Home>>Signaling Pathways>> GPCR/G protein>> GPCR19>>INT-777

INT-777 Sale

(Synonyms: 6ALPHA-乙基-23(S)-甲基胆酸,S-EMCA) 目录号 : GC13681

An agonist of TGR5

INT-777 Chemical Structure

Cas No.:1199796-29-6

规格 价格 库存 购买数量
10mM (in 1mL DMSO)
¥3,123.00
现货
2mg
¥2,100.00
现货
5mg
¥3,150.00
现货
10mg
¥4,500.00
现货
50mg
¥13,500.00
现货
100mg
¥18,900.00
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.

101

客户使用产品发表文献 1

产品文档

Quality Control & SDS

View current batch:

实验参考方法

Cell experiment [1]:

Cell lines

podocytes

Preparation Method

1.75 × 104 podocytes per well were seeded in 24-well microplates for 7 days. Baseline OCR was measured in control (vehicle) or INT-777 (10 μM) treated cells. To determine fatty acid β-oxidation cells were treated with BAS-complexed palmitate (0.5 mM) followed by treatment with control or INT-777 (10 μM).

Reaction Conditions

10 μM; 7 days

Applications

INT-777 increased mitochondrial oxygen consumption rate and suppressed mitochondrial ROS production as determined by Mito Sox staining. INT-777 also induced increases in mitochondrial p-AMPK and PGC-1α protein.

Animal experiment [2]:

Animal models

Sprague-Dawley rats

Preparation Method

A total volume of 30 μL vehicle (10% dimethyl sulfide) or INT-777 at three different doses (10 μg/kg, 30 μg/kg, and 90 μg/kg) were administered into the right and left nares, alternating 10 μL in one naris per 5 minutes. H89 was diluted in 10% dimethyl sulfide (DMSO) and was administrated by intraperitoneally (i.p.) at 1 h before SAH.

Dosage form

10 μg/kg, 30 μg/kg, and 90 μg/kg; Intranasal

Applications

The medium (30μg/kg) and high (90μg/kg) doses of INT-777 administration significantly improved the neurological deficits on modified Garcia test and beam balance test at 24 h after SAH compared with the vehicle group.

References:

[1]. Wang XX, et al. G Protein-Coupled Bile Acid Receptor TGR5 Activation Inhibits Kidney Disease in Obesity and Diabetes. J Am Soc Nephrol. 2016 May;27(5):1362-78.

[2]. Hu X, et al. INT-777 attenuates NLRP3-ASC inflammasome-mediated neuroinflammation via TGR5/cAMP/PKA signaling pathway after subarachnoid hemorrhage in rats. Brain Behav Immun. 2021 Jan;91:587-600.

产品描述

INT-777 is a novel specific semisynthetic TGR5 agonist used for alleviating cardiomyocyte injury and improving cognitive impairment and synaptic dysfunction in mice model of AD.[1][2]

In vitro, INT-777 induced a dose-dependent (10 μM to 60 μM) decrease in Isc, when INT-777 added on the serosal side of seromuscular stripped distal colon segments in Ussing chambers.[5] In vitro experiment it shown that treatment with 25 μm of INT-777 for 48 h in αTC1-6 cells as well as in mouse and human islets under high glucose conditions obviously increased PC1 mRNA. However, INT-777 (25 μm) had no effect on PC1 mRNA in cells exposed to low glucose conditions.[6] In vitro, combination 100 ng/ml LPS with treatment of 30 μM INT-777 dramatically decreased the transient increase in mRNA levels for Tnfα, monocyte chemoattractant protein-1 (Mcp-1), Il-6 and Il-1β in macrophages.[7]

In vivo efficacy test it indicated that treatment with 30μg/kg of INT-777 dramatically reduced NLRP3-ASC inflammasome activation in microglia, decreased brain edema and neuroinflammation, resulted in improved short-term neurobehavioral functions at 24 h after subarachnoid hemorrhage.[3] In vivo study it suggested that mice were treated with 1.5 or 3.0 μg/mouse INT-777, single intracerebroventricular (i.c.v.) injection of LPS obviously induced mouse behavioral impairments in Morris water maze, novel object recognition, and Y-maze avoidance tests were ameliorated.[7].

References:
[1]. Wu X, et al. Neuroprotective effects of INT-777 against Aβ1-42-induced cognitive impairment, neuroinflammation, apoptosis, and synaptic dysfunction in mice. Brain Behav Immun. 2018 Oct;73:533-545.
[2]. Deng L, et al. Activation of TGR5 Partially Alleviates High Glucose-Induced Cardiomyocyte Injury by Inhibition of Inflammatory Responses and Oxidative Stress. Oxid Med Cell Longev, 2019: 6372786.
[3]. Hu X, et al. INT-777 attenuates NLRP3-ASC inflammasome-mediated neuroinflammation via TGR5/cAMP/PKA signaling pathway after subarachnoid hemorrhage in rats. Brain Behav Immun. 2021 Jan;91:587-600.
[4]. Wu X, et al. Inhibitory effect of INT-777 on lipopolysaccharide-induced cognitive impairment, neuroinflammation, apoptosis, and synaptic dysfunction in mice. Prog Neuropsychopharmacol Biol Psychiatry. 2019 Jan 10;88:360-374.
[5]. Sorrentino G, et al. Bile Acids Signal via TGR5 to Activate Intestinal Stem Cells and Epithelial Regeneration. Gastroenterology. 2020 Sep;159(3):956-968.e8.
[6]. Kumar DP, et al. Activation of Transmembrane Bile Acid Receptor TGR5 Modulates Pancreatic Islet α Cells to Promote Glucose Homeostasis. J Biol Chem. 2016 Mar 25;291(13):6626-40.
[7]. Pols TW, et al. TGR5 activation inhibits atherosclerosis by reducing macrophage inflammation and lipid loading. Cell Metab. 2011 Dec 7;14(6):747-57.

INT-777 是一种新型特异性半合成 TGR5 激动剂,用于减轻 AD 小鼠模型的心肌细胞损伤,改善认知障碍和突触功能障碍。[1][2]

在体外,当 INT-777 添加到 Ussing 室的浆膜剥离的远端结肠段的浆膜侧时,INT-777 诱导 Isc 剂量依赖性(10 μM 至 60 μM)减少。[5] 体外实验表明,25 μm INT-777 处理 αTC1-6 细胞以及高糖条件下小鼠和人胰岛细胞 48 小时后,PC1 mRNA 明显增加。然而,INT-777 (25 μm) 对暴露于低葡萄糖条件的细胞中的 PC1 mRNA 没有影响。[6] 在体外,100 ng/ml LPS 与 30 μM INT-777 的组合显着降低巨噬细胞中 Tnfα、单核细胞趋化蛋白-1 (Mcp-1)、IL-6 和 Il-1β mRNA 水平的瞬时升高。[7]

体内药效试验表明,用 30μg/kg 的 INT-777 治疗可显着降低小胶质细胞中 NLRP3-ASC 炎性体的激活,减少脑水肿和神经炎症,从而改善蛛网膜下腔出血后 24 小时的短期神经行为功能。 [3] 体内研究表明,小鼠经 1.5 或 3.0 μg/小鼠 INT-777 处理后,单次侧脑室 (i.c.v.) 注射 LPS 可明显诱导小鼠在 Morris 水迷宫中的行为障碍,新颖的对象识别和 Y 迷宫回避测试得到改善。[7]

Chemical Properties

Cas No. 1199796-29-6 SDF
别名 6ALPHA-乙基-23(S)-甲基胆酸,S-EMCA
分子式 C27H46O5 分子量 450.65
溶解度 20mg/mL in DMSO,30mg/mL in DMF, 25mg/mL in Ethanol 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 2.219 mL 11.0951 mL 22.1902 mL
5 mM 0.4438 mL 2.219 mL 4.438 mL
10 mM 0.2219 mL 1.1095 mL 2.219 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

INT-777 attenuates NLRP3-ASC inflammasome-mediated neuroinflammation via TGR5/cAMP/PKA signaling pathway after subarachnoid hemorrhage in rats

Background: Inflammasome-mediated neuroinflammation plays an important role in the pathogenesis of early brain injury (EBI) following subarachnoid hemorrhage (SAH). The activation of the TGR5 receptor has been shown to be neuroprotective in a variety of neurological diseases. This study aimed to investigate the effects of the specific synthetic TGR5 agonist, INT-777, in attenuating NLRP3-ASC inflammasome activation and reducing neuroinflammation after SAH. Methods: One hundred and eighty-four male Sprague Dawley rats were used. SAH was induced by the endovascular perforation. INT-777 was administered intranasally at 1 h after SAH induction. To elucidate the signaling pathway involved in the effect of INT-777 on inflammasome activation during EBI, TGR5 knockout CRISPR and PKA inhibitor H89 were administered intracerebroventricularly and intraperitoneally at 48 h and 1 h before SAH. The SAH grade, short- and long-term neurobehavioral assessments, brain water content, western blot, immunofluorescence staining, and Nissl staining were performed. Results: The expressions of endogenous TGR5, p-PKA, and NLRP3-ASC inflammasome were increased after SAH. INT-777 administration significantly decreased NLRP3-ASC inflammasome activation in microglia, reduced brain edema and neuroinflammation, leading to improved short-term neurobehavioral functions at 24 h after SAH. The administration of TGR5 CRISPR or PKA inhibitor (H89) abolished the anti-inflammation effects of INT-777, on NLRP3-ASC inflammasome, pro-inflammatory cytokines (IL-6, IL-1β, and TNF-a), and neutrophil infiltration at 24 h after SAH. Moreover, early administration of INT-777 attenuated neuronal degeneration in hippocampus on 28 d after SAH. Conclusions: INT-777 attenuated NLRP3-ASC inflammasome-dependent neuroinflammation in the EBI after SAH, partially via TGR5/cAMP/PKA signaling pathway. Early administration of INT-777 may serve as a potential therapeutic strategy for EBI management in the setting of SAH.

INT-777 prevents cognitive impairment by activating Takeda G protein-coupled receptor 5 (TGR5) and attenuating neuroinflammation via cAMP/ PKA/ CREB signaling axis in a rat model of sepsis

Background: Survivors of sepsis must often endure significant cognitive and behavioral impairments after discharge, but research on the relevant mechanisms and interventions remains lacking. TGR5, a member of the class A GPCR family, plays an important role in many physiological processes, and recent studies have shown that agonists of TGR5 show neuroprotective effects in a variety of neurological disorders. To date, no studies have assessed the effects of TGR5 on neuroinflammatory, cognitive, or behavioral changes in sepsis models.
Methods: A total of 267 eight-week-old male Sprague-Dawley rats were used in this study. Sepsis was induced via cecal ligation and puncture (CLP). All animals received volume resuscitation. The rats were given TGR5 CRISPR oligonucleotide intracerebroventricularly 48 h before CLP surgery. INT-777 was administered intranasally 1 h after CLP, and the cAMP inhibitor, SQ22536, was administered intracerebroventricularly 1 h after CLP. Survival rate, bodyweight change, and clinical scores were assessed, and neurobehavioral tests, western blot, and immunofluorescence staining were performed. The cognitive function of rats was measured using the Morris water maze during 15-20 days after CLP.
Results: The expression of TGR5 in the rat hippocampus was upregulated, and peaked at 3 days after CLP. The survival rate of rats after CLP was less than 50%, and the growth rate, in terms of weight, was significantly decreased. While INT-777 treatment did not improve these changes, the treatment did reduce the clinical scores of rats at 24 h after CLP. On day 15 and later, the surviving mice completed a series of behavioral tests. CLP rats showed spatial and memory deficits and anxiety-like behaviors, but INT-777 treatment significantly improved these effects. Mechanistically, immunofluorescence analysis showed that INT-777 treatment reduced the number of microglia in the hippocampus, neutrophilic infiltration, and the expression of inflammatory factors after CLP in rats. Moreover, INT-777 treatment significantly increased the expression of TGR5, cAMP, p-PKA, and p-CREB, but downregulated the expression of IL-1β, IL-6, and TNF-α. CRISPR-mediated TGR5 knockdown and SQ22536 treatment abolished the neuroprotective effects of TGR5 activation after CLP.
Conclusion: This study demonstrates that INT-777 treatment reduced neuroinflammation and microglial cell activation, but improved cognitive impairment in the experimental sepsis rats. TGR5 has translational potential as a therapeutic target to improve neurological outcomes in sepsis survivors.

TGR5 Agonist INT-777 Alleviates Inflammatory Neurodegeneration in Parkinson's Disease Mouse Model by Modulating Mitochondrial Dynamics in Microglia

Parkinson's disease (PD) is one of the most common chronic progressive neurodegenerative diseases that affects both motor and non-motor functions. Bile acids modulate the immune system by targeting brain receptors. INT-777, a 6α-ethyl-23(S)-methyl derivative of cholic acid (S-EMCA), acts as an agonist for Takeda G protein-coupled receptor-5 (TGR5) and has neuroprotective properties. However, the effects of INT-777 on PD have not yet been investigated. In a subchronic PD model, mice treated with 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) developed motor deficits and cognitive impairment that were ameliorated after intranasal administration of INT-777. INT-777 prevented MPTP-induced neurodegeneration and microglia activation in the substantia nigra pars compacta, hippocampus, and cortical layer V. Based on bioinformatics and wet lab data, INT-777 inhibited microglia activation by suppressing the release of tumor necrosis factor alpha (TNF-α) in the hippocampus, along with secondary chemokines (C-C motif ligand 3 (CCL3) and CCL6) in these three brain regions. INT-777 inhibited TNF-α production by repairing mitochondrial damage, which was associated with nuclear factor-erythroid 2-related factor-2 (NRF2) activation and p62/LC3B-mediated autophagy. INT-777 reversed the downregulation of heme oxygenase-1 (HO1), NAD(P)H quinone oxidoreductase-1 (NQO1) and accumulation of p62 in microglia treated with 1-methyl-4-phenylpyridinium (MPP+). However, TGR5 knockdown in microglia abolished INT-777's inhibition of TNF-α release, resulting in neuronal death. Therefore, PD cognitive impairment is associated with hippocampal TNF-α elevation as a result of mitochondrial damage in microglia. Our data reveal the potential role of TGR5 in modulating inflammation-mediated neurodegeneration in PD, and provides new insights for bile acid metabolites as promising disease-modifying drugs for PD.

TGR5, Not Only a Metabolic Regulator

G-protein-coupled bile acid receptor, Gpbar1 (TGR5), is a member of G-protein-coupled receptor (GPCR) superfamily. High levels of TGR5 mRNA were detected in several tissues such as small intestine, stomach, liver, lung, especially in placenta and spleen. TGR5 is not only the receptor for bile acids, but also the receptor for multiple selective synthetic agonists such as 6α-ethyl-23(S)-methyl-cholic acid (6-EMCA, INT-777) and a series of 4-benzofuranyloxynicotinamde derivatives to regulate different signaling pathways such as nuclear factor κB (NF-κB), AKT, and extracellular signal-regulated kinases (ERK). TGR5, as a metabolic regulator, is involved in energy homeostasis, bile acid homeostasis, as well as glucose metabolism. More recently, our group and others have extended the functions of TGR5 to more than metabolic regulation, which include inflammatory response, cancer and liver regeneration. These findings highlight TGR5 as a potential drug target for different diseases. This review summarizes the basic information of TGR5 and its new functions.

FXR/TGR5 Dual Agonist Prevents Progression of Nephropathy in Diabetes and Obesity

Bile acids are ligands for the nuclear hormone receptor farnesoid X receptor (FXR) and the G protein-coupled receptor TGR5. We have shown that FXR and TGR5 have renoprotective roles in diabetes- and obesity-related kidney disease. Here, we determined whether these effects are mediated through differential or synergistic signaling pathways. We administered the FXR/TGR5 dual agonist INT-767 to DBA/2J mice with streptozotocin-induced diabetes, db/db mice with type 2 diabetes, and C57BL/6J mice with high-fat diet-induced obesity. We also examined the individual effects of the selective FXR agonist obeticholic acid (OCA) and the TGR5 agonist INT-777 in diabetic mice. The FXR agonist OCA and the TGR5 agonist INT-777 modulated distinct renal signaling pathways involved in the pathogenesis and treatment of diabetic nephropathy. Treatment of diabetic DBA/2J and db/db mice with the dual FXR/TGR5 agonist INT-767 improved proteinuria and prevented podocyte injury, mesangial expansion, and tubulointerstitial fibrosis. INT-767 exerted coordinated effects on multiple pathways, including stimulation of a signaling cascade involving AMP-activated protein kinase, sirtuin 1, PGC-1α, sirtuin 3, estrogen-related receptor-α, and Nrf-1; inhibition of endoplasmic reticulum stress; and inhibition of enhanced renal fatty acid and cholesterol metabolism. Additionally, in mice with diet-induced obesity, INT-767 prevented mitochondrial dysfunction and oxidative stress determined by fluorescence lifetime imaging of NADH and kidney fibrosis determined by second harmonic imaging microscopy. These results identify the renal signaling pathways regulated by FXR and TGR5, which may be promising targets for the treatment of nephropathy in diabetes and obesity.