51
|
Di Y, Gao Y, Gai X, Wang D, Wang Y, Yang X, Zhang D, Pan W, Yang X. Co-delivery of hydrophilic gemcitabine and hydrophobic paclitaxel into novel polymeric micelles for cancer treatment. RSC Adv 2017. [DOI: 10.1039/c7ra02909h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Schematic illustration of the preparation and intracellular performance of GEM–VE and PTX–VE loaded FA–PEG–VE micelle.
Collapse
Affiliation(s)
- Yan Di
- Department of Pharmaceutics
- School of Pharmacy
- Shenyang Pharmaceutical University
- Shenyang
- China
| | - Yunyun Gao
- Department of Pharmaceutics
- School of Pharmacy
- Shenyang Pharmaceutical University
- Shenyang
- China
| | - Xiumei Gai
- Department of Pharmaceutics
- School of Pharmacy
- Shenyang Pharmaceutical University
- Shenyang
- China
| | - Dun Wang
- Key Laboratory of Structure-Based Drug Design and Discovery
- Ministry of Education
- Shenyang Pharmaceutical University
- Shenyang
- China
| | - Yingying Wang
- Department of Pharmaceutics
- School of Pharmacy
- Shenyang Pharmaceutical University
- Shenyang
- China
| | - Xiaoguang Yang
- Key Laboratory of Structure-Based Drug Design and Discovery
- Ministry of Education
- Shenyang Pharmaceutical University
- Shenyang
- China
| | - Dan Zhang
- Liaoning Pharma-union Pharmaceutical Co. Ltd
- Benxi Economic Development Zone
- Benxi
- China
| | - Weisan Pan
- Department of Pharmaceutics
- School of Pharmacy
- Shenyang Pharmaceutical University
- Shenyang
- China
| | - Xinggang Yang
- Department of Pharmaceutics
- School of Pharmacy
- Shenyang Pharmaceutical University
- Shenyang
- China
| |
Collapse
|
52
|
Yang J, Bahreman A, Daudey G, Bussmann J, Olsthoorn RCL, Kros A. Drug Delivery via Cell Membrane Fusion Using Lipopeptide Modified Liposomes. ACS CENTRAL SCIENCE 2016; 2:621-630. [PMID: 27725960 PMCID: PMC5043431 DOI: 10.1021/acscentsci.6b00172] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Indexed: 05/27/2023]
Abstract
Efficient delivery of drugs to living cells is still a major challenge. Currently, most methods rely on the endocytotic pathway resulting in low delivery efficiency due to limited endosomal escape and/or degradation in lysosomes. Here, we report a new method for direct drug delivery into the cytosol of live cells in vitro and invivo utilizing targeted membrane fusion between liposomes and live cells. A pair of complementary coiled-coil lipopeptides was embedded in the lipid bilayer of liposomes and cell membranes respectively, resulting in targeted membrane fusion with concomitant release of liposome encapsulated cargo including fluorescent dyes and the cytotoxic drug doxorubicin. Using a wide spectrum of endocytosis inhibitors and endosome trackers, we demonstrate that the major site of cargo release is at the plasma membrane. This method thus allows for the quick and efficient delivery of drugs and is expected to have many invitro, ex vivo, and invivo applications.
Collapse
Affiliation(s)
| | | | - Geert Daudey
- Department of Supramolecular
Chemistry & Biomaterials, Leiden Institute of Chemistry, Leiden University, P.O.
Box 9502, Leiden, 2300 RA, The Netherlands
| | - Jeroen Bussmann
- Department of Supramolecular
Chemistry & Biomaterials, Leiden Institute of Chemistry, Leiden University, P.O.
Box 9502, Leiden, 2300 RA, The Netherlands
| | - René C. L. Olsthoorn
- Department of Supramolecular
Chemistry & Biomaterials, Leiden Institute of Chemistry, Leiden University, P.O.
Box 9502, Leiden, 2300 RA, The Netherlands
| | - Alexander Kros
- Department of Supramolecular
Chemistry & Biomaterials, Leiden Institute of Chemistry, Leiden University, P.O.
Box 9502, Leiden, 2300 RA, The Netherlands
| |
Collapse
|
53
|
Ahmed S, Fujita S, Matsumura K. Enhanced protein internalization and efficient endosomal escape using polyampholyte-modified liposomes and freeze concentration. NANOSCALE 2016; 8:15888-15901. [PMID: 27439774 DOI: 10.1039/c6nr03940e] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Here we show a new strategy for efficient freeze concentration-mediated cytoplasmic delivery of proteins, obtained via the endosomal escape property of polyampholyte-modified liposomes. The freeze concentration method successfully induces the efficient internalization of proteins simply by freezing cells with protein and nanocarrier complexes. However, the mechanism of protein internalization remains unclear. Here, we designed a novel protein delivery carrier by modifying liposomes through incorporating hydrophobic polyampholytes therein. These complexes were characterized for particle size, encapsulation efficiency, and cytotoxicity. Flow cytometry and microscopic analysis showed that the adsorption and internalization of protein-loaded polyampholyte-modified liposomes after freezing were enhanced compared with that observed in unfrozen complexes. Inhibition studies demonstrated that the internalization mechanism differs between unmodified and polyampholyte-modified liposomes. Furthermore, polyampholyte-modified liposomes exhibited high efficacy in facilitating endosomal escape to enhance protein delivery to the cytoplasm with low toxicity. These results strongly suggest that the freeze concentration-based strategy could be widely utilised for efficient cargo delivery into the cytoplasm in vitro not only in cancer treatment but also for gene therapy as well.
Collapse
Affiliation(s)
- Sana Ahmed
- School of Materials Science, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan.
| | | | | |
Collapse
|
54
|
Zubareva AA, Svirshchevskaya EV. Interactions of chitosan and its derivatives with cells (review). APPL BIOCHEM MICRO+ 2016. [DOI: 10.1134/s0003683816050185] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
55
|
Vidal F, Vásquez P, Díaz C, Nova D, Alderete J, Guzmán L. Mechanism of PAMAM Dendrimers Internalization in Hippocampal Neurons. Mol Pharm 2016; 13:3395-3403. [DOI: 10.1021/acs.molpharmaceut.6b00381] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Felipe Vidal
- Laboratory
of Molecular Neurobiology, Department of Physiology, Faculty of Biological
Sciences, University of Concepcion, Concepcion, Chile
| | - Pilar Vásquez
- Laboratory
of Molecular Neurobiology, Department of Physiology, Faculty of Biological
Sciences, University of Concepcion, Concepcion, Chile
| | - Carola Díaz
- Department
of Organic Chemistry, Faculty of Chemical Sciences, University of Concepcion, Concepcion, Chile
| | - Daniela Nova
- Laboratory
of Molecular Neurobiology, Department of Physiology, Faculty of Biological
Sciences, University of Concepcion, Concepcion, Chile
| | - Joel Alderete
- Department
of Organic Chemistry, Faculty of Chemical Sciences, University of Concepcion, Concepcion, Chile
| | - Leonardo Guzmán
- Laboratory
of Molecular Neurobiology, Department of Physiology, Faculty of Biological
Sciences, University of Concepcion, Concepcion, Chile
| |
Collapse
|
56
|
Lechanteur A, Furst T, Evrard B, Delvenne P, Hubert P, Piel G. PEGylation of lipoplexes: The right balance between cytotoxicity and siRNA effectiveness. Eur J Pharm Sci 2016; 93:493-503. [PMID: 27593989 DOI: 10.1016/j.ejps.2016.08.058] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/18/2016] [Accepted: 08/29/2016] [Indexed: 12/16/2022]
Abstract
The delivery of small interfering RNA (siRNA) is an attractive therapeutic approach to treat several pathologies, such as viral infections or cancers. However, the stability and the efficacy of these biotherapies are still a major obstacle to their use. Cationic liposomes (DOTAP/Chol/DOPE 1/0.75/0.5M ratio) have been complexed to siRNA (lipoplexes) in order to be administrated by the vaginal route, in the context of HPV16 induced cervical preneoplastic lesions. To overcome the constraint of the cervico-vaginal mucus, PEGylation is required to allow the diffusion of lipoplexes through it. Thereby, PEGylated lipoplexes coated with three types of polyethylene glycol (PEG) as DSPE-PEG2000, DSPE-PEG750 or C8-PEG2000-Ceramide (Ceramide-PEG2000) at different densities have been developed and characterized. PEGylated lipoplexes were successfully prepared and showed a hydrodynamic diameter around 200nm, appropriate for vaginal application. In vitro assays on HPV16 positive cell lines revealed that a positive charge of PEGylated lipoplexes allows a higher mRNA knockdown by siRNA. However, the cationic property is also associated to cytotoxicity. The addition of a high percentage of PEG prevented this toxicity but seemed also to reduce siRNA endosomal escape, probably by steric hindrance. The decreasing of PEG density of Ceramide-PEG2000 to 20% allows the release of siRNA and in consequence, biological activities, contrarily to DSPE-PEG. These results suggest that Ceramide-PEG is more appropriate for siRNA delivery compared to DSPE-PEG. In conclusion, the right balance between cytotoxicity and siRNA effectiveness has been found with the transfection of lipoplexes coated with 20% of Ceramide-PEG2000. This new nanovector could have a high potential against multiple mucosal diseases, such as human papillomavirus-induced genital lesions.
Collapse
Affiliation(s)
- Anna Lechanteur
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liège, Liège 4000, Belgium; Laboratory of Experimental Pathology, GIGA-Cancer, University of Liège, Liège 4000, Belgium.
| | - Tania Furst
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liège, Liège 4000, Belgium
| | - Brigitte Evrard
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liège, Liège 4000, Belgium
| | - Philippe Delvenne
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liège, Liège 4000, Belgium
| | - Pascale Hubert
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liège, Liège 4000, Belgium
| | - Géraldine Piel
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liège, Liège 4000, Belgium
| |
Collapse
|
57
|
Martínez-Negro M, Kumar K, Barrán-Berdón AL, Datta S, Kondaiah P, Junquera E, Bhattacharya S, Aicart E. Efficient Cellular Knockdown Mediated by siRNA Nanovectors of Gemini Cationic Lipids Having Delocalizable Headgroups and Oligo-Oxyethylene Spacers. ACS APPLIED MATERIALS & INTERFACES 2016; 8:22113-22126. [PMID: 27508330 DOI: 10.1021/acsami.6b08823] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The use of small interfering RNAs (siRNAs) to silence specific genes is one of the most promising approaches in gene therapy, but it requires efficient nanovectors for successful cellular delivery. Recently, we reported liposomal gene carriers derived from a gemini cationic lipid (GCL) of the 1,2-bis(hexadecyl dimethyl imidazolium) oligo-oxyethylene series ((C16Im)2(C2H4O)nC2H4 with n = 1, 2, or 3) and 1,2-dioleyol phosphatidylethanolamine as highly efficient cytofectins for pDNA. On the basis of the satisfactory outcomes of the previous study, the present work focuses on the utility of coliposomes of these gemini lipids with the biocompatible neutral lipid mono oleoyl glycerol (MOG) as highly potent vectors for siRNA cellular transport in the presence of serum. The (C16Im)2(C2H4O)nC2H4/MOG-siRNA lipoplexes were characterized through (i) a physicochemical study (zeta potential, cryo-transmission electron microscopy, small-angle X-ray scattering, and fluorescence anisotropy) to establish the relationship between size, structure, fluidity, and the interaction between siRNA and the GCL/MOG gene vectors and (ii) a biological analysis (flow cytometry, fluorescence microscopy, and cell viability) to report the anti-GFP siRNA transfections in HEK 293T, HeLa, and H1299 cancer cell lines. The in vitro biological analysis confirms the cellular uptake and indicates that a short spacer, a very low molar fraction of GCL in the mixed lipid, and a moderate effective charge ratio of the lipoplex yielded maximum silencing efficacy. At these experimental conditions, the siRNA used in this work is compacted by the GCL/MOG nanovectors by forming two cubic structures (Ia3d and Pm3n) that are correlated with excellent silencing activity. These liposomal nanocarriers possess high silencing activity with a negligible cytotoxicity, which strongly supports their practical use for in vivo knockdown studies.
Collapse
Affiliation(s)
- María Martínez-Negro
- Grupo de Química Coloidal y Supramolecular, Departamento de Química Física I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid , 28040 Madrid, Spain
| | | | - Ana L Barrán-Berdón
- Grupo de Química Coloidal y Supramolecular, Departamento de Química Física I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid , 28040 Madrid, Spain
| | | | | | - Elena Junquera
- Grupo de Química Coloidal y Supramolecular, Departamento de Química Física I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid , 28040 Madrid, Spain
| | | | - Emilio Aicart
- Grupo de Química Coloidal y Supramolecular, Departamento de Química Física I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid , 28040 Madrid, Spain
| |
Collapse
|
58
|
Zhang J, Liu D, Zhang M, Sun Y, Zhang X, Guan G, Zhao X, Qiao M, Chen D, Hu H. The cellular uptake mechanism, intracellular transportation, and exocytosis of polyamidoamine dendrimers in multidrug-resistant breast cancer cells. Int J Nanomedicine 2016; 11:3677-90. [PMID: 27536106 PMCID: PMC4977074 DOI: 10.2147/ijn.s106418] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Polyamidoamine dendrimers, which can deliver drugs and genetic materials to resistant cells, are attracting increased research attention, but their transportation behavior in resistant cells remains unclear. In this paper, we performed a systematic analysis of the cellular uptake, intracellular transportation, and efflux of PAMAM-NH2 dendrimers in multidrug-resistant breast cancer cells (MCF-7/ADR cells) using sensitive breast cancer cells (MCF-7 cells) as the control. We found that the uptake rate of PAMAM-NH2 was much lower and exocytosis of PAMAM-NH2 was much greater in MCF-7/ADR cells than in MCF-7 cells due to the elimination of PAMAM-NH2 from P-glycoprotein and the multidrug resistance-associated protein in MCF-7/ADR cells. Macropinocytosis played a more important role in its uptake in MCF-7/ADR cells than in MCF-7 cells. PAMAM-NH2 aggregated and became more degraded in the lysosomal vesicles of the MCF-7/ADR cells than in those of the MCF-7 cells. The endoplasmic reticulum and Golgi complex were found to participate in the exocytosis rather than endocytosis process of PAMAM-NH2 in both types of cells. Our findings clearly showed the intracellular transportation process of PAMAM-NH2 in MCF-7/ADR cells and provided a guide of using PAMAM-NH2 as a drug and gene vector in resistant cells.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang
| | - Dan Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang
| | - Mengjun Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang
| | - Yuqi Sun
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang; Department of Pharmaceutics, School of Pharmacy, Liaoning Medical University, Jinzhou, Liaoning Province, People's Republic of China
| | - Xiaojun Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang
| | - Guannan Guan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang
| | - Xiuli Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang
| | - Mingxi Qiao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang
| | - Dawei Chen
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang
| | - Haiyang Hu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang
| |
Collapse
|
59
|
Joshi A, Rastedt W, Faber K, Schultz AG, Bulcke F, Dringen R. Uptake and Toxicity of Copper Oxide Nanoparticles in C6 Glioma Cells. Neurochem Res 2016; 41:3004-3019. [DOI: 10.1007/s11064-016-2020-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 07/25/2016] [Accepted: 07/28/2016] [Indexed: 01/14/2023]
|
60
|
Mahlumba P, Choonara YE, Kumar P, du Toit LC, Pillay V. Stimuli-Responsive Polymeric Systems for Controlled Protein and Peptide Delivery: Future Implications for Ocular Delivery. Molecules 2016; 21:E1002. [PMID: 27483234 PMCID: PMC6273787 DOI: 10.3390/molecules21081002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/26/2016] [Accepted: 07/27/2016] [Indexed: 12/31/2022] Open
Abstract
Therapeutic proteins and peptides have become notable in the drug delivery arena for their compatibility with the human body as well as their high potency. However, their biocompatibility and high potency does not negate the existence of challenges resulting from physicochemical properties of proteins and peptides, including large size, short half-life, capability to provoke immune responses and susceptibility to degradation. Various delivery routes and delivery systems have been utilized to improve bioavailability, patient acceptability and reduce biodegradation. The ocular route remains of great interest, particularly for responsive delivery of macromolecules due to the anatomy and physiology of the eye that makes it a sensitive and complex environment. Research in this field is slowly gaining attention as this could be the breakthrough in ocular drug delivery of macromolecules. This work reviews stimuli-responsive polymeric delivery systems, their use in the delivery of therapeutic proteins and peptides as well as examples of proteins and peptides used in the treatment of ocular disorders. Stimuli reviewed include pH, temperature, enzymes, light, ultrasound and magnetic field. In addition, it discusses the current progress in responsive ocular drug delivery. Furthermore, it explores future prospects in the use of stimuli-responsive polymers for ocular delivery of proteins and peptides. Stimuli-responsive polymers offer great potential in improving the delivery of ocular therapeutics, therefore there is a need to consider them in order to guarantee a local, sustained and ideal delivery of ocular proteins and peptides, evading tissue invasion and systemic side-effects.
Collapse
Affiliation(s)
- Pakama Mahlumba
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Science, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa.
| | - Yahya E Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Science, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa.
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Science, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa.
| | - Lisa C du Toit
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Science, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa.
| | - Viness Pillay
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Science, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa.
| |
Collapse
|
61
|
Ren G, Jiang M, Xue P, Wang J, Wang Y, Chen B, He Z. A unique highly hydrophobic anticancer prodrug self-assembled nanomedicine for cancer therapy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:2273-2282. [PMID: 27389147 DOI: 10.1016/j.nano.2016.06.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 06/15/2016] [Accepted: 06/22/2016] [Indexed: 10/21/2022]
Abstract
In contrast with common thought, we generated highly hydrophobic anticancer prodrug self-assembled nanoparticles without the aid of surface active substances, based on the conjugation of docetaxel to d-α-tocopherol succinate. The reduction-sensitive prodrug was synthesized with a disulfide bond inserted into the linker and was compared with a control reduction-insensitive prodrug. The morphology and stability of self-assembled nanoparticles were investigated. Cytotoxicity and apoptosis assays showed that the reduction-sensitive nanoparticles had higher anticancer activity than the reduction-insensitive nanoparticles. The reduction-sensitive nanoparticles exhibited favorable in vivo antitumor activity and tolerance compared with docetaxel Tween80-containing formulation and the reduction-insensitive nanoparticles. Taken together, the unique nanomedicine demonstrated a number of advantages: (i) ease and reproducibility of preparation, (ii) high drug payload, (iii) superior stability, (iv) prolonged circulation, and (v) improved therapeutic effect. This highly reproducible molecular assembly strategy should motivate the development of new nanomedicines.
Collapse
Affiliation(s)
- Guolian Ren
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China; School of Pharmacy, Shanxi Medical University, Shanxi, China
| | - Mengjuan Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Peng Xue
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Jing Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Yongjun Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China.
| | - Bo Chen
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China.
| | - Zhonggui He
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
62
|
Real-time imaging and tracking of ultrastable organic dye nanoparticles in living cells. Biomaterials 2016; 93:38-47. [DOI: 10.1016/j.biomaterials.2016.03.045] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/23/2016] [Accepted: 03/30/2016] [Indexed: 12/30/2022]
|
63
|
Maussang D, Rip J, van Kregten J, van den Heuvel A, van der Pol S, van der Boom B, Reijerkerk A, Chen L, de Boer M, Gaillard P, de Vries H. Glutathione conjugation dose-dependently increases brain-specific liposomal drug delivery in vitro and in vivo. DRUG DISCOVERY TODAY. TECHNOLOGIES 2016; 20:59-69. [PMID: 27986226 DOI: 10.1016/j.ddtec.2016.09.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/27/2016] [Indexed: 06/06/2023]
Abstract
The blood-brain barrier (BBB) represents a major obstacle for the delivery and development of drugs curing brain pathologies. However, this biological barrier presents numerous endogenous specialized transport systems that can be exploited by engineered nanoparticles to enable drug delivery to the brain. In particular, conjugation of glutathione (GSH) onto PEGylated liposomes (G-Technology®) showed to safely enhance delivery of encapsulated drugs to the brain. Yet, understanding of the mechanism of action remains limited and full mechanistic understanding will aid in the further optimization of the technology. In order to elucidate the mechanism of brain targeting by GSH-PEG liposomes, we here demonstrate that the in vivo delivery of liposomal ribavirin is increased in brain extracellular fluid according to the extent of GSH conjugation onto the liposomes. In vitro, using the hCMEC/D3 human cerebral microvascular endothelial (CMEC) cell line, as well as primary bovine and porcine CMEC (and in contrast to non-brain derived endothelial and epithelial cells), we show that liposomal uptake occurs through the process of endocytosis and that the brain-specific uptake is also glutathione conjugation-dependent. Interestingly, the uptake mechanism is an active process that is temperature-, time- and dose-dependent. Finally, early endocytosis events rely on cytoskeleton remodeling, as well as dynamin- and clathrin-dependent endocytosis pathways. Overall, our data demonstrate that the glutathione-dependent uptake mechanism of the G-Technology involves a specific endocytosis pathway indicative of a receptor-mediated mechanism, and supports the benefit of this drug delivery technology for the treatment of devastating brain diseases.
Collapse
Affiliation(s)
- David Maussang
- to-BBB technologies BV (now 2-BBB Medicines BV), J.H. Oortweg 19, 2333 CH, Leiden, The Netherlands; Blood-Brain Barrier Research Group, Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Jaap Rip
- to-BBB technologies BV (now 2-BBB Medicines BV), J.H. Oortweg 19, 2333 CH, Leiden, The Netherlands
| | - Joan van Kregten
- to-BBB technologies BV (now 2-BBB Medicines BV), J.H. Oortweg 19, 2333 CH, Leiden, The Netherlands
| | - Angelique van den Heuvel
- to-BBB technologies BV (now 2-BBB Medicines BV), J.H. Oortweg 19, 2333 CH, Leiden, The Netherlands
| | - Susanne van der Pol
- Blood-Brain Barrier Research Group, Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Burt van der Boom
- to-BBB technologies BV (now 2-BBB Medicines BV), J.H. Oortweg 19, 2333 CH, Leiden, The Netherlands
| | - Arie Reijerkerk
- to-BBB technologies BV (now 2-BBB Medicines BV), J.H. Oortweg 19, 2333 CH, Leiden, The Netherlands
| | - Linda Chen
- to-BBB technologies BV (now 2-BBB Medicines BV), J.H. Oortweg 19, 2333 CH, Leiden, The Netherlands
| | - Marco de Boer
- to-BBB technologies BV (now 2-BBB Medicines BV), J.H. Oortweg 19, 2333 CH, Leiden, The Netherlands
| | - Pieter Gaillard
- to-BBB technologies BV (now 2-BBB Medicines BV), J.H. Oortweg 19, 2333 CH, Leiden, The Netherlands.
| | - Helga de Vries
- Blood-Brain Barrier Research Group, Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| |
Collapse
|
64
|
Srour B, Erhard B, Süss R, Hellwig P. Monitoring the pH Triggered Collapse of Liposomes in the Far IR Hydrogen Bonding Continuum. J Phys Chem B 2016; 120:4047-52. [DOI: 10.1021/acs.jpcb.6b03759] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Batoul Srour
- Laboratoire
de Bioélectrochimie et Spectroscopie, UMR 7140, Université de Strasbourg-CNRS, 1 Rue Blaise Pascal, Strasbourg 67070, France
| | - Birgit Erhard
- Institute
of Pharmaceutical Sciences, Department of Pharmaceutical Technology
and Biopharmacy and Freiburger Materialforschungszentrum (FMF), Albert Ludwig University Freiburg, Freiburg, Germany
| | - Regine Süss
- Institute
of Pharmaceutical Sciences, Department of Pharmaceutical Technology
and Biopharmacy and Freiburger Materialforschungszentrum (FMF), Albert Ludwig University Freiburg, Freiburg, Germany
| | - Petra Hellwig
- Laboratoire
de Bioélectrochimie et Spectroscopie, UMR 7140, Université de Strasbourg-CNRS, 1 Rue Blaise Pascal, Strasbourg 67070, France
| |
Collapse
|
65
|
Zhang X, Wang L, Zhou A, Zhou Q, Huang X. Alterations in cytosol free calcium in horseradish roots simultaneously exposed to lanthanum(III) and acid rain. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2016; 126:62-70. [PMID: 26720810 DOI: 10.1016/j.ecoenv.2015.12.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 12/11/2015] [Accepted: 12/14/2015] [Indexed: 05/29/2023]
Abstract
The extensive use of rare earth elements (REEs) has increased their environmental levels. REE pollution concomitant with acid rain in many agricultural regions can affect crop growth. Cytosol free calcium ions (Ca(2+)) play an important role in almost all cellular activities. However, no data have been reported regarding the role of cytosol free Ca(2+) in plant roots simultaneously exposed to REE and acid rain. In this study, the effects of exposures to lanthanum(III) and acid rain, independently and in combination, on cytosol free Ca(2+) levels, root activity, metal contents, biomass, cytosol pH and La contents in horseradish roots were investigated. The simultaneous exposures to La(III) and acid rain increased or decreased the cytosol free Ca(2+) levels, depending on the concentration of La(III), and these effects were more evident than independent exposure to La(III) or acid rain. In combined exposures, cytosol free Ca(2+) played an important role in the regulation of root activity, metal contents and biomass. These roles were closely related to La(III) dose, acid rain strength and treatment mode (independent exposure or simultaneous exposure). A low concentration of La(III) (20 mg L(-1)) could alleviate the adverse effects on the roots caused by acid rain, and the combined exposures at higher concentrations of La(III) and acid rain had synergic effects on the roots.
Collapse
Affiliation(s)
- Xuanbo Zhang
- State Key Laboratory of Food Science and Technology, School of Environment and Civil Engineering, Jiangnan University, Wuxi 214122, China
| | - Lihong Wang
- State Key Laboratory of Food Science and Technology, School of Environment and Civil Engineering, Jiangnan University, Wuxi 214122, China
| | - Anhua Zhou
- State Key Laboratory of Food Science and Technology, School of Environment and Civil Engineering, Jiangnan University, Wuxi 214122, China
| | - Qing Zhou
- State Key Laboratory of Food Science and Technology, School of Environment and Civil Engineering, Jiangnan University, Wuxi 214122, China.
| | - Xiaohua Huang
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Biomedical Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210046, China.
| |
Collapse
|
66
|
Hu B, Cheng R, Liu X, Pan X, Kong F, Gao W, Xu K, Tang B. A nanosensor for in vivo selenol imaging based on the formation of Au-Se bonds. Biomaterials 2016; 92:81-9. [PMID: 27043769 DOI: 10.1016/j.biomaterials.2016.03.030] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Revised: 03/17/2016] [Accepted: 03/17/2016] [Indexed: 12/22/2022]
Abstract
Selenol is a key metabolite of Na2SeO3 and plays an important role in many physiological and pathological processes. The real-time monitoring of selenol is of scientific interest for understanding the anti-cancer mechanism of Na2SeO3. Based on selenol's ability to specifically break AuS bonds and form more stable AuSe bonds on the surfaces of gold nanoparticles (AuNPs), we developed a novel near-infrared fluorescent nanosensor (Cy5.5-peptide-AuNPs) for detecting selenol. The nanosensor exhibited rapid response to selenol with high selectivity and sensitivity, and it was successfully used to image changes in the selenol level in HepG2 cells during Na2SeO3-induced apoptosis. Moreover, in vivo fluorescence imaging of selenol was obtained from H22 tumor-bearing mice injected with both the nanosensor and sodium selenite. The results showed that the tumor cell apoptosis induced by Na2SeO3 is correlated with high-level of selenol under hypoxic conditions. We believe that this nanosensor could serve as a powerful tool for monitoring selenol and exploring the physiological function of selenol in a variety of physiological and pathological contexts and that the probe-designed strategy will provide a new platform for research on relevant selenium chemistry.
Collapse
Affiliation(s)
- Bo Hu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, PR China
| | - Ranran Cheng
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, PR China
| | - Xiaojun Liu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, PR China
| | - Xiaohong Pan
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, PR China
| | - Fanpeng Kong
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, PR China
| | - Wen Gao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, PR China
| | - Kehua Xu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, PR China.
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, PR China.
| |
Collapse
|
67
|
Hu Y, Zhao Z, Ehrich M, Fuhrman K, Zhang C. In vitro controlled release of antigen in dendritic cells using pH-sensitive liposome-polymeric hybrid nanoparticles. POLYMER 2015; 80:171-179. [PMID: 26622069 DOI: 10.1016/j.polymer.2015.10.048] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A hybrid nanoparticle (NP) consisting of a pH sensitive lipid shell and a poly(lactic-co-glycolic) acid (PLGA) core was constructed. This hybrid NP has a mean size of 120.1 ± 8.8 nm and positively charged surface (zeta potential of 14.2 ± 1.4 mV). The lipid shell of the hybrid NP was quickly disintegrated in buffer with a pH of 5.5, which resembles the acidic environment of endosomes in dendritic cell (DC). Less than 20% of the antigen enclosed in pH-sensitive hybrid NP was released into human serum at physiological pH within 24 h, but more than 40% of the enclosed antigen was released within 8 h after pH was adjusted to 5.5. Fast uptake of the pH sensitive hybrid NP by DC was also observed. It was found that pH sensitive hybrid NP displayed faster degradation and antigen release compared to regular hybrid NPs after uptake by DC.
Collapse
Affiliation(s)
- Yun Hu
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - Zongmin Zhao
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - Marion Ehrich
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA 24061, USA
| | - Kristel Fuhrman
- Veterinary Medicine Experiment Station, Virginia Tech, Blacksburg, VA 24061, USA
| | - Chenming Zhang
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
68
|
Lopes-de-Araújo J, Neves AR, Gouveia VM, Moura CC, Nunes C, Reis S. Oxaprozin-Loaded Lipid Nanoparticles towards Overcoming NSAIDs Side-Effects. Pharm Res 2015; 33:301-14. [PMID: 26350105 DOI: 10.1007/s11095-015-1788-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 08/31/2015] [Indexed: 01/15/2023]
Abstract
PURPOSE Nanostructured Lipid Carriers (NLCs) loading oxaprozin were developed to address an effective drug packaging and targeted delivery, improving the drug pharmacokinetics and pharmacodynamics properties and avoiding the local gastric side-effects. Macrophages actively phagocyte particles with sizes larger than 200 nm and, when activated, over-express folate beta receptors - features that in the case of this work constitute the basis for passive and active targeting strategies. METHODS Two formulations containing oxaprozin were developed: NLCs with and without folate functionalization. In order to target the macrophages folate receptors, a DSPE-PEG2000-FA conjugate was synthesized and added to the NLCs. RESULTS These formulations presented a relatively low polydispersity index (approximately 0.2) with mean diameters greater than 200 nm and zeta potential inferior to -40 mV. The encapsulation efficiency of the particles was superior to 95% and the loading capacity was of 9%, approximately. The formulations retained the oxaprozin release in simulated gastric fluid (only around 10%) promoting its release on simulated intestinal fluid. MTT and LDH assays revealed that the formulations only presented cytotoxicity in Caco-2 cells for oxaprozin concentrations superior to 100 μM. Permeability studies in Caco-2 cells shown that oxaprozin encapsulation did not interfered with oxaprozin permeability (around 0.8 × 10(-5) cm/s in simulated intestinal fluid and about 1.45 × 10(-5) cm/s in PBS). Moreover, in RAW 264.7 cells NLCs functionalization promoted an increased uptake over time mainly mediated by a caveolae uptake mechanism. CONCLUSIONS The developed nanoparticles enclose a great potential for oxaprozin oral administration with significant less gastric side-effects.
Collapse
Affiliation(s)
- José Lopes-de-Araújo
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Ana Rute Neves
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Virgínia M Gouveia
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Catarina C Moura
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Cláudia Nunes
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| | - Salette Reis
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| |
Collapse
|
69
|
Mikhalin AA, Evdokimov NM, Frolova LV, Magedov IV, Kornienko A, Johnston R, Rogelj S, Tartis MS. Lipophilic prodrug conjugates allow facile and rapid synthesis of high-loading capacity liposomes without the need for post-assembly purification. J Liposome Res 2015; 25:232-260. [PMID: 25534989 PMCID: PMC4478286 DOI: 10.3109/08982104.2014.992022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Dihydropyridopyrazoles are simplified synthetic analogues of podophyllotoxin that can effectively mimic its molecular scaffold and act as potent mitotic spindle poisons in dividing cancer cells. However, despite nanomolar potencies and ease of synthetic preparation, further clinical development of these promising anticancer agents is hampered due to their poor aqueous solubility. In this article, we developed a prodrug strategy that enables incorporation of dihydropyridopyrazoles into liposome bilayers to overcome the solubility issues. The active drug was covalently connected to either myristic or palmitic acid anchor via carboxylesterase hydrolyzable linkage. The resulting prodrugs were self-assembled into liposome bilayers from hydrated lipid films using ultrasound without the need for post-assembly purification. The average particle size of the prodrug-loaded liposomes was about 90 nm. The prodrug incorporation was verified by differential scanning calorimetry, spectrophotometry and gel filtration reaching maximum at 0.3 and 0.35 prodrug/lipid molar ratios for myristic and palmitic conjugates, respectively. However, the ratio of 0.2 was used in the particle size and biological activity experiments to maintain long-term stability of the prodrug-loaded liposomes against phase separation during storage. Antiproliferative activity was tested against HeLa and Jurkat cancer cell lines in vitro showing that the liposomal prodrug retained antitubulin activity of the parent drug and induced apoptosis-mediated cancer cell death. Overall, the established data provide a powerful platform for further clinical development of dihydropyridopyrazoles using liposomes as the drug delivery system.
Collapse
Affiliation(s)
- Alexander A. Mikhalin
- Department of Biology, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA
- Department of Chemical Engineering, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA
| | - Nikolai M. Evdokimov
- Department of Chemistry, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA
| | - Liliya V. Frolova
- Department of Chemistry, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA
| | - Igor V. Magedov
- Department of Chemistry, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA
| | - Alexander Kornienko
- Department of Chemistry, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA
| | - Robert Johnston
- Department of Materials Engineering, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA
| | - Snezna Rogelj
- Department of Biology, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA
| | - Michaelann S. Tartis
- Department of Biology, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA
- Department of Materials Engineering, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA
- Department of Chemical Engineering, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA
| |
Collapse
|
70
|
Salem HF, Ahmed SM, Hassaballah AE, Omar MM. Targeting brain cells with glutathione-modulated nanoliposomes: in vitro and in vivo study. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:3705-27. [PMID: 26229435 PMCID: PMC4516201 DOI: 10.2147/dddt.s85302] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background The blood–brain barrier prevents many drug moieties from reaching the central nervous system. Therefore, glutathione-modulated nanoliposomes have been engineered to enhance the targeting of flucytosine to the brain. Methods Glutathione-modulated nanoliposomes were prepared by thin-film hydration technique and evaluated in the primary brain cells of rats. Lecithin, cholesterol, and span 65 were mixed at 1:1:1 molar ratio. The molar percentage of PEGylated glutathione varied from 0 mol% to 0.75 mol%. The cellular binding and the uptake of the targeted liposomes were both monitored by epifluorescent microscope and flow cytometry techniques. A biodistribution and a pharmacokinetic study of flucytosine and flucytosine-loaded glutathione–modulated liposomes was carried out to evaluate the in vivo brain-targeting efficiency. Results The size of glutathione-modulated nanoliposomes was <100 nm and the zeta potential was more than −65 mV. The cumulative release reached 70% for certain formulations. The cellular uptake increased as molar percent of glutathione increased to reach the maximum at 0.75 mol%. The uptake of the targeted liposomes by brain cells of the rats was three times greater than that of the nontargeted liposomes. An in vivo study showed that the relative efficiency was 2.632±0.089 and the concentration efficiency was 1.590±0.049, and also, the drug-targeting index was 3.670±0.824. Conclusion Overall, these results revealed that glutathione-PEGylated nanoliposomes enhance the effective delivery of flucytosine to brain and could become a promising new therapeutic option for the treatment of the brain infections.
Collapse
Affiliation(s)
- Heba F Salem
- Department of Pharmaceutics and Industrial Pharmacy, Beni-suef University, Assuit, Egypt
| | - Sayed M Ahmed
- Department of Industrial Pharmacy, Assiut University, Egypt
| | - Ashraf E Hassaballah
- Department of Clinical Pathology, Faculty of Medicine, Assiut University, Assuit, Egypt
| | - Mahmoud M Omar
- Department of Pharmaceutics and Industrial Pharmacy, Beni-suef University, Assuit, Egypt ; Department of Pharmaceutics and Industrial Pharmacy, Deraya University, Egypt
| |
Collapse
|
71
|
Abstract
Parthenolide (PTL) has shown great promise as a novel anti-leukemia agent as it selectively eliminates acute myeloid leukemia (AML) blast cells and leukemia stem cells (LSCs) while sparing normal hematopoietic cells. This success has not yet translated to the clinical setting because PTL is rapidly cleared from blood due to its hydrophobicity. To increase the aqueous solubility of PTL, we previously developed micelles formed from predominantly hydrophobic amphiphilic diblock copolymers of poly(styrene-alt-maleic anhydride)-b-poly(styrene) (e.g., PSMA100-b-PS258) that exhibit robust PTL loading (75%efficiency, 11% w/w capacity) and release PTL over 24 h. Here, PTL-loaded PSMA-b-PS micelles were thoroughly characterized in vitro for PTL delivery to MV4-11 AML cells. Additionally, the mechanisms governing micelle-mediated cytotoxicity were examined in comparison to free PTL. PSMA-b-PS micelles were taken up by MV4-11 cells as evidenced by transmission electron microscopy and flow cytometry. Specifically, MV4-11 cells relied on clathrin-mediated endocytosis, rather than caveolae-mediated endocytosis and macropinocytosis. In addition, PTL-loaded PSMA-b-PS micelles exhibited a dose-dependent cytotoxicity towards AML cells and were capable of reducing cell viability by 75% at 10 μM PTL, while unloaded micelles were nontoxic. At 10 μM PTL, the cytotoxicity of PTL-loaded micelles increased gradually over 24 h while free PTL achieved maximal cytotoxicity between 2 and 4 h, demonstrating micelle-mediated delivery of PTL to AML cells and stability of the drug-loaded micelle even in the presence of cells. Both free PTL and PTL-loaded micelles induced NF-κB inhibition at 10 μM PTL doses, demonstrating some mechanistic similarities in cytotoxicity. However, free PTL relied more heavily on exofacial free thiol interactions to induce cytotoxicity than PTL-loaded micelles; free PTL cytotoxicity was reduced by over twofold when cell surface free thiols were depleted, where PTL-loaded micelle doses were unaffected by cell surface thiol modulation. The physical properties, stability, and efficacy of PTL-loaded PSMA-b-PS micelles support further development of a leukemia therapeutic with greater bioavailability and the potential to eliminate LSCs.
Collapse
|
72
|
He X, Li L, Su H, Zhou D, Song H, Wang L, Jiang X. Poly(ethylene glycol)-block-poly(ε-caprolactone)-and phospholipid-based stealth nanoparticles with enhanced therapeutic efficacy on murine breast cancer by improved intracellular drug delivery. Int J Nanomedicine 2015; 10:1791-804. [PMID: 25784805 PMCID: PMC4356685 DOI: 10.2147/ijn.s75186] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Effective anticancer drug delivery to the tumor site without rapid body clearance is a prerequisite for successful chemotherapy. 1,2-distearoyl-sn-glycero-3-phospho-ethanolamine-N-(methoxy[polyethyleneglycol]-2000) (DSPE-PEG2000) has been widely used in the preparation of stealth liposomes. Although PEG chains can efficiently preserve liposomes from rapid clearance by the reticuloendothelial system (RES), its application has been hindered by poor cellular uptake and unsatisfactory therapeutic effect. METHODS To address the dilemma, we presented a facile approach to fabricate novel stealth nanoparticles generated by poly(ethylene glycol)-block-poly(ε-caprolactone) (PEG-b-PCL), soybean phosphatidylcholine (SPC), and cholesterol, namely LPPs (L represented lipid and PP represented PEG-b-PCL), for the delivery of anticancer drug paclitaxel (PTX). LPPs were prepared using the thin film hydration method. Two PEG-b-PCL polymers with different molecular weights (MW; PEG2000-b-PCL2000, MW: 4,000 Da and PEG5000-b-PCL5000, MW: 10,000 Da) were used to fabricate stealth nanoparticles. Conventional PEGylated liposome (LDP2000, L represented lipid and DP2000 represented DSPE-PEG2000) composed of SPC, cholesterol, and DSPE-PEG2000 was used as the control. The physical properties, cellular uptake, endocytosis pathway, cytotoxicity, pharmacokinetics, tumor accumulation, and anticancer efficacy of free PTX, PTX-loaded LPPs, and LDP2000 were systemically investigated after injection into 4T1 breast tumor-bearing mice. RESULTS LPPs were vesicles around 100 nm in size with negative zeta potential. With enhanced stability, LPPs achieved sustainable release of cancer therapeutics. The cellular uptake level was closely related to the PEG chain length of PEG-b-PCL; a shorter PEG chain resulted in higher cellular uptake. Moreover, the cellular internalization of LPP2000 modified by PEG2000-b-PCL2000 on 4T1 cells was 2.1-fold higher than LDP2000 due to the improved stability of LPP2000. The cytotoxicity of PTX-loaded LPP2000 was also higher than that of LDP2000 and LPP5000 as observed using a WST-8 assay, while blank LPPs showed negligible toxicity. Consistent with the results of the in vitro study, in vivo experiments showed that LPPs allowed significantly improved bioavailability and prolonged T1/2β as compared to free PTX injection. More importantly, LPPs mainly accumulated at the tumor site, probably due to the enhanced permeation and retention effect (EPR effect). As a nanomedicine, LPP2000 (tumor inhibition rate of 75.1%) significantly enhanced the therapeutic effect of PTX in 4T1 breast tumor-bearing mice by inhibiting tumor growth compared to LDP2000 and LPP5000 (tumor inhibition rates of 56.3% and 49.5%, respectively). CONCLUSION Modification of liposomes with PEG2000-b-PCL2000 can simultaneously improve drug accumulation at the target tumor site and tumor cells, showing great promise for utilization as a PEG modification tool in the fabrication of stealth nanoparticles for cancer chemotherapy.
Collapse
Affiliation(s)
- Xiaodan He
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Li Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Hong Su
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Dinglun Zhou
- West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Hongmei Song
- HitGen Ltd., Chengdu, Sichuan, People's Republic of China
| | - Ling Wang
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Xuehua Jiang
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
73
|
Fu H, Shi K, Hu G, Yang Y, Kuang Q, Lu L, Zhang L, Chen W, Dong M, Chen Y, He Q. Tumor-Targeted Paclitaxel Delivery and Enhanced Penetration Using TAT-Decorated Liposomes Comprising Redox-Responsive Poly(Ethylene Glycol). J Pharm Sci 2015; 104:1160-73. [DOI: 10.1002/jps.24291] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 10/25/2014] [Accepted: 11/06/2014] [Indexed: 12/27/2022]
|
74
|
Peters T, Grunewald C, Blaickner M, Ziegner M, Schütz C, Iffland D, Hampel G, Nawroth T, Langguth P. Cellular uptake and in vitro antitumor efficacy of composite liposomes for neutron capture therapy. Radiat Oncol 2015; 10:52. [PMID: 25889824 PMCID: PMC4349485 DOI: 10.1186/s13014-015-0342-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 01/29/2015] [Indexed: 11/21/2022] Open
Abstract
Background Neutron capture therapy for glioblastoma has focused mainly on the use of 10B as neutron capture isotope. However, 157Gd offers several advantages over boron, such as higher cross section for thermal neutrons and the possibility to perform magnetic resonance imaging during neutron irradiation, thereby combining therapy and diagnostics. We have developed different liposomal formulations of gadolinium-DTPA (Magnevist®) for application in neutron capture therapy of glioblastoma. The formulations were characterized physicochemically and tested in vitro in a glioma cell model for their effectiveness. Methods Liposomes entrapping gadolinium-DTPA as neutron capture agent were manufactured via lipid/film-extrusion method and characterized with regard to size, entrapment efficiency and in vitro release. For neutron irradiation, F98 and LN229 glioma cells were incubated with the newly developed liposomes and subsequently irradiated at the thermal column of the TRIGA reactor in Mainz. The dose rate derived from neutron irradiation with 157Gd as neutron capturing agent was calculated via Monte Carlo simulations and set in relation to the respective cell survival. Results The liposomal Gd-DTPA reduced cell survival of F98 and LN229 cells significantly. Differences in liposomal composition of the formulations led to distinctly different outcome in cell survival. The amount of cellular Gd was not at all times proportional to cell survival, indicating that intracellular deposition of formulated Gd has a major influence on cell survival. The majority of the dose contribution arises from photon cross irradiation compared to a very small Gd-related dose. Conclusions Liposomal gadolinium formulations represent a promising approach for neutron capture therapy of glioblastoma cells. The liposome composition determines the uptake and the survival of cells following radiation, presumably due to different uptake pathways of liposomes and intracellular deposition of gadolinium-DTPA. Due to the small range of the Auger and conversion electrons produced in 157Gd capture, the proximity of Gd-atoms to cellular DNA is a crucial factor for infliction of lethal damage. Furthermore, Gd-containing liposomes may be used as MRI contrast agents for diagnostic purposes and surveillance of tumor targeting, thus enabling a theranostic approach for tumor therapy.
Collapse
Affiliation(s)
- Tanja Peters
- Institute of Pharmacy and Biochemistry, Department of Biopharmaceutics and Pharmaceutical Technology, Johannes Gutenberg University Mainz, Staudingerweg 5, D-55128, Mainz, Germany.
| | - Catrin Grunewald
- Institute of Nuclear Chemistry, Johannes Gutenberg University Mainz, Fritz-Strassmann Weg 6, D-55128, Mainz, Germany.
| | - Matthias Blaickner
- AIT Austrian Institute of Technology, Health & Environment Department, Biomedical Systems, Donau-City-Strasse 1/2, A-1220, Vienna, Austria.
| | - Markus Ziegner
- AIT Austrian Institute of Technology, Health & Environment Department, Biomedical Systems, Donau-City-Strasse 1/2, A-1220, Vienna, Austria.
| | - Christian Schütz
- Institute of Nuclear Chemistry, Johannes Gutenberg University Mainz, Fritz-Strassmann Weg 6, D-55128, Mainz, Germany.
| | - Dorothee Iffland
- Institute of Nuclear Chemistry, Johannes Gutenberg University Mainz, Fritz-Strassmann Weg 6, D-55128, Mainz, Germany.
| | - Gabriele Hampel
- Institute of Nuclear Chemistry, Johannes Gutenberg University Mainz, Fritz-Strassmann Weg 6, D-55128, Mainz, Germany.
| | - Thomas Nawroth
- Institute of Pharmacy and Biochemistry, Department of Biopharmaceutics and Pharmaceutical Technology, Johannes Gutenberg University Mainz, Staudingerweg 5, D-55128, Mainz, Germany.
| | - Peter Langguth
- Institute of Pharmacy and Biochemistry, Department of Biopharmaceutics and Pharmaceutical Technology, Johannes Gutenberg University Mainz, Staudingerweg 5, D-55128, Mainz, Germany.
| |
Collapse
|
75
|
Zhang X, Li Y, Cheng Y, Tan H, Li Z, Qu Y, Mu G, Wang F. Tat PTD-endostatin: A novel anti-angiogenesis protein with ocular barrier permeability via eye-drops. Biochim Biophys Acta Gen Subj 2015; 1850:1140-9. [PMID: 25662072 DOI: 10.1016/j.bbagen.2015.01.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 12/28/2014] [Accepted: 01/30/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Endostatin, a specific inhibitor of endothelial cell proliferation and angiogenesis, has been proved to have effects on ocular neovascular diseases by intraocular injection. In order to increase its permeability to ocular barriers and make it effective on fundus oculi angiogenesis diseases via non-invasive administration (eye drops), endostatin was fused to Tat PTD via a genetic engineering method. METHODS Most of the Tat PTD- endostatin was expressed as inclusion bodies in Escherichia coli, so pure and active Tat PTD-endostatin was prepared by a series of operations, including inclusion body denaturation, refolding and chromatography. The anti-angiogenesis activity of Tat PTD-endostatin was investigated by cell proliferation experiments and chick embryo chorioallantoic membrane assay. In addition, its translocating ability and concrete entry mechanism into cells were also investigated by fluorescence microscope and flow cytometry. The penetrating ability to ocular barriers was also studied by immunohistochemistry. A mouse choroidal neovascularization model was established to investigate the pharmacodynamics of Tat PTD-endostatin. RESULTS The obtained Tat PTD-endostatin had excellent anti-angiogenesis activity and was superior to Es in cellular translocating. Macropinocytosis may be the dominant route of entry of Tat PTD-endostatin into cells. Tat PTD-endostatin could cross ocular barriers and arrive at the retina after eye-drop administration. In addition, it displayed inhibitory effects on choroidal neovascularization via eye drops. CONCLUSIONS Tat PTD-endostatin possessed excellent ocular penetrating ability and anti-angiogenesis effects. GENERAL SIGNIFICANCE Tat PTD is a promising ocular delivery tool, and Tat PTD-endostatin is a potential drug for curing fundus oculi angiogenesis diseases.
Collapse
Affiliation(s)
- Xinke Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yan Li
- Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yanna Cheng
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Haining Tan
- National Glycoengineering Research Center, Shandong University, Jinan, China
| | - Zhiwei Li
- Department of Ophthalmology, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yi Qu
- Department of Health Care, QiLu Hospital of Shandong University, Jinan, China
| | - Guoying Mu
- Department of Ophthalmology, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Fengshan Wang
- Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Shandong University, Jinan, China.
| |
Collapse
|
76
|
Shen Y, Wang J, Li Y, Tian Y, Sun H, Ammar O, Tu J, Wang B, Sun C. Co-delivery of siRNA and paclitaxel into cancer cells by hyaluronic acid modified redox-sensitive disulfide-crosslinked PLGA–PEI nanoparticles. RSC Adv 2015. [DOI: 10.1039/c5ra03085d] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Schematic diagram showing the structure of the co-delivery nano-complex and the process of entering tumor cells.
Collapse
Affiliation(s)
- Yan Shen
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
| | - Jue Wang
- National Institute for Food and Drug Control
- Beijing
- China
| | - Yanan Li
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
| | - Yu Tian
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
| | - Huimin Sun
- National Institute for Food and Drug Control
- Beijing
- China
| | - Ouahab Ammar
- Department of Pharmacy
- Institute of Medical Sciences
- Batna Elhadj Lakhdar University
- Algeria
| | - Jiasheng Tu
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
| | - Buhai Wang
- Department of Oncology
- Subei People's Hospital
- Yangzhou
- China
| | - Chunmeng Sun
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
| |
Collapse
|
77
|
Guilloteau N, Bienvenu C, Charrat C, Jiménez Blanco JL, Díaz-Moscoso A, Mellet CO, García Fernández JM, Vierling P, Di Giorgio C. Cell uptake mechanisms of glycosylated cationic pDNA–cyclodextrin nanoparticles. RSC Adv 2015. [DOI: 10.1039/c5ra00964b] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
β-Cyclodextrin-based glycoCDplexes are internalized through several redundant pathways whose relative prevalence depends on the coating sugar and on the cell line.
Collapse
Affiliation(s)
- Nicolas Guilloteau
- Institut de Chimie de Nice
- UMR 7272
- Université de Nice Sophia Antipolis
- CNRS
- F-06108 Nice
| | - Céline Bienvenu
- Institut de Chimie de Nice
- UMR 7272
- Université de Nice Sophia Antipolis
- CNRS
- F-06108 Nice
| | - Coralie Charrat
- Institut de Chimie de Nice
- UMR 7272
- Université de Nice Sophia Antipolis
- CNRS
- F-06108 Nice
| | - José L. Jiménez Blanco
- Departamento de Química Orgánica
- Facultad de Química
- Universidad de Sevilla
- E-41012 Sevilla
- Spain
| | - Alejandro Díaz-Moscoso
- Instituto de Investigaciones Químicas (IIQ)
- CSIC – Universidad de Sevilla
- E-41092 Sevilla
- Spain
| | - Carmen Ortiz Mellet
- Departamento de Química Orgánica
- Facultad de Química
- Universidad de Sevilla
- E-41012 Sevilla
- Spain
| | | | - Pierre Vierling
- Institut de Chimie de Nice
- UMR 7272
- Université de Nice Sophia Antipolis
- CNRS
- F-06108 Nice
| | - Christophe Di Giorgio
- Institut de Chimie de Nice
- UMR 7272
- Université de Nice Sophia Antipolis
- CNRS
- F-06108 Nice
| |
Collapse
|
78
|
Zhang W, Sun J, Fang W, Ai X, Cai C, Tang Y, Su X, Feng Z, Liu Y, Tao M, Yan X, Chen G, He Z. Nanomicelles based on X-shaped four-armed peglyated distearylglycerol as long circulating system for doxorubicin delivery. Eur J Pharm Sci 2015; 66:96-106. [DOI: 10.1016/j.ejps.2014.10.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 09/28/2014] [Accepted: 10/07/2014] [Indexed: 11/24/2022]
|
79
|
Angius F, Floris A. Liposomes and MTT cell viability assay: an incompatible affair. Toxicol In Vitro 2014; 29:314-9. [PMID: 25481524 DOI: 10.1016/j.tiv.2014.11.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 11/21/2014] [Accepted: 11/24/2014] [Indexed: 11/18/2022]
Abstract
The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay is commonly used to evaluate the cytotoxicity potential of drugs vehicled by liposomes. However, liposome delivering drugs could produce inconsistent values of MTT absorbance. On the basis of previous experiments demonstrating the MTT affinity for lipid droplets, this paper aims to show that empty-liposomes interfere, per se, on MTT assay due to its lipidic nature. This brings into question the use of MTT testing cytotoxicity when liposomes are involved in delivering drugs.
Collapse
Affiliation(s)
- Fabrizio Angius
- Department of Biomedical Sciences, University of Cagliari, Italy.
| | - Alice Floris
- Department of Life and Environment Sciences, University of Cagliari, Italy
| |
Collapse
|
80
|
Mauroy C, Castagnos P, Orio J, Blache MC, Rico-Lattes I, Teissié J, Rols MP, Blanzat M. Versatile cellular uptake mediated by catanionic vesicles: simultaneous spontaneous membrane fusion and endocytosis. Mol Pharm 2014; 12:103-10. [PMID: 25310849 DOI: 10.1021/mp500458f] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Lactose-derived catanionic vesicles offer unique opportunities to overcome cellular barriers. These potential nanovectors, very easy to formulate as drug delivery systems, are able to encapsulate drugs of various hydrophilicity. This article highlights versatile interaction mechanisms between these catanionic vesicles, labeled with hydrophilic and amphiphilic fluorescent probes, and a mammalian cell line, Chinese Hamster Ovary. Confocal microscopy and flow cytometry techniques show that these vesicles are internalized by cells through cellular energy dependent processes, as endocytosis, but are simultaneously able to spontaneously fuse with cell plasma membranes and release their hydrophilic content directly inside the cytosol. Such innovative and polyvalent nanovectors, able to deliver their content via different internalization pathways, would positively be a great progress for the coadministration of drugs of complementary efficiency.
Collapse
Affiliation(s)
- Chloé Mauroy
- Laboratoire des Interactions Moléculaires et Réactivité Chimique et Photochimique, UMR 5623 Université Paul Sabatier and Centre National de la Recherche Scientifique , 118 route de Narbonne, 31062 Toulouse, France
| | | | | | | | | | | | | | | |
Collapse
|
81
|
Stealth CD44-targeted hyaluronic acid supramolecular nanoassemblies for doxorubicin delivery: probing the effect of uncovalent pegylation degree on cellular uptake and blood long circulation. J Control Release 2014; 197:29-40. [PMID: 25449802 DOI: 10.1016/j.jconrel.2014.10.024] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 10/16/2014] [Accepted: 10/27/2014] [Indexed: 11/21/2022]
Abstract
Stealth active targeting nanoparticles (NPs) usually include two types of ligand sites: ligand anchored on distal ends of the polyethylene glycol (PEG) and ligand buried under pegylated layer. The latter typical case is hyaluronic acid (HA)-based NPs; however, there is little information available for the latter NPs about effect of the optimal density of surface PEG coating on the blood circulation time, cellular uptake and in vivo anticancer activity. Thus, in this study, in order to optimize the anticancer effects of HA-based NPs, we focus on how uncovalent pegylation degree modulates blood circulation time and cellular uptake of HA-based NPs. We firstly designed a new double-hydrophilic copolymer by conjugating HP-β-cyclodextrin with HA, and this carrier was further pegylated with adamantyl-peg (ADA-PEG) to form inclusion complex HA-HPCD/ADA-PEG, termed as HCPs. The supramolecular nanoassemblies were fabricated by host-guest and polar interactions between HCPs and doxorubicin (Dox), with vitamin E succinate (VES) being a nanobridge. Despite the active recognition between HA and CD44 receptor, the cellular uptake and targeting efficiency of HA-NPs decreased with the increasing peg density, demonstrating HA was partly buried by high density peg coating. However, the high density of peg coating was beneficial to long circulation time, tumor biodistribution and anticancer activity in vivo. NPs with 5% peg coating had the optimal cellular targeting efficiency in vitro and anticancer effects in vivo. The findings suggest that balancing long circulation property and cellular uptake is important to achieve the optimal antitumor efficacy for pegylated HA-based NPs, and that PEG coating densities cannot be extended beyond a certain density for shielding effect without compromising the efficacy of hyaluronic acid targeted delivery.
Collapse
|
82
|
Alves P, Hugo A, Szymanowski F, Tymczyszyn E, Pérez P, Coelho J, Simões P, Gómez-Zavaglia A. Stabilization of polymer lipid complexes prepared with lipids of lactic acid bacteria upon preservation and internalization into eukaryotic cells. Colloids Surf B Biointerfaces 2014; 123:446-51. [DOI: 10.1016/j.colsurfb.2014.09.043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/17/2014] [Accepted: 09/19/2014] [Indexed: 02/07/2023]
|
83
|
Abstract
In recent years, hundreds of genes have been linked to a variety of human diseases, and the field of gene therapy has emerged as a way to treat this wide range of diseases. The main goal of gene therapy is to find a gene delivery vehicle that can successfully target diseased cells and deliver therapeutic genes directly to their cellular compartment. The two main types of gene delivery vectors currently being investigated in clinical trials are recombinant viral vectors and synthetic nonviral vectors. Recombinant viral vectors take advantage of the evolutionarily optimized viral mechanisms to deliver genes, but they can be hard to specifically target in vivo and are also associated with serious side effects. Synthetic nonviral vectors are made out of highly biocompatible lipids or polymers, but they are much less efficient at delivering their genetic payload due to the lack of any active delivery mechanism. This mini review will introduce the current state of gene delivery in clinical trials, and discuss the specific challenges associated with each of these vectors. It will also highlight some specific gaps in knowledge that are limiting the advancement of this field and touch on the current areas of research being explored to overcome them.
Collapse
Affiliation(s)
- Yarong Liu
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California, USA
| | - Jennifer Rohrs
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Pin Wang
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California, USA
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
84
|
Stefanutti E, Papacci F, Sennato S, Bombelli C, Viola I, Bonincontro A, Bordi F, Mancini G, Gigli G, Risuleo G. Cationic liposomes formulated with DMPC and a gemini surfactant traverse the cell membrane without causing a significant bio-damage. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1838:2646-55. [PMID: 25017801 DOI: 10.1016/j.bbamem.2014.05.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 05/14/2014] [Accepted: 05/24/2014] [Indexed: 11/29/2022]
Abstract
Cationic liposomes have been intensively studied both in basic and applied research because of their promising potential as non-viral molecular vehicles. This work was aimed to gain more information on the interactions between the plasmamembrane and liposomes formed by a natural phospholipid and a cationic surfactant of the gemini family. The present work was conducted with the synergistic use of diverse experimental approaches: electro-rotation measurements, atomic force microscopy, ζ-potential measurements, laser scanning confocal microscopy and biomolecular/cellular techniques. Electro-rotation measurements pointed out that the interaction of cationic liposomes with the cell membrane alters significantly its dielectric and geometric parameters. This alteration, being accompanied by significant changes of the membrane surface roughness as measured by atomic force microscopy, suggests that the interaction with the liposomes causes locally substantial modifications to the structure and morphology of the cell membrane. However, the results of electrophoretic mobility (ζ-potential) experiments show that upon the interaction the electric charge exposed on the cell surface does not vary significantly, pointing out that the simple adhesion on the cell surface of the cationic liposomes or their fusion with the membrane is to be ruled out. As a matter of fact, confocal microscopy images directly demonstrated the penetration of the liposomes inside the cell and their diffusion within the cytoplasm. Electro-rotation experiments performed in the presence of endocytosis inhibitors suggest that the internalization is mediated by, at least, one specific pathway. Noteworthy, the liposome uptake by the cell does not cause a significant biological damage.
Collapse
Affiliation(s)
- E Stefanutti
- Dipartimento di Fisica, Sapienza Università di Roma, P.le Aldo Moro 2, 00185 Roma, Italy
| | - F Papacci
- Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, P.le Aldo Moro 5, 00185 Roma, Italy
| | - S Sennato
- Dipartimento di Fisica, Sapienza Università di Roma, P.le Aldo Moro 2, 00185 Roma, Italy; dCNR-IPCF, Sapienza Università di Roma, P.le Aldo Moro 2, 00185 Roma, Italy
| | - C Bombelli
- CNR, Istituto di Metodologie Chimiche and Dipartimento di Chimica Sapienza Università di Roma, P.le Aldo Moro 5, 00185 Roma, Italy
| | - I Viola
- National Nanotechnology Laboratory, Institute Nanoscience-CNR (NNL, CNR-NANO), I-73100 Lecce, Italy and c/o Dipartimento di Fisica, Sapienza Università di Roma, P.le Aldo Moro 2, 00185 Roma, Italy
| | - A Bonincontro
- Dipartimento di Fisica, Sapienza Università di Roma, P.le Aldo Moro 2, 00185 Roma, Italy
| | - F Bordi
- Dipartimento di Fisica, Sapienza Università di Roma, P.le Aldo Moro 2, 00185 Roma, Italy; dCNR-IPCF, Sapienza Università di Roma, P.le Aldo Moro 2, 00185 Roma, Italy; Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena, 291-00161 Roma, Italy
| | - G Mancini
- CNR, Istituto di Metodologie Chimiche and Dipartimento di Chimica Sapienza Università di Roma, P.le Aldo Moro 5, 00185 Roma, Italy
| | - G Gigli
- National Nanotechnology Laboratory, Institute Nanoscience-CNR (NNL, CNR-NANO), I-73100 Lecce, Italy and c/o Dipartimento di Fisica, Sapienza Università di Roma, P.le Aldo Moro 2, 00185 Roma, Italy; Università del Salento, Dip. di Matematica e Fisica Ennio de Giorgi and Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Lecce, Italy
| | - G Risuleo
- Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, P.le Aldo Moro 5, 00185 Roma, Italy.
| |
Collapse
|
85
|
Kong W, Liu R, Li H, Liu J, Huang H, Liu Y, Kang Z. High-bright fluorescent carbon dots and their application in selective nucleoli staining. J Mater Chem B 2014; 2:5077-5082. [DOI: 10.1039/c4tb00579a] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
86
|
Ishihara T, Nara S, Mizushima T. Interactions of Lecithinized Superoxide Dismutase with Serum Proteins and Cells. J Pharm Sci 2014; 103:1987-1994. [DOI: 10.1002/jps.24031] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 05/08/2014] [Accepted: 05/09/2014] [Indexed: 02/05/2023]
|
87
|
Ai X, Sun J, Zhong L, Wu C, Niu H, Xu T, Lian H, Han X, Ren G, Ding W, Wang J, Pu X, He Z. Star-Shape Redox-Responsive PEG-Sheddable Copolymer of Disulfide-Linked Polyethylene Glycol-Lysine-di-Tocopherol Succinate for Tumor-Triggering Intracellular Doxorubicin Rapid Release: Head-to-Head Comparison. Macromol Biosci 2014; 14:1415-28. [DOI: 10.1002/mabi.201400149] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 05/02/2014] [Indexed: 11/11/2022]
Affiliation(s)
- Xiaoyu Ai
- Department of Pharmaceutics, School of Pharmacy; Shenyang Pharmaceutical University; No. 103 Wenhua Road Shenyang 110016 P. R. China
| | - Jin Sun
- Department of Biopharmaceutics, School of Pharmacy; Shenyang Pharmaceutical University; No. 103 Wenhua Road Shenyang 110016 P. R. China
- Key Laboratory of Drug Delivery Technology and Pharmacokinetics; Tianjin Institute of Pharmaceutical Research; Tianjin P. R. China
| | - Lu Zhong
- Department of Pharmaceutics, School of Pharmacy; Shenyang Pharmaceutical University; No. 103 Wenhua Road Shenyang 110016 P. R. China
| | - Chunnuan Wu
- Department of Pharmaceutics, School of Pharmacy; Shenyang Pharmaceutical University; No. 103 Wenhua Road Shenyang 110016 P. R. China
| | - Handong Niu
- Department of Pharmaceutics, School of Pharmacy; Shenyang Pharmaceutical University; No. 103 Wenhua Road Shenyang 110016 P. R. China
| | - Tao Xu
- Department of Pharmaceutics, School of Pharmacy; Shenyang Pharmaceutical University; No. 103 Wenhua Road Shenyang 110016 P. R. China
| | - He Lian
- Department of Pharmaceutics, School of Pharmacy; Shenyang Pharmaceutical University; No. 103 Wenhua Road Shenyang 110016 P. R. China
| | - Xiaopeng Han
- Department of Pharmaceutics, School of Pharmacy; Shenyang Pharmaceutical University; No. 103 Wenhua Road Shenyang 110016 P. R. China
| | - Guolian Ren
- Department of Pharmaceutics, School of Pharmacy; Shenyang Pharmaceutical University; No. 103 Wenhua Road Shenyang 110016 P. R. China
| | - Wenya Ding
- Department of Pharmaceutics, School of Pharmacy; Shenyang Pharmaceutical University; No. 103 Wenhua Road Shenyang 110016 P. R. China
| | - Jia Wang
- Department of Pharmaceutics, School of Pharmacy; Shenyang Pharmaceutical University; No. 103 Wenhua Road Shenyang 110016 P. R. China
| | - Xiaohui Pu
- Pharmaceutical College of Henan University; Kaifeng 475004 P. R. China
| | - Zhonggui He
- Department of Pharmaceutics, School of Pharmacy; Shenyang Pharmaceutical University; No. 103 Wenhua Road Shenyang 110016 P. R. China
| |
Collapse
|
88
|
Sardan M, Kilinc M, Genc R, Tekinay AB, Guler MO. Cell penetrating peptide amphiphile integrated liposomal systems for enhanced delivery of anticancer drugs to tumor cells. Faraday Discuss 2014; 166:269-83. [PMID: 24611282 DOI: 10.1039/c3fd00058c] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Liposomes have been extensively used as effective nanocarriers, providing better solubility, higher stability and slower release of drugs compared to free drug administration. They are also preferred due to their nontoxic nature as well as their biodegradability and cell membrane mimicking abilities. In this study, we examined noncovalent integration of a cell penetrating arginine-rich peptide amphiphile into a liposomal formulation of negatively charged 1,2-dioleoyl-sn-glycero-3-[phospho-rac-(1-glycerol)] (DOPG) phospholipids in the presence of cholesterol due to its amphipathic character. We studied changes in the physical characteristics (size, surface potential and membrane polarity) of the liposomal membrane, as well as in the encapsulation of hydrophilic and hydrophobic agents due to peptide amphiphile incorporation. The activities of peptide integrated liposomal systems as drug delivery agents were investigated by using anticancer drugs, doxorubicin-HCI and paclitaxel. Enhancement in liposomal uptake due to arginine-rich peptide integration, and enhanced efficacy of the drugs were observed with peptide functionalized liposomes compared to free drugs.
Collapse
|
89
|
Gupta M, Chashoo G, Sharma PR, Saxena AK, Gupta PN, Agrawal GP, Vyas SP. Dual targeted polymeric nanoparticles based on tumor endothelium and tumor cells for enhanced antitumor drug delivery. Mol Pharm 2014; 11:697-715. [PMID: 24512060 DOI: 10.1021/mp400404p] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Some specific types of tumor cells and tumor endothelial cells represented CD13 proteins and act as receptors for Asn-Gly-Arg (NGR) motifs containing peptide. These CD13 receptors can be specifically recognized and bind through the specific sequence of cyclic NGR (cNGR) peptide and presented more affinity and specificity toward them. The cNGR peptide was conjugated to the poly(ethylene glycol) (PEG) terminal end in the poly(lactic-co-glycolic) acid PLGA-PEG block copolymer. Then, the ligand conjugated nanoparticles (cNGR-DNB-NPs) encapsulating docetaxel (DTX) were synthesized from preformed block copolymer by the emulsion/solvent evaporation method and characterized for different parameters. The various studies such as in vitro cytotoxicity, cell apoptosis, and cell cycle analysis presented the enhanced therapeutic potential of cNGR-DNB-NPs. The higher cellular uptake was also found in cNGR peptide anchored NPs into HUVEC and HT-1080 cells. However, free cNGR could inhibit receptor mediated intracellular uptake of NPs into both types of cells at 37 and 4 °C temperatures, revealing the involvement of receptor-mediated endocytosis. The in vivo biodistribution and antitumor efficacy studies indicated that targeted NPs have a higher therapeutic efficacy through targeting the tumor-specific site. Therefore, the study exhibited that cNGR-functionalized PEG-PLGA-NPs could be a promising approach for therapeutic applications to efficient antitumor drug delivery.
Collapse
Affiliation(s)
- Madhu Gupta
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S. Gour Vishwavidyalaya , Sagar-470003, M.P., India
| | | | | | | | | | | | | |
Collapse
|
90
|
Deno S, Takemoto N, Iwata H. Introduction of antioxidant-loaded liposomes into endothelial cell surfaces through DNA hybridization. Bioorg Med Chem 2014; 22:350-7. [DOI: 10.1016/j.bmc.2013.11.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 11/06/2013] [Accepted: 11/08/2013] [Indexed: 11/29/2022]
|
91
|
Zhang F, Lin G, Shao W, Yu Y, Zang L. Pharmacokinetics and Tissue Distribution of Vinorelbine Bitartrate after Intraveous Administration of Liposomal and Injectable Formulations. J DISPER SCI TECHNOL 2013. [DOI: 10.1080/01932691.2012.744691] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
92
|
Erdoğ A, Putra Limasale YD, Keskin D, Tezcaner A, Banerjee S. In Vitro Characterization of a Liposomal Formulation of Celecoxib Containing 1,2-Distearoyl-sn-Glycero-3-Phosphocholine, Cholesterol, and Polyethylene Glycol and its Functional Effects Against Colorectal Cancer Cell Lines. J Pharm Sci 2013; 102:3666-77. [DOI: 10.1002/jps.23674] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 07/01/2013] [Accepted: 07/03/2013] [Indexed: 12/19/2022]
|
93
|
Intracellular delivery of polymeric nanocarriers: a matter of size, shape, charge, elasticity and surface composition. Ther Deliv 2013; 4:705-23. [PMID: 23738668 DOI: 10.4155/tde.13.37] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent progress in drug discovery has enabled the targeting of specific intracellular molecules to achieve therapeutic effects. These next-generation therapeutics are often biologics that cannot enter cells by mere diffusion. Therefore, it is imperative that drug carriers are efficiently internalized by cells and reach specific target organelles before releasing their cargo. Nanoscale polymeric carriers are particularly suitable for such intracellular delivery. Although size and surface charge have been the most studied parameters for nanocarriers, it is now well appreciated that other properties, for example, particle shape, elasticity and surface composition, also play a critical role in their transport across physiological barriers. It is proposed that a multivariate design space that considers the interdependence of particle geometry with its mechanical and surface properties must be optimized to formulate drug nanocarriers for effective accumulation at target sites and efficient intracellular delivery.
Collapse
|
94
|
Luther EM, Petters C, Bulcke F, Kaltz A, Thiel K, Bickmeyer U, Dringen R. Endocytotic uptake of iron oxide nanoparticles by cultured brain microglial cells. Acta Biomater 2013; 9:8454-65. [PMID: 23727247 DOI: 10.1016/j.actbio.2013.05.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2012] [Revised: 05/17/2013] [Accepted: 05/21/2013] [Indexed: 11/29/2022]
Abstract
Microglia are the phagocytotic cells of the brain that respond rapidly to alterations in brain homeostasis. Since iron oxide nanoparticles (IONPs) are used for diagnostic and therapeutic applications in the brain, the consequences of an exposure of microglial cells to IONPs are of particular interest. To address this topic we have synthesized and characterized fluorescent BODIPY®-labelled IONPs (BP-IONPs). The average hydrodynamic diameter and the ζ-potential of BP-IONPs in water were ∼65 nm and -49 mV, respectively. Both values increased after dispersion of the particles in serum containing incubation medium to ∼130 nm and -8 mV. Exposure of cultured rat microglial cells with BP-IONPs caused a time-, concentration- and temperature-dependent uptake of the particles, as demonstrated by strong increases in cellular iron contents and cellular fluorescence. Incubation for 3h with 150 and 450 μM iron as BP-IONPs increased the cellular iron content from a low basal level of ∼50 nmol iron mg(-1) to 219±52 and 481±28 nmol iron (mg protein)(-1), respectively. These conditions did not affect cell viability, but exposure to higher concentrations of BP-IONPs or for longer incubation periods severely compromised cell viability. The BP-IONP fluorescence in viable microglial cells was co-localized with lysosomes. In addition, BP-IONP accumulation was lowered by 60% in the presence of the endocytosis inhibitors 5-(N-ethyl-N-isopropyl)amiloride, tyrphostin23 and chlorpromazin. These results suggest that the rapid accumulation of BP-IONPs by microglial cells is predominantly mediated by macropinocytosis and clathrin-mediated endocytosis, which direct the accumulated particles into the lysosomal compartment.
Collapse
Affiliation(s)
- Eva M Luther
- Center for Biomolecular Interactions Bremen, University of Bremen, P.O. Box 330440, D-28334 Bremen, Germany
| | | | | | | | | | | | | |
Collapse
|
95
|
Chen Y, Yang L, Huang S, Li Z, Zhang L, He J, Xu Z, Liu L, Cao Y, Sun L. Delivery system for DNAzymes using arginine-modified hydroxyapatite nanoparticles for therapeutic application in a nasopharyngeal carcinoma model. Int J Nanomedicine 2013; 8:3107-3118. [PMID: 23983464 PMCID: PMC3747848 DOI: 10.2147/ijn.s48321] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
DNAzymes are synthetic, single-stranded, catalytic nucleic acids that bind and cleave target mRNA in a sequence-specific manner, and have been explored for genotherapeutics. One bottleneck restricting their application is the lack of an efficient delivery system. As an inorganic nanomaterial with potentially wide application, nano-hydroxyapatite particles (nHAP) have attracted increasing attention as new candidates for nonviral vectors. In this study, we developed an nHAP-based delivery system and explored its cellular uptake mechanisms, intracellular localization, and biological effects. Absorption of arginine-modified nanohydroxyapatite particles (Arg-nHAP) and DZ1 (latent membrane protein 1 [LMP1]-targeted) reached nearly 100% efficiency under in vitro conditions. Using specific inhibitors, cellular uptake of the Arg-nHAP/DZ1 complex was shown to be mediated by the energy-dependent endocytosis pathway. Further, effective intracellular delivery and nuclear localization of the complex was confirmed by confocal microscopy. Biologically, the complex successfully downregulated the expression of LMP1 in nasopharyngeal carcinoma cells. In a mouse tumor xenograft model, the complex was shown to be delivered efficiently to tumor tissue, downregulating expression of LMP1 and suppressing tumor growth. These results suggest that Arg-nHAP may be an efficient vector for nucleic acid-based drugs with potential clinical application.
Collapse
Affiliation(s)
- Yan Chen
- Center for Molecular Medicine, Xiangya hospital, Changsha, People’s Republic of China
| | - Lifang Yang
- Center for Molecular Medicine, Xiangya hospital, Changsha, People’s Republic of China
- Cancer Research Institute, Changsha, People’s Republic of China
| | - Suping Huang
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, People’s Republic of China
| | - Zhi Li
- Center for Molecular Medicine, Xiangya hospital, Changsha, People’s Republic of China
| | - Lu Zhang
- Center for Molecular Medicine, Xiangya hospital, Changsha, People’s Republic of China
| | - Jiang He
- Center for Molecular Medicine, Xiangya hospital, Changsha, People’s Republic of China
| | - Zhijie Xu
- Cancer Research Institute, Changsha, People’s Republic of China
| | - Liyu Liu
- Cancer Research Institute, Changsha, People’s Republic of China
| | - Ya Cao
- Cancer Research Institute, Changsha, People’s Republic of China
| | - Lunquan Sun
- Center for Molecular Medicine, Xiangya hospital, Changsha, People’s Republic of China
| |
Collapse
|
96
|
Zidan AS, Spinks CB, Habib MJ, Khan MA. Formulation and transport properties of tenofovir loaded liposomes through Caco-2 cell model. J Liposome Res 2013; 23:318-26. [DOI: 10.3109/08982104.2013.810645] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
97
|
Charfi C, Levros LC, Edouard E, Rassart E. Characterization and identification of PARM-1 as a new potential oncogene. Mol Cancer 2013; 12:84. [PMID: 23902727 PMCID: PMC3750824 DOI: 10.1186/1476-4598-12-84] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 07/25/2013] [Indexed: 11/21/2022] Open
Abstract
Background The Graffi murine retrovirus is a powerful tool to find leukemia associated oncogenes. Using DNA microarrays, we recently identified several genes specifically deregulated in T- and B-leukemias induced by this virus. Results In the present study, probsets associated with T-CD8+ leukemias were analyzed and we validated the expression profile of the Parm-1 gene. PARM-1 is a member of the mucin family. We showed that human PARM-1 is an intact secreted protein accumulating predominantly, such as murine PARM-1, at the Golgi and in the early and late endosomes. PARM-1 colocalization with α-tubulin suggests that its trafficking within the cell involves the microtubule cytoskeleton. Also, the protein co-localizes with caveolin-1 which probably mediates its internalization. Transient transfection of both mouse and human Parm-1 cDNAs conferred anchorage- and serum-independent growth and enhanced cell proliferation. Moreover, deletion mutants of human PARM-1 without either extracellular or cytoplasmic portions seem to retain the ability to induce anchorage-independent growth of NIH/3T3 cells. In addition, PARM-1 increases ERK1/2, but more importantly AKT and STAT3 phosphorylation. Conclusions Our results strongly suggest the oncogenic potential of PARM-1.
Collapse
Affiliation(s)
- Cyndia Charfi
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Centre BioMed, Université du Québec à Montréal, Case Postale 8888, Succursale Centre-ville, Montréal, QC H3C-3P8, Canada
| | | | | | | |
Collapse
|
98
|
Yang F, Huang W, Li Y, Liu S, Jin M, Wang Y, Jia L, Gao Z. Anti-tumor effects in mice induced by survivin-targeted siRNA delivered through polysaccharide nanoparticles. Biomaterials 2013; 34:5689-99. [DOI: 10.1016/j.biomaterials.2013.03.047] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 03/15/2013] [Indexed: 12/20/2022]
|
99
|
Zheng C, Guo Q, Wu Z, Sun L, Zhang Z, Li C, Zhang X. Amphiphilic glycopolymer nanoparticles as vehicles for nasal delivery of peptides and proteins. Eur J Pharm Sci 2013; 49:474-82. [DOI: 10.1016/j.ejps.2013.04.027] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 03/28/2013] [Accepted: 04/24/2013] [Indexed: 11/25/2022]
|
100
|
Ma Y, Wang Z, Zhao W, Lu T, Wang R, Mei Q, Chen T. Enhanced bactericidal potency of nanoliposomes by modification of the fusion activity between liposomes and bacterium. Int J Nanomedicine 2013; 8:2351-60. [PMID: 23847417 PMCID: PMC3700910 DOI: 10.2147/ijn.s42617] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Pseudomonas aeruginosa represents a good model of antibiotic resistance. These organisms have an outer membrane with a low level of permeability to drugs that is often combined with multidrug efflux pumps, enzymatic inactivation of the drug, or alteration of its molecular target. The acute and growing problem of antibiotic resistance of Pseudomonas to conventional antibiotics made it imperative to develop new liposome formulations to overcome these mechanisms, and investigate the fusion between liposome and bacterium. Methods The rigidity, stability and charge properties of phospholipid vesicles were modified by varying the cholesterol, 1,2-dioleoyl-sn-glycero-3-phosphatidylethanolamine (DOPE), and negatively charged lipids 1,2-dimyristoyl-sn-glycero-3-phosphoglycerol sodium salt (DMPG), 1,2-dimyristoyl-sn-glycero-3-phopho-L-serine sodium salt (DMPS), 1,2-dimyristoyl-sn-glycero-3-phosphate monosodium salt (DMPA), nature phosphatidylserine sodium salt from brain and nature phosphatidylinositol sodium salt from soybean concentrations in liposomes. Liposomal fusion with intact bacteria was monitored using a lipid-mixing assay. Results It was discovered that the fluid liposomes-bacterium fusion is not dependent on liposomal size and lamellarity. A similar degree of fusion was observed for liposomes with a particle size from 100 to 800 nm. The fluidity of liposomes is an essential pre-request for liposomes fusion with bacteria. Fusion was almost completely inhibited by incorporation of cholesterol into fluid liposomes. The increase in the amount of negative charges in fluid liposomes reduces fluid liposomes-bacteria fusion when tested without calcium cations due to electric repulsion, but addition of calcium cations brings the fusion level of fluid liposomes to similar or higher levels. Among the negative phospholipids examined, DMPA gave the highest degree of fusion, DMPS and DMPG had intermediate fusion levels, and PI resulted in the lowest degree of fusion. Furthermore, the fluid liposomal encapsulated tobramycin was prepared, and the bactericidal effect occurred more quickly when bacteria were cultured with liposomal encapsulated tobramycin. Conclusion The bactericidal potency of fluid liposomes is dramatically enhanced with respect to fusion ability when the fusogenic lipid, DOPE, is included. Regardless of changes in liposome composition, fluid liposomes-bacterium fusion is universally enhanced by calcium ions. The information obtained in this study will increase our understanding of fluid liposomal action mechanisms, and help in optimizing the new generation of fluid liposomal formulations for the treatment of pulmonary bacterial infections.
Collapse
Affiliation(s)
- Yufan Ma
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|