Home>>Peptides>> Amyloid β protein>>Amyloid Beta-peptide (25-35) (human)

Amyloid Beta-peptide (25-35) (human) Sale

(Synonyms: BETA-淀粉样蛋白片断25-35,Gly-Ser-Asn-Lys-Gly-Ala-Ile-Ile-Gly-Leu-Met ) 目录号 : GP10082

淀粉样蛋白β肽(Aβ) (25-35)(human)是阿尔茨海默病淀粉样蛋白β肽的片段,具有神经毒性作用。

Amyloid Beta-peptide (25-35) (human) Chemical Structure

Cas No.:131602-53-4

规格 价格 库存 购买数量
1mg
¥431.00
现货
5mg
¥1,250.00
现货
10mg
¥2,184.00
现货
25mg
¥3,014.00
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

细胞实验 [1]:

细胞系

PC12 cells

实验方法

Amyloid-β-(25-35)聚集体的制备:将Amyloid-β-(25-35)溶解于浓度为1mM的去离子水中,过滤灭菌,37℃孵育4天诱导聚集体,-20℃保存备用。

将细胞以2个×104细胞/孔的密度接种到96孔培养板上。37℃稳定48h后,游离血清培养基中加入终浓度为20 μM的 Amyloid-β-(25-35)再培养24h。

实验条件

20 μM; 24 hours

应用

20 μM Amyloid-β-(25-35)处理PC12细胞24 h后,细胞活力降至对照组(100%)的67%,产生细胞毒性。

动物实验 [2]:

动物品系

Sprague-Dawley的雄性老鼠

实验方法

以15 nmol/5 μl剂量注射 Amyloid Beta-peptide (25-35) (human) 8 天后,测定足电刺激下大鼠的退缩、发声和跳跃阈值。

实验条件

15 nmol/5 μl; i.c.v.;8days

应用

单次脑室内注射15 nmol/大鼠 剂量的 Amyloid Beta-peptide (25-35) (human) 可显著降低步进被动回避任务的潜伏期。

References:

[1]. Xian YF, Lin ZX, et,al. Protective effect of isorhynchophylline against β-amyloid-induced neurotoxicity in PC12 cells. Cell Mol Neurobiol. 2012 Apr;32(3):353-60. doi: 10.1007/s10571-011-9763-5. Epub 2011 Nov 1. PMID: 22042506.

[2]. Yamaguchi Y, Kawashima S. Effects of amyloid-beta-(25-35) on passive avoidance, radial-arm maze learning and choline acetyltransferase activity in the rat. Eur J Pharmacol. 2001 Feb 2;412(3):265-72. doi: 10.1016/s0014-2999(01)00730-0. PMID: 11166290.

产品描述

Amyloid beta(Aβ)-peptide (25-35) (human) is an fragment of Alzheimer's Amyloid beta peptide which is commonly found in the brains of people with Alzheimer's disease (AD) and is a major component of Alzheimer's amyloid plaques[1-3]. Amyloid beta-peptide (25-35) (human) is considered a functional domain of Aβ due to its neurotoxic effects, and it is the bioactive region of beta-amyloid[4].

Amyloid beta-peptide (25-35) (human) treatment (20 μM, 6 hours) activated tau protein kinase I/glycogen synthase kinase-3beta (TPKI/GSK-3beta) in primary culture of hippocampal neurons [5]. The Amyloid beta-peptide (25-35) (human) (100nM, 1μM, 10μM, or 30μM) reduces the viability of cultured cerebral cortical rat neurons [6]. Amyloid beta-peptide (25-35) (human) (10μM) increased the amplitude of excitatory responses produced by local iontophoretic applications of glutamate and NMDA [7]. Treatment of PC12 cells with 20 μM of Amyloid beta-peptide (25-35) (human) for 24 h induced cytotoxicity as the cell viability was reduced to 67% of the control value (100%)[8].

A single intracerebroventricular (i.c.v.) injection of Amyloid beta-peptide (25-35) (human) (15 nmol/5 μl; i.c.v.;8days) at a dose of 15 nmol/rat induced a marked decrease in latency in step-through passive avoidance task[9].

References:

[1]. Nalbantoglu J. Beta-amyloid protein in Alzheimer's disease. Can J Neurol Sci. 1991 Aug;18(3 Suppl):424-7. doi: 10.1017/s0317167100032595. PMID: 1933692.

[2]. Rush DK, Aschmies S, et,al. Intracerebral beta-amyloid(25-35) produces tissue damage: is it neurotoxic? Neurobiol Aging. 1992 Sep-Oct;13(5):591-4. doi: 10.1016/0197-4580(92)90061-2. PMID: 1281289.

[3]. Mattson MP, Cheng B, et,al. beta-Amyloid peptides destabilize calcium homeostasis and render human cortical neurons vulnerable to excitotoxicity. J Neurosci. 1992 Feb;12(2):376-89. doi: 10.1523/JNEUROSCI.12-02-00376.1992. PMID: 1346802; PMCID: PMC6575616.

[4]. D'Errico, G; Vitiello, G; et,al. Interaction between Alzheimer's A(25-35) peptide and phospholipid bilayers: The role of cholesterol. Biochimica. Biophys. Acta (BBA) – Biomembr., 2008, 1778, 2710-2716.

[5]. Takashima A, Honda T, et,al. Activation of tau protein kinase I/glycogen synthase kinase-3beta by amyloid beta peptide (25-35) enhances phosphorylation of tau in hippocampal neurons. Neurosci Res. 1998 Aug;31(4):317-23. doi: 10.1016/s0168-0102(98)00061-3. PMID: 9809590.

[6]. Wang Y, Liu L, et,al. Mechanism of soluble beta-amyloid 25-35 neurotoxicity in primary cultured rat cortical neurons. Neurosci Lett. 2016 Apr 8;618:72-76. doi: 10.1016/j.neulet.2016.02.050. Epub 2016 Mar 3. PMID: 26940239.

[7]. Carette B, Poulain P, et,al. Electrophysiological effects of 25-35 amyloid-beta-protein on guinea-pig lateral septal neurons. Neurosci Lett. 1993 Mar 5;151(1):111-4. doi: 10.1016/0304-3940(93)90059-t. PMID: 8385758.

[8]. Xian YF, Lin ZX, Mao QQ, Ip SP, Su ZR, Lai XP. Protective effect of isorhynchophylline against β-amyloid-induced neurotoxicity in PC12 cells. Cell Mol Neurobiol. 2012 Apr;32(3):353-60. doi: 10.1007/s10571-011-9763-5. Epub 2011 Nov 1. PMID: 22042506.

[9]. Yamaguchi Y, Kawashima S. Effects of amyloid-beta-(25-35) on passive avoidance, radial-arm maze learning and choline acetyltransferase activity in the rat. Eur J Pharmacol. 2001 Feb 2;412(3):265-72. doi: 10.1016/s0014-2999(01)00730-0. PMID: 11166290.

淀粉样蛋白β (Aβ)肽(25-35)(human)是阿尔茨海默病淀粉样蛋白β肽的片段,通常存在于阿尔茨海默病(AD)患者的大脑中,是阿尔茨海默病淀粉样斑块的主要成分[1-3]。淀粉样蛋白β 肽(25-35)(human)由于其神经毒性作用被认为是Aβ的一个功能域,它是β -淀粉样蛋白的生物活性区域[4]

淀粉样蛋白β 肽(25-35)(human)处理(20 μM, 6h)激活了海马神经元原代培养的tau蛋白激酶I/糖原合成酶激酶3 β(TPKI/GSK-3beta) [5]。淀粉样蛋白β 肽(25-35)(human) (100nM, 1μM, 10μM或30μM)降低培养的大鼠大脑皮质神经元的活力[6]。淀粉样蛋白β 肽(25-35)(human) (10μM) 增加了局部应用谷氨酸和NMDA产生的兴奋反应的振幅[7]。20 μM淀粉样蛋白β 肽(25-35)(human) 处理PC12细胞24 h后,细胞活力降至对照组(100%)的67%,产生细胞毒性[8]

淀粉样蛋白β 肽(25-35)(human) (15 nmol/5 μl; i.c.v.;8days )可显著降低步进被动回避任务的潜伏期[9]

Chemical Properties

Cas No. 131602-53-4 SDF
别名 BETA-淀粉样蛋白片断25-35,Gly-Ser-Asn-Lys-Gly-Ala-Ile-Ile-Gly-Leu-Met
化学名 Amyloid Beta-peptide (25-35) (human)
Canonical SMILES CCC(C)C(C(=O)NC(C(C)CC)C(=O)NCC(=O)NC(CC(C)C)C(=O)NC(CCSC)C(=O)O)NC(=O)C(C)NC(=O)CNC(=O)C(CCCCN)NC(=O)C(CC(=O)N)NC(=O)C(CO)NC(=O)CN
分子式 C45H81N13O14S 分子量 1060.27
溶解度 ≥ 106mg/mL in DMSO 储存条件 Desiccate 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 0.9432 mL 4.7158 mL 9.4316 mL
5 mM 0.1886 mL 0.9432 mL 1.8863 mL
10 mM 0.0943 mL 0.4716 mL 0.9432 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

Epigenetic axis of SNHG19/miR-137/TNFAIP1 modulates amyloid beta peptide 25-35-induced SH-SY5Y cytotoxicity

Aims: In this study, the authors hypothesized that, in an in vitro Alzheimer's disease model, the epigenetic axis of SNHG19/hsa-miR-137 functionally regulates amyloid beta peptide 25-35 (Aβ25-35)-induced SH-SY5Y cytotoxicity. Methods: Dual luciferase activity assay demonstrated that SNHG19 could directly bind hsa-miR-137. In Aβ25-35-treated SH-SY5Y cells, SNHG19 was upregulated and hsa-miR-137 downregulated. Results: SNHG19 knockdown ameliorated Aβ25-35-induced SH-SY5Y cytotoxicity, then reversed by secondary hsa-miR-137 downregulation. TNFAIP1 was dynamically regulated by Aβ25-35 and gene modifications in SH-SY5Y cells. Finally, upregulation of TNFAIP1 reversed the protective effect of SNHG19 knockdown on Aβ25-35-induced cytotoxicity. Conclusions: The authors concluded that the epigenetic axis of SNHG19/hsa-miR-137/TNFAIP1 may functionally regulate Aβ25-35-induced SH-SY5Y cytotoxicity, thus making it a potential molecular target for Alzheimer's disease treatment.

Tachykinin Neuropeptides and Amyloid β (25-35) Assembly: Friend or Foe?

Amyloid β (Aβ) protein is responsible for Alzheimer's disease, and one of its important fragments, Aβ(25-35), is found in the brain and has been shown to be neurotoxic. Tachykinin neuropeptides, including Neuromedin K (NK), Kassinin, and Substance P, have been reported to reduce Aβ(25-35)'s toxicity in cells even though they share similar primary structures with Aβ(25-35). Here, we seek to understand the molecular mechanisms of how these peptides interact with Aβ(25-35) and to shed light on why some peptides with similar primary structures are toxic and others nontoxic. We use both experimental and computational methods, including ion mobility mass spectrometry and enhanced-sampling replica-exchange molecular dynamics simulations, to study the aggregation pathways of Aβ(25-35), NK, Kassinin, Substance P, and mixtures of the latter three with Aβ(25-35). NK and Substance P were observed to remove the higher-order oligomers (i.e., hexamers and dodecamers) of Aβ(25-35), which are related to its toxicity, although Substance P did so more slowly. In contrast, Kassinin was found to promote the formation of these higher-order oligomers. This result conflicts with what is expected and is elaborated on in the text. We also observe that even though they have significant structural homology with Aβ(25-35), NK, Kassinin, and Substance P do not form hexamers with a β-sheet structure like Aβ(25-35). The hexamer structure of Aβ(25-35) has been identified as a cylindrin, and this structure has been strongly correlated to toxic species. The reasons why the three tachykinin peptides behave so differently when mixed with Aβ(25-35) are discussed.

Curcumin and Homotaurine Suppress Amyloid-β25-35 Aggregation in Synthetic Brain Membranes

Amyloid-β (Aβ) peptides spontaneously aggregate into β- and cross-β-sheets in model brain membranes. These nanometer sized can fuse into larger micrometer sized clusters and become extracellular and serve as nuclei for further plaque and fibril growth. Curcumin and homotaurine represent two different types of Aβ aggregation inhibitors. While homotaurine is a peptic antiaggregant that binds to amyloid peptides, curcumin is a nonpeptic molecule that can inhibit aggregation by changing membrane properties. By using optical and fluorescent microscopy, X-ray diffraction, and UV-vis spectroscopy, we study the effect of curcumin and homotaurine on Aβ25-35 aggregates in synthetic brain membranes. Both molecules partition spontaneously and uniformly in membranes and do not lead to observable membrane defects or disruption in our experiments. Both curcumin and homotaurine were found to significantly reduce the number of small, nanoscopic Aβ aggregates and the corresponding β- and cross-β-sheet signals. While a number of research projects focus on potential drug candidates that target Aβ peptides directly, membrane-lipid therapy explores membrane-mediated pathways to suppress peptide aggregation. Based on the results obtained, we conclude that membrane active drugs can be as efficient as peptide targeting drugs in inhibiting amyloid aggregation in vitro.

Conformations and biological activities of amyloid beta peptide 25-35

Amyloid-beta (Abeta) peptide is commonly found in human Alzheimer's disease (AD) brain and is the main component of Alzheimer amyloid plaques. The predominant forms of Abeta in the human brain are Abeta(1-40) and Abeta(1-42), but Abeta(25-35) fragment, physiologically present in elderly people, is the more toxic region and has been recently found to play a relevant role in AD, due to its peculiar aggregation properties. In this work, we review the current understanding on the conformations and biological activity of Abeta(25-35) exploring aggregation, cytotoxic and neurodegenerative properties of this fundamental Abeta fragment, in order to provide an effective starting point to better approach a pathology spread and problematic as AD.

Structure of amyloid β25-35 in lipid environment and cholesterol-dependent membrane pore formation

The amyloid β (Aβ) peptide and its shorter variants, including a highly cytotoxic Aβ25-35 peptide, exert their neurotoxic effect during Alzheimer's disease by various mechanisms, including cellular membrane permeabilization. The intrinsic polymorphism of Aβ has prevented the identification of the molecular basis of Aβ pore formation by direct structural methods, and computational studies have led to highly divergent pore models. Here, we have employed a set of biophysical techniques to directly monitor Ca2+-transporting Aβ25-35 pores in lipid membranes, to quantitatively characterize pore formation, and to identify the key structural features of the pore. Moreover, the effect of membrane cholesterol on pore formation and the structure of Aβ25-35 has been elucidated. The data suggest that the membrane-embedded peptide forms 6- or 8-stranded β-barrel like structures. The 8-stranded barrels may conduct Ca2+ ions through an inner cavity, whereas the tightly packed 6-stranded barrels need to assemble into supramolecular structures to form a central pore. Cholesterol affects Aβ25-35 pore formation by a dual mechanism, i.e., by direct interaction with the peptide and by affecting membrane structure. Collectively, our data illuminate the molecular basis of Aβ membrane pore formation, which should advance both basic and clinical research on Alzheimer's disease and membrane-associated pathologies in general.