Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 12(1): 3527, 2022 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-35241750

RESUMO

Despite immense revolutionary therapeutics potential, sustaining release of active small interfering RNA (siRNA) remains an arduous challenge. The development of nanoparticles with siRNA sustained release capabilities provides an avenue to enhance the therapeutic efficacy of gene-based therapy. Herein, we present a new system based on the encapsulation of siRNA/chitosan-methacrylate (CMA) complexes into liposomes to form UV crosslinkable Nanolipogels (NLGs) with sustained siRNA-release properties in vitro. We demonstrated that the CMA nanogel in NLGs can enhance the encapsulation efficiency of siRNA and provide sustained release of siRNA up to 28 days. To understand the particle mechanism of cellular entry, multiple endocytic inhibitors have been used to investigate its endocytosis pathways. The study saw positively charged NLGs entering cells via multiple endocytosis pathways, facilitating endosomal escape and slowly releasing siRNA into the cytoplasm. Transfection experiments confirmed that the crosslinked NLG delivery system provides effective transfection and prolonged silencing effect up to 14 days in cell cultures. We expect that this sustained-release siRNA NLG platform would be of interest in both fundamental biological studies and in clinical applications to extend the use of siRNA-based therapies.


Assuntos
Quitosana , Nanopartículas , Quitosana/metabolismo , Preparações de Ação Retardada , Inativação Gênica , Metacrilatos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
2.
Expert Opin Drug Deliv ; 17(8): 1165-1176, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32484723

RESUMO

BACKGROUND: Nanoparticles that actively target tissues, with ligands attached at the extremity of polyethylene glycol (PEG) spacer, are a promising strategy to enhance target cell specificity and internalization. However, the interplay between the targeting ligands and the adjacent ligand-free PEG remains poorly understood. RESEARCH DESIGN AND METHODS: Experimentally, liposomes containing active folate ligands were firstly formulated and the optimum amount of ligand that yields the highest foam cell uptake was determined. Subsequently, ligand-free PEG was incorporated, and the effects of PEG lengths and concentrations on foam cell uptake were evaluated after the nanoparticles were incubated in human serum for 90 min. RESULTS: It was demonstrated that the targeting efficiency progressively decreased and was eventually annulled as PEG-to-ligand ratio was increased, with loss of targeting effect occurring at PEG-to-ligand ratio of >2 for PEG 750, >0.5 for PEG 2000 and <0.5 for PEG 5000. CONCLUSIONS: This work demonstrates that PEG-to-ligand ratio and serum coating on nanoparticle surface are both important features to be considered in the design of active targeting nanocarriers. This work also supports the development of novel active targeting nanotherapies for atherosclerosis.


Assuntos
Aterosclerose/tratamento farmacológico , Células Espumosas/metabolismo , Nanopartículas , Polietilenoglicóis/química , Animais , Ácido Fólico/metabolismo , Humanos , Ligantes , Lipossomos , Camundongos , Camundongos Knockout
3.
Adv Healthc Mater ; 9(14): e2000465, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32543010

RESUMO

Atherosclerosis is a chronic disease that can lead to life-threatening events such as myocardial infarction and stroke, is characterized by the build-up of lipids and immune cells within the arterial wall. It is understood that inflammation is a hallmark of atherosclerosis and can be a target for therapy. In support of this concept, an injectable nanoliposomal formulation encapsulating fluocinolone acetonide (FA), a corticosteroid, is developed that allows for drug delivery to atherosclerotic plaques while reducing the systemic exposure to off-target tissues. In this study, FA is successfully incorporated into liposomal nanocarriers of around 100 nm in size with loading efficiency of 90% and the formulation exhibits sustained release up to 25 d. The anti-inflammatory effect and cholesterol efflux capability of FA-liposomes are demonstrated in vitro. In vivo studies carried out with an apolipoprotein E-knockout (Apoe-/- ) mouse model of atherosclerosis show accumulation of liposomes in atherosclerotic plaques, colocalization with plaque macrophages and anti-atherogenic effect over 3 weeks of treatment. This FA-liposomal-based nanocarrier represents a novel potent nanotherapeutic option for atherosclerosis.


Assuntos
Aterosclerose , Placa Aterosclerótica , Animais , Apolipoproteínas E , Aterosclerose/tratamento farmacológico , Lipossomos , Macrófagos , Camundongos , Camundongos Knockout , Placa Aterosclerótica/tratamento farmacológico
4.
Nanoscale Adv ; 2(3): 1040-1045, 2020 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-36133062

RESUMO

We present studies of protein (insulin) efflux rates from nano-sized core-shell systems with a gelled core and a lipid bilayer (nanolipogels). The efflux control mechanism is the manipulation of mesh size, and we show that diffusion control via crosslinking is the dominant mechanism for efflux control. The concept is inspired by the macromolecular crowding effect in human cells, which may be considered as a physical network of undefined mesh size. Our bio-inspired system is made of chemically crosslinked water-swellable poly(ethylene glycol) diacrylate cores, whose mesh size can be manipulated to yield a quantifiable crowding effect that then leads to predictable release rates for biomacromolecules.

5.
Mar Drugs ; 17(6)2019 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-31242678

RESUMO

Approximately 4000 diseases are associated with malfunctioning genes in a particular cell type. Gene-based therapy provides a platform to modify the disease-causing genes expression at the cellular level to treat pathological conditions. However, gene delivery is challenging as these therapeutic genes need to overcome several physiological and intracellular barriers in order, to reach the target cells. Over the years, efforts have been dedicated to develop efficient gene delivery vectors to overcome these systemic barriers. Chitosan, a versatile polysaccharide, is an attractive non-viral vector material for gene delivery mainly due to its cationic nature, biodegradability and biocompatibility. The present review discusses the design factors that are critical for efficient gene delivery/transfection and highlights the recent progress of gene therapy using chitosan-based carriers.


Assuntos
Quitosana/química , Portadores de Fármacos/química , Animais , Técnicas de Transferência de Genes , Terapia Genética/métodos , Humanos , Polissacarídeos/química , Transfecção/métodos
6.
Expert Opin Drug Deliv ; 15(10): 937-949, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30173580

RESUMO

OBJECTIVES: Clinical translation of siRNA therapeutics has been severely limited due to the lack of stable and sustained siRNA delivery systems. Furthermore, when nanocarrier systems with siRNA are administered systemically to treat diseases, insufficient doses reach the target tissue. Here we report the successful development of a new nanocarrier system for the management of fibrosis. METHODS: The new carrier has a hydroxyapatite core, with alternating layers of siRNA and a cationic peptide. The siRNA used here targets secreted protein acidic and rich in cysteine (SPARC), a key matricellular protein involved in the regulation of collagen fibrillogenesis and assembly. We have also used FRET studies to elucidate the fate of the particles inside cells, including the mechanistic details of layer-by-layer detachment. RESULTS: In vitro studies using murine conjunctiva fibroblasts show sustained release over 2 weeks, and that such released siRNA sustained SPARC knockdown without affecting cell growth, and maintained siRNA presence in the cells for at least two weeks with a single-dose treatment. Release studies of siRNA from particles in vitro gave insight on how the particles delivered prolonged gene-silencing effects. CONCLUSION: A single treatment of the layer-by-layer nanoparticle designed can achieve sustained gene silencing over 2 weeks. Localized delivery of stabilized siRNA with sustained-release capabilities opens the door for many other applications of siRNA-based gene regulation.


Assuntos
Inativação Gênica , Nanopartículas , Osteonectina/genética , RNA Interferente Pequeno/administração & dosagem , Animais , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno/genética
7.
Br J Ophthalmol ; 102(10): 1460-1470, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30021812

RESUMO

BACKGROUND: To develop targeted antifibrotic therapy for glaucoma filtration surgery; this study determines the effectiveness of small interfering RNA (siRNA) to reduce in vivo secreted protein acidic and rich in cysteine (SPARC) expression using the mouse model of conjunctival scarring. METHODS: Experimental surgery was performed as described for the mouse model of conjunctival scarring. Scrambled (siScram) or Sparc (siSparc) siRNAs, loaded on layer-by-layer (LbL) nanoparticles, were injected into the conjunctiva immediately after surgery. Expression of Sparc, Col1a1, Fn1 and Mmp14 was measured by real-time PCR and immunoblotting on days 7 and 14 postsurgery. Live imaging of the operated eyes was performed using slit lamp, anterior segment-optical coherence tomography and confocal microscopy. Tissue pathology was evaluated by histochemical and immunofluorescent analyses of operated conjunctival cryosections. Tissue apoptosis was quantitated by annexin V assay. RESULTS : siSparc, delivered via expanded LbL nanoparticles, significantly inhibited Sparc transcription in both day 7 (2.04-fold) and day 14 (1.39-fold) treated tissues. Sparc suppression on day 7 was associated with a significant reduction of Col1a1 (2.52-fold), Fn1 (2.89-fold) and Mmp14 (2.23-fold) mRNAs. At the protein level, both SPARC and collagen 1A1 (COL1A1) were significantly reduced at both time points with siSparc treatment. Nanoparticles were visualised within cell-like structures by confocal microscopy, while overt tissue response or apoptosis was not observed. CONCLUSIONS : SPARC targeted therapy effectively reduced both SPARC and collagen production in the operated mouse conjunctiva. This proof-of-concept study suggests that targeted treatment of fibrosis in glaucoma surgery is safe and feasible, with the potential to extend to a range of potential genes associated with fibrosis.


Assuntos
Colágeno/metabolismo , Túnica Conjuntiva/patologia , Doenças da Túnica Conjuntiva/terapia , Córnea/metabolismo , Cirurgia Filtrante/efeitos adversos , Terapia Genética/métodos , Osteonectina/uso terapêutico , Animais , Células Cultivadas , Doenças da Túnica Conjuntiva/genética , Doenças da Túnica Conjuntiva/metabolismo , Córnea/patologia , Modelos Animais de Doenças , Citometria de Fluxo , Regulação da Expressão Gênica , Glaucoma/cirurgia , Immunoblotting , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Osteonectina/biossíntese , Osteonectina/genética , Complicações Pós-Operatórias , RNA/genética , Reação em Cadeia da Polimerase em Tempo Real , Tomografia de Coerência Óptica
8.
J Control Release ; 284: 39-48, 2018 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-29902484

RESUMO

This is a review of nanotherapeutic systems, specifically those that exhibit controlled release of the encapsulated bioactive compound. The survey includes the delivery of a range of bioactive compounds, from lipophilic small molecules to hydrophilic proteins and siRNA molecules. The research into enabling sustained delivery of these compounds from nanocarriers has been prolific, but clinical success has been harder to achieve. This is partly because achieving true sustained duration of action over several days is difficult when the carrier dimensions become less than about 400 nm, due to the much shorter diffusion path length compared to micron-sized carrier systems. Other options must be sought to control the efflux of incorporated bioactives, particularly when these bioactives have moderate to high hydrophilicity. A few of these options are discussed critically in this review. We also answer the question: is controlled release needed for nanotherapies? We present the case for controlled release in specific conditions, with two examples from our own work: one for treatment of glaucoma, and the second for inhibition of fibrosis following surgery. The former is sustaining the release of a small-molecule lipophilic drug, while the latter focusses on sustained siRNA delivery.


Assuntos
Preparações de Ação Retardada/química , Nanopartículas/química , Peptídeos/administração & dosagem , Preparações Farmacêuticas/administração & dosagem , Proteínas/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Animais , Humanos , Nanotecnologia/métodos , Interferência de RNA
9.
Small ; 10(9): 1790-8, 2014 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-24510544

RESUMO

Efficient and safe delivery systems for siRNA therapeutics remain a challenge. Elevated secreted protein, acidic, and rich in cysteine (SPARC) protein expression is associated with tissue scarring and fibrosis. Here we investigate the feasibility of encapsulating SPARC-siRNA in the bilayers of layer-by-layer (LbL) nanoparticles (NPs) with poly(L-arginine) (ARG) and dextran (DXS) as polyelectrolytes. Cellular binding and uptake of LbL NPs as well as siRNA delivery were studied in FibroGRO cells. siGLO-siRNA and SPARC-siRNA were efficiently coated onto hydroxyapatite nanoparticles. The multilayered NPs were characterized with regard to particle size, zeta potential and surface morphology using dynamic light scattering and transmission electron microscopy. The SPARC-gene silencing and mRNA levels were analyzed using ChemiDOC western blot technique and RT-PCR. The multilayer SPARC-siRNA incorporated nanoparticles are about 200 nm in diameter and are efficiently internalized into FibroGRO cells. Their intracellular fate was also followed by tagging with suitable reporter siRNA as well as with lysotracker dye; confocal microscopy clearly indicates endosomal escape of the particles. Significant (60%) SPARC-gene knock down was achieved by using 0.4 pmole siRNA/µg of LbL NPs in FibroGRO cells and the relative expression of SPARC mRNA reduced significantly (60%) against untreated cells. The cytotoxicity as evaluated by xCelligence real-time cell proliferation and MTT cell assay, indicated that the SPARC-siRNA-loaded LbL NPs are non-toxic. In conclusion, the LbL NP system described provides a promising, safe and efficient delivery platform as a non-viral vector for siRNA delivery that uses biopolymers to enhance the gene knock down efficiency for the development of siRNA therapeutics.


Assuntos
Inativação Gênica , Técnicas de Transferência de Genes , Nanopartículas/química , Osteonectina/genética , RNA Interferente Pequeno/metabolismo , Endocitose , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Humanos , Espaço Intracelular/metabolismo , Masculino , Nanopartículas/ultraestrutura , Proteínas de Neoplasias/metabolismo , Osteonectina/antagonistas & inibidores , Osteonectina/biossíntese , Osteonectina/ultraestrutura , Tamanho da Partícula , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Proteínas Ribossômicas/metabolismo , Eletricidade Estática
10.
Biomaterials ; 32(11): 2969-78, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21257200

RESUMO

This work developed a multimodal imaging system by co-encapsulating superparamagnetic iron oxides (IOs) and quantum dots (QDs) in the nanoparticles of poly (lactic acid) - d-α-tocopheryl polyethylene glycol 1000 succinate (PLA-TPGS) for concurrent imaging of the magnetic resonance imaging (MRI) and the fluorescence imaging to combine their advantages and to overcome their disadvantages as well as to promote a sustained and controlled imaging with passive targeting effects to the diseased cells. The QDs and IOs-loaded PLA-TPGS NPs were prepared by a modified nanoprecipitation method, which were then characterized for their size and size distribution, zeta potential and the imaging agent encapsulation efficiency. The transmission electron microscopy (TEM) images showed direct evidence for the well-dispersed distribution of the QDs and IOs within the PLA-TPGS NPs. The cellular uptake and the cytotoxicity of the PLA-TPGS NPs formulation of QDs and IOs were investigated in vitro with MCF-7 breast cancer cells, which were conducted in close comparison with the free QDs and IOs at the same agent dose. The Xenograft model was also conducted for biodistribution of the QDs and IOs-loaded PLA-TPGS NPs among the various organs, which showed greatly enhanced tumor imaging due to the passively targeting effects of the NPs to the tumor. Images of tumors were acquired in vivo by a 7T MRI scanner. Further ex vivo images of the tumors were obtained by confocal laser scanning microscopy. Such a multimodal imaging system shows great advantages of both contrast agents making the resultant probe highly sensitive with good depth penetration, which confirms the diagnosis obtained from each individual imaging. With therapeutics co-encapsulation and ligand conjugation, such nanoparticles system can realize a multi-functional system for medical diagnosis and treatment.


Assuntos
Compostos Férricos/química , Nanopartículas/química , Neoplasias/patologia , Polietilenoglicóis/química , Pontos Quânticos , Succinatos/química , Animais , Linhagem Celular Tumoral , Diagnóstico por Imagem/métodos , Feminino , Humanos , Camundongos , Camundongos SCID , Microscopia Eletrônica de Transmissão , Nanopartículas/ultraestrutura , Nanotecnologia/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...