Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Control Release ; 257: 32-39, 2017 07 10.
Article in English | MEDLINE | ID: mdl-28042083

ABSTRACT

Targeted delivery to the cell nucleus can enhance the efficiency of drugs with nuclear site of action (some anti-cancer agents, DNA drugs, etc.), and can reduce their toxicity. Such targeting can be attained using nano-drug delivery systems (nano-DDSs) decorated with nuclear targeting sequences (such as nuclear localization sequence peptides, NLS). Several types of nano-DDSs decorated with NLS peptides were designed, but their investigation usually did not include quantitate analysis of the decoration efficiency and its correlation with the nano-DDSs intracellular localization. Thus, the major mechanisms and limiting factors of the nano-DDSs nuclear targeting are largely unknown yet. In this study, we report quantitative data for specific nano-formulation (CdSe-ZnS quantum dots) that include the efficiencies of its decoration with NLS residues and of its nuclear and perinuclear targeting, and demonstrate correlation between these parameters. For instance, QDs decorated with 83, 246, and 265 NLS peptides accumulated efficiently in the nucleus of HeLa cells or its vicinity (an average of 30.4%, 43.3%, and 49.0% of the intracellular QDs, respectively). On the other hand, QDs decorated with 63, 231, and 308 scrambled peptides accumulated in the nucleus of HeLa cells or its vicinity to a much lower extent (an average of 17.3%, 21.1%, and 25.5% of the intracellular QDs, respectively). Thus, results of our study provide important insights into the structure-activity correlations (i.e., the relationships between the formulation properties and the intracellular fate of nano-DDSs) of nuclear-targeted drug delivery. We plan to apply the research tools that were developed in the course of this and our previous studies to investigate the nuclear and perinuclear targeting activities of different NLS sequences, and to investigate the effects of nano-DDSs size, charge, shape, decoration efficiency with nuclear targeting sequences, and other structural factors on nuclear and perinuclear targeting efficiency.


Subject(s)
Cell Nucleus/metabolism , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Nuclear Localization Signals/chemistry , Nuclear Localization Signals/pharmacokinetics , Quantum Dots/chemistry , Drug Delivery Systems , Endocytosis , HeLa Cells , Humans
2.
Int J Radiat Biol ; 92(11): 707-715, 2016 11.
Article in English | MEDLINE | ID: mdl-27010622

ABSTRACT

PURPOSE: We investigated the effect of incorporation of a nuclear localization signal (NLS) into a conjugate comprising the DNA binding ligand para-iodoHoechst (PIH) and octreotate on its DNA binding and affinity to the somatostatin receptor (SSTR). Confirmation of these properties would support development of similar conjugates labelled with Auger emitters for their potential in Auger endoradiotherapy. MATERIALS AND METHODS: We synthesized conjugates of PIH and octreotate (PO) or PIH and NLS (PN) and a conjugate comprising PIH, NLS and octreotate (PNO). DNA-binding characteristics of PIH and conjugates were assessed using synthetic DNA oligonucleotides employing spectrophotometric titration of ligand solutions with DNA. We used membranes from the type 2 SSTR (SSTR2) overexpressing human non-small cell lung cancer cell line A427-7 to investigate the binding affinity of PNO. RESULTS: We demonstrated PN and PNO retain specific high affinity DNA-binding properties observed for PIH, and acquire an additional non-specific binding capacity. No DNA binding was observed for PO. PNO retains its binding affinity for SSTR. CONCLUSIONS: The DNA-binding properties of PNO and its affinity for SSTR suggests that it could potentially be used for tumour-specific delivery of PIH labelled with an Auger emitter in SSTR expressing tumours.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , DNA, Neoplasm/metabolism , Iodine Radioisotopes/pharmacokinetics , Nuclear Localization Signals/pharmacokinetics , Peptides, Cyclic/pharmacokinetics , Receptors, Somatostatin/metabolism , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Line, Tumor , Electrons/therapeutic use , Humans , Iodine Radioisotopes/therapeutic use , Isotope Labeling/methods , Nuclear Localization Signals/chemistry , Subcellular Fractions/metabolism
3.
Nanoscale ; 7(14): 6104-13, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25773263

ABSTRACT

Strong blue fluorescent polyethylene glycol (PEG) anchored carbon nitride dots (CDs@PEG) with a high quantum yield (QY) of 75.8% have been synthesized by a one step hydrothermal treatment. CDs with a diameter of ca. 6 nm are well dispersed in water and present a graphite-like structure. Photoluminescence (PL) studies reveal that CDs display excitation-dependent behavior and are stable under various test conditions. Based on the as-prepared CDs, we designed novel cell nucleus targeting imaging carbon dots functionalized with a nuclear localization signal (NLS) peptide. The favourable biocompatibilities of CDs and NLS modified CDs (NLS-CDs) are confirmed by in vitro cytotoxicity assays. Importantly, intracellular localization experiments in MCF7 and A549 cells demonstrate that NLS-CDs could be internalized in the nucleus and show blue light, which indicates that CDs may serve as cell nucleus imaging probes.


Subject(s)
Cell Nucleus/metabolism , Materials Testing , Nitriles , Nuclear Localization Signals , Polyethylene Glycols , Quantum Dots/chemistry , Humans , MCF-7 Cells , Molecular Probes/chemistry , Molecular Probes/pharmacokinetics , Molecular Probes/pharmacology , Nitriles/chemistry , Nitriles/pharmacokinetics , Nitriles/pharmacology , Nuclear Localization Signals/chemistry , Nuclear Localization Signals/pharmacokinetics , Nuclear Localization Signals/pharmacology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/pharmacology
4.
Int J Nanomedicine ; 7: 5079-90, 2012.
Article in English | MEDLINE | ID: mdl-23049255

ABSTRACT

BACKGROUND: Supramolecular micelles as drug-delivery vehicles are generally unable to enter the nucleus of nondividing cells. In the work reported here, nuclear localization signal (NLS)-modified polymeric micelles were studied with the aim of improving nuclear drug delivery. METHODS: In this research, cholesterol-modified glycol chitosan (CHGC) was synthesized. NLS-conjugated CHGC (NCHGC) was synthesized and characterized using proton nuclear magnetic resonance spectroscopy, dynamic light scattering, and fluorescence spectroscopy. Doxorubicin (DOX), an anticancer drug with an intracellular site of action in the nucleus, was chosen as a model drug. DOX-loaded micelles were prepared by an emulsion/solvent evaporation method. The cellular uptake of different DOX formulations was analyzed by flow cytometry and confocal laser scanning microscopy. The cytotoxicity of blank micelles, free DOX, and DOX-loaded micelles in vitro was investigated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay in HeLa and HepG2 cells. RESULTS: The degree of substitution was 5.9 cholesterol and 3.8 NLS groups per 100 sugar residues of the NCHGC conjugate. The critical aggregation concentration of the NCHGC micelles in aqueous solution was 0.0209 mg/mL. The DOX-loaded NCHGC (DNCHGC) micelles were observed as being almost spherical in shape under transmission electron microscopy, and the size was determined as 248 nm by dynamic light scattering. The DOX-loading content of the DNCHGC micelles was 10.1%. The DOX-loaded micelles showed slow drug-release behavior within 72 hours in vitro. The DNCHGC micelles exhibited greater cellular uptake and higher amounts of DOX in the nuclei of HeLa cells than free DOX and DOX-loaded CHGC (DCHGC) micelles. The half maximal inhibitory concentration (IC(50)) values of free DOX, DCHGC, and DNCHGC micelles against HepG2 cells were 4.063, 0.591, and 0.171 µg/mL, respectively. Moreover, the IC(50) values of free DOX (3.210 µg/mL) and the DCHGC micelles (1.413 µg/mL) against HeLa cells were nearly 6.96- and 3.07-fold (P < 0.01), respectively, higher than the IC(50) value of the DNCHGC micelles (0.461 µg/mL). CONCLUSION: The results of this study suggest that novel NCHGC micelles could be a potential carrier for nucleus-targeting delivery.


Subject(s)
Cell Nucleus/chemistry , Cell Nucleus/metabolism , Chitosan/chemistry , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Nanocapsules/chemistry , Nuclear Localization Signals/pharmacokinetics , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/chemistry , Diffusion , HeLa Cells , Hep G2 Cells , Humans , Micelles , Nanocapsules/administration & dosage , Nuclear Localization Signals/chemistry , Polyethylene Glycols/chemistry
5.
Bioconjug Chem ; 22(8): 1567-75, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21688833

ABSTRACT

The nonviral vector with iodine-nuclear localization sequence (namely, NLS-I) targeting breast cancer cells was fabricated. Ternary complexes were formed via charge interactions among NLS-I peptides, PEI 1800, and DNA, and we investigated their cellular internalization, nuclear accumulation as well as transfection efficiency. All the experiments were assessed by employing MCF-7 cells that express sodium/iodide symporter and HeLa cells that lack the expression of the symporter. In MCF-7 cells, cell internalization and nuclear accumulation of NLS-I was markedly increased compared to that in NLS. In addition, compared to that of the PEI1800/DNA complex, PEI1800/DNA/NLS-I complexes exhibited much enhanced luciferase reporter gene expression by up to 130-fold. By contrast, in HeLa cells, the evident improvements of cellular internalization, nuclear accumulation, and transfection efficiency by NLS-I were not observed. This study demonstrates an alternative method to construct a nonviral delivery system for targeted gene transfer into breast cancer cells.


Subject(s)
Active Transport, Cell Nucleus , Breast Neoplasms/drug therapy , Genetic Therapy/methods , Iodine/metabolism , Nuclear Localization Signals/pharmacokinetics , Transfection/methods , Breast Neoplasms/pathology , Cell Line, Tumor , DNA/pharmacokinetics , DNA/therapeutic use , Female , Humans , Iodine/pharmacokinetics , Iodine/therapeutic use , Nuclear Localization Signals/metabolism , Nuclear Localization Signals/therapeutic use , Polyethyleneimine/pharmacokinetics , Polyethyleneimine/therapeutic use , Symporters/metabolism
6.
J Nucl Med ; 51(7): 1084-91, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20554744

ABSTRACT

UNLABELLED: (111)In-nuclear localization sequence-trastuzumab is a radioimmunotherapeutic agent consisting of trastuzumab modified with NLS peptides (CGYGPKKKRKVGG) and labeled with the Auger electron emitter (111)In. Our objectives were to evaluate the tumor growth-inhibitory properties and normal-tissue toxicity of (111)In-NLS-trastuzumab in mice after intraperitoneal administration. METHODS: The pharmacokinetics of (111)In-NLS-trastuzumab after intravenous (tail vein) or intraperitoneal injection in BALB/c mice were compared. Normal-tissue toxicity was determined in BALB/c mice at 2 wk after intraperitoneal injection of 3.7-18.5 MBq (4 mg/kg) of (111)In-NLS-trastuzumab by monitoring body weight, histopathologic examination of tissues, and hematology (white blood cell, platelet, red blood cell, and hemoglobin) or clinical biochemistry (alanine transaminase and creatinine) parameters. A no-observable-adverse-effect-level (NOAEL) dose was defined. Athymic mice bearing subcutaneous MDA-MB-361 or MDA-MB-231 human breast cancer xenografts (5.0 x 10(5) or 0.5 x 10(5) HER2/cell, respectively) were treated with a single NOAEL dose or 2 doses administered intraperitoneally and separated by 2 wk. Control groups were administered (111)In-trastuzumab, trastuzumab, nonspecific (111)In-NLS-human IgG (hIgG), or normal saline. RESULTS: The bioavailability of (111)In-NLS-trastuzumab after intraperitoneal injection was 0.7. The NOAEL dose was 9.25 MBq (4 mg/kg); doses greater than or equal to 18.5 MBq decreased white blood cell or platelet counts, and doses of 27.7 MBq decreased red blood cell counts. There was no increase in alanine transaminase or creatinine at any doses tested. There were no morphologic changes to the liver, kidneys, heart, or spleen or loss of body weight. A single dose of (111)In-NLS-trastuzumab (9.25 MBq)-compared with mice receiving (111)In-trastuzumab, trastuzumab, (111)In-NLS-hIgG, or normal saline-significantly slowed the rate of growth of MDA-MB-361 tumors over 60 d (0.014 d(-1) vs. 0.033 d(-1), 0.046 d(-1), 0.030 d(-1), and 0.061 d(-1), respectively; P < 0.05). (111)In-NLS-trastuzumab had no effect on the growth of MDA-MB-231 tumors. Two doses of (111)In-NLS-trastuzumab (9.25 MBq; 4 mg/kg) separated by 2 wk increased the survival of mice with MDA-MB-361 tumors, compared with mice treated with trastuzumab or normal saline (>140 d vs. 96 and 84 d, respectively; P < 0.001 or 0.027, respectively). CONCLUSION: (111)In-NLS-trastuzumab is a promising radioimmunotherapeutic agent that could be effective for treatment of HER2-overexpressing breast cancer in humans.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/toxicity , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Breast Neoplasms/radiotherapy , Nuclear Localization Signals/pharmacology , Nuclear Localization Signals/toxicity , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/toxicity , Receptor, ErbB-2/metabolism , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/metabolism , Female , Humans , Immunoglobulin G/chemistry , Indium Radioisotopes/pharmacokinetics , Injections, Intraperitoneal , Injections, Intravenous , Isotope Labeling , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Nuclear Localization Signals/pharmacokinetics , Radioimmunotherapy , Survival Analysis , Trastuzumab
7.
J Mater Sci Mater Med ; 20(1): 347-50, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18807262

ABSTRACT

Nanoparticles have been extensively used for a variety of biomedical applications and there is a growing need for highly specific and efficient delivery of the nanoparticles into target cells and subcellular location. We attempted to accomplish this goal by modifying gold particles with peptide motif's that are known to deliver a 'cargo' into chosen cellular location specifically, we intended to deliver nanogold particles into cells through chemical cross-linking with different peptides known to carry protein into cells. Our results suggest that specific sequence of such 'carrier peptides' can efficiently deliver gold nanoparticles into cells when chemically cross-linked with the metal particles.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/pharmacokinetics , Gold/chemistry , Gold/pharmacokinetics , Metal Nanoparticles/chemistry , Osteosarcoma/metabolism , Peptides/chemistry , Peptides/pharmacokinetics , Amino Acid Sequence , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacokinetics , Biological Transport, Active , Cell Line, Tumor , Cross-Linking Reagents , Drug Delivery Systems , Humans , Materials Testing , Metal Nanoparticles/ultrastructure , Microscopy, Electron, Transmission , Molecular Sequence Data , Nuclear Localization Signals/chemistry , Nuclear Localization Signals/pharmacokinetics , Osteosarcoma/ultrastructure , Subcellular Fractions/metabolism , Subcellular Fractions/ultrastructure
8.
Bioconjug Chem ; 16(3): 582-7, 2005.
Article in English | MEDLINE | ID: mdl-15898725

ABSTRACT

A trifunctional bioconjugate consisting of the SV40 nuclear localization signal (NLS) peptide, an aliphatic triamine ligand, and the DNA intercalating pyrene has been synthesized and quantitatively labeled with [(99m)Tc(OH(2))(3)(CO)(3)](+). The radiotoxicity of the resulting nucleus-targeting radiopharmaceutical on B16F1 mouse melanoma cells has been investigated to evaluate the activity of Auger and Coster-Kronig electrons on the viability of cells. We found a dose-dependent significant radiotoxicity of the nucleus-targeting radiopharmaceutical clearly related to the low energy decay of (99m)Tc. These principal results imply a possible therapeutic strategy based on the use of the low-energy Auger electron-emitting (99m)Tc radionuclide attached to nucleus-targeting molecules and comprising an intercalator. Highly efficient DNA targeting vectors could complement the usual role of (99m)Tc in diagnostic applications. The Auger electrons emitted by the (99m)Tc nuclide induce DNA damage leading ultimately, through a mitotic catastrophe pathway, to necrotic cell death. Non-DNA-targeting (99m)Tc complexes display much lower radiotoxicity.


Subject(s)
Intercalating Agents/pharmacokinetics , Intercalating Agents/toxicity , Nuclear Localization Signals/chemistry , Nuclear Localization Signals/pharmacokinetics , Organotechnetium Compounds/toxicity , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Dose-Response Relationship, Radiation , Intercalating Agents/chemistry , Mice , Molecular Structure , Organotechnetium Compounds/chemistry , Organotechnetium Compounds/pharmacokinetics
9.
Chem Biol ; 9(8): 943-8, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12204694

ABSTRACT

The cellular delivery of therapeutic agents and their localization within cells is currently a great challenge in medicinal chemistry. A few cationic peptides have shown a strong propensity to cross the cytoplasmic membrane and enter cells. Nuclear localization signal (NLS) sequences are a class of highly cationic peptides that may be exploited for cellular import of linked cargo. A series of NLS sequence peptides were investigated for entry into different cancer cell lines by flow cytometry and confocal microscopy. All NLS peptides demonstrated rapid accumulation within cells when added to the cellular media. Covalent adducts of proteins and oligonucleotides with NLS peptides were also effectively imported within cells. An understanding of the structural and mechanistic properties of these sequences will provide great potential for the rational design of efficient and selective peptidic delivery systems.


Subject(s)
Drug Delivery Systems/methods , Nuclear Localization Signals/pharmacokinetics , Cell Compartmentation , Flow Cytometry , Fluorescent Dyes , Humans , Microscopy, Confocal , Nuclear Localization Signals/chemistry , Oligonucleotides/chemistry , Oligonucleotides/pharmacokinetics , Transcription Factors/chemistry , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...