeting TLR and NOD signaling pathways exist but regrettably thus far none have proven efficacy from clinical trials.
Collapse
Affiliation(s)
- Niamh M Foley
- Department of Academic Surgery, University College Cork, Cork University Hospital, Cork, Ireland
| | - Jian Wang
- Department of Pediatric Surgery, Affiliated Children's Hospital, Soochow University, Suzhou, 215003 China
| | - H Paul Redmond
- Department of Academic Surgery, University College Cork, Cork University Hospital, Cork, Ireland
| | - Jiang Huai Wang
- Department of Academic Surgery, University College Cork, Cork University Hospital, Cork, Ireland
| |
Collapse
|
33
|
Bao GQ, He L, Lee D, D'Angelo J, Wang HC. An ongoing search for potential targets and therapies for lethal sepsis. Mil Med Res 2015; 2:20. [PMID: 26257917 PMCID: PMC4529709 DOI: 10.1186/s40779-015-0047-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 07/20/2015] [Indexed: 01/15/2023] Open
Abstract
Sepsis, which refers to a systemic inflammatory response syndrome resulting from a microbial infection, represents the leading cause of death in intensive care units. The pathogenesis of sepsis remains poorly understood although it is attributable to dysregulated immune responses orchestrated by innate immune cells that are sequentially released early (e.g., tumor necrosis factor(TNF), interleukin-1(IL-1), and interferon-γ(IFN-γ)) and late (e.g., high mobility group box 1(HMGB1)) pro-inflammatory mediators. As a ubiquitous nuclear protein, HMGB1 can be passively released from pathologically damaged cells, thereby converging infection and injury on commonly dysregulated inflammatory responses. We review evidence that supports extracellular HMGB1 as a late mediator of inflammatory diseases and discuss the potential of several Chinese herbal components as HMGB1-targeting therapies. We propose that it is important to develop strategies for specifically attenuating injury-elicited inflammatory responses without compromising the infection-mediated innate immunity for the clinical management of sepsis and other inflammatory diseases.
Collapse
Affiliation(s)
- Guo-Qiang Bao
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, NY 11030 USA.,The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030 USA.,Department of General Surgery, Tangdu Hospital, The 4th Military Medical University, Xi'an, Shaanxi 710032 China
| | - Li He
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - David Lee
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, NY 11030 USA
| | - John D'Angelo
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, NY 11030 USA
| | - Hai-Chao Wang
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, NY 11030 USA.,The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030 USA
| |
Collapse
|
34
|
Charoensup J, Sermswan RW, Paeyao A, Promakhejohn S, Punasee S, Chularari C, Krabkraikaew S, Lertanekawattana S, Wongratanacheewin S. High HMGB1 level is associated with poor outcome of septicemic melioidosis. Int J Infect Dis 2014; 28:111-6. [PMID: 25263503 DOI: 10.1016/j.ijid.2014.07.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Revised: 07/24/2014] [Accepted: 07/30/2014] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES A high level of HMGB1 (high-mobility group box 1) - a late onset inflammatory mediator - is a marker of lethal sepsis in several infectious diseases. The level of HMGB1 in the plasma of Burkholderia pseudomallei-infected patients was investigated together with the severity of the disease. The neutralization of HMGB1 to improve survival was also tested in a mouse model. METHODS HMGB1 levels in the plasma of 77 septic patients, 40 with B. pseudomallei infection and 37 with other bacterial infections, were determined by ELISA. Neutralizing antibody against purified recombinant HMGB1 was prepared in rabbits (rab-a-HMGB1) and its potential as an adjunct therapy was evaluated in B. pseudomallei-infected Balb/c mice treated with suboptimal doses of ceftazidime. RESULTS The plasma from B. pseudomallei-infected patients showed significantly higher HMGB1 levels than the plasma from other septic patients (median 11.1 ng/ml vs. 7.1 ng/ml). The HMGB1 level was significantly higher in patients with melioidosis who died than in those who survived (median 14.8 ng/ml vs. 9.2 ng/ml). Moreover, the HMGB1 level was significantly correlated with the clinical severity score (SOFA score). In the mouse study, although the rab-a-HMGB1 by itself could not improve the survival outcome of B. pseudomallei-infected mice, it could nevertheless enhance the effectiveness of suboptimal doses of ceftazidime in the treatment of these animals. CONCLUSION The level of HMGB1 in septic melioidosis patients can be used as a marker of late severe sepsis. Neutralizing antibody to HMGB1 may be used as an adjunct therapy to improve the outcome of melioidosis.
Collapse
Affiliation(s)
- Jaruek Charoensup
- Melioidosis Research Center, Khon Kaen University, Khon Kaen, Thailand
| | - Rasana W Sermswan
- Melioidosis Research Center, Khon Kaen University, Khon Kaen, Thailand; Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Arunee Paeyao
- Melioidosis Research Center, Khon Kaen University, Khon Kaen, Thailand
| | | | | | | | | | | | - Surasakdi Wongratanacheewin
- Melioidosis Research Center, Khon Kaen University, Khon Kaen, Thailand; Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.
| |
Collapse
|
35
|
Abstract
INTRODUCTION Sepsis refers to the host's deleterious and non-resolving systemic inflammatory response to microbial infections and represents the leading cause of death in the intensive care unit. The pathogenesis of sepsis is complex, but partly mediated by a newly identified alarmin molecule, the high mobility group box 1 (HMGB1). AREAS COVERED Here we review the evidence that support extracellular HMGB1 as a late mediator of experimental sepsis with a wider therapeutic window and discuss the therapeutic potential of HMGB1-neutralizing antibodies and small molecule inhibitors (herbal components) in experimental sepsis. EXPERT OPINION It will be important to evaluate the efficacy of HMGB1-targeting strategies for the clinical management of human sepsis in the future.
Collapse
Affiliation(s)
- Haichao Wang
- The Feinstein Institute for Medical Research and North Shore University Hospital, The Hofstra North Shore - LIJ School of Medicine, Laboratory of Emergency Medicine, North Shore-LIJ Health System , 350 Community Drive, Manhasset, NY 11030 , USA +1 516 562 2823 ; +1 516 562 1022 ;
| | | | | |
Collapse
|
36
|
Hirano Y, Takeuchi H, Suda K, Hagiwara T, Miyasho T, Kawamura Y, Yamada S, Oyama T, Takahashi T, Wada N, Saikawa Y, Ichihara A, Kitagawa Y. (Pro)renin receptor blocker improves survival of rats with sepsis. J Surg Res 2014; 186:269-277. [PMID: 24011922 DOI: 10.1016/j.jss.2013.08.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 07/22/2013] [Accepted: 08/05/2013] [Indexed: 01/27/2023]
Abstract
BACKGROUND The renin-angiotensin system (RAS) affects inflammatory responses during sepsis. Nonproteolytic activation of prorenin by the (pro)renin receptor has recently been shown to stimulate the tissue RAS. In the present study, the effect of (pro)renin receptor blocker (PRRB) pretreatment on sepsis in a rat cecal ligation and puncture (CLP) model was investigated. MATERIALS AND METHODS Male Sprague-Dawley rats underwent CLP and were randomly divided into two groups: PRRB-treated group and control peptide-treated group. Survival was analyzed for 7 d after CLP. The serum concentrations of cytokines and high-mobility group box chromosomal protein 1 (HMGB1) were measured at three time points (0, 3, and 6 h after CLP). Hematoxylin-eosin staining and immunohistochemical staining for nonproteolytically activated prorenin and HMGB1 were performed on the cecum to assess pathologic changes found 6 h after CLP. RESULTS Treatment with PRRB improved the survival rate of the post-CLP septic rats (P = 0.023). PRRB also significantly reduced serum tumor necrosis factor-α, interleukin-1β, and HMGB1 levels 6 h after CLP. In CLP rats that were treated with control peptide, the expression of activated prorenin was elevated in peritoneal foam cells. Moreover, expression of HMGB1 was increased in peritoneal inflammatory cells. In contrast, both were markedly suppressed in CLP rats that were treated with PRRB. CONCLUSIONS PRRB significantly improved the survival rate of rats with clinically relevant sepsis, possibly by attenuating a sepsis-induced systemic inflammatory response. We propose that overactivation of the RAS by activation of prorenin in foam cells may be a significant contributor to sepsis.
Collapse
Affiliation(s)
- Yuki Hirano
- Department of Surgery, School of Medicine, Keio University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kadowaki T, Morishita A, Niki T, Hara J, Sato M, Tani J, Miyoshi H, Yoneyama H, Masaki T, Hattori T, Matsukawa A, Hirashima M. Galectin-9 prolongs the survival of septic mice by expanding Tim-3-expressing natural killer T cells and PDCA-1+ CD11c+ macrophages. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2013; 17:R284. [PMID: 24321251 PMCID: PMC4056346 DOI: 10.1186/cc13147] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 11/26/2013] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Galectin-9 ameliorates various inflammatory conditions including autoimmune diseases by regulating T cell and macrophage/dendritic cell (DC) functions. However, the effect of galectin-9 on polymicrobial sepsis has not been assessed. METHODS We induced polymicrobial sepsis by cecal ligation and puncture (CLP) in mice. The survival rate was compared between galectin-9- and PBS-treated CLP mice. An ELISA was used to compare the levels of various cytokines in the plasma and culture supernatants. Fluorescence-activated cell sorting analysis was further performed to compare the frequencies of subpopulations of spleen cells. RESULTS Galectin-9 exhibited a protective effect in polymicrobial sepsis as demonstrated in galetin-9 transgenic mice and therapeutic galectin-9 administration. In contrast, such effect was not observed in nude mice, indicating the involvement of T cells in galectin-9-mediated survival prolongation. Galectin-9 decreased TNFα, IL-6, IL-10 and, high mobility group box 1 (HMGB1) and increased IL-15 and IL-17 plasma and spleen levels. Galectin-9 increased the frequencies of natural killer T (NKT) cells and PDCA-1+ CD11c+ macrophages (pDC-like macrophages) but did not change the frequency of CD4 or CD8 T cells, γδT cells or conventional DC. As expected, galectin-9 decreased the frequency of Tim-3+ CD4 T cells, most likely Th1 and Th17 cells. Intriguingly, many spleen NK1.1+ NKT cells and pDC-like macrophages expressed Tim-3. Galectin-9 increased the frequency of Tim-3-expressing NK1.1+ NKT cells and pDC-like macrophages. Galectin-9 further increased IL-17+ NK1.1+ NKT cells. CONCLUSION These data suggest that galectin-9 exerts therapeutic effects on polymicrobial sepsis, possibly by expanding NKT cells and pDC-like macrophages and by modulating the production of early and late proinflammatory cytokines.
Collapse
|
38
|
Li X, Wang LK, Wang LW, Han XQ, Yang F, Gong ZJ. Blockade of high-mobility group box-1 ameliorates acute on chronic liver failure in rats. Inflamm Res 2013; 62:703-9. [PMID: 23591781 DOI: 10.1007/s00011-013-0624-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 02/26/2013] [Accepted: 04/10/2013] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE High-mobility group box-1 (HMGB1) is identified as an extracellularly released mediator of inflammation. In this study, specific monoclonal anti-HMGB1 antibody was administered to rats with acute on chronic liver failure (ACLF) in order to evaluate the therapeutic efficacy of HMGB1 blockade. METHODS All animals were randomly divided into control group, model group and anti-HMGB1 antibody group. The changes in liver histology and apoptosis of liver tissue were detected by H&E staining and TUNEL assay, respectively. The serum levels of alanine aminotransferase (ALT), endotoxin, HMGB1, tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ) were examined. The hepatic levels of HMGB1, caspase3, Toll-like receptor 4 (TLR4) and p65 subunit of NF-κB (P65) were also determined. RESULTS Changes in liver pathology and liver cell apoptosis were greatly attenuated in the anti-HMGB1 antibody group compared with the model group. The serum levels of ALT, endotoxin, TNF-α, IFN-γ and HMGB1 were also decreased in the anti-HMGB1 antibody group. Furthermore, the hepatic levels of HMGB1, TLR4, caspase3 and P65 were also down-regulated by HMGB1 blockade. CONCLUSION Blockade of HMGB1 can confer a protective effect against ACLF in rats, even 24 h after induction of ACLF. The protective effect of HMGB1 blockade is associated with interactions of HMGB1 with the TLR4 signaling pathway and cytokine production.
Collapse
Affiliation(s)
- Xun Li
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | | | | | | | | | | |
Collapse
|
39
|
Mishra BB, Li Q, Steichen AL, Binstock BJ, Metzger DW, Teale JM, Sharma J. Galectin-3 functions as an alarmin: pathogenic role for sepsis development in murine respiratory tularemia. PLoS One 2013; 8:e59616. [PMID: 23527230 PMCID: PMC3603908 DOI: 10.1371/journal.pone.0059616] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 02/16/2013] [Indexed: 12/12/2022] Open
Abstract
Sepsis is a complex immune disorder with a mortality rate of 20–50% and currently has no therapeutic interventions. It is thus critical to identify and characterize molecules/factors responsible for its development. We have recently shown that pulmonary infection with Francisella results in sepsis development. As extensive cell death is a prominent feature of sepsis, we hypothesized that host endogenous molecules called alarmins released from dead or dying host cells cause a hyperinflammatory response culminating in sepsis development. In the current study we investigated the role of galectin-3, a mammalian β-galactoside binding lectin, as an alarmin in sepsis development during F. novicida infection. We observed an upregulated expression and extracellular release of galectin-3 in the lungs of mice undergoing lethal pulmonary infection with virulent strain of F. novicida but not in those infected with a non-lethal, attenuated strain of the bacteria. In comparison with their wild-type C57Bl/6 counterparts, F. novicida infected galectin-3 deficient (galectin-3−/−) mice demonstrated significantly reduced leukocyte infiltration, particularly neutrophils in their lungs. They also exhibited a marked decrease in inflammatory cytokines, vascular injury markers, and neutrophil-associated inflammatory mediators. Concomitantly, in-vitro pre-treatment of primary neutrophils and macrophages with recombinant galectin-3 augmented F. novicida-induced activation of these cells. Correlating with the reduced inflammatory response, F. novicida infected galectin-3−/− mice exhibited improved lung architecture with reduced cell death and improved survival over wild-type mice, despite similar bacterial burden. Collectively, these findings suggest that galectin-3 functions as an alarmin by augmenting the inflammatory response in sepsis development during pulmonary F. novicida infection.
Collapse
Affiliation(s)
- Bibhuti B. Mishra
- Department of Microbiology and Immunology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, United States of America
| | - Qun Li
- South Texas Center for Emerging Diseases and Department of Biology, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Anthony L. Steichen
- Department of Microbiology and Immunology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, United States of America
| | - Brandilyn J. Binstock
- Department of Microbiology and Immunology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, United States of America
| | | | - Judy M. Teale
- South Texas Center for Emerging Diseases and Department of Biology, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Jyotika Sharma
- Department of Microbiology and Immunology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, United States of America
- * E-mail:
| |
Collapse
|
40
|
Fujimura N, Obara H, Suda K, Takeuchi H, Miyasho T, Kawasako K, Du W, Yamada S, Ono S, Matsumoto K, Matsuda S, Yagi H, Kitago M, Shinoda M, Itano O, Tanabe M, Sakamoto M, Maruyama I, Kitagawa Y. Neutrophil elastase inhibitor improves survival rate after ischemia reperfusion injury caused by supravisceral aortic clamping in rats. J Surg Res 2013; 180:e31-e36. [PMID: 22595015 DOI: 10.1016/j.jss.2012.04.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 03/27/2012] [Accepted: 04/17/2012] [Indexed: 11/16/2022]
Abstract
BACKGROUND Sivelestat sodium hydrate is a specific neutrophil elastase inhibitor effective in acute lung injury (ALI) associated with systemic inflammatory response syndrome. Bowel ischemia reperfusion injury (IRI) induced by supravisceral aortic clamping is associated with an excessive systemic inflammatory response, resulting in remote organ damage, including ALI. In this study, we investigated whether sivelestat can attenuate neutrophil sequestration in the lung, alleviate ALI, and improve survival in a rat bowel IRI model. METHODS Adult male Sprague-Dawley rats underwent bowel IRI induced by supravisceral aortic clamping and were randomly assigned to receive sivelestat or saline (control) and monitored for survival. We randomly assigned other rats to undergo laparotomy alone (sham operation), IRI alone, or IRI and sivelestat treatment. We evaluated blood samples for organ function, cytokine levels, and neutrophil elastase activity after reperfusion. Organs were analyzed histologically. We also determined lung injury in another set of rats. RESULTS Bowel IRI induced a significant increase in serum variables indicative of organ function, cytokine concentrations, neutrophil elastase activity, and lung permeability and edema, which reflected the presence of both systemic inflammatory response syndrome and compensatory anti-inflammatory response syndrome. Treatment with sivelestat significantly improved survival rate, lung permeability and edema, and significantly decreased levels of creatinine, interleukin 6, interleukin 10, and neutrophil elastase activity. Histological studies showed that sivelestat-treated rats had less bowel IRI-induced damage to lung and liver tissue than controls. CONCLUSION In a rat model, administration of sivelestat attenuated the effects of bowel IRI induced by supravisceral aortic clamping, and improved the survival rate.
Collapse
Affiliation(s)
- Naoki Fujimura
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Human macrophage and dendritic cell-specific silencing of high-mobility group protein B1 ameliorates sepsis in a humanized mouse model. Proc Natl Acad Sci U S A 2012; 109:21052-7. [PMID: 23213216 DOI: 10.1073/pnas.1216195109] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Hypersecretion of cytokines by innate immune cells is thought to initiate multiple organ failure in murine models of sepsis. Whether human cytokine storm also plays a similar role is not clear. Here, we show that human hematopoietic cells are required to induce sepsis-induced mortality following cecal ligation and puncture (CLP) in the severely immunodeficient nonobese diabetic (NOD)/SCID/IL2Rγ(-/-) mice, and siRNA treatment to inhibit HMGB1 release by human macrophages and dendritic cells dramatically reduces sepsis-induced mortality. Following CLP, compared with immunocompetent WT mice, NOD/SCID/IL2Rγ(-/-) mice did not show high levels of serum HMGB1 or murine proinflammatory cytokines and were relatively resistant to sepsis-induced mortality. In contrast, NOD/SCID/IL2Rγ(-/-) mice transplanted with human hematopoietic stem cells [humanized bone marrow liver thymic mice (BLT) mice] showed high serum levels of HMGB1, as well as multiple human but not murine proinflammatory cytokines, and died uniformly, suggesting human cytokines are sufficient to induce organ failure in this model. Moreover, targeted delivery of HMGB1 siRNA to human macrophages and dendritic cells using a short acetylcholine receptor (AchR)-binding peptide [rabies virus glycoprotein (RVG)-9R] effectively suppressed secretion of HMGB1, reduced the human cytokine storm, human lymphocyte apoptosis, and rescued humanized mice from CLP-induced mortality. siRNA treatment was also effective when started after the appearance of sepsis symptoms. These results show that CLP in humanized mice provides a model to study human sepsis, HMGB1 siRNA might provide a treatment strategy for human sepsis, and RVG-9R provides a tool to deliver siRNA to human macrophages and dendritic cells that could potentially be used to suppress a variety of human inflammatory diseases.
Collapse
|
42
|
Kojima M, Tanabe M, Shinoda M, Yamada S, Miyasho T, Suda K, Hibi T, Obara H, Itano O, Kawachi S, Kitajima M, Maruyama I, Kitagawa Y. Role of high mobility group box chromosomal protein 1 in ischemia-reperfusion injury in the rat small intestine. J Surg Res 2012; 178:466-471. [PMID: 22494911 DOI: 10.1016/j.jss.2012.01.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 01/21/2012] [Accepted: 01/25/2012] [Indexed: 12/19/2022]
Abstract
BACKGROUND High mobility group box chromosomal protein 1 (HMGB1) has recently been shown to be an important late mediator of endotoxic shock and sepsis. The purpose of the present study was to investigate the role of HMGB1 in response to ischemia-reperfusion injury. METHODS Ischemia-reperfusion injury was induced in male Wistar rats by clamping the superior mesenteric artery for 60 min. Using this model, the serum concentrations and localization of HMGB1 were investigated. The histologic findings from reperfused intestines and the survival rates were compared between the anti-HMGB1 antibody treatment groups (group A treated with 6.0 mg/kg antibody and group B with 0.6 mg/kg antibody) and the control antibody treatment group (control group). RESULTS Serum HMGB1 concentrations increased early after reperfusion and peaked at 3 h. Immunohistochemistry for HMGB1 revealed a high degree of positive staining in the epithelial cells of the damaged villi. Anti-HMGB1 antibody treatment significantly reduced this damage (P < 0.05) and improved the 48-h survival rate (90% in group A versus 50% in the controls; P < 0.05). CONCLUSIONS These results suggest that HMGB1 plays a key role in small intestinal ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Masayuki Kojima
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kono H, Fujii H, Ogiku M, Hara M, Tsuchiya M, Ishii K, Hosomura N. The Kupffer Cell Inhibition Exacerbates but Splenectomy Prevents Mortality in a Rat Septic Peritonitis Model. J Surg Res 2012; 175:101-12. [DOI: 10.1016/j.jss.2011.02.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 01/24/2011] [Accepted: 02/17/2011] [Indexed: 11/16/2022]
|
44
|
Oshima G, Shinoda M, Tanabe M, Ebinuma H, Nishiyama R, Takano K, Yamada S, Miyasho T, Masugi Y, Matsuda S, Suda K, Fukunaga K, Matsubara K, Hibi T, Yagi H, Hayashida T, Yamagishi Y, Obara H, Itano O, Takeuchi H, Kawachi S, Saito H, Hibi T, Maruyama I, Kitagawa Y. Increased plasma levels of high mobility group box 1 in patients with acute liver failure. Eur Surg Res 2012; 48:154-162. [PMID: 22585050 DOI: 10.1159/000338363] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 03/07/2012] [Indexed: 11/19/2022]
Abstract
BACKGROUND High-mobility group box 1 (HMGB1) is a monocyte-derived late-acting inflammatory mediator, which is released in conditions such as shock, tissue injury and endotoxin-induced lethality. In this study, we determined the plasma and hepatic tissue levels of HMGB1 in patients with acute liver failure (ALF). PATIENTS AND METHODS We determined the plasma levels of HMGB1 and aspartate aminotransferase (AST) in 7 healthy volunteers (HVs), 40 patients with liver cirrhosis (LC), 37 patients with chronic hepatitis (CH), 18 patients with severe acute hepatitis (AH), and 14 patients with fulminant hepatitis (FH). The 14 patients with FH were divided into two subgroups depending upon the history of plasma exchange (PE) before their plasma sample collection. The hepatic levels of HMGB1 were measured in tissue samples from 3 patients with FH who underwent living-donor liver transplantation and from 3 healthy living donors. Hepatic tissue samples were also subjected to immunohistochemical examination for HMGB1. RESULTS The plasma levels of HMGB1 (ng/ml) were higher in patients with liver diseases, especially in FH patients with no history of PE, than in HVs (0.3 ± 0.3 in HVs, 4.0 ± 2.0 in LC, 5.2 ± 2.6 in CH, 8.6 ± 4.8 in severe AH, 7.8 ± 2.7 in FH with a history of PE, and 12.5 ± 2.6 in FH with no history of PE, p < 0.05 in each comparison). There was a strong and statistically significant relationship between the mean plasma HMGB1 level and the logarithm of the mean AST level (R = 0.900, p < 0.05). The hepatic tissue levels of HMGB1 (ng/mg tissue protein) were lower in patients with FH than in healthy donors (539 ± 116 in FH vs. 874 ± 81 in healthy donors, p < 0.05). Immunohistochemical staining for HMGB1 was strong and clear in the nuclei of hepatocytes in liver sections from healthy donors, but little staining in either nuclei or cytoplasm was evident in specimens from patients with FH. CONCLUSION We confirmed that plasma HMGB1 levels were increased in patients with ALF. Based on a comparison between HMGB1 contents in normal and ALF livers, it is very likely that HMGB1 is released from injured liver tissue.
Collapse
Affiliation(s)
- G Oshima
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Tadie JM, Bae HB, Deshane JS, Bell CP, Lazarowski ER, Chaplin DD, Thannickal VJ, Abraham E, Zmijewski JW. Toll-like receptor 4 engagement inhibits adenosine 5'-monophosphate-activated protein kinase activation through a high mobility group box 1 protein-dependent mechanism. Mol Med 2012; 18:659-68. [PMID: 22396017 DOI: 10.2119/molmed.2011.00401] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 03/01/2012] [Indexed: 12/29/2022] Open
Abstract
Despite the potent antiinflammatory effects of pharmacologically induced adenosine 5'-monophosphate kinase (AMPK) activation on Toll-like receptor 4 (TLR4)-induced cellular activation, there is little evidence that AMPK is activated during inflammatory conditions. In the present studies, we examined mechanisms by which TLR4 engagement may affect the ability of AMPK to become activated in neutrophils and macrophages under in vitro conditions and in the lungs during lipopolysaccharide (LPS)-induced acute lung injury. We found that incubation of neutrophils or macrophages with LPS diminished the ability of 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR) or hydrogen peroxide (H(2)O(2)) to activate AMPK. Although ratios of AMP to adenosine 5'-triphosphate (ATP) were increased in LPS-treated neutrophils and in the lungs of LPS exposed mice, a condition that should result in AMPK activation, no activation of AMPK was found. Immunocytochemistry and Western blot analysis revealed that nuclear to cytosolic translocation of the proinflammatory mediator high mobility group box 1 protein (HMGB1) correlated with inhibition of AMPK activation in LPS-stimulated macrophages. Moreover, while induced overexpression of HMGB1 resulted in inhibition of AMPK activation, Small interfering RNA (siRNA)-induced knockdown of HMGB1 was associated with enhanced activation of AMPK in macrophages incubated with AICAR. Increased interaction between liver kinase B1 (LKB1), an upstream activator of AMPK, and HMGB1 was found in LPS-stimulated macrophages and in the lungs of mice exposed to LPS. These results suggest that nuclear to cytoplasmic translocation of HMGB1 in TLR4-activated cells potentiates inflammatory responses by binding to LKB1, thereby inhibiting the antiinflammatory effects of AMPK activation.
Collapse
Affiliation(s)
- Jean-Marc Tadie
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kohno T, Anzai T, Kaneko H, Sugano Y, Shimizu H, Shimoda M, Miyasho T, Okamoto M, Yokota H, Yamada S, Yoshikawa T, Okada Y, Yozu R, Ogawa S, Fukuda K. High-mobility group box 1 protein blockade suppresses development of abdominal aortic aneurysm. J Cardiol 2012; 59:299-306. [DOI: 10.1016/j.jjcc.2012.01.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 12/09/2011] [Accepted: 01/09/2012] [Indexed: 01/22/2023]
|
47
|
Yoshikawa T, Takeuchi H, Suda K, Miyasho T, Yamada S, Okamoto M, Kawamura Y, Maruyama I, Kitajima M, Kitagawa Y. High-dose immunoglobulin preparations improve survival in a CLP-induced rat model of sepsis. Langenbecks Arch Surg 2012; 397:457-465. [PMID: 22116598 DOI: 10.1007/s00423-011-0878-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 11/13/2011] [Indexed: 12/29/2022]
Abstract
PURPOSE The efficacy of intravenous immunoglobulin G in the treatment of patients with severe sepsis or septic shock is still being debated. We investigated the impact of high-dose immunoglobulin administration on the survival rate and serum high-mobility group box chromosomal protein 1 (HMGB1) level in a rat model of sepsis created by cecal ligation and puncture (CLP). METHODS Rats received either CLP-induced sepsis or had additional immunoglobulin treatment in 1,500 or 300 mg/kg. After induction of sepsis and respective treatment conditions, pulmonary and renal tissues were examined histologically for pathological changes at postoperative hour (POH) 4, and serum cytokine and HMGB1 levels were measured at POH 4, 8, 20, and 44. Using other rats, we also observed the survival rate after CLP for 7 days. RESULTS Treatment with immunoglobulin significantly improved survival rate at postoperative day 7 (73% in the high-dose group vs. 33% in the control group; p = 0.037). The serum lactate dehydrogenase, endotoxin, creatinine, and blood urea nitrogen levels were significantly lower in the high-dose group than in the other groups. The serum HMGB1 level had increased at 4 h postoperatively in the control group (10.2 ± 3.3 ng/mL) and low-dose group (10.3 ± 4.0 ng/mL), but it was significantly reduced in the high-dose group (4.2 ± 0.8 ng/mL) compared with the control group (p = 0.03). CONCLUSIONS Our results suggest that high-dose immunoglobulin therapy may improve the serum endotoxin and HMGB1 levels and overall survival rate in sepsis by inhibiting the inflammation.
Collapse
Affiliation(s)
- Takahisa Yoshikawa
- Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Hashimoto T, Ishii J, Kitagawa F, Yamada S, Hattori K, Okumura M, Naruse H, Motoyama S, Matsui S, Tanaka I, Izawa H, Maruyama I, Nomura M, Ozaki Y. Circulating high-mobility group box 1 and cardiovascular mortality in unstable angina and non-ST-segment elevation myocardial infarction. Atherosclerosis 2012; 221:490-5. [PMID: 22369934 DOI: 10.1016/j.atherosclerosis.2012.01.040] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2011] [Revised: 01/24/2012] [Accepted: 01/24/2012] [Indexed: 11/17/2022]
Abstract
OBJECTIVE High-mobility group box 1 (HMGB1) is a damage-associated molecular pattern molecule, which suggests a potential role of this protein in the pathophysiology of acute coronary syndrome (ACS). Circulating HMGB1 has been shown to be independently associated with cardiac mortality in ST-segment elevation myocardial infarction. However, its prognostic value remains unclear in unstable angina and non-ST-segment elevation myocardial infarction (UA/NSTEMI). METHODS HMGB1, high-sensitivity C-reactive protein (hsCRP), cardiac troponin I and B-type natriuretic peptide concentrations were measured on admission in 258 consecutive patients (mean age of 67 years) hospitalized for UA/NSTEMI within 24h (mean, 7.4h) of the onset of chest symptoms. RESULTS A total of 38 (14.7%) cardiovascular deaths, including 10 in-hospital deaths, occurred during a median follow-up period of 49 months after admission. In a stepwise Cox regression analysis including 19 well-known clinical predictors of ACS, HMGB1 [relative risk (RR) 3.24 per 10-fold increment; P = 0.0003], cardiac troponin I (RR 1.83 per 10-fold increment, P = 0.0007), Killip class>1 (RR 4.67, P = 0.0001) and age (RR 1.05 per 1-year increment, P = 0.03), but not hsCRP, were independently associated with cardiovascular mortality. In-hospital and cardiovascular mortality rates were higher in patients with increased HMGB1 (≥ 2.4 ng/mL of median value) than those without increased HMGB1 (6.3% vs. 1.5%, P = 0.04; and 23% vs. 6.9%, P = 0.0003). CONCLUSION Circulating concentration of HMGB1 on admission may be a potential and independent predictor of cardiovascular mortality in patients hospitalized for UA/NSTEMI within 24h of onset.
Collapse
Affiliation(s)
- Tousei Hashimoto
- Department of Internal Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Vitali R, Stronati L, Negroni A, Di Nardo G, Pierdomenico M, del Giudice E, Rossi P, Cucchiara S. Fecal HMGB1 is a novel marker of intestinal mucosal inflammation in pediatric inflammatory bowel disease. Am J Gastroenterol 2011; 106:2029-2040. [PMID: 21788990 DOI: 10.1038/ajg.2011.231] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES High-mobility group box 1 (HMGB1) is a nuclear protein with functions in the regulation of transcription. In inflammatory conditions, HMGB1 is actively secreted from immune cells in the extracellular matrix, where it behaves as a proinflammatory cytokine. The aim of the present study was to investigate the role of HMGB1 in pediatric inflammatory bowel disease (IBD). METHODS We analyzed the stools of 19 children with Crohn's disease (CD), 21 with ulcerative colitis (UC), and 13 controls. The gene/protein expression levels of HMGB1 were assessed in bioptic specimens of all children using real-time PCR and western blot assay. Finally, intracellular localization of the protein was analyzed by western blot, after separation of nuclear and cytoplasmic extracts, and by immunohistochemistry. RESULTS HMGB1 protein levels were significantly increased (P<0.001) in the stools of patients, but were undetectable in the controls; fecal HMGB1 correlated well with fecal calprotectin levels (r: 0.77 in CD, r: 0.70 in UC; P<0.01); and mRNA and protein expression were unchanged in inflamed bioptic tissues compared with controls. However, by separately analyzing the nuclear and cytoplasmic fraction, we detected the cytoplasmic HMGB1 expression to be significantly enhanced (P<0.01) in the inflamed tissues of the patients. In addition, HMGB1 was significantly detected in 16 patients with inactive disease, whose endoscopic scores showed persisting inflammation, suggesting that it may be a sensitive marker of mucosal inflammation, although the disease is clinically inactive. CONCLUSIONS It was shown for the first time in our study that HMGB1 is secreted by human inflamed intestinal tissues and abundantly found in the stools of IBD patients. Hence, it can be considered as a novel marker for intestinal inflammation. We can also suggest that the presence of HMGB1 in large amounts in the fecal stream of IBD patients is mainly due to active secretion of the protein stored in the nucleus rather than a "de novo" synthesis.
Collapse
Affiliation(s)
- Roberta Vitali
- Department of Radiobiology and Human Health, ENEA, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Raby AC, Holst B, Davies J, Colmont C, Laumonnier Y, Coles B, Shah S, Hall J, Topley N, Köhl J, Morgan BP, Labéta MO. TLR activation enhances C5a-induced pro-inflammatory responses by negatively modulating the second C5a receptor, C5L2. Eur J Immunol 2011; 41:2741-52. [PMID: 21630250 PMCID: PMC3638321 DOI: 10.1002/eji.201041350] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 05/03/2011] [Accepted: 05/25/2011] [Indexed: 01/07/2023]
Abstract
TLR and complement activation ensures efficient clearance of infection. Previous studies documented synergism between TLRs and the receptor for the pro-inflammatory complement peptide C5a (C5aR/CD88), and regulation of TLR-induced pro-inflammatory responses by C5aR, suggesting crosstalk between TLRs and C5aR. However, it is unclear whether and how TLRs modulate C5a-induced pro-inflammatory responses. We demonstrate a marked positive modulatory effect of TLR activation on cell sensitivity to C5a in vitro and ex vivo and identify an underlying mechanistic target. Pre-exposure of PBMCs and whole blood to diverse TLR ligands or bacteria enhanced C5a-induced pro-inflammatory responses. This effect was not observed in TLR4 signalling-deficient mice. TLR-induced hypersensitivity to C5a did not result from C5aR upregulation or modulation of C5a-induced Ca2+ mobilization. Rather, TLRs targeted another C5a receptor, C5L2 (acting as a negative modulator of C5aR), by reducing C5L2 activity. TLR-induced hypersensitivity to C5a was mimicked by blocking C5L2 and was not observed in C5L2KO mice. Furthermore, TLR activation inhibited C5L2 expression upon C5a stimulation. These findings identify a novel pathway of crosstalk within the innate immune system that amplifies innate host defense at the TLR-complement interface. Unravelling the mutually regulated activities of TLRs and complement may reveal new therapeutic avenues to control inflammation.
Collapse
Affiliation(s)
- Anne-Catherine Raby
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Cardiff, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|