Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Small ; 20(15): e2308390, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38037673

RESUMEN

Compartments are a fundamental feature of life, based variously on lipid membranes, protein shells, or biopolymer phase separation. Here, this combines self-assembling bacterial microcompartment (BMC) shell proteins and liquid-liquid phase separation (LLPS) to develop new forms of compartmentalization. It is found that BMC shell proteins assemble at the liquid-liquid interfaces between either 1) the dextran-rich droplets and PEG-rich continuous phase of a poly(ethyleneglycol)(PEG)/dextran aqueous two-phase system, or 2) the polypeptide-rich coacervate droplets and continuous dilute phase of a polylysine/polyaspartate complex coacervate system. Interfacial protein assemblies in the coacervate system are sensitive to the ratio of cationic to anionic polypeptides, consistent with electrostatically-driven assembly. In both systems, interfacial protein assembly competes with aggregation, with protein concentration and polycation availability impacting coating. These two LLPS systems are then combined to form a three-phase system wherein coacervate droplets are contained within dextran-rich phase droplets. Interfacial localization of BMC hexameric shell proteins is tunable in a three-phase system by changing the polyelectrolyte charge ratio. The tens-of-micron scale BMC shell protein-coated droplets introduced here can accommodate bioactive cargo such as enzymes or RNA and represent a new synthetic cell strategy for organizing biomimetic functionality.


Asunto(s)
Proteínas Bacterianas , Dextranos , Proteínas Bacterianas/metabolismo
2.
Mol Pharm ; 20(3): 1696-1708, 2023 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-36707500

RESUMEN

Despite the great promise of antibiotic therapy in wound infections, antibiotic resistance stemming from frequent dosing diminishes drug efficacy and contributes to recurrent infection. To identify improvements in antibiotic therapies, new antibiotic delivery systems that maximize pharmacological activity and minimize side effects are needed. In this study, we developed elastin-like peptide and collagen-like peptide nanovesicles (ECnVs) tethered to collagen-containing matrices to control vancomycin delivery and provide extended antibacterial effects against methicillin-resistant Staphylococcus aureus (MRSA). We observed that ECnVs showed enhanced entrapment efficacy of vancomycin by 3-fold as compared to liposome formulations. Additionally, ECnVs enabled the controlled release of vancomycin at a constant rate with zero-order kinetics, whereas liposomes exhibited first-order release kinetics. Moreover, ECnVs could be retained on both collagen-fibrin (co-gel) matrices and collagen-only matrices, with differential retention on the two biomaterials resulting in different local concentrations of released vancomycin. Overall, the biphasic release profiles of vancomycin from ECnVs/co-gel and ECnVs/collagen more effectively inhibited the growth of MRSA for 18 and 24 h, respectively, even after repeated bacterial inoculation, as compared to matrices containing free vancomycin, which just delayed the growth of MRSA. Thus, this newly developed antibiotic delivery system exhibited distinct advantages for controlled vancomycin delivery and prolonged antibacterial activity relevant to the treatment of wound infections.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Infección de Heridas , Humanos , Vancomicina , Antibacterianos/farmacología , Liposomas/farmacología , Pruebas de Sensibilidad Microbiana , Colágeno/farmacología
3.
Bioconjug Chem ; 33(3): 452-462, 2022 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-35167278

RESUMEN

Naturally occurring protein nanocages are promising drug carriers because of their uniform size and biocompatibility. Engineering efforts have enhanced the delivery properties of nanocages, but cell specificity and high drug loading remain major challenges. Herein, we fused the SpyTag peptide to the surface of engineered E2 nanocages to enable tunable nanocage decoration and effective E2 cell targeting using a variety of SpyCatcher (SC) fusion proteins. Additionally, the core of the E2 nanocage incorporated four phenylalanine mutations previously shown to allow hydrophobic loading of doxorubicin and pH-responsive release in acidic environments. We functionalized the surface of the nanocage with a highly cell-specific epidermal growth factor receptor (EGFR)-targeting protein conjugate, 4GE11-mCherry-SC, developed previously in our laboratories by employing unnatural amino acid (UAA) protein engineering chemistries. Herein, we demonstrated the benefits of this engineered protein nanocage construct for efficient drug loading, with a straightforward method for removal of the unloaded drug through elastin-like polypeptide-mediated inverse transition cycling. Additionally, we demonstrated approximately 3-fold higher doxorubicin internalization in inflammatory breast cancer cells compared to healthy breast epithelial cells, leading to targeted cell death at concentrations below the IC50 of free doxorubicin. Collectively, these results demonstrated the versatility of our UAA-based EGFR-targeting protein construct to deliver a variety of cargoes efficiently, including engineered E2 nanocages capable of site-specific functionalization and doxorubicin loading.


Asunto(s)
Neoplasias de la Mama , Portadores de Fármacos , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos/métodos , Receptores ErbB , Femenino , Humanos , Ligandos
4.
Mol Pharm ; 19(2): 661-673, 2022 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-35040326

RESUMEN

Intracellular delivery of protein therapeutics remains a significant challenge limiting the majority of clinically available protein drugs to extracellular targets. Strategies to deliver proteins to subcellular compartments have traditionally relied on cell-penetrating peptides, which can drive enhanced internalization but exhibit unreliable activity and are rarely able to target specific cells, leading to off-target effects. Moreover, few design rules exist regarding the relative efficacy of various endosomal escape strategies in proteins. Accordingly, we developed a simple fusion modification approach to incorporate endosomolytic peptides onto epidermal growth factor receptor (EGFR)-targeted protein conjugates and performed a systematic comparison of the endosomal escape efficacy, mechanism of action, and capacity to maintain EGFR-targeting specificity of conjugates modified with four different endosomolytic sequences of varying modes of action (Aurein 1.2, GALA, HA2, and L17E). Use of the recently developed Gal8-YFP assay indicated that the fusion of each endosomolytic peptide led to enhanced endosomal disruption. Additionally, the incorporation of each endosomolytic peptide increased the half-life of the internalized protein and lowered lysosomal colocalization, further supporting the membrane-disruptive capacity. Despite this, only EGFR-targeted conjugates modified with Aurein 1.2 or GALA maintained EGFR specificity. These results thus demonstrated that the choice of endosomal escape moiety can substantially affect targeting capability, cytotoxicity, and bioactivity and provided important new insights into endosomolytic peptide selection for the design of targeted protein delivery systems.


Asunto(s)
Neoplasias de la Mama , Péptidos de Penetración Celular , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Péptidos de Penetración Celular/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Endosomas/metabolismo , Receptores ErbB/metabolismo , Femenino , Humanos
5.
Bioconjug Chem ; 31(10): 2272-2282, 2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-32931255

RESUMEN

Proteins have the capacity to treat a multitude of diseases both as therapeutics and as drug carriers due to their complex functional properties, specificity toward binding partners, biocompatibility, and programmability. Despite this, native proteins often require assistance to target diseased tissue due to poor pharmacokinetic properties and membrane impermeability. Functionalizing therapeutic proteins and drug carriers through direct conjugation of delivery moieties can enhance delivery capabilities. Traditionally, this has been accomplished through bioconjugation methods that have little control over the location or orientation of the modification, leading to highly heterogeneous products with varying activity. A multitude of promising site-specific protein conjugation methods have been developed to allow more tailorable display of delivery moieties and thereby enhance protein activity, circulation properties, and targeting specificity. Here, we focus on three particularly promising site-specific bioconjugation techniques for protein delivery: unnatural amino acid incorporation, Sortase-mediated ligation, and SpyCatcher/SpyTag chemistry. In this review, we highlight the promise of site-specific bioconjugation for targeted drug delivery by summarizing impactful examples in literature, considering important design principles when constructing bioconjugates, and discussing our perspectives on future directions.


Asunto(s)
Aminoácidos/química , Portadores de Fármacos/química , Proteínas/administración & dosificación , Aminoácidos/síntesis química , Animales , Técnicas de Química Sintética/métodos , Portadores de Fármacos/síntesis química , Sistemas de Liberación de Medicamentos , Humanos , Modelos Moleculares , Preparaciones Farmacéuticas/administración & dosificación
6.
Bioconjug Chem ; 30(2): 432-442, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30615416

RESUMEN

Proteins are ideal candidates for disease treatment because of their high specificity and potency. Despite this potential, delivery of proteins remains a significant challenge due to the intrinsic size, charge, and stability of proteins. Attempts to overcome these challenges have most commonly relied on direct conjugation of polymers and peptides to proteins via reactive groups on naturally occurring residues. While such approaches have shown some success, they allow limited control of the spacing and number of moieties coupled to proteins, which can hinder bioactivity and delivery capabilities of the therapeutic. Here, we describe a strategy to site-specifically conjugate delivery moieties to therapeutic proteins through unnatural amino acid (UAA) incorporation, in order to explore the effect of epidermal growth factor receptor (EGFR)-targeted ligand valency and spacing on internalization of proteins in EGFR-overexpressing inflammatory breast cancer (IBC) cells. Our results demonstrate the ability to enhance targeted protein delivery by tuning a small number of EGFR ligands per protein and clustering these ligands to promote multivalent ligand-receptor interactions. Furthermore, the tailorability of this simple approach was demonstrated through IBC-targeted cell death via the delivery of yeast cytosine deaminase (yCD), a prodrug converting enzyme.


Asunto(s)
Aminoácidos/metabolismo , Citosina Desaminasa/administración & dosificación , Proteínas Luminiscentes/administración & dosificación , Aminoácidos/química , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Química Clic , Citosina Desaminasa/química , Citosina Desaminasa/farmacocinética , Sistemas de Liberación de Medicamentos , Receptores ErbB/metabolismo , Femenino , Humanos , Ligandos , Proteínas Luminiscentes/química , Proteínas Luminiscentes/farmacocinética , Modelos Moleculares , Unión Proteica , Levaduras/enzimología , Proteína Fluorescente Roja
7.
Bioconjug Chem ; 29(11): 3691-3704, 2018 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-30350573

RESUMEN

Histone-inspired polymer assemblies (polyplexes) can regulate gene expression and subcellular transport in plasmids by harnessing the cellular machinery normally used for histone proteins. When grafted to polyplexes, histone tails promote nuclear accumulation, trigger plasmid DNA (pDNA) release, and enhance transcription. Herein, we developed multifunctional gold nanoparticles (AuNPs) decorated by histone motifs as histone-inspired scaffolds with improved pDNA binding, easy bioimaging, and increased potential for gene delivery and chromatin analysis applications. We hypothesized that polycationic AuNPs coupled to histone motifs would mimic the native presentation of these sequences on the histone octamer and thereby create structures with the capacity to both engage native histone effectors and condense pDNA into nucleosome-inspired nanostructures. AuNPs bearing ∼2 nm cores were prepared based on the well-established Brust-Schiffrin two-phase method involving tetrachloroaurate reduction in the presence of 1-pentanethiol. Solid phase peptide synthesis was employed to generate thiolated polycationic ligands and histone tail motifs, and the AuNPs and peptide ligands were combined in a two-step Murray place exchange reaction at various ratios to produce a collection of polycationic AuNPs modified with varying amounts of histone tails. Electron microscopy and thermal analyses demonstrated that these modified AuNPs exhibited tunable biochemical and biophysical properties that closely mimicked the properties of native histones. The histone-mimetic nanoscaffolds efficiently and sequence-specifically engaged histone effectors responsible for activating transcription. In addition, the nanoscaffolds condensed pDNA into complexes with high stability in the presence of physiological concentrations of heparin, a common extracellular polyanion. These combined properties of histone engagement and high stability led to a ∼6-fold enhancement in transfection efficiency as compared with typical polymeric transfection reagents, with the increased transfection efficiency correlated to the presence and amount of histone tails displayed on the surface of the nanoscaffolds. These findings demonstrate the utility of employing a biomimetic materials design approach to develop more effective and stable delivery vehicles for gene transfer and chromatin analysis applications.


Asunto(s)
Materiales Biomiméticos/química , ADN/administración & dosificación , Técnicas de Transferencia de Gen , Oro/química , Histonas/química , Nanopartículas del Metal/química , Animales , Células CHO , Cromatina/química , Cricetulus , ADN/química , ADN/genética , Nanopartículas del Metal/ultraestructura , Plásmidos/administración & dosificación , Plásmidos/química , Plásmidos/genética , Propiedades de Superficie , Transfección/métodos
8.
Biomacromolecules ; 18(6): 1814-1824, 2017 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-28441861

RESUMEN

The incorporation of anionic excipients into polyplexes is a promising strategy for modulating siRNA binding versus release and integrating diagnostic capabilities; however, specific design criteria and structure-function relationships are needed to facilitate the development of nanocarrier-based theranostics. Herein, we incorporated poly(acrylic acid) (PAA) and quantum dot (QD) excipients into photolabile siRNA polyplexes to increase gene silencing efficiencies by up to 100% and enable self-reporting of nanocarrier disassembly. Our systematic approach identified the functional relationships between gene silencing and key parameters such as excipient loading fractions and molecular weights that facilitated the establishment of design rules for optimization of nanocarrier efficacy. For example, we found that PAA molecular weights ∼10-20× greater than that of the coencapsulated siRNA exhibited the most efficient release and silencing. Furthermore, siRNA release assays and RNAi modeling allowed us to generate a PAA "heat map" that predicted gene silencing a priori as a function of PAA molecular weight and loading fraction. QDs further promoted selective siRNA release and provided visual as well as Förster resonance energy transfer (FRET)-based monitoring of the dynamic changes in nanostructure in situ. Moreover, even with the addition of anionic components, our formulations exhibited substantially improved stability and shelf life relative to typical formulations, with complete stability after a week of storage and full activity in the presence of serum. Taken together, this study enabled synergistic improvements in siRNA release and diagnostic capabilities, along with the development of mechanistic insights that are critical for advancing the translation of nucleic acid theranostics into the clinic.


Asunto(s)
Portadores de Fármacos , Nanopartículas/química , Polietilenglicoles/química , Puntos Cuánticos/química , Compuestos de Amonio Cuaternario/química , ARN Interferente Pequeño/genética , Animales , Composición de Medicamentos/métodos , Transferencia Resonante de Energía de Fluorescencia , Expresión Génica , Silenciador del Gen , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/antagonistas & inhibidores , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/genética , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Heparina/química , Luz , Ratones , Células 3T3 NIH , Nanopartículas/metabolismo , Nanopartículas/ultraestructura , Procesos Fotoquímicos , Puntos Cuánticos/metabolismo , Puntos Cuánticos/ultraestructura , ARN Interferente Pequeño/metabolismo
9.
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
10.
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
11.
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
12.
Chem Soc Rev ; 42(17): 7057-71, 2013 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-23403471

RESUMEN

Stimuli-responsive polymeric materials is one of the fastest growing fields of the 21st century, with the annual number of papers published more than quadrupling in the last ten years. The responsiveness of polymer solution assemblies and surfaces to biological stimuli (e.g. pH, reduction-oxidation, enzymes, glucose) and externally applied triggers (e.g. temperature, light, solvent quality) shows particular promise for various biomedical applications including drug delivery, tissue engineering, medical diagnostics, and bioseparations. Furthermore, the integration of copolymer architectures into stimuli-responsive materials design enables exquisite control over the locations of responsive sites within self-assembled nanostructures. The combination of new synthesis techniques and well-defined copolymer self-assembly has facilitated substantial developments in stimuli-responsive materials in recent years. In this tutorial review, we discuss several methods that have been employed to synthesize self-assembling and stimuli-responsive copolymers for biomedical applications, and we identify common themes in the response mechanisms among the targeted stimuli. Additionally, we highlight parallels between the chemistries used for generating solution assemblies and those employed for creating copolymer surfaces.


Asunto(s)
Polímeros/química , Sistemas de Liberación de Medicamentos , Enzimas/metabolismo , Glucosa/química , Glucosa/metabolismo , Concentración de Iones de Hidrógeno , Nanoestructuras/química , Oxidación-Reducción , Procesos Fotoquímicos , Solventes/química , Ingeniería de Tejidos
13.
ACS Appl Mater Interfaces ; 16(11): 13399-13410, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38466900

RESUMEN

Although lipid nanoparticles (LNPs) are the predominant nanocarriers for short-interfering RNA (siRNA) delivery, most therapies use nearly identical formulations that have taken 30 years to design but lack the diverse property ranges necessary for versatile application. This dearth in variety and the extended timeline for implementation are attributed to a limited understanding of how LNP properties facilitate overcoming biological barriers. Herein, a simple kinetic model was developed by using major rate-limiting steps for siRNA delivery, and this model enabled the identification of a critical parameter to predict LNP efficacy without extensive experimental testing. A volume-averaged log D, the "solubility" of charged molecules as a function of pH weighted by component volume fractions, resulted in a good correlation between LNP composition and siRNA delivery. Both the effects of modifying the structures of ionizable lipids and LNP composition on gene silencing were easily captured in the model predictions. Thus, this approach provides a robust LNP structure-activity relationship to dramatically accelerate the realization of effective LNP formulations.


Asunto(s)
Lípidos , Nanopartículas , Lípidos/química , Liposomas , ARN Interferente Pequeño/química , Nanopartículas/química
14.
Bioeng Transl Med ; 9(3): e10642, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38818118

RESUMEN

Chronic wounds are an unmet clinical need affecting millions of patients globally, and current standards of care fail to consistently promote complete wound closure and prevent recurrence. Disruptions in growth factor signaling, a hallmark of chronic wounds, have led researchers to pursue growth factor therapies as potential supplements to standards of care. Initial studies delivering growth factors in protein form showed promise, with a few formulations reaching clinical trials and one obtaining clinical approval. However, protein-form growth factors are limited by instability and off-target effects. Gene therapy offers an alternative approach to deliver growth factors to the chronic wound environment, but safety concerns surrounding gene therapy as well as efficacy challenges in the gene delivery process have prevented clinical translation. Current growth factor delivery and gene therapy approaches have primarily used single growth factor formulations, but recent efforts have aimed to develop multi-growth factor approaches that are better suited to address growth factor insufficiencies in the chronic wound environment, and these strategies have demonstrated improved efficacy in preclinical studies. This review provides an overview of chronic wound healing, emphasizing the need and potential for growth factor therapies. It includes a summary of current standards of care, recent advances in growth factor, cell-based, and gene therapy approaches, and future perspectives for multi-growth factor therapeutics.

15.
Curr Opin Biotechnol ; 86: 103070, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38354452

RESUMEN

Protein nanoparticles offer a highly tunable platform for engineering multifunctional drug delivery vehicles that can improve drug efficacy and reduce off-target effects. While many protein nanoparticles have demonstrated the ability to tolerate genetic and posttranslational modifications for drug delivery applications, this review will focus on three protein nanoparticles of increasing size. Each protein nanoparticle possesses distinct properties such as highly tunable stability, capacity for splitting or fusing subunits for modular surface decoration, and well-characterized conformational changes with impressive capacity for large protein cargos. While many of the genetic and posttranslational modifications leverage these protein nanoparticle's properties, the shared techniques highlight engineering approaches that have been generalized across many protein nanoparticle platforms.


Asunto(s)
Sistemas de Liberación de Medicamentos , Nanopartículas , Sistemas de Liberación de Medicamentos/métodos
17.
Annu Rev Chem Biomol Eng ; 14: 243-264, 2023 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-36888991

RESUMEN

From the first clinical trial by Dr. W.F. Anderson to the most recent US Food and Drug Administration-approved Luxturna (Spark Therapeutics, 2017) and Zolgensma (Novartis, 2019), gene therapy has revamped thinking and practice around cancer treatment and improved survival rates for adult and pediatric patients with genetic diseases. A major challenge to advancing gene therapies for a broader array of applications lies in safely delivering nucleic acids to their intended sites of action. Peptides offer unique potential to improve nucleic acid delivery based on their versatile and tunable interactions with biomolecules and cells. Cell-penetrating peptides and intracellular targeting peptides have received particular focus due to their promise for improving the delivery of gene therapies into cells. We highlight key examples of peptide-assisted, targeted gene delivery to cancer-specific signatures involved in tumor growth and subcellular organelle-targeting peptides, as well as emerging strategies to enhance peptide stability and bioavailability that will support long-term implementation.


Asunto(s)
Péptidos de Penetración Celular , Neoplasias , Ácidos Nucleicos , Humanos , Niño , Técnicas de Transferencia de Gen , Terapia Genética , Péptidos de Penetración Celular/química , Neoplasias/genética , Neoplasias/terapia , Sistemas de Liberación de Medicamentos
18.
J Mater Chem B ; 11(18): 3985-3993, 2023 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-37083736

RESUMEN

Protein therapeutics offer enormous clinical impact in treating a variety of diseases by offering high selectivity with limited off-target effects. However, delivery challenges severely hinder functional proteins from reaching their target cells and necessitate frequent administration. To address these problems, nanocarrier encapsulation can provide protease protection and enhanced targeted transportation of functional proteins to their intended disease site. Inspired by their viral analogues, virus-like particles (VLPs) are non-infectious viral capsids that have potential for drug delivery applications because of their shared structural characteristics, such as high loading capacity, particle stability, and structural uniformity. Here, we describe a modular hepatitis B virus (HBV) VLP delivery platform offering tunable modifications of both the exterior and interior viral capsid surfaces via SpyCatcher-SpyTag bioconjugation and a multi-expression system, respectively. This new platform facilitates modification with epidermal growth factor receptor (EGFR)-targeting proteins and encapsulation with both model green fluorescent protein (GFP) and prodrug-converting yeast cytosine deaminase (yCD) enzyme. The resultant targeted VLPs demonstrated enhanced uptake and toxicity in EGFR-overexpressing triple negative breast cancer (TNBC) cells in contrast to non-malignant breast epithelial cells.


Asunto(s)
Virus de la Hepatitis B , Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Proteínas Fluorescentes Verdes/genética , Saccharomyces cerevisiae , Receptores ErbB
19.
ACS Appl Mater Interfaces ; 15(13): 16434-16447, 2023 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-36961242

RESUMEN

Disruption in vascularization during wound repair can severely impair healing. Proangiogenic growth factor therapies have shown great healing potential; however, controlling growth factor activity and cellular behavior over desired healing time scales remains challenging. In this study, we evaluated collagen-mimetic peptide (CMP) tethers for their capacity to control growth factor gene transfer and growth factor activity using our recently developed gene-activated hyaluronic acid-collagen matrix (GAHCM). GAHCM was comprised of DNA/polyethyleneimine (PEI) polyplexes that were retained on hyaluronic acid (HA)-collagen hydrogels using CMPs. We hypothesized that using CMP-collagen tethers to control vascular endothelial growth factor-A (VEGF-A) gene delivery in fibroblasts would provide a powerful strategy to modulate the proangiogenic behaviors of endothelial cells (ECs) for blood vessel formation, resulting in enhanced wound repair. In co-culture experiments, we observed that CMP-modified GAHCM induced tunable gene delivery in fibroblasts as predicted, and correspondingly, VEGF-A produced by the fibroblasts led to increased growth and persistent migration of ECs for at least 7 days, as compared to non-CMP-modified GAHCM. Moreover, when ECs were exposed to fibroblast-containing VEGF-GAHCM with higher levels of CMP modification (50% CMP-PEI, or 50 CP), high CD31 expression was stimulated, resulting in the formation of an interconnected EC network with a significantly higher network volume and a larger diameter network structure than controls. Application of VEGF-GAHCM with 50 CP in murine splinted excisional wounds facilitated prolonged prohealing and proangiogenic responses resulting in increased blood vessel formation, improved granulation tissue formation, faster re-epithelialization, and overall enhanced repair. These findings suggest the benefits of CMP-collagen tethers as useful tools to control gene transfer and growth factor activity for improved treatment of wounds.


Asunto(s)
Factor A de Crecimiento Endotelial Vascular , Cicatrización de Heridas , Ratones , Animales , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ácido Hialurónico/química , Células Endoteliales/metabolismo , Colágeno/química
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA