Home>>Signaling Pathways>> Chromatin/Epigenetics>> PAD>>BB-Cl-Amidine

BB-Cl-Amidine Sale

目录号 : GC31755

BB-Cl-Amidine 是一种肽精氨酸脱氨酶 (PAD) 抑制剂。

BB-Cl-Amidine Chemical Structure

Cas No.:1802637-39-3

规格 价格 库存 购买数量
10mM (in 1mL DMSO)
¥1,355.00
现货
1mg
¥670.00
现货
5mg
¥1,339.00
现货
10mg
¥2,321.00
现货
25mg
¥5,266.00
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:

实验参考方法

Cell experiment [1]:

Cell lines

MCF10DCIS cells

Preparation Method

MCF10DCIS cells were grown in soft agar at different concentrations of BB-CL-AMIDINE (0 µM (DMSO), or 1 µM BB-CL-AMIDINE). After 2.5 weeks, individual colonies larger than 70 µm were counted.

Reaction Conditions

0 µM or 1 µM; 2.5 weeks

Applications

There was an average of 3,536 colonies in the DMSO control whereas only 1,967 colonies were seen in the BB-CL-AMIDINE treated group after 2.5 weeks of soft agar culture. This represents a 44% decrease in the average colony formation in the presence of 1 µM BB-CL-AMIDINE, indicating a significant tumorigenic inhibition of breast cancer cells (MCF10DCIS cells) by the PADI inhibitor.

Animal experiment [2]:

Animal models

Eight-week-old female NOD mice

Dosage form

1 μg/g; s.c.

Preparation method

Eight-week-old female NOD mice were used. Treatments involved subcutaneous injections with BB-Cl-amidine (1 μg/g body weight) or vehicle (25% DMSO in PBS) six times per week until 25 weeks of age for diabetes incidence or until 13 weeks of age for mechanistic studies

Applications

BB-Cl-amidine treatment fully prevented diabetes development, with all mice free of diabetes until 25 weeks of age, against 44% diabetes free in DMSO-treated mice.

References:

[1] Horibata S, et al. Utilization of the Soft Agar Colony Formation Assay to Identify Inhibitors of Tumorigenicity in Breast Cancer Cells. J Vis Exp. 2015 May 20;(99):e52727.

[2] Sodré FMC, et al. Peptidylarginine Deiminase Inhibition Prevents Diabetes Development in NOD Mice. Diabetes. 2021 Feb;70(2):516-528.

产品描述

BB-Cl-Amidine, a peptidylarginine deminase (PAD) inhibitor, is frequently used to study PAD function.[1]

In vitro experiment it shown that after 48 h of treatment with 0 to 20 μM BB-Cl-Amidine caused a dose-dependent decrease in cell viability in canine and feline mammary tumor cells.[1] In vitro, Cl-amidine and BB-Cl-Amidine show similar potencies and selectivities in U2OS cells, the cellular potency of BB-Cl-Amidine is increased by more than 20-fold, with EC50 values of 8.8±0.6 μM in U2OS osteosarcoma cells.[5]

In vivo efficacy test it exhibited that treatment with 1 μg/ml BB-Cl-Amidine intraperitoneally for two weeks in xenograft mice, BB-Cl-Amidine-treated tumors became crusty and the surrounding skin showed hair loss. There was an or a slight increase in apoptotic cells in the BB-Cl-Amidine-treated canine or feline xenograft tumors.[1] In vitro, at concentrations around 15-20 μM and 4 μM, respectively, BB-Cl-Amidine inhibited both PAD isoforms in a dose-dependent manner with 90% inhibition of PAD2 and PAD4.[2]

In vivo, arthritic mice were treated with 10 mg/kg BB-Cl-Amidine, there was a reduction in inflammation and joint destruction.[3] In vivo, treatment with 1 mg/kg BB-Cl-Amidine intraperitoneally and Ac-YVAD-cmk (a pyroptosis inhibitor) attenuated NET levels in BALF and neutrophil infiltration in alveoli. [4] In vivo, treatment with 1 mg/kg BB-Cl-Amidine subcutaneously obviously reduced splenomegaly in MRL/lpr mice and improved endothelium-dependent vasorelaxation.[5]

References:
[1] Ledet MM, et al. BB-Cl-Amidine as a novel therapeutic for canine and feline mammary cancer via activation of the endoplasmic reticulum stress pathway. BMC Cancer. 2018 Apr 12;18(1):412.
[2] Martín Monreal MT, et al. Applicability of Small-Molecule Inhibitors in the Study of Peptidyl Arginine Deiminase 2 (PAD2) and PAD4. Front Immunol. 2021 Oct 19;12:716250.
[3] Kawalkowska J, et al. Abrogation of collagen-induced arthritis by a peptidyl arginine deiminase inhibitor is associated with modulation of T cell-mediated immune responses. Sci Rep. 2016 May 23;6:26430.
[4] Li H, Li Y, et al. Neutrophil Extracellular Traps Augmented Alveolar Macrophage Pyroptosis via AIM2 Inflammasome Activation in LPS-Induced ALI/ARDS. J Inflamm Res. 2021 Sep 21;14:4839-4858.
[5] Knight JS, et al. Peptidylarginine deiminase inhibition disrupts NET formation and protects against kidney, skin and vascular disease in lupus-prone MRL/lpr mice. Ann Rheum Dis. 2015 Dec;74(12):2199-206.

BB-Cl-Amidine 是一种肽基精氨酸脱氨酶 (PAD) 抑制剂,常用于研究 PAD 功能。[1]

体外实验表明,在用 0 至 20 μM BB-Cl-Amidine 处理 48 小时后,犬和猫乳腺肿瘤细胞的细胞活力呈剂量依赖性降低。[1] 在体外,Cl-amidine 和 BB-Cl-Amidine 在 U2OS 细胞中表现出相似的效力和选择性,BB-Cl-Amidine 的细胞效力增加超过 20 倍,在 U2OS 中的 EC50 值为 8.8±0.6 μM骨肉瘤细胞.[5]

体内药效试验表明,在异种移植小鼠中用 1 μg/ml BB-Cl-Amidine 腹腔注射两周,BB-Cl-Amidine 处理的肿瘤变得结痂,周围皮肤出现脱发。在 BB-Cl-Amidine 处理的犬科动物或猫科动物异种移植肿瘤中,凋亡细胞有所增加或略有增加。[1] 在体外,浓度分别约为 15-20 μM 和 4 μM , BB-Cl-Amidine 以剂量依赖性方式抑制两种 PAD 亚型,对 PAD2 和 PAD4 的抑制率为 90%。[2]

在体内,用 10 mg/kg BB-Cl-Amidine 治疗关节炎小鼠,炎症和关节破坏减少。[3] 在体内,用 1 mg/kg 治疗腹膜内注射 BB-Cl-脒和 Ac-YVAD-cmk(一种细胞焦亡抑制剂)可减弱 BALF 中的 NET 水平和肺泡中的中性粒细胞浸润。 [4] 在体内,皮下注射 1 mg/kg BB-Cl-Amidine 可显着减少 MRL/lpr 小鼠的脾肿大并改善内皮依赖性血管舒张。[5]

Chemical Properties

Cas No. 1802637-39-3 SDF
Canonical SMILES O=C(C1=CC=C(C2=CC=CC=C2)C=C1)N[C@H](C3=NC4=CC=CC=C4N3)CCCNC(CCl)=N
分子式 C26H26ClN5O 分子量 459.97
溶解度 DMSO : 125 mg/mL (271.76 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 2.1741 mL 10.8703 mL 21.7405 mL
5 mM 0.4348 mL 2.1741 mL 4.3481 mL
10 mM 0.2174 mL 1.087 mL 2.1741 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

Inducement of ER Stress by PAD Inhibitor BB-Cl-Amidine to Effectively Kill AML Cells

Objective: Acute myeloid leukemia (AML) is a highly heterogeneous and recurrent hematological malignancy. Despite the emergence of novel chemotherapy drugs, AML patients' complete remission (CR) remains unsatisfactory. Consequently, it is imperative to discover new therapeutic targets or medications to treat AML. Such epigenetic changes like DNA methylation and histone modification play vital roles in AML. Peptidylarginine deminase (PAD) is a protein family of histone demethylases, among which the PAD2 and PAD4 expression have been demonstrated to be elevated in AML patients, thus suggesting a potential role of PADs in the development or maintenance of AML and the potential for the identification of novel therapeutic targets. Methods: AML cells were treated in vitro with the pan-PAD inhibitor BB-Cl-Amidine (BB-Cl-A). The AML cell lines were effectively induced into apoptosis by BB-Cl-A. However, the PAD4-specific inhibitor GSK484 did not. Results: PAD2 played a significant role in AML. Furthermore, we found that BB-Cl-A could activate the endoplasmic reticulum (ER) stress response, as evidenced by an increase in phosphorylated PERK (p-PERK) and eIF2α (p-eIF2α). As a result of the ER stress activation, the BB-Cl-A effectively induced apoptosis in the AML cells. Conclusion: Our findings indicated that PAD2 plays a role in ER homeostasis maintenance and apoptosis prevention. Therefore, targeting PAD2 with BB-Cl-A could represent a novel therapeutic strategy for treating AML.

BB-Cl-Amidine as a novel therapeutic for canine and feline mammary cancer via activation of the endoplasmic reticulum stress pathway

Background: Mammary cancer is highly prevalent in dogs and cats and results in a poor prognosis due to critically lacking viable treatment options. Recent human and mouse studies have suggested that inhibiting peptidyl arginine deiminase enzymes (PAD) may be a novel breast cancer therapy. Based on the similarities between human breast cancer and mammary cancer in dogs and cats, we hypothesized that PAD inhibitors would also be an effective treatment for mammary cancer in these animals.
Methods: Canine and feline mammary cancer cell lines were treated with BB-Cl-Amidine (BB-CLA) and evaluated for viability and tumorigenicity. Endoplasmic reticulum stress was tested by western blot, immunofluorescence, and quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). Canine and feline mammary cancer xenograft models were created using NOD scid gamma (NSG) mice, and were treated with BB-CLA for two weeks.
Results: We found that BB-CLA reduced viability and tumorigenicity of canine and feline mammary cancer cell lines in vitro. Additionally, we demonstrated that BB-CLA activates the endoplasmic reticulum stress pathway in these cells by downregulating 78 kDa Glucose-regulated Protein (GRP78), a potential target in breast cancer for molecular therapy, and upregulating the downstream target gene DNA Damage Inducible Transcript 3 (DDIT3). Finally, we established a mouse xenograft model of both canine and feline mammary cancer in which we preliminarily tested the effects of BB-CLA in vivo.
Conclusion: We propose that our established mouse xenograft models will be useful for the study of mammary cancer in dogs and cats, and furthermore, that BB-CLA has potential as a novel therapeutic for mammary cancer in these species.

Neutrophil Extracellular Traps Augmented Alveolar Macrophage Pyroptosis via AIM2 Inflammasome Activation in LPS-Induced ALI/ARDS

Background: Uncontrollable inflammation is a critical feature of gram-negative bacterial pneumonia-induced acute respiratory distress syndrome (ARDS). Both neutrophils and alveolar macrophages participate in inflammation, but how their interaction augments inflammation and triggers ARDS is unclear. The authors hypothesize that neutrophil extracellular traps (NETs), which are formed during neutrophil NETosis, partly cause alveolar macrophage pyroptosis and worsen the severity of ARDS.
Methods: The authors first analysed whether NETs and caspase-1 are involved in clinical cases of ARDS. Then, the authors employed a lipopolysaccharide (LPS)-induced ARDS model to investigate whether targeting NETs or alveolar macrophages is protective. The AIM2 sensor can bind to DNA to promote AIM2 inflammasome activation, so the authors studied whether degradation of NET DNA or silencing of the AIM2 gene could protect alveolar macrophages from pyroptosis in vitro.
Results: Analysis of aspirate supernatants from ARDS patients showed that NET and caspase-1 levels were correlated with the severity of ARDS and that the levels of NETs and caspase-1 were higher in nonsurvivors than in survivors. In vivo, the NET level and proportion of pyroptotic alveolar macrophages in bronchoalveolar lavage fluid (BALF) were obviously higher in LPS-challenged mice than in control mice 24 h after injury. Administration of DNase I (a NET DNA-degrading agent) and BB-Cl-amidine (a NET formation inhibitor) alleviated alveolar macrophage pyroptosis, and Ac-YVAD-cmk (a pyroptosis inhibitor) attenuated NET levels in BALF and neutrophil infiltration in alveoli. All treatments markedly attenuated the severity of ARDS. Notably, LPS causes NETs to induce alveolar macrophage pyroptosis, and degradation of NET DNA or silencing of the AIM2 gene protected against alveolar macrophage pyroptosis.
Conclusion: These findings shed light on the proinflammatory role of NETs in mediating the neutrophil-alveolar macrophage interaction, which influences the progression of ARDS.

Protein Arginine Deiminases (PADs): Biochemistry and Chemical Biology of Protein Citrullination

Proteins are well-known to undergo a variety of post-translational modifications (PTMs). One such PTM is citrullination, an arginine modification that is catalyzed by a group of hydrolases called protein arginine deiminases (PADs). Hundreds of proteins are known to be citrullinated and hypercitrullination is associated with autoimmune diseases including rheumatoid arthritis (RA), lupus, ulcerative colitis (UC), Alzheimer's disease, multiple sclerosis (MS), and certain cancers. In this Account, we summarize our efforts to understand the structure and mechanism of the PADs and to develop small molecule chemical probes of protein citrullination. PAD activity is highly regulated by calcium. Structural studies with PAD2 revealed that calcium-binding occurs in a stepwise fashion and induces a series of dramatic conformational changes to form a catalytically competent active site. These studies also identified the presence of a calcium-switch that controls the overall calcium-dependence and a gatekeeper residue that shields the active site in the absence of calcium. Using biochemical and site-directed mutagenesis studies, we identified the key residues (two aspartates, a cysteine, and a histidine) responsible for catalysis and proposed a general mechanism of citrullination. Although all PADs follow this mechanism, substrate binding to the thiolate or thiol form of the enzyme varies for different isozymes. Substrate-specificity studies revealed that PADs 1-4 prefer peptidyl-arginine over free arginine and certain citrullination sites on a peptide substrate. Using high-throughput screening and activity-based protein profiling (ABPP), we identified several reversible (streptomycin, minocycline, and chlorotetracycline) and irreversible (streptonigrin, NSC 95397) PAD-inhibitors. Screening of a DNA-encoded library and lead-optimization led to the development of GSK199 and GSK484 as highly potent PAD4-selective inhibitors. Furthermore, use of an electrophilic, cysteine-targeted haloacetamidine warhead to mimic the guanidinium group in arginine afforded several mechanism-based pan-PAD-inhibitors including Cl-amidine and BB-Cl-amidine. These compounds are highly efficacious in various animal models, including those mimicking RA, UC, and lupus. Structure-activity relationships identified numerous covalent PAD-inhibitors with different bioavailability, in vivo stability, and isozyme-selectivity (PAD1-selective: D-Cl-amidine; PAD2-selective: compounds 16-20; PAD3-selective: Cl4-amidine; and PAD4-selective: TDFA). Finally, this Account describes the development of PAD-targeted and citrulline-specific chemical probes. While PAD-targeted probes were utilized for identifying off-targets and developing high-throughput inhibitor screening platforms, citrulline-specific probes enabled the proteomic identification of novel diagnostic biomarkers of hypercitrullination-related autoimmune diseases.

Peptidylarginine deiminase inhibition disrupts NET formation and protects against kidney, skin and vascular disease in lupus-prone MRL/lpr mice

Objectives: An imbalance between neutrophil extracellular trap (NET) formation and degradation has been described in systemic lupus erythematosus (SLE), potentially contributing to autoantigen externalisation, type I interferon synthesis and endothelial damage. We have demonstrated that peptidylarginine deiminase (PAD) inhibition reduces NET formation and protects against lupus-related vascular damage in the New Zealand Mixed model of lupus. However, another strategy for inhibiting NETs--knockout of NOX2--accelerates lupus in a different murine model, MRL/lpr. Here, we test the effects of PAD inhibition on MRL/lpr mice in order to clarify whether some NET inhibitory pathways may be consistently therapeutic across models of SLE.
Methods: NET formation and autoantibodies to NETs were characterised in lupus-prone MRL/lpr mice. MRL/lpr mice were also treated with two different PAD inhibitors, Cl-amidine and the newly described BB-Cl-amidine. NET formation, endothelial function, interferon signature, nephritis and skin disease were examined in treated mice.
Results: Neutrophils from MRL/lpr mice demonstrate accelerated NET formation compared with controls. MRL/lpr mice also form autoantibodies to NETs and have evidence of endothelial dysfunction. PAD inhibition markedly improves endothelial function, while downregulating the expression of type I interferon-regulated genes. PAD inhibition also reduces proteinuria and immune complex deposition in the kidneys, while protecting against skin disease.
Conclusions: PAD inhibition reduces NET formation, while protecting against lupus-related damage to the vasculature, kidneys and skin in various lupus models. The strategy by which NETs are inhibited will have to be carefully considered if human studies are to be undertaken.