间尼索地平对野百合碱诱导的肺动脉高压的防治作用及机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
肺动脉高压(PAH)是以肺动脉压力升高以及血管痉挛、内膜增生和重构为主要特征的一种疾病。其发病机制尚未完全明确,目前观点认为,肺动脉高压形成的关键在于肺血管收缩和重构。肺动脉高压时,肺血管发生重构主要是由于血管平滑肌细胞增殖凋亡失衡引起的,因此抑制肺血管收缩,改善肺循环血流动力学以及阻止肺血管平滑肌细胞异常增殖是治疗肺动脉高压的关键。
     钙信号转导在肺动脉高压发生发展过程中起着非常重要的作用,细胞内钙离子浓度([Ca2+]i)的变化是触发细胞增殖相关信号转导的始动因素之一。在PAH发生过程中,5-HT等细胞因子激动相应的细胞表面受体后,都要通过胞质内游离钙浓度增高这一信号转导途径发挥作用,因此,[Ca2+]i的变化在PAH的形成过程中具有重要作用。
     钙拮抗剂是近年来临床常用的抗肺动脉高压药物,但关于其逆转肺动脉高压的确切机制尚不甚明了。间尼索地平(m-Nis)是我校药学院合成的二氢吡啶类钙拮抗剂,前期研究表明其具有良好的稳定性以及抑制血管收缩的作用。本实验以野百合碱(MCT)诱导的大鼠肺动脉高压为模型,观察m-Nis对肺动脉高压的防治作用及肺血管重构的改善作用。还通过细胞免疫化学、流式细胞术、激光共聚焦及细胞分子生物学等技术研究m-Nis对5-HT诱导的肺动脉平滑肌细胞增殖、迁移的影响,深入探讨其作用机制。
     第一部分m-Nis对野百合碱诱导的大鼠肺动脉高压的防治作用
     目的:研究m-Nis对野百合碱诱导的大鼠肺动脉高压的防治作用及其作用机制。
     方法:大鼠单剂量皮下注射MCT(60mg/kg)制备肺动脉高压模型。导管法测定肺血流动力学指标。光镜观察肺小动脉结构的改变。按试剂盒步骤测定血清中MDA含量及SOD活性。免疫印迹法测定PCNA、ERK1/2和p-ERK1/2的表达水平。免疫组化法观察5-HT和PCNA的表达。
     结果:
     1 m-Nis对野百合碱诱导的肺动脉高压大鼠平均肺动脉压(PAP)及其它血流动力学指标的影响
     模型组PAP(17.4±1.6 mmHg)比对照组(10.5±1.6 mmHg)显著升高(P<0.01);与模型组相比,m-Nis 0.5, 1.0和2.0 mg/kg组的PAP分别降至14.4±1.1,14.0±1.3及13.6±0.9mmHg(P<0.01)。而平均颈动脉压(SAP)及心率在各组之间并无显著差异。
     2 m-Nis对肺动脉高压大鼠右室肥厚的影响
     与对照组相比,模型组大鼠体重显著降低(P<0.05),右心室重量/体重(RV/BW)明显升高(从0.79±0.09升至1.05±0.09,P<0.01),m-Nis 0.5, 1.0和2.0mg/kg对大鼠体重并无改善作用,却使RV/BW分别降至0.90±0.04,0.83±0.07,0.85±0.09(P<0.05或P<0.01,与模型组相比)。RV/(LV+SP)测定结果表明:与对照组相比,模型组大鼠RV/(LV+SP)明显升高(从0.307±0.007升至0.408±0.015,P<0.01),而m-Nis 0.5, 1.0, 2.0mg/kg组的RV/(LV+SP)分别降至0.365±0.013, 0.341±0.023和0.353±0.021(P<0.01,与模型组相比)。
     3 m-Nis对肺动脉高压大鼠肺小动脉构型改变的影响
     光镜下观察HE染色切片,发现模型组肺小动脉中膜厚度增加,血管管腔狭窄甚至闭塞。与对照组相比,MT%显著增加(P<0.01),VA%明显减少(P<0.01),而m-Nis各组均不同程度地减轻肺组织损伤程度,肺小动脉中膜增厚程度也有明显减轻,与模型组相比, MT%明显降低(P < 0.05或P < 0.01),而VA%明显增加(P < 0.01)。
     4 m-Nis对肺动脉高压大鼠血清中SOD活性及MDA含量的影响
     与对照组相比,模型组大鼠血清中MDA含量显著升高(从6.32±0.27升至7.62±0.63nmol/ml,P<0.01),SOD活性明显降低(从163±10降至122±11 U/ml,P < 0.01)。而m-Nis 0.5,1.0,2.0mg/kg组均不同程度地逆转了上述反应,MDA含量分别降至6.49±0.82,6.76±0.38及6.40±0.60(P< 0.05),SOD活性相应升至142±12(P<0.05),149±8和146±11(P<0.01)。
     5 m-Nis对肺动脉高压大鼠肺组织中5-HT表达的影响
     与对照组相比,模型组大鼠肺组织中5-HT阳性细胞百分率显著增加(从4.2±2.1%增至14.2±3.0%,P<0.01)。m-Nis 0.5,1.0,2.0mg/kg组5-HT阳性细胞百分率分别降至8.6±1.3%,7.1±1.9%和7.2±1.0%(P<0.01,与模型组相比)。
     6 m-Nis对细胞增殖核抗原(PCNA)表达的影响
     PCNA免疫组化染色结果表明,与对照组相比,模型组大鼠肺组织中PCNA阳性细胞百分数显著增加(P<0.01),胞核染色较深。而m-Nis各组PCNA阳性细胞百分数均明显减少,与模型组相比有明显差异(P<0.01)。western blot结果分析表明,与对照组相比,模型组大鼠肺动脉中PCNA蛋白表达显著增加(P<0.01),而m-Nis 0.5,1.0和2.0mg/kg组PCNA蛋白表达水平明显降低(P <0.05,与模型组相比)。
     7 m-Nis对ERK1/2磷酸化水平的影响
     western blot结果表明,与对照组相比,模型组大鼠肺动脉中p-ERK1/2蛋白表达显著增加(P<0.01),各浓度m-Nis均能明显抑制其表达(P<0.01)。而总ERK1/2的表达水平在各组之间并无显著性差异。各组条带灰度值计算结果表明,模型组p-ERK1/2/ ERK1/2的比值明显增加(P<0.01,与对照组相比),而m-Nis可明显降低此比值(P<0.05,与模型组相比)。
     结论:m-Nis能明显降低肺动脉高压大鼠的平均肺动脉压,改善右室肥厚程度及肺小动脉构型改变,对野百合碱诱导的大鼠肺动脉高压有一定的防治作用,可能与其抗氧化作用,降低5-HT的表达及抑制ERK1/2 MAPK信号通路有关。
     第二部分m-Nis对5-HT诱导大鼠肺动脉平滑肌细胞增殖、迁移及细胞内游离钙浓度变化的影响
     目的:研究m-Nis对5-HT诱导大鼠肺动脉平滑肌细胞增殖、迁移及细胞内游离钙浓度变化的影响
     方法:组织块种植法原代培养大鼠PASMCs。MTT法测定PASMCs增殖。Transwell迁移小室(8μm孔径)测定PASMCs的迁移能力。流式细胞仪测定细胞周期。免疫细胞化学染色及western blot法测定PCNA蛋白表达。激光共聚焦显微镜检测平滑肌细胞内Ca2+的变化。定磷法测定CaN活性。western blot法测定ERK1/2及JNK的磷酸化水平。RT-PCR法检测c-fos、c-jun、CaM及CaN mRNA的表达。
     结果:
     1 m-Nis平对5-HT诱导的PASMCs增殖的影响
     MTT结果表明,与对照组比,5-HT组A值明显升高(从0.48±0.04升至0.74±0.09,P <0.01)。与5-HT组比,m-Nis 10-5,10-6,10-7,10-8 mol/L组A值分别下降至0.54±0.05,0.57±0.07,0.59±0.06(P <0.01),0.62±0.07 (P <0.05)。western blot结果显示,与对照组相比,5-HT组PASMCs的PCNA表达明显增加(P<0.01)。不同浓度m-Nis预处理,均明显降低5-HT诱导的PCNA的表达(P<0.05或P<0.01)。PCNA免疫细胞化学染色结果表明,5-HT处理组PCNA阳性细胞百分率明显高于对照组(P<0.01)。不同浓度m-Nis预处理后,均明显降低5-HT诱导的PCNA的阳性表达(P<0.01)。
     2 m-Nis对5-HT诱导的PASMCs细胞迁移的影响
     与对照组比,5-HT(1μmol/L)能明显促进PASMCs迁移,平均每个视野下的迁移细胞数从对照组的34±7增加至95±13(P<0.01)。与5-HT组比,m-Nis 10-5,10-6,10-7,10-8 mol/L组平均每个视野下的迁移细胞数分别下降至43±9,44±10,58±9,62±9(P <0.01)。
     3 m-Nis对PASMCs细胞周期的影响
     与对照组比,5-HT组G0/G1期PASMCs百分率明显减少(从84%降至55%,P < 0.01),S期PASMCs百分率明显增加(从4%增至21%,P < 0.01),PI值也相应增加(从16%增至45%,P <0.01),提示5-HT能诱导PASMCs增殖。与5-HT组比,m-Nis 10-5,10-6,10-7 mol/L组G0/ G1期PASMCs百分率分别增至78%(P < 0.01),71%和64%(P < 0.05),S期PASMCs百分率也分别降至6%,10%和11%(P < 0.05或P < 0.01),G2/M期细胞无显著变化,说明m-Nis作用后使细胞停滞于G0/G1期。PI值分别降至22%,29%和36%(P <0.05或P <0.01)。而m-Nis 10-8 mol/L对细胞周期及PI值均无明显影响。
     4 5-HT对培养的大鼠PASMCs内游离钙离子浓度的影响
     Flou-3/AM负载的肺动脉平滑肌细胞图象清晰,可较好地显示细胞内游离钙离子浓度的变化过程,在加入1μmol/L 5-HT后,荧光强度增强,表明细胞内钙离子浓度迅速升高,在90s左右达到峰值,然后减弱,在200s左右荧光强度达到稳定的状态,而后有一个较长的平台期。
     5 m-Nis对5-HT诱导大鼠PASMCs内[Ca2+]i升高的影响
     在5-HT组,达峰值时其相对荧光强度(FI-F0)/F0为132.4±4.3%,与对照组相比明显升高(P<0.01)。与5-HT组相比,m-Nis 10-5,10-6,10-7,10-8 mol/L均能明显抑制5-HT诱导的细胞内钙离子浓度的升高,达峰值时相对荧光强度(FI-F0)/F0分别为20.1±4.1%,33.6±9.0%,41.5±2.3%和54.7±2.2%(P<0.01)。
     6 m-Nis对5-HT诱导大鼠肺动脉平滑肌细胞CaN活性的影响
     与对照组相比,5-HT组CaN活性明显升高(从1.7±0.5升至3.7±0.8μmol Pi/mg pro/h,P < 0. 01),m-Nis 10-5, 10-6,10-7,10-8mol/L均不同程度地降低了5-HT诱导的PASMCs中CaN活性的升高(分别降至2.4±0.6,2.9±0.6,3.0±0.7和3.1±0.5,P< 0. 05或P<0.01)。
     7 m-Nis对5-HT诱导大鼠PASMCs内CaM、CaN mRNA表达的影响
     与对照组相比,1μmol/L 5-HT作用24小时后明显诱导大鼠PASMCs内CaM及CaN mRNA表达水平升高(P < 0.01),加入m-Nis 10-5,10-6,10-7 mol/L预处理均明显抑制了5-HT诱导的CaM mRNA表达水平的升高(P < 0.05或P < 0.01),而m-Nis 10-8 mol/L抑制作用不明显。m-Nis 10-5,10-6,10-7,10-8 mol/L均明显抑制了5-HT诱导的CaN mRNA表达水平的升高(P < 0.05或P < 0.01)。
     8 m-Nis对5-HT诱导的大鼠PASMCs中ERK1/2及JNK磷酸化的影响
     western blot结果显示,1μmol/L 5-HT在5至30分钟内诱导大鼠动脉平滑肌细胞ERK1/2及JNK磷酸化水平明显升高,在15分钟时磷酸化水平最高。而m-Nis可浓度依赖性地拮抗5-HT的作用。m-Nis 10-5,10-6,10-7 mol/L可明显抑制5-HT诱导的ERK1/2磷酸化水平的升高(P < 0.05或P < 0.01),而m-Nis 10-8 mol/L的抑制作用并不明显。10-8 ~10-5 mol/L m-Nis均可抑制5-HT诱导的JNK磷酸化水平的升高(P < 0.05或P < 0.01)。
     9 m-Nis对5-HT诱导的大鼠PASMCs中c-fos及c-jun mRNA表达的影响
     1μmol/L 5-HT在5至60分钟内能明显诱导大鼠肺动脉平滑肌细胞c-fos及c-jun mRNA表达水平升高,在30分钟时表达水平达最高。而不同浓度m-Nis均明显抑制了5-HT诱导的c-fos及c-jun mRNA表达水平的升高(P < 0.05或P < 0.01)。
     结论:(1)m-Nis对5-HT诱导的肺动脉平滑肌细胞增殖和迁移有明显的抑制作用,可能与其抑制增殖细胞核抗原(PCNA)表达及细胞周期进程有关。(2)m-Nis明显抑制了5-HT引起的[Ca2+]i的升高、CaM和CaNmRNA的表达以及CaN活性的升高;对5-HT诱导的ERK1/2, JNK磷酸化水平也有不同程度的抑制,还明显抑制了增殖相关基因c-fos, c-jun mRNA的表达水平。由此,我们推测m-Nis抗5-HT诱导的PASMCs增殖作用可能还与抑制细胞内[Ca2+]i的增高从而阻断了5-HT所介导的ERK1/2, JNK MAPK信号通路有关。
     第三部分RhoA/Rho激酶信号通路在5-HT刺激PASMCs增殖中的作用及m-Nis对此信号通路的影响
     目的:研究RhoA/Rho激酶信号通路在5-HT刺激PASMCs增殖中的作用及m-Nis对此信号通路的抑制作用。
     方法:组织块种植法原代培养大鼠PASMCs。MTT法测定PASMCs增殖。流式细胞仪测定细胞周期。western blot法测定PCNA蛋白表达及ERK1/2、JNK、MYPT1磷酸化水平。RT-PCR法检测RhoA、ROCK1、c-fos及c-jun mRNA的表达。结果:
     1法舒地尔对5-HT诱导的PASMCs增殖的影响
     MTT实验结果表明,与对照组比,5-HT组A值明显升高(从0.37±0.05升至0.61±0.03,P <0.01)。法舒地尔25、50、100μmol/L各组A值分别降至0.48±0.05,0.46±0.03和0.43±0.04(P <0.01)。western blot结果显示,与对照组比,5-HT组PCNA蛋白表达明显增多(P <0.01),而25、50、100μmol/L法舒地尔不同程度地降低了PCNA的蛋白表达(P<0.05或P<0.01)。
     2法舒地尔对5-HT作用下PASMCs细胞周期的影响
     与对照组比,5-HT组G0/G1期PASMCs百分率明显减少(从83%降至55%,P < 0.01), S期PASMCs百分率明显增加(从4%增至21%,P < 0.01),PI值也相应增加(从17%增至45%,P <0.01),提示5-HT能诱导PASMCs增殖。与5-HT组比,法舒地尔25,50,100μmol/L组G0/ G1期PASMCs百分率分别增至65%,70%和78%(P<0.05或P<0.01),S期PASMCs百分率也分别降至12%,11%和9%(P < 0.05或P < 0.01),PI值分别降至35%,30%和22%(P <0.05或P <0.01)。
     3法舒地尔对5-HT诱导的ROCK激活的影响
     与对照组比,1μmol/L 5-HT明显诱导了ROCK1 mRNA的表达(P < 0.01),法舒地尔25、50、100μmol/L预处理不同程度地降低了5-HT诱导的ROCK1 mRNA的表达(P < 0.05或P < 0.01)。western blot结果显示,1μmol/L 5-HT作用后明显升高了MYPT1的磷酸化水平,10分钟左右磷酸化水平最高(P < 0.01),法舒地尔25、50、100μmol/L不同程度地降低了5-HT诱导的MYPT1的磷酸化水平(P < 0.05或P < 0.01)。
     4法舒地尔对5-HT作用下ERK1/2及JNK磷酸化水平的影响
     与对照组比,5-HT作用15分钟明显诱导了ERK1/2的磷酸化(P<0.01),法舒地尔25、50、100μmol/L预处理对ERK1/2的磷酸化水平没有明显影响。而对于JNK的磷酸化水平,5-HT作用15分钟能明显诱导其升高(P < 0.01),法舒地尔25、50、100μmol/L预处理明显降低了JNK的磷酸化水平(P < 0.01)。另外,5-HT明显增加了核蛋白中p-ERK1/2的表达,15分钟左右表达水平最高(P <0.01),法舒地尔25、50、100μmol/L预处理明显降低了核蛋白中p-ERK1/2的表达(P < 0.01)。
     5法舒地尔对5-HT作用下c-fos和c-jun mRNA表达的影响
     与对照组比,5-HT作用30分钟明显诱导了c-fos及c-jun mRNA表达(P < 0.01)。法舒地尔25、50、100μmol/L预处理均明显抑制了5-HT诱导的c-fos (P < 0.01)及c-jun mRNA的表达(P < 0.05或P < 0.01)。
     6 m-Nis对5-HT作用下PASMCs中RhoA/ROCK通路的影响
     与对照组比,5-HT明显诱导了RhoA、ROCK1 mRNA的表达(P < 0.01),m-Nis10-5 mol/L明显降低5-HT诱导的RhoA mRNA的表达(P < 0.05)。m-Nis 10-5,10-6 mol/L还不同程度地降低了5-HT诱导的ROCK1 mRNA的表达(P < 0.05或P < 0.01)。western blot结果显示,5-HT作用10分钟明显升高了MYPT1的磷酸化水平(P < 0.01),m-Nis 10-5,10-6,10-7 mol/L预处理不同程度地降低了5-HT诱导的MYPT1的磷酸化水平(P < 0.05或P < 0.01),而m-Nis 10-8 mol/L对MYPT1的磷酸化水平无明显影响。
     结论:(1)5-HT明显诱导了大鼠PASMCs中Rho激酶的表达及激活;Rho激酶抑制剂法舒地尔对5-HT诱导的PASMCs的增殖,细胞周期进展有明显的抑制作用,还不同程度地抑制了JNK的磷酸化,ERK1/2的核转位以及增殖相关基因c-fos、c-jun mRNA的表达。说明RhoA/ROCK通路在5-HT诱导的PASMCs增殖中起着重要作用。(2)m-Nis不同程度地抑制了5-HT诱导的RhoA, ROCK1 mRNA的表达以及Rho激酶的活化标志p-MYPT1的蛋白表达。说明m-Nis抗5-HT诱导的PASMCs的增殖作用可能与抑制RhoA/ROCK信号通路有关。
     第四部分活性氧在5-HT刺激PASMCs增殖中的作用及m-Nis对此通路的影响
     目的:研究活性氧(ROS)在5-HT刺激PASMCs增殖中的作用和m-Nis对ROS的产生及其下游信号通路的影响。
     方法:组织块种植法原代培养大鼠PASMCs。激光共聚焦检测ROS生成量的变化。TAB法测定MDA含量。黄嘌呤氧化酶法测定SOD活性。MTT法测定PASMCs增殖。western blot法测定PCNA蛋白表达及ERK1/2、JNK磷酸化水平。
     结果:
     1 m-Nis对5-HT诱导的大鼠PASMCs内ROS生成的影响
     1μmol/L的5-HT处理不同时间组,荧光强度均增强,5-HT处理15分钟组荧光强度最强,达对照组的288±54%(P<0.01),然后减弱,在5-HT处理120和240分钟组荧光强度达到稳定的状态,但仍明显高于对照组(P<0.01)。m-Nis 10-5,10-6,10-7,10-8mol/L均能明显抑制5-HT诱导的ROS的生成,在5-HT处理15分钟时荧光强度分别降为对照组的157±29%,173±38%,178±43%和215±45%(P<0.01)。
     2 m-Nis对5-HT作用下大鼠PASMCs内SOD活性和MDA含量的影响
     与对照组相比,5-HT处理组SOD活性明显降低(P < 0. 01),MDA含量明显升高(P < 0. 01)。m-Nis 10-5,10-6,10-7mol/L预处理均不同程度地逆转了5-HT诱导的PASMCs SOD活性的降低(P < 0. 05或P < 0. 01),同时也降低了MDA的含量(P < 0. 05或P < 0. 01),而m-Nis 10-8mol/L对SOD的活性没有明显影响,但可降低MDA的含量(P < 0.05)。
     3 m-Nis平对H2O2诱导的大鼠PASMCs增殖的影响
     MTT实验结果表明,100μmol/L H2O2能明显促进大鼠PASMCs增殖,与对照组比,H2O2处理组A值明显提高(P <0.01)。与H2O2处理组比,m-Nis各组A值均不同程度降低(P <0.05或P <0.01)。western blot结果显示,与对照组相比,H2O2处理组PASMCs的PCNA表达明显增加(P<0.01)。不同浓度m-Nis预处理后,均明显降低H2O2诱导的PCNA的表达(P<0.05或P<0.01)。
     4 m-Nis对H2O2诱导的PASMCs中ERK1/2及JNK磷酸化水平的影响
     与对照组相比,100μmol/L H2O2作用15分钟时能明显升高大鼠脉平滑肌细胞ERK1/2及JNK磷酸化水平(P<0.01)。m-Nis 10-5,10-6,10-7 mol/L可明显抑制H2O2诱导的ERK1/2及JNK磷酸化水平的升高(P < 0.05或P < 0.01),而m-Nis 10-8mol/L仅明显抑制了ERK1/2磷酸化水平的升高(P < 0.05),对JNK磷酸化水平无明显影响。
     结论:(1)5-HT明显诱导了大鼠PASMCs中ROS的生成,H2O2不仅诱导了PASMCs增殖,还提高了ERK1/2及JNK的磷酸化水平。说明ROS在5-HT诱导的PASMCs增殖中起重要作用。(2)m-Nis不仅抑制了ROS的生成,还明显降低了H2O2诱导的PASMCs增殖及ERK1/2、JNK的磷酸化水平。说明m-Nis抗5-HT诱导的PASMCs增殖作用不仅与其抑制ROS的产生有关,还与其抑制ROS所介导的ERK1/2,JNK MAPK信号通路有关。
     总结
     1 m-Nis对野百合碱诱导的大鼠肺动脉高压有一定的防治作用,可能与其抗氧化作用,降低5-HT的表达及抑制ERK1/2 MAPK信号通路有关。
     2 m-Nis对5-HT诱导的肺动脉平滑肌细胞增殖和迁移有明显的抑制作用,可能与其抑制细胞内[Ca2+]i的增高从而阻断了5-HT所介导的ERK1/2,JNK MAPK信号通路有关。
     3 Rho激酶抑制剂法舒地尔明显抑制了5-HT诱导的PASMCs增殖,说明RhoA/ROCK通路在5-HT诱导的PASMCs增殖中起重要作用。
     4 m-Nis不同程度地抑制了5-HT诱导的RhoA, ROCK1 mRNA的表达以及Rho激酶的活化标志p-MYPT1的蛋白表达。说明m-Nis抗5-HT诱导的PASMCs的增殖作用可能与抑制RhoA/ROCK信号通路有关。
     5 ROS在5-HT诱导的PASMCs增殖中起重要作用。m-Nis抗5-HT诱导的PASMCs增殖作用不仅与其抑制ROS的产生有关,还与其抑制ROS所介导的ERK1/2,JNK MAPK信号通路有关。
Pulmonary artery hypertension (PAH) is a progressive and fatal disease characterized by an increase of vascular resistance, abnormal sustained pulmonary vasoconstriction, and progressive structural remodeling of pulmonary arteries, for which no clear pathogenesy has been developed. Recent studies suggest that constriction and remodeling of pulmonary arteries are the key point of PAH formation. The disequilibrium of proliferation of PASMCs and apoptosis results in pulmonary arteries remodeling. Therefore, the key points of PAH therapy include inhibiting the pulmonary artery constriction, improving the pulmonary circulation and preventing the proliferation of PASMCs.
     Calcium signals play an important role in the formation of PAH. Studies suggest that the elevation of [Ca2+]i is one of the most important initiation agents to trigger proliferation of PASMCs. In the progress of PAH, many cell factors including 5-HT can elevate the [Ca2+]i after they bind to their receptors. Therefore, the elevation of [Ca2+]i is important to PAH.
     Recently, calcium antagonists are frequently used in the treatment of PAH in clinic, but the clear mechanisms have not been elucidated. M-nisoldipine (m-Nis), a Ca2+ channel blocker of dihydropyridines (DHPs), is developed by Hebei Medical University. Early studies suggested that m-Nis had favourable stability and could inhibit vasoconstriction. Therefore, in the present study, monocrotaline (MCT) was use to induce PAH in rats, then the effect of m-Nis on PAH and pulmonary artery remodeling were studied. The methods such as immunhistochemistry, flow cytometry, confocal laser scanning microscope and molecular biology techniques were applied to explore the effect of m-Nis on 5-HT induced proliferation and migration of PASMCs and the mechanisms. Part 1 The effect of m-Nis on MCT-induced PAH in Rats
     Objective: To study the effect of m-Nis on MCT-induced PAH in rats.
     Methods: Rats were injected with a single dose (60mg/kg) of MCT subcutaneously to induce PAH. Pulmonary haemodynamic measurement and lung tissue morphological investigations were undertaken. The MDA production and SOD activity in the serum were tested. Expression of PCNA, ERK1 and p-ERK1/2 expression in pulmonary blood vessels was analyzed by western blot. The expression of 5-HT and PCNA in pulmonary tissues was observed by immunohistochemistry.
     Results:
     1 Effect of m-Nis on PAP and hemodynamic variables in MCT-induced PAH rats
     Compared with the control group, mean PAP in the MCT group was significantly elevated (from 10.5±1.6 to 17.4±1.6mmHg, P<0.01). Compared with the MCT group, mean PAP in m-Nis 0.5, 1.0 and 2.0 mg/kg groups was decreased to 14.4±1.1,14.0±1.3 and 13.6±0.9 mmHg(P<0.01), respectively。Yet, SAP and heart rate did not differ significantly among the five groups.
     2 Effect of m-Nis on right ventricle hypertrophy in MCT-induced PAH rats
     Body weight was significantly decreased by MCT injection (P<0.05). The ratio of right ventricular weight to body weight (RV/BW) was increased by MCT from 0.79±0.09 to 1.05±0.09 (P<0.01 vs control) and reduced by m-Nis 0.5, 1.0, 2.0mg/kg to 0.90±0.04 (P<0.05 vs MCT), 0.83±0.07 and 0.85±0.09 (P<0.01 vs MCT) respectively. Meanwhile, the right ventricular index RV/(LV+SP) was increased by MCT from 0.307±0.007 to 0.408±0.015 (P<0.01 vs control) and reduced obviously by m-Nis 0.5, 1.0, 2.0mg/kg to 0.365±0.013, 0.341±0.023 and 0.353±0.021 respectively (P<0.01 vs MCT).
     3 Effect of m-Nis on the morphological alterations of small pulmonary arteries
     Small pulmonary arteries in MCT group exhibited obvious medial wall thickening and lumen transverse area reducing, the MT% markedly increased, whereas the VA% decreased compared with those in control group (P<0.01). In m-Nis-treated groups, the pulmonary artery wall thickening was attenuated in different degrees, the MT% was decreased in m-Nis 0.5 mg/kg group (P<0.05) and m-Nis 1.0, 2.0 mg/kg groups (P<0.01) compared with that in MCT group. Meanwhile, the VA% was significantly increased in all the m-Nis groups compared with that in MCT group (P<0.01).
     4 Effect of m-Nis on the activity of SOD and the content of MDA
     Compared with control group, the content of MDA in MCT group was significantly elevated from 6.32±0.27 to 7.62±0.63 (P<0.01), and the activity of SOD was decreased from 163±10 to 122±11 (P<0.01). Compared with the MCT group, a significant reduction in MDA production was observed in all three m-Nis 0.5, 1.0 and 2.0mg/kg groups (from 7.62±0.63 to 6.49±0.82, 6.76±0.38 and 6.40±0.60 respectively) (P<0.05), which was associated with the increase of SOD activity (from 122±11 to 142±12, 149±8 and 146±11 respectively) (P<0.05 or P<0.01).
     5 5-HT immunohistochemistry assessment
     In MCT group, the percentage of 5-HT-positive cells (14.2±3.0%) was higher than that in control group (4.2±2.1%) (P<0.01), which was reduced to 8.6±1.3%,7.1±1.9% and 7.2±1.0% by m-Nis 0.5, 1.0 and 2.0mg/kg treatment (P<0.01 vs MCT group).
     6 Effect of m-Nis on the expression of PCNA
     From the result of immunohistological staining, we found that in MCT group the percentage of PCNA-positive cells was remarkably increased compared with the control group (P<0.01), which was decreased by pretreatment with m-Nis (P<0.01). Western blot analysis indicated that the expression of PCNA was significantly higher in MCT group than that in control group (P<0.01), which was markedly decreased (P<0.05 vs MCT group) by m-Nis 0.5, 1.0 and 2.0mg/kg pretreatment.
     7 Effect of m-Nis on the activation of ERK1/2
     Western blot analysis showed that p-ERK1/2 expression in MCT group exhibited a significant increase compared with that in control group (P<0.01), which was obviously decreased by m-Nis 0.5, 1.0 and 2.0mg/kg (P<0.01). About the expression of ERK1/2, no statistical difference was found among the five groups. So, the ratio of p-ERK1/2/ERK1/2 increased significantly in MCT group compared with that in control group (P<0.01), which was decreased significantly by m-Nis (P<0.05).
     Conclusion: M-Nis protects against MCT-induced pulmonary artery hypertension, which may be related to the antioxidation effect, the reduction of 5-HT exression and the suppression of the ERK/MAPK signal pathway. Part 2 The effect of m-Nis on 5-HT-induced proliferation, migration of PASMCs and change of [Ca2+]i
     Objective: To study the effect of m-Nis on 5-HT-induced proliferation, migration of PASMCs and change of [Ca2+]i.
     Methods: PASMCs were cultured with the explant technique. The proliferation of PASMCs was evaluated by MTT assay. Transwell chambers were used to detect the migration. Cell-cycle distribution was determined by flow cytometry assay. The expression of PCNA was evaluated by immunocytochemical stain and western blot. The change of [Ca2+]i was measured by confocal microscopy. The activity of CaN was measured using kits. The phosphorylation of ERK1/2 and JNK was detected by western blot. The mRNA expression of c-fos, c-jun, CaM and CaN was evaluated by RT-PCR.
     Results:
     1 Effect of m-Nis on 5-HT-induced proliferation of PASMCs
     The results of MTT assay suggested that 5-HT significantly increased the A value from 0.48±0.04 to 0.74±0.09 (P<0.01), which was decreased to 0.54±0.05,0.57±0.07,0.59±0.06 (P<0.01) and 0.62±0.07 (P<0.05) by m-Nis 10-5,10-6,10-7,10-8 mol/L, respectively. Similarly, western blot analysis of PCNA indicated that the expression of PCNA was significantly higher in 5-HT group than that in control group (P <0.01). Whereas, in four m-Nis treated groups, the level of PCNA was markedly decreased (P<0.05 or P<0.01). Next, the results of immunocytochemical stain suggested that in 5-HT group the percentage of PCNA-positive cells was remarkably increased compared with that in control group (P < 0.01), which was decreased by pretreatment with m-Nis 10-5,10-6,10-7 and 10-8 mol/L (P < 0.01).
     2 Effect of m-Nis on 5-HT-induced PASMCs migration
     Compared with control group, the number of migration cells increased from 34±7 to 95±13 (P<0.01) after 5-HT treatment, which was decreased to 43±9,44±10,58±9 and 62±9 (P <0.01) by m-Nis 10-5,10-6,10-7 and 10-8 mol/L pretreatment, respectively.
     3 Effect of m-Nis on cell-cycle progression
     The percentage of cells in S phase increased dramatically from 4% to 21% after 5-HT stimulation (P<0.01) and was accompanied by a reduction in the percentage of cells in G1 phase from 84% to 55% (P<0.01). M-Nis 10-5,10-6,10-7 mol/L markedly blocked 5-HT-induced cell-cycle progression, with the percentages of PASMCs entering the S phase decreasing to 6%, 10% and 11% (P<0.05 or P<0.01), and those arrested in the G0/G1 phase increasing to 78%, 71% and 64% respectively (P<0.05 or P<0.01). Meanwhile, the proliferation index increased significantly from 16% to 45% after 5-HT stimulation (P<0.01) and was decreased to 22%, 29% and 36% respectively by preincubation with m-Nis 10-5,10-6,10-7 mol/L (P<0.05 or P<0.01).
     4 Effect of 5-HT on fluorescence intensity of [Ca2+]i in rat PASMCs
     The fluorescence intensity that represented the [Ca2+]i of PASMCs was significantly increased and reached the peak at 90 s after treated with 5-HT. After that, the [Ca2+]i decreased gradually and reached to a stable state at 200 s after 5-HT treatment.
     5 Effect of m-Nis on [Ca2+]i in rat PASMCs
     The relative fluorescence intensity of intracellular Ca2+ was significantly increased from 3.0±1.0% to 132.4±4.3% after perfusion with 5-HT (P <0.01 vs control), which was decreased to 20.1±4.1%,33.6±9.0%,41.5±2.3% and 54.7±2.2% (P<0.01) respectively by preincubation with m-Nis 10-5,10-6,10-7 and 10-8 mol/L.
     6 Effect of m-Nis on the activity of CaN in rat PASMCs
     The activity of CaN was significantly increased from 1.7±0.5 to 3.7±0.8 by 5-HT treatment (P <0.01 vs control), which was decreased to 2.4±0.6, 2.9±0.6 (P<0.01), 3.0±0.7 and 3.1±0.5 (P<0.05) respectively by preincubation with m-Nis 10-5, 10-6, 10-7 and 10-8 mol/L.
     7 Effect of m-Nis on mRNA expression of CaM and CaN in rat PASMCs
     The mRNA expression of CaM and CaN increased obviously after 1μmol/L 5 -HT treatment for 24h (P<0.01), which was significantly reduced (P<0.05 or P<0.01) by pretreatment with m-Nis 10-5, 10-6 and 10-7 mol/L. M-Nis 10-8 mol/L also significantly reduced the CaN mRNA expression, but have no effect on CaM mRNA expression.
     8 Effect of m-Nis on the phosphorylation of ERK1/2 and JNK in rat PASMCs
     Treatment of cells with 1μmol/L 5-HT caused phosphorylation of ERK1/2 and JNK with a peak at 15 min. M-Nis 10-5, 10-6 and 10-7 mol/L pretreatment reduced 5-HT-induced phosphorylation of ERK1/2 and JNK obviously (P<0.05 or P<0.01), and m-Nis 10-8 mol/L also significantly reduced the JNK phosphorylation, but have no significant effect on ERK1/2 phosphorylation.
     9 Effect of m-Nis on mRNA expression of c-fos and c-jun in rat PASMCs
     Treatment of cells with 1μmol/L 5-HT increased mRNA expression of c-fos and c-jun with a peak at 30 min, which was decreased in different degree by m-Nis 10-5, 10-6, 10-7 and 10-8 mol/L pretreatment (P<0.05 or P<0.01).
     Conclusion: (1) M-Nis inhibited 5-HT-induced proliferation and migration of PASMCs, which may be related to the inhibition of PCNA expression and cell-cycle progression. (2) M-Nis inhibited 5-HT induced elevation of [Ca2+]i, mRNA expression of CaM, CaN, c-fos and c-jun, phosphorylation of ERK1/2 and JNK. So, the inhibition of m-Nis on proliferation of PASMCs may be related to the blockage of ERK1/2, JNK MAPK signal pathway through inhibiting the elevation of [Ca2+]i.
     Part 3 The activation of RhoA/ROCK signal pathway in 5-HT-induced proliferation of PASMCs and the inhibitory effect of m-Nis on this pathway
     Objective: To study the the activation of RhoA/ROCK signal pathway in 5-HT-induced proliferation of PASMCs and the inhibitory effect of m-Nis on this pathway.
     Methods: PASMCs were cultured with the explant technique. The proliferation of PASMCs was evaluated by MTT assay. Cell-cycle distribution was determined by flow cytometry assay. The expression of PCNA, the phosphorylation of ERK1/2, JNK and MYPT1 was detected by western blot. The mRNA expression of Rho A, ROCK1, c-fos and c-jun was evaluated by RT-PCR.
     Results:
     1 Effect of fasudil on 5-HT-induced proliferation of PASMCs
     The results of MTT assay suggested that 1μmol/L 5-HT significantly increased the A value from 0.37±0.05 to 0.61±0.031 (P<0.01), which was decreased to 0.48±0.05 , 0.46±0.03 and 0.43±0.04 by fasudil 25, 50, 100μmol/L pretreatment respectively (P<0.01). Similarly, western blot analysis of PCNA indicated that the expression of PCNA was higher in 5-HT group than that in control group (P <0.01), which was inhibited in different degree (P<0.05 or P<0.01) by pretreatment of PASMCs with fasudil.
     2 Effect of fasudil on cell-cycle progression
     The percentage of cells in S phase increased dramatically from 4% to 21% after 5-HT stimulation (P<0.01) and was accompanied by a reduction in the percentage of cells in G1 phase from 83% to 55% (P<0.01). Fasudil 25, 50, 100μmol/L markedly blocked 5-HT-induced cell-cycle progression, with the percentages of PASMCs entering the S phase decreasing to 12%, 11% and 9% (P<0.05 or P<0.01), and those arrested in the G0/G1 phase increasing to 65%, 70% and 78% respectively (P<0.05 or P<0.01). Meanwhile, the proliferation index increased dramatically from 17% to 45% after 5-HT stimulation (P < 0.01) and was decreased to 35%, 30% and 22% (P<0.05 or P<0.01) respectively by preincubation with fasudil 25, 50 and 100μmol/L.
     3 Fasudil inhibited 5-HT- mediated ROCK activation
     5-HT induced a significant upregulation of ROCK1 mRNA expression compared with that in quiescent PASMCs (P<0.01), and pretreatment with fasudil 25, 50, 100μmol/L resulted in a significant reduction in ROCK1 mRNA expression (P<0.05 or P<0.01). 5-HT caused phosphorylation of MYPT1, with a peak at 10 min (P<0.01), which was inhibited by fasudil in a concentration-dependent manner (P<0.05 or P<0.01).
     4 Effect of fasudil on ERK1/2 and JNK phosphorylation
     5-HT caused obvious phosphorylation of ERK1/2 and JNK with a peak at 15 min (P < 0.01). Pretreatment with fasudil 25, 50 and 100μmol/L did not block ERK1/2 phosphorylation, but reduced JNK phosphorylation obviously (P < 0.01). Next we examined the effect of fasudil on nuclear translocation of ERK1/2 induced by 5-HT. Result suggested that 5-HT increased nuclear translocation of ERK1/2 as determined by western blot analysis of nuclear-rich cellular fractions, with a peak at 15 min, consistent with its phosphorylation, which was reduced (P < 0.01) by pretreatment of cells with fasudil 25, 50 and 100μmol/L.
     5 Effect of fasudil on mRNA expression of c-fos and c-jun
     Treatment of cells with 1μmol/L 5-HT for 30 min increased mRNA expression of c-fos and c-jun (P< 0.01), which was decreased in different degree by fasudil 25, 50, 100μmo/L pretreatment (P < 0.05 or P<0.01).
     6 Effect of m-Nis on 5-HT-mediated RhoA/ROCK signal pathway
     5-HT induced a significant upregulation of mRNA expression of RhoA and ROCK1 (P < 0.01), which was reduced significantly (P < 0.05 or P < 0.01) by pretreatment with m-Nis 10-5 mol/L, m-Nis 10-6 mol/L also reduced ROCK1 mRNA expression (P < 0.05), but had no effect on RhoA mRNA expression. Phosphorylation of MYPT1 was induced by 5-HT with a peak at 10 min (P < 0.01), which was inhibited by m-Nis 10-5,10-6 and 10-7 mol/L in different degree (P < 0.05 or P < 0.01). However, m-Nis 10-8 mol/L had no effect on p-MYPT1 expression.
     Conclusion: (1) 5-HT mediated ROCK activation, fasudil not only inhibited 5-HT-induced proliferation of PASMCs and cell-cycle progression, but also reduced JNK phosphorylation, nuclear translocation of ERK1/2 and c-fos, c-jun mRNA expression, which suggested that RhoA/ROCK signal pathway played an important role in 5-HT-induced proliferation of PASMCs. (2) M-Nis significantly inhibited 5-HT-induced RhoA, ROCK1 mRNA expression and the phosphorylation of MYPT1. So we conclude that the inhibition of m-Nis on 5-HT-induced proliferation of PASMCs may be related to the blockage of RhoA/ROCK signal pathway.
     Part 4 Effect of ROS in 5-HT-induced proliferation of PASMCs and the inhibitory effect of m-Nis on this pathway
     Objective: To study the effect of ROS in 5-HT-induced proliferation of PASMCs and the inhibitory effect of m-Nis on this pathway.
     Methods: PASMCs were cultured with the explant technique. ROS was measured by confocal microscopy with DCFH-DA. The MDA production and SOD activity were measured using kits supplied by Nanjing Jiancheng Biotechnological Company. The proliferation of PASMCs was evaluated by MTT assay. The expression of PCNA, the phosphorylation of ERK1/2 and JNK were detected by western blot.
     Results:
     1 Effect of m-Nis on the production of ROS in rat PASMCs
     The fluorescence intensity of ROS was significantly increased and reached the peak at 15 min after the cells were treated with 5-HT (P<0.01). After that, the fluorescence intensity decreased gradually and reached to a stable state at 120 min and 240 min after 5-HT treatment, but still stronger than that in control group (P<0.01). Compared with control group, the fluorescence intensity of ROS increased by 288% 15 min after 5-HT treatment (P<0.01). Pretreatment with m-Nis 10-5, 10-6, 10-7 and 10-8 mol/L reduced the fluorescence intensity of ROS from 288±54% to 157±29%, 173±38%, 178±43% and 215±45%, respectively (P<0.01).
     2 Effect of m-Nis on the activity of SOD and the content of MDA
     Compared with the control group, the activity of SOD decreased obviously (P<0.01), and the content of MDA increased significantly (P<0.01) after 5-HT treatment. Compared with the 5-HT group, a significant reduction in MDA production was observed in m-Nis 10-5,10-6,10-7 mol/L groups (P<0.05 or P<0.01), which was associated with an increase in the SOD activity (P<0.05 or P<0.01). Meanwhile, m-Nis 10-8 mol/L also reduced the content of MDA (P<0. 05), but had no effect on the SOD activity.
     3 Effect of m-Nis on H2O2-induced proliferation of PASMCs
     The results of MTT assay suggested that 100μmol/L H2O2 significantly increased the A value (P<0.01), which was inhibited significantly (P <0.05 or P <0.01) by m-Nis 10-5,10-6,10-7 and 10-8 mol/L. Similarly, western blot analysis of PCNA indicated that the expression of PCNA was higher in H2O2 group than that in control group (P <0.01), which was also inhibited obviously by pretreatment of PASMCs with m-Nis (P<0.05 or P<0.01).
     4 Effect of m-Nis on H2O2-induced phosphorylation of ERK1/2 and JNK
     H2O2 caused obvious phosphorylation of ERK1/2 and JNK (P < 0.01), which was inhibited in different degree by m-Nis 10-5, 10-6 and 10-7 mol/L pretreatment (P < 0.05 or P < 0.01). Meanwhile, m-Nis 10-8 mol/L also inhibited the phosphorylation of ERK1/2 (P < 0.05), but had no effect on JNK activation.
     Conclusion: (1) 5-HT induced the production of ROS obviously. H2O2 not only mediated proliferation of PASMCs, but also induced ERK1/2 and JNK phosphorylation, which suggested that ROS played an important role in 5-HT-induced proliferation of PASMCs. (2) M-Nis not only inhibited 5-HT-induced production of ROS, but also reduced the proliferation of PASMCs and phosphorylation of ERK1/2 and JNK induced by H2O2. So we conclude that the inhibition of m-Nis on 5-HT-induced proliferation of PASMCs may be related to the blockage of ROS production and the downstream ERK1/2,JNK MAPK signal pathway.
     CONCLUSIONS
     1 M-Nis protects against MCT-induced pulmonary artery hypertension, which may be related to the antioxidation effect, the reduction of 5-HT exression and the suppression of the ERK/MAPK signal pathway.
     2 M-Nis inhibited 5-HT-induced proliferation and migration of PASMCs, which may be related to the blockage of ERK1/2, JNK MAPK signal pathway through inhibiting the elevation of [Ca2+]i.
     3 Fasudil inhibited 5-HT-induced proliferation of PASMCs obviously, which suggested that RhoA/ROCK signal pathway played an important role in 5-HT-induced proliferation of PASMCs.
     4 M-Nis inhibited 5-HT-induced RhoA, ROCK1 mRNA expression and the phosphorylation of MYPT1 in different degree. So we conclude that the inhibition of m-Nis on 5-HT-induced proliferation of PASMCs may be related to the blockage of RhoA/ROCK signal pathway.
     5 ROS played an important role in 5-HT-induced proliferation of PASMCs. The inhibition of m-Nis on 5-HT-induced proliferation of PASMCs may be related to the blockage of ROS production and the downstream ERK1/2,JNK MAPK signal pathway.
引文
1 Shujaat A, Minkin R, Eden E. Pulmonary hypertension and chronic cor pulmonale in COPD. Int J Chron Obstruct Pulmon Dis, 2007, 2: 273-282
    2 Alexandru B, Bogdan MA. Monocrotaline induces pulmonary hypertension in animal model. Pneumologia, 2001, 50: 85-89
    3 Eddahibi S, Guignabert C, Barlier-Mur AM, et al. Cross talk between endothelial and smooth muscle cells in pulmonary hypertension: critical role for serotonin-induced smooth muscle hyperplasia. Circulation, 2006, 113: 1857-1864
    4 KanaiY, HoriS, TanakaT, et al. Role of 5-hydroxytryptamine in the progression of monocrotaline induced pulmonary hypertension in rats. Cardiovasc Res, 1993, 27: 1619-1623
    5 Li M, Liu Y, Dutt P, et al. Inhibition of serotonin-induced mitogenesis, migration, and ERK MAPK nuclear translocation in vascular smooth muscle cells by atorvastatin. Am J Physiol Lung Cell Mol Physiol, 2007, 293: L463-471
    6 Liu Y, Suzuki YJ, Day RM, et al. Rho kinase-induced nuclear translocation of ERK1/ERK2 in smooth muscle cell mitogenesis caused by serotonin. Circ Res, 2004, 95: 579-586
    7 Okoro EO. Overlap in the pharmacology of L-type Ca2+-channel blockers and 5-HT2 receptor antagonists in rat aorta. J Pharm Pharmacol, 1999, 51: 953-957
    8 Okoro EO, Marwood JF. Effects of 5-HT2 receptor antagonists on responses to potassium depolarization, inratisolated aorta.Clin Exp Pharmacol Physiol, 1997, 24: 34-39
    9 Drummond RM, Wadsworth RM. In vitro effects of nifedipine on KCl and 5-hydroxytryptamine-induced contractions of the sheep coronary, cerebraland pulmonary arteries. Life Sci, 1994, 54: 1081-1090
    10 Abe K, Shimokawa H, Morikawa K, et al. Long-term treatment with a Rho-kinase inhibitor improves monocrotalin-induced fatal pulmonaryhypertension in rats. Circ Res, 2004, 94: 385-393
    11 Itoh T, Nagaya N, Fujii T, et al. A combination of oral sildenafil and beraprost ameliorated pulmonary hypertension in rats. Am J Respir Cirt Care Med, 2004, 169: 34-38
    12 Bath PJ, Kimpel CH, Roy S, et al. An improved mathematical approach for the assessment of the medical thickness of pulmonary arteries. Pathol Res Pract, 1993, 189: 567-576
    13 Zhang TT, Dai DZ. Therapeutic effects of nifedipine on pulmonary hypertension and mechanism involved. Prog Pharm Sci, 2003, 27: 227-229
    14 Wang HL. The serotonin receptor and transporter are potential therapeutic targets for pulmonary hypertension. Curr Opin Invest Drugs, 2004, 5: 963-966
    15 Miyata M, Ito M, Sasajima T, et al. Development of monocrotaline -induced pulmonary hypertension is attenuated by a serotonin receptor antagonist. Lung, 2000, 178: 63-73
    16 Morecroft I, Loughlin L, Nilsen M, et al. Functional interactions between 5-HT receptors and the 5-HT transporter in pulmonary arteries. J Pharmacol Exp Ther, 2005, 313: 539-548
    17 Herrick-Davis K, Grinde E, Harrigan TJ, et al. Inhibition of serotonin 5-hydroxytryptamine 2c receptor function through heterodimerization: receptor dimers bind two molecules of ligand and one G-protein. J Biol Chem, 2005, 280: 40144-40151
    18 Homma N, Nagaoka T, Morio Y, et al. Endothelin-1 and serotonin are involved in activation of RhoA/Rho kinase signaling in the chronically hypoxic hypertensive rat pulmonary circulation. J Cardiovasc Pharmacol, 2007, 50: 697-702
    19 Liu Y, Suzuki YJ, Day RM, et al. Rho kinase–induced nuclear translocation of ERK1/ERK2 in smooth muscle cell mitogenesis caused by serotonin. Circ Res, 2004, 95: 579-586
    20 Matthiesen S, Bahulayan A, Holz O, Racke K. MAPK pathway mediates muscarinic receptor-induced human lung fibroblast proliferation. Life Sci,2007, 80: 2259-2262
    21 Ghayor C, Rey A, Caverzasio J. Prostaglandin-dependent activation of ERK mediates cell proliferation induced by transforming growth factor beta in mouse osteoblastic cells. Bone, 2005, 36: 93-100
    22 Lee YS, Byun J, Kim JA, et al. Monocrotaline-induced pulmonary hypertension correlates with upregulation of connective tissue growth factor expression in the lung. Exp Mol Med, 2005, 37: 27-35
    23 Shan-hong GAO, Xiao-li, Li ZHANG, et al. Effect of nifedipine on proliferation of pulmonary artery smooth muscle cells. Journal of Shenyang Pharmaceutical University, 2004, 21: 442-446
    24 Lee SL, Wang WW, Finlay GA, et al. Serotonin stimulates mitogen-activated protein kinase activity through the formation of superoxide anion. Am J Physiol Lung Cell Mol Physiol, 1999, 277: 282-291
    25 Liu JQ, Sham JS, Shimoda LA, et al. Hypoxic constriction and reactive oxygen species in porcine distal pulmonary arteries. Am J Physiol Lung Cell Mol Physiol, 2003, 285: 322-333
    26 Marshall C, Mamary AJ, Verhoeven AJ, et al. Pulmonary artery NADPH-oxidase is activated in hypoxic pulmonary vasoconstriction. Am J Respir Cell Mol Biol, 1996, 15: 633-644
    1 Nakamura M. Chronic hypoxic pulmonary hypertension. Nippon Rinsho, 2001, 59(6): 1181-1185
    2 Suzuki YJ, Day RM, Tan CC, et al. Activation of GATA-4 by serotonin in pulmonary artery smooth muscle cells. J Biol Chem, 2003, 278(19): 17525-17531
    3 Wang CC, Gurevich I, Draznin B. Insulin affects vascular smooth muscle cell phenotype and migration via distinct signaling pathways. Diabetes, 2003, 52(10): 2562-2569
    4 Xu SZ, Zhang Y, Ren JY, et al. Effects of berberine on L- and T- type calcium channels in guinea pig ventricular myocytes. Acta Pharmacol Sina, 1997, 18(6): 515-518
    5 Li ZC, Zhang FQ, Song JC, et al. Therapeutic effects of DCDDP, a calcium channel blocker, on chronic pulmonary hypertension in rat. J Appl Physiol, 2002, 92(3): 997-1003
    6 Eddahibi S, Chaouat A, Morrell N, et al. Polymorphism of the serotonin transporter gene and pulmonary hypertension in chronic obstructive pulmonary disease. Circulation, 2003, 108: 1839-1844
    7 Seuwen K, Magnaldo I, Pouysségur J. Serotonin stimulates DNA synthesis in fibroblasts acting through 5-HT1B receptors coupled to a Gi-protein. Nature, 1988, 335(6187): 254-256
    8 Lange-Carter CA, Pleiman CM, Gardner AM, et al. A divergence in the MAP kinase regulatory network defined by MEK kinase and Raf. Science, 1993, 260(5106): 315-319
    9 Mauban JR, Remillard CV, Yuan JX. Hypoxic pulmonary vasoconstriction: role of ion channels. J Appl Physiol, 2005, 98(1): 415-420
    10 Paes-de-Carvalho R, Dias BV, Martins RA, et al. Activation of glutamate receptors promotes a calcium-dependent and transporter-mediated release of purines in cultured avian retinal cells: possible involvement of calcium/ calmodulin-dependent protein kinase II. Neurochem Int,2005,46(6):441-451
    11 Mukai K, Matsushima H, Ishii Y, et al. Effects of calcium on lens epithelial cells in rabbits. Nippon Ganka Gakkai Zasshi, 2006, 110(5): 361-369
    12 Wang C, Wang J, Zhao L, et al. Sildenafil inhibits human pulmonary artery smooth muscle cell proliferation by decreasing capacitative Ca2+ entry. J Pharmacol Sci, 2008, 108(1): 71-78
    13 Li B, Yang L, Shen J, et al. The antiproliferative effect of sildenafil on pulmonary artery smooth muscle cells is mediated via upregulation of mitogen-activated protein kinase phosphatase-1 and degradation of extracellular signal-regulated kinase 1/2 phosphorylation. Anesth Analg, 2007, 105(4): 1034-1041
    14 Li M, Liu Y, Dutt P, et al. Inhibition of serotonin-induced mitogenesis, migration, and ERK MAPK nuclear translocation in vascular smooth muscle cells by atorvastatin. Am J Physiol Lung Cell Mol Physiol, 2007, 293(2): 463-471
    15 Zheng S, Qian Z, Wen N, et al. Crocetin suppresses angiotensin II-induced vascular smooth-muscle cell proliferation through inhibition of ERK1/2 activation and cell-cycle progression. J Cardiovasc Pharmacol, 2007, 50(5): 519-525
    16 Owens GK. Regulation of differentiation of vascular smooth muscle cells. Physiol Rev, 1995, 75(3): 487-517
    17 Landxberg BR, Frishman WH, Lerrick K. Pathophysiology and pharmacological approaches for prevention of coronary artery restensis following coronary artery balloon angioplasty and related procedures. Prog Cardiovasc Dis, 1997, 39(4): 361-398
    18 Amano M, Fukata Y, Kaibuchi K. Regulation and functions of Rho-associated kinase. Exp Cell Res, 2000, 261(1): 44-51
    19 Fang LH, Kwon SC, Zhang YH, et al. Tyrosine kinase participates in vasoconstriction through a Ca(2+)-and myosin light chain phosphorylation independent pathway. FEBS Lett, 2002, 512(1-3): 282-286
    20 Kastan MB, Bartek J. Cell-cycle checkpoints and cancer. Nature, 2004, 432(7015): 316-323
    21 Lee SL, Wang WW, Finlay GA, et al. Serotonin stimulates mitogen-activated protein kinase activity through the formation of superoxide anion. Am J Physiol, 1999, 277(2 pt 1): 282-291
    22 Mizuno S, Kadowaki M, Demura Y, et al. p42/44 Mitogen-activated protein kinase regulated by p53 and nitric oxide in human pulmonary arterial smooth muscle cells. Am J Respir Cell Mol Biol, 2004, 31(2): 184-192
    23 Han HJ, Han JY, Heo JS, et al. ANG II-stimulated DNA synthesis is mediated by ANG II receptor-dependent Ca(2+)/PKC as well as EGF receptor-dependent PI3K/Akt/mTOR/p70S6K1 signal pathways in mouse embryonic stem cells. J Cell Physiol, 2007, 211(3): 618-629
    24 Matsushima H, Mukai K, Yoshida S, et al. Effects of calcium on human lens epithelial cells in vitro. Jpn J Ophthalmol, 2004, 48(2): 97-100
    25 Zhang S, Yang Y, Kone BC, et al. Insulin-stimulated cyclic guanosine monophosphate inhibits vascular smooth muscle cell migration by inhibiting Ca/calmodulin-dependent protein kinase II. Circulation, 2003, 107(11): 1539-1544
    26李映红,欧阳静萍,魏劲波.钙调神经磷酸酶在心血管系统中的作用.中国动脉硬化杂志, 2003, 11(5) : 477
    27 Lee SL, Simon AR, Wang WW, et al. H2O2 signals 5-HT-induced ERK MAP kinase activation and mitogenesis of smooth muscle cells. Am J Physiol Lung Cell Mol Physiol, 2001(3); 281: 646-652
    28 Fukushima M, Nakamuta M, Kohjima M, et al. Fasudil hydrochloride hydrate, a Rho-kinase(ROCK) inhibitor, suppresses collagen production and enhances collagenase activity inhepatic stellate cells. Liver Int, 2005, 25(4): 829-838
    29 Suggs WD, Olson SC, Madnani D, et al. Antisense oligonucleotides to c-fos and c-jun inhibit intimal thickening in a rat vein graft model. Surgery, 1999, 126(2): 443-449
    1 Fanburg BL, Lee SL. A new role for an old molecule: serotonin as a mitogen. Am J Physiol, 1997, 272 (5 Pt 1): L795-806
    2 Egermayer P, Town GI, Peacock AJ. Role of serotonin in the pathogenesis of acute and chronic pulmonary hypertension. Thorax, 1999, 54(2): 161-168
    3 Lee SL, Simon AR, Wang WW, et al. H2O2 signals 5-HT-induced ERK MAP kinase activation and mitogenesis of smooth muscle cells. Am J Physiol Lung Cell Mol Physiol, 2001, 281(3): L646-L652
    4 Amano M, Fukata Y, Kaibuchi K. Regulation and functions of Rhoassociated kinase. Exp Cell Res, 2000, 261(1): 44-51
    5 Brown JH, Del Re DP, Sussman MA. The Rac and Rho hall of fame: a decade of hypertrophic signaling hits. Circ Res, 2006, 98(6): 730-742
    6 Sakurada S, Takuwa N, Sugimoto N, et al. Ca2+-dependent activation of Rho and Rho kinase in membrane depolarization-induced and receptor stimulation-induced vascular smooth muscle contraction. Circ Res, 2003, 93(6): 548-556
    7 Gong MC, Fujihara H, Somlyo AV, et al. Translocation of rhoA associated with Ca2+ sensitization of smooth muscle. J Biol Chem, 1997, 272(16):10704-10709
    8 Wen JK, Han M. Comparative study of induction of iNOS mRNA expression in vascular cells of different species. Biochemistry (Mosc), 2000, 65(12): 1376-1379
    9 Liu Y, Suzuki YJ, Day RM, et al. Rho kinase-induced nuclear translocation of ERK1/ERK2 in smooth muscle cell mitogenesis caused by serotonin. Circ Res, 2004, 95(6): 579-586
    10 Ohtsu H, Mifune M, Frank GD, et al. Signal-crosstalk between Rho/ROCK and c-Jun NH2-terminal kinase mediates migration of vascular smooth muscle cells stimulated by angiotensin II. Arterioscler Thromb Vasc Biol, 2005, 25(9): 1831-1836
    11 Welsh CF, Roovers K, Villanueva J, et al. Timing of cyclin D1 expression within G1 phase is controlled by rho. Nat Cell Biol, 2001, 3(11): 950-957
    12 Kastan MB, Bartek J. Cell-cycle checkpoints and cancer. Nature, 2004, 432(7015): 316-323
    13 Ito M, Nakano T, Erdodi F, et al. Myosin phosphatase: structure, regulation and function. Mol Cell Biochem, 2004, 259(1-2): 197-209
    14 Lange-Carter CA, Pleiman CM, Gardner AM, et al. A divergence in the MAP kinase regulatory network defined by MEK kinase and Raf. Science, 1993, 260(5106), 315-319
    15 Liu Y, Suzuki YJ, Day RM, et al. Rho kinase-induced nuclear translocation of ERK1/ERK2 in smooth muscle cell mitogenesis caused by serotonin. Circ Res, 2004, 95(6): 579-586
    16 Suzuki YJ, Day RM, Tan CC, et al. Activation of GATA-4 by serotonin in pulmonary artery smooth muscle cells. J Biol Chem, 2003, 278(19): 17525-17531
    17 Huang C, Jacobson K, Schaller MD. MAP kinases and cell migration. J Cell Sci, 2004, 117(pt 20): 4619-4628
    18 Suggs WD, Olson SC, Madnani D, et al. Antisense oligonucleotides to c-fos and c-jun inhibit intimal thickening in a rat vein graft model. Surgery, 1999, 126(2): 443-449
    1 Li P, Dietz R, von Harsdorf R. Differential effect of hydrogen peroxide and superoxide anion on apoptosis and proliferation of vascular smooth muscle cells. Circulation, 1997, 96(10): 3602-3609
    2 Rao GN, Berk BC. Active oxygen species stimulate vascular smooth muscle cells growth and proto-oncogene expression. Circ Res, 1992, 70(3): 593-599
    3 Griendling KK, Minieri CA, Ollerenshaw JD, et al. AngiotensinⅡstimulates NADP and NADPH oxidase activity in cultured vascular smooth muscle cells. Circ Res, 1994, 74(6): 1141-1148
    4 Suzuki YJ, Day RM, Tan CC, et al. Activation of GATA-4 by serotonin in pulmonary artery smooth muscle cells. J Biol Chem, 2003, 278(19): 17525-17531
    5 Liu Y, Suzuki YJ, Day RM, et al. Rho kinase-induced nuclear translocation of ERK1/ERK2 in smooth muscle cell mitogenesis caused by serotonin. Circ Res, 2004, 95(6): 579-586
    6 Launay JM, HervéP, Peoc'h K, et al. Function of the serotonin 5-hydroxytryptamine 2B receptor in pulmonary hypertension.Nat Med, 2002, 8(10): 1129-1135
    7 Lum H, Roebuck KA. Oxidant stress and endothelial cell dysfunction. Am J Physiol Cell Physiol, 2001, 280(4): C719-741
    8 Tabet F, Savoia C, Schiffrin EL, et al. Differential calcium regulation by hydrogen peroxide and superoxide in vascular smooth muscle cells from spontaneously hypertensive rats. J Cardiovasc Pharmacol, 2004, 44(2): 200-208
    9 Manabe S, Okura T, Fukuoka T, et al. Antioxidative effects of azelnidipine on mesangial cell proliferation induced by highly concentrated insulin. Eur J Pharmacol, 2007, 567(3): 252-257
    10 Waypa GB, Guzy R, Mungai PT, et al. Increases in mitochondrial reactive oxygen species trigger hypoxia- induced calcium responses in pulmonaryartery smooth muscle cells. Circ Res, 2006, 99(9): 970-978
    11 Mason RP. Mechanisms of plaque stabilization for the dihydropyridine calcium channel blocker amlodipine: review of the evidence. Atherosclerosis, 2002, 165(2): 191-199
    12 Berkels R, Egink G, Marsen TA, et al. Nifedipine increases endothelial nitric oxide bioavailability by antioxidative mechanisms. Hypertension, 2001, 37(2): 240-245
    13 Hernández RH, Armas-Hernández MJ, Velasco M, et al. Calcium antagonists and atherosclerosis protection in hypertesion. Am J Ther, 2003, 10(6): 409-441
    14 Luoma JS, Stralin P, Marklund SL, et al. Expression of extracellular SOD and iNOS in macrophages and smooth muscle cells in human and rabbit atherosclerotic lesions: colocalization with epitopes characteristic of oxidized LDL and peroxynitritemodified proteins. Arterioscler Thromb Vasc Biol, 1998, 18(2): 157-167
    15 Dumont A, Hehner SP, Hofmann TG, et al. Hydrogen peroxide-induced apoptosis is CD95-independent, requires the release of mitochondria -derived reactive oxygen species and the activation of NF-kappaB. Oncogene, 1999, 18(3): 747-757
    16 Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature, 2001, 414(6865): 813-820
    1 Stenmark KR, McMurtry IF. Vascular remodeling versus vasoconstriction in chronic hypoxic pulmonary hypertension: A time for reappraisal. Cir Res, 2005, 97(2): 95-98
    2 Hoeper MM, Rubin LJ. Update in pulmonary hypertension 2005. Am J Respir Crit Care Med, 2006, 173(5): 499-505
    3 Galie N, Torbicki A, Barst R, et al. Guidelines on diagnosis and treatment of pulmonary arterial hypertension. The task force on diagnosis and treatment of pulmonary arterial hypertension of the European society of cardiology. European Heart Journal, 2004, 25(24): 2243-2278
    4 Budhiraja R, Tuder RM, Hassoun PM. Endothelial dysfunction in pulmonary hypertension. Circulation, 2004, 109(2): 159-165
    5 Opitz CF, Ewert R. Dual ET(A) /ET(B) vs. selective ET(A) endothelin receptor antagonism in patients with pulmonary hypertension. Eur J Clin Invest, 2006, 36 (supp 13): 1-9
    6 Mata-Greenwood E, Grobe A, Kumar S, et al. Cyclic stretch increases VEGF expression in pulmonary arterial smooth muscle cells via TGF-beta1 and reactive oxygen species: a requirement for NAD(P)H oxidase. Am J Physiol Lung Cell Mol Physiol, 2005, 289(2): 288-289
    7 Sobin SS, Chen PC. Ultrastructural changes in the pulmonary arterioles in acute hypoxia pulmonary hypertension in the rat. High Alt Med Biol, 2000, 1(4): 311-322
    8 Rabinovitch M. Molecular pathogenesis of pulmonary arterial hypertension. J Clin Invest, 2008, 118(7): 2372-2379
    9 Gurbanov E, Shiliang X. The key role of apoptosis in the pathogenesis and treatment of pulmonary hypertension. Eur J Cardiothorac Surg, 2006, 30(3): 499-507
    10 Riley DJ, Thakker Varia S. Wilson FJ, et al. Role of proteolysis and apoptosis in regression of pulmonary vascular remodeling. Physiol Res, 2000, 49(5): 577-585
    11 Stenmark KR, Gerasimovskaya E, Nemenoff RA, et al. Hypoxic Activation of Adventitial Fibroblasts: Role in Vascular Remodeling. Chest, 2002, 122(6 Suppl): 326S-334S
    12 Short M, Nemenoff RA, Zawada WM, et al. Hypoxia induces differentiation of pulmonary artery adventitial fibroblasts into myofibroblasts. Am J Physiol Cell Physiol, 2004, 286(2): C416-C425
    13 Newman JH, Fanburg BL, Archer SL, et al. Pulmonary arterialhypertension: future directions: Report of a national heart, lung and blood institute/office of rare diseases work shop. Circulation, 2004, 109(24): 2947-2952
    14 Cheever KH. An overview of pulmonary arterial hypertension: risks, pathogenesis, clinical manifestations, and management. J Cardiovas Nurs, 2005, 20(2): 108-116
    15 Herve P, Humbert M, Sitbon O, et al. Pathobiology of pulmonary hypertension: The role of platelets and thrombosis. Clin Chest Med, 2001, 22(3): 451-458
    16 Tuder RM, Cool CD , Geraci MW, et al. Prostacyclin synthase expression is decreased in lungs from patients with severe pulmonary hypertension. Am J Respir Crit Care Med, 1999, 159(6): 1925-1932
    17 Fisher KA, Serlin DM, Wilson KC, et al. Sarcoidosis - associated pulmonary hypertension: Outcome with long-term epoprostenol treatment. Chest, 2006, 130(5): 1481-1488
    18 Reddy MT, Patel H, Ventura HO. Successful transition from intravenous to inhaled prostacyclin in a patient with pulmonary hypertension and right ventricular failure. Congest Heart Fail, 2008, 14(5): 285-286
    19 Christman BW, McPherson CD, Newman JH, et al. Animbalance between the excretion of thromboxane and prostacyclin metabolites in pulmonary hypertension. N Engl J Med, 1992, 327(2): 70-75
    20 Browner NC, Dey NB, Bloch KD, et al. Regulation of cGMP-dependent protein kinase expression by soluble guanylyl cyclase in vascular smooth muscle cells. J Biol Chem, 2004, 279(45): 46631-46636
    21 Jacobs A, Preston IR, Gomberg Maitland M. Endothelin receptor antagonism in pulmonary arterial hypertension - a role for selective ET(A) inhibition? Curr Med Res Opin, 2006, 22(12): 2567-2574
    22 Kenyon KW, Nappi JM. Bosentan for the treatment of pulmonary arterial hypertension. Ann Pharmacother, 2003, 37(7): 1055-1062
    23 Hironaka E, HongoM, Sakai A, et al. Serotonin receptor antagonist inhibits monocrotaline induced pulmonary hypertension and prolongssurvival in rats. Cardiovas res, 2003, 60(3): 692-699
    24 Mair KM, MacLean MR, Morecroft I, et al. Novel interactions between the 5-HT transporter, 5-HT1B receptors and Rho kinase in vivo and in pulmonary fibroblasts. Br J Pharmacol, 2008, 155(4): 606-616
    25 Eddahibi S, Humbert M, Fadel E, et al. Serotonin transporter overexpression is responsible for pulmonary artery smooth muscle hyperplasia in primary pulmonary hypertension. J Clin Invest, 2001, 108(8): 1141-1150
    26 Liu Y, Fanburg BL. Serotonin-induced growth of pulmonary artery smooth muscle requires activation of phosphatidylinositol
    3-kinase/serine-threonine protein kinase B/mammalian target of rapamycin/p70 ribosomal S6 kinase 1. Am J Respir Cell Mol Bio, 2006, 34(2): 182-191
    27 Petkov V, Mosgoeller W, Ziesche R, et al. Vasoactive intestinal peptide as a new drug for treatment of primary pulmonary hypertension. J Clin Invest, 2003, 111(9): 1339-1346
    28 Kato K, Okuda M, Ishikawa H, et al. Oligohydramnios and pulmonary hypoplasia: a case in which involvement of an angiotensin II receptor antagonist was suspected. J Obstet Gynaecol Res, 2008, 34(2): 242-246
    29 NichollsM G, Lainchbury JG, LewisL K, et al. Bioactivity of adrenomedullin and proadrenomedullin N-terminal 20 peptide in man. Peptides, 2001, 22 (11): 1745-1752
    30 Christou H, Yoshida A, Arthur V, et al. Increase vascular endothelial growth factor production in the lungs of rats with hypoxia induced pulmonary hypertention. Am Respir Cell Mol Biol, 1998, 18(6): 768-776
    31 Oue T, Yoneda A, Shima H, et al. Increased vascular endothelial growth factor peptide and gene expression in hypoplastic lung in nitrofen induced congenital diaphragmatic hernia in rats. Pediatr Surg Int, 2002, 18(4): 221-226
    32 Barst RJ. PDGF signaling in pulmonary arterial hypertension. J Clin Invest, 2005, 115(10): 2691-2694
    33 Schermuly RT, Eva D, Ghofrani HA, et al. Reversal of experimental pulmonary hypertension by PDGF inhibition. J Clin Invest, 2005, 115(10): 2811-2821
    34 Strange JW, Wharton J, Phillips PG, et al. Recent insights into the pathogenesis and therapeutics of pulmonary hypertension. Clin Sci (Lond), 2002, 102(3): 253-268
    35 Cowan KN, Heilbut A, Humpl T, et al. Complete reversal of fatal pulmonary hypertension in rats by a serine elastase inhibitor. Nat Med, 2000, 6(6): 698-702
    36 Wallner K, Li C, Shah PK, et al. EGF-Like domain of tenascin-C is proapoptotic for cultured smooth muscle cells. Arterioscler Thromb Vasc Biol, 2004, 24(8): 1416-1421
    37 Schermuly RT, Kreisselmeier KP, Ghofrani HA, et al. Chronic sildenafil treatment inhibits monocrotaline-induced pulmonary hypertension in rats. Am J Respir Crit Care Med, 2004, 169(1): 39-45
    38 Fernandez Paltron C, Radomski MW, Davidge ST. Vascular matyix metalloproteinase-2 cleaves big endothelin-1 yielding a novel vasoconstrictor. Circ Res, 1999, 85(10): 906-911
    39 Bonnet S, Archer SL. Potassium channel diversity in the pulmonary arteries and pulmonary veins: Implications for regulation of the pulmonary vasculature in health and during pulmonary hypertension. Pharmacol Ther, 2007, 115(1): 56-69
    40 Moudgil R, Michelakis ED, Archer SL. The role of K+ channels in determining pulmonary vascular tone, oxygen sensing, cell proliferation, and apoptosis: Implications in hypoxic pulmonary vasoconstriction and pulmonary arterial hypertension. Microcirculation, 2006, 13(8): 615-632
    41 Platoshyn O,Yu Y, Golovina VA, et al. Chronic hypoxia decreases K channel expression and function in pulmonary artery myocytes. Am J Physiol Lung Cel Mol Physiol, 2001, 280(4): 801-812
    42 Weir EK, Reeve HL, Huang JM, et al. Anorexic agents Aminorex, Fenfluramine and Dexfenfluramine inhibit potassium current in ratpulmonary vascular smooth muscle and cause pulmonary vasoconstriction. Circulation, 1996, 94(9): 2216-2220
    43 Remilard CV, Yuan JX. Activation of K + channels: an essential pathway in programmed cell death. Am J Physiol Lung Cel Mol Physiol, 2004, 286(1): L49-L67
    44 Manban JR, Remillard CV, Yuan JX. Hypoxia pulmonary vasoconstriction: role of ion channels. J Appl Physiol, 2005, 98(1): 415-420
    45 Bakhramov A, Evans AM, Kozlowski RZ. Differential effects of hypoxia on intracellular concentration of myocytes isolated from different regions of the rat pulmonary arterial tree. Exp Physiol, 1998, 83(3): 337-347
    46 Morty RE, Neiman B, Kwap iszewska G, et al. Dysregulated bone morphogenetic protein signaling in monocrotaline-lnduced pulmonary arterial hypertension. Arterioscler Thromb Vasc Biol, 2007, 14(2): 254-259
    47 Morrel NW. Pulmonary hypertension due to BMPR2 Mutation: a new paradigm for tissue remodeling? Proc Am Thorac Soc, 2006, 3(8): 680-686
    48 Adnot S, Eddahibi S. Genetics and physiopathology of primary or secondary pulmonary artery hypertension. Bull Acad Natl Med, 2003, 187(8): 1529-1542
    49 Lane KB, Machado RD, Pauciulo MW, et al. Heterozygous germline mutations in BMPR2 encoding a TGF- beta recepor, cause familial primary pulmonary hypertension. The international PPH consortium. Nat Genet, 2000, 26(1): 81-84
    50 Yang Y, Long L, Southwood M, et al. Dysfunctional Smad signaling contributes to abnormal smooth muscle cell proliferation in familial pulmonary arterial hypertension. Circ Res, 2005, 96(10): 1053-1063
    51 Teichert-Kuliszewska K, Kutryk MJ, Kuliszewski MA. Bone morphogenetic protein receptor-2 signaling promotes pulmonary arterial endothelial cell survival. Circulation Research, 2006, 98(2): 209-217
    52 West J, Fagan K, Steudel W, et al. Pulmonary hypertension in transgenic mice expressing a dominant-negative BMPRII gene in smooth muscle. Circ Res, 2004, 94(8): 1109-1114
    53 Marcos E, Fadel E, Sanchez O, et al. Serotonin-induced smooth muscle hyperplasia in various forms of human pulmonary hypertension. Circ Res, 2004, 94(9): 1263-1270
    54 Guignabert C, Raffestin B, Benferhat R, et al. Serotonin transporter inhibition prevents and reverses monocrotaline-induced pulmonary hypertension in rats. Circulation, 2005, 111(21): 2812-2819
    55 Long L, MacLean MR, Jeffery TK, et al. Serotonin increases susceptibility to pulmonary hypertension in BMPR-2 deficient mice. Circ Res, 2006, 98(6): 818-827
    56 Stenmark KR, Fagan KA, Frid MG. Hypoxia-induced pulmonary vascular remodeling: cellular and molecular mechanisms. Circ Res, 2006, 99(7): 675-691
    57 Sakurada S, Okamoto H, Takuwa N, et al. Rho activation in excitatory agonist stimulated vascular smooth muscle. Am J Physiol Cell Physiol, 2001, 281(2): 571-578
    58 Yamakawa T, Tanaka S, Numaguchi K, et al. Involvement of Rho-kinase in angiotensin II-induced hypertrophy of rat vascular smooth muscle cells. Hypertension, 2000, 35(1 Pt 2): 313-318
    59 Takemoto M, Sun J, Hiroki J, et al. Rho-kinase mediates hypoxia-induced downregulation of endothelial nitric oxide synthase. Circulation, 2002, 106(1): 57-62
    60 Liu Y, Suzuki YJ, Day RM, et al. Rho kinase-induced nuclear translocation of ERK1/ERK2 in smooth muscle cell mitogenesis caused by serotonin. Circ Res, 2004, 95(6): 579-586
    61 Wang Z, Lanner MC, Jin N, et al. Hypoxia inhibits myosin phosphatase in pulmonary arterial smooth muscle cells: role of Rho kinase. Am J Respir Cell Mol Biol, 2003, 29(4): 465-471
    62 Fagan KA, Oka M, Bauer NR, et al. Attenuation of acute hypoxicpulmonary vasoconstriction and hypoxic pulmonary hypertension in mice by inhibition of Rhokinase. Am J Physiol Lung Cell Mol Physiol, 2004, 287(4): L656-664
    63 Takemoto M, Sun J, Hiroki J, et al. Rho kinase mediates hypoxia induced down regulation of endothelial nitric oxide synthase. Circulation, 2002, 106(1): 57-62
    64 Moon SH, Jenkins CM, Mancuso DJ, et al. Smooth muscle cell arachidonic acid release, migration, and proliferation are markedly attenuated in mice null for calcium-independent phospholipase A2beta. J Biol Chem. 2008, 283(49): 33975-33987
    65王颖,李智.肺动脉高压大鼠肺动脉平滑肌细胞ryanodine受体亚型的变化.中国药理学通报, 2000, 16(6): 628-630
    66 Shimoda LA, Sylvester JT, Sham JS. Mobilization of intracellular Ca2+ by endothelin-1 inratintra pulmonary arterial smooth muscle cells. Am J Physiol Lung Cell Mol Physiol, 2000, 278(1): L157-164
    67 Pype JL, Mak JCW, Dupont LJ, et al. Desensitization of the histamine H1-receptor and transcriptional down regulation of histamine H1-receptor gene expression in bovine tracheal smooth muscle. Br J Pharmacol, 1998, 125(7): 1477-1484
    68 Gelband CH, Gelband H. Ca2+ release from intracellular store is an initial step in hypoxic pulmonary vasoconstriction of rat pulmonary artery resistance vessels. Circulation, 1997, 96(10): 3647-3654
    69 Short AD, Bian J, Ghosh TK, et al. Intracellular Ca2+ pool content is linked to control of cell growth. Proc Natl Acad Sci USA, 1993, 90(11): 4986-4990

© 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