缝隙连接在损伤血管修复中作用的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
研究背景
     血管损伤性疾病如动脉粥样硬化、高血压及糖尿病血管并发症的发病率正逐年升高,是仅次肿瘤的的第二大致死因素,严重威胁着人类的健康。机械、炎性等损伤因素损伤血管内皮后可诱导炎性细胞浸入、血管平滑肌细胞增殖,最终导致粥样硬化、斑块破裂及血管再狭窄,是粥样硬化斑块形成的始动因素,也是血管损伤后不良修复反应的主要原因之一。内皮细胞损伤后,在生长因子、化学因子、切应力刺激及炎性介质等因素的作用下,血管平滑肌细胞由血管中层向内膜迁移、增殖、合成及分泌细胞外基质等是血管不良修复、新生内膜过度增生的病理生理学基础,也是动脉粥样硬化、再狭窄等血管损伤性疾病发生、发展的重要环节。抑制血管平滑肌的迁移、增殖是防治血管损伤性疾病的重要措施之一。因此,探讨血管平滑肌迁移、增殖的自身内在机制,从而寻找抑制血管平滑肌细胞迁移、增殖的措施,将会为血管损伤性疾病的防治提供新的策略。
     维持血管壁自身稳定需要血管壁细胞成分(如血管平滑肌细胞和内皮细胞)间的复杂的相互作用。已经证实,促进损伤血管的再内皮化、恢复损伤血管的内皮完整性能够有效抑制血管平滑肌细胞迁移、增殖,可抑制新生内膜的过度增生,最终可减少血管损伤性疾病的发生及抑制其发展。既往认为,血管内膜损伤后内皮修复只有依靠损伤血管内膜边缘的内皮细胞参与再生。近年来,实践证明邻近参与再生的内皮细胞丧失再生能力或修复的范围有限,不能有效修复损伤内皮。因此,探讨影响内皮损伤修复的机制对血管损伤性疾病的防治具有重要意义。
     损伤血管的再内皮化能够抑制血管平滑肌细胞的迁移及增殖,进而减少新生内膜形成、促进损伤血管的良性修复。但其作用的机制目前仍未完全清楚。既往认为主要机制是二者通过自分泌和旁分泌产生多种生物活性物质而相互调节。但在动物实验和体外实验中都发现,即使阻断这些作用途径,血管内皮和平滑肌细胞之间的相互调节作用并未完全消失,这提示血管内皮和平滑肌细胞之间还存在其它的联系通路。
     缝隙连接在维持组织动态平衡、调节细胞生长、发育及分化中具有重要作用。近年来研究发现,Cx在血管损伤性疾病的发生、发展过程中具有重要作用。在体外培养的大隐静脉血管中出现新生内膜过度增生,其中伴有Cx43表达上调。在动脉粥样硬化的早期斑块中Cx表达发生改变。在颈动脉损伤形成新生内膜中Cx43表达升高,而抑制Cx43的表达可抑制新生内膜的过度增生。但是,在这些过程中,Cx是通过形成缝隙连接介导的细胞间直接通讯还是通过Cx独立于缝隙连接的生物学功能来发挥作用,目前仍不清楚。另外,缝隙连接传递的信号介质目前也不清楚。
     研究目的
     缝隙连接介导的细胞间直接通讯可能在血管损伤修复中具有重要作用。本研究拟观察缝隙连接在内皮损伤修复、血管平滑肌细胞增殖及表型转化中的作用,并观察缝隙连接在血管损伤后新生内膜过度增生中的作用。另外,通过建立血管内皮细胞及平滑肌细胞双层共培养模型以观察缝隙连接在内皮细胞与血管平滑肌细胞相互调节中的作用,并初步探讨在内皮细胞与血管平滑肌细胞相互调节的过程中通过缝隙连接传递的信号介质及可能的机制,从而为寻找血管损伤性疾病的防治新策略提供实验依据。
     研究方法
     1.为了探讨内皮缝隙连接在介导细胞间通讯及在损伤血管内皮修复中的作用,本实验采用植块法培养大鼠主动脉内皮细胞,并用抗vWF免疫荧光染色及透射电镜观察来鉴定培养的大鼠主动脉内皮细胞,Annexin V/PI染色及流式细胞检测技术来检测内皮细胞凋亡,用体外血管新生试剂盒分析内皮细胞体外成管能力,细胞免疫荧光染色法检测大鼠主动脉内皮细胞缝隙连接蛋白37、40及47的表达,荧光漂白后恢复技术检测缝隙连接介导的细胞间通讯。机械划痕建立内皮损伤模型,每24h摄像一次以定量分析内皮损伤修复速度,并观察缝隙连接特异性阻断剂18α-甘草次酸(18α-GA)对内皮损伤后修复的影响。
     2.为探讨缝隙连接阻断剂对大鼠血管平滑肌细胞增殖及表型转化的影响,本实验采用贴块法培养大鼠主动脉平滑肌细胞,采用抗SMα-actin免疫荧光染色鉴定平滑肌细胞,细胞免疫荧光染色法及western blot检测大鼠主动脉平滑肌细胞缝隙连接蛋白43的表达,荧光漂白后恢复技术检测缝隙连接介导的细胞间通讯,四唑盐比色法测定细胞增殖能力以及采用逆转录聚合酶链式反应检测平滑肌α-肌动蛋白的表达,并观察缝隙连接特异性阻断剂观察18α-GA对上述指标的影响。
     3.为了观察缝隙连接在调节血管张力及在血管损伤修复中的作用,本实验取大鼠主动脉制成血管环分别测量在18α-GA作用前后血管环对N E和Ach反应性的变化,建立大鼠颈动脉损伤模型,给予生胃酮3mg/kg/d腹腔注射,对照组给予生理盐水腹腔注射(2ml/d),14天后处死动物,取目标血管段,用HE及DAPI-伊文思蓝染色观察新生内膜厚度,细胞免疫荧光染色法及western blot检测靶血管缝隙连接蛋白43的表达。
     4.为了探讨钙离子跨缝隙连接在内皮细胞与平滑肌细胞间的传递及其意义,本实验采用前述的植块法分别培养大鼠主动脉内皮及平滑肌细胞,建立内皮细胞与平滑肌细胞双层共培养模型,并用透射电镜观察两种细胞在transwell insert膜两侧生长情况,然后3H-TdR掺入法观察平滑肌细胞的增殖情况,将细胞负载Fluo-3后,在用ETA (10-5M)和/或ETB(10-5M)受体阻断剂处理一种细胞后用ET-1(10-7M)刺激另一种细胞在激光共聚焦显微镜测定未刺激细胞内荧光强度变化,并观察18α-GA及肝素对上述变化的影响。用RT-PCR测定在上述处理中刺激内皮细胞后平滑肌细胞内c-fos及c-jun mRNA表达的变化。
     主要结果
     1.内皮缝隙连接在介导细胞间通讯及在损伤血管内皮修复中的作用:成功培养了大鼠主动脉内皮细胞,vWF免疫荧光染色阳性、透射电镜观察可见WP小体。在内皮细胞中缝隙连蛋白37、40及47中均有表达。荧光物质能够通过缝隙连接在相邻细胞间进行传递,孤立细胞荧光恢复率显著低于相邻细胞(5.7±0.63 % vs. 82.26±1.68%,P<0.01);而18α-GA能够抑制缝隙连接介导的细胞间通讯,18α-GA组的荧光恢复率显著低于对照组(33.58±1.73% vs. 82.26±1.68%, P<0.01)。18α-GA可抑制内皮细胞的体外成管能力,与正常对照组相比有显著性差异(4.12±0.41 vs. 2.32±0.58, P<0.01)。在50μM浓度下,流式细胞检测结果显示18α-GA并不增加内皮细胞凋亡。内皮划痕宽度在对照组与18α-GA干预组之间无显著性差异(396.57±25.32μm vs. 370.12±19.4μm, P>0.05),在内皮损伤24h后,18α-GA组划痕宽度显著大于对照组(237.38±20.40μm vs. 126.29±21.40μm, P<0.05)。内皮损伤完全愈合时间,18α-GA显著多于对照组(4.2±0.2d vs. 2.6±0.3d,P<0.05)。
     2.缝隙连接阻断剂对大鼠血管平滑肌细胞增殖及表型转化的影响:成功培养了大鼠主动脉平滑肌细胞,SMα-actin免疫荧光染色阳性。大鼠血管平滑肌细胞体外培养3天后可表达缝隙连接蛋白43。荧光物质能够通过缝隙连接在相邻细胞间进行传递,孤立细胞荧光恢复率显著低于相邻细胞(7.3±0.58 % vs. 80.61±6.57 %,P<0.01);而18α-GA能够抑制缝隙连接介导的细胞间通讯,18α-GA组的荧光恢复率显著低于对照组(41.43±7.62% vs. 80.61±6.57%,P<0.05)。18α-GA并不诱导平滑肌细胞表达Cx43蛋白(0.85±0.06 vs 0.83±0.03,P>0.05),但18α-GA可抑制血管平滑肌细胞的增殖(与对照组相比,0.465±0.016 vs. 0.563±0.081,P<0.05),并且可促进平滑肌α-actin mRNA的表达(与对照组相比,1.384±0.13 vs.0.753±0.09,P<0.01)。
     3.缝隙连接在调节血管张力及在血管损伤修复中的作用:单纯给予18α-GA处理血管环并未出现明显的收缩或舒张反应。在对照组中,给予去甲肾上腺素或乙酰胆碱后,血管环发生明显的收缩或舒张反应,而经18α-GA预处理后,去甲肾上腺素或乙酰胆碱引起的血管环收缩或舒张反应幅度显著降低(NE: 0.60±0.03 vs. 0.21±0.04; Ach: 0.15±0.01 vs. 0.62±0.03;P<0.05)。损伤2周生胃酮干预组血管血管新生内膜明显减轻,血管腔狭窄较单纯损伤组轻。用新生内膜细胞核计数的方法评价新生内膜形成情况显示,生胃酮干预组新生内膜细胞核数量显著低于对照组(89±28. 40 vs. 236±15.04,n=5,P<0.01)。在球囊损伤后2周后血管组织切片免疫荧光染色发现,在形成的新生内膜中免疫荧光染色发现缝隙连接蛋白Cx43表达丰富。Western blot结果显示,对照组缝隙连接蛋白Cx43的表达显著高于生胃酮干预组(0.93±0.06 vs. 0.38±0.11, n=3,P<0.01)。
     4.钙离子跨缝隙连接在内皮细胞与平滑肌细胞间的传递:在共培养后第一天,EC/SMC组及SMC/SMC组平滑肌细胞3H-TdR掺入相比并无统计学差异(10900±1320 cpm/well 10430±1200, P>0.05)。第二天,EC/SMC组平滑肌细胞3H-TdR的掺入就明显小于SMC/SMC组(17200±1734 cpm/well vs. 20900±1659 cpm/well ),两者差异未达到统计学标准。到第三天这种差异更加明显,EC/SMC组平滑肌细胞3H-TdR的掺入为25800±1984 cpm/well,SMC/SMC组的则为33070±3569 cpm/well,两者相比有显著性差异(P<0.05)。在用18α-GA预处理24h后,EC/SMC共培养第三天后平滑肌细胞3H-TdR的掺入为明显高于未处理EC/SMC组,两者相比有显著性差异(30500±2650cpm/well vs. 25800±1984 cpm/well ,P<0.05)。在内皮细胞与平滑肌细胞双层共培养体系中,用ET-1刺激上层的内皮细胞,下层平滑肌细胞内游离钙浓度显著升高,与加药前相比有显著差异。加入18α-GA以阻断双层细胞间的肌内皮连接后,可抑制平滑肌细胞内游离钙浓度的升高(P<0.01)。而用肝素处理平滑肌细胞则对细胞内游离钙浓度无显著影响(P>0.05)。用ET-1刺激平滑肌细胞,下层内皮细胞内游离钙浓度显著升高,与加药前相比有显著差异。加入18α-GA以阻断双层细胞间的肌内皮连接后,可抑制平滑肌细胞内游离钙浓度的升高(P<0.01)。用肝素处理内皮细胞后可抑制细胞内游离钙浓度升高的幅度,但不能完全阻断其升高。在刺激内皮细胞后,平滑肌细胞内c-fos及c-jun mRNA表达显著增加,与未刺激内皮细胞相比有显著性差异(c-fos:0.957±0.051 vs. 0.682±0.038; c-jun:0.816±0.054 vs.0.467±0.032;P<0.01)。
     全文结论
     1.内皮细胞间存在缝隙连接介导的细胞间通讯,而这种通讯能够调节内皮损伤后的修复过程。
     2.缝隙连接介导的细胞间直接通讯能够调节平滑肌细胞的增殖能力以及平滑肌细胞由合成型向收缩型的转化。
     3.缝隙连接在生理条件下参与维持及调节血管张力,在病理条件能够促进血管损伤后新生内膜的过度增生,在血管损伤性疾病的发生、发展过程中具有重要作用。
     4.钙离子能跨缝隙连接在内皮细胞与平滑肌细胞间进行流动,是协调内皮细胞与平滑肌细胞功能的重要信号介质。钙离子跨缝隙连接由内皮细胞向平滑肌细胞流动的过程中不需IP3参与,而由平滑肌细胞向内皮细胞流动的过程中需IP3参与。钙离子由内皮细胞经缝隙连接进入平滑肌细胞后能促进平滑肌细胞中c-fos及c-jun的表达。
The prevalence of vascular injury diseases such as atherosclerosis, hypertension and the vascular complication of diabetes are increasing, and become second major cause leading to death after tumor. The injury and dysfunction of endothelial cells (ECs) by mechanical removal and inflammation,which induces a cascade of proinflammatory events resulting in infiltration of monocytic cells and smooth muscle cells(SMCs) proliferation, is a initial event of pathogenesis of atherosclerosis and a major cause of adverse repair response to vascular injury. After ECs injured, under the action of many factors such as growth factors, cytokines, chemical factors, inflammatory mediators and shear stress, vascular SMCs migrate from media to intima, proliferate, synthesize and secrete extracellular matrix. These processes are pathophysiological mechanism of vascular injury adverse repair and neointimal formation, and are also primary elements involving in that vascular injury diseases develop and progress. To inhibit the migration and proliferation of vascular SMCs is of improtance to treat vascular injury diseases. However, knowledge about its mechanism of these processes is still poor. Therefore, to explore itself internal mechanism of migrating and proliferating and to find a measure of inhibiting its migration and proliferation will provide a new strategy for preventing and treating vascular injury diseases.
     The maintenance of vessel wall homeostasis requires cellular interaction and coordination of the behavior of individual cells. It is established that measures to promote the reendothelization of injured vessel could effectively inhibit the migration and proliferation of vascular smooth muscle, subsequently reduce neointimal formation, and finally prevent vascular injury diseases from developing or progressing. In the past, it had been thought that endothelial repair only depended on the regeneration and migration of ECs at the edge of injured endothelium. But recently, it was proved that the adjacent ECs could not regenerate or its regenerative ability was limited and could not effectively repair the injured endothelium. Hence, to study the mechanism of endothelial wound repair is very important for preventing and treating vascular injury diseases.
     Reendothelization of injured vessel could inhibit SMC migration and proliferation, subsequently reduce neointimal formation and promote the benign repair of injured vessel. But the mechanisms still remain unclear. It has been thought that ECs and SMCs could interact through bioactive substances in an autocrine and/or paracrine way. But it is found in vivo and in vitro that interaction between ECs and SMCs didn’t disappear after blocking the pathway of autocrine and/or paracrine. It suggested that other pathway exist between two type cells besides autocrine and paracrine.
     Gap junctions play a key role in the maintenance of tissue homeostasis and the regulation of cellular growth, differentiation, and development. Recently, it was found that connexins is of very importance during the development and progression of vascular injury diseases. It was reported that upregulation of Cx43 protein was found in neointima of saphenous vein cultured in vitro and that the expression of Cx43 protein changed in the early stages of atherosclerosis. However, whether connexins act through the function dependent or independent on direct intercellular communication via gap junction remain unknown. And substances transferred between ECs and SMCs across gap junction are still unclear.
     Objectives
     Intercellular communication via gap junctions may play a key role on vascular injury repair. In this study, we attempted to explore the role of gap junctions on endothelial wound repair, the proliferation and phenotypic transition of SMCs and neointimal formation of vessel after balloon injury. Subsequently, we also tried to investigate if Ca2+ could transfer between ECs and SMCs across gap junctions during the processes of interaction of ECs and SMCs through a coculture system of ECs and SMCs. Lastly, we wanted to study a possible mechanism of Ca2+ transferred from ECs and SMCs across gap junctions would act on SMCs. And we hoped that this study could provide some experimental evidences for the strategy of preventing and treating vascular injury diseases.
     Methods
     1. Role of endothelial gap junctions in intercellular communication and vascular endothelial wound repair: Rat aortic ECs (RAECs) were cultured by explanted rat aortic wall tissue and were identified with cell immunofluorescence staining for vWF and transmission electron microscope. Cell immunofluorescence staining was applied to detect the expressions of connexin (Cx) 37, Cx40 and Cx47 in RAECs. Fluorescence redistribution after photobleaching (FRAP) was used to measure the communications between cells via gap junctions. Cell apoptosis was detected by Annexin V/PI staining combined with flow cytometry (FCM). The monolayer of cultured RAECs was scraped by a mechanical method and the endothelial wound healing rate was quantified by an analysis of the photographs taken every 24h after endothelial cell layers were wounded. Meanwhile, 18α-glycyrrhetinic acid (18α-GA), a specific blocker of gap junction, was administered to observe its effect on endothelial wound repair.
     2. Effect of the blocker of gap junction on phenotypic transition in cultured rat vascular SMCs: Rat aortic SMCs (RASMCs) were cultured by explanted rat aortic wall tissue and identified with cell immunofluorescence staining for SMα-actin. Cell immunofluorescence staining was applied to detect the expressions of connexin (Cx) 43 in RASMCs. FRAP was used to measure the communications between cells via gap junctions. MTT and RT-PCR were used to measure the proliferative capability of RASMCs and the expression of smooth muscle (SM)α-actin respectively. Meanwhile, 18α-glycyrrhetinic acid (18α-GA), a specific blocker of gap junction, was administered to observe its effect on the contents above.
     3. Role of gap junctions on the regulation of vascular tone and the repair of vascular wound: vascular rings from rat Carotid artery were made and used to compare to the changes of vascular response to NE or Ach with or without 18α-GA. The model of vascular injury was established with rat carotid balloon injury. And animals were administrated with intraperitoneal injections of carbenoxolone (3mg/kg.d) in carbenoxolone group or saline (2ml/d) in control group for 2weeks after carotid balloon injury. After 2 weeks, HE staining and DAPI-Evens blue double staining were applied to evaluate the neointimal formation of targeted vessels. And cell immunofluorescence staining and western blot were used to detect protein Cx43 expression on targeted vessels.
     4. To explore the transfer of Ca2+ between endothelia cells and SMCs across gap junctions and its implication: ECs and SMCs were cultured according to the protocols above. A coculture system was made by seeding ECs and SMCs on the both side of transwell insert with millipores respectively and observed with transmission electron microscope. Then, the proliferation of SMCs was evaluated by 3H-TdR incorporation. After loading both type cells with fluo-3 and pretreating ECs with ETB receptor blocker or SMCs with ETA(10-5M) and ETB (10-5M)receptor blocker, one type cells was stimulated with ET-1 and laser confocal scanning microscope was used to observe the change of immuofluorescence intensity in unstimulated other type cells. The effect of 18α-GA or heparin on above was also observed. After stimulating ECs according to above, the expression of c-fos and c-jun mRNA in unstimulated SMCs was detected with RT-PCR.
     Results
     1. Role of endothelial gap junctions in intercellular communication and vascular endothelial wound repair: In cultured RAECs, immunofluorescence staining for vWF was positive and Weibel-Palade bodies were found. Cx37, Cx40 and Cx47 were all expressed in RAECs. Fluorescent dye could only be transferred between conjugated cells, and mean fluorescence recovery rate in isolated cells were significantly lower compared with that in conjugated cells (5.7±0.63 % vs. 82.26±1.68%,P<0.01). Compared with in control group, mean fluorescence recovery rate in 18α-GA group were significant lower (33.58±1.73% vs. 82.26±1.68%, P<0.01). Therefore, 18α-GA could inhibit dye transfer between conjugated cells. FCS also showed that 18α-GA at a concentration of 50μM could not increase the apoptosis of RAECs (2.044 vs. 2.156, P>0.05). The scrape widths of endothelium were similar in two groups at the time of wound. However, at 24h after endothelial wound, the scrape widths in 18α-GA group were significantly bigger than those in control group(237.38±20.40μm vs. 126.29±21.40μm, P<0.05)。The times of the wound need to reach complete recover in 18α-GA group were significantly more than those in control group(4.2±0.2d vs.2.6±0.3d,P<0.05).
     2. Effect of the blocker of gap junction on phenotypic transition in cultured rat vascular SMCs: RASCMCs were cultured successfully with positive immunofluorescence staining for SMα-actin. At 3rd day after cultured, Cx43 was expressed in RASMCs. Fluorescent dye could only be transferred between conjugated cells, and mean fluorescence recovery rate in isolated cells were significantly lower compared with that in conjugated cells[(7.3±0.58 )% vs. ( 80.61±6.57 )%,P<0.01]. Compared to control group, mean fluorescence recovery rate in 18α-GA group were significant lower(41.43±7.62% vs. 80.61±6.57 % , P < 0.05). Therefore, 18α-GA could inhibit dye transfer between conjugate3.d cells. In this experiment, we found that 18α-GA couldn’t induce the expression of Cx43 protein in RASMCs(0.85±0.06 vs 0.83±0.03,P>0.05), but 18α-GA could inhibit the proliferation of RASMCs(0.465±0.016 vs. 0.563±0.081,P<0.05 vs. control group) and promote the expression of SMα-actin in cultured RASMCs(1.384±0.13 vs.0.753±0.09,P<0.01 vs. control group).
     3. Role of gap junctions on the regulation of vascular tone and the repair of vascular wound: 18α-GA alone didn’t cause significantly the response of vascular rings to systole or diastole. In control group, NE or Ach could induce vascular rings to contract or relax, but 18α-GA could inhibit the contraction or relaxation of vascular rings triggered by NE or Ach(NE: 0.60±0.03 vs. 0.21±0.04; Ach: 0.15±0.01 vs. 0.62±0.03; P<0.05). 2 weeks after carotid balloon injury, carbenoxolone could significantly reduce the neointimal formation and the stenosis of blood vessel lumen. When nuclei number of neointima was used to evaluate the neointimal formation, result suggested that nuclei number of neointima in carbenoxolone group was significantly lower than that in control group((89±28. 40 vs. 236±15.04,n=5,P<0.01). The expression of Cx43 protein in neointima was abundant. Western blot result also suggested that the expression of Cx 43 in carbenoxolone group was significantly lower than that in control group(0.93±0.06 vs. 0.38±0.11, n=3,P<0.01).
     4. To explore the transfer of Ca2+ between endothelia cells and SMCs across gap junctions and its implication: At first day after coculture, there was no significantly difference of 3H-TdR incorporation between EC/SMC and SMC/SMC(10900±1320 10430±1200, P>0.05). At second day, 3H-TdR incorporation in EC/SMC and SMC/SMC was 17200±1734 and 20900±1659 respectively. Although there were some difference, it didn’t reach statistical standard. At third day, there was significantly difference on 3H-TdR incorporation between EC/SMC and SMC/SMC (25800±1984 vs. 33070±3569, P<0.05). after pretreating EC/SMC cocultured for 3d with 18α-GA for 24h, 3H-TdR incorporation was significantly higher compared to EC/SMC which was not pretreated((30500±2650l vs. 25800±1984,P<0.05). In the coculture system, [Ca2+]i in SMCs increased following stimulation of endothelia cells with ET-1, and 18α-GA could inhibit the increase of [Ca2+], but heparin not. [Ca2+]i in ECs also increased following stimulation of SMCs with ET-1, and 18α-GA inhibit the increase of [Ca2+]i and heparin could inhibit the increase of [Ca2+]i partly. The expression of c-fos and c-jun mRNA in the unstimulated SMCs increased following stimulation of endothelia cells with ET-1, compared with no stimulation(c-fos:0.957±0.051 vs. 0.682±0.038; c-jun:0.816±0.054 vs.0.467±0.032;P<0.01)。.
     Conclusions
     1. There are intercellular communications via gap junctions among conjugated RAECs and this kind of intercellular communication might regulate the process of endothelial wound repair.
     2. Intercellular communications via gap junctions could influence smooth muscle cell proliferation and phenotypic transition of SMCs from synthetic to contractile state.
     3. Gap junctions participate in maintaining and regulating vascular tone under the physiological condition, and promote the neointimal formation after vascular injury under the pathological condition. Hence, gap junctions play a key role on the development and progression of vascular injury diseases.
     4. Ca2+ can flow between ECs and SMCs across gap junction and is an important signal mediator coordinates the behavior of ECs and SMCs. Ca2+ flow from SMCs to ECs need the involvement of IP3, but which don’t involve in Ca2+ flow from ECs to SMCs. Ca2+ flowing into SMCs from ECs via gap junctions increase the expression of c-fos and c-jun in SMCs.
引文
1. Loehr LR, Rosamond WD, Chang PP, et al. Heart Failure Incidence and Survival (from the Atherosclerosis Risk in Communities Study). Am J Cardiol. 2008,101(7):1016-22.
    2. Melo LG, Pachori AS, Kong D, et al. Molecular and cell-based therapies for protection, rescue, and repair of ischemic myocardium: reasons for cautious optimism. Circulation. 2004,109(20):2386-93.
    3. Zampetaki A, Kirton JP,Xu Q. Vascular Repair by Endothelial Progenitor Cells. Cardiovasc Res. 2008,
    4. Zargham R. Preventing restenosis after angioplasty: a multistage approach. Clin Sci (Lond). 2008,114(4):257-64.
    5. Tesfamariam B. Local vascular toxicokinetics of stent-based drug delivery. Toxicol Lett. 2007,168(2):93-102.
    6. Krotz F, Sohn HY, Klauss V, et al. Intracoronary brachytherapy--clinical state and pathophysiological considerations. Curr Pharm Des. 2005,11(4):421-33.
    7. Jahnke T, Schafer FK, Bolte H, et al. Short-term rapamycin for inhibition of neointima formation after balloon-mediated aortic injury in rats: is there a window of opportunity for systemic prophylaxis of restenosis? J Endovasc Ther. 2005,12(3):332-42.
    8. Parry TJ, Brosius R, Thyagarajan R, et al. Drug-eluting stents: sirolimus and paclitaxel differentially affect cultured cells and injured arteries. Eur J Pharmacol. 2005, 524(1-3):19-29.
    9. Seifert PS, Huibregtse BA, Polovick J, et al. Early vascular response to overlapped paclitaxel-eluting stents in swine coronary arteries. Cardiovasc Revasc Med. 2007,8(4):251-8.
    10. Shin DI, Kim PJ, Seung KB, et al. Drug-eluting stent implantation could be associated with long-term coronary endothelial dysfunction. Int Heart J. 2007,48(5):553-67.
    11. Lim SY, Jeong MH, Hong SJ, et al. Inflammation and delayed endothelization with overlapping drug-eluting stents in a porcine model of in-stent restenosis. Circ J. 2008,72(3):463-8.
    12. Bennett MR. Vascular pathology as a result of drug-eluting stents. Heart. 2007, 93(8):895-6.
    13. Doggrell SA. Sirolimus- or paclitaxel-eluting stents to prevent coronary artery restenosis. Expert Opin Pharmacother. 2004,5(11):2209-20.
    14. Lang NN,Newby DE. Emerging thrombotic effects of drug eluting stents. Arterioscler Thromb Vasc Biol. 2007,27(2):261-2.
    15. van Veen TA, van Rijen HV,Jongsma HJ. Physiology of cardiovascular gap junctions. Adv Cardiol. 2006,42(18-40.
    16. Kipshidze N, Dangas G, Tsapenko M, et al. Role of the endothelium in modulating neointimal formation: vasculoprotective approaches to attenuate restenosis after percutaneous coronary interventions. J Am Coll Cardiol. 2004,44(4):733-9.
    17. Ohno N, Itoh H, Ikeda T, et al. Accelerated reendothelialization with suppressed thrombogenic property and neointimal hyperplasia of rabbit jugular vein grafts by adenovirus-mediated gene transfer of C-type natriuretic peptide. Circulation. 2002,105(14):1623-6.
    18. Haefliger JA, Nicod P,Meda P. Contribution of connexins to the function of the vascular wall. Cardiovasc Res. 2004,62(2):345-56.
    19. Sohl G,Willecke K. Gap junctions and the connexin protein family. Cardiovasc Res. 2004,62(2):228-32.
    20. Deglise S, Martin D, Probst H, et al. Increased connexin43 expression in human saphenous veins in culture is associated with intimal hyperplasia. J Vasc Surg. 2005,41(6):1043-52.
    21. Blackburn JP, Peters NS, Yeh HI, et al. Upregulation of connexin43 gap junctions during early stages of human coronary atherosclerosis. Arterioscler Thromb Vasc Biol. 1995,15(8):1219-28.
    22. Chadjichristos CE, Matter CM, Roth I, et al. Reduced connexin43 expression limits neointima formation after balloon distension injury in hypercholesterolemic mice. Circulation. 2006,113(24):2835-43.
    23. Yeh HI, Lu CS, Wu YJ, et al. Reduced expression of endothelial connexin37 and connexin40 in hyperlipidemic mice: recovery of connexin37 after 7-day simvastatin treatment. Arterioscler Thromb Vasc Biol. 2003,23(8):1391-7.
    24. Severs NJ. Cardiovascular disease. Novartis Found Symp. 1999,219(188-206; discussion 206-11.
    25. Gotlieb AI. Endothelial Regulation of Vascular Repair: Role of bFGF in Paracrine Pathways. Endocr Pathol. 1997,8(2):129-135.
    26. Iyemere VP, Proudfoot D, Weissberg PL, et al. Vascular smooth muscle cell phenotypic plasticity and the regulation of vascular calcification. J Intern Med. 2006, 260(3):192-210.
    27. Tukaj C, Bohdanowicz J,Kubasik-Juraniec J. A scanning electron microscopic study of phenotypic plasticity and surface structural changes of aortal smooth muscle cells in primary culture. Folia Morphol (Warsz). 2002,61(4):191-8.
    28. Matsushita T, Rama A, Charolidi N, et al. Relationship of connexin43 expression to phenotypic modulation in cultured human aortic smooth muscle cells. Eur J Cell Biol. 2007,86(10):617-28.
    29. Isakson BE,Duling BR. Heterocellular contact at the myoendothelial junction influences gap junction organization. Circ Res. 2005,97(1):44-51.
    30.颜培华,李凤芝。大鼠主动脉内皮细胞培养及鉴定方法.军事医学科学院院刊. 1993,17(2):124-126.
    31.王培勇,孙秉庸。培养的肺动脉内皮细胞生长及生化特性的初步观察.第三军医大学学报. 1995,17(3):212-213。
    32. Reits EA,Neefjes JJ. From fixed to FRAP: measuring protein mobility and activity in living cells. Nat Cell Biol. 2001,3(6):E145-7.
    33.潘礼龙,戴敏,王伟。大鼠主动脉内皮细胞原代培养的研究。中国药理学通报. 2007,23(3):410-413。
    34. Suh SH, Vennekens R, Manolopoulos VG, et al. Characterisation of explanted endothelial cells from mouse aorta: electrophysiology and Ca2+ signalling. Pflugers Arch. 1999,438(5):612-20.
    35. Giovanni, E, Mann, et al.内皮细胞和平滑肌细胞氧化应激时Nrf2/ARE信号通路对抗氧化基因表达的调控:与动脉粥样硬化和先兆子痫的关系.生理学报. 2007,59(2):117-127.
    36. Simon AM,McWhorter AR. Role of connexin37 and connexin40 in vascular development. Cell Commun Adhes. 2003,10(4-6):379-85.
    37. Melo LG, Gnecchi M, Pachori AS, et al. Endothelium-targeted gene and cell-based therapies for cardiovascular disease. Arterioscler Thromb Vasc Biol. 2004,24(10):1761-74.
    38. Gumpricht E, Dahl R, Devereaux MW, et al. Licorice compounds glycyrrhizin and
    18beta-glycyrrhetinic acid are potent modulators of bile acid-induced cytotoxicity in rat hepatocytes. J Biol Chem. 2005,280(11):10556-63.
    39. Isakson BE, Kronke G, Kadl A, et al. Oxidized phospholipids alter vascular connexin expression, phosphorylation, and heterocellular communication. Arterioscler Thromb Vasc Biol. 2006,26(10):2216-21.
    40. Arakawa E,Hasegawa K. Benidipine, a calcium channel blocker, regulates proliferation and phenotype of vascular smooth muscle cells. J Pharmacol Sci. 2006,100(2):149-56.
    41. Kiyan J, Kusch A, Tkachuk S, et al. Rosuvastatin regulates vascular smooth muscle cell phenotypic modulation in vascular remodeling: role for the urokinase receptor. Atherosclerosis. 2007,195(2):254-61.
    42. Seyama Y,Wachi H. Atherosclerosis and matrix dystrophy. J Atheroscler Thromb. 2004,11(5):236-45.
    43. Hamaguchi A, Kim S, Ohta K, et al. Transforming growth factor-beta 1 expression and phenotypic modulation in the kidney of hypertensive rats. Hypertension. 1995, 26(1):199-207.
    44. Sobue K, Hayashi K,Nishida W. Molecular mechanism of phenotypic modulation of smooth muscle cells. Horm Res. 1998,50 Suppl 2(15-24.
    45. Kaplan-Albuquerque N, Garat C, Van Putten V, et al. Regulation of SM22 alpha expression by arginine vasopressin and PDGF-BB in vascular smooth muscle cells. Am J Physiol Heart Circ Physiol. 2003,285(4):H1444-52.
    46. Ratajska A, Campbell SE, Cleutjens JP, et al. Angiotensin II and structural remodeling of coronary vessels in rats. J Lab Clin Med. 1994,124(3):408-15.
    47. Frid MG, Shekhonin BV, Koteliansky VE, et al. Phenotypic changes of human smooth muscle cells during development: late expression of heavy caldesmon and calponin. Dev Biol. 1992,153(2):185-93.
    48. Yamamoto M, Yamamoto K,Noumura T. Type I collagen promotes modulation of cultured rabbit arterial smooth muscle cells from a contractile to a synthetic phenotype. Exp Cell Res. 1993,204(1):121-9.
    49. Kanzaki T, Shiina R, Saito Y, et al. Transforming growth factor-beta receptor andfibronectin expressions in aortic smooth muscle cells in diabetic rats. Diabetologia. 1997,40(4):383-91.
    50. Bingley JA, Hayward IP, Campbell JH, et al. Arterial heparan sulfate proteoglycans inhibit vascular smooth muscle cell proliferation and phenotype change in vitro and neointimal formation in vivo. J Vasc Surg. 1998,28(2):308-18.
    51. Schober A,Zernecke A. Chemokines in vascular remodeling. Thromb Haemost. 2007,97(5):730-7.
    52. Yeh HI, Lupu F, Dupont E, et al. Upregulation of connexin43 gap junctions between smooth muscle cells after balloon catheter injury in the rat carotid artery. Arterioscler Thromb Vasc Biol. 1997,17(11):3174-84.
    53. Rama A, Matsushita T, Charolidi N, et al. Up-regulation of connexin43 correlates with increased synthetic activity and enhanced contractile differentiation in TGF-beta- treated human aortic smooth muscle cells. Eur J Cell Biol. 2006,85(5):375-86.
    54. Matchkov VV, Rahman A, Bakker LM, et al. Analysis of effects of connexin-mimetic peptides in rat mesenteric small arteries. Am J Physiol Heart Circ Physiol. 2006,291(1):H357-67.
    55. Dora KA. Cell-cell communication in the vessel wall. Vasc Med. 2001,6(1):43-50.
    56. Hoepfl B, Rodenwaldt B, Pohl U, et al. EDHF, but not NO or prostaglandins, is critical to evoke a conducted dilation upon ACh in hamster arterioles. Am J Physiol Heart Circ Physiol. 2002,283(3):H996-H1004.
    57. Honing ML, Smits P, Morrison PJ, et al. Bradykinin-induced vasodilation of human forearm resistance vessels is primarily mediated by endothelium-dependent hyperpolarization. Hypertension. 2000,35(6):1314-8.
    58. Rubanyi GM. The role of endothelium in cardiovascular homeostasis and diseases. J Cardiovasc Pharmacol. 1993,22 Suppl 4(S1-14.
    59. Edwards DH, Chaytor AT, Bakker LM, et al. Modulation of gap-junction-dependent arterial relaxation by ascorbic acid. J Vasc Res. 2007,44(5):410-22.
    60.韩雅玲,康建,王效增, et al.低浓度脂多糖加速大鼠颈动脉球囊损伤后新生内膜的过度增生.中国病理生理杂志. 2005,21(5):849-854.
    61. Werner N, Junk S, Laufs U, et al. Intravenous transfusion of endothelial progenitor cells reduces neointima formation after vascular injury. Circ Res. 2003,93(2):e17-24.
    62. Brod J. Volume homeostasis and blood pressure regulation. Contrib Nephrol. 1981,25(36-43.
    63. Humphrey JD. Vascular adaptation and mechanical homeostasis at tissue, cellular, and sub-cellular levels. Cell Biochem Biophys. 2008,50(2):53-78.
    64. Wallez Y,Huber P. Endothelial adherens and tight junctions in vascular homeostasis, inflammation and angiogenesis. Biochim Biophys Acta. 2008,1778(3):794-809.
    65. Lee S, Chen TT, Barber CL, et al. Autocrine VEGF signaling is required for vascular homeostasis. Cell. 2007,130(4):691-703.
    66. Semenza GL. New insights into nNOS regulation of vascular homeostasis. J Clin Invest. 2005,115(11):2976-8.
    67. Hui DY. A no-no for NonO and JNK in extracellular matrix homeostasis and vascular stability. Arterioscler Thromb Vasc Biol. 2007,27(8):1677-8.
    68. de Wit C, Wolfle SE,Hopfl B. Connexin-dependent communication within the vascular wall: contribution to the control of arteriolar diameter. Adv Cardiol. 2006,42(268-83.
    69. Liao Y, Day KH, Damon DN, et al. Endothelial cell-specific knockout of connexin 43 causes hypotension and bradycardia in mice. Proc Natl Acad Sci U S A. 2001,98(17):9989-94.
    70. Earley S, Resta TC,Walker BR. Disruption of smooth muscle gap junctions attenuates myogenic vasoconstriction of mesenteric resistance arteries. Am J Physiol Heart Circ Physiol. 2004,287(6):H2677-86.
    71. Sandow SL, Looft-Wilson R, Doran B, et al. Expression of homocellular and heterocellular gap junctions in hamster arterioles and feed arteries. Cardiovasc Res. 2003,60(3):643-53.
    72. Griffith TM, Chaytor AT,Edwards DH. The obligatory link: role of gap junctional communication in endothelium-dependent smooth muscle hyperpolarization. Pharmacol Res. 2004,49(6):551-64.
    73. Larson DM, Haudenschild CC,Beyer EC. Gap junction messenger RNA expression by vascular wall cells. Circ Res. 1990,66(4):1074-80.
    74. Simon AM,McWhorter AR. Vascular abnormalities in mice lacking the endothelial gap junction proteins connexin37 and connexin40. Dev Biol. 2002,251(2):206-20.
    75. Severs NJ, Rothery S, Dupont E, et al. Immunocytochemical analysis of connexinexpression in the healthy and diseased cardiovascular system. Microsc Res Tech. 2001,52(3):301-22.
    76. Gonzalez D, Gomez-Hernandez JM,Barrio LC. Species specificity of mammalian connexin-26 to form open voltage-gated hemichannels. Faseb J. 2006,20(13):2329-38.
    77. Veenstra RD. Determining ionic permeabilities of gap junction channels. Methods Mol Biol. 2001,154(293-311.
    78. Chaytor AT, Bakker LM, Edwards DH, et al. Connexin-mimetic peptides dissociate electrotonic EDHF-type signalling via myoendothelial and smooth muscle gap junctions in the rabbit iliac artery. Br J Pharmacol. 2005,144(1):108-14.
    79. Danenberg HD, Grad E, Swaminathan RV, et al. Neointimal formation is reduced after arterial injury in human crp transgenic mice. Atherosclerosis. 2008,
    80. Sumi M, Sata M, Miura S, et al. Reconstituted high-density lipoprotein stimulates differentiation of endothelial progenitor cells and enhances ischemia-induced angiogenesis. Arterioscler Thromb Vasc Biol. 2007,27(4):813-8.
    81. Losordo DW, Isner JM,Diaz-Sandoval LJ. Endothelial recovery: the next target in restenosis prevention. Circulation. 2003,107(21):2635-7.
    82. Leon MB, Teirstein PS, Moses JW, et al. Localized intracoronary gamma-radiation therapy to inhibit the recurrence of restenosis after stenting. N Engl J Med. 2001,344(4):250-6.
    83. Bittl JA, Sanborn TA, Tcheng JE, et al. Clinical success, complications and restenosis rates with excimer laser coronary angioplasty. The Percutaneous Excimer Laser Coronary Angioplasty Registry. Am J Cardiol. 1992,70(20):1533-9.
    84. Sposito AC,Chapman MJ. Statin therapy in acute coronary syndromes: mechanistic insight into clinical benefit. Arterioscler Thromb Vasc Biol. 2002,22(10):1524-34.
    85. Yeh HI, Lee PY, Su CH, et al. Reduced expression of endothelial connexins 43 and 37 in hypertensive rats is rectified after 7-day carvedilol treatment. Am J Hypertens. 2006,19(2):129-35.
    86. Jiang JX,Gu S. Gap junction- and hemichannel-independent actions of connexins. Biochim Biophys Acta. 2005,1711(2):208-14.
    87. Sandow SL,Hill CE. Incidence of myoendothelial gap junctions in the proximal and distal mesenteric arteries of the rat is suggestive of a role in endothelium-derivedhyperpolarizing factor-mediated responses. Circ Res. 2000,86(3):341-6.
    88. Schmidt VJ, Wolfle SE, Boettcher M, et al. Gap junctions synchronize vascular tone within the microcirculation. Pharmacol Rep. 2008,60(1):68-74.
    89. Goldberg GS, Valiunas V,Brink PR. Selective permeability of gap junction channels. Biochim Biophys Acta. 2004,1662(1-2):96-101.
    90. Locke D, Stein T, Davies C, et al. Altered permeability and modulatory character of connexin channels during mammary gland development. Exp Cell Res. 2004,298(2):643-60.
    91. Lamboley M, Pittet P, Koenigsberger M, et al. Evidence for signaling via gap junctions from smooth muscle to endothelial cells in rat mesenteric arteries: possible implication of a second messenger. Cell Calcium. 2005,37(4):311-20.
    92.薛军辉,张作明,等.内皮细胞和平滑肌细胞在微孔聚碳酸酯膜上共培养模型方法.第四军医大学学报. 2002,23(5):390-393.
    93. Di Luozzo G, Bhargava J,Powell RJ. Vascular smooth muscle cell effect on endothelial cell endothelin-1 production. J Vasc Surg. 2000,31(4):781-9.
    94. Jaulmes A, Janvier B, Andreani M, et al. Autocrine and paracrine transcriptional regulation of type IIA secretory phospholipase A2 gene in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2005,25(6):1161-7.
    95. Aalkaer C,Nilsson H. Vasomotion: cellular background for the oscillator and for the synchronization of smooth muscle cells. Br J Pharmacol. 2005,144(5):605-16.
    96. Ungvari Z, Csiszar A,Koller A. Increases in endothelial Ca(2+) activate K(Ca) channels and elicit EDHF-type arteriolar dilation via gap junctions. Am J Physiol Heart Circ Physiol. 2002,282(5):H1760-7.
    97. Galie N, Manes A,Branzi A. The endothelin system in pulmonary arterial hypertension. Cardiovasc Res. 2004,61(2):227-37.
    98. Blomstrand F, Giaume C, Hansson E, et al. Distinct pharmacological properties of ET-1 and ET-3 on astroglial gap junctions and Ca(2+) signaling. Am J Physiol. 1999,277(4 Pt 1):C616-27.
    99. Taurin S, Dulin NO, Pchejetski D, et al. c-Fos expression in ouabain-treated vascular smooth muscle cells from rat aorta: evidence for an intracellular-sodium-mediated, calcium-independent mechanism. J Physiol. 2002,543(Pt 3):835-47.
    100. Fang LH, Zhang YH, Ma JJ, et al. Inhibitory effects of tetrandrine on the serum- and platelet-derived growth factor-BB-induced proliferation of rat aortic smooth muscle cells through inhibition of cell cycle progression, DNA synthesis, ERK1/2 activation and c-fos expression. Atherosclerosis. 2004,174(2):215-23.
    101. Nishimura J, Kobayashi S, Shikasho T, et al. Platelet-derived growth factor induces c-fos and c-myc mRNA in cultured rat aortic smooth muscle cells irrespective of intracellular Ca2+ levels. Jpn J Pharmacol. 1992,58 Suppl 2(413P.
    102. Rao GN, Alexander RW,Runge MS. Linoleic acid and its metabolites, hydroperoxyoctadecadienoic acids, stimulate c-Fos, c-Jun, and c-Myc mRNA expression, mitogen-activated protein kinase activation, and growth in rat aortic smooth muscle cells. J Clin Invest. 1995,96(2):842-7.
    1. Sohl G,Willecke K. Gap junctions and the connexin protein family. Cardiovasc Res. 2004,62(2):228-32.
    2. Gomes P, Srinivas SP, Van Driessche W, et al. ATP release through connexin hemichannels in corneal endothelial cells. Invest Ophthalmol Vis Sci. 2005,46(4):1208-18.
    3. Yeager M, Unger VM,Falk MM. Synthesis, assembly and structure of gap junction intercellular channels. Curr Opin Struct Biol. 1998,8(4):517-24.
    4. Yeager M,Nicholson BJ. Structure of gap junction intercellular channels. Curr Opin Struct Biol. 1996,6(2):183-92.
    5. Revel JP, Hoh JH, John SA, et al. Aspects of gap junction structure and assembly. Semin Cell Biol. 1992,3(1):21-8.
    6. Hwan Seul K,Beyer EC. Heterogeneous localization of connexin40 in the renal vasculature. Microvasc Res. 2000,59(1):140-8.
    7. Calabrese A, Guldenagel M, Charollais A, et al. Cx36 and the function of endocrine pancreas. Cell Commun Adhes. 2001,8(4-6):387-91.
    8. Personius KE, Chang Q, Mentis GZ, et al. Reduced gap junctional coupling leads to uncorrelated motor neuron firing and precocious neuromuscular synapse elimination. Proc Natl Acad Sci U S A. 2007,104(28):11808-13.
    9. Shelley J, Dedek K, Schubert T, et al. Horizontal cell receptive fields are reduced in connexin57-deficient mice. Eur J Neurosci. 2006,23(12):3176-86.
    10. Branes MC, Contreras JE,Saez JC. Activation of human polymorphonuclear cells induces formation of functional gap junctions and expression of connexins. Med Sci Monit. 2002,8(8):BR313-23.
    11. Stutenkemper R, Geisse S, Schwarz HJ, et al. The hepatocyte-specific phenotype of murine liver cells correlates with high expression of connexin32 and connexin26 but very low expression of connexin43. Exp Cell Res. 1992,201(1):43-54.
    12. Goldberg GS, Valiunas V,Brink PR. Selective permeability of gap junction channels. Biochim Biophys Acta. 2004,1662(1-2):96-101.
    13. Bennett MV, Rubin JB, Bargiello TA, et al. Structure-function studies of voltagesensitivity of connexins, the family of gap junction forming proteins. Jpn J Physiol. 1993,43 Suppl 1(S301-10.
    14. Nielsen PA, Beahm DL, Giepmans BN, et al. Molecular cloning, functional expression, and tissue distribution of a novel human gap junction-forming protein, connexin-31.9. Interaction with zona occludens protein-1. J Biol Chem. 2002,277(41):38272-83.
    15. Sohl G, Nielsen PA, Eiberger J, et al. Expression profiles of the novel human connexin genes hCx30.2, hCx40.1, and hCx62 differ from their putative mouse orthologues. Cell Commun Adhes. 2003,10(1):27-36.
    16. Hormuzdi SG, Pais I, LeBeau FE, et al. Impaired electrical signaling disrupts gamma frequency oscillations in connexin 36-deficient mice. Neuron. 2001,31(3):487-95.
    17. Toyofuku T, Yabuki M, Otsu K, et al. Intercellular calcium signaling via gap junction in connexin-43-transfected cells. J Biol Chem. 1998,273(3):1519-28.
    18. Oyamada M, Oyamada Y, Kaneko T, et al. Regulation of gap junction protein (connexin) genes and function in differentiating ES cells. Methods Mol Biol. 2002,185(63-9.
    19. Theis M, Speidel D,Willecke K. Astrocyte cultures from conditional connexin43- deficient mice. Glia. 2004,46(2):130-41.
    20. Naus CC, Bechberger JF, Zhang Y, et al. Altered gap junctional communication, intercellular signaling, and growth in cultured astrocytes deficient in connexin43. J Neurosci Res. 1997,49(5):528-40.
    21. Nishii K, Kumai M, Egashira K, et al. Mice lacking connexin45 conditionally in cardiac myocytes display embryonic lethality similar to that of germline knockout mice without endocardial cushion defect. Cell Commun Adhes. 2003,10(4-6):365-9.
    22. Hellmann P, Grummer R, Schirrmacher K, et al. Transfection with different connexin genes alters growth and differentiation of human choriocarcinoma cells. Exp Cell Res. 1999,246(2):480-90.
    23. Looft-Wilson RC, Payne GW,Segal SS. Connexin expression and conducted vasodilation along arteriolar endothelium in mouse skeletal muscle. J Appl Physiol. 2004,97(3):1152-8.
    24. Nodin C, Nilsson M,Blomstrand F. Gap junction blockage limits intercellular spreading of astrocytic apoptosis induced by metabolic depression. J Neurochem. 2005, 94(4):1111-23.
    25. Cusato K, Ripps H, Zakevicius J, et al. Gap junctions remain open during cytochrome c-induced cell death: relationship of conductance to 'bystander' cell killing. Cell Death Differ. 2006,13(10):1707-14.
    26. Cronier L, Bastide B, Defamie N, et al. Involvement of gap junctional communication and connexin expression in trophoblast differentiation of the human placenta. Histol Histopathol. 2001,16(1):285-95.
    27. Drumheller PD,Hubbell JA. Local modulation of intracellular calcium levels near a single-cell wound in human endothelial monolayers. Arterioscler Thromb. 1991,11(5):1258-65.
    28. McDonough WS, Johansson A, Joffee H, et al. Gap junction intercellular communication in gliomas is inversely related to cell motility. Int J Dev Neurosci. 1999,17(5-6):601-11.
    29. Sato H, Hagiwara H, Senba H, et al. The inhibitory effect of connexin 32 gene on metastasis in renal cell carcinoma. Mol Carcinog. 2007,
    30. Lewis BM, Pexa A, Francis K, et al. Adenosine stimulates connexin 43 expression and gap junctional communication in pituitary folliculostellate cells. Faseb J. 2006,20(14):2585-7.
    31. Niger C, Geneau G, Fiorini C, et al. Endothelin-1 inhibits human osteoblastic cell differentiation: influence of connexin-43 expression level. J Cell Biochem. 2008,103(1):110-22.
    32. Spinella F, Rosano L, Di Castro V, et al. Endothelin-1 decreases gap junctional intercellular communication by inducing phosphorylation of connexin 43 in human ovarian carcinoma cells. J Biol Chem. 2003,278(42):41294-301.
    33. Johnson TL,Nerem RM. Endothelial connexin 37, connexin 40, and connexin 43 respond uniquely to substrate and shear stress. Endothelium. 2007,14(4-5):215-26.
    34. Cherian PP, Siller-Jackson AJ, Gu S, et al. Mechanical strain opens connexin 43 hemichannels in osteocytes: a novel mechanism for the release of prostaglandin. Mol Biol Cell. 2005,16(7):3100-6.
    35. Inai T, Mancuso MR, McDonald DM, et al. Shear stress-induced upregulation of connexin 43 expression in endothelial cells on upstream surfaces of rat cardiac valves. Histochem Cell Biol. 2004,122(5):477-83.
    36. Cooper CD, Solan JL, Dolejsi MK, et al. Analysis of connexin phosphorylation sites. Methods. 2000,20(2):196-204.
    37. Lampe PD,Lau AF. The effects of connexin phosphorylation on gap junctional communication. Int J Biochem Cell Biol. 2004,36(7):1171-86.
    38. Abdelmohsen K, Patak P, Von Montfort C, et al. Signaling effects of menadione: from tyrosine phosphatase inactivation to connexin phosphorylation. Methods Enzymol. 2004,378(258-72.
    39. Isakson BE, Kronke G, Kadl A, et al. Oxidized phospholipids alter vascular connexin expression, phosphorylation, and heterocellular communication. Arterioscler Thromb Vasc Biol. 2006,26(10):2216-21.
    40. Laird DW. Connexin phosphorylation as a regulatory event linked to gap junction internalization and degradation. Biochim Biophys Acta. 2005,1711(2):172-82.
    41. Sitaramayya A, Crabb JW, Matesic DF, et al. Connexin 36 in bovine retina: lack of phosphorylation but evidence for association with phosphorylated proteins. Vis Neurosci. 2003,20(4):385-95.
    42. Burt JM,Steele TD. Selective effect of PDGF on connexin43 versus connexin40 comprised gap junction channels. Cell Commun Adhes. 2003,10(4-6):287-91.
    43. Moorby CD, Stoker M,Gherardi E. HGF/SF inhibits junctional communication. Exp Cell Res. 1995,219(2):657-63.
    44. Brink PR, Ricotta J,Christ GJ. Biophysical characteristics of gap junctions in vascular wall cells: implications for vascular biology and disease. Braz J Med Biol Res. 2000,33(4):415-22.
    45. Abrams CK, Bennett MV, Verselis VK, et al. Voltage opens unopposed gap junction hemichannels formed by a connexin 32 mutant associated with X-linked Charcot-Marie-Tooth disease. Proc Natl Acad Sci U S A. 2002,99(6):3980-4.
    46. Trexler EB, Bennett MV, Bargiello TA, et al. Voltage gating and permeation in a gap junction hemichannel. Proc Natl Acad Sci U S A. 1996,93(12):5836-41.
    47. Rubin JB, Verselis VK, Bennett MV, et al. A domain substitution procedure and its use to analyze voltage dependence of homotypic gap junctions formed by connexins 26 and
    32. Proc Natl Acad Sci U S A. 1992,89(9):3820-4.
    48. Christie TL, Mui R, White TW, et al. Molecular cloning, functional analysis, and RNAexpression analysis of connexin45.6: a zebrafish cardiovascular connexin. Am J Physiol Heart Circ Physiol. 2004,286(5):H1623-32.
    49. Jiang JX,Gu S. Gap junction- and hemichannel-independent actions of connexins. Biochim Biophys Acta. 2005,1711(2):208-14.
    50. Giovanni, E, Mann, et al.内皮细胞和平滑肌细胞氧化应激时Nrf2/ARE信号通路对抗氧化基因表达的调控:与动脉粥样硬化和先兆子痫的关系.生理学报. 2007,59(2):117-127.
    51. Mesnil M, Rideout D, Kumar NM, et al. Non-communicating human and murine carcinoma cells produce alpha 1 gap junction mRNA. Carcinogenesis. 1994, 15(8):1541-7.
    52. Huang TY, Hanani M, Ledda M, et al. Aging is associated with an increase in dye coupling and in gap junction number in satellite glial cells of murine dorsal root ganglia. Neuroscience. 2006,137(4):1185-92.
    53. Yamasaki H, Krutovskikh V, Mesnil M, et al. Role of connexin (gap junction) genes in cell growth control and carcinogenesis. C R Acad Sci III. 1999,322(2-3):151-9.
    54. Nambara C, Kawasaki Y,Yamasaki H. Role of the cytoplasmic loop domain of Cx43 in its intracellular localization and function: possible interaction with cadherin. J Membr Biol. 2007,217(1-3):63-9.
    55. Le AC,Musil LS. Normal differentiation of cultured lens cells after inhibition of gap junction-mediated intercellular communication. Dev Biol. 1998,204(1):80-96.
    56. Jiang JX,Goodenough DA. Heteromeric connexons in lens gap junction channels. Proc Natl Acad Sci U S A. 1996,93(3):1287-91.
    57. Andrade-Rozental AF, Rozental R, Hopperstad MG, et al. Gap junctions: the "kiss of death" and the "kiss of life". Brain Res Brain Res Rev. 2000,32(1):308-15.
    58. Nicholas TW, Read SB, Burrows FJ, et al. Suicide gene therapy with Herpes simplex virus thymidine kinase and ganciclovir is enhanced with connexins to improve gap junctions and bystander effects. Histol Histopathol. 2003,18(2):495-507.
    59. Tanaka T, Yamasaki H,Mesnil M. Stimulation of intercellular communication of poor-communicating cells by gap-junction-competent cells enhances the HSV-TK/GCV bystander effect in vitro. Int J Cancer. 2001,91(4):538-42.
    60. Robe PA, Princen F, Martin D, et al. Pharmacological modulation of the bystandereffect in the herpes simplex virus thymidine kinase/ganciclovir gene therapy system: effects of dibutyryl adenosine 3',5'-cyclic monophosphate, alpha-glycyrrhetinic acid, and cytosine arabinoside. Biochem Pharmacol. 2000,60(2):241-9.
    61. Figueroa XF, Isakson BE,Duling BR. Connexins: gaps in our knowledge of vascular function. Physiology (Bethesda). 2004,19(277-84.
    62. Pointis G. Connexin43: emerging role in erectile function. Int J Biochem Cell Biol. 2006,38(10):1642-6.
    63. Haefliger JA, Nicod P,Meda P. Contribution of connexins to the function of the vascular wall. Cardiovasc Res. 2004,62(2):345-56.
    64. Chaytor AT, Bakker LM, Edwards DH, et al. Connexin-mimetic peptides dissociate electrotonic EDHF-type signalling via myoendothelial and smooth muscle gap junctions in the rabbit iliac artery. Br J Pharmacol. 2005,144(1):108-14.
    65. Chaytor AT, Martin PE, Edwards DH, et al. Gap junctional communication underpins EDHF-type relaxations evoked by ACh in the rat hepatic artery. Am J Physiol Heart Circ Physiol. 2001,280(6):H2441-50.
    66. Mather S, Dora KA, Sandow SL, et al. Rapid endothelial cell-selective loading of connexin 40 antibody blocks endothelium-derived hyperpolarizing factor dilation in rat small mesenteric arteries. Circ Res. 2005,97(4):399-407.
    67.洪涛,汪阳,蒋丽萍, et al.缝隙连接阻断剂1-庚醇对脑血管痉挛的抑制作用.中华神经外科杂志. 2005,21(4):244-247.
    68. Morio Y, Carter EP, Oka M, et al. EDHF-mediated vasodilation involves different mechanisms in normotensive and hypertensive rat lungs. Am J Physiol Heart Circ Physiol. 2003,284(5):H1762-70.
    69. Yao J, Morioka T,Oite T. PDGF regulates gap junction communication and connexin43 phosphorylation by PI 3-kinase in mesangial cells. Kidney Int. 2000,57(5):1915-26.
    70. Spray DC. Hypertension in connexin40-null mice: a renin disorder. Kidney Int. 2007,72(7):781-2.
    71. Haefliger JA, Demotz S, Braissant O, et al. Connexins 40 and 43 are differentially regulated within the kidneys of rats with renovascular hypertension. Kidney Int. 2001,60(1):190-201.
    72. Haefliger JA, Castillo E, Waeber G, et al. Hypertension differentially affects theexpression of the gap junction protein connexin43 in cardiac myocytes and aortic smooth muscle cells. Adv Exp Med Biol. 1997,432(71-82.
    73. Tribulova N, Okruhlicova L, Novakova S, et al. Hypertension-related intermyocyte junction remodelling is associated with a higher incidence of low-K(+)-induced lethal arrhythmias in isolated rat heart. Exp Physiol. 2002,87(2):195-205.
    74. Haefliger JA,Meda P. Chronic hypertension alters the expression of Cx43 in cardiovascular muscle cells. Braz J Med Biol Res. 2000,33(4):431-8.
    75. Yeh HI, Lee PY, Su CH, et al. Reduced expression of endothelial connexins 43 and 37 in hypertensive rats is rectified after 7-day carvedilol treatment. Am J Hypertens. 2006,19(2):129-35.
    76. Chen HJ, Yao L, Chen TG, et al. Atorvastatin prevents connexin43 remodeling in hypertrophied left ventricular myocardium of spontaneously hypertensive rats. Chin Med J (Engl). 2007,120(21):1902-7.
    77. Krattinger N, Capponi A, Mazzolai L, et al. Connexin40 regulates renin production and blood pressure. Kidney Int. 2007,72(7):814-22.
    78. Haefliger JA, Krattinger N, Martin D, et al. Connexin43-dependent mechanism modulates renin secretion and hypertension. J Clin Invest. 2006,116(2):405-13.
    79. Danik SB, Rosner G, Lader J, et al. Electrical remodeling contributes to complex tachyarrhythmias in connexin43-deficient mouse hearts. Faseb J. 2007,
    80. Shaw RM,Rudy Y. Ionic mechanisms of propagation in cardiac tissue. Roles of the sodium and L-type calcium currents during reduced excitability and decreased gap junction coupling. Circ Res. 1997,81(5):727-41.
    81. Muller A, Schaefer T, Linke W, et al. Actions of the antiarrhythmic peptide AAP10 on intercellular coupling. Naunyn Schmiedebergs Arch Pharmacol. 1997,356(1):76-82.
    82. Kieval RS, Spear JF,Moore EN. Gap junctional conductance in ventricular myocyte pairs isolated from postischemic rabbit myocardium. Circ Res. 1992,71(1):127-36.
    83. Peters NS,Wit AL. Gap junction remodeling in infarction: does it play a role in arrhythmogenesis? J Cardiovasc Electrophysiol. 2000,11(4):488-90.
    84. Wong CW, Christen T, Roth I, et al. Connexin37 protects against atherosclerosis by regulating monocyte adhesion. Nat Med. 2006,12(8):950-4.
    85. Cai W, Ruan LM, Wang YN, et al. Effects of angiotensin II on connexin 43 of VSMCsin arteriosclerosis. J Zhejiang Univ Sci B. 2006,7(8):648-53.
    86. Chadjichristos CE, Derouette JP,Kwak BR. Connexins in atherosclerosis. Adv Cardiol. 2006,42(255-67.
    87. Galkina E,Ley K. Vascular adhesion molecules in atherosclerosis. Arterioscler Thromb Vasc Biol. 2007,27(11):2292-301.
    88. Kwak BR, Mulhaupt F, Veillard N, et al. Altered pattern of vascular connexin expression in atherosclerotic plaques. Arterioscler Thromb Vasc Biol. 2002,22(2): 225-30.
    89. Blackburn JP, Peters NS, Yeh HI, et al. Upregulation of connexin43 gap junctions during early stages of human coronary atherosclerosis. Arterioscler Thromb Vasc Biol. 1995,15(8):1219-28.
    90. Chanson M, Derouette JP, Roth I, et al. Gap junctional communication in tissue inflammation and repair. Biochim Biophys Acta. 2005,1711(2):197-207.
    91. Wong CW, Burger F, Pelli G, et al. Dual benefit of reduced Cx43 on atherosclerosis in LDL receptor-deficient mice. Cell Commun Adhes. 2003,10(4-6):395-400.
    92. Ramachandran S, Xie LH, John SA, et al. A novel role for connexin hemichannel in oxidative stress and smoking-induced cell injury. PLoS ONE. 2007,2(1):e712.
    93. Kwak BR, Veillard N, Pelli G, et al. Reduced connexin43 expression inhibits atherosclerotic lesion formation in low-density lipoprotein receptor-deficient mice. Circulation. 2003,107(7):1033-9.
    94. Plenz G, Ko YS, Yeh HI, et al. Upregulation of connexin43 gap junctions between neointimal smooth muscle cells. Eur J Cell Biol. 2004,83(10):521-30.
    95. Chadjichristos CE, Matter CM, Roth I, et al. Reduced connexin43 expression limits neointima formation after balloon distension injury in hypercholesterolemic mice. Circulation. 2006,113(24):2835-43.
    96. Liao Y, Regan CP, Manabe I, et al. Smooth muscle-targeted knockout of connexin43 enhances neointimal formation in response to vascular injury. Arterioscler Thromb Vasc Biol. 2007,27(5):1037-42.
    1. Loehr LR, Rosamond WD, Chang PP, et al. Heart failure incidence and survival (from the Atherosclerosis Risk in Communities study). Am J Cardiol. 2008,101(7):1016-22.
    2. Zampetaki A, Kirton JP,Xu Q. Vascular repair by endothelial progenitor cells. Cardiovasc Res. 2008,
    3. Melo LG, Pachori AS, Kong D, et al. Molecular and cell-based therapies for protection, rescue, and repair of ischemic myocardium: reasons for cautious optimism. Circulation. 2004,109(20):2386-93.
    4. Zargham R. Preventing restenosis after angioplasty: a multistage approach. Clin Sci (Lond). 2008,114(4):257-64.
    5. Bennett MV, Zheng X,Sogin ML. The connexins and their family tree. Soc Gen Physiol Ser. 1994,49(223-33.
    6. O'Brien J, Bruzzone R, White TW, et al. Cloning and expression of two related connexins from the perch retina define a distinct subgroup of the connexin family. J Neurosci. 1998,18(19):7625-37.
    7. Haefliger JA, Nicod P,Meda P. Contribution of connexins to the function of the vascular wall. Cardiovasc Res. 2004,62(2):345-56.
    8. Jiang JX,Gu S. Gap junction- and hemichannel-independent actions of connexins. Biochim Biophys Acta. 2005,1711(2):208-14.
    9. Hix LM, Lockwood SF,Bertram JS. Upregulation of connexin 43 protein expression and increased gap junctional communication by water soluble disodium disuccinate astaxanthin derivatives. Cancer Lett. 2004,211(1):25-37.
    10. Inai T, Mancuso MR, McDonald DM, et al. Shear stress-induced upregulation of connexin 43 expression in endothelial cells on upstream surfaces of rat cardiac valves. Histochem Cell Biol. 2004,122(5):477-83.
    11. Vine AL,Bertram JS. Upregulation of connexin 43 by retinoids but not by non-provitamin A carotenoids requires RARs. Nutr Cancer. 2005,52(1):105-13.
    12. Pandey NR, Benkirane K, Amiri F, et al. Effects of PPAR-gamma knock-down and hyperglycemia on insulin signaling in vascular smooth muscle cells from hypertensive rats. J Cardiovasc Pharmacol. 2007,49(6):346-54.
    13. Gomes P, Srinivas SP, Vereecke J, et al. Gap junctional intercellular communication in bovine corneal endothelial cells. Exp Eye Res. 2006,83(5):1225-37.
    14. Chadjichristos CE, Matter CM, Roth I, et al. Reduced connexin43 expression limits neointima formation after balloon distension injury in hypercholesterolemic mice. Circulation. 2006,113(24):2835-43.
    15. Carruba G, Cocciadiferro L, Bellavia V, et al. Intercellular communication and human hepatocellular carcinoma. Ann N Y Acad Sci. 2004,1028(202-12.
    16. Essalihi R, Ouellette V, Hao Dao H, et al. Phenotypic Modulation of Vascular Smooth Muscle Cells During Medial Arterial Calcification: a Role for Endothelin? J Cardiovasc Pharmacol. 2004,44(S147-S150.
    17. Hao H, Gabbiani G, Camenzind E, et al. Phenotypic modulation of intima and media smooth muscle cells in fatal cases of coronary artery lesion. Arterioscler Thromb Vasc Biol. 2006,26(2):326-32.
    18. Lien SC, Usami S, Chien S, et al. Phosphatidylinositol 3-kinase/Akt pathway is involved in transforming growth factor-beta1-induced phenotypic modulation of 10T1/2 cells to smooth muscle cells. Cell Signal. 2006,18(8):1270-8.
    19. Hayashi K, Takahashi M, Nishida W, et al. Phenotypic modulation of vascular smooth muscle cells induced by unsaturated lysophosphatidic acids. Circ Res. 2001, 89(3):251-8.
    20. Liu ZP, Wang Z, Yanagisawa H, et al. Phenotypic modulation of smooth muscle cellsthrough interaction of Foxo4 and myocardin. Dev Cell. 2005,9(2):261-70.
    21. Matsushita T, Rama A, Charolidi N, et al. Relationship of connexin43 expression to phenotypic modulation in cultured human aortic smooth muscle cells. Eur J Cell Biol. 2007,86(10):617-28.