SERCA2

SERCA2
  • 文章类型: Case Reports
    Darier病(DD)是一种常染色体显性疾病,归因于ATP2A2基因的致病性变异,可引起基于角质形成细胞断开和凋亡的孤立皮肤表现。到目前为止,尚未证明DD的系统性表现,尽管神经精神综合征的高发病率表明中枢神经系统受累。我们报道,致病性ATP2A2基因变异c.118G>A可能导致DD患者的心脏受累,由角质形成细胞和心肌细胞断开组成。他们共同的病理途径,仍未报告,通过皮肤和左心室心内膜活检记录,因为在皮肤表现后几十年出现心脏扩张和功能障碍。角质形成细胞的断开与外侧连接处的心肌细胞分离平行。心肌细胞分离与细胞紊乱有关,肌浆网扩张,心肌细胞凋亡增加。临床上,角化过度的皮肤丘疹与胸痛有关,严重的肌肉衰竭,服用氨茶碱后室性心律失常得到改善,一种增强SERCA2蛋白磷酸化的磷酸二酯酶抑制剂。心脏病理变化与皮肤中记录的相似,包括促进心前区疼痛和心律失常的心肌细胞断开。增强SERCA2蛋白磷酸化的磷酸二酯酶抑制剂可以基本上减轻症状。
    Darier disease (DD) is an autosomal dominant disorder due to pathogenic variants of the ATP2A2 gene that causes an isolated skin manifestation based on keratinocyte disconnection and apoptosis. Systemic manifestations of DD have not been demonstrated so far, although a high incidence of neuropsychiatric syndromes suggests an involvement of the central nervous system. We report that the pathogenic ATP2A2 gene variant c.118G>A may cause cardiac involvement in patients with DD, consisting of keratinocyte and cardiomyocyte disconnection. Their common pathologic pathway, still unreported, was documented by both skin and left ventricular endomyocardial biopsies because cardiac dilatation and dysfunction appeared several decades after skin manifestations. Keratinocyte disconnection was paralleled by cardiomyocyte separation at the lateral junction. Cardiomyocyte separation was associated with cell disarray, sarcoplasmic reticulum dilatation, and increased myocyte apoptosis. Clinically, hyperkeratotic skin papules are associated with chest pain, severe muscle exhaustion, and ventricular arrhythmias that improved following administration of aminophylline, a phosphodiesterase inhibitor enhancing SERCA2 protein phosphorylation. Cardiac pathologic changes are similar to those documented in the skin, including cardiomyocyte disconnection that promotes precordial pain and cardiac arrhythmias. Phosphodiesterase inhibitors that enhance SERCA2 protein phosphorylation may substantially attenuate the symptoms.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    内质网(ER)储存大量的钙(Ca2+),ERCa2+的控释调节了无数的细胞功能。虽然已知改变的ERCa2+稳态会诱导ER应激,ERCa2+失衡激活ER应激途径的机制知之甚少。基质相互作用分子STIM1和STIM2是两个结构同源的ER驻留Ca2传感器,可协同调节Ca2通过OraiCa2通道流入细胞质,以进行随后的转录和ERCa2再填充信号。这里,我们证明,在结直肠癌(CRC)中,STIM2而非STIM1的降低与患者预后不良相关.STIM2的丢失导致ERCa2+的SERCA2依赖性增加,增加的蛋白质翻译和转录和代谢重组支持增加的肿瘤大小,入侵,和转移。机械上,STIM2损失以Orai非依赖性方式激活cMyc和ER应激的PERK/ATF4分支。因此,STIM2和PERK/ATF4可用于预后或靶向治疗以抑制CRC肿瘤生长和转移。
    STIM2独立于Orai和SOCE调节ERCa2+稳态。结直肠癌细胞中STIM2下调导致ERCa2+增强,并与患者预后不良相关。STIM2下调诱导结直肠癌中PERK/ATF4依赖性内质网应激。增加的ER压力驱动结直肠癌代谢重编程,增长,和转移。
    The endoplasmic reticulum (ER) stores large amounts of calcium (Ca2+), and the controlled release of ER Ca2+ regulates a myriad of cellular functions. Although altered ER Ca2+ homeostasis is known to induce ER stress, the mechanisms by which ER Ca2+ imbalance activate ER stress pathways are poorly understood. Stromal-interacting molecules STIM1 and STIM2 are two structurally homologous ER-resident Ca2+ sensors that synergistically regulate Ca2+ influx into the cytosol through Orai Ca2+ channels for subsequent signaling to transcription and ER Ca2+ refilling. Here, we demonstrate that reduced STIM2, but not STIM1, in colorectal cancer (CRC) is associated with poor patient prognosis. Loss of STIM2 causes SERCA2-dependent increase in ER Ca2+, increased protein translation and transcriptional and metabolic rewiring supporting increased tumor size, invasion, and metastasis. Mechanistically, STIM2 loss activates cMyc and the PERK/ATF4 branch of ER stress in an Orai-independent manner. Therefore, STIM2 and PERK/ATF4 could be exploited for prognosis or in targeted therapies to inhibit CRC tumor growth and metastasis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    细胞穿透肽(CPPs)是短肽序列,其具有穿过细胞膜并递送货物的能力。尽管CPP以最小的脱靶效应完成这项任务至关重要,在许多情况下,这种行动没有得到强有力的筛选。我们目前研究了常用的CPPsTAT和聚精氨酸Arg9和Arg11是否对细胞Ca2稳态产生脱靶作用。在使用心脏左心室或比目鱼肌的肌细胞和匀浆的实验中,我们观察到与聚精氨酸CPPs孵育后,Ca2再循环到肌浆网(SR)的明显抑制作用。在这两个组织中,SRCa2+泄漏率保持不变,表明从细胞溶质中长期去除Ca2+源于对SRCa2+ATP酶2(SERCA2)的抑制。用TAT处理后没有发生这种抑制作用,或在表达SERCA1的趾长伸肌的制剂中。在过表达单个SERCA同工型的HEK细胞中的实验证实,聚精氨酸孵育特异性抑制SERCA2a和2b的活性,但不是SERCA1或3。SERCA2活性的衰减不依赖于磷脂的存在,基于ELISA的分析揭示了多聚精氨酸与蛋白质的致动器结构域之间的直接相互作用。表面等离子体共振实验证实了SERCA2的该区域内的强结合,以及两种物质之间的缓慢解离。基于这些观察,我们敦促谨慎使用聚精氨酸CPPs。的确,SERCA2在不同的细胞类型中表达,在实验和治疗环境中,应预期SERCA2结合和抑制的广泛后果.
    Cell-penetrating peptides (CPPs) are short peptide sequences that have the ability to cross the cell membrane and deliver cargo. Although it is critical that CPPs accomplish this task with minimal off-target effects, such actions have in many cases not been robustly screened. We presently investigated whether the commonly used CPPs TAT and the polyarginines Arg9 and Arg11 exert off-target effects on cellular Ca2+ homeostasis. In experiments employing myocytes and homogenates from the cardiac left ventricle or soleus muscle, we observed marked inhibition of Ca2+ recycling into the sarcoplasmic reticulum (SR) following incubation with polyarginine CPPs. In both tissues, the rate of SR Ca2+ leak remained unchanged, indicating that protracted Ca2+ removal from the cytosol stemmed from inhibition of the SR Ca2+ ATPase 2 (SERCA2). No such inhibition occurred following treatment with TAT, or in preparations from the SERCA1-expressing extensor digitorum longus muscle. Experiments in HEK cells overexpressing individual SERCA isoforms confirmed that polyarginine incubation specifically inhibited the activity of SERCA2a and 2b, but not SERCA1 or 3. The attenuation of SERCA2 activity was not dependent on the presence of phospholamban, and ELISA-based analyses rather revealed direct interaction between the polyarginines and the actuator domain of the protein. Surface plasmon resonance experiments confirmed strong binding within this region of SERCA2, and slow dissociation between the two species. Based on these observations, we urge caution when employing polyarginine CPPs. Indeed, as SERCA2 is expressed in diverse cell types, the wide-ranging consequences of SERCA2 binding and inhibition should be anticipated in both experimental and therapeutic settings.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    众所周知的胰岛素样生长因子1(IGF1)/IGF-1受体(IGF-1R)信号通路在许多肿瘤中过度表达,因此是癌症治疗的有吸引力的靶标。然而,由于与其他信号的串扰,结果往往令人失望。这里,我们报道,IGF-1R信号通过IGF-1R转位到ER中,以增强肌内质网钙ATP酶2(SERCA2)活性刺激肝细胞癌(HCC)细胞的生长。为了响应配体结合,IGF-1Rβ通过β-arrestin2(β-arr2)转位到ER中。质谱分析确定SERCA2为ERIGF-1Rβ的靶标。SERCA2活性在很大程度上依赖于ERIGF-1Rβ水平的增加。ERIGF-1Rβ磷酸化Tyr990上的SERCA2以增强其活性。SERCA2-Tyr990的突变破坏了ERIGF-1Rβ与SERCA2的相互作用,因此ERIGF-1Rβ不能促进SERCA2活性。SERCA2活性的增强引发了Ca2+ER的扰动,导致自噬增加。Thapsigargin阻断SERCA2和ERIGF-1R之间的相互作用,因此SERCA2活性,导致抑制HCC生长。总之,IGF-1R易位到ER触发Ca2+ER扰动,通过磷酸化HCC中Tyr990增强SERCA2活性。
    The well-known insulin-like growth factor 1 (IGF1)/IGF-1 receptor (IGF-1R) signaling pathway is overexpressed in many tumors, and is thus an attractive target for cancer treatment. However, results have often been disappointing due to crosstalk with other signals. Here, we report that IGF-1R signaling stimulates the growth of hepatocellular carcinoma (HCC) cells through the translocation of IGF-1R into the ER to enhance sarco-endoplasmic reticulum calcium ATPase 2 (SERCA2) activity. In response to ligand binding, IGF-1Rβ is translocated into the ER by β-arrestin2 (β-arr2). Mass spectrometry analysis identified SERCA2 as a target of ER IGF-1Rβ. SERCA2 activity is heavily dependent on the increase in ER IGF-1Rβ levels. ER IGF-1Rβ phosphorylates SERCA2 on Tyr990 to enhance its activity. Mutation of SERCA2-Tyr990 disrupted the interaction of ER IGF-1Rβ with SERCA2, and therefore ER IGF-1Rβ failed to promote SERCA2 activity. The enhancement of SERCA2 activity triggered Ca2+ER perturbation, leading to an increase in autophagy. Thapsigargin blocked the interaction between SERCA2 and ER IGF-1Rβ and therefore SERCA2 activity, resulting in inhibition of HCC growth. In conclusion, the translocation of IGF-1R into the ER triggers Ca2+ER perturbation by enhancing SERCA2 activity through phosphorylating Tyr990 in HCC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    终止密码子连读(TCR)是核糖体继续翻译mRNA超过终止密码子产生C-末端延伸的蛋白质同种型的过程。这里,我们展示了哺乳动物NNATmRNA中的TCR,编码NNAT,一种对神经元分化很重要的蛋白脂质。这是由正则终止密码子上游和下游的顺式作用RNA序列驱动的编程事件。并且受NONO的负调控,一种已知能促进神经元分化的RNA结合蛋白。与规范的同工型NNAT不同,我们确定TCR产品(NNATx)没有显示出可检测的相互作用与SERCA2Ca2+泵,不能增加细胞质Ca2+水平,因此不会增强Neuro-2a细胞中的神经元分化。此外,靶向经典终止密码子下游区域的反义寡核苷酸降低了NNAT的TCR,并增强了Neuro-2a细胞向胆碱能神经元的分化。此外,NNATx缺陷神经-2a细胞,使用CRISPR-Cas9产生,显示细胞质Ca2+水平增加,神经元分化增强。总的来说,这些结果表明NNAT的TCR对神经元分化的调节。重要的是,可以使用合成的反义寡核苷酸调节该过程。
    Termination codon readthrough (TCR) is a process in which ribosomes continue to translate an mRNA beyond a stop codon generating a C-terminally extended protein isoform. Here, we demonstrate TCR in mammalian NNAT mRNA, which encodes NNAT, a proteolipid important for neuronal differentiation. This is a programmed event driven by cis-acting RNA sequences present immediately upstream and downstream of the canonical stop codon and is negatively regulated by NONO, an RNA-binding protein known to promote neuronal differentiation. Unlike the canonical isoform NNAT, we determined that the TCR product (NNATx) does not show detectable interaction with the sarco/endoplasmic reticulum Ca2+-ATPase isoform 2 Ca2+ pump, cannot increase cytoplasmic Ca2+ levels, and therefore does not enhance neuronal differentiation in Neuro-2a cells. Additionally, an antisense oligonucleotide that targets a region downstream of the canonical stop codon reduced TCR of NNAT and enhanced the differentiation of Neuro-2a cells to cholinergic neurons. Furthermore, NNATx-deficient Neuro-2a cells, generated using CRISPR-Cas9, showed increased cytoplasmic Ca2+ levels and enhanced neuronal differentiation. Overall, these results demonstrate regulation of neuronal differentiation by TCR of NNAT. Importantly, this process can be modulated using a synthetic antisense oligonucleotide.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    钙(Ca2+)是中枢神经系统中一种关键的离子第二信使,受到一系列调节机制的控制。包括细胞器Ca2+商店,膜通道和泵,和细胞内Ca2+结合蛋白。毫不奇怪,Ca2+稳态紊乱与神经退行性疾病有关,如阿尔茨海默氏症和帕金森氏症。然而,Ca2稳态的畸变也与具有强烈神经发育成分的神经精神疾病有关,包括自闭症谱系障碍(ASD)注意缺陷多动障碍(ADHD)和精神分裂症(SCZ)。虽然已经广泛研究了质膜Ca2+通道和突触Ca2+结合蛋白,越来越多的证据表明细胞内Ca2+储存的重要作用,例如内质网(ER),神经发育异常。在目前的小型审查中,我们讨论了最近的发现,涉及关键的细胞内Ca2处理调节因子,如sarco-ERCa2ATPase2(SERCA2),ryanodine受体(RyRs),肌醇三磷酸受体(IP3Rs),和小白蛋白(PVALB),在ASD的出现中,SCZ,和ADHD。
    Calcium (Ca2+) comprises a critical ionic second messenger in the central nervous system that is under the control of a wide array of regulatory mechanisms, including organellar Ca2+ stores, membrane channels and pumps, and intracellular Ca2+-binding proteins. Not surprisingly, disturbances in Ca2+ homeostasis have been linked to neurodegenerative disorders, such as Alzheimer\'s and Parkinson\'s diseases. However, aberrations in Ca2+ homeostasis have also been implicated in neuropsychiatric disorders with a strong neurodevelopmental component including autism spectrum disorder (ASD) attention-deficit hyperactivity disorder (ADHD) and schizophrenia (SCZ). While plasma membrane Ca2+ channels and synaptic Ca2+-binding proteins have been extensively studied, increasing evidence suggests a prominent role for intracellular Ca2+ stores, such as the endoplasmic reticulum (ER), in aberrant neurodevelopment. In the context of the current mini-review, we discuss recent findings implicating critical intracellular Ca2+-handling regulators such as the sarco-ER Ca2+ ATPase 2 (SERCA2), ryanodine receptors (RyRs), inositol triphosphate receptors (IP3Rs), and parvalbumin (PVALB), in the emergence of ASD, SCZ, and ADHD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    血管紧张素转化酶(ACE)通过其N末端位点(ACE-N)将N-乙酰基-丝氨酰-天冬氨酰-赖氨酰-脯氨酸(Ac-SDKP)水解成非活性片段。我们以前表明,Ac-SDKP介导ACE抑制剂的心脏作用。由敲除ACE-N引起的内源性Ac-SDKP的生物利用度增加是否也改善了心肌梗死(MI)引起的心力衰竭(HF)的心脏功能尚不清楚。野生型(WT)和ACE-N敲除(ACE-NKO)小鼠通过结扎左前降支动脉进行MI,并用媒介物或Ac-SDKP(1.6mg/kg/天,s.c.)持续5周,之后进行超声心动图检查并收集左心室(LV)进行组织学和分子生物学研究.与WT相比,ACE-NKO小鼠在假手术组和MI组中显示增加的血浆Ac-SDKP浓度。外源性Ac-SDKP进一步增加其在WT和ACE-NKO中的循环浓度。MI后WT和ACE-NKO小鼠的缩短(SF)和射血(EF)分数均显着降低,但ACE-NKO小鼠表现出显著较少下降。外源性Ac-SDKP仅在WT中改善MI后的心脏功能,但在ACE-NKO小鼠中未能显示任何累加改善。肌内质网钙转运ATP酶(SERCA2),心脏功能和钙稳态的标志,MI后WT显着下降,但用Ac-SDKP抢救,而ACE-NKO小鼠显示较少的SERCA2表达缺失。我们的研究表明,ACE-N的基因缺失导致MI后小鼠左心室心功能的改善。这可能是由循环Ac-SDKP增加和SERCA2表达最低限度降低介导的。因此,针对ACE-N的特异性和选择性抑制剂的未来开发可能代表了一种新的方法,可以增加内源性Ac-SDKP,以保护心脏免受MI后重塑。
    Angiotensin-converting enzyme (ACE) hydrolyzes N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) into inactive fragments through its N-terminal site (ACE-N). We previously showed that Ac-SDKP mediates ACE inhibitors\' cardiac effects. Whether increased bioavailability of endogenous Ac-SDKP caused by knocking out ACE-N also improves cardiac function in myocardial infarction (MI)-induced heart failure (HF) is unknown. Wild-type (WT) and ACE-N knockout (ACE-NKO) mice were subjected to MI by ligating the left anterior descending artery and treated with vehicle or Ac-SDKP (1.6 mg/kg/day, s.c.) for 5 weeks, after which echocardiography was performed and left ventricles (LV) were harvested for histology and molecular biology studies. ACE-NKO mice showed increased plasma Ac-SDKP concentrations in both sham and MI group compared to WT. Exogenous Ac-SDKP further increased its circulating concentrations in WT and ACE-NKO. Shortening (SF) and ejection (EF) fractions were significantly decreased in both WT and ACE-NKO mice post-MI, but ACE-NKO mice exhibited significantly lesser decrease. Exogenous Ac-SDKP ameliorated cardiac function post-MI only in WT but failed to show any additive improvement in ACE-NKO mice. Sarcoendoplasmic reticulum calcium transport ATPase (SERCA2), a marker of cardiac function and calcium homeostasis, was significantly decreased in WT post-MI but rescued with Ac-SDKP, whereas ACE-NKO mice displayed less loss of SERCA2 expression. Our study demonstrates that gene deletion of ACE-N resulted in improved LV cardiac function in mice post-MI, which is likely mediated by increased circulating Ac-SDKP and minimally reduced expression of SERCA2. Thus, future development of specific and selective inhibitors for ACE-N could represent a novel approach to increase endogenous Ac-SDKP toward protecting the heart from post-MI remodeling.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目的:冷刺激触发白色脂肪组织转化为米色脂肪组织,能够不发抖的产热。然而,是什么过程驱动了米色脂肪中产热的激活,目前还不清楚。这里,我们研究了ER蛋白NNAT作为脂肪组织产热调节因子。
    方法:我们研究了响应环境温度变化的脂肪组织NNAT表达的调节。我们还使用Nnat无效小鼠和缺乏或过表达NNAT的原代脂肪细胞评估了NNAT在热调节中的功能作用。
    结果:冷暴露或β3-肾上腺素能激动剂治疗可降低小鼠脂肪组织NNAT的表达。Nnat在小鼠中的遗传破坏增强腹股沟脂肪组织产热。Nnat无效小鼠在存在和不存在UCP1的情况下都表现出改善的耐寒性。功能增益研究表明,Nnat的异位表达消除了米色脂肪细胞中肾上腺素能受体介导的呼吸。NNAT与脂肪细胞中的ERCa2-ATPase(SERCA)物理相互作用并抑制其活性,阻碍Ca2+传输和散热。我们进一步证明了NHLRC1,一种E3泛素蛋白连接酶参与NNAT的蛋白酶体降解,是由冷暴露或β3-肾上腺素能刺激引起的,从而在蛋白质水平提供调节控制。这有助于将冷刺激与脂肪组织中的NNAT降解联系起来,这反过来导致增强的SERCA活性。
    结论:我们的研究提示NNAT参与了脂肪细胞产热的调节。
    Cold stimuli trigger the conversion of white adipose tissue into beige adipose tissue, which is capable of non-shivering thermogenesis. However, what process drives this activation of thermogenesis in beige fat is not well understood. Here, we examine the ER protein NNAT as a regulator of thermogenesis in adipose tissue.
    We investigated the regulation of adipose tissue NNAT expression in response to changes in ambient temperature. We also evaluated the functional role of NNAT in thermogenic regulation using Nnat null mice and primary adipocytes that lack or overexpress NNAT.
    Cold exposure or treatment with a β3-adrenergic agonist reduces the expression of adipose tissue NNAT in mice. Genetic disruption of Nnat in mice enhances inguinal adipose tissue thermogenesis. Nnat null mice exhibit improved cold tolerance both in the presence and absence of UCP1. Gain-of-function studies indicate that ectopic expression of Nnat abolishes adrenergic receptor-mediated respiration in beige adipocytes. NNAT physically interacts with the ER Ca2+-ATPase (SERCA) in adipocytes and inhibits its activity, impairing Ca2+ transport and heat dissipation. We further demonstrate that NHLRC1, an E3 ubiquitin protein ligase implicated in proteasomal degradation of NNAT, is induced by cold exposure or β3-adrenergic stimulation, thus providing regulatory control at the protein level. This serves to link cold stimuli to NNAT degradation in adipose tissue, which in turn leads to enhanced SERCA activity.
    Our study implicates NNAT in the regulation of adipocyte thermogenesis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    化学耐药性仍然是成功治疗三阴性乳腺癌(TNBC)的主要障碍。识别可药用漏洞是TNBC治疗的重要目标。这里,我们报道SERCA2表达与人类患者的TNBC进展相关,促进TNBC细胞增殖,迁移和化学抗性。机械上,SERCA2通过LIR基序与LC3B相互作用,促进WIPI2非依赖性自噬体形成以诱导自噬。自噬介导的SERCA2降解通过Ca2+/CaMKK/CREB-1反馈诱导SERCA2反式激活。此外,我们发现靶向SERCA2的小分子RL71增强SERCA2-LC3B相互作用并诱导过度的自噬性细胞死亡.SERCA2表达的增加使TNBC细胞在体外和体内容易发生RL71诱导的自噬性细胞死亡。本研究阐明了TNBC细胞维持其高自噬活性以诱导化疗耐药的机制。并提示SERCA2表达增加是TNBC的药物脆弱性。
    Chemoresistance remains a major obstacle to successful treatment of triple negative breast cancer (TNBC). Identification of druggable vulnerabilities is an important aim for TNBC therapy. Here, we report that SERCA2 expression correlates with TNBC progression in human patients, which promotes TNBC cell proliferation, migration and chemoresistance. Mechanistically, SERCA2 interacts with LC3B via LIR motif, facilitating WIPI2-independent autophagosome formation to induce autophagy. Autophagy-mediated SERCA2 degradation induces SERCA2 transactivation through a Ca2+/CaMKK/CREB-1 feedback. Moreover, we found that SERCA2-targeting small molecule RL71 enhances SERCA2-LC3B interaction and induces excessive autophagic cell death. The increase in SERCA2 expression predisposes TNBC cells to RL71-induced autophagic cell death in vitro and in vivo. This study elucidates a mechanism by which TNBC cells maintain their high autophagy activity to induce chemoresistance, and suggests increased SERCA2 expression as a druggable vulnerability for TNBC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:鉴于微血管损伤在梗死形成和扩张中的重要性,开发针对心肌缺血/再灌注损伤(IRI)的微血管保护的治疗策略引起了极大的兴趣。这里,我们探讨了SGLT2抑制剂达格列净(DAPA)对IRI介导的心脏微血管功能障碍的保护作用的分子机制.方法:在体内评估DAPA效果,在接受IRI的小鼠中,在体外,在暴露于缺氧/复氧(H/R)的人冠状动脉内皮细胞(HCAECs)中。DAPA预处理减轻管腔狭窄,内皮肿胀,和IRI处理的小鼠心脏微血管的炎症。结果:在H/R攻击的HCAECs中,DAPA治疗改善内皮屏障功能,内皮型一氧化氮合酶(eNOS)活性,和血管生成能力,并通过阻止cofilin依赖性F-actin解聚和细胞骨架降解来抑制H/R诱导的细胞凋亡。抑制H/R诱导的黄嘌呤氧化酶(XO)激活和上调,Sarco(endo)质网钙ATP酶2(SERCA2)氧化和失活,在DAPA处理的HCAECs中进一步观察到细胞质钙超载。DAPA还抑制钙/钙调蛋白(CaM)依赖性激酶II(CaMKII)激活和cofilin磷酸化,H/R后保留的细胞骨架完整性和内皮细胞活力重要的是,在IRI治疗的SERCA2基因敲除小鼠中,DAPA对心脏微血管完整性和内皮细胞存活的有益作用在很大程度上被阻止.结论:这些结果表明,DAPA通过抑制XO-SERCA2-CaMKII-cofilin途径有效降低了IRI期间的心脏微血管损伤和内皮功能障碍。
    Background: Given the importance of microvascular injury in infarct formation and expansion, development of therapeutic strategies for microvascular protection against myocardial ischemia/reperfusion injury (IRI) is of great interest. Here, we explored the molecular mechanisms underlying the protective effects of the SGLT2 inhibitor dapagliflozin (DAPA) against cardiac microvascular dysfunction mediated by IRI. Methods: DAPA effects were evaluated both in vivo, in mice subjected to IRI, and in vitro, in human coronary artery endothelial cells (HCAECs) exposed to hypoxia/reoxygenation (H/R). DAPA pretreatment attenuated luminal stenosis, endothelial swelling, and inflammation in cardiac microvessels of IRI-treated mice. Results: In H/R-challenged HCAECs, DAPA treatment improved endothelial barrier function, endothelial nitric oxide synthase (eNOS) activity, and angiogenic capacity, and inhibited H/R-induced apoptosis by preventing cofilin-dependent F-actin depolymerization and cytoskeletal degradation. Inhibition of H/R-induced xanthine oxidase (XO) activation and upregulation, sarco(endo)plasmic reticulum calcium-ATPase 2 (SERCA2) oxidation and inactivation, and cytoplasmic calcium overload was further observed in DAPA-treated HCAECs. DAPA also suppressed calcium/Calmodulin (CaM)-dependent kinase II (CaMKII) activation and cofilin phosphorylation, and preserved cytoskeleton integrity and endothelial cell viability following H/R. Importantly, the beneficial effects of DAPA on cardiac microvascular integrity and endothelial cell survival were largely prevented in IRI-treated SERCA2-knockout mice. Conclusions: These results indicate that DAPA effectively reduces cardiac microvascular damage and endothelial dysfunction during IRI through inhibition of the XO-SERCA2-CaMKII-cofilin pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号