Home>>Signaling Pathways>> Endocrinology and Hormones>> Opioid Receptor>>Endomorphin 1 acetate

Endomorphin 1 acetate

目录号 : GC67956

Endomorphin 1 acetate,一种高度选择性的,高亲和力的 μ-opioid 受体激动剂 (Ki: 1.11 nM),对 kappa3结合位点具有高亲和力,Ki 值为 20 到 30 nM 之间。Endomorphin 1 acetate 具有镇痛特性。

Endomorphin 1 acetate Chemical Structure

Cas No.:1276123-71-7

规格 价格 库存 购买数量
10mg
¥693.00
现货
25mg
¥1,188.00
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:

产品描述

Endomorphin 1 acetate, a high affinity, highly selective agonist of the μ-opioid receptor (Ki: 1.11 nM), displays reasonable affinities for kappa3 binding sites, with Ki value between 20 and 30 nM. Endomorphin 1 acetate has antinociceptive properties[1][2][4].

Endomorphin 1 acetate inhibits Forskolin (1 μM) stimulated cyclic AMP formation with a pIC50 value of 8.03 in In CHOμ cells[5].
Endomorphin 1 (1-10 μM) acetate increases interleukin-8 secretion in Caco-2 cells[6].
Endomorphin 1 (1 μM) acetate inhibits excitatory transmission in adult rat substantia gelatinosa neurons[7].

Endomorphin 1 (i.c.v.) acetate shows antinociceptive properties in mice, with an ED50 value of 6.16 nM[2].
Endomorphin 1 (50 μg/kg, i.v., rats) acetate alleviates myocardial ischemia/reperfusion injury (MIRI) by inhibiting the inflammatory response[3].

Animal Model: ICR mice[2].
Dosage: 6.16 nM (ED50)
Administration: Intracerebroventricularly (i.c.v.) injection
Result: Inhibited dose-dependently the tail-flick response.
Animal Model: Rats[3].
Dosage: 50 μg/kg
Administration: Intravenously following LAD ligation for 25 min, subsequently the LAD was reperfused for 120 min.
Result: Alleviated MIRI by reducing the production of free radicals.
Dncreased LDH and CK-MB activities.
Increased SOD activity and decreased MDA content.
Decreased IL-6 and TNF-α plasma content.

[1]. Goldberg IE, et al. Pharmacological characterization of endomorphin-1 and endomorphin-2 in mouse brain. J Pharmacol Exp Ther. 1998 Aug;286(2):1007-13.
[2]. Tseng LF. The antinociceptive properties of endomorphin-1 and endomorphin-2 in the mouse. Jpn J Pharmacol. 2002 Jul;89(3):216-20.
[3]. Zhang WP, et al. Effects of endomorphin-1 postconditioning on myocardial ischemia/reperfusion injury and myocardial cell apoptosis in a rat model. Mol Med Rep. 2016 Oct;14(4):3992-8.
[4]. Koda Y, et al. Synthesis and in vitro evaluation of a library of modified endomorphin 1 peptides. Bioorg Med Chem. 2008 Jun 1;16(11):6286-96.
[5]. Harrison C, et al. The effects of endomorphin-1 and endomorphin-2 in CHO cells expressing recombinant mu-opioid receptors and SH-SY5Y cells. Br J Pharmacol. 1999 Sep;128(2):472-8.
[6]. Neudeck BL, et al. Endomorphin-1 alters interleukin-8 secretion in Caco-2 cells via a receptor mediated process. Immunol Lett. 2002 Dec 3;84(3):217-21.
[7]. Fujita T, et al. Inhibition by endomorphin-1 and endomorphin-2 of excitatory transmission in adult rat substantia gelatinosa neurons. Neuroscience. 2006;139(3):1095-105.

Chemical Properties

Cas No. 1276123-71-7 SDF Download SDF
分子式 C36H42N6O7 分子量 670.75
溶解度 DMSO : 250 mg/mL (372.72 mM; Need ultrasonic) 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 1.4909 mL 7.4543 mL 14.9087 mL
5 mM 0.2982 mL 1.4909 mL 2.9817 mL
10 mM 0.1491 mL 0.7454 mL 1.4909 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

Analgesic effect of Endomorphin-1

Acta Pharmacol Sin 2001 Nov;22(11):976-80.PMID:11749786doi

Aim: To study the analgesic effect of Endomorphin-1 (EM-1). Methods: The experiment was performed in rats and mice to study the analgesic effect of intraperitoneal (ip) injection of EM-1 with tail stimulation-vocalization test, writhing test, adjuvant arthritis, and neuropathic pain model and to compare it with the analgesic effects produced by intracerebroventricular (icv) and intrathecal (it) administrations. Results: 1) EM-1 raised the pain threshold dose-dependently in tail stimulation-vocalization test in rats and inhibited the writhing responses induced by ip acetic acid in mice. EM-1 also decreased the hyperalgesia in both adjuvant arthritis and neuropathic pain model. 2) The analgesic effect induced by central (icv and it) administration of EM-1 was faster and more powerful than that induced by peripheral (ip) administration. 3) The analgesic effect of EM-1 was reversed by naloxone (opioid receptor antagonist), as well as by cyprodime (mu-opioid receptor selective antagonist). Repeated administrations of EM-1 induced tolerance. Conclusion: EM-1 had a definite analgesic effect and the analgesic effect of EM-1 was mediated by central mu-opioid receptor.

Antinociception by a peripherally administered novel Endomorphin-1 analogue containing beta-proline

Eur J Pharmacol 2003 May 23;469(1-3):89-95.PMID:12782189DOI:10.1016/s0014-2999(03)01736-9.

We previously described a novel Endomorphin-1 analogue (Tyr-L-beta-Pro-Trp-Phe-NH(2); Endo1-beta-Pro) more resistant to enzymatic hydrolysis than Endomorphin-1 that acts as a mu-opioid receptor agonist. In this study we report that Endo1-beta-Pro, s.c. injected in the mouse, is an effective antinociceptive agent in the tail flick (ED(50)=9.2 mg/kg) and acetic acid-induced abdominal constriction (ED(50)=1.2 mg/kg) tests. Moreover, s.c. Endo1-beta-Pro significantly decreases, in the mouse, the gastrointestinal propulsion measured as transit of an orally administered charcoal meal (ED(50)=10.0 mg/kg). Subcutaneous beta-funaltrexamine or a high dose of the mu(1)-opioid receptor-selective antagonist naloxonazine (50 mg/kg) prevents the antinociceptive and antitransit action of Endo1-beta-Pro; moreover, these effects are partially blocked by i.c.v. naloxone or by i.p. naloxone methiodide, this latter does not readily cross the blood-brain barrier. On the contrary, the kappa-opioid receptor antagonist nor-binaltorphimine or the delta-opioid receptor antagonist naltrindole are ineffective Thus, Endo1-beta-Pro may act, preferentially, through central and peripheral mu(2)-opioid receptors to produce antinociception and to inhibit gastrointestinal transit. Endo1-beta-Pro is among the first Endomorphin-1 analogues showing antinociceptive activity after systemic administration. This compound will be extremely useful for exploring the pharmacological profile of endomorphins in vivo and confirms the potential therapeutic interest of endomorphin derivatives as novel analgesic agents.

Peripheral antinociceptive effects of the cyclic Endomorphin-1 analog c[YpwFG] in a mouse visceral pain model

Peptides 2010 Nov;31(11):2135-40.PMID:20713109DOI:10.1016/j.peptides.2010.08.005.

We previously described a novel cyclic Endomorphin-1 analog c[Tyr-D-Pro-D-Trp-Phe-Gly] (c[YpwFG]), acting as a mu-opioid receptor (MOR) agonist. This study reports that c[YpwFG] is more lipophilic and resistant to enzymatic hydrolysis than Endomorphin-1 and produces preemptive antinociception in a mouse visceral pain model when injected intraperitoneally (i.p.) or subcutaneously (s.c.) before 0.6% acetic acid, employed to evoke abdominal writhing (i.p. ED(50)=1.24 mg/kg; s.c. ED(50)=2.13 mg/kg). This effect is reversed by the selective MOR antagonist β-funaltrexamine and by a high dose of the mu(1)-opioid receptor-selective antagonist naloxonazine. Conversely, the kappa-opioid receptor antagonist nor-binaltorphimine and the delta-opioid receptor antagonist naltrindole are ineffective. c[YpwFG] produces antinociception when injected i.p. after acetic acid (ED(50)=4.80 mg/kg), and only at a dose of 20mg/kg did it elicit a moderate antinociceptive response in the mouse, evaluated by the tail flick assay. Administration of a lower dose of c[YpwFG] (10mg/kg i.p.) apparently produces a considerable part of antinociception on acetic acid-induced writhes through peripheral opioid receptors as this action is fully prevented by i.p. naloxone methiodide, which does not readily cross the blood-brain barrier; whereas this opioid antagonist injected intracerebroventricularly (i.c.v.) is not effective. Antinociception produced by a higher dose of c[YpwFG] (20mg/kg i.p.) is partially reversed by naloxone methiodide i.c.v. administered. Thus, only at the dose of 20mg/kg c[YpwFG] can produce antinociception through both peripheral and central opioid receptors. In conclusion, c[YpwFG] displays sufficient metabolic stability to be effective after peripheral administration and demonstrates the therapeutic potential of endomorphin derivatives as novel analgesic agents to control visceral pain.

A new concept affecting restoration of inflammation-reactive astrocytes

Neuroscience 2013 Oct 10;250:536-45.PMID:23892009DOI:10.1016/j.neuroscience.2013.07.033.

Long-lasting pain may partly be a consequence of ongoing neuroinflammation, in which astrocytes play a significant role. Following noxious stimuli, increased inflammatory receptor activity, influences in Na(+)/K(+)-ATPase activity and actin filament organization occur within the central nervous system. In astrocytes, the Ca(2+) signaling system, Na(+) transporters, cytoskeleton, and release of pro-inflammatory cytokines change during inflammation. The aim of this study was to restore these cell parameters in inflammation-reactive astrocytes. We found that the combination of (1) Endomorphin-1, an opioid agonist that stimulates the Gi/o protein of the μ-opioid receptor; (2) naloxone, an opioid antagonist that inhibits the Gs protein of the μ-opioid receptor at ultralow concentrations; and (3) levetiracetam, an anti-epileptic agent that counteracts the release of IL-1β, managed to activate the Gi/o protein and Na(+)/K(+)-ATPase activity, inhibit the Gs protein, and decrease the release of IL-1β. The cell functions of astrocytes in an inflammatory state were virtually restored to their normal non-inflammatory state and it could be of clinical significance and may be useful for the treatment of long-term pain.

Morphine and endomorphins differentially regulate micro-opioid receptor mRNA in SHSY-5Y human neuroblastoma cells

J Pharmacol Exp Ther 2003 Aug;306(2):447-54.PMID:12754318DOI:10.1124/jpet.103.048694.

A sensitive quantitative-competitive reverse transcriptase-polymerase chain reaction method was developed to measure micro-opioid receptor (MOR) mRNA expression in SHSY-5Y neuroblastoma cells. Differentiation of SHSY-5Y cells with either retinoic acid (RA) or 12-o-tetradecanoyl-phorbol-13-acetate (TPA) significantly increased MOR mRNA levels. Morphine treatment (10 microM) for 24 h decreased MOR mRNA levels in control, as well as RA- and TPA-differentiated cells. In contrast, chronic exposure to the opioid peptides Endomorphin-1 or endomorphin-2 significantly increased MOR mRNA levels in undifferentiated and RA-differentiated cells. An opioid antagonist, naloxone, reversed the morphine and Endomorphin-1 and -2 effects on MOR mRNA levels in undifferentiated SHSY-5Y cells, but naloxone had differential reversing effects on the agonists' regulation of MOR mRNA in RA- or TPA-differentiated cells. To investigate whether the changes in MOR mRNA expression paralleled changes in MOR receptor function, intracellular cAMP accumulation in SHSY-5Y cells was measured. After chronic treatment with morphine, forskolin-induced cAMP levels in SHSY-5Y cells were significantly higher than those of untreated control cells. In contrast, forskolin-induced cAMP accumulation levels were lower in cells treated with Endomorphin-1 or -2 than in untreated control cells. Together, our studies indicate that the opioid alkaloid morphine and the opioid peptides Endomorphin-1 and -2 differentially regulate MOR mRNA expression and MOR function in SHSY-5Y cells.