Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Mol Ther ; 29(5): 1729-1743, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33484965

RESUMEN

Extracellular vesicles (EVs) are an important intercellular communication system facilitating the transfer of macromolecules between cells. Delivery of exogenous cargo tethered to the EV surface or packaged inside the lumen are key strategies for generating therapeutic EVs. We identified two "scaffold" proteins, PTGFRN and BASP1, that are preferentially sorted into EVs and enable high-density surface display and luminal loading of a wide range of molecules, including cytokines, antibody fragments, RNA binding proteins, vaccine antigens, Cas9, and members of the TNF superfamily. Molecules were loaded into EVs at high density and exhibited potent in vitro activity when fused to full-length or truncated forms of PTGFRN or BASP1. Furthermore, these engineered EVs retained pharmacodynamic activity in a variety of animal models. This engineering platform provides a simple approach to functionalize EVs with topologically diverse macromolecules and represents a significant advance toward unlocking the therapeutic potential of EVs.


Asunto(s)
Vesículas Extracelulares/trasplante , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas/administración & dosificación , Proteínas Represoras/metabolismo , Animales , Comunicación Celular , Sistemas de Liberación de Medicamentos , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Femenino , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Ratones , Proteínas de Neoplasias/genética , Proteínas del Tejido Nervioso/genética , Proteínas Represoras/genética
2.
Biotechnol Bioeng ; 113(12): 2686-2697, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27241022

RESUMEN

Gene therapy platforms offer a variety of potentially effective solutions for development of targeted agents that can be exploited for cancer treatment. The physicochemical properties of nanocarriers can be tuned to enhance their localization in tumors, and cell specificity can also be increased by appropriate selection of gene targets. A relatively underexploited approach to enhance therapeutic selectivity in cancer tissues is the use of nanocarriers whose nuclear targeting and uptake are triggered by the altered expression of specific endomembrane trafficking proteins in cancer cells. Previously, we showed that histone 3 (H3) peptide-targeted DNA polyplexes traffic to the nucleus efficiently through caveolar endocytosis followed by transfer through the Golgi and endoplasmic reticulum (ER). We hypothesized that these polyplexes would exhibit enhanced activity in inflammatory breast cancer (IBC) cells, which overexpress caveolin-1 as part of their invasive phenotype, and we also posited that this targeting effect could be exploited to facilitate IBC-specific transfection and prodrug conversion in the presence of normal breast epithelial cells. Using cellular transfection experiments, function-blocking assays, and confocal imaging in both IBC SUM149 cell monocultures and IBC SUM149 co-cultures with MCF10A normal breast epithelial cells, we found that our H3-targeted polyplexes selectively transfected IBC SUM149 cells at a 4-fold higher level than normal breast epithelial cells. This selectivity and increased transfection were caused by a 2.2-fold overexpression of caveolin-1 in IBC SUM149 cells, which led to increased polyplex trafficking to the nucleus through the Golgi and ER. We also saw similar enhancements in cell selectivity and transfection when cells were transfected with a suicide gene/prodrug combination, as the increased expression of the suicide gene in IBC SUM149 cells led to a 55% decrease in viability in IBC SUM149 cells as compared to a 25% decrease in MCF10A cells. These findings demonstrate that differences in the expression of the endocytic membrane protein caveolin-1 can be exploited for cell-selective gene delivery, and ultimately, these gene-based targeting approaches may be useful in potential treatments for aggressive cancer types. Biotechnol. Bioeng. 2016;113: 2686-2697. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Caveolina 1/metabolismo , Histonas/metabolismo , Neoplasias Inflamatorias de la Mama/metabolismo , Neoplasias Inflamatorias de la Mama/terapia , Plásmidos/administración & dosificación , Profármacos/farmacocinética , Apoptosis/genética , Caveolina 1/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Marcación de Gen/métodos , Terapia Genética/métodos , Histonas/genética , Humanos , Neoplasias Inflamatorias de la Mama/patología , Nanocápsulas/química , Nanocápsulas/ultraestructura , Plásmidos/genética , Transfección/métodos , Resultado del Tratamiento
3.
J Gene Med ; 17(3-5): 69-79, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25731756

RESUMEN

BACKGROUND: The successful application of nonviral gene transfer technologies requires both improved understanding and control with respect to intracellular trafficking and release. However, the intracellular space is highly complex and hence well-defined, stable structures are necessary to probe the stages of the delivery pathway. Fluorescent labeling is a regularly used approach to monitor nonviral delivery and release, yet few studies investigate the effects of label incorporation on the structure and activity of gene-containing vehicles. METHODS: In the present study, the impacts of label incorporation on the assembly and gene transfer capacity of DNA polyplexes were determined through the utilization of a model DNA-polyethylenimine (PEI) delivery system. PEI was fluorescently labeled with the Oregon Green® dye prior to polyplex formation and delivery to CHO-K1 cells. RESULTS: The present study provides evidence showing that routine labeling strategies for polyplexes weakened DNA binding affinity, produced large quantities of extracellular structures and significantly increased intracellular polyplex aggregation. Additionally, cellular internalization studies showed that increased labeling fractions led to reductions in polyplex uptake as a result of weakened complexation. CONCLUSIONS: These results not only provide insight into the assembly of these structures, but also help to identify labeling strategies sufficient to preserve activity at the same time as enabling detailed studies of trafficking and disassembly.


Asunto(s)
Colorantes Fluorescentes/metabolismo , Técnicas de Transferencia de Gen , Espacio Intracelular/metabolismo , Polietileneimina/química , Animales , Células CHO , Cricetinae , Cricetulus , Dispersión Dinámica de Luz , Endocitosis , Etidio/metabolismo , Citometría de Flujo , Imagen Óptica , Transfección , Virus/metabolismo
4.
Mol Pharm ; 12(12): 4488-97, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26465823

RESUMEN

For successful gene delivery, plasmid DNA must be able to access the nucleus in order to be transcribed. Numerous studies have shown that gene delivery occurs more readily in dividing cells, which is attributed to increased nuclear access when the nuclear envelope disassembles during mitosis; however, nonviral carriers continue to have low transfection efficiencies and require large quantities of DNA per cell to achieve reasonable gene transfer, even in dividing cells. Therefore, we hypothesized that using histone-derived nuclear localization sequences (NLS)s to target polyplexes might enhance nuclear delivery by facilitating interactions with histone effectors that mediate nuclear partitioning and retention during mitosis. We discovered a novel interaction between polyplexes linked to histone 3 (H3) N-terminal tail peptides and the histone nuclear import protein importin-4, as evidenced by strong spatial colocalization as well as significantly decreased transfection when importin-4 expression was reduced. A fraction of the histone-targeted polyplexes was also found to colocalize with the retrotranslocon of the endoplasmic reticulum, Sec61. Super resolution microscopy demonstrated a high level of polyplex binding to chromatin postmitosis, and there also was a significant decrease in the amount of chromatin binding following importin-4 knockdown. These results provide evidence that natural histone effectors mediate both nuclear entry and deposition on chromatin by histone-targeted polyplexes, and a translocation event from the endoplasmic reticulum into the cytosol may occur before mitosis to enable the polyplexes to interact with these essential cytoplasmic proteins.


Asunto(s)
Núcleo Celular/metabolismo , Cromatina/metabolismo , Carioferinas/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Animales , Células CHO , Línea Celular , Cricetulus , ADN/metabolismo , Retículo Endoplásmico/metabolismo , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Histonas/metabolismo , Señales de Localización Nuclear/metabolismo , Plásmidos/metabolismo , Transfección/métodos
5.
J Gene Med ; 14(9-10): 580-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22976512

RESUMEN

BACKGROUND: Nonviral gene delivery has a limited efficacy partly as a result of poor nuclear delivery, yet an understanding of the mechanisms of nuclear entry is limited. The present study aimed to test the common hypothesis that most nonviral vehicles enter the nucleus during cell division. METHODS: Polystyrene particles with diameters of 24-200 nm and carboxylate or amine surface groups, were either used as is or, alternatively, were functionalized with carboxyl-, hydroxyl- or amine- terminated poly(ethylene glycol) (PEG) and subsequently microinjected into the cytoplasm of NIH/3T3 mouse fibroblast cells. The post-mitotic locations of the particles were analyzed and compared with the locations of cytoplasmically microinjected plasmid DNA (pDNA), pDNA polyplexes or nuclear localization signal (NLS)-functionalized pDNA polyplexes. RESULTS: We observed that all polystyrene particles as well as the NLS-free polyplexes were excluded from the nucleus post-mitosis. By contrast, free pDNA and pDNA polyplexes containing an NLS accumulated in the nucleus after division. CONCLUSIONS: These data suggest that biochemically specific modes of association with chromatin-associated proteins or other nuclear components are necessary for the nuclear inclusion of polyplexes and nanoparticles during mitosis.


Asunto(s)
Núcleo Celular/genética , ADN/administración & dosificación , Técnicas de Transferencia de Gen , Mitosis , Nanopartículas/administración & dosificación , Nanopartículas/química , Animales , Núcleo Celular/metabolismo , Citoplasma , ADN/química , ADN/metabolismo , Histonas/síntesis química , Ratones , Microinyecciones , Datos de Secuencia Molecular , Células 3T3 NIH , Señales de Localización Nuclear/química , Señales de Localización Nuclear/metabolismo , Tamaño de la Partícula , Polietilenglicoles/química , Poliestirenos/química , Poliestirenos/metabolismo
6.
Commun Biol ; 4(1): 497, 2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33888863

RESUMEN

Cyclic dinucleotide (CDN) agonists of the STimulator of InterferoN Genes (STING) pathway have shown immune activation and tumor clearance in pre-clinical models. However, CDNs administered intratumorally also promote STING activation leading to direct cytotoxicity of many cell types in the tumor microenvironment (TME), systemic inflammation due to rapid tumor extravasation of the CDN, and immune ablation in the TME. These result in a failure to establish immunological memory. ExoSTING, an engineered extracellular vesicle (EV) exogenously loaded with CDN, enhances the potency of CDN and preferentially activates antigen presenting cells in the TME. Following intratumoral injection, exoSTING was retained within the tumor, enhanced local Th1 responses and recruitment of CD8+ T cells, and generated systemic anti-tumor immunity to the tumor. ExoSTING at therapeutically active doses did not induce systemic inflammatory cytokines, resulting in an enhanced therapeutic window. ExoSTING is a novel, differentiated therapeutic candidate that leverages the natural biology of EVs to enhance the activity of CDNs.


Asunto(s)
Vesículas Extracelulares/fisiología , Vigilancia Inmunológica , Microambiente Tumoral/fisiología , Animales , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
7.
Curr Pharm Des ; 22(9): 1227-44, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26675220

RESUMEN

New biopharmaceutical molecules, potentially able to provide more personalized and effective treatments, are being identified through the advent of advanced synthetic biology strategies, sophisticated chemical synthesis approaches, and new analytical methods to assess biological potency. However, translation of many of these structures has been significantly limited due to the need for more efficient strategies to deliver macromolecular therapeutics to desirable intracellular sites of action. Engineered nanocarriers that encapsulate peptides, proteins, or nucleic acids are generally internalized into target cells via one of several endocytic pathways. These nanostructures, entrapped within endosomes, must navigate the intracellular milieu to orchestrate delivery to the intended destination, typically the cytoplasm or nucleus. For therapeutics active in the cytoplasm, endosomal escape continues to represent a limiting step to effective treatment, since a majority of nanocarriers trapped within endosomes are ultimately marked for enzymatic degradation in lysosomes. Therapeutics active in the nucleus have the added challenges of reaching and penetrating the nuclear envelope, and nuclear delivery remains a preeminent challenge preventing clinical translation of gene therapy applications. Herein, we review cutting-edge peptide- and polymer-based design strategies with the potential to enable significant improvements in biopharmaceutical efficacy through improved intracellular targeting. These strategies often mimic the activities of pathogens, which have developed innate and highly effective mechanisms to penetrate plasma membranes and enter the nucleus of host cells. Understanding these mechanisms has enabled advances in synthetic peptide and polymer design that may ultimately improve intracellular trafficking and bioavailability, leading to increased access to new classes of biotherapeutics.


Asunto(s)
Biofarmacia , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Portadores de Fármacos/farmacocinética , Sustancias Macromoleculares/administración & dosificación , Nanopartículas/química , Animales , Transporte Biológico , Humanos , Sustancias Macromoleculares/química
8.
Mol Ther Nucleic Acids ; 4: e226, 2015 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-25668340

RESUMEN

Nonviral gene delivery is a promising therapeutic approach because of its safety and controllability, yet limited gene transfer efficacy is a common issue. Most nonviral strategies rely upon endosomal escape designs; however, endosomal escape is often uncorrelated with improved gene transfer and membranolytic structures are typically cytotoxic. Previously, we showed that histone-targeted polyplexes trafficked to the nucleus through an alternative route involving caveolae and the Golgi and endoplasmic reticulum (ER), using pathways similar to several pathogens. We hypothesized that the efficacy of these polyplexes was due to an increased utilization of native vesicular trafficking as well as regulation by histone effectors. Accordingly, using confocal microscopy and cellular fractionation, we determined that a key effect of histone-targeting was to route polyplexes away from clathrin-mediated recycling pathways by harnessing endomembrane transfer routes regulated by histone methyltransferases. An unprecedented finding was that polyplexes accumulated in Rab6-labeled Golgi/ER vesicles and ultimately shuttled directly into the nucleus during ER-mediated nuclear envelope reassembly. Specifically, super resolution microscopy and fluorescence correlation spectroscopy unequivocally indicated that the polyplexes remained associated with ER vesicles/membranes until mitosis, when they were redistributed into the nucleus. These novel findings highlight alternative mechanisms to subvert endolysosomal trafficking and harness the ER to enhance gene transfer.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA