葛根芩连汤及其有效组分干预非酒精性脂肪性肝炎PPARγ的作用机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
本文通过文献综述、实验研究两方面阐述了现代医学和中医药对非酒精性脂肪性肝炎(NASH)的认识、临床与实验研究现状,探讨了葛根芩连汤及其有效组分干预非酒精性脂肪性肝炎PPARγ的作用机制研究,以达到探索古方新用,提升经方在该领域研究价值和临床治疗水平,同时为开发防治NASH有效的中药制剂提供新思路和实验依据。
     1文献综述
     通过近10年的国内外文献资料分析,归纳了NASH现代医学的病因、发病机制、治疗研究近况,中医药关于NASH病因病机、证候学、临床研究及NASH实验相关模型的研究进展。
     2葛根芩连汤对高脂饲料饲喂诱导的大鼠非酒精性脂肪性肝炎实验研究
     目的
     探讨葛根芩连汤对NASH大鼠模型的药效学作用及机制。
     方法
     以高脂饮食建立大鼠NASH模型,并于造模同时进行药物干预;实验第8周末进行血清ALT、AST、TG、HDL-C、LDL-C、TNF-a、IL-6、IL-10及肝组织TNF-a、IL-6、IL-10的测定,并进行肝组织PPARγ Western-blot与PCR检测,同时油红O、HE染色观察肝脏病理切片。
     结果
     (1)一般情况及病理形态学:模型组大鼠毛色枯黄黯淡,喜团聚蜷卧,饮食量较空白组减少,大便干稀不调,体重也随着造模时间逐渐增加,NAS组织学病理评分总分>5分(P<0.05);葛根芩连汤高、低剂量组脂肪变性、小叶内炎症、气球样变均明显下降,有显著性差异;葛根芩连汤高、低剂量组较模型组病理总分均显著下降<5分(P<0.05)。
     (2)肝脏酶学:模型组血清ALT、AST水平均明显高于空白组,有显著统计学差异(P<0.05),葛根芩连汤高、低剂量组血清ALT、AST水平均较模型组显著降低(P<0.05)。
     (3)血脂及肝脂:与空白组相比,模型组血清HDL显著降低,血清LDL、TG、TC明显增加(P<0.05)。葛根芩连汤高剂量组血清HDL水平与模型组相比增加明显(P<0.05);葛根芩连汤低剂量组与模型组相比,血清LDL水平下降明显(P<0.05);与模型组相比,葛根芩连汤高、低剂量组血清TC均降低明显(P<0.05);对于血清甘油三酯,各给药组与模型组之间无统计学意义(P>0.05)。
     (4)血清及肝组织IL-6、IL-10、TNF-a水平:与空白组相比,模型组血清及肝组织IL-10显著降低,血清IL-6、TNF-α明显增加(P<0.05)。与模型组相比,葛根芩连汤高、低剂量组可明显增加血清及肝组织IL-10含量,降低血清及肝组织IL-6、TNF-α水平(P     (5)肝组织PPARγ蛋白及mRNA表达:与空白组相比,模型组肝组织PPARγ蛋白表达及mRNA水平均降低(P<0.05);葛根芩连汤高、低剂量组与模型组相比,可明显改善肝组织PPAR γ蛋白表达及mRNA表达(P<0.05)。
     结论
     葛根芩连汤高、低剂量均能显著改善高脂饮食饲喂诱导的NASH大鼠肝组织病理,改善脂肪变、炎性因子浸润;能够降低血清转氨酶水平、改善血脂代谢紊乱;能够通过抑制促炎因子、提高抑炎因子两方面动态调控炎症的发展;同时通过对肝组织PPAR γ的调控达到干预大鼠NASH的作用。
     3游离脂肪酸诱导NASH体外模型的建立及动态监测
     目的
     研究游离脂肪酸诱导的非酒精性脂肪性肝炎体外模型,动态观测细胞脂肪变性、炎症、过氧化损伤及糖转运缺陷发生发展的特点。
     方法
     以HepG2细胞为实验对象,采用不同浓度游离脂肪酸(250μM,500μ M,1000μM)对细胞进行不同时间(12h,18h,24h)的干预刺激,并通过油红0染色,酶联免疫的方法分别对细胞内脂肪变及细胞培养上清液中IL-8, TNF-a, SOD, MDA, GLUT4进行测定评估。
     结果
     FFA作用的实验各组,随着时间的延长及浓度的增加,细胞脂肪样变、IL-8、TNF-a、SOD、MDA及GLUT4均有不同程度的改变,以24h0.5mM FFA、24h1mM FFA最为明显,与同期空白对照组相比,其在IL-8, TNF-a、SOD、MDA、GLUT4的表达方面均有统计学意义(P<0.05),油红0染色可见典型的“印戒”状。
     结论
     应用游离脂肪酸诱导建立体外NASH模型成模效果好,重复率高,并提示500μM FFA24h为佳。
     4葛根芩连汤对NASH细胞模型的干预研究
     目的
     探讨葛根芩连汤对非酒精性脂肪性肝炎体外模型的影响。
     方法
     用500μmol/L的FFA诱导HepG2细胞24h,建立NASH体外模型;用不同浓度的葛根芩连汤干预NASH体外模型,MTT法检测葛根芩连汤的安全药物浓度范围;油红0染色观察细胞内脂肪变;Elisa法检测细胞培养上清液中TNF-a、IL-8及GLUT4。
     结果
     (1)NASH细胞模型组培养液中TNF-α及IL-8含量明显高于空白对照组(P<0.01),GLUT4含量明显低于空白对照组。
     (2)NASH模型组中同时加入葛根芩连汤可明显改善细胞脂肪样变,减少TNF-αIL-8分泌,提高GLUT4含量。
     结论
     葛根芩连汤能够通过影响TNF-α、IL-8及GLUT4改善NASH体外模型炎症及糖脂代谢。
     5葛根素、小檗碱联合用药干预NASH细胞模型的的实验研究
     目的
     探讨葛根素、小檗碱联合用药干预非酒精性脂肪性肝炎体外模型的相关剂量及作用效果。
     方法
     用500μ mol/L的FFA诱导HepG2细胞24h,建立NASH体外模型;用不同浓度的葛根素、小檗碱单药及组合用药干预NASH体外模型。MTT法测定两种单体对非酒精性脂肪性肝病体外模型细胞安全药物浓度范围;油红0染色观察细胞内脂肪变;Elisa法检测细胞培养上清液中TNF-α、IL-8及GLUT4水平。
     结果
     (1)500μ mol/LFFA作用NASH模型组,可见细胞胞浆内被染成红色的颗粒状脂滴,并呈“印戒”状形态。不同浓度葛根素、小檗碱单用及联合用药(p500、p1000、p2000、b25、b50、b25p1000、b50p500,单位u mol/L)干预组,可见细胞内红色脂滴有所减少,单体联合用药组优于同浓度单独用药组,基本达到以小剂量联合不次于高剂量单独用药的目的。
     (2)500μ mol/L FFA刺激24h后的HepG2细胞,其细胞培养上清液中炎性因子TNF-α及IL-8含量明显增加,GLUT4含量明显下降,与空白组比,差异显著(P<0.05)。
     随着小檗碱、葛根素单独使用浓度的增加,IL-8水平较模型组明显降低,以b50、b100、p2000差异明显(P<0.05);组合用药后b25p1000、b50p500均较模型组有统计学意义(P<0.05),b25p1000较b25效果明显(P<0.05),b50p500较p500、p1000效果明显(P<0.05)。
     对于TNF-a,随着小檗碱、葛根素单独使用浓度的增加,其上清液含量显著降低,均较模型组用统计学差异(P<0.05);组合用药后,TNF-a含量的变化优于单独使用时,b25p1000较p1000、p2000效果明显(P<0.05),b50p500较b50效果明显(P<0.05)。
     对于GLUT4,随着小檗碱、葛根素单独使用浓度的增加,其上清液含量显著增加,以b50、b100、p500、p1000、p2000较模型组用统计学差异(P<0.05),且葛根素对于增加GLUT4含量作用优于小檗碱;组合用药后,GLUT4含量的变化优于小檗碱单独使用,b25p1000在加入葛根素后较b25、b50效果明显增强,b50p500较b50、b100效果明显增加(P<0.05)。
     结论
     葛根素、小檗碱组合用药在改善细胞病理、TNF-α、IL-8、GLUT4水平方面存在协同作用,其组合用药效果较单独用药效果增强。体外实验中,基本能够确定在一定范围内,小檗碱与葛根素组合比例在1:40-1:10为佳。
     6葛根素、小檗碱、黄芩苷联合用药干预NASH细胞模型的实验研究
     目的
     探讨葛根素、小檗碱、黄芩苷联合用药干预非酒精性脂肪性肝炎体外模型的相关剂量及作用效果。
     方法
     用500μ mol/L的FFA诱导HepG2细胞24h,建立NASH体外模型;用不同浓度的黄芩苷单药及葛根素、小檗碱、黄芩苷组合用药干预NASH体外模型。MTT法测定两种单体对非酒精性脂肪性肝病体外模型细胞安全药物浓度范围;油红0染色观察细胞内脂肪变;Elisa法检测细胞培养上清液中TNF-α及IL-8水平。
     结果
     (1)500μ mol/L FFA作用NASH模型组,可见细胞胞浆内被染成红色的颗粒状脂滴,并呈“印戒”状形态。不同浓度黄芩苷单用及葛根素、小檗碱、黄芩苷联合用药(bai25、bai50、bai100、bai200、b25p1000bai25、b25p1000bai50、b25p1000bai100、b50p500bai25、b50p500bai50、b50p500bai,单位100μ mol/L)干预组,可见细胞内红色脂滴有所减少,单体联合用药组效果明显。
     (2)500μ mol/L FFA刺激24h后的HepG2细胞,其细胞培养上清液中炎性因子TNF-α及工L-8含量明显增加,与空白组比差异显著(P,0.05)。
     随着黄芩苷单独使用浓度的增加,IL-8水平较模型组明显降低,以bai200差异明显(P<0.05);组合用药后b25p1000bai25、b25p1000bai50、b25p1000bai100、b50p500bai25、 b50p500bai50、b50p500bai100均较模型组有统计学意义(P,0.05),在各比例组合中,以b25p1000bai100、b50p500bai25、b50p500bai50、b50p500bai100效果最为明显(P<0.05)。
     对于TNF-a,随着黄芩苷单独使用浓度的增加,其上清液含量显著降低,均较模型组用统计学差异(P<0.05);组合用药后,b25p1000bai25、b25pl000bai50、b25p1000bai100、b50p500bai25、b50p500bai50、b50p500bai100均较模型组有统计学意义(P<0.05),且效果优于黄芩苷单独使用时。在各比例组合中,以b50p500bai50、b50p500bai100效果最为明显(P     结论
     葛根素、小檗碱、黄芩苷联合用药(b25p1000bai25、b25p1000bai50、b25p1000bai100、 b50p500bai25、b50p500bai50、b50p500bai100)在减少细胞内红色脂滴方面,存在显著协同作用;对于TNF-α、IL-8,通过单体组合使用,可在降低黄芩苷浓度的前提下保证干预效果。体外实验中,基本能够确定在一定范围内,小檗碱、葛根素、黄芩苷组合比例在1:10:1-1:10:2为佳。
     7PPAR γ与炎性因子在NASH细胞模型中的相关性研究
     目的
     明确PPAR y与炎性因子在NASH细胞模型中的关系。
     方法
     用500μ mol/L的FFA诱导HepG2细胞24h,建立NASH体外模型;造模同时给与不同浓度GW9662/罗格列酮刺激;Elisa法检测细胞培养上清液中TNF-α及IL-8水平,油红0染色测定细胞内脂滴。
     结果
     (1)与模型组比较,GW966210u M加入后,IL-8及TNF-a含量均增加,但未出现统计学意义。加入罗格列酮2μM后,IL-8及TNF-α含量显著降低,与模型组、gw10组相比,均具有统计学差异(P<0.05)。各单体与gw10组合中,IL-8及TNF-α含量均降低,与模型组、gw10组比较差异显著(P<0.05)。
     为了避免因模型带来的非GW9662因素造成的工L-8及TNF-α含量增加,补充了GW9662对未加入FFA诱导的“正常”细胞的作用。结果显示,不同浓度的GW9662均使“正常”细胞上清液中IL-8及TNF-α含量增加,与未加入GW9662组比,差异均具有统计学意义(P<0.05)。
     (2)读取OD值进行细胞内脂滴含量分析,GW9662不同浓度较模型组比,脂滴含量均升高,差异显著,具有统计学意义(P<0.05)。
     结论
     PPAR y阻断剂GW9662可以通过阻断PPAR y促进炎症发展;葛根素、小檗碱、黄芩苷具有潜在的PPAR γ激动剂的作用;PPAR γ与脂质沉积代谢密切相关。
     8葛根芩连汤有效组分对高脂饲料饲喂诱导的大鼠非酒精性脂肪性肝炎药效学观察
     目的
     明确葛根芩连汤有效组分(葛根素、小檗碱、黄芩苷)对高脂饲料饲喂诱导的大鼠非酒精性脂肪性肝炎药效学作用。
     方法
     以高脂饮食建立大鼠NASH模型,并于造模同时进行药物干预(通过正交表确定组数);实验第8周末进行血清ALT、AST、TG、HDL-C、LDL-C的测定,同时油红O、HE染色观察肝脏病理切片。
     结果
     (1)一般情况及病理形态学:模型组大鼠毛色黯淡枯黄,喜团聚蜷卧,饮食量较空白组减少,大便干稀不调,模型组肝组织HE染色存在明显的肝细胞脂肪变性,肝小叶界限不清,肝索结构紊乱,大量空泡及气球样变产生,中性粒细胞浸润,油红0染色可见大量红色脂滴,颜色较深,显示肝组织内脂滴弥漫、含量较多,肝小叶内脂滴融合成片;葛根芩连汤高、低剂量组HE染色病变有不同程度减轻,损伤面积明显减小,胞内炎性细胞浸润减少,空泡及气球样变减少,细胞及小叶结构不同程度恢复,油红0染色脂滴减少。
     (2)肝脏酶学:模型组血清ALT、AST均明显高于空白组,有显著差异(P<0.01),各给药组血清ALT、AST水平均较模型组均显著降低(P<0.01),其中F、G、H组在改善血清ALT、AST方面效果明显优于HH组(P<0.01);D、E组仅在改善AST方面优于HH组(P<0.01)
     (3)血脂及肝脂:与空白组相比,模型组HDL显著降低,LDL、TG、TC显著增高,具有统计学差异(P0.05)。对于血清总胆固醇,A、B、C、D、E、F、G、HH组与模型组之间均差异显著(P0.05)。
     结论
     葛根素为主药的单体组合,对肝组织病理改善,特别是脂滴改善最为明显;以抗炎类小檗碱、黄芩苷为主药的组合,对转氨酶影响最大;不同比例单体组合对血脂的调节都具有积极作用,但随着各单体所占比例的变化,并未见明确的规律性,一些组合的效能基本与葛根芩连汤高剂量组相当,提示组合明确的单体配伍可以在某些方面达到中药原方的效果。
Comprehensive learning of non-alcoholic steatohepatitis (NASH) in both modern medicine and traditional Chinese medicine (TCM) is demonstrated through literature review and experimental study in this paper. Efficacy and mechanism of GeGenQinLian Decoction and its componets are explored in in vivo and in vitro study. The research provided a basic reveal of the mechanism of GeGenQinLian Decoction and its componets, which may give a great contribution to clinical therapeutic levels and new grug discovery.
     1Literature review
     Through literature analysis in recent10years both at home and abroad, the etiology, pathogenesis, and treatment of modern medicine, also etiology and pathogenesis, syndrome differentiation, physical sciencetreatment progress of TCM have been review.
     2Effects and Mechinsm of GeGenQinLian Decoction in NASH Rats induced by High-Fat Diet
     Objective
     To investigate the efficacy and mechinsm of GeGenQinLian Decoction on rat with non-alcoholic steatohepatitis (NASH) induced by high-fat-diet.
     Method
     The SD rats were fed with high-fat-diet for8weeks to make NASH rat model. At the beginning of modeling, the treatment groups were intervened by GeGenQinLian Decoction in high and low doses. The change of serum transaminase, serum lipid, serum inflammation factors, liver inflammation factors and the liver pathology had been tested to observe the efficacy. Meanwhile, PPAR γ protein and mRNA in liver were also detected.
     Outcome
     (1) General condition and pathomorphology:The hepatic steatosis and inflammation were increased in model group, which were improved in the treatment groups, together with significantly reduced NAS score. And there was no significant difference between treatment groups.
     (2) Serum transaminase:Compared with blank control group, the serum ALT, AST in model group were obviously increased. The serum ALT and AST decreased both in high and low dose of GeGenQinLian Decoction group.
     (3) Lipid:Serum HDL-C was reduced, serum LDL-C, TG and TC were increased in the model group compared with blank control group. The high dose groups of GeGenQinLian Decoction increased the serum HDL-C compared with the model group, while the low dose of GeGenQinLian Decoction decreased the serum LDL-C significantly. Both the high and low dose of GeGenQinLian Decoction decreased serum TC.
     (4) Serum and liver IL-6, IL-10and TNF-α:Compared with blank control group, the serum and liver IL-6and TNF-α in model group were obviously increased, while the serum and liver IL-10decrease. GeGenQinLian Decoction can significantly increase the leverl of serum and liver IL-10and decrease the serum and liver IL-6and TNF-α.
     (5) liver PPAR γ protein and mRNA expression:Compared with blank control group, the liver PPAR γ protein and mRNA expressions were both declined, which were increased in GeGenQinLian Decoction groups.
     Conclusion
     GeGenQinLian Decoction can significantly improve liver pathology of the high-fat diet-induced NASH rats; improving steatosis and infiltration of inflammatory cytokines; reducing serum transaminase levels;improving dyslipidemia; and achieving intervention on NASH rats by PPAR γ regulation.
     3A Cell Model of Nonalcoholic Steatohepatitis Induced by FFA:Establishment and Dynamic Monitoring
     Objective
     To establish a cell model of nonalcoholic steatohepatitis (NASH) by free fatty acid (FFA) and to dynamically monitor the pathological features of steatosis, oxidative damage and inflammation in these cells.
     Methods
     Liver cells HepG2were cultured in DMEM medium supplemented with10%fetal bovine serum (FBS), penicillin (100units/ml) and streptomycin (100μg/ml) at37°C under5%CO2in a95%humidified atmosphere. For the treatment, palmitic and oleic acid0.1M stock solutions were prepare by dissolving FFA in DMSO (with a final concentration less than0.1%). The cells were exposed to increasing concentrations of a fresh mixture of exogenous FFA (250μmol/L,500μmol/L,1000μmol/L) in molar ratio1:2palmitic:oleic at different time points (12h,18,24h) respectively. Oil red0staining and the IL-8, TNF-α, SOD, MDA, GLUT4 in supernatant were evaluated.
     Outcome
     Steatosis could be seen in FFA groups to different extents. IL-8, TNF-α, SOD, MDA and GLUT4gradually differed according to the change of stimulation time by different FFA concentrations, especially in the groups with0.5mM and1mM FFA concentrations for24h, of which groups there could be seen a typical steatosis.
     Conclusion
     A cell model of NASH has been created successfully by FFA.
     4Research of Gegenqinlian Decoction on NASH in vitro
     Objective
     Free fatty acid induced NASH model was used to evaluate the effects of Gegenqinlian Decoction.
     Method
     Liver cells HepG2were cultured in DMEM medium supplemented with10%fetal bovine serum (FBS), penicillin (100units/ml) and streptomycin (100μg/ml) at37°C under5%CO2in a95%humidified atmosphere. The cells were exposed to a concentration of a fresh mixture of exogenous FFA (500μmol/L) in molar ratio1:2palmitic:oleic for24h. Different concentrations of Gegenqinlian Decoction were meanwhile added. MTT, Oil red0staining and the IL-8, TNF-α, GLUT4in supernatant were evaluated.
     Outcome
     (1) TNF-a and IL-8were significantly higher in model group than the control group (P<0.01), while the level of GLUT4was significantly lower than the control group (P<0.01).
     (2) Gegenqinlian Decoction can significantly improve cell degeneration and steatosis, reducing TNF-a, IL-8secretion and improving GLUT4content.
     Conclusion
     Gegenqinlian Decoction can improve lipid metabolism and inflammation through influencing TNF-a, IL-8and GLUT4.
     5Experimental Studies of the Combination Effects of Berberine and Puerarin on NASH in vitro
     Objective
     To discuss the combination effects of puerarin and berberine on NASH in vi tro.
     Method
     Liver cells HepG2were cultured in DMEM medium supplemented with10%fetal bovine serum (FBS), penicillin (100units/ml) and streptomycin (100μ g/ml) at37°C under5%CO2in a95%humidified atmosphere. The cells were exposed to a concentration of a fresh mixture of exogenous FFA (500μ mol/L) in molar ratio1:2palmitic:oleic for24h. Different concentrations of puerarin and berberine combination with different propotions were meanwhile added.MTT, Oil red0staining and the IL-8, TNF-a, GLUT4in supernatant were evaluated.
     Outcome
     (1) Compared with NASH model group, different concentrations of puerarin and berberine alone and combination (p500, p1000, p2000, b25, b50, b25p1000, b50p500, μmol/L) in the intervention group can decrease lipid droplets sidnificantly.
     (2) TNF-a and IL-8were significantly higher in model group than the control group (P<0.01), while the level of GLUT4was significantly lower than the control group(P<0.01). Different concentrations of puerarin and berberine combination can significantly improve cell degeneration and steatosis, reducing TNF-a, IL-8secretion and improving GLUT4content(P<0.01).
     Conclusion
     Puerarin and berberine combination can improve cellular pathology, influencing the levels of TNF-a, IL-8and GLUT4, which showed a synergistic effect and better than a single drug effect.
     6Experimental Studies of the Combination Effects of Berberine, Puerarin and baicalin on NASH in vitro
     Objective
     To discuss the combination effects of puerarin, berberine and baicalin on NASH in vitro.
     Method
     Liver cells HepG2were cultured in DMEM medium supplemented with10%fetal bovine serum (FBS), penicillin (100units/ml) and streptomycin (100μg/ml) at37°C under5%CO2in a95%humidified atmosphere. The cells were exposed to a concentration of a fresh mixture of exogenous FFA (500μmol/L) in molar ratio1:2palmitic:oleic for24h. Different concentrations of puerarin, berberine and baicalin combination with different propotions were meanwhile added.MTT, Oil red0staining and the IL-8and TNF-a in supernatant were evaluated.
     Outcome
     (1) Compared with NASH model group, different concentrations of puerarin, berberine and baicalin alone and combination (bai25, bai50, bai100, bai200, b25p1000bai25, b25p1000bai50, b25p1000bai100, b50p500bai25, b50p500bai50, b50p500bai100, μmol/L) in the intervention group can decrease lipid droplets sidnificantly.
     (2) TNF-a and IL-8were significantly higher in model group than the control group (P<0.01). Different concentrations of puerarin, berberine and baicalin combination can significantly improve cell degeneration and steatosis and reduce TNF-a, IL-8secretion (P<0.01).
     Conclusion
     Puerarin, berberine, and baicalin combination can improve cellular pathology, influencing the levels of TNF-a and IL-8, which showed a synergistic effect. The better proportion of berberine, puerarin, baicalin combination was1:10:1-1:10:2in vitro.
     7The Related Studies between PPAR γ and Inflammatory Factors on NASH in vitro
     Objective
     To clarify the relationship between PPAR γ and inflammatory factors.
     Method
     NASH model in vitro was established using500μmol/L FFA for-24h. The agonists of PPAR γ Rosiglitazone and antaonist of PPAR γ GW9662were meanwhile added.Oil red0staining and the IL-8and TNF-a in supernatant were evaluated.
     Outcome
     (1) Compared with NASH model group, the GW966210μmol/L group can increase IL-8and TNF-a level; while IL-8and TNF-a were significantly reduced after adding rosiglitazone2μmol/L (P<0.05). In the groups of GW966210μmol/L plus with each monomer, IL-8and TNF-a levels were lowerd compared with the model group and GW966210μmol/L group (P<0.05).
     In order to avoid an increase caused by model, GW9662was added to the "normal" cells without FFA. The results showed that different concentrations of GW9662increased IL-8and TNF-a levels compared with the group without GW9662(P<0.05).
     (2) The intracellular lipid droplets content was analysized by reading OD values, lipid droplets levels were increased significantly in groups with different concentrations GW9662compared with the model group (P<0.05).
     Conclusion
     PPAR γ antagonist GW9662can promote inflammation by blocking PPAR γ; puerarin, berberine and baicalin hold the potential effects as PPAR γ agonist; PPAR γ is closely related to lipid metabolism.
     8Effects of Components in GeGenQinLian Decoction in NASH Rats Induced by High-Fat Diet
     Objective
     To investigate the efficacy of components in GeGenQinLian Decoction (puerarin, berberine, baicalin) on rat with non-alcoholic steatohepatitis (NASH) induced by high-fat-diet.
     Method
     The SD rats were fed with high-fat-diet for8weeks to make NASH rat model. At the beginning of modeling, the treatment groups were intervened by the componts of GeGenQinLian Decoction (number of groups identified by orthogonal table). The change of serum transaminase, serum lipid and the liver pathology had been tested to observe the efficacy.
     Outcome
     (1) General condition and pathomorphology:The hepatic steatosis and inflammation were increased in model group, which were improved in the treatment groups. And there was no significant difference between the all treatment groups.
     (2) Serum transaminase:Compared with blank control group, the serum ALT, AST in model group were obviously increased (P<0.01). The serum ALT and AST decreased both in all treatment groups, in which F, G, H groups were better than HH gtoup in inproving ALT and AST, and D, E group were better than HH group in inproving AST (P<0.01).
     (3) Lipid:Serum HDL-C was reduced, serum LDL-C, TG and TC were increased in the model group compared with blank control group (P<0.01). The A, B, D, E, I, HH groups increased the serum HDL-C compared with the model group, in which HH group was better than B, E groups (P<0.01). Compared with the model group, each treatment groups decreased serum LDL levels significantly (P<0.01), in which the HH group was better than E, F, H, I groups (P<0.01). For the serum triglycerides, the level in A, C, D, E, F, G, HH groups decreased compared with model group (P<0.01). For the serum total cholesterol, the level in A, B, C, D, E, F, G, HH group decreased compared with model group (P<0.01).
     Conclusion
     GeGenQinLian Decoction can significantly improve of the high-fat diet-induced NASH rats and achieve intervention on NASH rats by PPAR γ regulation.
     Different proportions of monomer composition gave positive effects to the liver pathology, serum transaminase levels and dyslipidemia. With some certain propotion, it showd an equal effect to the high dose of GeGenQinLian Decoction, suggesting a clear monomer combination could achieve the effect at some aspects with the original formula in traditional Chinese medicine.
引文
[1]萧树东,许国铭主编.中华胃肠病学(第1版)[M].北京:人民卫生出版社出版,2008,591-597.
    [2]Stefano Bellentani, Mariano Marino. Epidemiology and natural history of non-alcoholic fatty liver disease (NAFLD) [J]. Annals of hepatology,2009, 8(Suppl 1):S4-8.
    [3]Brunt EM. Nonalcoholic steatohepatitis:pathologic features and differerntial diagnosis[J]. Seminar in diagnostic pathology,2005, 22(4):330-338.
    [4]Sanyal AJ. AGA technical review on nonalcoholic fatty liver disease [J]. Gastroenterology,2002,123:1705-1725.
    [5]Powell EE, Cooksley WG, Hanson R, et al.The natural history of nonalcoholic steatohepatitis:a follow-up study of forty-two patients for up to 21 years [J]. Hepatology,1990,11(1):74-80.
    [6]Caldwell SH, Crespo DM. The spectrum expanded:cryptogenic cirrhosis and the natural history of non-alcoholic fatty liver disease[J]. Journal of Hepatology,2004,40(4):578-584.
    [7]Smith BW, Adams LA. Non-alcoholic fatty liver disease [J]. Crit Rev Clin Lab Sci,2011,48(3):97-113.
    [8]Sheth SG, Gordon FD, Chopra S. Nonalcoholic steatohe patitis[J]. Annal of Internal Medicine,1997,126(2):137-145.
    [9]Clark JM, Brancati FL, Diehl AM. Nonalcoholic fatty liver disease[J]. Gastroenterology,2002,122(6):1649-1657.
    [10]Carulli L, Lonardo A, Lombardini S, et al. Gender, fatty liver and GGT[J]. Hepatology,2006,44(1):278-279.
    [11]McKenzie J, Fisher BM, Jaap AJ, et al. Effects of HRT on liver enzyme levels in women with type 2 diabetes:a randomized placebo-controlled trial [J]. Clinical Endocrinology (Oxf),2006,65(1):40-44.
    [12]Lemieux S, Prud'homme D, Bouchard C, et al. Sex differences in the relation of visceral adipose tissue accumulation to total body fatness[J]. The American Journal of Clinical Nutrition,1993,58(4):463-467.
    [13]Farrell GC, Larter CZ. Nonalcoholic fatty liver disease:from steatosis to cirrhosis[J]. Hepatology,2006,43 (2 Suppl 1):S99-S112.
    [14]de Alwis NM, Day CP. Non-alcoholic fatty liver disease:themist gradually clears[J].J Hepatol,2008,48 Suppl 1:S104-S112.
    [15]Angulo P. GI epidemiology:nonalcoholic fatty liver disease [J]. Aliment Pharmacol Ther,2007,25:883-889.
    [16]Hilden M, Christoffersen P, Juhl E, et al. Liver histology in a "normal" population:examinations of 503 consecutive fatal traffic casualties[J]. Scandinavian Journal of Gastroenterology,1977,12(5):593-597.
    [17]Petersen KF, Befroy D, Dufour S, et al. Mitochondrial dysfunction in the elderly:possible role in insulin resistance[J]. Science,2003,300(5622): 1140-1142.
    [18]Browning JD, Szczepaniak LS, Dobbins R, et al. Prevalence of hepatic steatosis in an urban population in the United States:impact of ethnicity[J]. Hepatology,2004,40(6):1387-1395.
    [19]Caldwell SH, Ikura Y, Iezzoni JC, et al. Has natural selection in human populations produced two types of metabolic syndrome (with and without fatty liver) [J]? Journal of Gastroenterology and Hepatology,2007,22(suppl 1): S11-S19.
    [20]Amarapurkar DN, Hashimoto E, Lesmana LA, et al. How common is non-alcoholic fatty liver disease in the Asia-Pacific region and are there local differences[J]? Journal of Gastroenterology and Hepatology,2007,22(6): 788-793.
    [21]Fan JG, Saibara T, Chitturi S, et al. What are the risk factors and settings of non-alcoholic fatty liver disease in Asia-Pacific[J]? Journal of Gastroenterology and Hepatology,2007,22(6):794-800.
    [22]Bedogni G, Miglioli L, Masutti F, et al. Prevalence of and risk factors for nonalcoholic fatty liver disease:the Dionysos nutrition and liver study[J]. Hepatology,2005,42(1):44-52.
    [23]Mathurin P, Gonzalez F, Kerdraon 0, et al. The evolution of severe steatosis after bariatric surgery is related to insulin resistance[J]. Gastroenterology,2006,130(6):1617-1624.
    [24]FARRELL GC, CHITTURI S, LAU GK, et al. Guidelines for the assessment and management of non-alcoholic fatty liver disease in the Asia-Pacific region executive summary[J]. J Gastroenterol Hepatol,2007,22(6):775-777.
    [25]Takeshi Okanoue, Atsushi Umemura, Kohichiroh Yasui et al. Nonalcoholic fatty liver disease and nonalcoholic steatohepatitis in Japan[J]. Journal of Gastroenterology and Hepatology,2011,26(Suppl 1):153-162.
    [26]Jian-Gao Fan, Geoffrey C. Farrell.Epidemiology of non-alcoholic fatty liver disease in China[J]. Journal of Hepatology,2009,50(1):204-210.
    [27]孙维会,褚爱霞,许琳,等.青岛市城阳区非酒精性脂肪肝患病率及危险因素分析[J].青岛医药卫生,2012,44(2):92-94.
    [28]赵红梅,张洁,杨小青,等.上海市陆家嘴地区公司职员非酒精性脂肪肝发病率及危险因素分析[J].同济大学学报(医学版),2012,33(5):110-114.
    [29]张泽高,薛黎,张跃新,等.乌鲁木齐市公务员非酒精性脂肪肝调查[J].现代预防医学,2012,39(8):1955-1957.
    [30]范歆,陈少科,唐晴,等.南宁市超重、肥胖儿童非酒精性脂肪肝患病情况分析[J].中国儿童保健杂志,2012,20(2):124-126.
    [31]Day CP. Natural history of NAFLD:remarkably benign in the absence of cirrhosis[J]. Gastroenterology,2005,129(1):375-378.
    [32]Harrison SA, Torgerson S, Hayashi PH. The natural history of nonalcoholic fatty liver disease:a clinical histopathological study[J]. The American Journal of Gastroenterology,2003,98(9):2042-2047.
    [33]Kashi MR, Torres DM, Harrison SA. Current and emerging therapies in nonalcoholic fatty liver disease[J]. Seminars in Liver Disease,2008,28(4): 396-406.
    [34]Hui JM, Kench JC, Chitturi S, et al. Leng-term outcomes of cirrhosis in nonalcoholic steatohepatitis compared with hepatitis C[J]. Hepatology,2003, 38(2):420-427.
    [35]Hashimoto E, Yatsuji S, KanedaH, et al. The characteristics and natural history of Japanese patients with nonalcoholic fatty liver disease[J]. Hepatology Research,2005,33 (2):72-76.
    [36]Sanyal AJ, Banas C, Sargeant C, et al. Similarities and differences in outcomes of cirrhosis due to nonalcoholic steatohepatitis and hepatitis C[J]. Hepatology,2006,43(4):682-689.
    [37]Lee RG. Nonalcoholic steatohepatitis:a study of 49 patients[J].Hum Pathol,1989,20(6):594-598.
    [38]Cortez-Pinto H, Baptista A, Camilo ME, et al. Nonalcoholic steatohepatitis-a long -term follow-up study:comparison with alcoholic hepatitis in am-bulatory and hospitalized patients [J]. Dig Dis Sci,2003,48(10): 1909-1913.
    [39]Adams LA, Sanderson S, Lindor KD, et al. The histological course of non-alcoholic fatty liver disease:a longitudinal study of 103 patients with sequential liver biopsies[J]. J Hepatol,2005,42(1):132-138.
    [40]Angulo P, Keach JC, Batts KP, et al. Independent predictors of liver fibrosis in patients with nonalcoholic steatohepatitis [J]. Hepatology,1999, 30(6):1356-1362.
    [41]Hui JM, Kench JG, Chitturi S, et al. Long-term outcomes of cirrhosis in nonalcoholic steatohepatitis compared with hepatitis C[J]. Hepatology,2003, 38(2):420-427.
    [42]Boppidi H, Daram SR. Nonalcoholic fatty liver disease:hepatic manifestation of obesity and the metabolic syndrome [J]. Postgrad Med,2008,120(2): E01-E07.
    [43]Bellentani S, Bedogni G, Miglioli L, et al. The epidemiology of fatty liver[J]. European Journal of Gastroenterology & Hepatology,2004,16(11): 1087-1093.
    [44]Jakobsen MU, Berentzen T, Sorensen TI, et al. Abdominal obesity and fatty liver [J]. Epidemiologic Reviews,2007,29:77-87.
    [45]Hamaguchi M, Kojima T,Takeda N, et al.The metabolic syndrome as a predictor of nonalcoholic fatty liver disease[J]. Ann Intern Med,2005,143(10): 722-728.
    [46]张颖,高庆伟,王淑兰.非酒精性脂肪性肝病病因分析[J].中国现代药物应用,2012,6(2):34-35.
    [47]曹创裕,曾峥,李瑜元.非酒精性脂肪性肝病的流行现状及自然病程调查[J].广东医学,2010,31(21):2850-2852.
    [48]李萍,傅晓英,徐谷根,等.广州地区健康体检人群非酒精性脂肪肝与代谢综合征的发病情况分析[J].当代医学,2012,18(5):162-163.
    [49]秦恳,周婷,吴琴琴,等.成都地区非酒精性脂肪肝与代谢综合征相关性研究[J].现代预防医学,2012,39(10):2395-2397.
    [50]李晓慧,李彬,孙光,等.高龄男性人群非酒精性脂肪性肝病的调查[J].实用糖尿病杂志,2012,8(1):55-56.
    [51]Harrison SA. Liver disease in patients with diabetes mellitus[J]. Journal of Clinical Gastroenterology,2006,40(1):68-76.
    [52]鲁超,邹宇宏,王建青,等.非酒精性脂肪性肝炎病因及发病机制研究进展[J].安徽医药,2006,10(2):81-84.
    [53]Jansen PL. Nonalcoholic steatohepatitis:diagnosis, pathogenesis, treatment and prognosis[J]. Ned Tijdschr Genecskd,2005,149(6):289-294.
    [54]Friis-Liby I, Aldenborg F, Jerlstad P, et al. High prevalence of the metabolic complication in patients with non-alcoholic fatty liver disease[J]. Scandinavian Journal of Gastroenterology,2004,39(9):864-869.
    [55]Angulo P. Nonalcoholic fatty liver disease [J]. The new England Journal of Medicine,2002,346(16):1221-1231.
    [56]Chitturi S, Abeygunasekera S, Farrell GC, et al. NASH and insulin resistance:insulin hypersecretion and specific association with the insulin resistance syndrome[J]. Hepatology,2002,35(2):373-379. [57]Bugianesi E, Gastaldelli A, Vanni E, et al. Insulin resistance in non-diabetic patients with non-alcoholic fatty liver disease:sites and mechanisms[J]. Diabetologia,2005,48(4):634-642.
    [58]Abdelmalek MF, Liu C, Shuster J, et al. Familial aggregation of insulin resistance in first-degree relatives of patients with nonalcoholic fatty liver disease[J]. Clinical Gastroenterology and Hepatology,2006,4(9):1162-1169.
    [59]Browning JD, Kumar KS, Saboorian MH, et al. Ethnic differences in the prevalence of cryptogenic cirrhosis[J]. The American Journal of Gastroenterology,2004,99 (2):292-298.
    [60]Struben VM, Hespenheide EE, Caldwell SH. Nonalcoholic steatohepatitis and cryptogenic cirrhosis within kindreds [J]. The American Journal of Medicine, 2000,108(1):9-13.
    [61]Norman D, Bardwell WA, Arosemena F, et al. Serum aminotransferase levels are associated with markers of hypoxia in patients with obstructive sleep apnea[J]. Sleep,2008,31(1):121-126.
    [62]Kallwitz ER, Herdegen J, Madura J, et al. Liver enzymes and histology in obese patients with obstructive sleep apnea[J]. Journal of Clinical Gastroenterology,2007,41(10):918-921.
    [63]Savransky V, Nanayakkara A, Vivero A, et al. Chronic intermittent hypoxia predisposes to liver injury [J]. Hepatology,2007,45(4):1007-1013.
    [64]Zamora-Valdes D, Mendez-Sanchez N. Experimental evidence of obstructive sleep apnea syndrome as a second hit accomplice in nonalcoholic steatohepatitis pathogenesis [J]. Annals of Hepatology,2007,6(4):281-283.
    [65]Solga SF, Diehl AM. Non-alcoholic fatty liver disease:lumen-liver interactions and possible role for probiotics[J]. Journal of Hepatology,2003, 38(5):681-687.
    [66]Day CP, James OF. Steatohepatitis:a tale of two "hits" [J]. Gastroenterology,1998,114:842-845.
    [67]Te Sligte K, Bourass I, Sels JP, et al. Non-alcoholic steatohepatitis: review of a growing medical problem[J]. Eur J Intern Med,2004,15(1):10-21.
    [68]Quigley EM, Marsh MN, Shaffer JL. Hepatobiliary complications of total parenteral nutrition[J]. Gastroenterology,1993,104:286-361.
    [69]George DK, Powell LW, Losowsky MS. The haemochromatosis gene:a co-factor for chronic liver disease[J]. J Gastroenterol Hepatol,1999,14: 745-749.
    [70]Bonkovsky HL, Jaward Q, Tortorelli K,et al. Non-alcoholic steatohepatitis and iron:increased prevalence of mutations of the HFE gene in non-alcoholic steatohepatitis[J]. J Hepatol,1999,31:421-429.
    [71]Mortimore M, Merryweather clarks AT, Robsion KJ, et al. The haemochromatosis gene:a global perspective and implications for the Asia-Pacific region [J]. J Gastroenterol Hepatol,1999,14:838-843.
    [72]Moriand R, Mortaji AM, Loreal 0, et al. HLA-H mutations in patients with iron overload and normal transferring saturation [J]. Hepatology,1997,26:199.
    [73]Mendler MH, Moirand R, Turlin B, et al. Study of HFE gene mutations in the dysmetabolic iron overload syndrome[J]. Hepatology,1998,28:419.
    [74]Gao B, Radaeva S, Park 0. Liver natural killer and natural killerT cells: immunobiology and emerging roles in liver diseases[J]. J Leukoc Biol,2009, 86(3):513-528.
    [75]Enomot o N, Ikejima K, Bradf ord BU, et al. Role of Kupffer cells and gut-derived endot oxins in al coholic liver injury [J]. J Gastroenterol Hepatol, 2000,15 (Suppl):D20-25.
    [76]Browning JD, Horton JD. Molecular mediators of hepatic steatosis and liver injury[J]. J Clin Invest,2004,114:147-152.
    [77]Gastaldelli A, Miyazaki Y, Mahankali A, et al. The effect of pioglitazone on the liver:role of adiponectin[J]. Diabetes Care,2006,29:2275-2281.
    [78]Belfort R, Harrison SA, Brown K, et al. Aplacebo-controlled trial of pioglitazone in subjects with nonalcoholic steatohepatitis [J]. N Engl J Med, 2006,355:2297-2307.
    [79]Jump BD, Botolin D, Wang Y, et al. Fatty acid regulation of hepatic gene transcription [J]. Nutr,2005,135(11):2503.
    [80]Yu-POTHS,YIN D, KRIS-ETHERTON PM, et al. Long-chain poly-unsaturated fatty acids upregulate LDL receptor protein expression in fib-reblasts and HepG2 Cells[J].Nutr,2005,135(11):2541-2545.
    [81]COLEMAN R A, LEE D P. Enzymes of triacylglycerol synthesis and their regulation [J].Prog Lipid Res,2004,43(2):134-176.
    [82]艾正琳,陈东风.SREBP-1c在大鼠非酒精性脂肪性肝病中的表达及意义[J].第三军医大学学报,2006,28(10):1063-1065.
    [83]Horton JD, Goldstein JL, Brown MS. SREBPs:activators of the complete program of cholesterol and fatty acid synthesis in the liver[J].Clin Invest, 2002,109:1125-1131.
    [84]郭启煜.胰岛素受体与胰岛素抵抗[J].海军总医院学报,1999,12(2):88-90.
    [85]Kandror K V, Coderre L, PushkinA V, Pilch PF. Comparison of glucosetransporter containing vesicles from rat fat and muscle tissues evidence for a unique endosomal compartment [J]. Biochem J,1995,307:383.
    [86]Watson R T, Pessin J. Intracellular Organization of Insulin Signaling and GLUT4 Translocation[J]. Recent Prog Horm Res,2001,56:175.
    [87]宋春宇,毕会民.葛根素对大鼠胰岛素刺激下骨骼肌细胞膜GLUT4蛋白含量的影响[J].中国中药杂志2004,29(2):172-175.
    [88]Pagano C,,Soardo G, Pilon C, et al. Increased serum resistin in nonalcoholic fatty liver disease is related to liver disease severity and not to insulin resistance[J]. Clin Endocrinol Metab,2006,91(3):1081-1086.
    [89]BajajM, Suraamornkul S, HardiesLJ, et al. Plasma resistin concentration, hepatic fat content, and hepatic and peripheral insulin resistance in pioglitazone-treated type II diabetic patients[J]. Int J Obes RelatMetab Disord,2004,28(6):783-789.
    [90]卢晓云,李运红,徐肇敏,等.脂肪因子在非酒精性脂肪肝发病中的变化[J].南京医科大学学报(自然科学版),2010,30(4):510-513.
    [91]Laubnerk, Kieffer T J, Lamn T, et al. Inhibition of preproinsulin gene expression by leotion induction of suppressor of cytokine signaling 3 in pancreatic beta-cells [J]. Diabetes,2005(54):3410-3417.
    [92]Huang W, Dedousis N, Bandi A, et al. Liver triglyceride secretion and lipid oxidative metabolism are rapidly altered by leptin invivo[J]. Endocrinology,2006,147 (3):1480-1487.
    [93]张自然,李良平,韩盛玺.非酒精性脂肪肝血清及肝组织瘦素、肿瘤坏死因子α、脂联素的变化及意义[J].西部医学,2009,21(2):192-195.
    [94]周婷婷,秦波.非酒精性脂肪肝患者血浆FGF-21水平及与肥胖、胰岛素抵抗关系研究[J].第三军医大学学报,2010,32(3):265-268.
    [95]尹彬彬,王蕾,宋莹,明盛金,李俊,尹维笳,王会岩.FGF-21的研究进展[J].吉林医药学院学报,2011,39(3):226-229.
    [96]Tsukamoto H.Iron regulation of hepatic macrophage TNF-alpha expression[J]. Free Radic Biol Med,2002,32:309-313.
    [97]Wigg AJ, Roberts-Thomson IC, Dymock RB, et al. The role of small intestinal bacterial overgrowth, intestinal permeability, endotoxaemia and tumour necrosis factor alpha in the pathogenesis of nonalcoholic steatoheoatitis[J].Gut,2001,48:206-211.
    [98]Bacon BR, Farahvash MJ, Janney CG, et al. Non alcoholic steatohepatitis: an expanded clinical entity[J]. Gastroenterology,1994,107:1103-1109.
    [99]Bonkovsky HL, Jawaid Q, Tortorelli K, et al. Nonalcoholic teatohepatitis and iron:increased prevalence of mutations of the HFE gene in nonalcoholic steatohepatitis[J]. J Hepatol,1999,31:421-429.
    [100]李蕾.非酒精性脂肪性肝病的治疗进展[J].世界临床药物,2008,29(12):7 16-721.
    [101]NairS, DiehlAM, W isemanM, et a.1 Metformin in the treatment of nonalcoholic steatohepatitis:A p ilot open label trial[J]. Aliment Pharma2colTher,2004,20:2.
    [102]Uygun A, KadayifciA, IsikAT, et a.1 Metformin in the treatment of patients with nonalcoholic steatohepatitis [J]. AlimentPharmacolTher,2004,19: 537.
    [103]Ramesh S, SanyalAJ. Evaluation and magnagement of nonalcoholic steatohepatitis[J]. J Hepato,1 2005,42:2212.
    [104]Chen R, Guo Q, Zhu W J, et al. Therapeutic efficacy of w-3 polyunsaturated fatty acid capsule in treatment of patients with non-alcoholic fatty liver disease [J]. Shi Jie Hua Ren Xiao Hua Za Zhi,2008,16(18):2002-2006.
    [105]FEDOR D, KELLY DS. Prevention of insulin resistance by n-3 ployunsaurated fatty acids [J]. Curr Opin Clin Nutr Metab Care,2009,12:138-146.
    [106]BUCKLEY JD, HOWE PR. Anti-obesity effects of long-chain omega -3 polyunsaturated fatty acid [J]. Obes Rev,2009,10:648-659.
    [107]Harrison SA, Torgerson S, HayashiP, eta.1 Vitamin E and vitamin C treatment improves fibrosis in patientswith nonalcoholic steatohepatitis[J]. Am JGastroentero,1 2003,98 (11):2485.
    [108]Dentico P, Volpe A, Buongiorn R, et al. Glutathione in the treatment of chronic fatty liver diseases[J]. Recenti Prog Med,1995,86(7-8):290-293.
    [109]Abdelmalek M F, Angulo P, Jorgensen R A, et al.Betain, a promising new agentof patientswith nonalcoholic steatoheptitis:results of a pilot study[J].Am J Gastroenterol,2001,96(9):2711-2717.
    [110]Zheng J. Study on the changes of TG, CHOT, LDL, ALT in serum of fatty liver suffering with intestinal dysbacteriosis[J]. Zhongguo Wei Sheng Tai Xue Za Zhi.2007,19(2):161-162. Chinese with abstract in English.
    [111]Adams L A, Zein C 0, Angulo L.Apilot trial of pentoxifylline,in nonalconolic steat ohepatitis[J]. Am J Gastroenterol,2004,99(12):2365-2368.
    [112]孙林林,石军,郝菁华,等.低分子肝素对高脂饮食大鼠非酒精性脂肪性肝炎模型的影响[J].山东大学学报(医学版),2010,48(11):58-62.
    [113]Hirose A, Ono M, Saibara T, et al. Angiotensin Ⅱ type 1 receptor blocker inhibits fibrosis in rat nonalcoholic steatohepatitis[J]. Hepatology,2007, 45(6):1375.
    [114]Harrison SA, Fecht W, Brunt EM, et al.Orlistat for overweight subjects with nonalcoholic steatohepatitis:a randomized, prospective trial[J]. Hepatology,2009,49(1):80.
    [115]Kenny PR, Brady DE, Torres DM, et al. Exenatide in the treatment of diabetic patients with non-alcoholic steatohepatitis:a case series[J].Am J Gastroenterol,2010,105(12):2707.
    [1]李军祥,陈润花,苏冬梅,等.中医药治疗非酒精性脂肪性肝病研究述评[J].世界华人消化杂志,2010,18(14):1443-1451.
    [2]张声生,王垂杰,沈洪主编.特色专科实用手册·消化病(第1版)[M].中国中医药出版社出版,2008,214-221.
    [3]季光,郑培永.医药治疗脂肪肝的研究评述[J].中医药学刊,2004,22(1):87-90.
    [4]李玉红,张伯礼,徐宗佩,等.脂肪肝中医药辨治系统评价[J].辽宁中医杂志, 2002,29(11):657-658.
    [5]朱文峰主编.国家标准应用·中医内科疾病诊疗常规[M].湖南科学技术出版社出版,1999,395-396.
    [6]程华焱,曾斌芳.脂肪肝中医病名的文献研究[J].新疆中医药,2008,26(6):12-14.
    [7]刘天虹.健脾益肾祛瘀法治疗脂肪肝34例[J].河北中医,2004,26(11):818.
    [8]中华中医药学会脾胃病分会.非酒精性脂肪性肝病中医诊疗共识意见[J].北京中医药,2011,30(2):83-86.
    [9]魏华凤,季光,邢练军.脂肪肝辨证分型规律的初步研究[J].辽宁中医杂志,2002,29(11):655-656.
    [10]王骏,魏玮.脂肪肝的中医辨证分型和血清生化指标之间的关系[J].中西医结合肝病杂志,2004,14(6):371-372.
    [11]杨钦河,周迎春,郭桃美,等.不同治法方药对脂肪肝大鼠血脂作用的比较研究[J].新中医,2004,36(5):74-75.
    [12]慕永平,都金星,刘平,刘平教授治疗非酒精性脂肪性肝炎遣方用药经验探析[J].中西医结合肝病杂志,2009,19(3):170-171.
    [13]陈润花,张海鹏,李军祥.李军祥教授治疗脂肪肝经验.光明中医,2008,4(23):426.
    [14]王雁翔,王灵台,高月求,等.脂肪肝中医证型流行病学调查及其中医病因病机初探[J].中国中西医结合杂志,2005,25(2):126-130.
    [15]李少东,李红山,冯琴,等.脂肪肝中医证型分类的文献分析[J].中西医结合肝病杂志,2006,16(4):255-257.
    [16]叶放,赵文霞.中医对非酒精性脂肪肝的研究现状[J].中国中西医结合消化杂志,2003,11(1):60-62.
    [17]蒋俊民,迟晓玲.广州与西宁地区非酒精性脂肪性肝病中医证型调查分析[J].江苏中医药,2008,40(9):30.
    [18]袁洋,卜平,孔桂美,等.112例非酒精性脂肪肝证候病机的研究[J].中西医结合肝病杂志,2007,17(1):40-42.
    [19]王雁翔,丁桂芳,陈理书,等.780例非酒精性脂肪肝中医证型流行病学调查[J].中西医结合肝病杂志,2007,17(6):364-365.
    [20]中华中医药学会脾胃病分会.非酒精性脂肪性肝病中医诊疗共识意见[J].中国中西医结合消化杂志,2010,18(4):276-279.
    [21]郭遂成.从瘀论治非酒精性脂肪肝80例[J].陕西中医,2006,27(11):1355-1356.
    [22]王亚平,要全保,张志银,等.益肝降脂方治疗痰瘀互结型非酒精性脂肪肝64 例[J].上海中医药杂志,2006,40(2):21-22.
    [23]凌嫚芝.桑明合剂治疗非酒精性脂肪肝60例[J].四川中医,2007,25(7):42-45.
    [24]黄育华,孙洁,徐建良,等.活血化痰颗粒治疗非酒精性脂肪性肝炎25例[J].中西医结合肝病杂志,2011,21(5):300-301.
    [25]李鑫,邢丽,孙丽华,等.降脂保肝汤治疗非酒精性脂肪性肝炎38例[J].辽宁中医药大学学报,2009,11(5):114-115.
    [26]陆文烈,赵柏良.化痰解郁法治疗非酒精性脂肪性肝炎40例[J].中医研究,2008,21(6):37-39.
    [27]刘洪波.脂平汤治疗非酒精性脂肪肝98例[J].河南中医,2006,26(8):33.
    [28]陈常云,郭彦敏,聂宝增,等.化痰祛脂汤治疗非酒精性脂肪肝临床研究[J].中国临床医生,2010,38(4):290.
    [29]焦宝娟,万树全.健脾利湿中药汤剂治疗非酒精性脂肪性肝病40例[J].中国临床医生2010,38(9):41-42.
    [30]欧阳亮,李贤军.健脾消滞饮治疗非酒精性脂肪性肝炎的临床观察[J].医学信息,2009,1(10):145.
    [31]林佑武,张诗军,陈泽雄,等.运脾通络方治疗非酒精性脂肪性肝炎疗效观察[J].中药材,2012,34(7):1158-1162.
    [32]季光,范建高,陈建杰,等.胆宁片治疗非酒精性脂肪性肝病(湿热型)的临床研究[J].中国中西医结合杂志,2005,25(6):485-489.
    [33]张北平,黄绍刚,黄穗平.疏肝降脂片治疗非酒精性脂肪肝38例[J].陕西中医,2002,6(9):881-882.
    [34]徐洪涛,马建,贺环宇.疏肝健脾清热利湿法对非酒精性脂肪性肝炎血清TNF-α的影响[J].航空航天医药,2010,21(10):1905.
    [35]王树林.补脾升阳泻火法治疗非酒精性脂肪性肝炎43例[J].山东中医杂志,2009,28(9):604-605。
    [36]王立新.辨病与辨证治疗非酒精性脂肪性肝炎临床疗效研[J].天津中医药,2009,26(4):289-290.
    [37]张卫星,吴忆东.补肝肾法治疗2型糖尿病性非酒精性脂肪肝50例[J].浙江中医杂志,2009,44(4):269.
    [38]陈丽华.肝脂康治疗非酒精性脂肪肝45例[J].光明中医,2008,23(3):319-320.
    [39]黄欣,张哲永.当飞利肝宁胶囊治疗非酒精性脂肪性肝病32例临床观察[J].中医杂志,2007,48(6):524-525.
    [40]邓银泉,范小芬,李友第.水飞蓟素治疗非酒精性脂肪性肝炎的临床观察[J].中 国中医药杂志,2005,30(13):1044-1045.
    [41]赵文霞,张玉禄.消脂护肝胶囊对非酒精性脂肪肝患者胰岛素抵抗及瘦素水平的影响[J].中医研究,2005,18(5):21-23.
    [42]金永日.六味五灵片治疗非酒精性脂肪性肝炎疗效观察[J].中国肝脏病杂志(电子版),2011,3(4):29-31.
    [43]张亚东.阿拓莫兰联合中药降脂汤治疗非酒精性脂肪性肝炎疗效观察[J].中国误诊学杂志,2006,6(13):2505-2506.
    [44]张小平,向龙珍,方园.中西医结合治疗非酒精性脂肪肝36例[J].湖南中医杂志,2006,22(5):54-55.
    [45]白洁,邓彩云,史美媛,等.中西医结合治疗非酒精性脂肪肝的临床观察[J].中国煤炭工业医学杂志,2005,8(8):910.
    [46]孙海潮,胡青海.中药离子导入治疗非酒精性脂肪性肝炎的临床应用[J].中国民族民间医药,2010,4:114-115.
    [47]孟胜喜.针刺治疗非酒精性脂肪性肝炎疗效观察[J].中国针灸,2009,29(8):616-618.
    [48]曾志华,冯雯琪,卓廉士.电针对非酒精性脂肪肝肝细胞色素P4502E1表达及氧化抗氧化的影响[J]第四军医大学学报,2008,29(11):994.
    [49]金建军,徐亚莉,郑昱.穴位注射对非酒精性脂肪肝血脂代谢及肝功能的影响[J].中国针灸,2006,26(2):100-102.
    [50]黄德亚,姚东坡,朱志义.“虎杖降脂汤”内服结合耳穴按压法治疗脂肪肝80例临床观察[J].江苏中医药,2008,40(4):44-45.
    [51]明玉华.隔药饼灸联合药物治疗非酒精性单纯性脂肪肝81例[J].中西医结合肝病杂志,2008,18(3):172-173.
    [52]周晓玲,谢胜,肖文胜,等.穴位埋线结合强肝消脂饮对非酒精性脂肪肝血清瘦素水平及胰岛素抵抗指数的影响[J].中国中医急症,2010,19(11):1845-1848.
    [53]黄振,宋双临.穴位埋线治疗非酒精性脂肪肝60例临床观察[J].山东中医药大学学报,2012,36(3):211-212.
    [54]杨露梅,许平,朱钰宝.整合疗法治疗非酒精性脂肪肝30例临床观察[J].河北中医,2006,28(5):348-349
    [55]朱代群,李蕾,曾昭萍.生物信息治疗仪联合中药离子导入治疗肝功能损害疗效观察[J].中国医师杂志,2008,10(5):646.
    [1]Alina P, Ralucaais, Vlad R. An overview of nonalcoholic steatohepatitis:past, present and puturedirections[J]. J Gastrointestin Liver Dis,2010,19(4):415.
    [2]翁小刚,朱晓新,杨庆,等.非酒精性脂肪肝的实验研究进展[J].中国实验方剂学杂志,2009,15(11):104.
    [3]London R, George J. Pathogenesis of NASH:animal models[J].Clin Liver Dis, 2007,11(1):55.
    [4]郑全喜,王昆,刘超.非酒精性脂肪性肝病动物模型的研究进展[J].中国实验方剂学杂志,2013,19(2):357-360.
    [5]Hatsugai K, Ohkohchi N, Fu kumori T, et al. Mechanism of primary graft non2fu nct ion in a rat model for fatty liver transplantation [J]. Trans pl Int, 2000,13(Suppl 1):S583-589.
    [6]Charles S Li eber, Maria A L eo, Ki M Mak, et al. Model of nonalcoholic steatohepatitis [J]. American J ou rnal of Clinical Nutrition,2004,79(3): 502-509.
    [7]Zvi Ack erman, Mor Oron Herman, Maria Grozovski Talma Rosenthal, et al. Fructose Induced fat ty liver di sease hepatic effect s of blood pressu re and plasma t riglyceride reduction[J]. Hypertension,2005,45(5):1012-1018.
    [8]Kirsch R, Clarkson V, Shephard EG,et al. Rodent nutritional model of non-alcoholic steatohepatitis:species, strain and sex difference studies[J]. J Gastroenterol Hepatol,2003,18(11):1272-1282.
    [9]Anstee Q M, Goldin R D. Mouse models in non-alcoholic fatty liver disease and steatohepatitis research [J]. Int J Exp Pathol,2006,87(1):1-16.
    [10]Xu ZJ, Fan JG, Ding XD, et al. Characterization of high-fat, diet-induced, non-alcoholic steatohepatitis withfibrosis in rats[J] Dig Dis Sci,2010,55(4): 931.
    [11]Chun g H, H ong DP, Kim H J, et al. Differential gene expression profil es in the s t eatos is/fib rosis model of rat liver by chronic adminis tration of carbon tetrachloride[J].Toxicol Appl Pharmacol,2005,208(3):242-254.
    [12]Ito M, Suzuk i J, Sasaki M, et al. Development of nonal coholic steatohepat it is model th rou gh comb inat ion of high2fat diet and tetracycline with morbid obesity in mice [J]. Hepatology Research,2006,34(2):92-98.
    [13]孙创斌.去脂软肝丸对四环素致小鼠脂肪肝模型肝脂的影响[J].云南中医学院学报,2002,325(1):1-4.
    [14]孙蓉.脂肝清颗粒对脂肪肝模型大鼠的影响[J].中药新药与临床药理,2003,14(5):313-316.
    [15]Fan CY, Pan J, Chu R, et al. Hepatocellular and hepatic peroxisomal alterations in mice with a disrupted peroxisomal fatty acyl-coenzyme A oxidase gene[J].J Biol Chem,1996,271:24698-24710.
    [16]Lin HZ, Yang SQ, Chuckaree C, et al. Met formin's reverses fatty liver dis ease in obes e 1 ept in2deficient mice [J]. Nature Medicine,2000,6(8):998-1003.
    [17]Shek hawat PS, Yang HS, Bennett MJ, et al. Carnitine content and expression of mitochondrial beta-oxidation enzymes inplacent as of wild 2 type (OCTN2 +/+) and OCTN2 Null (OCTN2 -/-) mice [J]. Pediatr Res,2004,56(3):323-328.
    [18]刘晓红,吕宗舜,王绪林.益肾软坚煎抑制脂肪肝脂质过氧化的实验研究[J].潍坊医学院学报,2006,28(4):276-277.
    [19]刘建忠,肖明中,韦丹,等.不同治则抗幼年大鼠非酒精性脂肪肝作用的实验比较研究[J].湖北中医杂志,2011,33(9):3-5.
    [20]张慧,冯琴,李红山,等.祛湿化瘀方对非酒精性脂肪性肝炎大鼠组织蛋白酶B和肿瘤坏死因子α表达的影响[J].中西医结合学报,2008,6(9):928-933.
    [21]梁惠卿,陈少东,张其清,等.茵陈蒿汤防治大鼠非酒精性脂肪性肝炎的实验研 究[J].光明中医,2009,24(2):212-214.
    [22]施军平,范建高,陈芝芸,等.理气化痰祛瘀方对非酒精性脂肪性肝炎大鼠肝组织PPAR和CPT-I表达的影响[J].中华中医药学刊,2008,26(8):1739-1743.
    [23]殷果华,赵和平.降脂益肝冲剂对非酒精性脂肪性肝炎大鼠肝脏PPARα mRNA表达的影响[J].中国药物与临床,2008,8(11):873-875.
    [24]孙凤霞,张姝媛,王宪波,等.清肝化痰活血方对大鼠非酒精性脂肪肝的防治研究[J].时珍国医国药,2010,21(11):2851-2853.
    [25]苏冬梅,李健,李军祥,等.健脾疏肝方对非酒精性脂肪性肝炎大鼠ADP、TNF-α的影响[J].北京中医药大学学报,2011,34(12):826-831.
    [26]周岳君,姚海清.脂易消对非酒精性脂肪肝炎大鼠肝组织APNmRNA的调控[J].浙江中医药大学学报,2012,36(3):298-302.
    [27]赵二劳,刘宣.山楂抗氧化性及其协同作用的研究[J].江西师范大学学报(自然科学版),2007,31(6):570-572.
    [28]范学辉,张清安.不同剂量山楂籽油连续给小鼠经口灌胃[J].江西农业学报,2007,19(10):111-112.
    [29]林秋实,陈吉樆.山楂及山楂黄酮预防大鼠脂质代谢紊乱的分子机制研究[J].营养学报,2000,22(2):131-136.
    [30]严茂祥,陈芝芸,高晓倩,蔡丹莉.山楂叶总黄酮对非酒精性脂肪性肝炎大鼠肝组织TNF-α、Leptin和IL-8表达的影响[J].中国中医药科技,2010,17(6):519-521.
    [31]刘涛,徐秋玲,赵岩,等.大黄素对非酒精性脂肪肝大鼠脂质水平及肝脏脂质代谢基因表达的影响[J].中草药,2010,41(9):1516-1518.
    [32]汪兴生,解光艳,史学礼,等.何首乌等对脂肪肝模型大鼠血液指标的影响[J].安徽中医学院学报,2006,25(5):39-40.
    [33]张荣,刘必旺,王永辉.决明子乙酸乙酯提取物对非酒精性脂肪肝大鼠的防治作用[J].山西中医,2009,25(7):45-47.
    [34]阎君宝,金龙,汪江碧.决明子抑制营养性肥化大鼠TPA、PAI、MDA及NO影响的研究[J].中国药房,2007,18(24):1847-1849.
    [35]王俊杰,舒洋,龙婷,等.荷叶黄酮治疗小鼠非酒精性脂肪肝的研究[J].中药药理与临床,2011,27(2):61-63.
    [36]刘兴,张利凤,高峰.银杏叶提取物对非酒精性脂肪性肝炎肝损伤的保护作用[J].实用医学杂志,2009,25(22):3770-3772.
    [37]李晶,冯五金.生山楂、泽泻、莪术对大鼠脂肪肝的影响及其交互作用的实验研究[J].山西中医,2006,22(3):57-59.
    [38]杨卫红.小柴胡汤及其药群配伍防治高脂血症性脂肪肝的实验研究.2008,6.
    [39]曾志华,曾明慧.电针对非酒精性脂肪肝胰岛素抵抗及肝细胞色素P450 2E1表 达的影响[J].江西中医药2010,41(336):48-51.
    [1]Tong Xiao-lin (仝小林), Zhao Lin-hua, Lian Feng-mei, et al. Clinical observations on the dose-effect relationship of Gegen Qin Lian Decoction on 54 out-patients with type 2 diabetes [J]. Journal of Traditional Chinese Medicine, 2011,31(1):56-59.
    [2]赵林华,姬航宇,仝小林,等.葛根芩连汤治疗糖尿病理论探讨[J].中华中医药杂志,2012,27(2):280-283.
    [3]王浩,徐明,王思程.葛根芩连汤对溃疡性结肠炎模型大鼠血清Bcl-2、TGF-β 1的影响[J].四川中医,2011,29(11):12-14.
    [4]陈润花,张海鹏,李军祥.李军祥教授治疗脂肪肝经验[J].光明中医,2008,4(23):426.
    [5]李军祥,陈润花,苏冬梅,等.中医药治疗非酒精性脂肪性肝病研究述评[J].世界 华人消化杂志,2010,18(14):1443-1451.
    [6]Kirsch R, Clarkson V, Shephard EG, et al. Rodent nutritional model of non-alcoholic steatohepatitis:species, strain and sex difference studies[J]. J Gastroenterol Hepatol,2003,18(11):1272-1282.
    [7]王允亮,诸葛丽,李军祥,等.调肝化浊解毒方对NASH大鼠的治疗作用及其机理研究[J].江苏中医药.2012,44(5):71-73.
    [8]钟岚.非酒精性脂肪肝动物模型[J].国外医学消化疾病分册,1999,19(3):175-178.
    [9]London RM, George J. Pathogenesis of NASH:animal models[J]. Clin Liver Dis,2007,11:55-74.
    [10]楼琦,石巧娟,郭红刚,等.非酒精性脂肪肝大鼠脂质代谢及病理变化的动态观察[J].中国比较医学杂志,2012,22(3):5-11.
    [11]非酒精性脂肪肝病的中西医结合诊治方案[C].中国中西医结合学会消化系统疾病专业委员.上海.第二十次全国中西医结合消化系统疾病学术会议论文汇编,2008.
    [12]Gastaldelli A, Miyazaki Y, Mahankali A, et al. The effect of pioglitazone on the liver:role of adiponectin[J]. Diabetes Care,2006,29:2275-2281.
    [13]Belfort R, Harrison SA, Brown K, et al. Aplacebo-controlled trial of pioglitazone in subjects with nonalcoholic steatohepatitis [J]. N Engl J Med, 2006,355:2297-2307.
    [1]潘雪丰,温彩霞,许建华.两种细胞系用于建立体外脂肪肝模型的比较[J].广东药学院学报,2010,26(1):85-89.
    [2]钟岚.非酒精性脂肪肝动物模型[J].国外医学消化疾病分册,1999,19(3):175-178.
    [3]London RM, George J. Pathogenesis of NASH:animal models[J]. Clin Liver Dis,2007,11:55-74.
    [4]楼琦,石巧娟,郭红刚,等.非酒精性脂肪肝大鼠脂质代谢及病理变化的动态观察[J].中国比较医学杂志,2012,22(3):5-11.
    [5]Yasuyuki Fujimoto, Jun Onoduka, Koichi J. Homma, et al. Long-Chain Fatty-Acids Induce Lipid Droplet Formation in a Cultured Human Hepatocyte in a Manner Dependent of Acyl-CoA Synthetase[J]. Biol Pharm Bull,2006,29(11):2174-2180.
    [6]Norberto C Chavez-Tapia, Natalia Rosso, Claudio Tiribelli. Effect of intracellular lipid accumulation in a new model of non-alcoholic fatty liver disease[J]. BMC Gastroenterol.2012,12:20.
    [1]Norberto C Chavez-Tapia, Natalia Rosso, Claudio Tiribelli. Effect of intracellular lipid accumulation in a new model of non-alcoholic fatty liver disease[J]. BMC Gastroenterol,2012,12:20.
    [2]Geoffrey C. Farrell, Derrick van Rooyen, Lay Gan, and Shivrakumar Chitturi. NASH is an Infl ammatory Disorder:Pathogenic, Prognostic and Therapeutic Implications [J]. Gut and Liver,2012,6(2):149-171.
    [3]郭启煜.胰岛素受体与胰岛素抵抗[J].海军总医院学报,1999,12(2):88-90.
    [4]宋春宇,毕会民.葛根素对大鼠胰岛素刺激下骨骼肌细胞膜GLUT4蛋白含量的影响[J].中国中药杂志,2004,29(2):172-175.
    [1]Norberto C Chavez-Tapia, Natalia Rosso, Claudio Tiribelli. Effect of intracellular lipid accumulation in a new model of non-alcoholic fatty liver disease[J]. BMC Gastroenterol,2012,12:20.
    [2]高风英.葛根素药理作用的研究进展[J].中草药,2003,34(12):附7-8.
    [3]李波,朱维良,陈凯先.小檗碱及其衍生物的研究进展[J].药学学报,2008,43(8):773-787.
    [1]Norberto C Chavez-Tapia, Natalia Rosso, Claudio Tiribelli. Effect of intracellular lipid accumulation in a new model of non-alcoholic fatty liver disease[J]. BMC Gastroenterol,2012,12:20.
    [2]Guo HX, Liu DH, Ma Y, et al. Long-term baicalin administration ameliorates metabolic disorders and hepatic steatosis in rats given a high-fat diet [J]. Acta Pharmacol Sin,2009,30(11):1505-1512.
    [1]Norberto C Chavez-Tapia, Natalia Rosso, Claudio Tiribelli. Effect of intracellular lipid accumulation in a new model of non-alcoholic fatty liver disease[J]. BMC Gastroenterol,2012,12:20.
    [2]Zhu J, Zhang J, Ji M, et al. The role of peroxisome proliferator-activated receptor and effects of its agonist, pioglitazone, on a rat model of optic nerve crush:PPAR γ in retinal neuroprotection[J]. PLoS One,2013,8(7):e68935.
    [3]Hahn SS, Tang Q, Zheng F, et al.GW1929 inhibits α 7 nAChR expression through PPAR γ -independent activation of p38 MAPK and inactivation of PI3-K/mTOR: The role of Egr-1[J]. Cell Signal,2014,26(4):730-739.
    [1]Kirsch R, Clarkson V, Shephard EG, et al. Rodent nutritional model of non-alcoholic steatohepatitis:species, strain and sex difference studies[J]. J Gastroenterol Hepatol,2003,18(11):1272-1282.
    [2]王允亮,诸葛丽,李军祥,等.调肝化浊解毒方对NASH大鼠的治疗作用及其机理研究.江苏中医药,2012,44(5):71-73.
    [3]冯高飞,杨钦河,张玉佩,等.黄连素配合高脂饮食控制对NAFLD大鼠肝组织PPARα mRNA及蛋白表达的影响[J].中药材,2012,35(4):629-634.
    [4]闫莉萍,陈舜宏,马小军,等.葛根素对膳食诱导的高胆固醇血症大鼠的血脂调节作用[J].中国临床药理学与治疗学,2006,11(5):574-577.
    [5]刘志勇,叶军,薛东英.黄芩苷对肝纤维化大鼠相关细胞因子表达的影响[J].中国当代医药,2013,20(12):18-21.

© 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