Home>>Signaling Pathways>> Immunology/Inflammation>> STING>>C-176 (STING inhibitor 1)

C-176 (STING inhibitor 1) Sale

(Synonyms: N-(4-碘苯基)-5-硝基呋喃-2-甲酰胺) 目录号 : GC33823

C-176 (STING inhibitor 1) 是一种强效共价小鼠 STING 抑制剂。

C-176 (STING inhibitor 1) Chemical Structure

Cas No.:314054-00-7

规格 价格 库存 购买数量
10mM (in 1mL DMSO)
¥982.00
现货
5mg
¥893.00
现货
10mg
¥1,250.00
现货
50mg
¥2,588.00
现货
100mg
¥4,016.00
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

Hcc-1806 cell line

Preparation Method

Cells were treated in C-176 (STING inhibitor 1) for 8 hours at the indicated concentration,

Reaction Conditions

10 µM C-176 (STING inhibitor 1) for 8hs

Applications

C-176 (STING inhibitor 1) induced apoptosis in HCC-1806 cells, C-176 (STING inhibitor 1) induced CHOP expression in HCC-1806 cells.

Animal experiment [2]:

Animal models

Trex1 - / - mice(2-5 weeks of age)

Preparation Method

For toxicology studies, 8-week-old mice were injected daily with 562.5 nmol of C-176 for 2 weeks. At day 14, blood samples were collected in lithium-heparin-coated tubes.

Dosage form

562.5 nmol of C-176 (STING inhibitor 1) for 2 weeks

Applications

Treatment of Trex1-/- mice with C-176 (STING inhibitor 1) resulted in a significant reduction in serum levels of type I IFNs and in a strong suppression of inflammatory parameters in the heart.We next conducted a three-month trial with C-176 (STING inhibitor 1) in Trex1-/- mice, which demonstrated marked amelioration of various signs of systemic inflammation. Thus, C-176 (STING inhibitor 1) attenuates STING-associated autoinflammatory disease in mice.

References:

[1]: Duan H, Li Y, et,al. Identification of 5-nitrofuran-2-amide derivatives that induce apoptosis in triple negative breast cancer cells by activating C/EBP-homologous protein expression. Bioorg Med Chem. 2015 Aug 1;23(15):4514-4521. doi: 10.1016/j.bmc.2015.06.011. Epub 2015 Jun 14. PMID: 26116180; PMCID: PMC5567983.
[2]: Haag SM, Gulen MF, et,al.Targeting STING with covalent small-molecule inhibitors. Nature. 2018 Jul;559(7713):269-273. doi: 10.1038/s41586-018-0287-8. Epub 2018 Jul 4. PMID: 29973723.

产品描述

C-176 (STING inhibitor 1) strongly reduces STING-mediated, but not RIG-I- or TBK1-mediated, IFNβ reporter activity. It directly targets mouse STING (mmSTING) but not human STING (hsSTING)[1].

Cytotoxicity against human HCC1806 cells assessed as reduction in cell viability incubated for 3 days by cell-titer-Glo reagent based assay, IC50 = 6.2 μM. C-176 (STING inhibitor 1) induced apoptosis in HCC-1806 cells, C-176 (STING inhibitor 1) induced CHOP expression in HCC-1806 cells[2].STING was mainly distributed in microglia, and microglial STING expression was significantly increased after SAH. Administration of C-176 (STING inhibitor 1) substantially attenuated SAH-induced brain edema and neuronal injury. More importantly, C-176 (STING inhibitor 1) significantly alleviated both short-term and persistent neurological dysfunction after SAH[3].Inhibition of mmSTING by C-176 (STING inhibitor 1) enhanced type H vessels' formation, implying osteogenesis promotion in bone healing (higher bone volume density and more OCN-positive cells). STING inhibition accelerates the bone healing process while enhancing type H vessel formation[7].

C-176 (STING inhibitor 1) covalently binds to Cys91 of STING preventing activation via blockade of palmitoylation at Cys91. Treatment of Trex-/- mice with C-176 (STING inhibitor 1) resulted in a significant reduction in serum levels of Type I Interferons and amelioration of systemic inflammation[1]. When explored the molecular mechanisms of STING in regulating lipopolysaccharide (LPS)-induced lung injury. Mice were pretreated with a STING inhibitor C-176 (STING inhibitor 1) (15, 30 mg/kg) before LPS inhalation to induce ALI. LPS inhalation significantly increased STING expression in the lung tissues, whereas C-176 (STING inhibitor 1) pretreatment dose-dependently suppressed the expression of STING, decreased the production of inflammatory cytokines including TNF-α, IL-6, IL-12, and IL-1β, and restrained the expression of chemokines and adhesion molecule vascular cell adhesion protein-1 (VCAM-1) in the lung tissues[4].Genetic deletion of Sting in Apoe-/- mice reduced atherosclerotic lesions in the aortic arch, lipid, and macrophage accumulation in plaques, and inflammatory molecule expression in the aorta. Pharmacological blockade of STING using a specific inhibitor, C-176 (STING inhibitor 1), ameliorated atherogenesis in Apoe-/- mice[5]. Treatment with the STING inhibitor, C-176, suppressed EBV-induced transformation in peripheral blood mononuclear cells. In an EBV-LPD mouse model, C-176 (STING inhibitor 1) treatment also inhibited tumor formation and prolonged survival[6].

References:
[1]: Haag SM, Gulen MF, et,al. Targeting STING with covalent small-molecule inhibitors. Nature. 2018 Jul;559(7713):269-273. doi: 10.1038/s41586-018-0287-8. Epub 2018 Jul 4. PMID: 29973723.
[2]: Duan H, Li Y, et,al. Identification of 5-nitrofuran-2-amide derivatives that induce apoptosis in triple negative breast cancer cells by activating C/EBP-homologous protein expression. Bioorg Med Chem. 2015 Aug 1;23(15):4514-4521. doi: 10.1016/j.bmc.2015.06.011. Epub 2015 Jun 14. PMID: 26116180; PMCID: PMC5567983.
[3]: Peng Y, Zhuang J, et,al. Stimulator of IFN genes mediates neuroinflammatory injury by suppressing AMPK signal in experimental subarachnoid hemorrhage. J Neuroinflammation. 2020 May 25;17(1):165. doi: 10.1186/s12974-020-01830-4. PMID: 32450897; PMCID: PMC7247752.
[4]: Li Y, Su J, et,al. Targeted next-generation sequencing of deaf patients from Southwestern China. Mol Genet Genomic Med. 2021 Apr;9(4):e1660. doi: 10.1002/mgg3.1660. Epub 2021 Mar 16. PMID: 33724713; PMCID: PMC8123756.
[5]: Pham PT, Fukuda D, et,al. STING, a cytosolic DNA sensor, plays a critical role in atherogenesis: a link between innate immunity and chronic inflammation caused by lifestyle-related diseases. Eur Heart J. 2021 Nov 7;42(42):4336-4348. doi: 10.1093/eurheartj/ehab249. PMID: 34226923.
[6]: Miyagi S, Watanabe T, et,al. A STING inhibitor suppresses EBV-induced B cell transformation and lymphomagenesis. Cancer Sci. 2021 Dec;112(12):5088-5099. doi: 10.1111/cas.15152. Epub 2021 Oct 11. PMID: 34609775; PMCID: PMC8645724.
[7]: Chen X, He W, et,al. STING inhibition accelerates the bone healing process while enhancing type H vessel formation. FASEB J. 2021 Nov;35(11):e21964. doi: 10.1096/fj.202100069RR. PMID: 34694030.

C-176(STING 抑制剂 1)强烈降低 STING 介导的,但不降低 RIG-I 或 TBK1 介导的 IFNβ 报告基因活性。它直接针对小鼠 STING (mmSTING) 而不是人类 STING (hsSTING)[1]

对人 HCC1806 细胞的细胞毒性通过基于细胞滴度-Glo 试剂的测定评估为细胞活力降低 3 天,IC50 = 6.2 μM。 C-176(STING抑制剂1)诱导HCC-1806细胞凋亡,C-176(STING抑制剂1)诱导HCC-1806细胞表达CHOP[2]。STING主要分布于小胶质细胞, SAH 后小胶质细胞 STING 表达显着增加。 C-176(STING 抑制剂 1)的给药显着减轻了 SAH 引起的脑水肿和神经元损伤。更重要的是,C-176(STING 抑制剂 1)显着缓解 SAH 后的短期和持续性神经功能障碍[3]。C-176(STING 抑制剂 1)增强 H 型血管对 mmSTING 的抑制作用' 形成,意味着骨愈合中的成骨促进(更高的骨体积密度和更多的 OCN 阳性细胞)。抑制 STING 可加速骨愈合过程,同时促进 H 型血管形成[7]

C-176(STING 抑制剂 1)共价结合 STING 的 Cys91,通过阻断 Cys91 处的棕榈酰化来防止激活。用 C-176(STING 抑制剂 1)处理 Trex-/- 小鼠可显着降低血清 I 型干扰素水平并改善全身炎症[1]。当探索 STING 调节脂多糖 (LPS) 诱导的肺损伤的分子机制时。在吸入 LPS 以诱导 ALI 之前,用 STING 抑制剂 C-176(STING 抑制剂 1)(15、30 mg/kg)预处理小鼠。 LPS 吸入显着增加肺组织中的 STING 表达,而 C-176(STING 抑制剂 1)预处理剂量依赖性地抑制 STING 的表达,减少炎症细胞因子的产生,包括 TNF-α、IL-6、IL-12 和IL-1β,抑制肺组织中趋化因子和粘附分子血管细胞粘附蛋白-1(VCAM-1)的表达[4]。Apoe-/-小鼠Sting基因缺失减少主动脉弓的动脉粥样硬化病变、斑块中的脂质和巨噬细胞聚集以及主动脉中炎症分子的表达。使用特异性抑制剂 C-176(STING 抑制剂 1)对 STING 进行药理学阻断,可改善 Apoe-/- 小鼠的动脉粥样硬化形成[5]。用 STING 抑制剂 C-176 治疗可抑制 EBV 诱导的外周血单核细胞转化。在 EBV-LPD 小鼠模型中,C-176(STING 抑制剂 1)治疗还可抑制肿瘤形成并延长生存期[6]

Chemical Properties

Cas No. 314054-00-7 SDF
别名 N-(4-碘苯基)-5-硝基呋喃-2-甲酰胺
Canonical SMILES O=C(C1=CC=C([N+]([O-])=O)O1)NC2=CC=C(I)C=C2
分子式 C11H7IN2O4 分子量 358.09
溶解度 DMSO : 150 mg/mL (418.89 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 2.7926 mL 13.963 mL 27.9259 mL
5 mM 0.5585 mL 2.7926 mL 5.5852 mL
10 mM 0.2793 mL 1.3963 mL 2.7926 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

STING inhibitor ameliorates LPS-induced ALI by preventing vascular endothelial cells-mediated immune cells chemotaxis and adhesion

Acta Pharmacol Sin 2022 Aug;43(8):2055-2066.PMID:PMC9343420DOI:10.1038/s41401-021-00813-2.

Acute lung injury (ALI) is a common and devastating clinical disorder featured by excessive inflammatory responses. Stimulator of interferon genes (STING) is an indispensable molecule for regulating inflammation and immune response in multiple diseases, but the role of STING in the ALI pathogenesis is not well elucidated. In this study, we explored the molecular mechanisms of STING in regulating lipopolysaccharide (LPS)-induced lung injury. Mice were pretreated with a STING inhibitor C-176 (15, 30 mg/kg, i.p.) before LPS inhalation to induce ALI. We showed that LPS inhalation significantly increased STING expression in the lung tissues, whereas C-176 pretreatment dose-dependently suppressed the expression of STING, decreased the production of inflammatory cytokines including TNF-α, IL-6, IL-12, and IL-1β, and restrained the expression of chemokines and adhesion molecule vascular cell adhesion protein-1 (VCAM-1) in the lung tissues. Consistently, in vitro experiments conducted in TNF-α-stimulated HMEC-1cells (common and classic vascular endothelial cells) revealed that human STING inhibitor H-151 or STING siRNA downregulated the expression levels of adhesion molecule and chemokines in HMEC-1cells, accompanied by decreased adhesive ability and chemotaxis of immunocytes upon TNF-α stimulation. We further revealed that STING inhibitor H-151 or STING knockdown significantly decreased the phosphorylation of transcription factor STAT1, which subsequently influenced its binding to chemokine CCL2 and adhesive molecule VCAM-1 gene promoter. Collectively, STING inhibitor can alleviate LPS-induced ALI in mice by preventing vascular endothelial cells-mediated immune cell chemotaxis and adhesion, suggesting that STING may be a promising therapeutic target for the treatment of ALI.

STING, a cytosolic DNA sensor, plays a critical role in atherogenesis: a link between innate immunity and chronic inflammation caused by lifestyle-related diseases

Eur Heart J 2021 Nov 7;42(42):4336-4348.PMID:34226923DOI:10.1093/eurheartj/ehab249.

Aims: Lifestyle-related diseases promote atherosclerosis, a chronic inflammatory disease; however, the molecular mechanism remains largely unknown. Endogenous DNA fragments released under over-nutrient condition provoke sterile inflammation through the recognition by DNA sensors. Here, we investigated the role of stimulator of interferon genes (STING), a cytosolic DNA sensor, in atherogenesis. Methods and results: Apolipoprotein E-deficient (Apoe-/-) mice fed a western-type diet (WTD), a hypercholesterolaemic mouse model, showed higher STING expression and markers for DNA damage such as γH2AX, p53, and single-stranded DNA (ssDNA) accumulation in macrophages in the aorta compared with wild-type (WT) mice. The level of cGAMP, a STING agonist, in the aorta was higher in Apoe-/- mice. Genetic deletion of Sting in Apoe-/- mice reduced atherosclerotic lesions in the aortic arch, lipid, and macrophage accumulation in plaques, and inflammatory molecule expression in the aorta compared with the control. Pharmacological blockade of STING using a specific inhibitor, C-176, ameliorated atherogenesis in Apoe-/- mice. In contrast, bone marrow-specific STING expression in Apoe-/- mice stimulated atherogenesis. Expression or deletion of STING did not affect metabolic parameters and blood pressure. In vitro studies revealed that STING activation by cGAMP or mitochondrial DNA accelerated inflammatory molecule expression (e.g. TNF-α or IFN-β) in mouse and human macrophages. Activation of nuclear factor-κB and TANK binding kinase 1 was involved in STING-associated vascular inflammation and macrophage activation. Furthermore, human atherosclerotic lesions in the carotid arteries expressed STING and cGAMP. Conclusion: Stimulator of interferon genes stimulates pro-inflammatory activation of macrophages, leading to the development of atherosclerosis. Stimulator of interferon genes signalling may serve as a potential therapeutic target for atherosclerosis.

STING mediates neuroinflammatory response by activating NLRP3-related pyroptosis in severe traumatic brain injury

J Neurochem 2022 Sep;162(5):444-462.PMID:35892155DOI:10.1111/jnc.15678.

Long-term neurological deficits after severe traumatic brain injury (TBI), including cognitive dysfunction and emotional impairments, can significantly impair rehabilitation. Glial activation induced by inflammatory response is involved in the neurological deficits post-TBI. This study aimed to investigate the role of the stimulator of interferon genes (STING)-nucleotide-binding oligomerization domain-like receptor pyrin domain-containing-3 (NLRP3) signaling in a rodent model of severe TBI. Severe TBI models were established using weight-drop plus blood loss reinfusion model. Selective STING agonist ADU-S100 or antagonist C-176 was given as a single dose after modeling. Further, NLRP3 inhibitor MCC950 or activator nigericin, or caspase-1 inhibitor VX765, was given as an intracerebroventricular injection 30 min before modeling. After that, a novel object recognition test, open field test, force swimming test, western blot, and immunofluorescence assays were used to assess behavioral and pathological changes in severe TBI. Administration of C-176 alleviated TBI-induced cognitive dysfunction and emotional impairments, neuronal loss, and inflammatory activation of glia cells. However, the administration of STING agonist ADU-S100 exacerbated TBI-induced behavioral and pathological changes. In addition, STING activation exacerbated pyroptosis-associated neuroinflammation via promoting glial activation, as evidenced by increased cleaved caspase-1 and GSDMD N-terminal expression. In contrast, the administration of C-176 showed anti-pyroptotic effects. The neuroprotective effects of C-176 were partially reversed by the NLRP3 activator, nigericin. Collectively, glial STING is responsible for neuroinflammation post-TBI. However, pharmacologic inhibition of STING led to a remarkable improvement of neuroinflammation partly through suppressing NLRP3 signaling. The STING-NLRP3 signaling is a potential therapeutic target in TBI-induced neurological dysfunction.

Lipotoxicity-induced mtDNA release promotes diabetic cardiomyopathy by activating the cGAS-STING pathway in obesity-related diabetes

Cell Biol Toxicol 2023 Feb;39(1):277-299.PMID:35235096DOI:10.1007/s10565-021-09692-z.

Diabetic cardiomyopathy (DCM) is characterized by lipid accumulation, mitochondrial dysfunction, and aseptic inflammatory activation. Mitochondria-derived cytosolic DNA has been reported to induce inflammation by activating cyclic GMP-AMP synthase (cGAS)/the stimulator of interferon genes (STING) pathway in the adipose, liver, and kidney tissues. However, the role of cytosolic mtDNA in the progression of DCM is unclear. In this study, with an obesity-related DCM mouse model established by feeding db/db mice with a high-fat diet (HFD), we observed increased mtDNA in the cytosol and activated cGAS-STING signaling pathway during DCM, as well as the downstream targets, IRF3, NF-κB, IL-18, and IL-1β. In a further study with a palmitic acid (PA)-induced lipotoxic cell model established in H9C2 cells, we revealed that the cytosolic mtDNA was the result of PA-induced overproduction of mitochondrial ROS, which also led to the activation of the cGAS/STING system and its downstream targets. Notably, treatment of extracted mtDNA alone was sufficient to activate the cGAS-STING signaling pathway in cultured H9C2 cells. Besides, both knockdown of STING in PA-induced H9C2 cells and inhibition of STING by C-176 injection in the DCM mouse model could remarkably block the inflammation and apoptosis of cardiomyocytes. In conclusion, our study elucidated the critical role of cytosolic mtDNA-induced cGAS-STING activation in the pathogenesis of obesity-related DCM and provided preclinical validation for using a STING inhibitor as a new potential therapeutic strategy for the treatment of DCM.

Stimulator of IFN genes mediates neuroinflammatory injury by suppressing AMPK signal in experimental subarachnoid hemorrhage

J Neuroinflammation 2020 May 25;17(1):165.PMID:32450897DOI:10.1186/s12974-020-01830-4.

Background: Neuroinflammation is closely associated with the poor prognosis in subarachnoid hemorrhage (SAH) patients. This study was aimed to determine the role of stimulator of IFN genes (STING), an essential regulator to innate immunity, in the context of SAH. Methods: A total of 344 male C57BL/6 J mice were subjected to endovascular perforation to develop a model of SAH. Selective STING antagonist C-176 and STING agonist CMA were administered at 30 min or 1 h post-modeling separately. To investigate the underlying mechanism, the AMPK inhibitor compound C was administered intracerebroventricularly at 30 min before surgery. Post-SAH assessments included SAH grade, neurological test, brain water content, western blotting, RT-PCR, and immunofluorescence. Oxygenated hemoglobin was introduced into BV2 cells to establish a SAH model in vitro. Results: STING was mainly distributed in microglia, and microglial STING expression was significantly increased after SAH. Administration of C-176 substantially attenuated SAH-induced brain edema and neuronal injury. More importantly, C-176 significantly alleviated both short-term and persistent neurological dysfunction after SAH. Meanwhile, STING agonist CMA remarkably exacerbated neuronal injury and deteriorated neurological impairments. Mechanically, STING activation aggravated neuroinflammation via promoting microglial activation and polarizing into M1 phenotype, evidenced by microglial morphological changes, as well as the increased level of microglial M1 markers including IL-1β, iNOS, IL-6, TNF-α, MCP-1, and NLRP3 inflammasome, while C-176 conferred a robust anti-inflammatory effect. However, all the mentioned beneficial effects of C-176 including alleviated neuroinflammation, attenuated neuronal injury and the improved neurological function were reversed by AMPK inhibitor compound C. Meanwhile, the critical role of AMPK signal in C-176 mediated anti-inflammatory effect was also confirmed in vitro. Conclusion: Microglial STING yielded neuroinflammation after SAH, while pharmacologic inhibition of STING could attenuate SAH-induced inflammatory injury at least partly by activating AMPK signal. These data supported the notion that STING might be a potential therapeutic target for SAH.