腺相关病毒介导的内皮型一氧化氮合酶在高血压基因治疗中的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
高血压是一种常见病、多发病,如何有效控制血压已成为医学界普遍关注的焦点之一。本研究旨在观察腺相关病毒载体介导的内皮型一氧化氮合酶(eNOS)基因对自发性高血压大鼠(SHR)血压的影响及对心脏、肾脏等靶器官的保护作用,为临床高血压基因治疗奠定理论基础。本研究以自发性高血压大鼠为研究对象,通过尾静脉注射腺相关病毒介导的eNOS(AAV-eNOS)进行干预,结果发现AAV-eNOS在SHR体内至少可稳定表达4周,经AAV-eNOS治疗2周后,血压可明显降低,并可持续至4周后,除明显的降压作用外,可明显降低心脏及肾脏组织胶原含量,延缓或抑制纤维化进程;同时,免疫组化及原位杂交结果显示,AAV-eNOS治疗后可明显降低心脏及肾脏组织中AT1 mRNA及蛋白表达水平,提示AAV-eNOS基因治疗高血压可能与其抑制靶器官AT1的表达、从而阻断Ang II的缩血管作用有关;TUNEL检测结果显示AAV-eNOS治疗可明显抑制心肌及肾脏组织细胞的凋亡,从而延缓器官纤维化进程,保护靶器官功能。因此,我们考虑AAV-eNOS可能成为高血压治疗的一个新手段。
Hypertension is defined as the abnormal occurrence which means the arterial blood pressure is above the normal value. How to control the blood pressure effectively has been one of the facal points which generally attract the attention in medical science because the hypertension has been become a common and frequent encountered disease. The present investigation aimed to observe the influence of blood pressure and the protection of the target organs by the endothelial nitricoxide synthase (eNOS) in spontaneous hypertension rat (SHR), and do some contribution to theory base for the clinic gene therapy to hypertension.
     The SHR rats were divided into SHR group, AAV-LacZ group, and AAV-eNOS group, and choose WKY rats as negative control group (WKY group) which has normal blood pressure. The AAV-eNOS group were injected AAV-eNOS 200μl which contains 1×1012 eNOS copy through tail vein, AAV-LacZ group were injected AAV-LacZ 200μl which contains no eNOS gene copy, and the rats in WKY and SHR group were injected 0.1 mol·L-1 PBS solution 200μl. The blood pressure were observed once a week till the 4th week. All the rats were sacrificed at the end of 4th week, and the heart and right kidney were collected into the 4 % polyoxymethylene solution. The immunohistochemistry and in situ hybridyzation were applied to detect eNOS and AT1 protein and mRNA expression, TUNEL were also used to evaluate the cell apoptosis in the heart and kidney. We got these results from this research:
     1. After the treatment of AAV-eNOS, there was no distinguished difference among the body weight, heart weight, and left venturicular weight in each group; also no obvious difference was found in cardiac index, left-ventricular index in all the groups.
     2. Two weeks after the treatment of AAV-eNOS, the blood pressure of AAV-eNOS group had been decreased significantly (P<0.05) compared with SHR group; after 4 weeks, the blood pressure of AAV-eNOS group had been decreased to normal level, compared with SHR group, there is significant difference (P<0.05).
     3. In WKY treatment group, the cardiac muscle fibers line up in order, transverse striations is clear, there is a clear nucleus, and no cell swelling, a wealth capillary network and a small number of fibroblasts could be seen between the cardiac muscle. In the SHR group, fibroblast proliferation of cardiac muscle, fibrous tissue accrementing obviously, myocardial fibrosis slightly, after AAV-eNOS treatment, myocardial fibrosis mitigate significantly. In WKY group, no obvious abnormality was found in glomerulus of kidney, nephric tubule and kidney arteriole. While in SHR group, the glomerulus of kidney generate fibrous degeneration and hyalinization (glomerulus of kidney express homogeneous red dye whith no structure), volume minification, peripheral glomcrulus hypertrophy comply, the wall of glomerulus arteriole became thickening, renal tubular cell swelling, lumina stenosis, interstitial fibrosis manifest in the area of serious pathological changes. The AAV-eNOS treatment group, the structure of glomerulus and nephric tubule is generally normal, the endomembrane of renal arterioles wall is smooth, interstitial fibrosis proliferation decreased significantly.
     4. Immunohistochemistry results: In WKY group, eNOS express signal could be seen in the myocardial cell endochylema,while in SHR group the express of eNOS is lower than WKY group significantly. The expression of eNOS significantly increased in AAV-eNOS treated group.
     The expression of eNOS protein in kidney showed no significant differences in each group. The positive signals of AT1 expression could be seen in the myocardial cell endochylema while in SHR group, the expression of AT1 is lower significantly than WKY group (P<0.05). The expression of AT1 significantly decreased in AAV-eNOS treated group compared with SHR group (P<0.05); The positive signals of AT 1 expression could be seen in renal tubular cells in WKY group and was lower than SHR group significantly (P<0.05).
     5. The results of eNOS in situ hybridization showed that, there was basic expression of eNOS mRNA in myocardial tissue of WKY group; The expression of eNOS mRNA was increased significantly in AAV-eNOS treatment group compared with SHR group (P<0.05).
     There was basic expression of eNOS mRNA in kidney in WKY group; The expression of eNOS mRNA increased significantly in AAV-eNOS treatment group compared with SHR group (P<0.05).
     6.The results of AT1 in situ hybridization showed that, there were basic expression of AT1 mRNA both in myocardial and kidney tissue in WKY group. The expression of AT1 mRNA significantly increased in the myocardial and renal interstitial tissue of SHR group compared with the WKY group (P<0.05), at the end of the experiment, The expression of eNOS mRNA in myocardial and renal interstitial tissue of AAV-eNOS treatment group was significantly lower than SHR group (P<0.05).
     7.The results of TUNEL show that:Compared with WKY group, The expression of staining positive signal in myocardial and renal interstitial tissue of SHR group increased significantly (P<0.05), while decreased significantly in AAV-eNOS group(P<0.05).
     Conclusions:
     1. The recombinant adenovirus vector carrying eNOS gene can be integrated into heart and kidney tissues of SHR and could be stablely expressed.
     2. After AAV-eNOS induced into SHR, it can reduce blood pressure effectively and sustain for more than 2 weeks.
     3. AAV-eNOS gene therapy can delay or inhibit fibrosis of the target organs caused by hypertension, and inhibit myocardial reconstruction and renal interstitial fibrosis.
     4. AAV-eNOS gene therapy successfully treating in hypertension may be related to inhibition of expression of AT1 in target organs, thereby block the vasoconstriction effects of Ang II.
     5. AAV-eNOS gene therapy can inhibit the apoptosis of heart and kidney and protect target organs.
引文
[1] Israili ZH, Hall WD. Cough and angioneurotic edema associated with angiotensin II converting enzyme inhibitor therapy. A review of the literature and pathophysiology[J]. Ann Intern Med, 1992,117(3):234-242.
    [2] Phillips MI. Gene therapy for hypertension: sense and antisense strategies[J]. Expert Opin Biol Ther, 2001,1(4):655-662.
    [3] Zhao C, Wang P, Xiao X, et al. Gene therapy with human tissue kallikrein reduces hypertension and hypednsulinemia in fructose-induced hypertensive rats[J]. Hypertension, 2003,42(5):1026-1033.
    [4]陈改玲,党爱民,张宇清,等.中国汉族原发性高血压患者缓激肽B2受体和内皮型一氧化氮合酶基因多态性与血管紧张素转换酶抑制剂降压疗效的关系[J].中国临床康复, 2006,10(4):19-22.
    [5] Giles RV, Ruddell CJ, Spiller DG, et a1. Single base discrimination for ribonuclease H dependent antisense effects within intact human leukemia cells[J]. Nucleic Acids Res, 1995,23(6):954-961.
    [6] Phillips MI, Kimura B. Gene therapy for hypertension: antisense inhibition of the renin-angiotensin system[J]. Methods Mol Med, 2005, 108(1):363-379.
    [7] Lizanecz E, Pásztor ET, Mohácsi A, et al. Mistyping of angiotensinogenM235T alleles[J]. Hypertens Res, 2006,29(3):197-201.
    [8] Sugano M, Tsuchida K, Sawada S, et al. Reduction of plasma angiotensin II to normal levels by antisense oligodeoxynucleotides against liver angiotensinogen cannot completely attenuate vascular remodeling in spontaneously hypertensive rats[J]. Hypertens, 2000,18(6):725-731.
    [9] Kagiyama S, Varela A, Phillips MI, et al. Antisense inhibition of brain renin-angiotensin system decreased blood pressure in chronic 2-kidney, 1 clip hypertensive rats[J]. Hypertension, 2001,37(2Part2):371-375.
    [10] Martinez JA, O'Connor DT. Somatic cell gene therapy for a trait as complex as hypertension[J]? Clin Invest, 1995,95(4):1426.
    [11] Gyurko R, Wielbo D, Phillips M. Antisense inhibition of AT1 receptor mRNA and angiotensinogen mRNA in the brain of spontaneously hypertension rats reduces hypertension of neurogenic origin[J]. Regul Pepr, 1993,49(2):167-174.
    [12] Phillips M. Gene therapy for hypertension: the preclinial data[J]. Hypertension, 2001,38(3part2):543-548.
    [13] Kimura B, Mohuczy D, Tang X, et al. Attenuation of hypertension and heart hypertrophy by adeno-associated virus delivering angiotensinogen antisense[J]. Hypertension, 2001,37(2Part2):376-380.
    [14] Gyurko R, Tran D, Phillips M. Time course of hypertension by antisenseoligonucleotides targeted to AT1 angiotensin receptor mRNA in spontaneously hypertension rats[J]. Am J Hypertension, 1997,10 (5pt2):56s-62s.
    [15] Wang H, Reaves PY, Gardon ML, et al. Angiotensin I-converting enzyme antisense gene therapy causes permanent antihypertensive effects in the SHR[J]. Hypertension, 2000,35(1Pt2):202-208.
    [16] Wang H, Katovich MJ, Gelband CH, et al. Sustained inhibition of angiotensin I-converting enzyme (ACE) expression and long-term antihypertensive action by virally mediated delivery of ACE antisense cDNA[J]. Circ Res, 1999,85(7):614-622.
    [17] Gelband CH, Wang H, Gardon ML,et al. Angiotensin I-converting enzyme antisense prevents altered renal vascular reactivity, but not high blood pressure, in spontaneously hypertensive rats[J]. Hypertension, 2000,35 (1Pt2):209-213.
    [18] Gyurko R, Wielbo D, Phillips MI. Antisense inhibition of AT1 receptor mRNA and angiotensinogen mRNA in the brain of spontaneously hypertensive rats reduces hypertension of neurogenic origin[J]. Regul Pept, 1993,49(2):167-174.
    [19] Meng H, Wielbo D, Gyurko R, et al. Antisense oligonucleotide to AT1 receptor mRNA inhibits central angiotensin induced thirst andvasopressin[J]. Regul Pept, 1994,54(2-3):543-551.
    [20] Iyer SN, Lu D, Katovich MJ, et al. Chronic control of high blood pressure in the spontaneously hypertensive rat by delivery of angiotensin type 1 receptor antisense[J]. Proc Natl Acad Sci U S A, 1996,93(18):9960-9965.
    [21] Yoneda M, Sanada H, Yatabe J, et al. Differential effects of angiotensin II type-1 receptor antisense oligonucleotides on renal function in spontaneously hypertensive rats[J]. Hypertension, 2005,46(1):58-65.
    [22] Yamakawa H, Phillips MI, et al. Intracisternal administration of Angiotensin II AT1 receptor antisense oligodeoxynucleotides protects against cerebral ischemia in spontaneously hypertensive rats[J]. Regul Pept, 2003,111(1-3):117-122.
    [23] Liang Y, Lin S, Zhou Y, et al. Beta-1 adrenergic receptor antisense- oligodeoxynucleotides ameliorates left ventricular remodeling in 2-Kidney, 1-Clip rats[J]. Biomed Sci, 2007,14(1):155-164.
    [24] Zhang YC, Bui JD, Shen L, et al. Antisense inhibition ofβ(1)-adrenergic receptor mRNA in a single dose produces a profound and prolonged reduction in high blood pressure in spontaneously hypertensive rats[J]. Circulation, 2000,101(6):682-688.
    [25] Yatabe J, Sanada H, Midorikawa S, et al. Effects of decreased renal cortical expression of G protein-coupled receptor kinase 4 and angiotensintype 1 receptors in rats[J]. Hypertens Res, 2008,31(7):1455-1464.
    [26] Mocellin S, Provenzano M. RNA interference: learning gene knock-down from cell physiology[J]. Transl Med, 2004,2(1):39.
    [27] Wang X, Skelley L, Cade R, et al. AAV delivery of mineralocorticoid receptor shRNA prevents progression of cold-induced hypertension and attenuates renal damage[J]. Gene Ther, 2006,13(3):1097-103.
    [28] Modlinger P, Chabrashvili T, Pritomhinder S, et al. RNA si-lencing in vivo reveals role of p22 phoxin rat angiotensin slow pressor response[J]. Hypertension, 2006,47(1):238-244.
    [29] Liu D, Scholze A, Zhu Z, et al. Increased transient receptor potential channel TRPC3 expression in spontaneously hypertensive rats[J]. Hypertension, 2005,18(5):1503-1507.
    [30] Liu D, Scholze A, Zhu Z, et al. Transient receptor potential channels in essential hypertension[J]. Hypertension, 2006,24(4):1105-1114.
    [31] He J, Bian Y, Gao F, et al. RNA interference targeting the ACE gene reduced blood pressure and improved myocardial remodelling in SHRs[J]. Clin Sci (Lond), 2009,116(3):249-255.
    [32] Ishimitsu T, Ono H, Minami J, et al. Pathophysiologic and therapeutic implications of adrenomedullin in cardiovascular disorders[J]. Pharmacol Ther, 2006,111(3):909-927.
    [33] Wei X, Zhao C, Jiang J, et al. Adrenomedullin gene delivery alleviates hypertension and its secondary injuries of cardiovascular system[J]. Hum Gene Ther, 2005,16(1):372-380.
    [34] Lin KF, Chao J, Chao L. Atrial natriuretic peptide gene delivery reduces stroke. induced mortality rate in DaM salt.sensitive rats[J]. Hypertension, 1999,33(1Pt2):219.
    [35] Chao J, Kato K, Zhang JJ, et al. Human adrenomedullin gene delivery protects against cardiovascular remodeling and renal injury[J]. Peptide, 2001,22(11):1731-1737.
    [36] Kobayashi N, Nishikimi T, Horinaka S, et al. Effects of TCV-116 on expression of NOS and adrenomedullin in failing heart of Dahl salt-sensitive rats[J]. Atherosclerosis, 2001,156(2):255-265.
    [37] McMurtryM S, Archer SL, Altieri DC, et al. Gene therapy targeting survivin selectively induces pulmonary vascu|ar apoptosis and reverses pulmonary arterial hypertension[J]. Clin Invest, 2005,l15(6):1479-1491.
    [38] Kate L, Disbert T, Lorraine M, et al. Gene Therapy for cardiovascular disease[J]. Biomed Biotechnol, 2003(2):138-148.
    [39] Wang T, Li H, Zhao C, et al. Recombinant adeno-associated virus- mediated kallikrein gene therapy reduces hypertension and attenuates its cardiovascular injuries[J]. Gene Ther, 2004,11(17):1342-1350.
    [40] Yan JT, Wang T, Li J, et al. Recombinant adeno-associated virus-mediated human kallikrein gene therapy prevents high-salt diet-induced hypertension without effect on basal blood pressure[J]. Acta Pharmacol Sin, 2008,29(7):808-814.
    [41] Falcon BL, Stewart JM, Bourassa E, et al. Angiotensin II type 2 receptor gene transfer elidts cardioprotective effects in an angiotensin l1 infusion rat model of hypertension[J]. Physiol Genomics, 2004,19(3):255-261.
    [42] Katovich MJ, Gelband CH, Reaves P, et al. Reversal of hypertension by angiotensin II type 1 receptor antisense gene therapy in the adult SHR[J]. Physiol, 1999,277(3Pt2):H1260-H1264.
    [43] Metcalfe BL, Huentelman MJ, Parilak LD, et al. Prevention of cardiac hypertrophy by angiotensinⅡtype 2 receptor gene transfer[J]. Hypertension, 2004,43(4):1233-1238.
    [44] Falcon BL, Stewart JM, Bourassa E, et al. AngiotensinⅡtype-receptor gene transfer elicits cardioprotective effects in an an- giotensinⅡinfusion rat model of hypertension[J]. Physiol Ge-nomics, 2004,19(1):255-261.
    [45]李涛,梁红雁,卢果,等.皮下移植心钠素基因表达细胞胶囊对高血压大鼠的降血压效应[J].中华医学杂志, 2002,82(16):1086-1089.
    [46] Rees DD, Palmer RMJ, Moncada S. Role of endothelium-derived nitric oxide in the regulation of blood pressure[J]. Proc Natl Acad Sci USA,1989,86(6):3375-3378.
    [47] Lin K F, Chao L. Prolonged reduction of high blood pressure with human nitric oxide synthase gene delivery[J]. Hypertension, 1997,30(1):307.
    [48]宋铁山,李旭光.导入人内皮型一氧化氮合酶基因对高盐饮食大鼠的血压及肾脏功能的影响[J].中华高血压杂志, 2007,15(4):303-306.
    [49] Ohashi Y, Kawashima S, Hirata K, et al. Hypotension and reduced nitric oxide elicited vasorelaxation in transgenic mice overexpressing endothelial nitric oxide synthase[J]. Clin Invest, 1998,102(7):2061-2071.
    [50]赵春霞,汪培华,李宏伟,等.人组织型激肽释放酶基因降低果糖诱导高血压大鼠的血压并改善高胰岛素血症[J].中国分子心脏病学杂志, 2002,2(4):3-8.
    [51] Marie J, Karel H, Jan B, et a1. Association ofthe Glu298 Asp polymorphism in the endothelial nitric oxide synthase gene with essential hypertension resistant to conventional therapy[J]. Biochem Biophys Res Commun, 2001,284(2):426.
    [52] Alexander MY, Brosnan MJ, Carlene AH, et a1. Gene transfer of endothelial nitrix oxide synthase improves nitric oxide-dependent endothelial function in a hypertensive rat mode[J]. Cardiovasc Res, 1999, 43(3):789.
    [53] Harrap SB. Where are all the blood-pressure genes[J]. Lancet,2003,36(8):2149-2151.
    [54] Schillinger KJ, Tsai SY, Taffet GE, et a1. Regulatable atrial natriuretic peptide gene therapy for hypertension[J]. Proc Natl Acad Sci USA, 2005, 102:13789-13794.
    [55] Furchgott RF, Zawadzki JV. The obligatory role of endothelial cells in the relaxation of arterial smooth muscle by acetylcholine[J]. Nature, 1980, 288(5789):373-376.
    [56] Palmer RM, Ferrige AG, et al. Nitric oxide release accounts for the biological activity of endothelium-derived relaxing factor[J]. Nature, 1987, 327(6122):524-526.
    [57] Palmer RM, Rees DD, Ashton DS, et al. L-arginine is the physiological precursor for the formation of nitric oxide in endothelium- dependent relaxation[J]. Biochem Biophys Res Commun, 1988,153(3):1251-1256.
    [58] Palmer RM, Ashton DS, Moncada S. Vascular endothelial cells synthesize nitric oxide from L-arginine[J]. Nature, 1988,333(6174):664-666.
    [59] Vo PA, Lad B, Tomlinson JA, et al. Auto regulatory role of endothelium- derived nitric oxide (NO) on lipopolysaccha ride- induced vascular inducible NO synthase expression and function[J]. Biol Chem, 2005, 280(8):7236-7243.
    [60] Golino P, Cappelli-BigazziM, Ambrosio G, et al. Endothelium- derivedrelaxing factor modulates platelet aggregation in an vivomodel of recurrent platelet activation[J]. CircRes, 1992,71(6):1447-1456.
    [61] Raij L. Nitric oxide in the pathogenesis of cardiac disease[J]. Clin Hypertens (Greenwich), 2006,8(12 Suppl):S30-S39.
    [62] Xiao J, Pang PK. Does a general alteration in nitric oxide synthesis system occur in spontaneously hypertensive rats[J]? Physiol, 1994,266 (1):H272-278.
    [63] Morishita R, Gibbons GH, Eullison KE, et al. Evidence for direct local effect of angiotensin in vascaular hypertrophy in vivo gene transfer of angiotensin converting enzyme[J]. Clin Invest, 1994,94(3):978-984.
    [64] Loibl S, Buck A, Strank C, et al. The role of early expression of inducible nitric oxide synthase in human breast cancer[J]. EurJCancer, 2005,41 (2):265-271.
    [65]陈世新,施静,邹晓静,等.大鼠局灶性脑缺血再灌注损伤中iNOS在大脑皮层和海马的表达[J].中国组织化学与细胞化学杂志, 2005;14 (4):395-398.
    [66] Rodrigo J, Fernández AP, Serrano J, et al. The role of free radicals in cerebral hypoxia and ischemia[J]. Free Radic BiolMed, 2005,39 (1):26-50.
    [67] Myatt L, Cui X. Oxidative stress in the placenta[J]. Histochem CellBiol, 2004,122(4):369-382.
    [68] Freeman SL, MacNaughton WK. Nitric oxide inhibitable isoforms of adenylate cyclase mediate epithelial secretory dysfunction following exposure to ionizing radiation[J]. Gut, 2004,53(2):214-221.
    [69] Cengel A, Sahinarslan A. Nitric oxide and cardiovascular system[J]. Anadolu KardiyolDerg, 2006,6(4):364-368.
    [70] Scatena R, Bottoni P, Martorana GE, et al. Nitric oxide donor drugs: an update on pathophysiology and therapeutical potential[J]. ExpertOpin InvestDrug, 2005,14(7):835-846.
    [71] Cirino G. Nitric oxide releasing drugs: from bench to bedside[J]. Dig Liver Dis, 2003,35(2 Suppl):S2-S8.
    [72] Yetik-Anacak G, Catravas JD. Nitric oxide and the endothelium: history and impact on cardiovascular disease[J]. Vasc Pharmacol, 2006,45 (5):268-276.
    [73] Martelli A, Rapposelli S, Calderone V. NO-releasing hybrids of cardiovascular drugs[J]. CurMed Chem, 2006,13(6):609-625.
    [74] Levy RM, Prince JM, Billiar TR. Nitric oxide: a clinical primer[J]. Crit Care Med, 2005,33(12 Suppl):S492-495.
    [75] Dichtl W, Dulak J, FrickM, et al. HMG-CoA reductase inhibitors regulate inflammatory transcription factors in human, endothelial and vascular smoothmuscle cells[J]. Arterioscler Thromb Vasc Biol, 2003,23(1):58-63.
    [76]黄震华.血管紧张肽转换酶抑制剂在冠心病治疗中的应用[J].中国新药与临床杂志, 2004,23(1):43-46.
    [77] Chen AF, O'Brien T, Katusic ZS. Transfer and expression of recombinant nitric oxide synthase genes in the cardiovascular system[J]. Trends Pharmacol Sci, 1998,19(7):276-286
    [78] Zanetti M, d'Uscio LV, Kovesdi I, et al. In vivo gene transfer of inducible nitric oxide synthase to carotid arteries from hypercholesterolemic rabbits[J]. Stroke, 2003,34(5):1293-1298.
    [79] Sen S, Conroy S, Hynes SO, et al. Gene delivery to the vasculaturemediated by low-titre adeno-associated virus serotypes 1 and 5[J]. GeneMed, 2008,10(2):143-151.
    [80] Muller DND, Echend R, Mervaala EM, et al. NF-kappa B inhibition ameliorate angiotensin II induced inflammatory damage in rats[J]. Hypertension, 2000,35(1):193-201.
    [81] Gomez GD, Largo R, Tejera N, et al. Activation of NF-κB in tubular epithelial cells of ras with intense proteinuria: Role of AngiotensinⅡand Endothelin-1[J]. Hypertension, 2001,37(4):1171-1178.
    [82] St-Louis J, Sicotte B, Bédard S, et al. Blockade of angiotensin receptor subtypes in arcuate uterine artery of pregnant and postpartum rats[J]. Hypertension, 2001,38(5):1017-1023.
    [83] Lehtonen JYA, Horiuchi M, Daviet L, et al. Activation of the denovo biosynthesis of sphingolipids mediates angiotensinⅡtype 2 receptor- induced apoptosis[J]. Biol Chem, 1999,274(4):16901-16906.
    [84] Shanley PF. The pathology of chronic renal ischemia[J]. Semin Nephrol 1996,16(1):21-32.
    [85] Lo M, Liu KL, Lantelme P, et al. Subtype of angiotensinⅡreceptors controls pressure-natriuesis in rats[J]. Clin Invest, 1995,95(3): 1394-1398.
    [86] Madrid MI, Garcia SM, Tornel J, et al. Effect of interaction between nitric oxide and angiotensinⅡon pressure diuresis[J]. Physiol, 1997,273 (5):R1676-R1680.
    [87] Phillips MI. Gene therapy for hypertension: antisense inhibition with adeno-associated viral vector delivery targeting angiotensin II type 1-receptor messenger ribonucleic acid[J]. Cardiol, 1998,82(10A): 60S-62S.
    [88] Joseph E, Rabinowitz A, Samulski RJ. Building a better vector: the manipulation of AAV virions[J]. Virology, 2000,278(1):301-308.
    [89] Ruiz OM, RuperezM, Esteban V, et al. Angiotensin II: a key factor in the inflammatory and fibrotic response in kidney diseases[J]. Nephrol Dial Transplant, 2006,21(l):16-20.
    [90] Fukuda S, Kaga S, Zhan L, et al. Resveratrola meliorates myocardialdamage by inducing vascular endothelial growth factor angiogenesis and tyrosine kinase receptor Flk-1[J]. Cell Biochem Biop, 2006,44(1):43-49.
    [91] Uemura R, Xu M, Ahmad N, et al. Bone marrow stem cells prevent left ventricular remodeling of ischemic heart through paracrine signaling[J]. Circ Res, 2006,98(6):1414-1421.
    [92] Johnston Cl, Risvanis J.Preclinieal Pharmacology of angiotensin Il receptor antagonists: Update and outstanding issues[J]. Hypertension, 1997,10(122):306-310.
    [93] Unger T, Culman J, Gohlke P. Angiotensin II receptor blockade and endorgan protection: pharmacological rationale and evidence[J]. Hypertension, 17:3-9.
    [94] Kubo T, Hagiwara Y. Enhanced activity of angiotensin II sensitive neuronsin the anterior hypothalamiearea of spontaneously hypertensive rats[J]. Brain Researeh, 2004,10(20):140-146.
    [95] HametP, Richard L, Dam TV, et al. Apoptosis in target organs of hypertension[J]. Hypertension, 1995,26(3):642-648
    [96]龙超良,汪海,肖文彬.高血压心血管重构及其药物治疗[J].军事医学科学院院刊, 1997,21:63-67.
    [97]张艳.高血压靶器官的细胞凋亡[J].国外医学内科学分册, 1997,24: 429-431.
    [98] Li Z, Bing OH, Long X, et al. Increased cardiomyocyte apoptosis during the transition to heart failure in the spontaneously hypertensive rat[J]. Physiol, 1997,272(5 Pt 2):H2313-H2319.
    [99] Moreau P, Tea BS, Dam TV, et al. Altered balance between cell replication and apoptosis in hearts and kidneys of newborn SHR[J]. Hypertension, 1997,30(3 Pt 2):720-724.
    [100] Hamet P, Moreau P, Dam TV, et al. The time window of apoptosis: a new component in the therapeutic strategy for cardiovascular remodeling[J]. Hypertension, 1996,14(Suppl 5):S65-S70.
    [101] Toru A, Nobukazu I.Different effets of angiotensin II and catecholamine on renal cell apoptosis and proliferation in rats[J]. Kidney Int. 2001:59(2): 645-653.
    [102] Pierre M, Tea BS. Altered balance between cell replication and apoptosis in hearts and kidnes of newborn SHR[J]. Hypertesion, 1997,30(Part2): 720-724.

© 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