用户名: 密码: 验证码:
外源性一氧化碳释放分子(CORM-2)对脓毒症早期肝脏炎症反应和JAK分子磷酸化的抑制作用及分子机制
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
背景与目的
     脓毒症(Spesis)是指由感染引起的全身性炎症反应综合征(SystemicInflammatory Response Syndrome,SIRS),是当前创伤、烧伤外科以及ICU所面临的棘手难题,其进一步发展所导致的脓毒性休克(septic shock,SS)、急性呼吸窘迫综合征(acute respiratory distress syndrome,ARDS)以及多脏器功能不全综合征(multiple organ dysfunction syndrome,MODS),已成为临床主要死亡原因之一。尽管现代外科监护技术和抗生素治疗有了很大进展,但脓毒症的死亡率仍然居高不下,因此脓毒症及其并发症尤其是MODS的防治,仍然是当代外科学领域的难题之一。
     肝脏作为机体的一个重要器官,在脓毒症的发生发展过程中起着至关重要的作用。有证据表明:肝脏拥有全身最多数量的巨噬细胞,负责清除来自肠道、脾脏的细菌和内毒素,肝巨噬细胞(Kuffer细胞)在脓毒症时合成和释放的多种急性时相蛋白和细胞因子,能使多种信号通路活化(包括januskinase/signal transducer andactivator of transcription-JAK/STAT通路)释放炎症介质且不断循环形成“瀑布样”级联反应,在脓毒症发生发展中起重要的作用。
     本研究即采用LPS刺激单核巨噬细胞细胞和小鼠盲肠结扎和穿孔(cecalligation and puncture,CLP)脓毒症模型,以外源性CO释放分子CORM-2进行早期干预,观察它对JAKs分子磷酸化的抑制作用,旨在研究其对脓毒症JAK/STAT通路的活化是否具有调节作用,并探讨可能的分子机制。
     方法
     研究分为2部分,第一部分,利用LPS刺激单核巨噬细胞(RAW264.7),建立细胞脓毒症模型作为脓毒症小鼠的肝脏离体模型,在离体方面研究炎症反应的机制以及外源性一氧化碳释放分子(CORM-2)所释放的CO对该模型的干预作用;第二部分,应用盲肠结扎穿孔(CLP)的C57BL/6小鼠复制脓毒症的在体模型,将小鼠随机分为4组(具体例数根据检测指标而定):假手术组(sham)、CLP组、CLP+iCORM组、CLP+CORM-2组,以肝脏的形态学、功能、细胞因子以及JAK/STAT表达水平为指标探讨CLP小鼠早期肝脏炎症反应的部分分子机制,评价了外源性一氧化碳释放分子(CORM-2)所释放的CO对CLP小鼠早期肝脏炎症反应的抑制作用。
     结果及结论
     1.单核巨噬细胞炎症模型:LPS刺激后细胞损伤明显,活性下降;炎症因子IL-1β、TNF-α相关炎性蛋白表达明显增加,JAK/STAT活性也显著增高。外源性CO的干预组炎症的反应较对照组明显减轻,说明外源性CO能在一定程度抑制小鼠脓毒症离体模型的炎症反应。
     2.CLP小鼠脓毒症模型:术后24h小鼠肝脏炎症细胞浸润明显,血浆中IL-1β、TNF-α等指标明显上升,ICAM-1、JAK/STAT蛋白表达明显增加。外源性CO的直接干预可以明显改善肝脏组织肿胀程度,减少验证细胞的浸润,抑制炎性反应,有效维护细胞功能。
     以上结果表明,外源性外源性一氧化碳释放分子能明显抑制JAKs分子磷酸化,继而抑制了JAK/STAT信号通路的活化,减少下游相关细胞因子的表达,有效防止严重感染时炎症反应的级联反应。证实了外源性一氧化碳的干预在一定程度上抑制了脓毒症早期肝脏炎症反应的发展。
Background and objective:
     Sepsis refers to infections caused by SIRS and is confirmed the existence of bacteria or highly suspicious of infection.Sepsis is the difficult problem faced by trauma,burns surgical and ICU and its further development due to septic shock (septic shock,SS),acute respiratory distress syndrome(acute respiratory distress syndrome,ARDS),as well as integrated multi-organ dysfunction syndrome (multiple organ dysfunction syndrome,MODS),has become a leading cause of death in septic patients.Despite modern surgical techniques,care and antibiotic treatment has made great progress,the mortality rate of sepsis is still high, therefore,the prevention and treatment of sepsis and its complications in particular MODS is still one of the problems among contemporary science. Intestine is an important organ of body and its important physiological functions can not be ignored.
     As an important organ,the Liver plays a vital role in the occurrence and development of sepsis and its important physiological functions can not be ignored,the acute-phase proteins and cytokines released by liver cell and kuffer cell play an important role in sepsis.The liver has the largest number of full-body macrophages,responsible for eliminating from the intestinal LPS and bacteria,so it plays an important role in the SIRS
     In this study,the CO-releasing molecule(CORM-2) was used as an exogenous source of CO.Through in vitro and in vivo experiments we studied whether the exogenous CO could inhibit the inflammatory response of the Liver of early sepsis and explored its mechanism.
     Method:
     The research is divided into 2 parts.In the first part,Monocyte-derived macrophages was stimulated LPS to establish cells sepsis model as a sepsis model of the Liver in vitro.The cells were used to study the mechanism of inflammatory response in vitro,as well as the intervention effect of the CO released by the CORM-2 on the role model.In the second part,use the C57BL/6 mice dealed with cecal ligation and puncture(CLP) as the in vivo model of sepsis.Mice were randomly divided into 4 groups(the number of specific cases may be based on the detection of targets):Sham-operated group,CLP group, CLP+iCORM group,CLP+CORM-2 group,the Liver morphology,function,and inflammatory cells in CLP mice was used as an index to explore the molecular mechanisms on early inflammatory responses in sepsis.The inhibition effect of exogenous carbon monoxide released by CORM-2 on the CLP mice's early liver inflammatory response was evaluated.
     Results and conclusions:
     1.Monocyte-derived macrophages Sepsis model:The phosphorylation levels of and the IL-1β,TNF-αlevel in the culture media in LPS-stimulated Monocyte-derived macrophages increased significantly.ICAM-1,JAK1/JAK3 protein expression was significantly increased.Inflammatory response in the exogenous CO intervention group was significantly reduced than in the control group and thus to some extent,exogenous CO can inhibit the inflammatory response in RAW264.7 model as in vitro model of the Liver sepsis.
     2.Sepsis model in CLP mice:24h after operation,the inflammatory cell infiltration of liver is obvious,plasma indicators increased significantly,ICAM-1, JAK1/JAK3 protein expression significantly increased.The direct intervention of exogenous CO can significantly improve the degree of liver tissue swelling, enhance peristaltic function,inhibit inflammatory response and protect cells. These results show that the cell and animal models of sepsis demonstrated that the intervention of exogenous carbon monoxide somewhat inhibited the development of inflammatory response of early sepsis in the the small intestine.
引文
1.Szabo G,Romics L Jr,Frendl G,Liver in sepsis and systemic inflammstory response syndrome.Clin Liver Dis,2002,6:1045.
    2.Mark GS,Brien JF,Nakatsu K,Mclavghlin BE.Does carbon monoxide have a physiological function.[J]Trends Pharmacol Sci.1991,12:185-188.
    3.Muenzer JT,Davis CG,Dunne BS,et al.Pneumonia alter cecal ligation and puncture:a clinically relevant "two-hit" model of sepsis.Shock,2006,26(6):565-570.
    4.Tang CW,Lan C,Liu R.Inereased activity of the intestinal mueosal mast cells in rats with multiple organ failure.Chin J Dig Dis.2004:5(2):81-6.
    5.王松柏,姚咏明,董宁,等。JAK/STAT通路介导脓毒症大鼠肝组织高迁移率族蛋白BlmRNA表达的研究。中国危重急救医学 2003,15(3):147-150.
    6.Lee C,LimH K,Sakong J,et al.Janus kinase-signal transducer and activator of transcription mediates phosphatidic acid-induced interleukin(IL)-1 and IL-6 roduction.Mol Pharmacol,2006,69(3):1041-1047.
    7.周凤桐,陶国才,王松柏,等.感染性休克大鼠JAK/STAT对组织IFN表达的影响.第三军医大学学报,2006,28(15):1585-1587.
    8.孙炳伟,陈曦,陈兆永,等.外源性一氧化碳释放分子—2对严重烧伤小鼠肝脏炎症反应的抑止作用.中华烧伤杂志,2007,23(3):179-184.
    9.孙炳伟,陈曦,陈兆永,等.外源途径一氧化碳抑制小鼠严重烧伤后早期肺损伤和炎症反应的分子机制.中华医学杂志,2007,87(44):3148-3151.
    10.Lee P J,Jiang B H,Chin B Y,et al.Hypoxia inducible factor-1 mediates transcriptional activation of the heme oxygenase-1 gene in response to hypoxia.J.Biol.Chem.1997,272:5375-5381.
    11.Maines M D.Heme oxygenase:function,multiplicity,regulatory mechanisms and clinical applications.FASEB J.1988,2:2557-2568.
    12.Kitada O.Expression of heme oxygenase-1 and its effects in allergic airway inflammation and hyperresiveness[J].Arerugi,2002,51(11):1095-1102.
    13.Furth R van ZA,H irsh JG,et al.The mononuclearphagocyte system:A new classification ofmacrophages,monocytes,and their p recurso rs.BullWHO,1972,46:846.
    14.姚春艳,姜丽娜,郭术俊,等.腹腔注射IL22对小鼠单核吞噬细胞吞噬功能的影响[J].山西医科大学学报,2007,38(3);202-203.
    15.CRUZ G V,PEREIRA P V,PATRICIO F J,et al.Increase o f c elularre cruitment,p hagocytosisa bility and nitric oxide productioni nduced by hydroalcoholic extract from Chenopodiuma mbrosioidesle aves[J].JE thnopharmacol,2007,111(1);148-154.
    16.PrabhakarNR,Dinerman JL,Agani FH,et al.Carbon monoxide:a role in carotid body chemoreception[J].Proc NatlAcad SciUSA,1995,92:1994-1997.
    17.郭志良.气体信使分子—一氧化亚氮与一氧化碳在生物医学中的研究现状[J].实用心脑肺血管病杂志,2000,8(2):118-121.
    18.STEFFEL J,LESCHER TF,TANNEr FC.Tissue factor in cardiovascular diseases:Molecular mechanisms and clinical imp lications[J].Circulation,2006,113:722-731.
    19.Sun,B.,et al.,Preconditioning of carbon monoxide releasing molecule-derived CO attenuates LPS-induced activation of HUVEC.Int J Biol Sci,2008.4(5):p.270-278.
    20.Sun BW,Sun ZW,Jin Q,et al.CO-releasing molecules(CORM-2)-liberated CO attenuates infiltration of leukocytes in the renal tissue of thermally injured mice.International J Biological Sci.2008;4(3):176-183.
    21.Sun,B.W.,et al.,CO liberated from CORM-2 modulates the inflammatory response in the liver of thermally injured mice.World J Gastroenterol,2008.14(4):p.547-553.
    22.Lee T,Chau LY.Heme oxygenase-1 mediates the anti-inflammatory effect of interleukin-10 in mice.Nat Med 2002;8:240-6.
    23.Otterbein LE,Soares M,Yamashita K,Bach FH.Heme oxygenase-1:unleashing the protective properties of heme.Trends Immunol 2003;24:449-455.
    24.Rodriguez-Gaspar M,Santolaria F,Jarque-Lopez A,et al.Prognostic value ofcytokines in SIRS general medical patients.Cytokine 2001;15(4):232-236.
    25.Parker SJ,Watkins PE.Experimental models of gram-negative sepsis[J].Br J Surg,2001,88(1):22-30.
    26.Sher A,Fioremino DF,Caspar,et al.Production of CD4+ lymphocytes correlates with dowmre gulation of the pholdne elminthm fection.Jm mural,1991:2713-2716.
    27.Mack VE,McCarter MD,Naema HA,et al.Dominance of Th2-type cytokines following severe injury.Arch Surg,1996,131:1303-1312.
    28.Nakao A,Kimizuka K,Stolz DB,Neto JS,Kaizu T,Choi AM,Uchiyama T,Zuckerbraun BS,Nalesnik MA,Otterbein LE,Murase N.Carbon monoxide inhalatio protects rat intesti nal grafts from ischemia/reperfusion injury.Am J Pathol 2003;163;1587-1598.
    29.Moore BA,Otterbein LE,Turler A,Choi AM,Bauer AJ.Inhaled carbon monoxide suppres se-s the development of postoperative ileus in the murine small intestine.Gastroenterology 2003;124;377-391.
    30.Gibbons SJ,Farrugia G.The role of carbon monoxide in the gastrointestinal tract.J Physiol 2004;556;325-336.
    31.Morisaki H,Katayama T,Kotake Y,Ito M,Tamatani T,Sakamoto S,Ishimura Y,Takeda J,Suematsu M.Roles of carbon monoxide in leukocyte and platelet dynamics in rat mesente ric during sevoflurane anesthesia.Attesthesiology 2001;95;192-199.
    32.Morisaki H,Katayama T,Kotake Y,Ito M,Handa M,Ikeda Y,Takeda J,Suematsu M.Carb onmonoxide-modulates endotoxin-induced microvascular leukocyte adhension through pla telet-dependent mechanisms.Attesthesiology 2002;97;701-709.
    33.Ko JS,Yang HR,Chang JY,Seo JK.Lactobacillus plantarum inhibits epithelial barrier dysfunction and interleukin-8 secretion induced by tumor necrosis factor-α.World J Gastroenterol 2007;13:1962-1965.
    34.Hayashi S,Takamiya R,Yamaguchi T,Matsumoto K,Tojo SJ,Tamatani T,et al.Induction of heme oxygenase-1 suppresses venular leukocyte adhesion elicited by oxidative stress.Role of bilirubin generatedby the enzyme.Circ Res 1999;85:663-671.
    35.Maier S.Cecal ligation and puncture versus colon ascendens stent peritonitis:two distinct animal models for polymicrubial sepsis[J].Shock,2004,21(6):505-511.
    36.Oberholzer A.Increased survival in sepsis by in vivo adenovirus-induced expression of IL-10 in dendritic cells[J].J Immunol,2002,168(4):3412-3418.
    37.Moore FA.The role of the gastrointestinal tract in post injury multiple organ failure[J].Am J Surg,1999,178(4):449-452.
    38.史洪涛,冷恩仁,陈东风.胃肠动力与肠源性内毒素血症关系的动物实验研究[J].第三军医大学学报,2001,23:1249-1250.
    39.姚咏明,盛志勇.我国创伤脓毒症基础研究新进展[J].中华创伤杂志,2003;19(1):9-12.
    40.Tracey KJ,Abraham E.From mouse to man:Or what have we learner about cytokine-based anti-inflammatory therapies[J].Shock,1999;11(2):224-225.
    41.Marrero MB,Venema VJ,He H,et al.Inhibition by the JAK/STAT pathway of IFNgamma and LPS-stimulated nitric oxide synthase induction in vascular smooth muscle cells[J].Biochem Biophys Rescommun,1998;252(2):508-512.
    42.Matsukawa A,Ksplan MH,Hogaboam CM,et al.Pivotal role of sigal though the JAK/STAT pathway[J].J Biol Chem,1999:274(23):16513-16512.
    43.Riley JK,Takeda K,Akira S,et al.Interleukin-10 receopter sigalling though the JAK/STAT pathway[J].J Biol Chem,1999:274(23):16513-16521.
    44.Reddy J,Chastegner P,Fiette L,et al.IL-2-induced tumor necrosis factor-beta expression:Futher annalysis in the IL-2 knockout model,and comparison with TNF-alpha,lymphotoxin-beta,TNFR1 and TNFR2 modulation[J].Inmmunology,2001:13(2):137-147.
    45.王松柏,姚咏明,陈劲松,等.严重腹腔感染大鼠JAK/STAT通路活化与多器官功能损害的关系[J].解放军医学杂志,2004;29(1):42-44.
    46.Canbay A,Feldstein AE,Higuchi H,Werneburg N,Grambihler A,BronkSF,Gores GJ.Kupffer cell engulfment of apoptotic bodies stimulates death ligand and cytokine expression[J].Hepatology.2003 Nov;38(5):1188-1198.
    47.姚胜,姚永明,梁华平.脓毒症时JAK/STAT通路活化及意义.中国危重病急救医学2001;13(9):559-562.
    48.Bone RC,Balk RA,Cerra FB,et al.ACCP/SCCM consensus conference:Definition for sepsis and organ failure and guidelines use of innovative therapies in sepsis[J].Chest,1992,101(6):164421655.
    49.Vencent JL,Moreno R,Takala J,et al.The SOFA(sepsis-related organ failure assessment) score to describe organ dysfunction/failure.Intensive Care Med,1996,22(5):707-710.
    50.王松柏,姚咏明,董宁,等.JAK/STAT通路介导脓毒症大鼠肝组织高迁移率族蛋白B1mRNA表达的研究.中国危重病急救医学 2003,15(3):147-150.
    51.Lee C,LimH K,Sakong J,et al.Janus kinase-signal transducer and activator of transcription mediates phosphatidic acid-induced interleukin(IL)-1 and IL-6 roduction.Mol Pharmacol,2006,69(3):1041-1047.
    52.Sarady JK,Zuckerbraun BS,Bilban M,et al.Carbon monoxide protection against endotoxic shock involves reciprocal effects on iNOS in the lung and liver.FASEB J,2004,18(7):854-856.
    53.Nakao A,Moore BA,Murase N,et al.Immunomodulatory effects of inhaled carbon monoxide on rat syngeneic small bowel graft motility.Gut,2003,52(9):1278-1285.
    54.Motterlini R,Mann BE,Johnson TR,et al.Bioactivity and pharmacological actions of carbon monoxide-releasing molecules.Curr Pharm Des,2003,9(30):2525-2539.
    55.姚咏明,盛志勇.我国创伤脓毒症基础研究新进展.中华创伤杂志,2003,19(1):9.
    56.姚胜,姚咏明,李红云等.烫伤脓毒症大鼠肝、肺组织STAT3活化规律及意义X. 解放军医学杂志, 2002, 27(9):763
    
    57. HUANG J S, CHUANG L Y, GUH J Y, et al. Effect of nitric oxide-cGMP-dependent protein kinase activation on advanced glycation end-product-induced proliferation in renal fibroblasts [ J ]. J Am Soc Nephrol, 2005,16(8) : 2318-2329.
    
    58. ARAKAWA T, MASAKI T, H IRA I T, et al, Activationof signal transducer and activator of transcrip tion 3 correlates with cell proliferation and renal injury in human glomerulonephritis[ J ]1.Nephrol Dial Transp lant, 2008,23(11): 3418-3426.
    
    59. KLOCKE R, KUHLMANN M T, SCOB IOALA S, et al,Granulocyte colony2stimulating factor ( G2CSF) for cardio and cerebrovascular regenerative app lications [ J ] .CurrMed Chem, 2008,15 (10): 968-977.
    
    60. LU Y, ZHOU J , XU C, et all JAK/STAT and P I3K/AKT pathways form a mutual transactivation loop and afford resistance to oxidative stress-induced apop tosis incardiomyocytes [ J ]. Cell Physiol Biochem, 2008, 21(4): 305-314.
    
    61. MART1NEZ-CHANTAR M L, VAZQUEZ-CHANTADAM, ARIZU, et al. Loss of the glycine N2methyltrans-ferase gene leads to steatosis and hepatocellular carcinoma in mice [ J ]. Hepatology, 2008, 47 (4 ) : 1191-1199.
    
    62. BAUTISTA D, BERMU' DEZ2SILVA F J , LASARTE J J,et al. Liver expression of p roteins controlling interferon-mediated signalling as p redictive factors in the response to therapy in patients with hepatitis C virus infection [ J ]. J Pathol, 2007, 213 (3): 347-355.
    
    63. CAO Q, MAK KM, L IEBER C S.Leptin represses matrix metallop roteinase-1 gene expression in LX2 human hepatic stellate cells [ J ]. J Hepatol, 2007, 46 ( 1) :124-133.
    
    64. WENG H, MERTENS P R, GRESSNER A M, et al. IFN-gamma abrogates p rofibrogenic TGF-beta signaling in liver by targeting exp ression of inhibitory and receptor Smads[ J ].J Hepatol, 2007, 46 (2 ): 295-303.
    
    65. CH IEN C M, YANG S H, L IN K L, et al.Novel indoloquinoline derivative, IQDMA, suppresses STAT5 phosphorylation and induces apop tosis in HL-60 cells[ J ]. Chem Biol Interact, 2008, 176 (1) : 40-47.
    
    66. ANTAO-MENEZES A, TURP IN E A, Bost P C, et al.STAT-1 signaling in human lung fibroblasts is induced by vanadium pentoxide through an IFN2beta autocrine loop [ J ] . J Immunol, 2008, 180 ( 6 ) : 4200-4207.
    
    67. WANG G, Q IAN P, JACKSON F R, et al. Sequential activation of JAKs, STATs and xanthine dehydrogenase /oxidase by hypoxia in lung microvascular endothelial cells[ J ] . Int J Biochem Cell Biol, 2008, 40 ( 3 ) :461-470.
    
    68. AUJLA S J , DUB IN P J , KOLLS J K Th17 cells and mucosal host defense [ J ] . Semin Immunol, 2007,19(6): 377-382.
    
    69. KIMURA A, NAKA T, NOHARA K, et all Aryl hydro-carbon recep tor regulates Statl activation and participates in the development of Th17 cells [ J ] . Proc Natl Acad Sci USA, 2008,105 (28): 9721-9726.
    
    70. GAGNON J , RAMANATHAN S, LEBLANC C, et al. IL-6, in synergy with IL-7 or IL-15, stimulates TCR-independent proliferation and functional differentiation of CD8 + T lymphocytes [ J ]. J Immunol, 2008,180(12): 7958-7968.
    
    71. OWAKI T, ASAKAWA M, MOR ISH IMA N, et al.STAT3 is indispensable to IL2272mediated cell p roliferation but not to IL-27-induced Th1 differentiation and suppression of proinflammatory cytokine production[ J ].J Immunol, 2008, 180 (5) : 2903-2911.
    [1]D.G.McCormack,S.Mehta,K.Tyml,et al.Pulmonary Microvascular Changes during Sepsis:Evaluation Using Intravital Videomicroscopy.[J].Microvascular Research 2000,60:131-140
    [2]郭志良.气体信使分子—氧化亚氮与一氧化碳在生物医学中的研究现状[J].实用心脑肺血管病杂志,2000,8(2):118-121.
    [3]Bing-Wei Sun,Qin Jin,Yan Sun,etal.Carbon liberated from CO-releasing molecules attenuates leukocyte infiltration in the small intestine of thermally injured mice.[J].World J Gastroenterol 2007 December 14;13(46):6183-6190
    [4]Sun,B.,et al.,Preconditioning of carbon monoxide releasing molecule-derived CO attenuates LPS-induced activation of HUVEC.Int J Biol Sci,2008.4(5):p.270-278.
    [5]PrabhakarNR,Dinerman JL,Agani FH,et al.Carbon monoxide:a role in carotid body chemoreception[J].Proc NatlAcad SciUSA,1995,92:1994-1997.
    [6]STEFFEL J,LESCHER TF,TANNEr FC.Tissue factor in cardiovascular diseases: Molecular mechanisms and clinical implications[J].Circulation,2006,113:722-731.
    [7]Sun,B.W.,et al.,CO liberated from CORM-2 modulates the inflammatory response in the liver of thermally injured mice.World J Gastroenterol,2008.14(4):p.547-553.
    [8]Canbay A,Feldstein AE,Higuchi H,Wemeburg N,Grambihler A,BronkSF,Gores GJ.Kupffer cell engulfment of apoptotic bodies stimulates death ligand and cytokine expression[J].Hepatology.2003 Nov;38(5):1188-1198.
    [9]姚胜,姚永明,梁华平.脓毒症时JAK/STAT通路活化及意义.中国危重病急救医学2001;13(9):559-562
    [10]Sun BW,Chen ZY,Chen X,et al.Attenuation of leukocytes sequestration by carbon monoxide-releasing molecules:liberated carbon monoxide in the liver of thermally injured mice[J].J Bum Care Res,2007,28(1):173-181.
    [11]Diamond P,Doran P,Brady H R et al.Suppressors of cytokine signaling(SOCS):putative modulators of cytokine bioactivity in health and disease.J Nephrol,2000;13:9
    [12]Naka T,Narazaki M,Hirata M,et al.Structure and function of a new STAT2induced STAT inhibitor[J].Nature,1997,387(6636):9242929.
    [13]Bingwei S,Hui S,Chang L,etal.Role of CO-Releasing Molecules Liberated CO in Attenuating Leukocytes Sequestration and Inflammatory Responses in the Lung of Thermally Injured Mice[J].Journal of Surgical Res,2007,139:128-135
    [14]尹文,杨剑虹,虎晓岷等,急性肺损伤肺泡巨噬细胞NF-κB激活通路的实验研究.中国急救医学2003年5月第23卷第5期2003,5(23):281-283
    [15]武志远,樊寻梅.脓毒症抗炎治疗新靶点——晚发炎症介质HMGB1.[J].Chin J Emerg Med,December 2005,14:(12)1051-1052
    [16]外源途径一氧化碳抑制小鼠严重烧伤后早期肺损伤和炎症反应的分子机制,中华烧伤杂志,2007,2319
    [17]赵峰,郑曙云,徐建国.组织因子与脓毒症的关系[J].医学研究生学报,2008,21(4):435-437.
    [18]Price GC,Thomp son SA,Kam PC.Tissue factor and tissue factor pathway inhibitor[J].Anaesthesia,2004,59(5):483-492.
    [19]Napoleone E,Di Santo A,Bastone A,et al.Long pentraxin PTX3 up-regulates tissue factor expression in human endothelial cells:a novellink between vascular inflammation and clotting activation[J].Arterioscler Thromb Vasc Biol,2002,22(5):782-787.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700