calcium-activated potassium channels

钙激活钾通道
  • 文章类型: Journal Article
    钙激活钾通道(KCa)是钙信号通路的重要参与者,因为它们能够通过增加细胞内游离钙浓度而被激活。KCa通道参与正常和病理生理条件下的细胞过程的调节,包括生物转化。以前,使用膜片钳,我们记录了人类慢性粒细胞白血病K562细胞质膜中的KCa电流,其活性受机械敏感性钙渗透通道的局部Ca2进入控制。这里,我们进行了KCa通道的分子和功能鉴定,并揭示了它们在增殖中的作用,K562细胞的迁移和侵袭。使用组合方法,我们确定了细胞质膜中SK2,SK3和IK通道的功能活性。选择性SK和IK通道抑制剂,分别是阿帕明和TRAM-34,减少了增殖,人类骨髓性白血病细胞的迁移和侵袭能力。同时,K562细胞的活力不受KCa通道抑制剂的影响。Ca2成像显示,SK和IK通道抑制剂均影响Ca2进入,这可能是观察到的K562细胞病理生理反应抑制的基础。我们的数据表明,SK/IK通道抑制剂可用于减缓在质膜中表达功能活性KCa通道的慢性髓性白血病K562细胞的增殖和扩散。
    Calcium-activated potassium channels (KCa) are important participants in calcium signaling pathways due to their ability to be activated by an increase in intracellular free calcium concentration. KCa channels are involved in the regulation of cellular processes in both normal and pathophysiological conditions, including oncotransformation. Previously, using patch-clamp, we registered the KCa currents in the plasma membrane of human chronic myeloid leukemia K562 cells, whose activity was controlled by local Ca2+ entry via mechanosensitive calcium-permeable channels. Here, we performed the molecular and functional identification of KCa channels and have uncovered their role in the proliferation, migration and invasion of K562 cells. Using a combined approach, we identified the functional activity of SK2, SK3 and IK channels in the plasma membrane of the cells. Selective SK and IK channel inhibitors, apamin and TRAM-34, respectively, reduced the proliferative, migratory and invasive capabilities of human myeloid leukemia cells. At the same time, the viability of K562 cells was not affected by KCa channel inhibitors. Ca2+ imaging showed that both SK and IK channel inhibitors affect Ca2+ entry and this could underlie the observed suppression of pathophysiological reactions of K562 cells. Our data imply that SK/IK channel inhibitors could be used to slow down the proliferation and spreading of chronic myeloid leukemia K562 cells that express functionally active KCa channels in the plasma membrane.
    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
    钾(K+)通道建立并维持大多数活细胞的静息潜能。它们的活性主要受膜电压或跨细胞膜的K+梯度调节。然而,许多细胞也表达小电导钙激活钾(SK)通道,它们具有独特的能力来翻译细胞内第二信使水平的变化,Ca2+对膜K+电导的变化,因此,静息膜电位。本文回顾了该结构,存在,分布,和SK通道的功能,他们的药理调制,以及它们在健康和疾病中的作用,强调伤害性和疼痛。
    Potassium (K+) channels establish and maintain the resting potential of most living cells. Their activity is predominantly regulated by the membrane voltage or the K+ gradient across the cell membrane. However, many cells also express small-conductance calcium-activated potassium (SK) channels, which have the unique ability to translate changes in the level of the intracellular second messenger, Ca2+ to changes in the membrane K+ conductance and, therefore, the resting membrane potential. This article reviews the structure, presence, distribution, and function of SK channels, their pharmacological modulation, and their role in health and disease, emphasizing nociception and pain.
    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
    背景:S1P(鞘氨醇-1-磷酸)已被报道具有血管舒张特性,但是潜在的途径在很大程度上是未知的。
    方法:用离体小鼠肠系膜动脉和内皮细胞模型测定S1P诱导的血管舒张功能,细胞内钙,膜电位,和钙激活钾通道(KCa2.3和KCa3.1)。评估内皮S1PR1(1型S1P受体)缺失对血管舒张和血压的影响。
    结果:接受急性S1P刺激的肠系膜动脉显示出剂量依赖性血管舒张反应,通过阻断内皮KCa2.3或KCa3.1通道而减弱。在培养的人脐静脉内皮细胞中,在用升高的胞浆Ca2激活KCa2.3/KCa3.1后,S1P刺激了立即膜电位超极化。Further,长期S1P刺激以剂量和时间依赖性方式增强人脐静脉内皮细胞中KCa2.3和KCa3.1的表达,通过破坏S1PR1-Ca2信号或下游Ca2激活的钙调磷酸酶/NFAT(活化T细胞的核因子)信号而消除。结合基于生物信息学的结合位点预测和染色质免疫沉淀分析,我们发现,在人脐静脉内皮细胞中,S1P/S1PR1的慢性激活促进NFATc2核易位,并与KCa2.3和KCa3.1基因的启动子区结合,从而上调这些通道的转录.内皮细胞S1PR1的缺失降低了肠系膜动脉中KCa2.3和KCa3.1的表达,并加剧了血管紧张素II输注小鼠的高血压。
    结论:本研究为KCa2.3/KCa3.1激活的内皮依赖性超极化在响应S1P的血管舒张和血压稳态中的机制作用提供了证据。这种机制证明将有助于开发与高血压相关的心血管疾病的新疗法。
    S1P (sphingosine-1-phosphate) has been reported to possess vasodilatory properties, but the underlying pathways are largely unknown.
    Isolated mouse mesenteric artery and endothelial cell models were used to determine S1P-induced vasodilation, intracellular calcium, membrane potentials, and calcium-activated potassium channels (KCa2.3 and KCa3.1 [endothelial small- and intermediate-conductance calcium-activated potassium channels]). Effect of deletion of endothelial S1PR1 (type 1 S1P receptor) on vasodilation and blood pressure was evaluated.
    Mesenteric arteries subjected to acute S1P stimulation displayed a dose-dependent vasodilation response, which was attenuated by blocking endothelial KCa2.3 or KCa3.1 channels. In cultured human umbilical vein endothelial cells, S1P stimulated immediate membrane potential hyperpolarization following activation of KCa2.3/KCa3.1 with elevated cytosolic Ca2+. Further, chronic S1P stimulation enhanced expression of KCa2.3 and KCa3.1 in human umbilical vein endothelial cells in dose- and time-dependent manners, which was abolished by disrupting either S1PR1-Ca2+ signaling or downstream Ca2+-activated calcineurin/NFAT (nuclear factor of activated T-cells) signaling. By combination of bioinformatics-based binding site prediction and chromatin immunoprecipitation assay, we revealed in human umbilical vein endothelial cells that chronic activation of S1P/S1PR1 promoted NFATc2 nuclear translocation and binding to promoter regions of KCa2.3 and KCa3.1 genes thus to upregulate transcription of these channels. Deletion of endothelial S1PR1 reduced expression of KCa2.3 and KCa3.1 in mesenteric arteries and exacerbated hypertension in mice with angiotensin II infusion.
    This study provides evidence for the mechanistic role of KCa2.3/KCa3.1-activated endothelium-dependent hyperpolarization in vasodilation and blood pressure homeostasis in response to S1P. This mechanistic demonstration would facilitate the development of new therapies for cardiovascular diseases associated with hypertension.
    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
    尽管在早期阿尔茨海默病(AD)患者中发现了嗅觉缺陷,潜在机制尚不清楚.在这里,我们研究了人类淀粉样蛋白β(Aβ)低聚物是否以及如何影响成年小鼠梨状皮层(PC)切片中的神经活性。我们发现寡聚Aβ1-42降低了前PC中锥体神经元的兴奋性。该作用未被谷氨酸或GABAA受体拮抗剂阻断,表明Aβ1-42诱导的低活动独立于谷氨酸能和GABA能传递。有趣的是,兴奋性低下被5-羟色胺(5-HT)阻断,被5-HT2C受体拮抗剂阻断,磷脂酶C(PLC),和钙激活钾(BK)通道。此外,在存在BK通道阻断剂的情况下,Aβ1-42寡聚体无法增加K通道电流。最后,5-HT2C受体拮抗剂改善APP/PS1小鼠的嗅觉记忆和气味辨别。上述数据表明Aβ破坏了通过5-HT-5-HT2C受体-PLC-BK通道途径从PC输出的嗅觉信息。这项研究表明,血清素能调节是AD中嗅觉损伤的潜在新治疗靶标。
    Although olfactory deficits have been found in patients with early-stage Alzheimer\'s disease (AD), the underlying mechanisms remain unclear. Here we investigated whether and how human amyloid β (Aβ) oligomers affect neural activity in the piriform cortex (PC) slices of adult mice. We found that oligomeric Aβ1-42 decreased the excitability of pyramidal neurons in the anterior PC. The effect was not blocked by glutamate or GABAA receptor antagonists, suggesting that Aβ1-42-induced hypoactivity is independent of glutamatergic and GABAergic transmission. Interestingly, the hypoexcitability was occluded by serotonin (5-HT) and blocked by antagonists of 5-HT2C receptors, phospholipase C (PLC), and calcium-activated potassium (BK) channels. Furthermore, Aβ1-42 oligomers failed to increase K+-channel currents in the presence of a BK channel blocker. Finally, 5-HT2C receptor antagonist improved olfactory memory and odor discrimination in APP/PS1 mice. The above data indicate that Aβ disrupts olfactory information output from the PC via the 5-HT-5-HT2C receptor-PLC-BK channel pathway. This study reveals that serotonergic modulation is a potential novel therapeutic target for olfactory damage in AD.
    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
    在兴奋细胞强烈极化的膜中,通过弱去极化刺激激活T型Ca2通道(TTCC)允许Ca2流入,进一步放大膜去极化,因此,“招募”更高的阈值电压门控通道,以促进动作电位激发。尽管如此,TTCC在可兴奋和不可兴奋细胞的质膜中执行其他功能,其中它们调节与细胞周期和细胞命运相关的许多生化途径。此外,在过去20年中获得的数据表明,TTCC参与了肿瘤生物学,将它们指定为有希望的化疗靶标。然而,它们在癌细胞稳定去极化的膜中的活性,其中大多数电压门控通道处于失活(非导通)状态,是反直觉的。在这里,我们讨论了在癌细胞中,弱的超极化刺激会增加开放TTCC的比例,与Ca2+依赖的K+通道相关,可能会严重增强膜超极化和通过不同电压无关的Ca2通道进入Ca2的驱动力。现有证据还表明,TTCC参与具有信令效应器的正反馈电路,尽管它们的可用性较低,但仍可能保证促增殖和促生存途径的开关样激活。在非兴奋膜的背景下解开TTCC的操作方式可能有助于新型抗癌方法的开发。
    In the strongly polarized membranes of excitable cells, activation of T-type Ca2+ channels (TTCCs) by weak depolarizing stimuli allows the influx of Ca2+ which further amplifies membrane depolarization, thus \"recruiting\" higher threshold voltage-gated channels to promote action potential firing. Nonetheless, TTCCs perform other functions in the plasma membrane of both excitable and non-excitable cells, in which they regulate a number of biochemical pathways relevant for cell cycle and cell fate. Furthermore, data obtained in the last 20 years have shown the involvement of TTCCs in tumor biology, designating them as promising chemotherapeutic targets. However, their activity in the steadily-depolarized membranes of cancer cells, in which most voltage-gated channels are in the inactivated (nonconducting) state, is counter-intuitive. Here we discuss that in cancer cells weak hyperpolarizing stimuli increase the fraction of open TTCCs which, in association with Ca2+-dependent K+ channels, may critically boost membrane hyperpolarization and driving force for Ca2+ entry through different voltage-independent Ca2+ channels. Available evidence also shows that TTCCs participate in positive feedback circuits with signaling effectors, which may warrant a switch-like activation of pro-proliferative and pro-survival pathways in spite of their low availability. Unravelling TTCC modus operandi in the context of non-excitable membranes may facilitate the development of novel anticancer approaches.
    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
    在阻力动脉中,内皮依赖性超极化(EDH)介导的血管舒张在糖尿病中被抑制。我们假设KCa通道衍生的EDH的下调减少了糖尿病中运动引起的血管舒张和血流再分布。为了检验这个假设,我们评估了后肢肌肉收缩后的血管功能,以及KCa通道在麻醉ZFDM中的贡献,2型糖尿病代谢病大鼠。我们还测试了运动训练是否改善了血管反应。使用体内微血管造影,在节律性肌肉收缩之前和之后观察后肢脉管系统(每3秒0.5s破伤风,20倍)由坐骨神经刺激(40Hz)引起。通过超声流量计同时测量后肢的股骨血流量。在存在和不存在阿帕明和炭毒素的情况下研究了KCa通道的贡献。我们发现,与瘦对照fa/大鼠相比,ZFDMfa/fa大鼠的小动脉段水平对肌肉收缩的血管和血流反应显着受损。fa/fa中KCa通道的贡献也小于fa/大鼠。fa/fa大鼠进行12周的低强度运动训练显示,血管和血流对肌肉收缩的反应发生了微小变化。然而,运动训练中对肌肉收缩的反应中KCa衍生成分比久坐的fa/fa大鼠大得多。这些数据表明,在这种代谢疾病大鼠模型中,运动训练增加了KCa通道在内皮依赖性血管舒张机制中的贡献,以维持血管和血流对肌肉收缩的反应。要点:2型糖尿病患者的微血管功能障碍会损害运动过程中的血流再分布,限制骨骼肌的功能,并可能导致早期疲劳。内皮依赖性超极化(EDH),介导阻力动脉的血管舒张,已知在患有糖尿病的动物中抑郁。这里,我们报道,与久坐的大鼠相比,ZFDM大鼠的低强度运动训练增加了血管扩张剂对肌肉收缩的反应中KCa通道衍生成分,部分是由于KCNN3表达的增加。这些结果表明,低强度运动训练可通过EDH的上调改善糖尿病代谢性疾病中骨骼肌收缩过程中的血流再分布。
    In resistance arteries, endothelium-dependent hyperpolarization (EDH)-mediated vasodilatation is depressed in diabetes. We hypothesized that downregulation of KCa channel derived EDH reduces exercise-induced vasodilatation and blood flow redistribution in diabetes. To test this hypothesis, we evaluated vascular function in response to hindlimb muscle contraction, and the contribution of KCa channels in anaesthetised ZFDM, metabolic disease rats with type 2 diabetes. We also tested whether exercise training ameliorated the vascular response. Using in vivo microangiography, the hindlimb vasculature was visualized before and after rhythmic muscle contraction (0.5 s tetanus every 3 s, 20 times) evoked by sciatic nerve stimulation (40 Hz). Femoral blood flow of the contracting hindlimb was simultaneously measured by an ultrasonic flowmeter. The contribution of KCa channels was investigated in the presence and absence of apamin and charybdotoxin. We found that vascular and blood flow responses to muscle contraction were significantly impaired at the level of small artery segments in ZFDM fa/fa rats compared to its lean control fa/+ rats. The contribution of KCa channels was also smaller in fa/fa than in fa/+ rats. Low-intensity exercise training for 12 weeks in fa/fa rats demonstrated minor changes in the vascular and blood flow response to muscle contraction. However, the KCa-derived component in the response to muscle contraction was much greater in exercise trained than in sedentary fa/fa rats. These data suggest that exercise training increases the contribution of KCa channels among endothelium-dependent vasodilatory mechanisms to maintain vascular and blood flow responses to muscle contraction in this metabolic disease rat model. KEY POINTS: Microvascular dysfunction in type 2 diabetes impairs blood flow redistribution during exercise and limits the performance of skeletal muscle and may cause early fatigability. Endothelium-dependent hyperpolarization (EDH), which mediates vasodilatation in resistance arteries, is known to be depressed in animals with diabetes. Here, we report that low-intensity exercise training in ZFDM rats increased the KCa channel-derived component in the vasodilator responses to muscle contraction compared to that in sedentary rats, partly as a result of the increase in KCNN3 expression. These results suggest that low-intensity exercise training improves blood flow redistribution in contracting skeletal muscle in metabolic disease with diabetes via upregulation of EDH.
    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
    Acetylsalicylic acid (aspirin) exhibits a broad range of activities, including analgesic, antipyretic, and antiplatelet properties. Recent clinical studies also recommend aspirin prophylaxis in women with a high risk of pre-eclampsia, a major complication of pregnancy characterized by hypertension. We investigated the effect of aspirin on mesenteric resistance arteries and found outdiscovered the molecular mechanism underlying this action. Aspirin (10-12-10-6 M) was tested on pregnant rat mesenteric resistance arteries by a pressurized arteriography. Aspirin was investigated in the presence of several inhibitors of: (a) nitric oxide synthase (L-NAME 2 × 10-4 M); (b) cyclooxygenase (Indomethacin, 10-5 M); (c) Ca2+-activated K+ channels (Kca): small conductance (SKca, Apamin, 10-7 M), intermediate conductance (IKca, TRAM34, 10-5 M), and big conductance (BKca, paxilline, 10-5 M); and (d) endothelial-derived hyperpolarizing factor (high KCl, 80 mM). Aspirin caused a concentration-dependent vasodilation. Aspirin-vasodilation was abolished by removal of endothelium or by high KCl. Furthermore, preincubation with either apamin plus TRAM-34 or paxillin significantly attenuated aspirin vasodilation (p < 0.05). For the first time, we showed that aspirin induced endothelium-dependent vasodilation in mesenteric resistance arteries through the endothelial-derived hyperpolarizing factor (EDHF) and calcium-activated potassium channels. By activating this molecular mechanism, aspirin may lower peripheral vascular resistance and be beneficial in pregnancies complicated by hypertension.
    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
    孕烯醇酮是一种调节神经胶质生长和分化的神经类固醇,神经元放电,和几个大脑功能,这些影响归因于孕烯醇酮对神经元和神经胶质细胞本身的作用。尽管脑循环对脑功能的重要作用,而且孕烯醇酮是一种血管活性剂,孕烯醇酮对脑动脉的作用尚不清楚。这里,我们获得了麻醉的雄性和雌性C57BL/6J小鼠对孕烯醇酮在大脑中动脉(MCA)直径上的体内浓度反应曲线。在雄性和雌性动物中,孕烯醇酮(1nM-100μM)以浓度依赖性方式收缩MCA,其最大效果达到~22-35%的直径减少。孕烯醇酮的作用在完整和去内皮化的情况下被复制,体外加压的MCA节段与孕烯醇酮在完整和去内皮化的MCA中引起类似的收缩。1μM帕索林消除了神经类固醇作用,大电导(BK)的Ca2-和电压门控K通道的选择性阻断剂。细胞附着,在从小鼠MCAs中新鲜分离的平滑肌细胞上的膜片钳记录表明,在体外和体内(10μM)收缩MCAs的浓度下,孕烯醇酮降低BK活性(NPo),NPo的平均跌幅达到24.2%。脑动脉的孕烯醇酮收缩的浓度依赖性和完整细胞中BK活性的抑制与无细胞中获得的数据平行,由内而外的补丁,在10μM孕烯醇酮时达到最大抑制。MCA平滑肌BKs包括通道形成α(slo1蛋白)和调节β1亚基,分别由KCNMA1和KCNMB1编码。然而,在KCNMB1-/-小鼠的MCA心肌细胞中,孕烯醇酮驱动的NPo降低仍然很明显。将slo1通道重建为人造通道后,二元磷脂双层,10μM孕烯醇酮引起slo1NPo抑制,与天然膜中看到的相似。最后,孕烯醇酮未能抑制KCNMA1-/-小鼠的MCA。总之,孕烯醇酮独立于神经元收缩MCA,胶质,内皮和循环因子,以及细胞完整性,细胞器,复杂的膜细胞结构,和胞质信号的持续存在。相反,这种作用涉及直接抑制SMBK通道,它不需要β1亚基,但通过通道形成α亚基对神经类固醇的直接感知来介导。
    Pregnenolone is a neurosteroid that modulates glial growth and differentiation, neuronal firing, and several brain functions, these effects being attributed to pregnenolone actions on the neurons and glial cells themselves. Despite the vital role of the cerebral circulation for brain function and the fact that pregnenolone is a vasoactive agent, pregnenolone action on brain arteries remain unknown. Here, we obtained in vivo concentration response curves to pregnenolone on middle cerebral artery (MCA) diameter in anesthetized male and female C57BL/6J mice. In both male and female animals, pregnenolone (1 nM-100 μM) constricted MCA in a concentration-dependent manner, its maximal effect reaching ~22-35% decrease in diameter. Pregnenolone action was replicated in intact and de-endothelialized, in vitro pressurized MCA segments with pregnenolone evoking similar constriction in intact and de-endothelialized MCA. Neurosteroid action was abolished by 1 μM paxilline, a selective blocker of Ca2+ - and voltage-gated K+ channels of large conductance (BK). Cell-attached, patch-clamp recordings on freshly isolated smooth muscle cells from mouse MCAs demonstrated that pregnenolone at concentrations that constricted MCAs in vitro and in vivo (10 μM), reduced BK activity (NPo), with an average decrease in NPo reaching 24.2%. The concentration-dependence of pregnenolone constriction of brain arteries and inhibition of BK activity in intact cells were paralleled by data obtained in cell-free, inside-out patches, with maximal inhibition reached at 10 μM pregnenolone. MCA smooth muscle BKs include channel-forming α (slo1 proteins) and regulatory β1 subunits, encoded by KCNMA1 and KCNMB1, respectively. However, pregnenolone-driven decrease in NPo was still evident in MCA myocytes from KCNMB1-/- mice. Following reconstitution of slo1 channels into artificial, binary phospholipid bilayers, 10 μM pregnenolone evoked slo1 NPo inhibition which was similar to that seen in native membranes. Lastly, pregnenolone failed to constrict MCA from KCNMA1-/- mice. In conclusion, pregnenolone constricts MCA independently of neuronal, glial, endothelial and circulating factors, as well as of cell integrity, organelles, complex membrane cytoarchitecture, and the continuous presence of cytosolic signals. Rather, this action involves direct inhibition of SM BK channels, which does not require β1 subunits but is mediated through direct sensing of the neurosteroid by the channel-forming α subunit.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Ca2+-activated potassium (KCa) channels of small and intermediate conductance influence proliferation, apoptosis, and cell metabolism. We analysed whether prolonged activation of KCa channels by zoxazolamine (ZOX) induces differentiation of mouse embryonic stem (ES) cells towards cardiomyocytes. ZOX treatment of ES cells dose-dependent increased the number and diameter of cardiac foci, the frequency of contractions as well as mRNA expression of the cardiac transcription factor Nkx-2.5, the cardiac markers cardiac troponin I (cTnI), α-myosin heavy chain (α-MHC), ventricular myosin light chain-2 (MLC2v), and the pacemaker hyperpolarization-activated, cyclic nucleotide-gated 4 channel (HCN4). ZOX induced hyperpolarization of membrane potential due to activation of IKCa, raised intracellular Ca2+ concentration ([Ca2+]i) and nitric oxide (NO) in a Ca2+-dependent manner. The Ca2+ response to ZOX was inhibited by chelation of Ca2+ with BAPTA-AM, release of Ca2+ from intracellular stores by thapsigargin and the phospholipase C (PLC) antagonist U73,122. Moreover, the ZOX-induced Ca2+ response was blunted by the purinergic receptor antagonist pyridoxalphosphate-6-azophenyl-2\',4\'-disulfonic acid (PPADS) as well as the specific P2Y1 antagonist MRS 2,179, suggesting purinergic receptor-stimulated signal transduction. Consequently, ZOX initiated ATP release from differentiating ES cells, which was inhibited by the chloride channel inhibitor NPPB and the gap junction inhibitor carbenoxolone (CBX). The stimulation of cardiomyogenesis by ZOX was blunted by the nitric oxide synthase (NOS) inhibitor l-NAME, as well as CBX and NPPB. In summary, our data suggest that ZOX enhances cardiomyogenesis of ES cells by ATP release presumably through gap junctional hemichannels, purinergic receptor activation and intracellular Ca2+ response, thus promoting NO generation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Arginine vasopressin (AVP) and oxytocin (OT) are nonapeptides that bind to G-protein coupled receptors and influence social behaviors. Consensus mammalian AVP and OT (Leu8-OT) sequences are highly conserved. In marmosets, an amino acid change in the 8th position of the peptide (Pro8-OT) exhibits unique structural and functional properties. There is ∼85 % structural homology between the OT receptor (OTR) and vasopressin 1a receptor (V1aR) resulting in significant cross-reactivity between the ligands and receptors. Chinese hamster ovary (CHO) cells expressing marmoset (mV1aR), macaque (qV1aR), or human vasopressin receptor 1a (hV1aR) were used to assess AVP, Leu8-OT and Pro8-OT pharmacological profiles. To assess activation of Gq, functional assays were performed using Fluo-3 to measure ligand-induced Ca2+ mobilization. In all three V1aR-expressing cell lines, AVP was more potent than the OT ligands. To assess ligand-induced hyperpolarization, FLIPR Membrane Potential (FMP) assays were performed. In all three V1aR lines, AVP was more potent than the OT analogs. The distinctive U-shaped concentration-response curve displayed by AVP may reflect enhanced desensitization of the mV1aR and hV1aR, which is not observed with qV1aR. Evaluation of Ca2+-activated potassium (K+) channels using the inhibitors apamin, paxilline, and TRAM-34 demonstrated that both intermediate and large conductance Ca2+-activated K+ channels contributed to membrane hyperpolarization, with different pharmacological profiles identified for distinct ligand-receptor combinations. Taken together, these data suggest differences in ligand-receptor signaling that may underlie differences in social behavior. Integrative studies of behavior, genetics and ligand-receptor interaction will help elucidate the connection between receptor pharmacology and social behaviors.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号