101
|
Máca J, Burša F, Ševčík P, Sklienka P, Burda M, Holub M. Alarmins and Clinical Outcomes After Major Abdominal Surgery-A Prospective Study. J INVEST SURG 2016; 30:152-161. [PMID: 27689623 DOI: 10.1080/08941939.2016.1231855] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE Tissue injury causing immune response is an integral part of surgical procedure. Evaluation of the degree of surgical trauma could help to improve postoperative management and determine the clinical outcomes. MATERIALS AND METHODS We analyzed serum levels of alarmins, including S100A5, S100A6, S100A8, S100A9, S100A11, and S100A12; high-mobility group box 1; and heat-shock protein 70, after elective major abdominal surgery (n = 82). Blood samples were collected for three consecutive days after surgery. The goals were to evaluate the relationships among the serum levels of alarmins and selected surgical characteristics and to test potential of alarmins to predict the clinical outcomes. RESULTS Significant, positive correlations were found for high-mobility group box 1 with the length of surgery, blood loss, and intraoperative fluid intake for all three days of blood sampling. The protein S100A8 serum levels showed positive correlations with intensive care unit length of stay, 28-day and in-hospital mortality. The protein S100A12 serum levels had significant, positive correlations with intensive care unit length of stay, 28-day mortality, and in-hospital mortality. We did not find significant differences in alarmin levels between cancer and noncancer subjects. CONCLUSION The high-mobility group box 1 serum levels reflect the degree of surgical injury, whereas proteins S100A8 and S100A12 might be considered good predictors of major abdominal surgery morbidity and mortality.
Collapse
Affiliation(s)
- Jan Máca
- a University of Ostrava , Ostrava , Czech Republic.,b University Hospital of Ostrava , Ostrava , Czech Republic
| | - Filip Burša
- a University of Ostrava , Ostrava , Czech Republic.,b University Hospital of Ostrava , Ostrava , Czech Republic
| | - Pavel Ševčík
- a University of Ostrava , Ostrava , Czech Republic.,b University Hospital of Ostrava , Ostrava , Czech Republic
| | - Peter Sklienka
- a University of Ostrava , Ostrava , Czech Republic.,b University Hospital of Ostrava , Ostrava , Czech Republic
| | - Michal Burda
- c University of Ostrava , Institute for Research and Applications of Fuzzy Modeling , Ostrava , Czech Republic
| | - Michal Holub
- d Univerzita Karlova v Praze , First Faculty Of Medicine , Praha , Czech Republic.,e Military Hospital of Prague , Prague , Czech Republic
| |
Collapse
|
102
|
Li W, Wu AH, Zhu S, Li J, Wu R, D'Angelo J, Wang H. EGCG induces G-CSF expression and neutrophilia in experimental sepsis. Immunol Res 2016; 63:144-52. [PMID: 26293782 DOI: 10.1007/s12026-015-8681-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
A major green tea component, epigallocatechin-3-gallate (EGCG), has been proven protective against lethal sepsis in experimental setting, but its protective mechanisms remain incompletely understood. Here, we provide evidence to support EGCG's capacities in stimulating G-CSF production and neutrophilia in vivo. In an animal model of sepsis, EGCG significantly elevated peritoneal levels of G-CSF and several chemokines (e.g., MCP-1/CCL2 and MIP-1γ/CCL9), and consequently increased peritoneal neutrophil numbers (neutrophilia) at a late stage. In vitro, EGCG divergently affected HMGB1-mediated production of several chemokines: reducing CXCL15 and RANTES/CCL5, but elevating G-CSF and MIP-1α/CCL3 production by peritoneal macrophages. Similarly, it significantly induced the expression and secretion of G-CSF and MIP-1α/CCL3 in human peripheral blood mononuclear cells. Based on our preliminary data, it may be important to search for anti-inflammatory and G-CSF-stimulating agents for the clinical management of inflammatory diseases.
Collapse
Affiliation(s)
- Wei Li
- Department of Emergency Medicine, North Shore University Hospital, 350 Community Drive, Manhasset, NY, 11030, USA
- The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Andrew H Wu
- Department of Emergency Medicine, North Shore University Hospital, 350 Community Drive, Manhasset, NY, 11030, USA
- The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Shu Zhu
- Department of Emergency Medicine, North Shore University Hospital, 350 Community Drive, Manhasset, NY, 11030, USA
- The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Jianhua Li
- The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Rong Wu
- The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - John D'Angelo
- Department of Emergency Medicine, North Shore University Hospital, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Haichao Wang
- Department of Emergency Medicine, North Shore University Hospital, 350 Community Drive, Manhasset, NY, 11030, USA.
- The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
103
|
Shimizu T, Yamakuchi M, Biswas KK, Aryal B, Yamada S, Hashiguchi T, Maruyama I. HMGB1 is secreted by 3T3-L1 adipocytes through JNK signaling and the secretion is partially inhibited by adiponectin. Obesity (Silver Spring) 2016; 24:1913-21. [PMID: 27430164 DOI: 10.1002/oby.21549] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/13/2016] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Obesity is a chronic inflammatory disease, and adipocytes contribute to obesity-associated inflammation by releasing inflammatory mediators. High mobility group box 1 (HMGB1), a highly conserved DNA-binding protein, mainly localized to cell nuclei, has been recently recognized as an innate pro-inflammatory mediator when released extracellularly. It was hypothesized that HMGB1 is an adipocytokine that acts as an innate pro-inflammatory mediator in white adipose tissue (WAT) of patients with obesity and is associated with insulin resistance. Additionally, it was hypothesized that HMGB1 secretion is regulated by adiponectin. METHODS 3T3-L1 cells were differentiated into mature adipocytes. After tumor necrosis factor-α (TNF-α) stimulation, HMGB1 in culture media was measured. Localizations of HMGB1 in 3T3-L1 adipocytes and human WAT were examined by immunostaining. RESULTS HMGB1 was secreted from TNF-α-induced 3T3-L1 adipocytes through JNK signaling. HMGB1-activated MAP kinases (ERK1/2, JNK) and suppressed insulin-stimulated Akt phosphorylation in 3T3-L1 adipocytes. The cytoplasm in 3T3-L1 adipocytes and adipocytes of WAT from a patient with obesity was intensely stained with HMGB1. Adiponectin partially inhibited TNF-α-induced HMGB1 secretion from 3T3-L1 adipocytes. CONCLUSIONS These findings suggest that HMGB1 is a pro-inflammatory adipocytokine involved in WAT inflammation and insulin resistance in patients with obesity, which may contribute to the progression of metabolic syndrome, and that adiponectin protects against HMGB1-induced adipose tissue inflammation.
Collapse
Affiliation(s)
- Toshiaki Shimizu
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Munekazu Yamakuchi
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kamal Krishna Biswas
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh
| | - Bibek Aryal
- Cardiovascular and Gastroenterological Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | | | - Teruto Hashiguchi
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Ikuro Maruyama
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
104
|
Shi Z, Chapes SK, Ben-Arieh D, Wu CH. An Agent-Based Model of a Hepatic Inflammatory Response to Salmonella: A Computational Study under a Large Set of Experimental Data. PLoS One 2016; 11:e0161131. [PMID: 27556404 PMCID: PMC4996536 DOI: 10.1371/journal.pone.0161131] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 07/29/2016] [Indexed: 01/04/2023] Open
Abstract
We present an agent-based model (ABM) to simulate a hepatic inflammatory response (HIR) in a mouse infected by Salmonella that sometimes progressed to problematic proportions, known as "sepsis". Based on over 200 published studies, this ABM describes interactions among 21 cells or cytokines and incorporates 226 experimental data sets and/or data estimates from those reports to simulate a mouse HIR in silico. Our simulated results reproduced dynamic patterns of HIR reported in the literature. As shown in vivo, our model also demonstrated that sepsis was highly related to the initial Salmonella dose and the presence of components of the adaptive immune system. We determined that high mobility group box-1, C-reactive protein, and the interleukin-10: tumor necrosis factor-α ratio, and CD4+ T cell: CD8+ T cell ratio, all recognized as biomarkers during HIR, significantly correlated with outcomes of HIR. During therapy-directed silico simulations, our results demonstrated that anti-agent intervention impacted the survival rates of septic individuals in a time-dependent manner. By specifying the infected species, source of infection, and site of infection, this ABM enabled us to reproduce the kinetics of several essential indicators during a HIR, observe distinct dynamic patterns that are manifested during HIR, and allowed us to test proposed therapy-directed treatments. Although limitation still exists, this ABM is a step forward because it links underlying biological processes to computational simulation and was validated through a series of comparisons between the simulated results and experimental studies.
Collapse
Affiliation(s)
- Zhenzhen Shi
- Health Care Operations Resource Center, Department of Industrial and Manufacturing Systems Engineering, Kansas State University, Manhattan, Kansas, United States of America
| | - Stephen K. Chapes
- Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
| | - David Ben-Arieh
- Health Care Operations Resource Center, Department of Industrial and Manufacturing Systems Engineering, Kansas State University, Manhattan, Kansas, United States of America
| | - Chih-Hang Wu
- Health Care Operations Resource Center, Department of Industrial and Manufacturing Systems Engineering, Kansas State University, Manhattan, Kansas, United States of America
| |
Collapse
|
105
|
Factors affecting biomarkers of endothelial and alveolar epithelial dysfunction. Intensive Care Med 2016; 42:2111-2112. [PMID: 27535563 DOI: 10.1007/s00134-016-4460-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2016] [Indexed: 10/21/2022]
|
106
|
Role of cellular events in the pathophysiology of sepsis. Inflamm Res 2016; 65:853-868. [PMID: 27392441 DOI: 10.1007/s00011-016-0970-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 05/11/2016] [Accepted: 06/25/2016] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Sepsis is a dysregulated host immune response due to an uncontrolled infection. It is a leading cause of mortality in adult intensive care units globally. When the host immune response induced against a local infection fails to contain it locally, it progresses to sepsis, severe sepsis, septic shock and death. METHOD Literature survey was performed on the roles of different innate and adaptive immune cells in the development and progression of sepsis. Additionally, the effects of septic changes on reprogramming of different immune cells were also summarized to prepare the manuscript. FINDINGS Scientific evidences to date suggest that the loss of balance between inflammatory and anti-inflammatory responses results in reprogramming of immune cell activities that lead to irreversible tissue damaging events and multi-organ failure during sepsis. Many surface receptors expressed on immune cells at various stages of sepsis have been suggested as biomarkers for sepsis diagnosis. Various immunomodulatory therapeutics, which could improve the functions of immune cells during sepsis, were shown to restore immunological homeostasis and improve survival in animal models of sepsis. CONCLUSION In-depth and comprehensive knowledge on the immune cell activities and their correlation with severity of sepsis will help clinicians and scientists to design effective immunomodulatory therapeutics for treating sepsis.
Collapse
|
107
|
Nakamura T, Yoshioka T, Yamada S, Miyasho T, Sakakibara N, Hatanaka D. High-mobility group box-1 release into fetal circulation from umbilical cord tissue and amniotic epithelium in fetal ischemia. Pediatr Int 2016; 58:631-4. [PMID: 27097754 DOI: 10.1111/ped.12883] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 12/01/2015] [Accepted: 12/09/2015] [Indexed: 11/29/2022]
Abstract
We report the case of a baby with low birthweight born by emergency caesarean section at 33 weeks 2 days' gestation due to placental abruption. High-mobility group box-1 (HMGB-1) and interleukin-17 concentration in the umbilical cord blood were high at 55.7 ng/mL and 50.7 pg/mL, respectively. On immunostaining of umbilical cord and amniotic epithelium, HMGB-1 was identified in the nuclei of vascular endothelial cells and cytoplasm of the surrounding cells in the umbilical cord. This suggests that, in the present case of placental abruption and subsequent ischemic placenta and fetus, the high level of HMGB-1 observed was due to the release of HMGB-1 into the umbilical cord blood from the vascular endothelial cells of the umbilical cord.
Collapse
Affiliation(s)
- Toshihiko Nakamura
- Division of Neonatology, Center for Maternal Fetal and Neonatal Medicine, National Hospital Organization Nishisaitama Chuo National Hospital, Tokorozawa, Japan
| | - Toshirou Yoshioka
- Division of Neonatology, Center for Maternal Fetal and Neonatal Medicine, National Hospital Organization Nishisaitama Chuo National Hospital, Tokorozawa, Japan
| | - Shingo Yamada
- Division of Development, Shino-Test Corporation, Sagamihara, Kanagawa, Japan
| | - Taku Miyasho
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Nana Sakakibara
- Division of Neonatology, Center for Maternal Fetal and Neonatal Medicine, National Hospital Organization Nishisaitama Chuo National Hospital, Tokorozawa, Japan
| | - Daishuke Hatanaka
- Division of Neonatology, Center for Maternal Fetal and Neonatal Medicine, National Hospital Organization Nishisaitama Chuo National Hospital, Tokorozawa, Japan
| |
Collapse
|
108
|
Liu J, Zhang BL, Sun CL, Wang J, Li S, Wang JF. High mobility group box1 protein is involved in acute inflammation induced by Clostridium difficile toxin A. Acta Biochim Biophys Sin (Shanghai) 2016; 48:554-62. [PMID: 27151296 DOI: 10.1093/abbs/gmw038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/31/2016] [Indexed: 12/31/2022] Open
Abstract
High mobility group box1 (HMGB1), as a damage-associated inflammatory factor, contributes to the pathogenesis of numerous chronic inflammatory and autoimmune diseases. In this study, we explored the role of HMGB1 in CDI (Clostridium difficile infection) by in vivo and in vitro experiments. Our results showed that HMGB1 might play an important role in the acute inflammatory responses to C. difficile toxin A (TcdA), affect early inflammatory factors, and induce inflammation via the HMGB1-TLR4 pathway. Our study provides the essential information for better understanding the molecular mechanisms of CDI and the potential new therapeutic strategies for the treatment of this infection.
Collapse
Affiliation(s)
- Ji Liu
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Bei-Lei Zhang
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Chun-Li Sun
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Jun Wang
- Shenzhen Huada Gene Research Institute, Shenzhen 518083, China
| | - Shan Li
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China Guangdong Province Key Laboratory of Fermentation and Enzyme Engineering, South China University of Technology, Guangzhou 510006, China
| | - Ju-Fang Wang
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
109
|
Sohun M, Shen H. The implication and potential applications of high-mobility group box 1 protein in breast cancer. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:217. [PMID: 27386491 PMCID: PMC4916368 DOI: 10.21037/atm.2016.05.36] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 04/14/2016] [Indexed: 01/09/2023]
Abstract
High-mobility group box 1 protein (HMGB1) is a highly conserved, non-histone and ubiquitous chromosomal protein found enriched in active chromatin forming part of the high mobility group family of proteins and is encoded by the HMGB1 gene (13q12) in human beings. It has various intranuclear and extracellular functions. It plays an important role in the pathogenesis of many diseases including cancer. In 2012, there was approximately 1.67 million new breast cancer cases diagnosed which makes it the second most frequent cancer in the world after lung cancer (25% of all cancers) and the commonest cancer among women. Both pre-clinical and clinical studies have suggested that HMGB1 might be a useful target in the management of breast cancer. This review summarises the structure and functions of HMGB1 and its dual role in carcinogenesis both as a pro-tumorigenic and anti-tumorigenic factor. It also sums up evidence from in vitro and in vivo studies using breast cancer cell lines and samples which demonstrate its influence in radiotherapy, chemotherapy and hormonal therapy in breast cancer. It may have particular importance in HER2 positive and metastatic breast cancer. It might pave the way for new breast cancer treatments through development of novel drugs, use of microRNAs (miRNAs), targeting breast cancer stem cells (CSCs) and breast cancer immunotherapy. It may also play a role in determining breast cancer prognosis. Thus HMGB1 may open up novel avenues in breast cancer management.
Collapse
Affiliation(s)
- Moonindranath Sohun
- Department of Oncology, the Affiliated People's Hospital, Jiangsu University, Zhenjiang 212013, China
| | - Huiling Shen
- Department of Oncology, the Affiliated People's Hospital, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
110
|
Adiponectin Inhibits LPS-Induced HMGB1 Release through an AMP Kinase and Heme Oxygenase-1-Dependent Pathway in RAW 264 Macrophage Cells. Mediators Inflamm 2016; 2016:5701959. [PMID: 27313399 PMCID: PMC4904123 DOI: 10.1155/2016/5701959] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 05/10/2016] [Indexed: 12/30/2022] Open
Abstract
High mobility group protein B1 (HMGB1) is a late inflammatory mediator that exaggerates septic symptoms. Adiponectin, an adipokine, has potent anti-inflammatory properties. However, possible effects of adiponectin on lipopolysaccharide- (LPS-) induced HMGB1 release are unknown. The aim of this study was to investigate effects of full length adiponectin on HMGB1 release in LPS-stimulated RAW 264 macrophage cells. Treatment of the cells with LPS alone significantly induced HMGB1 release associated with HMGB1 translocation from the nucleus to the cytosol. However, prior treatment with adiponectin suppressed LPS-induced HMGB1 release and translocation. The anti-inflammatory cytokine interleukin- (IL-) 10 similarly suppressed LPS-induced HMGB1 release. Adiponectin treatment decreased toll-like receptor 4 (TLR4) mRNA expression and increased heme oxygenase- (HO-) 1 mRNA expression without inducing IL-10 mRNA, while IL-10 treatment decreased TLR2 and HMGB1 mRNA expression and increased the expression of IL-10 and HO-1 mRNA. Treatment with the HO-1 inhibitor ZnPP completely prevented the suppression of HMGB1 release by adiponectin but only partially inhibited that induced by IL-10. Treatment with compound C, an AMP kinase (AMPK) inhibitor, abolished the increase in HO-1 expression and the suppression of HMGB1 release mediated by adiponectin. In conclusion, our results indicate that adiponectin suppresses HMGB1 release by LPS through an AMPK-mediated and HO-1-dependent IL-10-independent pathway.
Collapse
|
111
|
McClellan SA, Ekanayaka SA, Li C, Jiang X, Barrett RP, Hazlett LD. Thrombomodulin Protects Against Bacterial Keratitis, Is Anti-Inflammatory, but Not Angiogenic. Invest Ophthalmol Vis Sci 2016; 56:8091-100. [PMID: 26720461 DOI: 10.1167/iovs.15-18393] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Thrombomodulin (TM) is a multidomain, transmembrane protein with anti-inflammatory properties. Thrombomodulin domain (D) 1 is lectin-like, interacting with Lewis Y antigen on lipopolysaccharide, and with HMGB1, while TMD23 is associated with angiogenic and anti-inflammatory functions. Thus, we tested if TM is protective against Pseudomonas aeruginosa keratitis and whether it enhanced corneal vascularity. METHODS Eyes of C57BL/6 (B6) mice were injected with recombinant TM (rTM), rTMD1, or PBS subconjunctivally before and intraperitoneally after infection with P. aeruginosa. Clinical scores, photography with a slit lamp, RT-PCR, ELISA, myeloperoxidase (MPO) assay, viable bacterial plate counts, and India ink perfusion were used to assess the disease response and corneal vascularity (rTM only). RESULTS Recombinant TM versus PBS treatment reduced clinical scores and corneal opacity. Corneal mRNA levels for HMGB1 were unchanged, but proinflammatory molecules IL-1β, CXCL2, NF-κB, TLR4, and RAGE were decreased; anti-inflammatory molecules SIGIRR and ST2 were increased. ELISA confirmed the mRNA data for HMGB1, IL-1β, and CXCL2 proteins. Both neutrophil influx and viable bacterial plate counts also were decreased after rTM treatment. Protein levels for angiogenic molecules VEGF, VEGFR-1, and VEGFR-2 were measured at 5 days post infection and were not different or reduced significantly after rTM treatment. Further, perfusion with India ink revealed similar vessel ingrowth between the two groups. Similar studies were performed with rTMD1, but disease severity, mRNA, proteins, MPO, and plate counts were not changed from controls. CONCLUSIONS These data provide evidence that rTM treatment is protective against bacterial keratitis, does not reduce HMGB1, and is not angiogenic.
Collapse
|
112
|
High Mobility Group Box-1 Promotes Inflammation-Induced Lymphangiogenesis via Toll-Like Receptor 4-Dependent Signalling Pathway. PLoS One 2016; 11:e0154187. [PMID: 27100831 PMCID: PMC4839690 DOI: 10.1371/journal.pone.0154187] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 04/11/2016] [Indexed: 01/28/2023] Open
Abstract
Lymphangiogenesis in inflammation has received considerable attention in recent years. Administration of modulating lymphangiogenesis provides more possibilities of treating inflammation-associated diseases. However, the main mediators and factors governing inflammation-induced lymphangiogenesis (ILA) are yet to be defined. Here, we explored the role of HMGB1-TLR4 signalling pathway in modulating inflammation-induced lymphangiogenesis and its underlying mechanisms using an ILA mouse model and 2 cell lines. Our results show that HMGB1 promoted VEGF-C-induced HDLECs proliferation in a dose-dependent manner and TLR4 mediates HMGB1-induced LECs proliferation and tube formation in vitro. And in vivo, rHMGB1 treatment significantly promoted ILA, and the promoting effects was inhibited notably when HMGB1-TLR4 was blocked. HMGB1-associated ILA is primarily dependent on TLR4 but not on TLR2. In mechanisms, the recruitment and activation of CD11b+ cells are important cellular mechanisms in HMGB1-TLR4 associated ILA, and multiple key pro-lymphangiogenesis molecules mediates HMGB1-TLR4 associated ILA, including VEGF-C/VEGFR3, inflammatory factors IL-1β and TNF-α, MMP-2 and MMP-9 and NF-κB p65. In conclusion, HMGB1-associated ILA is primarily dependent on TLR4, and CD11b+ cells and multiple molecular mechanisms mediate HMGB1-TLR4 associated ILA. Furthermore, the ILA can be effectively modulated by HMGB1-TLR4 signalling. Consequently, administration of modulating ILA through HMGB1-TLR4 pathway may provide us more possibilities of treating inflammation and lymphangiogenesis associated diseases.
Collapse
|
113
|
Nguyen A, Bhavsar S, Riley E, Caponetti G, Agrawal D. Clinical Value of High Mobility Group Box 1 and the Receptor for Advanced Glycation End-products in Head and Neck Cancer: A Systematic Review. Int Arch Otorhinolaryngol 2016; 20:382-389. [PMID: 27746844 PMCID: PMC5063723 DOI: 10.1055/s-0036-1583168] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 02/28/2016] [Indexed: 01/04/2023] Open
Abstract
Introduction High mobility group box 1 is a versatile protein involved in gene transcription, extracellular signaling, and response to inflammation. Extracellularly, high mobility group box 1 binds to several receptors, notably the receptor for advanced glycation end-products. Expression of high mobility group box 1 and the receptor for advanced glycation end-products has been described in many cancers. Objectives To systematically review the available literature using PubMed and Web of Science to evaluate the clinical value of high mobility group box 1 and the receptor for advanced glycation end-products in head and neck squamous cell carcinomas. Data synthesis A total of eleven studies were included in this review. High mobility group box 1 overexpression is associated with poor prognosis and many clinical and pathological characteristics of head and neck squamous cell carcinomas patients. Additionally, the receptor for advanced glycation end-products demonstrates potential value as a clinical indicator of tumor angiogenesis and advanced staging. In diagnosis, high mobility group box 1 demonstrates low sensitivity. Conclusion High mobility group box 1 and the receptor for advanced glycation end-products are associated with clinical and pathological characteristics of head and neck squamous cell carcinomas. Further investigation of the prognostic and diagnostic value of these molecules is warranted.
Collapse
Affiliation(s)
- Austin Nguyen
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, United States
| | - Sheila Bhavsar
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, United States
| | - Erinn Riley
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, United States
| | - Gabriel Caponetti
- Department of Pathology, Creighton University School of Medicine, Omaha, Nebraska, United States
| | - Devendra Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, United States
| |
Collapse
|
114
|
Fontaine M, Lepape A, Piriou V, Venet F, Friggeri A. Innate danger signals in acute injury: From bench to bedside. Anaesth Crit Care Pain Med 2016; 35:283-92. [PMID: 26987739 DOI: 10.1016/j.accpm.2015.10.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/08/2015] [Accepted: 10/08/2015] [Indexed: 11/24/2022]
Abstract
The description of the systemic inflammatory response syndrome (SIRS) as a reaction to numerous insults marked a turning point in the understanding of acute critical states, which are intensive care basic cases. This concept highlighted the final inflammatory response features whichever the injury mechanism is: infectious, or non-infectious such as extensive burns, traumas, major surgery or acute pancreatitis. In these cases of severe non-infectious insult, many endogenous mediators are released. Like infectious agents components, they can activate the immune system (via common signaling pathways) and initiate an inflammatory response. They are danger signals or alarmins. These molecules generally play an intracellular physiological role and acquire new functions when released in extracellular space. Many progresses brought new information on these molecules and on their function in infectious and non-infectious inflammation. These danger signals can be used as biomarkers and provide new pathophysiological and therapeutic approaches, particularly for immune dysfunctions occurring after an acute injury. We present herein the danger model, the main danger signals and the clinical consequences.
Collapse
Affiliation(s)
- Mathieu Fontaine
- Burn Intensive Care Unit, centre hospitalier Saint-Joseph-Saint-Luc, 20, quai Claude-Bernard, 69007 Lyon, France; EAM 4174 « Hemostasis, inflammation and sepsis », hospices civils de Lyon, université Claude-Bernard Lyon I, 69008 Lyon, France.
| | - Alain Lepape
- EAM 4174 « Hemostasis, inflammation and sepsis », hospices civils de Lyon, université Claude-Bernard Lyon I, 69008 Lyon, France; Intensive Care Unit, centre hospitalier Lyon Sud, 165, chemin du Grand-Revoyet, 69495 Pierre-Bénite cedex, France
| | - Vincent Piriou
- EAM 4174 « Hemostasis, inflammation and sepsis », hospices civils de Lyon, université Claude-Bernard Lyon I, 69008 Lyon, France; Intensive Care Unit, centre hospitalier Lyon Sud, 165, chemin du Grand-Revoyet, 69495 Pierre-Bénite cedex, France
| | - Fabienne Venet
- EAM 4174 « Hemostasis, inflammation and sepsis », hospices civils de Lyon, université Claude-Bernard Lyon I, 69008 Lyon, France; Immunology Laboratory, hôpital Édouard-Herriot, hospices civils de Lyon, 5, place d'Arsonval, 69437 Lyon cedex 03, France
| | - Arnaud Friggeri
- Intensive Care Unit, centre hospitalier Lyon Sud, 165, chemin du Grand-Revoyet, 69495 Pierre-Bénite cedex, France
| |
Collapse
|
115
|
Behringer M, Kilian Y, Montag J, Geesmann B, Mester J. Plasma concentration of high-mobility group box 1 (HMGB1) after 100 drop to vertical jumps and after a 1200-km bicycle race. Res Sports Med 2016; 24:119-29. [DOI: 10.1080/15438627.2015.1126275] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- M. Behringer
- Institute of Training Science and Sport Informatics, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933 Cologne, Germany
| | - Y. Kilian
- Institute of Training Science and Sport Informatics, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933 Cologne, Germany
| | - J. Montag
- Institute of Training Science and Sport Informatics, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933 Cologne, Germany
| | - B. Geesmann
- Institute of Biochemistry, German Sport University, Cologne 50933, Germany
| | - J. Mester
- Institute of Training Science and Sport Informatics, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933 Cologne, Germany
| |
Collapse
|
116
|
Liu Z, Wang Z, Han G, Huang L, Jiang J, Li S. Ketamine attenuates high mobility group box-1–induced inflammatory responses in endothelial cells. J Surg Res 2016; 200:593-603. [DOI: 10.1016/j.jss.2015.08.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 08/02/2015] [Accepted: 08/19/2015] [Indexed: 02/06/2023]
|
117
|
Yang YI, Woo JH, Seo YJ, Lee KT, Lim Y, Choi JH. Protective Effect of Brown Alga Phlorotannins against Hyper-inflammatory Responses in Lipopolysaccharide-Induced Sepsis Models. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:570-8. [PMID: 26730445 DOI: 10.1021/acs.jafc.5b04482] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Brown algae have been recognized as a food ingredient and health food supplement in Japan and Korea, and phlorotannins are unique marine phenol compounds produced exclusively by brown algae. Sepsis is a whole-body inflammatory condition with a mortality rate of 30-40%. Here, we investigated the effects of a phlorotannin-rich extract of the edible brown alga Ecklonia cava against hyper-inflammatory response in LPS-induced septic shock mouse model. E. cava extract significantly increased the survival rate and attenuated liver and kidney damage in the mice. In addition, E. cava attenuated serum levels of NO, PGE2, and HMGB-1. In macrophages, treatment with E. cava extract down-regulated iNOS, COX-2, TNF-α, IL-6, and HMGB-1. In addition, E. cava suppressed the NIK/TAK1/IKK/IκB/NFκB pathway. Moreover, E. cava increased Nrf2 and HO-1 expression. HO-1 knockdown using siRNA restored the extract-suppressed NO and PGE2 production. Dieckol, a major compound in the extract, reduced mortality, tissue toxicity, and serum levels of the inflammatory factors in septic mice. These data suggest that brown algae phlorotannins suppress septic shock through negative regulation of pro-inflammatory factors via the NIK/TAK1/IKK/IκB/NFκB and Nrf2/HO-1 pathways.
Collapse
Affiliation(s)
- Yeong-In Yang
- Department of Life & Nanopharmaceutical Science and ‡Department of Food and Nutrition, Kyung Hee University , 26 Kyunghee-daero, Dongdaemoon-gu, Seoul 130-701, South Korea
| | - Jeong-Hwa Woo
- Department of Life & Nanopharmaceutical Science and ‡Department of Food and Nutrition, Kyung Hee University , 26 Kyunghee-daero, Dongdaemoon-gu, Seoul 130-701, South Korea
| | - Yun-Ji Seo
- Department of Life & Nanopharmaceutical Science and ‡Department of Food and Nutrition, Kyung Hee University , 26 Kyunghee-daero, Dongdaemoon-gu, Seoul 130-701, South Korea
| | - Kyung-Tae Lee
- Department of Life & Nanopharmaceutical Science and ‡Department of Food and Nutrition, Kyung Hee University , 26 Kyunghee-daero, Dongdaemoon-gu, Seoul 130-701, South Korea
| | - Yunsook Lim
- Department of Life & Nanopharmaceutical Science and ‡Department of Food and Nutrition, Kyung Hee University , 26 Kyunghee-daero, Dongdaemoon-gu, Seoul 130-701, South Korea
| | - Jung-Hye Choi
- Department of Life & Nanopharmaceutical Science and ‡Department of Food and Nutrition, Kyung Hee University , 26 Kyunghee-daero, Dongdaemoon-gu, Seoul 130-701, South Korea
| |
Collapse
|
118
|
Walshe CM, Laffey JG, Kevin L, O’Toole D. Sepsis protects the myocardium and other organs from subsequent ischaemic/reperfusion injury via a MAPK-dependent mechanism. Intensive Care Med Exp 2015; 3:35. [PMID: 26215802 PMCID: PMC4513033 DOI: 10.1186/s40635-014-0035-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 12/09/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Sepsis has been shown to precondition the intact heart against ischaemia/reperfusion (IR) injury, and prior endotoxin exposure of cells in in vitro models has shown evidence of protection against subsequent simulated ischaemia. Our aim in this study is to validate these findings and further investigate the signaling pathways involved. METHODS Adult male Sprague Dawley rats were randomised to control (n = 7) or caecal ligation and perforation (CLP)-induced sepsis (n = 7). Hearts were harvested at 48 h, suspended in Langendorff mode and subjected to 30-min global ischaemia followed by 90-min reperfusion. In subsequent experiments, designed to determine the mechanisms by which sepsis protected against ischaemic injury, endotoxin-stimulated isolated cardiomyocytes, pulmonary A549 cells and renal HK2 cells were subjected to normoxic and hypoxic conditions. The roles of key pathways, including mitogen-activated protein (MAP) kinases extracellular-regulated protein kinase (ERK) 1/2, p38 MAPK (p38), c-Jun NH2-terminal protein kinase (JNK)), and nuclear factor-kappaB (NF-κB) were examined. RESULTS Systemic sepsis protected isolated hearts from subsequent ischaemic/reperfusion-induced injury, enhancing functional recovery on reperfusion [developed left ventricular pressure ((d)LVP) mean(SE) 66.63(±10.7) mmHg vs. 54.13(±9.9) mmHg; LVPmax at 60 min 67.29(±11.9) vs. 72.48(±9.3), sepsis vs. control] despite significantly reduced baseline LV function in CLP animals (p < 0.001). Septic preconditioning significantly reduced infarct size after IR injury (p < 0.05). Endotoxin exposure protected isolated cardiomyocytes against hypoxia-induced cell death (p < 0.001). This effect appeared mediated in part via the p38, JNK and NF-κB pathways, but was independent of the ERK pathway, and did not appear to be mediated via HMGB1. The preconditioning effect of endotoxin was also demonstrated in isolated kidney and lung cells, suggesting that this preconditioning effect of sepsis is not confined to the myocardium. CONCLUSIONS Sepsis preconditions the isolated rat heart against myocardial IR injury. These effects appeared to be mediated in part via the p38, JNK and NF-κB and pathways, but were independent of the ERK and HMGB pathways.
Collapse
Affiliation(s)
- Criona M Walshe
- Department of Anaesthesia, Galway University Hospitals and National University of Ireland, University Road, Galway, Ireland
| | - John G Laffey
- Department of Anaesthesia, Galway University Hospitals and National University of Ireland, University Road, Galway, Ireland
| | - Leo Kevin
- Department of Anaesthesia, Galway University Hospitals and National University of Ireland, University Road, Galway, Ireland
| | - Daniel O’Toole
- Department of Anaesthesia, Galway University Hospitals and National University of Ireland, University Road, Galway, Ireland
| |
Collapse
|
119
|
Zheng YJ, Xu WP, Ding G, Gao YH, Wang HR, Pan SM. Expression of HMGB1 in septic serum induces vascular endothelial hyperpermeability. Mol Med Rep 2015; 13:513-21. [PMID: 26572550 DOI: 10.3892/mmr.2015.4536] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 09/09/2015] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the effects of high mobility group protein B1 (HMGB1), which is expressed in the serum of patients with sepsis, on vascular endothelial permeability. Sera from patients with sepsis were used to treat endothelial cells (ECs), and the effect on endothelial permeability was evaluated using immunofluorescence. The morphologies of endothelial cytoskeletal actin and vascular endothelial (VE)‑cadherin were assessed using laser scanning confocal microscopy. The protein expression levels of HMGB1, B‑cell lymphoma 2 (BCL‑2) and BCL‑2‑associated X protein (BAX) were detected using western blotting. EC apoptosis was measured using flow cytometry. The results demonstrated that HMGB1 was significantly expressed in the serum 24 h following the onset of sepsis, and the expression levels peaked at 48 h, which were sustained until 96 h post‑onset. Compared with the control group, treatment of the ECs with 20% septic serum in vitro significantly increased endothelial monolayer permeability (P<0.01), markedly induced transcellular filamentous (F)‑actin rearrangement with stress fiber formation, and resulted in the localization of VE‑cadherin fragmentations at the cell borders with increased gaps between ECs. Furthermore, flow cytometry showed that the apoptotic rate of ECs was significantly increased following treatment with septic serum. In addition, the expression levels of BAX were significantly increased, whereas the expression levels of BCL‑2 were significantly decreased. Pretreatment with an HMGBI inhibitor (ethyl pyruvate; 5 µM) 24 h prior to treatment with the septic serum attenuated the effects of septic serum treatment. Together, these findings suggested that treatment of ECs with sera from patients with sepsis may induce the loss of vascular endothelial monolayer integrity, elicit the formation of endothelial F‑actin stress fibers and initiate VE‑cadherin redistribution, which may be attributed to high levels of HMGB1 in the serum. This mechanism also appears to involve changes in the activation of BAX and BCL‑2, resulting in EC apoptosis.
Collapse
Affiliation(s)
- Yun-Jiang Zheng
- Emergency Department, Chongming Branch, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 202150, P.R. China
| | - Wei-Ping Xu
- Administration Division, Chongming Branch, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 202150, P.R. China
| | - Gang Ding
- Department of Oncology, Chongming Branch, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 202150, P.R. China
| | - Yu-Hua Gao
- Administration Division, Chongming Branch, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 202150, P.R. China
| | - Hai-Rong Wang
- Emergency Department, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Shu-Ming Pan
- Emergency Department, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| |
Collapse
|
120
|
Mathematical Model of Innate and Adaptive Immunity of Sepsis: A Modeling and Simulation Study of Infectious Disease. BIOMED RESEARCH INTERNATIONAL 2015; 2015:504259. [PMID: 26446682 PMCID: PMC4584099 DOI: 10.1155/2015/504259] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 04/07/2015] [Accepted: 04/15/2015] [Indexed: 12/14/2022]
Abstract
Sepsis is a systemic inflammatory response (SIR) to infection. In this work, a system dynamics mathematical model (SDMM) is examined to describe the basic components of SIR and sepsis progression. Both innate and adaptive immunities are included, and simulated results in silico have shown that adaptive immunity has significant impacts on the outcomes of sepsis progression. Further investigation has found that the intervention timing, intensity of anti-inflammatory cytokines, and initial pathogen load are highly predictive of outcomes of a sepsis episode. Sensitivity and stability analysis were carried out using bifurcation analysis to explore system stability with various initial and boundary conditions. The stability analysis suggested that the system could diverge at an unstable equilibrium after perturbations if rt2max (maximum release rate of Tumor Necrosis Factor- (TNF-) α by neutrophil) falls below a certain level. This finding conforms to clinical findings and existing literature regarding the lack of efficacy of anti-TNF antibody therapy.
Collapse
|
121
|
Geng Y, Ma Q, Liu YN, Peng N, Yuan FF, Li XG, Li M, Wu YS, Li BL, Song WB, Zhu W, Xu WW, Fan J, Su L. Heatstroke induces liver injury via IL-1β and HMGB1-induced pyroptosis. J Hepatol 2015; 63:622-33. [PMID: 25931416 DOI: 10.1016/j.jhep.2015.04.010] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 03/28/2015] [Accepted: 04/02/2015] [Indexed: 01/06/2023]
Abstract
BACKGROUND & AIMS Liver injury is a common complication of heat stroke (HS), and often constitutes a direct cause for patient death. The cellular and molecular mechanism underlying HS-induced liver injury remains unclear. Recent evidence indicates that inflammasome plays an important role in mediating sterile inflammation triggered by tissue damage. Using a rat HS model, we identified a novel mechanism by which inflammasome-dependent interleukin-1β (IL-1β) activation and hepatocyte pyroptosis mediate HS-induced liver injury. METHODS To induce HS, rats were subjected to heat exposure. Inhibition of inflammasomes was achieved by RNA silencing and pharmacologic inhibitor prior to heat exposure. Inflammasome assembly, caspase-1 activation, histological changes, as well as serum levels of liver enzymes were measured. RESULTS We demonstrated that the onset of HS activated inflammasome in the liver as evidenced by increased capase-1 activity and the association of inflammasome components NOD-like receptor family pyrin domain containing 3 (Nlrp3) and apoptosis speck-like protein containing a caspase-recruitment domain (ASC); and the activated inflammasome, in turn, induced IL-1β activation and hepatocyte pyroptosis, and subsequent augmented liver injury. HS-induced hepatocyte inflammasome activation seems to be high-mobility group box 1 (HMGB1) dependent. Inhibition of Nlrp3, caspase-1, or HMGB1 prevented HS-induced liver inflammation and ameliorated liver injury. CONCLUSIONS These findings demonstrate an important role of HMGB1 in mediating inflammasome activation in the development of liver injury following HS, and suggest that targeting inflammasome may represent a novel therapeutic strategy to limit cell death and prevent liver failure after HS.
Collapse
Affiliation(s)
- Yan Geng
- Department of Gastroenterology, 303 Hospital of People's Liberation Army, Nanning 530021, PR China; Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou 510010, PR China.
| | - Qiang Ma
- State Key Laboratory of Organ Failure Research, Institute of Antibody Engineering, Southern Medical University, Guangzhou 510515, PR China.
| | - Ya-Nan Liu
- Department of Graduate School, Southern Medical University, Guangzhou 510515, PR China
| | - Na Peng
- Department of Graduate School, Southern Medical University, Guangzhou 510515, PR China; Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou 510010, PR China
| | - Fang-Fang Yuan
- Department of Graduate School, Southern Medical University, Guangzhou 510515, PR China; Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Xing-Gui Li
- Department of Neurology, The Fifth People's Hospital of Chongqing, Chongqing 400062, PR China; Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou 510010, PR China
| | - Ming Li
- State Key Laboratory of Organ Failure Research, Institute of Antibody Engineering, Southern Medical University, Guangzhou 510515, PR China
| | - Ying-Song Wu
- State Key Laboratory of Organ Failure Research, Institute of Antibody Engineering, Southern Medical University, Guangzhou 510515, PR China
| | - Bing-ling Li
- Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou 510010, PR China
| | - Wei-bing Song
- Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou 510010, PR China
| | - Wei Zhu
- Department of Toxicology, Guangzhou Center for Disease Control and Prevention, Guangzhou 510515, PR China
| | - Wei-Wen Xu
- State Key Laboratory of Organ Failure Research, Institute of Antibody Engineering, Southern Medical University, Guangzhou 510515, PR China
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - Lei Su
- Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou 510010, PR China.
| |
Collapse
|
122
|
Akai S, Uematsu Y, Tsuneyama K, Oda S, Yokoi T. Kupffer cell-mediated exacerbation of methimazole-induced acute liver injury in rats. J Appl Toxicol 2015; 36:702-15. [DOI: 10.1002/jat.3202] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 05/18/2015] [Accepted: 05/29/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Sho Akai
- Department of Drug Safety Sciences, Division of Clinical Pharmacology; Nagoya University Graduate School of Medicine; 65 Tsurumai-cho Showa-ku Nagoya 466-8550 Japan
| | - Yasuaki Uematsu
- Department of Drug Safety Sciences, Division of Clinical Pharmacology; Nagoya University Graduate School of Medicine; 65 Tsurumai-cho Showa-ku Nagoya 466-8550 Japan
| | - Koichi Tsuneyama
- Department of Molecular and Environmental Pathology; Institute of Health Biosciences Tokushima University Graduate School; Kuramoto Tokushima 770-8503 Japan
| | - Shingo Oda
- Department of Drug Safety Sciences, Division of Clinical Pharmacology; Nagoya University Graduate School of Medicine; 65 Tsurumai-cho Showa-ku Nagoya 466-8550 Japan
| | - Tsuyoshi Yokoi
- Department of Drug Safety Sciences, Division of Clinical Pharmacology; Nagoya University Graduate School of Medicine; 65 Tsurumai-cho Showa-ku Nagoya 466-8550 Japan
| |
Collapse
|
123
|
Role of plasma high mobility group box-1 in disseminated intravascular coagulation with leukemia. Thromb Res 2015; 136:422-6. [PMID: 26049171 DOI: 10.1016/j.thromres.2015.05.030] [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] [Received: 02/05/2015] [Revised: 05/15/2015] [Accepted: 05/27/2015] [Indexed: 11/23/2022]
Abstract
INTRODUCTION High mobility group box 1(HMGB1) is a DNA-binding protein which can act as a proinflammatory cytokine when released by necrotic cells, monocytes or macrophages. It also plays a role in the coagulation activation and several tumors including leukemia. The objective of this study was to investigate the role of HMGB1 in the diagnosis of DIC with leukemia. METHODS 89 subjects with leukemia in Wuhan Union Hospital were prospectively recruited. Among them, 83 cases were suspected of DIC, while the other 6 were the negative controls. Their clinical data, laboratory tests and plasma samples were collected or measured respectively. Accordingly, we made scores for these subjects by the Japanese Ministry of Health and Welfare (JMHW) criteria. RESULTS This study demonstrated that the plasma levels of HMGB1 were higher in the DIC group than non-DIC (115.16 ng/ml vs. 63.94 ng/ml, p=0.003). The similar results were achieved in infected or non-infected groups. And along with the increase of DIC scores, the levels of HMGB1 increased gradually (p=0.006). In addition, HMGB1 was an independent factor in the diagnosis of DIC with leukemia(p<0.05). The diagnostic sensitivity of HMGB1 was high (Se=90.32%), and there was a tendency of increased HMGB1 levels in the pre-DIC patients. Three of these six pre-DIC patients were diagnosed as DIC by the new revised scoring system which contained HMGB1. Finally, the HMGB1 levels were significantly higher in patients with organ failures (SOFA≥2) than those without (118.76 vs. 72.75, p=0.032). CONCLUSION The increased plasma levels of HMGB1 were related tightly to the diagnosis and severity of DIC in leukemia patients. Furthermore, the diagnostic sensitivity of HMGB1 was high. So HMGB1 in plasma is a helpful molecular marker, and can be added in the scoring system for the early diagnosis of DIC with leukemia.
Collapse
|
124
|
Ishii K, Takeuchi H, Fukunaga K, Hirano Y, Suda K, Hagiwara T, Miyasho T, Yamada S, Nakamura R, Takahashi T, Wada N, Kawakubo H, Saikawa Y, Omori T, Betsuyaku T, Ichihara A, Kitagawa Y. Attenuation of lipopolysaccharide-induced acute lung injury after (pro)renin receptor blockade. Exp Lung Res 2015; 41:199-207. [PMID: 25844689 DOI: 10.3109/01902148.2014.993444] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE/AIM We performed a randomized, prospective animal study to investigate whether inhibiting the renin-angiotensin system with a (pro)renin receptor blocker (PRRB) prevents acute lung injury (ALI) in a rodent model. MATERIALS We used Thirty-six male Sprague-Dawley rats. We administered lipopolysaccharide (LPS; 2 mg/kg) intratracheally with or without PRRB pretreatment (1 mg/kg/d). METHODS We performed bronchoalveolar lavage (BAL) and lung removal at 4 h after LPS administration and measured levels of inflammatory cytokines, high mobility group box 1 (HMGB-1) protein, and total protein in bronchoalveolar lavage fluid (BALF). Myeloperoxidase (MPO) activity was detected in lung tissue homogenates using a sensitive ELISA. We performed hematoxylin and eosin staining and immunohistochemical staining for nonproteolytically activated prorenin in the left lung. RESULTS The PRRB decreased leukocyte counts and total protein, tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-2, IL-6, and IL-10 levels in the BALF and MPO activity in lung tissue. The PRRB reduced interstitial edema, hemorrhage, and the neutrophil count in the lung tissues. Consistent with the reduction in lung tissue damage, immunohistochemical staining showed that the PRRB decreased the amount of nonproteolytically activated prorenin. CONCLUSIONS The PRRB blocked LPS-induced inflammatory response in the lung and protected against ALI. Therefore, it is a potential therapeutic agent for preventing ALI.
Collapse
Affiliation(s)
- Kenjiro Ishii
- 1Department of Surgery, School of Medicine, Keio University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
125
|
Im KI, Kim N, Lim JY, Nam YS, Lee ES, Kim EJ, Kim HJ, Kim SH, Cho SG. The Free Radical Scavenger NecroX-7 Attenuates Acute Graft-versus-Host Disease via Reciprocal Regulation of Th1/Regulatory T Cells and Inhibition of HMGB1 Release. THE JOURNAL OF IMMUNOLOGY 2015; 194:5223-32. [PMID: 25911749 DOI: 10.4049/jimmunol.1402609] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 03/27/2015] [Indexed: 01/27/2023]
Abstract
Graft-versus-host disease (GVHD) is a major complication associated with allogeneic hematopoietic stem cell transplantation. Despite the prominent role of the adaptive immune system, the importance of controlling the innate immune system in the pathogenesis of GVHD has recently been rediscovered. High-mobility group box 1 (HMGB1) is a crucial damage-associated molecular pattern signal that functions as a potent innate immune mediator in GVHD. In the present study, we investigated treatment of experimental GVHD through HMGB1 blockade using the compound cyclopentylamino carboxymethylthiazolylindole (NecroX)-7. Treated animals significantly attenuated GVHD-related mortality and inhibited severe tissue damage. These protective effects correlated with the decrease in HMGB1 expression and lower levels of reactive oxidative stress. Additionally, NecroX-7 inhibited the HMGB1-induced release of TNF and IL-6, as well as the expression of TLR-4 and receptor for advanced glycation end products. We also observed increased regulatory T cell numbers, which may be associated with regulation of differentiation signals independent of HMGB1. Taken together, these data indicate that NecroX-7 protects mice against lethal GVHD by reciprocal regulation of regulatory T/Th1 cells, attenuating systemic HMGB1 accumulation and inhibiting HMGB1-mediated inflammatory response. Our results indicate the possibility of a new use for a clinical drug that is effective for the treatment of GVHD.
Collapse
Affiliation(s)
- Keon-Il Im
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Nayoun Kim
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Jung-Yeon Lim
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Young-Sun Nam
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Eun-Sol Lee
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Eun-Jung Kim
- Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Hyoung Jin Kim
- Strategy and Development, LG Life Sciences Ltd., Seoul 2305-738, Korea; and
| | - Soon Ha Kim
- Strategy and Development, LG Life Sciences Ltd., Seoul 2305-738, Korea; and
| | - Seok-Goo Cho
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea; Department of Hematology, Catholic Blood and Marrow Transplantation Center, Seoul St. Mary's Hospital, Catholic University of Korea College of Medicine, Seoul 137-701, Korea
| |
Collapse
|
126
|
Zhang S, Hwaiz R, Luo L, Herwald H, Thorlacius H. STAT3-dependent CXC chemokine formation and neutrophil migration in streptococcal M1 protein-induced acute lung inflammation. Am J Physiol Lung Cell Mol Physiol 2015; 308:L1159-67. [PMID: 25840996 DOI: 10.1152/ajplung.00324.2014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 04/01/2015] [Indexed: 01/05/2023] Open
Abstract
Streptococcus pyogenes cause infections ranging from mild pharyngitis to severe streptococcal toxic shock syndrome (STSS). The M1 serotype of Streptococcus pyogenes is most frequently associated with STSS. Herein, it was hypothesized that STAT3 signaling might be involved in M1 protein-evoked lung inflammation. The STAT3 inhibitor, S3I-201, was administered to male C57Bl/6 mice before iv challenge with M1 protein. Bronchoalveolar fluid and lung tissue were harvested for quantification of STAT3 activity, neutrophil recruitment, edema, and CXC chemokine formation. Neutrophil expression of Mac-1 was quantified by use of flow cytometry. Levels of IL-6 and HMGB1 were determined in plasma. CXCL2-induced neutrophil chemotaxis was studied in vitro. Administration of S3I-201 markedly reduced M1 protein-provoked STAT3 activity, neutrophil recruitment, edema formation, and inflammatory changes in the lung. In addition, M1 protein significantly increased Mac-1 expression on neutrophils and CXC chemokine levels in the lung. Treatment with S3I-201 had no effect on M1 protein-induced expression of Mac-1 on neutrophils. In contrast, inhibition of STAT3 activity greatly reduced M1 protein-induced formation of CXC chemokines in the lung. Interestingly, STAT3 inhibition markedly decreased plasma levels of IL-6 and HMGB1 in animals exposed to M1 protein. Moreover, we found that S3I-201 abolished CXCL2-induced neutrophil migration in vitro. In conclusion, these novel findings indicate that STAT3 signaling plays a key role in mediating CXC chemokine production and neutrophil infiltration in M1 protein-induced acute lung inflammation.
Collapse
Affiliation(s)
- Songen Zhang
- Department of Clinical Sciences, Section for Surgery, Lund University, Malmö, Sweden and
| | - Rundk Hwaiz
- Department of Clinical Sciences, Section for Surgery, Lund University, Malmö, Sweden and
| | - Lingtao Luo
- Department of Clinical Sciences, Section for Surgery, Lund University, Malmö, Sweden and
| | - Heiko Herwald
- Division of Infection Medicine, Lund University, Lund, Sweden
| | - Henrik Thorlacius
- Department of Clinical Sciences, Section for Surgery, Lund University, Malmö, Sweden and
| |
Collapse
|
127
|
Wang C, Wang H, Chang DY, Hao J, Zhao MH, Chen M. High mobility group box 1 contributes to anti-neutrophil cytoplasmic antibody-induced neutrophils activation through receptor for advanced glycation end products (RAGE) and Toll-like receptor 4. Arthritis Res Ther 2015; 17:64. [PMID: 25889374 PMCID: PMC4382936 DOI: 10.1186/s13075-015-0587-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 02/25/2015] [Indexed: 12/19/2022] Open
Abstract
Introduction High mobility group box-1 (HMGB1), a typical damage-associated molecular pattern (DAMP) protein, is associated with inflammatory conditions and tissue damage. Our recent study found that circulating HMGB1 levels could reflect the disease activity of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). The current study aimed to investigate whether HMGB1 participated in ANCA-induced neutrophil activation, which is one of the most important pathogenic aspects in the development of AAV. Methods The various effects of HMGB1 in ANCA-induced neutrophil activation were measured. Antagonists for relevant receptors and signaling molecules were employed. Results ANCA antigens translocation on neutrophils primed with HMGB1 was significantly higher than non-primed neutrophils. The levels of respiratory burst and degranulation increased significantly in HMGB1-primed neutrophils activated with ANCA-positive IgG, as compared with non-primed neutrophils. Furthermore, blocking Toll-like receptor 4 (TLR4) and receptor for advanced glycation end products (RAGE), rather than TLR2, resulted in a significant decrease in HMGB1-induced ANCA antigens translocation, respiratory burst and degranulation. Similar effects were also found when blocking MyD88 and NF-κB. Conclusions HMGB1 could prime neutrophils by increasing ANCA antigens translocation, and the primed neutrophils could be further induced by ANCA, resulting in the respiratory burst and degranulation. This process is TLR4- and RAGE-dependent through the MyD88/NF-κB pathway. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0587-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chen Wang
- Department of Medicine, Renal Division, Peking University First Hospital; Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, 8, Xishiku Street, Beijing, 100034, China.
| | - Huan Wang
- Department of Medicine, Renal Division, Peking University First Hospital; Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, 8, Xishiku Street, Beijing, 100034, China.
| | - Dong-Yuan Chang
- Department of Medicine, Renal Division, Peking University First Hospital; Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, 8, Xishiku Street, Beijing, 100034, China.
| | - Jian Hao
- Department of Medicine, Renal Division, Peking University First Hospital; Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, 8, Xishiku Street, Beijing, 100034, China.
| | - Ming-Hui Zhao
- Department of Medicine, Renal Division, Peking University First Hospital; Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, 8, Xishiku Street, Beijing, 100034, China.
| | - Min Chen
- Department of Medicine, Renal Division, Peking University First Hospital; Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, 8, Xishiku Street, Beijing, 100034, China.
| |
Collapse
|
128
|
Wang JL, Wu DW, Cheng ZZ, Han WZ, Xu SW, Sun NN. Expression of high mobility group box - B1 (HMGB-1) and matrix metalloproteinase-9 (MMP-9) in non-small cell lung cancer (NSCLC). Asian Pac J Cancer Prev 2015; 15:4865-9. [PMID: 24998555 DOI: 10.7314/apjcp.2014.15.12.4865] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE This study evaluated the expression level of high mobility group box-B1 (HMGB-1) and matrix metalloproteinase-9 (MMP-9) in non-small cell lung cancer (NSCLC) inmorder to reveal any relation with development and prognosis. METHODS NSCLC and normal tissues were selected from 30 patients at age of 30- 73, and used for RT-PCR and Western blot analyses of HMGB-1. A total of 100 paraffin embedded NSCLC tissues were also isolated from patients through surgical resection, and used for detection of HMGB-1 by immunohistochemistry. In addition, 50 samples were also applied for MMP-9 detection, and 30 normal tissues were considered as controls. Correlation analysis of HMGB-1 and MMP-9 was carried out by Pearsons correlation coefficient. RESULTS The average expression level of HMGB-1 in NSCLC patients was significantly higher than in normal lung tissues. In addition, patients in III-IV period exhibit significantly higher positive rate of HMGB- 1 when compared with I-II period cases. Furthermore, a positive correlation with HMGB-1 was found in the expression of MPP-9. CONCLUSION HMGB-1 was highly expressed in NSCLC, which may become a prognostic and predictive marker for NSCLC. Besides, MPP-9 was positively correlated with HMGB-1.
Collapse
Affiliation(s)
- Jing-Luan Wang
- Department of Critical Care Medicine, Qilu Hospital, Shandong University, Jinan, China E-mail :
| | | | | | | | | | | |
Collapse
|
129
|
Ma C, Wang Y, Dong L, Li M, Cai W. Anti-inflammatory effect of resveratrol through the suppression of NF-κB and JAK/STAT signaling pathways. Acta Biochim Biophys Sin (Shanghai) 2015; 47:207-13. [PMID: 25651848 DOI: 10.1093/abbs/gmu135] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Resveratrol, the most important ingredient extracted from Polygonum cuspidatum, exerts cytoprotective effects via anti-inflammatory actions in vitro. In this study, we investigated this effect of resveratrol on the lipopolysaccharide (LPS)-induced inflammatory response and its underlying molecular mechanism of action in RAW264.7 murine macrophages. Results showed that resveratrol down-regulated the expression of inducible nitric oxide synthase (iNOS) and interleukin-6 (IL-6), therefore, suppressed the production of nitric oxide and the secretion of IL-6 in LPS-stimulated RAW264.7 cells in a dose-dependent manner. Resveratrol also inhibited the translocation of high-mobility group box 1 (HMGB1) from the nucleus to the cytoplasm and of nuclear transcription factor kappa-B (NF-κB) p65 from the cytoplasm to the nucleus; it suppressed the phosphorylation of IκBα. Furthermore, these actions were mediated by suppressing the phosphorylation of signal transducer and activator of transcription (STAT)-1 and -3. In conclusion, these data indicate that resveratrol exerts anti-inflammatory effects, at least in part by reducing the release of HMGB1 and modulating the NF-κB and Janus kinase/STAT signaling pathways. Resveratrol could potentially be developed as a useful agent for the chemoprevention of inflammatory diseases.
Collapse
Affiliation(s)
- Chunfang Ma
- The Second Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou 310005, China
| | - Yin Wang
- The Second Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou 310005, China
| | - Lei Dong
- The Second Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou 310005, China
| | - Minjing Li
- The Second Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou 310005, China
| | - Wanru Cai
- The Second Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou 310005, China
| |
Collapse
|
130
|
Abe A, Kuwata T, Yamauchi C, Higuchi Y, Ochiai A. High Mobility Group Box1 (HMGB1) released from cancer cells induces the expression of pro-inflammatory cytokines in peritoneal fibroblasts. Pathol Int 2015; 64:267-75. [PMID: 24965109 DOI: 10.1111/pin.12167] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Accepted: 04/13/2014] [Indexed: 11/28/2022]
Abstract
High Mobility Group Box1 protein (HMGB1), one of the mediators of inflammation, is associated with tumorigenesis. The HMGB1-Receptor for advanced glycation end-products (RAGE) in gastric adenocarcinoma tissues promoted gastric cancer growth, however, there are no reports concerning the relationship between the expression of HMGB1 in gastric cancer and cancer-related inflammation. Fibroblasts exist most abundantly on cancer tissue where inflammation occurs. So, we studied the effects of HMGB1 released from cancer cells on the fibroblasts. The expression of HMGB1 in cancer cells and nuclear factor-kappa B (NF-kB) in fibroblasts were evaluated immunohistochemically in human gastric cancer specimens. Cytoplasmic HMGB1 expression in the cancer cells and nuclear translocation of NF-kB in fibroblasts were detected at deeper invasion. To determine whether HMGB1 released from cancer cells induces the expression of pro-inflammatory cytokines in fibroblasts, we analyzed the activation of Toll-like receptor (TLR) signaling. Fibroblasts stimulated by recombinant HMGB1 and the HSC44PE-conditioned medium showed the phosphorylation of Interleukin-1 receptor associated-kinase 4 (IRAK4), nuclear translocation of NF-kB, and enhanced pro-inflammatory cytokine expression. Treatment with HSC44PE-conditioned-medium transfected with siRNA-HMGB1 reduced the expressions of pro-inflammatory cytokines in the fibroblasts. We propose that HMGB1 released from cancer cells induces the expression of pro-inflammatory cytokines in peritoneal fibroblasts through the HMGB1-TLR2/4 pathway.
Collapse
Affiliation(s)
- Anna Abe
- Laboratory of Cancer Biology, Department of Integrated Bioscience, Graduate School of Frontier Science, The University of Tokyo; Pathology Division, Research Center for Innovative Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | | | | | | | | |
Collapse
|
131
|
Opatrna S, Popperlova A, Kalousová M, Zima T. Low glucose degradation product peritoneal dialysis regimen is associated with lower plasma EN-RAGE and HMGB-1 proinflammatory ligands of receptor for advanced glycation end products. Ther Apher Dial 2015; 18:309-16. [PMID: 24965297 DOI: 10.1111/1744-9987.12103] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Intraperitoneal glucose degradation products (GDP) load influences systemic advanced glycation end products (AGEs) but the effects on soluble receptor for AGEs (s-RAGE) and its proinflammatory ligands: extracellular newly identified receptor for advanced glycation end-products binding protein(EN-RAGE) and high mobility group box-1 protein (HMGB-1) are unknown. We aimed to compare plasma and peritoneal s-RAGE, EN-RAGE and HMGB-1 between three peritoneal dialysis (PD) prescription regimens with different intraperitoneal GDP loads. High GDP load (glucose-lactate PD fluid, D; N = 8) was compared with a low (glucose-bicarbonate/lactate with icodextrin for overnight dwell, E; N = 9) and a very low GDP load (glucose-bicarbonate/lactate, P; N = 16). D group demonstrated higher plasma EN-RAGE, 77.8 ng/mL, vs. both E, 11.2, P < 0.001 and P, 27.0, P < 0.001 as well as higher plasma HMGB-1, 2.2 ng/mL vs. both E, 1.1, P < 0.01 and P, 1.5, P < 0.01. Plasma s-RAGE, which did not differ between D, E and P, correlated with its effluent levels. Patients with faster peritoneal transport (D/Pcr > 0.65) tended to have higher plasma s-RAGE compared to slow transporters (2300 vs. 1762 pg/mL, P = 0.056). Peritoneal clearance of s-RAGE and EN-RAGE was higher with E compared to both D and P (P < 0.001 resp. P < 0.01). Subgroup of PD patients with CRP above median demonstrated higher plasma HMGB-1 and EN-RAGE, P < 0.05 for both. A lower intraperitoneal GDP load is associated with decreased plasma levels of EN-RAGE and HMGB-1. Peritoneal transport, microinflammation and the capability of icodextrin to increase peritoneal clearance of middle molecular weight substances might also exert an effect on plasma s-RAGE and its proinflammatory ligands levels.
Collapse
Affiliation(s)
- Sylvie Opatrna
- Medicine I, Charles University Medical School, Pilsen, Czech Republic
| | | | | | | |
Collapse
|
132
|
Gao R, Ma Z, Hu Y, Chen J, Shetty S, Fu J. Sirt1 restrains lung inflammasome activation in a murine model of sepsis. Am J Physiol Lung Cell Mol Physiol 2015; 308:L847-53. [PMID: 25659903 DOI: 10.1152/ajplung.00274.2014] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 02/02/2015] [Indexed: 02/07/2023] Open
Abstract
Excessive inflammation is a major cause of organ damage during sepsis. The elderly are highly susceptible to sepsis-induced organ injury. Sirt1 expression is reduced during aging. In the present study, we investigated the role of Sirt1, a histone deacetylase, in controlling inflammatory responses in a murine sepsis model induced by cecal ligation and puncture (CLP). We examined lung inflammatory signaling in inducible Sirt1 knockout (Sirt1(-/-)) mice and wild-type littermates (Sirt1(+/+)) after CLP. Our results demonstrated that Sirt1 deficiency led to severe lung inflammatory injury. To further investigate molecular mechanisms of Sirt1 regulation of lung inflammatory responses in sepsis, we conducted a series of experiments to assess lung inflammasome activation after CLP. We detected increased lung inflammatory signaling including NF-κB, signal transducer and activator of transcription 3, and ERK1/2 activation in Sirt1(-/-) mice after CLP. Furthermore, inflammasome activity was increased in Sirt1(-/-) mice after CLP, as demonstrated by increased IL-1β and caspase-7 cleavage and activation. Aggravated inflammasome activation in Sirt1(-/-) mice was associated with the increased production of lung proinflammatory mediators, including ICAM-1 and high-mobility group box 1, and further disruption of tight junctions and adherens junctions, as demonstrated by dramatic reduction of lung claudin-1 and vascular endothelial-cadherin expression, which was associated with the upregulation of matrix metallopeptidase 9 expression. In summary, our results suggest that Sirt1 suppresses acute lung inflammation during sepsis by controlling inflammasome activation pathway.
Collapse
Affiliation(s)
- Rong Gao
- Center for Research on Environmental Disease, University of Kentucky, Lexington, Kentucky; The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Zhongsen Ma
- The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Yuxin Hu
- Center for Research on Environmental Disease, University of Kentucky, Lexington, Kentucky; The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Jiao Chen
- Center for Research on Environmental Disease, University of Kentucky, Lexington, Kentucky
| | - Sreerama Shetty
- Center for Biomedical Research, University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Jian Fu
- Center for Research on Environmental Disease, University of Kentucky, Lexington, Kentucky; Graduate Center for Toxicology, College of Medicine, University of Kentucky, Lexington, Kentucky;
| |
Collapse
|
133
|
Hongo K, Kazama S, Tsuno NH, Ishihara S, Sunami E, Kitayama J, Watanabe T. Immunohistochemical detection of high-mobility group box 1 correlates with resistance of preoperative chemoradiotherapy for lower rectal cancer: a retrospective study. World J Surg Oncol 2015; 13:7. [PMID: 25622595 PMCID: PMC4417215 DOI: 10.1186/1477-7819-13-7] [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: 08/30/2014] [Accepted: 11/18/2014] [Indexed: 12/29/2022] Open
Abstract
Background High-mobility group box 1 (HMGB1) is a nucleoprotein that is related to inflammation. It has been implicated in a variety of biologically important processes, including transcription, DNA repair, differentiation, development, and extracellular signaling. Recently, its important role in the process of tumor invasion, metastasis, and resistance to anti-cancer therapies has been demonstrated. In this study, we aimed to investigate the correlation of HMGB1 expression and resistance of rectal cancer patients to chemoradiotherapy (CRT) prior to curative operation. Methods We retrospectively reviewed the data of 75 lower rectal cancer patients without complete pathological response who had received preoperative CRT and had undergone curative resection at the University of Tokyo Hospital between May 2003 and June 2010. HMGB1 expression in surgically resected specimens was evaluated using immunohistochemical detection and specimens were classified into high or low HMGB1 expression groups. Clinicopathologic features, degree of tumor reduction, regression of tumor grade, and patient survival were compared between the groups using non-paired Student’s t-tests and Kaplan-Meier analysis. Results A total of 52 (69.3%) patients had high HMGB1 expression, and 23 (30.7%) had low expression. HMGB1 expression was significantly correlated with histologic type (P = 0.02), lymphatic invasion (P = 0.02), and venous invasion (P = 0.05). Compared to patients with low HMGB1 expression, those with high expression had a poorer response to CRT, in terms of tumor reduction ratio (42.2 versus 28.9%, respectively; P <0.01) and post-CRT histological tumor regression grade (56.5 versus 30.8% grade 2; respectively; P = 0.03). However, no significant correlation was found between HMGB1 expression and recurrence-free and overall survival rates. Conclusions HMGB1 expression may be one of the key factors regulating the response of rectal cancer to preoperative CRT in terms of tumor invasiveness and resistance to therapy.
Collapse
Affiliation(s)
- Kumiko Hongo
- Department of Surgical Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Shinsuke Kazama
- Department of Surgical Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Nelson H Tsuno
- Department of Transfusion Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Soichiro Ishihara
- Department of Surgical Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Eiji Sunami
- Department of Surgical Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Joji Kitayama
- Department of Surgical Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Toshiaki Watanabe
- Department of Surgical Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
134
|
Zhang X, Luan Z, Liang Y, Liu Y, Ma X. Downregulation of high mobility group box 1 attenuates the severity of acute lung injury in endotoxemic mice. Mol Med Rep 2015; 11:4513-7. [PMID: 25625772 DOI: 10.3892/mmr.2015.3251] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 01/07/2015] [Indexed: 11/06/2022] Open
Abstract
High mobility group box 1 (HMGB1) is a systemic inflammation‑associated cytokine mediator. The aim of the present study was to examine the effect of the downregulation of HMGB1 in a lipopolysaccharide (LPS)‑induced mouse model of acute lung injury (ALI). It was identified that serum levels of tumor necrosis factor‑α and interleukin‑1β, lung myeloperoxidase activity and malondialdehyde content, as well as lung wet/dry weight ratios were all increased following LPS challenge. However, LPS‑mediated increases in these parameters were significantly downregulated in HMGB1 small interfering (si)RNA‑treated mice versus the negative control siRNA‑treated mice. In addition, the administration of HMGB1 siRNA in LPS‑treated mice resulted in a decreased DNA binding activity of nuclear factor‑κB (NF‑κB) in the lung. It was demonstrated that downregulation of HMGB1 decreases inflammation and the severity of sepsis associated with ALI, possibly via inhibiting the NF‑κB DNA‑binding activity. The present data support HMGB1 as a contributor to the pathogenesis of LPS‑induced sepsis and ALI.
Collapse
Affiliation(s)
- Xiaojuan Zhang
- Intensive Care Unit, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zhenggang Luan
- Intensive Care Unit, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yingjian Liang
- Intensive Care Unit, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yina Liu
- Intensive Care Unit, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xiaochun Ma
- Intensive Care Unit, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
135
|
Abstract
Danger-associated molecular patterns (DAMPs) are nuclear or cytoplasmic proteins that are released from the injured tissues and activate the innate immune system. Mitochondrial DNA (mtDNA) is a novel DAMP that is released into the extracellular milieu subsequent to cell death and injury. We hypothesized that cell death within the central nervous system in children with traumatic brain injury (TBI) would lead to the release of mtDNA into the cerebrospinal fluid (CSF) and has the potential to predict the outcome after trauma. Cerebrospinal fluid was collected from children with severe TBI who required intracranial pressure monitoring with Glasgow Coma Scale (GCS) scores of 8 or less via an externalized ventricular drain. Control CSF was obtained in children without TBI or meningoencephalitis who demonstrated no leukocytes in the diagnostic lumbar puncture. The median age for patients with TBI was 6.3 years, and 62% were male. The common mechanisms of injury included motor vehicle collision (35.8%), followed by falls (21.5%) and inflicted TBI (19%); six children (14.2%) died during their intensive care unit course. The mean CSF mtDNA concentration was 1.10E+05 ± 2.07E+05 and 1.63E+03 ± 1.80E+03 copies/μL in the pediatric TBI and control populations, respectively. Furthermore, the mean CSF mtDNA concentration in pediatric patients who later died or had severe disability was significantly higher than that of the survivors (1.63E+05 ± 2.77E+05 vs. 5.05E+04 ± 6.21E+04 copies/μL) (P < 0.0001). We found a significant correlation between CSF mtDNA and high mobility group box 1, another prototypical DAMP, concentrations (ρ = 0.574, P < 0.05), supporting the notion that both DAMPs are increased in the CSF after TBI. Our data suggest that CSF mtDNA is a novel DAMP in TBI and appears to be a useful biomarker that correlates with neurological outcome after TBI. Further inquiry into the components of mtDNA that modulate the innate immune response will be helpful in understanding the mechanism of local and systemic inflammation after TBI.
Collapse
|
136
|
Bobek D, Grčević D, Kovačić N, Lukić IK, Jelušić M. The presence of high mobility group box-1 and soluble receptor for advanced glycation end-products in juvenile idiopathic arthritis and juvenile systemic lupus erythematosus. Pediatr Rheumatol Online J 2014; 12:50. [PMID: 25516724 PMCID: PMC4267139 DOI: 10.1186/1546-0096-12-50] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 11/05/2014] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The involvement of high mobility group box-1 (HMGB1) in various inflammatory and autoimmune diseases has been documented but clinical trials on the contribution of this pro-inflammatory alarmin in children with juvenile idiopathic arthritis (JIA) and systemic lupus erythematosus (SLE) are basically absent. To address the presence of HMGB1 and a soluble receptor for advanced glycation end products (sRAGE) in different subtypes of JIA and additionally in children with SLE, we enrolled a consecutive sample of children harvested peripheral blood as well as synovial fluids (SF) at diagnosis and correlated it with ordinary acute-phase reactants and clinical markers. METHODS Serum and synovial fluids levels of HMGB1 and sRAGE in total of 144 children (97 with JIA, 19 with SLE and 27 healthy controls) were determined by ELISA. RESULTS The children with JIA and those with SLE were characterised by significantly higher serum levels of HMGB1 and significantly lower sRAGE levels compared to the healthy controls. A positive correlation between serum HMGB1 and ESR, CRP, α2 globulin was found while serum sRAGE levels were inversely correlated with the same inflammatory markers in children with JIA. Additionally, high level of serum HMGB1 was related to hepatosplenomegaly or serositis in systemic onset JIA. CONCLUSION The inverse relationship of the HMGB1 and its soluble receptor RAGE in the blood and SF indicates that inflammation triggered by alarmins may play a role in pathogenesis of JIA as well as SLE. HMGB1 may serve as an inflammatory marker and a potential target of biological therapy in these patients. Further studies need to show whether the determination of HMGB1 levels in patients with JIA can be a useful guideline for detecting disease activity.
Collapse
Affiliation(s)
- Dubravka Bobek
- Department of Pediatrics, Division of Pediatric Rheumatology and Immunology, University Hospital Centre Zagreb, University of Zagreb School of Medicine, Kišpatićeva 12, 10000 Zagreb, Croatia
| | - Danka Grčević
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Nataša Kovačić
- Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ivan Krešimir Lukić
- Department of Research in Biomedicine, and Health, University of Split School of Medicine, Split, Croatia
| | - Marija Jelušić
- Department of Pediatrics, Division of Pediatric Rheumatology and Immunology, University Hospital Centre Zagreb, University of Zagreb School of Medicine, Kišpatićeva 12, 10000 Zagreb, Croatia
| |
Collapse
|
137
|
Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, Huang J, Yu Y, Fan XG, Yan Z, Sun X, Wang H, Wang Q, Tsung A, Billiar TR, Zeh HJ, Lotze MT, Tang D. HMGB1 in health and disease. Mol Aspects Med 2014; 40:1-116. [PMID: 25010388 PMCID: PMC4254084 DOI: 10.1016/j.mam.2014.05.001] [Citation(s) in RCA: 731] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/05/2014] [Indexed: 12/22/2022]
Abstract
Complex genetic and physiological variations as well as environmental factors that drive emergence of chromosomal instability, development of unscheduled cell death, skewed differentiation, and altered metabolism are central to the pathogenesis of human diseases and disorders. Understanding the molecular bases for these processes is important for the development of new diagnostic biomarkers, and for identifying new therapeutic targets. In 1973, a group of non-histone nuclear proteins with high electrophoretic mobility was discovered and termed high-mobility group (HMG) proteins. The HMG proteins include three superfamilies termed HMGB, HMGN, and HMGA. High-mobility group box 1 (HMGB1), the most abundant and well-studied HMG protein, senses and coordinates the cellular stress response and plays a critical role not only inside of the cell as a DNA chaperone, chromosome guardian, autophagy sustainer, and protector from apoptotic cell death, but also outside the cell as the prototypic damage associated molecular pattern molecule (DAMP). This DAMP, in conjunction with other factors, thus has cytokine, chemokine, and growth factor activity, orchestrating the inflammatory and immune response. All of these characteristics make HMGB1 a critical molecular target in multiple human diseases including infectious diseases, ischemia, immune disorders, neurodegenerative diseases, metabolic disorders, and cancer. Indeed, a number of emergent strategies have been used to inhibit HMGB1 expression, release, and activity in vitro and in vivo. These include antibodies, peptide inhibitors, RNAi, anti-coagulants, endogenous hormones, various chemical compounds, HMGB1-receptor and signaling pathway inhibition, artificial DNAs, physical strategies including vagus nerve stimulation and other surgical approaches. Future work further investigating the details of HMGB1 localization, structure, post-translational modification, and identification of additional partners will undoubtedly uncover additional secrets regarding HMGB1's multiple functions.
Collapse
Affiliation(s)
- Rui Kang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| | - Ruochan Chen
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Qiuhong Zhang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Wen Hou
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Sha Wu
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Lizhi Cao
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jin Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yan Yu
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xue-Gong Fan
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhengwen Yan
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA; Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Xiaofang Sun
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Experimental Department of Institute of Gynecology and Obstetrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510510, China
| | - Haichao Wang
- Laboratory of Emergency Medicine, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - Qingde Wang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Allan Tsung
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Herbert J Zeh
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Daolin Tang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| |
Collapse
|
138
|
Yang PS, Kim DH, Lee YJ, Lee SE, Kang WJ, Chang HJ, Shin JS. Glycyrrhizin, inhibitor of high mobility group box-1, attenuates monocrotaline-induced pulmonary hypertension and vascular remodeling in rats. Respir Res 2014; 15:148. [PMID: 25420924 PMCID: PMC4248446 DOI: 10.1186/s12931-014-0148-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 11/10/2014] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND High mobility group box-1 (HMGB1), a proinflammatory cytokine, plays a pivotal role in tissue remodeling and angiogenesis, both of which are crucial for the pathogenesis of pulmonary arterial hypertension. In this study, we explored the relationship between HMGB1 and pulmonary hypertension and whether glycyrrhizin, an inhibitor of HMGB1, attenuates disease progression in an animal model of pulmonary hypertension induced by monocrotaline sodium (MCT). METHODS After inducing pulmonary hypertension through a single subcutaneous injection of MCT (60 mg/kg) to Sprague-Dawley rats, we administered daily intraperitoneal injections of either glycyrrhizin (GLY, 50 mg/kg), an inhibitor of HMGB1, or saline (control) for either 4 or 6 weeks. RESULTS Expression levels of HMGB1 in serum increased from the second week after MCT injection and remained elevated throughout the experiment periods. Lung tissue levels of HMGB1 assessed by immunohistochemical staining at 4 weeks after MCT injection also increased. Chronic inhibition of HMGB1 by GLY treatment reduced the MCT-induced increase in right ventricular (RV) systolic pressure, RV hypertrophy (ratio of RV to [left ventricle + septum]), and pulmonary inflammation. MCT-induced muscularization of the pulmonary artery was also attenuated in the GLY-treated group. As assessed 6 weeks after MCT injection, the GLY-treated group exhibited increased survival (90% [18 of 20]) when compared with the control group (60% [12 of 20]; p =0.0027). CONCLUSIONS Glycyrrhizin, an inhibitor of HMGB1, attenuates pulmonary hypertension progression and pulmonary vascular remodeling in the MCT-induced pulmonary hypertension rat model. Further studies are needed to confirm the potential of HMGB1 as a novel therapeutic target for pulmonary hypertension.
Collapse
Affiliation(s)
- Pil-Sung Yang
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University Health System, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea.
| | - Dae-Hoon Kim
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University Health System, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea.
| | - Yong Joon Lee
- Departments of Microbiology, Yonsei University College of Medicine, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea.
| | - Sang-Eun Lee
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University Health System, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea.
| | - Won Jun Kang
- Departments of Nuclear Medicine, Yonsei University College of Medicine, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea.
| | - Hyuk-Jae Chang
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University Health System, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea. .,Severance Biomedical Science Institute, Yonsei University Health System, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea.
| | - Jeon-Soo Shin
- Departments of Microbiology, Yonsei University College of Medicine, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea. .,Severance Biomedical Science Institute, Yonsei University Health System, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea.
| |
Collapse
|
139
|
Yu SL, Wong CK, Szeto CC, Li EK, Cai Z, Tam LS. Members of the receptor for advanced glycation end products axis as potential therapeutic targets in patients with lupus nephritis. Lupus 2014; 24:675-86. [PMID: 25411258 DOI: 10.1177/0961203314559631] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 10/20/2014] [Indexed: 12/22/2022]
Abstract
The relationship of inflammation and the expression of full-length receptor for advanced glycation end products (flRAGE) on monocytes, plasma levels of RAGE ligand high mobility group box protein 1 (HMGB1), soluble RAGE (sRAGE) and endogenous secretory RAGE (esRAGE) was assessed to elucidate the effect of HMGB1/DNA/RAGE-mediated innate inflammatory responses in patients with lupus nephritis. Cell surface expression of flRAGE was elevated on the monocytes of lupus patients, correlated with plasma HMGB1 levels. Plasma sRAGE level negatively correlated with systemic lupus erythematosus (SLE) disease activity index. Plasma esRAGE level was significantly lower in SLE patients with flare while esRAGE/sRAGE ratio negatively correlated with complement C3 level. HMGB1 alone could moderately induce ex vivo IL-6 production from monocytes, resulting in activation of intracellular p38 mitogen-activated protein kinase (MAPK), c-Jun N-terminal kinase and nuclear factor (NF)-κB. Moreover, toll-like receptor-9 ligand together with HMGB1 exhibited a synergistic effect on IL-6 and IL-12p70 secretions and the phosphorylation of p38 MAPK and NF-κB. Therefore, over-expression of flRAGE in lupus may lead to the amplification of RAGE ligands-mediated inflammatory responses through the activation of p38 MAPK and NF-κB. Plasma sRAGE may serve as a potential biomarker for disease activity and a future therapeutic target in SLE.
Collapse
Affiliation(s)
- S L Yu
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China Department of Rheumatology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University (GZMU), Guangdong, China
| | - C K Wong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong Institute of Chinese Medicine and State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Hong Kong Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - C C Szeto
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - E K Li
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Z Cai
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - L S Tam
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| |
Collapse
|
140
|
Hirose T, Hamaguchi S, Matsumoto N, Irisawa T, Seki M, Tasaki O, Hosotsubo H, Yamamoto N, Yamamoto K, Akeda Y, Oishi K, Tomono K, Shimazu T. Presence of neutrophil extracellular traps and citrullinated histone H3 in the bloodstream of critically ill patients. PLoS One 2014; 9:e111755. [PMID: 25392950 PMCID: PMC4230949 DOI: 10.1371/journal.pone.0111755] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 09/30/2014] [Indexed: 12/30/2022] Open
Abstract
Neutrophil extracellular traps (NETs), a newly identified immune mechanism, are induced by inflammatory stimuli. Modification by citrullination of histone H3 is thought to be involved in the in vitro formation of NETs. The purposes of this study were to evaluate whether NETs and citrullinated histone H3 (Cit-H3) are present in the bloodstream of critically ill patients and to identify correlations with clinical and biological parameters. Blood samples were collected from intubated patients at the time of ICU admission from April to June 2011. To identify NETs, DNA and histone H3 were visualized simultaneously by immunofluorescence in blood smears. Cit-H3 was detected using a specific antibody. We assessed relationships of the presence of NETs and Cit-H3 with the existence of bacteria in tracheal aspirate, SIRS, diagnosis, WBC count, and concentrations of IL-8, TNF-α, cf-DNA, lactate, and HMGB1. Forty-nine patients were included. The median of age was 66.0 (IQR: 52.5-76.0) years. The diagnoses included trauma (7, 14.3%), infection (14, 28.6%), resuscitation from cardiopulmonary arrest (8, 16.3%), acute poisoning (4, 8.1%), heart disease (4, 8.1%), brain stroke (8, 16.3%), heat stroke (2, 4.1%), and others (2, 4.1%). We identified NETs in 5 patients and Cit-H3 in 11 patients. NETs and/or Cit-H3 were observed more frequently in "the presence of bacteria in tracheal aspirate" group (11/22, 50.0%) than in "the absence of bacteria in tracheal aspirate" group (4/27, 14.8%) (p<.01). Multiple logistic regression analysis showed that only the presence of bacteria in tracheal aspirate was significantly associated with the presence of NETs and/or Cit-H3. The presence of bacteria in tracheal aspirate may be one important factor associated with NET formation. NETs may play a pivotal role in the biological defense against the dissemination of pathogens from the respiratory tract to the bloodstream in potentially infected patients.
Collapse
Affiliation(s)
- Tomoya Hirose
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- * E-mail:
| | - Shigeto Hamaguchi
- Division of Infection Control and Prevention, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Naoya Matsumoto
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Taro Irisawa
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masafumi Seki
- Division of Infection Control and Prevention, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Osamu Tasaki
- Department of Emergency Medicine, Unit of Clinical Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hideo Hosotsubo
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Norihisa Yamamoto
- Division of Infection Control and Prevention, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kouji Yamamoto
- Department of Medical Innovation, Osaka University Hospital, Osaka, Japan
| | - Yukihiro Akeda
- International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Kazunori Oishi
- International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Kazunori Tomono
- Division of Infection Control and Prevention, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takeshi Shimazu
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
141
|
Distinctive colonic mucosal cytokine signature in new-onset, untreated pediatric Crohn disease. J Pediatr Gastroenterol Nutr 2014; 59:553-61. [PMID: 25000355 DOI: 10.1097/mpg.0000000000000480] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVE The aim of the study was to compare the colonic mucosal immune response in children with new, untreated Crohn disease (CD-New), CD in remission (CD-Remission), and unaffected children (CTRL [controls]). METHODS We performed flow cytometry of mitogen-stimulated colonic lamina propria mononuclear cells isolated from colonic biopsies and 72-hour biopsy explant cultures, and analyzed the supernatant by an unbiased multiplex cytokine array of 45 analytes. RESULTS Thirty-six children were studied (mean age 14 ± 3 years, 14 girls): 12 CD-New, 11 CD-Remission, and 13 CTRL. We found that stimulation of lamina propria mononuclear cells isolated from colonic biopsies induced comparable intracellular cytokine levels of interferon (IFN-γ), interleukin (IL)-17, and tumor necrosis factor (TNF)-α in T cells from CD-New, CD-Remission, and CTRL, suggesting that mucosal innate inflammation plays a larger role than activated T cells in CD-New. To measure factors released during the ongoing inflammatory response in CD-New, we cultured colonic biopsy explants and uncovered 13/45 factors that were significantly higher in CD-New versus CD-Remission, whereas 10 were increased in CD-New over CTRL. Ingenuity Pathway Analysis software revealed the anticipated interconnectivity of TNF-α, IL-6, and CSF-2 in CD-New of the colon. A novel subnetwork of chemokines was, however, evident, whereas IL-17a appeared as a peripheral factor. Principal component analysis and hierarchal clustering showed that CD-New and CD-Remission separated into distinct subgroups based on the 13 factors. CONCLUSIONS At diagnosis of inflammatory bowel disease, the colonic cytokine response contains a predominance of innate immune factors, with chemoattractants and vascular adhesion molecules playing a central role.
Collapse
|
142
|
Paris G, Pozharskaya T, Asempa T, Lane AP. Damage-associated molecular patterns stimulate interleukin-33 expression in nasal polyp epithelial cells. Int Forum Allergy Rhinol 2014; 4:15-21. [PMID: 24574111 DOI: 10.1002/alr.21237] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 07/18/2013] [Accepted: 08/07/2013] [Indexed: 02/02/2023]
Abstract
BACKGROUND Chronic rhinosinusitis with nasal polyps (CRSwNP) is a disorder characterized by eosinophilic inflammation and local T-helper 2 (Th2) cytokine production. Innate lymphoid cells that elaborate Th2 cytokines have recently been characterized within nasal polyps. These cells can be activated by the epithelial cell-derived cytokine interleukin-33 (IL-33). The objective of this study is to determine whether 2 molecules associated with tissue damage (high mobility group box-1 [HMGB-1] and adenosine triphosphate [ATP]) elicit expression of IL-33 in sinonasal epithelial cells (SNECs) derived from recalcitrant CRSwNP patients. METHODS Ethmoid tissue was obtained from 8 recalcitrant CRSwNP and 9 control subjects during endoscopic sinus surgery (ESS). Tissue was prepared for immunohistochemistry and for SNEC air-liquid interface culture. After exposure to either HMGB1 or ATP in vitro, SNECs were processed for messenger RNA (mRNA) extraction and immunocytochemistry. IL-33 levels were determined by real-time polymerase chain reaction (PCR) and by immunochemical staining with anti-IL-33 antibody. RESULTS Intranuclear IL-33 is normally expressed in basal epithelial cells, but is present in more apical cells and outside the nucleus in CRSwNP. Exposure of SNECs to HMGB-1 or ATP resulted in a statistically significant increase in IL-33 mRNA expression in SNECs derived from recalcitrant CRSwNP patients. This increase was reflected at the protein level by immunochemical staining of IL-33. CONCLUSION Tissue damage is a nonspecific trigger of epithelial IL-33 production in treatment-recalcitrant polyps, which may be responsible for perpetuating eosinophilic inflammation in CRSwNP. This common pathway may help explain why multiple environmental and infectious agents have been implicated in CRSwNP exacerbation.
Collapse
|
143
|
Lee W, Ku SK, Jeong TC, Lee S, Bae JS. Ginsenosides Inhibit HMGB1-induced Inflammatory Responses in HUVECs and in Murine Polymicrobial Sepsis. B KOREAN CHEM SOC 2014. [DOI: 10.5012/bkcs.2014.35.10.2955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
144
|
High-mobility group box 1 (HMGB1) impaired cardiac excitation–contraction coupling by enhancing the sarcoplasmic reticulum (SR) Ca2+ leak through TLR4–ROS signaling in cardiomyocytes. J Mol Cell Cardiol 2014; 74:260-73. [DOI: 10.1016/j.yjmcc.2014.06.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 05/04/2014] [Accepted: 06/08/2014] [Indexed: 10/25/2022]
|
145
|
Xu H, Li H, Qu Y, Zheng J, Lu J. High mobility group box 1 release from cholangiocytes in patients with acute-on-chronic liver failure. Exp Ther Med 2014; 8:1178-1184. [PMID: 25187820 PMCID: PMC4151661 DOI: 10.3892/etm.2014.1904] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 07/29/2014] [Indexed: 12/29/2022] Open
Abstract
High mobility group box chromosomal protein 1 (HMGB1) is an important proinflammatory molecule in a number of inflammatory disorders, but little is known about its role in acute-on-chronic liver failure (ACLF). To elucidate the role of HMGB1 in ACLF, the expression of HMGB1 in liver specimens from patients with ACLF was investigated. Immunohistochemical staining was performed to confirm the expression and subcellular localization of HMGB1 in liver specimens obtained from 13 patients with ACLF caused by hepatitis B virus (HBV) infection, 20 patients with chronic viral hepatitis B and 20 healthy controls. In addition, TFK-1 cells (human cholangiocarcinoma cell line) were stimulated with lipopolysaccharide (LPS) or tumor necrosis factor (TNF)-α. The extracellular level of HMGB1 in the culture medium was then determined by ELISA, and cell viability was also examined. In patients with ACLF caused by HBV infection, HMGB1 was found mainly in the cholangiocytes, and cytoplasmic translocation was observed in the cholangiocytes in the liver specimens. In the TFK-1 cell cultures, HMGB1 levels gradually increased from as early as 4 h after stimulation with LPS or TNF-α until the end of the stimulation. LPS and TNF-α actively induced the cytoplasmic translocation of the HMGB1 protein in TFK-1 cells. These data suggest that HMGB1 plays a critical role in the systemic inflammation associated with ACLF.
Collapse
Affiliation(s)
- Heng Xu
- Hepatology and Cancer Biotherapy Ward, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China ; Department of Pathophysiology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Hongxia Li
- Hepatology and Cancer Biotherapy Ward, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Yachao Qu
- Hepatology and Cancer Biotherapy Ward, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Junfu Zheng
- Hepatology and Cancer Biotherapy Ward, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Jun Lu
- Hepatology and Cancer Biotherapy Ward, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| |
Collapse
|
146
|
|
147
|
Sadamura-Takenaka Y, Ito T, Noma S, Oyama Y, Yamada S, Kawahara KI, Inoue H, Maruyama I. HMGB1 promotes the development of pulmonary arterial hypertension in rats. PLoS One 2014; 9:e102482. [PMID: 25032709 PMCID: PMC4102514 DOI: 10.1371/journal.pone.0102482] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 06/19/2014] [Indexed: 11/18/2022] Open
Abstract
Rationale Pulmonary arterial hypertension (PAH) is characterized by increased pulmonary vascular resistance leading to right ventricular failure and death. Recent studies have suggested that chronic inflammatory processes are involved in the pathogenesis of PAH. However, the molecular and cellular mechanisms driving inflammation have not been fully elucidated. Objectives To elucidate the roles of high mobility group box 1 protein (HMGB1), a ubiquitous DNA-binding protein with extracellular pro-inflammatory activity, in a rat model of PAH. Methods Male Sprague-Dawley rats were administered monocrotaline (MCT). Concentrations of HMGB1 in bronchoalveolar lavage fluid (BALF) and serum, and localization of HMGB1 in the lung were examined over time. The protective effects of anti-HMGB1 neutralizing antibody against MCT-induced PAH were tested. Results HMGB1 levels in BALF were elevated 1 week after MCT injection, and this elevation preceded increases of other pro-inflammatory cytokines, such as TNF-α, and the development of PAH. In contrast, serum HMGB1 levels were elevated 4 weeks after MCT injection, at which time the rats began to die. Immunohistochemical analyses indicated that HMGB1 was translocated to the extranuclear space in periarterial infiltrating cells, alveolar macrophages, and bronchial epithelial cells of MCT-injected rats. Anti-HMGB1 neutralizing antibody protected rats against MCT-induced lung inflammation, thickening of the pulmonary artery wall, and elevation of right ventricular systolic pressure, and significantly improved the survival of the MCT-induced PAH rats. Conclusions Our results identify extracellular HMGB1 as a promoting factor for MCT-induced PAH. The blockade of HMGB1 activity improved survival of MCT-induced PAH rats, and thus might be a promising therapy for the treatment of PAH.
Collapse
Affiliation(s)
- Yukari Sadamura-Takenaka
- Department of Pulmonary Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takashi Ito
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Satoshi Noma
- Department of Pulmonary Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yoko Oyama
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | | | - Ko-ichi Kawahara
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka, Japan
| | - Hiromasa Inoue
- Department of Pulmonary Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Ikuro Maruyama
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
- * E-mail:
| |
Collapse
|
148
|
Jessop F, Holian A. Extracellular HMGB1 regulates multi-walled carbon nanotube-induced inflammation in vivo. Nanotoxicology 2014; 9:365-72. [PMID: 24983895 DOI: 10.3109/17435390.2014.933904] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Endotoxin is often used to activate NF-κB in vitro when assessing NLRP3 inflammasome activation by various exogenous particles including nanoparticles. However, the endogenous source of this signal 1 is unknown. High-mobility group box 1 (HMGB1) is known to play a critical role in acute lung injury, however the potential contribution of the alarmin HMGB1 to NLRP3 Inflammasome activation has not been determined in response to nanoparticles in vivo. In this study, the ability of multi-walled carbon nanotubes (MWCNT) to cause release of HMGB1 in vitro and in vivo, as well as the potential of HMGB1 to function as signal 1 in vitro and in vivo, was determined. HMGB1 activity in vivo was assessed by administration of HMGB1 neutralization antibodies following MWCNT exposure. Caspase-1(-/-) mice were utilized to elucidate the dependence of HMGB1 secretion on NLRP3 inflammasome activity. MWCNT exposure increased extracellular HMGB1 levels in primary alveolar macrophages from C57Bl/6 mice and C10 mouse epithelial cell culture supernatants, and in C57Bl/6 mouse lung lavage fluid. MWCNT-induced HMGB1 secretion was dependent upon caspase-1. HMGB1 increased MWCNT-induced IL-1β release from macrophages in vitro, and neutralization of extracellular HMGB1 reduced MWCNT-induced IL-1β secretion in vivo. HMGB1 neutralization was accompanied with overall decreased inflammation. In summary, this study suggests extracellular HMGB1 participates in NLRP3 inflammasome activity and regulates IL-1β associated sterile inflammation induced by MWCNT.
Collapse
Affiliation(s)
- Forrest Jessop
- Center for Environmental Health Sciences, University of Montana , Missoula, MT , USA
| | | |
Collapse
|
149
|
Wang C, Gou SJ, Chang DY, Yu F, Zhao MH, Chen M. Association of circulating level of high mobility group box 1 with disease activity in antineutrophil cytoplasmic autoantibody-associated vasculitis. Arthritis Care Res (Hoboken) 2014; 65:1828-34. [PMID: 24591411 DOI: 10.1002/acr.22187] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Accepted: 05/23/2013] [Indexed: 11/08/2022]
Abstract
OBJECTIVE High mobility group box 1 (HMGB-1), a kind of proinflammatory mediator, is associated with inflammatory conditions and tissue damage. Previous studies have reported that circulating HMGB-1 levels in patients with active antineutrophil cytoplasmic autoantibody (ANCA)-associated vasculitis (AAV) were associated with renal manifestations and burdens of granulomatous inflammation. The current study aimed to investigate whether circulating HMGB-1 levels were associated with disease activity in AAV. METHODS Plasma samples from 74 patients with AAV in active stage and 65 patients with AAV in remission were collected. The plasma levels of HMGB-1 were determined by enzyme-linked immunosorbent assay. Associations between plasma levels of HMGB-1 with clinical and pathologic parameters were analyzed. RESULTS Plasma levels of HMGB-1 in active AAV patients were significantly higher than those in normal controls and AAV patients in remission (median 6.11 [interquartile range (IQR) 3.25-12.79] ng/ml versus median 1.12 [IQR 0.53-1.39] ng/ml, P< 0.001; median 6.11 [IQR 3.25-12.79] ng/ml versus median 3.04 [IQR 1.97-4.63] ng/ml, P < 0.001, respectively). Correlation analysis showed that plasma levels of HMGB-1 correlated with initial serum creatinine (r = 0.275, P = 0.018), estimated glomerular filtration rate (r = -0.277, P = 0.017), the Birmingham Vasculitis Activity Score (r = 0.308, P = 0.008), and C-reactive protein level (r = 0.309, P = 0.008). Among the patients with myeloperoxidase (MPO)-ANCA, those within the first quartile of plasma HMGB-1 levels had a significantly lower level of MPO-ANCA than those within the other 3 quartiles. CONCLUSION Circulating HMGB-1 levels might reflect the disease activity and renal involvement of AAV vasculitis.
Collapse
Affiliation(s)
- Chen Wang
- Peking University First Hospital, Beijing, China
| | | | | | | | | | | |
Collapse
|
150
|
Abstract
Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain receptors (NLRs) are families of pattern recognition receptors that, together with inflammasomes, sense and respond to highly conserved pathogen motifs and endogenous molecules released upon cell damage or stress. Evidence suggests that TLRs, NLRs and the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome have important roles in kidney diseases through regulation of inflammatory and tissue-repair responses to infection and injury. In this Review, we discuss the pathological mechanisms that are related to TLRs, NLRs and NLRP3 in various kidney diseases. In general, these receptors are protective in the host defence against urinary tract infection, but can sustain and self-perpetuate tissue damage in sterile inflammatory and immune-mediated kidney diseases. TLRs, NLRs and NLRP3, therefore, have become promising drug targets to enable specific modulation of kidney inflammation and suppression of immunopathology in kidney disease.
Collapse
|