Home>>Signaling Pathways>> Ubiquitination/ Proteasome>> Autophagy>>PX-478 2HCl

PX-478 2HCl Sale

目录号 : GC11031

PX-478是一种选择性抑制剂,可以抑制常规和低氧诱导的HIF-1α水平

PX-478 2HCl Chemical Structure

Cas No.:685898-44-6

规格 价格 库存 购买数量
10mM (in 1mL DMSO)
¥693.00
现货
5mg
¥599.00
现货
10mg
¥1,050.00
现货
50mg
¥2,405.00
现货
100mg
¥3,938.00
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.

105

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

EC109 and EC9706 cell lines

Preparation Method

Both cell lines were cultured in RPMI 1640 supplemented with 10% FBS at 37°C in a 5% CO2 humidified incubator. Cells were starved for 24 h in medium containing 0.1% FBS prior to treatment.

Reaction Conditions

PX-478 was added to the cultures to a final concentration of 20 umol/L after serum starvation overnight.

Applications

PX-478 could inhibit normoxic and hypoxia-induced HIF-1α expression as well as COX-2 and PD-L1 expression in vitro at a dose of 20 μM in EC9706 and EC109 cells. Targeted HIF-1α can effectively reduce their expression.

Animal experiment [1]:

Animal models

6-week-old female BALB/c nude mice, 18 ± 2 g, SPF grade

Preparation Method

EC109 cells were re-suspended in PBS at 107/mL. A total of 12 BABL/c nude female mice were used. Mice were injected with EC109 cells in the right flank area to establish the tumor xenograft. The 12 tumor-bearing mice in each group were randomly divided into the control group and the PX-478 groups. All the drugs were delivered via p.o. gavage administration.

Dosage form

30 mg/kg every other day

Applications

PX-478 shows a wide effect on tumor development, such as suppressing proliferation, promoting cells apoptosis, inhibiting EMT process and arresting cell cycle. In vivo mouse xenograft models showed the inhibitory effect on tumor growth. In addition, it can reduce COX-2 and PD-L1 expression.

References:

[1].Zhu Y, et al. Inhibition of HIF-1α by PX-478 suppresses tumor growth of esophageal squamous cell cancer in vitro and in vivo. Am J Cancer Res. 2017 May 1;7(5):1198-1212.

产品描述

PX-478 is a selective inhibitor that suppresses constitutive and hypoxia-induced HIF-1α levels. PX-478 inhibits HIF-1α at multiple levels including inhibition of HIF-1α translation and reduction in HIF-1α mRNA levels.[1] PX-478 also has been shown antitumor activity against several aggressive human tumor xenografts.[2]

In vitro and in vivo study demonstrated that PX-478 could inhibit normoxic and hypoxia-induced HIF-1α expression as well as inflammatory molecule COX-2 and immunosuppressive molecule PD-L1 expression. Therefore, PX-478 shows a wide effect on tumor development, such as suppressing proliferation, promoting cells apoptosis, inhibiting EMT process and arresting cell cycle. In vivo experiment indicated the inhibitory effect of PX-478 on tumor growth. [1]

PX-478是一种选择性抑制剂,可以抑制常规和低氧诱导的HIF-1α水平。PX-478在多个层面上抑制HIF-1α,包括抑制HIF-1α翻译和降低HIF-1α mRNA水平。[1]此外,PX-478还显示出对几种侵袭性人类肿瘤异种移植的抗肿瘤活性。[2]

体外和体内研究表明,PX-478可以抑制正常氧和低氧诱导的HIF-1α表达以及炎性分子COX-2和免疫抑制分子PD-L1的表达。因此,PX-478对肿瘤发展具有广泛影响,如抑制增殖、促进细胞凋亡、抑制EMT过程并阻止细胞周期。体内实验显示了PX-478对肿瘤生长的抑制作用。[1]

References:
[1].Zhu Y, et al. Inhibition of HIF-1α by PX-478 suppresses tumor growth of esophageal squamous cell cancer in vitro and in vivo. Am J Cancer Res. 2017 May 1;7(5):1198-1212.
[2].Palayoor ST, et al. PX-478, an inhibitor of hypoxia-inducible factor-1alpha, enhances radiosensitivity of prostate carcinoma cells. Int J Cancer. 2008 Nov 15;123(10):2430-7.

Chemical Properties

Cas No. 685898-44-6 SDF
化学名 (S)-4-(2-amino-2-carboxyethyl)-N,N-bis(2-chloroethyl)aniline oxide dihydrochloride
Canonical SMILES ClCC[N+](C1=CC=C(C[C@@](N)([H])C(O)=O)C=C1)([O-])CCCl.Cl.Cl
分子式 C13H20Cl4N2O3 分子量 394.12
溶解度 79mg/mL in DMSO (200.45mM), 79mg/mL in Water (200.45mM), 79mg/mL in Ethanol (200.45mM) 储存条件 Store at -20°C, stored under nitrogen, 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 2.5373 mL 12.6865 mL 25.373 mL
5 mM 0.5075 mL 2.5373 mL 5.0746 mL
10 mM 0.2537 mL 1.2686 mL 2.5373 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

HIF-1α inhibitor PX-478 preserves pancreatic β cell function in diabetes

Sci Transl Med2022 Mar 30;14(638):eaba9112.PMID: 35353540DOI: 10.1126/scitranslmed.aba9112

During progression of type 2 diabetes, pancreatic β cells are subjected to sustained metabolic overload. We postulated that this state mediates a hypoxic phenotype driven by hypoxia-inducible factor-1α (HIF-1α) and that treatment with the HIF-1α inhibitor PX-478 would improve β cell function. Our studies showed that the HIF-1α protein was present in pancreatic β cells of diabetic mouse models. In mouse islets with high glucose metabolism, the emergence of intracellular Ca2+ oscillations at low glucose concentration and the abnormally high basal release of insulin were suppressed by treatment with the HIF-1α inhibitor PX-478, indicating improvement of β cell function. Treatment of db/db mice with PX-478 prevented the rise of glycemia and diabetes progression by maintenance of elevated plasma insulin concentration. In streptozotocin-induced diabetic mice, PX-478 improved the recovery of glucose homeostasis. Islets isolated from these mice showed hallmarks of improved β cell function including elevation of insulin content, increased expression of genes involved in β cell function and maturity, inhibition of dedifferentiation markers, and formation of mature insulin granules. In response to PX-478 treatment, human islet organoids chronically exposed to high glucose presented improved stimulation index of glucose-induced insulin secretion. These results suggest that the HIF-1α inhibitor PX-478 has the potential to act as an antidiabetic therapeutic agent that preserves β cell function under metabolic overload.

Effects and Mechanism of Action of PX-478 in Oxygen-Induced Retinopathy in Mice

Ophthalmic Res2020;63(2):182-193.PMID: 31955159DOI: 10.1159/000504023

Importance: Retinopathy of prematurity (ROP) is an important risk factor for blindness in children due to neovascularization (NV). Hypoxia stimulates the formation of NV, as retinal hypoxia affects the stability and function of hypoxia-inducible factor (HIF) transcription factors. The purpose of this study is to study the mechanism of ROP and provide theoretical basis for clinical treatment of ROP.
Objective: In the present study, we used a mouse model of oxygen-induced retinopathy (OIR) to demonstrate the effects of the HIF-1α inhibitor PX-478 on OIR, and to determine its mechanism of action, to provide a theoretical basis for the clinical treatment of ROP.
Materials and methods: The OIR mouse model was induced by exposing neonatal mouse pups and their mothers to 75 ± 5% oxygen from postnatal day 7 (P7) to P12, before being returned to room air from P12 to P17. Flat mount analyses were performed at P12 and P17. Hif1a, Hif2a, Hif3a, and Vegfa mRNA were detected by reverse transcription-polymerase chain reaction in OIR mice at P12 and P17. Hif1a and Vegfa mRNA were detected in OIR mice at P12 and P17 treatment with PX-478. Western blot analyses were used to assess the levels of HIF-1α, VEGF-A, and EPO before and after treatment with PX-478 at P12 and P17.
Results: Hif1a mRNA was increased in OIR mice at P12 and P17, while Vegfa mRNA was increased at P12 and P17. HIF-1α, VEGF-A, and EPO protein levels were increased in OIR mice at P12 and P17, as compared to control mice at the same age (all p < 0.05). Inhibition of HIF-1α by injection of PX-478 in OIR mice (P9-P16) caused a decrease in the retinal avascular area at P12 and P17 (both p < 0.05), NV areas at P17 (p < 0.05), Vegfa mRNA decreased at P12 and P17, as compared to control mice (p < 0.05), and VEGF-A and EPO protein levels at P12 and P17, as compared to control mice. Our study found that there were PX-478 both retina and vitreous body of OIR. Inhibition of HIF-1α by injection of PX-478 in OIR mice caused a decrease in the retinal avascular area at P12 and P17, NV areas decreased at P17, VEGF-A and EPO protein levels at P12 and P17. Endothelial cell migration assays and cell tube formation indication PX-478 attenuate cell migration and significantly weakened the cell cavity formation under the condition of hypoxia.
Conclusion: HIF-1α plays a main role in OIR and can be considered a therapeutic target in OIR by suppressing downstream angiogenic factors, PX-478 decreasing the retinal avascular area and NV.

Dichloroacetate and PX-478 exhibit strong synergistic effects in a various number of cancer cell lines

BMC Cancer2021 Apr 30;21(1):481.PMID: 33931028DOI: 10.1186/s12885-021-08186-9

Background: One key approach for anticancer therapy is drug combination. Drug combinations can help reduce doses and thereby decrease side effects. Furthermore, the likelihood of drug resistance is reduced. Distinct alterations in tumor metabolism have been described in past decades, but metabolism has yet to be targeted in clinical cancer therapy. Recently, we found evidence for synergism between dichloroacetate (DCA), a pyruvate dehydrogenase kinase inhibitor, and the HIF-1α inhibitor PX-478. In this study, we aimed to analyse this synergism in cell lines of different cancer types and to identify the underlying biochemical mechanisms.
Methods: The dose-dependent antiproliferative effects of the single drugs and their combination were assessed using SRB assays. FACS, Western blot and HPLC analyses were performed to investigate changes in reactive oxygen species levels, apoptosis and the cell cycle. Additionally, real-time metabolic analyses (Seahorse) were performed with DCA-treated MCF-7 cells.
Results: The combination of DCA and PX-478 produced synergistic effects in all eight cancer cell lines tested, including colorectal, lung, breast, cervical, liver and brain cancer. Reactive oxygen species generation and apoptosis played important roles in this synergism. Furthermore, cell proliferation was inhibited by the combination treatment.
Conclusions: Here, we found that these tumor metabolism-targeting compounds exhibited a potent synergism across all tested cancer cell lines. Thus, we highly recommend the combination of these two compounds for progression to in vivo translational and clinical trials.

Single-Cell RNA Sequencing to Dissect the Immunological Network of Autoimmune Myocarditis

Circulation2020 Jul 28;142(4):384-400.PMID: 32431172DOI: 10.1161/CIRCULATIONAHA.119.043545

Background: Myocarditis can develop into dilated cardiomyopathy, which may require heart transplantation. The immunological network of myocarditis phases remains unknown. This study aimed to investigate the immunological network during the transition from myocarditis to cardiomyopathy and to identify the genes contributing to the inflammatory response to myocarditis.
Methods: Mice were treated with myosin heavy chain-α peptides to generate an experimental autoimmune myocarditis (EAM) model. We performed single-cell RNA sequencing analysis of Cd45+ cells extracted from mouse hearts during different EAM phases, including normal control, acute inflammatory, subacute inflammatory, and myopathy phases. Human heart tissues were collected from the surgically removed hearts of patients who had undergone heart transplantation.
Results: We identified 26 cell subtypes among 34 665 cells. Macrophages constituted the main immune cell population at all disease phases (>60%), and an inflammation-associated macrophage cluster was identified in which the expression of Hif1a-regulated genes was upregulated. The neutrophil population was increased after the induction of EAM, and neutrophils then released Il-1 to participate in the EAM process. T cells were observed at the highest percentage at the subacute inflammatory phase. T-helper 17 cells, in which the expression of Hif1a-regulated genes was upregulated, constituted the main T-cell population detected at the acute inflammatory phase, whereas regulatory T cells were the main T-cell population detected at the subacute inflammatory phase, and γδ T cells releasing Il-17 were the main T-cell population observed at the myopathy phase. Moreover, the Hif1a expression level correlated with the extent of inflammation. In addition, PX-478 could alleviate the inflammatory responses of the different EAM phases. Last, HIF1A was expressed at higher levels in patients with acute autoimmune myocarditis than in patients with dilated cardiomyopathy and healthy control subjects.
Conclusions: We present here a comprehensive single-cell landscape of the cardiac immune cells in different EAM phases. In addition, we elucidate the contribution of Hif1a to the inflammatory response through the regulation of immune cell activity, particularly of macrophage cluster 2 and T-helper 17 cells. Moreover, an Hif1a inhibitor alleviated inflammatory cell infiltration of the EAM model and may serve as a potential therapeutic target in the clinic.

A novel approach to cancer therapy using PX-478 as a HIF-1α inhibitor

Arch Pharm Res2011 Oct;34(10):1583-5.PMID: 22076756DOI: 10.1007/s12272-011-1021-3

Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor produced by tumor cells under hypoxic conditions, and a key regulator of a number of genes important in cancer biology. Over-expression of HIF-1α in human tumors is associated with poor prognosis and poor therapeutic outcomes and HIF-1α has been suggested as a novel target for cancer therapy. This article provides a review of PX-478 as the first novel HIF-1α inhibitor in clinical stage for the treatment of solid tumors.