期刊论文详细信息
Journal of Nanobiotechnology
Activation of caspase-dependent apoptosis by intracellular delivery of cytochrome c-based nanoparticles
Kai Griebenow1  Myraida Morales2  Jessica Méndez1  Anna Molina1  Yamixa Delgado3  Tania González-Robles1  Cindy M Figueroa1  Moraima Morales-Cruz3 
[1] Departments of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan 00931, PR, USA;Department of Graduate Studies, University of Puerto Rico, Río Piedras Campus, Río Piedras Campus, San Juan 00931, PR, USA;Department of Biology, University of Puerto Rico, Río Piedras Campus, San Juan 00931, PR, USA
关键词: Triggered release;    Passive targeting;    PLGA;    Protein nanoparticles;    Drug delivery;   
Others  :  1146161
DOI  :  10.1186/s12951-014-0033-9
 received in 2014-05-30, accepted in 2014-08-23,  发布年份 2014
PDF
【 摘 要 】

Background

Cytochrome c is an essential mediator of apoptosis when it is released from the mitochondria to the cytoplasm. This process normally takes place in response to DNA damage, but in many cancer cells (i.e., cancer stem cells) it is disabled due to various mechanisms. However, it has been demonstrated that the targeted delivery of Cytochrome c directly to the cytoplasm of cancer cells selective initiates apoptosis in many cancer cells. In this work we designed a novel nano-sized smart Cytochrome c drug delivery system to induce apoptosis in cancer cells upon delivery.

Results

Cytochrome c was precipitated with a solvent-displacement method to obtain protein nanoparticles. The size of the Cytochrome c nanoparticles obtained was 100-300 nm in diameter depending on the conditions used, indicating good potential to passively target tumors by the Enhanced Permeability and Retention effect. The surface of Cytochrome c nanoparticles was decorated with poly (lactic-co-glycolic) acid-SH via the linker succinimidyl 3-(2-pyridyldithio) propionate to prevent premature dissolution during delivery. The linker connecting the polymer to the protein nanoparticle contained a disulfide bond thus allowing polymer shedding and subsequent Cytochrome c release under intracellular reducing conditions. A cell-free caspase-3 assay revealed more than 80% of relative caspase activation by Cytochrome c after nanoprecipitation and polymer modification when compared to native Cytochrome c. Incubation of HeLa cells with the Cytochrome c based-nanoparticles showed significant reduction in cell viability after 6 hours while native Cytochrome c showed none. Confocal microscopy confirmed the induction of apoptosis in HeLa cells when they were stained with 4¿,6-diamidino-2-phenylindole and propidium iodide after incubation with the Cytochrome c-based nanoparticles.

Conclusions

Our results demonstrate that the coating with a hydrophobic polymer stabilizes Cytochrome c nanoparticles allowing for their delivery to the cytoplasm of target cells. After smart release of Cytochrome c into the cytoplasm, it induced programmed cell death.

【 授权许可】

   
2014 Morales-Cruz et al.; licensee BioMed Central Ltd.

【 预 览 】
附件列表
Files Size Format View
20150403093341971.pdf 2548KB PDF download
Figure 8. 18KB Image download
Figure 7. 24KB Image download
Figure 6. 15KB Image download
Figure 5. 50KB Image download
Figure 4. 19KB Image download
Figure 3. 57KB Image download
Figure 2. 18KB Image download
Figure 1. 50KB Image download
【 图 表 】

Figure 1.

Figure 2.

Figure 3.

Figure 4.

Figure 5.

Figure 6.

Figure 7.

Figure 8.

【 参考文献 】
  • [1]Danhier F, Feron O, Preat V: To exploit the tumor microenviroment: passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. J Control Release 2010, 148:135-146.
  • [2]Fang J, Nakamura H, Maeda H: The EPR effect: unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect. Adv Drug Deliv Rev 2011, 63:136-151.
  • [3]Morachis JM, Mahmoud EA, Almutairi A: Physical and chemical strategies for therapeutic delivery by using polymeric nanoparticles. Pharmacol Rev 2012, 64:505-519.
  • [4]Morigi V, Tocchio A, Bellavite-Pellegrini C, Sakamoto JH, Arnone M, Tasciotti E: Nanotechnology in medicine: from inception to market domination. J Drug Deliv 2012, 2012:1-7.
  • [5]Lanni JS, Lowe SW, Licitra EJ, Liu JO, Jacks T: p53-independent apoptosis induced by paclitaxel through an indirect mechanism. Proc Natl Acad Sci U S A 1997, 94:9679-9683.
  • [6]Ryan KM, Phillips AC, Vousden KH: Regulation and function of the p53 tumor suppressor protein. Curr Opin Cell Biol 2001, 13:332-337.
  • [7]Santra S, Kaittanis C, Perez JM: Cytochrome C encapsulating theranostic nanoparticles: a novel bifunctional system for targeted delivery of therapeutic membrane-impermeable proteins to tumors and imaging of cancer therapy. Mol Pharm 2010, 7:1209-1222.
  • [8]Yamada Y, Harashima H: Mitochondrial drug delivery systems for macromolecule and their therapeutic application to mitochondrial diseases. Adv Drug Deliv Rev 2008, 60:1439-1462.
  • [9]Solá RJ, Griebenow K: Effects of glycosylation on the stability of protein pharmaceuticals. J Pharm Sci 2009, 98:1223-1245.
  • [10]Manning MC, Chou DK, Murphy BM, Payne RW, Katayama DS: Stability of protein pharmaceuticals: an update. Pharm Res 2010, 27:544-575.
  • [11]Brown LR: Commercial challenges of protein drug delivery. Expert Opin Drug Deliv 2005, 2:29-42.
  • [12]Slowing II, Trewyn BG, Lin VSY: Mesoporous silica nanoparticles for intracellular delivery of membrane-impermeable proteins. J Am Chem Soc 2007, 129:8845-8849.
  • [13]Solaro R: Targeted delivery of protein drugs by nanocarriers. Materials 2010, 3:1928-1980.
  • [14]Morales-Cruz M, Flores-Fernández GM, Morales-Cruz M, Orellano EA, Rodriguez-Martinez JA, Ruiz M, Griebenow K: Two-step nanoprecipitation for the production of protein-loaded PLGA nanospheres. Results Pharma Sci 2012, 2:79-85.
  • [15]Méndez J, Morales-Cruz M, Delgado Y, Figueroa CM, Orellano EA, Morales M, Monteagudo A, Griebenow K: Delivery of chemically glycosylated cytochrome c immobilized in mesoporous silica nanoparticles induces apoptosis in hela cancer cells. Mol Pharm 2014, 11:102-111.
  • [16]Langer K, Balthasar S, Vogel V, Dinauer N, von Briesen H, Schubert D: Optimization of the preparation process for human serum albumin (HSA) nanoparticles. Int J Pharm 2003, 257:169-180.
  • [17]Jahanshahi M, Babaei Z: Protein nanoparticle: a unique system as drug delivery vehicles. Afr J Biotechnol 2008, 7:4926-4934.
  • [18]Zhao M, Biswas A, Hu B, Joo K, Wang P, Gu Z, Tang Y: Redox-responsive nanocapsules for intracellular protein delivery. Biomaterials 2011, 32:5223-5230.
  • [19]Shive M, Anderson J: Biodegradation and biocompatibility of PLA and PLGA microspheres. Adv Drug Deliv Rev 1997, 28:5-24.
  • [20]Lü JM, Wang X, Marin-Muller C, Wang H, Lin PH, Yao Q, Chen C: Current advances in research and clinical applications of PLGA based nanotechnology. Expert Rev Mol Diagn 2009, 4:325-341.
  • [21]Nitta SK, Numata K: Biopolymer-based nanoparticles for drug/gene delivery and tissue engineering. Int J Mol Sci 2013, 14:1629-1654.
  • [22]Torchilin V: Tumor delivery of macromolecular drugs based on the EPR effect. Adv Drug Deliv Rev 2011, 63:131-135.
  • [23]Kim KY: Nanotechnology platforms and physiological challenges for cancer therapeutics. Nanomedicine 2007, 3:103-110.
  • [24]Griebenow K, Diaz-Laureano Y, Santos AM, Montañez-Clemente I, Rodriguez L, Vidal M, Barletta G: Improved enzyme activity and enantioselectivity in organic solvents by methyl-?-cyclodextrin. J Am Chem Soc 1999, 121:8157-8163.
  • [25]Davies AM, Guillemette JG, Smith M, Greenwood C, Thurgood AG, Mauk AG, Moore GR: Redesign of the interior hydrophilic region of mitochondrial cytochrome c by site-directed mutagenesis. Biochemistry 1993, 32:5431-5435.
  • [26]Aachmann FL, Otzen DE, Larsen KL, Wimmer L: Structural background of cyclodextrin¿protein interactions. Protein Eng 2003, 16:905-912.
  • [27]Cooper A: Effect of cyclodextrins on the thermal stability of globular proteins. J Am Chem Soc 1992, 114:9208-9209.
  • [28]Méndez J, Monteagudo A, Griebenow K: Stimulus-responsive controlled release system by covalent immobilization of an enzyme into mesoporous silica nanoparticles. Bioconjug Chem 2012, 23:698-704.
  • [29]Besaratinia A, Pfeifer GP: A review of mechanisms of acrylamide carcinogenicity. Carcinogenesis 2007, 28:519-528.
  • [30]Yu B, Zhang Y, Zheng W, Fan C, Chen T: Positive surface charge enhances selective cellular uptake and anticancer efficacy of selenium nanoparticles. Inorg Chem 2012, 51:8956-8963.
  • [31]Choi SY, Jang SH, Park J, Jeong S, Park JH, Ock KS, Lee K, Yang SI, Joo SW, Ryu PD, Lee SY: Cellular uptake and cytotoxicity of positively charged chitosan gold nanoparticles in human lung adenocarcinoma cells. J Nanopart Res 2012, 14:1234.
  • [32]Panyam J, Zhou WZ, Prabha S, Sahoo SK, Labhasetwar V: Rapid endo-lysosomal escape of poly (DL-lactide-co-glycolide) nanoparticles: implications for drug and gene delivery. FASEB J 2002, 16:1217-1226.
  • [33]Castellanos IJ, Griebenow K: Improved ?-chymotrypsin stability upon encapsulation in PLGA microspheres by solvent replacement. Pharm Res 2003, 20:1873-1880.
  • [34]Bratton SB, Salvesen GS: Regulation of the apaf-1-caspase-9 apoptosome. J Cell Sci 2010, 123:3209-3214.
  • [35]Shacter E, Williams JA, Hinson RM, Senturker S, Lee YJ: Oxidative stress interferes with cancer chemotherapy: inhibition of lymphoma cell apoptosis and phagocytosis. Blood 2000, 96:307-313.
  文献评价指标  
  下载次数:36次 浏览次数:25次