首页> 美国卫生研究院文献>other >Bisphosphonate-Functionalized Hydroxyapatite Nanoparticles for the Delivery of the Bromodomain Inhibitor JQ1 in the Treatment of Osteosarcoma
【2h】

Bisphosphonate-Functionalized Hydroxyapatite Nanoparticles for the Delivery of the Bromodomain Inhibitor JQ1 in the Treatment of Osteosarcoma

机译:双膦酸酯功能化羟基磷灰石纳米颗粒的溴结构域抑制剂JQ1的输送治疗骨肉瘤。

代理获取
本网站仅为用户提供外文OA文献查询和代理获取服务,本网站没有原文。下单后我们将采用程序或人工为您竭诚获取高质量的原文,但由于OA文献来源多样且变更频繁,仍可能出现获取不到、文献不完整或与标题不符等情况,如果获取不到我们将提供退款服务。请知悉。

摘要

Osteosarcoma (OS) is one of the most common neoplasia among children, and its survival statistics have been stagnating since the combinatorial anticancer therapy triad was first introduced. Here, we report on the assessment of the effect of hydroxyapatite (HAp) nanoparticles loaded with medronate, the simplest bisphosphonate, as a bone-targeting agent and JQ1, a small-molecule bromodomain inhibitor, as a chemotherapeutic in different 2D and 3D K7M2 OS in vitro models. Both additives decreased the crystallinity of HAp, but the effect was more intense for medronate because of its higher affinity for HAp. As the result of PO43−-NH+ binding, JQ1 shielded the surface phosphates of HAp and pushed its surface charge to more positive values, exhibiting the opposite effect from calcium-blocking medronate. In contrast to the faster and more exponential release of JQ1 from monetite, its release from HAp nanoparticles followed a zero-order kinetics, but 98% of the payload was released after 48 h. The apoptotic effect of HAp nanoparticles loaded with JQ1, with medronate and with both JQ1 and medronate, was selective in 2D culture: pronounced against the OS cells and nonexistent against the healthy fibroblasts. While OS cell invasion was significantly inhibited by all of the JQ1-containing HAp formulations, that is, with and without medronate, all of the combinations of the targeting compound, medronate, and the chemotherapeutic, JQ1, delivered using HAp, but not HAp alone, inhibited OS cell migration from the tumor spheroids. JQ1 delivered using HAp had an effect on tumor migration, invasion, and apoptosis even at extremely low, subnanomolar concentrations, at which no effect of JQ1 per se was observed, meaning that this form of delivery could help achieve a multifold increase of this drug’s efficacy. More than 80% of OS cells internalized JQ1-loaded HAp nanoparticles after 24 h of coincubation, suggesting that this augmentation of the activity of JQ1 may be due to the intracellular delivery and sustained release of the drug enabled by HAp. In addition to the reduction of the OS cell viability, the reduction of the migration and invasion radii was observed in OS tumor spheroids challenged with even JQ1-free medronate-functionalized HAp nanoparticles, demonstrating a definite anticancer activity of medronate alone when combined with HAp. The effect of medronate-functionalized JQ1-loaded HAp nanoparticles was most noticeable against OS cells differentiated into an osteoblastic lineage, in which case they surpassed in effect pure JQ1 and medronate-free compositions. The activity of JQ1 was mediated through increased Ezrin expression and decreased RUNX2 expression and was MYC and FOSL1 independent, but these patterns of gene expression changed in cells challenged with the nanoparticulate form of delivery, having been accompanied by the upregulation of RUNX2 and downregulation of Ezrin in OS cells treated with medronate-functionalized JQ1-loaded HAp nanoparticles.
机译:骨肉瘤(OS)是儿童中最常见的肿瘤,自从首次引入组合抗癌治疗三联症以来,其生存统计一直停滞不前。在这里,我们报告了在不同的2D和3D K7M2 OS中,最简单的双膦酸盐甲磺酸盐作为骨靶向剂和小分子溴结构域抑制剂JQ1作为羟磷灰石(HAp)纳米颗粒的疗效评估报告体外模型。两种添加剂均降低了HAp的结晶度,但由于其对HAp的亲和力更高,因此对甲膦酸盐的作用更为强烈。由于PO4 3 − -NH + 的结合,JQ1屏蔽了HAp的表面磷酸盐,并将其表面电荷推高至正值,与钙离子相反。阻止甲酸盐。与JQ1从透纳石中更快,更指数地释放相比,其从HAp纳米颗粒中释放的过程遵循零级动力学,但是98%的有效载荷在48小时后释放。装有JQ1,甲酸盐和JQ1和甲酸盐的HAp纳米颗粒的凋亡作用在2D培养中具有选择性:对OS细胞明显,对健康的成纤维细胞不存在。尽管所有含JQ1的HAp制剂均能显着抑制OS细胞的侵袭,也就是说,在有或没有甲磺酸盐的情况下,靶向化合物,甲酸盐和化学治疗剂JQ1的所有组合均使用HAp递送,但未单独使用HAp抑制了OS细胞从肿瘤球的迁移。即使在极低的亚纳摩尔浓度下,使用HAp递送的JQ1也会对肿瘤迁移,侵袭和细胞凋亡产生影响,在这种浓度下,未观察到JQ1本身的作用,这意味着这种递送形式可以帮助实现该药物功效的倍数提高。共孵育24小时后,超过80%的OS细胞将装载JQ1的HAp纳米颗粒内在化,这表明JQ1活性的这种增强可能是由于HAp能够实现药物的细胞内递送和持续释放。除了降低OS细胞的生存力外,在甚至连无JQ1的甲酸酯盐官能化的HAp纳米颗粒挑战的OS肿瘤球体中也观察到迁移和侵袭半径的降低,这表明单独的甲磺酸盐与HAp结合时具有确定的抗癌活性。甲磺酸盐官能化的装载JQ1的HAp纳米颗粒对分化为成骨细胞谱系的OS细胞的影响最为明显,在这种情况下,它们实际上超过了纯JQ1和无甲酸盐的组合物。 JQ1的活性是通过增加Ezrin表达和降低RUNX2表达来介导的,并且是MYC和FOSL1独立的,但是这些基因表达模式在受到纳米颗粒递送形式挑战的细胞中发生了改变,并伴有RUNX2的上调和Ezrin的下调。甲磺酸盐功能化的JQ1加载的HAp纳米颗粒处理的OS细胞中的细胞凋亡

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
代理获取

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号