强的松在土三七诱导的肝窦阻塞综合征中的治疗作用及其机制探讨
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
第一章土三七诱导小鼠HSOS模型的建立与评价
     目的:采用PBS、野百合碱、环磷酰胺、吗替麦考酚酯、云南三七、土三七小鼠灌胃法,比较各组小鼠HSOS发生率,以求成功构建稳定可复制的HSOS建模方法,并给予综合评价。
     方法:160只清洁级健康雌性昆明小鼠随机分为六组:PBS正常对照组、野百合碱组、环磷酰胺组、吗替麦考酚酯组、云南三七组和土三七组。分别予以野百合碱灌胃7天,环磷酰胺隔日灌胃30天,PBS、吗替麦考酚酯、云南三七、土三七灌胃30天后处死小鼠。检测血液中血细胞计数、肝功能情况;测肝重/体重;留取肝组织行HE染色、Masson染色,用修改后的Deleve评分标准进行病理学评分。
     结果:
     1.野百合碱组、环磷酰胺组和土三七组分别有23只、5只和24只小鼠成模,PBS正常对照组、云南三七组和MMF组小鼠未诱导出HSOS。
     2.与PBS正常对照组比较,野百合碱组及土三七组小鼠体重有增加(P<0.05),其中以土三七组小鼠体重增加更为明显;环磷酰胺组小鼠体重明显下降(P<0.05);而MMF组和云南三七组小鼠体重与PBS正常对照组无明显差异(P>0.05)。
     3.与PBS正常对照组对比,野百合碱组和土三七组小鼠血细胞中RBC及PLT计数下降,WBC计数增加(P<0.05);云南三七组RBC计数稍有增加,WBC和PLT计数稍有减少,但差异并无统计学意义(P>0.05);环磷酰胺组和MMF组小鼠均出现三系减少,且以环磷酰胺组减少得更为明显(P<0.05)。
     4.与PBS正常对照组比较,各模型组小鼠肝指数、ALT、AST、 TBIL均明显升高,ALB明显下降(P<0.05);MMF组小鼠肝功能较PBS正常对照组亦存在一定程度损害,但明显轻于环磷酰胺组(P<0.05);云南三七组小鼠肝功能变化情况与正常对照PBS正常对照组比较无明显差异(P>0.05)。
     5.根据修改后的Deleve评分标准,野百合碱组小鼠多为轻-中度HSOS改变,土三七组及环磷酰胺组小鼠多表现为中-重度HSOS。
     结论:
     1.野百合碱、环磷酰胺、土三七均能成功诱导小鼠HSOS模型,其中环磷酰胺法成模率低于野百合碱和土三七灌胃法,云南三七和MMF未能成功诱导出小鼠HSOS模型。
     2.野百合碱、土三七诱导的小鼠HSOS模型均符合人类HSOS病理改变,其中野百合碱可诱导急性HSOS模型,土三七可诱导慢性HSOS模型,为HSOS的进一步研究提供了理想动物模型。
     3.与野百合碱比较,土三七诱导的HSOS动物模型在进行药物建模机制、药物干预研究方面更为理想。
     4.与土三七比较,云南三七未能成功诱导出小鼠HSOS模型,说明云南三七无明显肝毒性,临床用药相对安全。
     5.与环磷酰胺比较,吗替麦考酚酯的骨髓抑制及肝毒性均较轻,是相对安全的免疫抑制剂。
     第二章强的松对土三七诱导HSOS的防治疗效观察
     目的:探讨不同剂量强的松对土三七诱导小鼠HSOS的防治作用。
     方法:115只清洁级健康雌性昆明小鼠随机分为四组:PBS正常对照组、土三七组、低剂量强的松干预组(5mg·kg-1·d-1)和高剂量强的松干预组(10mg·kg-1·d-1)分别灌胃30天后处死小鼠。检测血液中血细胞计数、肝功能情况;测肝重/体重;留取肝组织行HE染色、Masson染色,用修改后的Deleve评分标准进行病理学评分;并用免疫组织化学法和RT-PCR法检测肝组织的TGF-β1和CTGF表达水平。
     结果:
     1.土三七组、低剂量强的松干预组和高剂量强的松干预组分别有24只、5只和7只小鼠成模,PBS正常对照组无一成模。
     2.与土三七组比较,两组强的松干预组小鼠RBC及PLT计数稍有增加,WBC计数减少,ALB下降明显轻于土三七组,TBIL增高程度亦低于土三七组(P<0.05),ALT和AST无明显差异,不同剂量干预组间无明显差异(P>0.05);低剂量强的松干预组小鼠肝指数明显下降(P<0.01),但高剂量强的松干预组小鼠肝指数改变不明显(P>0.05)。
     3.根据修改后的Deleve评分标准,土三七组小鼠多表现为中-重度HSOS,强的松干预组多表现为轻-中度,无重度HSOS发生,其中以低剂量组平均评分更低(P>0.05)。
     4.肝组织TGF-β1和CTGF表达:免疫组化和RT-PCR均显示两组强的松干预组与土三七组比较,TGF-β1和CTGF表达明显减少(P<0.05),不同剂量干预组间无明显差异(P>0.05)。
     结论:
     1.强的松干预可降低土三七诱导的小鼠HSOS发生,改善小鼠一般情况和肝功能水平,减轻肝窦内皮细胞损害及肝窦纤维化等肝脏病理组织学损伤。
     2.与高剂量强的松(10mg·kg-1·d-1)比较,低剂量强的松(5mg·kg-1·d-1)干预土三七诱导的HSOS小鼠模型,在改善肝窦纤维化方面并无显著差异,但就血药浓度及远期副作用考虑,5mg·kg-1·d-1可能为更为理想的防治剂量。
     3. TGF-β1和CTGF的表达水平可反映HSOS的病情程度、病理改变及临床转归等情况,对观察疗效、评估预后等有重要意义。
     第三章强的松对土三七诱导HSOS防治机制的探讨
     目的:研究强的松对炎性因子及致纤因子的表达及调控机制,从ECM的合成与降解、参与肝窦纤维化形成的细胞因子角度探讨强的松作为HSOS治疗药物的可能性及作用途径和机制,为HSOS的治疗提供进一步的理论和实验依据。
     方法:115只清洁级健康雌性昆明小鼠随机分为四组:PBS正常对照组、土三七组、低剂量强的松干预组(5mg·kg-1·d-1)和高剂量强的松干预组(10mg·kg-1·d-1)分别灌胃30天后处死小鼠。应用RT-PCR法检测肝组织中MMP-1、TIMP-1、MCP-1、TNF-α、IL-1β和NF-κBp65mRNA的表达,western blot法检测TNF-α、TGF-β1和CTGF蛋白的表达。
     结果:
     1. RT-PCR结果:土三七组小鼠肝组织中MMP-1、TIMP-1、 MCP-1、TNF-α、IL-1β、NF-κBp65mRNA的表达较PBS正常对照组显著增高(P<0.01),强的松干预后肝组织中NF-κBp65、TNF-α、IL-1β、 MCP-1mRNA的表达较模型组下降(P<0.05)。与PBS正常对照组比较,土三七组MMP-1/TIMP-1显著减小(P<0.01),强的松干预后MMP-1/TIMP-1较土三七组明显上升(P<0.05)。不同剂量干预组间无明显差异(P>0.05)。
     2.Western blot结果:土三七组小鼠肝组织中TNA-α、TGF-β1和CTGF蛋白的表达较PBS正常对照组显著增高(P<0.01),强的松干预后肝组织中TNA-α、TGF-β1和CTGF蛋白的表达较模型组下降(P<0.05),且不同剂量干预组间无明显差异(P>0.05)。
     结论:强的松在土三七诱导HSOS模型过程中,可能通过下调核转录因子NF-κB的表达,调控趋化蛋白MCP-1和炎症因子TNF-α和IL-1p,进而减少HSC的活化,抑制TGF-β1、CTGF、TIMP-1合成,促进MMP-1降解ECM,降低SEC损害及ECM沉积,减轻炎症和纤维化,起到有效防治HSOS的作用。
Part1Establishment and evaluation of mice model of hepatic sinusoidal obstruction syndrome induced by Gynura segetum
     Objective:To establish and evaluate mice model of hepatic sinusoidal obstruction syndrome (HSOS) induced by Gynura segetum. Phosphate buffer (PBS) was used as the control group. Mice models of HSOS induced by Gynura segetum, monocrotaline, cyclophophamide and mycophenolate mofetil were evaluated in order to choose a more favourable model for further research about the disease.
     Methods:160female KunMing mice were randomly divided into six groups. PBS, monocrotaline, cyclophosphamide, mycophenolate mofetil (MMF), Yunnan Sanqi, and gynura segetum was intragastric administered. The mice in cyclophosphamide group were administered every other day, meanwhile other groups were administered every day. They were sacrificed on day30,7,30,30,30,30. The weight of the liver and body were measured; blood samples were collected to determine cytometry, alanine aminotransferase (ALT), aspartate aminotransferase (AST),albumin (ALB), total bilirubin(TBIL). Liver were sectioned and stained using HE and Masson to observe the pathological changes, and the changes on light microscopy were assessed by a modified Deleve scoring system.
     Results:
     1.23mice in group monocrotaline,5in group cyclophosphamide, and24in group gynura segetum developed HSOS. Mice in group PBS, Yunnan Sanqi and mycophenolate mofetil failed to show any manifestations of HSOS.
     2. Compared with the controls, mice in group monocrotaline and group gynura segetum showed increased the weight of body (P<0.05) especially in group gynura segetum. Meanwhile, the body weight were significantly decreased in group cyclophosphamide (P<0.05). There were no significant differences between the MMF and Yunnan Sanqi group compared with the controls (P>0.05)
     3. Compared with the controls, the value of WBC increased in group monocrotaline and gynura segetum, while the values of RBC and PLT reduced significantly (P<0.05). the value of RBC slightly increased in group Yunnan Sanqi, while the values of WBC and PLT reduced moderatly(P>0.05)without any statistical significance. Hypocytosis was found in mice gavaged with cyclophosphamide and mycophenolate mofetil, with more significant reduction in mice gavaged with cyclophosphamide (P<0.05)
     4. Compare with PBS group, the model groups had significant increased in the level of liver index, serum ALT, AST, TBIL and decreased in the level of serum ALB (P<0.05). Although, the liver function in group MMF had injured at a certain level compare with PBS group, obviously better than in group cyclophosphamide.
     5. Most of gynura segetum and cyclophosphamide group were in moderate or severe HSOS, while monocrotaline group were in mild or moderate HSOS by a modified Deleve scoring system.
     Conclusion:
     1. Monocrotaline, cyclophosphamide and gynura segetum could induce the model of HSOS successfully. The model induced by gynura segetum and monocrotaline were higher than cyclophosphamide. Yunnan Sanqi and MMF failed to induce the model of HSOS.
     2. The model induced by gynura segetum exhibited the clinical and histogical features of human HSOS, and this model may provide a new animal model experimental approach for the further study of the human HSOS. Monocrotaline could induce the acute HSOS model, while gynura segetum could induce the chronic ones.
     3. Compared with monocrotaline, gynura segetum induced more reliable animal models of HSOS in research of mechanism and effects of medicine.
     4. Compared with gynura segetum, Yunnan Sanqi had no hepatotoxicity, and more safe in clinical use.
     5. Compared with cyclophosphamide, MMF is considered to be a immunosuppressive safe drugs without obvious hepatotoxicity and bone marrow suppression.
     Part2Therapeutic effect of prednisone on mice model of hepatic sinusoidal obstruction syndrome induced by Gynura segetum
     Objective:To investigate the possible therapeutic effect of prednisone with different doses on HSOS induced by Gynura segetum.
     Methods:115female KM mice were randomly divided into4groups, they were gavaged with30ml·kg-1·d-1PBS (group A),30g·kg-1·d-1Gynura segetum (group B),30g·kg-1·d-1Gynura segetum+5mg·kg-1·d-1prednisone (groupC), or30g·kg-1·d-1Gynura segetum+10mg·kg-1·d-1prednisone (groupD). All the mice were sacrificed on the30day. Blood samples were collected to determine cytometry, alanine aminotransferase (ALT), aspartate aminotransferase (AST), albumin (ALB), total bilirubin(TBIL). Liver were sectioned and stained using HE and Masson to observe the pathological changes, and the changes on light microscopy were assessed by a modified Deleve scoring system. The expression of TGF-β1and CTGF was determined by immunohistochemical staining and RT-PCR.
     Results:
     1.24mice in group B,5in group C and7in group D developed HSOS. Compared with the controls, they had increased WBC、liver ratio、 TBIL、DBIL、ALT、AST and decreased RBC、PLT、ALB (P<0.05). Administration of prednisone improved the clinical signs and biochemistry parameters without dose-dependent manner.
     2. Most mice of group B developed moderate or severe HSOS, with those in group C and D were in mild or moderate HSOS by a modified Deleve scoring system. The value of group C was lower (P<0.05).
     3. Compared with group B, the expression of TGF-β1and CTGF decreased in groups C and D (P<0.05), and there was no significant difference between groups C and D (P>0.05). These changes were in accordance with the histopathologic changes.
     Conclusion
     Prednisone with different doses has therapeutic effect on HSOS, improving clinical signs and liver function and independent of dose. The expression of TGF-β1and CTGF is associated with the clinical course and severity of HSOS and response to treatment, so it can be an index for monitoring.
     Part3The possible mechanism of the effect of prednisone on mice models of HSOS induced by Gynura segetum
     Objective:To investigate the possible mechanism of the therapeutic effect of prednisone on HSOS induced by Gynura segetum.
     Methods:115female KM mice were randomly divided into4groups, they were gavaged with30ml·kg-1·d-1PBS (group A),30g·kg-1·d-1Gynura segetum (group B),30g·kg-1·d-1Gynura segetum+5mg·kg-1·d-1prednisone (groupC), or30g·kg-1·d-1Gynura segetum+10mg·kg-1·d-1prednisone (groupD). All the mice were sacrificed on the30day. The livers were harvested. The expression of MMP-1、TIMP-1、MCP-1、 TNF-α、IL-1β and NF-κBp65mRNA were determined by RT-PCR and the expression of TNF-α、TGF-β1and CTGF were determined by western blot.
     Results:
     RT-PCR:Compared with the controls, the expression of MMP-1, TIMP-1, MCP-1, TNF-a, IL-1β and NF-KBp65mRNA increased significantly in group B (P<0.01). Prednisone reduced the mRNA expression of MMP-1、TIMP-1、MCP-1、TNF-α、IL-1β and NF-κBP65in group C and D (P<0.05). Compared with the controls, the expression of MMP-1/TIMP-1decreased significantly in group B (P<0.01) while MMP-1/TIMP-1significantly increased (P<0.05). No significant difference between group C and D (P>0.05)
     Western blot:Compared with the controls, the expression of TNF-α、 TGF-β1and CTGF protein increased significantly in group B (P<0.01) Prednisone reduced the protein expression of TNF-α、TGF-β1and CTGF in group C and D (P<0.05). No significant difference between group C and D (P>0.05)
     Conclusion:Prednisone has therapeutic effect on HSOS, the possible mechanism may be that prednisone could reduce inflammation and fobrosis by down regulate the expression of NF-κB, reduce MCP-1, TNF-a and IL-1(3, thereby reducing the activation of HSC, inhibit synthesis of TGF-β, CTGF, TIMP-1, promote MMP-1degradating ECM, and reducing SEC damage and ECM deposition.
引文
[1]Bras G, Jelliffe DB, Stuart KL. Veno-occlusive disease of liver with nonportal type of cirrhosis, occurring in Jamaica. AMA Arch Pathol. Apr 1954;57(4):285-300.
    [2]Helmy A. Review article:updates in the pathogenesis and therapy of hepatic sinusoidal obstruction syndrome. Aliment Pharmacol Ther. Jan 1 2006;23(1):11-25.
    [3]马新光.肝小静脉闭塞病的彩超表现(附16例分析).浙江临床医学.2008(05):593-594.
    [4]刘健红,濮翔科,吴云飞.肝内静脉闭塞病的临床与病理诊断.实用肝脏病杂志.2008(02):107-109.
    [5]黄焕军,刘瑶,林菊生,等.土三七致肝小静脉闭塞病诊断及治疗体会.临床消化病杂志.2008(01):26-28+32.
    [6]黄纪国.土三七致肝小静脉闭塞病8例临床诊治体会.现代中西医结合杂志.2009((04):412.
    [7]张满萍,石红荣,章玉珍.过量服用菊叶三七致肝小静脉闭塞病的护理体会.浙江创伤外科.2009(01):90.
    [8]房龙,樊艳华,王晓娣,等.肝小静脉闭塞病2例并文献复习.胃肠病学和肝病学杂志.2008(06):513-516.
    [9]李尚日,杨玲,熊莉娜,等.肝小静脉闭塞病误诊1例报告并文献复习.中西医结合肝病杂志.2007(04):251-252.
    [10]李晨,梁雪松,李成忠.菊科三七诱导肝窦阻塞综合征1例及文献复习.肝脏.2010(05):395-397.
    [11]钟剑峰,童照威.肝小静脉闭塞病5例临床分析.现代实用医学.2010(07):787-788.
    [12]李玉香,杜娜,王峰,等.土三七引起急性肝小静脉闭塞症2例报告.临床肝胆病杂志.2006(04):319-320.
    [13]Dai X, Yu YC, Ren TH, et al. Gynura root induces hepatic veno-occlusive disease:A case report and review of the literature. World J Gastroentero. Mar 14 2007;13 (10):1628-1631.
    [14]Chen M, Cai J, Du Q. Hepatic veno-occlusive disease associated with the use of Gynura segetum. Bur J Intern Med. Dec 2007;18(8):609.
    [15]李杨,许建明.中草药致肝小静脉闭塞病11例临床分析.临床肝胆病杂 志.2011(02):140-142.
    [16]徐小俊,陈洪潭,单国栋.土三七致肝窦阻塞综合征42例诊治分析.中华危重症医学杂志(电子版).2010(03):178-180.
    [17]徐海波.土三七致肝小静脉闭塞病14例临床分析.中国乡村医药.2010(09):45.
    [18]Brice K, Valerie B, Claire G, et al. Risk-adjusted monitoring of veno-occlusive disease following Bayesian individualization of busulfan dosage for bone marrow transplantation in paediatrics. Pharmacoepidemiol Drug Saf. Feb 2008;17(2):135-143.
    [19]Cacchione A, LeMaitre A, Couanet DV, et al. Risk factors for hepatic veno-occlusive disease:a retrospective unicentric study in 116 children autografted after a high-dose BU-thiotepa regimen. Bone Marrow Transplant. Oct 2008;42(7):449-454.
    [20]Carreras E, Rosinol L, Terol MJ, et al. Veno-occlusive disease of the liver after high-dose cytoreductive therapy with busulfan and melphalan for autologous blood stem cell transplantation in multiple myeloma patients. Biol Blood Marrow Transplant. Dec 2007:13(12):1448-1454.
    [21]DeLeve LD, Shulman HM, McDonald GB. Toxic injury to hepatic sinusoids:sinusoidal obstruction syndrome (veno-occlusive disease). Semin Liver Dis. Feb 2002;22(1):27-42.
    [22]Richardson P, Guinan E. The pathology, diagnosis, and treatment of hepatic veno-occlusive disease:current status and novel approaches. Br J Haematol. Dec 1999;107 (3):485-493.
    [23]Shulman HM, Fisher LB, Schoch HG, et al. Veno-occlusive disease of the liver after marrow transplantation:histological correlates of clinical signs and symptoms. Hepatology. May 1994; 19(5):1171-1181.
    [24]DeLeve LD, McCuskey RS, Wang X, et al. Characterization of a reproducible rat model of hepatic veno-occlusive disease. Hepatology. Jun 1999;29(6):1779-1791.
    [25]Al Beihany A, Al Omar H, Sahovic E, et al. Successful treatment of hepatic veno-occlusive disease after myeloablative allogeneic hematopoietic stem cell transplantation by early administration of a short course of methylprednisolone. Bone Marrow Transplant. Feb 2008;41(3):287-291.
    [26]Sayer HG, Will U, Schilling K, et al. Hepatic veno-occlusive disease (VOD) with complete occlusion of liver venules after tandem autologous stem cell transplantation-- successful treatment with high-dose methylprednisolone and defibrotide. J Cancer Res Clin Oncol. Mar 2002;128(3):148-152.
    [27]Khoury H, Adkins D, Brown R, et al. Does early treatment with high-dose methylprednisolone alter the course of hepatic regimen-related toxicity? Bone Marrow Transpl. Apr 2000;25(7):737-743.
    [28]Akyuz C, CaClar K, Emir S, et al. High-dose methylprednisolone treatment of hepatic veno-occlusive disease in a child with Wilms tumor. Pediatr Hematol Oncol. Jun 2003;20 (4):345-349.
    [29]Willmot FC, Robertson GW. Senecio disease, or cirrhosis of the liver due to Senecio poisoning. Lancet.1920;2:848-849.
    [30]de Jonge ME, Huitema AD, Beijnen JH, et al. High exposures to bioactivated cyclophosphamide are related to the occurrence of veno-occlusive disease of the liver following'nigh-dose chemotherapy. Br J Cancer. May 8 2006;94(9):1226-1230.
    [31]Deeg HJ, Shulman HM, Schmidt E, et al. Marrow graft rejection and veno-occlusive disease of the liver in patients with aplastic anemia conditioned with cyclophosphamide and cyclosporine. Transplantation. Nov 1986:42(5):497-501.
    [32]Copelan EA, Penza SL, Theil KS, et al. The influence of early transplantation, age, GVHD prevention regimen, and other factors on outcome of allogeneic transplantation for CML following BuCy. Bone Marrow Transplant. Nov 2000:26(10):1037-1043.
    [33]de Mattos AM, Olyaei AJ, Bennett WM. Nephrotoxicity of immunosuppressive drugs:long-term consequences and challenges for the future. Am J Kidney Dis. Feb 2000;35(2):333-346.
    [34]Slevers TM, Rossi SJ, Ghobrial RM, et al. Mycophenolate mofetil. Pharmacotherapy. Nov-Dec 1997;17(6):1178-1197.
    [35]Allison AC, Eugui EM. Mycophenolate mofetil and its mechanisms of action. Immunopharmacology. May 2000;47(2-3):85-118.
    [36]陈卫星,杨铭,虞朝辉,等.土三七致肝小静脉闭塞病2例.中华肝脏病杂志.2005(05):394-395.
    [37]吴国琳,余国友,陈玖.中药土三七致肝小静脉闭塞病临床分析.中国中药杂志.2008(20):2402-2404.
    [38]李泓,张顺财,石虹.土三七致肝小静脉闭塞病2例及文献复习.中国临床医学.2009(05):739-741.
    [39]高新生,肖绍树,贺降福.土三七生物碱成分分析及肝小静脉闭塞病小鼠模型建立.中国中西医结合消化杂志.2006(05):311-313.
    [40]Stegelmeier BL, Edgar JA, Colegate SM, et al. Pyrrolizidine alkaloid plants, metabolism and toxicity. J Nat Toxins. Feb 1999;8(1):95-116.
    [41]Couet CE, Crews C, Hanley AB. Analysis, separation, and bioassay of pyrrolizidine alkaloids from comfrey (Symphytum officinale). Nat Toxins.1996;4(4):163-167.
    [42]Dai N, Yu YC, Ren TH, et al. Gynura root induces hepatic veno-occlusive disease:a case report and review of the literature. World J Gastroenterol. Mar 14 2007;13(10):1628-1631.
    [43]Lin G, Wang JY, Li N, et al. Hepatic sinusoidal obstruction syndrome associated with consumption of Gynura segetum. J Hepatol. Apr 2011; 54 (4):666-673.
    [44]Selzer G, Parker RG, Sapeika N. An experimental study of senecio poisoning in rats. Br J Exp Pathol. Feb 1951;32(1):14-20.
    [45]Schoental R, Magee PN. Further observations on the subacute and chronic liver changes in rats after a single dose of various pyrrolizidine (Senecio) alkaloids. J Pathol Bacteriol. Oct 1959;78:471-482.
    [46]Hill KR. Hepatic veno-occlusive disease produced experimentally in rats by the injection of monocrotaline. Lancet. Mar 22 1958;1(7021):623.
    [47]Perazzo J, Eizayaga F, Romay S, et al. An experimental model of liver damage and portal hypertension induced by a single dose of monocrotaline. Hepatogastroenterology. Jan-Feb 1999;46(25):432-435.
    [48]Czauderna P, Chyczewski L, Lech K, et al. Experimental model of hepatic venoocclusive disease (VOD) caused by dactinomycin--preliminary report about hepatoprotective effect of amifostine. Med Sci Monit. May-Jun 2000;6(3):446-453.
    [49]Borowska H, Arciszewska E, Czauderna P, et al. Dactinomycin-induced veno-occlusive disease in rats. The hepatoprotective action of amifostine. Evaluation in a light and electron microscope. Folia Morphol (Warsz). Feb 2004;63(1):91-93.
    [50]Epstein RB, Min KW, Anderson SL, et al. A canine model for hepatic venoocclusive disease. Transplantation. Jul 1992;54(1):12-16.
    [51]Jones RJ, Lee KS, Beschorner WE, et al. Venoocclusive disease of the liver following bone marrow transplantation. Transplantation. Dec 1987:44(6):778-783.
    [52]McDonald GB, Hinds MS, Fisher LD, et al. Veno-occlusive disease of the liver and multiorgan failure after bone marrow transplantation: a cohort study of 355 patients. Ann Intern Med. Feb 15 1993:118(4):255-267.
    [53]McDonald GB, Sharma P, Matthews DE, et al. Venocclusive disease of the liver after bone marrow transplantation:diagnosis, incidence, and predisposing factors. Hepatology. Jan-Feb 1984;4(1):116-122.
    [54]Litzow MR, Repoussis PD, Schroeder G, et al. Veno-occlusive disease of the liver after blood and marrow transplantation:analysis of pre- and post-transplant risk factors associated with severity and results of therapy with tissue plasminogen activator. Leuk Lymphoma. Nov 2002:43(11):2099-2107.
    [55]Gooley TA, Rajvanshi P, Schoch HG, et al. Serum bilirubin levels and mortality after myeloablative allogeneic hematopoietic cell transplantation. Hepatology. Feb 2005;41 (2):345-352.
    [56]Hayashi Y, Hussa JF, Lalich JJ. Cor pulmonalo in rats. Lab Invest. Jun 1967:16(6):875-881.
    [57]Kalayoglu-Besisik S, Yenerel MN, Caliskan Y, et al. Time-related changes in the incidence, severity, and clinical outcome of hepatic veno-occlusive disease in hematopoietic stem cell transplantation patients during the past 10 years. Transplant Proc. Jun 2005:37(5):2285-2289.
    [58]de Fontbrune FS, Mal H, Dauriat G, et al. Veno-occlusive disease of the liver after lung transplantation. Am J Transplant. Sep 2007; 7 (9):2208-2211.
    [59]Membreno FE, Ortiz J, Foster PF, et al. Liver transplantation for sinusoidal obstructive syndrome (veno-occlusive disease):case report with review of the literature and the UNOS database. Clin Transplant. Jul-Aug 2008;22(4):397-404.
    [60]Perkins JD. Sinusoidal obstructive syndrome:a rare and stressful indication for liver transplantation. Liver Transpl. Aug 2008;14(8):1219-1220.
    [61]Azoulay D, Castaing D, Lemoine A, et al. Transjugular intrahepatic portosystemic shunt (TIPS) for severe veno-occlusive disease of the liver following bone marrow transplantation. Bone Marrow Transplant. May 2000;25(9):987-992.
    [62]Rajvanshi P, McDonald GB. Expanding the use of transjugular intrahepatic portosystemic shunts for veno-occlusive disease. Liver Transplant. Feb 2001;7(2):154-157.
    [63]Gressner AM, Weiskirchen R. Modern pathogenetic concepts of liver fibrosis suggest stellate cells and TGF-beta as major players and therapeutic targets. J Cell Mol Med. Jan-Mar 2006;10(1):76-99.
    [64]Rachfal AW, Brigstock DR. Structural and functional properties of CCN proteins. Vitam Horm.2005:70:69-103.
    [65]Sun K, Wang Q, Huang XH. PPAR gamma inhibits growth of rat hepatic stellate cells and TGF beta-induced connective tissue growth factor expression. Acta Pharmacol Sin. Jun 2006;27(6):715-723.
    [66]Chopra R, Eaton JD, Grassi A, et al. Defibrotide for the treatment of hepatic veno-occlusive disease:results of the European compassionate-use study. Br J Haematol. Dec 2000; 111(4):1122-1129.
    [67]Corbacioglu S, Greil J, Peters C, et al. Defibrotide in the treatment of children with veno-occlusive disease (VOD):a retrospective multicentre study demonstrates therapeutic efficacy upon early intervention. Bone Marrow Transplant. Jan 2004;33(2):189-195.
    [68]Richardson P, Soiffer RJ, Antin JH, et al. Defibrotide (DF) for the treatment of severe veno-occlusive disease (sVOD) and multi-organ failure (MOF) post SCT:Final results of a multi-center, randomized, dose-finding trial. Blood. Nov 16 2006;108(11):17a-18a.
    [69]Bearman SI, Lee JL, Baron AE, et al. Treatment of hepatic venocclusive disease with recombinant human tissue plasminogen activator and heparin in 42 marrow transplant patients. Blood. Mar 11997:89(5):1501-1506.
    [70]Ruutu T, Eriksson B, Remes K, et al. Ursodeoxycholic acid for the prevention of hepatic complications in allogeneic stem cell transplantation. Blood. Sep 15 2002;100(6):1977-1983.
    [71]Essell JH, Schroeder MT, Harman GS, et al. Ursodiol prophylaxis against hepatic complications of allogeneic bone marrow transplantation. A randomized, double-blind, placebo-controlled trial. Ann Intern Med. Jun 15 1998;128(12 Pt 1):975-981.
    [72]Ishida H, Nakayasu H, Nukaya H, et al. Study of the pharmacological effect of the bile salt, sodium scymnol sulfate, from Rhizoprionodon acutus. Ⅲ. Protective effect of scymnol against vascular endothelial cell damage in a rat peripheral arterial occlusion model. Biol Pharm Bull. Aug 1999;22(8):822-827.
    [73]Neuman MG, Shear NH, Bellentani S, et al. Role of cytokines in ethanol-induced cytotoxicity in vitro in Hep G2 cells. Gastroenterology. Jul 1998;115(1):157-166.
    [74]Gluckman E, Jolivet I, Scrobohaci ML, et al. Use of prostaglandin E1 for prevention of liver veno-occlusive disease in leukaemic patients treated by allogeneic bone marrow transplantation. Br J Haematol. Mar 1990;74(3):277-281.
    [75]Bearman SI, Shen DD, Hinds MS, et al. A phase Ⅰ/Ⅱ study of prostaglandin El for the prevention of hepatic venocciusive disease after bone marrow transplantation. Br J Haematol. Aug 1993:84(4):724-730.
    [76]Pihusch V, Pihusch M, Penovici M, et al. Transforming growth factor beta-1 released from platelets contributes to hypercoagulability in veno-occlusive disease following hematopoetic stem cell transplantation. Thromb Res.2005;116(3):233-240.
    [77]Bradbury DA, Newton R, Zhu YM, et al. Effect of bradykinin, TGF-beta 1, IL-1 beta, and hypoxia on COX-2 expression in pulmonary artery smooth muscle cells. Am J Physiol-Lung C. Oct 2002;283(4):L717-L725.
    [78]Humbert M, Morrell NW, Archer SL, et al. Cellular and molecular pathobiology of pulmonary arterial hypertension. J Am Coll Cardiol. Jun 16 2004:43(12 Suppl S):13S-24S.
    [79]Shulman HM, Gown AM, Nugent DJ. Hepatic veno-occlusive disease after bone marrow transplantation. Immunohistochemical identification of the material within occluded central venules. Am J Pathol. Jun 1987;127(3):549-558.
    [80]DeLeve LD. Dacarbazine toxicity in murine liver cells:a model of hepatic endothelial injury and glutathione defense. J Pharmacol Exp Ther. Mar 1994;268(3):1261-1270.
    [81]Meresse V, Hartmann O, Vassal G, et al. Risk factors for hepatic veno-occlusive disease after high-dose busulfan-containing regimens followed by autologous bone marrow transplantation:a study in 136 children. Bone Marrow Transplant. Aug 1992:10(2):135-141.
    [82]Hiller O, Lichte A, Oberpichler A, et al. Matrix metalloproteinases collagenase-2, macrophage elastase, collagenase-3, and membrane type 1-matrix metalloproteinase impair clotting by degradation of fibrinogen and factor Ⅻ. J Biol Chem. Oct 20 2000:275(42):33008-33013.
    [83]Nagase H, Visse R, Murphy G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc Res. Feb 15 2006:69(3):562-573.
    [84]Visse R, Nagase H. Matrix metalloproteinases and tissue inhibitors of metal loproteinases:structure, function, and biochemistry. Circ Res. May 2 2003:92(8):827-839.
    [85]Stolow MA, Bauzon DD, Li J, et al. Identification and characterization of a novel collagenase in Xenopus laevis:possible roles during frog development. Mol Biol Cell. Oct 1996:7(10):1471-1483.
    [86]Patterson ML, Atkinson SJ, Knauper V, et al. Specific collagenolysis by gelatinase A, MMP-2, is determined by the hemopexin domain and not the fibronectin-like domain. FEBS Lett. Aug 17 2001:503(2-3):158-162.
    [87]Strieter RM, Wiggins R, Phan SH, et al. Monocyte chemotactic protein gene expression by cytokine-treated human fibroblasts and endothelial cells. Biochem Biophys Res Commun. Jul 31 1989;162(2):694-700.
    [88]Bissell DM, Friedman SL, Maher JJ, et al. Connective tissue biology and hepatic fibrosis:report of a conference. Hepatology. Mar 1990; 11 (3):488-498.
    [89]Hellerbrand C, Jobin C, Iimuro Y, et al. Inhibition of NFkappaB in activated rat hepatic stellate cells by proteasome inhibitors and an IkappaB super-repressor. Hepatology. May 1998; 27(5):1285-1295.
    [90]Luckey SW, Peterson DR. Activation of Kupffer cells during the course of carbon tetrachloride-induced liver injury and fibrosis in rats. Exp Viol Pathol. Dec 2001; 71 (3):226-240.
    [91]Hideshima T, Chauhan D, Richardson P, et al. NF-kappa B as a therapeutic target in multiple myeloma. Journal of Biological Chemistry. May 10 2002;277(19):16639-16647.
    [92]Friedman SL. Seminars in medicine of the Beth Israel Hospital, Boston. The cellular basis of hepatic fibrosis. Mechanisms and treatment strategies. N Engl J Med. Jun 24 1993;328(25):1828-1835.
    [93]Pardo A, Selman M. MMP-1:the elder of the family. Int J Biochem Cell Biol. Feb 2005;37(2):283-288.
    [94]Han YP. Matrix metalloproteinases, the pros and cons, in liver fibrosis. J Gastroenterol Hepatol. Oct 2006;21 Suppl 3:S88-91.
    [95]Zuo F, Kaminski N, Eugui E, et al. Gene expression analysis reveais matrilysin as a key regulator of pulmonary fibrosis in mice and humans. Proc Natl Acad Sci U S A. Apr 30 2002;99(9):6292-6297.
    [96]Selman M, Ruiz V, Cabrera S, et al. TIMP-1,-2,-3, and -4 in idiopathic pulmonary fibrosis. A prevailing nondegradative lung microenvironment? Am J Physiol Lung Cell Mol Physiol. Sep 2000; 279 (3):L562-574.
    [97]Murata K, Kamata Y, Munakata H, et al. Immunohistochemical study on liver fibrosis in biliary atresia. Hepatogastroenterology. Jan-Feb 2008:55(81):179-183.
    [98]Kobayashi H, Li ZX, Yamataka A, et al. Clinical evaluation of serum levels of matrix metalloproteinases and tissue inhibitors of metalloproteinases as predictors of progressive fibrosis in postoperative biliary atresia patients. J Pediatr Surg. Jul 2002:37(7):1030-1033.
    [99]Knittel T, Mehde M, Grundmann A, et al. Expression of matrix metalloproteinases and their inhibitors during hepatic tissue repair in the rat. Histochem Cell Biol. Jun 2000;113(6):443-453.
    [100]Overall CM, Wrana JL, Sodek J. Transcriptional and post-transcriptional regulation of 72-kDa gelatinase/type IV collagenase by transforming growth factor-beta 1 in human fibroblasts. Comparisons with collagenase and tissue inhibitor of matrix metalloproteinase gene expression. J Bio] Chem. Jul 25 1991:266(21):14064-14071.
    [101]Roeb E, Graeve L, Hoffmann R, et al. Regulation of tissue inhibitor of metalloproteinases-1 gene expression by cytokines and dexamethasone in rat hepatocyte primary cultures. Hepatology. Dec 1993;18(6):1437-1442.
    [102]Kordula T, Guttgemann I, Rose-John S, et al. Synthesis of tissue inhibitor of metalloproteinase-1 (TIMP-1) in human hepatoma cells (HepG2). Up-regulation by interleukin-6 and transforming growth factor beta 1. FEBS Lett. Nov 23 1992:313(2):143-147.
    [103]Gomez DE, Alonso DF, Yoshiji H, et al. Tissue inhibitors of metalloproteinases:structure, regulation and biological functions. Eur J Cell Biol. Oct 1997;74(2):111-122.
    [104]Arthur MJ, Mann DA, Iredale JP. Tissue inhibitors of metalloproteinases, hepatic stellate cells and liver fibrosis. J Gastroenterol Hepatol. Sep 1998; 13 Suppl:S33-38.
    [105]Van Coil] ie E, Van Damme J, Opdenakker G. The MCP/eotaxin subfamily of CC chemokines. Cytokine Growth Factor Rev. Mar 1999; 10(1):61-86.
    [106]Ikura M, Kay LE, Bax A. A novel approach for sequential assignment of 1H,13C, and 15N spectra of proteins:heteronuclear triple-resonance three-dimensional NMR spectroscopy. Application to calmodulin. Biochemistry. May 15 1990;29(19):4659-4667.
    [107]Marra F, Romanelli RG, Giannini C, et al. Monocyte chemotactic protein-1 as a chemoattractant for human hepatic stellate cells. Hepatology. Jan 1999;29(1):140-148.
    [108]Seki E, de Minicis S, Inokuchi S, et al. CCR2 promotes hepatic fibrosis in mice. Hepatology. Jul 2009;50(1):185-197.
    [109]Clement S, Juge-Aubry C, Sgroi A, et al. Monocyte chemoattractant protein-1 secreted by adipose tissue induces direct lipid accumulation in hepatocytes. Hepatology. Sep 2008;48(3):799-807.
    [110]Tang W, Ziring D, Gershman G, et al. Role of macrophages and stellate cells in the pathogenesis of veno-occlusive disease:an electron microscopic case study. Exp Mol Pathol. Dec 2003;75(3):201-209.
    [111]Tsukamoto H. Cytokine regulation of hepatic stellate cells in liver fibrosis. Alcohol Clin Exp Res. May 1999;23(5):911-916.
    [112]Basak C, Pathak SK, Bhattacharyya A, et al. NF-kappaB-and C/EBPbuta-driven interleukin-lbeta gene expression and PAK1-mediated caspase-1 activation play essential roles in interleukin-lbeta release from Helicobacter pylori lipopolysaccharide-stimulated macrophages. J Biol Chem. Feb 11 2005;280(6):4279-4288.
    [113]Dinarello CA. Interleukin-1 and interleukin-1 antagonism. Blood. Apr 15 1991;77(8):1627-1652.
    [114]Neurath MF, Becker C, Barbulescu K. Role of NF-kappaB in immune and inflammatory responses in the gut. Gut. Dec 1998;43(6):856-860.
    [115]Rogler G, Brand K, Vogl D, et al. Nuclear factor kappaB is activated in macrophages and epithelial cells of inflamed intestinal mucosa. Gastroenterology. Aug 1998;115(2):357-369.
    [116]Blom IE, van Dijk AJ, de Weger RA, et al. Identification of human ccn2 (connective tissue growth factor) promoter polymorphisms. Mol Pathol. Jun 2001;54(3):192-196.
    [117]Grotendorst GR. Connective tissue growth factor:a mediator of TGF-beta action on fibroblasts. Cytokine Growth Factor Rev. Sep 1997;8(3):171-179.
    [118]Healy E, Brady HR. Role of tubule epithelial cells in the pathogenesis of tubulointerstitial fibrosis induced by glomerular disease. Curr Opin Nephrol Hypertens. Sep 1998;7(5):525-530.
    [119]杨丽,霍继荣,欧大联.土三七致肝小静脉闭塞症临床与诊断.肝脏.2009(05):433-434.
    [120]杨丽,朱洪怡,陈哲,等.3种药物诱导肝小静脉闭塞病小鼠模型的比较.中华肝脏病杂志.2010:18(12):944-945.
    [121]陈哲,霍继荣,朱洪怡,等.内皮损伤相关因子在小鼠肝小静脉闭塞病模型中的表达.胃肠病学.2010;15(10):604-608.
    [122]Chen Z, Huo JR. Hepatic veno-occlusive disease associated with toxicity of pyrrolizidine alkaloids in herbal preparations. Neth J Med. Jun 2010;68(6):252-260.
    [123]Chen Z, Huo JR, Zhu HY. Effect of ligustrazine on mice model of hepatic veno-occlusive disease induced by Sedum aizoon. J Gastroenterol Hepatol. Jan 20 2011.
    [1]Wadleigh M, Ho V, Momtaz P, et al. Hepatic veno-occlusive disease: pathogenesis, diagnosis and treatment. Curr Opin Hematol. Nov 2003;10(6):451-462.
    [2]Willmot FC, Robertson GW. Senecio disease, or cirrhosis of the liver due to Senecio poisoning. Lancet.1920;2:848-849.
    [3]Bras G, Jelliffe DB, Stuart KL. Veno-occlusive disease of liver with nonportal type of cirrhosis, occurring in Jamaica. AMA Arch Pathol. Apr 1954;57(4):285-300.
    [4]Hill KR. Hepatic veno-occlusive disease produced experimentally in rats by the injection of monocrotaline. Lancet. Mar 22 1958;1(7021):623.
    [5]候景贵,夏玉亭,余昌时,等.肝小静脉闭塞病.中华内科杂志.1980;19(3):187-191.
    [6]Helmy A. Review article:updates in the pathogenesis and therapy of hepatic sinusoidal obstruction syndrome. Aliment Pharmacol Ther. Jan 1 2006;23(1):11-25.
    [7]Berk PD, Popper H, Krueger GR, et al. Veno-occlusive disease of the liver after allogeneic bone marrow transplantation:possible association with graft-versus-host disease. Ann Intern Med. Feb 1979;90(2):158-164.
    [8]Jacobs P, Miller JL, Uys CJ, et al. Fatal veno-occlusive disease of the liver after chemotherapy, whole-body irradiation and bone marrow transplantation for refractory acute leukaemia. S Afr Med J. Jan 6 1979;55(1):5-10.
    [9]McDonald GB, Hinds MS, Fisher LD, et al. Veno-occlusive disease of the liver and multiorgan failure after bone marrow transplantation:a cohort study of 355 patients. Ann Intern Med. Feb 15 1993;118(4):255-267.
    [10]DeLeve LD. Cellular target of cyclophosphamide toxicity in the murine liver: role of glutathione and site of metabolic activation. Hepatology. Oct 1996;24(4):830-837.
    [11]Meresse V, Hartmann O, Vassal G, et al. Risk factors for hepatic veno-occlusive disease after high-dose busulfan-containing regimens followed by autologous bone marrow transplantation:a study in 136 children. Bone Marrow Transplant. Aug 1992; 10(2):135-141.
    [12]Carreras E, Bertz H, Arcese W, et al. Incidence and outcome of hepatic veno-occlusive disease after blood or marrow transplantation:a prospective cohort study of the European Group for Blood and Marrow Transplantation. European Group for Blood and Marrow Transplantation Chronic Leukemia Working Party. Blood. Nov 15 1998;92(10):3599-3604.
    [13]Izaki T, Inomata Y, Asonuma K, et al. Early graft failure due to a veno-occlusive disease after a pediatric living donor liver transplantation. Pediatr Transplant. Jun 2004;8(3):301-304.
    [14]Jones RJ, Lee KS, Beschorner WE, et al. Venoocclusive disease of the liver following bone marrow transplantation. Transplantation. Dec 1987;44(6):778-783.
    [15]Roeder E. Medicinal plants in China containing pyrrolizidine alkaloids. Pharmazie. Oct 2000;55(10):711-726.
    [16]王强,鲁重美,郭涛.含吡咯烷生物碱的中草药与肝小静脉闭塞病.临床消化病杂志.2008(01):22-25.
    [17]DeLeve LD, McCuskey RS, Wang X, et al. Characterization of a reproducible rat model of hepatic veno-occlusive disease. Hepatology. Jun 1999;29(6):1779-1791.
    [18]高新生,肖绍树,贺降福.土三七生物碱成分分析及肝小静脉闭塞病小鼠模型建立.中国中医结合消化杂志.2006(05):311-313.
    [19]Cavet J, Middleton PG, Segall M, et al. Recipient tumor necrosis factor-alpha and interleukin-10 gene polymorphisms associate with early mortality and acute graft-versus-host disease severity in HLA-matched sibling bone marrow transplants. Blood. Dec 1 1999;94(11):3941-3946.
    [20]Kallianpur AR, Hall LD, Yadav M, et al. The hemochromatosis C282Y allele: a risk factor for hepatic veno-occlusive disease after hematopoietic stem cell transplantation. Bone Marrow Transplant. Jun 2005;35(12):1155-1164.
    [21]Beutler E, Gelbart T, Kondo T, et al. The molecular basis of a case of gamma-glutamylcysteine synthetase deficiency. Blood. Oct 15 1999;94(8):2890-2894.
    [22]Poonkuzhali B, Chandy M, Srivastava A, et al. Glutathione S-transferase activity influences busulfan pharmacokinetics in patients with beta thalassemia major undergoing bone marrow transplantation. Drug Metab Dispos. Mar 2001;29(3):264-267.
    [23]Shulman HM, Gown AM, Nugent DJ. Hepatic veno-occlusive disease after bone marrow transplantation. Immunohistochemical identification of the material within occluded central venules. Am J Pathol. Jun 1987;127(3):549-558.
    [24]DeLeve LD. Dacarbazine toxicity in murine liver cells:a model of hepatic endothelial injury and glutathione defense. J Pharmacol Exp Ther. Mar 1994;268(3):1261-1270.
    [25]Shulman HM, Fisher LB, Schoch HG, et al. Veno-occlusive disease of the liver after marrow transplantation:histological correlates of clinical signs and symptoms. Hepatology. May 1994; 19(5):1171-1181.
    [26]DeLeve LD. Ito Y, Bethea NW, et al. Embolization by sinusoidal lining cells obstructs the microcirculation in rat sinusoidal obstruction syndrome. Am J Physiol Gastrointest Liver Physiol. Jun 2003;284(6):G1045-1052.
    [27]Coppell JA, Brown SA, Perry DJ. Veno-occlusive disease:cytokines, genetics, and haemostasis. Blood Rev. Jun 2003;17(2):63-70.
    [28]Gharib MI, Bulley SR, Doyle JJ, et al. Venous occlusive disease in children. Thromb Res.2006; 118(1):27-38.
    [29]Iguchi A, Kobayashi R, Yoshida M, et al. Vascular endothelial growth factor (VEGF) is one of the cytokines causative and predictive of hepatic veno-occlusive disease (VOD) in stem cell transplantation. Bone Marrow Transplant. Jun 2001;27(11):1173-1180.
    [30]Bearman SI. The syndrome of hepatic veno-occlusive disease after marrow-transplantation. Blood. Jun 1 1995;85(11):3005-3020.
    [31]Litzow MR, Repoussis PD, Schroeder G, et al. Veno-occlusive disease of the liver after blood and marrow transplantation:analysis of pre-and post-transplant risk factors associated with severity and results of therapy with tissue plasminogen activator. Leuk Lymphoma. Nov 2002;43(11):2099-2107.
    [32]Gooley TA, Rajvanshi P, Schoch HG, et al. Serum bilirubin levels and mortality after myeloablative allogeneic hematopoietic cell transplantation. Hepatology. Feb 2005;41(2):345-352.
    [33]Tanikawa S, Mori S, Ohhashi K, et al. Predictive markers for hepatic veno-occlusive disease after hematopoietic stem cell transplantation in adults: a prospective single center study. Bone Marrow Transplant. Oct 2000;26(8):881-886.
    [34]Eltumi M, Trivedi P, Hobbs JR, et al. Monitoring of veno-occlusive disease after bone marrow transplantation by serum aminopropeptide of type Ⅲ procollagen. Lancet. Aug 28 1993;342(8870):518-521.
    [35]DeLeve LD, Shulman HM, McDonald GB. Toxic injury to hepatic sinusoids: sinusoidal obstruction syndrome (veno-occlusive disease). Semin Liver Dis. Feb 2002;22(1):27-42.
    [36]Rio B, Bauduer F, Arrago JP, et al. N-terminal peptide of type Ⅲ procollagen: a marker for the development of hepatic veno-occlusive disease after BMT and a basis for determining the timing of prophylactic heparin. Bone Marrow Transplant. Jun 1993; 11(6):471-472.
    [37]Nurnberger W, Michelmann I, Burdach S, et al. Endothelial dysfunction after bone marrow transplantation:increase of soluble thrombomodulin and PAI-1 in patients with multiple transplant-related complications. Ann Hematol. Feb 1998;76(2):61-65.
    [38]Petaja J, Pitkanen S, Vettenranta K, et al. Serum tumor marker CA 125 is an early and sensitive indicator of veno-occlusive disease in children undergoing bone marrow transplantation. Clin Cancer Res. Feb 2000;6(2):531-535.
    [39]Carreras E, Garcia-Pagan JC, Bosch J, et al. Transvenous liver biopsies in marrow transplant recipients. Transplantation. Dec 15 1995;60(11):1375.
    [40]Shulman HM, Gooley T, Dudley MD, et al. Utility of transvenous liver biopsies and wedged hepatic venous pressure measurements in sixty marrow transplant recipients. Transplantation. Apr 15 1995;59(7):1015-1022.
    [41]Senzolo M, Burra P, Cholongitas E, et al. The transjugular route:the key hole to the liver world. Dig Liver Dis. Feb 2007;39(2):105-116.
    [42]Bearman SI, Anderson GL, Mori M, et al. Venoocclusive disease of the liver: development of a model for predicting fatal outcome after marrow transplantation. J Clin Oncol. Sep 1993; 11 (9):1729-1736.
    [43]柴国君,洪丽霞,刘迎娣.肝静脉造影测压在肝小静脉闭塞病中的应用价值.武警医学.2007(11):828-830.
    [44]Cholongitas E, Quaglia A, Samonakis D, et al. Transjugular liver biopsy:how good is it for accurate histological interpretation? Gut. Dec 2006;55(12):1789-1794.
    [45]Blostein MD, Paltiel OB, Thibault A, et al. A comparison of clinical criteria for the diagnosis of veno-occlusive disease of the liver after bone marrow transplantation. Bone Marrow Transplant. Nov 1992;10(5):439-443.
    [46]McDonald GB, Sharma P, Matthews DE, et al. Venocclusive disease of the liver after bone marrow transplantation:diagnosis, incidence, and predisposing factors. Hepatology. Jan-Feb 1984;4(1):116-122.
    [47]彭涛,刘玉兰Budd-Chiari综合征、肝小静脉闭塞病与肝硬化的鉴别.胃肠病学.2007(12):770-773.
    [48]Corbacioglu S, Greil J, Peters C, et al. Defibrotide in the treatment of children with veno-occlusive disease (VOD):a retrospective multicentre study demonstrates therapeutic efficacy upon early intervention. Bone Marrow Transplant. Jan 2004;33(2):189-195.
    [49]Bairey O, Kirgner I, Yakobi M, et al. Clinical severe hepatic venoocclusive disease during induction treatment of acute monoblastic leukemia managed with defibrotide. Am J Hematol. Apr 2002;69(4):281-284.
    [50]Chopra R, Eaton JD, Grassi A, et al. Defibrotide for the treatment of hepatic veno-occlusive disease:results of the European compassionate-use study. Br J Haematol. Dec 2000; 111 (4):1122-1129.
    [51]Richardson P, Soiffer RJ. Antin JH, et al. Defibrotide (DF) for the treatment of severe veno-occlusive disease (sVOD) and multi-organ failure (MOF) post SCT:Final results of a multi-center, randomized, dose-finding trial. Blood. Nov 162006;108(11):17a-18a.
    [52]Simon M, Hahn T. Ford LA, et al. Retrospective multivariate analysis of hepatic veno-occlusive disease after blood or marrow transplantation:possible beneficial use of low molecular weight heparin. Bone Marrow Transplant. Mar 2001;27(6):627-633.
    [53]Imran H, Tleyjeh IM, Zirakzadeh A, et al. Use of prophylactic anticoagulation and the risk of hepatic veno-occlusive disease in patients undergoing hematopoietic stem cell transplantation:a systematic review and meta-analysis. Bone Marrow Transplant. Apr 2006;37(7):677-686.
    [54]Batsis I, Yannaki E, Kaloyannidis P, et al. Veno-occlusive disease prophylaxis with fresh frozen plasma and heparin in bone marrow transplantation. Thromb Res.2006; 118(5):611-618.
    [55]Bearman SI, Lee JL, Baron AE, et al. Treatment of hepatic venocclusive disease with recombinant human tissue plasminogen activator and heparin in 42 marrow transplant patients. Blood. Mar 1 1997;89(5):1501-1506.
    [56]Bajwa RP, Cant AJ, Abinun M, et al. Recombinant tissue plasminogen activator for treatment of hepatic veno-occlusive disease following bone marrow transplantation in children:effectiveness and a scoring system for initiating treatment. Bone Marrow Transplant. Apr 2003;31(7):591-597.
    [57]Ruutu T, Eriksson B, Remes K, et al. Ursodeoxycholic acid for the prevention of hepatic complications in allogeneic stem cell transplantation. Blood. Sep 15 2002; 100(6):1977-1983.
    [58]Khoury H, Adkins D, Brown R, et al. Does early treatment with high-dose methylprednisolone alter the course of hepatic regimen-related toxicity? Bone Marrow Transpl. Apr 2000;25(7):737-743.
    [59]Akyuz C, CaClar K, Emir S, et al. High-dose methylprednisolone treatment of hepatic veno-occlusive disease in a child with Wilms tumor. Pediatr Hematol Oncol. Jun 2003;20(4):345-349.
    [60]A1 Beihany A, Al Omar H, Sahovic E, et al. Successful treatment of hepatic veno-occlusive disease after myeloablative allogeneic hematopoietic stem cell transplantation by early administration of a short course of methylprednisolone. Bone Marrow Transplant. Feb 2008;41(3):287-291.
    [61]房龙,樊艳华,王晓娣,等.肝小静脉闭塞病2例并文献复习.胃肠病学和肝病学杂志.2008(06):513-516.
    [62]Essell JH, Schroeder MT, Harman GS, et al. Ursodiol prophylaxis against hepatic complications of allogeneic bone marrow transplantation. A randomized, double-blind, placebo-controlled trial. Ann Intern Med. Jun 15 1998;128(12 Pt 1):975-981.
    [63]Ishida H, Nakayasu H, Nukaya H, et al. Study of the pharmacological effect of the bile salt, sodium scymnol sulfate, from Rhizoprionodon acutus. Ⅲ. Protective effect of scymnol against vascular endothelial cell damage in a rat peripheral arterial occlusion model. Biol Pharm Bull. Aug 1999;22(8):822-827.
    [64]Neuman MG, Shear NH, Bellentani S, et al. Role of cytokines in ethanol-induced cytotoxicity in vitro in Hep G2 cells. Gastroenterology. Jul 1998;115(1):157-166.
    [65]Gluckman E, Jolivet I, Scrobohaci ML, et al. Use of prostaglandin E1 for prevention of liver veno-occlusive disease in leukaemic patients treated by allogeneic bone marrow transplantation. Br J Haematol. Mar 1990;74(3):277-281.
    [66]Bearman SI, Shen DD, Hinds MS, et al. A phase I/II study of prostaglandin E1 for the prevention of hepatic venocclusive disease after bone marrow transplantation. Br J Haematol. Aug 1993;84(4):724-730.
    [67]Ringden O, Remberger M, Lehmann S, et al. N-acetylcysteine for hepatic veno-occlusive disease after allogeneic stem cell transplantation. Bone Marrow Transplant. May 2000;25(9):993-996.
    [68]Tefferi A, Kumar S, Wolf RC, et al. Charcoal hemofiltration for hepatic veno-occlusive disease after hematopoietic stem cell transplantation. Bone Marrow Transplant. Nov 2001;28(10):997-999.
    [69]Chen Z, Huo JR, Zhu HY. Effect of ligustrazine on mice model of hepatic veno-occlusive disease induced by Sedum aizoon. J Gastroenterol Hepatol. Jan 202011.
    [70]赵婷,吴彤,陆道培.新疆紫草的乙醇提取物抗肝窦阻塞综合征作用的实验研究.中国药学杂志.2005(21).
    [71]张智勤,赵俊彪,潘锦辉,等.中西医结合治疗肝小静脉闭塞病17例.浙江中医杂志.2007(09):526.