Home>>Signaling Pathways>> Metabolism>> Ferroptosis>>RSL3

RSL3 Sale

(Synonyms: (1S,3R)-RSL3) 目录号 : GC12431

谷胱甘肽过氧化物酶4抑制剂

RSL3 Chemical Structure

Cas No.:1219810-16-8

规格 价格 库存 购买数量
10mM (in 1mL DMSO)
¥1,295.00
现货
5mg
¥893.00
现货
10mg
¥1,575.00
现货
25mg
¥3,507.00
现货
50mg
¥5,891.00
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.

104

客户使用产品发表文献 4

产品文档

Quality Control & SDS

View current batch:

实验参考方法

Cell experiment [1]:

Cell lines

BJ-TERT/LT/ST/RASV12 and DRD cells

Preparation method

Soluble in DMSO. General tips for obtaining a higher concentration: Please warm the tube at 37 ℃ for 10 minutes and/or shake it in the ultrasonic bath for a while. Stock solution can be stored below -20℃ for several months.

Reacting condition

5 μg/ml, 2 days

Applications

RSL3 displayed synthetic lethality with oncogenic RAS in both BJ-TERT/LT/ST/RASV12 and DRD cells. RSL3 inhibited the growth of BJ-TERT/LT/ST/RASV12 and DRD cells as low as 10 ng/ml and started to kill sensitive cells as early as 8 hr after treatment. Longer treatment with RSL3 had little effect on the viability of cells lacking oncogenic RAS. RSL3 induced rapid and nonapoptotic cell death in oncogenic ras containing tumorigenic cells.

Animal experiment [2]:

Animal models

Athymic nude mice xenografted with BJeLR cells

Dosage form

Subcutaneous injection (s.c.), 100 mg/kg, twice each week for 2 weeks.

Application

RSL3 prevented tumor growth in a xenograft model. (1S, 3R)-RSL3 significantly prevented tumor growth. (1S, 3R)-RSL3 significantly reduced tumor volume via the induction of ferroptosis. Intraperitoneal injection of (1S, 3R)-RSL3 showed no toxicity up to 400 mg/kg dose, which suggested that (1S, 3R)-RSL3 was well tolerated.

Other notes

Please test the solubility of all compounds indoor, and the actual solubility may slightly differ with the theoretical value. This is caused by an experimental system error and it is normal.

References:

[1]. Yang W S, Stockwell B R. Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells[J]. Chemistry & biology, 2008, 15(3): 234-245.

[2]. Yang W S, SriRamaratnam R, Welsch M E, et al. Regulation of ferroptotic cancer cell death by GPX4[J]. Cell, 2014, 156(1): 317-331.

产品描述

RSL3 is identified as a potent ferroptosis-triggering agent, which is dependent on the activity of GPX4. RSL3 behaved as an inhibitor of GPX4, reducing the expression of GPX4 and inducing ferroptotic death of head and neck cancer cell[1]

In vitro study demonstrated that RSL3 have a degree of synthetic lethality with oncogenic RAS. RSL3 showed rapid and potent ability to induce synthetic lethality with oncogenic RAS. RSL3 inhibited the growth of BJ-TERT/LT/ST/RASV12 and DRD cells as low as 10 ng/ml and started to kill sensitive cells as early as 8 h after treatment. The growth inhibitory effect and the selectivity of RSL3 were confirmed by trypan blue exclusion assay, which demonstrated the potency and selectivity of RSL3. Moreover, longer treatment with RSL3 had little effect on the viability of cells lacking oncogenic RAS, confirming the qualitative nature of RSL3’s selectivity.[2]

In vivo study determined that RSL3-induced ferroptosis could be enhanced by cetuximab with the mechanism of suppressing the Nrf2/HO-1 axis. A DLD-1 xenograft nude mouse model was established to further explored whether cetuximab promotes RSL3-induced ferroptosis in vivo. All the mice survived well after cell implantation or were treated with vehicle, RSL3, cetuximab, or RSL3 in combination with cetuximab. However, administration of RSL3 alone and treatment with both RSL3 and cetuximab led to a decrease in tumour size and tumour volume. Meanwhile, the relative levels of Nrf2 and HO-1 were decreased and that keap1 expressed was relatively increased after co-treatment with RSL3 and cetuximab.[3]

RSL3被认为是一种强效的诱导铁死亡剂,其作用依赖于GPX4的活性。RSL3表现出抑制GPX4的作用,降低了GPX4的表达,并引发头颈癌细胞的铁死亡[1]

体外研究表明,RSL3与致癌的RAS具有一定程度的合成致死性。 RSL3显示出快速而强大的能力,可以诱导与致癌RAS的合成致死性。 RSL3抑制BJ-TERT / LT / ST / RASV12和DRD细胞的生长,低至10 ng/ml,并在治疗后8小时开始杀死敏感细胞。 通过尝试蓝染色排除实验证实了RSL3的生长抑制作用和选择性,这证明了RSL3的效力和选择性。 此外,较长时间使用RSL3对缺乏致癌RAS细胞的存活率几乎没有影响,从而确认了其选择性质量。 [2]

实验结果表明,通过抑制Nrf2/HO-1轴的机制,Cetuximab可以增强RSL3诱导的铁死亡。为了进一步探究Cetuximab是否能促进体内RSL3诱导的铁死亡,研究人员建立了一个DLD-1异种移植裸鼠模型。所有小鼠在细胞移植或接受车载剂、RSL3、Cetuximab或两者联合治疗后都存活良好。然而,单独使用RSL3和同时使用RSL3和Cetuximab治疗均导致肿瘤大小和体积减小。与此同时,在与RSL3和Cetuximab联合处理后,Nrf2和HO-1相对水平降低,并且Keap1表达相对增加。[3]

References:
[1]. Sui X, et al. RSL3 Drives Ferroptosis Through GPX4 Inactivation and ROS Production in Colorectal Cancer. Front Pharmacol. 2018 Nov 22;9:1371.
[2]. Shin D, et al. Nrf2 inhibition reverses resistance to GPX4 inhibitor-induced ferroptosis in head and neck cancer. Free Radic Biol Med. 2018 Dec;129:454-462.
[3]. Yang J, et al. Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer. Cell Death Dis. 2021 Nov 13;12(11):1079.

Chemical Properties

Cas No. 1219810-16-8 SDF
别名 (1S,3R)-RSL3
化学名 (1S,3R)-methyl 2-(2-chloroacetyl)-1-(4-(methoxycarbonyl)phenyl)-2,3,4,9-tetrahydro-1H-pyrido[3,4-b]indole-3-carboxylate
Canonical SMILES COC(C1=CC=C([C@@]2([H])C3=C(C4=CC=CC=C4N3)C[C@](N2C(CCl)=O)([H])C(OC)=O)C=C1)=O
分子式 C23H21ClN2O5 分子量 440.88
溶解度 ≥ 125.4 mg/mL in DMSO 储存条件 Store at -20°C, sealed storage, away from moisture
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 2.2682 mL 11.341 mL 22.6819 mL
5 mM 0.4536 mL 2.2682 mL 4.5364 mL
10 mM 0.2268 mL 1.1341 mL 2.2682 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

RSL3 Drives Ferroptosis Through GPX4 Inactivation and ROS Production in Colorectal Cancer

Front Pharmacol2018 Nov 22;9:1371.PMID: 30524291DOI: 10.3389/fphar.2018.01371

Ferroptosis is an iron-dependent, oxidative cell death, and is characterized by iron-dependent accumulation of reactive oxygen species (ROS) within the cell. It has been implicated in various human diseases, including cancer. Recently, ferroptosis, as a non-apoptotic form of cell death, is emerging in specific cancer types; however, its relevance in colorectal cancer (CRC) is unexplored and remains unclear. Here, we showed that ferroptosis inducer RSL3 initiated cell death and ROS accumulation in HCT116, LoVo, and HT29 CRC cells over a 24 h time course. Furthermore, we found that ROS levels and transferrin expression were elevated in CRC cells treated with RSL3 accompanied by a decrease in the expression of glutathione peroxidase 4 (GPX4), indicating an iron-dependent cell death, ferroptosis. Overexpression GPX4 resulted in decreased cell death after RSL3 treatment. Therefore, RSL3 was able to induce ferroptosis on three different CRC cell lines in vitro in a dose- and time-dependent manner, which was due to increased ROS and an increase in the cellular labile iron pool. Moreover, this effect was able to be reversed by overexpression of GPX4. Taken together, our results suggest that the induction of ferroptosis contributed to RSL3-induced cell death in CRC cells and ferroptosis may be a pervasive and dynamic form of cell death for cancer treatment.

Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer

Cell Death Dis2021 Nov 13;12(11):1079.PMID: 34775496DOI: 10.1038/s41419-021-04367-3

Cetuximab is approved for the treatment of metastatic colorectal cancer (mCRC) with RAS wild-type. Nevertheless, the prognosis remains poor and the effectiveness of cetuximab is limited in KRAS mutant mCRC. Recently, emerging evidence has shown that ferroptosis, a newly discovered form of nonapoptotic cell death, is closely related to KRAS mutant cells. Here, we further investigated whether cetuximab-mediated regulation of p38/Nrf2/HO-1 promotes RSL3-induced ferroptosis and plays a pivotal role in overcoming drug resistance in KRAS mutant colorectal cancer (CRC). In our research, we used two KRAS mutant CRC cell lines, HCT116 and DLD-1, as models of intrinsic resistance to cetuximab. The viability of cells treated with the combination of RSL3 and cetuximab was assessed by the CCK-8 and colony formation assays. The effective of cetuximab to promote RSL3-induced ferroptosis was investigated by evaluating lipid reactive oxygen species accumulation and the expression of the malondialdehyde and the intracellular iron assay. Cetuximab therapy contributed to regulating the p38/Nrf2/HO-1 axis, as determined by western blotting and transfection with small interfering RNAs. Cetuximab promoted RSL3-induced ferroptosis by inhibiting the Nrf2/HO-1 in KRAS mutant CRC cells, and this was further demonstrated in a xenograft nude mouse model. Our work reveals that cetuximab enhances the cytotoxic effect of RSL3 on KRAS mutant CRC cells and that cetuximab enhances RSL3-induced ferroptosis by inhibiting the Nrf2/HO-1 axis through the activation of p38 MAPK.

Microglia and macrophage exhibit attenuated inflammatory response and ferroptosis resistance after RSL3 stimulation via increasing Nrf2 expression

J Neuroinflammation2021 Oct 30;18(1):249.PMID: 34717678DOI: 10.1186/s12974-021-02231-x

Background: Many neurological diseases involve neuroinflammation, during which overproduction of cytokines by immune cells, especially microglia, can aggregate neuronal death. Ferroptosis is a recently discovered cell metabolism-related form of cell death and RSL3 is a well-known inducer of cell ferroptosis. Here, we aimed to investigate the effects of RSL3 in neuroinflammation and sensitivity of different type of microglia and macrophage to ferroptosis.
Methods: Here, we used quantitative RT-PCR analysis and ELISA analysis to analyze the production of proinflammatory cytokine production of microglia and macrophages after lipopolysaccharides (LPS) stimulation. We used CCK8, LDH, and flow cytometry analysis to evaluate the sensitivity of different microglia and macrophages to RSL3-induced ferroptosis. Western blot was used to test the activation of inflammatory signaling pathway and knockdown efficiency. SiRNA-mediated interference was conducted to knockdown GPX4 or Nrf2 in BV2 microglia. Intraperitoneal injection of LPS was performed to evaluate systemic inflammation and neuroinflammation severity in in vivo conditions.
Results: We found that ferroptosis inducer RSL3 inhibited lipopolysaccharides (LPS)-induced inflammation of microglia and peritoneal macrophages (PMs) in a cell ferroptosis-independent manner, whereas cell ferroptosis-conditioned medium significantly triggered inflammation of microglia and PMs. Different type of microglia and macrophages showed varied sensitivity to RSL3-induced ferroptosis. Mechanistically, RSL3 induced Nrf2 protein expression to inhibit RNA Polymerase II recruitment to transcription start site of proinflammatory cytokine genes to repress cytokine transcription, and protect cells from ferroptosis. Furthermore, simultaneously injection of RSL3 and Fer-1 ameliorated LPS-induced neuroinflammation in in vivo conditions.
Conclusions: These data revealed the proinflammatory role of ferroptosis in microglia and macrophages, identified RSL3 as a novel inhibitor of LPS-induced inflammation, and uncovered the molecular regulation of microglia and macrophage sensitivity to ferroptosis. Thus, targeting ferroptosis in diseases by using RSL3 should consider both the pro-ferroptosis effect and the anti-inflammation effect to achieve optimal outcome.

NCOA4-mediated ferritinophagy promotes ferroptosis induced by erastin, but not by RSL3 in HeLa cells

Biochim Biophys Acta Mol Cell Res2021 Feb;1868(2):118913.PMID: 33245979DOI: 10.1016/j.bbamcr.2020.118913

Ferroptosis is a regulated cell death characterized by a lethal accumulation of lipid peroxides due to an increase of intracellular iron and a decrease of antioxidant capacity. The reduction of antioxidant activity is obtained by using chemical agents, such as erastin and RSL3, the first one inhibiting the transmembrane cystine-glutamate antiporter causing a cysteine and glutathione depletion and the second one inactivating directly the glutathione peroxidase 4 (GPX4) respectively. The role of iron and its related proteins in supporting the formation of lipid peroxides, is not completely understood hence to try to shed light on it we generated HeLa clones with altered ferritinophagy, the ferritin degradation process, by knocking-out or overexpressing Nuclear Receptor Coactivator 4 (NCOA4), the ferritin autophagic cargo-receptor. NCOA4 deficiency abolished ferritinophagy increasing ferritin level and making the cells more resistant to erastin, but unexpectedly more sensitive to RSL3. Interestingly, we found that erastin promoted ferritinophagy in HeLa cells expressing NCOA4, increasing the free iron, lipid peroxidation and the sensitivity to ferroptosis. In contrast, RSL3 did not modulate ferritinophagy, while NCOA4 overexpression delayed RSL3-induced cell death suggesting that RSL3 mechanism of action is independent of ferritin degradation process. Therefore, the ferritin-iron release in the execution of ferroptosis seems to depend on the inducing compound, its target and downstream pathway of cell death activation.

RSL3 Drives Ferroptosis through NF- κ B Pathway Activation and GPX4 Depletion in Glioblastoma

Oxid Med Cell Longev2021 Dec 26;2021:2915019.PMID: 34987700DOI: 10.1155/2021/2915019

Glioblastoma, the most aggressive form of malignant glioma, is very difficult to treat because of its aggressively invasive nature and high recurrence rates. RAS-selective lethal 3 (RSL3), a well-known inhibitor of glutathione peroxidase 4 (GPX4), could effectively induce oxidative cell death in glioblastoma cells through ferroptosis, and several signaling pathways are involved in this process. However, the role of the nuclear factor kappa-B (NF-κB) pathway in glioblastoma cell ferroptosis has not yet been investigated. Therefore, we aimed to clarify the underlying mechanism of the NF-κB pathway in RSL3-induced ferroptosis in glioblastoma cells. We found that RSL3 led to an increase in lipid ROS concentration and downregulation of ferroptosis-related proteins such as GPX4, ATF4, and SLC7A11 (xCT) in glioblastoma cells. Additionally, the NF-κB pathway was activated by RSL3, and its inhibition by BAY 11-7082 could alleviate ferroptosis. The murine xenograft tumor model indicated that NF-κB pathway inhibition could mitigate the antitumor effects of RSL3 in vivo. Furthermore, we found that GPX4 knockdown could not effectively induce ferroptosis. However, NF-κB pathway activation coupled with GPX4 silencing induced ferroptosis. Additionally, ATF4 and xCT expression might be regulated by the NF-κB pathway. Collectively, our results revealed that the NF-κB pathway plays a novel role in RSL3-induced ferroptosis in glioblastoma cells and provides a new therapeutic strategy for glioblastoma treatment.