cannabinoid receptors

大麻素受体
  • 文章类型: Journal Article
    大麻素和阿片样物质受体活性可以通过多种翻译后机制来调节,包括相互作用复合物的形成。这项研究检查了内源性和外源性伴侣参与调节大麻素CB1受体(CB1R)的丰度和活性,δ阿片受体(DOR),和CB1R-DOR相互作用复合物。关注内源性蛋白伴侣,即受体转运蛋白(RTPs),我们检查了小鼠脊髓中的相对mRNA表达,发现与其他RTPs相比,RTP4的表达水平更高。接下来,我们通过操纵细胞系中的RTP4表达来评估RTP4对受体丰度的影响.RTP4的过表达导致增加,敲低导致CB1R水平降低,DOR,和CB1R-DOR相互作用复合物;这伴随着信号传导的平行变化。使用受体选择性拮抗剂检查了小分子亲脂性配体充当外源伴侣的能力。长期治疗导致受体丰度和活性增加,而mRNA没有变化,支持作为药理伴侣的作用。最后,大麻二酚(CBD)的作用,一种小分子配体和大麻的主要活性成分,对小鼠的受体丰度和活性进行了检查。我们发现CBD给药导致小鼠脊髓中受体丰度和活性增加。一起,这些结果突出了伴侣(蛋白质和小分子)在调节CB1R的水平和活性中的作用,DOR,和它们相互作用的复合物可能通过包括受体成熟和运输的机制。意义陈述本研究强调了伴侣(内源性和小膜可渗透分子)在调节CB1R水平中的作用,DOR,以及它们相互作用的复合体。这些伴侣可以被开发为涉及这些受体的病理的治疗剂。
    Cannabinoid and opioid receptor activities can be modulated by a variety of posttranslational mechanisms including the formation of interacting complexes. This study examines the involvement of endogenous and exogenous chaperones in modulating the abundance and activity of cannabinoid CB1 receptor (CB1R), delta opioid receptor (DOR), and CB1R-DOR interacting complexes. Focussing on endogenous protein chaperones namely receptor transporter proteins (RTPs), we examined relative mRNA expression in the mouse spinal cord and found RTP4 to be expressed at higher levels compared to other RTPs. Next, we assessed the effect of RTP4 on receptor abundance by manipulating RTP4 expression in cell lines. Overexpression of RTP4 causes an increase and knock-down causes a decrease in the levels of CB1R, DOR, and CB1R-DOR interacting complexes; this is accompanied by parallel changes in signaling. The ability of small molecule lipophilic ligands to function as exogenous chaperones was examined using receptor-selective antagonists. Long term treatment leads to increases in receptor abundance and activity with no changes in mRNA supporting a role as pharmacological chaperones. Finally, the effect of cannabidiol (CBD), a small molecule ligand and a major active component of Cannabis, on receptor abundance and activity in mice was examined. We find that CBD administration leads to increases in receptor abundance and activity in mouse spinal cord. Together, these results highlight a role for chaperones (proteins and small molecules) in modulating levels and activity of CB1R, DOR, and their interacting complexes potentially through mechanisms including receptor maturation and trafficking. Significance Statement This study highlights a role for chaperones (endogenous and small membrane-permeable molecules) in modulating levels of CB1R, DOR, and their interacting complexes. These chaperones could be developed as therapeutics for pathologies involving these receptors.
    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
    Gi偶联受体,特别是大麻素受体(CBRs),被认为是治疗脑部病变的透视目标,包括癫痫。然而,CBR激动剂抗惊厥作用的确切机制尚不清楚.我们已经发现,WIN55,212-2(CBR激动剂)抑制了双微胶或NH4Cl在大鼠海马神经元-神经胶质培养物的神经元中诱导的Ca2离子细胞内浓度的同步振荡(癫痫样活性)。正如我们所证明的,WIN55,212-2效应由CB1R受体介导。激动剂抑制电压门控钙通道介导的Ca2+流入,但不改变NMDA介导的流入,AMPA,和红藻氨酸受体。我们还发现磷脂酶C(PLC),蛋白激酶C(PKC),和G蛋白偶联的向内整流K通道(GIRK通道)参与了CB1R激活对癫痫样活性的抑制作用的分子机制。因此,我们的结果表明,CB1R激动剂的抗癫痫作用是由不同的细胞内信号级联介导的,包括非规范PLC/PKC相关途径。
    Gi-coupled receptors, particularly cannabinoid receptors (CBRs), are considered perspective targets for treating brain pathologies, including epilepsy. However, the precise mechanism of the anticonvulsant effect of the CBR agonists remains unknown. We have found that WIN 55,212-2 (a CBR agonist) suppresses the synchronous oscillations of the intracellular concentration of Ca2+ ions (epileptiform activity) induced in the neurons of rat hippocampal neuron-glial cultures by bicuculline or NH4Cl. As we have demonstrated, the WIN 55,212-2 effect is mediated by CB1R receptors. The agonist suppresses Ca2+ inflow mediated by the voltage-gated calcium channels but does not alter the inflow mediated by NMDA, AMPA, and kainate receptors. We have also found that phospholipase C (PLC), protein kinase C (PKC), and G-protein-coupled inwardly rectifying K+ channels (GIRK channels) are involved in the molecular mechanism underlying the inhibitory action of CB1R activation against epileptiform activity. Thus, our results demonstrate that the antiepileptic action of CB1R agonists is mediated by different intracellular signaling cascades, including non-canonical PLC/PKC-associated pathways.
    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
    靶向内源性大麻素(eCB)信号系统以缓解疼痛是一种重要的治疗选择,现在才开始进行机械探索。在这次审查中,我们专注于两种最近受到赞赏的基于大麻素的靶向策略,用大麻二酚(CBD)和含有6种(ABHD6)抑制剂的a/b-水解酶结构域治疗,它们具有令人兴奋的潜力,可以通过不同的作用机制(MOA)缓解疼痛,并且不会中毒。我们回顾了关于植物来源的大麻素疼痛的证据,重点是CBD及其在疼痛途径中表达的多个分子靶标。我们还讨论了eCB信号在调节疼痛反应中的功能,以及针对ABHD6(一种2-花生四酰基甘油(2-AG)水解酶)的抑制剂的治疗前景。最后,我们讨论了新型大麻素生物传感器,可以利用GRABeCB2.0以使得能够在电路特异性水平上发现由大麻素调节的靶标。重要性声明大麻已被人类用作有效的药物数千年,包括疼痛管理。最近的证据强调了调节内源性大麻素信号传导的化合物的治疗潜力。具体来说,大麻二酚和ABHD6酶抑制剂代表了通过不同的调节疼痛途径中内源性大麻素信号来缓解疼痛的有希望的策略,不醉人,行动机制。
    Targeting the endocannabinoid (eCB) signaling system for pain relief is an important treatment option that is only now beginning to be mechanistically explored. In this review, we focus on two recently appreciated cannabinoid-based targeting strategies, treatments with cannabidiol (CBD) and a/b-hydrolase domain containing 6 (ABHD6) inhibitors, which have the exciting potential to produce pain relief through distinct mechanisms of action (MOA) and without intoxication. We review evidence on plant-derived cannabinoids for pain, with an emphasis on CBD and its multiple molecular targets expressed in pain pathways. We also discuss the function of eCB signaling in regulating pain responses and the therapeutic promises of inhibitors targeting ABHD6, a 2-arachidonoylglycerol (2-AG) hydrolyzing enzyme. Finally, we discuss how the novel cannabinoid biosensor, GRABeCB2.0, may be leveraged to enable the discovery of targets modulated by cannabinoids at a circuit-specific level. Significance Statement Cannabis has been used by humans as an effective medicine for millennia, including for pain management. Recent evidence emphasizes the therapeutic potential of compounds that modulate endocannabinoid signaling. Specifically, cannabidiol and inhibitors of the enzyme ABHD6 represent promising strategies to achieve pain relief by modulating endocannabinoid signaling in pain pathways via distinct, non-intoxicating, mechanisms of action.
    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
    几个世纪以来,大麻及其产品一直用于医疗和娱乐目的。最近大麻的广泛合法化极大地扩大了其在美国的使用范围,除了青少年。同时,数十年的研究提高了我们对大麻药理学的认识,特别是与大麻成分相互作用的内源性大麻素系统。这项研究揭示了多种目标和方法来操纵用于治疗用途的系统并改善大麻毒性或大麻使用障碍。研究还引发了新的问题,强调了其广泛使用的潜在风险,特别是在大脑发育的关键窗口期或每日服用大剂量含有高含量D9四氢大麻酚(THC)的大麻时暴露的持久后果。在这里,我们重点介绍了当前对大麻的神经科学研究,这些研究揭示了滥用的治疗机会和潜在的不利后果,并指出了可以指导未来研究的知识空白。重要性声明大麻的使用随着其可用性的增加而升级。在这里,我们强调了大麻研究的挑战以及我们对大麻药理学及其目标内源性大麻素系统的知识的差距。需要解决这些差距的未来研究,以便可以利用内源性大麻素系统进行安全有效的使用。
    Cannabis and its products have been used for centuries for both medicinal and recreational purposes. The recent widespread legalization of cannabis has vastly expanded its use in the United States across all demographics except for adolescents. Meanwhile decades of research have advanced our knowledge of cannabis pharmacology and particularly of the endocannabinoid system with which the components of cannabis interact. This research has revealed multiple targets and approaches for manipulating the system for therapeutic use and to ameliorate cannabis toxicity or cannabis use disorder. Research has also led to new questions that underscore the potential risks of its widespread use, particularly the enduring consequences of exposure during critical windows of brain development or for consumption of large daily doses of cannabis with high content D9 tetrahydrocannabinol (THC). Here we highlight current neuroscience research on cannabis that has shed light on therapeutic opportunities and potential adverse consequences of misuse and point to gaps in knowledge that can guide future research. Significance Statement Cannabis use has escalated with its increased availability. Here we highlight the challenges of cannabis research and the gaps in our knowledge of cannabis pharmacology and of the endocannabinoid system that it targets. Future research that addresses these gaps is needed so that the endocannabinoid system can be leveraged for safe and effective use.
    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
    大麻,通常被认为是全球使用最广泛的非法精神活性物质,在几个国家和地区,其娱乐和医疗用途的法律地位发生了变化。这一变化揭示了将大麻消费与各种血管状况联系起来的新证据。具体来说,大麻的使用与动脉粥样硬化有关联,连同动脉炎等疾病,可逆性血管痉挛,和主动脉瘤或夹层的事件。最近的研究已经开始揭示大麻素化合物与动脉粥样硬化发展的连接机制。众所周知,大麻素的主要生物学作用通过激活1型和2型大麻素受体起作用。操纵内源性大麻素系统,无论是遗传还是药理学,正在成为解决与肥胖相关的代谢功能障碍的一种有前途的方法。此外,大量研究已经证明了大麻素的血管舒张特性和潜在的动脉粥样硬化保护作用.在临床前试验中,正在探索大麻二酚作为野百合碱诱导的肺动脉高压的治疗选择。尽管现有文献表明大麻素在动脉粥样硬化的发病机理中具有直接作用,大麻素与其他血管疾病之间的相关性仅在某些病例系列或观察性研究中报道,其作用和确切机制尚不清楚。因此,有必要总结和更新以前发表的研究。本文旨在总结大麻使用与血管疾病关系的最新临床和实验研究成果。它还试图阐明这些关联背后的潜在机制,在这个不断发展的研究领域提供当前知识的全面视图。
    Cannabis, often recognized as the most widely used illegal psychoactive substance globally, has seen a shift in its legal status in several countries and regions for both recreational and medicinal uses. This change has brought to light new evidence linking cannabis consumption to various vascular conditions. Specifically, there is an association between cannabis use and atherosclerosis, along with conditions such as arteritis, reversible vasospasm, and incidents of aortic aneurysm or dissection. Recent research has started to reveal the mechanisms connecting cannabinoid compounds to atherosclerosis development. It is well known that the primary biological roles of cannabinoids operate through the activation of cannabinoid receptor types 1 and 2. Manipulation of the endocannabinoid system, either genetically or pharmacologically, is emerging as a promising approach to address metabolic dysfunctions related to obesity. Additionally, numerous studies have demonstrated the vasorelaxant properties and potential atheroprotective benefits of cannabinoids. In preclinical trials, cannabidiol is being explored as a treatment option for monocrotaline-induced pulmonary arterial hypertension. Although existing literature suggests a direct role of cannabinoids in the pathogenesis of atherosclerosis, the correlation between cannabinoids and other vascular diseases was only reported in some case series or observational studies, and its role and precise mechanisms remain unclear. Therefore, it is necessary to summarize and update previously published studies. This review article aims to summarize the latest clinical and experimental research findings on the relationship between cannabis use and vascular diseases. It also seeks to shed light on the potential mechanisms underlying these associations, offering a comprehensive view of current knowledge in this evolving field of study.
    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
    镰状细胞病(SCD)患者经常经历慢性疼痛以及不可预测的急性疼痛发作,这极大地影响了他们的生活质量和预期寿命。目前SCD相关疼痛的治疗策略主要依赖于阿片类镇痛药,疗效有限,并引起严重的不良反应。大麻已经成为一种潜在的替代品,但其疗效仍不确定。在这项研究中,我们研究了Δ9-四氢大麻酚(THC)的抗伤害作用,大麻令人陶醉的成分,在雄性HbSS小鼠中,表达>99%的人类镰刀血红蛋白,和雄性HbAA小鼠,它表达正常的人类血红蛋白A,作为一个控制。急性THC给药(0.1-3mg-kg-1,腹膜内,i.p.)剂量依赖性地降低HbSS的机械和冷超敏反应,但不是HbAA小鼠。在甩尾试验中,THC(1和3mg-kg-1,腹膜内)在HbSS小鼠中产生了实质性的抗伤害作用。相比之下,THC(1mg-kg-1,i.p.)不会改变焦虑样行为(升高加迷宫)或长期记忆(24小时新颖物体识别)。亚慢性THC治疗(1和3mg-kg-1,i.p.)可持续缓解机械超敏反应,但导致HbSS小鼠对冷超敏反应的耐受性。一起,本研究发现THC是治疗SCD慢性疼痛的一种可能选择.需要进一步研究以阐明其作用机制以及与其他大麻成分的可能相互作用。意义声明该研究使用人源化小鼠模型探索了THC在缓解镰状细胞病(SCD)中的疼痛中的功效。研究结果表明,急性THC可降低SCD小鼠的机械和冷超敏反应,而不会影响情绪和认知功能障碍。亚慢性THC治疗可持续缓解机械性超敏反应,但会导致冷超敏反应耐受性。这些结果提供了对THC作为SCD替代疼痛管理选项的潜力的见解,强调它的优点和局限性。
    People with sickle cell disease (SCD) often experience chronic pain as well as unpredictable episodes of acute pain, which significantly affect their quality of life and life expectancy. Current treatment strategies for SCD-associated pain primarily rely on opioid analgesics, which have limited efficacy and cause serious adverse effects. Cannabis has emerged as a potential alternative, yet its efficacy remains uncertain. In this study, we investigated the antinociceptive effects of Δ9-tetrahydrocannabinol (THC), cannabis\' intoxicating constituent, in male HbSS mice, which express >99% human sickle hemoglobin, and male HbAA mice, which express normal human hemoglobin A, as a control. Acute THC administration (0.1-3 mg-kg-1, intraperitoneal, i.p.) dose-dependently reduced mechanical and cold hypersensitivity in HbSS, but not HbAA mice. In the tail-flick assay, THC (1 and 3 mg-kg-1, i.p.) produced substantial antinociceptive effects in HbSS mice. By contrast, THC (1 mg-kg-1, i.p.) did not alter anxiety-like behavior (elevated plus maze) or long-term memory (24-h novel object recognition). Subchronic THC treatment (1 and 3 mg-kg-1, i.p.) provided sustained relief of mechanical hypersensitivity but led to tolerance in cold hypersensitivity in HbSS mice. Together, the findings identify THC as a possible therapeutic option for the management of chronic pain in SCD. Further research is warranted to elucidate its mechanism of action and possible interaction with other cannabis constituents. Significance Statement The study explores THC\'s efficacy in alleviating pain in sickle cell disease (SCD) using a humanized mouse model. Findings indicate that acute THC administration reduces mechanical and cold hypersensitivity in SCD mice without impacting emotional and cognitive dysfunction. Subchronic THC treatment offers sustained relief of mechanical hypersensitivity but leads to cold hypersensitivity tolerance. These results offer insights into THC\'s potential as an alternative pain management option in SCD, highlighting both its benefits and limitations.
    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
    氧化应激,纤维化,来自AGE-RAGE相互作用的炎性小体激活有助于糖尿病心肌病(DCM)的形成和进展。我们的研究揭示了β-石竹烯(BCP)对激活CB2受体对抗糖尿病并发症的影响,并研究了小鼠中潜在的细胞信号传导途径。通过用链脲佐菌素注射液喂养高脂饮食来建立DCM的小鼠模型。糖尿病发展后,动物接受12周口服BCP治疗,剂量为50mg/kg/体重.BCP治疗显示糖耐量显著改善,胰岛素抵抗,和提高糖尿病动物的血清胰岛素水平。BCP治疗可有效逆转心脏重塑并恢复磷酸化肌钙蛋白I和SERCA2a的表达。超微结构检查显示用BCP处理的DCM小鼠的心肌细胞损伤减少。发现保存的肌细胞与DCM小鼠心脏中AGE/RAGE的表达降低有关。BCP处理通过抑制NOX4的表达和激活PI3K/AKT/Nrf2信号传导来减轻氧化应激。BCP通过抑制TGF-β/Smad信号抑制DCM小鼠的心肌纤维化和内皮-间质转化(EndMT)。Further,BCP处理抑制了DCM小鼠的NLRP3炎性体激活,并减轻了对胰腺组织的细胞损伤,胰岛素阳性细胞数量显着增加。为了证明BCP的CB2受体依赖性机制,另一组DCM小鼠用AM630,一种CB2受体拮抗剂AM630预处理,观察到AM630消除了BCP对DCM小鼠的有益作用.一起来看,BCP通过介导CB2受体依赖性机制显示出保护DCM小鼠心肌和胰腺的潜力。意义声明1.β-石竹烯(BCP),大麻素2型受体(CB2R)激动剂。2.BCP通过激活小鼠中的CB2R减轻糖尿病性心肌病3.BCP对CB2R的激活显示出对纤维化和炎性体激活的强保护作用4.它在小鼠中调节AGE/RAGE和PI3K/Nrf2/Akt信号传导。
    Oxidative stress, fibrosis, and inflammasome activation from AGE-RAGE interaction contribute to diabetic cardiomyopathy (DCM) formation and progression. Our study revealed the impact of β-caryophyllene (BCP) on activating CB2 receptors against diabetes complications and investigated the underlying cell signaling pathways in mice. The murine model of DCM was developed by feeding high-fat diet with streptozotocin injections. After the development of diabetes, the animals received a 12-week oral BCP treatment at a dosage of 50 mg/kg/body weight. BCP treatment showed significant improvement in glucose tolerance, insulin resistance, and enhanced serum insulin levels in diabetic animals. BCP treatment effectively reversed the heart remodeling and restored the phosphorylated troponin I and SERCA2a expression. Ultrastructural examination showed reduced myocardial cell injury in DCM mice treated with BCP. The preserved myocytes were found associated with reduced expression of AGE/RAGE in DCM mice hearts. BCP treatment mitigated oxidative stress by inhibiting expression of NOX4 and activating PI3K/AKT/Nrf2 signaling. BCP suppressed cardiac fibrosis and endothelial-to-mesenchymal transition (EndMT) in DCM mice by inhibiting TGF-β/Smad signaling. Further, BCP treatment suppressed NLRP3 inflammasome activation in DCM mice and alleviated cellular injury to the pancreatic tissues evidenced by significant elevation of the number of insulin-positive cells. To demonstrate CB2 receptor dependent mechanism of BCP, another group of DCM mice were pretreated with AM630, a CB2 receptor antagonist AM630 and AM630 was observed to abrogate the beneficial effects of BCP in DCM mice. Taken together, BCP showed the potential to protect the myocardium and pancreas of DCM mice mediating CB2 receptor dependent mechanisms. Significance Statement 1. β-caryophyllene (BCP), a cannabinoid type 2 receptor (CB2R) agonist. 2. BCP attenuates diabetic cardiomyopathy via activating CB2R in mice 3. CB2R activation by BCP shows strong protection against fibrosis and inflammasome activation 4. It regulates AGE/RAGE and PI3K/Nrf2/Akt signaling in mice.
    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
    大麻素受体(CBRs)CB1和CB2的生物信息学分析揭示了它们的详细结构,进化,和内源性大麻素系统(ECS)内的生理意义。该研究强调了这些受体的进化保守性,这通过包括人类在内的不同物种的序列比对得到了证明。两栖动物,和鱼。两种CBR都具有七个跨膜(TM)螺旋的结构标志,A类G蛋白偶联受体(GPCRs)的特征,这对它们的信号功能至关重要。该研究报告两个CBR序列之间的相似性为44.58%,这表明尽管它们的进化路径和生理角色可能不同,他们的结构有相当大的保护作用。像KEGG这样的路径数据库,Reactome,和WikiPathways被用来确定受体在各种信号通路中的参与。本研究中整合的途径分析提供了大麻素相关信号通路复杂网络中CBRs相互作用的详细视图。高分辨率晶体结构(PDBID:CB1为5U09,CB2为5ZTY)提供了准确的结构信息,显示受体的结合袋体积和表面积,对于配体相互作用至关重要。这些受体的天然序列和它们的工程伪CBRs(p-CBRs)之间的比较显示出高度的序列同一性,证实在受体-配体相互作用研究中使用p-CBRs的有效性。这种综合分析增强了对大麻素受体结构和功能动力学的理解,强调它们的生理作用和它们作为ECS内治疗靶点的潜力。
    The bioinformatic analysis of cannabinoid receptors (CBRs) CB1 and CB2 reveals a detailed picture of their structure, evolution, and physiological significance within the endocannabinoid system (ECS). The study highlights the evolutionary conservation of these receptors evidenced by sequence alignments across diverse species including humans, amphibians, and fish. Both CBRs share a structural hallmark of seven transmembrane (TM) helices, characteristic of class A G-protein-coupled receptors (GPCRs), which are critical for their signalling functions. The study reports a similarity of 44.58 % between both CBR sequences, which suggests that while their evolutionary paths and physiological roles may differ, there is considerable conservation in their structures. Pathway databases like KEGG, Reactome, and WikiPathways were employed to determine the involvement of the receptors in various signalling pathways. The pathway analyses integrated within this study offer a detailed view of the CBRs interactions within a complex network of cannabinoid-related signalling pathways. High-resolution crystal structures (PDB ID: 5U09 for CB1 and 5ZTY for CB2) provided accurate structural information, showing the binding pocket volume and surface area of the receptors, essential for ligand interaction. The comparison between these receptors\' natural sequences and their engineered pseudo-CBRs (p-CBRs) showed a high degree of sequence identity, confirming the validity of using p-CBRs in receptor-ligand interaction studies. This comprehensive analysis enhances the understanding of the structural and functional dynamics of cannabinoid receptors, highlighting their physiological roles and their potential as therapeutic targets within the ECS.
    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:内源性大麻素,存在于整个中枢神经系统(CNS),能激活CB1和CB2受体。CB1和CB2激动剂表现出广泛的抗炎特性,表明了它们治疗炎症性疾病的潜力。然而,仔细评估滥用的可能性是必要的。方法:本研究评估了Lenabasum的滥用潜力,参与者(n=56)支持娱乐性大麻使用的选择性CB2受体激动剂。将三种剂量的lenabasum(20、60和120mg)与安慰剂进行比较,和纳比酮(3和6mg)。主要终点是双极性药物喜欢视觉模拟量表(VAS)的峰值效应(Emax)。评估次要VAS和药代动力学(PK)终点和不良事件。结果:Lenabasum安全且耐受性良好。与安慰剂相比,20mg剂量的lenabasum没有增加药物喜欢度的评级,并且对其他VAS终点没有明显影响。用60和120mglenabasum观察到药物喜好评级的剂量依赖性增加。纳比隆3mg和6mg的药物喜好和所有其他VAS结果最大,这是目前FDA批准的药物。结论:在目标治疗剂量(20mg)下,Lenabasum并未引起对药物喜好的主观评价。然而,与安慰剂相比,lenabasum的超治疗剂量(60和120mg)确实引起了药物喜欢的主观评分。尽管两种剂量的lenabasum与3mg和6mg的nabilone相比,药物喜好评分较低,提示Lenabasum确实有滥用的可能,在临床应用中应谨慎使用.重要性声明这项工作提供了证据,表明在有娱乐性大麻使用史的人中,Lenabasum是安全且耐受性良好的,尽管它确实显示出滥用的可能性。这项工作支持lenabasum用于潜在治疗适应症的进一步开发。
    Background: Endocannabinoids, which are present throughout the central nervous system (CNS), can activate CB1 and CB2 receptors. CB1 and CB2 agonists exhibit broad anti-inflammatory properties, suggesting their potential to treat inflammatory diseases. However, careful evaluation of abuse potential is necessary. Methods: This study evaluated the abuse potential of lenabasum, a selective CB2 receptor agonist in participants (n=56) endorsing recreational cannabis use. Three doses of lenabasum (20, 60, and 120mg) were compared to placebo, and nabilone (3 and 6mg). The primary endpoint was the peak effect (Emax) on a bipolar Drug Liking visual analog scale (VAS). Secondary VAS and pharmacokinetic (PK) endpoints and adverse events were assessed. Results: Lenabasum was safe and well tolerated. Compared to placebo, a 20mg dose of lenabasum did not increase ratings of Drug Liking and had no distinguishable effect on other VAS endpoints. Dose-dependent increases in ratings of Drug Liking were observed with 60 and 120mg lenabasum. Drug Liking and all other VAS outcomes were greatest for nabilone 3mg and 6mg, which is a currently FDA-approved medication. Conclusions: At a target therapeutic dose (20mg), lenabasum did not elicit subjective ratings of Drug Liking. However, supratherapeutic doses of lenabasum (60 and 120mg) did elicit subjective ratings of Drug Liking compared to placebo. Although both doses of lenabasum were associated with lower ratings of Drug Liking compared to 3mg and 6mg of nabilone, suggesting that lenabasum does have abuse potential and should be used cautiously in clinical settings. Significance Statement This work provides evidence that in people with a history of recreational cannabis use, lenabasum was safe and well-tolerated, although it did demonstrate abuse potential. This work supports further development of lenabasum for potential therapeutic indications.
    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
    大麻素及其受体在胃肠(GIT)蠕动和肠屏障通透性的调节中起重要作用。这篇综述严格评估了有关内源性大麻素和植物大麻素对GIT功能的作用机制和生物学作用以及这些化合物的潜在治疗应用的最新知识。离体和体内临床前数据的结果表明,大麻素可以抑制和刺激肠道蠕动,取决于各种因素。内源性大麻素以大麻素(CB)受体特异性方式影响蠕动;然而,它们与瞬时受体电位阳离子通道亚家族V成员1(TRPV1)系统之间也存在重要的相互作用。植物大麻素如Δ9-四氢大麻酚(THC)和大麻二酚(CBD)主要通过CB1受体影响肠道运动。还发现它们可以改善肠道屏障的完整性,主要通过CB1受体刺激,也可以通过蛋白激酶A(PKA),丝裂原相关蛋白激酶(MAPK),和腺苷酸环化酶信号通路,以及通过影响紧密连接(TJ)蛋白的表达。据推测,大麻素在GIT疾病中的抗炎作用是通过降低炎症因子如髓过氧化物酶(MPO)活性和调节细胞因子水平而发生的。总之,使用大麻素作为GIT疾病治疗的组成部分是有前景的。
    Cannabinoids and their receptors play a significant role in the regulation of gastrointestinal (GIT) peristalsis and intestinal barrier permeability. This review critically evaluates current knowledge about the mechanisms of action and biological effects of endocannabinoids and phytocannabinoids on GIT functions and the potential therapeutic applications of these compounds. The results of ex vivo and in vivo preclinical data indicate that cannabinoids can both inhibit and stimulate gut peristalsis, depending on various factors. Endocannabinoids affect peristalsis in a cannabinoid (CB) receptor-specific manner; however, there is also an important interaction between them and the transient receptor potential cation channel subfamily V member 1 (TRPV1) system. Phytocannabinoids such as Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD) impact gut motility mainly through the CB1 receptor. They were also found to improve intestinal barrier integrity, mainly through CB1 receptor stimulation but also via protein kinase A (PKA), mitogen-associated protein kinase (MAPK), and adenylyl cyclase signaling pathways, as well as by influencing the expression of tight junction (TJ) proteins. The anti-inflammatory effects of cannabinoids in GIT disorders are postulated to occur by the lowering of inflammatory factors such as myeloperoxidase (MPO) activity and regulation of cytokine levels. In conclusion, there is a prospect of utilizing cannabinoids as components of therapy for GIT disorders.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号