Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Rep ; 12(1): 10550, 2022 06 22.
Article in English | MEDLINE | ID: mdl-35732805

ABSTRACT

Carbon dots (CDs) are categorized as an emerging class of zero-dimension nanomaterials having high biocompatibility, photoluminescence, tunable surface, and hydrophilic property. CDs, therefore, are currently of interest for bio-imaging and nano-medicine applications. In this work, polyethylene glycol functionalized CDs (CD-PEG) were prepared from oil palm empty fruit bunch by a one-pot hydrothermal technique. PEG was chosen as a passivating agent for the enhancement of functionality and photoluminescence properties of CDs. To prepare the CDs-PEG, the effects of temperature, time, and concentration of PEG were investigated on the properties of CDs. The as-prepared CDs-PEG were characterized by several techniques including dynamic light scattering, high-resolution transmission electron microscopy, X-ray photoelectron spectroscopy, fluorescence spectroscopy, Raman spectroscopy, Fourier-transform infrared spectroscopy and Thermogravimetric analysis. The as-prepared CDs under hydrothermal condition at 220 °C for 6 h had spherical morphology with an average diameter of 4.47 nm. Upon modification, CDs-PEG were photo-responsive with excellent photoluminescence property. The CDs-PEG was subsequently used as a drug carrier for doxorubicin [DOX] delivery to CaCo-2, colon cancer cells in vitro. DOX was successfully loaded onto CDs-PEG surface confirmed by FT-IR and Matrix-Assisted Laser Desorption/Ionization Time-of-Flight Mass Spectrometer (MALDI-TOF/MS) patterns. The selective treatment of CDs-PEG-DOX against the colorectal cancer cells, , relative to normal human fibroblast cells was succesfully demonstrated.


Subject(s)
Colonic Neoplasms , Quantum Dots , Caco-2 Cells , Carbon/chemistry , Colonic Neoplasms/drug therapy , Doxorubicin , Fluorescent Dyes/chemistry , Humans , Polyethylene Glycols/chemistry , Quantum Dots/chemistry , Spectroscopy, Fourier Transform Infrared , Theranostic Nanomedicine
2.
Carbohydr Polym ; 277: 118858, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34893265

ABSTRACT

We report herein the development of the novel nanohybrids of gold nanoparticles reduced/stabilized/coated with collagen (AuNPs@collagen) in the first layer and subsequently modified with biotin-quat188-chitosan (Bi-QCS) in the outer layer for 5-fluorouracil (5-FU) delivery to improve cellular uptake and promote specific cell targeting of the nanocarrier. The fabrication of the layer-by-layer technique on the surface of gold nanoparticles (AuNPs) can overcome the limitation of poor drug loading capacity of the classic AuNPs from 64.67% to 87.46%. The AuNPs@collagen coated by the Bi-QCS exhibits strong electrostatic interactions between drug anion (5-FU) and amine groups of the modified chitosan as well as hydrogen bonding. Furthermore, the Bi-QCS-AuNPs@collagen demonstrated a significantly higher anti-inflammatory activity in RAW264.7 macrophage cell line. The Bi-QCS-AuNPs@collagen enhanced the activity of 5-FU approximately 3.3-fold (HeLa) and 6.2-fold (A549), compared to the free 5-Fluorouracil. According to these results, it is very promising that Bi-QCS-AuNPs@collagen can be used as an effective drug delivery carrier in the future.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Drug Delivery Systems , Fluorouracil/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Antimetabolites, Antineoplastic/chemical synthesis , Antimetabolites, Antineoplastic/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Chitosan/chemistry , Collagen/chemistry , Drug Liberation , Drug Screening Assays, Antitumor , Fluorouracil/chemical synthesis , Fluorouracil/chemistry , Gold/chemistry , Humans , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Metal Nanoparticles/chemistry , Mice , Molecular Structure , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Particle Size , RAW 264.7 Cells
3.
Nanotechnology ; 31(43): 435101, 2020 Oct 23.
Article in English | MEDLINE | ID: mdl-32647102

ABSTRACT

Bone morphogenic protein-2 (BMP-2) knuckle epitope peptide has been recently discovered and known to activate chondrogenesis. However, the applications of this soluble peptide remain very limited due to rapid diffusion resulting in poor cellular uptake into target cells. We herein designed nanoparticles made from hyaluronic acid functionalized gold nanorods (GNRs) to conjugate with thiolated BMP-2 knuckle epitope peptide via a two-step reaction. Hyaluronic acid was modified to have thiol functional groups to replace the cetyl trimethylammonium bromide ligands on the surface of GNRs. The thiolated peptides were subsequently reacted with hyaluronic acid on the surface on GNRs via a maleimide-hydrazide crosslinker. The conjugation was confirmed by the change of surface charge of GNRs and the plasmon shift. A colorimetric peptide assay suggested more than 69% of the thiolated peptides were conjugated with the hyaluronic acid coated gold nanorods. Moreover, in vitro cell viability showed that BMP-2 conjugated hyaluronic acid functionalized gold nanorods (B2HGR) were cytocompatible and did not cause cytotoxicity to fibroblast cells. The B2HGRs also significantly promote cellular uptake of the BMP-2 peptides in both human mesenchymal stem cells and porcine chondrocytes due to multivalent ligand binding to the BMP receptors on the cell surface resulting in receptor-mediated endocytosis. The enhanced cellular uptake was clearly observed under a confocal microscope resulting in the significant activation of type II collagen gene expression and glucosaminoglycan secretion in those cells. Furthermore, our delivery system is a proof-of-concept of using scaffolds in combination with nanodelivery platform to enhance cartilaginous repair. The peptide loading capacity and the release is not limited by the scaffolds. Therefore, our delivery platform has potential applications for cartilage regeneration in a preclinical and clinical setting in the future.


Subject(s)
Bone Morphogenetic Protein 2/administration & dosage , Chondrogenesis/drug effects , Drug Carriers/chemistry , Hyaluronic Acid/chemistry , Nanotubes/chemistry , Peptides/administration & dosage , Animals , Bone Morphogenetic Protein 2/pharmacokinetics , Bone Morphogenetic Protein 2/pharmacology , Cell Line , Gold , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Peptides/pharmacokinetics , Peptides/pharmacology , Swine
4.
Int J Biol Macromol ; 162: 561-577, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-32553955

ABSTRACT

This study aimed to develop new organic/inorganic nanohybrids of targeted pullulan derivative/gold nanoparticles (FA-PABA-Q188-PUL@AuNPs) to improve the selectivity and efficacy of drugs. The chemical structure of targeted pullulan derivative, folic acid-decorated para-aminobenzoic acid-quat188-pullulan (FA-PABA-Q188-PUL), was designed for reducing, stabilizing, capping, and functionalizing AuNPs. Here, the key factors, including pH, temperature, and FA-PABA-Q188-PUL concentrations, were systematically optimized to control the morphology, size, and functionalization of multifunctional FA-PABA-Q188-PUL@AuNPs. Spherical FA-PABA-Q188-PUL@AuNPs obtained by a green, simple, and bio-inspired strategy under the optimum conditions were thoroughly characterized and had an average size of 12.6 ± 1.5 nm. The anticancer drug DOX was successfully loaded on monodispersed FA-PABA-Q188-PUL@AuNPs and the system exhibited excellent intracellular uptake, specificity, and physicochemical properties. The pH-responsive DOX release from FA-PABA-Q188-PUL@AuNPs-DOX showed fast release (85% after 72 h) under acidic conditions. Furthermore, FA-PABA-Q188-PUL@AuNPs-DOX enhanced the anticancer activity of DOX toward Chago-k1 cancer cells up to 4.8-fold and showed less cytotoxicity toward normal cells than free DOX. The FA-PABA-Q188-PUL@AuNPs-DOX induced the death of cells by increasing late apoptotic cells (26.4%) and arresting the cell cycle at S-G2/M phases. These results showed that innovative FA-PABA-Q188-PUL@AuNPs should be considered as new candidate platforms for anticancer drug delivery systems.


Subject(s)
Doxorubicin , Drug Carriers , Glucans , Gold , Metal Nanoparticles , Nanocomposites , Neoplasms , A549 Cells , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Glucans/chemistry , Glucans/pharmacokinetics , Glucans/pharmacology , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , Hep G2 Cells , Humans , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Nanocomposites/chemistry , Nanocomposites/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology
5.
Carbohydr Polym ; 230: 115625, 2020 Feb 15.
Article in English | MEDLINE | ID: mdl-31887856

ABSTRACT

A new positively charged nanoemulsion using quaternized chitosan (QCS) as a protective layer was developed to improve the stability and bioactivity of lipophilic active components. The anti-inflammatory Plai extract was chosen as both an active ingredient and an oil phase of the system. Compared with chitosan-coated nanoemulsion (NE2-CS) and uncoated nanoemulsion (NE1), the QCS coating could improve the stability of the Plai extract during 28 days. The particle size of NE1 increased from 141 nm to 202 nm after coating with QCS, whereas zeta potential changed from -22.03 mV for NE1 to 20.23 mV for NE2-QCS, confirming the presence of QCS. A clear improvement in anti-inflammatory, anti-cancer, and transdermal properties of Plai extract was verified for NE2-QCS, which could be due to the NEs' fineness and the permanent positive charge of the protective layer. Therefore, we suggested that QCS-coated NEs can be used as an effective transdermal delivery system for lipophilic active components.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Antineoplastic Agents/administration & dosage , Chitosan/analogs & derivatives , Nanoparticles/chemistry , Plant Extracts/administration & dosage , Skin Absorption , Zingiberaceae/chemistry , Anti-Inflammatory Agents/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Cell Line , Cell Line, Tumor , Drug Liberation , Emulsions/chemistry , Humans , Plant Extracts/pharmacokinetics , Static Electricity
6.
Nanomedicine (Lond) ; 15(1): 77-92, 2020 01.
Article in English | MEDLINE | ID: mdl-31868112

ABSTRACT

Aim: We aim to demonstrate that a local nanoparticle-mediated hyperthermia can effectively eliminate tumor-associated Tregs and thereby boost checkpoint blockade-based immunotherapy. Materials & methods: Photothermal therapy (PTT), mediated with systemically administered stealthy iron-oxide nanoparticles, was applied to treat BALB/c mice bearing 4T1 murine breast tumors. Flow cytometry was applied to evaluate both Treg and CD8+ T-cell population. Tumor growth following combination therapy of both PTT and anti-CTLA-4 was further evaluated. Results: Our data reveal that tumor-associated Tregs can be preferentially depleted via iron-oxide nanoparticles-mediated PTT. When combining PTT with anti-CTLA-4 immunotherapy, we demonstrate a significant inhibition of syngeneic 4T1 tumor growth. Conclusion: This study offers a novel strategy to overcome Treg-mediated immunosuppression and thereby to boost cancer immunotherapy.


Subject(s)
Breast Neoplasms/therapy , CTLA-4 Antigen/immunology , Immunotherapy , T-Lymphocytes, Regulatory/immunology , Animals , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/drug effects , CTLA-4 Antigen/antagonists & inhibitors , Combined Modality Therapy , Disease Models, Animal , Female , Ferric Compounds/chemistry , Ferric Compounds/pharmacology , Humans , Hyperthermia, Induced/methods , Immune Tolerance/drug effects , Immune Tolerance/immunology , Mice , Nanoparticles/chemistry , Phototherapy , Tumor Microenvironment/drug effects
7.
ACS Appl Mater Interfaces ; 11(27): 23858-23869, 2019 Jul 10.
Article in English | MEDLINE | ID: mdl-31245984

ABSTRACT

Although the functionalization of magnetic nanoparticles (MNPs) with biomolecules has been widely explored for various biological applications, achieving efficient bioconjugations with a wide range of biomolecules through a single, universal, and versatile platform remains a challenge, which may significantly impact their applications' outcomes. Here, we report a novel MNP platform composed of Au@Fe core/satellite nanoparticles (CSNPs) for versatile and efficient bioconjugations. The engineering of the CSNPs is facilely formed through the self-assembly of ultrasmall gold nanoparticles (AuNPs, 2-3 nm in diameter) around MNPs with a polysiloxane-containing polymer coating. The formation of the hybrid magnetic nanostructure is revealed by absorption spectroscopy, dynamic light scattering (DLS), transmission electron microscopy (TEM), element analysis using atomic absorption spectroscopy, and vibrating sample magnetometer. The versatility of biomolecule loading to the CSNP is revealed through the bioconjugation of a wide range of relevant biomolecules, including streptavidin, antibodies, peptides, and oligonucleotides. Characterizations including DLS, TEM, lateral flow strip assay, fluorescence assay, giant magnetoresistive nanosensor array, high-performance liquid chromatography, and absorption spectrum are performed to further confirm the efficiency of various bioconjugations to the CSNP. In conclusion, this study demonstrates that the CSNP is a novel MNP-based platform that offers versatile and efficient surface functionalization with various biomolecules.


Subject(s)
Coated Materials, Biocompatible/chemistry , Gold/chemistry , Iron/chemistry , Magnetite Nanoparticles , Metal Nanoparticles , Animals , Cattle , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/ultrastructure , Metal Nanoparticles/chemistry , Metal Nanoparticles/ultrastructure , Mice , Mice, Inbred BALB C , Particle Size
8.
Clin Cancer Res ; 24(17): 4242-4255, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29769207

ABSTRACT

Purpose: The response rates of Head and Neck Squamous Cell Carcinoma (HNSCC) to checkpoint blockade are below 20%. We aim to develop a mechanism-based vaccine to prevent HNSCC immune escape.Experimental Design: We performed RNA-Seq of sensitive and resistant HNSCC cells to discover central pathways promoting resistance to immune killing. Using biochemistry, animal models, HNSCC microarray, and immune cell deconvolution, we assessed the role of SOX2 in inhibiting STING-type I interferon (IFN-I) signaling-mediated antitumor immunity. To bypass SOX2-potentiated STING suppression, we engineered a novel tumor antigen-targeted nanosatellite vehicle to enhance the efficacy of STING agonist and sensitize SOX2-expressing HNSCC to checkpoint blockade.Results: The DNA-sensing defense response is the most suppressed pathway in immune-resistant HNSCC cells. We identified SOX2 as a novel inhibitor of STING. SOX2 facilitates autophagy-dependent degradation of STING and inhibits IFN-I signaling. SOX2 potentiates an immunosuppressive microenvironment and promotes HNSCC growth in vivo in an IFN-I-dependent fashion. Our unique nanosatellite vehicle significantly enhances the efficacy of STING agonist. We show that the E6/E7-targeted nanosatellite vaccine expands the tumor-specific CD8+ T cells by over 12-fold in the tumor microenvironment and reduces tumor burden. A combination of nanosatellite vaccine with anti-PD-L1 significantly expands tumor-specific CTLs and limits the populations expressing markers for exhaustion, resulting in more effective tumor control and improved survival.Conclusions: SOX2 dampens the immunogenicity of HNSCC by targeting the STING pathway for degradation. The nanosatellite vaccine offers a novel and effective approach to enhance the adjuvant potential of STING agonist and break cancer tolerance to immunotherapy. Clin Cancer Res; 24(17); 4242-55. ©2018 AACR.


Subject(s)
Cancer Vaccines/immunology , Membrane Proteins/genetics , SOXB1 Transcription Factors/genetics , Squamous Cell Carcinoma of Head and Neck/immunology , Animals , Autophagy/immunology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Cancer Vaccines/pharmacology , Drug Resistance, Neoplasm/immunology , Gene Expression Regulation, Neoplastic , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Immune Tolerance , Immunotherapy , Interferon Type I/genetics , Interferon Type I/immunology , Membrane Proteins/immunology , Mice , Nanostructures/administration & dosage , Nanostructures/chemistry , SOXB1 Transcription Factors/immunology , Squamous Cell Carcinoma of Head and Neck/prevention & control , Squamous Cell Carcinoma of Head and Neck/therapy , Tumor Microenvironment/immunology
9.
Acta Pharmacol Sin ; 38(6): 754-763, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28392567

ABSTRACT

Exosomes, a subgroup of extracellular vesicles (EVs), have been recognized as important mediators of long distance intercellular communication and are involved in a diverse range of biological processes. Because of their ideal native structure and characteristics, exosomes are promising nanocarriers for clinical use. Exosomes are engineered at the cellular level under natural conditions, but successful exosome modification requires further exploration. The focus of this paper is to summarize passive and active loading approaches, as well as specific exosome modifications and examples of the delivery of therapeutic and imaging molecules. Examples of exosomes derived from a variety of biological origins are also provided. The biocompatible characteristics of exosomes, with suitable modifications, can increase the stability and efficacy of imaging probes and therapeutics while enhancing cellular uptake. Challenges in clinical translation of exosome-based platforms from different cell sources and the advantages of each are also reviewed and discussed.


Subject(s)
Drug Delivery Systems , Exosomes/chemistry , Nanomedicine , Drug Carriers/chemistry , Drug Carriers/metabolism , Exosomes/metabolism , Humans , Nanoparticles/chemistry , Nanoparticles/metabolism
10.
Small ; 13(6)2017 Feb.
Article in English | MEDLINE | ID: mdl-27873448

ABSTRACT

The asymmetrical features and unique properties of multibuilding block Janus nanostructures (JNSs) provide superior functions for biomedical applications. However, their production process is very challenging. This problem has hampered the progress of JNS research and the exploration of their applications. In this study, an asymmetrical multibuilding block gold/iron oxide JNS has been generated to enhance photothermal effects and display colored Brownian motion in an optical trap. JNS is formed by seed-mediated self-assembly of nanoparticle-loaded thermocleavable micelles, where the hydrophobic backbones of the polymer are disrupted at high temperatures, resulting in secondary self-assembly and structural rearrangement. The JNS significantly enhances photothermal effects compared to their homogeneous counterpart after near-infrared (NIR) light irradiation. The asymmetrical distribution of gold and iron oxide within JNS also generates uneven thermophoretic force to display active colored Brownian rotational motion in a single-beam gradient optical trap. These properties indicate that the asymmetrical JNS could be employed as a strong photothermal therapy mediator and a fuel-free nanoscale Janus motor under NIR light.


Subject(s)
Light , Metal Nanoparticles/chemistry , Motion , Optical Tweezers , Temperature , Cell Line, Tumor , Color , Ferric Compounds/chemistry , Gold/chemistry , Humans , Metal Nanoparticles/ultrastructure , Polymers/chemical synthesis , Polymers/chemistry
11.
Nanotechnology ; 24(35): 355101, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-23940104

ABSTRACT

We report and demonstrate biomedical applications of a new technique--'living' PEGylation--that allows control of the density and composition of heterobifunctional PEG (HS-PEG-R; thiol-terminated poly(ethylene glycol)) on gold nanoparticles (AuNPs). We first establish 'living' PEGylation by incubating HS-PEG5000-COOH with AuNPs (∼20 nm) at increasing molar ratios from zero to 2000. This causes the hydrodynamic layer thickness to differentially increase up to 26 nm. The controlled, gradual increase in PEG-COOH density is revealed after centrifugation, based on the ability to re-suspend the pellet and increase the AuNP absorption. Using a fluorescamine-based assay we quantify differential HS-PEG5000-NH2 binding to AuNPs, revealing that it is highly efficient until AuNP saturation is reached. Furthermore, the zeta potential incrementally changes from -44.9 to +52.2 mV and becomes constant upon saturation. Using 'living' PEGylation we prepare AuNPs with different ratios of HS-PEG-RGD (RGD: Arg-Gly-Asp) and incubate them with U-87 MG (malignant glioblastoma) and non-target cells, demonstrating that targeting ligand density is critical to maximizing the efficiency of targeting of AuNPs to cancer cells. We also sequentially control the HS-PEG-R density to develop multifunctional nanoparticles, conjugating positively charged HS-PEG-NH2 at increasing ratios to AuNPs containing negatively charged HS-PEG-COOH to reduce uptake by macrophage cells. This ability to minimize non-specific binding/uptake by healthy cells could further improve targeted nanoparticle efficacy.


Subject(s)
Gold/metabolism , Nanoparticles/metabolism , Oligopeptides/metabolism , Polyethylene Glycols/metabolism , Biological Transport , Cell Line, Tumor , Drug Delivery Systems , Glioblastoma/drug therapy , Glioblastoma/metabolism , Gold/chemistry , Humans , Macrophages/metabolism , Nanoparticles/chemistry , Oligopeptides/chemistry , Polyethylene Glycols/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...