Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biomed Microdevices ; 21(2): 38, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30937546

RESUMEN

Presently, cardiovascular interventions such as stent deployment and balloon angioplasty are performed under x-ray guidance. However, x-ray fluoroscopy has poor soft tissue contrast and is limited by imaging in a single plane, resulting in imprecise navigation of endovascular instruments. Moreover, x-ray fluoroscopy exposes patients to ionizing radiation and iodinated contrast agents. Magnetic resonance imaging (MRI) is a safe and enabling modality for cardiovascular interventions. Interventional cardiovascular MR (iCMR) is a promising approach that is in stark contrast with x-ray fluoroscopy, offering high-resolution anatomic and physiologic information and imaging in multiple planes for enhanced navigational accuracy of catheter-based devices, all in an environment free of radiation and its deleterious effects. While iCMR has immense potential, its translation into the clinical arena is hindered by the limited availability of MRI-visible catheters, wire guides, angioplasty balloons, and stents. Herein, we aimed to create application-specific, devices suitable for iCMR, and demonstrate the potential of iCMR by performing cardiovascular catheterization procedures using these devices. Tools, including catheters, wire guides, stents, and angioplasty balloons, for endovascular interventions were functionalized with a polymer coating consisting of poly(lactide-co-glycolide) (PLGA) and superparamagnetic iron oxide (SPIO) nanoparticles, followed by endovascular deployment in the pig. Findings from this study highlight the ability to image and properly navigate SPIO-functionalized devices, enabling interventions such as successful stent deployment under MRI guidance. This study demonstrates proof-of-concept for rapid prototyping of iCMR-specific endovascular interventional devices that can take advantage of the capabilities of iCMR.


Asunto(s)
Procedimientos Endovasculares/instrumentación , Imagen por Resonancia Magnética Intervencional/instrumentación , Nanopartículas de Magnetita/química , Animales , Catéteres , Femenino , Procesamiento de Imagen Asistido por Computador , Masculino , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Porcinos
2.
Mol Pharm ; 15(5): 1814-1825, 2018 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-29537266

RESUMEN

Selective drug accumulation in the malignant tissue is a prerequisite for effective cancer treatment. However, most drug molecules and their formulated particles are blocked en route to the destiny tissue due to the existence of multiple biological and physical barriers including the tumor microvessel endothelium. Since the endothelial cells on the surface of the microvessel wall can be modulated by inflammatory cytokines and chemokines secreted by the tumor or stromal cells, an effective drug delivery approach is to enhance interaction between the drug particles and the unique spectrum of surface proteins on the tumor endothelium. In this study, we performed in vivo screening for thioaptamers that bind to the bone marrow endothelium with specificity in a murine model of lymphoma with bone marrow involvement (BMI). The R1 thioaptamer was isolated based on its high homing potency to bones with BMI, and 40-60% less efficiency in accumulation to healthy bones. In cell culture, R1 binds to human umbilical vein endothelial cells (HUVEC) with a high affinity ( Kd ≈ 3 nM), and the binding affinity can be further enhanced when cells were treated with a mixture of lymphoma cell and bone marrow cell conditioned media. Cellular uptake of R1 is through clathrin-mediated endocytosis. Conjugating R1 on to the surface of liposomal doxorubicin nanoparticles resulted in 2-3-fold increase in drug accumulation in lymphoma BMI. Taking together, we have successfully identified a thioaptamer that preferentially binds to the endothelium of lymphoma BMI. It can serve as an affinity moiety for targeted delivery of drug particles to the disease organ.


Asunto(s)
Aptámeros de Nucleótidos/farmacología , Células de la Médula Ósea/efectos de los fármacos , Médula Ósea/efectos de los fármacos , ADN/administración & dosificación , Linfoma/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Animales , Línea Celular , Línea Celular Tumoral , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos/métodos , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones SCID , Polietilenglicoles/farmacología
3.
Pharmacol Res ; 113(Pt A): 92-99, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27546164

RESUMEN

Various nanodelivery systems have been designed to release therapeutic agents upon contact with specific enzymes. However, enzyme-triggered release typically takes place in the tissue interstitium, thereby resulting in the extracellular delivery of drugs. Here, we have designed an enzyme-stimulated multistage vector (ESMSV), which enables stimulus-triggered release of drug-encapsulated nanoparticles from a microparticle. Specifically, polymeric nanoparticles with a surface matrix metalloproteinase-2 (MMP2) peptide substrate were conjugated to the surface of porous silicon microparticles. In the presence of MMP2, the polymeric nanoparticles were released into the tumor interstitium. This platform can be used to attain triggered drug release, while simultaneously facilitating the cellular internalization of drugs. The results indicate that nanoparticle release was MMP2-specific and resulted in improved intracellular uptake of hydrophobic agents in the presence of MMP2. Furthermore, in a mouse model of melanoma lung metastasis, systemic delivery of ESMSVs caused a substantial increase in intracellular accumulation of agents in cancer cells in comparison to delivery with non-stimulus-responsive particles.


Asunto(s)
Portadores de Fármacos/química , Enzimas/química , Neoplasias/tratamiento farmacológico , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/química , Animales , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos/métodos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Desnudos , Nanopartículas/administración & dosificación , Nanopartículas/química , Tamaño de la Partícula , Polietilenglicoles/química , Polímeros/química
4.
Small ; 10(19): 3943-53, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24867543

RESUMEN

The ideal scaffold for regenerative medicine should concurrently mimic the structure of the original tissue from the nano- up to the macroscale and recapitulate the biochemical composition of the extracellular matrix (ECM) in space and time. In this study, a multiscale approach is followed to selectively integrate different types of nanostructured composite microspheres loaded with reporter proteins, in a multi-compartment collagen scaffold. Through the preservation of the structural cues of the functionalized collagen scaffold at the nano- and microscale, its macroscopic features (pore size, porosity, and swelling) are not altered. Additionally, the spatial confinement of the microspheres allows the release of the reporter proteins in each of the layers of the scaffold. Finally, the staged and zero-order release kinetics enables the temporal biochemical patterning of the scaffold. The versatile manufacturing of each component of the scaffold results in the ability to customize it to better mimic the architecture and composition of the tissues and biological systems.


Asunto(s)
Materiales Biocompatibles/química , Biomimética , Microesferas , Colágeno/química , Matriz Extracelular/metabolismo , Genes Reporteros , Humanos , Cinética , Ácido Láctico/química , Microscopía Electrónica de Rastreo , Microscopía Fluorescente , Nanoestructuras/química , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Porosidad , Silicio/química , Andamios del Tejido/química
5.
Nanomedicine ; 10(5): 879-88, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24566273

RESUMEN

Endogenous peptides that represent biological and pathological information of disease have attracted interest for diagnosis. However, the extraction of those low abundance peptides is still a challenge because of the complexity of human bodily fluids (HBF). Hepcidin, a peptide hormone, has been recognized as a biomarker for iron-related diseases. There is no rapid and reliable way to enrich them from HBF. Here we describe a peptide extraction approach based on nanoporous silica thin films to successfully detect hepcidin from HBF. Cooperative functions of nanopore to biomolecule, including capillary adsorption, size-exclusion and electrostatic interaction, were systematically investigated to immobilize the target peptide. To promote this new approach to clinical practices, we further applied it to successfully assay the hepcidin levels in HBF provided by healthy volunteers and patients suffering from inflammation. Our finding provides a high-throughput, rapid, label-free and cost-effective detection method for capturing and quantifying low abundance peptides from HBF. FROM THE CLINICAL EDITOR: Diagnosing diseases with low concentration peptide biomarkers remains challenging. This team of authors describes a peptide extraction approach based on nanoporous silica thin films to successfully detect low concentrations of hepcidin from human body fluids collected from 119 healthy volunteers and 19 inflammation patients.


Asunto(s)
Biomarcadores/análisis , Líquidos Corporales/química , Hepcidinas/análisis , Nanoporos , Humanos , Membranas Artificiales , Péptidos/química , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
6.
J Microencapsul ; 31(5): 501-7, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24654943

RESUMEN

CONTEXT: Celastrol, a natural compound derived from the herb Tripterygium wilfordii, is known to have anticancer activity, but is not soluble in water. OBJECTIVE: Formation of celastrol liposomes, to avoid the use of toxic solubilising agents. MATERIALS AND METHODS: Two different formulations of PEGylated celastrol liposomes were fabricated. Liposomal characteristics and serum stability were determined using dynamic light scattering. Drug entrapment efficacy and drug release were measured spectrophotometrically. Cellular internalisation and anticancer activity was measured in prostate cancer cells. RESULTS: Liposomal celastrol displayed efficient serum stability, cellular internalisation and anticancer activity, comparable to that of the free drug reconstituted in dimethyl sulfoxide. DISCUSSION AND CONCLUSION: Liposomal celastrol can decrease the viability of prostate cancer cells, while eliminating the need for toxic solubilising agents.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Tripterygium/química , Triterpenos/administración & dosificación , Triterpenos/farmacología , Antineoplásicos Fitogénicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Liposomas , Masculino , Triterpenos Pentacíclicos , Próstata/citología , Próstata/efectos de los fármacos , Próstata/patología , Neoplasias de la Próstata/patología , Triterpenos/química
7.
Anal Bioanal Chem ; 405(5): 1547-57, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23090650

RESUMEN

Novel drug delivery systems capable of continuous sustained release of therapeutics have been studied extensively for use in the prevention and management of chronic diseases. The use of these systems holds promise as a means to achieve higher patient compliance while improving therapeutic index and reducing systemic toxicity. In this work, an implantable nanochannel drug delivery system (nDS) is characterized and evaluated for the long-term sustained release of atorvastatin (ATS) and trans-resveratrol (t-RES), compounds with a proven role in managing atherogenic dyslipidemia and promoting cardioprotection. The primary mediators of drug release in the nDS are nanofluidic membranes with hundreds of thousands of nanochannels (up to 100,000/mm(2)) that attain zero-order release kinetics by exploiting nanoconfinement and molecule-to-surface interactions that dominate diffusive transport at the nanoscale. These membranes were characterized using gas flow analysis, acetone diffusion, and scanning and transmission electron microscopy (SEM, TEM). The surface properties of the dielectric materials lining the nanochannels, SiO(2) and low-stress silicon nitride, were further investigated using surface charge analysis. Continuous, sustained in vitro release for both ATS and t-RES was established for durations exceeding 1 month. Finally, the influence of the membranes on cell viability was assessed using human microvascular endothelial cells. Morphology changes and adhesion to the surface were analyzed using SEM, while an MTT proliferation assay was used to determine the cell viability. The nanochannel delivery approach, here demonstrated in vitro, not only possesses all requirements for large-scale high-yield industrial fabrication, but also presents the key components for a rapid clinical translation as an implantable delivery system for the sustained administration of cardioprotectants.


Asunto(s)
Anticolesterolemiantes/administración & dosificación , Sistemas de Liberación de Medicamentos/instrumentación , Implantes de Medicamentos/química , Ácidos Heptanoicos/administración & dosificación , Membranas Artificiales , Pirroles/administración & dosificación , Estilbenos/administración & dosificación , Vasodilatadores/administración & dosificación , Atorvastatina , Línea Celular , Supervivencia Celular , Difusión , Diseño de Equipo , Humanos , Nanoestructuras/química , Resveratrol
8.
Aviat Space Environ Med ; 83(11): 1025-36, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23156089

RESUMEN

One purpose of the International Space Station (ISS) is to explore powerful new areas of biomedical science in microgravity. Recent advances in nanotechnology applied to medicine--what we now refer to as nano-medicine--and regenerative medicine have enormous untapped potential for future space and terrestrial medical applications. Novel means for drug delivery and nanoscale screening tools will one day benefit astronauts venturing to Mars and places beyond, while the space laboratory will foster advances in nanotechnologies for diagnostic and therapeutic tools to help our patients here on Earth. Herein we review a series of nanotechnologies and selected regenerative medical approaches and highlight key areas of ongoing and future investigation that will benefit both space and terrestrial medicine. These studies target significant areas of human disease such as osteoporosis, diabetes, radiation injury, and many others.


Asunto(s)
Medicina Aeroespacial , Nanotecnología , Medicina Regenerativa , Sistemas de Liberación de Medicamentos , Corazón , Humanos , Espectrometría de Masas , Membranas Artificiales , Nanoestructuras , Proteínas/química , Proteómica , Andamios del Tejido
9.
Cancer Sci ; 102(7): 1247-52, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21447010

RESUMEN

Cancer is a leading cause of morbidity and mortality worldwide, with recent advancements resulting in modest impacts on patient survival. Nanomedicine represents an innovative field with immense potential for improving cancer treatment, having ushered in several established drug delivery platforms. Nanoconstructs such as liposomes are widely used in clinics, while polymer micelles are in advanced phases of clinical trials in several countries. Currently, the field of nanomedicine is generating a new wave of nanoscale drug delivery strategies, embracing trends that involve the functionalization of these constructs with moieties that enhance site-specific delivery and tailored release. Herein, we discuss several advancements in established nanoparticle technologies such as liposomes, polymer micelles, and dendrimers regarding tumor targeting and controlled release strategies, which are being incorporated into their design with the hope of generating a more robust and efficacious nanotherapeutic modality. We also highlight a novel strategy known as multistage drug delivery; a rationally designed nanocarrier aimed at overcoming numerous biological barriers involved in drug delivery through the decoupling of various tasks that comprise the journey from the moment of systemic administration to arrival at the tumor site.


Asunto(s)
Antineoplásicos/administración & dosificación , Nanomedicina , Neoplasias/tratamiento farmacológico , Animales , Preparaciones de Acción Retardada , Dendrímeros , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Humanos , Liposomas , Micelas , Nanopartículas
10.
Anal Chem ; 83(8): 3096-103, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21434670

RESUMEN

The lack of a viable theory for describing diffusivity when fluids are confined at the micro- and nanoscale [Ladero et al. Chem. Eng. Sci.2007, 62, 666-678; Deen AIChE J.1987, 33, 1409-1425] has necessitated accurate measurement of diffusivity (D) [Jin and Chen Chromatographia2000, 52, 17-21; Nie et al. Science1994, 266, 1018-1021; Durand et al. Anal. Chem.2009, 81, 5407-5412], crucial for a host of micro- and nanofluidic technologies [Grattoni et al. Curr. Pharm. Biotechnol.2010, 11, 343-365]. We demonstrate a rapid and agile method for the direct measurement of diffusivity in a system possessing 10(4) to 10(5) precisely fabricated channels with characteristic sizes (ß) ranging from micro- to nanometers. Custom chambers allowed us to measure the diffusivity in a closed unperturbed system using UV/vis spectroscopy. D was measured for rhodamine B (RhoB) in aqueous solution in channels of 200 and 1 µm, as well as 13 and 5.7 nm. The observed logarithmic scaling of diffusivity with ß, in close agreement with prior experiments, but far from theoretical prediction, surprisingly highlights that diffusivity is significantly altered even at the microscale. Accurate measurement of D by reducing the size of the source reservoir by 3 orders of magnitude (from 150 µL to 910 nL) proves that a substantial reduction in measurement time (from 7 days to 40 min) can be achieved. Our design thus is ready for rapid translation into a standard analytical tool--useful for multiple applications.


Asunto(s)
Técnicas Analíticas Microfluídicas/métodos , Nanotecnología/métodos , Rodaminas/análisis , Difusión , Diseño de Equipo , Membranas Artificiales , Técnicas Analíticas Microfluídicas/instrumentación , Nanoestructuras/química , Nanotecnología/instrumentación , Tamaño de la Partícula , Propiedades de Superficie
11.
Pharm Res ; 28(7): 1520-30, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21607779

RESUMEN

PURPOSE: Protein therapeutics often require repeated administrations of drug over a long period of time. Protein instability is a major obstacle to the development of systems for their controlled and sustained release. We describe a surface modification of nanoporous silicon particles (NSP) with an agarose hydrogel matrix that enhances their ability to load and release proteins, influencing intracellular delivery and preserving molecular stability. METHODS: We developed and characterized an agarose surface modification of NSP. Stability of the released protein after enzymatic treatment of loaded particles was evaluated with SDS-page and HPLC analysis. FITC-conjugated BSA was chosen as probe protein and intracellular delivery evaluated by fluorescence microscopy. RESULTS: We showed that agarose coating does not affect NSP protein release rate, while fewer digestion products were found in the released solution after all the enzymatic treatments. Confocal images show that the hydrogel coating improves intracellular delivery, specifically within the nucleus, without affecting the internalization process. CONCLUSIONS: This modification of porous silicon adds to its tunability, biocompatibility, and biodegradability the ability to preserve protein integrity during delivery without affecting release rates and internalization dynamics. Moreover, it may allow the silicon particles to function as protein carriers that enable control of cell function.


Asunto(s)
Preparaciones de Acción Retardada , Estabilidad de Medicamentos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Nanopartículas/química , Sefarosa/química , Células Cultivadas , Electroforesis en Gel de Poliacrilamida , Humanos , Microscopía Confocal , Porosidad , Silicio/química , Propiedades de Superficie
12.
Pharm Res ; 28(2): 292-300, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20593302

RESUMEN

PURPOSE: The purpose of this study is to demonstrate the long-term, controlled, zero-order release of low- and high-molecular weight chemotherapeutics through nanochannel membranes by exploiting the molecule-to-surface interactions presented by nanoconfinement. METHODS: Silicon membranes were produced with nanochannels of 5, 13 and 20 nm using standardized industrial microfabrication techniques. The study of the diffusion kinetics of interferon α-2b and leuprolide was performed by employing UV diffusion chambers. The released amount in the sink reservoir was monitored by UV absorbance. RESULTS: Continuous zero-order release was demonstrated for interferon α-2b and leuprolide at release rates of 20 and 100 µg/day, respectively. The release rates exhibited by these membranes were verified to be in ranges suitable for human therapeutic applications. CONCLUSIONS: Our membranes potentially represent a viable nanotechnological approach for the controlled administration of chemotherapeutics intended to improve the therapeutic efficacy of treatment and reduce many of the side effects associated with conventional drug administration.


Asunto(s)
Sistemas de Liberación de Medicamentos/instrumentación , Nanoestructuras/química , Nanotecnología/instrumentación , Nanotecnología/métodos , Antineoplásicos/administración & dosificación , Preparaciones de Acción Retardada/administración & dosificación , Difusión , Humanos , Interferón alfa-2 , Interferón-alfa/administración & dosificación , Leuprolida/administración & dosificación , Membranas Artificiales , Neoplasias/tratamiento farmacológico , Tamaño de la Partícula , Proteínas Recombinantes
13.
Biomaterials ; 271: 120719, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33652266

RESUMEN

Carbon fibers reinforced polymers (CFRPs) are prolifically finding applications in the medical field, moving beyond the aerospace and automotive industries. Owing to its high strength-to-weight ratio, lightness and radiolucency, CFRP-based materials are emerging to replace traditional metal-based medical implants. Numerous types of polymers matrices can be incorporated with carbon fiber using various manufacturing methods, creating composites with distinct properties. Thus, prior to biomedical application, comprehensive evaluation of material properties, biocompatibility and safety are of paramount importance. In this study, we systematically evaluated a series of novel CFRPs, aiming at analyzing biocompatibility for future development into medical implants or implantable drug delivery systems. These CFRPs were produced either via Carbon Fiber-Sheet Molding Compound or Fused Deposition Modelling-based additive manufacturing. Unlike conventional methods, both fabrication processes afford high production rates in a time-and cost-effective manner. Importantly, they offer rapid prototyping and customization in view of personalized medical devices. Here, we investigate the physicochemical and surface properties, material mutagenicity or cytotoxicity of 20 CFRPs, inclusive of 2 surface finishes, as well as acute and sub-chronic toxicity in mice and rabbits, respectively. We demonstrate that despite moderate in vitro physicochemical and surface changes over time, most of the CFRPs were non-mutagenic and non-cytotoxic, as well as biocompatible in small animal models. Future work will entail extensive material assessment in the context of orthopedic applications such as evaluating potential for osseointegration, and a chronic toxicity study in a larger animal model, pigs.


Asunto(s)
Materiales Biocompatibles , Polímeros , Animales , Materiales Biocompatibles/toxicidad , Carbono , Fibra de Carbono , Ratones , Oseointegración , Prótesis e Implantes , Conejos , Porcinos
14.
Lab Chip ; 10(22): 3074-83, 2010 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-20697650

RESUMEN

This manuscript demonstrates a mechanically robust implantable nanofluidic membrane capable of tunable long-term zero-order release of therapeutic agents in ranges relevant for clinical applications. The membrane, with nanochannels as small as 5 nm, allows for the independent control of both dosage and mechanical strength through the integration of high-density short nanochannels parallel to the membrane surface with perpendicular micro- and macrochannels for interfacing with the ambient solutions. These nanofluidic membranes are created using precision silicon fabrication techniques on silicon-on-insulator substrates enabling exquisite control over the monodispersed nanochannel dimensions and surface roughness. Zero-order release of analytes is achieved by exploiting molecule to surface interactions which dominate diffusive transport when fluids are confined to the nanoscale. In this study we investigate the nanofluidic membrane performance using custom diffusion and gas testing apparatuses to quantify molecular release rate and process uniformity as well as mechanical strength using a gas based burst test. The kinetics of the constrained zero-order release is probed with molecules presenting a range of sizes, charge states, and structural conformations. Finally, an optimal ratio of the molecular hydrodynamic diameter to the nanochannel dimension is determined to assure zero-order release for each tested molecule.


Asunto(s)
Sistemas de Liberación de Medicamentos/instrumentación , Membranas Artificiales , Técnicas Analíticas Microfluídicas/instrumentación , Nanoestructuras/química , Nanotecnología/instrumentación , Animales , Bovinos , Dextranos , Difusión , Fluoresceína-5-Isotiocianato/análogos & derivados , Glucosa , Técnicas Analíticas Microfluídicas/métodos , Microscopía de Fuerza Atómica , Microscopía Electrónica de Rastreo , Modelos Químicos , Nanotecnología/métodos , Tamaño de la Partícula , Albúmina Sérica Bovina , Propiedades de Superficie
15.
Nanotechnology ; 21(41): 415302, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20834118

RESUMEN

We demonstrate porous silica surface modification, combined with microcontact printing, as an effective method for enhanced protein patterning and adsorption on arbitrary surfaces. Compared to conventional chemical treatments, this approach offers scalability and long-term device stability without requiring complex chemical activation. Two chemical surface treatments using functionalization with the commonly used 3-aminopropyltriethoxysilane (APTES) and glutaraldehyde (GA) were compared with the nanoporous silica surface on the basis of protein adsorption. The deposited thickness and uniformity of porous silica films were evaluated for fluorescein isothiocyanate (FITC)-labeled rabbit immunoglobulin G (R-IgG) protein printed onto the substrates via patterned polydimethlysiloxane (PDMS) stamps. A more complete transfer of proteins was observed on porous silica substrates compared to chemically functionalized substrates. A comparison of different pore sizes (4-6 nm) and porous silica thicknesses (96-200 nm) indicates that porous silica with 4 nm diameter, 57% porosity and a thickness of 96 nm provided a suitable environment for complete transfer of R-IgG proteins. Both fluorescence microscopy and atomic force microscopy (AFM) were used for protein layer characterizations. A porous silica layer is biocompatible, providing a favorable transfer medium with minimal damage to the proteins. A patterned immunoassay microchip was developed to demonstrate the retained protein function after printing on nanoporous surfaces, which enables printable and robust immunoassay detection for point-of-care applications.


Asunto(s)
Inmunoglobulina G/metabolismo , Nanoporos , Nanotecnología/métodos , Dióxido de Silicio/química , Adsorción , Animales , Fluoresceína-5-Isotiocianato , Inmunoensayo , Microscopía de Fuerza Atómica , Nanoporos/ultraestructura , Nitrógeno/química , Tamaño de la Partícula , Poloxámero/química , Conejos , Propiedades de Superficie , Difracción de Rayos X
16.
Eur J Pharm Biopharm ; 151: 61-72, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32283213

RESUMEN

Inflammatory bowel disease (IBD) affects a confined area of the intestine and, therefore, administration of drugs via oral route is preferable. However, obstacles such as changes in the pH along gastrointestinal tract (GIT), enzymatic activity, and intraluminal pressure may cause low drug availability in the target tissue when delivered orally. Previous studies have pointed out the benefits of using micron-sized particles for targeting inflamed intestinal mucosa and nanoparticles for delivery of anti-inflammatory agents to the affected epithelial cells. We hypothesized that by combining the benefits of micro- and nano- particles, we could create a more efficient delivery system for budesonide, a glucocorticosteroid commonly used for anti-inflammatory IBD therapy. The aim of this study was to develop a novel multistage system for oral delivery designed to increase concentrations budesonidein the inflamed intestinal tissue. The multistage system consists of Stage 1 mesoporous silicon microparticles (S1MP) loaded with stage 2 poly-lactic-glycolic acid (PLGA) budesonide-encapsulating nanoparticles (BNP). BNP were efficiently loaded into S1MP (loading efficiency of 45.9 ± 14.8%) due to the large pore volume and high surface area of S1MP and exhibited controlled release profiles with enhanced drug dissolution rate in biologically relevant pHs. Due to the robustness in acidic pH and their geometry, S1MP protected the loaded budesonide in the acidic (gastric) pH with only 20% release. This allowed for the prolonged release of the BNP in the higher pH conditions (intestinal pH). The sustained release of BNP could facilitate accumulation in the inflamed tissue, enabling BNP to penetrate inflamed mucosa and release active budesonide to the target site. The multistage systems of S1MP and BNP were further evaluated in three-dimensional (3D) in vitro model of IBD and were found to (1) increase accumulation of BNP in the inflamed areas, (2) restore the barrier function of Caco-2 inflamed monolayer, and (3) significantly reduce pro-inflammatory cytokine release almost to the level of the healthy control.


Asunto(s)
Budesonida/química , Budesonida/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Silicio/química , Antiinflamatorios/química , Antiinflamatorios/farmacología , Células CACO-2 , Línea Celular Tumoral , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacología , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos/fisiología , Humanos , Concentración de Iones de Hidrógeno , Inflamación/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Mucosa Intestinal/metabolismo , Nanopartículas/química , Tamaño de la Partícula , Solubilidad
17.
Angew Chem Int Ed Engl ; 48(5): 872-97, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19142939

RESUMEN

The application of nanotechnology concepts to medicine joins two large cross-disciplinary fields with an unprecedented societal and economical potential arising from the natural combination of specific achievements in the respective fields. The common basis evolves from the molecular-scale properties relevant to the two fields. Local probes and molecular imaging techniques allow surface and interface properties to be characterized on a nanometer scale at predefined locations, while chemical approaches offer the opportunity to elaborate and address surfaces, for example, for targeted drug delivery, enhanced biocompatibility, and neuroprosthetic purposes. However, concerns arise in this cross-disciplinary area about toxicological aspects and ethical implications. This Review gives an overview of selected recent developments and applications of nanomedicine.


Asunto(s)
Nanomedicina , Animales , Materiales Biocompatibles/efectos adversos , Materiales Biocompatibles/química , Usos Diagnósticos de Compuestos Químicos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Humanos , Nanomedicina/métodos , Nanopartículas/efectos adversos , Nanopartículas/química , Preparaciones Farmacéuticas/química , Propiedades de Superficie
18.
Theranostics ; 8(1): 31-44, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29290791

RESUMEN

Aptamers have the potential to be used as targeting ligands for cancer treatment as they form unique spatial structures. Methods: In this study, a DNA aptamer (T1) that accumulates in the tumor microenvironment was identified through in vivo selection and validation in breast cancer models. The use of T1 as a targeting ligand was evaluated by conjugating the aptamer to liposomal doxorubicin. Results: T1 exhibited a high affinity for both tumor cells and polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). Treatment with T1 targeted doxorubicin liposomes triggered apoptosis of breast cancer cells and PMN-MDSCs. Suppression of PMN-MDSCs, which serve an immunosuppressive function, leads to increased intratumoral infiltration of cytotoxic T cells. Conclusion: The cytotoxic and immunomodulatory effects of T1-liposomes resulted in superior therapeutic efficacy compared to treatment with untargeted liposomes, highlighting the promise of T1 as a targeting ligand in cancer therapy.


Asunto(s)
Aptámeros de Nucleótidos/metabolismo , Doxorrubicina/análogos & derivados , Células Supresoras de Origen Mieloide/metabolismo , Células A549 , Animales , Antígeno CD11b/metabolismo , Línea Celular Tumoral , Doxorrubicina/química , Doxorrubicina/farmacología , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Supresoras de Origen Mieloide/efectos de los fármacos , Polietilenglicoles/química , Polietilenglicoles/farmacología , Receptores de Quimiocina/metabolismo
19.
Curr Opin Chem Biol ; 10(1): 11-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16418011

RESUMEN

Nanotechnology-based platforms for the high-throughput, multiplexed detection of proteins and nucleic acids in heretofore unattainable abundance ranges promise to bring substantial advances in molecular medicine. The emerging approaches reviewed in this article, with reference to their diagnostic potential, include nanotextured surfaces for proteomics, a two-particle sandwich assay for the biological amplification of low-concentration biomolecular signals, and silicon-based nanostructures for the transduction of molecular binding into electrical and mechanical signals, respectively.


Asunto(s)
Biopolímeros/química , Técnicas y Procedimientos Diagnósticos/instrumentación , Nanotecnología/métodos , Humanos , Nanoestructuras/química , Nanotecnología/instrumentación , Análisis por Matrices de Proteínas/instrumentación , Análisis por Matrices de Proteínas/métodos , Sensibilidad y Especificidad , Silicio/química , Propiedades de Superficie
20.
Biomaterials ; 28(3): 550-8, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16987550

RESUMEN

Reverse phase protein microarrays (RPMA) enable high throughput screening of posttranslational modifications of important signaling proteins within diseased cells. One limitation of protein-based molecular profiling is the lack of a PCR-like intrinsic amplification system for proteins. Enhancement of protein microarray sensitivities is an important goal, especially because many molecular targets within patient tissues are of low abundance. The ideal array substrate will have a high protein-binding affinity and low intrinsic signal. To date, nitrocellulose-coated glass has provided an effective substrate for protein binding in the microarray format when using chromogenic detection systems. As fluorescent systems, such as quantum dots, are explored as potential reporter agents, the intrinsic fluorescent properties of nitrocellulose-coated glass slides limit the ability to image microarrays for extended periods of time where increases in net sensitivity can be attained. Silicon, with low intrinsic autofluorescence, is being explored as a potential microarray surface. Native silicon has low binding potential. Through titrated reactive ion etching (RIE), varying surface areas have been created on silicon in order to enhance protein binding. Further, via chemical modification, reactive groups have been added to the surfaces for comparison of relative protein binding. Using this combinatorial method of surface roughening and surface coating, 3-aminopropyltriethoxysilane (APTES) and mercaptopropyltrimethoxysilane (MPTMS) treatments were shown to transform native silicon into a protein-binding substrate comparable to nitrocellulose.


Asunto(s)
Análisis por Matrices de Proteínas/instrumentación , Análisis por Matrices de Proteínas/métodos , Silicio/química , Adsorción , Albúminas/química , Animales , Biotinilación , Colodión/química , Humanos , Iones , Compuestos de Organosilicio , Propilaminas , Proteínas/química , Silanos/química , Especificidad por Sustrato , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA