首页> 外文会议>World biomaterials congress >Light-activatable polymeric nanoparticles for intracellular delivery in cancer stem cells
【24h】

Light-activatable polymeric nanoparticles for intracellular delivery in cancer stem cells

机译:可光活化的聚合物纳米颗粒用于癌症干细胞的细胞内递送

获取原文

摘要

Introduction: Nanoparticles (NP) may be used as vehicles for the intracellular delivery of biomolecules and ultimately to modulate the activity of cancer cells. Here we present a novel light-dissociable polymeric NP formulation that has approximately 140nm in diameter, positive net charge, and disassemble when exposed to UV light (365nm) or blue light (405nm) releasing retinoic acid (RA). These NPs can be taken up rapidly (4h) by acute myeloid leukemia (AML) cells (K562, NB4, U937, THP1 and AML stem cells). Importantly, our results show that the light activation of NPs contribute for the differentiation of these cells at levels not observed using formulations that release passively RA or by conventional chemotherapy. Also our data shows that the activation of the NP can be done in vivo at the bone marrow (BM), using exousgenous light activation in NOD/SCID mice. This study highlights the importance of drug spatial positioning and concentration to treat leukemia. Experimental Methods: Diameter, counts and zeta potential of NPs in response to light over time were recorded by DLS. The in vitro effects of RA-loaded NPs on: (ⅰ) the erythroid differentiation of K562 at day 9 was assessed by cytochemical staining with benzidine solution, (ⅱ) the granulocytic differentiation at day 6 and 3 for human NB4 and U937 (PLZF/RARA), respectively, was assessed by flow cytometry using CD13 and CD11b as a marker, (ⅲ) reduction of stem cell colonies in AML stem cells was assessed by colony-forming cell (CFC, 2 weeks) and long-term culture initiating cell assays (LTC-IC, 7 weeks). The in vivo effects of RA-NP on homing, engraftment and differentiation of AML cells was performed by injecting intravenously NP-loaded THP1 cells in NOD/SCID mice. At day 6 the NP were light activated and 72h later mice were sacrificed for analysis. The effects at the calvaria BM were accessed by flow cytometry at day 6 and by ex-vivo imaging of the calvaria at day 9, using CD45 and CD11b markers. Three independent runs were done for each experimental group and for each run three technical replicates were performed. Results and Discussion: The activation of NPs promotes rapid and efficient RA delivery. Light-activated RA-NPs induce 1.92 (± 0.17) times higher levels of erythroid differentiation in K562,1.19 (±0.01) times of granulocytic differentiation in NB4 and 1.45 (±0.03) times of granulocytic differentiation in U937 cells as compared to non-activated NPs. In addition, activated RA-NPs induce 1.83 (±0.17) times higher levels of erythroid differentiation in K562,1.12 (±0.01) times of granulocytic differentiation in NB4 and 1.44 (± 0.04) times of granulocytic differentiation in U937 cells as compared to 1 mM RA in solution. It should be noted that RA-NPs contain -10 times less RA than the one used in solution. This experimental result is particularly important in the RA-low sensitive cell line U937-PLZF/RARA where the high RA intracellular release is able to overcome the low sensitivity of the cell. AML stem cells treated with light-activated RA+-NPs showed 69.6 ± 9.2 % (CFC) and 61.8 ± 10.1 % (LTC-IC) less colonies. In agreement with the in vitro results, we were also able to induce differentiation of THP1 cells in the calvaria BM niche without affecting homing or engraftment. These results indicate that the enhanced intracellular release and the kinetic control mediated by the light triggered-NPs may be an effective strategy to treat leukemia. Conclusion: The spatio-temporal control of the intracellular release of RA is important to control the differentiation of leukemic (stem) cells.
机译:简介:纳米粒子(NP)可以用作生物分子在细胞内递送的载体,并最终调节癌细胞的活性。在这里,我们介绍了一种新型的可光解离的聚合物NP制剂,该制剂的直径约为140nm,带正净电荷,当暴露于紫外线(365nm)或蓝光(405nm)时会分解成视黄酸(RA),从而分解。这些NP可被急性髓细胞白血病(AML)细胞(K562,NB4,U937,THP1和AML干细胞)迅速摄取(4小时)。重要的是,我们的结果表明,NPs的光活化以这些水平导致了这些细胞的分化,而使用被动释放RA的制剂或常规化学疗法未观察到该水平。我们的数据还表明,使用NOD / SCID小鼠中的外源光激活,可以在骨髓(BM)体内进行NP激活。这项研究强调了药物空间定位和浓度对治疗白血病的重要性。实验方法:用DLS记录随时间变化的NPs的直径,数量和zeta电位。负载RA的NPs对(ⅰ)第9天K562的红系分化的体外作用是通过联苯胺溶液的细胞化学染色评估的;(ⅱ)人NB4和U937在第6天和第3天的粒细胞分化(PLZF /使用CD13和CD11b作为标记,分别通过流式细胞术评估RARA)。(ⅲ)通过集落形成细胞(CFC,2周)和长期培养起始细胞评估AML干细胞中干细胞集落的减少分析(LTC-IC,7周)。通过在NOD / SCID小鼠中静脉内注射NP负载的THP1细胞,RA-NP对AML细胞的归巢,移入和分化产生了体内效应。在第6天,NP被光激活,并且72小时后处死小鼠用于分析。在第6天通过流式细胞术以及在第9天通过体外成像颅盖骨来获得对颅盖骨BM的作用,使用CD45和CD11b标记。每个实验组进行了三个独立的运行,并且每个运行进行了三个技术重复。结果与讨论:NPs的激活促进快速有效的RA递送。与未活化的RA-NP相比,光活化的RA-NPs在K562中诱导的红细胞分化水平高1.92(±0.17)倍,在NB4中诱导的粒细胞分化高1.19(±0.01)倍,而在U937细胞中则是1.45(±0.03)倍。激活的NP。此外,与1相比,活化的RA-NPs在K562中诱导的红细胞分化水平高1.83(±0.17)倍,在NB4中的颗粒细胞分化诱导1.12(±0.01)倍,而在U937细胞中则是1.44(±0.04)倍。溶液中的mM RA。应该注意的是,RA-NP所含的RA比溶液中所用的RA少-10倍。该实验结果在RA低敏感性细胞系U937-PLZF / RARA中尤其重要,其中高RA细胞内释放能够克服细胞的低敏感性。用光活化的RA + -NP处理的AML干细胞集落减少了69.6±9.2%(CFC)和61.8±10.1%(LTC-IC)。与体外结果一致,我们还能够在颅盖BM壁iche中诱导THP1细胞分化,而不会影响归巢或植入。这些结果表明,由光触发的NPs介导的增强的细胞内释放和动力学控制可能是治疗白血病的有效策略。结论:时空控制类风湿关节炎的细胞内释放对于控制白血病(干)细胞的分化具有重要意义。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

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