301
|
Wang Q, van Hoecke M, Tang X, Lee H, Zheng Z, Swanson RA, Yenari MA. Pyruvate protects against experimental stroke via an anti-inflammatory mechanism. Neurobiol Dis 2009; 36:223-31. [PMID: 19635562 PMCID: PMC2742567 DOI: 10.1016/j.nbd.2009.07.018] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Revised: 07/16/2009] [Accepted: 07/17/2009] [Indexed: 01/09/2023] Open
Abstract
Pyruvate, a key intermediate in glucose metabolism, was explored as a potential treatment in models of experimental stroke and inflammation. Pyruvate was administered to rodents after the onset of middle cerebral artery occlusion (MCAO). Since the extent of inflammation is often proportional to the size of the infarct, we also studied a group of animals given lipopolysaccharide (LPS) to cause brain inflammation without cell death. Following MCAO, pyruvate did not affect physiological parameters but significantly reduced infarct volume, improved behavioral tests and reduced numbers of neutrophils, microglial and NFkappaB activation. Animals given LPS showed increased microglial and NFkappaB activation which was almost completely abolished by pyruvate. Lactate, a major metabolite of pyruvate, was increased after pyruvate administration. However, administration of lactate itself did not have any anti-inflammatory effects. Pyruvate protects against ischemia possibly by blocking inflammation, but lactate itself does not appear to explain pyruvate's anti-inflammatory properties.
Collapse
Affiliation(s)
- Qing Wang
- Department of Neurology, University of California, San Francisco & the San Francisco Veterans Affairs Medical Center, California 94121, USA
- Dept. Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Michael van Hoecke
- Department of Neurology, University of California, San Francisco & the San Francisco Veterans Affairs Medical Center, California 94121, USA
| | - Xiannan Tang
- Department of Neurology, University of California, San Francisco & the San Francisco Veterans Affairs Medical Center, California 94121, USA
| | - Hokyou Lee
- Department of Neurology, University of California, San Francisco & the San Francisco Veterans Affairs Medical Center, California 94121, USA
| | - Zheng Zheng
- Department of Neurology, University of California, San Francisco & the San Francisco Veterans Affairs Medical Center, California 94121, USA
| | - Raymond A. Swanson
- Department of Neurology, University of California, San Francisco & the San Francisco Veterans Affairs Medical Center, California 94121, USA
| | - Midori A. Yenari
- Department of Neurology, University of California, San Francisco & the San Francisco Veterans Affairs Medical Center, California 94121, USA
| |
Collapse
|
302
|
Doi K, Leelahavanichkul A, Yuen PST, Star RA. Animal models of sepsis and sepsis-induced kidney injury. J Clin Invest 2009; 119:2868-78. [PMID: 19805915 DOI: 10.1172/jci39421] [Citation(s) in RCA: 415] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Sepsis is characterized by a severe inflammatory response to infection, and its complications, including acute kidney injury, can be fatal. Animal models that correctly mimic human disease are extremely valuable because they hasten the development of clinically useful therapeutics. Too often, however, animal models do not properly mimic human disease. In this Review, we outline a bedside-to-bench-to-bedside approach that has resulted in improved animal models for the study of sepsis - a complex disease for which preventive and therapeutic strategies are unfortunately lacking. We also highlight a few of the promising avenues for therapeutic advances and biomarkers for sepsis and sepsis-induced acute kidney injury. Finally, we review how the study of drug targets and biomarkers are affected by and in turn have influenced these evolving animal models.
Collapse
Affiliation(s)
- Kent Doi
- Department of Nephrology and Endocrinology, University of Tokyo, Tokyo, Japan
| | | | | | | |
Collapse
|
303
|
Sepsis, ghrelin, the cholinergic anti-inflammatory pathway, gut mucosal hyperpermeability, and high-mobility group box 1*. Crit Care Med 2009; 37:2483-5. [DOI: 10.1097/ccm.0b013e3181abf5f4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
304
|
|
305
|
Devi Ramnath R, Weing S, He M, Sun J, Zhang H, Singh Bawa M, Bhatia M. Inflammatory mediators in sepsis: Cytokines, chemokines, adhesion molecules and gases. ACTA ACUST UNITED AC 2009. [DOI: 10.1080/17471060500435662] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
306
|
Cohen J. Recent Developments in the Identification of Novel Therapeutic Targets for the Treatment of Patients with Sepsis and Septic Shock. ACTA ACUST UNITED AC 2009; 35:690-6. [PMID: 14620156 DOI: 10.1080/00365540310016358] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The overall mortality in patients with sepsis is approximately 30%; this figure increases to 50% or higher in patients with septic shock, and sepsis continues to be seen as a major clinical challenge. The recent success of several important trials has fuelled interest in further therapeutic developments. Here, I review the many different strategies that are being investigated, focusing in particular on those that are in late pre-clinical or early clinical development. These can be broadly divided into three groups: strategies aimed at bacterial targets, strategies aimed at disorders of immune regulation in the host, and finally, other novel strategies based on modifying host response; which, if any, of these will prove successful in large clinical trials is unknown. Nevertheless, the fact that sepsis has finally proved tractable as a target for new drug development lends support to those who believe that at least some of the compounds identified in this paper will prove to have clinical benefit.
Collapse
Affiliation(s)
- Jonathan Cohen
- Division of Medicine, Brighton & Sussex Medical School, Brighton, UK.
| |
Collapse
|
307
|
Anti-inflammatory resuscitation improves survival in hemorrhage with trauma. ACTA ACUST UNITED AC 2009; 66:1632-9; discussion 1639-40. [PMID: 19509625 DOI: 10.1097/ta.0b013e3181a5b179] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Hemorrhage is a common cause of death despite the recent advances in resuscitation and critical care. Conventional resuscitation fluids are designed to reestablish tissue perfusion, but they fail to prevent systemic inflammation. Indeed, resuscitation can promote inflammatory responses, which can be more dangerous than the original hemorrhage. This consideration is relevant in critical care where hemorrhage is normally associated with collateral trauma that can exacerbate the inflammatory responses during resuscitation. Here, we analyzed whether ethyl pyruvate could provide a therapeutic anti-inflammatory potential during resuscitation in experimental hemorrhage with trauma. METHODS Adult male Sprague-Dawley rats were subjected to trauma induced by closed femur fracture. Then, the animals were immediately subjected to lethal hemorrhage during 15 minutes to reach a mean arterial blood pressure of 35 mm Hg to 40 mm Hg and subsequent maintenance of this mean arterial blood pressure for another 15 minutes. Resuscitation was limited to 15 mL/kg Hextend with or without ethyl pyruvate. RESULTS Resuscitation with conventional fluids reestablished normal tissue perfusion, but still more than 60% of the animals died. Resuscitation with ethyl pyruvate protected all the animals from lethal hemorrhage with trauma. Trauma exacerbated tumor necrosis factor (TNF) levels in the serum, the spleen, and the heart. Ethyl pyruvate blunted TNF levels in the serum and all the organs but particularly in the lung and the liver during resuscitation. TNF levels in the lung, spleen, and the liver of those animals resuscitated with ethyl pyruvate were statistically similar to those in control animals. CONCLUSION Ethyl pyruvate may attenuate systemic inflammatory responses during resuscitation and improve survival in experimental models of hemorrhage with trauma.
Collapse
|
308
|
Effect of ethyl pyruvate on physical and immunological barriers of the small intestine in a rat model of sepsis. ACTA ACUST UNITED AC 2009; 66:1355-64. [PMID: 19430239 DOI: 10.1097/ta.0b013e31817d0568] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Physical and immunologic barriers of the small intestine play an important role in development and treatment of sepsis, so a rat model of sepsis was used to investigate the effect of ethyl pyruvate (EP) on the barriers. METHODS Male Wistar rats were divided into sham operated, cecal ligation and puncture, and EP groups. Survival and bacterial translocation were measured in response to EP administration. Physical barrier (including mitochondria of enterocyte, tight junction, microvilli, and the grade of small intestinal mucosa damage) and immunologic barrier (including distribution of T-cell subgroups in small intestinal villi, proportion of T-cell subgroups in mesenteric lymph nodes and spleens, proliferation and cytokines release of splenocytes) were determined by electron and light microscopy, immunohistochemistry, flow cytometry, and enzyme-linked immunosorbent assay, respectively. RESULTS Sepsis induced morphologic alteration and immunosuppression in the small intestine. EP administration can prevent these changes, especially immunologic change. Distribution of CD4+ T cells in villi, proportions of CD4+ T cells in mesenteric lymph nodes and spleens, and proliferative capacity of splenocytes were increased in rats treated with EP. Interferon-gamma and Interleukin-4 release were also modulated. Moreover, EP improved survival from 37.1% to 57.1% and reduced bacterial translocation. CONCLUSIONS EP administration ameliorated physical and immunologic barriers dysfunction of small intestine in a rat model of sepsis. EP may be used to treat sepsis as an immunologic modulator.
Collapse
|
309
|
Yang R, Miki K, Oksala N, Nakao A, Lindgren L, Killeen ME, Mennander A, Fink MP, Tenhunen J. Bile high-mobility group box 1 contributes to gut barrier dysfunction in experimental endotoxemia. Am J Physiol Regul Integr Comp Physiol 2009; 297:R362-9. [PMID: 19494177 DOI: 10.1152/ajpregu.00184.2009] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lipopolysaccharide (LPS) is an important factor in sepsis. LPS given by intraperitoneal injection induces intestinal hyperpermeability and bacterial translocation in animals and stimulates hepatic Kupffer cells to release TNF-alpha into the bile. This study aims to test the hypothesis that in response to LPS stimulation, hepatic Kupffer cells and extrahepatic macrophages release a large amount of the inflammatory cytokine high-mobility group box 1 (HMGB1) into the bile and that bile containing HMGB1 contributes to gut barrier dysfunction in experimental endotoxemia. To test this, rat common bile ducts were catheterized and bile flow rate was monitored before and during the LPS administration. Eight hours after LPS challenge, anti-HMGB1 neutralizing antibody or nonimmune (sham) IgG was injected into the duodenal lumen of endotoxemic rats; normal mice were also gavaged with normal or endotoxemic rat bile (bile collected from LPS-treated rats). We found that after LPS challenge, the bile flow rate in rats was significantly decreased at the 4- to 12-h time points, TNF-alpha concentration in the bile was markedly elevated at the 3- to 4-h time points, and bile HMGB1 levels were significantly increased at the 8- to 12-h time points. Duodenal injection with anti-HMGB1 antibody reversed LPS-induced gut barrier dysfunction in rats. In addition, feeding endotoxemic rat bile to normal mice significantly increased both mucosal permeability and bacterial translocation. The increase in permeability and bacterial translocation was reversible following removal of HMGB1 from the endotoxemic rat bile. These findings document that bile HMGB1 mediates gut barrier dysfunction in experimental endotoxemia.
Collapse
Affiliation(s)
- Runkuan Yang
- Dept. of Critical Care Medicine, Univ. of Pittsburgh Medical School, 1055 Scaife Hall, 3550 Terrace St., Pittsburgh, PA 15261, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
310
|
Bennett-Guerrero E, Swaminathan M, Grigore AM, Roach GW, Aberle LG, Johnston JM, Fink MP. A Phase II Multicenter Double-Blind Placebo-Controlled Study of Ethyl Pyruvate in High-Risk Patients Undergoing Cardiac Surgery With Cardiopulmonary Bypass. J Cardiothorac Vasc Anesth 2009; 23:324-9. [PMID: 18835526 DOI: 10.1053/j.jvca.2008.08.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2008] [Indexed: 11/11/2022]
|
311
|
Ethyl pyruvate modulates adhesive and secretory reactions in human lung epithelial cells. Life Sci 2009; 84:805-9. [DOI: 10.1016/j.lfs.2009.03.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Revised: 03/20/2009] [Accepted: 03/24/2009] [Indexed: 11/21/2022]
|
312
|
Davé SH, Tilstra JS, Matsuoka K, Li F, DeMarco RA, Beer-Stolz D, Sepulveda AR, Fink MP, Lotze MT, Plevy SE. Ethyl pyruvate decreases HMGB1 release and ameliorates murine colitis. J Leukoc Biol 2009; 86:633-43. [PMID: 19454652 DOI: 10.1189/jlb.1008662] [Citation(s) in RCA: 143] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Signals from stressed cells and the enteric microbiota activate macrophages and dendritic cells and mediate intestinal inflammation. HMGB1 serves as an immunogenic stimuli causing release of inflammatory cytokines by myeloid cells. Ethyl pyruvate inhibits secretion of HMGB1 and improves survival in models of endotoxemia and hemorrhagic shock. We reasoned that ethyl pyruvate may be protective in colitis, which involves similar inflammatory pathways. In IL-10(-/-) mice with established chronic colitis, ethyl pyruvate administration ameliorated colitis and reduced intestinal cytokine production. IL-10(-/-) mice demonstrated increased intestinal HMGB1 expression and decreased expression of RAGE compared with wild-type mice. Fecal HMGB1 levels were decreased in ethyl pyruvate-treated mice. Furthermore, ethyl pyruvate induced HO-1 expression in intestinal tissue. In TNBS-induced colitis, intrarectal administration of ethyl pyruvate resulted in amelioration of colitis and reduced intestinal cytokine production. In LPS-activated murine macrophages, ethyl pyruvate decreased expression of IL-12 p40 and NO production but did not affect IL-10 levels. Ethyl pyruvate did not inhibit nuclear translocation of NF-kappaB family members but attenuated NF-kappaB DNA binding. Additionally, ethyl pyruvate induced HO-1 mRNA and protein expression and HO-1 promoter activation. Moreover, ethyl pyruvate prevented nuclear-to-cytoplasmic translocation of HMGB1. In conclusion, the HMGB1/RAGE pathway has pathophysiologic and diagnostic significance in experimental colitis. Ethyl pyruvate and other strategies to inhibit HMGB1 release and function represent promising interventions in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Shaival H Davé
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
313
|
Zhu S, Ashok M, Li J, Li W, Yang H, Wang P, Tracey KJ, Sama AE, Wang H. Spermine protects mice against lethal sepsis partly by attenuating surrogate inflammatory markers. Mol Med 2009; 15:275-82. [PMID: 19593412 DOI: 10.2119/molmed.2009.00062] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Accepted: 04/29/2009] [Indexed: 11/06/2022] Open
Abstract
The pathogenesis of sepsis is partly attributable to dysregulated inflammatory response mediated by pathogen-associated molecular patterns (PAMPs) (for example, endotoxin) and damage-associated molecular patterns (DAMPs) (for example, high-mobility group box 1 [HMGB1]). An endogenous ubiquitous polyamine, spermine, inhibits endotoxin-induced cytokine release in vitro, but its capacities to attenuate sepsis- and HMGB1-induced inflammatory responses was previously unknown. We thus tested the hypothesis that spermine protects mice against lethal sepsis by attenuating sepsis-induced local and systemic inflammatory responses. Intraperitoneal (i.p.) administration of spermine (10 mg/kg, twice daily, for 3 d) conferred a significant protection against lethal sepsis. The protective effects were associated with a significant reduction in peritoneal and serum levels of several surrogate markers of sepsis (for example, Interleukin-6 [IL-6], keratinocyte-derived chemokine [KC], monocytes chemoattractant protein-1 [MCP-1], macrophage inflammatory protein-2 [MIP-2], tissue inhibitor of metalloproteinase-1 [TIMP-1], soluble tumor necrosis factor-alpha receptor I [sTNFRI], and soluble tumor necrosis factor-alpha receptor II [sTNFRII]) during a late stage of sepsis. In vitro, spermine effectively inhibited HMGB1-induced release of the above surrogate markers in peritoneal macrophages. Thus, spermine confers protection against lethal sepsis partly by attenuating sepsis- and HMGB1-induced inflammatory responses.
Collapse
Affiliation(s)
- Shu Zhu
- Department of Emergency Medicine, North Shore University Hospital, New York University School of Medicine, Manhasset, New York 11030, United States of America
| | | | | | | | | | | | | | | | | |
Collapse
|
314
|
Wang P, Gong G, Wei Z, Li Y. Ethyl pyruvate prevents intestinal inflammatory response and oxidative stress in a rat model of extrahepatic cholestasis. J Surg Res 2009; 160:228-35. [PMID: 19628226 DOI: 10.1016/j.jss.2009.03.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2008] [Revised: 03/01/2009] [Accepted: 03/13/2009] [Indexed: 01/01/2023]
Abstract
BACKGROUND Ringer's ethyl pyruvate solution (REPS) has been shown to ameliorate liver injury in a murine model of extrahepatic cholestasis. The goal of the present investigation was to gain additional information about whether infusing REPS instead of Ringer's lactate solution (RLS) after inducing obstructive jaundice would be beneficial to intestinal barrier function, inflammatory response, and oxidative stress. METHODS Male Sprague Dawley rats were divided into three groups: Group Sham (n=6), sham-treated controls; Group RLS (n=9), common bile duct ligation (CBDL) plus RLS; and Group REPS (n=9), CBDL plus REPS. On 14 d after BDL, the rats were sacrificed and intestinal permeability was analyzed. Ileal IL-6 and TNF-alpha levels, malondialdehyde (MDA), glutathione (GSH), myeloperoxidase (MPO), and NF-kappaB activity were determined. Histologic examination and apoptosis of ileum were also examined. RESULTS Relative to sham-treated controls, CBDL in RLS-treated rats were associated with increased intestinal permeability to FITC-labeled dextran (4.51+/-0.85 versus 0.44+/-0.18, P<0.01), histopathologic damage and apoptosis (68.4+/-13.4 versus 6.7+/-1.9 pre-1000 villi cells, P<0.01). IL-6 and TNF-alpha level, MDA, MPO, and NF-kappaB activity in ileal tissues were also promoted, along with decreased GSH levels. Treatment with REPS significantly decreased intestinal permeability (3.37+/-0.71, P<0.01) and apoptosis (42.8+/-14.3 pre-1000 villi cells, P<0.01). Other changes were also significantly attenuated by treatment with REPS after CBDL. CONCLUSIONS The present study demonstrates that administration of REPS, but not RLS, maintains intestinal barrier function and reduces intestinal oxidative damage, inflammatory response, and apoptosis in cholestatic rats. This effect of ethyl pyruvate may be useful for preventing intestinal injury in patients with biliary obstruction.
Collapse
Affiliation(s)
- Pengfei Wang
- Department of Intensive Care Unit, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | | | | |
Collapse
|
315
|
Kabarowski JH. G2A and LPC: regulatory functions in immunity. Prostaglandins Other Lipid Mediat 2009; 89:73-81. [PMID: 19383550 DOI: 10.1016/j.prostaglandins.2009.04.007] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Revised: 04/10/2009] [Accepted: 04/13/2009] [Indexed: 02/07/2023]
Abstract
The G2A receptor was originally identified by virtue of its transcriptional induction in murine B lymphoid cells in response to oncogenic transformation and treatment with various DNA-damaging agents. While preliminary characterization of cellular responses to G2A overexpression in fibroblastic cell lines suggested that this receptor may negatively regulate cell growth under conditions of proliferative and genotoxic stress, subsequent studies driven by the discovery of lysophosphatidylcholine (LPC) as a regulator of G2A signaling in immunoregulatory cells point to an important role for this receptor in innate and adaptive immunity.
Collapse
Affiliation(s)
- Janusz H Kabarowski
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294-2170, USA.
| |
Collapse
|
316
|
Kamau E, Takhampunya R, Li T, Kelly E, Peachman KK, Lynch JA, Sun P, Palmer DR. Dengue virus infection promotes translocation of high mobility group box 1 protein from the nucleus to the cytosol in dendritic cells, upregulates cytokine production and modulates virus replication. J Gen Virol 2009; 90:1827-1835. [PMID: 19369409 DOI: 10.1099/vir.0.009027-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
High mobility group box 1 (HMGB1) protein functions in regulation of transcription, cellular activation and pro-inflammatory responses. However, the potential role of HMGB1 during viral infection has not been investigated. This study attempted to elucidate whether the HMGB1-mediated inflammatory response contributes to the pathogenesis of dengue virus (DENV) infection. Our data showed that HMGB1 was released at low DENV infection levels (m.o.i. of 1) under non-necrotic conditions by human dendritic cells (DCs). When DENV-infected DCs were co-cultured with autologous T cells, there was increased production of HMGB1 by both cell types. HMGB1 regulated tumour necrosis factor alpha, interleukin (IL)-6, IL-8 and alpha interferon secretion in DENV-infected DCs. Additionally, increased HMGB1 production was associated with reduced DENV replication titres in DCs. These results suggest that HMGB1 production influences DENV infection in susceptible hosts.
Collapse
Affiliation(s)
- Edwin Kamau
- Division of Viral Diseases, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Ratree Takhampunya
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC 20057, USA
| | - Tao Li
- Division of Viral Diseases, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Eileen Kelly
- Division of Viral Diseases, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Kristina K Peachman
- Division of Retrovirology, Walter Reed Army Institute of Research, Rockville, MD 21910, USA
| | - Julia A Lynch
- Division of Viral Diseases, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Peifang Sun
- Viral and Rickettsial Diseases Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - Dupeh R Palmer
- Division of Viral Diseases, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| |
Collapse
|
317
|
Tsoyi K, Lee TY, Lee YS, Kim HJ, Seo HG, Lee JH, Chang KC. Heme-oxygenase-1 induction and carbon monoxide-releasing molecule inhibit lipopolysaccharide (LPS)-induced high-mobility group box 1 release in vitro and improve survival of mice in LPS- and cecal ligation and puncture-induced sepsis model in vivo. Mol Pharmacol 2009; 76:173-82. [PMID: 19366789 DOI: 10.1124/mol.109.055137] [Citation(s) in RCA: 157] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
We examined our hypothesis that heme-oxygenase-1 (HO-1)-derived carbon monoxide (CO) inhibits the release of high-mobility group box 1 (HMGB1) in RAW264.7 cells activated with lipopolysaccharide (LPS) in vitro and in LPS- or cecal ligation and puncture (CLP)-induced septic mice in vivo, so that HO-1 induction or CO improves survival of sepsis in rodents. We found that pretreatment with HO-1 inducers (hemin, cobalt protoporphyrin IX) or transfection of HO-1 significantly inhibited HMGB1 release, which was blocked by HO-1 small interfering RNA, in cells activated by LPS. Carbon monoxide-releasing molecule 2 (CORM-2) but not bilirubin or deferoxamine inhibited HMGB1 release in LPS-activated macrophages. Oxyhemoglobin reversed the effect of HO-1 inducers on HMGB1 release. Translocation of HMGB1 from nucleus to cytosol was significantly inhibited by HO-1 inducers, CORM-2, or HO-1 transfection. Neutralizing antibodies to tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, interferon-beta, and N(omega)-nitro-L-arginine methyl ester hydrochloride but not N-[2-(cyclohexyloxyl)-4-nitrophenyl]-methane sulfonamide (NS-398) significantly inhibited HMGB1 release in LPS-activated cells. Production of TNF-alpha, IL-1beta, and IFN-beta was significantly reduced by pretreatment of HO-1 inducers, CORM-2, or HO-1 transfection in LPS-activated cells. Plasma levels of HMGB1 in mice challenged with LPS or CLP were significantly reduced by the administration of HO-1 inducers or CORM-2, which was accompanied by either reduction (pretreatment) or no change (delayed administration) of serum TNF-alpha and IL-1beta levels. Regardless of pretreatment or delayed administration, CORM-2 and hemin rescued mice from lethal endotoxemia and sepsis induced by LPS or CLP. Taken together, we concluded that HO-1-derived CO reduces HMGB1 release in LPS-activated cells and LPS- or CLP-induced animal model of sepsis.
Collapse
Affiliation(s)
- Konstantin Tsoyi
- Department of Pharmacology, School of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
318
|
Yang R, Miki K, He X, Killeen ME, Fink MP. Prolonged treatment with N-acetylcystine delays liver recovery from acetaminophen hepatotoxicity. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2009; 13:R55. [PMID: 19358737 PMCID: PMC2689502 DOI: 10.1186/cc7782] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2009] [Revised: 03/23/2009] [Accepted: 04/09/2009] [Indexed: 12/16/2022]
Abstract
Introduction Acetaminophen (APAP) toxicity is the most common cause of acute liver failure in the US and Europe. Massive hepatocyte necrosis is the predominant feature of APAP-induced acute liver injury (ALI). Liver regeneration is a vital process for survival after a toxic insult, it occurs at a relative late time point after the injurious phase. Currently, N-acetylcysteine (NAC), a glutathione precursor, is the antidote for acetaminophen overdose. However, NAC is effective only for patients who present within hours of an acute overdose, and is less effective for late-presenting patients. It is possible that in delayed patients, previously reduced endogenous glutathione (GSH) level has restored and prolonged treatment with NAC might be toxic and impair liver regeneration. Therefore, we hypothesize that prolonged treatment with NAC impairs liver regeneration in ALI induced by APAP. Methods ALI was induced in C57BL/6 male mice by a single dose of APAP (350 mg/kg) by intraperitoneal injection. After two hours of APAP challenge, the mice were given 100 mg/kg NAC dissolved in 0.6 mL saline, or saline treatment every 12 hours for a total of 72 hours. Results Seventy-two hours after APAP challenge, compared with saline treatment, NAC treatment significantly increased serum transaminases (alanine transaminase/aspartate aminotransferase), induced evident hepatocyte vacuolation in the periportal area and delayed liver regeneration seen in histopathology. This detrimental effect was associated with reduced hepatic nuclear factor (NF)-κB DNA binding and decreased expression of cell cycle protein cyclin D1, two important factors in liver regeneration. Conclusions Prolonged treatment with NAC impairs liver regeneration in ALI induced by APAP.
Collapse
Affiliation(s)
- Runkuan Yang
- Department of Critical Care Medicine, University of Pittsburgh Medical School, 3550 Terrace Street, Pittsburgh, PA 15261, USA.
| | | | | | | | | |
Collapse
|
319
|
Abstract
OBJECTIVES Severe hemorrhage is a common cause of death despite the recent advances in critical care. Conventional resuscitation fluids are designed to reestablish tissue perfusion, but they fail to prevent lethal inflammatory responses. Our previous studies indicate that ethyl pyruvate (EP) inhibits tumor necrosis factor (TNF) production from macrophages. Here, we analyze whether EP can provide a therapeutic anti-inflammatory value to resuscitation fluids. DESIGN Laboratory animal experiments. SETTING Animal research laboratory at university medical school. SUBJECTS Adult male Sprague-Dawley rats. INTERVENTIONS Lethal hemorrhage over 15 minutes to reach a mean arterial blood pressure of 35-40 mm Hg and subsequent maintenance of this mean arterial blood pressure for another 15 minutes. Resuscitation was limited to 15 mL/kg Hextend with or without EP. RESULTS Resuscitation with Hextend supplemented with EP rescued all the animals from lethal hemorrhage. Unlike conventional fluids, EP inhibited the production of inflammatory and cardiodepressant factors such as TNF and high mobility group B protein-1. From a pharmacologic perspective, resuscitation with EP was particularly effective inhibiting TNF production in the spleen and the heart. Unlike other anti-inflammatory strategies, EP mitigated systemic inflammation through a mechanism independent of the spleen. At the molecular level, EP inhibited both poly(ADP-ribose) polymerase and p65RelA DNA binding without affecting IkappaBalpha activation. CONCLUSIONS EP may be a promising anti-inflammatory supplement to improve survival during resuscitation in critical care.
Collapse
|
320
|
The effect of high-mobility group box 1 protein on activity of regulatory T cells after thermal injury in rats. Shock 2009; 31:322-9. [PMID: 18665051 DOI: 10.1097/shk.0b013e3181834070] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The aim of the present study was to investigate in vivo the effect of high-mobility group box 1 protein (HMGB1) on activity of regulatory T cells (Tregs) and the influence on T-cell-mediated immunity after thermal injury. Male Wistar rats were randomly divided into four groups as follows: sham burn group, burn group, burn with ethyl pyruvate treatment group, and burn with antireceptor for advanced glycation end products (RAGE) antibody treatment group, and they were killed on postburn days 1, 3, 5, and 7, respectively, with eight animals at each time point. Magnetic cell sorting microbeads were used to isolate splenic Tregs and a column of nylon wool to obtain T cells. Phenotypes, including cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), forkhead/winged helix transcription factor p3 (Foxp3), RAGE, and IL-2Ralpha, were analyzed by flow cytometry. Levels of HMGB1, IL-10, IL-2, IL-4 and interferon gamma were determined by enzyme-linked immunosorbent assay kits, and real-time reverse transcription-polymerase chain reaction was performed to detect mRNA expression of IL-10, IL-2, and IL-2Ralpha. Serum HMGB1 levels were significantly elevated during postburn days 1 to 7. In the burn group, CTLA-4 and Foxp3 expression levels of Tregs were strongly enhanced in comparison to the sham-injured group, and the capacity of Tregs to produce IL-10 was markedly increased. Administration of ethyl pyruvate to inhibit HMGB1 or anti-RAGE antibody could significantly decrease expression levels of CTLA-4, Foxp3 on Tregs, and IL-10 production after burns. Simultaneously, proliferative activity and expression levels of IL-2 and IL-2Ralpha of T cell were restored. The excessively released HMGB1 might stimulate CD4+CD25+Treg activity via binding RAGE on the surface of Tregs and trigger a shift of T(H)1 to T(H)2 with suppression of T-lymphocyte immune function after burn injury.
Collapse
|
321
|
Tang D, Kang R, Xiao W, Zhang H, Lotze MT, Wang H, Xiao X. Quercetin prevents LPS-induced high-mobility group box 1 release and proinflammatory function. Am J Respir Cell Mol Biol 2009; 41:651-60. [PMID: 19265175 DOI: 10.1165/rcmb.2008-0119oc] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The pathogenesis of sepsis is mediated in part by the pathogen-associated molecular pattern molecule bacterial endotoxin, which stimulates macrophages to sequentially release early (e.g., TNF-alpha, IL-1beta) and late (e.g., high-mobility group box [HMGB] 1 protein) proinflammatory mediators. The recent discovery of HMGB1 as a late mediator of lethal sepsis has prompted investigation into development of several new experimental therapeutics that limit release, either blocking HMGB1 itself or its nominal receptors. Quercetin was recently identified as an experimental therapeutic that significantly protects against oxidative injury. Here, we report that quercetin attenuates lethal systemic inflammation caused by endotoxemia, even if treatment is started after the early TNF response. Quercetin treatment reduced circulating levels of HMGB1 in animals with established endotoxemia. In macrophage cultures, quercetin inhibited release as well as the cytokine activities of HMGB1, including limiting the activation of mitogen-activated protein kinase and NF-kappaB, two signaling pathways that are critical for HMGB1-induced subsequent cytokine release. Quercetin and autophagic inhibitor, wortmannin, inhibited LPS-induced type-II microtubule-associated protein 1A/1B-light chain 3 production and aggregation, as well as HMGB1 translocation and release, suggesting a potential association between autophagy and HMGB1 release. Quercetin delivery, a strategy to pharmacologically inhibit HMGB1 release that is effective at clinically achievable concentrations, now warrants further evaluation in sepsis and other systemic inflammatory disorders.
Collapse
Affiliation(s)
- Daolin Tang
- Laboratory of Shock, Department of Pathophysiology, Xiangya School of Medicine, Changsha, Hunan, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
322
|
Kim JH, Kim SJ, Lee IS, Lee MS, Uematsu S, Akira S, Oh KI. Bacterial endotoxin induces the release of high mobility group box 1 via the IFN-beta signaling pathway. THE JOURNAL OF IMMUNOLOGY 2009; 182:2458-66. [PMID: 19201901 DOI: 10.4049/jimmunol.0801364] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Sepsis is a devastating condition characterized by a systemic inflammatory response. Recently, high mobility group box 1 (HMGB1) was identified as a necessary and sufficient mediator of the lethal systemic inflammation caused by sepsis. However, despite its clinical importance, the mechanism of HMGB1 release has remained to be elusive. In this study, we demonstrate that the IFN-beta-mediated JAK/STAT pathway is essential for LPS or Escherichia coli-induced HMGB1 release, which is dependent on Toll/IL-1R domain-containing adapter-inducing IFN-beta adaptor. Additionally, we show that NO acts as a downstream molecule of the IFN-beta signaling. Furthermore, the JAK inhibitor treatment as well as the STAT-1 or IFN-beta receptor deficiency reduced HMGB1 release in a murine model of endotoxemia. Our results suggest that HMGB1 release in sepsis is dependent on the IFN-beta signaling axis; thus, therapeutic agents that selectively inhibit IFN-beta signaling could be beneficial in the treatment of sepsis.
Collapse
Affiliation(s)
- Ju-Hyun Kim
- Department of Pathology, Hallym University College of Medicine, Chuncheon, Korea
| | | | | | | | | | | | | |
Collapse
|
323
|
Pan HF, Wu GC, Li WP, Li XP, Ye DQ. High Mobility Group Box 1: a potential therapeutic target for systemic lupus erythematosus. Mol Biol Rep 2009; 37:1191-5. [PMID: 19247800 DOI: 10.1007/s11033-009-9485-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Accepted: 02/19/2009] [Indexed: 11/26/2022]
Abstract
High Mobility Group Box 1 (HMGB1) is a nuclear protein participating in chromatin architecture and transcriptional regulation. Recently, there is increasing evidence that HMGB1 contributes to the pathogenesis of chronic inflammatory and autoimmune diseases due to its pro-inflammatory and immunostimulatory properties. Elevated expression of HMGB1 was found in the sera of patients and mice with systemic lupus erythematosus (SLE). In addition, it has been shown that HMGB1 may act as a proinflammatory mediator in antibody-induced kidney damage in SLE. All theses findings suggest that HMGB1 have important biological effects in autoimmunity that might be a promising therapeutic target for SLE. In this review, we will briefly discuss the biological features of HMGB1 and summarize recent advances on the role of HMGB1 in the pathogenesis and treatment of SLE.
Collapse
Affiliation(s)
- Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui, People's Republic of China.
| | | | | | | | | |
Collapse
|
324
|
Parrish WR, Gallowitsch-Puerta M, Czura CJ, Tracey KJ. Experimental therapeutic strategies for severe sepsis: mediators and mechanisms. Ann N Y Acad Sci 2009; 1144:210-36. [PMID: 19076379 DOI: 10.1196/annals.1418.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Severe sepsis is the leading cause of mortality in intensive care units. The limited ability of current therapies to reduce sepsis mortality rates has fueled research efforts for the development of novel treatment strategies. Through the close collaboration between clinicians and scientists, progress can be seen in the struggle to develop effective therapeutic approaches for the treatment of sepsis and other immune and inflammatory disorders. Indeed, significant advances in intensive care, such as lung protective mechanical ventilation, improved antibiotics, and superior monitoring of systemic perfusion, are improving patient survival. Nonetheless, specific strategies that target the pathophysiological disorders in sepsis patients are essential to further improve clinical outcomes. This article reviews current clinical management approaches and experimental interventions that target pleiotropic or late-acting inflammatory mediators like caspases, C5a, MIF, and HMGB1, or the body's endogenous inflammatory control mechanisms such as the cholinergic anti-inflammatory pathway. These inflammatory mediators and anti-inflammatory mechanisms, respectively, show significant potential for the development of new experimental therapies for the treatment of severe sepsis and other infectious and inflammatory disorders.
Collapse
Affiliation(s)
- William R Parrish
- The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | | | | | | |
Collapse
|
325
|
Tetrandrine Increased the Survival Rate of Mice With Lipopolysaccharide-Induced Endotoxemia. ACTA ACUST UNITED AC 2009; 66:411-7. [DOI: 10.1097/ta.0b013e31815ebae9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
326
|
Ethyl pyruvate improves survival in awake hemorrhage. J Mol Med (Berl) 2009; 87:423-33. [PMID: 19172241 DOI: 10.1007/s00109-009-0441-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2008] [Revised: 12/10/2008] [Accepted: 01/02/2009] [Indexed: 10/21/2022]
Abstract
Classical experimental models of hemorrhage are characterized by the use of anesthetics that may interfere with the typical immune responses and pathology of hemorrhage/resuscitation. Thus, therapeutic strategies successful in anesthetized animals might not be beneficial in clinical trials. In this study, we analyzed whether ethyl pyruvate could provide therapeutic benefits during resuscitation in awake (unanesthetized) hemorrhage. Our results indicate that hemorrhage in unanesthetized animals required approximately 25% higher blood withdrawal than anesthetized animals to achieve the same targeted mean arterial blood pressure. Resuscitation with Hextend reestablished circulatory volume and improved survival during resuscitation of awake rodents. Yet, over 75% of the animals resuscitated with Hextend died within the first hours after hemorrhage. Resuscitation with Hextend containing 50 mM ethyl pyruvate protected over 87% of the animals. This survival benefit did not correlate with significant changes in the metabolic markers but with an anti-inflammatory potential during resuscitation. Unlike classical hemorrhage in anesthetized animals, ethyl pyruvate reestablished mean arterial blood pressure significantly earlier than Hextend in unanesthetized rodents. Unanesthetized animals showed twofold higher serum tumor necrosis factor (TNF)-alpha than anesthetized animals subjected to the same blood pressure. This process was not due to the response of a single organ, but affected all the analyzed organs including the lung, heart, spleen, and liver. Although resuscitation with Hextend failed to attenuate systemic TNF-alpha levels, it inhibited TNF-alpha levels in the lung, heart, and liver but not in the spleen. Unlike Hextend, resuscitation with ethyl pyruvate prevented high serum TNF-alpha levels and blunted TNF-alpha responses in all the organs including the spleen. These studies indicate that the inflammatory responses in anesthetized animals differ from that in unanesthetized animals and that awake hemorrhage can provide advantages in the study of anti-inflammatory strategies during resuscitation. Ethyl pyruvate may attenuate systemic inflammatory responses during resuscitation and improve survival in experimental models of awake hemorrhage.
Collapse
|
327
|
Intracellular NAD levels regulate tumor necrosis factor protein synthesis in a sirtuin-dependent manner. Nat Med 2009; 15:206-10. [PMID: 19151729 DOI: 10.1038/nm.1906] [Citation(s) in RCA: 213] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2008] [Accepted: 12/04/2008] [Indexed: 01/22/2023]
Abstract
Tumor necrosis factor (TNF) synthesis is known to play a major part in numerous inflammatory disorders, and multiple transcriptional and post-transcriptional regulatory mechanisms have therefore evolved to dampen the production of this key proinflammatory cytokine. The high expression of nicotinamide phosphoribosyltransferase (Nampt), an enzyme involved in the nicotinamide-dependent NAD biosynthetic pathway, in cells of the immune system has led us to examine the potential relationship between NAD metabolism and inflammation. We show here that intracellular NAD concentration promotes TNF synthesis by activated immune cells. Using a positive screen, we have identified Sirt6, a member of the sirtuin family, as the NAD-dependent enzyme able to regulate TNF production by acting at a post-transcriptional step. These studies reveal a previously undescribed relationship between metabolism and the inflammatory response and identify Sirt6 and the nicotinamide-dependent NAD biosynthetic pathway as novel candidates for immunointervention in an inflammatory setting.
Collapse
|
328
|
Ethyl pyruvate reduces the development of zymosan-induced generalized inflammation in mice. Crit Care Med 2009; 37:270-82. [DOI: 10.1097/ccm.0b013e318192fa63] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
329
|
Abstract
Advances in intensive care and antibiotics have prevented the spread of some infections, though sepsis mortality rates remain high. With failure of over thirty clinical trials, sepsis remains a scientific and clinical challenge in modern medicine. Sepsis is defined by the clinical signs of a systemic inflammatory response to infection. "Severe sepsis" is when these symptoms are associated with multiple organ dysfunction. These definitions of sepsis may be too broad and common to heterogeneous groups of patients who do not necessarily have the same disorder. This consideration has become especially evident in the clinical trials that have failed to obtain consistent results in similar studies of patients diagnosed with severe sepsis. In these trials, patients with infections caused by different microorganisms, and affecting different organs, have been combined under the general diagnosis of severe sepsis. The situation is analogous to attempting a clinical trial based on the general definition of cancer, combining all patients with tumor independent of the type of malignancy. In this consideration, it would not be very surprising that activated protein C, the only treatment in sepsis approved by the Food and Drug Administration, is projected for use in only a small subset of patients with severe sepsis. This article reviews novel inflammatory molecular aspects and the experimental anti-inflammatory strategies for sepsis, as they may represent particular pathological processes in specific subsets of patients.
Collapse
Affiliation(s)
- Luis Ulloa
- Laboratory of Anti-inflammatory Signaling and Surgical Immunology, Department of Surgery, UMDNJ-New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA.
| | | | | | | |
Collapse
|
330
|
Unal B, Karabeyoglu M, Huner T, Canbay E, Eroglu A, Yildirim O, Dolapci M, Bilgihan A, Cengiz O. Ethyl pyruvate protects colonic anastomosis from ischemia-reperfusion injury. Surg Innov 2008; 16:21-5. [PMID: 19064591 DOI: 10.1177/1553350608328584] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Ethyl pyruvate is a simple derivative in Ca(+2)- and K(+)-containing balanced salt solution of pyruvate to avoid the problems associated with the instability of pyruvate in solution. It has been shown to ameliorate the effects of ischemia-reperfusion (I/R) injury in many organs. It has also been shown that I/R injury delays the healing of colonic anastomosis. In this study, the effect of ethyl pyruvate on the healing of colon anastomosis and anastomotic strength after I/R injury was investigated. Anastomosis of the colon was performed in 32 adult male Wistar albino rats divided into 4 groups of 8 individuals: (1) sham-operated control group (group 1); (2) 30 minutes of intestinal I/R by superior mesenteric artery occlusion (group 2); (3) I/R+ ethyl pyruvate (group 3), ethyl pyruvate was administered as a 50-mg/kg/d single dose; and (4) I/R+ ethyl pyruvate (group 4), ethyl pyruvate administration was repeatedly (every 6 hours) at the same dose (50 mg/kg). On the fifth postoperative day, animals were killed. Perianastomotic tissue hydroxyproline contents and anastomotic bursting pressures were measured in all groups. When the anastomotic bursting pressures and tissue hydroxyproline contents were compared, it was found that they were decreased in group 2 when compared with groups 1, 3, and 4 (P < .05). Both anastomotic bursting pressure (P = .005) and hydroxyproline content (P < .001) levels were found to be significantly increased with ethyl pyruvate administration when compared with group 2. When ethyl pyruvate administration doses were compared, a significant difference was not observed (P > .05). Ethyl pyruvate significantly prevents the delaying effect of I/R injury on anastomotic strength and healing independent from doses of administration.
Collapse
Affiliation(s)
- B Unal
- Department of Surgery, Faculty of Medicine, Inonu University, Malatya, Turkey
| | | | | | | | | | | | | | | | | |
Collapse
|
331
|
Relationship between high-mobility group box 1 protein release and T-cell suppression in rats after thermal injury. Shock 2008; 30:449-55. [PMID: 18277947 DOI: 10.1097/shk.0b013e3181672495] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
To study whether high-mobility group box 1 protein (HMGB1) has an effect on T-cell-mediated immunity secondary to burn injury, 96 male Wistar rats weighing 250 to 300 g were randomly divided into three groups as follows:sham burn group, burn group, and burn with ethyl pyruvate treatment group, and they were killed on postburn days (PBDs)1, 3, 5, and 7, respectively, with 8 animals at each time point. Columns of nylon wool were used to isolate splenic T cells. T-Cell proliferation was analyzed with thiazolyl blue and expression of IL-2 receptor alpha (IL-2Ralpha) on the surface of T cell with flow cytometry. Levels of HMGB1 were determined using Western blot analysis. IL-2, soluble IL-2R, IL-4, and interferon-gamma were determined with enzyme-linked immunosorbent assay kits. Gene expressions of HMGB1, IL-2, and IL-2R were assessed using reverse-transcription polymerase chain reaction, and activation of nuclear factor of activated T cell was determined with gel mobility shift assay. The levels of HMGB1 in plasma were significantly elevated on PBDs 1 to 5. Significant proliferation of splenic T cells and IL-2, as well as IL-2Ralpha expression on T cells, were simultaneously suppressed to a certain extent on PBDs 1 to 7. Nuclear factor of activated T-cell activity of splenic T cells was markedly down-regulated on PBDs 1 to 3. Administration of ethyl pyruvate to inhibit HMGB1 can significantly restore proliferative activity, nuclear factor of activated T-cell activity, and expression levels of IL-2 and IL-2Ralpha on T cells. High-mobility group box 1 protein released after major burns might be associated with the pathogenesis of immunosuppression in splenic T lymphocytes in rats.
Collapse
|
332
|
Chung KY, Park JJ, Kim YS. The role of high-mobility group box-1 in renal ischemia and reperfusion injury and the effect of ethyl pyruvate. Transplant Proc 2008; 40:2136-8. [PMID: 18790172 DOI: 10.1016/j.transproceed.2008.06.040] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE High mobility group box-1(HMGB1) was identified as a DNA-binding protein that functions as a cofactor for proper transcriptional regulation in somatic cells. Extracellular HMGB1 acts as a potent proinflammatory cytokine that contributes to the pathogenesis of diverse inflammatory and infectious disorders. Ethyl pyruvate (EP), a stable aliphatic ester derived from pyruvic acid, was first described as a pharmacological inhibitor of HMGB1 secretion. We designed this study to identify changes in HMGB1 expression in rat kidney tissues after ischemia reperfusion injury and effects of EP on the expression of HMGB1. MATERIALS AND METHODS Sprague-Dawley rats (200-300 g) were subjected to 40 minutes of renal warm ischemia. The animals were divided into 3 groups: sham group without warm ischemia, EP group (EP given before ischemia), and ischemic control group. Kidneys were harvested and serum creatinine and TNF-alpha measured at 6 hours, 1 day, 3 days, and 5 days after reperfusion. We performed immunohistochemical staining of HMGB1. RESULTS Serum creatinine and TNF-alpha level were elevated in the ischemic control group and the EP injection group. In the EP injection group, serum creatinine and TNF-alpha levels were lower than the ischemic control group. In the 40-minute ischemia-reperfusion model, HMGB1 expression increased at 6 hours after reperfusion and decreased gradually at 1, 3, and 5 days after reperfusion. HMGB1 expression was more distinct at the outer medullary area. intraperitoneal EP injection had no effect on HMGB1 expression. CONCLUSION From these results, we deduced that the preventive effect of EP on rat kidney ischemia-reperfusion injury was not due to the decreased expression of HMGB1 but the prevention of HMGB1 release.
Collapse
Affiliation(s)
- K-Y Chung
- Department of Surgery, Ewha Womans University, School of Medicine, Ewha Medical Research Center, Seoul, Korea.
| | | | | |
Collapse
|
333
|
Tokumaru O, Kuroki C, Yoshimura N, Sakamoto T, Takei H, Ogata K, Kitano T, Nisimaru N, Yokoi I. Neuroprotective effects of ethyl pyruvate on brain energy metabolism after ischemia-reperfusion injury: a 31P-nuclear magnetic resonance study. Neurochem Res 2008; 34:775-85. [PMID: 18985448 DOI: 10.1007/s11064-008-9871-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2008] [Indexed: 12/20/2022]
Abstract
The neuroprotective effects of ethyl pyruvate (EP), a stable derivative of pyruvate, on energy metabolism of rat brain exposed to ischemia-reperfusion stress were investigated by (31)P-nuclear magnetic resonance ((31)P-NMR) spectroscopy. Recovery level of phosphocreatine after ischemia was significantly greater when superfused with artificial cerebrospinal fluid (ACSF) with 2 mM EP than when superfused with ACSF without EP. EP was neuroprotective against ischemia only when administered before the ischemic exposure. Intracellular pH during ischemia was less acidic when superfused ahead of time with EP. EP did not show neuroprotective effects in neuron-rich slices pretreated with 100 microM fluorocitrate, a selective glial poison. It was suggested that both the administration of EP before ischemic exposure and the presence of astrocytes are required for EP to exert neuroprotective effects. We suggest the potential involvement of multiple mechanisms of action, such as less acidic intracellular pH, glial production of lactate, and radical scavenging ability.
Collapse
Affiliation(s)
- Osamu Tokumaru
- Department of Physiology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita, 879-5593, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
334
|
Abstract
Sepsis refers to a systemic inflammatory response syndrome resulting from a microbial
infection. The inflammatory response is partly mediated by innate immune cells (such as
macrophages, monocytes and neutrophils), which not only ingest and eliminate invading
pathogens but also initiate an inflammatory response upon recognition of
pathogen-associated molecular patterns (PAMPs). The prevailing theories of sepsis as a
dysregulated inflammatory response, as manifested by excessive release of inflammatory
mediators such as tumour necrosis factor and high-mobility group box 1 protein (HMGB1),
are supported by extensive studies employing animal models of sepsis. Here we review
emerging evidence that support extracellular HMGB1 as a late mediator of experimental
sepsis, and discuss the therapeutic potential of several HMGB1-targeting agents (including
neutralising antibodies and steroid-like tanshinones) in experimental sepsis.
Collapse
|
335
|
Cai B, Chen F, Ji Y, Kiss L, de Jonge WJ, Conejero-Goldberg C, Szabo C, Deitch EA, Ulloa L. Alpha7 cholinergic-agonist prevents systemic inflammation and improves survival during resuscitation. J Cell Mol Med 2008; 13:3774-85. [PMID: 19602049 PMCID: PMC3046874 DOI: 10.1111/j.1582-4934.2008.00550.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Severe haemorrhage is a common cause of death despite the recent advances in critical care. Conventional resuscitation fluids are designed to re-establish tissue perfusion, but they fail to prevent inflammatory responses during resuscitation. Our previous studies indicated that the vagus nerve can modulate systemic inflammation via the alpha7 nicotinic acetylcholine receptor (alpha7nAchR). Here, we report that the alpha7nAChR-agonist, GTS, restrains systemic inflammation and improves survival during resuscitation. Resuscitation with GTS rescued all the animals from lethal haemorrhage in a concentration-dependent manner. Unlike conventional resuscitation fluids, GTS inhibited the production of characteristic inflammatory and cardiodepressant factors including tumour necrosis factor (TNF) and high mobility group B protein-1 (HMGB1). Resuscitation with GTS was particularly effective in restraining systemic TNF responses and inhibiting its production in the spleen. At the molecular level, GTS inhibited p65RelA but not RelB NF-kappaB during resuscitation. Unlike non-specific nicotinic agonists, GTS inhibited serum protein TNF levels in both normal and splenectomized, haemorrhagic animals. Resuscitation with GTS inhibited poly(ADP-ribose) polymerase and systemic HMGB1 levels. Our studies suggest that GTS provides significant advantages as compared with non-specific nicotinic agonists, and it could be a promising anti-inflammatory supplement to improve survival during resuscitation.
Collapse
Affiliation(s)
- Bolin Cai
- Laboratory of Anti-inflammatory Signaling and Surgical Immunology, UMDNJ-New Jersey Medical School, Newark, NJ 07103, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
336
|
Leelahavanichkul A, Yasuda H, Doi K, Hu X, Zhou H, Yuen PST, Star RA. Methyl-2-acetamidoacrylate, an ethyl pyruvate analog, decreases sepsis-induced acute kidney injury in mice. Am J Physiol Renal Physiol 2008; 295:F1825-35. [PMID: 18922884 DOI: 10.1152/ajprenal.90442.2008] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We tested the anti-inflammatory agent methyl-2-acetamidoacrylate (M2AA), an ethyl pyruvate analog, in a cecal ligation-and-puncture (CLP) model of sepsis in CD-1 mice. M2AA administration at the time of CLP improved survival, renal function, kidney histology, liver injury, and splenocyte apoptosis, and lowered cytokine levels (TNF-alpha, IL-6, IFN-gamma, and IL-10). When M2AA treatment was delayed 6 h (but not 12 h), M2AA still significantly reduced kidney dysfunction, liver injury, splenocyte apoptosis, and cytokine levels. NF-kappaB, a M2AA target, was transiently activated in spleen, peaking at 6 h; kidney and liver NF-kappaB increased steadily with a plateau at 12-24 h. M2AA reduced NF-kappaB activation in spleen at 6 h and in kidney and liver at 24 h. Splenectomy diminished the ability of M2AA to reduce cytokines, especially IL-6, but M2AA still decreased kidney and liver dysfunction, suggesting that splenic NF-kappaB is not central to M2AA action. In contrast, beneficial effects of chloroquine on cytokines and organ damage were neutralized by splenectomy, demonstrating a spleen-specific chloroquine target. Because M2AA and chloroquine act differently, we tested this combination. Survival at 96 h was highest with combination therapy (57%) vs. chloroquine (38%), M2AA (47.6%), or vehicle (5%). The benefit of combination therapy over chloroquine or M2AA alone did not reach statistical significance, indicating potential mechanistic overlap. We conclude that the transient target(s) for M2AA responsible for the narrow 6-h therapeutic window is not splenic NF-kappaB. Identifying this new target and downstream signaling pathways could lengthen the therapeutic window and improve combination therapy with chloroquine.
Collapse
|
337
|
Kim HS, Cho IH, Kim JE, Shin YJ, Jeon JH, Kim Y, Yang YM, Lee KH, Lee JW, Lee WJ, Ye SK, Chung MH. Ethyl pyruvate has an anti-inflammatory effect by inhibiting ROS-dependent STAT signaling in activated microglia. Free Radic Biol Med 2008; 45:950-63. [PMID: 18625301 DOI: 10.1016/j.freeradbiomed.2008.06.009] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Revised: 05/28/2008] [Accepted: 06/03/2008] [Indexed: 11/29/2022]
Abstract
Ethyl pyruvate (EP) has been demonstrated to have an anti-inflammatory function. However, the molecular mechanisms underlying the anti-inflammatory action of EP are largely unknown. We here show that EP exerts its anti-inflammatory effect by inhibiting ROS-dependent STAT signaling through its antioxidant activity, like vitamin C or N-acetyl-L-cysteine. The inhibition of STAT1 and STAT3 by EP prevented their translocation to the nucleus and consequently inhibited expression of iNOS and COX-2 by inhibiting STAT1- and STAT3-mediated transcriptional activity, followed by changes in chromatin conformation via deacetylation of histones H3 and H4 in both gene promoters. EP also suppressed transcripts of other STAT-responsive inflammatory genes such as IL-1beta, IL-6, TNF-alpha, and MCP-1. We further found that the mechanism of inhibition of STAT1 and STAT3 by EP is due to inhibition of JAK2 through Rac1 inactivation and SOCS1 induction. These findings offer new therapeutic possibilities for EP based on a better understanding of the mechanism underlying the action of EP.
Collapse
Affiliation(s)
- Hong Sook Kim
- Department of Pharmacology, Seoul National University College of Medicine, Yongon-dong 28, Chungno-gu, Seoul 110-799, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
338
|
Pahuja M, Tran C, Wang H, Yin K. Alveolar macrophage suppression in sepsis is associated with high mobility group box 1 transmigration. Shock 2008; 29:754-60. [PMID: 18004227 DOI: 10.1097/shk.0b013e31815d0c8f] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Macrophage dysfunction occurs late in sepsis and is implicated in increased mortality. Interferon gamma (IFN-gamma) stimulates transmigration of high mobility group box 1 (HMGB-1) from the nucleus into cytoplasm of macrophages and subsequent release. Because HMGB-1 release also occurs late, and because one of the actions of HMGB-1 in the nucleus is to enhance transcription factors, we investigated if HMGB-1 transmigration is involved in macrophage suppression in sepsis. Alveolar macrophages were isolated 12 and 24 h from sham controls, cecal ligation and puncture (CLP), and CLP rats given IFN-gamma antibody (1.2 mg/kg, i.v.). All injections were given immediately after surgery. At 12 h, 60% of cells from sham controls had HMGB-1 located primarily in the nucleus, whereas 35% of cells had diffuse staining in both cytoplasm and nucleus. In CLP rats, HMGB-1 was located predominantly in the cytoplasm of 37% of cells, and 48% had diffuse staining, whereas in IFN-gamma antibody (Ab)-treated rats, HMGB-1 was located predominantly in the nucleus of 56% of cells, whereas 32% had diffuse staining. At 24 h, most cells from CLP rats (82%) had HMGB-1 located in the cytoplasm, whereas in contrast, HMGB-1 was located in the nucleus of 80% and 82% of cells from sham control and IFN-gamma Ab-treated rats, respectively. Gene expression of TNF-alpha was not significantly changed 12 h after surgery, but at 24 h, alveolar macrophages from CLP rats had reduced gene expression of TNF-alpha. Interferon gamma Ab treatment prevented the reduction in TNF-alpha gene expression. TNF-alpha release was not altered at 12 h. At 24 h, LPS-stimulated release of TNF-alpha was decreased in macrophages from CLP rats compared with sham controls. Interferon gamma Ab treatment prevented the decrease in LPS-stimulated TNF-alpha release. The results suggest that alveolar macrophage suppression after CLP is associated with HMGB-1 transmigration out of the cell nucleus and provides evidence that intranuclear HMGB-1 may play an integral role in macrophage activation in sepsis.
Collapse
Affiliation(s)
- Madhuri Pahuja
- Department of Cell Biology, University of Medicine and Dentistry, NJ-School of Osteopathic Medicine, Stratford, NJ 08084, USA
| | | | | | | |
Collapse
|
339
|
Abstract
Sepsis, a systemic inflammatory response to infection, is a leading cause of death in intensive care units. Recent investigations into the pathogenesis of sepsis reveal a biphasic inflammatory process. An early phase is characterized by pro-inflammatory cytokines (e.g. tumour necrosis factor-alpha), whereas a late phase is mediated by an inflammatory high-mobility group box 1 and an anti-inflammatory interleukin-10. Inflammation aberrantly activates coagulation cascades as sepsis progresses. This dual inflammatory response concomitant with dysregulated coagulation partially accounts for unsuccessful anti-cytokine therapies that have solely targeted early pro-inflammatory mediators (e.g. tumour necrosis factor-alpha). In contrast, activated protein C, which modifies both inflammatory and coagulatory pathways, has improved survival in patients in severe sepsis. Inhibition of the late mediator high-mobility group box 1 improves survival in established sepsis in pre-clinical studies. In addition, recent advances in molecular medicine have shed light on two novel experimental interventions against sepsis. Accelerated apoptosis of lymphocytes has been shown to play an important role in organ dysfunction in sepsis and techniques to suppress apoptosis have improved survival rate in sepsis models. The vagus nerve system has also been shown to suppress innate immune response through endogenous release and exogenous administration of cholinergic agonists, ameliorating inflammation and lethality in sepsis models.
Collapse
|
340
|
Chorny A, Anderson P, Gonzalez-Rey E, Delgado M. Ghrelin protects against experimental sepsis by inhibiting high-mobility group box 1 release and by killing bacteria. THE JOURNAL OF IMMUNOLOGY 2008; 180:8369-77. [PMID: 18523304 DOI: 10.4049/jimmunol.180.12.8369] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Sepsis, a life-threatening complication of infections and the most common cause of death in intensive care units, is characterized by a hyperactive and out-of-balance network of endogenous proinflammatory cytokines. None of the current therapies are entirely effective, illustrating the need for novel therapeutic approaches. Ghrelin (GHR) is an orexigenic peptide that has emerged as a potential endogenous anti-inflammatory factor. In this study, we show that the delayed administration of GHR protects against the mortality in various models of established endotoxemia and sepsis. The therapeutic effect of GHR is mainly mediated by decreasing the secretion of the high mobility box 1 (HMGB1), a DNA-binding factor that acts as a late inflammatory factor critical for sepsis progression. Macrophages seem to be the major cell targets in the inhibition of HMGB1 secretion, in which GHR blocked its cytoplasmic translocation. Interestingly, we also report that GHR shows a potent antibacterial activity in septic mice and in vitro. Remarkably, GHR also reduces the severity of experimental arthritis and the release of HMGB1 to serum. Therefore, by regulating crucial processes of sepsis, such as the production of early and late inflammatory mediators by macrophages and the microbial load, GHR represents a feasible therapeutic agent for this disease and other inflammatory disorders.
Collapse
Affiliation(s)
- Alejo Chorny
- Institute of Parasitology and Biomedicine, Consejo Superior de Investigaciones Cientificas, Granada, Spain
| | | | | | | |
Collapse
|
341
|
PACAP inhibit the release and cytokine activity of HMGB1 and improve the survival during lethal endotoxemia. Int Immunopharmacol 2008; 8:1646-51. [PMID: 18713653 DOI: 10.1016/j.intimp.2008.07.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2008] [Revised: 07/16/2008] [Accepted: 07/22/2008] [Indexed: 12/22/2022]
Abstract
The pathogenesis of sepsis is mediated in part by bacterial endotoxin (lipopolysaccharide; LPS), which stimulates macrophages/monocytes to sequentially release early (e.g., TNF-alpha, IL-1beta) and late [e.g., high mobility group box 1 (HMGB1) protein] pro-inflammatory cytokines. Specifically targeting early mediators has not been effective clinically, in part, because peak mediator activity often has passed before therapy can be initiated. Recent discovery of HMGB1 as a late mediator of lethal sepsis has provided a new target for the treatment of septic shock. Here, we demonstrate that pituitary adenylate cyclase-activating polypeptide (PACAP), an endogenous neuropeptide, significantly attenuated circulating HMGB1 levels and increased survival in animals with established endotoxemia, even if treatment began after acute cytokine response has occurred. In vitro, PACAP suppressed LPS-induced HMGB1 release from macrophages/monocytes, even when given 2-4 h after LPS stimulation. PACAP also suppressed HMGB1 release induced by TNF-alpha or IFN-gamma. Moreover, PACAP inhibits HMGB1-induced cytokine release in vitro and in vivo. These results indicate that PACAP inhibits the release and pro-inflammatory activity of HMGB1 and improves survival during lethal endotoxemia, which confirms this peptide as a candidate for therapy of septic shock.
Collapse
|
342
|
Yang ZY, Ling Y, Yin T, Tao J, Xiong JX, Wu HS, Wang CY. Delayed ethyl pyruvate therapy attenuates experimental severe acute pancreatitis via reduced serum high mobility group box 1 levels in rats. World J Gastroenterol 2008; 14:4546-50. [PMID: 18680237 PMCID: PMC2731284 DOI: 10.3748/wjg.14.4546] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of delayed ethyl pyruvate (EP) delivery on distant organ injury, survival time and serum high mobility group box 1 (HMGB1) levels in rats with experimental severe acute pancreatitis (SAP).
METHODS: A SAP model was induced by retrograde injection of artificial bile into the pancreatic ducts of rats. Animals were divided randomly into three groups (n = 32 in each group): sham group, SAP group and delayed EP treatment group. The rats in the delayed EP treatment group received EP (30 mg/kg) at 12 h, 18 h and 30 h after induction of SAP. Animals were sacrificed, and samples were obtained at 24 h and 48 h after induction of SAP. Serum HMGB1, aspartate aminotransferase (AST), alanine aminotransferase (ALT), blood urea nitrogen (BUN), and creatinine (Cr) levels were measured. Lung wet-to-dry-weight (W/D) ratios and histological scores were calculated to evaluate lung injury. Additional experiments were performed between SAP and delayed EP treatment groups to study the influence of EP on survival times of SAP rats.
RESULTS: Delayed EP treatment significantly reduced serum HMGB1 levels, and protected against liver, renal and lung injury with reduced lung W/D ratios (8.22 ± 0.42 vs 9.76 ± 0.45, P < 0.01), pulmonary histological scores (7.1 ± 0.7 vs 8.4 ± 1.1, P < 0.01), serum AST (667 ± 103 vs 1 368 ± 271, P < 0.01), ALT (446 ± 91 vs 653 ± 98, P < 0.01) and Cr (1.2 ± 0.3 vs 1.8 ± 0.3, P < 0.01) levels. SAP rats had a median survival time of 44 h. Delayed EP treatment significantly prolonged median survival time to 72 h (P < 0.01).
CONCLUSION: Delayed EP therapy protects against distant organ injury and prolongs survival time via reduced serum HMGB1levels in rats with experimental SAP. EP may potentially serve as an effective new therapeutic option against the inflammatory response and multiple organ dysfunction syndrome (MODS) in SAP patients.
Collapse
|
343
|
Lee J, Kwon W, Jo Y, Suh G, Youn Y. Protective effects of ethyl pyruvate treatment on paraquat-intoxicated rats. Hum Exp Toxicol 2008; 27:49-54. [PMID: 18480149 DOI: 10.1177/0960327108088976] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Although, numerous studies have attempted to reduce the oxygen radical injury induced by the antioxidants in paraquat intoxication, these antioxidant therapies have showed few survival benefits. Ethyl pyruvate (EP) may function as an effective scavenger of oxygen radicals, an anti-inflammatory agent and an energy source in many ischemia reperfusion models. The aim of this study was to evaluate the antioxidant and anti-inflammatory effects of EP on the lung and the liver tissues in paraquat-intoxicated rats. Rats were randomly given either a low (2 mg/kg i.p.) or high (40 mg/kg i.p.) EP dose, 30 min before or 1 h after paraquat (50 mg/kg i.p.) administration, and subsequently killed at 6 and 24 h. Glutathione (GSH) and malondialdehyde (MDA) levels of the lungs and the livers, and plasma nitric oxide (NO) concentrations were measured. Pretreatment of EP significantly decreased the MDA level in the lung and the liver tissues. EP also significantly decreased plasma NO concentrations at 6 h. EP pretreatment, however, failed to show significant change in GSH concentration. In post-treatment of EP, MDA levels in the lung tissue and plasma NO levels were significantly decreased. In conclusion, EP decreased the lipid peroxidation and seemed to exert an anti-inflammatory action in the paraquat intoxication rat model.
Collapse
Affiliation(s)
- Jh Lee
- Department of Emergency Medicine, Dongguk University College of Medicine, Gyeongsanbuk-do, Korea
| | | | | | | | | |
Collapse
|
344
|
Alleva LM, Budd AC, Clark IA. Systemic Release of High Mobility Group Box 1 Protein during Severe Murine Influenza. THE JOURNAL OF IMMUNOLOGY 2008; 181:1454-9. [DOI: 10.4049/jimmunol.181.2.1454] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
345
|
Hollenbach M, Hintersdorf A, Huse K, Sack U, Bigl M, Groth M, Santel T, Buchold M, Lindner I, Otto A, Sicker D, Schellenberger W, Almendinger J, Pustowoit B, Birkemeyer C, Platzer M, Oerlecke I, Hemdan N, Birkenmeier G. Ethyl pyruvate and ethyl lactate down-regulate the production of pro-inflammatory cytokines and modulate expression of immune receptors. Biochem Pharmacol 2008; 76:631-44. [PMID: 18625205 DOI: 10.1016/j.bcp.2008.06.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2008] [Revised: 05/26/2008] [Accepted: 06/09/2008] [Indexed: 02/03/2023]
Abstract
Esters of alpha-oxo-carbonic acids such as ethyl pyruvate (EP) have been demonstrated to exert inhibitory effects on the production of anti-inflammatory cytokines. So far, there is no information about effects, if any, of ethyl lactate (EL), an obviously inactive analogue of EP, on inflammatory immune responses. In the present study, we provide evidence that the anti-inflammatory action of alpha-oxo-carbonic acid esters is mediated by inhibition of glyoxalases (Glo), cytosolic enzymes that catalyse the conversion of alpha-oxo-aldehydes such as methylglyoxal (MGO) into the corresponding alpha-hydroxy acids using glutathione as a cofactor. In vitro enzyme activity measurements revealed the inhibition of human Glo1 by alpha-oxo-carbonic acid esters, whilst alpha-hydroxy-carbonic acid esters such as EL were not inhibitory. In contrast, both EP and EL were shown to suppress the Lipopolysaccharide (LPS)-induced production of pro-inflammatory cytokines such as tumor necrosis factor-alpha, interleukin (IL)-1beta, IL-6 and IL-8 from human immunocompetent cells, and modulated the expression of the immune receptors HLA-DR, CD14 and CD91 on human monocytes. Here, we show a crossing link between glyoxalases and the immune system. The results described herein introduce glyoxalases as a possible target for therapeutic approaches of immune suppression.
Collapse
Affiliation(s)
- Marcus Hollenbach
- University of Leipzig, Institute of Biochemistry, Johannisallee 30, 04103 Leipzig, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
346
|
Yuan Z, Chen J, Zhang Y, Peng Y. Construction and characterization of the HMGB1 mutant as a competitive antagonist to HMGB1 induced cytokines release. Biochem Biophys Res Commun 2008; 372:703-7. [PMID: 18515078 DOI: 10.1016/j.bbrc.2008.05.115] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Accepted: 05/17/2008] [Indexed: 11/16/2022]
Abstract
Recent studies indicate that the High Mobility Group Box-1 protein (HMGB1) acts as a potent proinflammatory cytokine that contributes to the pathogenesis of diverse inflammatory and infectious disorders. The proinflammatory cytokine activity of HMGB1 has become a therapeutic target. In this study, we cloned the cDNA encoding human HMGB1 and constructed HMGB1 mutants using a one-step opposite direction PCR. The binding of the HMGB1 mutants to THP-1 cell and the cytokine activities of these HMGB1 mutants were observed. Results showed that the HMGB1 Mut (102-105), one of the HMGB1 mutants, in which amino acids 102-105 (FFLF) were replaced with two Glys, significantly decreased the full-length HMGB1 protein induced TNF-alpha release in human monocyte cultures. The results indicate that we have developed a novel recombinant HMGB1 mutant that competitively antagonizes the proinflammatory activity of HMGB1. This may be of significant importance in providing a new anti-inflammatory strategy for the treatment of severe sepsis and other inflammatory disorders.
Collapse
Affiliation(s)
- Zhiqiang Yuan
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, 30 Gaotanyan Street, Chongqing 400038, China.
| | | | | | | |
Collapse
|
347
|
Fink MP. Bench-to-bedside review: High-mobility group box 1 and critical illness. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2008; 11:229. [PMID: 17903310 PMCID: PMC2556731 DOI: 10.1186/cc6088] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
High-mobility group box 1 (HMGB1) is a DNA-binding protein that also exhibits proinflammatory cytokine-like activity. HMGB1 is passively released by necrotic cells and also is actively secreted by immunostimulated macrophages, dendritic cells, and enterocytes. Although circulating HMGB1 levels are increased relative to healthy controls in patients with infections and severe sepsis, plasma or serum HMGB1 concentrations do not discriminate reliably between infected and uninfected critically ill patients. Nevertheless, administration of drugs that block HMGB1 secretion or of anti-HMGB1 neutralizing antibodies has been shown to ameliorate organ dysfunction and/or improve survival in numerous animal models of critical illness. Because HMGB1 tends to be released relatively late in the inflammatory response (at least in animal models of endotoxemia or sepsis), this protein is an attractive target for the development of new therapeutic agents for the treatment of patients with various forms of critical illness.
Collapse
Affiliation(s)
- Mitchell P Fink
- Department of Critical Care Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15261, USA.
| |
Collapse
|
348
|
Johansson AS, Johansson-Haque K, Okret S, Palmblad J. Ethyl pyruvate modulates acute inflammatory reactions in human endothelial cells in relation to the NF-kappaB pathway. Br J Pharmacol 2008; 154:1318-26. [PMID: 18500358 DOI: 10.1038/bjp.2008.201] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND AND PURPOSE Endothelial cell activation plays a critical role in regulating leukocyte recruitment during inflammation and infection. Ethanol (EtOH) reduces host defence systems, including cell adhesion. However, well-known side effects of EtOH limit its clinical use as an anti-inflammatory drug. Instead, ethyl pyruvate (EtP) may represent a better alternative. Here, we compared effects of EtP and EtOH on neutrophil recruitment and activation of human umbilical vein endothelial cells (HUVECs). EXPERIMENTAL APPROACH Adhesion of neutrophils to HUVEC monolayers, surface expression of intercellular cell adhesion molecule, E-selectin, vascular cell adhesion molecule, release of interleukin (IL)-8 and granulocyte colony-stimulating factor (G-CSF) from HUVECs were assessed as well as translocation of interleukin-1 receptor-associated kinase (IRAK-1), the nuclear factor-kappa B (NF-kappaB) subunits p50, p65 and IkappaB-alpha. NF-kappaB activation was analysed with a luciferase reporter plasmid. Cells were stimulated with IL-1beta, lipopolysaccharide (LPS) or tumour necrosis factor-alpha. KEY RESULTS EtP was several-fold more potent than EtOH in reducing adhesion of neutrophils to activated HUVECs, generation of IL-8 or G-CSF and surface expression of the adhesion molecules. This last reaction was decreased by EtP even when added after cytokines or LPS. Translocation of IRAK-1, IkappaBalpha and the NF-kappaB p65 subunit to the HUVEC nucleus was inhibited by EtP for all stimuli, whereas the diminished p50 translocation was stimulus specific. When p65 was constitutively expressed in Cos7 cells, stimulation of an NF-kappaB-dependent reporter gene was not affected by EtP, suggesting that EtP acted upstream of gene activation. CONCLUSIONS AND IMPLICATIONS EtP impedes adhesive, secretory and signalling events typical of the early inflammatory response in endothelial cells, suggesting EtP as a possible treatment for acute inflammatory conditions.
Collapse
Affiliation(s)
- A-S Johansson
- Center for Inflammation and Hematology Research at Department of Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden.
| | | | | | | |
Collapse
|
349
|
Abstract
PURPOSE OF REVIEW The intention of this article is to review endotoxin, host response to endotoxin, clinical significance of endotoxemia, past failed therapies targeting endotoxin, current therapeutic efforts in this area and the authors' opinion on the future of such therapy. RECENT FINDINGS Endotoxin or lipopolysaccharide is implicated in the activation of cytokine release with the potential to lead to severe sepsis. Therapies targeting endotoxin are very appealing and remain a matter of study and debate. Antiendotoxin antibody studies did not show consistent benefit to warrant its approval for use. Lipid A analog, phospholipid emulsion, and ethyl pyruvate are currently being evaluated for potential clinical use. Polymyxin B as an antiendotoxin strategy has an unacceptable toxicity profile for routine use as an intravenous agent and its use in plasmapheris is too cumbersome. Curcumin and lipopolysaccharide binding peptides, although having a potentially desirable effect on ameliorating endotoxin toxicity, remain to be shown effective in clinical trials. The development of a vaccine against endotoxin carries promise. SUMMARY The benefits of therapies targeting endotoxin remain to be elucidated. Clinical trials targeting populations with documented endotoxemia are more likely to provide an adequate test of this therapeutic approach. Prophylaxis of high-risk populations should also be considered.
Collapse
|
350
|
Yang R, Shaufl AL, Killeen ME, Fink MP. Ethyl pyruvate ameliorates liver injury secondary to severe acute pancreatitis. J Surg Res 2008; 153:302-9. [PMID: 19027919 DOI: 10.1016/j.jss.2008.04.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Revised: 03/07/2008] [Accepted: 04/04/2008] [Indexed: 12/19/2022]
Abstract
BACKGROUND Ethyl pyruvate (EP) is capable of significantly decreasing serum alanine aminotransferase and reducing hepatic necrosis in a murine model of severe acute pancreatitis (SAP); however, the working mechanism is still unclear. This study aims to elucidate the underlying mechanism of EP solution ameliorating SAP-induced liver injury and provide a new therapeutic agent to treat liver injury. MATERIALS AND METHODS Acute necrotizing pancreatitis was induced in C57Bl/6 male mice by feeding the animals a choline-deficient diet supplemented with 0.5% ethionine for 24 h; then the animals were challenged with 7 hourly 50 mug/kg cerulein i.p. injections and a single i.p. injection of Escherichia coli lipopolysaccharide (4 mg/kg). Two hours after the injection of lipopolysaccharide, 40 mg/kg EP, the same volume of Ringers lactate solution (RLS), or saline solution were i.p. injected to animals of EP, RLS, and control groups every 6 h for a total 48-h period. RESULTS When mice were treated with EP, hepatic mRNA expression of tumor necrosis factor-alpha, interleukin-6, inducible nitric oxide synthase, and cyclooxygenase-2 was significantly lower than that in pancreatitis mice treated with RLS. Compared to RLS treatment, treatment with EP significantly decreased the number of inflammatory cell infiltration and markedly inhibited hepatic nuclear factor-kappa B DNA binding; EP therapy dramatically inhibited high motility group B1 release from inflamed hepatic tissue and significantly decreased the concentration of hepatic tissue malondialdehyde, an oxidative stress parameter. EP treatment also significantly improved body circulating blood volume. CONCLUSION EP is a potent anti-inflammatory and anti-oxidative agent to ameliorate hepatic local inflammatory response and resultantly decreases liver injury secondary to SAP.
Collapse
Affiliation(s)
- Runkuan Yang
- Department of Critical Care Medicine, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania 15261, USA.
| | | | | | | |
Collapse
|