Home>>Signaling Pathways>> Immunology/Inflammation>> NO Synthase>>Tat-NR2B9c

Tat-NR2B9c Sale

(Synonyms: Tat-NR2Bct; NA-1) 目录号 : GC30774

Tat-NR2B9c (Tat-NR2Bct; NA-1) 是一种突触后密度 95 (PSD-95) 抑制剂,对 PSD-95d2 (PSD-95 PDZ domain 2) 和 PSD-95d1 的 EC50 值为 6.7 nM 和 670 nM , 分别。

Tat-NR2B9c Chemical Structure

Cas No.:500992-11-0

规格 价格 库存 购买数量
1mg
¥625.00
现货
5mg
¥1,607.00
现货
10mg
¥2,499.00
现货
25mg
¥4,909.00
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.

101

客户使用产品发表文献 1

产品文档

Quality Control & SDS

View current batch:

实验参考方法

Cell experiment [1]:

Cell lines

Neuronal cells

Preparation Method

Studies were initiated in cultures at days 10 to 12 in vitro by exchanging the culture medium with a low-magnesium balanced salt solution.10 Peptides and drugs were added from concentrated stocks 15 minutes before the addition of NMDA.

Reaction Conditions

Treated neuronal cell cultures with 100 μmol/L NMDA for 30 minutes together with Tat-NR2B9c over a range of concentrations: 0, 0.05, 0.1, 0.5 μmol/L.

Applications

Tat-NR2B9c showed neuroprotectant efficacy in patients with acute stroke. Tat-NR2B9c is designed to uncouple NO production from NMDAr activation by blocking PSD-95 binding to NMDAr and nNOS. Tat-NR2B9c also prevents NMDA-induced superoxide p47phox formation by blocking phosphorylation.

Animal experiment [2]:

Animal models

Timed pregnant CD1 mice (7-day-old (P7) pups of either sex)

Preparation Method

Tat-NR2B9c was administered intraperitoneally at a single dose of 15 μg/g body weight in 100–120 μl of saline.

Dosage form

15 μg/g

Applications

Tat-NR2B9c has a neuroprotective effect in the neonatal mouse hypoxic-ischemic brain injury model of stroke. In addition, Tat-NR2B9c reduced brain damage caused by hypoxic-ischemic injury and showed the potential to promote long-term recovery. Tat-NR2B9c would be effective in treating or preventing perinatal and neonatal hypoxic-ischemic brain injury, as well as its related brain disorders.

References:

[1]. Chen Y, et al. Tat-NR2B9c prevents excitotoxic neuronal superoxide production. J Cereb Blood Flow Metab. 2015 May;35(5):739-42.

[2]. Xu B, et al. Neuroprotective Effects of a PSD-95 Inhibitor in Neonatal Hypoxic-Ischemic Brain Injury. Mol Neurobiol. 2016 Nov;53(9):5962-5970.

产品描述

Tat-NR2B9c is designed to prevent nitric oxide (NO) production by preventing postsynaptic density protein 95 (PSD-95) binding to N-methyl-D-aspartate (NMDA) receptors and neuronal nitric oxide synthase. Tat-NR2B9c dissociates NMDA glutamate receptors from downstream excitotoxic signaling pathways without affecting normal glutamate receptor function. Neuroprotective effects of Tat-NR2B9c have been demonstrated in a diverse range of stroke models in several species including rodents, primates, and humans. Moreover, Tat-NR2B9c peptide has shown clinical efficacy as a neuroprotective agent in acute stroke.[1][2]

In vitro study indicated that Tat-NR2B9c have no measurable effect on the rate or magnitude of NMDA-induced calcium influx. However, Tat-NR2B9c prevented NMDA-induced DNA breaks, and the neuronal death could be significantly reduced by Tat-NR2B9c. Tat-NR2B9c also prevented NMDA-induced superoxide p47phox formation by blocking phosphorylation, and neuroprotective effect of Tat-NR2B9c may be partly or wholly attributable to its suppression of NOX2 activation. In addition, Tat-NR2B9c, which targets the PDZ domain of PSD-95, disrupts the functional coupling between NR2B and NOX2.[2]

In vivo experiments demonstrated that Tat-NR2B9c would be effective in treating or preventing perinatal and neonatal hypoxic-ischemic brain injury, as well as its related brain disorders. Results indicated that Tat-NR2B9c reduced brain damage caused by hypoxic-ischemic injury when administered either before or after ischemia and improved post-HI neurobehavioral outcomes when delivered before or after ischemia. Moreover, Tat-NR2B9c might exert neuroprotective effects through the promotion of pro-survival signaling and inhibition of pro-apoptotic signaling.[1]

References:
[1]. Xu B, et al. Neuroprotective Effects of a PSD-95 Inhibitor in Neonatal Hypoxic-Ischemic Brain Injury. Mol Neurobiol. 2016 Nov;53(9):5962-5970.
[2]. Chen Y, et al. Tat-NR2B9c prevents excitotoxic neuronal superoxide production. J Cereb Blood Flow Metab. 2015 May;35(5):739-42.

Tat-NR2B9c 旨在通过阻止突触后密度蛋白 95 (PSD-95) 与 N-甲基-D-天冬氨酸 (NMDA) 受体和神经元一氧化氮结合来阻止一氧化氮 (NO) 的产生氧化物合成酶。 Tat-NR2B9c 将 NMDA 谷氨酸受体与下游兴奋性毒性信号通路解离,而不影响正常的谷氨酸受体功能。 Tat-NR2B9c 的神经保护作用已在多个物种(包括啮齿动物、灵长类动物和人类)的各种中风模型中得到证实。此外,Tat-NR2B9c 肽作为急性中风的神经保护剂已显示出临床疗效。[1][2]

体外研究表明,Tat-NR2B9c 对 NMDA 诱导的钙内流的速率或幅度没有可测量的影响。然而,Tat-NR2B9c 阻止了 NMDA 诱导的 DNA 断裂,并且 Tat-NR2B9c 可以显着减少神经元死亡。 Tat-NR2B9c 还通过阻断磷酸化阻止 NMDA 诱导的超氧化物 p47phox 形成,Tat-NR2B9c 的神经保护作用可能部分或全部归因于其抑制 NOX2 激活。此外,靶向 PSD-95 的 PDZ 结构域的 Tat-NR2B9c 破坏了 NR2B 和 NOX2 之间的功能耦合。[2]

体内实验表明,Tat-NR2B9c 可有效治疗或预防围产期和新生儿缺氧缺血性脑损伤及其相关脑部疾病。结果表明,Tat-NR2B9c 在缺血前或缺血后给药可减少缺氧缺血性损伤引起的脑损伤,在缺血前或缺血后给药可改善 HI 后的神经行为结果。此外,Tat-NR2B9c 可能通过促进促生存信号传导和抑制促凋亡信号传导发挥神经保护作用。[1]

Chemical Properties

Cas No. 500992-11-0 SDF
别名 Tat-NR2Bct; NA-1
Canonical SMILES Tyr-Gly-Arg-Lys-Lys-Arg-Arg-Gln-Arg-Arg-Arg-Lys-Leu-Ser-Ser-Ile-Glu-Ser-Asp-Val
分子式 C105H188N42O30 分子量 2518.88
溶解度 Water : ≥ 50 mg/mL (19.85 mM) 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 0.397 mL 1.985 mL 3.97 mL
5 mM 0.0794 mL 0.397 mL 0.794 mL
10 mM 0.0397 mL 0.1985 mL 0.397 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

Tat-HA-NR2B9c attenuate oxaliplatin-induced neuropathic pain

Exp Neurol2019 Jan;311:80-87.PMID: 30253135DOI: 10.1016/j.expneurol.2018.09.014

Oxaliplatin is a commonly used chemotherapy drug, which can produce acute and chronic peripheral neurotoxicity. Currently, there is no good therapeutic drug in clinic. Excessive stimulation of N-methyl-d-aspartate receptors (NMDARs) is crucial for the transmission of pain signals. However, directly inhibiting NMDARs can cause severe side effects because they have key physiological functions in the Central nervous system (CNS). Several years ago, we prepared a polypeptide Tat-HA-NR2B9c which can disturb NMDARs-postsynaptic density protein-95 (PSD-95) interaction. In this study, we studied whether Tat-HA-NR2B9c could be an effective treatment for oxaliplatin-induced neuropathic pain. To conform it, a rat model of oxaliplatin-induced neuropathic was established, and analgesic effect of Tat-HA-NR2B9c was studied. Here, we show that oxaliplatin induces the interaction of NMDARs with PSD-95. Uncoupling the complex by Tat-HA-NR2B9c has potent analgesic effect in oxaliplatin-induced cold hyperalgesia and mechanical allodynia without suppressing general behavioral. Tat-HA-NR2B9c neither inhibits NMDARs function nor impacts antitumor activity of oxaliplatin. Thus, this new drug may serve as a treatment for oxaliplatin-induced neuropathic pain, perhaps without major side effects.

Tat-NR2B9c prevents excitotoxic neuronal superoxide production

J Cereb Blood Flow Metab2015 May;35(5):739-42.PMID: 25669908DOI: 10.1038/jcbfm.2015.16

The Tat-NR2B9c peptide has shown clinical efficacy as a neuroprotective agent in acute stroke. Tat-NR2B9c is designed to prevent nitric oxide (NO) production by preventing postsynaptic density protein 95 (PSD-95) binding to N-methyl-D-aspartate (NMDA) receptors and neuronal nitric oxide synthase; however, PSD-95 is a scaffolding protein that also couples NMDA receptors to other downstream effects. Here, using neuronal cultures, we show that Tat-NR2B9c also prevents NMDA-induced activation of neuronal NADPH oxidase, thereby blocking superoxide production. Given that both superoxide and NO are required for excitotoxic injury, the neuroprotective effect of Tat-NR2B9c may alternatively be attributable to uncoupling neuronal NADPH oxidase from NMDA receptor activation.

Conjugation of Therapeutic PSD-95 Inhibitors to the Cell-Penetrating Peptide Tat Affects Blood-Brain Barrier Adherence, Uptake, and Permeation

Pharmaceutics2020 Jul 14;12(7):661.PMID: 32674358DOI: 10.3390/pharmaceutics12070661

Novel stroke therapies are needed. Inhibition of the interaction between the postsynaptic density-95 (PSD-95)/disc large/ZO-1 (PDZ) domains of PSD-95 and the N-methyl-D-aspartate (NMDA) receptor has been suggested as a strategy for relieving neuronal damage. The peptides NR2B9c and N-dimer have been designed to hinder this interaction; they are conjugated to the cell-penetrating peptide Tat to facilitate blood-brain barrier (BBB) permeation and neuronal uptake. Tat-N-dimer exhibits 1000-fold better target affinity than Tat-NR2B9c, but the same magnitude of improvement is not observed in terms of therapeutic effect. Differences in BBB permeation by Tat-NR2B9c and Tat-N-dimer may explain this difference, but studies providing a direct comparison of Tat-NR2B9c and Tat-N-dimer are lacking. The aim of the present study was therefore to compare the BBB uptake and permeation of Tat-NR2B9c and Tat-N-dimer. The peptides were conjugated to the fluorophore TAMRA and their chemical stability assessed. Endothelial membrane association and cell uptake, and transendothelial permeation were estimated using co-cultures of primary bovine brain capillary endothelial cells and rat astrocytes. In vivo BBB permeation was demonstrated in mice using two-photon microscopy imaging. Tissue distribution was evaluated in mice demonstrating brain accumulation of TAMRA-Tat (0.4% ID/g), TAMRA-Tat-NR2B9c (0.3% ID/g), and TAMRA-Tat-N-dimer (0.25% ID/g). In conclusion, we demonstrate that attachment of NR2B9c or N-dimer to Tat affects both the chemical stability and the ability of the resulting construct to interact with and permeate the BBB.

Efficacy of the PSD95 inhibitor Tat-NR2B9c in mice requires dose translation between species

J Cereb Blood Flow Metab2016 Mar;36(3):555-61.PMID: 26661213DOI: 10.1177/0271678X15612099

Tat-NR2B9c, a clinical-stage stroke neuroprotectant validated in rats and primates, was recently deemed ineffective in mice. To evaluate this discrepancy, we conducted studies in mice subjected to temporary middle cerebral artery occlusion (tMCAO) for either 30 or 60 min according to the established principles for dose-translation between species. Tat-NR2B9c treatment reduced infarct volume by by 24.5% (p = 0.49) and 26.0% (p = 0.03) for 30 and 60 min tMCAO, respectively, at the rat-equivalent dose of 10 nMole/g, but not at the previously reported 3 nMole/g in mice. Dose translation is thus critical when preclinical experiments are conducted in new species.

Delayed Administration of Tat-HA-NR2B9c Promotes Recovery After Stroke in Rats

Stroke2015 May;46(5):1352-8.PMID: 25851770DOI: 10.1161/STROKEAHA.115.008886

Background and purpose: Previous studies reported that Tat-NR2B9c, a peptide disrupting the N-methyl-d-aspartate receptor-postsynaptic density protein-95 interaction, reduced ischemic damage in the acute phase after stroke. However, its effect in the subacute phase is unknown. The aim of this study is to determine whether disrupting the N-methyl-d-aspartate receptor-postsynaptic density protein-95 interaction in the subacute phase promotes recovery after stroke.
Methods: Studies were performed on Sprague-Dawley rats or nNOS(-/-) mice, and experimental ischemic stroke was induced by middle cerebral artery occlusion. Animals were treated with drugs starting at day 4 after ischemia. Sensorimotor functions and spatial learning and memory ability were assessed after drug treatment. Then, rats were euthanized for morphological observation and biochemical tests.
Results: Disrupting the N-methyl-d-aspartate receptor-postsynaptic density protein-95 interaction with Tat-HA-NR2B9c significantly ameliorated the ischemia-induced impairments of spatial memory and sensorimotor functions in rats during subacute stage but did not improve stroke outcome in nNOS(-/-) mice. Consistent with the functional recovery, Tat-HA-NR2B9c substantially increased neurogenesis in the dentate gyrus and dendritic spine density of mature neurons in the motor cortex of rats, meanwhile, reversed the ischemia-induced formation of S-nitrosylation-cyclin-dependent kinase 5 and increased cyclin-dependent kinase 5 activity in ipsilateral hippocampus. However, directly blocking N-methyl-d-aspartate receptors with MK-801 or Ro 25-6981 did not show the beneficial effects above.
Conclusions: Dissociating N-methyl-d-aspartate receptor-postsynaptic density protein-95 coupling by Tat-HA-NR2B9c in the subacute phase after stroke promotes functional recovery, probably because of that it increases neurogenesis and dendritic spine density of mature neurons via regulating cyclin-dependent kinase 5 in the ischemic brain.