VGSCs

VGSC
  • 文章类型: Journal Article
    疼痛,影响全球数百万人的复杂和令人衰弱的状况,是一个重要的问题,尤其是在术后恢复的背景下。这篇全面的综述探讨了疼痛的复杂性及其全球影响,强调电压门控钠通道(VGSC或NaV通道)的调节是疼痛管理的有希望的途径,目的是减少对阿片类药物的依赖。本文深入研究了特定NaV同工型的作用,特别是疼痛过程中的NaV1.7,NaV1.8和NaV1.9,并讨论了钠通道阻滞剂的发展以精确地靶向这些同工型。传统局部麻醉药和选择性NaV亚型抑制剂,尽管在疼痛管理中表现出不同的功效,在系统分布和潜在副作用方面面临挑战。该综述强调了纳米医学在改善局部麻醉药递送方面的潜力,毒素和选择性NaV亚型抑制剂,用于在疼痛部位的靶向和持续释放。这一创新战略旨在提高药物的生物利用度,尽量减少全身暴露,优化治疗结果,为从外科手术中恢复或患有慢性疼痛的患者提供安全的疼痛管理和提高生活质量的重要希望。
    Pain, a complex and debilitating condition affecting millions globally, is a significant concern, especially in the context of post-operative recovery. This comprehensive review explores the complexity of pain and its global impact, emphasizing the modulation of voltage-gated sodium channels (VGSC or NaV channels) as a promising avenue for pain management with the aim of reducing reliance on opioids. The article delves into the role of specific NaV isoforms, particularly NaV 1.7, NaV 1.8, and NaV 1.9, in pain process and discusses the development of sodium channel blockers to target these isoforms precisely. Traditional local anesthetics and selective NaV isoform inhibitors, despite showing varying efficacy in pain management, face challenges in systemic distribution and potential side effects. The review highlights the potential of nanomedicine in improving the delivery of local anesthetics, toxins and selective NaV isoform inhibitors for a targeted and sustained release at the site of pain. This innovative strategy seeks to improve drug bioavailability, minimize systemic exposure, and optimize therapeutic outcomes, holding significant promise for secure pain management and enhancing the quality of life for individuals recovering from surgical procedures or suffering from chronic 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
    当其他疼痛管理方法失败并证明无效时,阿片类药物可用于治疗化疗引起的神经性疼痛(CINP)。然而,他们在CINP中的使用通常被认为是二线或辅助治疗,因为他们的中心副作用和长期使用的耐受性发展。与传统的基于CNS的方法相比,靶向外周部位可能会提供一些优势,因为外周靶点会在其源处调节疼痛信号。从而以更高的特异性缓解疼痛,疗效和尽量减少与非现场中枢神经系统活动相关的不良反应。因此,本研究旨在研究洛哌丁胺的作用,一种外周作用的μ阿片受体激动剂,对紫杉醇诱导的大鼠神经病理性疼痛的影响及其机制。洛哌丁胺治疗明显减弱机械,和冷超敏反应,并在神经病大鼠中产生明显的位置偏爱行为,表明其具有治疗诱发和自发性疼痛的潜力。更重要的是,在幼稚大鼠中,洛哌丁胺治疗没有产生对药物配对室的位置偏好,这表明其非成瘾性镇痛潜力。Further,分子研究显示,离子通道如TRPA1,TRPM8的表达增加;电压门控钠通道(VGSCs)和神经炎症标志物在背根神经节(DRG)和腰(L4-L5)脊髓的神经病变大鼠,洛哌丁胺治疗后显著下调。这些发现共同表明,外周μ阿片受体的激活通过下调TRP通道和VGSC以及抑制氧化-亚硝基应激和神经炎性级联反应,有助于改善神经病大鼠的诱发和自发性疼痛。
    Opioids are employed in the management of chemotherapy-induced neuropathic pain (CINP) when other pain management approaches have failed and proven ineffective. However, their use in CINP is generally considered as a second-line or adjunctive therapy owing to their central side effects and development of tolerance with their long-term usage. Targeting peripheral sites may offer several advantages over the conventional CNS-based approaches as peripheral targets modulate pain signals at their source, thereby relieving pain with higher specificity, efficacy and minimizing adverse effects associated with off-site CNS actions. Therefore, present study was designed with an aim to investigate the effect of loperamide, a peripherally acting mu-opioid receptor agonist, on paclitaxel-induced neuropathic pain in rats and elucidate its underlying mechanism. Loperamide treatment significantly attenuated mechanical, and cold hypersensitivity and produced significant place preference behaviour in neuropathic rats indicating its potential to treat both evoked and spontaneous pain. More importantly, loperamide treatment in naïve rats did not produce place preference to drug-paired chamber pointing towards its non-addictive analgesic potential. Further, molecular investigations revealed increased expression of ion channels such as TRPA1, TRPM8; voltage-gated sodium channels (VGSCs) and neuroinflammatory markers in the dorsal root ganglion (DRG) and lumbar (L4-L5) spinal cord of neuropathic rats, which was significantly downregulated upon loperamide treatment. These findings collectively suggest that activation of peripheral mu-opioid receptors contributes to the amelioration of both evoked and spontaneous pain in neuropathic rats by downregulating TRP channels and VGSCs along with suppression of oxido-nitrosative stress and neuroinflammatory cascade.
    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:雷公藤。f.由于其良好的抗炎和镇痛活性,已广泛应用于临床。然而,其应用受到潜在的毒性和副作用的限制。
    目的:该研究旨在确定雷公藤主要单体的药理活性和心脏毒性的机制。
    方法:数据库分析预测离子通道可能是雷公藤的潜在靶标。单体(雷公藤甲素,celastrol,去甲基zeylasal,和wilforgine)对Nav1.5和Nav1.7蛋白的预测和检测。然后,我们使用福尔马林诱导的疼痛模型,并评估心率和心肌酶谱,以研究体内各单体的镇痛活性和心脏毒性。
    结果:所有四种单体都能够以不同的结合能与Nav1.7和Nav1.5结合,并随后抑制Nav1.7和Nav1.5的峰值电流。这些单体都对福尔马林引起的疼痛表现出镇痛作用;因此,我们假设Nav1.7是重要的镇痛靶点之一.去甲基肾小球降低心率,增加肌酸激酶-MB的水平,因此提示潜在的心脏风险;数据提示对Nav1.5的抑制作用可能是其心脏毒性的重要因素。
    结论:我们的发现为进一步筛选活性水平较高,毒性水平较低的活性单体提供了重要的理论依据。
    BACKGROUND: Tripterygium wilfordii Hook. f. has been widely used in clinical practice due to its good anti-inflammatory and analgesic activities. However, its application is limited by potential toxicity and side effects.
    OBJECTIVE: The study aimed to identify the mechanisms responsible for the pharmacological activity and cardiotoxicity of the main monomers of Tripterygium wilfordii.
    METHODS: Database analysis predicted that ion channels may be potential targets of Tripterygium wilfordii. The regulatory effects of monomers (triptolide, celastrol, demethylzeylasteral, and wilforgine) on protein Nav1.5 and Nav1.7 were predicted and detected by Autodock and patch clamping. Then, we used the formalin-induced pain model and evaluated heart rate and myocardial zymograms to investigate the analgesic activity and cardiotoxicity of each monomer in vivo.
    RESULTS: All four monomers were able to bind to Nav1.7 and Nav1.5 with different binding energies and subsequently inhibited the peak currents of both Nav1.7 and Nav1.5. The monomers all exhibited analgesic effects on formalin-induced pain; therefore, we hypothesized that Nav1.7 is one of the key analgesic targets. Demethylzeylasteral reduced heart rate and increased the level of creatine kinase-MB, thus suggesting a potential cardiac risk; data suggested that the inhibitory effect on Nav1.5 might be an important factor underlying its cardiotoxicity.
    CONCLUSIONS: Our findings provide an important theoretical basis for the further screening of active monomers with higher levels of activity and lower levels of toxicity.
    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
    电压门控钠通道(VGSC)是许多疗法的靶标。膜电位的变化发生在整个细胞周期,然而,很少关注VGSC和Na+的作用,K+-ATP酶。我们假设除了DNA和细胞膜加倍以预期细胞分裂之外,VGSC和Na+应该加倍,K+-ATP酶与非分裂细胞比较。我们通过关联VGSCs或Na+的免疫细胞荧光标记,在8个永生化细胞系中测试了这一假设。K+-ATP酶与碘化丙啶或DAPI荧光使用流式细胞术和成像。在S至M期期间VGSC的细胞表面表达是在G0-G1期期间看到的两倍。相比之下,Na+,K+-ATP酶表达仅增加1.5倍。增加与通道或泵的基线表达无关。VGSC和Na+的变化,K+-ATP酶的表达对我们了解钠在控制细胞周期中的作用以及针对Na处理系统的这些组件的处理的可变性都有影响。
    Voltage-gated sodium channels (VGSCs) are the target for many therapies. Variation in membrane potential occurs throughout the cell cycle, yet little attention has been devoted to the role of VGSCs and Na+,K+-ATPases. We hypothesized that in addition to doubling DNA and cell membrane in anticipation of cell division, there should be a doubling of VGSCs and Na+,K+-ATPase compared to non-dividing cells. We tested this hypothesis in eight immortalized cell lines by correlating immunocytofluorescent labeling of VGSCs or Na+,K+-ATPase with propidium iodide or DAPI fluorescence using flow cytometry and imaging. Cell surface expression of VGSCs during phases S through M was double that seen during phases G0-G1. By contrast, Na+,K+-ATPase expression increased only 1.5-fold. The increases were independent of baseline expression of channels or pumps. The variation in VGSC and Na+,K+-ATPase expression has implications for both our understanding of sodium\'s role in controlling the cell cycle and variability of treatments targeted at these components of the Na+ handling system.
    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
    延胡索王伟,一种传统的中草药,已被用作镇痛药数千年,它也促进血液循环。在这项研究中,从中药数据库和分析平台(TCMSP)获得了33延胡索生物碱活性成分。共有543个疼痛相关目标,1774个心律失常目标,使用瑞士目标预测获得了这些活性成分的642个潜在目标,GeneCards,开放目标平台,和治疗目标数据库。通过维恩图可视化了50个相交目标,KEGG和GO途径富集分析。分析认为钠离子通道可能是延胡索活性成分作为镇痛和抗心律失常药物的潜在靶点。分子对接表明,33个组分可以以不同的结合能与Nav1.7和Nav1.5(离子通道蛋白的两种亚型)结合。在膜片钳研究中,二氢血根碱和二氢白屈菜红碱,两种单体具有最强的结合效应,可以抑制峰值电流并促进Nav1.5的激活和失活阶段。同时,二氢血根碱和二氢白屈菜红碱也可以抑制峰值电流并促进Nav1.7的活化阶段。因此,这项研究的结果为将来使用中药治疗疼痛和心血管疾病提供了有价值的信息。
    Corydalis yanhusuo W. T. Wang, a traditional Chinese herbal medicine, has been used as an analgesic for thousands of years and it also promotes blood circulation. In this study, 33 Corydalis yanhusuo alkaloid active components were acquired from Traditional Chinese Medicine Database and Analysis Platform (TCMSP). A total of 543 pain-related targets, 1774 arrhythmia targets, and 642 potential targets of these active components were obtained using Swiss Target Prediction, GeneCards, Open Target Platform, and Therapeutic Target Database. Fifty intersecting targets were visualized through a Venn diagram, KEGG and GO pathway enrichment analysis. The analysis proposed that sodium ion channels are likely potential targets of Corydalis yanhusuo active components as analgesia and anti-arrhythmia agents. Molecular docking showed that the 33 components could bind to Nav1.7 and Nav1.5 (two subtypes of ion channel proteins) with different binding energies. In a patch clamp study, dihydrosanguinarine and dihydrochelerythrine, two monomers with the strongest binding effects, could inhibit the peak currents and promote both activation and inactivation phases of Nav1.5. Meanwhile, dihydrosanguinarine and dihydrochelerythrine could also inhibit peak currents and promote the activation phase of Nav1.7. Therefore, the findings from this study provide valuable information for future uses of traditional Chinese medicines to treat pain and cardiovascular disease.
    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
    三叉神经痛(TN)是临床上常见的一种外周神经痛,通常很难治愈。常见的镇痛药物很难达到理想的镇痛效果。Syb-prII-1是从ButhusmartensiKarsch(BmK)的蝎毒中分离出的β型蝎子神经毒素。它对电压门控钠通道(VGSCs)有重要影响,特别是与Nav1.8和Nav1.9密切相关。探讨Syb-prII-1对TN是否有良好的镇痛作用,我们建立了SpragueDawley(SD)大鼠眶下神经慢性压迫性损伤(IoN-CCI)模型。行为,电生理学,蛋白质印迹,等方法对模型进行了验证。发现Syb-prII-1可以显着缓解IoN-CCI大鼠的疼痛行为。给予Syb-prII-1后,在IoN-CCI损伤后,丝裂原活化蛋白激酶(MAPKs)途径的磷酸化水平呈剂量依赖性下降.此外,Syb-prII-1(4.0mg/kg)可以显著改变Nav1.8的稳态激活和失活曲线。Nav1.9的稳态激活和失活曲线与Nav1.8相似,但无显著差异。据推测,它可能起到辅助作用。绑定模式,临界残基,用计算模拟方法阐明了Syb-prII-1和VSD2rNav1.8的特定相互作用类型。我们的结果表明,Syb-prII-1可以通过作用于Nav1.8靶标为TN提供潜在的治疗。
    Trigeminal neuralgia (TN) is a common type of peripheral neuralgia in clinical practice, which is usually difficult to cure. Common analgesic drugs are difficult for achieving the desired analgesic effect. Syb-prII-1 is a β-type scorpion neurotoxin isolated from the scorpion venom of Buthus martensi Karsch (BmK). It has an important influence on the voltage-gated sodium channel (VGSCs), especially closely related to Nav1.8 and Nav1.9. To explore whether Syb-prII-1 has a good analgesic effect on TN, we established the Sprague Dawley (SD) rats\' chronic constriction injury of the infraorbital nerve (IoN-CCI) model. Behavioral, electrophysiological, Western blot, and other methods were used to verify the model. It was found that Syb-prII-1 could significantly relieve the pain behavior of IoN-CCI rats. After Syb-prII-1 was given, the phosphorylation level of the mitogen-activated protein kinases (MAPKs) pathway showed a dose-dependent decrease after IoN-CCI injury. Moreover, Syb-prII-1(4.0 mg/kg) could significantly change the steady-state activation and inactivation curves of Nav1.8. The steady-state activation and inactivation curves of Nav1.9 were similar to those of Nav1.8, but there was no significant difference. It was speculated that it might play an auxiliary role. The binding mode, critical residues, and specific interaction type of Syb-prII-1 and VSD2rNav1.8 were clarified with computational simulation methods. Our results indicated that Syb-prII-1 could provide a potential treatment for TN by acting on the Nav1.8 target.
    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
    Voltage-gated sodium channels (VGSCs) are considered to be one of the most important ion channels given their remarkable physiological role. VGSCs constitute a family of large transmembrane proteins that allow transmission, generation, and propagation of action potentials. This occurs by conducting Na+ ions through the membrane, supporting cell excitability and communication signals in various systems. As a result, a wide range of coordination and physiological functions, from locomotion to cognition, can be accomplished. Drugs that target and alter the molecular mechanism of VGSCs\' function have highly contributed to the discovery and perception of the function and the structure of this channel. Among those drugs are various marine toxins produced by harmful microorganisms or venomous animals. These toxins have played a key role in understanding the mode of action of VGSCs and in mapping their various allosteric binding sites. Furthermore, marine toxins appear to be an emerging source of therapeutic tools that can relieve pain or treat VGSC-related human channelopathies. Several studies documented the effect of marine toxins on VGSCs as well as their pharmaceutical applications, but none of them underlined the principal marine toxins and their effect on VGSCs. Therefore, this review aims to highlight the neurotoxins produced by marine animals such as pufferfish, shellfish, sea anemone, and cone snail that are active on VGSCs and discuss their pharmaceutical values.
    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
    Visceral pain is one of the most common symptoms associated with functional gastrointestinal (GI) disorders. Although the origin of these symptoms has not been clearly defined, the implication of both the central and peripheral nervous systems in visceral hypersensitivity is well established. The role of several pathways in visceral nociception has been explored, as well as the influence of specific receptors on afferent neurons, such as voltage-gated sodium channels (VGSCs). VGSCs initiate action potentials and dysfunction of these channels has recently been associated with painful GI conditions. Current treatments for visceral pain generally involve opioid based drugs, which are associated with important side-effects and a loss of effectiveness or tolerance. Hence, efforts have been intensified to find new, more effective and longer-lasting therapies. The implication of VGSCs in visceral hypersensitivity has drawn attention to tetrodotoxin (TTX), a relatively selective sodium channel blocker, as a possible and promising molecule to treat visceral pain and related diseases. As such, here we will review the latest information regarding this toxin that is relevant to the treatment of visceral pain and the possible advantages that it may offer relative to other treatments, alone or in combination.
    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
    疼痛通常会导致生理和心理上的一系列异常变化,这可能导致疾病甚至死亡。药物治疗是疼痛缓解和管理的最基本和最常用的方法。有趣的是,目前,据报道,数百种中药被用于缓解疼痛,其中大多数是单体制剂,已经发展成为新的止痛药。延胡索是这些药物之一的代表,可用于缓解疼痛。
    目的:本研究旨在确定延胡索衍生单体的镇痛作用和潜在靶标,并探讨任何可能的相关心脏危险因素。
    方法:在本研究中,来自延胡索的四种单体(四氢巴马汀,Corydaline,普罗托品,脱氢紫藤碱)进行了体内测试,使用福尔马林诱导的疼痛模型来确定它们的镇痛特性。还使用全细胞膜片钳记录和心肌酶测定来确定它们的潜在靶标。
    结果:结果表明,所有单体均表现出镇痛活性,并抑制峰值电流,促进了Nav1.7的激活和失活阶段,这表明Nav1.7可能参与了延胡索的镇痛机制。Protopine增加肌酸激酶-MB(CK-MB)的水平并抑制峰值电流,促进了Nav1.5的激活和失活阶段,表明Nav1.5可能参与了与普托品治疗相关的心脏风险。
    结论:这些数据表明,延胡索乙素的镇痛效果最好,心脏风险最低。因此,电压门控钠通道(VGSCs)可能是延胡索的主要作用靶点。这项研究,因此,为未来研究和使用中药缓解疼痛提供了有价值的信息。
    UNASSIGNED: Pain often causes a series of abnormal changes in physiology and psychology, which can lead to disease and even death. Drug therapy is the most basic and commonly used method for pain relief and management. Interestingly, at present, hundreds of traditional Chinese medicines have been reported to be used for pain relief, most of which are monomer preparations, which have been developed into new painkillers. Corydalis yanhusuo is a representative of one of these medicines and is available for pain relief.
    OBJECTIVE: This study aims to determine the analgesic effect and the potential targets of the monomers derived from Corydalis yanhusuo, and to explore any possible associated cardiac risk factors.
    METHODS: In this study, four monomers derived from Corydalis yanhusuo (tetrahydropalmatine, corydaline, protopine, dehydrocorydaline) were tested in vivo, using the formalin-induced pain model to determine their analgesic properties. Their potential targets were also determined using whole cell patch clamp recordings and myocardial enzyme assays.
    RESULTS: The results showed that all monomers showed analgesic activity and inhibited the peak currents, promoted the activation and inactivation phases of Nav1.7, which indicating that Nav1.7 might be involved in the analgesic mechanism of Corydalis yanhusuo. Protopine increased the level of creatine kinase-MB (CK-MB) and inhibited the peak currents, promoted the activation and inactivation phases of Nav1.5, indicating that Nav1.5 might be involved in the cardiac risk associated with protopine treatment.
    CONCLUSIONS: These data showed that tetrahydropalmatine produced the best analgesic effect and the lowest cardiac risk. Thus, voltage gated sodium channels (VGSCs) might be the main targets associated with Corydalis yanhusuo. This study, therefore, provides valuable information for future studies and use of traditional Chines medicines for the alleviation of pain.
    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
    电压门控钠通道(VGSCs),在各种类型的癌症如乳腺癌中异常表达,前列腺癌,肺癌,还有宫颈癌,参与了侵袭和迁移的转移过程。Nav1.5是由SCN5A编码的VGSC的成孔α亚基。各种研究表明,Nav1.5通常作为其新生儿剪接形式,在转移性乳腺癌细胞中高表达。Nav1.5的异常激活和表达引发多种细胞机制,包括改变H+流出,促进上皮-间质转化(EMT)和半胱氨酸组织蛋白酶的表达,增强乳腺癌细胞的体内外转移和侵袭力。这里,我们系统回顾了有关Nav1.5在乳腺癌中的促转移作用及其潜在机制的最新可用数据.我们总结了影响乳腺癌细胞Nav1.5表达的因素,并讨论Nav1.5受体阻滞剂作为乳腺癌治疗候选药物的潜力。
    Voltage-gated sodium channels (VGSCs), which are abnormally expressed in various types of cancers such as breast cancer, prostate cancer, lung cancer, and cervical cancer, are involved in the metastatic process of invasion and migration. Nav1.5 is a pore-forming α subunit of VGSC encoded by SCN5A. Various studies have demonstrated that Nav1.5, often as its neonatal splice form, is highly expressed in metastatic breast cancer cells. Abnormal activation and expression of Nav1.5 trigger a variety of cellular mechanisms, including changing H+ efflux, promoting epithelial-to-mesenchymal transition (EMT) and the expression of cysteine cathepsin, to potentiate the metastasis and invasiveness of breast cancer cells in vitro and in vivo. Here, we systematically review the latest available data on the pro-metastatic effect of Nav1.5 and its underlying mechanisms in breast cancer. We summarize the factors affecting Nav1.5 expression in breast cancer cells, and discuss the potential of Nav1.5 blockers serving as candidates for breast cancer treatment.
    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