内吞相关蛋白调控AngⅡ诱导心肌肥大作用机制的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
有研究证明,受体介导的配体内吞可以降低细胞膜表面受体的数量,使受体配体复合物进入胞内溶酶体降解并发生解离,引起受体功能的下调和再循环至细胞膜表面。另外,还有学者认为内吞进入细胞内的配体受体复合物可能参与了下游信号的转导与调控,发挥其生理学及病理生理学作用。血管紧张素II(Angiotensin II,Ang II)是肾素-血管紧张素系统(Renin-angiotensin system,RAS)最主要的效应分子,具有收缩血管效应、调控醛固酮分泌等生理功能。Ang II与血管紧张素II 1型受体(Angiotensin II 1 type receptor,ATl receptor)在细胞膜表面结合,一方面激活下游信号转导途径发挥生物学作用,另一方面快速的以网格蛋白包被小泡的方式发生内吞,其中组成网格蛋白包被的主要衔接蛋白为衔接蛋白复合体2(AP2)。最近的研究表明,AngII与细胞膜表面AT1受体结合,刺激心肌细胞肥大和(或)成纤维细胞增生,是导致心肌肥厚的主要原因之一。但是,ATl受体介导的AngⅡ内吞是否也参与了心肌肥厚的形成尚需进一步地研究。本实验利用流式细胞术、激光共聚焦显微镜、RNAi、RT-PCR及Western-blot等技术,研究发现AngII可以引起H9C2细胞体积增加;p38的磷酸化形式增多,NF-κB(p65)入核增多和氯离子通道蛋白ClC-2表达增加;同时使细胞进入S期的比例增加,G1期的比例降低,提示细胞DNA和蛋白合成增加。应用内吞特异性抑制剂PAO可以抑制上述细胞体积和信号途径的改变,与应用AT1受体拮抗剂(ARB)candesartan的作用一致,即PAO与Candesartan都可抑制AngII的促细胞肥大增生作用。初步证明AngII在细胞膜上与AT1受体结合后,在激活下游心肌肥大相关信号因子的同时,发生快速的受体内吞,这种配体介导的受体内吞在进入细胞内可能依然发挥着病理生理学作用。本实验又通过RNAi技术敲除AP2,抑制网格蛋白介导的内吞,发现可以抑制AT1受体介导的AngII内吞,并影响细胞体积,细胞周期及相关信号蛋白的改变,但对ClC-2表达无明显影响。因此我们得出初步结论,ATl受体介导的AngII内吞,不但使配体受体复合物进入细胞内,发生降解和受体配体的解离,及受体的再循环,而且内吞入胞内的受体在细胞内继续发挥其生物学功能,持续激活下游信号,通过影响细胞周期的改变,引起细胞的肥大增生。但其具体作用机制还需要进一步研究探讨。
Objective
     Myocyte hypertrophy can increase the risk of cardiac ischemia, left ventricular dysfunction and congestive heart failure, and it is a risk factor of leading to sudden cardiac death. Angiotensin II(Ang II) is the most important effect molecule of myocyte hypertrophy in renin-angiotensin system. Ang II binding its angiotensin II type 1(AT1) receptor in cell surface, can active downstream signal transduction pathways, and stimulate myocyte hypertrophy and/or fibroblast proliferation , leading to cardiac hypertrophy. While Ang II active AT1 receptor, the ligand and receptor complax endocytosis by clathrin-coated vesicle. However , the role of model, pathway and signal transductiong of AT1 receptor-mediated endocytosis in cardiac hypertrophy formation is not clear. Previous studies suggest that endocytosis can decrease the number of receptor of cell surface, promote degradation of receptor ligand complex in lysosome, and cause receptor function down-regulation and receptor recycling. However, some people have found, the ligand receptor complex through endocytosis can continue to active downstream signals, play a role of pathophysiology. Therefore , based on the view, we inhibited the AngII AT1 receptor-mediated endocytosis by endocytosis inhibitors ((phenylarsine oxide, PAO) and RNAi endocytosis-related protein (adaptor protein 2, AP2), observed the AngII induced myocardial cell hypertrophy and hyperplasia associated signaling pathway for the pathogenesis of cardiovascular diseases and provided a theoretical basis for clinical treatment.
     Methods:
     By rat myocardial cell line (H9C2), through AngII, endocytosis inhibitor (PAO), angiotensin II receptor blocker (candesartan) and RNAi silencing endocytosis-related protein(AP2), observed the following points:
     (1) H9C2 cells treated with candesartan and the PAO 30min, then added Ang II 24h, cells were collected and analyzed cell volume changes by flow cytometry.
     (2) H9C2 cells treated with candesartan and the PAO 30min, then added AngⅡ30min, cells were collected and extracted total protein, cytoplasmic and nuclear proteins from the cells, observed the changes of phosph-p38 and the distribution of NF-κB (p65) in cytoplasm and nucleus.
     (3) H9C2 cell synchronized by serum starvation, treated with candesartan and the PAO 30min, then added Ang II 24h, PI staining, detected change of cell cycle (G1, G2, S and M phase) by flow cytometry.
     (4) Construction of AP2 interference vector and detected silence effect. According to rat AP2-μ2 subunit gene sequences from Gene Bank and RNAi design principles, we designed and constructed three pSH1Si-AP2 expression vector. By transfecting H9C2 cells, silence effect of AP2 mRNA and protein expression was detected by RT-PCR and Western Blotting.
     (5) H9C2 cells treated with AngII at different time points (5min, 10min, 15min and 30min) and at different concentrations (10-6M ,10-7M and 10-8M), the morphology change of AT1 receptor-mediated endocytosis of AngⅡwere detected by the immunofluorescence technique and laser confocal microscope.
     (6) H9C2 cells treated with PAO, candesartan, and transfected pSH1Si-AP2 plasmid, the morphology change of AT1 receptor-mediated endocytosis of AngⅡwere detected by laser confocal microscope.
     (7) The AngII-induced cell volume, p38 phosphorylation, NF-κB (p65) of nuclear transfer and cell cycle changes was observed by inhibiting the expression of AP2 protein of H9C2 cells.
     Results:
     Testing cell survival by MTT, H9C2 cells survival had no effect at PAO 0.25μM and candesartan 40μM, so we choose the drug dose of above in experiment. H9C2 cell treated with Ang II. AngII also induced activation of MAPK signaling pathway throught p38 phosphorylation; and allowed NF-κB (p65) moving into the nucleus. Candesartan can inhibit the above change and decrease p38 phosphorylation, p65 nuclear transfer reduction by inhibiting Ang II and AT1 receptor binding at cell membrane. PAO also inhibited Ang II-induced p38 phosphorylation and p65 nuclear translocation. Cell cycle changes were observed in H9C2 cells treated with Ang II that G0/G1 phase cells decreased, and G1 / S phase cells increased by flow cytometry. Candesartan and PAO can inhibit above change of cell cycle.
     AP2 RNAi expression vector was successfully constructed by sequencing non-base mismatch and loss. pSH1Si-AP2 plasmid-1 plasmid can inhibit the AP2 of mRNA and protein expression by transfected AP2 RNAi expression vector in H9C2 cells. It suggested that AP2 RNAi vectors can be successfully silence the expression of AP2. After treating with AngII, AT1 receptor endocytosis quickly occur by immunofluorescence in H9C2 cells, and the most efficient endocytosis occur in Ang II concentration of 10-7M for 15min. Candesartan and PAO can inhibit AT1 receptor endocytosis. PSH1Si-AP2 plasmid-1 can also inhibit the AT1 receptor-mediated Ang II endocytosis after transient transfection of H9C2 cells, suggesting that endocytosis of Ang II and AT1 receptor mainly by clathrin-coated vesicles. At the same time, AP2 silence can suppress cell volume increase induced by Ang II, p38 phosphorylation and p65 nuclear transfer, also inhibit Ang II-induced cell cycle changes.
     Conclusion:
     (1). By flow cytometry, Ang II can increase the volume of H9C2 cells, successed copy the model of cell hypertrophy and hyperplasia. Candesartan and PAO can inhibit the AngII role in promoting cell hypertrophy. Candesartan for ligand binding receptor antagonist, directly block the downstream signal transduction, inhibition of cell hypertrophy, PAO mainly endocytosis inhibitors, considered by blocking endocytosis , affected downstream signal transduction, and inhibition of cell hypertrophy.
     (2). Ang II induced H9C2 cell activation of MAPK signaling pathway, increased p38 phosphorylation and NF-κB (p65) into the nucleus. Candesartan can inhibit the activation of these signaling pathways, by inhibiting the AT1 receptor and Ang II binding at membrane, it suggested that Ang II promotes H9C2 cells hypertrophy by activating p38 sigal and affecting the NF-κB (p65) into the nucleus.
     (3). PAO also can inhibit Ang II-induced p38 phosphorylation and p65 nuclear transfer, because PAO was the inhibitor for the AT1 receptor endocytosis, it suggested that inhibiting the AT1 receptor-mediated Ang II endocytosis may also prevent the MAPK and activation of NF-κB pathway, endocytosis of the ligand receptor complex is likely to continue the signal transduction pathway.
     (4). Ang II can decrease the proportion of H9C2 cells of G0/G1 phase cells, and increased the proportion of G1 / S phase, suggesting that Ang II promote DNA and protein synthesis by influencing the cell cycle, causing cell hypertrophy and hyperplasia. Similar with candesartan, PAO also can inhibit change of the cell cycle, suggesting that inhibition of receptor endocytosis also affected the changes of cell cycle by Ang II induced.
     (5). After treating with AngII, AT1 receptor endocytosis quickly occur by immunofluorescence in H9C2 cells, and the most efficient endocytosis occur in Ang II concentration of 10-7M for 15min. Candesartan can inhibit AT1 receptor endocytosis by inhibiting ligand and receptor binding. PAO also inhibit endocytosis by inhibitiong of receptor intracellular domain tyrosine kinase phosphorylation.
     (6). pSH1Si-AP2 plasmid-1 can also inhibit the AT1 receptor-mediated Ang II endocytosis after transient transfection of H9C2 cells, suggesting that endocytosis of Ang II and AT1 receptor mainly by clathrin-coated vesicles. At the same time, AP2 silence can suppress cell volume increase induced by Ang II, p38 phosphorylation and p65 nuclear transfer, also inhibit Ang II-induced cell cycle changes, suggesting that the ligand receptor complex endocytosis into the cells may also be involved in activation of downstream signal transduction pathways, and continue to play its biological role.
     Ang II and AT1 receptors, through activation of MAPK and NF-κB signal transduction pathways, affect the cell cycle changes, and promote cell hypertrophy and hyperplasia; PAO and AP2 RNAi inhibited AT1 receptor-mediated Ang II endocytosis ,at the same time, affected the MAPK and NF-κB pathway activation and cell cycle changes, inhibited the AngII role in promoting cell hypertrophy and hyperplasia, indicating that AT1 receptor-mediated Ang II endocytosis process may also continue to activate downstream signal transduction pathways, involved in the formation of myocardial hypertrophy mechanism.
引文
[1] Hunt SA, Abraham WT, Chin MH, et al. American College of Cardiology; American Heart Association Task Force on Practice Guidelines; American College of Chest Physicians; International Society for Heart and Lung Transplantation; Heart Rhythm Society. ACC/AHA 2005 Guideline Update for the Diagnosis and Management of Chronic Heart Failure in the Adult: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (Writing Committee to Update the 2001 Guidelines for the Evaluation and Management of Heart Failure): developed in collaboration with the American College of Chest Physicians and the International Society for Heart and Lung Transplantation: endorsed by the Heart Rhythm Society[J]. Circulation, 2005, 112(12): e154-e235.
    [2]中华医学会心血管病学分会.2007年中国慢性心力衰竭诊断和治疗指南[J].中华心血管病杂志, 2007, 35(12): 1076-1096.
    [3] Colucci WS. Molecular and cellular mechanisms of myocardial failure[J]. Am J Cardiol, 1997, 80(11A): 15L-25L.
    [4] Levy D, Garrison RJ, Savage DD, et al. Prognostic implications of echocardiographically determined left ventricular mass in the Framingham Heart Study[J]. N Engl J Med, 1990, 322:1561–1566.
    [5] Baker KM, Aceto JF. AngiotensinⅡetimulation of protein synthesis and cell growth in chick heart cellls[J]. Am J Physiol, 1990, 259(2 Pt 2): H610-8.
    [6] Dostal DE, Baker KM. Angiotensin II stimulation of left ventricular hypertrophy in adult rat heart. Mediation by the AT1 receptor[J]. Am J Hypertens, 1992, 5(5 Pt 1): 276-80.
    [7] Obaya AJ, Sedivy JM. Regulation of cyclin-CDK activity in mammalian cells[J]. Cell Mol life Sci, 2002, 59:126.
    [8] Morgan DO. Principles of CDK regulation[J]. Nature, 1995, 374(6518):131-4.
    [9] Preisig P A, Franch H A. Renal epithelial cell hyperplasia and hypertrophy[J].Semin Nephrol, 1995, 15: 327–340.
    [10] Meikrantz W Schegal R. Apoptosis and the cell cycle[J]. J Cell Biochem, 1995, 58: 160–174.
    [11] Kim S, Iwao H. Molecular and cellular mechanisms of angiotensin II-mediated cardiovascular and renal disease[J]. Pharmacol Rev, 2000, 52(1): 11-34.
    [12] Griendling KK, Lasségue B, Alexander RW. Angiotensin receptors and their therapeutic implications[J]. Annu Rev Pharmacol Toxicol, 1996, 36: 281–306.
    [13] Sp?t A, Enyedi P, Hajnóczky, et al. Generation and role of calcium signal in adrenal glomerulosa cells[J]. Exp Physiol, 1991, 76: 859–85.
    [14] Goodfriend TL, Elliott ME, Catt KJ. Drug therapy– angiotensin receptors and their antagonists[J]. New Engl J Med, 1996, 334: 1649–54.
    [15] Johnston CI. Angiotensin receptor antagonists: focus on losartan[J]. Lancet, 1995, 346: 1403–7.
    [16] Paul M, Poyan MA, Kreutz R. Physiology of local reninangiotensin systems[J]. Physiol Rev, 2006, 86: 747-803.
    [17] Pinto YM, Buikema H, van Gilst WH, et al. Cardiovascular endorgan damage in Ren-2 transgenic rat compared to spontaneously hypertensive rats[J]. J Mol Med, 1997, 75: 371–377.
    [18] De Gasparo M, Catt KJ, Inagami T, et al. International Union of Pharmacology. XXIII. The angiotensin II receptors[J]. Pharmacol Rev, 2000, 52: 415–472.
    [19] Zhuo JL, Li XC. Novel roles of intracrine angiotensin II and signalling mechanisms in kidney cells[J]. J Renin Angiotensin Aldosterone Syst, 2007, 8(1): 23-33.
    [20] Gilmore TD. Introduction to NF-κB: players, pathways, perspectives[J]. Oncogene, 2006, 25: 6680–6684.
    [21] Hoffmann A, Levchenko A, Scott ML, et al. The IκB-NF-κB signaling module: temporal control and selective gene activation[J]. Science, 2002, 298: 1241–1245.
    [22] Li XC, Zhuo JL.Nuclear factor-kappaB as a hormonal intracellular signaling molecule: focus on angiotensin II-induced cardiovascular and renal injury[J].Curr Opin Nephrol Hypertens, 2008, 17(1): 37-43.
    [23] Hunyady L, Balla T, Catt KJ. The ligand binding site of the angiotensin AT1 receptor[J]. Trends Pharmacol Sci, 1996, 17: 135–40.
    [24] JIA L ZHUO, OSCAR A CARRETERO, XIAO C LI. Effects of AT1 Receptor-Mediated Endocytosis of Extracellular Ang II on Activation of Nuclear Factor-κB in Proximal Tubule Cells[J]. Ann N Y Acad Sci, 2006, 1091: 336–345.
    [25] Berk BC. Angiotensin II signal transduction in vascular smooth muscle: pathways activated by specific tyrosine kinases[J]. J Am Soc Nephrol, 1999, 10: S62–8.
    [26] Aohi H, Izumo S, Sadoshima J. Angiotensin II activates RhoA incardiac myocytes: a critical role of RhoA in angiotensin II-induced premyofibril formation[J]. Circ Res, 1998, 82:666–76.
    [27] Boulay G, Chrétien L, Richard DE, et al. Short-term desensitization of the angiotensin II receptor of bovine adrenal glomerulosa cells corresponds to a shift from a high to a low affinity state[J]. Endocrinology, 1994, 135: 2130–6.
    [28] Tang H, Guo DF, Porter JP, et al. Role of cytoplasmic tail of the type 1A angiotensin II receptor in agonistand phorbol ester-induced desensitization[J]. Circ Res, 1998, 82: 523–31.
    [29] Thomas WG. Regulation of angiotensin II type 1 (AT1) receptor function[J]. Regul Pept, 1999, 79: 9–23.
    [30] Nouet S, Nahmias C. Signal transduction from the angiotensin II AT2 receptor[J]. Trends Endocrinol Metab, 2000, 11:1–6.
    [31] Utsunomiya H, Nakamura M, Kakudo K, et al. AngiotensinⅡAT2 receptor localization in cardiovascular tissues by its antibody developed in AT2 gene-deleted mice[J]. 2005; 126(3): 155-161
    [32] Widdop RE, Jones ES, Hannan RE, et al. Angiotensin AT2 receptors: cardiovascular hope or hype[J]? J Mol Cell Cardiol, 2003, 140(5): 809-24.
    [33] Thomas WG, Thekkumkara TJ, Baker KM. Molecular mechanisms of angiotensin II (AT1A) receptor endocytosis[J]. Clin Exp Pharmacol Physiol, 1996,Suppl. 3: S74–80.
    [34] Carey RM. Update on the role of the AT2 receptor[J]. Curr Opin Nephrol Hypertens, 2005, 14(1):67-71.
    [35] Pucell AG, Hodges JC, Sen I, et al. Biochemical properties of the ovarian granulosa cell type 2-angiotensin II receptor[J]. Endocrinology, 1991, 128: 1947–59.
    [36] Hunyady L, Bor M, Balla T, et al. Identification of a cytoplasmic Ser–Thr–Leu motif that determines agonist-induced internalization of the AT1 angiotensin receptor[J]. J Biol Chem, 1994, 269: 31378–82.
    [37] Hein L, Meinel L, Pratt RE, et al. Intracellular trafficking of angiotensin II and its AT1 and AT2 receptors: evidence for selective sorting of receptor and ligand[J]. Mol Endocrinol, 1997, 11: 1266–77.
    [38] Trowbridge IS, Collawn JF, Hopkins CR. Signal-dependent membrane protein trafficking in the endocytic pathway[J]. Annu Rev Cell Biol, 1993, 9: 129–61.
    [39] B?hm SK, Grady EF, Bunnett NW. Regulatory mechanisms that modulate signalling by G-protein-coupled receptors[J]. Biochem J, 1997, 322: 1–18.
    [40] Mukherjee S, Ghosh RN, Maxfield FR. Endocytosis[J]. Physiol Rev, 1997, 77: 759–803.
    [41] Anderson RGW. Caveolae: where incoming and outgoing messengers meet[J]. Proc Natl Acad Sci USA, 1993, 90:10909–13.
    [42] Montesano R, Orci L, Robert A, et al. Non-coated membrane invaginations are involved in binding and internalization of cholera and tetanus toxins[J]. Nature, 1982, 296:651–3.
    [43] Schmid SL. Cladmncnmed vesicle formation and protein sorting;an integrated process[J]. An- Rev Biochem, 1997, 66:511-548.
    [44] Pearse BMF, Robinson MS. Clathrin, adaptors, and sorting[J]. Annu Rev Cell Biol, 1990, 6: 151–71.
    [45] Marsh M, McMahon HT. The structural era of endocytosis[J]. Science, 1999, 285: 215–20.
    [46] Hunyady L, Merelli F, Baukal AJ, et al. Agonist- induced endocytosis and signalgeneration in adrenal glomerulosa cells. A potential mechanism for receptor-operated calcium entry[J]. J Biol Chem, 1991, 266:2783–8.
    [47] Ferguson SSG, Zhang J, Barak LS, et al. G-protein-coupled receptor kinases and arrestins: regulators of G-protein-coupled receptor sequestration[J]. Biochem Soc Trans, 1996, 24:953–9.
    [48] Cao TT, Mays RW, Von Zastrow M. Regulated endocytosis of G-protein- coupled receptors by a bichemically and functionally distinct subpopulation of clathrin-coated pits[J]. J Biol Chem, 1998, 273:24592–602.
    [49] Ferguson SSG, Zhang J, Barak LS, et al. Pleiotropic role for GRKs and b-arrestins in receptor regulation[J]. News Physiol Sci, 1997, 12: 145–52.
    [50] Bünemann M, Lee KB, Pals-Rylaarsdam R, et al. Desensitization of G-protein- coupled receptors in the cardiovascular system[J]. Annu Rev Physiol, 1999, 61: 169–92.
    [51] Claing A, Perry SJ, Achiriloaie M, et al. Multiple endocytic pathways of G proteincoupled receptors delineated by GIT1 sensitivity[J]. Proc Natl Acad Sci USA, 2000, 97: 1119–24.
    [52] Goodman Jr. OB, Krupnick JG, Santini F, et al. b-Arrestin acts as a clathrin adaptor in endocytosis of theα2-adrenergic receptor[J]. Nature, 1996, 383: 447–50.
    [53] Ferguson SSG, Downey III WE, Colapietro AM, et al. Role of b-arrestin in mediating agonistpromoted G protein-coupled receptor internalization[J]. Science, 1996, 271: 363–6.
    [54] Parton RG. Caveolae and caveolins[J]. Curr Opin Cell Biol, 1996, 8: 542–8.
    [55] Anderson RG. The caveolae membrane system[J]. Annu Rev Biochem, 1998, 67: 199–225.
    [56] Henley JR, Krueger EW, Oswald BJ, et al. Dynaminmediated internalization of caveolae[J]. J Cell Biol, 1998, 141: 85–99.
    [57] Oh P, McIntosh DP, Schnitzer JE. Dynamin at the neck of caveolae mediates their budding to form transport vesicles by GTP-driven fission from the plasma membrane of endothelium[J]. J Cell Biol, 1998, 141: 101–14.
    [58] Raposo G, Andre C, Benedetti EL, et al. Redistribution of muscarinic acetylcholine-receptors on humanfibroblasts induced by regulatory ligands[J]. Biol Cell, 1987, 60: 117–24.
    [59] Raposo G, Dunia I, Delavierklutchko C, et al. Internalization of beta-adrenergic -receptor in A431 cells involves non-coated vesicles[J]. Eur J Cell Biol, 1989, 50:340–52.
    [60] Richardson JB, Beaulnes A. The cellular site of action of angiotensin[J]. Cell Biol, 1971, 51: 419–32.
    [61] Robertson Jr. AL, Khairallah PA. Angiotensin II: rapid localization in nuclei of smooth and cardiac muscle[J]. Science, 1971, 172: 1138–9.
    [62] Bianchi C, Gutkowska J, De Le′an A, et al. Fate of [125I]angiotensin II in adrenal zona glomerulosa cells[J]. Endocrinology 1986, 118: 2605–7.
    [63] Bianchi C, Gutkowska J, Charbonneau C, et al. Internalization and lysosomal associationof [125I]angiotensin II in norepinephrine-containing cells of the rat adrenal medulla[J]. Endocrinology, 1986; 119: 1873–5.
    [64] Anderson KM, Murahashi T, Dostal DE, et al. Morphological and biochemical analysis of angiotensin II internalization in cultured rat aortic smooth muscle cells[J]. Am J Physiol Cell Physiol, 1993, 264: C179–88.
    [65] Richard DE, Chrétien L, Caron M, et al. Stimulation of the angiotensin II type I receptor on bovine adrenal glomerulosa cells activates a temperature-sensitive internalization- recycling pathway[J]. Mol Cell Endocrinol, 1997, 129: 209–18.
    [66] Griendling KK, Delafontaine P, Rittenhouse SE, et al. MA, Alexander RW. Correlation of receptor sequestration with sustained diacylglycerol accumulation in angiotensin II-stimulated cultured vascular smooth muscle cells[J]. J Biol Chem, 1987, 262: 14555–62.
    [67] Becker BN, Cheng H-F, Harris RC. Apical ANG II-stimulated PLA2 activity and Na+ flux: a potential role for Ca 2+ -independent PLA2 [J]. Am J Physiol, 1997, 273: F554–62.
    [68] Lázari MDM, Porto CS, Freymüller E, et al. Receptor-mediated endocytosis of angiotensin II in rat myometrial cells[J]. Biochem Pharmacol, 1997, 54:399–408.
    [69] Schelling JR, Hanson AS, Marzec R, et al. Cytoskeleton-dependent endocytosis is required for apical type 1 angiotensin II receptor-mediated phospholipase C activation in cultured rat proximal tubule cells[J]. J Clin Invest, 1992, 90: 2472–80.
    [70] Becker BN, Cheng HF, Burns KD, et al. Polarized rabbit type 1 angiotensin II receptors manifest differential rates of endocytosis and recycling[J]. Am J Physiol Cell Physiol, 1995, 269: C1048–56.
    [71] Becker BN, Kondo S, Chen J-K, et al. Tyrosine kinase inhibition affects type 1 angiotensin II receptor internalization[J]. J Recept Signal Transduct Res, 1999, 19: 975–93.
    [72] Harris RC. Potential mechanisms and physiologic actions of intracellular angiotensin II[J]. Am J Med Sci, 1999, 318: 374–9.
    [73] Conchon S, Monnot C, Teutsch B, et al. Internaliza tion of the rat AT1a and AT1b receptors: pharmacological and functional requirements[J]. FEBS Lett, 1994, 349: 365–70.
    [74] Chaki S, Guo D-F, Yamano Y, et al. Role of carboxyl tail of the rat angiotensin II type 1A receptor in agonist-induced internalization of the receptor[J]. Kidney Int, 1994, 46: 1492–5.
    [75] Hunyady L, Tian Y, Sandberg K, et al. Divergent conformational requirements for angiotensin II receptor internalization and signaling[J]. Kidney Int, 1994, 46: 1496–8.
    [76] Hunyady L, Baukal AJ, Balla T, et al. Independence of type 1 angiotensin II receptor endocytosis from G protein coupling and signal stransduction[J]. J Biol Chem, 1994, 269: 24798–804.
    [77] Thomas WG, Thekkumkara TJ, Motel TJ, et al. Stable expression of a truncated AT 1A receptor in CHO-K1 cells. The carboxyl-terminal region directs agonist-induced internalization but not receptor signaling or desensitization[J]. J Biol Chem, 1995, 270: 207–13.
    [78] Hunyady L, Bor M, Balla T, et al. Critical role of a conserved intramembranetyrosine residue in angiotensin II receptor activation[J]. J Biol Chem, 1995, 270: 9702–5.
    [79] Hunyady L, Bor M, Baukal AJ, et al. A conserved NPLFY sequence contributes to agonist binding and signal transduction but is not an internalization signal for the type 1 angiotensin II receptor[J]. J Biol Chem, 1995, 270: 16602–9.
    [80] Thomas WG, Baker KM, Motel TJ, et al. Angiotensin II receptor endocytosis involves two distinct regions of the cyto plasmic tail. A role for residues on the hydrophobic face of a putative amphipathic helix[J]. J Biol Chem ,1995, 270: 22153–9.
    [81] Kapas S, Hinson JP, Puddefoot JR, et al. Internaliza tion of the type I angiotensin II receptor (AT1) is required for protein kinase C activation but not for inositol trisphosphate release in the angiotensin II stimulated rat adrenal zona glomerulosa cell[J]. Biochem Biophys Res Commun, 1994, 204: 1292–8.
    [82] Hunyady L, Zhang M, Jagadeesh G, et al. Dependence of agonist activation on a conserved apolar residue in the third intracellular loop of the AT1 angiotensin receptor[J]. Proc Natl Acad Sci USA, 1996, 93: 10040–5.
    [83] Conchon S, Peltier N, Corvol P, et al. A noninternalized nondesensitized truncated AT1A receptor transduces an amplified ANG II signal[J]. Am J Physiol Endocrinol Metab, 1998, 274: E336–45.
    [84] Heuser JE, Anderson RG. Hypertonic media inhibit receptor-mediated endocytosis by blocking clathrin-coated pit formation[J]. J Cell Biol, 1989, 108: 389–400.
    [85] Hunyady L, Gáborik Z, Mihalik B, et al. Dynamin dependent mechanism of internalization of the AT1a angiotensin receptor (abstract) [J]. Mol Biol Cell (Suppl), 1999, 10: 316a.
    [86] Chun MY, Liyanage UK, Lisanti MP, et al. Signal-transduction of a G-protein-coupled receptor in caveolae– colocalization of endothelin and its receptor with caveolin[J]. Proc Natl Acad Sci USA, 1994, 91: 11728–32.
    [87] Haasemann M, Cartaud J, Muller-Esterl W, et al. Agonistinduced redistribution of bradykinin B2 receptor in caveolae[J]. J Cell Sci, 1998, 111: 917–28.
    [88] Feron O, Smith TW, Michel T, et al. Dynamic targeting of the agonist-stimulated m2 muscarinic acetylcholine receptor to caveolae in cardiac myocytes[J]. J Biol Chem, 1997, 272: 17744–8.
    [89] Ishizaka N, Griendling KK, Lassegue B, et al. Angiotensin II type 1 receptor: relationship with caveolae and caveolin after initial agonist stimulation[J]. Hypertension, 1998, 32: 459–66.
    [90] Lasley RD, Narayan P, Uittenbogaard A, et al. Activated cardiac adenosine A(1) receptors translocate out of caveolae[J]. J Biol Chem, 2000, 275: 4417–21.
    [91] Okamoto Y, Ninomiya H, Miwa S, et al. Cholesterol oxidation switches the internalization pathway of endothelin receptor type A from caveolae to clathrin-coated pits in Chinese hamster ovary cells[J]. J Biol Chem, 2000, 275: 6439–46.
    [92] Pike LJ, Casey L. Localization and turnover of phosphatidylinositol 4,5-bisphosphate in caveolin-enriched membrane domains[J]. J Biol Chem, 1996, 271: 26453–6.
    [93] Zhang J, Ferguson SG, Barak LS, et al. Dynamin andβ-arrestin reveal distinct mechanisms for G protein-coupled receptor internalization[J]. J Biol Chem, 1996, 271: 18302–5.
    [94] Crozat A, Penhoat A, Saez JM. Processing of angiotensin II (A-II) and (Sar1, Ala8)A-II by cultured bovine adrenocortical cells[J]. Endocrinology, 1986, 118 :2312–8.
    [95] Bihoreau C, Monnot C, Davies E, et al. Mutation of Asp74 of the rat angiotensin II receptor confers changes in antagonist affinities and abolishes G-protein coupling[J]. Proc Natl Acad Sci USA, 1993, 90: 5133–7.
    [96] Balmforth AJ, Lee AJ, Bajaj BPS, et al. Functional domains of the C-terminus of the rat angioten sin AT1A receptor[J]. Eur J Pharmacol Mol Pharmacol, 1995, 291: 135–41.
    [97] Chen W-J, Goldstein JL, Brown MS. NPXY, a sequence often found in cytoplasmic tails, is required for coated pit-mediated internaliza tion of the low density lipoprotein receptor[J]. J Biol Chem, 1990, 265: 3116–23.
    [98] SCHMID S L. Clathrin-coated vesicle formation and protein sorting: an integrated process[J]. Annual Review Biochemistry, 1997, 66 : 511-548.
    [99] Haucke V, De Camilli P. AP-2 recmitment to synaptotagmin stimulated by tyrosine-based endocytic motifs[J]. Science, 1999, 285: 1268-1271.
    [00] KIRCHHAUSEN T. Clathrin adaptors really adapt [J]. Cell, 2002, 109: 413-416.
    [101] COLLINS B M, McCOY A J, KENT H M, et al . Molecular architecture and functional model of the endocytic AP2 complex [J ] . Cell, 2002, 109: 523-535.
    [102] Owen PJ, Ecans PR. A structural explanation for the recognition of tyrosine-based endocytotic signals [J]. Science, 1998, 282: 1327-1332.
    [103] Chen H, Fre S, Slepnev VI, et al. Epsin is an EH-domain-binding protein implicated in clathrinmediated endocytosis[J]. Nature, 1998, 394: 793–7.
    [104] Nesterov A, Carter RE, Sorkina T, et al. Inhibition of the receptor-binding function of clathrin adaptor protein AP-2 by dominant-negative mutantσ2 subunit and its effects on endoagonist cytosis[J]. EMBO J, 1999, 18: 2489–99.
    [105] Dell’Angelica EC, Klumperman J, Stoorvogel W, et al. Association of the AP-3 adaptor complex with clathrin[J]. Science, 1998, 280: 431–4. 106. Pearse BM, Smith CJ, Owen DJ. Clathrin coat construction in endocytosis[J]. Curr Opin Struct Biol, 2000, 10 (2): 220–8.
    [107] Laporte SA, Oakley RH, Zhang J, et al. The beta2-adrenergic receptor/beta-arrestin complex recruits the clathrin adaptor AP-2 during endocytosis[J]. Proc Natl Acad Sci USA, 1999, 96: 3712–7.
    [108] Zhang J, Barak LS, Anborgh PH, et al. Cellular trafficking of G protein-coupled receptor/beta-arrestin endocytic complexes[J]. J Biol Chem, 1999, 274: 10999–11006.
    [109] Ullian ME, Linas SL. Role of receptor cycling in the regulation of angiotensin II surface receptor number and angiotensin II uptake in rat vascular smooth muscle cells[J]. J Clin Invest, 1989, 84: 840–6.
    [110] Krupnick JG, Benovic JL. The role of receptor kinases and arrestins in G protein-coupled receptor regulation[J]. Annu Rev Pharmacol Toxicol, 1998, 38: 289–319.
    [111] Lefkowitz RJ. G protein-coupled receptors. III. New roles for receptor kinases and beta-arrestins in receptor signaling and desensitization[J]. J Biol Chem, 1998, 273: 18677–80.
    [112] Schelling JR, Linas SL. Angiotensin II-dependent proximal tubule sodium transport requires receptor-mediated endocytosis[J]. Am J Physiol Cell Physiol, 1994, 266: C669–75.
    [113] Vinson GP, Ho MM, Puddefoot JR, et al. The relationship between the adrenal tissue renin–angiotensin system, internalization of the type I angiotensin II receptor (AT1) and angiotensin II function in the rat adrenal zona glomerulosa cell[J]. Adv Exp Med Biol, 1995, 377: 319–29.
    [114] Vinson GP, Ho MM, Puddefoot JR, et al. Internalisation of the type I angiotensin II receptor (AT1) and angiotensin II function in the rat adrenal zona glomerulosa cell[J]. Endocr Res, 1995, 21: 211–7.
    [115] Smith RD, Hunyady L, Olivares-Reyes JA, et al. Agonist-induced phosphorylation of the angiotensin AT1a receptor is localized to a serine / threonine-rich region of its cytoplasmic tail[J]. Mol Pharmacol, 1998, 54: 935–41.
    [116] Gáborik Z, Mihalik B, Jayadev S, et al. Requirement of membrane-proximal amino acids in the carboxyl-terminal tail for expression of the rat AT1a angiotensin receptor[J]. FEBS Lett, 1998, 428: 147–51.
    [117] Karnik SS, Husain A, Graham RM. Molecular determinants of peptide and non-peptide binding to the AT1 receptor[J]. Clin Exp Pharmacol Physiol, 1997, Suppl. 3: S58–66.
    [118] Bevan AP, Drake PG, Bergeron JJM, et al. Intracellular signal transduction: the role of endosomes[J]. Trends Endocrinol Metab, 1998, 7: 13–21.
    [119] Daaka Y, Luttrell LM, Ahn S, et al. Essential role for G protein-coupled receptor endocytosis in the activation of mitogen-activated protein kinase[J]. J Biol Chem, 1998; 273, 685–8.
    [120] Cheng ZJ, Zhao J, Sun Y, et al. Beta-arrestin differentially regulates the chemokine receptor CXCR4-mediated signaling and receptor internalization, andthis implicates multiple interaction sites between beta-arrestin and CXCR4[J]. J Biol Chem, 2000, 275: 2479–85.
    [121] Ignatova EG, Belcheva MM, Bohn LM, et al. Requirement of receptor internalization for opioid stimulation of mitogen-activated protein kinase: biochemical and immunofluorescence confocal microscopic evidence[J]. J Neurosci, 1999, 19: 56–63.
    [122] Schramm NL, Limbird LE. Stimulation of mitogen-activated protein kinase by G protein-coupled alpha(2)-adrenergic receptors does not require agonist-elicited endocytosis[J]. J Biol Chem, 1999, 274: 24935–40.
    [123] DeGraff JL, Gagnon AW, Benovic JL, et al. Role of arrestins in endocytosis and signaling of alpha2-adrenergic receptor subtypes[J]. J Biol Chem, 1999, 274: 11253–9.
    [124] Tang H, Nishishita T, Fitzgerald T, et al. Inhibition of AT1 receptor internalization by concanavalin A blocks angiotensin II-induced ERK activation in vascular smooth muscle cells[J]. J Biol Chem, 2000, 275: 13420–6.
    [125] Hausdorff WP, Sung J, Caron MG, et al. Recent molecular analyses ofβ-adrenergic receptor phosphorylation, sequestration and down regulation[J]. Asian Pac J Pharm, 1992, 7: 149–58.
    [126] Jockers R, Angers S, Da Silva A, et al. Beta(2)-adrenergic receptor down- regulation. Evidence for a pathway that does not require endocytosis[J]. J Biol Chem, 1999, 274: 28900–8.
    [127] Quan A, Baum M. Endogenous production of angiotensin II modulates rat proximal tubule transport[J]. J Clin Invest, 1996, 97: 2878–82.
    [128] Zou L-X, Imig JD,Von Thun AM, et al. Receptor-mediated intrarenal angiotensin II augmentation in angiotensin II-infused rats[J]. Hypertension, 1996, 28: 669–77.
    [129] Zou L-X, Hymel A, Imig JD, et al. Renal accumulation of circulating angiotensin II in angiotensin II-infused rats[J]. Hypertension, 1996, 27: 658–62.
    [130] Van Kats JP, De Lannoy LM, Danser AHJ, et al. Angiotensin II type 1 (AT1) receptor-mediated accumulation of angiotensin II in tissues and its intracellularhalf-life in vivo[J]. Hypertension, 1997, 30: 42–9.
    [131] Mercure C, Ramla D, Garcia R, et al. Evidence for intracellular generation of angiotensin II in rat juxtaglomerular cells[J]. FEBS Lett, 1998, 422: 395–9.
    [132] Urata H, Khosla MC, Bumpus F, et al. Evidence for extracellular, but not intracellular, generation of angiotensin II in the rat adrenal zona glomerulosa[J]. Proc Natl Acad Sci USA, 1988, 85: 8251–5.
    [133] Haller H, Lindschau C, Erdmann B, et al. Effects of intracellular angiotensin II in vascular smooth muscle cells[J]. Circ Res, 1996, 79: 765–72.
    [134] Sen I, Bull HG, Soffer RL. Isolation of an angiotensin II-binding protein from liver[J]. Proc Natl Acad Sci USA, 1984, 81: 1679–83.
    [135] Kiron MAR, Soffer RL. Purification and properties of a soluble angiotensin II-binding protein from rabbit liver[J]. J Biol Chem, 1989, 264: 4138–42.
    [136] Sugiura N, Hagiwara H, Hirose S. Molecular cloning of porcine soluble angiotensin-binding protein[J]. J Biol Chem, 1992, 267: 18067–72.
    [137] McKie N, Dando PM, Rawlings ND, et al. Thimet oligopeptidase: similarity to‘soluble angiotensin II-binding protein’and some corrections to the published amino acid sequence of the rat testis enzyme[J]. Biochem J, 1993, 295: 57–60.
    [138] Tang S-S, Rogg H, Schumacher R, et al. Characterization of nuclear angiotensin II-binding sites in rat liver and comparison with plasma membrane receptors[J]. Endocrinology, 1992, 131: 374–80.
    [139] Lu D, Yang H, Shaw G, et al. Angiotensin II-induced nuclear targeting of the angiotensin type 1 (AT1) receptor in brain neurons[J]. Endocrinology, 1998, 139: 365–75.
    [140] Jiménez E, Vinson GP, Montiel M. Angiotensin II (AII)-binding sites in nuclei form rat liver: partial characterization of the mechanism of AII accumulation in nuclei[J]. J Endocrinol, 1994, 143: 449–53.
    [141] Re R, Parab M. Effect of angiotensin II on RNA synthesis by isolated nuclei[J]. Life Sci, 1984, 34: 647–51.
    [142] Eggena P, Zhu JH, Clegg K, et al. Nuclear angiotensin receptors induce transcription of renin and angiotensinogen mRNA[J]. Hypertension, 1993, 22:496–501.
    [143] JENTSCH T J,STEIN V,WEINREICH F,et al. Molecular structure and physiological function of chloride channels[J].Physiol Rev, 2002, 82: 503-568.
    [144] Duan DY, Liu LL, Bozeat N,et al. Functional role of anion channels in cardiac diseases. Acta Pharmacol Sin. 2005; 26(3):265-278.

© 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