化纤汤对博莱霉素致大鼠肺纤维化干预作用的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的:肺间质纤维化是以弥漫性肺泡炎和肺泡结构紊乱最终导致肺间质纤维化为主要特征的疾病。目前其发病机制尚未完全阐明,又无确切有效的药物治疗。探讨中医药对肺纤维化的防治具有广阔的前景和深远的意义。本课题采用大鼠一次性气管内滴注博莱霉素法造模,通过观察大鼠肺组织的形态学变化、肺泡Ⅱ型上皮细胞凋亡率变化、血清中IL-6和TNF-α的表达水平、肺组织中CTGF的表达、肺组织匀浆中SOD和GSH-PX的活性及MDA的含量,来探讨肺间质纤维化的发病机制,并观察具有“益气通阳、行气活血化痰”功效的化纤汤对这些指标的影响,以探讨化纤汤对大鼠肺纤维化的保护作用及可能机制,为寻求中医中药对肺纤维化的防治进行实验研究。
     方法:健康Wistar大鼠240只,体重180-220克,随机分为正常对照组(A组)、模型组(B组)、强的松干预组(C组)、化纤汤大剂量(D组)、中剂量组(E组)、小剂量(F组)干预组共6组。A、B、C、D四组每组各48只(其中24只用于肺泡Ⅱ型上皮细胞的提取),E、F组各24只。采用一次性气管内滴注博莱霉素(5mg/kg·bw)法造模,各组均于造模后第二天开始灌胃给药。C组每日一次给予醋酸强的松混悬液(0.32mg/ml)灌胃,D、E、F组每日一次给予化纤汤(浓度分别是2g/ml、1g/ml、0.5g/ml)灌胃,A、B组每日给予一次等容积的生理盐水灌胃,其中强的松和化纤汤用量相当于成人常用量,按体表面积折算。分别于第7、14、28天随机处死A、B、C、D四组的大鼠各8只,取材用于肺泡Ⅱ型上皮细胞的分离和纯化;其余各组大鼠亦于第7、14、28天随机处死8只,用于形态学观察及血清、组织匀浆的制备。①在光镜及电镜下观察肺组织病理形态学的变化及超微结构的改变;②用流式细胞仪检测肺泡Ⅱ型上皮细胞凋亡率的变化情况。③用ELISA法检测血清中IL-6、TNF-α表达水平的变化;④用免疫组化SABC法检测肺组织中CTGF的表达;⑤分别用TBA法和比色法检测肺组织匀浆中SOD和GSH-PX的活性及MDA的含量。
     结果:①肺大体形态改变:各时间点,A组大鼠双肺呈粉红色,表面光滑,弹性好。B组,第7天,双肺见密集的点状出血灶样瘀斑,肺体积增大;第14天,双肺呈灰白色,局部可见大小不等的结节样改变及陈旧性出血点,肺组织较硬;第28天,双肺苍白,体积缩小,触及较硬,表面结节样改变及条索状凹沟。与B组比较,第7天时,C、D、E、F组两肺外观病理变化明显好于B组,两肺稍暗红,弹性可,其中D、C、E组明显好于F组;第14-28天,D、E组大部分动物两肺颜色稍暗,弹性尚可,偶可见部分肺叶有散在的点状灰色斑块,F、C与B组病变相近。②形态学改变:第7天,B组大鼠肺泡壁增厚,伴有炎性细胞浸润,间质内成纤维细胞增多,病灶内肺泡萎缩,肺泡内大量炎性细胞浸润;第14天,肺泡间隔明显增宽,成纤维化和胶原基质明显增多;第28天,肺泡结构破坏,肺泡壁显著增厚,肺间质纤维化瘢痕形成,毛细血管腔闭塞。超微病理示:B组大鼠第7天,肺泡Ⅱ型上皮细胞增生、肿胀,线粒体肿胀,嵴缩短、断裂或消失,甚至空泡样变性,板层小体数量增多,但是多呈空泡样,细胞微绒毛破坏、稀少或消失;第14-28天,肺泡Ⅱ型细胞数量较第7天减少,核形态不规则,线粒体嵴断裂或消失,大多呈空泡样变性,数量明显减少,板层小体量也减少,大多呈空泡样变。与B组比较,这些病理变化,第7天,D组改善最明显,其次为C、E、F组;第14天,D组改善最明显,其次为E、C、F组;第28天,D组改善最明显,其次为E、F、C组,其中第7天、28天肺泡炎、肺纤维化Szapiel分级比较具有显著性差异(P<0.01或0.05)。③与A组比较,各时间点,各组大鼠肺泡Ⅱ型上皮细胞凋亡率显著升高(P<0.05)。与B组比较,第7天时,C、D两组凋亡率均有显著降低(P<0.05);第14-28天,C组肺泡Ⅱ型上皮细胞凋亡率有所下降,但统计学无显著差异(P>0.05),D组肺泡Ⅱ型上皮细胞凋亡率有显著减少(P<0.05)。④与A组比较,各时间点各组大鼠血清中TNF-α、IL-6水平明显增高。与B组比较,第7天,C、D、E、F各组大鼠血清中TNF-α、IL-6的水平显著降低(P<0.01或0.05);第14-28天,D、E组大鼠血清中TNF-α、IL-6水平明显降低(P<0.01或0.05),C、F组虽有降低,但比较差异无统计学意义(P>0.05)。⑤与A组比较,各时间点各组大鼠肺组织中CTGF的表达明显增高(P<0.01)。与B组比较,各时间点,C、D、E各组CTGF表达均低于B组(P<0.01或0.05),F组低于B组,但比较差异无统计学意义(P>0.05)。⑥与A组比较,各时间点,B、C、D、E、F各组大鼠肺组织中GSH-PX、SOD的活性均降低,MDA的含量均增高,存在显著差异(P<0.01或0.05)。与B组比较,各时间点,D、E组大鼠GSH-PX与SOD的活性均升高,MDA含量均降低,存在显著差异(P<0.01或0.05);第7-14天,C组大鼠GSH-PX与SOD的活性均升高,MDA含量均降低,存在明显差异(P<0.05),第28天,C组与B组无明显差异(P>0.05)。各时间点,F组与B组比较无明显差异(P>0.05)。
     结论:①模型组病理特征变化与文献报道一致,说明造模成功;化纤汤对肺纤维化发展各个阶段的病理变化有明显的改善作用,且大、中剂量组明显优于小剂量组。②ATⅡ细胞过度凋亡可能促进肺泡炎症反应并导致炎症后纤维增生;化纤汤对ATⅡ细胞可能有抗凋亡作用,可能通过抑制ATⅡ细胞的过度凋亡而起到保护和修复肺泡结构及功能的作用,从而减轻BLM对大鼠肺组织的损伤,延缓并减轻肺纤维化的发生发展。③化纤汤能降低肺纤维化大鼠血清中TNF-α、IL-6水平,可能通过下调上述细胞因子水平而对博莱霉素致大鼠的肺损伤起保护作用,减轻肺纤维化形成的程度,化纤汤大、中剂量组疗效优于小剂量组。④CTGF可能在肺泡炎、肺纤维化的发生发展过程中起着重要的作用,其表达程度与肺纤维化的程度密切相关;化纤汤可能通过抑制CTGF的表达对肺组织起着修复及保护作用。⑤化纤汤能调节肺纤维化大鼠体内自由基水平,减轻自由基对肺组织结构的氧化损伤,而且化纤汤大、中剂量组的作用优于小剂量组。
Object: Interstitial pulmonary fibrosis is a kind of disease mainly characterized by inflammation and structural destroy of alveolus, which finally changed into interstitial fibrosis. The therapeutic approach to this not particularly effective because of its unclear mechanism. Tranditional Chinese medicine has reached a several achievements in improving symptoms and reducing death rate, and it's necessary to summarize and improve the methods of TCM for this disease. Huaxian decoction(HD), a prescription of tonifying qi and regulating yang, smoothing qi activing blood and eliminating sputum, has shown a bright future in clinical study of pulmonary fibrosis. In this article, the rat model was induced by blyomicin and we evaluated HD's curative effect objectively and explore its way and mechanism by observing the changing of apoptosis of ATⅡcells, the content of TNF-αand IL-6 in sera, the expression of CTGF, the level of SOD, GSH-PX and MDA in lung, the morphological alternation of the lung tissue.
     Methods: Two hundreds and forty healthy Wistar rats, weight from 180 to 220gram were randomly divided into six groups: normal control group(group A, n=48), model group(group B, n=48), prednisone group(group C, n=48), Chinese drugs of high(group D, n=48), moderate (group E, n=24) and low dosage (group F, n=24). The rat model was induced by blyomicin and respective medicine was given to each group. Every group was subdivided into the three groups which was killed in the seventh, fourteenth, and twenty-eighth day respectively.①The rat's lung tissue section was observed under light microscope and electron microscope to study the morphological alternation.②Find the changes of apoptosis of ATⅡcells by flow cytometry.③Assay the sera level of TNF-αand IL-6 by ELISA method.④Detect the expression of CTGF in the lung tissue with immunohistochemical method of analysis.⑤TBA method and colorimetric method were used to measure the content of SOD, GSH-PX and MDA in lung tissue respectively.
     Results:①Changes of pathologic morphosis under light microscope in the rat lung fibrosis model: in the 7th day after model was made, interval between alveolus became thicker,, the number of fibroblast increased and alveoli in the focus of infection were atrophied and lots of inflammation cells infiltrated; in the 14th day after model was made, interval between alveolus became thicker and thicker, fibroblast and collagen matrix increased markedly; in the 28th day after model was made, alveoli structure was damaged, the wall of alveoli became thicker remarkably, interstitial fibrotic cicatrix came into being and capillary blocked. Changes under electron microscope in the 7th and 14th day showed that mitochondria of typeⅡalveolar (epithelial) cells became hyperplastic and swollen and its cristae decurtated, collapsed and vanished. Quantities of lamellar body decreased and became vacuole. Microvilli damaged, lessened and disappeared. In the 28th day, quantity of typeⅡalveolar (epithelial) cells decreased more than that in the 7th day or in the 14th day. Cristae in mitochondria and lamellar body damaged and degenerated like vacuole. In the 7th day, amelioration of pathological changes from the most obviously to the most slightly was D, C, E, F, and that in the 14th day was D, E, C, F, that in the 28th day was D, E, F, C. There was significant difference among Szapiel grade of alveolitis and fibrosis in each group (P<0.01 or 0.05).②the changes of apoptosis of ATⅡcells in group C, D in each phase was lower than that in groupB (P<0.05).③The sera level of TNF-αand IL-6 in group C, D, E, F in each phase was lower than that in group B respectively(P<0.01) in the 7th, but only that in group D, E was higher than that in group B in the 14th -28th (P<0.01 or 0.05).④The expression of CTGF in the lung tissue in the group C, D, E in each phase was lower than that in groupB respectively (P<0.01 or 0.05).⑤Content of MDA in lung tissue in group C, D, E, F in each phase was lower than that in group B respectively(P<0.01or0.05), while that of GSH-PX and SOD was higher(P<0.01, 0.05). In the 7th and 14th day, content of MDA in lung tissue of degressive sequence was F, C, E, D, and in the 28th day was C, F, E, D (P<0.01or 0.05), while that of GSH-PX was opposite.
     Conclusion:①The model was made successfully according to its corresponding manifestation of pathology to human's lung fibrosis and other reports.②HD has therapeutic effect on lung fibrosis in each phase.③HD can decrease the apoptosis of ATⅡcells.④HD can regulate the disbalance of cytokine such as decreacing sera TNF-α, IL-6 level.⑤HD can inhibit the expression of CTGF in lung tissue.⑥HD can decrease harmful free radical and increase beneficial free radical scavenger in lung tissue.
引文
1. American Thoracic Society(ATS). Idiopathic pulmonary fibrosis: diagnosis and treatment. International Consensus Statement. Am J Respir Crit Care Med, 2000, 161:646-664
    2.李仪奎.中药药理实验方法学.上海:上海科学技术出版社,1991.36
    3. Szapiel SV, Elson NA, Fulmer JD, etal. Bleomycin-induced interstitial pulmonary disease in the nude, athymic mouse. Am Rev Respir Dis, 1979, 120(4):893-899
    4. Thrall RS. Differential cellular analysis of bronchoalveolar lavage fluid obtained aat various stages during the development of bleomycininduced pulmonary in the rat. Am Tev Respir Dis, 1982, 126:488
    5. Clark JG. BLM-induced pulmonary fibrosis in hamster. J Clin Invest, 1983, 72:2082-2088
    6. Thrall R, Swendsen CL, Shannon TH, et al. Correlation of change in pulmonary surfactant phospholipids with compliance in bleomycin induced pulmonary fibrosis in the rat. Am Respir Dis, 1987, 136:113-117
    7.赵俭,王红曼,王化洲.川芎嗪对BLM所致大鼠肺纤维化保护作用的电镜研究[J].2006,27(2):11-13
    8.王响英,吴淑燕,李苏安,等.实验性大鼠肺纤维化病理形态及超微结构观察[J].苏州大学学报(医学版),2005,25(3):379-382
    9. Kumar RK. Morphological methods for assessment of fibrosis. Methods Mol Med. 2005, 117:179-188
    10.张天嵩,韩镭.近十年来中医药治疗肺间质纤维化临床研究进展[J].浙江中医杂志,2002,35:452
    1. Barbas-Filho JV, Ferreira MA, Sesso A, et al. Evidence of typeⅡ pneumoeyte apoptosis in the pathogenesis of idiopathic pulmonary fibrosis(IPF)/usual interstitisl pneumonia(UIP). J Clin Pathol, 2001, 54:132-138
    2. HengartherMO. The. biochemistryofa poptosis [J].Nature, 2000, 407 (6805):770-776
    3. Dobbs LG, Gonzalez R, Williams MC.An imporoved method for isolating type 2 cells in high yield and purity. Am Rev Rispir Dis, 1986, 134-141
    4.卢韶华,张农,张秀荣,等.博莱霉素诱导大鼠实验性肺纤维化肺 泡Ⅱ型上皮细胞MMP-2和MTI-MMPmRNA表达的动态变化[J].复旦大学学报(医学科学版),2001,28(6):501-504
    5. KasperM, Haroske G. Alterations in the alveolar epithelium after injury leading to pulmonary fibrosis. Histol Histopathol, 1996, 11:463-483
    6.崔光彬,魏经国,王玮,等.博莱霉素对肺泡Ⅱ型上皮细胞增殖及功能的影响[J].第四军医大学学报,2001,22(12):1073-1076
    7.孔璐,王志刚,牛建昭,等.博莱霉素对大鼠肺泡Ⅱ型上皮细胞功能的损伤[J].中国医学科学院学报,2005,27(2):81-85
    8. Lesur O, Arsalane K, Lung alveolar epithelial cell migiration in vitro: modulators and regulation processes [J]. Am J Physiol, 1996, 330:L311-319
    9. Kremler SG, Vmstesd TM, Phelps DS, Am J Physiol, 1997, 272(5):996-1004
    10. Cott G, Westcott JY, Voelkel NF et al. Am J Physiol, 1990, 258(4): 179-187
    11.孔璐,王继峰,牛建昭.肺纤维化与细胞凋亡[J].中华结核和呼吸杂志,2004,27(6):368-370
    12. FineA, Janssen-Heiningger Y, Soultanakis RP, et al. Apoptosis in lung pathophysilogy[J]. Am J Physiol Lung Cell Mol Physiol, 2000, 279(3):423-427
    13. Liying W James M A, Yon R, et al. Potential role of apoptosis macrophages in pulmonary inglammation and fibrosis[J]. J Cellular Physiol, 2003, 194(2):215-224
    14. Michael J, Mabe JR, Mechanisms and consequences of silica-induced apoptosis[J]. Toxicological Sciences, 2003, 76:1-2]
    15. KuwanoK, HagimotoN, TanakaT, et al. Expression of apoptosis regulatory genes in epithelial cells in pulmonary fibrosis in mice[J]. Pathol, 2000, 190(2):221-229.
    16. Barbas-Filho JV, Ferreira MA, Sesso A, et al. Evidence of typeⅡ pneumocyte apoptosis in the pathogenesis of idiopathic pulmonary fibrosis (IPF)/usual interstitial pneumonia(UIP)[J]. J Clin Pathol, 2001, 54(2):132-138
    17. Aljandali A, Pollack H, Yeldandi A, Asbestos causes apoptosis in alveolar epithelial cells: roles of iron-induced free radicals[J]. J Lab Clin Med, 2001, 137(5):330-339
    18. FineA, Janssen-HeiningerY, Soultanakis RP, et al. Apoptosis in lung pathophysiology[J]. AmJ Physiol Lung Cell Mol Physiol, 2000, 279(3):L423-427)
    1. Bissonnette E, Rola PM. Pulmonary inflammation and fibrosis in a murine model of asbestosis snd silicosis possible role of tumor necrosis factor. Inflammation, 1989, 13(3):329-339
    2. Khalil N, Bereznay, Sporn M, et al. Macrophage production of transforming growth factor and fibroblast collagen synthesis in chronic pulmonary inflammation.J Ecp Med.1992, 170:727-737
    3. Chensue SW, et al. Type1/type2 cytokine paradigin and the progression of pulmonary fibrosis. Chest. 2001, 120(1):5-8]
    4. Piguet PF, RibauxC, karpuzV, et al. Expression and localization of tumor necrosis factor-alpha and its mRNA in idiopathic pulmonary fibrosis[J]. Am J Pathol, 1993,143:651-655.
    5. Jirk FR, Podor TJ, Hirano T, et al. Bacterial and inflammatory mediators augment IL-6 secretion by human endothelial cells[J]. Immunnol, 1989,142(1): 144-148
    6. Kapanci Y, Desmouliere A, Pache JC, et al. Cytoskeletal pretein modulation in pulmonary alveolar myo-fibroblasis during idiopathic pulmonary fibrosis factor alpha[J]. Am J Respir Crit Care Med, 1995,152:22163-22169.
    7. Wang R, Alam G, Zagrariya A, et al. Apoptosis of lung epithelial cells in response to TNF-alpha requires angiotension Ⅱ generation denovo[J]. J Cell Physiol,2000,185:253-259.
    8. Kuwano K, Kunitake R, Kawasaki M, et al. P21 wafl/cipl/sdil and p53 expression in association with DNA strand breaks in idiopathic pulmonary fibrosis [J]. Am J Respir Crit Care Med,1996,154(2ptl):477-483.
    9.彭丽萍,左梦华,迟宝荣.甲基强的松龙治疗特发性肺间质纤维化其前后TNF-α和IL-8的变化[J].吉林大学学报(医学版),2004,30(2):286-288
    10.邵洪波,吴厚生,倪赞明.SLE患者外周血单个核细胞分泌IL-6和IgG的研究[J].中国免疫学杂志,1993,9(2):121-123
    1. Blom IE, Van Dijk A J, WietenL, et al. In vitroevidence for differential involvement of CTGF, TGF-β and PDGF-BB in mesangial response to injury. Nephrol Dial Transplant, 2001,16(6):1139-1148
    2. Grocendorst GR. Connective Tissue Growth Factor:a mediator of TGF-beta action on fibrolasts Cytokine Growth Factor Rev, 1997,8(3): 171-179
    3. Bramdham DM,Igarashi A, Potter RL, et al. Connective tissue growth factor: a cysteine-rich mitogen secreted by human vascuclar endothelial cells is related to the SRC-induced immediate early gene product CEF-10[J].J Cell Biol,1991,114(6):1285-1294
    4. Holmes A, Abraham DJ, Susan S, et al. CTGF and AMADs, Maintenance of sclerpderma phenotype is independent of SMADs signaling [J].J Biol Chem,2001,276:10594-10601
    5. Kireeva ML, Latinkic BV, Kolesnikova TV, et al. Cyrb1 and Fispl2 are both ECM-associated dignaling mokecules: activities, metabolism and localization during development[J]. Exp Cell Res, 1997,233(1):63-77
    6. Flanders KC, Smsds as a mediator of the fibrotic response. Int J Exp Pathol,2004,85:47-64]
    7. Grotendoral GR, Rahmanie H, Duncan MR Combinatorial aignaling pathways delermine fibroblast proliferation and myofibroblast differentialion. FASEBJ, 2004,8:469-479]
    8. Crean JKG, FinelayD,Murphy M, et al. The role of P42/P44 MAPK and protein kinase B in connective tissue growth factor induced extracelluar matrix protein Production, cell migration, and action cytoskeletal rearrangement in human mesangial cells[J],J Biol Chem,2002, 277 (46):44187-44194
    9. Mori T, KawaraS, Shinozki M, et al. Role and interaction of connective tissue growth factor with transforming growth factor-β in persistent fibrosis:a mouse fibrosis model[J]. J Cell Physiol, 1999,181 (1): 153-159.
    10.叶旭军,徐启勇,叶燕青.结缔组织生长因子在大鼠肺纤维化模型中的蛋白表达[J].微循环学杂志,2004,14(1):16-17
    1.赵亚滨,张玉霞,王常慧,等.N-乙酰半胱氨酸对博莱霉素大鼠肺脏氧化.还原状态的影响[J].中国医科大学学报,2002,1(6):422-425
    2. Inghilleri S, Morbini P, Oggionni T, Barui S, Fenoglio C. In situassessment of oxidant and nitrogenic stress in bleomycin pulmonary fibrosis,Histochem Cell Biol,2005, Nov 24:1-9
    3. Cheryl LF, Lisa MS, Tim Do. Extracellular superoxide dismutatse in biology and medicine[J]. Free Rad Bid Med, 2003, 35(3):236-256
    4. Hong JS, KoHH, HanES, et al. Inhibition of bleomycin-induced cell death in rat alveolar marcophages and human lung epithelial cells by ambroxol[J]. Biochem Pharmacol,2003,66(7): 1297-1306
    5. Weiss SJ. Tissue destruction by neutrophils[J]. N Engl J Med,1989, 320:365-376
    6.胡一鸿,牛健康.超氧化物歧化酶研究进展[J].生物学教学,2005,30(1):2-4
    1. Clark JG, Kostal KM, Marino BA. Bleomycin-induced pulmonary fibrosis in hamsters. An alveolar macrophage product increases fibroblast prostaglandin E2 and cyclic adenosine monophosphate and suppresses fibroblast proliferation and collagen production[J]. J Clin Invest, 1983,72(6):2082-2091.
    2. Thrall R, Swendsen CL, Shannon TH, et al. Correlation of change in pulmonary surfactant phospholipids with compliance in bleomycin induced pulmonary fibrosis in the rat[J]. Am Respir Dis, 1987, 136:113-117
    3.赵俭,王红曼,王化洲.川芎嗪对BLM所致大鼠肺纤维化保护作用的电镜研究.2006,27(2):11-13
    4.王响英,吴淑燕,李苏安,等.实验性大鼠肺纤维化病理形态及超微结构观察[J].苏州大学学报(医学版),2005,25(3):379-382
    5.张天嵩,韩镭.近十年来中医药治疗肺间质纤维化临床研究进展[J].浙江中医杂志,2002,35:452
    6. Fine A, Janssen-Heininger Y, Soultanakis RP, et al. Apoptosis in lung pathophysiology[J]. Am J Physiol Lung Cell Mol Physiol. 2000,279(3):L423-427
    7. Kuwano K, Hagimoto N, Kawasaki M, et al. Essential roles of the Fas-Fas ligand pathway in the development of pulmonary fibrosis[J]. J Clin Invest, 1999,104(1):13-19.
    8. Kuwano K, Kunitake R, Maeyama T, et al. Attenuation of bleomycin-induced pneumopathy in mice by a caspase inhibitor[J]. Am J Physiol Lung Cell Mol Physiol. 2001,280:L316-325
    9. Barbas-Filho JV, Ferreira MA, Sesso A, et al. Evidence of typeⅡ pneumoeyte apoptosis in the pathogenesis of idiopathic pulmonary fibrosis(IPF)/usual interstitisl pneumonia(UIP) [J]. J Clin Pathol, 2001,54:132-138
    10.赵勇,曾庆富,钱仲裴.实验性矽肺早期病变中肺泡Ⅱ型细胞增生的病理学意义[J].中华劳动卫生职业病杂志,2001,19(4):295-297
    11.赵亚滨,张玉霞,王常慧,等.N-乙酰半胱氨酸对博莱霉素大鼠肺脏氧化-还原状态的影响[J].中国医科大学学报,2002,31(6):422-425
    12. Inghilleri S, Morbini P, Oggionni T, Barui S, Fenoglio C. In situassessment of oxidant and nitrogenic stress in bleomycin pulmonary fibrosis, Histochem Cell Biol, 2005, Nov 24:1-9
    13. Cheryl LF, Lisa MS, Tim Do. Extracellular superoxide dismutatse in biology and medicine[J]. Free Rad Bid Med, 2003,35(3):236-256
    14. Weiss SJ. Tissue destruction by neutrophils[J]. N Engl J Med,1989,320:365-376
    15. Kang H, Ahn KS, Cho C, et al. Immunomodulatory effect of Astragali radix tract on murine Th1/Th2 cell linage development[J]. Biol Pharma Bulletin, 2004,27(12):1946-1950]
    16.蒋云峰.黄芪桃红汤治疗特发性肺纤维化24例[J].吉林中医药.2003,23(11):1
    17.戴令娟,侯杰,蔡后荣.中药和中西药结合治疗肺纤维化的实验研究[J].中国中西医结合杂志,2004,24(2):130-132
    18.颜润,俞雷,张义雄,等.黄芪当归合剂对大鼠肾间质纤维化的影响及 机制探讨[J].贵州医药,2006,30(1):8-10]
    19.周贤、戴立里,贾丽萍,等.黄芪注射液对肝纤维化抑制作用的实验研究[J].中华肝病杂志,2005,13(8):575-578
    20.蒋春明,张苗,孙峥.黄芪对腹膜间皮细胞致纤维化细胞生长因子分泌与表达的影响[J].医学研究生学报,2005,18(11):972-976
    21. Yang YZ, Jin PY, Gu Q, etal. Effects of astragalus membrane-cells on natural killer cell activity and induction of A- and C- interfere on in-patients with coxsackie B viral myocarditis. Chin Meal J, 1990,103 (4):304-307
    22.孙成文,钟国赣,江岩,等.黄芪多糖抗氧化损伤的作用[J].中国药理学通报,1996,12(2):161-163
    23.何明,张晓梅,苑惠青,等.银杏黄酮甙治疗肺间质纤维化疗效与免疫细胞因子相关性研究[J].北京中医药大学学报(中医临床版),2005.5:6-9
    24.何明,张晓梅,苑惠青,等.银杏叶提取物治疗肺间质纤维化的临床研究[J].中国中西医结合杂志,2005,25(3):201-204
    25.易文龙,陈建明.水蛭、桃仁、黄芪、当归合剂对小鼠血吸虫性肝纤维化的影响[J].中国血吸虫病防治杂志,2004,16(1):322341
    26.盛丽,王莉,姚岚,等.水蛭、疏血通、甘利欣对博莱霉素小鼠肺纤维化的干预作用[J].中国工业医学杂志,2006,19(3):185-187
    27. Kumar RK. Morphological for assessment of fibrosis. Methods Mol Med,2005,117:179-188
    1.中华医学会呼吸病学分会.特发性肺(间质)纤维化诊断和治疗指南(草案)[J].中华结核和呼吸杂志,2002,25(7):387-388
    2.李芳,李廷谦.特发性肺间质纤维化的现代治疗研究进展[J].国外医学—呼吸分册,2003,23(1):45-49
    3. Travis WD, Matsui K, Moss J, et al. Idiopathic nonspecific interstitial pneumonia: prognostic significance of celluar and fibrosing patterns. Am J Surg Pathol, 2000,24(1): 19-33
    4.柴文戍,李永春,刘玉玲,等.博莱霉素致肺纤维化大鼠形态学变化的实验研究[J].中国实验动物学报,2003,11(2):78-80
    5.何冰,李桂莲,张烨,等.特发性肺纤维化的电镜观察及发病机理的探讨[J].中华结核和呼吸杂志,1999,22(1):27-29
    6.曾庆富,牛海艳.肺纤维化机制的研究进展[J].中华病理学杂志,2001.30(5):371-373
    7. Hagimoto BD. Apoptosis in lung fiSrosis and repair[J]. Chest, 2002, 122(6Suppl):S293-298
    8.佟振月,侯立新,马跃文,等.肺纤维化细胞间黏附分子-1基因产物表达的作用[J].中华结核和呼吸杂志,2002,23(6):363-364
    9.赵勇,曾庆富,钱仲裴.实验性矽肺早期病变中肺泡Ⅱ型细胞增生的病理学意义[J].中华劳动卫生职业病杂志,2001,19(4):295-297
    10. Westergren Thorssor GA. Altered expression of small proteoglycans, collagen and TGF-β in developing Bleomyin-induced pulmonary fibrosis in rats. J Clin Imest,1993,92(3):632-637
    11. Sanner BM, Kollhosser P, Buechner N, et al. Influence of treatment on leptin levels in patients with obstructive sleep apnoea. Eur Respir J, 2004,23 (4):601- 604.
    12.吴晓梅,王欣燕,陈复辉.两种细胞因子与特发性肺纤维化的关系[J]. 中国危重病急救学,2003,15(6):362-364.
    13. Bramdham DM, Igarashi A, Potter RL, et al. Connective tissue growth factor: a cysteine-rich mitogen secreted by human vascuclar endothelial cells is related to the SRC-induced immediate early gene product CEF-10[J]. J Cell Biol,1991,114(6):1285-1294
    14. Crean JKG, FinelayD, Murphy M, et al. The role of P42/P44 MAPK and protein kinase B in connective tissue growth factor induced extracelluar matrix protein Production, cell migration, and action cytoskeletal rearrangement in human mesangial cells[J], J Biol Chem, 2002,277(46):44187-44194
    15. Mori T, Kawara S, Shinozki M, et al. Role and interaction of connective tissue growth factor with transforming growth factor- β in persistent fibrosis: a mouse fibrosis model[J]. J Cell Physiol, 1999, 181(1): 153-159.
    16.叶旭军,徐启勇,叶燕青.结缔组织生长因子在大鼠肺纤维化模型中的蛋白表达[J].微循环学杂志,2004,14(1):16-17
    17.邵洪波,吴厚生,倪赞明.SLE患者外周血单个核细胞分泌IL-6和IgG的研究[J].中国免疫学杂志,1993,9(2):121-123
    18. Piguet PF, RibauxC, karpuzV, et al. Expression and localization of tumor necrosis factor-alpha and its mRNA in idiopathic pulmonary fibrosis[J]. Am J Pathol, 1993,143:651-655.
    19. Jirk FR, Podor TJ, Hirano T, et al. Bacterial and inflammatory mediators augment IL-6 secretion by human endothelial cells[J]. Immunnol, 1989,142(1):144-148
    20. Kapanci Y, Desmouliere A, Pache JC, et al. Cytoskeletal pretein modulation in pulmonary alveolar myo-fibroblasis during idiopathic pulmonary fibrosis factor alpha[J]. Am J Respir Crit Care Med, 1995,152:22163-22169.
    21. Wang R, Alam G, Zagrariya A, et al. Apoptosis of lung epithelial cells in response to TNF-alpha requires angiotension Ⅱ generation denovo[J]. J Cell Physiol,2000,185:253-259.
    22. Kuwano K, Kunitake R, Kawasaki M, et al. P21 wafl/cipl/sdil and p53 expression in association with DNA strand breaks in idiopathic pulmonary fibrosis [J]. Am J Respir Crit Care Med, 1996,154(2ptl):477-483.
    23. Strieter RM. Mechanisms of pulmonary fibrosis: conference summary. Chest, 2001,120(Suppl):77S-85S.
    24. Hung M, et al.[J]. J Clin Invest, 2002,109(7):931-937]
    25. Lee CG, et al. [J]. J Exp Med, 2001,194(6):809-821.]
    26. Kesne MP, et al.[J]. Am J Physiol Lung Cell Mol Physiol, 2001,281(1):L92-97.
    27. Madri J, Furthmayr OH. Collagen polymorphism in the lung. An immunohistochemical of study pulmonary fibrosis, Hum Pathol, 1985,4:353-366
    28. Raghow T, Streker LJ, Hudson LD et al. Extra cellular matrix in normal and fibrotic human lungs. Am Rev Respir Dis, 1985,131:281-289
    29.赵亚滨,张玉霞,王常慧,等.N-乙酰半胱氨酸对博莱霉素大鼠肺脏氧化-还原状态的影响[J].中国医科大学学报,2002,31(6):422-425
    30. Weiss SJ. Tissue destruction by neutrophils[J]. N Engl J Med,1989;320:365-376
    31. Entzian P, Schlaak M, Seitzer U, et al. Antiinflammatory and antifibrotic properties of colchicine: implications for idiopathic pulmonary fibrosis [J]. Lung, 1997,175:41-51.
    32. Douglas W, Ryu J, Swensen P, et al. Colchicine versus prednisone in the treatment of idiopathic pulmonary fibrosis. A randomized prospective study. Members of the Lung Study Group[J]. Am J Respir Crit Care Med,1998,158:220-225.
    33.陈宁,张石革.特发性肺间质纤维化的研究及其药物疗效的回顾[J].中国全科医学,2003,7(7):588
    34. Gillissen A, Nowak D. Characterization of N-acetylcysteine and am broxol in anti-oxidant therapy. Respir Med,1998,92(4):609-623
    35. Hagiwara SI, Ishii Y, Kitamura S, et al. Aeroslized administration of N-acctylaysteine attenuates lung fibrosis induced by bleomycin in mice. Am J Respir Crit Care Med, 2000, 162(1):225-231
    36. Behr J, Maier K, Degenkolb B, et al. Antioxidative and clinical effects of high-dose N-acetyl cysteine in fibrosing alveolitis. Adjunctive therapy to maintenance immunosuppression. Am J Respir Crit Card Med,1997,156(6):1897-1901
    37. Azuma A, nukiwa T, Tsuboj E, et al. Double-blind placebo-controlled trial of pirfenidone in patients with idiopathic pulmonary fibrosis[J]. Am J Respir Crit Care Med. 2005,171: 1040-1047.
    38. Piguet PF, Vesin C, Grau GE. Interleukin-1 receptor antagonist(IL-1ra) prevents or cures pulmonary fibrosis elicited in mice by bleomycin or silica. Cytokine, 1993,5(1):57-61].
    39. Ziesche R, Hotbauer E, Wittmann K, et al. Apreliminary study of long term treatment with interferon-gamma-1b and low dose prednisolone in patients with idiopathic pulmonary fibrosis. N Engl J Med,1999, 341:1264-1269
    40. Hagimoto N, Kuwano K, Inoshima I, et al. TGF-betal as an enhancer of Fas-mediaed apoptosis of lung epithelial cells. J Immunol, 2002,168:6470-6478
    41. Laurent P. Recognition and treatment of idiopathic pulmonary fibrosis [J]. Drugs, 1998,55:555-562
    42. Snell GI, Waiters EH, Kotsimbos TC, et al. Idiopathic pulmonary fibrosis: In need of focused and systematic management. Med J Aust. 2001,174:137-140
    1.张纾难.肺间质纤维化中医治疗概述.现代药学、预防医学及中医进展(中医分册),科学技术文献出版社,2006,10(1):21-32
    2.陈瑞,王鹏.浅谈肺痹与肺纤维化的关系[J].中国中医药信息杂志,2005,12(6):90-91
    3.陈金亮,王殿华,等.络病理论与肺纤维化的关系探析[J].中医药学刊,2004,3(22)3:
    4.吴银根,张天嵩.络病理论指导肺纤维化中医证治探析[J].中医药学刊,2005,23(1):14-15
    5.蔡代仲,翟华强,等.论上盛下虚为肺纤维化基本病机[J].中国中医药信息杂志,2000,9(11)9:
    6.仝润芍.血府逐淤汤加减治疗弥漫性间质性肺炎46例[J].河南中医,1996,16(4):233
    7.邱志楠,潘俊辉,等.中西医结合治疗特发性肺间质纤维化68例临床观察[J].中国中医急症,2002,4;11(2)
    8.王新华,张弘,等.特发性肺纤维化的中西医治疗[J].贵阳中医学院学报,2000,12;22(4)
    9.宋建平.《金匮要略》所论短气与特发性肺纤维化[J].南京中医药大学学报,1998,11(6)
    10.张天嵩,董洪涛.特发性肺纤维化的中医病机探讨[J].中医函授通讯,1999,6;18(3):
    11.李素云,朱佳,等.抗纤颗粒治疗弥漫性肺间质纤维化极其对细胞因子的影响[J].中国中医药信息杂志,2002,9(5):122-125
    12.智屹惠.曹世宏教授论治肺间质纤维化[J].南京中医药大学学报(自然科学版),2001,5;17(3)
    13.杨效华,张晓梅,等.肺间质纤维化的中医药治疗[J].中国临床医生, 2002,30(6):213-215
    14.彭玉华.特发性肺纤维化中医药治疗思路与方法[J].中医药学刊,2003,21(10):335
    16.孔祥文.特发性肺纤维化的中医治疗[J].现代中西医结合杂志,2003,12(1):108
    17.王海彤,武维屏.中医药治疗弥漫性肺间质纤维化信息讨论[J].中国中医药信息杂志,1996,3(7):25-26
    19.王书臣,崔天红,等.弥漫性肺间质性肺炎62例[J].中医杂志,1998,39(9):62-64
    21.张天嵩,马君,等.赵子贤辨治弥漫性肺间质纤维化经验[J].浙江中医杂志,1998,33(3):61-62
    22.张纾难,李兰群,等.益气润肺化淤解毒法治疗特发性肺纤维化临床研究[J].北京中医药大学学报,1999,22(3):57-60
    23.张天嵩,赵子贤,等.补气通肺汤治疗特发性肺纤维化12例[J].浙江中医杂志,1999,34(2):54-55
    24.唐玲华,李春生.特发性弥漫性肺间质纤维化13例治疗报告[J].中医杂志,1997,38(1):34-36
    26.刘建博,刘小虹,等.肺康颗粒治疗弥漫性肺间质纤维化临床观察[J].中国中医急症,2001,10(2):77
    27.刘三都,鲍崇忠,等.肺乐宁抗结核性肺纤维化临床研究[J].贵阳中医学院学报,1999,21(4):11-12
    28.欧阳修河,胡翠花.川芎嗪治疗特发性肺纤维化临床观察[J].实用医学杂志,1998,14(7):93-94
    29.陆思静,刘又宁,王代洲.丹参对博莱霉素所致大鼠肺纤维化保护作用的研究[J].中国临床药理与治疗学,2005,10(5):514-517
    30.李戎,闫智勇,唐勇,等.针灸治疗肺纤维化的疗效观察[J].针灸临 床杂志,2004,20(2):74-76
    31.李戎,闫智勇,唐勇,等.肺俞、膏肓俞、四花穴灸治肺痿(肺纤维化)沿革[J].中国针灸,2004,24(6):429
    32.李戎.论艾灸膏肓俞治疗肺纤维化的中医理论基础与实施依据[J].辽宁中医杂志,2004,31(4):291
    33.张捷,安继红,张伟.刺五加对肺纤维化大鼠BALF中IL-6抑制作用的研究[J].中国免疫学杂志,1995,11(5):300-303
    34.金洪,田英麟,姚汉德,等.汉防己阻断平阳霉素所致肺纤维化的实验研究[J].中华结核和呼吸杂志,1991,14(6):359-360
    35.陈建,何冰,刘新民,等.银杏叶制剂治疗肺间质纤维化的实验研究[J].中国中西医结合杂志,2000,20(6):441-443
    36.钟殿胜,朱元钰,郭子健,等.雷公藤T4单体治疗特发性肺纤维化的实验研究[J].中华内科杂志,1997,36(8):546-547
    37.张炜,毕小利,马文欢.生地对特发性肺纤维化基质重建调控[J].中国中医基础医学杂志,2001,7(6):434-436
    38.蒋玉宇,曹世宏,孙子凯.当归干预肺间质纤维化的作用途径研究[J].辽宁中医杂志,2006,33(11):1501-1502
    39.董静,罗桂林,苗维纳.丹参对实验性肺纤维化小鼠病理变化和核因子κB表达的影响[J].中国药理学通报,2003,19(12):1428-1431.
    40.宋月莲,郑应昭,李素慧.中药桃仁对体外纤维母细胞增生的抑制作用的实验研究[J].中西医结合眼科杂志,1995,(1):1—3.
    41.陈翠菊,方伟.药物治疗急性呼吸窘迫综合征的研究进展[J].中国综合临床,2005,21(5):471
    42.龚捷宁,卞慧敏,刘学风.养肺活血汤对实验性肺纤维化大鼠血液流变学的影响[J].中药药理与临床,2000,16(6):34-46
    43.徐杰,刘兴奎.血府逐淤汤防治平阳霉素引起肺纤维化的实验研究 [J].中医药信息,2000,(1):49-50
    44.俞发荣,张启,石军年,等.中药抗纤Ⅰ、Ⅱ号对大鼠肺纤维化影响的实验研究[J].甘肃科学学报,2001,13(1):62-67
    45.宋建平,李瑞琴,李伟,等.乌梢散对肺纤维化模型作用机理研究[J].中医药学刊,2001,19(5):515-517