Home>>Signaling Pathways>> Metabolism>> Ferroptosis>>Ferrostatin-1 (Fer-1)

Ferrostatin-1 (Fer-1) Sale

(Synonyms: Fer-1) 目录号 : GC10380

一种铁死亡抑制剂

Ferrostatin-1 (Fer-1) Chemical Structure

Cas No.:347174-05-4

规格 价格 库存 购买数量
10mM (in 1mL DMSO)
¥473.00
现货
5mg
¥326.00
现货
25mg
¥1,197.00
现货
50mg
¥2,265.00
现货
100mg
¥4,116.00
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.

136

客户使用产品发表文献 35

质量管理

产品文档

Quality Control & SDS

View current batch:

实验参考方法

Cell experiment [1]:

Cell lines

SH-S5Y

Preparation Method

Soluble in DMSO (up to 100 mg/ml) or in Ethanol (up to 100 mg/ml).

Reaction Conditions

1 μM, 24 h

Applications

Ferrostatin-1 was able to inhibit a non-apoptotic cell death named ferroptosis. The neuroprotective role of ferrostatin-1 under rotenone-induced oxidative stress in dopaminergic neuroblastoma cells (SH-SY5Y) was reported. Besides, Ferrostatin-1 inhibited the ROS/RNS generated under rotenone insult in SH-SY5Y cells.

Animal experiment [2]:

Animal models

C57BL/6 (ICH, Intracerebral hemorrhage)

Preparation Method

10 μM Ferrostatin-1 in 1 μl 0.01% DMSO in saline

Dosage form

1 pmol of Ferrostatin-1, collagenase injection

Applications

Ferrostatin-1, a specific inhibitor of ferroptosis, was Administrated to prevent neuronal death and reduced iron deposition induced by hemoglobin in organotypic hippocampal slice cultures (OHSCs). Mice treated with ferrostatin-1 after ICH exhibited marked brain protection and improved neurologic function. Additionally, Ferrostatin-1 reduced lipid reactive oxygen species production and attenuated the increased expression level of PTGS2 and its gene product cyclooxygenase-2 ex vivo and in vivo.

References:

[1]. Kabiraj P, et al. The neuroprotective role of ferrostatin-1 under rotenone-induced oxidative stress in dopaminergic neuroblastoma cells. Protein J. 2015 Oct;34(5):349-58.

[2]. Li Q, et al. Inhibition of neuronal ferroptosis protects hemorrhagic brain. JCI Insight. 2017 Apr 6;2(7):e90777.

产品描述

Ferrostatin-1 is a potent inhibitor of ferroptosis with an EC50 of 60 nM.

Ferrostatin-1, as an molecular inhibitor, can block ferroptosis. Ferrostatins are believed to act by preventing oxidative damage to membrane lipids. Ferrostatin-1, an arylalkylamine with antioxidative properties, was identified as one of the first inhibitors of ferroptosis. Ferrostatin-1 attenuates oxidative, iron-dependent cell death in cancer cells treated with small molecules such as erastin. [3]

Ferrostatin-1, an inhibitor of ferroptosis, has an EC50 of 60 nM (HT-1080). Besides, Ferrostatin-1 was able to inhibit a non-apoptotic cell death named ferroptosis. The neuroprotective role of Ferrostatin-1 under rotenone-induced oxidative stress in dopaminergic neuroblastoma cells (SH-SY5Y) was reported. Ferrostatin-1 inhibited the ROS/RNS generated under rotenone insult in SH-SY5Y cells. The effective role of Ferrostatin-1 in ER stress mediated activation of apoptotic pathway was confirmed. Additionally, Ferrostatin-1 mitigated rotenone-induced α-syn aggregation was also reported.[1]

Ferrostatin-1, a specific inhibitor of ferroptosis, was Administrated to prevent neuronal death and reduced iron deposition induced by hemoglobin in organotypic hippocampal slice cultures (OHSCs). Mice treated with ferrostatin-1 after ICH exhibited marked brain protection and improved neurologic function. Additionally, Ferrostatin-1 reduced lipid reactive oxygen species production and attenuated the increased expression level of PTGS2 and its gene product cyclooxygenase-2 ex vivo and in vivo. For in vivo experiments, Ferrostatin-1 was injected 1 pmol (10 μM Ferrostatin-1 in 1 μl 0.01% DMSO in saline) into the striatum immediately after collagenase injection or into the cerebral ventricle 2 hours after collagenase injection. The coordinates for cerebral ventricle injection were: 1.0 mm lateral, 0.5 mm posterior, and 2.5 mm in depth relative to the bregma. [2]

References:
[1] Kabiraj P, et al. The neuroprotective role of ferrostatin-1 under rotenone-induced oxidative stress in dopaminergic neuroblastoma cells. Protein J. 2015 Oct;34(5):349-58.
[2] Li Q, et al. Inhibition of neuronal ferroptosis protects hemorrhagic brain. JCI Insight. 2017 Apr 6;2(7):e90777. doi: 10.1172/jci.insight.90777.
[3] Hofmans S, et al. Novel Ferroptosis Inhibitors with Improved Potency and ADME Properties. J Med Chem. 2016 Mar 10;59(5):2041-53.

Ferrostatin-1是一种有效的铁死亡抑制剂,EC50为60纳摩尔。

Ferrostatin-1是一种分子抑制剂,可以阻止铁死亡。相信Ferrostatins通过防止膜脂质的氧化损伤来发挥作用。 Ferrostatin-1是一种具有抗氧化性能的芳基烷胺,被认为是铁死亡的第一个抑制剂之一。在使用小分子如erastin处理癌细胞时,Ferrostatin-1减轻了氧化、依赖于铁离子的细胞死亡。[3]

Ferrostatin-1是一种抑制铁死亡的药物,其EC50为60 nM(HT-1080)。此外,Ferrostatin-1还能够抑制一种非凋亡性细胞死亡方式——铁死亡。报道了在罗滕酮诱导氧化应激下对多巴胺能神经母细胞瘤细胞(SH-SY5Y)中Ferrostatin-1的神经保护作用。Ferrostatin-1可以抑制SH-SY5Y细胞在罗滕酮刺激下产生的ROS/RNS。确认了Ferrostatin-1在内质网应激介导的凋亡通路激活中的有效作用。此外,报道了Ferrostatin-1缓解罗滕酮诱导α-syn聚集的作用。[1]

Ferrostatin-1是一种特定的铁死亡抑制剂,被用来预防血红蛋白诱导的神经元死亡和减少器官培养海马切片中的铁沉积。给予Ferrostatin-1治疗后,ICH小鼠表现出明显的脑保护作用和改善神经功能。此外,Ferrostatin-1还能够减少脂质活性氧产生,并在体内外减弱PTGS2及其基因产物环氧合酶2表达水平的增加。对于体内实验,将Ferrostatin-1注射到胶原酶注射后立即进入纹状体或在胶原酶注射后2小时注入侧脑室中。侧脑室注射坐标为:相对于bregma点位移动 1.0 mm 横向、0.5 mm 后方、深度为 2.5 mm 。[2]

参考文献:
[1] Kabiraj P等。铁死亡素-1在罗滕酮诱导的多巴胺能神经母细胞瘤细胞氧化应激下的神经保护作用。蛋白质学杂志,2015年10月;34(5):349-58。
[2] Li Q等。抑制神经元铁死亡可保护出血性脑损伤。JCI Insight. 2017年4月6日;2(7):e90777. doi: 10.1172/jci.insight.90777。
[3] Hofmans S等。具有改进效力和ADME特性的新型铁死亡素抑制剂。医药化学杂志,2016年3月10日;59(5):2041-53。

Chemical Properties

Cas No. 347174-05-4 SDF
别名 Fer-1
Canonical SMILES NC1=C(NC2CCCCC2)C=CC(C(OCC)=O)=C1
分子式 C15H22N2O2 分子量 262.35
溶解度 125mg/ml in DMSO 储存条件 4°C, protect from light
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 3.8117 mL 19.0585 mL 38.117 mL
5 mM 0.7623 mL 3.8117 mL 7.6234 mL
10 mM 0.3812 mL 1.9059 mL 3.8117 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

Ferrostatin-1 alleviates lipopolysaccharide-induced acute lung injury via inhibiting ferroptosis

Background: Ferroptosis is a newly recognized type of cell death, which is different from traditional necrosis, apoptosis or autophagic cell death. However, the position of ferroptosis in lipopolysaccharide (LPS)-induced acute lung injury (ALI) has not been explored intensively so far. In this study, we mainly analyzed the relationship between ferroptosis and LPS-induced ALI. Methods: In this study, a human bronchial epithelial cell line, BEAS-2B, was treated with LPS and ferrostatin-1 (Fer-1, ferroptosis inhibitor). The cell viability was measured using CCK-8. Additionally, the levels of malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), and iron, as well as the protein level of SLC7A11 and GPX4, were measured in different groups. To further confirm the in vitro results, an ALI model was induced by LPS in mice, and the therapeutic action of Fer-1 and ferroptosis level in lung tissues were evaluated. Results: The cell viability of BEAS-2B was down-regulated by LPS treatment, together with the ferroptosis markers SLC7A11 and GPX4, while the levels of MDA, 4-HNE and total iron were increased by LPS treatment in a dose-dependent manner, which could be rescued by Fer-1. The results of the in vivo experiment also indicated that Fer-1 exerted therapeutic action against LPS-induced ALI, and down-regulated the ferroptosis level in lung tissues. Conclusions: Our study indicated that ferroptosis has an important role in the progression of LPS-induced ALI, and ferroptosis may become a novel target in the treatment of ALI patients.

Ferrostatin-1 alleviates lipopolysaccharide-induced cardiac dysfunction

Cardiac dysfunction is a common complication of sepsis, and is attributed to severe inflammatory responses. Ferroptosis is reported to be involved in sepsis-induced cardiac inflammation. Therefore, we speculated that ferrostatin-1 (Fer-1), a ferroptosis inhibitor, improves cardiac dysfunction caused by sepsis. An intraperitoneal injection of lipopolysaccharide (LPS) was performed to induce a rat cardiac dysfunction model. Echocardiography, cardiac histopathology, biochemical and western blot results were analyzed. Twelve hours after the LPS injection, LPS-treated rats exhibited deteriorating cardiac systolic function, increased levels of cardiac injury markers and levels of ferroptosis markers prostaglandin endoperoxide synthase 2 (PTGS2). Additionally, LPS increased iron deposition in the myocardium, with downregulating ferroportin (FPN, SLC40A1) and transferrin receptor (TfR)expression, and upregulating ferritin light chain (FTL) and ferritin heavy chain (FTH1) expression. Meanwhile, LPS also increased lipid peroxidation in the rat heart by decreasing the expression of glutathione peroxidase 4 (GPX4). Moreover, the expression of inflammatory cytokines, such as tumor necrosis-alpha (TNF-α), interleukin-1 (IL-1β), and interleukin-6 (IL-6), and inflammatory cell infiltration were also increased following LPS challenge. Finally, the abovementioned adverse effects of LPS were relieved by Fer-1 except for TfR expression. Mechanistically, Fer-1 significantly reduced the levels of toll-like receptor 4 (TLR4), phospho-nuclear factor kappa B (NF-κB), and phospho-inhibitor of kappa Bα (IκBα) in LPS-treated rats. In summary, these findings imply that Fer-1 improved sepsis-induced cardiac dysfunction at least partially via the TLR4/NF-κB signaling pathway.

Insight into the mechanism of ferroptosis inhibition by ferrostatin-1

Ferroptosis is a form of cell death primed by iron and lipid hydroperoxides and prevented by GPx4. Ferrostatin-1 (fer-1) inhibits ferroptosis much more efficiently than phenolic antioxidants. Previous studies on the antioxidant efficiency of fer-1 adopted kinetic tests where a diazo compound generates the hydroperoxyl radical scavenged by the antioxidant. However, this reaction, accounting for a chain breaking effect, is only minimally useful for the description of the inhibition of ferrous iron and lipid hydroperoxide dependent peroxidation. Scavenging lipid hydroperoxyl radicals, indeed, generates lipid hydroperoxides from which ferrous iron initiates a new peroxidative chain reaction. We show that when fer-1 inhibits peroxidation, initiated by iron and traces of lipid hydroperoxides in liposomes, the pattern of oxidized species produced from traces of pre-existing hydroperoxides is practically identical to that observed following exhaustive peroxidation in the absence of the antioxidant. This supported the notion that the anti-ferroptotic activity of fer-1 is actually due to the scavenging of initiating alkoxyl radicals produced, together with other rearrangement products, by ferrous iron from lipid hydroperoxides. Notably, fer-1 is not consumed while inhibiting iron dependent lipid peroxidation. The emerging concept is that it is ferrous iron itself that reduces fer-1 radical. This was supported by electroanalytical evidence that fer-1 forms a complex with iron and further confirmed in cells by fluorescence of calcein, indicating a decrease of labile iron in the presence of fer-1. The notion of such as pseudo-catalytic cycle of the ferrostatin-iron complex was also investigated by means of quantum mechanics calculations, which confirmed the reduction of an alkoxyl radical model by fer-1 and the reduction of fer-1 radical by ferrous iron. In summary, GPx4 and fer-1 in the presence of ferrous iron, produces, by distinct mechanism, the most relevant anti-ferroptotic effect, i.e the disappearance of initiating lipid hydroperoxides.

Ferritinophagy is involved in the zinc oxide nanoparticles-induced ferroptosis of vascular endothelial cells

Zinc oxide nanoparticles (ZnONPs) hold great promise for biomedical applications. Previous studies have revealed that ZnONPs exposure can induce toxicity in endothelial cells, but the underlying mechanisms have not been fully elucidated. In this study, we report that ZnONPs can induce ferroptosis of both HUVECs and EA.hy926 cells, as evidenced by the elevation of intracellular iron levels, lipid peroxidation and cell death in a dose- and time-dependent manner. In addition, both the lipid reactive oxygen species (ROS) scavenger ferrostatin-1 and the iron chelator deferiprone attenuated ZnONPs-induced cell death. Intriguingly, we found that ZnONPs-induced ferroptosis is macroautophagy/autophagy-dependent, because the inhibition of autophagy with a pharmacological inhibitor or by ATG5 gene knockout profoundly mitigated ZnONPs-induced ferroptosis. We further demonstrated that NCOA4 (nuclear receptor coactivator 4)-mediated ferritinophagy (autophagic degradation of the major intracellular iron storage protein ferritin) was required for the ferroptosis induced by ZnONPs, by showing that NCOA4 knockdown can reduce the intracellular iron level and lipid peroxidation, and subsequently alleviate ZnONPs-induced cell death. Furthermore, we showed that ROS originating from mitochondria (mtROS) probably activated the AMPK-ULK1 axis to trigger ferritinophagy. Most importantly, pulmonary ZnONPs exposure caused vascular inflammation and ferritinophagy in mice, and ferrostatin-1 supplementation significantly reversed the vascular injury induced by pulmonary ZnONPs exposure. Overall, our study indicates that ferroptosis is a novel mechanism for ZnONPs-induced endothelial cytotoxicity, and that NCOA4-mediated ferritinophagy is required for ZnONPs-induced ferroptotic cell death.Abbreviations: 3-MA: 3-methyladenine; ACTB: Actin beta; AMPK: AMP-activated protein kinase; ATG: Autophagy-related; BafA1: Bafilomycin A1; CQ: Choloroquine; DFP: Deferiprone; FACS: Fluorescence-activated cell sorting; Fer-1: Ferrostatin-1; FTH1: Ferritin heavy chain 1; GPX4: Glutathione peroxidase 4; GSH: Glutathione; IREB2/IRP2: Iron responsive element binding protein 2; LIP: Labile iron pool; MAP1LC3B/LC3B: Microtubule associated protein 1 light chain 3 beta; MTOR: Mechanistic target of rapamycin kinase; NCOA4: Nuclear receptor coactivator 4; NFE2L2/NRF2: Nuclear factor, erythroid 2 like 2; PGSK: Phen Green? SK; ROS: Reactive oxygen species; siRNA: Small interfering RNA; SQSTM1/p62: Sequestosome 1; TEM: Transmission electron microscopy; ULK1: Unc-51 like autophagy activating kinase 1; ZnONPs: Zinc oxide nanoparticles.

Ferritinophagy-mediated ferroptosis is involved in sepsis-induced cardiac injury

Ferroptosis is a reactive oxygen species (ROS)- and iron-dependent form of regulated cell death (RCD), playing critical roles in organ injury and targeting therapy of cancers. Previous studies have demonstrated that ferroptosis participates in the development of cardiomyopathy including cardiac hypertrophy, diabetic cardiomyopathy and doxorubicin-induced cardiotoxicity. However, the role of ferroptosis in sepsis-induced cardiac injury remains unclear. This study aimed to explore the role and underlying mechanism of ferroptosis on lipopolysaccharide (LPS)-induced cardiac injury. Mice were injected with LPS (10 mg/kg) for 12 h to generate experimental sepsis. Ferrostatin-1 (Fer-1) and Dexrazoxane (DXZ) were used to suppress ferroptosis of mice with sepsis-induced cardiac injury. LPS increased the levels of ferroptotic markers involving prostaglandin endoperoxide synthase 2 (PTGS2), malonaldehyde (MDA) and lipid ROS, apart from resulting in obvious mitochondria damage, which were alleviated by Fer-1 and DXZ. In vitro experiments showed that Fer-1 inhibited LPS-induced lipid peroxidation and injury of H9c2 myofibroblasts while erastin and sorafenib aggravated LPS-induced ferroptosis. Additionally, Fer-1 and DXZ improved survival rate and cardiac function of mice with sepsis. Mechanistically, LPS increased the expression of nuclear receptor coactivator 4 (NCOA4) and the level of intracellular Fe2+ but decreased the level of ferritin. NCOA4 could directly interact with ferritin and degrade it in a ferritinophagy-dependent manner, which subsequently released a great amount of iron. Cytoplasmic Fe2+ further activated the expression of siderofexin (SFXN1) on mitochondrial membrane, which in turn transported cytoplasmic Fe2+ into mitochondria, giving rise to the production of mitochondrial ROS and ferroptosis. Based on these findings, we concluded that ferritinophagy-mediated ferroptosis is one of the critical mechanisms contributing to sepsis-induced cardiac injury. Targeting ferroptosis in cardiomyocytes may be a therapeutic strategy for preventing sepsis in the future.