CaMKIIδ

CaMKII δ
  • 文章类型: Journal Article
    心力衰竭(HF)是全球发病率和死亡率的主要原因。RNA结合蛋白被确定为心脏疾病的调节因子;DDX5(死盒解旋酶5)是许多RNA过程的主要调节因子,尽管其在心脏生理学中的功能尚不清楚。
    我们评估了人衰竭心脏和小鼠HF模型中的DDX5表达。为了研究DDX5在心脏中的功能,我们设计了心肌细胞特异性Ddx5基因敲除小鼠。我们使用腺相关病毒血清型9在心肌细胞中过表达DDX5,并进行横向主动脉缩窄以建立小鼠HF模型。随后使用免疫沉淀-质谱法研究了下划线的机制,RNA测序,选择性剪接分析,和RNA免疫沉淀测序。
    我们筛选了鼠HF和人扩张型心肌病样本的转录组数据库,发现DDX5在两者中均显著下调。Ddx5的心肌细胞特异性缺失导致心脏功能降低的HF,扩大的心腔,和增加小鼠的纤维化。DDX5过表达改善了患有横主动脉缩窄诱导的HF的小鼠的心脏功能并防止了不利的心脏重塑。此外,蛋白质组学研究表明DDX5参与心肌细胞的RNA剪接。我们发现DDX5调节Ca2+/钙调蛋白依赖性蛋白激酶IIδ(CamkIIδ)的异常剪接,从而阻止CaMKIIδA的产生,它通过Cacna1c的丝氨酸残基磷酸化L型钙通道,导致Ca2+稳态受损。与此相符,我们发现DDX5耗竭心肌细胞细胞内Ca2+瞬变增加,肌浆网Ca2+含量增加.使用CaMKIIδA的腺相关病毒血清型9敲除部分挽救了Ddx5敲除小鼠的心脏功能障碍和HF。
    这些发现揭示了DDX5通过调节心肌细胞的可变剪接在维持钙稳态和心脏功能方面的作用,确定DDX5作为HF治疗干预的潜在目标。
    UNASSIGNED: Heart failure (HF) is a leading cause of morbidity and mortality worldwide. RNA-binding proteins are identified as regulators of cardiac disease; DDX5 (dead-box helicase 5) is a master regulator of many RNA processes, although its function in heart physiology remains unclear.
    UNASSIGNED: We assessed DDX5 expression in human failing hearts and a mouse HF model. To study the function of DDX5 in heart, we engineered cardiomyocyte-specific Ddx5 knockout mice. We overexpressed DDX5 in cardiomyocytes using adeno-associated virus serotype 9 and performed transverse aortic constriction to establish the murine HF model. The mechanisms underlined were subsequently investigated using immunoprecipitation-mass spectrometry, RNA-sequencing, alternative splicing analysis, and RNA immunoprecipitation sequencing.
    UNASSIGNED: We screened transcriptome databases of murine HF and human dilated cardiomyopathy samples and found that DDX5 was significantly downregulated in both. Cardiomyocyte-specific deletion of Ddx5 resulted in HF with reduced cardiac function, an enlarged heart chamber, and increased fibrosis in mice. DDX5 overexpression improved cardiac function and protected against adverse cardiac remodeling in mice with transverse aortic constriction-induced HF. Furthermore, proteomics revealed that DDX5 is involved in RNA splicing in cardiomyocytes. We found that DDX5 regulated the aberrant splicing of Ca2+/calmodulin-dependent protein kinase IIδ (CamkIIδ), thus preventing the production of CaMKIIδA, which phosphorylates L-type calcium channel by serine residues of Cacna1c, leading to impaired Ca2+ homeostasis. In line with this, we found increased intracellular Ca2+ transients and increased sarcoplasmic reticulum Ca2+ content in DDX5-depleted cardiomyocytes. Using adeno-associated virus serotype 9 knockdown of CaMKIIδA partially rescued the cardiac dysfunction and HF in Ddx5 knockout mice.
    UNASSIGNED: These findings reveal a role for DDX5 in maintaining calcium homeostasis and cardiac function by regulating alternative splicing in cardiomyocytes, identifying the DDX5 as a potential target for therapeutic intervention in HF.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    阻塞性睡眠呼吸暂停(OSA)与各种病理有关,包括心律失常,如心房颤动。OSA的具体治疗选择主要限于对症治疗。我们先前表明,活性氧(ROS)的产生增加通过Ca2/钙调蛋白依赖性蛋白激酶IIδ(CaMKIIδ)通过电压依赖性Na通道刺激晚期钠电流,从而增加心律失常的倾向。然而,对心房细胞内Na+稳态的影响从未被证实。此外,患者通常表现出广泛的合并症,这使得很难确定OSA单独的影响。
    我们分析了OSA对ROS产生的影响,胞质Na+水平,从无合并症的OSA小鼠模型中分离出的心房心肌细胞中的自发性心律失常率。
    通过将聚四氟乙烯(PTFE)注射到舌头中,在C57BL/6野生型和CaMKIIδ敲除小鼠中诱导了OSA。8周后,通过激光扫描共聚焦显微镜分析其心房心肌细胞的胞浆和线粒体ROS的产生.通过落射荧光显微镜对胞质Na浓度和心律失常进行定量。
    PTFE处理导致胞浆和线粒体ROS产生增加。重要的是,在PTFE处理的小鼠中,细胞溶质Na+浓度在各种刺激频率下急剧增加,而CaMKIIδ敲除小鼠受到保护。因此,在野生型PTFE小鼠中,自发Ca2释放事件的速率增加,而在CaMKIIδ敲除小鼠中受到阻碍。
    在OSA小鼠模型中,心房Na浓度和自发Ca2+释放事件的倾向以CaMKIIδ依赖性方式较高,可能有治疗意义。
    UNASSIGNED: Obstructive sleep apnea (OSA) has been linked to various pathologies, including arrhythmias such as atrial fibrillation. Specific treatment options for OSA are mainly limited to symptomatic approaches. We previously showed that increased production of reactive oxygen species (ROS) stimulates late sodium current through the voltage-dependent Na+ channels via Ca2+/calmodulin-dependent protein kinase IIδ (CaMKIIδ), thereby increasing the propensity for arrhythmias. However, the impact on atrial intracellular Na+ homeostasis has never been demonstrated. Moreover, the patients often exhibit a broad range of comorbidities, making it difficult to ascertain the effects of OSA alone.
    UNASSIGNED: We analyzed the effects of OSA on ROS production, cytosolic Na+ level, and rate of spontaneous arrhythmia in atrial cardiomyocytes isolated from an OSA mouse model free from comorbidities.
    UNASSIGNED: OSA was induced in C57BL/6 wild-type and CaMKIIδ-knockout mice by polytetrafluorethylene (PTFE) injection into the tongue. After 8 weeks, their atrial cardiomyocytes were analyzed for cytosolic and mitochondrial ROS production via laser-scanning confocal microscopy. Quantifications of the cytosolic Na+ concentration and arrhythmia were performed by epifluorescence microscopy.
    UNASSIGNED: PTFE treatment resulted in increased cytosolic and mitochondrial ROS production. Importantly, the cytosolic Na+ concentration was dramatically increased at various stimulation frequencies in the PTFE-treated mice, while the CaMKIIδ-knockout mice were protected. Accordingly, the rate of spontaneous Ca2+ release events increased in the wild-type PTFE mice while being impeded in the CaMKIIδ-knockout mice.
    UNASSIGNED: Atrial Na+ concentration and propensity for spontaneous Ca2+ release events were higher in an OSA mouse model in a CaMKIIδ-dependent manner, which could have therapeutic implications.
    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/钙调蛋白依赖性蛋白激酶II(CaMKII)已被证明在病毒性心肌炎(VMC)中异常激活,但其亚型CaMKIIδ在VMC中的作用尚不清楚。通过柯萨奇病毒B3(CVB3)处理诱导VMC小鼠和心肌细胞模型。接受假手术的小鼠和盐水处理的心肌细胞作为对照。体重,生存,左心室射血分数(LVEF),测量缩短分数(LVFS),并进行HE染色以评估VMC小鼠模型和假对照的心功能。ELISA法检测血清或细胞上清液中的炎症因子。CaMKIIδ的表达,含Toll/白介素-1受体结构域的衔接蛋白(TIRAP),胰岛素样生长因子2mRNA结合蛋白2(IGF2BP2),核因子NF-κB(NF-κB)信号,通过定量实时聚合酶链反应(qRT-PCR)或Westernblot检查炎症因子。CCK-8,EdU,和流式细胞术用于评估细胞行为。免疫共沉淀(Co-IP),RNA免疫沉淀(RIP),和RNA下拉来验证分子相互作用。进行甲基化RNA免疫沉淀(MeRIP)以测量特定分子的N6-甲基腺苷(m6A)水平。CaMKIIδ在VMC小鼠和CVB3处理的原代心肌细胞中上调,其中敲除提高了细胞活力,扩散,并在体外抑制细胞凋亡,从而减轻体内心肌炎。CaMKIIδ的稳定性归因于通过m6A修饰的IGF2BP2的存在。CaMKIIδ的缺失通过负向和直接调节TIRAP来抑制NF-κB途径,从而参与炎性损伤。CaMKIIδ,由m6A阅读器IGF2BP2稳定,通过与TIRAP相互作用调节NF-κB途径以改变细胞活力,扩散,和细胞凋亡,从而影响VMC结果。
    Calcium/calmodulin-dependent protein kinase II (CaMKII) has been demonstrated to be aberrantly activated in viral myocarditis (VMC), but the role of its subtype CaMKIIδ in VMC remains unclear.VMC mice and cardiomyocytes models were induced by Coxsackievirus B3 (CVB3) treatment. Mice that underwent sham surgery and saline-treated cardiomyocytes served as controls. Body weight, survival, left ventricular ejection fraction (LVEF), and fractional shortening (LVFS) were measured, and HE staining was performed to evaluate heart function in VMC mice model and sham control. Inflammation factors in serum or cell supernatant were detected by ELISA. Expressions of CaMKIIδ, Toll/interleukin-1 receptor domain containing adaptor protein (TIRAP), insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2), nuclear factor NF-kappaB (NF-κB) signals, and inflammation factors were examined by quantitative real time polymerase chain reaction (qRT-PCR) or western blot. CCK-8, EdU, and flow cytometry were used to evaluate cell behaviors. Co-immunoprecipitation (Co-IP), RNA immunoprecipitation (RIP), and RNA pull-down were utilized to validate molecule interaction. Methylated RNA immunoprecipitation (MeRIP) was performed to measure N6-methyladenosine (m6A) level of specific molecule.CaMKIIδ was upregulated in VMC mice and CVB3-treated primary cardiomyocytes, of which knockdown improved cell viability, proliferation, and suppressed cell apoptosis in vitro, thereby alleviating myocarditis in vivo. The stability of CaMKIIδ was attributed to the presence of IGF2BP2 through m6A modification. Loss of CaMKIIδ repressed NF-κB pathway via negatively and directly regulating TIRAP to be involved in inflammatory damage.CaMKIIδ, stabilized by m6A reader IGF2BP2, modulated NF-κB pathway via interacting with TIRAP to alter cell viability, proliferation, and apoptosis, thereby affecting VMC outcome.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    长时间刺激β-肾上腺素能受体(β-AR)可导致交感神经过度活动,引起病理性心脏肥大和纤维化,最终导致心力衰竭。最近的研究表明,异常的蛋白质泛素化可能有助于心脏肥大和重塑的发病机制。在这项研究中,我们证明了去泛素酶的缺乏,含Josephin结构域的蛋白2(JOSD2),改善异丙肾上腺素(ISO)-和心肌梗死(MI)-诱导的心脏肥大,纤维化,以及体外和体内功能障碍。相反,JOSD2过表达加重ISO诱导的心脏病理学。通过全面的质谱分析,我们确定JOSD2与钙-钙调蛋白依赖性蛋白激酶II(CaMKIIδ)相互作用。JOSD2直接水解CaMKIIδ上的K63连接的聚泛素链,从而增加CaMKIIδ的磷酸化并导致钙处理不当,肥大,和心肌细胞纤维化。体内实验表明,CAMKIIδ抑制剂KN-93可以逆转JOSD2过表达诱导的心脏重塑。总之,我们的研究强调了JOSD2在通过调节CaMKIIδ泛素化介导ISO诱导的心脏重塑中的作用,并表明其作为对抗疾病的治疗靶点的潜力。请检查并确认作者及其各自的从属关系已被正确识别,并在必要时进行修改。都检查过了。
    Prolonged stimulation of β-adrenergic receptor (β-AR) can lead to sympathetic overactivity that causes pathologic cardiac hypertrophy and fibrosis, ultimately resulting in heart failure. Recent studies suggest that abnormal protein ubiquitylation may contribute to the pathogenesis of cardiac hypertrophy and remodeling. In this study, we demonstrated that deficiency of a deubiquitinase, Josephin domain-containing protein 2 (JOSD2), ameliorated isoprenaline (ISO)- and myocardial infarction (MI)-induced cardiac hypertrophy, fibrosis, and dysfunction both in vitro and in vivo. Conversely, JOSD2 overexpression aggravated ISO-induced cardiac pathology. Through comprehensive mass spectrometry analysis, we identified that JOSD2 interacts with Calcium-calmodulin-dependent protein kinase II (CaMKIIδ). JOSD2 directly hydrolyzes the K63-linked polyubiquitin chains on CaMKIIδ, thereby increasing the phosphorylation of CaMKIIδ and resulting in calcium mishandling, hypertrophy, and fibrosis in cardiomyocytes. In vivo experiments showed that the cardiac remodeling induced by JOSD2 overexpression could be reversed by the CaMKIIδ inhibitor KN-93. In conclusion, our study highlights the role of JOSD2 in mediating ISO-induced cardiac remodeling through the regulation of CaMKIIδ ubiquitination, and suggests its potential as a therapeutic target for combating the disease. Please check and confirm that the authors and their respective affiliations have been correctly identified and amend if necessary. All have been checked.
    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
    肥胖会增加年轻人动脉高血压的风险,并通过产生氧化应激而促进早发性心肌病。在这个意义上,早年不分青红皂白地食用蔗糖和果糖含糖饮料会导致肥胖,然而,当存在发展为高血压的遗传倾向时,其对心脏的影响尚不清楚。我们比较了蔗糖的影响,果糖,以及它们在断奶雄性自发性高血压大鼠中的组合,以确定遗传性高血压之间的关系,肥胖,以及这些糖的消耗对心脏肥大程度的影响,氧化应激和Ca2+/钙调蛋白依赖性蛋白激酶Ⅱδ氧化。组织学,生物化学,和Westernblot研究在治疗开始后12周进行。我们发现长期食用蔗糖或果糖会导致肥胖,加剧遗传性动脉高血压引起的代谢改变,并增加心脏氧化应激,Ca2+/钙调蛋白依赖性蛋白激酶Ⅱδ氧化与心肌肥厚.尽管如此,当蔗糖和果糖一起食用时,代谢改变恶化,并伴有扩张型心肌病。这些数据表明,从具有动脉高血压遗传易感性的大鼠的母体断奶开始,蔗糖和果糖的联合消耗会加速心肌病的进展,从而导致早期扩张型心肌病。
    Obesity increases the risk of arterial hypertension in young adults and favors an early-onset cardiomyopathy by generating oxidative stress. In this sense, indiscriminate consumption of sucrose and fructose sweetened beverages from early ages causes obesity, however its consequences on the heart when there is a genetic predisposition to develop hypertension are not clear. We compared the effects of sucrose, fructose, and their combination in weanling male spontaneously hypertensive rats to determine the relationship between genetic hypertension, obesity, and consumption of these sugars on the degree of cardiac hypertrophy, oxidative stress and Ca2+/calmodulin dependent protein kinase II delta oxidation. Histological, biochemical, and Western blot studies were performed 12 weeks after treatment initiation. We found that chronic consumption of sucrose or fructose leads to obesity, exacerbates genetic arterial hypertension-induced metabolic alterations, and increases cardiac oxidative stress, Ca2+/calmodulin dependent protein kinase II delta oxidation and cardiac hypertrophy. Nonetheless, when sucrose and fructose are consumed together, metabolic alterations worsen and are accompanied by dilated cardiomyopathy. These data suggest that sucrose and fructose combined consumption starting from maternal weaning in rats with genetic predisposition to arterial hypertension accelerates the progression of cardiomyopathy resulting in an early dilated cardiomyopathy.
    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
    坏死是一种新发现的细胞死亡机制,其关键调节作用归因于受体相互作用蛋白激酶(RIPKs)RIPK1和RIPK3的相互作用。Ca2+/钙调蛋白依赖性蛋白激酶(CaMKII)是一种新发现的RIPK3底物,它的选择性剪接在心血管疾病中起着根本性的作用。在本研究中,我们旨在探讨CaMKIIδ坏死和选择性剪接在心肌肥厚中的作用和机制。对野生型和基因敲除小鼠进行横向主动脉缩窄(TAC)建立心肌肥厚模型。3周后,超声心动图,心脏指数,心肌细胞的横截面积,肥大基因表达,心肌损伤,和纤维进行了评估。此外,我们检测了炎症因子(IL-6和TNF-α)的水平,并检测了坏死相关蛋白RIPK3,RIPK1和磷酸化MLKL的表达。同时,我们测试了剪接因子ASF/SF2和SC-35的表达水平,试图探索CaMKIIδ。可变剪接障碍与剪接因子ASF/SF2和SC-35表达水平的关系.Further,我们还调查了CaMKII的激活,氧化应激,和线粒体超微结构。此外,给野生型小鼠施用携带RIPK3的重组腺相关病毒(AAV),然后进行TAC手术以构建心肌肥大模型,3周后对上述指标进行检测。结果表明,RIPK3缺乏可以减轻心功能不全,心肌损伤,坏死加重,TAC手术诱导心肌肥厚小鼠的CaMKII激活。尾静脉注射AAV可以逆转心功能不全,心肌损伤,坏死加重,和心肌肥厚小鼠的CaMKII激活。这些成果证明RIPK3可作为防治心肌肥厚的份子介入靶点。
    Necroptosis is a newly discovered mechanism of cell death, and its key regulatory role is attributed to the interaction of receptor-interacting protein kinases (RIPKs) RIPK1 and RIPK3. Ca2+/calmodulin-dependent protein kinase (CaMKII) is a newly discovered RIPK3 substrate, and its alternative splicing plays a fundamental role in cardiovascular diseases. In the present study, we aimed to explore the role and mechanism of necroptosis and alternative splicing of CaMKIIδ in myocardial hypertrophy. Transverse aortic constriction (TAC) was performed on wild-type and knockout mice to establish the model of myocardial hypertrophy. After 3 weeks, echocardiography, cardiac index, cross-sectional area of myocardial cells, hypertrophic gene expression, myocardial damage, and fibers were assessed. Moreover, we detected the levels of inflammatory factors (IL-6 and TNF-α) and examined the expressions of necroptosis-related proteins RIPK3, RIPK1, and phosphorylated MLKL. Meanwhile, we tested the expression levels of splicing factors ASF/SF2 and SC-35 in an attempt to explore CaMKII δ. The relationship between variable splicing disorder and the expression levels of splicing factors ASF/SF2 and SC-35. Further, we also investigated CaMKII activation, oxidative stress, and mitochondrial ultrastructure. In addition, wild-type mice were administered with a recombinant adeno-associated virus (AAV) carrying RIPK3, followed by TAC surgery to construct a model of myocardial hypertrophy, and the above-mentioned indicators were tested after 3 weeks. The results showed that RIPK3 deficiency could alleviate cardiac dysfunction, myocardial injury, aggravation of necrosis, and CaMKII activation induced by TAC surgery in mice with myocardial hypertrophy. Tail vein injection of AAV could reverse cardiac dysfunction, myocardial damage, aggravation of necrosis, and CaMKII activation in mice with myocardial hypertrophy. These results proved that RIPK3 could be used as a molecular intervention target for the prevention and treatment of myocardial hypertrophy.
    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:环孢素A(CsA)诱导的心肌间质纤维化和心肌肥大是众所周知的现象;然而,CsA心脏毒性的基本机制尚不清楚.本研究评估了转化生长因子-β(TGF-β)/Smad3/miR-29b信号通路和CaMKIIδ同工型基因表达在CsA暴露单独或联合适度运动下心脏重塑中的作用。
    方法:将24只雄性Wistar大鼠分为对照组,环孢菌素(30毫克/千克体重),和环孢素运动组。
    结果:治疗42天后,研究结果表明,miR-29和miR-30b-5p基因表达显着下降,Smad3,钙/钙调蛋白依赖性蛋白激酶IIδ(CaMKIIδ)亚型的基因表达增加,基质金属蛋白酶(MMPs),TGF-β的蛋白表达,心脏组织蛋白羰基和氧化LDL(Ox-LDL),与对照组相比,CsA治疗组的血浆LDL和胆固醇水平。CsA组表现出更大的组织学心脏变化,如纤维化,坏死,出血,浸润的白细胞,和左心室重量/心脏重量比对照组。此外,与CsA组相比,适度运动和CsA组合相对改善了基因表达变化和组织学变化。
    结论:TGF-β-Smad3-miR-29和CaMKIIδ亚型可能主要参与CsA暴露导致的心脏纤维化和肥大的进展,为CsA引起的心脏组织副作用的发病机制和治疗提供了新的见解。
    BACKGROUND: Cyclosporine A (CsA)-induced cardiac interstitial fibrosis and cardiac hypertrophy are highly known phenomena; however, the basic mechanisms of CsA cardiotoxicity are unclear. The present study evaluated the role of the Transforming growth factor-beta (TGF-β)/Smad3/miR-29b signaling pathway and CaMKIIδ isoforms gene expression in cardiac remodeling under CsA exposure alone or combined with moderate exercise.
    METHODS: A total of 24 male Wistar rats were divided into control, cyclosporine (30 mg/kg BW), and cyclosporine-exercise groups.
    RESULTS: After 42 days of treatment, the findings revealed a significant decline in miR-29 and miR-30b-5p gene expression and an increase in gene expression of Smad3, calcium/calmodulin-dependent protein kinaseIIδ (CaMKIIδ) isoforms, Matrix Metalloproteinases (MMPs), protein expression of TGF-β, heart tissue protein carbonyl and oxidized LDL (Ox-LDL), and plasma LDL and cholesterol levels in the CsA-treated group compared to the control group. The CsA group presented greater histological heart changes such as fibrosis, necrosis, hemorrhage, infiltrated leukocyte, and left ventricular weight/heart weight than the control group. Moreover, combined moderate exercise and CsA relatively improved gene expression changes and histological alternations compared to the CsA group.
    CONCLUSIONS: TGF-β-Smad3-miR-29 and CaMKIIδ isoforms may mainly contribute to the progression of heart fibrosis and hypertrophy due to CsA exposure, providing new insight into the pathogenesis and treatment of CsA-induced side effects on the heart tissue.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    长QT综合征(LQTS)是人类遗传性心脏病,可引起危及生命的心律失常,包括心脏猝死。普遍存在的Ca2+敏感蛋白钙调蛋白(CaM)的突变与LQTS相关,但是这些突变导致不规则心跳的分子机制尚不完全清楚。这里,我们使用多学科方法,包括蛋白质生物物理学,结构生物学,共聚焦成像和膜片钳电生理学来确定疾病相关CaM突变E140G对CaM结构和功能的影响。我们提供了新的数据,表明突变调节的CaMKIIδ激酶活性随着酶自磷酸化速率的显着降低而受损。我们报告了与CaMKIIδ肽复合的LQTS相关CaM变体的第一个高分辨率晶体结构,这显示了显著的结构差异,与野生型复合物相比。此外,我们证明E140G突变显著破坏了Cav1.2Ca2+/CaM依赖性失活,而心脏ryanodine受体(RyR2)活性未受影响。此外,我们表明LQTS相关突变改变了CaM的Ca2+结合特性,二级结构含量和与参与激发-收缩耦合的关键伙伴的相互作用(CaMKIIδ,Cav1.2,RyR2)。总之,LQTS相关CaM突变E140G严重影响CaM的结构-功能关系及其对CaMKIIδ和Cav1.2的调控这提供了对CaM介导的心律失常的分子因素的重要见解,对CaMKIIδ具有重要作用。
    Long QT syndrome (LQTS) is a human inherited heart condition that can cause life-threatening arrhythmia including sudden cardiac death. Mutations in the ubiquitous Ca2+-sensing protein calmodulin (CaM) are associated with LQTS, but the molecular mechanism by which these mutations lead to irregular heartbeats is not fully understood. Here, we use a multidisciplinary approach including protein biophysics, structural biology, confocal imaging, and patch-clamp electrophysiology to determine the effect of the disease-associated CaM mutation E140G on CaM structure and function. We present novel data showing that mutant-regulated CaMKIIδ kinase activity is impaired with a significant reduction in enzyme autophosphorylation rate. We report the first high-resolution crystal structure of a LQTS-associated CaM variant in complex with the CaMKIIδ peptide, which shows significant structural differences, compared to the WT complex. Furthermore, we demonstrate that the E140G mutation significantly disrupted Cav1.2 Ca2+/CaM-dependent inactivation, while cardiac ryanodine receptor (RyR2) activity remained unaffected. In addition, we show that the LQTS-associated mutation alters CaM\'s Ca2+-binding characteristics, secondary structure content, and interaction with key partners involved in excitation-contraction coupling (CaMKIIδ, Cav1.2, RyR2). In conclusion, LQTS-associated CaM mutation E140G severely impacts the structure-function relationship of CaM and its regulation of CaMKIIδ and Cav1.2. This provides a crucial insight into the molecular factors contributing to CaM-mediated arrhythmias with a central role for CaMKIIδ.
    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
    动脉粥样硬化(AS)是涉及许多细胞的慢性进行性炎性疾病。miR-145-5p介导人主动脉血管平滑肌细胞(HAVSMCs)的生物学表型并影响AS的进展,但潜在的机制需要进一步研究。
    从患者血浆和动脉中提取总RNA以测定miR-145-5p的表达。通过生物信息学方法预测CaMKIIδ通路和基因是miR-145-5p的靶标。miR-145-5p与CaMKIIδ之间的相互作用通过RT-qPCR和双荧光素酶报告分析系统进行确认。蛋白质印迹分析,免疫荧光染色,透射电子显微镜(TEM)和用mCherry-GFP-LC3慢病毒载体转导的HAVSMC的蛋白质示踪,以确定miR-145-5p影响动脉粥样硬化疾病过程的机制。
    冠状动脉狭窄患者的血液和动脉标本中miR-145-5p的表达下调。相应地,在低氧HAVSMC中,CaMKIIδ上调,miR-145-5p下调。预测并确认CaMKIIδ为miR-145-5p的下游靶标。此外,CaMKIIδ通过激活AMPK/mTOR/ULK1信号通路诱导自噬相关蛋白的上调。此外,我们证实miR-145-5p通过与CaMKIIδ3'UTR中的特定序列结合而抑制CaMKIIδ的表达并影响自噬。至关重要的是,通过下调miR-145-5p促进CaMKIIδ,然后通过AMPK/mTOR/ULK1信号通路激活HAVSMC中的自噬以影响AS进展。
    miR-145-5p调节CaMKIIδ,导致HAVSMC中的自噬改变。这种改变在AS进展中起重要作用。
    Atherosclerosis (AS) is a chronic progressive inflammatory disease involving many cells. miR-145-5p mediates the biological phenotypes of human aortic vascular smooth muscle cells (HAVSMCs) and influences the progression of AS, but the potential mechanism needs further study.
    Total RNA was extracted from patient plasma and arteries to determine the expression of miR-145-5p. The CaMKIIδ pathway and genes were predicted as the target of miR-145-5p by bioinformatics approaches. The interaction between miR-145-5p and CaMKIIδ was confirmed by RT-qPCR and Dual Luciferase Reporter Assay System. Western blot analysis, immunofluorescence staining, transmission electron microscopy (TEM) and protein tracing on HAVSMCs transduced with mCherry-GFP-LC3 lentiviral vectors to determine the mechanism by which miR-145-5p affects the atherosclerotic disease process.
    The expression of miR-145-5p was downregulated in blood and arteries specimens of patients with coronary stenosis. Correspondingly, CaMKIIδ was upregulated and miR-145-5p was downregulated in hypoxic HAVSMCs. CaMKIIδ was predicted and confirmed as a downstream target of miR-145-5p. In addition, CaMKIIδ induced the upregulation of autophagy-related proteins by activating the AMPK/mTOR/ULK1 signalling pathway. Moreover, we confirmed that miR-145-5p inhibits CaMKIIδ expression by binding to a specific sequence in the CaMKIIδ 3\' UTR and affects autophagy. Crucially, CaMKIIδ was promoted by the downregulation of miR-145-5p and then activating autophagy in HAVSMCs through the AMPK/mTOR/ULK1 signalling pathway to affect the AS progress.
    miR-145-5p regulates CaMKIIδ, leading to altered autophagy in HAVSMCs. This alteration plays an important role in AS progression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    心肌缺血/再灌注(I/R)损伤对人类健康构成重大威胁。高水平的活性氧(ROS)和钙超载是I/R中心肌损害的首要原因。萝卜硫烷(SFN)以其有前途的抗氧化作用而闻名。SFN是否对I/R具有心肌保护作用在很大程度上是未知的。本研究旨在探讨SFN是否可以保护心肌免受I/R损伤。我们发现用SFN预处理的小鼠或细胞显示出改善的心脏功能和细胞存活。SFN处理抑制缺氧再灌注(H/R)诱导的炎性细胞因子的产生和细胞内钙的增加,同时线粒体膜电位得到有效维持。转录组分析表明,在I/R心肌中,SFN处理下调了CaMKIIδ的表达,而CaMKIIN2,CaMKII的抑制剂,被上调了。敲除CaMKIIN2不仅导致总CaMKIIδ和磷酸化CaMKIIδ的水平升高,而且还阻断了SFN对H/R细胞的促存活作用。此外,CaMKIIN2过表达足以抑制CaMKIIδ活化并改善H/R下的细胞存活。一起来看,这项研究表明,SFN通过上调CaMKIIN2和下调CaMKIIδ对I/R损伤具有心脏保护作用。
    Myocardial ischemia/reperfusion (I/R) injury poses a significant threat to human health. High level of reactive oxygen species (ROS) and calcium overload are the foremost causes of myocardial damage in I/R. Sulforaphane (SFN) is known for its promising antioxidant effect. Whether or not SFN has myocardial protective effect against I/R is largely unknown. This study aimed to investigate if SFN can protect myocardium from I/R injury. We found that mice or cells pre-treated with SFN showed improved cardiac functions and cell survival. SFN treatment inhibited the production of inflammatory cytokines and the increase of intracellular calcium induced by hypoxia-reperfusion (H/R), while mitochondria membrane potential was effectively maintained. Transcriptome analysis showed that CaMKIIδ expression was down-regulated by SFN treatment in I/R myocardium, while CaMKIIN2, the inhibitor of CaMKII, was upregulated. Knockdown of CaMKIIN2 not only led to increased level of total CaMKIIδ and the phosphorylated CaMKIIδ but also blocked the pro-survival effect of SFN for H/R cells. Moreover, CaMKIIN2 overexpression was sufficient to suppress CaMKIIδ activation and improve cell survival under H/R. Taken together, this study demonstrated that SFN exerts cardioprotective effect toward I/R injury through upregulating CaMKIIN2 and down-regulating CaMKIIδ.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号