Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Small ; 12(11): 1479-88, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26797709

RESUMEN

Scaffolds functionalized with delivery systems for the release of growth factors is a robust strategy to enhance tissue regeneration. However, after implantation, macrophages infiltrate the scaffold, eventually initiating the degradation and clearance of the delivery systems. Herein, it is hypothesized that fully embedding the poly(d,l-lactide-co-glycolide acid) microspheres (MS) in a highly structured collagen-based scaffold (concealing) can prevent their detection, preserving the integrity of the payload. Confocal laser microscopy reveals that non-embedded MS are easily internalized; when concealed, J774 and bone marrow-derived macrophages (BMDM) cannot detect them. This is further demonstrated by flow cytometry, as a tenfold decrease is found in the number of MS engulfed by the cells, suggesting that collagen can cloak the MS. This correlates with the amount of nitric oxide and tumor necrosis factor-α produced by J774 and BMDM in response to the concealed MS, comparable to that found for non-functionalized collagen scaffolds. Finally, the release kinetics of a reporter protein is preserved in the presence of macrophages, only when MS are concealed. The data provide detailed strategies for fabricating three dimensional (3D) biomimetic scaffolds able to conceal delivery systems and preserve the therapeutic molecules for release.


Asunto(s)
Materiales Biomiméticos/química , Ácido Láctico/química , Macrófagos/metabolismo , Microesferas , Ácido Poliglicólico/química , Andamios del Tejido/química , Adsorción , Animales , Endocitosis , Genes Reporteros , Mediadores de Inflamación/metabolismo , Cinética , Macrófagos/ultraestructura , Ratones , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Transducción de Señal
2.
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
3.
Sci Rep ; 11(1): 5107, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33658580

RESUMEN

In the field of oncology research, a deeper understanding of tumor biology has shed light on the role of environmental conditions surrounding cancer cells. In this regard, targeting the tumor microenvironment has recently emerged as a new way to access this disease. In this work, a novel extracellular matrix (ECM)-targeting nanotherapeutic was engineered using a lipid-based nanoparticle chemically linked to an inhibitor of the ECM-related enzyme, lysyl oxidase 1 (LOX), that inhibits the crosslinking of elastin and collagen fibers. We demonstrated that, when the conjugated vesicles were loaded with the chemotherapeutic epirubicin, superior inhibition of triple negative breast cancer (TNBC) cell growth was observed both in vitro and in vivo. Moreover, in vivo results displayed prolonged survival, minimal cytotoxicity, and enhanced biocompatibility compared to free epirubicin and epirubicin-loaded nanoparticles. This all-in-one nano-based ECM-targeting chemotherapeutic may provide a key-enabling technology for the treatment of TNBC.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Anticuerpos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Epirrubicina/administración & dosificación , Liposomas/química , Nanopartículas/química , Proteína-Lisina 6-Oxidasa/antagonistas & inhibidores , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Anticuerpos/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimioterapia Combinada/métodos , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/enzimología , Femenino , Humanos , Ratones , Ratones Desnudos , Proteína-Lisina 6-Oxidasa/inmunología , Distribución Tisular , Resultado del Tratamiento , Neoplasias de la Mama Triple Negativas/patología , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Sci Rep ; 10(1): 172, 2020 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-31932600

RESUMEN

Despite recent advances in drug delivery, the targeted treatment of unhealthy cells or tissues continues to remain a priority. In cancer (much like other pathologies), delivery vectors are designed to exploit physical and biological features of unhealthy tissues that are not always homogenous across the disease. In some cases, shifting the target from unhealthy tissues to the whole organ can represent an advantage. Specifically, the natural organ-specific retention of nanotherapeutics following intravenous administration as seen in the lung, liver, and spleen can be strategically exploited to enhance drug delivery. Herein, we outline the development of a cell-based delivery system using macrophages as a delivery vehicle. When loaded with a chemotherapeutic payload (i.e., doxorubicin), these cellular vectors (CELVEC) were shown to provide continued release within the lung. This study provides proof-of-concept evidence of an alternative class of biomimetic delivery vectors that capitalize on cell size to provide therapeutic advantages for pulmonary treatments.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Biomimética , Doxorrubicina/administración & dosificación , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Pulmón/metabolismo , Macrófagos/química , Animales , Antibióticos Antineoplásicos/farmacocinética , Doxorrubicina/farmacocinética , Liberación de Fármacos , Liposomas , Pulmón/citología , Masculino , Ratones , Ratones Desnudos , Distribución Tisular
5.
Biomater Sci ; 8(1): 333-341, 2020 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-31714542

RESUMEN

In the last decades, several approaches were developed to design drug delivery systems to address the multiple biological barriers encountered after administration while safely delivering a payload. In this scenario, bio-inspired and bio-mimetic approaches have emerged as promising solutions to evade the mononuclear phagocytic system while simultaneously negotiating the sequential transport across the various biological barriers. Leukocytes freely circulate in the bloodstream and selectively target the inflamed vasculature in response to injury, infection, and cancer. Recently we have shown the use of biomimetic nanovesicles, called leukosomes, which combine both the physical and biological properties of liposomes and leukocytes, respectively, to selectively deliver drugs to the inflamed vasculature. Here we report the use of leukosomes to target and deliver doxorubicin, a model chemotherapeutic, to tumors in syngeneic murine models of breast cancer and melanoma. Exploiting the inflammatory pathway responsible for recruiting immune cells to the site of injury, leukosomes exhibited increased targeting of cancer vasculature and stroma. Furthermore, delivery of doxorubicin with leukosomes enabled significant tumor growth inhibition compared with free doxorubicin in both breast and melanoma tumors. This study demonstrates the promise of using biomimetic nanovesicles for effective cancer management in solid tumors.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/uso terapéutico , Melanoma/tratamiento farmacológico , Nanopartículas/química , Animales , Materiales Biomiméticos/química , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/química , Doxorrubicina/farmacología , Femenino , Estimación de Kaplan-Meier , Leucocitos/química , Liposomas/química , Melanoma/mortalidad , Melanoma/patología , Ratones , Trasplante Homólogo
6.
Sci Rep ; 7(1): 2147, 2017 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-28526874

RESUMEN

Failure to remove a retrievable inferior vena cava (IVC) filter can cause severe complications with high treatment costs. Polydioxanone (PPDO) has been shown to be a good candidate material for resorbable IVC filters. However, PPDO is radioluscent under conventional imaging modalities. Thus, the positioning and integrity of these PPDO filters cannot be monitored by computed tomography (CT) or x-ray. Here we report the development of radiopaque PPDO IVC filters based on gold nanoparticles (AuNPs). Commercially available PPDO sutures were infused with AuNPs. Scanning electron microscopy analysis confirmed the presence of AuNP on the surface of PPDO. Micro-CT and x-ray images of the AuNP-infused PPDO sutures showed significant signal enhancement compared to untreated PPDO sutures. Elemental analysis showed that gold loading exceeded 2000 ppm. Tensile strength and in vitro cytotoxicity showed no significant difference between AuNP-infused and untreated PPDO. In a 10-week stability study, neither the gold content nor the radiopacity of the infused PPDO sutures significantly changed in the first 6 weeks. The increased attenuation of AuNP-infused PPDO sutures indicates their major advantage as a radiopaque resorbable filter material, as the radiopacity allows monitoring of the position and integrity of the filter, thereby increasing its safety and efficacy.


Asunto(s)
Oro , Nanopartículas del Metal , Filtros de Vena Cava , Supervivencia Celular , Oro/química , Humanos , Nanopartículas del Metal/química , Nanopartículas del Metal/ultraestructura , Polidioxanona/química , Embolia Pulmonar/etiología , Embolia Pulmonar/prevención & control , Radiografía , Suturas/efectos adversos , Resistencia a la Tracción , Vena Cava Inferior/cirugía , Microtomografía por Rayos X
7.
Biomaterials ; 82: 168-77, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26761780

RESUMEN

Recently, engineering the surface of nanotherapeutics with biologics to provide them with superior biocompatibility and targeting towards pathological tissues has gained significant popularity. Although the functionalization of drug delivery vectors with cellular materials has been shown to provide synthetic particles with unique biological properties, these approaches may have undesirable immunological repercussions upon systemic administration. Herein, we comparatively analyzed unmodified multistage nanovectors and particles functionalized with murine and human leukocyte cellular membrane, dubbed Leukolike Vectors (LLV), and the immunological effects that may arise in vitro and in vivo. Previously, LLV demonstrated an avoidance of opsonization and phagocytosis, in addition to superior targeting of inflammation and prolonged circulation. In this work, we performed a comprehensive evaluation of the importance of the source of cellular membrane in increasing their systemic tolerance and minimizing an inflammatory response. Time-lapse microscopy revealed LLV developed using a cellular coating derived from a murine (i.e., syngeneic) source resulted in an active avoidance of uptake by macrophage cells. Additionally, LLV composed of a murine membrane were found to have decreased uptake in the liver with no significant effect on hepatic function. As biomimicry continues to develop, this work demonstrates the necessity to consider the source of biological material in the development of future drug delivery carriers.


Asunto(s)
Materiales Biocompatibles/toxicidad , Materiales Biomiméticos/toxicidad , Inmunidad Innata/inmunología , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Nanocápsulas/toxicidad , Animales , Células Cultivadas , Ratones , Ratones Endogámicos BALB C
8.
J Biomed Mater Res A ; 102(10): 3540-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25269799

RESUMEN

The careful scrutiny of drug delivery systems is essential to evaluate and justify their potential for the clinic. Among the various studies necessary for preclinical testing, the impact of degradation is commonly overlooked. In this article, we investigate the effect of fabrication (porosity and nucleation layer) and environment (buffer and pH) factors on the degradation kinetics of multistage nanovectors (MSV) composed of porous silicon. The degradation by-products of MSV were exposed to endothelial cells and analyzed for detrimental effects on cellular internalization, architecture, proliferation, and cell cycle. Increases in porosity resulted in accelerated degradation exhibiting smaller-sized particles at comparable times. Removal of the nucleation layer (thin layer of small pores formed during the initial steps of etching) triggered a premature collapse of the entire central porous region of MSV. Variations in buffers prompted a faster degradation rate yielding smaller MSV within faster time frames, whereas increases in pH stimulated erosion of MSV and thus faster degradation. In addition, exposure to these degradation by-products provoked negligible impact on the proliferation and cell cycle phases on primary endothelial cells. In this study, we propose methods that lay the foundation for future investigations toward understanding the impact of the degradation of drug delivery platforms.


Asunto(s)
Materiales Biocompatibles/química , Portadores de Fármacos/química , Nanopartículas/química , Tampones (Química) , Endocitosis , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Concentración de Iones de Hidrógeno , Nanopartículas/ultraestructura , Porosidad
9.
Nat Nanotechnol ; 8(1): 61-8, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23241654

RESUMEN

The therapeutic efficacy of systemic drug-delivery vehicles depends on their ability to evade the immune system, cross the biological barriers of the body and localize at target tissues. White blood cells of the immune system--known as leukocytes--possess all of these properties and exert their targeting ability through cellular membrane interactions. Here, we show that nanoporous silicon particles can successfully perform all these actions when they are coated with cellular membranes purified from leukocytes. These hybrid particles, called leukolike vectors, can avoid being cleared by the immune system. Furthermore, they can communicate with endothelial cells through receptor-ligand interactions, and transport and release a payload across an inflamed reconstructed endothelium. Moreover, leukolike vectors retained their functions when injected in vivo, showing enhanced circulation time and improved accumulation in a tumour.


Asunto(s)
Biomimética/métodos , Leucocitos/química , Membranas Artificiales , Modelos Biológicos , Nanopartículas/química , Animales , Transporte Biológico , Adhesión Celular , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Leucocitos/metabolismo , Hígado/química , Hígado/metabolismo , Neoplasias Hepáticas Experimentales/química , Neoplasias Hepáticas Experimentales/metabolismo , Ratones , Ratones Endogámicos C57BL , Fagocitosis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA