Receptor, Adenosine A1

受体,腺苷 A1
  • 文章类型: Journal Article
    介导全身性炎症进入脑实质以诱发脓毒症相关性脑病(SAE)的分子途径仍然难以捉摸。这里,我们报道,在小鼠外周脂多糖(LPS)诱发的全身性炎症(6hpi)的前6小时,腺苷的血浆水平迅速增加并增强了中枢细胞外腺苷的张力,然后通过触发早期星形胶质细胞反应性引起神经炎症。星形细胞Gi蛋白偶联A1腺苷受体(A1ARs)的特异性消融可预防这种早期反应并降低炎症因子的水平(例如,CCL2,CCL5和CXCL1)在星形胶质细胞中,从而减轻小胶质反应,改善血脑屏障破坏,外周免疫细胞浸润,神经元功能障碍,和老鼠的抑郁样行为。在LPS注射2和4hpi时,A1AR缺陷星形胶质细胞中Gi信号的化学遗传刺激可以恢复神经炎症和抑郁样行为,强调星形胶质细胞而不是小胶质细胞是神经炎症的早期驱动因素。我们的结果确定了早期星形胶质细胞对周围和中枢腺苷水平的反应性是驱动SAE的重要途径,并强调了靶向A1AR进行治疗干预的潜力。
    Molecular pathways mediating systemic inflammation entering the brain parenchyma to induce sepsis-associated encephalopathy (SAE) remain elusive. Here, we report that in mice during the first 6 hours of peripheral lipopolysaccharide (LPS)-evoked systemic inflammation (6 hpi), the plasma level of adenosine quickly increased and enhanced the tone of central extracellular adenosine which then provoked neuroinflammation by triggering early astrocyte reactivity. Specific ablation of astrocytic Gi protein-coupled A1 adenosine receptors (A1ARs) prevented this early reactivity and reduced the levels of inflammatory factors (e.g., CCL2, CCL5, and CXCL1) in astrocytes, thereby alleviating microglial reaction, ameliorating blood-brain barrier disruption, peripheral immune cell infiltration, neuronal dysfunction, and depression-like behaviour in the mice. Chemogenetic stimulation of Gi signaling in A1AR-deficent astrocytes at 2 and 4 hpi of LPS injection could restore neuroinflammation and depression-like behaviour, highlighting astrocytes rather than microglia as early drivers of neuroinflammation. Our results identify early astrocyte reactivity towards peripheral and central levels of adenosine as an important pathway driving SAE and highlight the potential of targeting A1ARs for therapeutic intervention.
    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
    代谢(功能障碍)相关的脂肪性肝炎(MASH)是代谢(功能障碍)相关的脂肪肝(MAFLD)缺乏批准的临床药物的晚期阶段。腺苷A1受体(A1R),属于G蛋白偶联受体(GPCRs)超家族,主要分布在中枢神经系统和主要外周器官中,具有广泛的生理功能;然而,肝A1R在MAFLD中的确切作用尚不清楚.这里,我们报道,A1R的肝脏特异性耗竭加重,而过表达减弱小鼠饮食诱导的代谢相关脂肪肝(MAFL)/MASH.机械上,肝A1R的激活促进甾醇调节元件结合蛋白(SREBP)裂解激活蛋白(SCAP)与螯合体1(SQSTM1)的竞争性结合,而不是导致溶酶体中SCAP降解的蛋白激酶A(PKA)。减少的SCAP阻碍SREBP1c/2成熟并因此抑制从头脂肪生成和炎症。在MAFL/MASH和高脂饮食(HFD)喂养的小鼠中观察到较高的肝A1R表达,这被认为是生理适应性反应,因为A1R激动剂以A1R依赖性方式减弱MAFL/MASH。这些结果强调肝A1R是MAFL/MASH治疗的潜在靶标。
    Metabolic (dysfunction)-associated steatohepatitis (MASH) is the advanced stage of metabolic (dysfunction)-associated fatty liver disease (MAFLD) lacking approved clinical drugs. Adenosine A1 receptor (A1R), belonging to the G-protein-coupled receptors (GPCRs) superfamily, is mainly distributed in the central nervous system and major peripheral organs with wide-ranging physiological functions; however, the exact role of hepatic A1R in MAFLD remains unclear. Here, we report that liver-specific depletion of A1R aggravates while overexpression attenuates diet-induced metabolic-associated fatty liver (MAFL)/MASH in mice. Mechanistically, activation of hepatic A1R promotes the competitive binding of sterol-regulatory element binding protein (SREBP) cleavage-activating protein (SCAP) to sequestosome 1 (SQSTM1), rather than protein kinase A (PKA) leading to SCAP degradation in lysosomes. Reduced SCAP hinders SREBP1c/2 maturation and thus suppresses de novo lipogenesis and inflammation. Higher hepatic A1R expression is observed in patients with MAFL/MASH and high-fat diet (HFD)-fed mice, which is supposed to be a physiologically adaptive response because A1R agonists attenuate MAFL/MASH in an A1R-dependent manner. These results highlight that hepatic A1R is a potential target for MAFL/MASH therapy.
    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
    咖啡因消耗通过阻断腺苷信号抑制针灸镇痛作用。然而,现有证据仍有争议。因此,这项研究旨在研究腺苷A1受体(A1R)在中等剂量咖啡因诱导的消除A1R基因敲除小鼠(A1R-/-)针刺镇痛作用中的作用。我们通过在野生型(WT)和A1R-/-小鼠中测量机械和热痛阈值并施用A1R和A2AR拮抗剂,评估了A1R在生理感官知觉中的作用及其与咖啡因的相互作用。建立甲醛和完全弗氏佐剂(CFA)诱导的炎性疼痛模型,以探讨中等剂量咖啡因对WT和A1R-/-小鼠疼痛感知和针刺镇痛的影响。此外,在股二头肌中进行了C纤维反射肌电图检查,以验证A1R在咖啡因诱导的针刺镇痛阻滞中的作用。我们发现A1R对于生理感觉和福尔马林和CFA诱导的超敏反应是不必要的。然而,A1R的基因缺失损害了A1R-/-小鼠在生理或炎性疼痛条件下针刺的镇痛作用。在生理条件下,急性中等剂量咖啡因给药可引起机械和热痛觉过敏,但在福尔马林和CFA引起的炎性疼痛中却没有。此外,咖啡因能显著抑制WT小鼠生理性和炎性疼痛的EA镇痛,与A1R拮抗剂相当。相反,A1R缺失会损害EA的镇痛作用,并降低咖啡因在生理条件和炎性疼痛中对EA镇痛的抑制作用。中等剂量的咖啡因给药通过阻断A1R降低了EA诱导的镇痛作用。总的来说,我们的研究表明,在针灸治疗期间应避免摄入咖啡因。结果:通过阻断A1R,中等剂量的咖啡因注射在福尔马林和CFA诱导的炎性疼痛小鼠模型中减弱了EA诱导的镇痛作用。这凸显了在针灸治疗期间监测咖啡因摄入量的重要性。
    Caffeine consumption inhibits acupuncture analgesic effects by blocking adenosine signaling. However, existing evidence remains controversial. Hence, this study aimed to examine the adenosine A1 receptor (A1R) role in moderate-dose caffeine-induced abolishing effect on acupuncture analgesia using A1R knockout mice (A1R-/-). We assessed the role of A1R in physiological sensory perception and its interaction with caffeine by measuring mechanical and thermal pain thresholds and administering A1R and adenosine 2A receptor antagonists in wild-type (WT) and A1R-/- mice. Formalin- and complete Freund\'s adjuvant (CFA)-induced inflammatory pain models were recruited to explore moderate-dose caffeine effect on pain perception and acupuncture analgesia in WT and A1R-/- mice. Moreover, a C-fiber reflex electromyogram in the biceps femoris was conducted to validate the role of A1R in the caffeine-induced blockade of acupuncture analgesia. We found that A1R was dispensable for physiological sensory perception and formalin- and CFA-induced hypersensitivity. However, genetic deletion of A1R impaired the antinociceptive effect of acupuncture in A1R-/- mice under physiological or inflammatory pain conditions. Acute moderate-dose caffeine administration induced mechanical and thermal hyperalgesia under physiological conditions but not in formalin- and CFA-induced inflammatory pain. Moreover, caffeine significantly inhibited electroacupuncture (EA) analgesia in physiological and inflammatory pain in WT mice, comparable to that of A1R antagonists. Conversely, A1R deletion impaired the EA analgesic effect and decreased the caffeine-induced inhibitory effect on EA analgesia in physiological conditions and inflammatory pain. Moderate-dose caffeine administration diminished the EA-induced antinociceptive effect by blocking A1R. Overall, our study suggested that caffeine consumption should be avoided during acupuncture treatment. PERSPECTIVE: Moderate-dose caffeine injection attenuated EA-induced antinociceptive effect in formalin- and CFA-induced inflammatory pain mice models by blocking A1R. This highlights the importance of monitoring caffeine intake during acupuncture treatment.
    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
    目的:为了研究鞘内注射CCPA的效果,腺苷A1受体激动剂,环磷酰胺(CYP)致膀胱炎大鼠排尿功能的研究.
    方法:将30只8周龄SD大鼠随机分为对照组(n=15)和膀胱炎组(n=15)。通过单次腹腔注射CYP(200mg/kg,溶解在生理盐水中)。对照组大鼠腹腔注射生理盐水。PE10导管通过L3-4椎间隙到达L6-S1脊髓水平,鞘内注射。腹腔注射后48小时,进行尿动力学测试,以观察鞘内给药10%二甲基亚砜(载体)和1nmolCCPA对排尿参数的影响,包括基础压(BP),阈值压力(TP),最大排尿压力(MVP),收缩间期(ICI),作废卷(VV),剩余体积(RV),膀胱容量(BC),和排尿效率(VE)。通过苏木精-伊红染色(HE染色)研究膀胱炎大鼠膀胱的组织学变化。此外,采用Westernblot和免疫荧光法研究两组大鼠脊髓背侧L6-S1中腺苷A1受体的表达。
    结果:HE染色显示粘膜下出血,水肿,膀胱炎大鼠膀胱壁炎性细胞浸润。尿动力学试验显示血压显著升高,TP,膀胱炎大鼠的MVP和RV,而ICI,VV,BC和VE显著下降,表明膀胱过度活动。CCPA抑制对照组和膀胱炎大鼠的排尿反射,TP显著增加,ICI,VV,BC,和VE,但对血压没有显著影响,MVP和RV。Westernblot和免疫荧光显示,对照组和膀胱炎大鼠L6-S1脊髓背侧腺苷A1受体的表达无明显差异。
    结论:本研究结果表明鞘内注射腺苷A1受体激动剂CCPA可缓解CYP诱导的膀胱过度活动。此外,我们的研究结果表明,腰骶脊髓的腺苷A1受体可能是治疗膀胱过度活动症的一个有希望的靶点.
    OBJECTIVE: To investigate the effect of intrathecal administration of CCPA, an adenosine A1 receptor agonist, on voiding function in rats with cystitis induced by cyclophosphamide (CYP).
    METHODS: Thirty 8-week-old Sprague Dawley rats were randomly divided into a control group (n = 15) and a cystitis group (n = 15). Cystitis was induced by a single intraperitoneal injection of CYP (200 mg/kg, dissolved in physiological saline) in rats. Control rats were injected intraperitoneally with physiological saline. The PE10 catheter reached the level of L6-S1 spinal cord through L3-4 intervertebral space for intrathecal injection. Forty-eight hours after intraperitoneal injection, urodynamic tests were conducted to observe the effect of intrathecal administration of 10% dimethylsulfoxide (vehicle) and 1 nmol CCPA on micturition parameters, including basal pressure (BP), threshold pressure (TP), maximal voiding pressure (MVP), intercontraction interval (ICI), voided volume (VV), residual volume (RV), bladder capacity (BC), and voiding efficiency (VE). Histological changes of the bladder of cystitis rats were studied through hematoxylin-eosin staining (HE staining). Moreover, Western blot and immunofluorescence were used to study the expression of adenosine A1 receptor in the L6-S1 dorsal spinal cord in both groups of rats.
    RESULTS: HE staining revealed submucosal hemorrhage, edema, and inflammatory cell infiltration in the bladder wall of cystitis rats. The urodynamic test showed significant increase in BP, TP, MVP and RV in cystitis rats, while ICI, VV, BC and VE decreased significantly, indicating bladder overactivity. CCPA inhibited the micturition reflex in both control and cystitis rats, and significantly increased TP, ICI, VV, BC, and VE, but had no significant effect on BP, MVP and RV. Western blot and immunofluorescence showed that there was no significant difference in the expression of adenosine A1 receptor in the L6-S1 dorsal spinal cord between the control and cystitis rats.
    CONCLUSIONS: The findings of this study suggest that intrathecal administration of the adenosine A1 receptor agonist CCPA alleviates CYP-induced bladder overactivity. Furthermore, our results indicate that the adenosine A1 receptor in the lumbosacral spinal cord may be a promising target for treatment of bladder overactivity.
    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
    腺苷A1受体在调节游离脂肪酸(FFA)水平中起重要作用,并且代表代谢紊乱的有吸引力的靶标。尽管在利用有效(正构)A1受体激动剂调节异常FFA水平方面取得了显着进展,A1受体变构调节对脂质稳态的影响研究较少。在此,我们试图探索变构调节剂对A1受体正构激动剂在体外和体内调节脂解过程中的作用。我们检查了在不存在或存在变构调节剂(2-氨基-4,5-二甲基-3-噻吩基)-[3-(三氟甲基)-苯基]甲酮(PD81,723)的情况下,选择性A1受体激动剂2-氯-N6-环戊基腺苷(CCPA)在大鼠脂肪细胞膜上的结合动力学。我们还检查了PD81,723对介导CCPA诱导的cAMP积累抑制的变构效应,体外和体内模型中的HSL(激素敏感性脂肪酶)磷酸化和FFA产生。我们的结果表明,PD81,723减缓了CCPA从A1受体的解离,which,因此,通过下调cAMP/HSL途径增强CCPA的抗脂解作用。我们的研究举例说明了A1受体变构调节剂作为代谢疾病治疗替代方案的应用。
    The adenosine A1 receptor plays important roles in tuning free fatty acid (FFA) levels and represents an attractive target for metabolic disorders. Though remarkable progress has been achieved in the exploitation of effective (orthosteric) A1 receptor agonists in modulating aberrant FFA levels, the effect of A1 receptor allosteric modulation on lipid homeostasis is less investigated. Herein we sought to explore the effect of an allosteric modulator on the action of an A1 receptor orthosteric agonist in regulating the lipolytic process in vitro and in vivo. We examined the binding kinetics of a selective A1 receptor agonist 2-chloro-N6-cyclopentyladenosine (CCPA) in the absence or presence of an allosteric modulator (2-amino-4,5-dimethyl-3-thienyl)-[3-(trifluoromethyl)-phenyl]methanone (PD81,723) on rat adipocyte membranes. We also examined the allosteric effects of PD81,723 on mediating the CCPA-induced inhibition of cAMP accumulation, HSL (hormone-sensitive lipase) phosphorylation and FFA production in in vitro and in vivo models. Our results demonstrated that PD81,723 slowed down the dissociation of CCPA from the A1 receptor, which, consequently, potentiated the antilipolytic action of CCPA through downregulating the cAMP/HSL pathway. Our study exemplified the application of A1 receptor allosteric modulators as an alternative for metabolic disease treatments.
    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
    丘脑前核的深部脑刺激(DBS),是耐药癫痫患者的有效治疗方法,然而,其作用机制仍然难以捉摸。腺苷激酶(ADK),腺苷的关键负调节剂,是癫痫发生的潜在调节剂。DBS已被证明可以增加腺苷水平,可能通过A1受体(A1Rs)抑制癫痫发作。我们调查了DBS是否可以阻止疾病进展以及腺苷机制的潜在参与。
    对照组,SE(癫痫持续状态)组,SE-DBS组,SE-sham-DBS组纳入本研究。毛果芸香碱诱发癫痫持续状态一周后,SE-DBS组大鼠给予DBS治疗4周。通过视频EEG监测大鼠。用组织化学和蛋白质印迹测试ADK和A1Rs,分别。
    与SE组和SE-sham-DBS组比较,DBS可以减少自发性复发性癫痫发作(SRS)的频率和发作间癫痫放电的数量。DPCPX,A1-R拮抗剂,逆转DBS对发作间癫痫放电的影响。此外,DBS抑制ADK的过表达和A1Rs的下调。
    结果表明,DBS可以通过抑制ADK和激活A1Rs来降低癫痫大鼠的SRS。A1Rs可能是DBS治疗癫痫的潜在靶点。
    Deep brain stimulation (DBS) of the anterior nucleus of the thalamus, is an effective therapy for patients with drug-resistant epilepsy, yet, its mechanism of action remains elusive. Adenosine kinase (ADK), a key negative regulator of adenosine, is a potential modulator of epileptogenesis. DBS has been shown to increase adenosine levels, which may suppress seizures via A1 receptors (A1 Rs). We investigated whether DBS could halt disease progression and the potential involvement of adenosine mechanisms.
    Control group, SE (status epilepticus) group, SE-DBS group, and SE-sham-DBS group were included in this study. One week after a pilocarpine-induced status epilepticus, rats in the SE-DBS group were treated with DBS for 4 weeks. The rats were monitored by video-EEG. ADK and A1 Rs were tested with histochemistry and western blot, respectively.
    Compared with the SE group and SE-sham-DBS group, DBS could reduce the frequency of spontaneous recurrent seizures (SRS) and the number of interictal epileptic discharges. The DPCPX, an A1 R antagonist, reversed the effect of DBS on interictal epileptic discharges. In addition, DBS inhibited the overexpression of ADK and the downregulation of A1 Rs.
    The findings indicate that DBS can reduce SRS in epileptic rats via inhibition of ADK and activation of A1 Rs. A1 Rs might be a potential target of DBS for the treatment of epilepsy.
    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:奥沙利铂是一种常用的铂类化疗药物,而周围神经毒性是一种广泛观察到的不良反应,缺乏令人满意的治疗策略。作为共同神经病表型基础的不同腺苷受体通过不同的病理生理机制发挥不同的作用。在这项研究中,我们研究了腺苷受体A1(A1R)在奥沙利铂诱导的神经性疼痛中的作用及其在有效治疗策略中的潜在用途.
    方法:我们建立了奥沙利铂诱导的神经病理性疼痛模型,模拟了化疗给药方式,观察了相关的神经病理性行为表型和相关机制。
    结果:每周5次注射奥沙利铂2周,在小鼠中诱发严重和持续的神经性疼痛表型。在此过程中,脊髓背角中的A1R表达下降。针对A1R的药物干预证实了其在这一过程中的重要性。机械上,A1R表达的缺失主要归因于其在星形胶质细胞中的表达减少。与药理学结果一致,奥沙利铂诱导的神经性疼痛表型被A1R通过慢病毒载体在星形胶质细胞中的特异性治疗干预阻断,谷氨酸代谢相关蛋白的表达上调。神经性疼痛可以通过该途径通过药理学或星形细胞干预来缓解。
    结论:这些数据揭示了与奥沙利铂诱导的外周神经性疼痛有关的特定腺苷受体信号通路,这与星形胶质细胞A1R信号通路的抑制有关。这可能为奥沙利铂化疗期间观察到的神经性疼痛的治疗和管理提供新的机会。本文受版权保护。保留所有权利。
    Oxaliplatin is a commonly used platinum chemotherapy drug, whereas peripheral neurotoxicity is a widely observed adverse reaction lacking a satisfactory therapeutic strategy. Different adenosine receptors underlying the common neuropathic phenotype play different roles through varied pathophysiological mechanisms. In this study, we investigated the role of adenosine receptor A1 (A1R) in oxaliplatin-induced neuropathic pain and its potential use in an effective therapeutic strategy.
    We established an oxaliplatin-induced neuropathic pain model simulating the mode of chemotherapy administration and observed the related neuropathic behavioral phenotype and implicated mechanisms.
    Five weekly injections of oxaliplatin for 2 weeks induced a severe and persistent neuropathic pain phenotype in mice. A1R expression in the spinal dorsal horn decreased during this process. Pharmacological intervention against A1R verified its importance in this process. Mechanistically, the loss of A1R expression was mainly attributed to its decreased expression in astrocytes. Consistent with the pharmacological results, the oxaliplatin-induced neuropathic pain phenotype was blocked by specific therapeutic interventions of A1R in astrocytes via lentiviral vectors, and the expression of glutamate metabolism-related proteins was upregulated. Neuropathic pain can be alleviated by pharmacological or astrocytic interventions via this pathway.
    These data reveal a specific adenosine receptor signaling pathway involved in oxaliplatin-induced peripheral neuropathic pain, which is related to the suppression of the astrocyte A1R signaling pathway. This may provide new opportunities for the treatment and management of neuropathic pain observed during oxaliplatin chemotherapy.
    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
    我们之前的研究发现,腺苷A1受体(A1R)的激活诱导δ阿片受体(DOR)的磷酸化和其下游信号分子的脱敏,cAMP和Akt。为了进一步研究A1R激动剂对DOR信号的影响及其机制,我们研究了A1R激活对其激动剂N6-环己基腺苷(CHA)结合DOR介导的Raf-1/MEK/ERK激活的影响,发现长时间CHA暴露导致DOR介导的Raf-1/MEK/ERK信号通路下调。CHA处理时间依赖性减弱[D-Pen2,5]脑啡肽(DPDPE)诱导的Raf-1-Ser338磷酸化,DOR的特定激动剂,并进一步引起DOR激动剂激活的Raf-1/MEK/ERK信号通路的下调。此外,CHA暴露时间依赖性地诱导Raf-1-Ser289/296/301的磷酸化,这些磷酸化位点受负反馈调节,从而抑制MEK/ERK途径的激活,这种作用可以被MEK抑制剂U0126阻断。最后,我们证明了Raf-1/MEK/ERK级联的异源脱敏在DOR激动剂的抗伤害性作用的调节中是必不可少的,通过证实CHA的预处理可以抑制这种作用。因此,我们得出结论,A1R的激活通过Raf-1/MEK/ERK级联的异源脱敏抑制DOR介导的MAPK信号通路,这是ERK介导的Raf-1-Ser289/296/301磷酸化的结果,由A1R的激活介导。
    Our previous study found that activation of adenosine A1 receptor (A1R) induced phosphorylation of delta opioid receptor (DOR) and desensitization of its downstream signaling molecules, cAMP and Akt. To further investigate the effect of A1R agonist on DOR signaling and the underlying mechanism, we examined the effect of A1R activation upon binding of its agonist N6-cyclohexyl-adenosine (CHA) on DOR-mediated Raf-1/MEK/ERK activation, and found that prolonged CHA exposure resulted in downregulation of DOR-mediated Raf-1/MEK/ERK signaling pathway. CHA-treatment time dependently attenuated Raf-1-Ser338 phosphorylation induced by [D-Pen2,5] enkephalin (DPDPE), a specific agonist of DOR, and further caused downregulation of the Raf-1/MEK/ERK signaling pathway activated by DOR agonist. Moreover, CHA exposure time-dependently induced the phosphorylation of Raf-1-Ser289/296/301, the inhibitory phosphorylation sites that were regulated by negative feedback, thereby inhibiting activation of the MEK/ERK pathway, and this effect could be blocked by MEK inhibitor U0126. Finally, we proved that the heterologous desensitization of the Raf-1/MEK/ERK cascade was essential in the regulation of anti-nociceptive effect of DOR agonists by confirming that such effect was inhibited by pretreatment of CHA. Therefore, we conclude that the activation of A1R inhibits DOR-mediated MAPK signaling pathway via heterologous desensitization of the Raf-1/MEK/ERK cascade, which is a result of ERK-mediated Raf-1-Ser289/296/301 phosphorylation mediated by activation of A1R.
    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
    The full activation process of G protein-coupled receptor (GPCR) plays an important role in cellular signal transduction. However, it remains challenging to simulate the whole process in which the GPCR is recognized and activated by a ligand and then couples to the G protein on a reasonable simulation timescale. Here, we developed a molecular dynamics (MD) approach named supervised (Su) Gaussian accelerated MD (GaMD) by incorporating a tabu-like supervision algorithm into a standard GaMD simulation. By using this Su-GaMD method, from the active and inactive structure of adenosine A1 receptor (A1R), we successfully revealed the full activation mechanism of A1R, including adenosine (Ado)-A1R recognition, preactivation of A1R, and A1R-G protein recognition, in hundreds of nanoseconds of simulations. The binding of Ado to the extracellular side of A1R initiates conformational changes and the preactivation of A1R. In turn, the binding of Gi2 to the intracellular side of A1R causes a decrease in the volume of the extracellular orthosteric site and stabilizes the binding of Ado to A1R. Su-GaMD could be a useful tool to reconstruct or even predict ligand-protein and protein-protein recognition pathways on a short timescale. The intermediate states revealed in this study could provide more detailed complementary structural characterizations to facilitate the drug design of A1R in the future.
    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
    作为神经调质,腺苷及其受体是睡眠-觉醒调节的介质。在我们的生物信息学分析中已经预测了CREB1和抑郁症之间的推定相关性,并且它的表达也被预测为响应于睡眠剥夺而上调。因此,这项研究旨在阐述A1和A2A腺苷受体以及CREB1相关机制,这些机制是快速眼动睡眠剥夺(REMSD)在慢性不可预测的轻度应激(CUMS)诱导的抑郁样行为大鼠中的抗抑郁作用。给模型大鼠注射腺苷A1受体拮抗剂DPCPX或腺苷A2A受体拮抗剂ZM241385以评估腺苷受体在抑郁症中的作用。此外,CREB1和YAP1的异位表达和耗竭实验也在体内和体外进行。发现REMSD减轻了CUMS大鼠的抑郁样行为,如自发活动增加所示,蔗糖消耗和百分比,缩短了逃避延迟和不动持续时间。同时,A1或A2A腺苷受体拮抗剂否定了REMSD的抗抑郁作用。REMSD增强腺苷受体的激活,促进CREB1的磷酸化,从而增加CREB1的表达。此外,CREB1的过表达激活了YAP1/c-Myc轴,从而减轻了抑郁样行为。总的来说,我们的结果为理解REMSD的抗抑郁作用提供了新的机制见解,这与腺苷受体和CREB1/YAP1/c-Myc轴的激活有关。
    As neuromodulators, adenosine and its receptors are mediators of sleep-wake regulation. A putative correlation between CREB1 and depression has been predicted in our bioinformatics analyses, and its expression was also predicted to be upregulated in response to sleep deprivation. Therefore, this study aims to elaborate the A1 and A2A adenosine receptors and CREB1-associated mechanism underlying the antidepressant effect of rapid eye movement sleep deprivation (REMSD) in rats with chronic unpredictable mild stress (CUMS)-induced depressive-like behaviors. The modeled rats were injected with adenosine A1 receptor antagonist DPCPX or adenosine A2A receptor antagonist ZM241385 to assess the role of adenosine receptors in depression. In addition, ectopic expression and depletion experiments of CREB1 and YAP1 were also conducted in vivo and in vitro. It was found that REMSD alleviated depressive-like behaviors in CUMS rats, as shown by increased spontaneous activity, sucrose consumption and percentage, and shortened escape latency and immobility duration. Meanwhile, A1 or A2A adenosine receptor antagonists negated the antidepressant effect of REMSD. REMSD enhanced adenosine receptor activation and promoted the phosphorylation of CREB1, thus increasing the expression of CREB1. In addition, the overexpression of CREB1 activated the YAP1/c-Myc axis and consequently alleviated depressive-like behaviors. Collectively, our results provide new mechanistic insights for an understanding of the antidepressant effect of REMSD, which is associated with the activation of adenosine receptors and the CREB1/YAP1/c-Myc axis.
    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