首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 109 毫秒
1.
Jiang QS  Huang XN  Yang GZ  Dai ZK  Zhou QX  Shi JS  Wu Q 《生理学报》2005,57(6):742-748
利用野百合碱(monocrotaline,MCT)诱导大鼠右心室肥厚模型和培养乳鼠心肌细胞,研究前列腺素F2α(prostaglandin F2α,PGF2α)在心肌肥厚中的作用及钙调神经磷酸酶(calcineurin,CaN)信号通路征其中的作用。在雄性Sprague-Dawley大鼠中,用MCT(60mg/kg)单次i.p.诱导右心室肥厚,同时用塞来旨布(20mg/kg)预防/治疗给药2周。用病理检测、电镜观察等方法观察心肌肥厚时组织病理改变;EIA试剂盒检测心肌组织PGF2α含量;RT-PCR检测心房钠尿肽(atrial natriuretic peptide,ANP)和CaNmRNA的表达;用蛋白免疫印迹法检测CaN及其下游因了NFAT3和GATA4蛋门质的表达。以心肌细胞直径、蛋白含量和ANP mRNA表达的变化为0.1μmol/L PGF2α诱导心肌细胞肥大的指标。以CaN mRNA表达作为该信号通路的主要指标,并观察CaN抑制剂环孢素A对PGF2α所致心肌细胞肥人和CaN mRNA表达的影响。结果显示:MCT注射2周(M2W组),右心室肥厚指数(RVHI)、右心室/体重比及肺重/体重比分别增加了47%、53%和118%;注射后4周(M4W组)增加了64%、94%及156%。电镜观察发现右心室组织损伤。同时,右心室组织PGF2α含量在M2W和M4W组分别增加了44%和51%,与RVHI、ANP和CaN的mRNA表达,及CaN/NFAT3/GATA4的蛋白质表达均呈正相关。环氧酶抑制剂塞来昔布预防和治疗给药均明显改善MCT诱导的组织病理学改变。在高体细胞培养中,PGF2α(0.1μmol/L)明显使心肌细胞增大,蛋白质含量增加,ANP和CaN mRNA表达增强:同时,CaN抑制剂环孢素A明显抑制PGF2α诱导的心肌细胞肥大和CaN mRNA表达。上述结果提示:心肌组织局部PGF2α参与了MCT诱导的心肌肥厚过程,CaN信号通路是其细胞内信号转导通路之一。  相似文献   

2.
林瑶  牛勃  解军  颜真 《生命科学研究》2006,10(3):224-227
采用差速贴壁法体外原代培养大鼠心肌细胞;NPY刺激培养的心肌细胞增殖;RNA干涉特异性抑制CaN的活性,阻断NPY刺激的心肌细胞中Ca2 /CaM-CaN信号转导通路;观察对CaN活性、表达水平和心肌细胞蛋白合成速率的变化.实验结果显示NPY可增加心肌细胞的CaN活性和表达,加快细胞内蛋白合成速率.RNA干涉抑制CaN活性后,明显降低NPY刺激的蛋白合成速率.CaN参与了NPY刺激的心肌细胞增殖,RNA干涉通过抑制CaN的活性可阻断N PY诱导的心肌细胞肥大Ca2 /CaM-CaN通路.  相似文献   

3.
目的:研究在压力负荷下辛伐他汀对钙调神经磷酸酶介导的心肌肥大的影响.方法:选用SD大鼠随机分为3组:假手术组(n=10)、单纯模型组(n=10)和辛伐他汀组(n=10).大鼠通过腹主动脉缩窄建立压力超负荷模型,8周后测定左室重量指数,B超检测左室形态结构,Westernblot检测心肌CaN蛋白表达,RT-PCR法检测心肌CaNmRNA水平.结果:①单纯模型组和辛伐他汀组心肌肥厚指数明显高于假手术组,辛伐他汀组心肌肥厚指数明显低于单纯模型组(P<0.05).②单纯模型组和辛伐他汀组心肌CaN蛋白及CaNmRNS表达水平高于假手术组(P<0.05),辛伐他汀组低于单纯模型组(P<0.05).结论:辛伐他汀可能参与干预钙调神经磷酸酶介导的通路从而抑制心肌肥厚的形成.  相似文献   

4.
Sheng HZ  Zhang JN  Yang D  Yang GP  Xu JD  Chen XJ 《生理学报》2003,55(5):594-598
本文观察了钙调神经磷酸酶(calcineurin,CaN)在肾血管性高血压大鼠肥厚心肌中的表达和活性以及CaN抑制剂——环孢菌素A(cyclosporine A,CsA)对逆转心肌肥厚的影响。利用两肾一夹肾血管性高血压大鼠心肌肥厚模型,观察大鼠心肌肥厚程度、CaN mRNA和蛋白质表达及CaN活性的改变。结果显示:大鼠左室重与胫骨长度的比值和光镜下心肌细胞横截面积在两肾一夹2月和3月组都较相应假手术组增高(P<0.05),CsA组大鼠左室重与胫骨长度比值、心肌细胞横截面积较两肾一夹2月和3月组均显著下降(P<0.05),与假手术组无显著性差异。大鼠心肌CaN mRNA和蛋白质表达及CaN活性在两肾一夹2月和3月组均高于相应假手术组(P<0.05),在CsA组低于两肾一夹2月和3月组(P<0.05)。这些结果提示,CaN参与肾血管性高血压大鼠心肌肥厚发展,抑制CaN活性可逆转心肌肥厚。  相似文献   

5.
目的:研究钙调神经磷酸酶(CaN)信号通路在肿瘤坏死因子-α(TNF-α)诱导心肌细胞肥大中的作用。方法:Lowry法测心肌细胞蛋白含量;计算机图象分析系统测心肌细胞体积;[3H]-亮氨酸掺入法测心肌细胞蛋白合成;Till阳离子测定系统观察胞内[Ca2+]i瞬变;Western blot法测定CaN的表达。结果:①CaN特异性抑制剂CsA(0.2μmol/L)明显抑制TNF-α(100μg/L)诱导的心肌细胞蛋白含量、蛋白合成和细胞体积增大,但对正常心肌细胞生长无影响。②CaN特异性抑制剂CsA(0.2μmol/L)明显降低TNF-α诱导的心肌细胞内钙离子浓度([Ca2+]i)瞬变幅度增高。③TNF-α明显增强心肌细胞内CaN的表达。结论:TNF-α可能通过引起心肌细胞[Ca2+]i升高,促进CaN表达诱导心肌细胞肥大。  相似文献   

6.
钙调神经磷酸酶的研究进展   总被引:8,自引:0,他引:8  
钙调神经磷酸酶(CaN)是一种受Ca2+/钙调素调节的丝/苏氨酸蛋白磷酸酶,广泛存在于哺乳动物的组织细胞中,作为Ca2+信号下游的一种效应分子,参与多种细胞功能的调节.在T细胞活化的信号传导中起到调节枢纽的作用;在神经递质的释放、突触可塑性方面亦有重要的调节作用.新近的研究表明,CaN在心肌肥厚的发生发展中起到中心作用.对CaN的分子结构、酶学特性、组织分布、信号传导及生物学功能方面的研究进展进行了介绍.  相似文献   

7.
肌纤基因调节因子(myofibrillogenesis regulator1,MR1)是首次从人体骨骼肌cDNA文库中分离得到的基因.以前的研究证明MR1能够介导血管紧张素Ⅱ(AngⅡ)诱导的体内外心肌肥厚效应,但相关分子机制有待进一步阐明.利用腺病毒载体在小鼠中沉默MR1表达,利用基因芯片对比检查了小鼠心肌基因表达谱的变化.结果发现,在AngⅡ诱导心肌肥厚的小鼠,沉默MR1前后出现明显的信号通路方面的变化和基因表达差异,其中沉默MR1后表达降低90%以上的基因有39个,而表达升高10倍以上的基因有5个.在这些基因中,对与心肌肥厚密切相关的基因进行了定量RT-PCR检测,以进一步验证基因芯片的结果,发现沉默MR1后HSP72和硫氧还蛋白1(Trx1)均表达升高,而钙调神经磷酸酶β(CnAβ)和β肌球蛋白(β-myosin)的基因表达则受抑制.这些信号通路和基因均与AngⅡ诱导的心肌肥厚有一定的关系,为揭示MR1在AngⅡ所致心肌肥厚中的作用和分子机制提供了新的证据.  相似文献   

8.
病理性心肌肥厚是心肌细胞受到多种因素刺激后所产生的失代偿性反应,最终可演变为心力衰竭,甚至诱发猝死。鉴于其复杂的病理过程,具体发病机制至今尚未完全阐明,但既有研究已明确有丝分裂原活化蛋白激酶信号通路、Ca~(2+)介导的信号通路、蛋白激酶信号通路、Janus激酶/信号转导子和转录激活子信号通路和MicroRNAs信号通路在调控心肌肥厚的进程中起着至关重要的作用。现就相关信号通路在心肌肥厚发生、进展及预后中所起作用的最新研究进展予以综述。  相似文献   

9.
NF-κB是一种重要的核转录因子,在TNF、IL-1等因子的刺激下通过上调HIF-1α、IL-1β、IFN-γ等的表达来调节细胞增殖、凋亡、炎症、免疫应答等病理生理过程。近年来已有大量研究表明NF-κB的激活与病理性心肌肥厚的发生密切相关。病理性心肌肥厚是指由于病理性刺激如血流动力学超负荷、心肌细胞损伤等导致的心肌肥厚,是心血管疾病发生过程中的一种重要病理生理表现。本文重点阐述了NF-κB在病理性心肌肥厚发展过程中的作用以及相关的信号通路和分子机制,为心血管疾病的临床治疗提供新思路。  相似文献   

10.
糖尿病心肌病相关信号通路的研究进展   总被引:1,自引:0,他引:1  
尹茂山  牟艳玲 《生命科学》2014,(10):1084-1089
糖尿病心肌病是一种独立、特异的心肌病,与糖尿病患者发生心力衰竭和死亡率升高密切相关。高血糖引起的心血管并发症涉及心肌病变和血管病变、心肌细胞结构的改变、信号通路和炎症因子的改变等,导致心肌纤维化、心肌肥厚、心脏肥大、心力衰竭和心律失常。综述了糖尿病心肌病发病机制中研究较多的几条信号通路,探究各信号通路在糖尿病心肌病发生、发展过程中对心脏的保护(损伤)作用的相关研究进展。  相似文献   

11.
The PTEN/PI3K signaling pathway regulates a vast array of fundamental cellular responses. We show that cardiomyocyte-specific inactivation of tumor suppressor PTEN results in hypertrophy, and unexpectedly, a dramatic decrease in cardiac contractility. Analysis of double-mutant mice revealed that the cardiac hypertrophy and the contractility defects could be genetically uncoupled. PI3Kalpha mediates the alteration in cell size while PI3Kgamma acts as a negative regulator of cardiac contractility. Mechanistically, PI3Kgamma inhibits cAMP production and hypercontractility can be reverted by blocking cAMP function. These data show that PTEN has an important in vivo role in cardiomyocyte hypertrophy and GPCR signaling and identify a function for the PTEN-PI3Kgamma pathway in the modulation of heart muscle contractility.  相似文献   

12.
Isorhamnetin, a flavonoid compound extracted from the Chinese herb Hippophae rhamnoides L., is well known for its anti-inflammatory, anti-oxidative, anti-adipogenic, anti-proliferative, and anti-tumor activities. However, the role of isorhamnetin in cardiac hypertrophy has not been reported. The aims of the present study were to find whether isorhamnetin could alleviate cardiac hypertrophy and to define the underlying molecular mechanisms. Here, we investigated the effects of isorhamnetin (100 mg/kg/day) on cardiac hypertrophy induced by aortic banding in mice. Cardiac hypertrophy was evaluated by echocardiographic, hemodynamic, pathological, and molecular analyses. Our data demonstrated that isorhamnetin could inhibit cardiac hypertrophy and fibrosis 8 weeks after aortic banding. The results further revealed that the effect of isorhamnetin on cardiac hypertrophy was mediated by blocking the activation of phosphatidylinositol 3-kinase–AKT signaling pathway. In vitro studies performed in neonatal rat cardiomyocytes confirmed that isorhamnetin could attenuate cardiomyocyte hypertrophy induced by angiotensin II, which was associated with phosphatidylinositol 3-kinase–AKT signaling pathway. In conclusion, these data indicate for the first time that isorhamnetin has protective potential for targeting cardiac hypertrophy by blocking the phosphatidylinositol 3-kinase–AKT signaling pathway. Thus, our study suggests that isorhamnetin may represent a potential therapeutic strategy for the treatment of cardiac hypertrophy and heart failure.  相似文献   

13.
Pressure overload-induced cardiac hypertrophy results in a pathological type of hypertrophy with activation of signaling cascades like the extracellular signal-regulated kinase (ERK) pathway, which promotes negative cardiac remodeling and decreased contractile function. In contrast, thyroid hormone mediates a physiological type of hypertrophy resulting in enhanced contractile function. In addition, thyroid hormone action is diminished in pressure overload-induced cardiac hypertrophy. We hypothesized that thyroid hormone status modulates ERK activity and that administration of thyroid hormone could alter the activity of this kinase in cardiac hypertrophy induced by pressure overload. ERK is activated by phosphorylation; accordingly, we investigated phosphorylation of ERK in hearts of control, hypothyroid, and hyperthyroid mice. In addition, the effect of T3 treatment on ERK phosphorylation in hypertrophied hearts from transverse aortic-constricted (TAC) mice was investigated. Results showed that phosphorylated ERK (p-ERK) was decreased by 25% in hyperthyroid mice. In contrast, hypothyroid mice presented increased p-ERK by 80%. TAC mice presented a greater than fourfold increase of p-ERK compared with control mice. Interestingly, T3 administration dramatically canceled TAC-induced ERK phosphorylation (36% lower compared with control). Raf-1 is upstream of the ERK pathway. TAC mice presented a 45% increase in phospho-Raf-1 (Ser338). T3 treatment inhibited this effect of pressure overload and further decreased p-Raf-1 (Ser338) by 37%, compared with control. Overexpression of thyroid hormone receptor-α in cultured cardiomyocytes potentiated the inhibitory effect of T3 on ERK phosphorylation. We concluded that thyroid hormone has an inhibitory effect on the Raf-1/ERK pathway. Furthermore, treatment of TAC mice with T3 inhibited Raf-1/ERK pathway by a thyroid hormone receptor-dependent mechanism.  相似文献   

14.
Hong HM  Song EJ  Oh E  Kabir MH  Lee C  Yoo YS 《Proteomics》2011,11(2):283-297
It is well known that the two chemical compounds endothelin-1 (ET-1) and isoproterenol (ISO) can individually induce cardiac hypertrophy through G protein-coupled receptors in cardiomyocytes. However, the cardiac hypertrophy signaling pathway activated by ET-1 and ISO is not well defined. Therefore, we investigated the protein expression profile and signaling transduction in HL-l cardiomyocyte cells treated with ET-1 and ISO. Following separation of the cell lysates by using 2-DE and silver staining, we identified 16 protein spots that were differentially expressed as compared to the controls. Of these 16 spots, three changed only after treatment with ET-1, whereas four changed only after treatment with ISO, suggesting that these two stimuli could induce different signaling pathways. In order to reveal the differences between ET-1- and ISO-induced signaling, we studied the different events that occur at each step of the signaling pathways, when selected biocomponents were blocked by inhibitors. Our results indicated that ET-1 and ISO used different pathways for phosphorylation of glycogen synthase kinase-3β (GSK3β). ET-1 mainly used the mitogen-activated protein kinase and phosphatidylinositol-3-kinase/AKT pathways to activate GSK3β, whereas under ISO stimulation, only the phosphatidylinositol-3-kinase/AKT pathway was required to trigger the GSK3β pathway. Furthermore, the strength of the GSK3β signal in ISO-induced cardiac hypertrophy was stronger than that in ET-1-induced cardiac hypertrophy. We found that these two agonists brought about different changes in the protein expression of HL-1 cardiomyocytes through distinct signaling pathways even though the destination of the two signaling pathways was the same.  相似文献   

15.
Increased oxidative stress has been associated with the pathogenesis of chronic cardiac hypertrophy and heart failure. Since allicin suppresses oxidative stress in vitro and in vivo, we hypothesized that allicin would inhibit cardiac hypertrophy through blocking oxidative stress-dependent signaling. We examined this hypothesis using primary cultured cardiac myocytes and fibroblasts and one well-established animal model of cardiac hypertrophy. Our results showed that allicin markedly inhibited hypertrophic responses induced by Ang II or pressure overload. The increased reactive oxygen species (ROS) generation and NADPH oxidase activity were significantly suppressed by allicin. Our further investigation revealed this inhibitory effect on cardiac hypertrophy was mediated by blocking the activation of ROS-dependent ERK1/2, JNK1/2 and AKT signaling pathways. Additional experiments demonstrated allicin abrogated inflammation and fibrosis by blocking the activation of nuclear factor-κB and Smad 2/3 signaling, respectively. The combination of these effects resulted in preserved cardiac function in response to cardiac stimuli. Consequently, these findings indicated that allicin protected cardiac function and prevented the development of cardiac hypertrophy through ROS-dependent mechanism involving multiple intracellular signaling.  相似文献   

16.
Breviscapine is a mixture of flavonoid glycosides extracted from the Chinese herbs. Previous studies have shown that breviscapine possesses comprehensive pharmacological functions. However, very little is known about whether breviscapine have protective role on cardiac hypertrophy. The aim of the present study was to determine whether breviscapine attenuates cardiac hypertrophy induced by angiotensin II (Ang II) in cultured neonatal rat cardiac myocytes in vitro and pressure‐overload‐induced cardiac hypertrophy in mice in vivo. Our data demonstrated that breviscapine (2.5–15 µM) dose‐dependently blocked cardiac hypertrophy induced by Ang II (1 µM) in vitro. The results further revealed that breviscapine (50 mg/kg/day) prevented cardiac hypertrophy induced by aortic banding as assessed by heart weight/body weight and lung weight/body weight ratios, echocardiographic parameters, and gene expression of hypertrophic markers. The inhibitory effect of breviscapine on cardiac hypertrophy is mediated by disrupting PKC‐α‐dependent ERK1/2 and PI3K/AKT signaling. Further studies showed that breviscapine inhibited inflammation by blocking NF‐κB signaling, and attenuated fibrosis and collagen synthesis through abrogating Smad2/3 signaling. Therefore, these findings indicate that breviscapine, which is a potentially safe and inexpensive therapy for clinical use, has protective potential in targeting cardiac hypertrophy and fibrosis through suppression of PKC‐α‐dependent signaling. J. Cell. Biochem. 109: 1158–1171, 2010. © 2010 Wiley‐Liss, Inc.  相似文献   

17.
18.
Transforming growth factor-β family cytokines have diverse actions in the maintenance of cardiac homeostasis. Follistatin-like 3 (Fstl3) is an extracellular regulator of certain TGF-β family members, including activin A. The aim of this study was to examine the role of Fstl3 in cardiac hypertrophy. Cardiac myocyte-specific Fstl3 knock-out (KO) mice and control mice were subjected to pressure overload induced by transverse aortic constriction (TAC). Cardiac hypertrophy was assessed by echocardiography and histological and biochemical methods. KO mice showed reduced cardiac hypertrophy, pulmonary congestion, concentric LV wall thickness, LV dilatation, and LV systolic dysfunction after TAC compared with control mice. KO mice displayed attenuated increases in cardiomyocyte cell surface area and interstitial fibrosis following pressure overload. Although activin A was similarly up-regulated in KO and control mice after TAC, a significant increase in Smad2 phosphorylation only occurred in KO mice. Knockdown of Fstl3 in cultured cardiomyocytes inhibited PE-induced cardiac hypertrophy. Conversely, adenovirus-mediated Fstl3 overexpression blocked the inhibitory action of activin A on hypertrophy and Smad2 activation. Transduction with Smad7, a negative regulator of Smad2 signaling, blocked the antihypertrophic actions of activin A stimulation or Fstl3 ablation. These findings identify Fstl3 as a stress-induced regulator of hypertrophy that controls myocyte size via regulation of Smad signaling.  相似文献   

19.
Obesity is associated with heart failure and cardiac hypertrophy. Adiponectin has been shown to play a protective role for cardiovascular diseases. The β-catenin signaling pathway is deeply involved in cardiac hypertrophy. However, the effect of adiponectin on β-catenin signaling has not been investigated in cardiac hypertrophy. Present study aimed to clarify the involvement of adiponectin and β-catenin signaling pathway in the mouse model of angiotensin II (AngII)-induced cardiac hypertrophy. In hearts of Wild type (WT) mice, AngII dose-dependently augmented cytosolic β-catenin protein level. WT and adiponectin knockout (Adipo-KO) mice were administered with AngII at 2.4 mg/kg/day for 14 days and were also injected with adenovirus expressing the adiponectin (Ad-Adipo) or the β-galactosidase (Ad-βgal). Cardiac mRNA levels relating to hypertrophy and β-catenin signaling were increased in Adipo-KO mice and these changes were reversed by Ad-Adipo. Phosphorylation of Akt was increased in Adipo-KO mice and such increases were reversed by Ad-Adipo. Furthermore, the phosphorylation of glycogen synthase kinase 3β (GSK3β) at Ser9 and cytosolic β-catenin level were increased in Adipo-KO mice and they were significantly reduced by Ad-Adipo treatment. Phosphorylation of mammalian target of rapamycin (mTOR) was reduced by Ad-Adipo-mediated adiponectin supplementation in WT and Adipo-KO mice. The current study suggests that adiponectin attenuates AngII-induced cardiac hypertrophic signals partly through Akt/GSK3β/β-catenin and Akt/mTOR pathways.  相似文献   

20.
Tumor necrosis factor superfamily ligands provoke a dilated cardiac phenotype signal through a common scaffolding protein termed tumor necrosis factor receptor-associated factor 2 (Traf2); however, Traf2 signaling in the adult mammalian cardiac hypertrophy is not fully understood. This study was aimed to identify the effect of Traf2 on cardiac hypertrophy and the underlying mechanisms. A significant up-regulation of Traf2 expression was observed in mice failing hearts. To further investigate the role of Traf2 in cardiac hypertrophy, we used cultured neonatal rat cardiomyocytes with gain and loss of Traf2 function and cardiac-specific Traf2-overexpressing transgenic (TG) mice. In cultured cardiomyocytes, Traf2 positively regulated angiotensin II (Ang II)-mediated hypertrophic growth, as detected by [3H]-Leucine incorporation, cardiac myocyte area, and hypertrophic marker protein levels. Cardiac hypertrophy in vivo was produced by constriction of transverse aortic (TAC) in TG mice and their wild-type controls. The extent of cardiac hypertrophy was evaluated by echocardiography as well as by pathological and molecular analyses of heart samples. Traf2 overexpression in the heart remarkably enhanced cardiac hypertrophy, left ventricular dysfunction in mice in response to TAC. Further analysis of the signaling pathway in vitro and in vivo suggested that these adverse effects of Traf2 were associated with the activation of AKT/glycogen synthase kinase 3β (GSK3β). The present study demonstrates that Traf2 serves as a novel mediator that enhanced cardiac hypertrophy by activating AKT/GSK3β signaling.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号