当前位置: X-MOL 学术Artif. Cells Nanomed. Biotechnol. › 论文详情
Our official English website, www.x-mol.net, welcomes your feedback! (Note: you will need to create a separate account there.)
Intracellular synthesis of gold nanoparticles by Gluconacetobacter liquefaciens for delivery of peptide CopA3 and ginsenoside and anti-inflammatory effect on lipopolysaccharide-activated macrophages.
Artificial Cells, Nanomedicine, and Biotechnology ( IF 4.5 ) Pub Date : 2020-04-21 , DOI: 10.1080/21691401.2020.1748639
Ying Liu 1 , Haribalan Perumalsamy 1 , Chang Ho Kang 2 , Seung Hyun Kim 1 , Jae-Sam Hwang 3 , Sung-Cheol Koh 4 , Tae-Hoo Yi 1, 2 , Yeon-Ju Kim 1, 2
Affiliation  

Probiotic Gluconacetobacter strains are intestinal microbes with beneficial effects on human health. Recently, researchers have used these strains to biosynthesize metal and non-metal nanoparticles for treating various chronic diseases. Despite their importance in nanotechnology, gold nanoparticles (AuNPs) biosynthesized by Gluconacetobacter species have not been clearly identified for treating inflammation and inflammation-associated diseases. While ginsenoside CK has strong pharmaceutical activity, it also has strong cytotoxicity and hydrophobicity which is hurdle to make formulation. Peptide-nanoparticle hybrids are gaining increasing attention for their potential biomedical applications, including human inflammatory diseases. Herein, we developed peptide CopA3 surface conjugated and ginsenoside compound K (CK) loaded gold nanoparticles (GNP-CK-CopA3), which intracellularly synthesised by the probiotic Gluconacetobacter liquefaciens kh-1, to target lipopolysaccharide (LPS)-activated RAW264.7 macrophages. The synthetic GNP-CK-CopA3 was characterised by various instrumental techniques. The results of our cellular uptake and MTT assays exhibited obvious drug intracellular delivery without significant cytotoxicity. In addition, pre-treatment with GNP-CK-CopA3 significantly ameliorated LPS-induced nitric oxide (NO) and reactive oxygen species (ROS) production and suppressed the mRNA and protein expression of pro-inflammatory cytokines in macrophages. Furthermore, GNP-CK-CopA3 efficiently inhibited the activation of the nuclear factor-κB (NF-κB) and mitogen-activating protein kinase (MAPK) signalling pathways. Taken together, our findings highlight the potential of using peptide-nanoparticle hybrids in the development of anti-inflammatory approaches and providing the experimental foundation for further application.

中文翻译:

液化葡糖杆菌对金纳米颗粒的细胞内合成,用于肽CopA3和人参皂苷的递送以及对脂多糖激活的巨噬细胞的抗炎作用。

益生菌糖醋杆菌菌株是对人体健康有益的肠道微生物。最近,研究人员已使用这些菌株生物合成金属和非金属纳米粒子,以治疗各种慢性疾病。尽管它们在纳米技术中具有重要意义,但尚未明确鉴定由糖醋杆菌属物种生物合成的金纳米颗粒(AuNP)用于治疗炎症和与炎症相关的疾病。人参皂苷CK虽然具有很强的药物活性,但它也具有很强的细胞毒性和疏水性,因此难以配制。肽-纳米粒子杂交体因其潜在的生物医学应用(包括人类炎症性疾病)而受到越来越多的关注。在这里 我们开发了肽CopA3表面共轭和负载人参皂苷化合物K(CK)的金纳米颗粒(GNP-CK-CopA3),该糖纳米颗粒由益生糖液杆菌kh-1胞内合成,以靶向脂多糖(LPS)活化的RAW264.7巨噬细胞。合成的GNP-CK-CopA3通过各种仪器技术进行了表征。我们的细胞摄取和MTT分析的结果显示出明显的药物细胞内递送,而没有明显的细胞毒性。另外,用GNP-CK-CopA3预处理可显着改善LPS诱导的一氧化氮(NO)和活性氧(ROS)的产生,并抑制巨噬细胞中促炎细胞因子的mRNA和蛋白质表达。此外,GNP-CK-CopA3有效抑制核因子-κB(NF-κB)和有丝分裂原激活蛋白激酶(MAPK)信号通路的激活。综上所述,我们的发现突出了在消炎方法开发中使用肽-纳米颗粒杂化物的潜力,并为进一步应用提供了实验基础。
更新日期:2020-05-06
down
wechat
bug