Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Opt Express ; 30(18): 32051-32060, 2022 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-36242274

RESUMEN

We demonstrate a short-wave infrared computed tomography method. It uses a fiber-coupled 1.44µm super-luminescent diode as light source, a PbSe photodiode as infrared detector, and an electronically controlled rotation and translation stage for high-speed Radon scanning. It is a safe and low power nondestructive testing method that can be used for the detection of plastic polymers, biological tissue and other materials that visible light cannot penetrate. We analyze the theoretical resolution of the method and build a short-wave infrared computed tomography system, which realizes the tomography and 3D reconstruction of black plastic bottles and artificial blood vessels. The measured resolution reaches10µm.


Asunto(s)
Sustitutos Sanguíneos , Radón , Plásticos , Tomografía Computarizada por Rayos X
2.
Mol Pharm ; 18(3): 928-939, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33427470

RESUMEN

Photodynamic therapy (PDT) is promising for clinical cancer therapy; however, the efficacy was limited as an individual treatment regimen. Here, an approach synergistically combining PDT and nitric oxide (NO) gas therapy along with destruction of the tumor extracellular matrix (ECM) was presented to eliminate cancer. Specifically, the NO donor l-arginine (l-Arg) and the photosensitizer indocyanine green (ICG) were co-encapsulated in poly(lactic-glycolic acid) (PLGA) nanoparticles and then loaded into the poly(ε-caprolactone)-poly(ethylene glycol)-poly(ε-caprolactone) (PCL-PEG-PCL) hydrogel to develop an injectable, thermosensitive dual drug delivery system (PLGA@ICG@l-Arg/Gel). Significantly, reactive oxygen species (ROS) produced by PLGA@ICG@l-Arg/Gel under near-infrared (NIR) light irradiation could not only result in the apoptosis of cancer cells but also oxidize l-Arg to generate NO, which could suppress the proliferation of cancer cells. Moreover, ROS could further oxidize NO to generate peroxynitrite anions (ONOO-). ONOO- could activate matrix metalloproteinases (MMPs), which notably degraded collagen in ECM so as to damage the tumor microenvironment. PLGA@ICG@l-Arg/Gel significantly increased the antitumor efficacy against highly malignant 4T1 tumors in mice. Taken together, PLGA@ICG@l-Arg/Gel is a multifunctional platform that provides a novel strategy for cancer treatment with cascade amplification of the ROS oxidation effect, which holds great potential in clinical translation.


Asunto(s)
Arginina/química , Colágeno/metabolismo , Hidrogeles/administración & dosificación , Verde de Indocianina/química , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Óxido Nítrico/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Femenino , Hidrogeles/química , Ratones , Ratones Endogámicos BALB C , Neoplasias/metabolismo , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/química , Fototerapia/métodos , Poliésteres/química , Polietilenglicoles/química , Microambiente Tumoral/efectos de los fármacos
3.
J Am Soc Nephrol ; 31(10): 2292-2311, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32769144

RESUMEN

BACKGROUND: Progressive fibrosis is the underlying pathophysiological process of CKD, and targeted prevention or reversal of the profibrotic cell phenotype is an important goal in developing therapeutics for CKD. Nanoparticles offer new ways to deliver antifibrotic therapies to damaged tissues and resident cells to limit manifestation of the profibrotic phenotype. METHODS: We focused on delivering plasmid DNA expressing bone morphogenetic protein 7 (BMP7) or hepatocyte growth factor (HGF)-NK1 (HGF/NK1) by encapsulation within chitosan nanoparticles coated with hyaluronan, to safely administer multifunctional nanoparticles containing the plasmid DNA to the kidneys for localized and sustained expression of antifibrotic factors. We characterized and evaluated nanoparticles in vitro for biocompatibility and antifibrotic function. To assess antifibrotic activity in vivo, we used noninvasive delivery to unilateral ureteral obstruction mouse models of CKD. RESULTS: Synthesis of hyaluronan-coated chitosan nanoparticles containing plasmid DNA expressing either BMP7 or NGF/NKI resulted in consistently sized nanoparticles, which-following endocytosis driven by CD44+ cells-promoted cellular growth and inhibited fibrotic gene expression in vitro. Intravenous tail injection of these nanoparticles resulted in approximately 40%-45% of gene uptake in kidneys in vivo. The nanoparticles attenuated the development of fibrosis and rescued renal function in unilateral ureteral obstruction mouse models of CKD. Gene delivery of BMP7 reversed the progression of fibrosis and regenerated tubules, whereas delivery of HGF/NK1 halted CKD progression by eliminating collagen fiber deposition. CONCLUSIONS: Nanoparticle delivery of HGF/NK1 conveyed potent antifibrotic and proregenerative effects. Overall, this research provided the proof of concept on which to base future investigations for enhanced targeting and transfection of therapeutic genes to kidney tissues, and an avenue toward treatment of CKD.


Asunto(s)
Antifibrinolíticos/administración & dosificación , Proteína Morfogenética Ósea 7/genética , Técnicas de Transferencia de Gen , Factor de Crecimiento de Hepatocito/genética , Nanopartículas Multifuncionales , Insuficiencia Renal Crónica/terapia , Animales , Técnicas de Cultivo de Célula , Quitosano , Modelos Animales de Enfermedad , Ácido Hialurónico , Ratones , Polímeros
4.
Mol Pharm ; 17(9): 3513-3525, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32787283

RESUMEN

Poly[lactic-co-(glycolic acid)] (PLGA) is arguably one of the most versatile synthetic copolymers used for biomedical applications. In vivo delivery of multiple substances including cells, pharmaceutical compounds, and antigens has been achieved by using PLGA-based micro-/nanoparticles although, presently, the exact biological impact of PLGA particles on the immune system remains controversial. Type 1 diabetes (T1D) is one subtype of diabetes characterized by the attack of immune cells against self-insulin-producing pancreatic islet cells. Considering the autoimmune etiology of T1D and the recent use of PLGA particles for eliciting desired immune responses in various aspects of immunotherapy, for the present study, a combination of Ins29-23 peptide (a known autoantigen of T1D) and PLGA microparticles was selected for T1D prevention assessment in nonobese diabetic (NOD) mice, a well-known animal model with spontaneous development of T1D. Thus, inoculation of PLGA microparticles + Ins29-23 completely prevented T1D development, significantly better than untreated controls and mice treated by either PLGA microparticles or Ins29-23 per se. Subsequent mechanistic investigation further revealed a facilitative role of PLGA microparticles in immune tolerance induction. In summary, our data demonstrate an adjuvant potential of PLGA microparticles in tolerance induction and immune remodulation for effective prevention of autoimmune diseases such as T1D.


Asunto(s)
Adyuvantes Inmunológicos/química , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Tolerancia Inmunológica/efectos de los fármacos , Insulina/inmunología , Microplásticos/química , Fragmentos de Péptidos/inmunología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Adyuvantes Inmunológicos/farmacología , Animales , Antígenos/inmunología , Células Cultivadas , Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Femenino , Tolerancia Inmunológica/inmunología , Inmunidad/efectos de los fármacos , Inmunidad/inmunología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/inmunología , Ratones , Ratones Endogámicos NOD , Nanopartículas/química
5.
Biomacromolecules ; 21(3): 1243-1253, 2020 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-32045224

RESUMEN

Multifunctional tissue adhesives with excellent adhesion, antibleeding, anti-infection, and wound healing properties are desperately needed in clinical surgery. However, the successful development of multifunctional tissue adhesives that simultaneously possess all these properties remains a challenge. We have prepared a novel chitosan-based hydrogel adhesive by integration of hydrocaffeic acid-modified chitosan (CS-HA) with hydrophobically modified chitosan lactate (hmCS lactate) and characterized its gelation time, mechanical properties, and microstructure. Tissue adhesion properties were evaluated using both pigskin and intestine models. In situ antibleeding efficacy was demonstrated via the rat hemorrhaging liver and full-thickness wound closure models. Good antibacterial activity and anti-infection capability toward S. aureus and P. aeruginosa were confirmed using in vitro contact-killing assays and an infected pigskin model. The result of coculturing with 3T3 fibroblast cells indicated that the hydrogels have no significant cytotoxicity. Most importantly, the biocompatible and biodegradable CS-HA/hmCS lactate hydrogel was able to close the wound in a sutureless way and promote wound healing. Our results demonstrate that this hydrogel has great promise for sutureless closure of surgical incisions.


Asunto(s)
Quitosano , Adhesivos Tisulares , Adhesivos/farmacología , Animales , Antibacterianos/farmacología , Hidrogeles/farmacología , Ratas , Staphylococcus aureus , Adhesivos Tisulares/farmacología
6.
Soft Matter ; 16(8): 2141-2148, 2020 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-32016231

RESUMEN

Poly(l-lactic acid) (PLLA) scaffolds have been used in regenerative medicine, however, they commonly suffer from low flexibility, restricting their application in the repair and reconstruction of soft tissues. In this study, poly(l-lactide-co-ε-caprolactone) (PLCL) copolymers were examined to modulate the elasticity of PLLA with the random presence of CL units in PLLA. Thermodynamic analysis revealed that the introduction of PCL could significantly decrease the melting point and glass transition temperature of PLLA, benefiting the extrusion and printing of PLCL. Diverse scaffolds with designed architectures including porous cubes with or without large holes, cambered plates with holes and round tubes could be easily constructed by 3D printing. In the process of elastic deformation, the maximum elastic stress of the copolymer scaffold was obviously increased from 19.6 to 31.5 MPa when the relative content of PCL was increased to 70%, while the elongation at break was evidently increased from 388% to about 1974%. The Young's modulus of PLCL was also significantly decreased (P < 0.05) in comparison with that of PLLA. PLCL scaffolds have good platelet and endotheliocyte adhesion ability and no obvious hemolysis was observed. In vivo subcutaneous implantation of PLCL scaffolds demonstrated superior biocompatibility. Collectively, this work highlights that copolymerization of PCL segments into PLLA is an effective approach to tune the 3D printability and the stiffness and elasticity of PLLA scaffolds. PLCL scaffolds hold great promise for the regeneration of soft tissues including but not limited to cartilage, myocardium, muscle, tendon and nervous tissues.


Asunto(s)
Poliésteres/química , Andamios del Tejido/química , Animales , Materiales Biocompatibles/química , Fenómenos Biomecánicos , Plaquetas/citología , Adhesión Celular , Proliferación Celular , Elasticidad , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Impresión Tridimensional , Conejos , Ingeniería de Tejidos/instrumentación
7.
Arterioscler Thromb Vasc Biol ; 38(7): e117-e134, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29853570

RESUMEN

OBJECTIVE: The objective of this study was to develop small-diameter vascular grafts capable of eluting SDF (stromal cell-derived factor)-1α-derived peptide and SP (substance P) for in situ vascular regeneration. APPROACH AND RESULTS: Polycaprolactone (PCL)/collagen grafts containing SP or SDF-1α-derived peptide were fabricated by electrospinning. SP and SDF-1α peptide-loaded grafts recruited significantly higher numbers of mesenchymal stem cells than that of the control group. The in vivo potential of PCL/collagen, SDF-1, and SP grafts was assessed by implanting them in a rat abdominal aorta for up to 4 weeks. All grafts remained patent as observed using color Doppler and stereomicroscope. Host cells infiltrated into the graft wall and the neointima was formed in peptides-eluting grafts. The lumen of the SP grafts was covered by the endothelial cells with cobblestone-like morphology, which were elongated in the direction of the blood flow, as discerned using scanning electron microscopy. Moreover, SDF-1α and SP grafts led to the formation of a confluent endothelium as evaluated using immunofluorescence staining with von Willebrand factor antibody. SP and SDF-1α grafts also promoted smooth muscle cell regeneration, endogenous stem cell recruitment, and blood vessel formation, which was the most prominent in the SP grafts. Evaluation of inflammatory response showed that 3 groups did not significantly differ in terms of the numbers of proinflammatory macrophages, whereas SP grafts showed significantly higher numbers of proremodeling macrophages than that of the control and SDF-1α grafts. CONCLUSIONS: SDF-1α and SP grafts can be potential candidates for in situ vascular regeneration and are worthy for future investigations.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Aorta Abdominal/cirugía , Implantación de Prótesis Vascular/instrumentación , Prótesis Vascular , Quimiocina CXCL12/farmacología , Materiales Biocompatibles Revestidos , Colágeno Tipo I/química , Neovascularización Fisiológica/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Poliésteres/química , Sustancia P/farmacología , Inductores de la Angiogénesis/química , Animales , Aorta Abdominal/diagnóstico por imagen , Aorta Abdominal/patología , Aorta Abdominal/fisiopatología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL12/química , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Neointima , Fragmentos de Péptidos/química , Diseño de Prótesis , Ratas Sprague-Dawley , Sustancia P/química , Factores de Tiempo , Ultrasonografía Doppler en Color , Grado de Desobstrucción Vascular , Remodelación Vascular
8.
Nano Lett ; 18(7): 4377-4385, 2018 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-29932335

RESUMEN

Dendritic cells (DCs) are increasingly used in cancer vaccines due to their ability to regulate T-cell immunity. Major limitations associated with the present DC adoptive transfer immunotherapy are low cell viability and transient duration of transplanted DCs at the vaccination site and the lack of recruitment of host DCs, leading to unsatisfactory T-cell immune response. Here, we developed a novel vaccine nodule comprising a simple physical mixture of the peptide nanofibrous hydrogel, anti-PD-1 antibodies, DCs, and tumor antigens. Upon subcutaneous injection, the vaccine nodule maintained the viability and biological function including the antigen uptake and maturation of encapsulated DCs and simultaneously recruited a number of host DCs and promoted the drainage of activated DCs to lymph nodes, resulting in enhanced proliferation of antigen-specific splenocytes and provoking potent cellular immune responses. Compared with adoptive transfer of DCs and subcutaneous administration of antigen vaccine, such a vaccine nodule shows superior antitumor immunotherapy efficiency in both prophylactic and therapeutic tumor models including delayed tumor growth and prolonged mice survival due to effective stimulation of antitumor T-cell immunity and increased infiltration of activated CD8+ effector T-cells in the tumor. Our findings provide a simple and robust vaccination strategy for DC-based vaccines and also a unique vaccine product for stimulating and enhancing T-cell immunity, holding great promise for immunotherapy against cancer and infectious diseases.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Neoplasias/terapia , Linfocitos T/inmunología , Vacunas contra el Cáncer/uso terapéutico , Ingeniería Celular , Células Dendríticas/citología , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/uso terapéutico , Neoplasias/inmunología , Péptidos/inmunología , Péptidos/uso terapéutico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología
9.
Mol Pharm ; 15(2): 508-518, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29323913

RESUMEN

Currently, development of subunit vaccine based on recombinant antigens or peptides has gradually become an important alternative option for traditional vaccine. However, induction of potent immune response with desired efficacy remains a major challenge. The nanoparticle-based antigen delivery system has been considered a potential carrier system to improve the efficacy of subunit vaccine. In the present study, we have designed an immune-stimulatory delivery system by conjugating three-armed PLGA to PEG via the peroxalate ester bond which is sensitive to hydrogen peroxide (H2O2), a major reactive oxygen species (ROS). Hyaluronic acid (HA), a ligand for CD44 receptors was also modified onto the outer shell of the 3s-PLGA-PEG nanoparticles to promote immune cell uptake. For in vitro and in vivo immune response assessment, a model antigen ovalbumin (OVA) was enclosed within the core of the 3s-PLGA-PEG nanoparticles to form 3s-PLGA-PO-PEG/HA nanoparticles (PHO NPs). Our results showed that the PHO NPs enhanced dendritic cell maturation, antigen uptake, and antigen presentation in vitro, likely due to enhanced lysosomal escape. In vivo experiments further revealed that the PHO nanovaccine robustly promoted OVA-specific antibody production and T cell response accompanied by modest stimulation of memory T cells. In summary, the ROS-responsive PHO NPs with modified HA may be an effective vehicle antigen delivery system to promote antigen-induced immune response.


Asunto(s)
Antígenos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Receptores de Hialuranos/metabolismo , Nanopartículas/química , Vacunas de Subunidad/inmunología , Animales , Antígenos/inmunología , Línea Celular , Células Dendríticas , Femenino , Receptores de Hialuranos/inmunología , Ácido Hialurónico/química , Peróxido de Hidrógeno/química , Inmunogenicidad Vacunal , Ratones , Ratones Endogámicos C57BL , Nanopartículas/administración & dosificación , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Poliésteres , Polietilenglicoles , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Vacunas de Subunidad/administración & dosificación
10.
Angew Chem Int Ed Engl ; 57(50): 16396-16400, 2018 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-30341792

RESUMEN

Liposomes have been used as popular drug delivery systems for cancer therapy. However, it is difficult to track traditional liposome delivery systems in an efficient and stable fashion to assess their delivery efficacy and biodistribution after administration. Meanwhile, conventional fluorescent liposomes containing optical tracers face the challenge of aggregation-caused quenching. Herein, we report a strategy for the integration of an aggregation-induced emission fluorogen with a liposome to yield an AIEgen-lipid conjugate, termed "AIEsome". The AIEsome exhibits bright red fluorescence along with great photostability and biocompatibility, and can be used for in vitro cancer cell labeling and in vivo tumor targeting. Meanwhile, benefiting from the excellent photosensitizing ability of the AIEgen and its good oxygen exposure in aqueous media, the AIEsome also performs well in efficient photodynamic therapy (PDT) for both in vitro cancer cell ablation and in vivo antitumor therapy after white light illumination.


Asunto(s)
Colorantes Fluorescentes/administración & dosificación , Lípidos/química , Liposomas/química , Neoplasias Mamarias Animales/diagnóstico por imagen , Neoplasias Mamarias Animales/tratamiento farmacológico , Fármacos Fotosensibilizantes/administración & dosificación , Animales , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Femenino , Colorantes Fluorescentes/farmacocinética , Colorantes Fluorescentes/uso terapéutico , Ratones , Imagen Óptica , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacocinética , Fármacos Fotosensibilizantes/uso terapéutico , Distribución Tisular
11.
Mol Pharm ; 14(5): 1760-1770, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28296410

RESUMEN

In this study, the photochemical internalization (PCI) technique was adopted in a nanoparticle-based antigen delivery system to enhance antigen-specific CD8+ T cell immune response for cancer immunotherapy. Pheophorbide A, a hydrophobic photosensitizer, grafted with polyethylenimine (PheoA-PEI) with endosome escape activity and near-infrared imaging capability was prepared. A model antigen ovalbumin (OVA) was then complexed with PheoA-PEI to form PheoA-PEI/OVA nanoparticles (PheoA-PEI/OVA NPs) that are responsive to light. Flow cytometry analysis revealed increased endocytosis in a murine dendritic cell line (DC2.4) that was treated with PheoA-PEI/OVA NPs compared to free OVA. Generation of reactive oxygen species (ROS) in DC2.4 cells was also confirmed quantitatively and qualitatively using 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA). Confocal laser scanning microscopy (CLSM) further demonstrated that the PheoA-PEI/OVA NPs enhanced cytosolic antigen release after light stimulation. Moreover, PheoA-PEI/OVA NP treated DC2.4 cells exhibited enhanced cross-presentation to B3Z T cell hybridoma in vitro after light irradiation, substantially increased compared to those treated with free OVA. Consistently, in vivo results revealed upregulation of CD3+CD8+T lymphocytes in tumors of mice treated with dendritic cells plus PheoA-PEI/OVA NPs and light irradiation. The activated T cell response is partly responsible for the inhibitory effect on E.G7 tumor growth in mice immunized with dendritic cells plus PheoA-PEI/OVA NPs and light irradiation. Our results demonstrate the feasibility to enhance antigen-specific CD8+ T cell immune response by light-responsive nanoparticle-based vaccine delivery for cancer immunotherapy.


Asunto(s)
Clorofila/análogos & derivados , Células Dendríticas/metabolismo , Inmunoterapia/métodos , Nanopartículas/química , Polietileneimina/química , Animales , Linfocitos T CD8-positivos/metabolismo , Línea Celular , Línea Celular Tumoral , Clorofila/química , Femenino , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Especies Reactivas de Oxígeno/metabolismo , Oxígeno Singlete/metabolismo
12.
J Am Soc Nephrol ; 27(8): 2357-69, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26869006

RESUMEN

Low cell retention and engraftment after transplantation limit the successful application of stem cell therapy for AKI. Engineered microenvironments consisting of a hydrogel matrix and growth factors have been increasingly successful in controlling stem cell fate by mimicking native stem cell niche components. Here, we synthesized a bioactive hydrogel by immobilizing the C domain peptide of IGF-1 (IGF-1C) on chitosan, and we hypothesized that this hydrogel could provide a favorable niche for adipose-derived mesenchymal stem cells (ADSCs) and thereby enhance cell survival in an AKI model. In vitro studies demonstrated that compared with no hydrogel or chitosan hydrogel only, the chitosan-IGF-1C hydrogel increased cell viability through paracrine effects. In vivo, cotransplantation of the chitosan-IGF-1C hydrogel and ADSCs in ischemic kidneys ameliorated renal function, likely by the observed promotion of stem cell survival and angiogenesis, as visualized by bioluminescence imaging and attenuation of fibrosis. In conclusion, IGF-1C immobilized on a chitosan hydrogel provides an artificial microenvironment for ADSCs and may be a promising therapeutic approach for AKI.


Asunto(s)
Lesión Renal Aguda/terapia , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Trasplante de Células Madre Mesenquimatosas , Tejido Adiposo/citología , Animales , Quitosano , Terapia Combinada , Hidrogel de Polietilenoglicol-Dimetacrilato , Ratones
13.
J Mater Sci Mater Med ; 27(1): 17, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26676863

RESUMEN

Star-shaped block copolymers based on poly(D,L-lactide-co-glycolide) (PLGA) and poly(ethylene glycol) (PEG) (st-PLGA-PEG) were synthesized with structural variation on arm numbers in order to investigate the relationship between the arm numbers of st-PLGA-PEG copolymers and their micelle properties. st-PLGA-PEG copolymers with arm numbers 3, 4 and 6 were synthesized by using different cores such as trimethylolpropane, pentaerythritol and dipentaerythritol, and were characterized by nuclear magnetic resonance and gel permeation chromatography. The critical micelle concentration decreased with increasing arm numbers in st-PLGA-PEG copolymers. The doxorubicin-loaded st-PLGA-PEG micelles were prepared by a modified nanoprecipitation method. Micellar properties such as particle size, drug loading content and in vitro drug release behavior were investigated as a function of the number of arms and compared with each other. The doxorubicin-loaded 4-arm PLGA-PEG micelles were found to have the highest cellular uptake efficiency and cytotoxicity compared with 3-arm PLGA-PEG micelles and 6-arm PLGA-PEG micelles. The results suggest that structural tailoring of arm numbers from st-PLGA-PEG copolymers could provide a new strategy for designing drug carriers of high efficiency. Structural tailoring of arm numbers from star shaped-PLGA-PEG copolymers (3-arm/4-arm/6-arm-PLGA-PEG) could provide a new strategy for designing drug carriers of high efficiency.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Portadores de Fármacos , Ácido Láctico , Micelas , Polietilenglicoles , Ácido Poliglicólico , Células HeLa , Humanos , Técnicas In Vitro , Microscopía Confocal , Copolímero de Ácido Poliláctico-Ácido Poliglicólico
14.
Nanotechnology ; 26(11): 115101, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25708980

RESUMEN

Nanoscale drug delivery platforms have been developed over the past four decades that have shown promising clinical results in several types of cancer and inflammatory disorders. These nanocarriers carrying therapeutic payloads are maximizing the therapeutic outcomes while minimizing adverse effects. Yet one of the major challenges facing drug developers is the dilemma of premature versus on-demand drug release, which influences the therapeutic regiment, efficacy and potential toxicity. Herein, we report on redox-sensitive polymer-drug conjugate micelles for on-demand intracellular delivery of a model active agent, curcumin. Biodegradable methoxy poly(ethylene glycol)-poly(lactic acid) copolymer (mPEG-PLA) was conjugated with curcumin via a disulfide bond or ester bond (control), respectively. The self-assembled redox-sensitive micelles exhibited a hydrodynamic size of 115.6 ± 5.9 (nm) with a zeta potential of -10.6 ± 0.7 (mV). The critical micelle concentration was determined at 6.7 ± 0.4 (µg mL(-1)). Under sink conditions with a mimicked redox environment (10 mM dithiothreitol), the extent of curcumin release at 48 h from disulfide bond-linked micelles was nearly three times higher compared to the control micelles. Such rapid release led to a lower half maximal inhibitory concentration (IC50) in HeLa cells at 18.5 ± 1.4 (µg mL(-1)), whereas the IC50 of control micelles was 41.0 ± 2.4 (µg mL(-1)). The cellular uptake study also revealed higher fluorescence intensity for redox-sensitive micelles. In conclusion, the redox-sensitive polymeric conjugate micelles could enhance curcumin delivery while avoiding premature release, and achieving on-demand release under the high glutathione concentration in the cell cytoplasm. This strategy opens new avenues for on-demand drug release of nanoscale intracellular delivery platforms that ultimately might be translated into pre-clinical and future clinical practice.


Asunto(s)
Portadores de Fármacos , Micelas , Nanotecnología/métodos , Materiales Biocompatibles/química , Dominio Catalítico , Supervivencia Celular , Curcumina/química , Citoplasma/metabolismo , Disulfuros/química , Glutatión/química , Células HeLa , Humanos , Hidrodinámica , Concentración de Iones de Hidrógeno , Inflamación , Concentración 50 Inhibidora , Lactatos/química , Espectroscopía de Resonancia Magnética , Microscopía Electrónica de Transmisión , Peso Molecular , Nanomedicina/métodos , Nanopartículas/química , Oxidación-Reducción , Tamaño de la Partícula , Polietilenglicoles/química , Polímeros/química , Temperatura
15.
Nanotechnology ; 26(27): 275101, 2015 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-26066389

RESUMEN

Poor aqueous solubility, potential degradation, rapid metabolism and elimination lead to low bioavailability of pleiotropic impotent curcumin. Herein, we report two types of acid-responsive polymeric micelles where curcumin was encapsulated via both covalent and non-covalent modes for enhanced loading capacity and on-demand release. Biodegradable methoxy poly(ethylene glycol)-poly(lactic acid) copolymer (mPEG-PLA) was conjugated with curcumin via a hydrazone linker, generating two conjugates differing in architecture (single-tail versus double-tail) and free curcumin was encapsulated therein. The two micelles exhibited similar hydrodynamic size at 95 ± 3 nm (single-tail) and 96 ± 3 nm (double-tail), but their loading capacities differed significantly at 15.0 ± 0.5% (w/w) (single-tail) and 4.8 ± 0.5% (w/w) (double-tail). Under acidic sink conditions (pH 5.0 and 6.0), curcumin displayed a faster release from the single-tail nanocarrier, which was correlated to a low IC50 of 14.7 ± 1.6 (µg mL(-1)) compared to the value of double-tail micelle (24.9 ± 1.3 µg mL(-1)) in HeLa cells. The confocal imaging and flow cytometry analysis demonstrated a superior capability of single-tail micelle for intracellular curcumin delivery, which was a consequence of the higher loading capacity and lower degree of mPEG surface coverage. In conclusion, the dual loading mode is an effective means to increase the drug content in the micellar nanocarriers whose delivery efficiency is highly dependent on its polymer-drug conjugate architecture. This strategy offers an alternative nanoplatform for intracellularly delivering impotent hydrophobic agents (i.e. curcumin) in an efficient stimuli-triggered way, which is valuable for the enhancement of curcumin's efficacy in managing a diverse range of disorders.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Curcumina/química , Portadores de Fármacos/química , Lactatos/química , Micelas , Nanopartículas/química , Polietilenglicoles/química , Curcumina/farmacología , Células HeLa , Humanos
16.
Biomacromolecules ; 15(8): 3128-38, 2014 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-25054812

RESUMEN

Reasonably structural design of nanoparticles (NPs) to combine functions of prolonged systemic circulation, enhanced tumor targeting and specific intracellular drug release is crucial for antitumor drug delivery. Combining advantages of Arg-Gly-Asp (RGD) for active tumor targeting, zwitterionic polycarboxybetaine methacrylate (PCB) for prolonged systemic circulation, poly(2-(diisopropylamino) ethyl methacrylate) (PDPA) for acid-triggered intracellular release, novel RGD-PCB-b-PDPA (RGD-PCD) block copolymers were prepared via reversible addition-fragmentation chain transfer (RAFT) polymerization and followed by functionalization with RGD. Doxorubicine (DOX) was encapsulated within the RGD-PCD NPs as model medicine (RGD-PCD/DOX NPs). With ultra pH-sensitivity of PDPA, the drug release was restrained at pH 7.4 for only 24% within 36 h, which was increased to 60% at pH 6.0 within 24 h, and released more rapidly at pH 5.0 for 100% within 5 h, indicating that the RGD-PCD/DOX NPs were able to turn drug release "off" at neutral pH (e.g., systemic circulation) whereas "on" under acidic conditions (e.g., inside endo/lysosomes). Furthermore, the results of fluorescence microscopy and flow cytometry analysis demonstrated improved internalization of RGD-PCD/DOX NPs in HepG2 cells via integrin-mediated endocytosis with rapid DOX release intracellularly. Consequently, the RGD-PCD/DOX NPs showed considerable cytotoxicity against HepG2 and HeLa cells in comparison with free DOX. Importantly, the RGD-PCD/DOX NPs exhibited little protein adsorption property with excellent serum stability, which led to prolonged systemic circulation and enhanced tumor accumulation in tumor-bearing nude mice. Therefore, this multifunctional RGD-PCD NPs, which represented the flexible design approach, showed great potential for the development of novel nanocarriers in tumor-targeted drug delivery.


Asunto(s)
Portadores de Fármacos/química , Liberación de Fármacos , Integrinas/química , Nanopartículas/química , Polímeros/química , Animales , Betaína/química , Doxorrubicina/farmacología , Endocitosis/efectos de los fármacos , Células HeLa , Células Hep G2 , Humanos , Concentración de Iones de Hidrógeno , Ratones Desnudos , Oligopéptidos/química , Ácidos Polimetacrílicos/química
17.
J Nanosci Nanotechnol ; 14(5): 3305-12, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24734545

RESUMEN

Polyamidoamine (PAMAM) dendrimers have been widely used as drug carriers, non-viral gene vectors and imaging agents. However, the use of dendrimers in biological system is constrained because of inherent toxicity and organ accumulation. In this study, the strategy of acetylation and PEGylation-acetylation was used to minimize PAMAM dendrimers toxicities and to improve their biodistribution and pharmacokinetics for medical application. PEGylated-acetylated PAMAM (G4-Ac-PEG) dendrimers were synthesized by PEGylation of acetylated PAMAM dendrimer of generation 4 (G4) with acetic anhydride and polyethylene glycol (PEG) 3.4 k. To investigate the cytotoxicity and in vivo biodistribution of the conjugates, in vitro cell viability analysis, Iodine-125 (125I) imaging, tissue distribution and hematoxylin-eosin (HE) staining were performed. We find that acetylation and PEGylation-acetylation essentially eliminates the inherent dendrimer cytotoxicity in vitro. Planar gamma (gamma) camera imaging revealed that all the conjugates were slowly eliminated from the body, and higher abdominal accumulation of acetylation PAMAM dendrimer was observed. Tissue distribution analysis showed that PEGylated-acetylated dendrimers have longer blood retention and lower accumulation in organs such as the kidney and liver than the non-PEGylated-acetylated dendrimers, but acetylation only can significantly increase the accumulation of G4 in the kidney and decrease the concentration in blood. Histology results reveal that no obvious damage was observed in all groups after high dose administration. This study indicates that PEGylation-acetylation could improve the blood retention, decrease organ accumulation, and improve pharmacokinetic profile, which suggests that PEGylation-acetylation provides an alternative method for PAMAM dendrimers modification.


Asunto(s)
Dendrímeros/síntesis química , Dendrímeros/farmacocinética , Polietilenglicoles/química , Acetilación , Animales , Dendrímeros/administración & dosificación , Células HEK293 , Humanos , Infusiones Intravenosas , Radioisótopos de Yodo/administración & dosificación , Masculino , Ratones , Ratones Endogámicos BALB C , Distribución Tisular
18.
Adv Healthc Mater ; 13(20): e2304675, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38688026

RESUMEN

The mitochondrial enzyme arginase-2 (Arg-2) is implicated in the pathophysiology of contrast-induced acute kidney injury (CI-AKI). Therefore, Arg-2 represents a candid target for CI-AKI prevention. Here, layer-by-layer (LbL) assembled renal-targeting polymeric nanoparticles are developed to efficiently deliver small interfering RNA (siRNA), knockdown Arg-2 expression in renal tubules, and prevention of CI-AKI is evaluated. First, near-infrared dye-loaded poly(lactic-co-glycolic acid) (PLGA) anionic cores are electrostatically coated with cationic chitosan (CS) to facilitate the adsorption and stabilization of Arg-2 siRNA. Next, nanoparticles are coated with anionic hyaluronan (HA) to provide protection against siRNA leakage and shielding against early clearance. Sequential electrostatic layering of CS and HA improves loading capacity of Arg-2 siRNA and yields LbL-assembled nanoparticles. Renal targeting and accumulation is enhanced by modifying the outermost layer of HA with a kidney targeting peptide (HA-KTP). The resultant kidney-targeting and siRNA loaded nanoparticles (PLGA/CS/HA-KTP siRNA) exhibit proprietary accumulation in kidneys and proximal tubular cells at 24 h post-tail vein injection. In iohexol-induced in vitro and in vivo CI-AKI models, PLGA/CS/HA-KTP siRNA delivery alleviates oxidative and nitrification stress, and rescues mitochondrial dysfunction while reducing apoptosis, thereby demonstrating a robust and satisfactory therapeutic effect. Thus, PLGA/CS/HA-KTP siRNA nanoparticles offer a promising candidate therapy to protect against CI-AKI.


Asunto(s)
Lesión Renal Aguda , Arginasa , Medios de Contraste , Nanopartículas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , ARN Interferente Pequeño , Nanopartículas/química , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/prevención & control , Lesión Renal Aguda/metabolismo , Animales , ARN Interferente Pequeño/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Medios de Contraste/química , Ratones , Arginasa/metabolismo , Arginasa/genética , Quitosano/química , Técnicas de Silenciamiento del Gen , Riñón/metabolismo , Riñón/efectos de los fármacos , Riñón/patología , Ácido Hialurónico/química , Masculino , Humanos , Ácido Láctico/química , Ácido Poliglicólico/química , Nanopartículas Capa por Capa
19.
ACS Nano ; 18(33): 22104-22121, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39102149

RESUMEN

Digital light processing (DLP) bioprinting, known for its high resolution and speed, enables the precise spatial arrangement of biomaterials and has become integral to advancing tissue engineering and regenerative medicine. Nevertheless, inherent light scattering presents significant challenges to the fidelity of the manufactured structures. Herein, we introduce a photoinhibition strategy based on Rutin nanoparticles (Rnps), attenuating the scattering effect through concurrent photoabsorption and free radical reaction. Compared to the widely utilized biocompatible photoabsorber tartrazine (Tar), Rnps-infused bioink enhanced printing speed (1.9×), interlayer homogeneity (58% less overexposure), resolution (38.3% improvement), and print tolerance (3× high-precision range) to minimize trial-and-error. The biocompatible and antioxidative Rnps significantly improved cytocompatibility and exhibited resistance to oxidative stress-induced damage in printed constructs, as demonstrated with human induced pluripotent stem cell-derived endothelial cells (hiPSC-ECs). The related properties of Rnps facilitate the facile fabrication of multimaterial, heterogeneous, and cell-laden biomimetic constructs with intricate structures. The developed photoinhibitor, with its profound adaptability, promises wide biomedical applications tailored to specific biological requirements.


Asunto(s)
Bioimpresión , Luz , Nanopartículas , Rutina , Humanos , Rutina/química , Rutina/farmacología , Nanopartículas/química , Ingeniería de Tejidos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos
20.
Nat Commun ; 15(1): 1377, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38355941

RESUMEN

Injectable biomaterials have garnered increasing attention for their potential and beneficial applications in minimally invasive surgical procedures and tissue regeneration. Extracellular matrix (ECM) hydrogels and porous synthetic polymer microspheres can be prepared for injectable administration to achieve in situ tissue regeneration. However, the rapid degradation of ECM hydrogels and the poor injectability and biological inertness of most polymeric microspheres limit their pro-regenerative capabilities. Here, we develop a biomaterial system consisting of elastic porous poly(l-lactide-co-ε-caprolactone) (PLCL) microspheres mixed with ECM hydrogels as injectable composites with interleukin-4 (IL-4) and insulin-like growth factor-1 (IGF-1) dual-release functionality. The developed multifunctional composites have favorable injectability and biocompatibility, and regulate the behavior of macrophages and myogenic cells following injection into muscle tissue. The elicited promotive effects on tissue regeneration are evidenced by enhanced neomusle formation, vascularization, and neuralization at 2-months post-implantation in a male rat model of volumetric muscle loss. Our developed system provides a promising strategy for engineering bioactive injectable composites that demonstrates desirable properties for clinical use and holds translational potential for application as a minimally invasive and pro-regenerative implant material in multiple types of surgical procedures.


Asunto(s)
Materiales Biocompatibles , Matriz Extracelular , Masculino , Ratas , Animales , Porosidad , Microesferas , Hidrogeles , Ingeniería de Tejidos/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA