Cdc42相关蛋白4对转化生长因子β1诱导肾小管上皮细胞—间充质转分化的影响和机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
慢性肾脏病(chronic kidney disease, CKD)不可逆的最终结局是终末期肾功能衰竭(end stage renal disease, ESRD),各种进行性慢性肾脏病发展为终末期肾功能衰竭的共同的病理特征是肾小管间质纤维化(tubular interstitial fibrosis,TIF),其对慢性肾脏病的预防和治疗与肾小球硬化相比更为重要。在肾脏疾病进展的过程中,肾间质纤维化程度与肾功能的减退密切相关。其中,肾小管上皮细胞-间充质细胞转分化(Epithelial to mesenchymal transition, EMT)是肾小管间质纤维化过程中的关键事件。EMT的完成需要数天时间,经过一系列复杂的过程,主要包括以下四个经典的步骤:早期细胞间粘附功能的丧失,a-SMA开始表达和肌动蛋白重组,肾小管基底膜的破坏和细胞转移和侵袭力增加。TGF-β1是参与EMT发生、发展的重要因子,TGF-β1可通过诱导EMT参与肾间质纤维化的进展,研究发现其信号传导需要多种介质的参与,包括Smads和非Smads途径,后者主要有PI3K/Akt、MAPK、ERK、p38MAPK、JNK、MEK、PKA、PKC和RhoGTPase等,而RhoGTPase分子机制研究较少。Cdc42相关蛋白4(Cdc42 interacting protein-4,CIP4)是属于RhoGTPase蛋白,含Bin/amphiphysin/Rvs (BAR)结构域蛋白超家族,主要调节肌动蛋白的聚合和细胞膜的动力学、稳定张力纤维丝,调节细胞内胞吞和胞吐等胞内、外物质的转运,故在细胞骨架的构建、细胞黏附、细胞内运输、信号转导、细胞周期等多种生物学方面发挥重要作用。过量表达的CIP4会引起细胞黏附功能的丧失、细胞转化、癌细胞的浸袭和转移。由于纤维化疾病与肿瘤一样都存在细胞转分化过程,因此我们推测在肾间质纤维化过程中CIP4高表达可能促进了肾小管EMT,进而加速了慢性纤维化的发展。本课题探讨了CIP4在TGF-β1诱导的肾小管EMT中的作用及其机制:通过构建5/6肾大部切除和TGF-β1诱导的肾小管上皮细胞-间充质转分化模型,拟构建高表达CIP4质粒稳定转染至肾肾小管上皮细胞,设计CIP4-siRNA,敲除CIP4基因,研究CIP4在慢性肾脏病肾小管间质纤维化中的表达及作用,以及CIP4与极性蛋白Par6的关系。
     第一部分Cdc42相关蛋白4(CIP4)与肾间质纤维化之间的关系
     目的观察5/6肾大部切除大鼠肾脏中和TGF-β1诱导的人肾小管上皮细胞(HK-2)Cdc42相关蛋白4(CIP4)的表达,并探讨内源性CIP4表达与肾脏间质纤维化的关系,为研究CIP4在肾小管上皮细胞-间充质转分化(EMT)过程中的作用提供基础。
     方法选择雄性SD大鼠20只(含正常组10只),采用5/6肾大部切除建立慢性肾脏病肾间质纤维化模型,按常规把SD大鼠饲养12周后,用自动生化仪检测大鼠血清尿素氮(BUN)、肌酐(Scr),Masson染色检测各组SD大鼠肾小球硬化和小管间质纤维化的程度,应用免疫组化SABC法检测CIP4在各组大鼠肾脏组织的分布变化,蛋白质免疫印迹法测定CIP4蛋白在肾脏组织中表达水平。用RT-PCR和Western blot法检测TGF-β1刺激的HK-2细胞12-72h后,各组细胞内CIP4的mRNA和蛋白表达的变化。
     结果CIP4蛋白在假手术组表达较少,仅分布于肾小管,而5/6肾大部切除组CIP4表达明显增多,几乎所有小管细胞均有表达。RT-PCR和Westernblot示,肾衰组CIP4蛋白的表达较假手术组明显增加(P<0.05)。体外实验CIP4在TGF-β1刺激的HK-2细胞内表达上调,24h时升高有显著意义(P<0.05),一直到72h仍在升高。
     结论通过体内和体外实验研究发现:CIP4在肾间质纤维化中表达增高,TGF-β1可能是其增高的原因。
     第二部分Cdc42相关蛋白4对TGF-β1诱导的肾小管上皮细胞-间充质转分化的影响
     目的构建CIP4稳定转染的HK-2细胞系,观察CIP4对HK-2细胞上皮细胞-间充质转分化(EMT)的影响,设计一条特异性沉默CIP4表达的RNA片段(siRNA-CIP4),将其转染TGF-β1刺激的HK-2细胞,观察CIP4对TGF-β1刺激的HK-2细胞上皮细胞-间充质转分化(EMT)的影响,并探讨PI3K/Akt信号通路在CIP4产生中的作用。
     方法根据GenBank里人CIP4 cDNA序列,设计一对含Not I酶切位点的特异引物,从人肾脏组织中通过RT-PCR技术得到的全长CIP4-cDNA,利用Not I酶切位点将全长CIP4-cDNA克隆到pcDNA3.1载体上,经过限制性酶切、生物公司测序、鉴定正确后,将pcDNA3.1/CIP4稳定转染至HK-2细胞。蛋白质免疫印迹法检测转染细胞的CIP4、E-cadherin和a-SMA的蛋白表达变化,共聚焦显微镜检测各组细胞内E-cadherin和a-SMA蛋白分布的改变。根据GenBank里人CIP4 cDNA序列,设计一条CIP4-siRNA片段,特异性沉默CIP4基因,然后将其用Lipofactamine2000转染TGF-β1刺激的HK-2细胞,Western blot检测各组细胞内CIP4、E-cadherin和a-SMA蛋白的表达变化,共聚焦显微镜检测E-cadherin和a-SMA蛋白的分布,通过P13K/Akt特异性抑制剂’Wortmannin 1μM干预TGF-β1刺激的HK-2细胞48h, Western blot检测各组细胞内CIP4表达的变化。
     结果肾小管上皮细胞经CIP4高表达的质粒转染后,上皮细胞形态明显发生改变,长梭形细胞替代了类圆形的肾小管上皮细胞,其细胞标志物E-cadherin蛋白表达减少,肌成纤维细胞标志a-SMA的表达增多(P<0.05), CIP4-siRNA转染TGF-β1刺激的HK-2细胞,上述相同的细胞形态改变出现逆转,细胞逐渐恢复成类圆形正常小管细胞,与TGF-β1刺激组相比,E-cadherin蛋白表达明显增多,a-SMA蛋白表达显著减少(P<0.05)。CIP4基因敲除逆转了TGF-β1诱导的肾小管EMT。以上结果证实CIP4能够促进肾小管上皮细胞—间充质细胞转分化。Wortmannin抑制TGF-β1刺激的HK-2细胞48小时后,CIP4蛋白表达显著减少。
     结论TGF-β1通过PI3K/Akt途径产生大量的CIP4,CIP4进一步参与TGF-β1诱导的肾小管上皮细胞EMT。
     第三部分Cdc42相关蛋白4对TGF-β1诱导的大鼠肾小管上皮细胞-间充质转分化的影响及其与极性蛋白Par6关系
     目的观察CIP4对TGF-β1诱导的大鼠肾小管上皮细胞-间充质转分化(EMT)的影响,并探讨极性蛋白Par6在TGF-β1诱导的肾小管EMT中的作用。
     方法TGF-β1 20μg/L作用72h诱导大鼠肾小管上皮细胞(NRK-52E)转分化,用Western blot法检测细胞内CIP4、E-cadherin和α-SMA的蛋白表达及Par6蛋白及其磷酸化水平,免疫共沉淀检测CIP4与Par6蛋白的结合状况,免疫荧光共聚焦检测Par6、E-cadherin和α-SMA的分布。根据Genbank里大鼠Par6的完全cDNA序列,设计一条Par6-siRNA片段,特异性沉默Par6的表达,通过Lipofactamine2000将Par6-siRNA转染至TGF-β1刺激的NRK-52E细胞,检测正常细胞、TGF-β1刺激细胞、par6-siRNA转染的TGF-β1刺激细胞内CIP4、E-cadherin和α-SMA的蛋白改变;同时设计根据Genbank里大鼠CIP4的完全cDNA序列,设计CIP4-siRNA,利用Lipofactamine2000(?)将其转染至TGF-β1刺激的NRK-52E细胞,检测正常细胞、TGF-β1刺激细胞、CIP4-siRNA转染的TGF-β1刺激细胞内Par6、P-Par6、E-cadherin和α-SMA的蛋白改变;免疫荧光共聚焦检测E-cadherin和α-SMA的分布。
     结果TGF-β1刺激72h后NRK-52E细胞上皮细胞标志E-cadherin(?)减少,并出现肌成纤维细胞标志α-SMA,提示TGF-β1 20μg/L能够促进肾小管上皮细胞向间充质转分化。随着NRK-52E发生EMT, CIP4蛋白表达增高,CIP4与Par6结合增强,Par6磷酸化水平显著增强。Par6-siRNA、CIP4-siRNA分别转染TGF-β1刺激细胞,磷酸化Par6均减少,细胞内E-cadherin蛋白表达增加,恢复到NRK-52E细胞膜和细胞连接处,α-SMA相应减少,Par6-siRNA和CIP4-siRNA转染逆转了TGF-β1诱导的NRK-52E细胞EMT。
     结论Cdc42相关蛋白4(CIP4)在TGF-β1诱导的肾小管EMT细胞内表达增高,通过极性蛋白Par6,促进了TGF-β1诱导大鼠肾小管上皮细胞-间充质细胞转分化。
Tubular interstitial fibrosis(TIF) is a common pathological characteristics and an inevitable outcome of all kinds of chronic kidney disease (CKD) progress to end-stage renal failure. Tubular epithelial-mesenchymal transition (TEMT) contributes significantly to the onset and pathogenesis of renal fibrosis. This phenotype has been shown to a complicated multistep process instead of simply a morphological transition, which consists of four key steps:loss of epithelial cell adhesion, de novo a-SMA expression and actin reorganization, disruption of tubular basement membrane, and enhanced cell migration and invasion, transforming growth factor-β1(TGF-β1), as a sole one, initiates and completes the entire course of EMT. TGF-β1 mediates the EMT process via intracellular pathways, including the canonical Smad pathway, phosphatidylinositol-3-kinase/Akt (PI3K/AKT) pathway, mitogen-activted protein kinase (MAPK) pathway, integrinlinked kinases(ERK) pathway, p38MAPK pathway, c-Jun N-terminal kinases (JNK) pathway and Rho-like GTPase signaling pathway. While the Smad, MAPK and PI3K/AKT pathways have been well described, less is known about Rho GTPase signaling molecular mechanism.
     Cdc42 interacting protein-4 (CIP4) is a small Rho GTPase, belongs to the Bin/ amphiphysin/Rvs (BAR) domain protein superfamily, which involved in membrane remodeling in various cellular pathways ranging from endocytosis and cytokinesis, T-tubule formation to cell migration and neuromorphogenesis. Thus, CIP4 plays important roles in the many aspects of biological fuctions such as the construction of cytoskeleton, cell adhesion, intracellular transportation, signal transduction and cell cycles. In human renal tumor cells, large amount of CIP4 splicing variant has been produced to promote the (3-catenin tyrosine phosphorylation and lead to renal tumor cells metastasis. The activated CIP4 can also induce cellular transformation and cell-cell interactions, the overexpression of CIP4 in pancreatic tumor cells and breast cancer cells have been demonstrated to be able to promote their migration and invasion. As the migration and invasion of cancer cells is a process analogous to that observed during renal epithelial-mesenchymal transition (EMT), the data above suggests that CIP4 may play as an important Rho GTPase signaling molecule to promote renal tubular EMT. In present study, we examined whether TGF-β1-induced EMT is mediated by CIP4. For this purpose, we measured CIP4 in the fibrotic renal tissue in a model of 5/6 nephrectomy, this model associated with EMT induced by over-expression of TGF-beta, and in vitro, we detected CIP4 in HK-2 cells treated with TGF-β1, and examined the effect of CIP4-siRNA on TGF-β1-induced EMT. As the polarity protein Par6 was identified as a key regulator in epithelial cell polarity and tight-junction, the interaction between CIP4 and Par6 was subsequently detected.
     Par I Relationship of CIP4 and Tubulointerstitial fibrosis
     Objective To study the expression and significance of Cdc42 interacting protein-4 (CIP4) in the model of chronic renal failure of 5/6 nephrectomized rats and the model of EMT of HK-2 cells induced by TGF-(31.
     Methods 20 SD rats were involved in this experiment cotaining 10 in the normal group. The model of chronic kidney disease was induced by a operation of 5/6 nephrectomy in rat. The expression of CIP4 was detected by Immunohistochemical SABC method at 12 weeks, and Western Blot was used to evaluate the proteins of CIP4. In vitro, CIP4 in HK-2 cells induced by TGF-β1 for 12,24,48,72 h were also detected by Western blot and RT-PCR.
     Results Immunohistochemical assay showed that rat CIP4 (rCIP4) as extensively expressed in renal tubules in 5/6-nephrectomized rats, whereas there was little staining in sham-operated rats at 12 weeks. The mRNA and proteins of CIP4 was also notably up-regulated in HK-2 cells induced by TGF-β1.
     Conclusion TGF-β1 could significantly improve the level of CIP4 in renal tubular epithelial cells both in vivo and in vitro.
     PartⅡCIP4 Regulates TGF-β1-Induced Renal Epithelial to Mesenchymal Transiformation
     Objective To observe the effect of CIP4 on human renal tubular epithelial to mesenchymal transition induced by TGF-β1 and to study the mechanism of its generation.
     Methods According to CIP4(human Cdc42 interacting protein-4) cDNA sequence in genebank (NM_004240), Reverse transcription polymerase chain reaction (RT-PCR) was used to amplify the full-length CIP4 by utilizing two primers including restriction sites Not I. After purification by RT-PCR, the amplified cDNA was inserted into the pcDNA 3.1 plasmid at the Not I site. To carry on, the recombined plasmids pcDNA 3.1/CIP4 were transfected into HK-2 cells. Western blot or confocal microscope was performed to detect the phenotypic changes correlated to EMT in pcDNA3.1/CIP4 transfected HK-2 cells, as indicated the changes in epithelial and mesenchymal protein E-cadherin and a-SMA respectively. One set of siRNA oligos specific for CIP4 were designed based on the full CIP4 sequence in Genbank, Then HK-2 cells induced by TGF-β1 were transfected with CIP4-siRNA via Lipofactamine 2000.Western blot were used to evaluate the protein expressions of CIP4, E-cadherin andα-SMA respectively in control cells, TGF-β1 treated cells, siRNA transfected cells. The distribution of E-cadherin and a-SMA was observed by confocal microscope. After we interferenced HK-2 cells stimulated with TGF-β1 with specific inhibitor of PI3K/Akt (Wortmannin) 1μM for 48 hours, Western blotting was used to detect the CIP4 protein in control cells and Interferenced cells.
     Results HK-2 cells acquired phenotypic changes correlated with EMT by transfection of CIP4. The normal HK-2 and pcDNA3.1/Zeo transfected cells were similar in morphology, presenting typical morphology of epithelial cells of round or ovoid shape, and they grew by adhering to the flask wall. As compared with normal HK-2 and pcDNA3.1/Zeo transfected cells, the cells transfected with hCIP4 were altered in morphology, presenting long fusiform and widened inter-cellular space, which was similar to the morphology of myofibroblast. A cell-cell adhesion molecule, E-cadherin present in the plasma membrane of most epithelial cells, was decreased and formed zipper-like patterns at cell borders in CIP4-transfected cells. a-SMA, markers of mesenchymal cells, were increased in CIP4-transfected cells. It show that TGF-β1 can induce EMT, after transfected with CIP4-siRNA, the protein expression of E-cadherin was increased, and a-SMA was decreased, the EMT induced by TGF-β1 was effectively reversed. With treatment of Wortmannin, the expression of CIP4 was decreased.
     Conclusion TGF-β1 produced a large number of CIP4 via PI3K/Akt pathway in renal tubular EMT and CIP4 further participate in TGF-β1 induced EMT.
     PartⅢCdc42 Interacting Protein-4 Regulates Transforming Growth Factor-β1-induced Renal Tubular Epithelial-Mesenchymal Transition through the Polarity Protein Par6
     Objective To observe the effect of CIP4 on the transdifferentiation of rat renal tubular epithelial cells (NRK-52E) induced by TGF-β1, and to search for the influence of the polarity protein Par6 on the TGF-β1-mediated transdifferentiation of NRK-52E cells.
     Methods In vitro, the rat tubular epithelial cells (NRK-52E) were cultured with 20μg/ L TGF-β1 for 72 h. Western blot or confocal microscope was used to detected the phenotypic changes associated with EMT in TGF-(31-treated NRK-52E cells, as illustrated by alterations in epithelial and mesenchymal protein E-cadherin and a-SMA, also as demonstrated the level of protein CIP4 and Par6 phosphorylation. To investigate whether CIP4 interacts with Par6, immunoprecipitation were used to evaluate the combination CIP4 and Par6. To further demonstrate the CIP4-induced EMT related to the activation of the polarity protein Par6, According to Par6 cDNA sequence (NM_001003653) and CIP4 cDNA (NM_ 053920.1) in genebank, a Par6-siRNA and a CIP4-siRNA were devised and synthesised by Shanghai Invitrogen company. The Par6-siRNA and CIP4-siRNA were transfected into TGF-β1-treated cells respectively, and the phenotype changes associated with TGF-β1-induced EMT were then detected by confocal microscope or Western blot.
     Results TGF-β1 induced phenotypic alternations correlate with EMT in NRK-52E cells. E-cadherin molecule, a cell-cell adhesiontake on in the plasma membrane of most epithelial cells, was decreased and shaped zipper-like patterns at cell borders in TGF-β1-treated cells. a-SMA, markers of mesenchymal cells, were increased in TGF-β1-treated NRK-52E cells. With treatment of Par6-siRNA and CIP4-siRNA respectively, the phosphorylated Par6 was reduced and the losting E-cadherin was re-expressed and re-localized at cell boundary in TGF-β1-treated cells, on the other hand a-SMA were deceased.
     Conclusion The overexpression of CIP4 is directly involved in TGF-β1-induced tubular EMT through the polarity Par6.
引文
1. Eddy A A. Molecular basis of renal fibrosis. Pediatr Nephrol,2000,15:290-301.
    2. Kalluri R, Neilson EG. Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest,2003,112:1776-1784.
    3. Iwano M, Plieth D, Danoff TM, et al. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest,2002,110:341-350.
    4. Li J, Qu X, Bertram JF, et al. Endothelial-myofibroblast transition contributes to the early development of diabetic renal interstitial fibrosis in streptozotocin-induced diabetic mice. Am J Pathol,2009,175:1380-1388.
    5. Liu Y. New insights into epithelial-mesenchymal transition in kidney fibrosis. J Am Soc Nephrol,2010,21:212-222.
    6. Zavadil J, Bottinger EP. TGF-beta and epithelial-to-mesenchymal transitions. Oncogene,2005,24:5764-5774.
    7. Zheng G, Lyons JG, Tan TK, et al. Disruption of E-cadherin by matrix metalloprotein-ase directly mediates epithelial-mesenchymal transition downstream of transforming growth factor-betal in renal tubular epithelial cells. Am J Pathol,2009,175:580-591.
    8. Barrios-Rodiles M, Brown KR, Ozdamar B, et al. High-throughput mapping of a dynamic signaling network in mam-malian cells. Science,2005,307:1621-1625.
    9. Zhang YE. Non-Smad pathways in TGF-p signaling. Cell Res,2009,19:128-139.
    10. Aspenstrom P. A Cdc42 target protein with homology to the non-kinase domain of FER has a potential role in regulating the actin cytoskeleton. Curr Biol,1997,7: 479-487.
    11. Holbert S, Dedeoglu A, Humbert S, et al. Cdc42-interacting protein4 binds to huntingtin:neuropathologic and biological evidence for a role in Huntington's disease. Proc Natl Acad Sci USA,2003,100:2712-2717.
    12. Takano K, Toyooka K, Suetsugu S. EFC/F-BAR proteins and the N-WASP-WIP complex induce membrane curvature-dependent actin polymerization. EMBO J,2008, 27:2817-2828.
    13. Toguchi M, Richnau N, Ruusala A, et al. Members of the CIP4 family of proteins participate in the regulation of platelet-derived growth factor receptor-beta-dependent actin reorganization and migration. Biol Cell,2010,102:215-230.
    14. Tsuji E, Tsuji Y, Fujiwara T,et al. Splicing variant of Cdc42 interacting protein-4 disrupts beta-catenin-mediated cell-cell adhesion:expression and function in renal cell carcinoma. Biochem and Biophys Res Commun,2006,339:1083-1088.
    15. Pichot CS, Arvanitis C, Hartig SM, et al. Cdc42-Interacting Protein 4 Promotes Breast Cancer Cell Invasion and Formation of Invadopodia through Activation of N-WASp. Cancer Res,2010,70:8347-8356.
    16. Roignot J, Taieb D, Suliman M, et al. CIP4 is a new ArgBP2 interacting protein that modulates the ArgBP2 mediated control of WAVE1 phosphorylation and cancer cell migration. Cancer Lett,2010,288:116-123.
    17. Guarino M. Epithelial-mesenchymal transition and tumour invasion. Int J Biochem Cell Biol,2007,39:2153-2160.
    18. Leibfried A, Fricke R, Morgan MJ, et al. Drosophila Cip4 and WASp define a branch of the Cdc42-Par6-aPKC pathway regulating E-cadherin endocytosis, Curr Biol,2008, 18:1639-1648.
    19. Harris KP, Tepass U. Cdc42 and Par proteins stabilize dynamic adherens junctions in the Drosophila neuroectoderm through regulation of apical endocytosis. J Cell Biol, 2008,183:1129-1143.
    20. Wirtz-Peitz F, Zallen JA. Junctional trafficking and epithelial morphogenesis. Curr Opin Genet Dev,2009,19:350-356.
    21. Lu H, Bilder D. Endocytic control of epithelial polarity and proliferation in Drosophila. Nat Cell Biol,2005,7:1232-1239.
    22. Ozdamar B, Bose R, Barrios-Rodiles M, et al. Regulation of the polarity protein Par6 by TGFbeta receptors controls epithelial cell plasticity. Science,2005,307:1603-1609.
    1 Wang L, Rudert WA, Grishin A, et al. Identification and geneticanalysis of human and mouse activated Cdc42 interacting protein-4 isoforms. Biochem Biophys Res Commun,2002,293:1426-1430.
    2 Aspenstrom P. A Cdc42 target protein with homology to the nonkinase domain of FER has a potential role in regulating the actin cytoskeleton. Curr Biol,1997,7: 479-487.
    3 Dombrosky-Ferlan P, Grishin A, Botelho RJ, et al. Felic (CIP4b), a novel binding partner with the Src kinase Lyn and Cdc42, localizes to the phagocytic cup. Blood, 2003,101:2804-2809.
    4 Tsuji E, Tsuji Y, Fujiwara T, et al. Splicing variant of Cdc42 interacting protein-4 disrupts β-catenin-mediated cell-cell adhesion:expression and function in renal cell carcinoma.Biochem Biophys Res Commun,2006,339:1083-1088.
    5 Ayton GS, Lyman E, Krishna V, et al. New insights into BAR domaininduced membrane remodeling. Biophys J,2009,97:1616-1625.
    6 Frost A, Perera R, Roux A, et al. Structural basis of membrane invagination by F-BAR domains. Cell,2008,132:807-817.
    7 Bu W, Chou AM, Lim KB, et al. The Toca-1-NWASP complex links filopodial formation to endocytosis. J Biol Chem,2009,284:11622-11636.
    8 Hartig SM, Ishikura S, Hicklen RS, et al. The F-BAR protein CIP4 promotes GLUT4 endocytosis through bidirectional interactions with N-WASp and Dynamin-2. J Cell Sci,2009,122:2283-2291.
    9 Kamioka Y, Fukuhara S, Sawa H, et al. A novel dynamin-associating molecule, formin-binding protein 17, induces tubularmembrane invaginations and participates in endocytosis. J Biol Chem,2004,279:40091-40099.
    10 Shimada A, Niwa H, Tsujita K, et al. Curved EFC/F-BAR-domain dimmers are joined end to end into a filament for membrane invagination in endocytosis. Cell, 2007,129:761-772.
    11 Tsujita K, Suetsugu S, Sasaki N, et al. Coordination between the actin cytoskeleton and membrane deformation by a novel membrane tubulation domain of PCH proteins is involved in endocytosis. J Cell Biol,2006,172:269-279.
    12 Holbert S, Dedeoglu A, Humbert S, et al. Cdc42-interacting protein 4 binds to huntingtin: neuropathologic and biological evidence for a role in Huntington's disease. Proc Natl Acad Sci USA,2003,100:2712-2717.
    13 Dombrosky-Ferian P, Grishin A,Botelho R.J, et al. Felic (CIP4b), a novel binding partner with the Src kinase Lyn and Cdc42, localizes to the phagocytic cup. Blood, 2003,101:2804-2809.
    14 Pichot CS, Arvanitis C, Hartig SM, et al. Cdc42-Interacting Protein 4 Promotes Breast Cancer Cell Invasion and Formation of Invadopodia through Activation of N-WASp. Cancer Res,2010,70:8347-8356.
    15 Roignot J, Taieb D, Suliman M, et al. CIP4 is a new ArgBP2 interacting protein that modulates the ArgBP2 mediated control of WAVE1 phosphorylation and cancer cell migration. Cancer Lett,2010,288:116-123.
    16 Vignjevic D, Montagnac G. Reorganisation of the dendritic actin network during cancer cell migration and invasion. Semin Cancer Biol,2008,18:12-22.
    17 Li JH, Wang W, Huang XR,et al. Advanced glycation end products induce tubular epithelial-myofibroblast transition through the RAGE-ERK1/2 MAP kinase signaling pathway. Am J Pathol,2004,164:1389-1397.
    18 Rastaldi MP. Epithelial to mesenchymal transition and its implications for the development of renal tubulointerstitial fibrosis. J Nephrol,2006,19:407-412.
    19 Yang J, Liu Y:Dissection of key events in tubular epithelial to myofibroblast transition
    20 its implications in renal interstitial fibrosis. Am J Pathol,2007,159:1465-1475.
    21 位红兰,曾锐,张娟,等.肾间质纤维化中DJ-1抑制抗纤维化因子PTEN的表达.中华肾脏病杂志,2009,25:782-787.
    22 Ng YY, Huang TP, Yang WC, et al. Tubular epithelial-myofibroblast transdifferentiation in
    23 progressive tubulointerstitial fibrosis in 5/6 nephrectomized rats. Kidney Int, 98;:864-876.
    24 Fricke R, Gohl C, Dharmalingam E, Grevelhorster A, et al. Drosophila Cip4/Toca-1 integrates membrane trafficking and actin dynamics through WASP and SCAR/WAVE. Curr Biol,2009,19:1429-3147.
    25 Leibfried A, Fricke R, Morgan MJ,et al. Drosophila Cip4 and WASp define a branch of the Cdc42-Par6-aPKC pathway regulating E-cadherin endocytosis.Curr Biol,2008, 18:1639-1648.
    26 Itoh T, Hasegawa J, Tsujita K, et al. The tyrosine kinase Fer is a downstream target of the PLD-PA pathway that regulates cell migration. Sci Signal,2009,2:ra52.
    27 Jun HS, Hwang K, Kim Y, et al. High-fat diet alters PP2A, TC10, and CIP4 expression in visceral adipose tissue of rats. Obesity (Silver Spring).2008,16:1226-1231.
    28 Zeng R, Yao Y, Han M, et al. Biliverdin reductase mediates hypoxia-induced EMT via PI3-kinase and Akt. J Am Soc Nephrol.2008,19:380-387.
    1 Eddy AA. Molecular basis of renal fibrosis. Pediatr Nephrol,2000,15:290-301.
    2 Iwano M, Neilson SG. Mechanisms of tubulointerstitial fibrosis. Curr Opin Nephrol Hypertens,2004,13:279-284.
    3 Heidland A, Sebekova K, Paczek L, et al. Renal fibrosis:role of impaired proteolysis and potential therapeutic strategies. Kidney Int Suppl,1997,62:S32-35.
    4 Liu Y. Epithelial to mesenchymal transition in renal fibrogenesis:Pathologic significance, molecular mechanism, and therapeutic intervention. J Am Soc Nephrol, 2004,15:1-12.
    5 Strutz FM. EMT and proteinuria as progression factors. Kidney Int,2009,75:475-481.
    6 Iwano M, Plieth D, Danoff TM, et al. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest,2002,110:341-350.
    7 Rastaldi MP, Ferrario F, Giardino L,et al. Epithelial-mesenchymal transition of tubular epithelial cells in human renal biopsies. Kidney Int,2002,62:137-146.
    8 Hertig A, Anglicheau D, Verine J, et al. Early epithelialp henotypic changes predict graft fibrosis. J Am Soc Nephrol,2008,19:1584-1591.
    9 Strutz F, Neilson EG. New insights into mechanisms of fibrosis in immune renal injury.Springer Semin Immunopathol,2003,24:459-476.
    10 Gore-Hyer E, Shegogue D, Markiewicz M et al. TGF-beta and CTGF have overlapping and distinct fibrogenic effects on human renal cells. Am J Physiol Renal Physiol,2002; 283:F707-716.
    11 Zavadil J, Bottinger EP. TGF-beta and epithelial-to-mesenchymal transitions. Oncogene, 2005,24:5764-5774.
    12 Lan HY. Tubular epithelial-myofibroblasttransdifferentiation mechanismsin proximal tubule cells. Curr Opin Nephrol Hypertens,2003,12:25-29.
    13 Dai C, Yang J, Liu Y.Transforming growth factor-betal potentiates renal tubular epithelial cell death by a mechanism independent of Smad signaling. J Biol Chem,2003, 278:12537-12545.
    14 Eric G Neilson. Setting a trap for tissue fibrosis. Nat Med,2005,11:387-393.
    15 Li Y, Yang J, Dai C, et al. Role for integrin-linked kinase in mediating tubular epithelial to mesenchymal transition and renal interstitial fibrigenesis. J Chin Invest, 2003,112:503-516
    16 Docherty NG, O'Sullivan OE, et al. TGF-betal-induced EMT can occur independently of its proapoptotic effects and is aided by EGF receptor activation. Am J Physiol Renal Physiol,2006,290:F1202-12
    17 Liu Y. Renal fibrosis:new insights into the pathogenesis and therapeutics. Kidney Int, 2006,69,213-217.
    18 Itoh T, Hasegawa J, Tsujita K, et al. The tyrosine kinase Fer is a downstream target of the PLD-PA pathway that regulates cell migration. Sci Signal,2009,8; 2:ra52.
    19 Jun HS, Hwang K, Kim Y, et al. High-fat diet alters PP2A, TC10, and CIP4 expression in visceral adipose tissue of rats. Obesity (Silver Spring),2008,16:1226-1231.
    20 Zeisberg M, Duffield JS. Resolved:EMT produces fibroblasts in the kidney. J Am Soc Nephrol,2010,21:1247-1253.
    21 Liu Y. Epithelial to mesenchymal transition in renal fibrogenesis:pathologic significance, molecular mechanism, and therapeutic intervention. J Am Soc Nephrol, 2004,15:1-12.
    22 Ozdamar B, Bose R, Barrios-Rodiles M, et al. Regulation of the polarity protein Par6 by TGFbeta receptors controls epithelial cell plasticity.Science,2005,307:1603-1609
    23 Xiangyang Wang, Jing Nie, Qin Zhou, et al. Downregulation of Par-3 expression and disruption of Par complex integrity by TGF-β during the process of epithelial to mesenchymal transition in rat proximal epithelial cells. Biochimica et Biophysica Acta, 1782:51-59.
    24 Zhang YE. Non-Smad pathways in TGF-beta signaling. Cell Res.2009,19:128-139.
    25位红兰,梁萍萍,李娣昕,等.DJ-癌基因在肾间质纤维化中表达的实验研究.中华肾脏病杂志,2009,25:288-492.
    26位红兰,曾锐,张娟,等.肾间质纤维化中DJ-1抑制抗纤维化因子PTEN的表达.中华肾脏病杂志,2009,25:782-787.
    27 Edlund S, Landstrom M, Heldin CH, et al. Smad7 is required for TGF-b-induced activation of the small GTPase Cdc42. J Cell Sci,2003,117:1835-1846.
    28 Qian Y, Zhong X, Flynn DC, et al. ILK mediates actin filament rearrangements and cell migration and invasion through PI3K/Akt/Racl signaling. Oncogene.2005,24:3154-3165.
    29 Holbert S, Dedeoglu A, Humbert S, et al. Cdc42-interacting protein 4 binds to huntingtin:neuropathologic and biological evidence for a role in Huntington's disease. Proc Natl Acad Sci USA,2003,100:2712-2717.
    30 Tsuji E, Tsuji Y, Fujiwara T, et al. Splicing variant of Cdc42 interacting protein-4 disrupts beta-catenin- mediated cell-cell adhesion:expression and function in renal cell carcinoma. Biochem and Biophys Res Commun,2006,339:1083-1088.
    31 Tian L, Nelson DL, Stewart DM. Cdc42-interacting protein 4 mediates binding of the Wiskott-Aldrich syndrome protein to microtubules. J Biol Chem,2000,275:7854-7861.
    32 Wirtz-Peitz F, Zallen JA. Junctional trafficking and epithelial morphogenesis. Curr Opin Genet Dev,2009,19:350-356.
    33 Leibfried A, Fricke R, Morgan MJ, et al. Drosophila Cip4 and WASp define a branch of the Cdc42-Par6- aPKC pathway regulating E-cadherin endocytosis. EMBO J,2008,18: 1639-1648.
    1 Grunert S, Jechlinger M, Beug H. Diverse cellular and molecular mechanisms contribute to epithelial plasticity and metastasis. Nat Rev Mol Cell Biol,2003,4: 657-665.
    2 Baum B, Settleman J, Quinlan MP. Transitions between epithelial and mesenchymal states in development and disease. Semin Cell Dev Biol,2008,19:294-308.
    3 Shi Y, Massague J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell,2003,13; 113:685-700.
    4 Derynck R, Zhang YE. Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature,2003,425:577-584.
    5 Ozdamar B, Bose R, Barrios-Rodiles M, et al. Regulation of the polarity protein Par6 by TGFbeta receptors controls epithelial cell plasticity.Science,2005,307:1603-1609.
    6 Lin D, Edwards AS, Fawcett JP. A mammalian PAR-3-PAR-6 complex implicated in Cdc42/Racl and aPKC signalling and cell polarity. Nat Cell Biol,2000,2:540-547.
    7 Joberty G, Petersen C, Gao L,et al. The cell-polarity protein Par6 links Par3 and atypical protein kinase C to Cdc42. Nat Cell Biol,2000,2:531-539.
    8 Wirtz-Peitz F, Zallen JA. Junctional trafficking and epithelial morphogenesis. Curr Opin Genet Dev,2009,19:350-356.
    9 Lu H, Bilder D. Endocytic control of epithelial polarity and proliferation in Drosophila. Nat Cell Biol,2005,7:1232-1239.
    10 Aspenstrom P. A Cdc42 target protein with homology to the non-kinase domain of FER has a potential role in regulating the actin cytoskeleton. Curr Biol,1997,7:479-487.
    11 Leibfried A, Fricke R, Morgan MJ, et al. Drosophila Cip4 and WASp define a branch of the Cdc42-Par6-aPKC pathway regulating E-cadherin endocytosis. Curr Biol,2008, 18:1639-1648.
    12 Kalluri R, Neilson EG. Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest,2003,112:1776-1784.
    13 Zeng R, Han M, Luo Y, et al. Role of Sema4C in TGF-{beta} 1-induced mitogen-activated protein kinase activation and epithelial-mesenchymal transition in renal tubular epithelial cells. Nephrol Dial Transplant,2011,26:1149-1156.
    14 Bottinger EP, Bitzer M. TGF-beta signaling in renal disease. J Am Soc Nephrol,2002, 13:2600-2610.
    15 Barrios-Rodiles M, Brown KR, Ozdamar B, et al.High-throughput mapping of a dynamic signaling network in mammalian cells. Science,2005,307:1621-1625.
    16 Gao L and Macara IG. Isoforms of the polarity protein par6 have distinct 711 functions. J Biol Chem,2004,279:41557-41562.
    17 Hurd TW, Gao L, Roh MH, et al. Direct interaction of two polarity complexes implicated in epithelial tight junction assembly. Nat Cell Biol,2003,5:137-142
    18 Lemmers C, Michel D, Lane-Guermonprez L, et al. CRB3 binds directly to Par6 and regulates the morphogenesis of the tight junctions in mammalian epithelial cells. Mol Biol Cell,2004,15:1324-1333.
    19 Karp CM, Tan TT, Mathew R,et al. Role of the polarity determinant crumbs in suppressing mammalian epithelial tumor progression. Cancer Res,2008,68: 4105-4115.
    20 Whiteman EL, Liu CJ, Fearon ER, et al. The transcription factor snail represses Crumbs3 expression and disrupts apico-basal polarity complexes. Oncogene,2008,27: 3875-3879.
    21 Wang Z, Sandiford S, Wu C, Li SS. Numb regulates cell-cell adhesion and polarity in response to tyrosine kinase signalling.EMBO J,2009,28:2360-2373.
    22 Wang X, Nie J, Zhou Q, et al. Downregulation of Par-3 expression and disruption of Par complex integrity by TGF-beta during the process of epithelial to mesenchymal transition in rat proximal epithelial cells. Biochim Biophys Acta,2008,1782:51-59.
    23 Townsend TA, Wrana JL, Davis GE, et al. Trans forming growth factor-beta-stimulated endocardial cell transformation is dependent on Par6c regulation of RhoA.J Biol Chem, 2008,283:13834-13841.
    24 Joberty G, Petersen C, Gao L, et al. The cell-polarity protein Par6 links Par3 and atypical protein kinase C to Cdc42. Nat Cell Biol,2000,2:531-539.
    25 Johansson A, Driessens M, and Aspenstrom P. The mammalian homologue of the Caenorhabditis elegans polarity protein PAR-6 is a binding partner for the Rho GTPases Cdc42 and Racl. J Cell Sci,2000,113:3267-3275.
    26 Hugo H, Ackland ML, Blick T, et al. Epithelial-mesenchymal and esenchymal-epithelial transitions in carcinoma progression. J Cell Physiol,2007,213:374-383.
    27 Guarino M. Epithelial-mesenchymal transition and tumour invasion. Int J Biochem Cell Biol 2007,39:2153-2160.
    28 Mandal M, Myers JN, Lippman SM, et al. Epithelial to mesenchymal transition in head and neck squamous carcinoma: association of Src activation with E-cadherin down-regulation, vimentin expression, and aggressive tumor features. Cancer,2008, 112:2088-2100.
    29 Pieczynski J, Margolis B.Protein complexes that control renal epithelial polarity. Am J Physiol Renal Physiol,2011,300:F589-601.
    30 Assemat E, Bazellieres E, Pallesi-Pocachard E, et al. Polarity complex proteins. Biochim Biophys Acta.2008,1778:614-630.
    31 Leibfried A, Fricke R, Morgan MJ, et al. Drosophila Cip4 and WASp define a branch of the Cdc42-Par6-aPKC pathway regulating E-cadherin endocytosis, Curr Biol,2008, 18:1639-1648.
    32 Wirtz-Peitz F, Zallen JA. Junctional trafficking and epithelial morphogenesis. Curr Opin Genet Dev,2009,19:350-356.
    1 Nelson WJ. Adaptation of core mechanisms to generate cell polarity. Nature,2003,422: 766-774.
    2 Shin K, Fogg V, Margolis B. Tight junctions and cell polarity. Annual 1030 Review of Cell and Developmental Biology,2006,22:207-235.
    3 O'Brien L, Zegers MMP, Mostov K. Opinion: Building epithelial architecture:insights from three-dimensional culture models. Nature Reviews Molecular Cell Biology,2002, 3:531-537.
    4 Takai Y, Miyoshi J, Ikeda W, et al. Nectins and nectin-like molecules:roles in contact inhibition of cell movement and proliferation. Nature Reviews Molecular Cell Biology, 2008,9:603-615.
    5 Furuse M. Molecular basis of the core structure of tight junctions. Cold Spring Harbor perspectives in biology,2010,2:002907-002907.
    6 Steed E, Balda M, and Matter K. Dynamics and functions of tight junctions. Trends in cell biology 2010,20:142-149.
    7 McNeill H. Planar cell polarity and the kidney. Journal of the American Society of Nephrology,2009,20:2104-2111.
    8 Goldstein B, Macara I. The PAR proteins:fundamental players in animal cell polarization. Developmental cell,2007,13:609-622.
    9 Lynch AM Hardin J. The assembly and maintenance of epithelial junctions in C. elegans. Front Bio sci,2009,14:1414-1432.
    10 Etemad-Moghadam B, Guo S, Kemphues KJ. Asymmetrically distributed PAR-3 protein contributes to cell polarity and spindle alignment in early C. elegans embryos.Cell,1995,83:743-752.
    11 Kemphues KJ, Priess JR, Morton DG, et al. Identification of genes required for cytoplasmic ocalization in early C. elegans embryos. Cell,1998,52:311-320,
    12 Gao L, Macara IG, Joberty G. Multiple splice variants of Par3 and of a novel related gene, Par3L, produce proteins with different binding properties. Gene,2002,294: 99-107.
    13 Kohjima M, Noda Y, Takeya R, et al. PAR3beta, a novel homologue of the cell polarity protein PAR3, localizes to tight junctions. Biochem Biophys Res Commun, 2002,299:641-646.
    14 Suzuki A, Yamanaka T, Hirose T, et al. Atypical protein kinase C is involved in the evolutionarily conserved par protein complex and plays a critical role in establishing epithelia-specific junctional structures. J Cell Biol,2001,152:1183-1196.
    15 Hung TJ, Kemphues KJ. PAR-6 is a conserved PDZ domain-containing protein that colocalizes with PAR-3 in Caenorhabditis elegans embryos. Development,1999,126: 127-135.
    16 Izumi Y, Hirose T, Tamai Y, et al. An atypical PKC directly associates and colocalizes at the epithelial tight junction with ASIP, a mammalian homologue of Caenorhabditis elegans polarity protein PAR-3. J Cell Biol,1998,143:95-106.
    17 Joberty G, Petersen C, Gao L, et al. The cell-polarity protein Par6 links Par3 and atypical protein kinase C to Cdc42. Nat Cell Biol,2000,2:531-539.
    18 Horikoshi Y, Suzuki A, Yamanaka T, et al. Interaction between PAR-3 and the aPKC-PAR-6 complex is indispensable for apical domain development of epithelial cells. J Cell Sci,2009,122:1595-1606.
    19 Lin D, Edwards AS, Fawcett JP, et al. A mammalian PAR-3-PAR-6 complex implicated in Cdc42/Racl and aPKC signalling and cell polarity. Nat Cell Biol,2000,2: 540-547.
    20 Mizuno K, Suzuki A, Hirose T, et al. Self-association of PAR-3-mediated by the conserved N-terminal domain contributes to the development of epithelial tight junctions. J Biol Chem,2003,278:31240-31250.
    21 Grzeschik N, Parsons L, Allott M, et al. Lgl, aPKC,and Crumbs regulate the Salvador/Warts/Hippo pathway through two distinct mechanisms. Current biology, 2010,20:573-581.
    22 Montcouquiol M, Sans N, Huss D, et al. Asymmetric localization of Vangl2 and Fz3 indicate novel mechanisms for planar cell polarity in mammals. J Neurosci,2006,26: 5265-5275.
    23 Nagai-Tamai Y, Mizuno K, Hirose T, et al. Regulated protein-protein interaction between aPKC and PAR-3 plays an essential role in the polarization of epithelial cells. Genes Cells,2002,7:1161-1171.
    24 Suzuki A, Ishiyama C, Hashiba K, et al. aPKC kinase activity is required for the asymmetric differentiation of the premature junctional complex during epithelial cell polarization. J Cell Sci,2002,115:3565-3573.
    25 Walther RF, Pichaud F. Crumbs/DaPKC-Dependent Apical Exclusion of Bazooka Promotes Photoreceptor Polarity Remodeling. Current biology,2010,20:1065-1074.
    26 Benton R, St Johnston D. Drosophila PAR-1 and 14-3-3 inhibit Bazooka/PAR-3 to establish complementary cortical domains in polarized cells. Cell,2003,115:691-704.
    27 Hurd TW, Fan S, Liu CJ, et al. Phosphorylation-dependent binding of 14-3-3 to the polarity protein Par3 regulates cell polarity in mammalian epithelia. Curr Biol,2003,13: 2082-2090.
    28 Wang Y, Du D, Fang L, et al. Tyrosine phosphorylated Par3 regulates epithelial tight junction assembly promoted by EGFR signaling. EMBO J,2006,25:5058-5070.
    29 Traweger A, Wiggin G, Taylor L, et al. Protein phosphatase regulates the phosphorylation state of the polarity scaffold Par-3. Proc Natl Acad Sci,2008, 105:10402-10407.
    30 Iden S, Collard J. Crosstalk between small GTPases and polarity proteins in cell polarization. Nature Reviews Molecular Cell Biology,2008,9:846-859.
    31 Chen X Macara IG. Par-3 controls tight junction assembly through the Rac exchange factor Tiaml. Nat Cell Biol,2005,7:262-269.
    32 Nakayama M, Goto TM, Sugimoto M, et al. Rho-kinase phosphorylates PAR-3 and disrupts PAR complex formation. Dev Cell,2008,14:205-215.
    33 Sugimoto M, Inoko A, Shiromizu T, et al. The keratin-binding protein Albatross regulates polarization of epithelial cells. J Cell Biol.2008,183:19-28,.
    34 Feng W, Wu H, Chan LN, et al. Par-3-mediated junctionallocalization of the lipid phosphatase PTEN is required for cell polarity establishment. J Biol Chem,2008,283: 23440-23449.
    35 Wu H, Feng W, Chen J, et al. PDZ domains of Par-3 as potential phosphoinositide signaling integrators. Mol Cell,2007,28:886-898.
    36 Ebnet K, Suzuki A, Horikoshi Y, et al. The cell polarity protein ASIP/PAR-3 directly associates with junctional adhesion molecule (JAM). EMBO J,2001,20:3738-3748.
    37 Itoh M, Sasaki H, Furuse M, et al. Junctional adhesion molecule (JAM) binds to PAR-3: a possible mechanism for the recruitment of PAR-3 to tight junctions. J Cell Biol,2001, 54:91-97.
    38 Gao L and Macara IG. Isoforms of the polarity protein par6 have distinct functions. J Biol Chem,2004,279:41557-41562.
    39 Hurd TW, Gao L, Roh MH, et al. Direct interaction of two polarity complexes implicated in epithelial tight junction assembly. Nat Cell Biol,2003,5:137-142.
    40 Lemmers C, Michel D, Lane-Guermonprez L, et al. CRB3 binds directly to Par6 and regulates the morphogenesis of the tight junctions in mammalian epithelial cells. Mol Biol Cell,2004,15:1324-1333.
    41 Wang Q, Hurd TW, Margolis B. Tight junction protein Par6 interacts with can evolutionarily conserved region in the amino terminus of PALS1/stardust. J Bio Chem, 2004,279:30715-30721.
    42 Johansson A, Driessens M, Aspenstrom P. The mammalian homologue of the Caenorhabditis elegans polarity protein PAR-6 is a binding partner for the Rho GTPases Cdc42 and Racl. J Cell Sci,2000,113 (18):3267-3275.
    43 Yamanaka T, Horikoshi Y, Suzuki A, et al. PAR-6 regulates αPKC activity in a novel way and mediates cell-cell contact-induced formation of the epithelial junctional complex. Genes Cells,2001,6:721-731.
    44 Gao L, Macara IG. Isoforms of the polarity protein par6 have distinct functions. J Biol Chem,2004,279:41557-41562.
    45 Garrard SM, Capaldo CT, Gao L, et al. Structure of Cdc42 in a complex with the GTPase-binding domain of the cell polarity protein, Par6. EMBO J,2003, 22:1125-1133.
    46 Peinado H, Olmeda D, and Cano A. Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype. Nat Rev Cancer,2007,7:415-428.
    47 Yamanaka T, Horikoshi Y, Sugiyama Y, et al. Mammalian Lgl forms a protein complex withPAR-6 and aPKC independently of PAR-3 to regulate epithelial cell polarity. Curr Biol,2003,13:734-743.
    48 Gao L, Joberty G, and Macara IG. Assembly of epithelial tight junctions is negatively regulated by Par6. Curr Biol,2002,12:221-225.
    49 Nolan ME, Aranda V, Lee S, et al. The polarity protein Par6 induces cell proliferation and is overexpressed in breast cancer. Cancer Res,2008,68:8201-8209.
    50 Akimoto K, Mizuno K, Osada S, et al. A new member of the third class in the protein kinase C family, PKC lambda, expressed dominantly in an undifferentiated mouse embryonal carcinoma cell line and also in many tissues and cells. J Biol Chem,1994, 269:12677-12683.
    51 lant PJ, Fawcett JP, Lin DC, et al. A polarity complex of mPar-6 and atypical PKC binds, phosphorylates and regulates mammalian Lgl. Nat Cell Bio,2003,15:301-308.
    52 Yamanaka T, Horikoshi Y, Sugiyama Y, et al. Mammalian Lgl forms a protein complex with PAR-6 and PKC independently of PAR-3 to regulate epithelial cell polarity. Curr Biol,2003,13:734-743.
    53 Morais-de-Sa E, Mirouse V, St Johnston D. aPKC phosphorylation of Bazooka defines the apical/lateral border in Drosophila epithelial cells. Cell,2010,141:509-523.
    54 Shen L, Turner J. Intercellular junctions:actin the PARt. Current biology.2008,18: R1014-R1017.
    55 Shivas J, Morrison H, Bilder D, et al. Polarity and endocytosis:reciprocal regulation. Trends in cell biology,2010,20:445-452.
    56 Leitges M, Sanz L, Martin P, et al. Targeted disruption of the zeta PKC gene results in the impairment of the NF-kappa B pathway. Molecular cell,2001,8:771-780.
    57 Imai F, Hirai S-i, Akimoto K, et al. Inactivation of aPKClambda results in the loss of adherens junctions in neuroepithelial cells without affecting neurogenesis in mouse neocortex. Development 2006,133:1735-1744.
    58 Hirose T, Satoh D, Kurihara H, et al. An essential role of the universal polarity protein, aPKClambda, on the maintenance of podocyte slit diaphragms. PloS one,2009; 4: e4194-e4194.
    59 Huber T, Hartleben B, Winkelmann K, et al. Loss of podocyte aPKClambda/iota causes polarity defects and nephrotic syndrome. Journal of the American Society of Nephrology,2009,20:798-806.
    60 Hartleben B, Schweizer H, Lbben P, et al. Neph-Nephrin proteins bind the Par3-Par6-atypical protein kinase C (aPKC) complex to regulate podocyte cell polarity. The Journal of biological chemistry.2008,283:23033-23038.
    61 Chaffer CL, Thompson EW, Williams ED. Mesenchymal to epithelial transition in development and disease. Cells Tissues Organs,2007,185:7-19.
    62 Thiery J, Acloque H, Huang YJ. Epithelial-mesenchymal transitions in development and disease. Cell,2009,139:871-890.
    63 Schmidt-Ott K, Yang J, Chen X, et al. Novel regulators of kidney development from the tips of the ureteric bud. Journal of the American Society of Nephrology,2005,16: 1993-2002.
    64 Schluter MA and Margolis B. Apical lumen formation in renal epithelia. J Am Soc Nephrol,2009,20:1444-1452.
    65 Peinado H, Olmeda D, Cano A. Snail, Zeb and bHLH factors in tumour progression:an alliance against the epithelial phenotype? Nat Rev Cancer,2007,7:415-428
    66 Ikenouchi J, Matsuda M, Furuse M. et al. Regulation of tight junctions during the epithelium-mesenchyme transition:direct repression of the gene expression of claudins/occludin by Snail. J Cell Sci,2003,116:1959-1967.
    67 Aigner K, Dampier B, Descovich L. et al, The transcription factor ZEB1 (deltaEF1) promotes tumour cell dedifferentiation by repressing master regulators of pithelial polarity. Oncogene,2007,26:6979-6988.
    68 Caruana G. Genetic studies define MAGUK proteins as regulators of epithelial cell polarity. The International journal of developmental biology,2002,46:511-518.
    69 Ozdamar B, Bose R, Barrios-Rodiles M, et al. Regulation of the polarity protein Par6 by TGFbeta receptors controls epithelial cell plasticity. Science,2005,307:1603-1609.
    70 Aranda V, Haire T, Nolan M, et al.Par6-aPKC uncouples ErbB2 induced disruption of polarized epithelial organization from proliferation control. Nature Cell Biology,2006, 8:1235-1245.
    71 Neilson EG. Mechanisms of disease:Fibroblasts--a new look at an old problem.Nature clinical practice nephrology,2006,2:101-108.
    72 Liu Y. New insights into epithelial-mesenchymal transition in kidney fibrosis. Journal of the American Society of Nephrology,2010,21:212-222.

© 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