Home>>(Gly¹⁴)-Humanin (human)

(Gly¹⁴)-Humanin (human) Sale

目录号 : GA20196

护脑素既是细胞内蛋白又是分泌蛋白。

(Gly¹⁴)-Humanin (human) Chemical Structure

Cas No.:330936-70-4

规格 价格 库存 购买数量
0.5mg
¥2,880.00
现货
1mg
¥4,942.00
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

EMP(from Primary HUVECs )

Preparation Method

For cells receiving (Gly14)-Humanin (human), cells were pretreated with (Gly14)-Humanin (human) for 3 hours.

Reaction Conditions

1 µM (Gly14)-Humanin (human) for 3 hours

Applications

(Gly14)-Humanin (human) can attenuate HG-induced apoptosis of HUVECs cells by reducing the expression of Mir-155 in EMPs.

Animal experiment [2]:

Animal models

10-12-week-old adult male CD-1 mice weighing 25-30 g

Preparation Method

(Gly14)-Humanin (human) -treated groups were administered 0.1 µg of (Gly14)-Humanin (human) in 5 µL of saline intraventricularly (i.c.v., coordinates 1 mm posterior, 1 mm lateral, 2.5 mm deep to bregma) before TBI or 1 µg in 100 µL of saline intraperitoneally (i.p.) post TBI.

Dosage form

0.1 µg of (Gly14)-Humanin (human) in 5 µL saline intraperitoneally post TBI.

Applications

(Gly14)-Humanin (human) reduced TBI-induced cell damage.

References:

[1]: Shen MY, Wang M, et,al. [Gly14]-Humanin Ameliorates High Glucose-Induced Apoptosis by Inhibiting the Expression of MicroRNA-155 in Endothelial Microparticles. Diabetes Metab Syndr Obes. 2021 May 24;14:2335-2347. doi: 10.2147/DMSO.S306026. PMID: 34079312; PMCID: PMC8163639.
[2]: Wang T, Zhang L, et,al. [Gly14]-Humanin reduces histopathology and improves functional outcome after traumatic brain injury in mice. Neuroscience. 2013 Feb 12;231:70-81. doi: 10.1016/j.neuroscience.2012.11.019. Epub 2012 Nov 20. PMID: 23178909.

产品描述

Humanin is both an intracellular protein and a secreted protein. It is the first newly discovered peptide encoded in the mitochondrial genome, which exerts robust protective effects against a myriad of cytotoxic stimuli in many cell types[2]. Replacement of serine at position 14 of the HN peptide chain (Ser14) with glycine (Gly) leads to the formation of (Gly14)-Humanin (human), which has cytoprotective activity enhanced by 1000-fold when compared with that of Humanin[3].

(Gly14)-Humanin (human) reduced Dil-ox-LDL accumulation in the RAW 264.7 macrophages. (Gly14)-Humanin (human) could inhibit ox-LDL-induced macrophage-derived foam cell formation, which occurs because of a decrease in lipid uptake and an increase in cholesterol efflux from macrophage cells[7]. (Gly14)-Humanin (human) pretreatment inhibits HG-induced apoptosis of vascular endothelial cells by down-regulating miR-155, which was transferred to adjacent vascular endothelial cells via cell-derived EMPs. EMPs could serve as a potential tool for the specific delivery of miRNAs, contributing to a stronger therapeutic effect of (Gly14)-Humanin (human) in anti-diabetic vascular complications therapy[1]. (Gly14)-Humanin (human) could restore cathepsin D activity and protein level in HUVECs to repair lysosomal functional damage induced by Ox-LDL, further repairing Ox-LDL-induced autophagic damage in HUVECs[4].

Pretreatment with (Gly14)-Humanin (human) not only protected spatial memory but also rescued STAT3 from Aβ-induced disruption; and the neuroprotective effects of (Gly14)-Humanin (human) were effectively counteracted by genistein, a specific tyrosine kinase inhibitor. Sequence 31-35 in Aβ is the shortest active center responsible for the neurotoxicity of Aβ from molecule to behavior; and (Gly14)-Humanin (human) protects spatial learning and memory in rats against Aβ-induced insults; and probably involves the activation of tyrosine kinases and subsequent beneficial modulation of STAT3 and caspase3[5]. Mice administered (Gly14)-Humanin (human) intracerebroventricularly (i.c.v.) prior to TBI had decreased cells with plasmalemma permeability in the injured cortex and hippocampus, reduced brain lesion volume, improved motor performance and ameliorated performance in the Morris water maze test post TBI. Reduced lesion volume was also observed even when (Gly14)-Humanin (human) was administered intraperitoneally (i.p.) at 1h and 2h post TBI, and minor amelioration in motor and Morris water maze test deficits was also observed. (Gly14)-Humanin (human) treatment improved morphological and functional outcomes after TBI in mice and the protective effect of (Gly14)-Humanin (human) against TBI may be associated with down-regulating apoptosis and autophagy[6].

References:
[1]: Shen MY, Wang M, et,al. [Gly14]-Humanin Ameliorates High Glucose-Induced Apoptosis by Inhibiting the Expression of MicroRNA-155 in Endothelial Microparticles. Diabetes Metab Syndr Obes. 2021 May 24;14:2335-2347. doi: 10.2147/DMSO.S306026. PMID: 34079312; PMCID: PMC8163639.
[2]: Kim SJ, Mehta HH, et,al. Mitochondrial peptides modulate mitochondrial function during cellular senescence. Aging (Albany NY). 2018 Jun 10;10(6):1239-1256. doi: 10.18632/aging.101463. PMID: 29886458; PMCID: PMC6046248.
[3]: Hashimoto Y, Niikura T, et,al. A rescue factor abolishing neuronal cell death by a wide spectrum of familial Alzheimer's disease genes and Abeta. Proc Natl Acad Sci U S A. 2001 May 22;98(11):6336-41. doi: 10.1073/pnas.101133498. Erratum in: Proc Natl Acad Sci U S A 2001 Oct 23;98(22):12854. PMID: 11371646; PMCID: PMC33469.
[4]: Ding Y, Feng Y, et,al. [Gly14]-humanin restores cathepsin D function via FPRL1 and promotes autophagic degradation of Ox-LDL in HUVECs. Nutr Metab Cardiovasc Dis. 2020 Nov 27;30(12):2406-2416. doi: 10.1016/j.numecd.2020.07.022. Epub 2020 Jul 25. PMID: 32917500.
[5]: Yuan L, Liu XJ, et,al. [Gly14]-Humanin Protects Against Amyloid β Peptide-Induced Impairment of Spatial Learning and Memory in Rats. Neurosci Bull. 2016 Aug;32(4):374-82. doi: 10.1007/s12264-016-0041-x. Epub 2016 Jun 15. PMID: 27306655; PMCID: PMC5563781.
[6]: Wang T, Zhang L, et,al. [Gly14]-Humanin reduces histopathology and improves functional outcome after traumatic brain injury in mice. Neuroscience. 2013 Feb 12;231:70-81. doi: 10.1016/j.neuroscience.2012.11.019. Epub 2012 Nov 20. PMID: 23178909.
[7]: Zhu WW, Wang SR, et,al. Gly[14]-humanin inhibits ox-LDL uptake and stimulates cholesterol efflux in macrophage-derived foam cells. Biochem Biophys Res Commun. 2017 Jan 1;482(1):93-99. doi: 10.1016/j.bbrc.2016.10.138. Epub 2016 Nov 1. PMID: 27815075.

Humanin 既是细胞内蛋白又是分泌蛋白。它是第一个新发现的在线粒体基因组中编码的肽,对许多细胞类型的无数细胞毒性刺激具有强大的保护作用[2]。 HN 肽链 14 位丝氨酸 (Ser14) 被甘氨酸 (Gly) 取代导致 (Gly14)-Humanin (human) 的形成,与 Humanin 相比,其细胞保护活性增强了 1000 倍[3].

(Gly14)-Humanin(人类)减少了 RAW 264.7 巨噬细胞中的 Dil-ox-LDL 积累。 (Gly14)-Humanin(人)可抑制 ox-LDL 诱导的巨噬细胞衍生泡沫细胞形成,这是由于巨噬细胞脂质摄取减少和胆固醇流出增加所致[7] . (Gly14)-Humanin(人)预处理通过下调 miR-155 抑制 HG 诱导的血管内皮细胞凋亡,miR-155 通过细胞衍生的 EMP 转移到邻近的血管内皮细胞。 EMP 可作为特异性递送 miRNA 的潜在工具,有助于增强 (Gly14)-Humanin (human) 在抗糖尿病血管并发症治疗中的疗效[1]。 (Gly14)-Humanin (human) 可恢复 HUVECs 组织蛋白酶 D 活性和蛋白水平,修复 Ox-LDL 诱导的溶酶体功能损伤,进一步修复 Ox-LDL 诱导的 HUVECs 自噬损伤[4] .

用 (Gly14)-Humanin(人)预处理不仅可以保护空间记忆,还可以从 Aβ 诱导的破坏中拯救 STAT3; (Gly14)-Humanin(人)的神经保护作用被染料木黄酮(一种特定的酪氨酸激酶抑制剂)有效抵消。 Aβ中的31-35序列是负责Aβ从分子到行为的神经毒性的最短活性中心; (Gly14)-Humanin(人类)保护大鼠的空间学习和记忆免受 Aβ 诱导的损伤;并且可能涉及酪氨酸激酶的激活和随后对 STAT3 和 caspase3 的有益调节[5]。在 TBI 前脑室内 (i.c.v.) 施用 (Gly14)-Humanin(人)的小鼠在受损皮层和海马体中具有质膜通透性的细胞减少、脑损伤体积减小、运动性能改善以及 TBI 后莫里斯水迷宫测试中的性能改善。即使在 TBI 后 1 小时和 2 小时腹膜内 (i.p.) 施用 (Gly14)-Humanin(人)时,也观察到损伤体积减小,并且还观察到运动和莫里斯水迷宫测试缺陷的轻微改善。 (Gly14)-Humanin(人)治疗改善了小鼠 TBI 后的形态学和功能结果,并且(Gly14)-Humanin(人)对 TBI 的保护作用可能与下调细胞凋亡和自噬有关[6]< /sup>.

Chemical Properties

Cas No. 330936-70-4 SDF
分子式 C118H202N34O31S2 分子量 2657.25
溶解度 Soluble in DMSO 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 0.3763 mL 1.8816 mL 3.7633 mL
5 mM 0.0753 mL 0.3763 mL 0.7527 mL
10 mM 0.0376 mL 0.1882 mL 0.3763 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

Protection effect of [Gly14]-Humanin from apoptosis induced by high glucose in human umbilical vein endothelial cells

Diabetes Res Clin Pract2014 Dec;106(3):560-6.PMID: 25451915DOI: 10.1016/j.diabres.2014.09.020

Aims: Humanin (HN) is known for its anti-apoptotic functions in neuronal cells. In this study, we sought to investigate the protective effect of [Gly14]-Humanin (HNG) in high glucose (HG)-induced apoptosis of human umbilical vein endothelial cells (HUVECs).
Methods: 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to examine cell viability, DNA chromatin morphology was assessed using Hoechst 33342 staining, and the generation of intracellular reactive oxygen species (ROS) was assessed using the fluorescent probe dichlorofluorescein diacetate (DCFH-DA). The expression of poly ADP-ribose polymerase (PARP), the pro-apoptotic protein bax and the anti-apoptotic protein bcl-2 were examined using western blot analysis. The mRNA level of bax and bcl-2 were detected by quantitative Real-Time PCR.
Results: Compared with treatment with HG 72h, pretreatment with HNG for 3h significantly increased cell viability (P<0.001), reduced nuclear fluorescence of HUVECs (P<0.05), the levels of cleaved PARP (P<0.05), ROS formation (P<0.05) and the ratio of bax/bcl-2 (P<0.05) compared with treatment with HG for 72h. Quantitative Real-Time PCR showed that mRNA level of bax reduced (P<0.05) and mRNA level of bcl-2 increased (P<0.05) after pretreatment with HNG.
Conclusions: Our results imply that HNG can protect HUVECs from apoptosis induced by HG through the bax/bcl-2 pathway.

[Gly14]-Humanin Ameliorates High Glucose-Induced Apoptosis by Inhibiting the Expression of MicroRNA-155 in Endothelial Microparticles

Diabetes Metab Syndr Obes2021 May 24;14:2335-2347.PMID: 34079312DOI: 10.2147/DMSO.S306026

Background: Humanin, a newly emerging endogenously expressed cytoprotective peptide, has been shown to have anti-apoptotic properties effects by protecting neuronal cells injury. Endothelial microparticles (EMPs) are considered as vital mediators in intercellular communication. EMPs may regulate various physiological and pathological processes by transferring mRNAs and microRNAs (miRNAs) to recipient cells.
Methods: EMPs were isolated from human umbilical vein endothelial cells (HUVECs) by ultracentrifugation. EMPs were characterized by transmission electron microscopy and nanoparticle tracking analyses. Observation of EMPs uptake into HUVECs and the number of EMPs were realized by confocal microscopy. The expression of miR-155 was examined using real-time PCR. Cell apoptosis was examined by flow cytometry assay.
Results: We found that high glucose (HG) increased the number of EMPs and upregulated the expression of miR-155 contained within EMPs, which was mitigated by HNG pretreatment. miR-155 overexpression in EMPs reversed the effects of HNG pretreatment and increased apoptosis of target cells. Effects of HNG pretreatment on HG-treated endothelial cells (ECs) were mitigated after miR-155 mimic transfection into HUVECs while were augmented after miR-155 inhibitor transfection into HUVECs.
Conclusion: HNG inhibited HG-induced apoptosis of ECs and the effect of HNG may be mediated by inhibiting the transfer of EMPs miR-155 from HG-induced HUVECs to normal cells. This study provides a new direction for biological products related to humanin to treat vascular complications associated with all forms of diabetes mellitus.

[Gly14]-Humanin restores cathepsin D function via FPRL1 and promotes autophagic degradation of Ox-LDL in HUVECs

Nutr Metab Cardiovasc Dis2020 Nov 27;30(12):2406-2416.PMID: 32917500DOI: 10.1016/j.numecd.2020.07.022

Background and aim: Abnormal aggregation of oxidized low-density lipoprotein (Ox-LDL) in vascular endothelial cells (VECs) is one of the major pathological changes in atherosclerotic lesions. Our research aimed to assess the mechanism of humanin (HN) in promoting autophagic degradation of Ox-LDL in HUVECs.
Methods and results: Flow cytometry and lipid quantitation results showed that Ox-LDL caused lipid and cholesterol accumulation in HUVECs. Western blot results showed that Ox-LDL increased the expression of autophagy-related proteins P62 and LC3-II in a concentration-dependent manner. The cathepsin D activity assay showed that Ox-LDL inhibited the function of cathepsin D. HNG pretreatment reduced lipid and cholesterol aggregation in HUVECs induced by Ox-LDL, increased LC3-II protein level, decreased P62 protein content, and reversed Ox-LDL-induced cathepsin D functional impairment. Inhibition of the FPRL1 pathway by FPRL1 siRNA or the FPRL1-specific inhibitor Boc-MLF blocked all HNG's protective effects. These results indicate that HNG could restore cathepsin D activity and protein level in HUVECs to repair lysosomal functional damage induced by Ox-LDL, further repairing Ox-LDL-induced autophagic damage in HUVECs.
Conclusion: HNG restores the activity of Ox-LDL-induced damaged lysosomal enzyme cathepsin D through its membrane protein receptor FPRL1 to promote autophagic degradation of Ox-LDL in HUVECs.

[Gly14]-Humanin Prevents Lipid Deposition and Endothelial Cell Apoptosis in a Lectin-like Oxidized Low-density Lipoprotein Receptor-1-Dependent Manner

Lipids2019 Nov;54(11-12):697-705.PMID: 31574567DOI: 10.1002/lipd.12195

Oxidized low-density lipoprotein (Ox-LDL) may induce apoptosis and dysfunction of vascular endothelial cells, contributing to the initiation and development of atherosclerosis and lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) plays a central role in Ox-LDL uptake in the course of atherogenesis. Humanin (HN), a mitochondrial-derived peptide, was recently demonstrated to exert a protective role against endothelial dysfunction and Ox-LDL-induced progression of atherosclerosis. The HN analog HNGF6A (HNG) modulates cholesterol metabolism in macrophage RAW 264.7 cells. However, whether HNG affects Ox-LDL metabolism in endothelial cells is unknown. In this study, we investigated the effect of HNG on Ox-LDL accumulation in human umbilical vein endothelial cell (HUVEC) and its underlying mechanisms. HUVEC were preincubated with HNG for 1 h before addition of Ox-LDL. Total cholesterol content was measured by using a tissue total cholesterol assay kit and flow cytometry. Cell viability was measured by CCK8 assay. Protein content was examined by Western blot assays. Flow cytometry was used to identify apoptotic cells. Flow cytometry and tissue total cholesterol assays showed that HNG reduced Ox-LDL accumulation in HUVEC. In addition, HNG inhibited Ox-LDL-induced apoptosis of HUVEC. Western blot results showed that HNG reduced LOX-1 protein content. However, when LOX-1 was knocked down or inhibited, the effect of HNG in reducing Ox-LDL aggregation and apoptosis in HUVEC disappeared. Our study demonstrated that HNG reduces lipid aggregation and apoptosis in HUVEC in a LOX-1-dependent manner.

Protective effects of [Gly14]-Humanin on beta-amyloid-induced PC12 cell death by preventing mitochondrial dysfunction

Neurochem Int2010 Feb;56(3):417-23.PMID: 19941922DOI: 10.1016/j.neuint.2009.11.015

Mitochondrial dysfunction is a hallmark of beta-amyloid (Abeta)-induced neuronal toxicity in Alzheimer's disease (AD), and is considered as an early event in AD pathology. Humanin (HN) and its derivative, [Gly14]-Humanin (HNG), are known for their ability to suppress neuronal death induced by AD-related insults in vitro and in vivo. In the present study, we investigated the neuroprotective effects of HNG on Abeta(25-35)-induced toxicity and its potential mechanisms in PC12 cells. Exposure of PC12 cells to 25 microM Abeta(25-35) caused significant viability loss and cell apoptosis. In addition, decreased mitochondrial membrane potential and increased cytochrome c releases from mitochondria were also observed after Abeta(25-35) exposure. All these effects induced by Abeta(25-35) were markedly reversed by HNG. Pretreatment with 100 nM HNG 6h prior to Abeta(25-35) exposure significantly elevated cell viability, reduced Abeta(25-35)-induced cell apoptosis, stabilized mitochondrial membrane potential, and blocked cytochrome c release from mitochondria. Furthermore, HNG also ameliorated the Abeta(25-35)-induced Bcl-2/Bax ratio reduction and decreased caspase-3 activity in PC12 cells. These results demonstrate that HNG could attenuate Abeta(25-35)-induced PC12 cell injury and apoptosis by preventing mitochondrial dysfunction. Furthermore, these data suggest that mitochondria are involved in the protective effect of HNG against Abeta(25-35).