Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
J Nanobiotechnology ; 21(1): 5, 2023 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-36597089

RESUMEN

The aggressive proliferation of tumor cells often requires increased glucose uptake and excessive anaerobic glycolysis, leading to the massive production and secretion of lactate to form a unique tumor microenvironment (TME). Therefore, regulating appropriate lactate levels in the TME would be a promising approach to control tumor cell proliferation and immune suppression. To effectively consume lactate in the TME, lactate oxidase (LOX) and catalase (CAT) were displayed onto Aquifex aeolicus lumazine synthase protein nanoparticles (AaLS) to form either AaLS/LOX or AaLS/LOX/CAT. These complexes successfully consumed lactate produced by CT26 murine colon carcinoma cells under both normoxic and hypoxic conditions. Specifically, AaLS/LOX generated a large amount of H2O2 with complete lactate consumption to induce drastic necrotic cell death regardless of culture condition. However, AaLS/LOX/CAT generated residual H2O2, leading to necrotic cell death only under hypoxic condition similar to the TME. While the local administration of AaLS/LOX to the tumor site resulted in mice death, that of AaLS/LOX/CAT significantly suppressed tumor growth without any severe side effects. AaLS/LOX/CAT effectively consumed lactate to produce adequate amounts of H2O2 which sufficiently suppress tumor growth and adequately modulate the TME, transforming environments that are favorable to tumor suppressive neutrophils but adverse to tumor-supportive tumor-associated macrophages. Collectively, these findings showed that the modular functionalization of protein nanoparticles with multiple metabolic enzymes may offer the opportunity to develop new enzyme complex-based therapeutic tools that can modulate the TME by controlling cancer metabolism.


Asunto(s)
Nanopartículas , Neoplasias , Animales , Ratones , Ácido Láctico , Catalasa , Microambiente Tumoral , Peróxido de Hidrógeno , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Nanopartículas/uso terapéutico , Línea Celular Tumoral
2.
Biomacromolecules ; 23(12): 5193-5201, 2022 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-36378752

RESUMEN

With the continued advancement in the design and engineering of hydrogels for biomedical applications, there is a growing interest in imparting stimuli-responsiveness to the hydrogels in order to control their physicomechanical properties in a more programmable manner. In this study, an in situ forming hydrogel is developed by cross-linking alginate with an elastin-like polypeptide (ELP). Lysine-rich ELP synthesized by recombinant DNA technology is reacted with alginate presenting an aldehyde via Schiff base formation, resulting in facile hydrogel formation under physiological conditions. The physicomechanical properties of alginate-ELP hydrogels can be controlled in a wide range by the concentrations of alginate and ELP. Owing to the thermoresponsive properties of the ELP, the alginate-ELP hydrogels undergo swelling/deswelling near the physiological temperature. Taking advantage of these highly attractive properties of alginate-ELP, drug release kinetics were measured to evaluate their potential as a thermoresponsive drug delivery system. Furthermore, an ex vivo model was used to demonstrate the minimally invasive tissue injectability.


Asunto(s)
Elastina , Hidrogeles , Hidrogeles/química , Liberación de Fármacos , Elastina/química , Péptidos/química , Temperatura , Cinética
3.
J Nanobiotechnology ; 20(1): 387, 2022 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-35999603

RESUMEN

The plant toxin ricin, especially its cytotoxic A chain (RTA), can be genetically engineered with targeting ligands to develop specific anti-cancer recombinant immunotoxins (RITs). Here, we used affibody molecules targeting two cancer biomarkers, the receptors HER2 and EGFR, along with the KDEL signal peptide to construct two cancer-specific ricin-based RITs, HER2Afb-RTA-KDEL and EGFRAfb-RTA-KDEL. The affibodies successfully provided target-specificity and subsequent receptor-mediated endocytosis and the KDEL signal peptide routed the RITs through the retrograde transport pathway, effectively delivering RTA to the cytosol as well as avoiding the alternate recycling pathway that typical cancer cells frequently have. The in vivo efficacy of RITs was enhanced by introducing the albumin binding domain (AlBD) to construct AlBD/HER2Afb/RTA-KDEL. Systemic administration of AlBD-containing RITs to tumor-bearing mice significantly suppressed tumor growth without any noticeable side-effects. Collectively, combining target-selective affibody molecules, a cytotoxic RTA, and an intracellularly designating peptide, we successfully developed cancer-specific and efficacious ricin-based RITs. This approach can be applied to develop novel protein-based "magic bullets" to effectively suppress tumors that are resistant to conventional anti-cancer drugs.


Asunto(s)
Inmunotoxinas , Neoplasias , Ricina , Animales , Apoptosis , Endocitosis , Inmunotoxinas/metabolismo , Inmunotoxinas/farmacología , Ratones , Neoplasias/tratamiento farmacológico , Señales de Clasificación de Proteína , Ricina/farmacología , Ricina/toxicidad
4.
Biomacromolecules ; 22(6): 2649-2658, 2021 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-34060808

RESUMEN

The successful development of targeted nanoparticle (NP)-based therapeutics depends on the effective conjugation of targeting ligands to the NP. However, conventional methods based on chemical reactive groups such as N-hydroxysuccinimide, 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide, and maleimide have several limitations, including low binding efficiency, complex reaction methods, long reaction times, and reduced activity of the targeting ligand. In this study, we developed a novel method for conjugating targeting ligands to albumin NPs using the recently developed bacterial superglue the SpyTag/SpyCatcher (ST/SC) ligation system. This method involves a rapid one-step conjugation process with almost 100% efficiency. Albumin NPs conjugated to human epidermal growth factor receptor 2 (HER2) affibody molecules using the ST/SC system showed strong binding to HER2-overexpressing cells. In addition, NPs encapsulated with indocyanine green accumulated in cells overexpressing HER2 and exhibited superior photothermal treatment effects. Thus, surface functionalization of NPs using the ST/SC reaction may be used to develop new nanosystems that exhibit improved therapeutic benefits.


Asunto(s)
Nanopartículas , Terapia Fototérmica , Albúminas , Línea Celular Tumoral , Humanos , Ligandos , Receptor ErbB-2
5.
Biomacromolecules ; 22(7): 3028-3039, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-34142815

RESUMEN

Protein cage nanoparticles have a unique spherical hollow structure that provides a modifiable interior space and an exterior surface. For full application, it is desirable to utilize both the interior space and the exterior surface simultaneously with two different functionalities in a well-combined way. Here, we genetically engineered encapsulin protein cage nanoparticles (Encap) as modular nanoplatforms by introducing a split-C-intein (IntC) fragment and SpyTag into the interior and exterior surfaces, respectively. A complementary split-N-intein (IntN) was fused to various protein cargoes, such as NanoLuc luciferase (Nluc), enhanced green fluorescent protein (eGFP), and Nluc-miniSOG, individually, which led to their successful encapsulation into Encaps to form Cargo@Encap through split intein-mediated protein ligation during protein coexpression and cage assembly in bacteria. Conversely, the SpyCatcher protein was fused to various protein ligands, such as a glutathione binder (GST-SC), dimerizing ligands (FKBP12-SC and FRB-SC), and a cancer-targeting affibody (SC-EGFRAfb); subsequently, they were displayed on Cargo@Encaps through SpyTag/SpyCatcher ligation to form Cargo@Encap/Ligands in a mix-and-match manner. Nluc@Encap/glutathione-S-transferase (GST) was effectively immobilized on glutathione (GSH)-coated solid supports exhibiting repetitive and long-term usage of the encapsulated luciferases. We also established luciferase-embedded layer-by-layer (LbL) nanostructures by alternately depositing Nluc@Encap/FKBP12 and Nluc@Encap/FRB in the presence of rapamycin and applied enhanced green fluorescent protein (eGFP)@Encap/EGFRAfb as a target-specific fluorescent imaging probe to visualize specific cancer cells selectively. Modular functionalization of the interior space and the exterior surface of a protein cage nanoparticle may offer the opportunity to develop new protein-based nanostructured devices and nanomedical tools.


Asunto(s)
Nanopartículas , Neoplasias , Colorantes Fluorescentes , Humanos , Inteínas , Ligandos
6.
Anal Chem ; 92(7): 5473-5481, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32142265

RESUMEN

In general immunoassays, secondary antibodies are covalently linked with enzymes and bind to the Fc region of target-bound primary antibodies to amplify signals of low-abundant target molecules. The antibodies themselves are obtained from large mammals and are further modified with enzymes. In this study, we developed novel recombinant immunoglobulin G (IgG)-binding luciferase-based signal amplifiers (rILSAs) by genetically fusing luciferase (Nluc) with antimouse IgG1 nanobody (MG1Nb) and antibody-binding domain (ABD), individually or together, in a mix-and-match manner. We obtained three different highly pure rILSAs in large quantities using a bacterial overexpression system and one-step purification. Mouse-specific rILSA, MG1Nb-Nluc, and rabbit-specific rILSA, Nluc-ABD, selectively bound to target-molecule-bound mouse IgG1 and rabbit IgG primary antibodies, whereas the bispecific rILSA, MG1Nb-Nluc-ABD, mutually bound to both mouse IgG1 and rabbit IgG primary antibodies. All rILSAs exhibited an outstanding signal-amplifying capability comparable to those of conventional horseradish-peroxidase-conjugated secondary antibodies, regardless of the target molecules, in various immunoassay formats, such as enzyme-linked immunosorbent assay, Western blot, and lateral flow assays. Each rILSA was selected for its own individual purpose and applied to various types of target analytes, in combination with a variety of target-specific primary antibodies, effectively minimizing the use of animals as well as reducing the costs and time associated with the production and chemical conjugation of signal-amplifying enzymes.


Asunto(s)
Inmunoensayo/métodos , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Luciferasas/metabolismo , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Animales , Especificidad de Anticuerpos , Ratones
7.
Biotechnol Bioeng ; 116(11): 2843-2851, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31329283

RESUMEN

Protein cage nanoparticles are widely used as targeted delivery nanoplatforms, because they have well-defined symmetric architectures, high biocompatibility, and enough plasticity to be modified to produce a range of different functionalities. Targeting peptides and ligands are often incorporated on the surface of protein cage nanoparticles. In this research, we adopted the SpyTag/SpyCatcher protein ligation system to covalently display target-specific affibody molecules on the exterior surface of bacteriophage P22 virus-like particles (VLP) and evaluated their modularity and efficacy of targeted delivery. We genetically introduced the 13 amino acid SpyTag peptide into the C-terminus of the P22 capsid protein to construct a target-tunable nanoplatform. We constructed two different SpyCatcher-fused affibody molecules as targeting ligands, SC-EGFRAfb and SC-HER2Afb, which selectively bind to EGFR and HER2 surface markers, respectively. We produced target-specific P22 VLP-based delivery nanoplatforms for the target cell lines by selectively combining SpyTagged P22 VLP and SC-fused affibody molecules. We confirmed its target-switchable modularity through cell imaging and verified the target-specific drug delivery efficacy of the affibody molecules displaying P22 VLP using cell viability assays. The P22 VLP-based delivery nanoplatforms can be used as multifunctional delivery vehicles by ligating other functional proteins, as well as affibody molecules. The interior cavity of P22 VLP can be also used to load cargoes like enzymes and therapeutic proteins. We anticipate that the nanoplatforms will provide new opportunities for developing target-specific functional protein delivery systems.


Asunto(s)
Antineoplásicos Inmunológicos , Bacteriófago P22 , Sistemas de Liberación de Medicamentos , Nanopartículas/química , Anticuerpos de Cadena Única , Virión , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/farmacología , Bacteriófago P22/química , Bacteriófago P22/genética , Línea Celular Tumoral , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/farmacología , Virión/química , Virión/genética
8.
Small ; 14(35): e1801488, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30066359

RESUMEN

Fabrication of functional nanostructures is a prominent issue in nanotechnology, because they often exhibit unique properties that are different from the individual building blocks. Protein cage nanoparticles are attractive nanobuilding blocks for constructing nanostructures due to their well-defined symmetric spherical structures, polyvalent nature, and functional plasticity. Here, a lumazine synthase protein cage nanoparticle is genetically modified to be used as a template to generate functional nanobuilding blocks and covalently display enzymes (ß-lactamase) and protein ligands (FKBP12/FRB) on its surface, making dual-functional nanobuilding blocks. Nanoreaction clusters are subsequently created by ligand-mediated alternate deposition of two complementary building blocks using layer-by-layer (LbL) assemblies. 3D nanoreaction clusters provide enhanced enzymatic activity compared with monolayered building block arrays. The approaches described here may provide new opportunities for fabricating functional nanostructures and nanoreaction clusters, leading to the development of new protein nanoparticle-based nanostructured biosensor devices.


Asunto(s)
Nanoestructuras/química , Nanotecnología/métodos , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Ligandos , Nanoestructuras/ultraestructura , Péptidos/química , Multimerización de Proteína , Pteridinas/metabolismo , Sirolimus/farmacología , Proteína 1A de Unión a Tacrolimus/metabolismo
9.
Biomacromolecules ; 19(7): 2896-2904, 2018 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-29847113

RESUMEN

The selective detection of specific cells of interest and their effective visualization is important but challenging, and fluorescent cell imaging with target-specific probes is commonly used to visualize cell morphology and components and to track cellular processes. Multiple displays of two or more targeting ligands on a polyvalent single template would make it possible to construct versatile multiplex fluorescent cell imaging probes that can visualize two or more target cells individually without the need for a set of individual probes. To achieve this goal, we used encapsulin, a new class of protein cage nanoparticles, as a template and implanted dual targeting capability by presenting two different affibody molecules on a single encapsulin protein cage nanoparticle post-translationally. Encapsulin was self-assembled from 60 identical subunits to form a hollow and symmetric spherical structure with a uniform size. We genetically inserted SpyTag peptides onto the encapsulin surface and prepared various SpyCatcher-fused proteins, such as fluorescent proteins and targeting affibody molecules. We successfully displayed fluorescent proteins and affibody molecules together on a single encapsulin in a mix-and-match manner post-translationally using bacterial superglue, the SpyTag/SpyCatcher ligation system, and demonstrated that these dual functional encapsulins can be used as target-specific fluorescent cell imaging probes. Dual targeting protein cage nanoparticles were further constructed by ligating two different affibody molecules onto the encapsulin surface with fluorescent dyes, and they effectively recognized and bound to two individual targeting cells independently, which could be visualized by selective colors on demand.


Asunto(s)
Proteínas Bacterianas/química , Nanopartículas/química , Proteínas Bacterianas/genética , Línea Celular , Colorantes Fluorescentes/química , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Humanos , Células MCF-7 , Microscopía Fluorescente/métodos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Thermotoga maritima/enzimología
10.
Adv Exp Med Biol ; 1064: 27-43, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30471024

RESUMEN

Protein cage nanoparticles are made of biomaterials, proteins, and have well-defined cage-like architectures designed and built by nature. They are composed of multiple copies of one or a small number of chemically identical subunits having a highly uniform nano-size and symmetric structure. Protein cage nanoparticles have genetic and chemical plasticity amenable to simultaneously introducing multiple cell-specific targeting ligands, diagnostic agents, and their corresponding therapeutic agents at desired sites depending on its purpose. A wide range of protein cage nanoparticles, such as ferritin, lumazine synthase, encapsulin, and virus-like particles, has been extensively explored and utilized in biomedical fields as effective delivery nanoplatforms of diagnostics and/or therapeutics. Highly biocompatible and plastic protein cage nanoparticles may provide a new paradigm for developing simple, but versatile in vivo delivery systems.


Asunto(s)
Sistemas de Liberación de Medicamentos , Nanopartículas , Preparaciones Farmacéuticas , Proteínas/química , Ferritinas/química , Ligandos , Complejos Multienzimáticos/química
11.
J Am Chem Soc ; 139(35): 12121-12124, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28814075

RESUMEN

Transition-metal-catalyzed or metal-free azide-alkyne cycloadditions are methods to access 1,4- or 1,5-disubstituted 1,2,3-triazoles. Although the copper-catalyzed cycloaddition to access 1,4-disubstituted products has been applied to biomolecular reaction systems, the azide-alkyne cycloaddition to access the complementary 1,5-regioisomers under aqueous and ambient conditions remains a challenge due to limited substrate scope or moisture-/air-sensitive catalysts. Herein, we report a method to access 1,5-disubstituted 1,2,3-triazoles using a Cp2Ni/Xantphos catalytic system. The reaction proceeds both in water and organic solvents at room temperature. This protocol is simple and scalable with a broad substrate scope including both aliphatic and aromatic substrates. Moreover, triazoles attached with carbohydrates or amino acids are prepared via this cycloaddition.

12.
Bioconjug Chem ; 27(9): 2007-13, 2016 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-27504746

RESUMEN

Specific recognitions of pathogen associated molecular patterns by Toll-like receptors (TLRs) initiate dendritic cell (DC) activation, which is critical for coordinating innate and adaptive immune responses. Imidazoquinolines as small-molecule TLR7 agonists often suffer from prompt dissemination and short half-life in the bloodstream, preventing their localization to the corresponding receptors and effective DC activation. We postulated that covalent incorporation of imidazoquinoline moieties onto the surface of biocompatible nanoparticles (∼30 nm size) would enhance their chemical stability, cellular uptake efficiency, and adjuvanticity. The fully synthetic adjuvant-nanocomplexes led to successful DC activation at lower nanomolar doses compared with free small-molecule agonists. Once a model antigen such as ovalbumin was used for immunization, we found that the nanocomplexes promoted an unusually strong cytotoxic T lymphocyte response, revealing their unique immunostimulatory capacity benefiting from multivalency and efficient transport to endosomal TLR7.


Asunto(s)
Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Nanopartículas/química , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Relación Dosis-Respuesta a Droga
13.
Sensors (Basel) ; 15(5): 10481-510, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25951336

RESUMEN

Surface plasmon resonance (SPR) is a label-free detection method which has emerged during the last two decades as a suitable and reliable platform in clinical analysis for biomolecular interactions. The technique makes it possible to measure interactions in real-time with high sensitivity and without the need of labels. This review article discusses a wide range of applications in optical-based sensors using either surface plasmon resonance (SPR) or surface plasmon resonance imaging (SPRI). Here we summarize the principles, provide examples, and illustrate the utility of SPR and SPRI through example applications from the biomedical, proteomics, genomics and bioengineering fields. In addition, SPR signal amplification strategies and surface functionalization are covered in the review.


Asunto(s)
Técnicas Biosensibles/métodos , Resonancia por Plasmón de Superficie/métodos
14.
Biomacromolecules ; 15(10): 3794-801, 2014 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-25180761

RESUMEN

Protein cage nanoparticles are excellent candidates for use as multifunctional delivery nanoplatforms because they are built from biomaterials and have a well-defined structure. A novel protein cage nanoparticle, encapsulin, isolated from thermophilic bacteria Thermotoga maritima, is prepared and developed as a versatile template for targeted delivery nanoplatforms through both chemical and genetic engineering. It is pivotal for multifunctional delivery nanoplatforms to have functional plasticity and versatility to acquire targeting ligands, diagnostic probes, and drugs simultaneously. Encapsulin is genetically engineered to have unusual heat stability and to acquire multiple functionalities in a precisely controlled manner. Hepatocellular carcinoma (HCC) cell binding peptide (SP94-peptide, SFSIIHTPILPL) is chosen as a targeting ligand and displayed on the surface of engineered encapsulin (Encap_loophis42C123) through either chemical conjugation or genetic insertion. The effective and selective targeted delivery of SP94-peptide displaying encapsulin (SP94-Encap_loophis42C123) to HepG2 cells is confirmed by fluorescent microscopy imaging. Aldoxorubicin (AlDox), an anticancer prodrug, is chemically loaded to SP94-Encap_loophis42C123 via thiol-maleimide Michael-type addition, and the efficacy of the delivered drugs is evaluated with a cell viability assay. SP94-Encap_loophis42C123-AlDox shows comparable killing efficacy with that of free drugs without the platform's own cytotoxicity. Functional plasticity and versatility of the engineered encapsulin allow us to introduce targeting ligands, diagnostic probes, and therapeutic reagents simultaneously, providing opportunities to develop multifunctional delivery nanoplatforms.


Asunto(s)
Nanopartículas/administración & dosificación , Proteínas/administración & dosificación , Proteínas/genética , Carcinoma Hepatocelular/tratamiento farmacológico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Células Hep G2 , Humanos , Ligandos , Neoplasias Hepáticas/tratamiento farmacológico , Microscopía Fluorescente/métodos , Péptidos/administración & dosificación , Péptidos/genética , Ingeniería de Proteínas/métodos
15.
Microb Cell Fact ; 13: 68, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24885571

RESUMEN

BACKGROUND: Inclusion bodies (IBs) were generally considered to be inactive protein deposits and did not hold any attractive values in biotechnological applications. Recently, some IBs of recombinant proteins were confirmed to show their functional properties such as enzyme activities, fluorescence, etc. Such biologically active IBs are not commonly formed, but they have great potentials in the fields of biocatalysis, material science and nanotechnology. RESULTS: In this study, we characterized the IBs of DL4, a deletion variant of green fluorescent protein which forms active intracellular aggregates. The DL4 proteins expressed in Escherichia coli were exclusively deposited to IBs, and the IBs were estimated to be mostly composed of active proteins. The spectral properties and quantum yield of the DL4 variant in the active IBs were almost same with those of its native protein. Refolding and stability studies revealed that the deletion mutation in DL4 didn't affect the folding efficiency of the protein, but destabilized its structure. Analyses specific for amyloid-like structures informed that the inner architecture of DL4 IBs might be amorphous rather than well-organized. The diameter of fluorescent DL4 IBs could be decreased up to 100-200 nm by reducing the expression time of the protein in vivo. CONCLUSIONS: To our knowledge, DL4 is the first GFP variant that folds correctly but aggregates exclusively in vivo without any self-aggregating/assembling tags. The fluorescent DL4 IBs have potentials to be used as fluorescent biomaterials. This study also suggests that biologically active IBs can be achieved through engineering a target protein itself.


Asunto(s)
Proteínas Fluorescentes Verdes/metabolismo , Cuerpos de Inclusión/metabolismo , Escherichia coli/metabolismo , Proteínas Fluorescentes Verdes/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Cinética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Microscopía Confocal , Mutación , Nanopartículas/química , Nanopartículas/metabolismo , Pliegue de Proteína , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación , Temperatura , Factores de Tiempo
16.
Appl Microbiol Biotechnol ; 98(19): 8281-90, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24965559

RESUMEN

Based on recent developments, virus-like particles (VLPs) are considered to be perfect candidates as nanoplatforms for applications in materials science and medicine. To succeed, mass production of VLPs and self-assembly into a correct form in plant systems are key factors. Here, we report expression of synthesized coat proteins of the three viruses, Brome mosaic virus, Cucumber mosaic virus, and Maize rayado fino virus, in Nicotiana benthamiana and production of self-assembled VLPs by transient expression system using agroinfiltration. Each coat protein was synthesized and cloned into a pBYR2fp single replicon vector. Target protein expression in cells containing p19 was fourfold higher than that of cells lacking p19. After agroinfiltration, protein expression was analyzed by SDS-PAGE and quantitative image analyzer. Quantitative analysis showed that BMVCP, CMVCP, and MRFVCP concentrations were 0.5, 1.0, and 0.8 mg · g(-1) leaf fresh weight, respectively. VLPs were purified by sucrose cushion ultracentrifugation and then analyzed by transmission electron microscopy. Our results suggested that BMVCP and CMVCP proteins expressed in N. benthamiana leaves were able to correctly self-assemble into particles. Moreover, we evaluated internal cavity accessibility of VLPs to load foreign molecules. Finally, plant growth conditions after agroinfiltration are critical for increasing heterologous protein expression levels in a transient expression system.


Asunto(s)
Proteínas de la Cápside/metabolismo , Vectores Genéticos/genética , Nicotiana/genética , Replicón , Virión/metabolismo , Biotecnología , Bromovirus/genética , Bromovirus/metabolismo , Proteínas de la Cápside/genética , Cucumovirus/genética , Cucumovirus/metabolismo , Expresión Génica , Vectores Genéticos/metabolismo , Nicotiana/metabolismo , Tymoviridae/genética , Tymoviridae/metabolismo , Virión/genética
17.
Nanomedicine ; 10(3): 561-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24262997

RESUMEN

We utilized ferritin protein cage nanoparticles (FPCN) as antigen delivery nanoplatforms for DC-based vaccine development and investigated DC-mediated antigen-specific immune responses. Antigenic peptides, OT-1 (SIINFEKL) or OT-2 (ISQAVHAAHAEINEAGR) which are derived from ovalbumin, were genetically introduced either onto the exterior surface or into the interior cavity of FPCN. FPCN carrying antigenic peptides (OT-1-FPCN and OT-2-FPCN) were effectively delivered to DCs and processed within endosomes. Delivered antigenic peptides, OT-1 or OT-2, to DCs successfully induced antigen-specific CD8(+) or CD4(+) T cell proliferations both in vitro and in vivo. Naïve mice immunized with OT-1-FPCN efficiently differentiated OT-1 specific CD8(+) T cells into functional effector cytotoxic T cells resulting in selective killing of antigen-specific target cells. Effective differentiation of proliferated OT-2 specific CD4(+) T cells into functional CD4(+) Th1 and Th2 cells was confirmed with the productions of IFN-γ/IL-2 and IL-10/IL-13 cytokines, respectively. FROM THE CLINICAL EDITOR: In this study, the authors utilized ferritin protein cage nanoparticles as antigen delivery nanoplatforms for dendritic cell-based vaccine development and investigated DC-mediated antigen-specific immune responses using strong model antigens derived from ovalbumin, suggesting potential future clinical applicability of this or similar techniques.


Asunto(s)
Antígenos/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Células Dendríticas/inmunología , Ferritinas/química , Nanopartículas/química , Ovalbúmina/administración & dosificación , Secuencia de Aminoácidos , Animales , Antígenos/química , Antígenos/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Células Cultivadas , Citocinas/inmunología , Células Dendríticas/citología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Ovalbúmina/química , Ovalbúmina/inmunología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Células TH1/citología , Células TH1/inmunología , Células Th2/citología , Células Th2/inmunología
18.
Int J Biol Macromol ; 261(Pt 1): 129622, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38266854

RESUMEN

Intracellular protein delivery systems have great potential in the fields of therapeutics development and biomedical research. However, targeted delivery, passing through the cell membrane without damaging the cells, and escaping from endosomal entrapment of endocytosed molecular cargos are major challenges of the system. Here, we present a novel intracellular protein delivery system based on modularly engineered botulinum neurotoxin type A (BoNT/A). LHNA domain, consisting of light chain and endosomal escape machinery of BoNT/A, was genetically fused with SpyCatcher (SC) and EGFR targeting affibody (EGFRAfb) to create SC-LHNA-EGFRAfb, a target-specific and protein cargo-switchable BoNT/A-based intracellular protein delivery platform. SC-LHNA-EGFRAfb was purely purified in large quantities, efficiently ligated with multiple ST-fused protein cargos individually, generating a variety of protein cargo-containing intracellular delivery complexes, and successfully delivered ligated protein cargos into the cytosol of target cells via receptor-mediated endocytosis, followed by endosomal escape and subsequent cytosolic delivery. SC-LHNA-EGFRAfb enhanced intracellular delivery efficiency of protein toxin, gelonin, by approximately 100-fold, highlighting the crucial roles of EGFRAfb and LHNA domain as a targeting ligand and an endosomal escape machinery, respectively, in the delivery process. The BoNT-based plug-and-deliverable intracellular protein delivery system has the potential to expand its applications in protein therapeutics and manipulating cellular processes.


Asunto(s)
Toxinas Botulínicas Tipo A , Citosol/metabolismo , Endosomas/metabolismo , Endocitosis
19.
Biomacromolecules ; 14(10): 3515-22, 2013 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-23962280

RESUMEN

Supramolecular nanogel, a physically cross-linked nanosize hydrogel, spontaneously self-assembles in aqueous solution via secondary interactions and is thus of great interest in nanomedicine as a drug carrier. We developed a versatile method for supramolecular nanogel self-assembled by electrostatic interaction between positive surfactant micelles and negative polypeptides. Core-shell-like structures of supramolecular nanogels provide stable hydrophobic pockets that prevent simple diffusion of hydrophobic guest molecules, resulting in high encapsulation stability. The size of the supramolecular nanogels can be systematically controlled by varying the size of the surfactant micelles. Furthermore, noncovalently encapsulated dye molecules can be released in response to matrix metalloproteinases highly overexpressed in tumor tissues, potentially providing tumor-triggered targeting.


Asunto(s)
Antineoplásicos/farmacología , Productos Biológicos/química , Doxorrubicina/farmacología , Péptidos/química , Polietilenglicoles/química , Polietileneimina/química , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Productos Biológicos/síntesis química , Proteínas Sanguíneas/química , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Ensayos de Selección de Medicamentos Antitumorales , Células HeLa , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Sustancias Macromoleculares/síntesis química , Sustancias Macromoleculares/química , Micelas , Modelos Moleculares , Estructura Molecular , Nanogeles , Péptidos/síntesis química , Polietilenglicoles/síntesis química , Polietileneimina/síntesis química , Electricidad Estática , Relación Estructura-Actividad , Tensoactivos/química , Células Tumorales Cultivadas
20.
Biomacromolecules ; 14(7): 2332-9, 2013 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-23758486

RESUMEN

P22 viral capsids and ferritin protein cages are utilized as templating macromolecules to conjugate Gd(III)-chelating agent complexes, and we systematically investigates the effects of the macromolecules' size and the conjugation positions of Gd(III)-chelating agents on the magnetic resonance (MR) relaxivities and the resulting image contrasts. The relaxivity values of the Gd(III)-chelating agent-conjugated P22 viral capsids (outer diameter: 64 nm) are dramatically increased as compared to both free Gd(III)-chelating agents and Gd(III)-chelating agent-conjugated ferritins (outer diameter: 12 nm), suggesting that the large sized P22 viral capsids exhibit a much slower tumbling rate, which results in a faster T1 relaxation rate. Gd(III)-chelating agents are attached to either the interior or exterior surface of P22 viral capsids and the conjugation positions of Gd(III)-chelating agents, however, do not have a significant effect on the relaxivity values of the macromolecular conjugates. The contrast enhancement of Gd(III)-chelating agent-conjugated P22 viral capsids is confirmed by in vitro phantom imaging at a short repetition times (TR) and the potential usage of Gd(III)-chelating agent-conjugated P22 viral capsids for in vivo MR imaging is validated by visualizing a mouse's intravascular system, including the carotid, mammary arteries, the jugular vein, and the superficial vessels of the head at an isotropic resolution of 250 µm.


Asunto(s)
Bacteriófago P22 , Vasos Sanguíneos/anatomía & histología , Cápside , Medios de Contraste/química , Imagen por Resonancia Magnética/métodos , Animales , Quelantes/química , Ferritinas/química , Gadolinio/química , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA