用户名: 密码: 验证码:
角蛋白18及其磷酸化在HBV感染慢性肝病及肝细胞凋亡中的作用研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
角蛋白18(keratin 18,K18)是构成成熟肝细胞骨架的主要中间纤维丝蛋白之一,其主要作用是保护肝细胞免受各种机械性和非机械性的损伤。K18的突变、结构的重组以及磷酸化的改变会影响肝细胞的完整性,引起肝细胞的损伤。此外,K18除了发挥抗机械性损伤作用之外还参与着重要的细胞信号传导作用,包括Fas/FasL、TRADD等相关的凋亡通路都与K18及其磷酸化有关。已有研究发现K18磷酸化水平在慢性丙型肝炎时出现增高,并且增高的程度与肝病进程呈正相关。在我国乙型肝炎病毒(HBV)感染是慢性肝病的最常见病因,关于HBV感染的肝病进展与K18及其磷酸化之间关系的研究在国内外未见报道。因此,研究K18及其磷酸化对揭示HBV感染慢性肝病肝损伤及肝细胞凋亡的机制具有重要意义,对于临床制定个体化的治疗方案、判定预后都具有重要的参考价值和临床应用前景。
     p53是重要的促凋亡基因,p53及其介导的凋亡通路在HBV感染慢性肝病的肝细胞损伤中发挥着重要的作用。ASPP2(Apoptosis Stimulating Protein 2 of P53, ASPP2)是p53凋亡刺激蛋白家族的重要成员,能够与p53结合选择性地提高p53的促凋亡活性。我们在前期的工作中应用酵母双杂交技术发现,ASPP2能够与K18结合。这一发现对于揭示K18及其磷酸化与肝细胞凋亡的关系和探索其作用方式提供了重要线索。本研究围绕K18及其磷酸化在HBV感染慢性肝病和肝细胞凋亡中的作用及其作用机制进行了系统研究。
     1、角蛋白18磷酸化在HBV感染慢性肝病中的作用及其意义
     为了研究K18及其磷酸化在慢性乙型肝炎(chronic hepatitis B, CHB)病程进展中的变化及其意义,我们收集了40例慢性乙肝患者肝穿刺组织,21例肝硬化肝组织(其中静止性肝硬化11例,活动性肝硬化10例)和5例慢性重型肝炎肝组织,以12名健康肝移植供体肝组织作为对照。首先应用免疫荧光技术,分析了不同慢性肝病进程当中K18及其磷酸化在肝细胞内的表达和定位,结果发现,K18在正常肝组织中表达水平较高,呈网状分布;慢性肝炎中表达水平无显著变化;但在肝硬化肝组织和慢性重型肝炎肝组织中,其组织正常结构被破坏,许多新生胆管细胞中K18水平较高;K18 Ser33和Ser52在正常肝组织中磷酸化水平较低,但是随着肝损伤程度的增加,在新生胆管细胞中磷酸化水平明显增高。
     应用Western Blotting的方法检测肝组织中K18磷酸化水平,分组分析K18磷酸化水平与慢乙肝病程进展的关系。
     (1)将肝组织按照疾病进展分为正常组织,慢性肝炎和肝硬化组。结果发现,K18 Ser52磷酸化水平在正常肝组织中较低,而慢性肝炎组增高,至肝硬化组织达到最高水平;K18 Ser33磷酸化水平在慢性肝炎和肝硬化两组均显著高于正常组织,但两组之间无显著性差异。
     (2)进一步将40例慢性乙型肝炎患者的肝组织按照Ishak组织学评分的高低分为3组,即轻度损伤(MiH)组、中度损伤(MeH)组、重度损伤(AdH)组,肝硬化组织按照炎症程度分为静止性肝硬化和活动性肝硬化,五组之间进行比较研究其K18磷酸化水平的差异。结果发现,K18 Ser52磷酸化水平随着肝病进程而逐渐增加,从正常肝组织到活动性肝硬化,相邻两组之间有显著性差异。K18 Ser52磷酸化水平变化趋势是:正常组织< MiH < MeH < AdH <静止性肝硬化<活动性肝硬化。K18 Ser33位磷酸化水平从正常组织到活动性肝硬化亦呈上升趋势,在慢性肝炎肝组织中,从轻度损伤至重度损伤,K18 Ser33磷酸化增加明显(p<0.005),但在静止性肝硬化组织中磷酸化水平较低,其水平与MiH组相当(p=0.750),而慢肝中重度损伤组磷酸化水平与活动性肝硬化相当(p=0.715),即K18 Ser33磷酸化水平变化趋势是:正常组织< MiH≈静止性肝硬化< MeH < AdH≈活动性肝硬化。
     (3)为了进一步探究K18磷酸化与整体肝组织损伤的关系,我们分析了K18磷酸化与临床上最常用的反映肝功能损伤的生化指标谷丙转氨酶(ALT)之间的关系。将本研究中所有慢性乙肝患者的肝组织根据其ALT水平进行分组(1,ALT<40U/L;2,ALT:40~200 U/L;3,ALT>200 U/L),分别与正常肝组织进行比较。结果显示K18 Ser52磷酸化在正常肝组织组、ALT<40U/L组、40~200 U/L组和>200 U/L组中的均值分别为0.73,0.72,1.77和2.71。其中后两组与前两组比较均有显著性差异,而正常组织和ALT<40 U/L组比较无显著性差异(p=0.976),表明在HBV感染但肝功能未受损伤的肝组织中K18 Ser52磷酸化水平并不增加。提示K18 Ser52磷酸化水平反映了肝组织的损伤情况,但与HBV感染本身并无直接联系。而K18 Ser33位磷酸化水平随着肝功能水平变化增高明显,在四组中均值分别为1.02,1.96,2.53和3.00,各组之间存在显著性差异。值得注意的是,在ALT<40 U/L组,K18 Ser33位磷酸化水平也出现增高,与正常相比差异显著。这一结果提示我们,K18 Ser33位磷酸化可能与HBV感染有关系。
     (4)为了进一步研究HBV感染对于K18磷酸化水平的影响,我们选取了HBV感染但是肝功能水平正常的慢性乙肝携带者的肝组织标本,冰冻切片后分别进行了HBsAg抗体和K18两种磷酸化抗体的双染免疫荧光检测,结果发现,HBsAg在HBV感染的肝细胞胞浆中表达,呈现大面积红染,而在正常对照肝组织中未见染色。K18 Ser33磷酸化恰恰在HBV感染的细胞中出现异常增高,而K18 Ser52磷酸化水平仍然较低,与周围细胞无差异。
     本研究表明:K18磷酸化水平是慢性乙型肝炎肝损伤的重要指标;K18 Ser52位磷酸化是慢性乙型肝炎肝病进展的判定指标;K18 Ser33位磷酸化可能是HBV感染的早期现象,并且是肝组织炎症损伤程度的判定指标。
     2、角蛋白18与p53凋亡刺激蛋白2(ASPP2)在肝细胞凋亡中的作用研究
     我们前期的研究发现,K18能够和p53凋亡刺激蛋白(ASPP2)结合。为进一步证实这种结合关系并研究其对于肝细胞凋亡的意义,我们从三个方面进行了研究。
     (1)K18及其磷酸化和ASPP2在肝细胞凋亡中的作用研究:应用免疫荧光双染发现,在正常HepG2细胞中,K18和ASPP2均表达在细胞浆中,给予药物处理的细胞发生凋亡时,ASPP2进入到细胞核中,发生核转位。应用不同浓度的甲基甲烷磺酸盐(MMS)作用于HepG2细胞,用Annexin V/PI染色流式细胞法检测细胞凋亡水平,研究K18及其磷酸化和ASPP2在肝细胞凋亡不同状态下的水平变化。结果发现,K18总体水平随着药物浓度增加而降低,ASPP2和K18 Ser52位磷酸化水平随着药物浓度增加而增加,K18 Ser33磷酸化在低浓度药物作用下水平增高,但在高浓度药物作用下反而降低。以上结果说明,ASPP2在肝细胞发生凋亡时出现核转位,从而进入到细胞核中参与p53介导的细胞凋亡通路,而K18的Ser33或Ser52磷酸化水平的变化与肝细胞凋亡有密切关系。
     (2)ASPP2与K18结合功能区域的确定:采用重组的GST-mASPP2,ASPP2 N-末端和C-末端融合蛋白分别与K18融合蛋白进行免疫共沉淀。实验结果发现ASPP2的N-末端能够与K18结合。进一步将ASPP2的N-末端逐步截短,表达了ASPP2 N-末端的截短小片段融合蛋白,再次与K18进行体外免疫沉淀反应,结果发现,ASPP2的N-末端位于83-105个氨基酸残基之间正是与K18结合的关键部位,而其下游的氨基酸残基则与结合能力有关系。应用293T细胞分别高效转染K18-pcDNA4或K18 S33A、K18 S52A,提取细胞总蛋白应用GST-mASPP2融合蛋白进行免疫沉淀反应,结果发现,ASPP2能够与K18结合,但在转染突变K18的细胞中结合降低,在转染S33A突变K18的细胞中结合最少。提示这种结合可能与33位的丝氨酸磷酸化有关系。为了研究这种结合与肝病进展的关系,我们提取了活动性肝硬化和静止性肝硬化肝组织的总蛋白,利用免疫共沉淀技术分析了K18和ASPP2的结合水平与肝病进展的关系,结果发现,ASPP2在静止性肝硬化中与K18的结合更强,在活动性肝硬化结合显著减少。说明,在肝组织受到严重损伤时,ASPP2与K18的结合减少。
     (3)K18与ASPP2结合作用方式的初步研究:将细胞共转染ASPP2和K18或S33A,S52A定点突变的K18,给予细胞药物刺激,转染S33A K18的细胞发生凋亡时,ASPP2全部进入到细胞核中,而转染K18和S52A K18的细胞发生凋亡时,ASPP2部分进入到核中,大部分仍然位于细胞浆中,说明K18 Ser33位磷酸化与ASPP2的核转位有关。应用报告基因分析法研究K18对ASPP2-p53及其调控的下游分子Bax转录活性的影响。结果发现:ASPP2的存在能够显著提高p53刺激的Bax-luc报告基因活性,当加入过量野生型K18后,Bax-luc报告基因活性大大减低,提示K18可能参与了抑制ASPP2的促p53转录激活作用;当用K18 S33A质粒替代K18后,Bax-luc报告基因活性水平与ASPP2单独存在时大致相同。这一结果再次提示K18 Ser33位磷酸化能够促进K18与ASPP2的结合,从而间接抑制ASPP2与p53的结合而发挥凋亡抑制作用。
     上述实验研究表明:K18磷酸化在肝细胞凋亡中发挥着重要的作用;正常情况下K18在细胞浆中以Ser33位磷酸化依赖的方式结合ASPP2,在细胞遇到轻度损伤性刺激时,K18 Ser33和Ser52位磷酸化水平均出现增高,使得ASPP2与K18的结合水平增加;但当细胞受到严重刺激时,K18 Ser33位磷酸化水平反而降低,导致ASPP2与K18的结合变得松弛,ASPP2进入到细胞核与p53结合中发挥其促凋亡的作用。
Keratin 18 (K18) is one of the major intermediate filament proteins forming the cytoskeleton of human mature hepatocytes, and its main function is to protect hepatocytes from mechanical and non-mechanical injuries. Mutations, structural reorganization, and changes in phosphorylation in K18 may affect the integrity of hepatocytes and cause their injury. In addition to its role in resisting mechanical injury, K18 also participates in some key signal transduction activities, for example, apoptosis pathways such as Fas/FasL, TRADD have been found to be related to K18 phosphorylation. Studies have shown that K18 phosphorylation level increases in chronic liver diseases with hepatitis C virus (HCV) infection, and the degree of increase is positively associated with liver disease progression. In China , hepatitis B virus (HBV) infection is the most common cause of chronic liver diseases, but there has been no report on the relation of K18 and its phosphorylation to chronic liver diseases with HBV infection. Therefore the study of this relationship will help to reveal the mechanisms of liver injury and hepatocyte apoptosis in chronic liver injury with HBV infection, and it can also be valuable for clinicians in making individualized treatment plan and prognosis.
     P53 is an important pro-apoptosis gene; p53-mediated apoptosis pathway plays an important role in hepatocytes injury in chronic liver diseases with HBV infection. Apoptosis Stimulating Protein 2 of P53 (ASPP2) is an important member of the p53 apoptosis stimulating proteins family and it can bind with p53 and selectively increase the pro-apoptotic activity of p53. In our previous work, we found by yeast two-hybrid technique that ASPP2 can bind with K18, which provides a key clue in revealing the relationship between K18 phosphorylation and hepatocyte apoptosis and explore the underlying mechanism. The current study systemically investigated the role of K18 and its phosphorylation in HBV infection related chronic liver diseases and hepatocyte apoptosis and its mechanism of action.
     1. The function and significance of K18 phosphorylation in HBV infection related chronic liver diseases:
     To investigate changes of K18 phosphorylation in the progression of chronic B type hepatitis (CHB) and its significance, liver tissue specimens were collected from 12 healthy donors (used as control), 40 chronic hepatitis B patients, 21 cirrhosis patients (11 inactive, 10 active) and 5 patients of severe chronic hepatitis. The cellular location of K18 expression and its phosphorylation level in different stages of CHB was first analyzed with immnofluorescence staining. In normal liver tissues, K18 expression level was high and distribute in a net form. Its expression level did not change much in chronic hepatitis. However in cirrhosis and chronic severe hepatitis, normal structure of liver tissue was damaged and K18 was expressed at high levels in proliferative bile duct epithelial cells. The phosphorylation of K18 Ser-33 and Ser-52 was low in normal liver tissues but as liver injury intensified the level increased markedly in proliferative bile duct epithelial cells.
     Western Blotting was used to analyze K18 phosphorylation in liver tissues.
     (1) Liver tissues were divided into normal, chronic hepatitis and cirrhosis. K18 Ser52 phosphorylation showd low level in normal liver, increased in chronic hepatitis and reached high level in cirrhosis. K18 Ser-33 phosphorylation in CHB and cirrhosis were both higher than normal tissue, but showed no evident difference between the two groups.
     (2) Chronic hepatitis was further divided into three groups (minimal histological lesions, MiH; medium lesions, MeH; advanced lesions, AdH) and cirrhosis were divided into inactive cirrhosis (inactive-LC) and active cirrhosis (active-LC). K18 phosphorylations were compared among the five groups. K18 Ser-52 phosphorylation was increased along with the progression of liver disease. From normal liver to active cirrhosis, there are distinctive differences between consecutive groups. The relative level of Ser52 phosphorylation follows the trend of Ser52 normal < MiH < MeH < AdH < inactive LC < active LC. K18 Ser33 phosphorylation also displayed increasing trend from normal tissue to active cirrhosis. From MiH to AdH in chronic hepatitis, K18 Ser33 phosphorylation increased evidently (p<0.005). But K18 Ser33 phosphorylation showed low level in inactive-LC and was almost equivalent to MiH(P = 0.750). K18 Ser33 phosphorylation level in AdH was also equivalent to active-LC (P = 0.715). The relative level of Ser33 phosphorylation follows the trend of normal < MiH≈inactive LC < MeH < AdH≈active LC.
     (3) To further investigate the relationship of K18 phosphorylation and the total injury in liver tissue, K18 phosphorylation was analyzed to compare with the most commonly used clinical marker ALT. 40 CHB livers were divided into 3 subgroups (1, ALT < 40 U/L; 2, 40≤ALT <200 U/L; 3, ALT≥200 U/L). The mean relative levels of Ser52 phosphorylation in normal control liver and the three groups were: 0.73, 0.72, 1.77 and 2.71. Ser52 phosphorylation was equivalent in the control and subgroup 1 (P = 0.976), while significantly increased subgroups 1 and 2 (P < 0.001), and subgroups 2 and 3 (P < 0.001) . The positive correlation between levels of phosphorylation and ALT activities suggested that the phosphorylation of Ser52 in K18 was progressively related to liver injuries but had no direct relation with HBV infection. K18 Ser33 phosphorylation increased along with ALT, the mean relative level of the four subgroups were: 1.02, 1.96, 2.53 and 3.00. It was notable that K18 Ser33 phosphorylation increased in subgroup 2 and had significant difference to normal liver. This results indicated that K18 Ser33 phosphorylation might be related to HBV infection, therefore, K18 Ser-33 still increased in non-injured liver.
     (4) To further investigate the effect of HBV infection on K18 phosphorylation, a liver specimen respectively from a randomly selected inactive HBV carrier and a healthy donor was sectioned, and probed for HBsAg and phosphorylated Ser33 or Ser52. Strong signals of phosphorylated Ser33 were restricted to hepatocytes with strong positive detection of HBsAg. However, only basal levels of Ser33 phosphorylation were detected in the control. Basal levels of Ser52 phosphorylation were observed in both HBV positive and healthy samples. These results reiterated the conclusion that phosphorylation at Ser33 in K18 was an indicator of HBV infection.
     This study indicated: K18 phosphorylations were important injure markers of CHB. K18 Ser52 phosphorylation was a marker of progression of CHB; K18 Ser33 phosphorylation may be an early indicator of HBV infection and marker of liver inflammation.
     2. Study of the function of Keratin 18 and p53 apoptosis stimulate protein ASPP2 in hepatocyte apoptosis
     K18 was found in our previous work to bind to ASPP2 by Yeast two-hybrid. To further confirm the interaction of the two proteins and investigate the significance, we studied from three aspects.
     (1) Study of the function of K18 phosphorylation and ASPP2 in apoptosis of hepatocytes: It was found by immnofluorescence double-staining that ASPP2 and K18 both located in cytoplasm in normal cells. But when cells were treated with drugs and induced into apoptosis, ASPP2 entered into nucleus. HepG2 cells were then treated with MMS with different concentration, apoptosis were stainedby Annexin V/PI and analyzed by Flow cytometry. It was found that total K18 decreased but ASPP2 and K18 Ser52 phosphorylation increased along with the increase of cell apoptosis level. Ser33 phosphorylatoin increased under low level MMS, but decreased under high level MMS. The results above indicated: when hepatocytes were apoptosis, ASPP2 entered into nucleus to participate in p53-mediated apoptosis pathway. K18 Ser33 or Ser52 phosphorylation had close relationship with hepatocytes apoptosis.
     (2) Determination of binding domain of ASPP2 and K18: Recombination GST-mASPP2, ASPP2 N-terminal and C-terminal fusion proteins were prepared to Co-IP with K18 fusion protein in vitro. It was found that ASPP2 N-terminal can bind with K18. Then ASPP2 N-terminal was further truncated. The small fragment fusion proteins were expressed to Co-IP with K18. 83-105aa of ASPP2 N-terminal was found to be the key domain of binding. The followed amino acid residues of N-terminal were correlated with binding ability. 293T cells were transfected with K18 ,S33A K18 or S52A K18, then total protein were extracted to Co-IP with ASPP2 in semi-in vivo. It was found that ASPP2 could combine with K18, but the hybrid signal was much lower in cells transfected with mutated K18. The binding signal in cells transfected with S33A K18 was the lowest, which suggested that Ser33 phosphorylation was related with binding of ASPP2 and K18. To investigate the relationship of the combination with the progression of liver disease, total protein of active cirrhosis and inactive cirrhosis tissues were extracted. Then the relationship between the combination of ASPP2 and K18 and the progression of liver disease were analyzed by Co-IP. It was found that ASPP2 binded with K18 much more in inactive cirrhosis, but the hybrid signal was barely detected in active cirrhosis. It indicated that binding of ASPP2 and K18 decreased in serious injury in liver.
     (3) Investigation of relationship between combination of K18 and ASPP2 and p53 apoptosis pathway: HepG2 cells were co-transfected with ASPP2 and K18 or S33A K18 or S52A K18. When cells were treated with drugs, ASPP2 entered into nucleus totally in apoptosis cells co-transfected with K18 S33A and ASPP2, but entered into nucleus much less in apoptosis cells co-transfected with K18 S52A and ASPP2 or K18 and ASPP2. This result indicated that K18 Ser33 phosphorylation had close relationship with ASPP2 nucleus translocation. Effect of K18 on ASPP2-p53 and its regulated Bax transcription activity were studied by report gene analysis. It was found that ASPP2 increased the activity of Bax-luc report gene but the activity decreased when excessive wild K18 was added, which indicated that K18 may inhibit the pro-apoptotic effect of ASPP2. When K18 was replaced with S33A K18, Bax report gene activity was almost the same with transfected with ASPP2 alone. This result again confirmed that K18 Ser33 phosphorylation could significantly enhance the combination of K18 and ASPP2, therefore indirectly inhibit ASPP2 binding with p53 and exert its apoptosis inhibition function.
     The studies above indicated: K18 and its phosphorylation played important role in hepatocytes apoptosis; under normal circumstances, K18 in cytoplasm combined with ASPP2 in a Ser33 phosphorylation-dependent manner. When cells were suffered with mild injury, K18 Ser33 and Ser52 phosphorylation both increased to enhance the binding of ASPP2 and K18, But when cells suffered from serious injury, K18 Ser33 phosphorylation decreased to make ASPP2 depart from K18 and enter into nucleus to exert its pro-apoptosis function.
引文
[1] Van Eyken P, Desmet VJ, Cytokeratins and the liver, Liver, 1993, 13(3):113-22.
    [2] Omary MB, Ku NO, Toivola DM, Keratins: guardians of the liver, Hepatology, 2002, 35(2): 251-257.
    [3] Ku NO, Wright TL, Terrault NA, Gish R, Omary MB, Mutation of Human Keratin 18 in Association with Cryptogenic Cirrhosis, J Clin Invest, 1997, 99(1): 19–23.
    [4] Zhong B, Strnad P, Selmi C, Invernizzi P, Tao GZ, Caleffi A, Chen M, Bianchi I, Podda M, Pietrangelo A, Gershwin ME, Omary MB, Keratin variants are overrepresented in primary biliary cirrhosis and associate with disease severity, Hepatology, 2009, 50(2):546-554.
    [5] Kumemura H, Harada M, Yanagimoto C, Koga H, Kawaguchi T, Hanada S, Taniguchi E, Ueno T, Sata M, Mutation in keratin 18 induces mitochondrial fragmentation in liver-derived epithelial cells, Biochem Biophys Res Commun, 2008, 29;367(1):33-40.
    [6] Strnad P, Tao GZ, Zhou Q, Harada M, Toivola DM, Brunt EM, Omary MB, Keratin mutation predisposes to mouse liver fibrosis and unmasks differential effects of the carbon tetrachloride and thioacetamide models, Gastroenterology, 2008, 134(4):1169-79.
    [7] Ku NO, Lim JK, Krams SM, Esquivel CO, Keeffe EB, Wright TL, Parry DA, Omary MB, Keratins as susceptibility genes for end-stage liver disease, Gastroenterology, 2005, 129(3):885-893.
    [8] Ku NO, Gish R, Wright TL, Omary MB, Keratin 8 mutations in patients with cryptogenic liver disease, N Engl J Med, 2001, 344(21): 1580–1587.
    [9] Ku NO, Darling JM, Krams SM, Esquivel CO, Keeffe EB, Sibley RK, Lee YM, Wright TL, Omary MB, Keratin 8 and 18 mutations are risk factors for developing liver disease of multiple etiologies, Proc Natl Acad Sci U S A, 2003, 100(10): 6063–6068
    [10] Lau SK, Prakash S, Geller SA, Alsabeh R, Comparative immunohistochemical profile of hepatocellular carcinoma, cholangiocarcinoma, and metastatic adenocarcinoma, Hum Pathol, 2002, 33(12): 1175–1181.
    [11] Feldstein AE, Wieckowska A, Lopez AR, Liu YC, Zein NN, McCullough AJ, Cytokeratin-18 fragment levels as noninvasive biomarkers for nonalcoholicsteatohepatitis: a multicenter validation study, Hepatology, 2009, 50(4):1072-1078.
    [12] Hofer S, Brenner T, Bopp C, Steppan J, Lichtenstern C, Weitz J, Bruckner T, Martin E, Hoffmann U, Weigand MA, Cell death serum biomarkers are early predictors for survival in severe septic patients with hepatic dysfunction, Crit Care, 2009, 13(3):R93.
    [13] Chou CF, Riopel CL, Rott LS, Omary MB, A significant soluble keratin fraction in‘simple’epithelial cells: Lack of an apparent phosphorylation and glycosylation role in keratin solubility, J Cell Sci, 1993, 105(Pt2):433-445
    [14] Caulin C, Ware CF, Magin TM, Oshima RG, Keratin-dependent, epithelial resistance to tumor necrosis factor-induced apoptosis, J Cell Biol, 2000, 149(1):17–22.
    [15] Gilbert S, Loranger A, Daigle N, Marceau N, Simple epithelium keratins 8 and 18 provide resistance to Fas-mediated apoptosis. The protection occurs through a receptor-targeting modulation, J Cell Biol, 2001, 154(4): 763–774.
    [16] Gilbert S, Loranger A, Marceau N, Keratins modulate c- Flip/extracellular signal-regulated kinase 1 and 2 antiapoptotic signaling in simple epithelial cells, Mol Cell Biol, 2004, 24(16):7072–7081 .
    [17] Inada H, Izawa I, Nishizawa M, Fujita E, Kiyono T, Takahashi T, Momoi T, Inagaki M, Keratin attenuates tumor necrosis factor-induced cytotoxicity through association with TRADD, J Cell Biol, 2001, 155(3):415-426.
    [18] Coulombe PA, Omary MB, Hard and soft principles defining the structure, function and regulation of keratin intermediate filaments, Curr Opin Cell Biol, 2002, 14(1): 110-122.
    [19] Caulín C, Salvesen GS, Oshima RG, Caspase cleavage of keratin 18 and reorganization of intermediate filaments during epithelial cell apoptosis J Cell Biol, 1997, 138(6):1379-1394.
    [20] Ku NO, Omary MB, Keratins turn over by ubiquitination in a phosphorylation-modulated fashion, J Cell Biol, 2000, 149(3):547-552.
    [21] Omary MB, Ku NO, Liao J, Price D, Keratin modifications and solubility properties in epithelial cells and in vitro, Subcell Biochem, 1998, 31:105-140.
    [22] Ku NO, Liao J, Omary MB, Phosphorylation of human keratin 18 serine 33 regulates binding to 14-3-3 proteins, EMBO J, 1998, 17(7): 1892-1906.
    [23] Ku NO, Michie S, Resurreccion EZ, Broome RL, Omary MB, Keratin binding to 14-3-3 proteins modulates keratin filaments and hepatocyte mitotic progression,Proc Natl Acad Sci U S A, 2002, 99(7) :4373– 4378.
    [24] Stumptner C, Omary MB, Fickert P, Denk H, Zatloukal K, Hepatocyte cytokeratins are hyperphosphorylated at multiple sites in human alcoholic hepatitis and in a mallory body mouse model, Am J Pathol, 2000, 156(1):77–90.
    [25] Zatloukal K, Stumptner C, Lehner M, Denk H, Baribault H, Eshkind LG, Franke WW, Cytokeratin 8 protects from hepatotoxicity, and its ratio to cytokeratin 18 determines the ability of hepatocytes to form Mallory bodies, Am J Pathol, 2000, 156(4):1263–1274.
    [26] Fickert P, Trauner M, Fuchsbichler A, Stumptner C, Zatloukal K, Denk H, Mallory body formation in primary biliary cirrhosis is associated with increased amounts and abnormal phosphorylation and ubiquitination of cytokeratins, J Hepatol, 2003, 38(4):387–394.
    [27] Toivola DM, Ku NO, Resurreccion EZ, Nelson DR, Wright TL, Omary MB, Keratin 8 and 18 Hyperphosphorylation Is a Marker of Progression of Human Liver Disease, Hepatology, 2004, 40(2):459–466.
    [28] Green DR, Kroemer G, Cytoplasmic functions of the tumour suppressor p53, Nature, 2009, 458(7242):1127-1130.
    [29] Kruse JP, Gu W, Modes of p53 regulation, Cell, 2009, 137(4):609-622.
    [30] Lassus P, Ferlin M, Piette J, Hibner U, Anti-apoptotic activity of low levels of wild-type p53, Embo J, 1996, 15(17): 4566-4573.
    [31] Vousden KH, Lu X, Live or let die : the cell’s response to P53 [J ], Nat Rev Cancer, 2002, 2(8) :594-604.
    [32] Ayed A, Mulder FA, Yi GS, Lu Y, Kay LE, Arrowsmith CH, Latent and active P53 are identical in conformation, Nat Struct Biol, 2001, 8(9): 730-732.
    [33] Vogelstein B, Lane D, Levine AJ, Surfing the P53 network [J ], Nature, 2000, 408(6810) :307-310.
    [34] Kampa KM, Bonin M, Lopez CD, New insights into the expanding complexity of the tumor suppressor ASPP2, Cell Cycle, 2009, 8(18):2871-2876.
    [35]中华医学会传染病与寄生虫病学分会,肝病学分会,病毒性肝炎防治方案,中华肝脏病杂志,2000, 8(6):324-329。
    [36] Ishak K, Baptista A, Bianchi L, Callea F, De Groote J, Gudat F, Denk H, Desmet V, Korb G, MacSween RN, Histological grading and staging of chronic hepatitis, J Hepatol, 1995, 22(6):696-699.
    [37] Moll R, Franke WW, Schiller DL, Geiger B, Krepler R, The catalog of humancytokeratins: patterns of expression in normal epithelia, tumors and cultured cells, Cell, 1982, 31(1): 11–24.
    [38] Ku NO, Zhou X, Toivola DM, Omary MB, The cytoskeleton of digestive epithelia in health and disease, Am J Physiol, 1999, 277(6 Pt 1):G1108-1137.
    [39] Franke WW, Denk H, Kalt R, Schmid E, Biochemical and immunological identification of cytokeratin proteins present in hepatocytes of mammalian liver tissue, Exp Cell Res, 1981, 131(2):299-318.
    [40] Fuchs E, Weber K, Intermediate filaments: structure, dynamics, function, and disease, Annu Rev Biochem, 1994, 63: 345–382.
    [41] Omary MB, Ku NO, Tao GZ, Toivola DM, Liao J, 'Heads and tails' of intermediate filament phosphorylation: multiple sites and functional insights, Trends Biochem Sci, 2006, 31(7): 383?394.
    [42] Ku NO, Liao J, Chou CF, Omary MB, Implications of intermediate filament protein phosphorylation, Cancer Metastasis Rev, 1996, 15(4): 429?444.
    [43] Zatloukal K, Stumptner C, Lehner M, Denk H, Baribault H, Eshkind LG, Franke WW, Cytokeratin 8 protects from hepatotoxicity, and its ratio to cytokeratin 18 determines the ability of hepatocytes to form Mallory bodies, Am J Pathol, 2000, 156(4): 1263–1274.
    [44] Stumptner C, Omary MB, Fickert P, Denk H, Zatloukal K, Hepatocyte cytokeratins are hyperphosphorylated at multiple sites in human alcoholic hepatitis and in a mallory body mouse model, Am J Pathol, 2000, 156(1): 77?90.
    [45] Liao J, Omary MB, 14-3-3 proteins Associate with Phosphorylated Simple Epithelial Keratins during Cell Cycle Progression and Act as a Solubility Cofactor, J Cell Biol, 1996, 133(2):345-357.
    [46] Ku NO, Michie SA, Soetikno RM, Resurreccion EZ, Broome RL, Omary MB, Mutation of a major keratin phosphorylation site predisposes to hepatotoxic injury in transgenic mice, J Cell Biol, 1998, 143(7):2023–2032.
    [47] Ku NO, Soetikno RM, Omary MB, Keratin mutation in transgenic mice predisposes to Fas but not TNF-induced apoptosis and massive liver injury, Hepatology, 2003, 37(5):1006-1014.
    [48] Leifeld L, Kothe S, S?hl G, Hesse M, Sauerbruch T, Magin TM, Spengler U, Keratin 18 provides resistance to Fas-mediated liver failure in mice, Eur J Clin Invest, 2009, 39(6):481-488.
    [49] Gilbert S, Ruel A, Loranger A, Marceau N, Switch in Fas-activated deathsignaling pathway as result of keratin 8/18-intermediate filament loss, Apoptosis, 2008, 13(12):1479–1493.
    [50] Herrmann H, Hesse M, Reichenzeller M, Aebi U, Magin TM, Functional complexity of intermediate filament cytoskeletons: from structure to assembly to gene ablation, Int Rev Cytol, 2003, 223: 83–175.
    [51] Lenhoff RJ, Summers J, Construction of avian hepadnavirus variants with enhanced replication and cytopathicity in primary hepatocytes, J Virol, 1994, 68(9): 5706-5713.
    [52] Bruss V, Revisiting the cytopathic effect of hepatitis B virus infection, Hepatology, 2002, 36(6): 1327-1329,
    [53] Foo NC, Ahn BY, Ma X, Hyun W, Yen TS, Cellular vacuolization and apoptosis induced by hepatitis B virus large surface protein, Hepatology, 2002, 36(6): 1400-1407.
    [54] Su F, Theodosis CN, Schneider RJ, Role of NF-kappaB and myc proteins in apoptosis induced by hepatitis B virus HBx protein, J Virol, 2001, 75(1): 215-225.
    [55] Kim KH, Seong BL, Pro-apoptotic function of HBV X protein is mediated by interaction with c-FLIP and enhancement of death-inducing signal, EMBO J, 2003, 22(9): 2104-2116.
    [56] Chirillo P, Pagano S, Natoli G, Puri PL, Burgio VL, Balsano C, Levrero M, The hepatitis B virus X gene induces p53-mediated programmed cell death, Proc Natl Acad Sci USA, 1997, 94(15): 8162-8167.
    [57] Song le H, Binh VQ, Duy DN, Bock TC, Kremsner PG, Luty AJ, Mavoungou E, Variations in the serum concentrations of soluble Fas and soluble Fas ligand in Vietnamese patients infected with hepatitis B virus, J Med Virol, 2004, 73(2):244–249.
    [58] Fang JW, Shen WW, Meager A, Lau JY, Activation of the tumor necrosis factor-alpha system in the liver in chronic hepatitis B virus infection, Am J Gastroenterol, 1996, 91(4):748–753.
    [59] Qu J, Lin J, Zhang S, Zhu Z, Ni C, Zhang S, Gao H, Zhu M, HBV DNA can bind to P53 protein and influence p53 transactivation in hepatoma cells, Biochem Biophys Res Commun, 2009, 386 (3):504–509.
    [60] Zhao J, Wu G, Bu F, Lu B, Liang A, Cao L, Tong X, Lu X, Wu M, Guo Y, Epigenetic silence of ankyrin-repeat-containing, SH3-domain-containing, and proline-rich-region- containing protein 1 (ASPP1) and ASPP2 genes promotes tumorgrowth in hepatitis B virus-positive hepatocellular carcinoma, Hepatology, 2010, 51(1):142-153.
    [61] Bargonetti J, Manfredi JJ, Multiple roles of the tumor suppressor p53, Curr Opin Oncol, 2002, 14 (1) :86-91.
    [62] Dewantoro O, Gani RA, Akbar N, Hepatocarcinogenesis in viral Hepatitis B infection: the role of HBx and p53, Acta Med Indones, 2006, 38(3):154-159.
    [63] Fang L, Li G, Liu G, Lee SW, Aaronson SA, p53 induction of heparin-binding EGF-like growth factor counteracts p53 growth suppression through activation of MAPK and PI3K/Akt signaling cascades, EMBO J, 2001, 20(8):1931-1939.
    [64] Bensaad K, Tsuruta A, Selak MA, Vidal MN, Nakano K, Bartrons R, Gottlieb E, Vousden KH, TIGAR, a p53-inducible regulator of glycolysis and apoptosis, Cell, 2006, 126(1):107-120.
    [65] Mori T, Okamoto H, Takahashi N, Ueda R, Okamoto T, Aberrant overexpression of 53BP2 mRNA in lung cancer cell lines, FEBS Lett, 2000, 465 (2-3):124–128.
    [66] Sullivan A, Lu X, ASPP: a new family of oncogenes and tumour suppressor genes, Br J Cancer, 2007, 96(2):196-200.
    [67] Ao Y, Rohde LH, Naumovski L, p53-interacting protein 53BP2 inhibits clonogenic survival and sensitizes cells to doxorubicin but not paclitaxel-induced apoptosis, Oncogene, 2001, 20(21): 2720-2725.
    [68] Iwabuchi K, Li B, Massa HF, Trask BJ, Date T, Fields S, Stimulation of p53-mediated transcriptional activation by the p53 binding protein, 53BP1 and 53BP2, J Biol Chem, 1998, 273(40):26061-26068.
    [69] Samuels-Lev Y, O'Connor DJ, Bergamaschi D, Trigiante G, Hsieh JK, Zhong S, Campargue I, Naumovski L, Crook T, Lu X, ASPP proteins specifically stimulate the apoptotic function of p53, Mol Cell, 2001. 8(4):781-794.
    [70] Ahn J, Byeon IJ, Byeon CH, Gronenborn AM, Insight into the structural basis of pro- and antiapoptotic p53 modulation by ASPP proteins, J Biol Chem, 2009, 284(20):13812-13822.
    [71] Lassus P, Ferlin M, Piette J, Hibner U, Anti-apoptotic activity of low levels of wild-type P53, EMBO J, 1996, 15(17): 4566-4573.
    [72] Kim S, Wong P, Coulombe PA, A keratin cytoskeletal protein regulates protein synthesis and epithelial cell growth, Nature, 2006, 441(7091):362-365.
    [73] Izawa I, Nishizawa M, Ohtakara K, Ohtsuka K, Inada H, Inagaki M,Identification of Mrj, a DnaJ/Hsp40 family protein, as a keratin 8/18 filament regulatory protein, J Biol Chem, 2000, 275(44):34521–34527.
    [74] Inada H, Izawa I, Nishizawa M, Fujita E, Kiyono T, Takahashi T, Momoi T, Inagaki M, Keratin attenuates tumor necrosis factor-induced cytotoxicity through association with TRADD, J Cell Biol, 2001, 155(3):415–426.
    [75] Naumovski L and Cleary ML, The P53-binding protein 53BP2 also interacts with Bc12 and impedes cell cycle progression at G2/M, Mol Cell Biol, 1996, 16(7): 3884-3892.
    [76] Tidow H, Andreeva A, Rutherford TJ, Fersht AR, Solution structure of ASPP2 N-terminal domain (N-ASPP2) reveals a ubiquitin-like fold, J Mol Biol, 2007, 371 (4): 948–958.
    [77] Robinson RA, Lu X, Jones EY, Siebold C, Biochemical and structural studies of ASPP proteins reveal differential binding to p53, p63, and p73, Structure, 2008, 16(2):259-268.
    [78] Gorina S, Pavletich NP, Structure of the p53 tumor suppressor bound to the ankyrin and SH3 domains of 53BP2, Science, 1996, 274 (5289):1001–1005.
    [79] Naumovski L, Cleary ML, The p53-binding protein 53BP2 also interacts with Bc12 and impedes cell cycle progression at G2/M, Mol Cell Biol, 1996, 7 (16): 3884–3892.
    [80] Helps NR, Barker HM, Elledge SJ, Cohen PT, Protein phosphatase 1 interacts with p53BP2, a protein which binds to the tumour suppressor p53, FEBS Lett, 1995, 377(3):295-300.
    [81] Cao Y, Hamada T, Matsui T, Date T, Iwabuchi K, Hepatitis C virus core protein interacts with p53-binding protein, 53BP2/Bbp/ASPP2, and inhibits p53-mediated apoptosis, Biochem Biophys Res Commun, 2004, 315(4):788-795.
    [82] Lassus P, Ferlin M, Piette J, Hibner U, Anti-apoptotic activity of low levels of wild-type P53, EMBO J, 1996, 15(17): 4566-4573.
    [83] Sachdev S, Hoffmann A, Hannink M, Nuclear localization of IkappaB alpha is mediated by the second ankyrin repeat: the IkappaB alpha ankyrin repeats define a novel class of cis-acting nuclear import sequences, Mol Cell Biol, 1998, 18(5):2524-2534.
    [84] Rotem S, Katz C, Benyamini H, Lebendiker M, Veprintsev D, Rüdiger S, Danieli T, Friedler A, The structure and interactions of the proline-rich domain of ASPP2, J Biol Chem, 2008, 283(27):18990-18999.
    [1] Moll R, Franke WW, Schiller DL, Geiger B, Krepler R, The catalog of human cytokeratins: patterns of expression in normal epithelia, tumors and cultured cells, Cell, 1982, 31(1): 11–24.
    [2] Herrmann H, Hesse M, Reichenzeller M, Aebi U, Magin TM, Functional complexity of intermediate filament cytoskeletons: from structure to assembly to gene ablation, Int Rev Cytol, 2003, 223:83-175.
    [3] Schweizer J, Bowden PE, Coulombe PA, Langbein L, Lane EB, Magin TM, Maltais L, Omary MB, Parry DA, Rogers MA, Wright MW, New consensus nomenclature for mammalian keratins, J Cell Biol, 2006, 174(2):169-174.
    [4] Coulombe PA, Wong P, Cytoplasmic intermediate filaments revealed as dynamic and multipurpose scaffolds, Nat Cell Biol, 2004, 6(8):699-706.
    [5] Omary MB, Ku NO, Tao GZ, Toivola DM, Liao J, 'Heads and tails' of intermediate filament phosphorylation: multiple sites and functional insights, Trends Biochem Sci, 2006, 31(7): 383?394.
    [6] Omary MB, Ku NO, Toivola DM, Keratins: guardians of the liver, Hepatology, 2002, 35(2): 251-257.
    [7] Pallari HM, Eriksson JE, Intermediate filaments as signaling platforms, Sci STKE, 2006, 19(366):pe53.
    [8] Maddox P, Sasieni P, Szarewski A, Anderson M, Hanby A, Differential expression of keratins 10, 17, and 19 in normal cervical epithelium, cervical intraepithelial neoplasia, and cervical carcinoma, J Clin Pathol, 1999, 52(1):41-46.
    [9] Steinert PM, Idler WW, Zhou XM, Structural and functional implication of amino acid sequences of keratin intermediate filament sub unit, Ann NY Acad Sci, 1985, 455:451-461.
    [10] Mclachlan AD, Coiled coil formation and sequence regularities in the helical region of alpa-keratin, J Mol Biol, 1978, 124(1):297-304.
    [11] Fuchs E, Weber K, Intermediate filaments: structure, dynamics, function, and disease, Annu Rev Biochem, 1994, 63:345-382.
    [12] Caulín C, Salvesen GS, Oshima RG, Caspase cleavage of keratin 18 and reorganization of intermediate filaments during epithelial cell apoptosis, J Cell Biol, 1997, 138(6):1379-1394.
    [13] Kumemura H, Harada M, Yanagimoto C, Koga H, Kawaguchi T, Hanada S, Taniguchi E, Ueno T, Sata M, Mutation in keratin 18 induces mitochondrialfragmentation in liver-derived epithelial cells, Biochem Biophys Res Commun, 2008, 367(1):33-40.
    [14] Kulesh DA, Cece?a G, Darmon YM, Vasseur M, Oshima RG, Posttranslational regulation of keratins: degradation of mouse and human keratins 18 and 8, Mol Cell Biol, 1989, 9(4):1553-1565.
    [15] Baribault H, Price J, Miyai K, Oshima RG, Mid-gestational lethality in mice lacking keratin 8, Genes Dev, 1993, 7(7A):1191-1202.
    [16] Magin TM, Schr?der R, Leitgeb S, Wanninger F, Zatloukal K, Grund C, Melton DW, Lessons from keratin 18 knockout mice: formation of novel keratin filaments, secondary loss of keratin 7 and accumulation of liver-specific keratin 8-positive aggregates, J Cell Biol, 1998, 140(6):1441-1451.
    [17] Tamai Y, Ishikawa T, B?sl MR, Mori M, Nozaki M, Baribault H, Oshima RG, Taketo MM, Cytokeratins 8 and 19 in the mouse placental development, J Cell Biol, 2000, 151(3):563-572.
    [18] Hesse M, Franz T, Tamai Y, Taketo MM, Magin TM, Targeted deletion of keratins 18 and 19 leads to trophoblast fragility and early embryonic lethality, EMBO J, 2000, 19(19):5060-5070.
    [19] Ku NO, Michie S, Oshima RG, Omary MB, Chronic hepatitis, hepatocyte fragility, and increased soluble phosphoglycokeratins in transgenic mice expressing a keratin 18 conserved arginine mutant, J Cell Biol, 1995, 131(5):1303-1314.
    [20] Ku NO, Michie SA, Soetikno RM, Resurreccion EZ, Broome RL, Oshima RG, Omary MB, Susceptibility to hepatotoxicity in transgenic mice that express a dominant-negative human keratin 18 mutant, J Clin Invest, 1996, 98(4):1034-1046.
    [21] Ku NO, Wright TL, Terrault NA, Gish R, Omary MB, Mutation of human keratin 18 in association with cryptogenic cirrhosis, J Clin Invest, 1997. 99(1):19-23.
    [22] Ku NO, Gish R, Wright TL, Omary MB, Keratin 8 mutations in patients with cryptogenic liver disease, N Engl J Med, 2001, 344(21):1580-1587.
    [23] Zhong B, Strnad P, Selmi C, Invernizzi P, Tao GZ, Caleffi A, Chen M, Bianchi I, Podda M, Pietrangelo A, Gershwin ME, Omary MB, Keratin variants are overrepresented in primary biliary cirrhosis and associate with disease severity, Hepatology, 2009, 50(2):546-554.
    [24] Ku NO, Lim JK, Krams SM, Esquivel CO, Keeffe EB, Wright TL, Parry DA, Omary MB, Keratins as susceptibility genes for end-stage liver disease, Gastroenterology, 2005, 129(3):885-893.
    [25] Ku NO, Darling JM, Krams SM, Esquivel CO, Keeffe EB, Sibley RK, Lee YM, Wright TL, Omary MB, Keratin 8 and 18 mutations are risk factors for developing liver disease of multiple etiologies, Proc Natl Acad Sci U S A, 2003, 100(10):6063-6068.
    [26] Malhi H, Gores GJ, Cellular and molecular mechanisms of liver injury, Gastroenterology, 2008, 134(6):1641-1654.
    [27] Ku NO, Liao J, Omary MB, Apoptosis generates stable fragments of human type I keratins, J Biol Chem, 1997, 272(52):33197-33203.
    [28] Leers MP, Bj?rklund V, Bj?rklund B, J?rnvall H, Nap M, An immunohistochemical study of the clearance of apoptotic cellular fragments, Cell Mol Life Sci, 2002, 59(8):1358-1365.
    [29] Feldstein AE, Wieckowska A, Lopez AR, Liu YC, Zein NN, McCullough AJ, Cytokeratin-18 fragment levels as noninvasive biomarkers for nonalcoholic steatohepatitis: a multicenter validation study, Hepatology, 2009, 50(4):1072-1078.
    [30] Yilmaz Y, Kedrah AE, Ozdogan O, Cytokeratin-18 fragments and biomarkers of the metabolic syndrome in nonalcoholic steatohepatitis, World J Gastroenterol, 2009, 15(35):4387-4391.
    [31] Tsutsui M, Tanaka N, Kawakubo M, Sheena Y, Horiuchi A, Komatsu M, Nagaya T, Joshita S, Umemura T, Ichijo T, Matsumoto A, Yoshizawa K, Aoyama T, Tanaka E, Sano K, Serum Fragmented Cytokeratin 18 Levels Reflect the Histologic Activity Score of Nonalcoholic Fatty Liver Disease More Accurately Than Serum Alanine Aminotransferase Levels, J Clin Gastroenterol, 2010, Jan 25. [Epub ahead of print]
    [32] Rupa JD, de Bru?ne AP, Gerbers AJ, Leers MP, Nap M, Kessels AG, Schutte B, Arends JW, Simultaneous detection of apoptosis and proliferation in colorectal carcinoma by multiparameter flow cytometry allows separation of high and low-turnover tumors with distinct clinical outcome, Cancer, 2003, 97(10):2404-2411.
    [33] Ludwig J, Viggiano TR, McGill DB, Oh BJ, Nonalcoholic steatohepatitis: Mayo Clinic experiences with a hitherto unnamed disease, Mayo Clin Proc, 1980, 55(7):434-438.
    [34] Baptista A, Bianchi L, Groote J, Alcoholic liver disease: morphological manifestations. Review by an international group, Lancet, 1981, 1(8222):707-711.
    [35] Burt AD, Mutton A, Day CP, Diagnosis and interpretation of steatosis and steatohepatitis, Semin Diagn Pathol, 1998, 15(4):246-258.
    [36] Hall P, Pathological spectrum of alcoholic liver disease. In Pathology andPathogenesis: Alcoholic Liver Disease, Hall P (ed), Edward Arnold, 1995 34:41–68.
    [37] James OW, Day CP, Non-alcoholic steatohepatitis (NASH): a disease of emerging identity and importance, J Hepatol, 1998, 29(3):495-501.
    [38] Jansen PL, Nonalcoholic steatohepatitis, Neth J Med, 2004, 62(7):217-224.
    [39] Stumptner C, Omary MB, Fickert P, Denk H, Zatloukal K, Hepatocyte cytokeratins are hyperphosphorylated at multiple sites in human alcoholic hepatitis and in a mallory body mouse model, Am J Pathol, 2000, 156(1):77-90.
    [40] Hazan R, Denk H, Franke WW, Lackinger E, Schiller DL, Change of cytokeratin organization during development of Mallory bodies as revealed by a monoclonal antibody, Lab Invest, 1986, 54(5):543-553.
    [41] Zatloukal K, Fesus L, Denk H, Tarcsa E, Spurej G, B?ck G, High amount of epsilon-(gamma-glutamyl)lysine cross-links in Mallory bodies, Lab Invest, 1992, 66(6):774-777.
    [42] Fickert P, Trauner M, Fuchsbichler A, Stumptner C, Zatloukal K, Denk H, Mallory body formation in primary biliary cirrhosis is associated with increased amounts and abnormal phosphorylation and ubiquitination of cytokeratins, J Hepatol, 2003, 38(4):387-394.
    [43] Fickert P, Trauner M, Fuchsbichler A, Stumptner C, Zatloukal K, Denk H, Cytokeratins as targets for bile acid-induced toxicity, Am J Pathol, 2002, 160(2):491-499.
    [44] Lau SK, Prakash S, Geller SA, Alsabeh R, Comparative immunohistochemical profile of hepatocellular carcinoma, cholangiocarcinoma, and metastatic adenocarcinoma, Hum Pathol, 2002, 33(12):1175-1181.
    [45] Caulin C, Ware CF, Magin TM, Oshima RG, Keratin-dependent, epithelial resistance to tumor necrosis factor-induced apoptosis, J Cell Biol, 2000, 149(1):17-22.
    [46] Gilbert S, Loranger A, Daigle N, Marceau N, Simple epithelium keratins 8 and 18 provide resistance to Fas-mediated apoptosis. The protection occurs through a receptor-targeting modulation, J Cell Biol, 2001, 154(4):763-773.
    [47] Gilbert S, Loranger A, Marceau N, Keratins modulate c-Flip/extracellular signal-regulated kinase 1 and 2 antiapoptotic signaling in simple epithelial cells, Mol Cell Biol, 2004, 24(16):7072-7081.
    [48] Inada H, Izawa I, Nishizawa M, Fujita E, Kiyono T, Takahashi T, Momoi T, Inagaki M, Keratin attenuates tumor necrosis factor-induced cytotoxicity through association with TRADD, J Cell Biol, 2001, 155(3):415-426.
    [49] Gilbert S, Ruel A, Loranger A, Marceau N, Switch in Fas-activated death signaling pathway as result of keratin 8/18-intermediate filament loss, Apoptosis, 2008, 13(12):1479-1493.
    [50] Tzivion G, Gupta VS, Kaplun L, Balan V, 14-3-3 proteins as potential oncogenes, Semin Cancer Biol, 2006, 16(3):203-213.
    [51] Coulombe PA, Omary MB, 'Hard' and 'soft' principles defining the structure, function and regulation of keratin intermediate filaments, Curr Opin Cell Biol, 2002, 14(1):110-122.
    [52] Omary MB, Ku NO, Liao J, Price D, Keratin modifications and solubility properties in epithelial cells and in vitro, Subcell Biochem, 1998, 31:105-140.
    [53] Ku NO, Liao J, Omary MB, Phosphorylation of human keratin 18 serine 33 regulates binding to 14-3-3 proteins, EMBO J, 1998, 17(7):1892-1906.
    [54] Ku NO, Michie S, Resurreccion EZ, Broome RL, Omary MB, Keratin binding to 14-3-3 proteins modulates keratin filaments and hepatocyte mitotic progression, Proc Natl Acad Sci U S A, 2002, 99(7):4373-4378.
    [55] Ku NO, Azhar S, Omary MB, Keratin 8 phosphorylation by p38 kinase regulates cellular keratin filament reorganization: modulation by a keratin 1-like disease-causing mutation, J Biol Chem, 2002, 277(13):10775-10782.
    [56] Ku NO, Omary MB, Keratins Turn Over by Ubiquitination in a Phosphorylation-modulated Fashion, J Cell Biol, 2000, 149(3):547-552.
    [57] Toivola DM, Ku NO, Resurreccion EZ, Nelson DR, Wright TL, Omary MB, Keratin 8 and 18 hyperphosphorylation is a marker of progression of human liver disease, Hepatology, 2004, 40(2):459-466.
    [58] Ichiro I ,Masaki I, Regulatory mechanisms and functions of intermediate filaments: A study using site- and phosphorylation state-specific antibodies, Cancer Sci, 2006, 97(3):167-174.
    [59] Nelson DR, Lauwers GY, Lau JY, Davis GL, Interleukin 10 treatment reduces fibrosis in patients with chronic hepatitis C: a pilot trial of interferon nonresponders, Gastroenterology, 2000, 118(4):655-660.
    [60] Ku NO, Omary MB, Identification of the major physiologic phosphorylation site of human cytokeratin 18: potential kinases and a role in filament reorganization, J Cell Biol, 1994, 127(1):161-171.
    [61] Ku NO, Liao J, Omary MB, Phosphorylation of human keratin 18 serine 33 regulates binding to 14–3-3 proteins, EMBO J, 1998, 17(7):1892-1906.
    [62] Tsunoo C, Harwood TR, Arak S, Yokoo H, Cytoskeletal alterations leading to Mallory body formation in livers of mice fed 3,5-diethoxycarbonyl-1,4- dihydrocollidine, J Hepatol, 1987, 5(1):85-97.
    [63] Shi Y, Sun S, Liu Y, Li J, Zhang T, Wu H, Chen X, Chen D, Zhou Y, Keratin 18 phosphorylation as a progression marker of chronic hepatitis B, Virol J, 2010, 7(1):70. [Epub ahead of print]
    [64] Hoek JB, Thomas AP, Rooney TA, Higashi K, Rubin E, Ethanol and signal transduction in the liver, FASEB J, 1992, 6(7):2386-2396.
    [65] Chen J, Ishac EJ, Dent P, Kunos G, Gao B, Effects of ethanol on mitogen-activated protein kinase and stress-activated protein kinase cascades in normal and regenerating liver, Biochem J, 1998, 334 ( Pt 3):669-676.
    [66] Zatloukal K, Stumptner C, Fuchsbichler A, Fickert P, Lackner C, Trauner M, Denk H, The keratin cytoskeleton in liver diseases, J Pathol, 2004, 204(4):367-376.
    [67] Zatloukal K, Stumptner C, Lehner M, Denk H, Baribault H, Eshkind LG, Franke WW, Cytokeratin 8 protects from hepatotoxicity, and its ratio to cytokeratin 18 determines the ability of hepatocytes to form Mallory bodies, Am J Pathol, 2000, 156(4):1263-1274.
    [68] Magin TM, Schr?der R, Leitgeb S, Wanninger F, Zatloukal K, Grund C, Melton DW, Lessons from keratin 18 knockout mice: formation of novel keratin filaments, secondary loss of keratin 7 and accumulation of liver-specific keratin 8-positive aggregates, J Cell Biol, 1998, 140(6):1441-1451.
    [69] Parry DA, Marekov LN, Steinert PM, Subfilamentous protofibril structures in fibrous proteins: cross-linking evidence for protofibrils in intermediate filaments, J Biol Chem, 2001, 276(42):39253-39258.
    [70] Cadrin M, Anderson NM, Aasheim LH, Kawahara H, Franks DJ, French SW, Modifications in cytokeratin and actin in cultured liver cells derived from griseofulvin-fed mice, Lab Invest, 1995, 72(4):453-460.
    [71] Fickert P, Trauner M, Fuchsbichler A, Stumptner C, Zatloukal K, Denk H, Mallory body formation in primary biliary cirrhosis is associated with increased amounts and abnormal phosphorylation and ubiquitination of cytokeratins, J Hepatol, 2003, 38(4):387-394.
    [72] Fausther M, Villeneuve L, Cadrin M, Heat shock protein 70 expression, keratin phosphorylation and Mallory body formation in hepatocytes from griseofulvin-intoxicated mice, Comp Hepatol, 2004, 3(1):5.
    [73] Harada M, Strnad P, Resurreccion EZ, Ku NO, Omary MB, Keratin 18 overexpression but not phosphorylation or filament organization blocks mouse Mallory body formation, Hepatology, 2007, 45(1):88-96.

© 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