Home>>Signaling Pathways>> Immunology/Inflammation>> Innate Immunity>>SRI-011381

SRI-011381 Sale

目录号 : GC45831

A TGF-β signaling activator

SRI-011381 Chemical Structure

Cas No.:1629138-41-5

规格 价格 库存 购买数量
10mM (in 1mL DMSO)
¥433.00
现货
1mg
¥242.00
现货
5mg
¥535.00
现货
10mg
¥924.00
现货
25mg
¥1,918.00
现货
50mg
¥3,083.00
现货
100mg
¥4,997.00
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

SRI-011381 is an activator of TGF-β signaling.1 It reduces increases in cell death and the number of dystrophic neurites induced by amyloid-β (1-42) in primary mouse embryonic forebrain neurons when used at a concentration of 3 μM. SRI-011381 (2 and 5 μM) increases phagocytosis of Aβ42 by greater than 20% in J774A.1 and THP-1 macrophages. It increases contextual fear conditioning freezing time and spontaneous alternations in the Y-maze, indicating prevention of memory deficits, in the APP751Lon,Swe transgenic mouse model of Alzheimer's disease when administered at a dose of 10 mg/kg for 10 weeks.

Chemical Properties

Cas No. 1629138-41-5 SDF
Canonical SMILES O=C(NC1CCCCC1)N(CC2CCNCC2)CC3=CC=CC=C3
分子式 C20H31N3O 分子量 329.5
溶解度 DMF: 15mg/mL,DMSO: 20mg/mL,Ethanol: 25mg/mL,Ethanol:PBS (pH 7.2) (1:4): 0.2mg/mL 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 3.0349 mL 15.1745 mL 30.349 mL
5 mM 0.607 mL 3.0349 mL 6.0698 mL
10 mM 0.3035 mL 1.5175 mL 3.0349 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

Astrocytic YAP protects the optic nerve and retina in an experimental autoimmune encephalomyelitis model through TGF-β signaling

Theranostics 2021 Jul 25;11(17):8480-8499.PMID:34373754DOI:10.7150/thno.60031.

Rationale: Optic neuritis is one of main symptoms in multiple sclerosis (MS) that causes visual disability. Astrocytes are pivotal regulators of neuroinflammation in MS, and astrocytic yes-associated protein (YAP) plays a critical role in neuroinflammation. Meanwhile, YAP signaling is involved in visual impairment, including glaucoma, retinal choroidal atrophy and retinal detachment. However, the roles and underlying mechanisms of astrocytic YAP in neuroinflammation and demyelination of MS-related optic neuritis (MS-ON) remains unclear. Methods: To assess the functions of YAP in MS-ON, experimental autoimmune encephalomyelitis (EAE, a common model of MS) was established, and mice that conditional knockout (CKO) of YAP in astrocytes, YAPGFAP-CKO mice, were successfully generated. Behavior tests, immunostaining, Nissl staining, Hematoxylin-Eosin (HE) staining, TUNEL staining, Luxol Fast Blue (LFB) staining, electron microscopy (EM), quantitative real-time PCR (qPCR), gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) by RNA sequencing were used to examine the function and mechanism of YAP signaling based on these YAPGFAP-CKO mice and EAE model mice. To further explore the potential treatment of YAP signaling in EAE, EAE mice were treated with various drugs, including SRI-011381 that is an agonist of transforming growth factor-β (TGF-β) pathway, and XMU-MP-1 which inhibits Hippo kinase MST1/2 to activate YAP. Results: We found that YAP was significantly upregulated and activated in the astrocytes of optic nerve in EAE mice. Conditional knockout of YAP in astrocytes caused more severe inflammatory infiltration and demyelination in optic nerve, and damage of retinal ganglion cells (RGCs) in EAE mice. Moreover, YAP deletion in astrocytes promoted the activation of astrocytes and microglia, but inhibited the proliferation of astrocytes of optic nerve in EAE mice. Mechanically, TGF-β signaling pathway was significantly down-regulated after YAP deletion in astrocytes. Additionally, both qPCR and immunofluorescence assays confirmed the reduction of TGF-β signaling pathway in YAPGFAP-CKO EAE mice. Interestingly, SRI-011381 partially rescued the deficits in optic nerve and retina of YAPGFAP-CKO EAE mice. Finally, activation of YAP signaling by XMU-MP-1 relieved the neuroinflammation and demyelination in optic nerve of EAE mice. Conclusions: These results suggest astrocytic YAP may prevent the neuroinflammatory infiltration and demyelination through upregulation of TGF-β signaling and provide targets for the development of therapeutic strategies tailored for MS-ON.

Botox-A improve the thyroid-associated ophthalmopathy (TAO) orbital fibroblast activation through inhibiting the TGF-β/Smad signaling

Exp Eye Res 2022 Apr;217:108971.PMID:35108585DOI:10.1016/j.exer.2022.108971.

The activation of orbital fibroblasts can result in fibrosis, finally contributing to thyroid-associated ophthalmopathy (TAO) progression. Although the effect of BTX-A on the treatment of TAO-related strabismus and upper eyelid retraction has long been recognized in clinical work, the underlying mechanism of BTX-A improving TAO-related strabismus and upper eyelid retraction has not been uncovered yet. In the present study, we successfully isolated and authenticated normal and TAO orbital fibroblasts. Compared with PBS, BTX-A and TACA exerted similar inhibitory effects on TAO orbital fibroblast proliferation and ECM production. TGF-β stimulation induced the proliferation and ECM production by TAO orbital fibroblast, which was significantly inhibited by BTX-A or TACA treatment. Under TGF-β stimulation, the inhibitory effects of BTX-A or TACA treatment on TAO orbital fibroblast proliferation and ECM production were reversed by TGF-β/Smad signaling agonist SRI-011381. Collectively, BTX-A inhibited TGF-β-induced TAO orbital fibroblast activation through inhibiting the TGF-β/Smad signaling. Considering that TACA shows no satisfactory curative effects on symptoms closely related to the function of extraocular muscles, such as eye movement and diplopia, BTX-A might be a promising agent in TAO treatment.

Recombinant TSG-6 protein inhibits the growth of capsule fibroblasts in frozen shoulder via suppressing the TGF-β/Smad2 signal pathway

J Orthop Surg Res 2021 Sep 15;16(1):564.PMID:34526039DOI:10.1186/s13018-021-02705-x.

Background: The tumor necrosis factor-stimulated gene-6 (TSG-6) has been confirmed to inhibit inflammation. It is now generally accepted that local inflammatory stimulation around shoulder capsule causes proliferative fibrosis. This study aims to investigate the mechanism of recombinant TSG-6 protein inhibiting the growth of capsule fibroblasts in frozen shoulder via the TGF-β/Smad2 signal pathway. Methods: Human frozen shoulder capsule tissue was taken for primary and passage culture, and the 3rd generation fibroblasts from pathological frozen shoulder capsule were treated with different concentrations of recombinant TSG-6 protein, or with TGF-β1 agonist SRI-011381. Immunoconfocal analysis was used to identify the isolated fibroblasts, and MTT assay, colony formation assay, and flow cytometry were used to detect the viability, proliferation, and apoptosis rate of fibroblast. The contents of fibrosis and inflammation indexes COL1A1, TNF-α, IL-6, and IL-1β in the cell supernatant were detected using ELISA and then further examined by qRT-PCR. The expression of Bax, Bcl-2, and proteins related to TGF-β/Smad2 pathway were detected by Western Blot. Results: Compared with the blank control group, fibroblasts intervened with TSG-6 (2 μg and 5 μg) showed significantly decreased viability and proliferation ability and enhanced cell apoptosis, concurrent with the reductions in Bcl-2 expression; COL1A1, TNF-α, IL-6, and IL-1β levels; and the expression of TGF-β1 and phosphorylated Smad22, and an increase in Bax expression, while SRI-011381 treatment would reverse the effect of recombinant TSG-6 protein. Conclusion: Recombinant TSG-6 protein inhibited the growth of primary fibroblasts from human frozen shoulder capsule by suppressing the TGF-β/Smad2 signaling pathway.

Bone mesenchymal stem cell-derived extracellular vesicles promote the repair of intervertebral disc degeneration by transferring microRNA-199a

Cell Cycle 2021 Feb;20(3):256-270.PMID:33499725DOI:10.1080/15384101.2020.1863682.

Extracellular vesicles (EVs) secreted by bone marrow mesenchymal stem cells (BMSCs) protect intervertebral disc degeneration (IDD) by regulating nucleus pulposus cell (NPC) apoptosis. But the mechanism of BMSCs-EVs-microRNA (miR)-199a in IDD remains unclear. In this study, after the acquisition and identification of BMSCs and BMSCs-EVs, IDD mouse model was established and treated with BMSCs-EVs. The pathological changes of NPCs, positive expression of MMP-2, MMP-6 and TIMP1, and the senescence and apoptosis of NPCs were evaluated. Microarray analysis was employed to analyze the differentially expressed miRs and genes after EV treatment. NPCs were treated with EVs/miR-199a/TGF-β agonist SRI-011381. The positive expression of col II and Aggrecan was assessed. The target gene and downstream pathway of miR-199a were analyzed. In vivo experiment, after BMSCs-EV treatment, MMP-2, MMP-6, TIMP1 and TUNEL-positive cells in IDD mice were decreased, and miR-199a was increased. In vitro experiments, the expression of col Ⅱ and Aggrecan, SA-β gal positive cells and apoptosis rate of NPCs were decreased after EV intervention. The protective effect of BMSCs-EVs on NPCs was impaired by reducing miR-199a carried by EVs. miR-199a could target GREM1 to inactivate the TGF-β pathway. miR-199a carried by BMSCs-EVs promotes IDD repair by targeting GREM1 and downregulating the TGF-β pathway. Our work confers a promising therapeutic strategy for IDD.

Down-regulation of Gremlin1 inhibits inflammatory response and vascular permeability in chronic idiopathic urticaria through suppression of TGF-β signaling pathway

Gene 2020 Sep 25;756:144916.PMID:32580008DOI:10.1016/j.gene.2020.144916.

Chronic idiopathic urticaria (CIU) is an unfavorable skin condition which could be maintained for six weeks or longer time. Gremlin1 (GREM1) was recently applied in treatments of many diseases. However, the possible regulatory mechanism of GREM1 in CIU remained unclear. This study aimed to explore the regulatory effects of GREM1 on the inflammatory response and vascular permeability mediated by mast cells of CIU via TGF-β signaling pathway. Initially, microarray analysis was used to identify CIU-related differentially expressed genes and the potential mechanism of this gene. A mouse model of CIU was established. To explore the functional role of GREM1 in CIU, the modeled mice were then injected with GREM1-siRNA, SRI-011381 (the activator of TGF-β signaling pathway), or both, followed by serum test, and immunoglobulin detection. The levels of inflammatory factors and tryptase, β-hexosaminase, histamine in the serum were detected. Besides, vascular endothelial cell permeability and the target relation between GREM1 and TGF-β were also examined. Mice injected with SRI-011381 exhibited higher levels of tryptase, β-hexosaminase, histamine, inflammation-related factors and increased vascular endothelial cell permeability, while GREM1-silenced mice yet expressed opposite tendency. Silencing of GREM1 was demonstrated to inhibit the TGF-β signaling pathway. Taken together, our results demonstrated that down-regulation of GREM1 could potentially impede inflammatory response and vascular permeability by suppressing TGF-β signaling pathway. GREM1 may promote the development of prognosis management and therapeutic treatment in CIU.