Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nano Lett ; 21(4): 1839-1847, 2021 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-33586442

RESUMO

Nanosubstrate engineering is an established approach for modulating cellular responses, but it remains infrequently exploited to facilitate the intracellular delivery of nanoparticles (NPs). We report nanoscale roughness of the extracellular environment as a critical parameter for regulating the cellular uptake of NPs. After seeding cells atop a substrate that contains randomly immobilized gold NPs (termed AuNP-S) with sub-10 nm surface roughness, we demonstrate that such cells internalize up to ∼100-fold more poly(ethylene glycol)-coated AuNPs (Au@PEG NPs) than those cells seeded on a conventional flat culture plate. Our result is generalizable to 4 different cell types and Au@PEG NPs modified with 13 different hydrocarbyl functional groups. Conditioning cells to AuNP-S not only leads to upregulation of clathrin- and integrin-related genes, but also supports elevated uptake of Au@PEG NPs via clathrin-mediated endocytosis. Our data suggest a simple and robust method for boosting the intracellular delivery of nanomedicines by nanosubstrate engineering.


Assuntos
Nanopartículas Metálicas , Nanopartículas , Clatrina , Endocitose , Ouro , Polietilenoglicóis
2.
Biochemistry ; 60(13): 1019-1030, 2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33169977

RESUMO

Designing nanoparticles (NPs) with desirable cell type-specific exocytosis properties, say promoting their exocytosis from scavenging cell types (e.g., macrophages and endothelial cells) or suppressing their exocytosis from target disease cell types (e.g., cancer cells), improves the application of nanomedicines. However, the design parameters available for tuning the exocytosis of NPs remain scarce in the "nano-cell" literature. Here, we demonstrate that surface modification of NPs with hydrocarbyl functional groups, commonly found in biomolecules and NP-based drug carriers, is a critical parameter for tuning the exocytosis of NPs from RAW264.7 macrophages, C166 endothelial cells, and HeLa epithelial cancer cells. To exclude the effect of hydrophobicity, we prepare a collection of hydrophilic NPs that bear a gold NP (AuNP) core, a dense polyethylene glycol (PEG) shell, and different types of hydrocarbyl groups (X) that are attached to the distal end of the PEG strands (termed "Au@PEG-X NPs"). For all three cell types tested, modification of NPs with straight-chain dodecane leads to a >10-fold increase in the level of cellular uptake, drastically higher than those of all other types of X tested. However, the probability of exocytosis of NPs significantly depends on the types of cell and X. Notably, NPs modified with cyclododecanes are most likely to be exocytosed by RAW264.7 and C166 cells (but not HeLa cells), accompanied by the release of intralumenal vesicles to the extracellular milieu. These data suggest a reductionist approach for rationally assembling bionanomaterials for nanomedicine applications by using hydrocarbyl functional groups as building blocks.


Assuntos
Exocitose , Nanopartículas/química , Animais , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Células Endoteliais/metabolismo , Ouro/química , Células HeLa , Humanos , Nanopartículas Metálicas/química , Camundongos , Tamanho da Partícula , Polietilenoglicóis/química , Células RAW 264.7 , Propriedades de Superfície
3.
Acc Chem Res ; 52(6): 1519-1530, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31058496

RESUMO

Advances in nanotechnology have empowered the design of bionanomaterials by assembling different types of natural biomolecules (e.g., nucleic acids, proteins, and lipids) as building blocks into nanoparticles (NPs) of 1-100 nm in diameter. Such bionanomaterials form the basis of useful nanomedicine applications, such as targeted delivery, gene regulation, molecular diagnostics, and immunomodulation. To achieve optimal performance in these applications, it is imperative that the NPs be delivered effectively to the organs, tissues, and cells of interest. A rational approach to facilitating the delivery of NPs is to develop a detailed and comprehensive understanding in their fundamental interactions with the biological system (or nano-bio interactions). Rigorous nano-bio research can provide mechanistic insights for circumventing the bottlenecks associated with inefficient and nonspecific delivery of NPs, catalyzing the clinical translation of nanomedicines. Cationic liposomes and lipid NPs are conventional carriers of therapeutic cargoes into cells due to their high ability to penetrate the cell membrane, a barrier comprised by an anionic phospholipid bilayer. Yet, cationic NPs tend to cause cytotoxicity and immune responses that may hamper their clinical translation. Contrary to cationic NPs, non-cationic NPs (be they near-neutral or anionic in surface charge) generally exhibit higher biocompatibility but enter mammalian cells in much less pronounced amounts. Intriguingly, some types of non-cationic NPs exhibit high biocompatibility and cellular uptake properties, all attractive features for intracellular delivery. In this Account, we present our studies of the interactions of non-cationic bionanomaterials with cells (or nano-cell interactions). To start with, we introduce the use of near-neutral poly(ethylene glycol)-coated NPs for probing the roles of two rarely explored physicochemical parameters on cellular uptake, namely, extracellular compression and alkylation. We next present the nano-cell interactions of two representative types of anionic bionanomaterials that effectively enter mammalian cells and have found widespread applications in the past decade, including DNA-coated NPs and polydopamine (PDA)-coated NPs. In our cell-based studies, we dissect the route of intracellular trafficking, pathway proteins that dictate cellular uptake, and trafficking of NPs. We further touch on our recent quantitative analysis of the cellular-level distribution of NPs in various organs and tissues of diseased animal models. Our results offer important design rules of NPs for achieving effective intracellular delivery and may even guide us to explore nanomedicine applications that we did not conceive before, such as using DNA-coated NPs for targeting atherosclerotic plaques and PDA-coated plasmonic nanoworms for photothermal killing of cancer cells. We conclude with our perspectives in elucidating nano-bio interactions via a reductionist approach, calling for closer attention to the role of functional groups and more refined studies on the organelle-level distribution of NPs and the genetic basis of in vivo distribution of NPs.


Assuntos
Transporte Biológico/fisiologia , Nanopartículas/metabolismo , Animais , Linhagem Celular Tumoral , DNA/química , Endocitose/fisiologia , Feminino , Ouro/química , Humanos , Indóis/química , Masculino , Camundongos , Polietilenoglicóis/química , Polímeros/química
4.
Langmuir ; 34(46): 14033-14045, 2018 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-30360612

RESUMO

Polydopamine (PDA)-coated nanoparticles are adhesive bionanomaterials widely utilized in intracellular applications, yet how their adhesiveness affects their colloidal stability and their interactions with serum proteins and mammalian cells remain unclear. In this work, we systematically investigate the combined effects of dopamine (DA) concentration and polymerization time (both reaction parameters spanning 2 orders of magnitude) on the morphological diversity of PDA-coated nanoparticles by coating PDA onto gold nanoparticle cores. Independent of the DA concentration, Au@PDA NPs remain largely aggregated upon several hours of limited polymerization; interestingly, extended polymerization for 2 days or longer yield randomly aggregated NPs, nearly monodisperse NPs, or worm-like NP chains in the ascending order of DA concentration. Upon exposure to serum proteins, the specific type of proteins adsorbed to the Au@PDA NPs strongly depends upon the DA concentration. As DA concentration increases, less albumin and more hemoglobin subunits adhere. Moreover, cellular uptake is a strong function of polymerization time. Serum-stabilized Au@PDA NPs prepared by limited polymerization enter Neuro-2a and HeLa cancer cells more abundantly than those prepared by extended polymerization. Our data underscore the importance of DA concentration and polymerization time for tuning the morphology and degree of intracellular delivery of PDA-coated nanostructures.


Assuntos
Ouro/química , Indóis/química , Nanopartículas Metálicas/química , Polímeros/química , Coroa de Proteína/química , Adsorção , Transporte Biológico , Dopamina/química , Células HeLa , Humanos , Indóis/metabolismo , Polímeros/metabolismo
5.
Angew Chem Int Ed Engl ; 57(12): 3064-3068, 2018 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-29364558

RESUMO

Functional siRNAs are employed as cross-linkers to direct the self-assembly of DNA-grafted polycaprolactone (DNA-g-PCL) brushes to form spherical and nanosized hydrogels via nucleic acid hybridization in which small interfering RNAs (siRNAs) are fully embedded and protected for systemic delivery. Owing to the existence of multivalent mutual crosslinking events inside, the crosslinked nanogels with tunable size exhibit not only good thermostability, but also remarkable physiological stability that can resist the enzymatic degradation. As a novel siRNA delivery system with spherical nucleic acid (SNA) architecture, the crosslinked nanogels can assist the delivery of siRNAs into different cells without any transfection agents and achieve the gene silencing effectively both in vitro and in vivo, through which a significant inhibition of tumor growth is realized in the anticancer treatment.


Assuntos
DNA/química , Sistemas de Liberação de Medicamentos , Neoplasias Experimentais/tratamento farmacológico , Polietilenoglicóis/química , Polietilenoimina/química , RNA Interferente Pequeno/administração & dosagem , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células HeLa , Humanos , Camundongos , Nanogéis , Neoplasias Experimentais/patologia , Poliésteres/química , RNA Interferente Pequeno/química , RNA Interferente Pequeno/farmacologia , RNA Interferente Pequeno/uso terapêutico
6.
Small ; 12(37): 5178-5189, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27442290

RESUMO

Biomedical applications of non-spherical nanoparticles such as photothermal therapy and molecular imaging require their efficient intracellular delivery, yet reported details on their interactions with the cell remain inconsistent. Here, the effects of nanoparticle geometry and receptor targeting on the cellular uptake and intracellular trafficking are systematically explored by using C166 (mouse endothelial) cells and gold nanoparticles of four different aspect ratios (ARs) from 1 to 7. When coated with poly(ethylene glycol) strands, the cellular uptake of untargeted nanoparticles monotonically decreases with AR. Next, gold nanoparticles are functionalized with DNA oligonucleotides to target Class A scavenger receptors expressed by C166 cells. Intriguingly, cellular uptake is maximized at a particular AR: shorter nanorods (AR = 2) enter C166 cells more than nanospheres (AR = 1) and longer nanorods (AR = 4 or 7). Strikingly, long targeted nanorods align to the cell membrane in a near-parallel manner followed by rotating by ≈90° to enter the cell via a caveolae-mediated pathway. Upon cellular entry, targeted nanorods of all ARs predominantly traffic to the late endosome without progressing to the lysosome. The studies yield important materials design rules for drug delivery carriers based on targeted, anisotropic nanoparticles.


Assuntos
Endocitose , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Ouro/química , Nanotubos/química , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular , DNA/metabolismo , Endocitose/efeitos dos fármacos , Células Endoteliais/ultraestrutura , Camundongos , Modelos Biológicos , Nanotubos/toxicidade , Nanotubos/ultraestrutura , Polietilenoglicóis/química
7.
Curr Opin Biotechnol ; 84: 103013, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37897860

RESUMO

Polydopamine (PDA) nanoparticles (NPs) have diverse nanomedicine applications owing to their biocompatibility and abundant entry to cells. Yet, our knowledge in their interactions with cells was infrequently studied until recent years. This review presents the latest insights into the cell-nano interactions of PDA NPs, including their 'self-targeting' to dopamine receptors for cellular entry without the aid of ligands, in vitro 'self-therapeutic' cellular responses (antiferroptosis, macrophage polarization, and modulation of mitochondrial bioenergetics) in the absence of drugs, and in vivo cellular localization and pharmacological properties upon various routes of administration. This review concludes with our perspectives on the therapeutic promise of PDA NPs and the need for studies on PDA biochemistry, biodegradability, and protein adsorption.


Assuntos
Nanopartículas , Polímeros , Polímeros/química , Nanopartículas/química , Indóis/química , Indóis/farmacologia
8.
ACS Nano ; 16(2): 2032-2045, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35137580

RESUMO

Understanding the exocytosis of nanoparticles (NPs) from cells is valuable because it informs design rules of NPs that support desirable cellular retention for nanomedicine applications, but investigations into the mechanism for the exocytosis of NPs remain scarce. We elucidate the mechanism for the exocytosis of dodecyl-terminated, polyethylene glycol-coated gold NPs (termed "dodecyl-PEG-AuNP"). The Au core enables ultrastructural differentiation of the exocytosed NPs from the nearby extracellular vesicles (EVs). The PEG shell prevents interparticle agglomeration or aggregation that disfavors exocytosis. The minute amounts of alkyl chains on the PEG shell not only promote cellular uptake but also improve exocytosis by up to 4-fold higher probability and upregulate exocytosis- and vesicle-related genes. After entering Kera-308 keratinocytes and trafficking to multivesicular bodies and lysosomes, these NPs exit the cell predominantly via unconventional exocytosis, accompanied by enhanced secretion of sub-100 nm, CD81-enriched exosomes. The pathway for NP exocytosis and subpopulation of EVs that are secreted alongside the exocytosed NPs depends on dodecyl loading. This work provides insights into dissecting the mechanism of NP exocytosis and its relationship with EV secretion.


Assuntos
Vesículas Extracelulares , Nanopartículas Metálicas , Nanopartículas , Animais , Exocitose , Ouro/química , Nanopartículas Metálicas/química , Nanopartículas/química , Polietilenoglicóis/química
9.
Biomater Sci ; 10(1): 189-201, 2021 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-34817474

RESUMO

A series of glutathione (GSH)-responsive polydopamine (PDA) nanoparticles (NPs) were prepared using a disulfide-linked dopamine dimer as starting material, of which the size could be tuned systematically by adjusting the amount of ammonia solution used. Molecules of a phthalocyanine (Pc)-based photosensitiser and an epidermal growth factor receptor (EGFR)-targeting peptide were then sequentially immobilised on the surface of the NPs through coupling with the surface functionalities of PDA. The immobilised Pc molecules in the resulting nanosystem were photodynamically inactive due to the strong self-quenching effect and the quenching by the PDA core. Upon exposure to GSH in phosphate-buffered saline or EGFR-positive cancer cells, namely A549 and A431 cells, the NPs were disassembled through cleavage of the disulfide linkages to release the Pc molecules, thereby restoring their fluorescence emission and singlet oxygen generation. The NPs with the smallest size (ca. 200 nm in diameter) exhibited the highest cellular uptake and high photocytotoxicity with IC50 values as low as 0.05 µM based on Pc. These NPs could also accumulate and be activated in the tumour of A431 tumour-bearing nude mice, lighting up the tumour with fluorescence over a period of 72 h and completely eradicating the tumour through laser irradiation for 10 min (675 nm, 20 J cm-2). The results suggest that these biodegradable and versatile PDA-based NPs can serve as a promising nanoplatform for fabrication of advanced photosensitisers for targeted photodynamic therapy.


Assuntos
Antineoplásicos/farmacologia , Nanopartículas , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Animais , Linhagem Celular Tumoral , Glutationa , Indóis , Camundongos , Camundongos Nus , Polímeros
10.
Nanoscale ; 13(37): 15899-15915, 2021 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34522935

RESUMO

A thioketal-linked dimer of 3,4-dihydroxy-L-phenylalanine was prepared which underwent self-polymerisation in the presence of doxorubicin (Dox) in an ethanol/water (1 : 4, v/v) mixture with ammonia. The resulting Dox-encapsulated polydopamine (PDA) nanoparticles were further conjugated with molecules of a zinc(II) phthalocyanine (Pc)-based photosensitiser and a peptide containing the heptapeptide QRHKPRE sequence (labelled as QRH) that can target the epidermal growth factor receptor (EGFR) overexpressed in cancer cells. Upon internalisation into these cells through receptor-mediated endocytosis, these nanoparticles labelled as PDA-Dox-Pc-QRH were disassembled gradually via cleavage of the thioketal linkages by the intrinsic intracellular reactive oxygen species (ROS). The stacked Pc molecules were then disaggregated, resulting in activation of their photosensitising property upon irradiation. The ROS generated by the activated Pc promoted further degradation of the nanoparticles and release of Dox, thereby enhancing cell death by synergistic chemo and photodynamic therapy. Systemic injection of PDA-Dox-Pc-QRH into EGFR-overexpressed tumour-bearing nude mice led to targeted delivery to the tumour, and subsequent light irradiation caused complete tumour ablation without inducing notable toxicity.


Assuntos
Nanopartículas , Fotoquimioterapia , Animais , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Indóis , Camundongos , Camundongos Nus , Polímeros , Espécies Reativas de Oxigênio
11.
Nanoscale ; 13(13): 6499-6512, 2021 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-33885529

RESUMO

A novel nanosystem of polydopamine-coated gold nanorods (AuNR@PDA) immobilised with molecules of hairpin DNA-conjugated distyryl boron dipyrromethene (DSBDP) was designed and fabricated for detection of microRNA-21 (miR-21). By using this oncogenic stimulus, the photodynamic effect of the DSBDP-based photosensitiser was also activated. In the presence of miR-21, the fluorescence intensity of the nanosystem was increased due to the dissociation of the conjugate from AuNR@PDA upon hybridisation. The intracellular fluorescence intensity triggered by intracellular miR-21 was in the order: MCF-7 > HeLa > HEK-293, which was in accordance with their miR-21 expression levels. The specificity was demonstrated by comparing the results with those of an analogue with a scrambled DNA sequence. The nanosystem could also result in miR-21-mediated photodynamic eradication of miR-21-overexpressed MCF-7 cells. After intravenous injection of the nanosystem into HeLa tumour-bearing nude mice, the fluorescence intensity of the tumour was increased over 24 h and was about 3-fold stronger than that of the scrambled analogue. Upon irradiation, the nanosystem could also greatly reduce the size of the tumour without causing significant tissue damage in the major organs. The overall results showed that this nanoplatform can serve as a specific and potent theranostic agent for simultaneous miR-21 detection and miR-21-mediated photodynamic therapy.


Assuntos
MicroRNAs , Nanotubos , Fotoquimioterapia , Animais , Boro , DNA , Ouro , Células HEK293 , Humanos , Indóis , Camundongos , Camundongos Nus , MicroRNAs/genética , Polímeros , Porfobilinogênio/análogos & derivados
12.
Chem Commun (Camb) ; 56(54): 7439-7442, 2020 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-32494799

RESUMO

A multitude of maleimides are grafted onto the backbone of a phosphorothioate antisense oligonucleotide (ASO) to generate the construct of maleimide-grafted ASO (Mal-g-ASO). Through click conjugation with cell membrane thiols that triggers endocytosis-independent cellular internalization, Mal-g-ASO exhibited enhanced cellular uptake efficiency, resulting in a remarkable improvement of ASO-based gene silencing.


Assuntos
Inativação Gênica , Maleimidas/química , Oligonucleotídeos Antissenso/metabolismo , Animais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/metabolismo , Linhagem Celular , Endocitose , Corantes Fluorescentes/química , Humanos , Camundongos , Microscopia Confocal , Oligonucleotídeos Antissenso/química , Fosfatos/química , Ratos
13.
ACS Appl Mater Interfaces ; 11(15): 13888-13904, 2019 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-30516979

RESUMO

Many nanoparticle-based carriers to atherosclerotic plaques contain peptides, lipoproteins, and sugars, yet the application of DNA-based nanostructures for targeting plaques remains infrequent. In this work, we demonstrate that DNA-coated superparamagnetic iron oxide nanoparticles (DNA-SPIONs), prepared by attaching DNA oligonucleotides to poly(ethylene glycol)-coated SPIONs (PEG-SPIONs), effectively accumulate in the macrophages of atherosclerotic plaques following an intravenous injection into apolipoprotein E knockout (ApoE-/-) mice. DNA-SPIONs enter RAW 264.7 macrophages faster and more abundantly than PEG-SPIONs. DNA-SPIONs mostly enter RAW 264.7 cells by engaging Class A scavenger receptors (SR-A) and lipid rafts and traffic inside the cell along the endolysosomal pathway. ABS-SPIONs, nanoparticles with a similarly polyanionic surface charge as DNA-SPIONs but bearing abasic oligonucleotides also effectively bind to SR-A and enter RAW 264.7 cells. Near-infrared fluorescence imaging reveals evident localization of DNA-SPIONs in the heart and aorta 30 min post-injection. Aortic iron content for DNA-SPIONs climbs to the peak (∼60% ID/g) 2 h post-injection (accompanied by profuse accumulation in the aortic root), but it takes 8 h for PEG-SPIONs to reach the peak aortic amount (∼44% ID/g). ABS-SPIONs do not appreciably accumulate in the aorta or aortic root, suggesting that the DNA coating (not the surface charge) dictates in vivo plaque accumulation. Flow cytometry analysis reveals more pronounced uptake of DNA-SPIONs by hepatic endothelial cells, splenic macrophages and dendritic cells, and aortic M2 macrophages (the cell type with the highest uptake in the aorta) than PEG-SPIONs. In summary, coating nanoparticles with DNA is an effective strategy of promoting their systemic delivery to atherosclerotic plaques.


Assuntos
DNA/química , Compostos Férricos/química , Nanopartículas de Magnetita/química , Administração Intravenosa , Animais , Meios de Contraste/química , Meios de Contraste/farmacocinética , Fígado/patologia , Macrófagos/citologia , Macrófagos/metabolismo , Nanopartículas de Magnetita/administração & dosagem , Nanopartículas de Magnetita/análise , Masculino , Camundongos , Camundongos Knockout , Microscopia Confocal , Oligonucleotídeos/química , Placa Aterosclerótica/diagnóstico por imagem , Placa Aterosclerótica/patologia , Polietilenoglicóis/química , Células RAW 264.7 , Espectroscopia de Luz Próxima ao Infravermelho , Distribuição Tecidual
14.
ACS Nano ; 12(3): 2088-2093, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29485854

RESUMO

A longstanding and widely accepted bottleneck in the targeted delivery of intravenously injected nanoparticles lies in their clearance by macrophages in the liver and spleen. In this Perspective, we call for deeper understanding of the critical role of endothelial cells in the sequestration of nanoparticles in vivo. In this issue of ACS Nano, Campbell et al. used a combination of real-time imaging and genome-editing methods to demonstrate that stabilin-2 is an important receptor for removing anionic liposomes from blood circulation in a zebrafish model. Such mechanistic insights at the molecular level will provide a more holistic picture of the in vivo sequestration of administered nanoparticles beyond the cellular level and pose valuable design considerations for redistributing nanoparticles in vivo.


Assuntos
Células Endoteliais/metabolismo , Fígado/metabolismo , Macrófagos/metabolismo , Nanopartículas/metabolismo , Animais , Receptores de Hialuronatos/metabolismo , Lipossomos/metabolismo , Nanopartículas/administração & dosagem , Fagocitose , Baço/metabolismo , Peixe-Zebra/metabolismo
15.
Int J Pharm ; 532(1): 647-655, 2017 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-28827202

RESUMO

Spherical nucleic acid gold nanoparticles represent a unique nanotechnology in which the spherical arrangement of oligonucleotides enables the nanoparticles to be efficiently internalized into cells expressing scavenger receptors class A (SR-A). Herein, we seek to replace the gold core with a biodegradable polymeric construct and explore their potential applications in targeted drug delivery. Oligonucleotide-conjugated poly(ethylene glycol)-block-poly(ε-caprolactone) was synthesized and characterized by 1H NMR and gel electrophoresis. This polymer was applied to fabricate micellar nanoparticles (OLN-NPs) by an anti-solvent method. These nanoparticles have a mean particle size about 58.1nm with a narrow size distribution (PDI <0.2) and they were also non-cytotoxic. Relative to non-targeted NPs, OLN-NPs exhibited substantially better uptake (3.94×) in a mouse endothelial cell line (C166), attributing to lipid-raft-mediated endocytosis via SR-A. To explore the potential applications of OLN-NPs as drug carriers, paclitaxel, a poorly soluble anti-angiogenic compound, was selected as the model. OLN-NPs increased the solubility of paclitaxel by at least 300×. The boosted drug solubility in conjunction with improved cellular uptake translated into enhanced in vitro efficacy in the inhibition of angiogenesis. In conclusions, OLN-NPs show considerable promise in targeted drug delivery and their potential applications should be further investigated.


Assuntos
Sistemas de Liberação de Medicamentos , Lactonas/química , Nanopartículas/química , Oligonucleotídeos/química , Polietilenoglicóis/química , Receptores Depuradores/metabolismo , Animais , Linhagem Celular , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA