Home>>Signaling Pathways>> GPCR/G protein>> Adrenergic Receptor>>Bufuralol

Bufuralol

(Synonyms: 丁呋洛尔; Ro 3-4787) 目录号 : GC67692

Bufuralol (Ro 3-4787) 是一种有效的非选择性,具有口服活性的 β-肾上腺素受体 (β-adrenoreceptor) 拮抗剂,具有部分激动剂活性。Bufuralol hydrochloride 是一种 CYP2D6 探针的底物。

Bufuralol Chemical Structure

Cas No.:54340-62-4

规格 价格 库存 购买数量
1mg
¥1,759.00
现货
5mg
¥3,870.00
现货
10mg
¥6,210.00
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:

产品描述

Bufuralol (Ro 3-4787) is a potent non-selective, orally active β-adrenoreceptor antagonist with partial agonist activity. Bufuralol hydrochloride is a CYP2D6 probe substrate[1][2][3][4].

Bufuralol (Ro 3-4787) 广泛用于表征 CYP2D6 活性,并具有 CYP2D6 底物特有的芳香环和碱性氮[3]

Bufuralol (Ro 3-4787) 代谢由 NADPH 介导表现出双相动力学,其效率低于在和猴子肠道中观察到的氢过氧化异丙苯 (CuOOH) 存在的情况,这与在肝脏中的观察结果一致[4]

[1]. T H Pringle, et al. Pharmacodynamic and pharmacokinetic studies on bufuralol in man. Br J Clin Pharmacol. 1986 Nov;22(5):527-34.
[2]. Jie Cai, et al. Effects of 22 Novel CYP2D6 Variants Found in the Chinese Population on the Bufuralol and Dextromethorphan Metabolisms In Vitro. Basic Clin Pharmacol Toxicol. 2016 Mar;118(3):190-9.
[3]. Sarah M Glass, et al. CYP2D6 Allelic Variants *34, *17-2, *17-3, and *53 and a Thr309Ala Mutant Display Altered Kinetics and NADPH Coupling in Metabolism of Bufuralol and Dextromethorphan and Altered Susceptibility to Inactivation by SCH 66712. Drug Metab Dispos. 2018 Aug;46(8):1106-1117.
[4]. T Prueksaritanont, et al. (+)-bufuralol 1'-hydroxylation activity in human and rhesus monkey intestine and liver. Biochem Pharmacol. 1995 Oct 26;50(9):1521-5.

Chemical Properties

Cas No. 54340-62-4 SDF Download SDF
别名 丁呋洛尔; Ro 3-4787
分子式 C16H23NO2 分子量 261.36
溶解度 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 3.8261 mL 19.1307 mL 38.2614 mL
5 mM 0.7652 mL 3.8261 mL 7.6523 mL
10 mM 0.3826 mL 1.9131 mL 3.8261 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

(+)-bufuralol 1'-hydroxylation activity in human and rhesus monkey intestine and liver

Biochem Pharmacol 1995 Oct 26;50(9):1521-5.PMID:7503805DOI:10.1016/0006-2952(95)02052-7.

(+)-Bufuralol 1'-hydroxylation, a commonly used marker of hepatic CYP2D6 activity, was investigated in human and rhesus monkey intestinal microsomes and compared with that in hepatic microsomes. The cumene hydroperoxide (CuOOH)-mediated metabolism of (+)-bufuralol suggested that at least two enzymes were responsible for Bufuralol 1'-hydroxylation in both human and monkey intestinal microsomes. In contrast, the kinetics of the CuOOH-mediated metabolism in human and monkey livers were monophasic. The Km values for the higher affinity component of the intestinal enzyme(s) of both species were similar to, while the corresponding Vmax values were much lower than, those obtained with the livers. Bufuralol metabolism mediated by NADPH exhibited biphasic kinetics and was less efficient than that observed in the presence of CuOOH in both human and monkey intestines, in agreement with the observations in the livers. Inhibition of Bufuralol hydroxylase activity in the intestine and liver preparations from the same species by known CYP2D6 inhibitors/substrates was qualitatively similar. Quinidine was the most potent inhibitor of (+)-bufuralol 1'-hydroxylation in all tissues studied. Western immunoblots using anti-CYP2D6 peptide antibody revealed a protein band in human and monkey intestinal microsomes of the same molecular weight as that observed in the liver preparations. The intestinal CYP2D protein content appeared to be much less than that of liver, and correlated with the (+)-bufuralol hydroxylase activity. Immunoinhibition studies indicated significant (up to 50%) inhibition of the CuOOH-mediated (+)-bufuralol metabolism in human and monkey intestines only by anti-CYP2D6, and not by anti-CYP2A6, or anti-CYP2E1. Inhibition of the Bufuralol 1'-hydroxylase activity by anti-rat CYP3A1 was only slight (20%) in human, but marked (60-65%) in monkey intestinal microsomes. The hepatic metabolism of (+)-bufuralol in humans and monkeys was only inhibited (75%) by anti-CYP2D6, but not by anti-CYP3A1. Overall, the results suggest that (1) tissue and species differences exist in the catalysis of (+)-bufuralol 1'-hydroxylation, and (2) CYP2D6-related enzymes are partially or primarily responsible for the Bufuralol hydroxylase activity in human and monkey intestines or monkey liver.

Stereoselectivity in the oxidation of Bufuralol, a chiral substrate, by human cytochrome P450s

Chirality 2003 May 5;15(4):333-9.PMID:12666241DOI:10.1002/chir.10212.

Bufuralol (BF), a nonselective beta-adrenoceptor blocking agent, has a chiral center in its molecule, yielding the enantiomers 1'R-BF and 1'S-BF. beta-Adrenoceptor blocking potency is much higher in 1'S-BF than in 1'R-BF. One of the metabolic pathways of BF is 1"-hydroxylation of an ethyl group attached at the aromatic 7-position forming a carbinol metabolite (1"-hydroxybufuralol, 1"-OH-BF), and further oxidation (or dehydrogenation) produces a ketone metabolite (1-oxobufuralol, 1"-Oxo-BF). Both 1"-OH-BF and 1"-Oxo-BF are known to have beta-adrenoceptor blocking activities comparable to or higher than those of the parent drug. The 1"-hydroxylation introduces another chiral center into the BF molecule and four 1"-OH-BF diastereomers are formed from BF racemate in mammals, including humans, making elucidation of the metabolic profiles complicated. HPLC methods employing derivatization, reversed phase, or chiral columns have been developed to efficiently separate the four 1"-OH-BF diastereomers formed from BF enantiomers or racemate. Accumulated in vitro experimental results revealed that 1'R-BF is a much more preferential substrate than 1'S-BR for BF 1"-hydroxylation in human liver microsomes. Kinetic studies using recombinant human cytochrome P450 (CYP) enzymes indicate that CYP2D6 serves as a major BF 1"-hydroxylase and that CYP1A2 and CYP2C19 also contribute to BF 1"-hydroxylation in human livers. This mini-review summarizes the knowledge reported so far on the pharmacology of BF and its metabolites and the profiles of BF metabolism, especially focusing on the stereoselectivity in the oxidation of BF mainly in human livers and recombinant CYP enzymes.

Bufuralol hydroxylation by cytochrome P450 2D6 and 1A2 enzymes in human liver microsomes

Mol Pharmacol 1994 Sep;46(3):568-77.PMID:7935340doi

Bufuralol 1'-hydroxylation is a prototypical reaction catalyzed by cytochrome P450 (P450) 2D6, an enzyme known to show debrisoquine/sparteine-type genetic polymorphism in humans. In the present study we further examined the roles of several human P450 enzymes, as well as P450 2D6, in the hydroxylation of (+/-)-bufuralol, using liver microsomes from several human samples and human P450 enzymes expressed in human lymphoblastoid cell lines or Escherichia coli. Kinetic analysis of Bufuralol 1'-hydroxylation by liver microsomes showed that there were different Km and Vmax values in seven human samples examined; low Km values (approximately 0.05 mM) were observed in four samples (including sample HL-18), high Km values (approximately 0.25 mM) in two samples (including sample HL-67), and an intermediate Km value (approximately 0.1 mM) in one sample. Quinidine and anti-rat P450 2D1 antibody almost completely inhibited Bufuralol 1'-hydroxylation in human sample HL-18 at a substrate concentration of 0.4 mM, whereas these effects were not so drastic when liver microsomes from human sample HL-67 were used. In contrast, a very low concentration (< 10 microM) of alpha-naphthoflavone or anti-human P450 1A2 antibody significantly inhibited Bufuralol 1'-hydroxylation catalyzed by human sample HL-67, but not HL-18, with 0.4 mM Bufuralol. When the relative contents of P450 2D6 and P450 1A2 in 20 human samples were determined, Bufuralol 1'-hydroxylation in samples containing large amounts of P450 2D6 tended to be more sensitive to quinidine, whereas the P450 1A2-rich samples were highly susceptible to alpha-naphthoflavone. However, at low substrate concentrations Bufuralol 1'-hydroxylation was shown to be catalyzed principally by P450 2D6, based on the inhibitory effects of anti-rat P450 2D1 antibody and quinidine, in both human samples HL-18 and HL-67. At least five other, minor, Bufuralol products were formed by human liver microsomes, in addition to 1'-hydroxybufuralol. Two of them were identified as 4- and 6-hydroxybufuralol by 1H NMR spectroscopy and mass spectrometry. The formation of the 4- and 6-hydroxylated products was suggested to be catalyzed by P450 1A2, based on the results of correlation with P450 1A2 contents in 60 human samples and inhibition by anti-P450 1A2 and alpha-naphthoflavone. Purified recombinant P450 1A2 (expressed in E. coli) produced 1'-, 4-, and 6-hydroxybufuralol in a reconstituted system, although P450 2D6 (expressed in human lymphoblast cell lines) was found to catalyze only Bufuralol 1'-hydroxylation.(ABSTRACT TRUNCATED AT 400 WORDS)

Pharmacodynamic and pharmacokinetic studies on Bufuralol in man

Br J Clin Pharmacol 1986 Nov;22(5):527-34.PMID:2878678DOI:10.1111/j.1365-2125.1986.tb02931.x.

Observations were made in eight subjects who exercised before and at 1, 2, 4, 6, 8 and 24 h after the double-blind oral administration of placebo, Bufuralol 7.5, 15, 30, 60 and 120 mg and propranolol 40 and 160 mg. The exercise heart rate remained constant after placebo. Bufuralol 7.5 mg and propranolol 40 mg reduced exercise heart rate up to 6 and 8 h respectively after dosing but Bufuralol 15, 30, 60 and 120 mg and propranolol 160 mg were still active at 24 h. The lowest exercise heart rate occurred at 2 h after all active treatments. Bufuralol 60 and 120 mg produced similar reduction in exercise tachycardia as propranolol 40 mg but less than propranolol 160 mg. Plasma levels of Bufuralol and its two major metabolites were measured. The peak plasma concentrations of Bufuralol occurred at 1.5 h after 7.5 mg and at 2 h after the other doses of Bufuralol. In six subjects the plasma elimination half-life of Bufuralol was 2.61 +/- 0.18 h and in the other three subjects 4.85 +/- 0.35 h. There was a corresponding longer time to peak concentration and plasma elimination half-life of the two metabolites in these three subjects. These findings show that Bufuralol is a potent beta-adrenoceptor antagonist with partial agonist activity. It has a long duration of action and there is bimodal metabolism of the drug in man.

Bufuralol metabolism in human liver: a sensitive probe for the debrisoquine-type polymorphism of drug oxidation

Eur J Clin Invest 1984 Jun;14(3):184-9.PMID:6147254DOI:10.1111/j.1365-2362.1984.tb01121.x.

The genetically controlled polymorphism causing decreased metabolism of debrisoquine is closely related to that of the metabolism of Bufuralol and numerous other drugs and has important clinical consequences. A sensitive in vitro assay was developed which quantifies the production of 1'-hydroxy-bufuralol (carbinol) from Bufuralol in human liver microsomes. Initial formation rates of carbinol suggested Michaelis-Menten kinetics with an apparent KM of 61 and 171 mumol l-1 and Vmax of 3.2 and 5.8 nmol mg-1 microsomal protein h-1 in two human liver samples. The Vmax in microsomes of thirty-two liver samples was 4.2 +/- 1.0 (SD) nmol carbinol mg-1 protein h-1. Metabolism of debrisoquine in vivo, as expressed by the 'metabolic ratio' of debrisoquine over 4-OH debrisoquine correlated (r = -0.65, P less than 0.01; n = 18) with carbinol production rate in microsomes in vitro. Microsomes of one individual identified as poor metabolizer of debrisoquine in vivo showed reduction of carbinol formation to 1.97 nmol mg-1 h-1. Mixing his microsomes with those of an extensive metabolizer resulted in additive formation of carbinol excluding mediation of the defect by a soluble inhibitor. These data support the concept of a primary defect in microsomal oxidation of Bufuralol. The described assay offers a sensitive tool to investigate the molecular mechanism of the 'debrisoquine polymorphism'.