Home>>Signaling Pathways>> Proteases>> Mitochondrial Metabolism>>Mito-TEMPO

Mito-TEMPO Sale

目录号 : GC31682

Mito-TEMPO 是一种线粒体靶向的超氧化物歧化酶模拟物,具有超氧化物和烷基自由基清除特性。

Mito-TEMPO Chemical Structure

Cas No.:1334850-99-5

规格 价格 库存 购买数量
10mM (in 1mL DMSO)
¥808.00
现货
5mg
¥720.00
现货
10mg
¥1,152.00
现货
50mg
¥3,420.00
现货
100mg
¥5,400.00
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.

103

客户使用产品发表文献 3

产品文档

Quality Control & SDS

View current batch:

实验参考方法

Cell experiment [1]:

Cell lines

Human neuroblastoma cells (SH-SY5Y )

Preparation Method

SH-SY5Y cells were treated with 100 μM glutamate in the presence or absence of Mito-Tempo for 24 h. The cell viability was assessed by the mitochondrial performance, using the MTT reduction assay.

Reaction Conditions

50, 100 µM Mito-Tempo for 24h.

Applications

10 and 100 μM glutamate significantly reduced the number of the living cell to 88.86 ± 3.45% and 51.12 ± 2.97%. 50 and 100 μM Mito-Tempo significantly restored cell viability to 82.90 ± 1.78% and 93.56 ± 2.85%, respectively, indicating that Mito-Tempo could protect the cells from glutamate toxicity.

Animal experiment [2]:

Animal models

C57BL/6 mice

Preparation Method

Lipopolysaccharide- (LPS-) induced experimental sepsis mouse model was established by injecting male mice intraperitoneally with LPS. Mito-TEMPO was intraperitoneally injected 1 h prior to LPS injection.

Dosage form

20 mg/kg Mito-TEMPO, intraperitoneal(i.p.) injection

Applications

In the LPS+Mito-TEMPO group, the levels of alanine transaminase(ALT) and aspartate transaminase (AST) were approximately 1.5- and 2-fold lower compared with those in the LPS group. Mito-TEMPO pretreatment protects mice against LPS-Induced acute liver injury.

References:

[1]. Mukem S, Thongbuakaew T, et al. Mito-Tempo suppresses autophagic flux via the PI3K/Akt/mTOR signaling pathway in neuroblastoma SH-SY5Y cells. Heliyon. 2021 Jun 15;7(6):e07310.

[2]. Wang PF, Xie K, et al. Hepatoprotective Effect of Mitochondria-Targeted Antioxidant Mito-TEMPO against Lipopolysaccharide-Induced Liver Injury in Mouse. Mediators Inflamm. 2022 Jun 20;2022:6394199.

产品描述

Mito-Tempo is a mitochondria-targeted antioxidant with effective superoxide scavenging properties, which converts toxic superoxide molecules into hydrogen peroxide or oxygen and subsequently detoxified to oxygen and water by catalase or glutathione peroxidase[1].

In vitro, Mito-Tempo has a greater protective effect by enhancing superoxide dismutase (SOD) activity and PI3K/AKT/mTOR phosphorylation. Glutamate-exposed cells significantly increased cellular oxidative stress by enhancing ROS production. Glutamate treatment also increased LDH release follows the loss of mitochondrial membrane potential, caused cell viability loss. Treatment with Mito-Tempo not only attenuated the generation of ROS and improved mitochondrial membrane potential but also reduced the neurotoxicity of glutamate in a concentration-dependent manner, which leads to increased cell viability and decreased LDH release[2]

In vivo,Mito-TEMPO pretreatment inhibited inflammation, attenuated LPS-induced liver injury, and enhanced the antioxidative capability in septic mice, as evidenced by the decreased MDA content and the increased SOD activity. In addition, Mito-TEMPO restored mitochondrial size and improved mitochondrial function. Finally, we found that the levels of pyroptosis-related proteins in the liver of LPS-treated mice were lower after pretreatment with Mito-TEMPO [3]

References:
[1]. Liang HL, Sedlic F, et al. SOD1 and MitoTEMPO partially prevent mitochondrial permeability transition pore opening, necrosis, and mitochondrial apoptosis after ATP depletion recovery. Free Radic Biol Med. 2010 Nov 30;49(10):1550-60.
[2]. Mukem S, Thongbuakaew T, et al. Mito-Tempo suppresses autophagic flux via the PI3K/Akt/mTOR signaling pathway in neuroblastoma SH-SY5Y cells. Heliyon. 2021 Jun 15;7(6):e07310.
[3]. Mukem S, Thongbuakaew T, et al. Mito-Tempo suppresses autophagic flux via the PI3K/Akt/mTOR signaling pathway in neuroblastoma SH-SY5Y cells. Heliyon. 2021 Jun 15;7(6):e07310.

Mito-Tempo 是一种靶向线粒体的抗氧化剂,具有有效的超氧化物清除特性,可将有毒的超氧化物分子转化为过氧化氢或氧气,随后通过过氧化氢酶或谷胱甘肽过氧化物酶解毒为氧气和水[1] .

在体外,Mito-Tempo 通过增强超氧化物歧化酶 (SOD) 活性和 PI3K/AKT/mTOR 磷酸化而具有更强的保护作用。暴露于谷氨酸的细胞通过增强 ROS 的产生显着增加细胞氧化应激。谷氨酸处理还增加了线粒体膜电位丧失后 LDH 的释放,导致细胞活力丧失。 Mito-Tempo 处理不仅减少了 ROS 的产生并改善了线粒体膜电位,而且以浓度依赖性方式降低了谷氨酸的神经毒性,从而导致细胞活力增加和 LDH 释放减少[2]

在体内,Mito-TEMPO 预处理可抑制炎症,减轻 LPS 诱导的肝损伤,并增强脓毒症小鼠的抗氧化能力,这可以通过降低 MDA 含量和增加 SOD 活性来证明。此外,Mito-TEMPO 恢复了线粒体大小并改善了线粒体功能。最后,我们发现在用 Mito-TEMPO [3]

预处理后,LPS 处理的小鼠肝脏中焦亡相关蛋白的水平较低

Chemical Properties

Cas No. 1334850-99-5 SDF
Canonical SMILES [O]N1C(C)(C)CC(NC(C[P+](C2=CC=CC=C2)(C3=CC=CC=C3)C4=CC=CC=C4)=O)CC1(C)C.[Cl-]
分子式 C29H35ClN2O2P 分子量 510.03
溶解度 DMSO : 125 mg/mL (245.08 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 1.9607 mL 9.8033 mL 19.6067 mL
5 mM 0.3921 mL 1.9607 mL 3.9213 mL
10 mM 0.1961 mL 0.9803 mL 1.9607 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

Mito-TEMPO Alleviates Renal Fibrosis by Reducing Inflammation, Mitochondrial Dysfunction, and Endoplasmic Reticulum Stress

Background: Renal fibrosis is a common pathological symptom of chronic kidney disease (CKD). Many studies support that mitochondrial dysfunction and endoplasmic reticulum (ER) stress are implicated in the pathogenesis of CKD. In our study, we investigated the benefits and underlying mechanisms of Mito-TEMPO on renal fibrosis in 5/6 nephrectomy mice. Methods: Mice were randomly divided into five groups as follows: control group, CKD group, CKD + Mito-TEMPO (1 mg·kg-1·day-1) group, CKD + Mito-TEMPO (3 mg·kg-1·day-1) group, and Mito-TEMPO group (3 mg·kg-1·day-1). Renal fibrosis was evaluated by PAS, Masson staining, immunohistochemistry, and real-time PCR. Oxidative stress markers such as SOD2 activity and MDA level in serum and isolated mitochondria from renal tissue were measured by assay kits. Mitochondrial superoxide production was evaluated by MitoSOX staining and Western blot. Mitochondrial dysfunction was assessed by electron microscopy and real-time PCR. ER stress-associated protein was measured by Western blot. Results: Impaired renal function and renal fibrosis were significantly improved by Mito-TEMPO treatment. Furthermore, inflammation cytokines, profibrotic factors, oxidative stress markers, mitochondrial dysfunction, and ER stress were all increased in the CKD group. However, these effects were significantly ameliorated in the Mito-TEMPO treatment group. Conclusions: Mito-TEMPO ameliorates renal fibrosis by alleviating mitochondrial dysfunction and endoplasmic reticulum stress possibly through the Sirt3-SOD2 pathway, which sheds new light on prevention of renal fibrosis in chronic kidney disease.

Mito-TEMPO protects against bisphenol-A-induced testicular toxicity: an in vivo study

Bisphenol-A (BPA) is a common environmental toxin which alters testicular function in both animals and humans. BPA exerts its cytotoxic potential by altering mitochondrial oxidative stress and functioning. Therefore, protecting mitochondria from oxidative stress may prevent BPA-induced testicular damage. In the present study, modulation of BPA toxicity by mitochondria-targeted antioxidant, mito-TEMPO was studied in male Wistar rats. Rats were administered mito-TEMPO (0.1 mg/kg b.w, i.p.) twice a week, followed by BPA (10 mg/kg b.w., orally) once a week for 4 weeks. After 4 weeks, sperm parameters were evaluated in the testis along with histopathological analysis. The mitochondrial oxidative stress, mitochondrial membrane potential (MMP) and enzymatic activity of mitochondrial complex II and IV were estimated in the testicular tissue. Pretreatment of mito-TEMPO protected animals from toxic effects of BPA as indicated by the normalization of sperm parameters and preserved histoarchitecture of testis. BPA treatment to animals significantly increased mitochondrial reactive oxygen species (ROS) and lipid peroxidation (LPO). A significant decrease in the activity of mitochondrial complex II was also observed after BPA exposure whereas, mitochondrial complex II activity was increased. In addition, an increase in MMP was also observed in BPA group. Mito-TEMPO successfully normalized mitochondrial ROS and LPO formation. Similar normalization effect was also noted in the activity of mitochondrial complex II, complex IV, and MMP. Results suggested that mito-TEMPO pretreatment significantly protected BPA-induced oxidative stress and thereby mito-TEMPO effectively prevented testicular damage.

Mito-Tempo suppresses autophagic flux via the PI3K/Akt/mTOR signaling pathway in neuroblastoma SH-SY5Y cells

The generation of excessive mitochondrial reactive oxygen species (mtROS) is associated with glutamate-stimulated neurotoxicity and pathogenesis of Alzheimer's disease (AD). Impaired mitochondrial function is accompanied with oxidative stress that is a significant contributor to initiate autophagy, but the underlying mechanisms are not fully understood. The present study aimed to investigate the neuroprotective effects of Mito-Tempo on glutamate-induced neuroblastoma SH-SY5Y cell toxicity. SH-SY5Y cells were treated with 100 μM glutamate in the presence or absence of 50 and 100 μM Mito-Tempo for 24 h. Changes in cell viability were measured by MTT assay. Cytotoxicity and intracellular ROS accumulation were also evaluated using lactate dehydrogenase (LDH) activity assay and 2,7-dichlorofluorescein diacetate (DCFDA) Reactive Oxygen Species Assay kit, respectively. Mitochondrial membrane potential was analyzed by tetraethylbenzimidazoly-lcarbocyanine iodide (JC-1) staining. Expression of PI3K/AKT/mTOR pathway and autophagy markers, including LC3 (LC3-I/-II) and p62 (SQSTM1) were performed using Western blot analysis. Our results demonstrated that glutamate-exposed cells significantly increased cellular oxidative stress by enhancing ROS production. Glutamate treatment also increased LDH release follows the loss of mitochondrial membrane potential, caused cell viability loss. Treatment with Mito-Tempo not only attenuated the generation of ROS and improved mitochondrial membrane potential but also reduced the neurotoxicity of glutamate in a concentration-dependent manner, which leads to increased cell viability and decreased LDH release. Mito-Tempo has a greater protective effect by enhancing superoxide dismutase (SOD) activity and PI3K/AKT/mTOR phosphorylation. Moreover, Mito-Tempo treatment altered the autophagy process resulting in the decline in the ratio of the autophagy markers LC3-I/-II and p62 (SQSTM1). We propose that Mito-Tempo can improve neuronal properties against glutamate cytotoxicity through its direct free radical scavenging activity and inhibit excessive autophagy signaling pathway, therefore, allow for further studies to investigate the therapeutic potentials of Mito-Tempo in animal disease models and human.

Mitochondrial ROS promote mitochondrial dysfunction and inflammation in ischemic acute kidney injury by disrupting TFAM-mediated mtDNA maintenance

Aims: Ischemia-reperfusion injury (IRI)-induced acute kidney injury (IRI-AKI) is characterized by elevated levels of reactive oxygen species (ROS), mitochondrial dysfunction, and inflammation, but the potential link among these features remains unclear. In this study, we aimed to investigate the specific role of mitochondrial ROS (mtROS) in initiating mitochondrial DNA (mtDNA) damage and inflammation during IRI-AKI. Methods: The changes in renal function, mitochondrial function, and inflammation in IRI-AKI mice with or without mtROS inhibition were analyzed in vivo. The impact of mtROS on TFAM (mitochondrial transcription factor A), Lon protease, mtDNA, mitochondrial respiration, and cytokine release was analyzed in renal tubular cells in vitro. The effects of TFAM knockdown on mtDNA, mitochondrial function, and cytokine release were also analyzed in vitro. Finally, changes in TFAM and mtDNA nucleoids were measured in kidney samples from IRI-AKI mice and patients. Results: Decreasing mtROS levels attenuated renal dysfunction, mitochondrial damage, and inflammation in IRI-AKI mice. Decreasing mtROS levels also reversed the decrease in TFAM levels and mtDNA copy number that occurs in HK2 cells under oxidative stress. mtROS reduced the abundance of mitochondrial TFAM in HK2 cells by suppressing its transcription and promoting Lon-mediated TFAM degradation. Silencing of TFAM abolished the Mito-Tempo (MT)-induced rescue of mitochondrial function and cytokine release in HK2 cells under oxidative stress. Loss of TFAM and mtDNA damage were found in kidneys from IRI-AKI mice and AKI patients. Conclusion: mtROS can promote renal injury by suppressing TFAM-mediated mtDNA maintenance, resulting in decreased mitochondrial energy metabolism and increased cytokine release. TFAM defects may be a promising target for renal repair after IRI-AKI.

Mitochondria-targeted antioxidant, mito-TEMPO mitigates initiation phase of N-Nitrosodiethylamine-induced hepatocarcinogenesis

Targeting mitochondrial oxidative stress during initial stages of hepatocarcinogenesis can be an effective and promising strategy to prevent hepatocellular carcinoma (HCC). In the present study, mitochondria targeted antioxidant, mito-TEMPO was administered to male BALB/c mice at a dosage 0.1 mg/kg b.w. (intraperitoneal) twice a week, followed by single N-Nitrosodiethylamine (NDEA) intraperitoneal injection (10 mg/kg b.w.). After 24 h of NDEA administration, animals were sacrificed, blood and liver tissue were collected. Liver injury markers, histoarchitecture, antioxidant defence status, mitochondrial reactive oxygen species (ROS), lipid peroxidation (LPO), mitochondrial dysfunction analysis, and mitochondrial membrane potential were investigated. Mito-TEMPO pre-treatment protected animals from the damaging effects of NDEA as observed by normalization of liver injury markers. NDEA metabolism resulted in a significantly increased intracellular and mitochondrial ROS generation with concomitant increase in LPO formation. The activity of mitochondrial complex I, complex II, malate dehydrogenase were significantly reduced and mitochondrial membrane potential was increased. Mito-TEMPO effectively scavenged NDEA-induced ROS generation and reduced LPO formation. A significant improvement was also observed in the activity of mitochondrial complex I, complex II, malate dehydrogenase and normalisation of mitochondrial membrane potential. Results suggested that mito-TEMPO had significant impact on the initiation phase of hepatocarcinogensis which could be one of the reason for its reported chemopreventive effect.