期刊文献+

In vivo evaluation of riboflavin receptor targeted fluorescent USPIO in mice with prostate cancer xenografts 被引量:4

In vivo evaluation of riboflavin receptor targeted fluorescent USPIO in mice with prostate cancer xenografts
原文传递
导出
摘要 Riboflavin (Rf) receptors bind and translocate Rf and its phosphorylated forms (e.g. flavin mononucleotide, FMN) into cells where they mediate various cellular metabolic pathways. Previously, we showed that FMN-coated ultrasmall superparamagnetic iron oxide (FLUSPIO) nanoparticles are suitable for labeling metabolically active cancer and endothelial cells in vitro. In this study, we focused on the in vivo application of FLUSPIO using prostate cancer xenografts. Size, charge, and chemical composition of FLUSPIO were evaluated. We explored the in vitro specificity of FLUSPIO for its cellular receptors using magnetic resonance imaging (MRI) and Prussian blue staining. Competitive binding experiments were performed in vivo by injecting free FMN in excess. Bio-distribution of FLUSPIO was determined by estimating iron content in organs and tumors using a colorimetric assay. AFM analysis and zeta potential measurements revealed a particulate morphology approximately 20-40 nm in size and a negative zeta potential (-24.23±0.15 mV) in water. X-ray photoelectron spectroscopy and time-of-flight secondary ion mass spectrometry data confirmed FMN present on the USPIO nanoparticle surface. FLUSPIO uptake in prostate cancer cells and human umbilical vein endothelial cells was significantly higher than that of control USPIO, while addition of excess of free FMN reduced accumulation. Similarly, in vivo MRI and histology showed specific FLUSPIO uptake by prostate cancer cells, tumor endothelial cells, and tumor-associated macrophages. Besides prominent tumor accumulation, FLUSPIO accumulated in the liver, spleen, lung, and skin. Hence, our data strengthen our hypothesis that targeting riboflavin receptors is an efficient approach to accumulate nanomedicines in tumors opening perspectives for the development of diagnostic and therapeutic systems. Riboflavin (Rf) receptors bind and translocate Rf and its phosphorylated forms (e.g. flavin mononucleotide, FMN) into cells where they mediate various cellular metabolic pathways. Previously, we showed that FMN-coated ultrasmall superparamagnetic iron oxide (FLUSPIO) nanoparticles are suitable for labeling metabolically active cancer and endothelial cells in vitro. In this study, we focused on the in vivo application of FLUSPIO using prostate cancer xenografts. Size, charge, and chemical composition of FLUSPIO were evaluated. We explored the in vitro specificity of FLUSPIO for its cellular receptors using magnetic resonance imaging (MRI) and Prussian blue staining. Competitive binding experiments were performed in vivo by injecting free FMN in excess. Bio-distribution of FLUSPIO was determined by estimating iron content in organs and tumors using a colorimetric assay. AFM analysis and zeta potential measurements revealed a particulate morphology approximately 20-40 nm in size and a negative zeta potential (-24.23±0.15 mV) in water. X-ray photoelectron spectroscopy and time-of-flight secondary ion mass spectrometry data confirmed FMN present on the USPIO nanoparticle surface. FLUSPIO uptake in prostate cancer cells and human umbilical vein endothelial cells was significantly higher than that of control USPIO, while addition of excess of free FMN reduced accumulation. Similarly, in vivo MRI and histology showed specific FLUSPIO uptake by prostate cancer cells, tumor endothelial cells, and tumor-associated macrophages. Besides prominent tumor accumulation, FLUSPIO accumulated in the liver, spleen, lung, and skin. Hence, our data strengthen our hypothesis that targeting riboflavin receptors is an efficient approach to accumulate nanomedicines in tumors opening perspectives for the development of diagnostic and therapeutic systems.
出处 《Nano Research》 SCIE EI CAS CSCD 2016年第5期1319-1333,共15页 纳米研究(英文版)
关键词 flavin mononucleotide iron oxide nanoparticles magnetic resonance imaging molecular imaging riboflavin carrier protein riboflavin transporters flavin mononucleotide,iron oxide nanoparticles,magnetic resonance imaging,molecular imaging,riboflavin carrier protein,riboflavin transporters
  • 相关文献

参考文献35

  • 1Mamede, A. C.; Tavares, S. D.; Abrantes, A. M.; Trindade, J.; Maia, J. M.; Botelho, M. F. The role of vitamins in cancer: A review. Nutr. Cancer 2011, 63, 479~494.
  • 2Russell-Jones, G.; McTavish, K.; McEwan, J.; Rice, J.; Nowotnik, D. Vitamin-mediated targeting as a potential mechanism to increase drug uptake by mmours. 31 lnorg. Biochem. 2004, 98, 1625 1633.
  • 3Giancaspero, T. A.; Busco, G.; Panebianco, C.; Carmone, C.; Miccolis, A.; Liuzzi, G. M.; Colella, M.; Barile, M. FAD synthesis and degradation in the nucleus create a local flavin cofactor pool. J. Biol. Chem. 2013, 288, 2906%29080.
  • 4Becker, K.; Schirmer, M.; Kanzok, S.; Schirmer, R. H. Flavins and flavoenzymes in diagnosis and therapy. In Flavoprotein Protocols; Chapman, S. K.; Reid, G. A., Eds.; Humana Press: Totowa, N.J., 1999; pp 229 245.
  • 5Rao, P. N.; Levine, E.; Myers, M. O.; Prakash, V.; Watson, J. Stolier, A.; Kopicko, J, J.; Kissinger, P.; Raj, S. G.; Raj, M. H Elevation of serum riboflavin carrier protein in breast cancer Cancer Epidemiol. Biomarkers Prev. 1999, 8, 985-990.
  • 6Bareford, L. M.; Swaan, P. W. Endocytic mechanisms for targeted drug delivery. Adv. Drug Deliv. Rev. 2007, 59, 748-758.
  • 7Bareford, L. M.; Avaritt, B. R.; Ghandehari, H.; Nan, A.; Swaan, P. W. Riboflavin-targeted polymer conjugates for breast tumor delivery. Pharm. Res. 2013, 30, 1799-1812.
  • 8Huang, S. N.; Phelps, M. A.; Swaan, P. W. Involvement of endocytie organelles in the subcellular trafficking and localization of riboflavin. J. Pharma~ol. Exp. Ther. 2003, 306, 681-687.
  • 9Pedrolli, D. B.; Jankowitsch, F.; Schwarz, J.; Langer, S.; Nakanishi, S.; Frei, E.; Mack, M. Riboflavin analogs as antiinfectives: Occurrence, mode of action, metabolism and resistance. Curr. Pharm. Des. 2013, 19, 2552-2560.
  • 10Chen, C.; Ke, J. Y.; Zhou, X. E.; Yi, W.; Brunzelle, J. S.; Li, J.; Yong, E. L.; Xu, H. E.; Melcher, K. Structural basis for molecular recognition of folio acid by folate receptors. Nature 2013, 500, 486-489.

同被引文献9

引证文献4

二级引证文献1

相关作者

内容加载中请稍等...

相关机构

内容加载中请稍等...

相关主题

内容加载中请稍等...

浏览历史

内容加载中请稍等...
;
使用帮助 返回顶部