Home>>Signaling Pathways>> Others>> Others>>D-Lin-MC3-DMA

D-Lin-MC3-DMA Sale

(Synonyms: 4-(N,N-二甲基氨基)丁酸(二亚油基)甲酯) 目录号 : GC35879

Dilinoleylmethyl-4-dimethylaminobutyrate (D-Lin-MC3-DMA) 是有效的体内 siRNA 递送载体。

D-Lin-MC3-DMA Chemical Structure

Cas No.:1224606-06-7

规格 价格 库存 购买数量
10mM (in 1mL DMSO)
¥1,266.00
现货
1mg
¥320.00
现货
5mg
¥896.00
现货
10mg
¥1,295.00
现货
25mg
¥2,590.00
现货
50mg
¥3,850.00
现货
100mg
¥5,950.00
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

HeLa cells

Preparation Method

Luciferase expression following incubation of cultured HeLa cells with LNP-pCI-FLuc (0.029 mg DNA per µmol lipid) for 24 h at pDNA concentrations of 0.75-6.0 µg/ml. LNP lipid composition D-Lin-MC3-DMA/HL/cholesterol/PEG-lipid (50/10/38.5/1.5; mol/mol)( D-Lin-MC3-DMA).

Reaction Conditions

LNP lipid composition MC3/HL/cholesterol/PEG-lipid (50/10/38.5/1.5; mol/mol).

Applications

D-Lin-MC3-DMA is an ionizable cationic lipid that is an effective carrier for delivering siRNA.

Animal experiment [2]:

Animal models

8-10 week old, female C57BL/6 mice

Preparation Method

LNP-siRNA systems containing Factor VII siRNA( D-Lin-MC3-DMA ) were diluted to the appropriate concentrations in sterile PBS immediately prior to use and the formulations were administered intravenously via the lateral tail vein in a total volume of 10 ml/kg.

Dosage form

Molar ratio of the four lipid MC3, DSPC, cholesterol and PEG-lipid (50/10/38.5/1.5).

Applications

In the murine FVII model, an ED50 of 0.005 mg kg 1 siRNA was achieved in the optimized composition( D-Lin-MC3-DMA), which represented a sixfold improvement relative to the 40/10/40/10 molar ratio composition.

References:

[1]. Kulkarni JA, Myhre JL,et,al. Design of lipid nanoparticles for in vitro and in vivo delivery of plasmid DNA. Nanomedicine. 2017 May;13(4):1377-1387. doi: 10.1016/j.nano.2016.12.014. Epub 2016 Dec 28. PMID: 28038954.
[2]. Jayaraman M, Ansell SM, et,al.Maximizing the potency of siRNA lipid nanoparticles for hepatic gene silencing in vivo. Angew Chem Int Ed Engl. 2012 Aug 20;51(34):8529-33. doi: 10.1002/anie.201203263. Epub 2012 Jul 10. PMID: 22782619; PMCID: PMC3470698.

产品描述

Dilinoleylmethyl-4-dimethylaminobutyrate (D-Lin-MC3-DMA) are potent siRNA delivery vehicles in vivo[1].

D-Lin-MC3-DMA is an effective carrier for delivering siRNA in HeLa cells[1]. Differences in gene silencing in two distinct liver cell populations when siRNA was delivered with LNPs containing the ionizable cationic lipid MC3(D-Lin-MC3-DMA) or ALC-0315[1]. D-Lin-MC3-DMA, which is practically uncharged at physiological pH, can become ionized once endocytosed and entrained in the early endosome compartment. Because these early endosomes are expected to contain negatively charged phosphatidylserine lipids, one may speculate that the LNP cargo might escape by LNPs adhering to and fusing with the early endosomal membrane through electrostatic means[5,6]. D-Lin-MC3-DMA -LNPs were close to neutral charge with a zeta-potential of 5±3 mV. While D-Lin-MC3-DMA -LNPs are neutrally charged at physiological pH, they are known to be positively charged at endosomal pH[7]. LNPs were synthesized by the microfluidic mixing technique and are composed of ionizable cationic lipid (D-Lin-MC3-DMA), a phospholipid, cholesterol, and poly(ethylene glycol) (PEG), as well as encapsulated cargoes that are either phosphorothioated siRNA (50 or 100%) or mRNA. LNPs form physically stable complexes with bioactive drug siRNA for a period of 94 days[8].

In the murine FVII model, an ED50 of 0.005 mg kg 1 siRNA was achieved in the optimized composition(D-Lin-MC3-DMA), which represented a sixfold improvement relative to the 40/10/40/10 molar ratio composition[2]. 2nd generation LNP containing D-Lin-MC3-DMA were more than two orders of magnitude more potent than 1st generation LNP containing DLinDMA[4].

References:
[1]. Kulkarni JA, Myhre JL, et,al.Design of lipid nanoparticles for in vitro and in vivo delivery of plasmid DNA. Nanomedicine. 2017 May;13(4):1377-1387. doi: 10.1016/j.nano.2016.12.014. Epub 2016 Dec 28. PMID: 28038954.
[2]. Jayaraman M, Ansell SM, et,al. Maximizing the potency of siRNA lipid nanoparticles for hepatic gene silencing in vivo. Angew Chem Int Ed Engl. 2012 Aug 20;51(34):8529-33. doi: 10.1002/anie.201203263. Epub 2012 Jul 10. PMID: 22782619; PMCID: PMC3470698.
[3]. Ferraresso F, Strilchuk AW, et,al.Comparison of DLin-MC3-DMA and ALC-0315 for siRNA Delivery to Hepatocytes and Hepatic Stellate Cells. Mol Pharm. 2022 Jul 4;19(7):2175-2182. doi: 10.1021/acs.molpharmaceut.2c00033. Epub 2022 May 31. PMID: 35642083; PMCID: PMC9621687.
[4]. Akinc A, Maier MA, et,al. The Onpattro story and the clinical translation of nanomedicines containing nucleic acid-based drugs. Nat Nanotechnol. 2019 Dec;14(12):1084-1087. doi: 10.1038/s41565-019-0591-y. PMID: 31802031.
[5]. Yanez Arteta M, Kjellman T, et,al. Successful reprogramming of cellular protein production through mRNA delivered by functionalized lipid nanoparticles. Proc Natl Acad Sci U S A. 2018 Apr 10;115(15):E3351-E3360. doi: 10.1073/pnas.1720542115. Epub 2018 Mar 27. PMID: 29588418; PMCID: PMC5899464.
[6]. Chan CL, Majzoub RN, et,al. Endosomal escape and transfection efficiency of PEGylated cationic liposome-DNA complexes prepared with an acid-labile PEG-lipid. Biomaterials. 2012 Jun;33(19):4928-35. doi: 10.1016/j.biomaterials.2012.03.038. Epub 2012 Apr 1. PMID: 22469293; PMCID: PMC3337860.
[7]. Maugeri M, Nawaz M, et,al. Linkage between endosomal escape of LNP-mRNA and loading into EVs for transport to other cells. Nat Commun. 2019 Sep 24;10(1):4333. doi: 10.1038/s41467-019-12275-6. PMID: 31551417; PMCID: PMC6760118.
[8]. Viger-Gravel J, Schantz A, et,al.Structure of Lipid Nanoparticles Containing siRNA or mRNA by Dynamic Nuclear Polarization-Enhanced NMR Spectroscopy. J Phys Chem B. 2018 Feb 22;122(7):2073-2081. doi: 10.1021/acs.jpcb.7b10795. Epub 2018 Feb 9. PMID: 29332384.

Dilinoleylmethyl-4-dimethylaminobutyrate (D-Lin-MC3-DMA) 是有效的体内 siRNA 递送载体[1]

D-Lin-MC3-DMA 是在 HeLa 细胞中递送 siRNA 的有效载体[1]。当 siRNA 与含有可电离阳离子脂质 MC3(D-Lin-MC3-DMA) 或 ALC-0315[1] 的 LNP 一起递送时,两个不同肝细胞群中基因沉默的差异。 D-Lin-MC3-DMA 在生理 pH 下几乎不带电荷,一旦被内吞并夹带在早期内体隔室中,就会电离。由于预计这些早期内体含有带负电荷的磷脂酰丝氨酸脂质,因此可以推测 LNP 货物可能通过静电方式粘附并与早期内体膜融合的 LNP 逃逸[5,6]。 D-Lin-MC3-DMA -LNP 接近中性电荷,zeta 电位为 5±3 mV。虽然 D-Lin-MC3-DMA -LNP 在生理 pH 值下带中性电荷,但已知它们在核内体 pH 值下带正电荷 [7]。 LNPs 通过微流体混合技术合成,由可电离的阳离子脂质 (D-Lin-MC3-DMA)、磷脂、胆固醇和聚乙二醇 (PEG) 以及硫代磷酸化 siRNA 封装的货物组成(50 或 100%)或 mRNA。 LNPs 与生物活性药物 siRNA 形成物理稳定的复合物,持续时间为 94 天[8]。

在小鼠 FVII 模型中,优化组合物 (D-Lin-MC3-DMA) 的 ED50 为 0.005 mg kg 1 siRNA,相对于 40/10/40/10 摩尔比提高了六倍组成[2]。含有 D-Lin-MC3-DMA 的第 2 代 LNP 比含有 DLinDMA 的第 1 代 LNP 强两个数量级以上[4]。

Chemical Properties

Cas No. 1224606-06-7 SDF
别名 4-(N,N-二甲基氨基)丁酸(二亚油基)甲酯
Canonical SMILES O=C(OC(CCCCCCCC/C=C\C/C=C\CCCCC)CCCCCCCC/C=C\C/C=C\CCCCC)CCCN(C)C
分子式 C43H79NO2 分子量 642.09
溶解度 DMSO: 250 mg/mL (389.35 mM); Ethanol: ≥ 60 mg/mL (93.44 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 1.5574 mL 7.7871 mL 15.5741 mL
5 mM 0.3115 mL 1.5574 mL 3.1148 mL
10 mM 0.1557 mL 0.7787 mL 1.5574 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

In Vitro Inhibition of Replication of Dengue Virus Serotypes 1-4 by siRNAs Bound to Non-Toxic Liposomes

Viruses 2022 Feb 7;14(2):339.PMID:35215929DOI:10.3390/v14020339.

Dengue virus is a ssRNA+ flavivirus, which produces the dengue disease in humans. Currently, no specific treatment exists. siRNAs regulate gene expression and have been used systematically to silence viral genomes; however, they require controlled release. Liposomes show favorable results encapsulating siRNA for gene silencing. The objective herein was to design and evaluate in vitro siRNAs bound to liposomes that inhibit DENV replication. siRNAs were designed against DENV1-4 from conserved regions using siDirect2.0 and Web-BLOCK-iT™ RNAiDesigner; the initial in vitro evaluation was carried out through transfection into HepG2 cells. siRNA with silencing capacity was encapsulated in liposomes composed of D-Lin-MC3-DMA, DSPC, Chol. Cytotoxicity, hemolysis, pro-inflammatory cytokine release and antiviral activity were evaluated using plaque assay and RT-qPCR. A working concentration of siRNA was established at 40 nM. siRNA1, siRNA2, siRNA3.1, and siRNA4 were encapsulated in liposomes, and their siRNA delivery through liposomes led to a statistically significant decrease in viral titers, yielded no cytotoxicity or hemolysis and did not stimulate release of pro-inflammatory cytokines. Finally, liposomes were designed with siRNA against DENV, which proved to be safe in vitro.

DLin-MC3-Containing mRNA Lipid Nanoparticles Induce an Antibody Th2-Biased Immune Response Polarization in a Delivery Route-Dependent Manner in Mice

Pharmaceutics 2023 Mar 21;15(3):1009.PMID:36986871DOI:10.3390/pharmaceutics15031009.

mRNA-based vaccines have made a leap forward since the SARS-CoV-2 pandemic and are currently used to develop anti-infectious therapies. If the selection of a delivery system and an optimized mRNA sequence are two key factors to reach in vivo efficacy, the optimal administration route for those vaccines remains unclear. We investigated the influence of lipid components and immunization route regarding the intensity and quality of humoral immune responses in mice. The immunogenicity of HIV-p55Gag encoded mRNA encapsulated into D-Lin-MC3-DMA or GenVoy-ionizable lipid-based LNPs was compared after intramuscular or subcutaneous routes. Three sequential mRNA vaccines were administrated followed by a heterologous boost composed of p24-HIV protein antigen. Despite equivalent IgG kinetic profiles of general humoral responses, IgG1/IgG2a ratio analysis showed a Th2/Th1 balance toward a Th1-biased cellular immune response when both LNPs were administrated via the intramuscular route. Surprisingly, a Th2-biased antibody immunity was observed when DLin-containing vaccine was injected subcutaneously. A protein-based vaccine boost appeared to reverse this balance to a cellular-biased response correlated to an increase in antibody avidity. Our finding suggests that the intrinsic adjuvant effect of ionizable lipids appears to be dependent on the delivery route used, which could be relevant to reach potent and long-lasting immunity after mRNA-based immunization.