Home>>Signaling Pathways>> Neuroscience>> GPR139>>JNJ-63533054

JNJ-63533054 Sale

(Synonyms: 3-氯-N-[2-氧代-2-[[(1S)-1-苯基乙基]氨基]乙基]苯甲酰胺) 目录号 : GC19209

A potent agonist of GPR139

JNJ-63533054 Chemical Structure

Cas No.:1802326-66-4

规格 价格 库存 购买数量
5mg
¥441.00
现货
10mg
¥756.00
现货
50mg
¥2,867.00
现货
100mg
¥4,715.00
现货

电话:400-920-5774 Email: sales@glpbio.cn

Customer Reviews

Based on customer reviews.

Sample solution is provided at 25 µL, 10mM.

产品文档

Quality Control & SDS

View current batch:

实验参考方法

Cell experiment [1]:

Cell lines

HEK293-T cells expressing zebrafish GPR139

Preparation Method

The cells were serum starved in the media with 0.5% FBS for 18-20h, and then treated with the vehicle (control) or various concentrations of JNJ-63533054 in the media for 6h.

Reaction Conditions

4-128nM in 0.5% DMSO, 6h

Applications

JNJ-63533054 induced luciferase activity against zebrafish GPR139 in a concentration-dependent manner. For zebrafish GPR139, the maximal induction of 3.3-fold of vehicle control was achieved at a concentration of 128nM. From the dose-response curve, the half effective maximal concentration (EC50) values of JNJ-63533054 to zebrafish GPR139 was 3.91nM.

Animal experiment [2]:

Animal models

Male Sprague-Dawley rats, 350-450g

Preparation Method

Separate groups of rats were orally dosed at 2 hours into the light phase with the GPR139 agonist JNJ-63533054 (3, 10, and 30mg/kg, n=8), its less active enantiomer JNJ-63770044 (10mg/kg, n=7), and L-Trp (200mg/kg, n=7) or vehicle (0.5% hydroxypropyl methylcellulose in suspension) administered in the same animals receiving each dose.

Dosage form

3, 10, and 30mg/kg, orally dosed

Applications

The results are presented for the first hour after dosing based on the short-lasting effects observed. Compared with vehicle, JNJ-63533054 induced a dose-dependent reduction in locomotor activity in the first hour after the treatment that reached significance from the dose of 10mg/kg onward. In contrast, at the same dose of 10mg/kg, its less active enantiomer did not modify locomotor activity. For comparison, L-Trp was tested in the same experimental conditions and spontaneous locomotor activity was not affected at 200mg/kg.

References:

[1]. Roy N, Ogawa S, et al. Habenula GPR139 is associated with fear learning in the zebrafish. Sci Rep. 2021;11(1):5549. Published 2021 Mar 10.

[2]. Liu C, Bonaventure P, et al. GPR139, an Orphan Receptor Highly Enriched in the Habenula and Septum, Is Activated by the Essential Amino Acids L-Tryptophan and L-Phenylalanine. Mol Pharmacol. 2015;88(5):911-925.

产品描述

JNJ-63533054 is a potent and selective agonist of hGPR139 with an EC50 = 16nM[1,2]. JNJ-63533054 is an excellent candidate to explore the unknown in vivo function of central GPR139[3]

JNJ-63533054 specifically activated human GPR139 in the calcium mobilization (EC50=16±6nM) and GTPγS binding (EC50=17±4nM) assays[1]. JNJ-63533054 effectively binds and acts as an agonist to zebrafish GPR139 with EC50 of 3.91nM[4]

JNJ-63533054 activated the rat and mouse GPR139 receptor with similar potency (rat EC50=63±24nM, mouse EC50=28±7nM)[1]. JNJ-63533054(oral dose of 10mg/kg) crossed the blood-brain barrier in both male mice and male rats, and the brain to plasma ratio was close to 1 in mouse and slightly higher in rat. In the marble burying test, JNJ-63533054(10mg/kg p.o.) produced a small anxiolytic-like effect, with no interaction with fluoxetine, and no effect in elevated plus maze (EPM)[3]. Administration of a high dose (1μg/g BW) of JNJ-6353305 had no effect on locomotor activity, and fear response, but fear-conditioned place avoidance was diminished; zebrafish treated with a lower dose(0.1μg/g BW) of GPR139 agonist exhibited avoidance to the contextual compartments[4]. JNJ-63533054(oral dose of 3-30mg/kg) dose-dependently reduced non-rapid eye movement (NREM) latency and increased NREM sleep duration without altering rapid eye movement (REM) sleep when acutely administered at the beginning of the light phase. This effect progressively dissipated upon 7-day repeated dosing[5]. Systemic administration of JNJ-63533054 (30mg/kg, p.o.) reversed compulsive-like alcohol drinking and decreases withdrawal-induced hyperalgesia in alcohol-dependent rats that exhibit symptoms of alcohol dependence[6]

References:
[1]. Liu C, Bonaventure P, et al. GPR139, an Orphan Receptor Highly Enriched in the Habenula and Septum, Is Activated by the Essential Amino Acids L-Tryptophan and L-Phenylalanine. Mol Pharmacol. 2015;88(5):911-925.
[2]. Dvorak CA, Coate H, et al. Identification and SAR of Glycine Benzamides as Potent Agonists for the GPR139 Receptor. ACS Med Chem Lett. 2015;6(9):1015-1018. Published 2015 Jul 20.
[3]. Shoblock JR, Welty N, et al. In vivo Characterization of a Selective, Orally Available, and Brain Penetrant Small Molecule GPR139 Agonist. Front Pharmacol. 2019;10:273. Published 2019 Mar 21.
[4]. Roy N, Ogawa S, et al. Habenula GPR139 is associated with fear learning in the zebrafish. Sci Rep. 2021;11(1):5549. Published 2021 Mar 10.
[5]. Wang L, Dugovic C, et al. Putative role of GPR139 on sleep modulation using pharmacological and genetic rodent models. Eur J Pharmacol. 2020;882:173256.
[6]. Kononoff J, Kallupi M, et al. Systemic and Intra-Habenular Activation of the Orphan G Protein-Coupled Receptor GPR139 Decreases Compulsive-Like Alcohol Drinking and Hyperalgesia in Alcohol-Dependent Rats. eNeuro. 2018;5(3):ENEURO.0153-18.2018. Published 2018 Jul 2.

JNJ-63533054 是一种有效的选择性 hGPR139 激动剂,EC50 = 16nM[1,2]。 JNJ-63533054 是探索中枢 GPR139 未知体内功能的极佳候选者[3]

JNJ-63533054 在钙动员 (EC50=16±6nM) 和 GTPγS 结合 (EC50=17±4nM) 测定中特异性激活人 GPR139[1]。 JNJ-63533054 有效结合并作为斑马鱼 GPR139 的激动剂,EC50 为 3.91nM[4]

JNJ-63533054 激活大鼠和小鼠 GPR139 受体具有相似的效力(大鼠 EC50=63±24nM,小鼠 EC50=28±7nM) [1]. JNJ-63533054(口服剂量10mg/kg)在雄性小鼠和雄性大鼠体内均可透过血脑屏障,小鼠脑浆比接近1,大鼠略高。在大理石掩埋试验中,JNJ-63533054(10mg/kg p.o.) 产生了轻微的抗焦虑样作用,与氟西汀没有相互作用,在高架十字迷宫 (EPM) 中也没有作用[3]。给予高剂量(1μg/g BW)的 JNJ-6353305 对运动活动和恐惧反应没有影响,但恐惧条件性位置回避减少;用较低剂量 (0.1μg/g BW) 的 GPR139 激动剂处理的斑马鱼表现出对上下文隔室的回避[4]。 JNJ-63533054(口服剂量为 3-30 毫克/千克)在光照阶段开始时急性给药时,剂量依赖性地减少非快速眼动 (NREM) 潜伏期并增加 NREM 睡眠持续时间,而不改变快速眼动 (REM) 睡眠.这种效果在重复给药 7 天后逐渐消失[5]。 JNJ-63533054(30mg/kg,口服)的全身给药可逆转表现出酒精依赖症状的酒精依赖大鼠的强迫性饮酒并减少戒断诱导的痛觉过敏[6]

Chemical Properties

Cas No. 1802326-66-4 SDF
别名 3-氯-N-[2-氧代-2-[[(1S)-1-苯基乙基]氨基]乙基]苯甲酰胺
Canonical SMILES O=C(NCC(N[C@H](C1=CC=CC=C1)C)=O)C2=CC=CC(Cl)=C2
分子式 C17H17ClN2O2 分子量 316.78
溶解度 DMSO : ≥ 50 mg/mL (157.84 mM);Water : < 0.1 mg/mL (insoluble) 储存条件 Store at -20°C
General tips 请根据产品在不同溶剂中的溶解度选择合适的溶剂配制储备液;一旦配成溶液,请分装保存,避免反复冻融造成的产品失效。
储备液的保存方式和期限:-80°C 储存时,请在 6 个月内使用,-20°C 储存时,请在 1 个月内使用。
为了提高溶解度,请将管子加热至37℃,然后在超声波浴中震荡一段时间。
Shipping Condition 评估样品解决方案:配备蓝冰进行发货。所有其他可用尺寸:配备RT,或根据请求配备蓝冰。

溶解性数据

制备储备液
1 mg 5 mg 10 mg
1 mM 3.1568 mL 15.7838 mL 31.5676 mL
5 mM 0.6314 mL 3.1568 mL 6.3135 mL
10 mM 0.3157 mL 1.5784 mL 3.1568 mL
  • 摩尔浓度计算器

  • 稀释计算器

  • 分子量计算器

质量
=
浓度
x
体积
x
分子量
 
 
 
*在配置溶液时,请务必参考产品标签上、MSDS / COA(可在Glpbio的产品页面获得)批次特异的分子量使用本工具。

计算

动物体内配方计算器 (澄清溶液)

第一步:请输入基本实验信息(考虑到实验过程中的损耗,建议多配一只动物的药量)
给药剂量 mg/kg 动物平均体重 g 每只动物给药体积 ul 动物数量
第二步:请输入动物体内配方组成(配方适用于不溶于水的药物;不同批次药物配方比例不同,请联系GLPBIO为您提供正确的澄清溶液配方)
% DMSO % % Tween 80 % saline
计算重置

Research Update

Habenula GPR139 is associated with fear learning in the zebrafish

G-protein coupled receptor 139 (GPR139) is an evolutionarily conserved orphan receptor, predominantly expressing in the habenula of vertebrate species. The habenula has recently been implicated in aversive response and its associated learning. Here, we tested the hypothesis that GPR139 signalling in the habenula may play a role in fear learning in the zebrafish. We examined the effect of intraperitoneal injections of a human GPR139-selective agonist (JNJ-63533054) on alarm substance-induced fear learning using conditioned place avoidance paradigm, where an aversive stimulus is paired with one compartment, while its absence is associated with the other compartment of the apparatus. The results indicate that fish treated with 1 ?g/g body weight of GPR139 agonist displayed no difference in locomotor activity and alarm substance-induced fear response. However, avoidance to fear-conditioned compartment was diminished, which suggests that the agonist blocks the consolidation of contextual fear memory. On the other hand, fish treated with 0.1 ?g/g body weight of GPR139 agonist spent a significantly longer time in the unconditioned neutral compartment as compared to the conditioned (punished and unpunished) compartments. These results suggest that activation of GPR139 signalling in the habenula may be involved in fear learning and the decision-making process in the zebrafish.

Putative role of GPR139 on sleep modulation using pharmacological and genetic rodent models

GPR139 is a G-protein coupled receptor expressed in circumventricular regions of the habenula and septum. Amino acids L-tryptophan and L-phenylalanine have been shown to activate GPR139 at physiologically relevant concentrations. The aim of the present study was to investigate the role of GPR139 on sleep modulation using pharmacological and genetic (GPR139 knockout mice, KO) rodent models. To evaluate the effects of GPR139 pharmacological activation on sleep, rats were orally dosed with the selective GPR139 agonist JNJ-63533054 (3-30 mg/kg). When acutely administered at the beginning of the light phase, the GPR139 agonist dose-dependently reduced non-rapid eye movement (NREM) latency and increased NREM sleep duration without altering rapid eye movement (REM) sleep. This effect progressively dissipated upon 7-day repeated dosing, suggesting functional desensitization. Under baseline conditions, GPR139 KO mice spent less time in REM sleep compared to their wild type littermates during the dark phase, whereas NREM sleep was not altered. Under conditions of pharmacologically enhanced monoamine endogenous tone, GPR139 KO mice showed a blunted response to citalopram or fluoxetine induced REM sleep suppression and an attenuated response to the wake promoting effect of amphetamine. These findings indicate an emerging role of GPR139 in the modulation of sleep states.

Re-evaluation of Adrenocorticotropic Hormone and Melanocyte Stimulating Hormone Activation of GPR139 in Vitro

It is now well established that GPR139, a G-protein coupled receptor exclusively expressed in the brain and pituitary, is activated by the essential amino acids L-tryptophan (L-Trp) and L-phenylalanine (L-Phe) via Gαq-coupling. The in vitro affinity and potency values of L-Trp and L-Phe are within the physiological concentration ranges of L-Trp and L-Phe. A recent paper suggests that adrenocorticotropic hormone (ACTH), α and β melanocyte stimulating hormones (α-MSH and β-MSH) and derivatives α-MSH1-9/α-MSH1-10 can also activate GPR139 in vitro. We tested this hypothesis using guanosine 5'-O-(3-[35S]thio)-triphosphate binding (GTPγS), calcium mobilization and [3H]JNJ-63533054 radioligand binding assays. In the GTPγS binding assay, α-MSH, α-MSH1-9/α-MSH1-10, and β-MSH had no effect on [35S]GTPγS incorporation in cell membranes expressing GPR139 up to 30 μM in contrast to the concentration dependent activation produced by L-Trp, JNJ-63533054, and TC-09311 (two small molecule GPR139 agonists). ACTH slightly decreased the basal level of [35S]GTPγS incorporation at 30 μM. In the GPR139 radioligand binding assay, a moderate displacement of [3H]JNJ-63533054 binding by ACTH and β-MSH was observed at 30 μM (40 and 30%, respectively); α-MSH, α-MSH1-9/α-MSH1-10 did not displace any specific binding at 30 μM. In three different host cell lines stably expressing GPR139, α-MSH, and β-MSH did not stimulate calcium mobilization in contrast to L-Trp, JNJ-63533054, and TC-09311. ACTH, α-MSH1-9/α-MSH1-10 only weakly stimulated calcium mobilization at 30 μM (<50% of EC100). We then co-transfected GPR139 with the three melanocortin (MC) receptors (MC3R, MC4R, and MC5R) to test the hypothesis that ACTH, α-MSH, and β-MSH might stimulate calcium mobilization through a MCR/GPR139 interaction. All three MC peptides stimulated calcium response in cells co-transfected with GPR139 and MC3R, MC4R, or MC5R. The MC peptides did not stimulate calcium response in cells expressing MC3R or MC5R alone consistent with the Gs signaling transduction pathway of these receptors. In agreement with the previously reported multiple signaling pathways of MC4R, including Gq transduction pathway, the MC peptides produced a calcium response in cells expressing MC4R alone. Together, our findings do not support that GPR139 is activated by ACTH, α-MSH, and β-MSH at physiologically relevant concentration but we did unravel an in vitro interaction between GPR139 and the MCRs.

In vivo Characterization of a Selective, Orally Available, and Brain Penetrant Small Molecule GPR139 Agonist

Recently, our group along with another demonstrated that GPR139 can be activated by L-phenylalanine (L-Phe) and L-tryptophan (L-Trp) at physiologically relevant concentrations. GPR139 is discretely expressed in brain, with highest expression in medial habenula. Not only are the endogenous ligands catecholamine/serotonin precursors, but GPR139 expressing areas can directly/indirectly regulate the activity of catecholamine/serotonin neurons. Thus, GPR139 appears expressed in an interconnected circuit involved in mood, motivation, and anxiety. The aim of this study was to characterize a selective and brain penetrant GPR139 agonist (JNJ-63533054) in relevant in vivo models. JNJ-63533054 was tested for its effect on c-fos activation in the habenula and dorsal striatum. In vivo microdialysis experiments were performed in freely moving rats to measure basal levels of serotonin or dopamine (DA) in prefrontal cortex (mPFC) and nucleus accumbens (NAc). Finally, the compound was profiled in behavioral models of anxiety, despair, and anhedonia. The agonist (10-30 mg/kg, p.o.) did not alter c-fos expression in medial habenula or dorsal striatum nor neurotransmitter levels in mPFC or NAc. JNJ-63533054 (10 mg/kg p.o.) produced an anhedonic-like effect on urine sniffing, but had no significant effect in tail suspension, with no interaction with imipramine, no effect on naloxone place aversion, and no effect on learned helplessness. In the marble burying test, the agonist (10 mg/kg p.o.) produced a small anxiolytic-like effect, with no interaction with fluoxetine, and no effect in elevated plus maze (EPM). Despite GPR139 high expression in medial habenula, an area with connections to limbic and catecholaminergic/serotoninergic areas, the GPR139 agonist had no effect on c-fos in medial habenula. It did not alter catecholamine/serotonin levels and had a mostly silent signal in in vivo models commonly associated with these pathways. The physiological function of GPR139 remains elusive.

Systemic and Intra-Habenular Activation of the Orphan G Protein-Coupled Receptor GPR139 Decreases Compulsive-Like Alcohol Drinking and Hyperalgesia in Alcohol-Dependent Rats

GPR139 is an orphan G protein-coupled receptor (GPCR) that is expressed mainly in the brain, with the highest expression in the medial habenula. The modulation of GPR139 receptor function has been hypothesized to be beneficial in the treatment of some mental disorders, but behavioral studies have not yet provided causal evidence of the role of GPR139 in brain dysfunction. Because of the high expression of GPR139 in the habenula, a critical brain region in addiction, we hypothesized that GPR139 may play role in alcohol dependence. Thus, we tested the effect of GPR139 receptor activation using the selective, brain-penetrant receptor agonist JNJ-63533054 on addiction-like behaviors in alcohol-dependent male rats. Systemic administration of JNJ-63533054 (30 mg/kg but not 10 mg/kg, p.o.) reversed the escalation of alcohol self-administration in alcohol-dependent rats, without affecting water or saccharin intake in dependent rats or alcohol intake in nondependent rats. Moreover, systemic JNJ-63533054 administration decreased withdrawal-induced hyperalgesia, without affecting somatic signs of alcohol withdrawal. Further analysis demonstrated that JNJ-63533054 was effective only in a subgroup of dependent rats that exhibited compulsive-like alcohol drinking. Finally, site-specific microinjection of JNJ-63533054 in the habenula but not interpeduncular nucleus (IPN) reduced both alcohol self-administration and withdrawal-induced hyperalgesia in dependent rats. These results provide robust preclinical evidence that GPR139 receptor activation reverses key addiction-like behaviors in dependent animals, suggest that GPR139 may be a novel target for the treatment of alcohol use disorder, and demonstrate that GPR139 is functionally relevant in regulating mammalian behavior.