Home>>Signaling Pathways>> Immunology/Inflammation>> TLR>>Poly(I:C)

Poly(I:C) Sale

(Synonyms: 聚肌苷-聚胞苷酸复合物; Polyinosinic-polycytidylic acid) 目录号 : GC14710

Poly(I:C)是一种合成的双链 RNA (dsRNA),它是一种 Toll 样受体 3 (TLR3) 激动剂。

Poly(I:C) Chemical Structure

Cas No.:24939-03-5

规格 价格 库存 购买数量
10mM (in 1mL Water)
¥389.00
现货
10mg
¥714.00
现货
50mg
¥2,200.00
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

BEAS-2B cells

Preparation Method

BEAS-2B cells were seeded in 6-well plates one day before Poly (I:C) treatment. One hour before infection at multiplicity of infection (MOI) 0.01, 1, or 5, the cells were pre-treated with 4 µg/ml of Poly (I:C) or left untreated. After adsorption, the cells were maintained in the medium with or without Poly (I: C) (4 µg/ml). The CPE was observed at 24, 48, and 72 h postinfection (p.i.) under a microscope.

Reaction Conditions

4 µg/ml Poly (I: C) for 24/48/72 h

Applications

Poly (I:C) suppressed cytopathic effects (CPE) induced by CHIKV infection in BEAS-2B cells in the presence of Poly (I:C) and inhibited the replication of CHIKV in the cells. The virus titers of Poly (I:C)-treated cells were much lower compared with those of untreated cells. CHIKV infection and Poly (I:C) treatment of BEAS-2B cells induced the production of IFN-β and increased the expression of anti-viral genes, including IFN-α, IFN-β, MxA, and OAS. Both Poly (I:C) and CHIKV infection upregulate the expression of TLR3 in BEAS-2B cells.

Animal experiment [2]:

Animal models

C57BL/6J mouse

Preparation Method

Injection of MC38 cells into C57BL/6J mice and the subsequent intraperitoneal injections of Poly(I:C),Mice were treated with 50 µg Poly(I:C) by intraperitoneal injection on days 8, 11, 14, 17, 20 and 23.

Dosage form

50 µg Poly(I:C) by intraperitoneal injection on days 8, 11, 14, 17, 20 and 23.

Applications

Poly(I:C)-activated macrophages displayed enhanced phagocytosis after CD47 blockade. Compared with mice receiving monotherapy, those received Poly(I:C) in combination with anti-CD47 mAb exhibited significantly inhibited tumor growth. Moreover, in vivo delivery of Poly(I:C) plus anti-CD47 mAb to tumor-bearing mice altered the tumor immune microenvironment.

References:

[1]: Li YG, Siripanyaphinyo U, et,al. Poly (I:C), an agonist of toll-like receptor-3, inhibits replication of the Chikungunya virus in BEAS-2B cells. Virol J. 2012 Jun 14;9:114. doi: 10.1186/1743-422X-9-114. PMID: 22698190; PMCID: PMC3490739.
[2]: Zhong C, Wang L, et,al. Poly(I:C) enhances the efficacy of phagocytosis checkpoint blockade immunotherapy by inducing IL-6 production. J Leukoc Biol. 2021 Dec;110(6):1197-1208. doi: 10.1002/JLB.5MA0421-013R. Epub 2021 May 14. PMID: 33988261.

产品描述

Poly (I:C), a synthetic double-stranded RNA (dsRNA) analog, is an immunostimulant that acts as the most potent interferon (IFN) inducer. Toll-like receptor- 3 (TLR3) agonist[1].

Poly (I:C) suppressed cytopathic effects (CPE) induced by CHIKV infection in BEAS-2B cells in the presence of Poly (I:C) and inhibited the replication of CHIKV in the cells. Both Poly (I:C) and CHIKV infection upregulate the expression of TLR3 in BEAS-2B cells[1].DsRNA poly(I:C) up-regulated the expression of IFNβ and apoptosis-associated genes in cervical cancer cells, activating both intrinsic and extrinsic apoptotic pathways, and eventually inducing cell death[8].Stable maturation of DC can be simply induced by the addition of polyriboinosinic polyribocytidylic acid (poly(I:C))[3].Poly(I:C) decreased the rate of successful in vitro fertilization via DNA damage and abnormal spindle morphology at the first cleavage and inhibited early embryonic development by inducing immune response and promoting blastocyst cell apoptosis[9]. Mixtures that included the TLR7/8 agonists R848 or CL075, combined with the Poly (I:C), yield 3-d mature dendritic cells [2].

Poly(I:C)-activated macrophages displayed enhanced phagocytosis after CD47 blockade. Compared with mice receiving monotherapy, those received Poly(I:C) in combination with anti-CD47 mAb exhibited significantly inhibited tumor growth[4]. In vivo, poly(I:C) enhanced cell surface expression of Mac-1 on neutrophils in mice and facilitated their infiltration to lung tissues. Poly(I:C) also downregulated thrombomodulin expression in mouse tissues and reduced its circulating soluble level in plasma[5].1.25 and 5 mg/kg poly(I:C) preconditioning significantly reduced myocardial infarct size and cardiac dysfunction. Moreover, poly(I:C) significantly promoted cell survival by restoring autophagy flux and then regulating it to an adequate level Increased autophagy protein Beclin1 and LC3II together with p62 degradation after additional chloroquine[6].In mice,following Poly I:C exposure, a significant decrease in DA-D2 receptor binding, reduction in corpus callosum calibre and MOG immunolabelling indicating demyelination and a significant decrease in locomotor activity, neuromuscular strength and motor coordination signify motor deficits and hypokinetic influence of early life viral infection[7].

聚(I:C)是一种合成的双链RNA类似物,是一种免疫刺激剂,可作为最强效的干扰素(IFN)诱导剂。它是Toll样受体-3(TLR3)激动剂[1]。

在存在Poly(I:C)的情况下,Poly(I:C)抑制了CHIKV感染引起的BEAS-2B细胞细胞病变效应(CPE),并抑制了病毒在细胞中的复制。Poly(I:C)和CHIKV感染都上调了BEAS-2B细胞中TLR3的表达[1]。DsRNA poly(I:C)上调了宫颈癌细胞中IFNβ和与凋亡相关基因的表达,激活内源性和外源性凋亡途径,并最终诱导细胞死亡[8]。通过添加聚核苷酸聚核苷酸酰化肽(poly(I:C))可以简单地诱导DC稳定成熟[3]。Poly(I:C)通过引发免疫反应和促进囊胚细胞凋亡,在第一次分裂时降低体外受精成功率并抑制早期胚胎发育[9]。包含TLR7/8激动剂R848或CL075以及Poly (I: C)混合物可产生3天成熟树突状细胞 [2]。

在CD47阻断后,聚(I:C)激活的巨噬细胞表现出增强的吞噬作用。与单一治疗组相比,接受Poly(I:C)和抗CD47 mAb联合治疗的小鼠明显抑制了肿瘤生长[4]。在体内,Poly(I:C)增强了小鼠中中性粒细胞上Mac-1的细胞表面表达,并促进它们浸润到肺组织中。Poly(I:C)还下调了小鼠组织中凝血调节蛋白原模因素(thrombomodulin)的表达,并降低其循环可溶性水平[5]。1.25和5 mg/kg Poly(I:C)预处理显著减少心肌梗死面积和心功能障碍。此外,Poly(I:C)通过恢复自噬通路并将其调节到适当水平来显著促进细胞存活,在额外使用氯喹后增加自噬蛋白Beclin1和LC3II以及p62降解[6]。在小鼠身上,在经历聚I:C暴露后,DA-D2受体结合量显着降低、胼胝体直径缩小、MOG免疫标记表明脱髓鞘,以及运动活动度、神经肌肉力量和运动协调性显着降低,这表明早期病毒感染对运动功能的影响[7]。

References:
[1]: Li YG, Siripanyaphinyo U, et,al. Poly (I:C), an agonist of toll-like receptor-3, inhibits replication of the Chikungunya virus in BEAS-2B cells. Virol J. 2012 Jun 14;9:114. doi: 10.1186/1743-422X-9-114. PMID: 22698190; PMCID: PMC3490739.
[2]: Spranger S, Javorovic M, et,al. Generation of Th1-polarizing dendritic cells using the TLR7/8 agonist CL075. J Immunol. 2010 Jul 1;185(1):738-47. doi: 10.4049/jimmunol.1000060. Epub 2010 May 28. PMID: 20511554.
[3]: Verdijk RM, Mutis T, et,al. Polyriboinosinic polyribocytidylic acid (poly(I:C)) induces stable maturation of functionally active human dendritic cells. J Immunol. 1999 Jul 1;163(1):57-61. PMID: 10384099.
[4]: Zhong C, Wang L, et,al. Poly(I:C) enhances the efficacy of phagocytosis checkpoint blockade immunotherapy by inducing IL-6 production. J Leukoc Biol. 2021 Dec;110(6):1197-1208. doi: 10.1002/JLB.5MA0421-013R. Epub 2021 May 14. PMID: 33988261.
[5]: Cai X, Panicker SR, et,al.Protective Role of Activated Protein C against Viral Mimetic Poly(I:C)-Induced Inflammation. Thromb Haemost. 2021 Nov;121(11):1448-1463. doi: 10.1055/s-0041-1726093. Epub 2021 Mar 11. PMID: 33706396; PMCID: PMC8433266.
[6]: Chen E, Chang H, et,al. Poly(I:C) attenuates myocardial ischemia/reperfusion injury by restoring autophagic function. FASEB J. 2022 May;36(5):e22317. doi: 10.1096/fj.202101220RR. PMID: 35438806.
[7]:Singh B, Dhuriya YK, et,al. Early life exposure to poly I:C impairs striatal DA-D2 receptor binding, myelination and associated behavioural abilities in rats. J Chem Neuroanat. 2021 Dec;118:102035. doi: 10.1016/j.jchemneu.2021.102035. Epub 2021 Sep 28. PMID: 34597812.
[8]: Meng X, Cui X, et,al. poly(I:C) synergizes with proteasome inhibitors to induce apoptosis in cervical cancer cells. Transl Oncol. 2022 Apr;18:101362. doi: 10.1016/j.tranon.2022.101362. Epub 2022 Feb 9. PMID: 35151092; PMCID: PMC8842080.
[9]: Wang Z, Chen S, et,al. Poly(I:C) exposure during in vitro fertilization disrupts first cleavage of mouse embryos and subsequent blastocyst development. J Reprod Immunol. 2022 Jun;151:103635. doi: 10.1016/j.jri.2022.103635. Epub 2022 Apr 30. PMID: 35525084.

Chemical Properties

Cas No. 24939-03-5 SDF
别名 聚肌苷-聚胞苷酸复合物; Polyinosinic-polycytidylic acid
化学名 ((2R,3S,4R,5R)-3,4-dihydroxy-5-(6-oxo-3H-purin-9(6H)-yl)tetrahydrofuran-2-yl)methyl dihydrogen phosphate compound with ((2R,3S,4S,5R)-5-(4-amino-2-oxopyrimidin-1(2H)-yl)-3,4-dihydroxytetrahydrofuran-2-yl)methyl dihydrogen phosphate (1:1)
Canonical SMILES O[C@@H]([C@H](N1C(NC=NC2=O)=C2N=C1)O[C@@H]3COP(O)(O)=O)[C@@H]3O.O[C@H]([C@@H](COP(O)(O)=O)O[C@H]4N5C=CC(N)=NC5=O)[C@@H]4O
分子式 C19H27N7O16P2 分子量 671.41
溶解度 ≥ 21.5mg/mL in Water 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 1.4894 mL 7.447 mL 14.894 mL
5 mM 0.2979 mL 1.4894 mL 2.9788 mL
10 mM 0.1489 mL 0.7447 mL 1.4894 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

Exploiting poly(I:C) to induce cancer cell apoptosis

TLR3 belong to the Toll-like receptors family, it is mainly expressed on immune cells where it senses pathogen-associated molecular patterns and initiates innate immune response. TLR3 agonist poly(I:C) was developed to mimic pathogens infection and boost immune system activation to promote anti-cancer therapy. Accordingly, TLR agonists were included in the National Cancer Institute list of immunotherapeutic agents with the highest potential to cure cancer. Besides well known effects on immune cells, poly(I:C) was also shown, in experimental models, to directly induce apoptosis in cancer cells expressing TLR3. This review presents the current knowledge on the mechanism of poly(I:C)-induced apoptosis in cancer cells. Experimental evidences on positive or negative regulators of TLR3-mediated apoptosis induced by poly(I:C) are reported and strategies are proposed to successfully promote this event in cancer cells. Cancer cells apoptosis is an additional arm offered by poly(I:C), besides activation of immune system, for the treatment of various type of cancer. A further dissection of TLR3 signaling would contribute to greater resolution of the critical steps that impede full exploitation of the poly(I:C)-induced apoptosis. Experimental evidences about negative regulator of poly(I:C)-induced apoptotic program should be considered in combinations with TLR3 agonists in clinical trials.

Particulate formulations for the delivery of poly(I:C) as vaccine adjuvant

Current research and development of antigens for vaccination often center on purified recombinant proteins, viral subunits, synthetic oligopeptides or oligosaccharides, most of them suffering from being poorly immunogenic and subject to degradation. Hence, they call for efficient delivery systems and potent immunostimulants, jointly denoted as adjuvants. Particulate delivery systems like emulsions, liposomes, nanoparticles and microspheres may provide protection from degradation and facilitate the co-formulation of both the antigen and the immunostimulant. Synthetic double-stranded (ds) RNA, such as polyriboinosinic acid-polyribocytidylic acid, poly(I:C), is a mimic of viral dsRNA and, as such, a promising immunostimulant candidate for vaccines directed against intracellular pathogens. Poly(I:C) signaling is primarily dependent on Toll-like receptor 3 (TLR3), and on melanoma differentiation-associated gene-5 (MDA-5), and strongly drives cell-mediated immunity and a potent type I interferon response. However, stability and toxicity issues so far prevented the clinical application of dsRNAs as they undergo rapid enzymatic degradation and bear the potential to trigger undue immune stimulation as well as autoimmune disorders. This review addresses these concerns and suggests strategies to improve the safety and efficacy of immunostimulatory dsRNA formulations. The focus is on technological means required to lower the necessary dosage of poly(I:C), to target surface-modified microspheres passively or actively to antigen-presenting cells (APCs), to control their interaction with non-professional phagocytes and to modulate the resulting cytokine secretion profile.

Intratumoral combination therapy with poly(I:C) and resiquimod synergistically triggers tumor-associated macrophages for effective systemic antitumoral immunity

Background: Tumor-associated macrophages (TAMs) play a key immunosuppressive role that limits the ability of the immune system to fight cancer and hinder the antitumoral efficacy of most treatments currently applied in the clinic. Previous studies have evaluated the antitumoral immune response triggered by (TLR) agonists, such as poly(I:C), imiquimod (R837) or resiquimod (R848) as monotherapies; however, their combination for the treatment of cancer has not been explored. This study investigates the antitumoral efficacy and the macrophage reprogramming triggered by poly(I:C) combined with R848 or with R837, versus single treatments.
Methods: TLR agonist treatments were evaluated in vitro for toxicity and immunostimulatory activity by Alamar Blue, ELISA and flow cytometry using primary human and murine M-CSF-differentiated macrophages. Cytotoxic activity of TLR-treated macrophages toward cancer cells was evaluated with an in vitro functional assay by flow cytometry. For in vivo experiments, the CMT167 lung cancer model and the MN/MCA1 fibrosarcoma model metastasizing to lungs were used; tumor-infiltrating leukocytes were evaluated by flow cytometry, RT-qPCR, multispectral immunophenotyping, quantitative proteomic experiments, and protein-protein interaction analysis.
Results: Results demonstrated the higher efficacy of poly(I:C) combined with R848 versus single treatments or combined with R837 to polarize macrophages toward M1-like antitumor effectors in vitro. In vivo, the intratumoral synergistic combination of poly(I:C)+R848 significantly prevented tumor growth and metastasis in lung cancer and fibrosarcoma immunocompetent murine models. Regressing tumors showed increased infiltration of macrophages with a higher M1:M2 ratio, recruitment of CD4+ and CD8+ T cells, accompanied by a reduction of immunosuppressive CD206+ TAMs and FOXP3+/CD4+ T cells. The depletion of both CD4+ and CD8+ T cells resulted in complete loss of treatment efficacy. Treated mice acquired systemic antitumoral response and resistance to tumor rechallenge mediated by boosted macrophage cytotoxic activity and T-cell proliferation. Proteomic experiments validate the superior activation of innate immunity by poly(I:C)+R848 combination versus single treatments or poly(I:C)+R837, and protein-protein-interaction network analysis reveal the key activation of the STAT1 pathway.
Discussion: These findings demonstrate the antitumor immune responses mediated by macrophage activation on local administration of poly(I:C)+R848 combination and support the intratumoral application of this therapy to patients with solid tumors in the clinic.

Poly I:C promotes malate to enhance innate immune response against bacterial infection

Polyinosinic-polycytidylic acid (poly I:C) is a synthetic analog of double-stranded RNA (dsRNA) that activates anti-infective innate immunity. The underlying mechanisms are identified as targeting pattern recognition receptors and Th1-inducing. However, whether poly I:C manipulates metabolism to implement this anti-infective function is unknown. Here, GC-MS based metabolomics was used to characterize metabolic profiles induced by different doses of poly I:C. Analysis on the dose-dependent metabolomes shows that elevation of the TCA cycle and malate with the increasing dose of ploy I:C forms the most characteristic feature of the poly I:C stimulation. Exogenous malate activates the TCA cycle and elevates survival of zebrafish infected with Vibrio alginolyticus, which is related to the elevated expression of il-1b, il-6, il-8, tnf-a, and c3b. These results reveal a previously unknown regulation of poly I:C that boosts the TCA cycle to enhance innate immunity against bacterial infection.

Poly I:C-induced tumor cell apoptosis mediated by pattern-recognition receptors

Poly I:C is a synthetic dsRNA that can imitate a viral infection and elicit host immune responses by triggering specific pattern-recognition receptors (PRRs) such as toll-like receptor 3 and retinoic acid inducible gene I(RIG-I)-like receptors, including RIG-I and melanoma differentiation-associated gene 5. Activation of these PRRs by poly I:C triggers a signal transduction cascade that results in the activation of NF-κB and production of type I interferon. Poly I:C has been used as a vaccine adjuvant for cancer immunotherapy for several decades. Evidence from recent studies indicates that poly I:C can directly induce apoptosis in several types of tumor cells, thus providing a new therapeutic approach for cancer treatment. However, the molecular mechanism underlying the induction of apoptosis by poly I:C is still unclear. In this review, we summarize the current knowledge of poly I:C-induced tumor cell apoptosis, focusing on the key molecules and pathways involved in this process.