β-adrenergic receptor

β - 肾上腺素能受体
  • 文章类型: Journal Article
    心脏纤维化的特征是过度增殖,心脏成纤维细胞(CFs)的细胞外基质(ECM)的过度转分化和过度沉积。心脏交感神经激活是心肌纤维化的主要原因之一。同时,心脏纤维化通常伴随着心脏炎症,通过介导炎性细胞因子分泌加速纤维化。最近,Janus激酶/信号转导子和转录激活因子(JAK/STAT3)信号通路在心脏纤维化进展过程中的重要作用已得到证实。因此,JAK/STAT3信号通路被认为是心脏纤维化的潜在治疗靶点。巴利替尼(BR),一种新型的JAK1/2抑制剂,已经报道了抗纤维化在多种纤维化疾病中的优异效果。然而,关于BR是否以及如何改善由慢性交感神经激活引起的心脏纤维化知之甚少。异丙肾上腺素(ISO),β-肾上腺素能受体(β-AR)非选择性激动剂,用于调节小鼠的慢性交感神经激活。不出所料,我们的结果证明BR改善了ISO诱导的心功能不全。同时,BR减轻ISO诱导的小鼠心脏纤维化和心脏炎症。此外,BR还抑制ISO诱导的心脏成纤维细胞活化和巨噬细胞促炎分泌。至于机制研究,BR通过JAK2/STAT3和PI3K/Akt信号降低ISO诱导的心肌成纤维细胞,同时通过JAK1/STAT3和NF-κB信号降低ISO诱导的巨噬细胞促炎分泌。总之,BR减轻由慢性交感神经激活引起的心脏纤维化和炎症。潜在机制涉及BR介导的JAK1/2/STAT3、PI3K/Akt和NF-κB信号传导的抑制。
    Cardiac fibrosis is characterized by the over-proliferation, over-transdifferentiation and over-deposition of extracellular matrix (ECM) of cardiac fibroblasts (CFs). Cardiac sympathetic activation is one of the leading causes of myocardial fibrosis. Meanwhile, cardiac fibrosis is often together with cardiac inflammation, which accelerates fibrosis by mediating inflammatory cytokines secretion. Recently, the Janus kinase/signal transducer and activator of transcription (JAK/STAT3) signaling pathway has been confirmed by its vital role during the progression of cardiac fibrosis. Thus, JAK/STAT3 signaling pathway is thought to be a potential therapeutic target for cardiac fibrosis. Baricitinib (BR), a novel JAK1/2 inhibitor, has been reported excellent effects of anti-fibrosis in multiple fibrotic diseases. However, little is known about whether and how BR ameliorates cardiac fibrosis caused by chronic sympathetic activation. Isoproterenol (ISO), a β-Adrenergic receptor (β-AR) nonselective agonist, was used to modulate chronic sympathetic activation in mice. As expected, our results proved that BR ameliorated ISO-induced cardiac dysfunction. Meanwhile, BR attenuated ISO-induced cardiac fibrosis and cardiac inflammation in mice. Moreover, BR also inhibited ISO-induced cardiac fibroblasts activation and macrophages pro-inflammatory secretion. As for mechanism studies, BR reduced ISO-induced cardiac fibroblasts by JAK2/STAT3 and PI3K/Akt signaling, while reduced ISO-induced macrophages pro-inflammatory secretion by JAK1/STAT3 and NF-κB signaling. In summary, BR alleviates cardiac fibrosis and inflammation caused by chronic sympathetic activation. The underlying mechanism involves BR-mediated suppression of JAK1/2/STAT3, PI3K/Akt and NF-κB signaling.
    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
    交感神经-β-肾上腺素能受体(βAR)系统的激活是心脏病的标志,具有促进心力衰竭(HF)发作和进展的不良后果。使用β-阻断药物已成为HF的一线治疗。过去十年见证了研究的进展,证明了Hippo途径在心肌病和HF中的关键作用。临床研究表明,在几种类型的人类心肌病中,心肌Hippo通路激活/YAP-TEAD1失活。心脏Hippo信号的实验性激活或YAP-TEAD1的抑制已被证明会导致扩张型心肌病,并伴有严重的线粒体功能障碍和代谢重编程。研究还令人信服地表明,βAR的刺激激活了心脏Hippo途径,并使下游效应分子YAP/TAZ失活。有强有力的证据表明βAR-Hippo信号传导导致HF的不良后果。除了促进心肌细胞死亡和纤维化,最近的进展是线粒体功能障碍和βAR-Hippo通路信号介导的代谢重编程的证明。心脏βAR-Hippo信号的激活在下调一系列线粒体和代谢基因方面是有效的,而促炎因子和促纤维化因子的表达上调。βAR-Hippo通路信号的偶联是由几种激酶介导的,机械传导和/或Ca2+信号,并可被β-拮抗剂阻断。βAR信号和Hippo途径的趋同的证明对更好地理解增强的交感神经活动的作用具有重要意义。β-拮抗剂的功效,以及针对HF这一途径的代谢疗法。在这篇综述中,我们总结了该领域的进展,并讨论了未来的研究方向。
    Activation of the sympatho-β-adrenergic receptor (βAR) system is the hallmark of heart disease with adverse consequences that facilitate the onset and progression of heart failure (HF). Use of β-blocking drugs has become the front-line therapy for HF. Last decade has witnessed progress in research demonstrating a pivotal role of Hippo pathway in cardiomyopathy and HF. Clinical studies have revealed myocardial Hippo pathway activation/YAP-TEAD1 inactivation in several types of human cardiomyopathy. Experimental activation of cardiac Hippo signaling or inhibition of YAP-TEAD1 have been shown to leads dilated cardiomyopathy with severe mitochondrial dysfunction and metabolic reprogramming. Studies have also convincingly shown that stimulation of βAR activates cardiac Hippo pathway with inactivation of the down-stream effector molecules YAP/TAZ. There is strong evidence for the adverse consequences of the βAR-Hippo signaling leading to HF. In addition to promoting cardiomyocyte death and fibrosis, recent progress is the demonstration of mitochondrial dysfunction and metabolic reprogramming mediated by βAR-Hippo pathway signaling. Activation of cardiac βAR-Hippo signaling is potent in downregulating a range of mitochondrial and metabolic genes, whereas expression of pro-inflammatory and pro-fibrotic factors are upregulated. Coupling of βAR-Hippo pathway signaling is mediated by several kinases, mechanotransduction and/or Ca2+ signaling, and can be blocked by β-antagonists. Demonstration of the converge of βAR signaling and Hippo pathway bears implications for a better understanding on the role of enhanced sympathetic nervous activity, efficacy of β-antagonists, and metabolic therapy targeting this pathway in HF. In this review we summarize the progress and discuss future research directions in this field.
    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
    β-肾上腺素能受体(βARs)是G蛋白偶联受体(GPCRs),介导儿茶酚胺激素诱导的应激反应,如心率升高。除了那些与质膜结合的物质,内膜βARs也有信号传导能力。βAR途径的失调是严重病理状况的基础。新出现的证据表明,在较深的内膜βARs信号中的病理分子特征,可能导致心肌细胞肥大和凋亡等疾病。然而,缺乏控制内膜β1ARs的方法阻碍了信号与病理学的联系。由β1AR-儿茶酚胺相互作用引起,我们设计了一种有效的光不稳定的前配体(OptoIso),使用蓝光刺激仅在内膜区域触发βAR信号。OptoIso不仅在几秒钟内进行蓝光脱保护,而且还可以有效地进入细胞,并允许仅在内膜上检查G蛋白异源三聚体的激活。OptoIso还允许以天然保真度在选定的单细胞中光学激活质膜βAR信号,可以通过终止蓝光来逆转。因此,OptoIso将是一种有价值的实验工具,可以在用户定义的内膜或质膜区域中以天然保真度对未修饰细胞中的βAR信号进行时空控制。
    Beta-adrenergic receptors (βARs) are G protein-coupled receptors (GPCRs) that mediate catecholamine hormone-induced stress responses, such as elevation of heart rate. Besides those that are plasma membrane-bound, endomembrane βARs are also signaling competent. Dysregulation of βAR pathways underlies severe pathological conditions. Emerging evidence indicates pathological molecular signatures in deeper endomembrane βARs signaling, likely contributing to conditions such as cardiomyocyte hypertrophy and apoptosis. However, the lack of approaches to control endomembrane β1ARs has impeded linking signaling with pathology. Informed by the β1AR-catecholamine interactions, we engineered an efficient photolabile proligand (OptoIso) to trigger βAR signaling exclusively in endomembrane regions using blue light stimulation. Not only does OptoIso undergo blue light deprotection in seconds, but also efficiently enters cells and allows examination of G protein heterotrimer activation exclusively at endomembranes. OptoIso also allows optical activation of plasma membrane βAR signaling in selected single cells with native fidelity, which can be reversed by terminating blue light. Thus, OptoIso will be a valuable experimental tool to elicit spatial and temporal control of βAR signaling in user-defined endomembrane or plasma membrane regions in unmodified cells with native fidelity.
    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:心力衰竭(HF)通常伴有交感神经的激活,交感神经的过度激活也促进了心脏重塑和心脏功能障碍。在异丙肾上腺素(ISO)诱导的动物模型中,常伴有心肌肥厚,纤维化,和炎症。白细胞免疫球蛋白样受体B4a(Lilrb4a)是一种免疫抑制性调节受体,在心血管疾病中起着至关重要的作用。然而,Lilrb4a对ISO诱导小鼠模型室性心律失常的影响尚不清楚。
    目的:本研究的目的是探讨Lilrb4a在ISO诱导的心律失常性重塑中的作用及其分子机制。
    方法:Lilrb4a敲除小鼠和Lilrb4a过表达小鼠输注ISO(15mg/kg/24h,4周)。超声心动图和组织学用于评估心肌肥厚和心脏结构重塑。体表心电图和电生理检查用于评估心脏电重构和室性心律失常(VAs)的敏感性。qRT-PCR和WesternBlot检测离子通道蛋白和信号通路蛋白的表达水平。
    结果:结果发现ISO诱导心脏肥大,纤维化,和炎症,并导致电重塑和VAs的发生。Lilrb4a减轻了心脏结构和电重构,并通过功能增益或功能丧失方法在ISO诱导的小鼠中防止了VA的发生。机制是Lilrb4a抑制TAK1介导的NF-kB信号和p38信号激活。
    结论:Lilrb4a通过调节NF-kB信号和p38信号激活,减轻与心脏纤维化和炎症相关的心功能不全和异丙肾上腺素诱导的心律失常性重塑。
    BACKGROUND: Heart failure is usually accompanied by activation of the sympathetic nerve, and excessive activation of the sympathetic nerve promotes cardiac remodeling and cardiac dysfunction. In the isoproterenol (ISO)-induced animal model, it is often accompanied by myocardial hypertrophy, fibrosis, and inflammation. Leukocyte immunoglobulin-like receptor B4a (Lilrb4a), an immunosuppressive regulatory receptor, plays a vital role in cardiovascular disease. However, the effect of Lilrb4a on ventricular arrhythmia in an ISO-induced mouse model remains unclear.
    OBJECTIVE: The purpose of this study was to explore the role and molecular mechanism of Lilrb4a in ISO-induced arrhythmogenic remodeling.
    METHODS: Lilrb4a knockout mice and Lilrb4a overexpression mice were infused with ISO (15 mg/kg per 24 hours, 4 weeks). Echocardiography and histology evaluations of myocardial hypertrophy and cardiac structural remodeling were conducted. Surface electrocardiography and electrophysiologic examination were used to evaluate cardiac electrical remodeling and susceptibility to ventricular arrhythmias. Quantitative reverse transcriptase-polymerase chain reaction analysis and Western blotting were used to detect the expression levels of ion channel proteins and signal pathway proteins.
    RESULTS: The results discovered that ISO induced cardiac hypertrophy, fibrosis, and inflammation and led to electrical remodeling and the occurrence of ventricular arrhythmias. Lilrb4a alleviated cardiac structural and electrical remodeling and protected against the occurrence of ventricular arrhythmias in ISO-induced mice by gain-of-function or loss-of-function approaches. The mechanism is that Lilrb4a inhibited NF-κB signaling and MAPK signaling activation mediated by transforming growth factor kinase 1.
    CONCLUSIONS: Lilrb4a alleviates cardiac dysfunction and ISO-induced arrhythmogenic remodeling associated with cardiac fibrosis and inflammation through the regulation of NF-κB signaling and MAPK signaling activation.
    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
    肾上腺素能途径代表神经系统和免疫系统之间的主要沟通渠道。在炎症期间,血液单核细胞在组织内迁移并分化为巨噬细胞,极化为M1或M2巨噬细胞,具有组织损伤或修复特性,分别。这项研究调查了β-肾上腺素能受体(β-AR)阻断药物普萘洛尔是否调节单核细胞到巨噬细胞的分化过程,并进一步影响巨噬细胞向M1和M2样表型的极化。在存在或不存在普萘洛尔的情况下与M-CSF一起培养6天人单核细胞,然后向M1促炎状态或M2抗炎状态活化。单核细胞在分化成巨噬细胞期间长期暴露于普萘洛尔,可促进M1标记CD16和M2标记CD206和CD163以及过氧化物酶体增殖物激活受体的表达增加。它还增加了内吞作用和IL-10的释放,而减少了生理活性氧。暴露于普萘洛尔分化的巨噬细胞的促炎条件导致抗炎促进作用。在分子水平上,普萘洛尔上调氧化应激调节剂NRF2,血红素加氧酶1和NQO1的表达。通过调节巨噬细胞的活动,普萘洛尔可能是一种新型的抗炎和免疫调节化合物,在几种炎症性疾病中具有相关的治疗潜力。
    Adrenergic pathways represent the main channel of communication between the nervous system and the immune system. During inflammation, blood monocytes migrate within tissue and differentiate into macrophages, which polarize to M1 or M2 macrophages with tissue-damaging or -reparative properties, respectively. This study investigates whether the β-adrenergic receptor (β-AR)-blocking drug propranolol modulates the monocyte-to-macrophage differentiation process and further influences macrophages in their polarization toward M1- and M2-like phenotypes. Six-day-human monocytes were cultured with M-CSF in the presence or absence of propranolol and then activated toward an M1 pro-inflammatory state or an M2 anti-inflammatory state. The chronic exposure of monocytes to propranolol during their differentiation into macrophages promoted the increase in the M1 marker CD16 and in the M2 markers CD206 and CD163 and peroxisome proliferator-activated receptor ɣ expression. It also increased endocytosis and the release of IL-10, whereas it reduced physiological reactive oxygen species. Exposure to the pro-inflammatory conditions of propranolol-differentiated macrophages resulted in an anti-inflammatory promoting effect. At the molecular level, propranolol upregulated the expression of the oxidative stress regulators NRF2, heme oxygenase-1 and NQO1. By contributing to regulating macrophage activities, propranolol may represent a novel anti-inflammatory and immunomodulating compound with relevant therapeutic potential in several inflammatory diseases.
    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:病理性心脏重构,包括心脏肥大和纤维化,是心力衰竭发展的关键病理过程。然而,有效的治疗方法是有限的。β-肾上腺素能受体是调节心脏功能的关键信号分子。G-α相互作用蛋白(GAIP)-相互作用蛋白,C末端1(GIPC1)是一种多功能支架蛋白,可直接与β1-肾上腺素能受体(β1-肾上腺素能受体)的C末端结合。然而,人们对它在心脏功能中的作用知之甚少。因此,我们研究了GIPC1在心脏重塑中的作用及其潜在的分子机制.
    方法:通过腹膜内注射异丙肾上腺素14d或横向主动脉缩窄手术8周建立小鼠病理性心脏重塑。使用Myh6驱动的心肌细胞特异性GIPC1条件敲除(GIPC1cKO)小鼠和腺相关病毒9(AAV9)介导的GIPC1过表达小鼠。使用超声心动图评估GIPC1对心脏重塑的影响,组织学,和生化分析。
    结果:GIPC1表达在心脏重塑模型中持续降低。GIPC1cKO小鼠表现出自发性异常,包括心脏肥大,纤维化,和收缩功能障碍.相比之下,AAV9介导的GIPC1在心脏中的过表达减弱了异丙肾上腺素诱导的小鼠病理性心脏重塑。机械上,GIPC1与β1-肾上腺素能受体相互作用,并通过阻止其泛素化和降解来稳定其表达,维持β1-肾上腺素受体/β2-肾上腺素受体的平衡,并抑制丝裂原活化蛋白激酶信号通路的过度活化。
    结论:这些结果表明GIPC1具有心脏保护作用,是治疗心脏重塑和心力衰竭的一个有希望的治疗靶点。
    OBJECTIVE: Pathological cardiac remodelling, including cardiac hypertrophy and fibrosis, is a key pathological process in the development of heart failure. However, effective therapeutic approaches are limited. The β-adrenergic receptors are pivotal signalling molecules in regulating cardiac function. G-alpha interacting protein (GAIP)-interacting protein, C-terminus 1 (GIPC1) is a multifunctional scaffold protein that directly binds to the C-terminus of β1-adrenergic receptor (β1-adrenergic receptor). However, little is known about its roles in heart function. Therefore, we investigated the role of GIPC1 in cardiac remodelling and its underlying molecular mechanisms.
    METHODS: Pathological cardiac remodelling in mice was established via intraperitoneal injection of isoprenaline for 14 d or transverse aortic constriction surgery for 8 weeks. Myh6-driving cardiomyocyte-specific GIPC1 conditional knockout (GIPC1 cKO) mice and adeno-associated virus 9 (AAV9)-mediated GIPC1 overexpression mice were used. The effect of GIPC1 on cardiac remodelling was assessed using echocardiographic, histological, and biochemical analyses.
    RESULTS: GIPC1 expression was consistently reduced in the cardiac remodelling model. GIPC1 cKO mice exhibited spontaneous abnormalities, including cardiac hypertrophy, fibrosis, and systolic dysfunction. In contrast, AAV9-mediated GIPC1 overexpression in the heart attenuated isoproterenol-induced pathological cardiac remodelling in mice. Mechanistically, GIPC1 interacted with the β1-adrenergic receptor and stabilised its expression by preventing its ubiquitination and degradation, maintaining the balance of β1-adrenergic receptor/β2-adrenergic receptor, and inhibiting hyperactivation of the mitogen-activated protein kinase signalling pathway.
    CONCLUSIONS: These results suggested that GIPC1 plays a cardioprotective role and is a promising therapeutic target for the treatment of cardiac remodelling and heart failure.
    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: Preprint
    β-肾上腺素能受体(βARs)是G蛋白偶联受体(GPCRs),介导儿茶酚胺诱导的应激反应,如心率增加和支气管扩张。除了来自细胞表面的信号,βAR还从细胞内膜(内膜)传播非规范的信号传导活性。这些受体途径的失调是严重病理状况的基础。过度的βAR刺激与心脏肥大有关,导致心力衰竭,而受损的刺激会导致战斗或飞行应激反应和稳态受损。除了质膜βAR,新出现的证据表明,潜在的病理意义更深的内膜βARs,如诱导心肌细胞肥大和凋亡,潜在的心力衰竭.然而,缺乏仅在亚细胞区室中控制其信号传导的方法阻碍了内膜βAR信号传导与病理学的联系。由β1AR-儿茶酚胺相互作用引起,我们设计了一个有效的照片不稳定,受保护的羟基β1AR前配体(OptoIso)在细胞表面触发βAR信号,以及蓝光刺激下的专属内膜区域。OptoIso不仅在几秒钟内进行蓝光脱保护,但它也可以有效地进入细胞,并允许仅在内膜上检查G蛋白异源三聚体的激活。除了其在未修饰细胞中βARs的光学询问中的应用外,鉴于其控制深层细胞器βAR信号的能力,OptoIso将是一个有价值的实验工具。
    Beta-adrenergic receptors (βARs) are G protein-coupled receptors (GPCRs) that mediate catecholamine-induced stress responses, such as heart rate increase and bronchodilation. In addition to signals from the cell surface, βARs also broadcast non-canonical signaling activities from the cell interior membranes (endomembranes). Dysregulation of these receptor pathways underlies severe pathological conditions. Excessive βAR stimulation is linked to cardiac hypertrophy, leading to heart failure, while impaired stimulation causes compromised fight or flight stress responses and homeostasis. In addition to plasma membrane βAR, emerging evidence indicates potential pathological implications of deeper endomembrane βARs, such as inducing cardiomyocyte hypertrophy and apoptosis, underlying heart failure. However, the lack of approaches to control their signaling in subcellular compartments exclusively has impeded linking endomembrane βAR signaling with pathology. Informed by the β1AR-catecholamine interactions, we engineered an efficiently photo-labile, protected hydroxy β1AR pro-ligand (OptoIso) to trigger βAR signaling at the cell surface, as well as exclusive endomembrane regions upon blue light stimulation. Not only does OptoIso undergo blue light deprotection in seconds, but it also efficiently enters cells and allows examination of G protein heterotrimer activation exclusively at endomembranes. In addition to its application in the optical interrogation of βARs in unmodified cells, given its ability to control deep organelle βAR signaling, OptoIso will be a valuable experimental tool.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    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
    三阴性乳腺癌(TNBC),最具侵袭性的乳腺癌亚型,以雌激素受体(ER)不出现为特征,孕激素受体(PR),和人表皮生长因子受体2(HER2)。临床上,TNBC的特点是存活率低,治疗效果不佳,高侵略性,缺乏靶向治疗。在过去的几十年里,许多针对TNBC靶向治疗的临床试验正在进行中.虽然有些班级,如聚(ADP核糖)聚合酶(PARP)抑制剂和免疫疗法,已经显示出积极的治疗结果,然而,临床效果并不令人满意。此外,耐药性的发展是许多靶向单一疗法中观察到的主要模式。TNBC的异质性可能是临床获益有限的原因。因此,,有必要识别新的治疗靶点以解决上述限制.在这种情况下,在TNBC治疗时代,一些可以解决上述问题的新靶点正在出现,其中包括缺氧诱导因子(HIF-1α),基质金属蛋白酶9(MMP-9),肿瘤坏死因子-α(TNF-α),β-肾上腺素能受体(β-AR),电压门控钠通道(VGSC),和细胞周期调节剂。目前,我们总结了正在进行的临床试验,并讨论了TNBC治疗的新治疗靶点.
    Triple-negative Breast Cancer (TNBC), the most aggressive breast cancer subtype, is characterized by the non-appearance of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Clinically, TNBC is marked by its low survival rate, poor therapeutic outcomes, high aggressiveness, and lack of targeted therapies. Over the past few decades, many clinical trials have been ongoing for targeted therapies in TNBC. Although some classes, such as Poly (ADP Ribose) Polymerase (PARP) inhibitors and immunotherapies, have shown positive therapeutic outcomes, however, clinical effects are not much satisfiable. Moreover, the development of drug resistance is the major pattern observed in many targeted monotherapies. The heterogeneity of TNBC might be the cause for limited clinical benefits. Hence,, there is a need for the potential identification of new therapeutic targets to address the above limitations. In this context, some novel targets that can address the above-mentioned concerns are emerging in the era of TNBC therapy, which include Hypoxia Inducible Factor (HIF-1α), Matrix Metalloproteinase 9 (MMP-9), Tumour Necrosis Factor-α (TNF-α), β-Adrenergic Receptor (β-AR), Voltage Gated Sodium Channels (VGSCs), and Cell Cycle Regulators. Currently, we summarize the ongoing clinical trials and discuss the novel therapeutic targets in the management of TNBC.
    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
    β2-肾上腺素受体(β2-AR)激动剂用于治疗哮喘和慢性阻塞性肺疾病,但也在包括癌症在内的其他复杂疾病中发挥作用,糖尿病和心脏病。由于β2-AR在各种细胞和组织中的细胞和分子机制仍然非常难以捉摸,我们开发了具有高时空分辨率的调查工具。合成了几种具有纳摩尔活性的光可转换β2-AR激动剂。具有合理转换的β2-AR的最有效激动剂是一位数纳摩尔活性物质,反式-on芳基唑并吡唑的肾上腺素衍生物,具有有价值的光药理学性质,可用于进一步的生物学研究,具有与天然配体肾上腺素的高度结构。
    β2 -adrenergic receptor (β2 -AR) agonists are used for the treatment of asthma and chronic obstructive pulmonary disease, but also play a role in other complex disorders including cancer, diabetes and heart diseases. As the cellular and molecular mechanisms in various cells and tissues of the β2 -AR remain vastly elusive, we developed tools for this investigation with high temporal and spatial resolution. Several photoswitchable β2 -AR agonists with nanomolar activity were synthesized. The most potent agonist for β2 -AR with reasonable switching is a one-digit nanomolar active, trans-on arylazopyrazole-based adrenaline derivative and comprises valuable photopharmacological properties for further biological studies with high structural accordance to the native ligand adrenaline.
    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