Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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
2.
J Colloid Interface Sci ; 652(Pt A): 184-194, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37595436

RESUMEN

The development of quantum dot (QD)-based modular bioprobe that has a compact size and enable a facile conjugation of various biofunctional groups is in high demand. To address this, we surface engineered QDs with zwitterion polymer ligands to have an inherent compact size and derivatized them sequentially with the recombinant proteins SpyCatcher/SpyTag (SC/ST) to use their protein ligation system. SC/ST spontaneously form one complex through the isopeptide bond between them. SC-conjugated QDs (QD-SC) were used as base building blocks. Then, ST-biomolecules were added for modular biofunctionalization. We synthesized compact sized (∼15 nm) QD-SC-ST-affibody (antibody-mimicking small protein for tumor detection) conjugates, which showed successful cell-receptor targeting. The target cell-receptor could be easily tuned by changing the type of ST-affibody. We also demonstrated that anti-human-chorionic-gonadotropin mouse IgG1 antibodies can be labeled on the QD surface by mixing QD-SC with the ST-MG1Nb (mouse-IgG1-specific protein). The immunoassay performance of the antibody-labeled QDs was evaluated using a pregnancy test kit, displaying equivalent detection sensitivity to a commercially available kit. This study proposed an innovative strategy for QD biofunctionalization in a modular manner, which can be expanded to a diverse range of colloidal particle derivatization.


Asunto(s)
Puntos Cuánticos , Ratones , Animales , Polímeros , Proteínas Recombinantes/química , Inmunoglobulina G
3.
Int J Biol Macromol ; 251: 126299, 2023 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-37573903

RESUMEN

In conventional immunoassays, a secondary antibody is used to amplify the signal generated by the binding of the primary antibody to the target analyte. Due to concerns regarding animal use and cost-inefficiency of secondary antibody productions, there is a significant demand for the development of recombinant secondary antibody mimics (rSAMs). Here, we developed rSAMs using a signal-generating enzyme, monomeric alkaline phosphatase (mALP), and antibody-binders, including monomeric streptavidin (mSA2) and mouse IgG1- or rabbit IgG-binding nanobodies (MG1Nb or RNb). The mALP-MG1Nb, mALP-RNb, and mALP-mSA2 were genetically constructed and produced in large quantities using bacterial overexpression systems, which reduced manufacturing costs and time without the use of animals. Each rSAM exhibited high and selective binding to its respective primary antibody, generating linear band signals corresponding to the amounts of target analytes in western blots. The rSAMs also successfully generated sigmoidal signal curves that increased as the sample concentration increased. Moreover, they generated stronger signals than conventional ALP-conjugated secondary antibodies and SA, particularly in the medium to high sample concentration range, in both indirect and sandwich-type indirect ELISAs at the same sample concentration. The rSAMs we developed here may provide new insights to develop novel immunoassay-based analytical and diagnostic tools.

4.
Int J Biol Macromol ; 231: 123577, 2023 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36758763

RESUMEN

Aggressive tumor formation often leads to excessive anaerobic glycolysis and massive production and accumulation of lactate in the tumor microenvironment (TME). To significantly curb lactate accumulation in TME, in this study, lactate oxidase (LOX) was used as a potential therapeutic enzyme and signal regulatory protein α variant (vSIRPα) as a tumor cell targeting ligand. SpyCatcher protein and SpyTag peptide were genetically fused to LOX and vSIRPα, respectively, to form SC-LOX and ST-vSIRPα and tumor-targeting LOX/vSIRPα conjugates were constructed via a SpyCatcher/SpyTag protein ligation system. LOX/vSIRPα conjugates selectively bound to the CD47-overexpressing mouse melanoma B16-F10 cells and effectively consumed lactate produced by the B16-F10 cells, generating adequate amounts of hydrogen peroxide (H2O2), which induces drastic necrotic tumor cell death. Local treatments of B16-F10 tumor-bearing mice with LOX/vSIRPα conjugates significantly suppressed B16-F10 tumor growth in vivo without any severe side effects. Tumor-targeting vSIRPα may allow longer retention of LOX in tumor sites, effectively consuming surrounding lactate in TME and locally generating adequate amounts of cytotoxic H2O2 to suppress tumor growth. The approach restraining the local lactate concentration and H2O2 in TME using LOX and vSIRPα could offer new opportunities for developing enzyme/targeting ligand conjugate-based therapeutic tools for tumor treatment.


Asunto(s)
Peróxido de Hidrógeno , Neoplasias , Animales , Ratones , Peróxido de Hidrógeno/metabolismo , Ligandos , Necrosis , Ácido Láctico , Microambiente Tumoral
5.
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
6.
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
7.
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
8.
J Control Release ; 349: 367-378, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35809662

RESUMEN

The TNF-related apoptosis-inducing ligand (TRAIL) is a promising anticancer drug candidate because it selectively binds to the proapoptotic death receptors, which are frequently overexpressed in a wide range of cancer cells, subsequently inducing strong apoptosis in these cells. However, the therapeutic benefit of TRAIL has not been clearly proven, mainly because of its poor pharmacokinetic characteristics and frequent resistance to its application caused by the activation of a survival signal via the EGF/epidermal growth factor receptor (EGFR) signaling pathway. Here, a lumazine synthase protein cage nanoparticle isolated from Aquifex aeolicus (AaLS) was used as a multiple ligand-displaying nanoplatform to display polyvalently both TRAIL and EGFR binding affibody molecules (EGFRAfb) via a SpyTag/SpyCatcher protein-ligation system, to form AaLS/TRAIL/EGFRAfb. The dual-ligand-displaying AaLS/TRAIL/EGFRAfb exhibited a dramatically enhanced cytotoxicity on TRAIL-resistant and EGFR-overexpressing A431 cancer cells in vitro, effectively disrupting the EGF-mediated EGFR survival signaling pathway by blocking EGF/EGFR binding as well as strongly activating both the extrinsic and intrinsic apoptotic pathways synergistically. The AaLS/TRAIL/EGFRAfb selectively targeted A431 cancer cells in vitro and actively reached the tumor sites in vivo. The A431 tumor-bearing mice treated with AaLS/TRAIL/EGFRAfb exhibited a significant suppression of the tumor growth without any significant side effects. Collectively, these findings showed that the AaLS/TRAIL/EGFRAfb could be used as an effective protein-based therapeutic for treating EGFR-positive cancers, which are difficult to manage using mono-therapeutic approaches.


Asunto(s)
Antineoplásicos , Nanopartículas , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis , Línea Celular Tumoral , Factor de Crecimiento Epidérmico , Receptores ErbB/metabolismo , Ligandos , Ratones , Receptores de Muerte Celular , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
9.
Biosens Bioelectron ; 213: 114441, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-35696868

RESUMEN

Lateral flow assays (LFA) enable development of portable and rapid diagnostic kits; however, their capacity to detect low levels of disease markers remains poor. Here, we report a highly sensitive pregnancy test kit as a proof of concept, by combining brush-type ligand-coated quantum beads (B-type QBs) and nanobody, which can control the antibody orientation and enhance sensitivity. The brush-type ligand provided excellent dispersion stability and high-binding capacity toward antibody. Fc-binding nanobody increased the antigen-binding capacity of conjugated antibodies on the B-type QBs. To facilitate convenient acquisition of the LFA results, we developed a smartphone-based reader with a 3D-printed optical imaging module, and validated the diagnostic performance of the sensing platform. The pregnancy test kit achieved a 5.1 pg mL-1 limit of detection, corresponding to the levels for early-stage detection of heart disease and malaria. Our LFA application can potentially be expanded to diagnosis other diseases by simply changing the antibody pair in the kit.


Asunto(s)
Técnicas Biosensibles , Pruebas de Embarazo , Anticuerpos , Técnicas Biosensibles/métodos , Femenino , Humanos , Ligandos , Embarazo
10.
Sci Rep ; 12(1): 1005, 2022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-35046461

RESUMEN

The pandemic of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has caused a public health emergency, and research on the development of various types of vaccines is rapidly progressing at an unprecedented development speed internationally. Some vaccines have already been approved for emergency use and are being supplied to people around the world, but there are still many ongoing efforts to create new vaccines. Virus-like particles (VLPs) enable the construction of promising platforms in the field of vaccine development. Here, we demonstrate that non-infectious SARS-CoV-2 VLPs can be successfully assembled by co-expressing three important viral proteins membrane (M), envelop (E) and nucleocapsid (N) in plants. Plant-derived VLPs were purified by sedimentation through a sucrose cushion. The shape and size of plant-derived VLPs are similar to native SARS-CoV-2 VLPs without spike. Although the assembled VLPs do not have S protein spikes, they could be developed as formulations that can improve the immunogenicity of vaccines including S antigens, and further could be used as platforms that can carry S antigens of concern for various mutations.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , Proteínas M de Coronavirus/inmunología , Proteínas de la Nucleocápside de Coronavirus/inmunología , SARS-CoV-2/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Proteínas Viroporinas/inmunología , COVID-19/prevención & control , COVID-19/virología , Vacunas contra la COVID-19/administración & dosificación , Proteínas M de Coronavirus/genética , Proteínas M de Coronavirus/metabolismo , Proteínas de la Nucleocápside de Coronavirus/genética , Proteínas de la Nucleocápside de Coronavirus/metabolismo , Humanos , Nicotiana/inmunología , Nicotiana/metabolismo , Nicotiana/virología , Vacunas de Partículas Similares a Virus/genética , Vacunas de Partículas Similares a Virus/metabolismo , Proteínas Viroporinas/genética , Proteínas Viroporinas/metabolismo
11.
BMB Rep ; 55(4): 175-180, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34814976

RESUMEN

Peptides are gaining substantial attention as therapeutics for human diseases. However, they have limitations such as low bioavailability and poor pharmacokinetics. Periostin, a matricellular protein, can stimulate the repair of ischemic tissues by promoting angiogenesis. We have previously reported that a novel angiogenic peptide (amino acids 142-151) is responsible for the pro-angiogenic activity of periostin. To improve the in vivo delivery efficiency of periostin peptide (PP), we used proteins self-assembled into a hollow cage-like structure as a drug delivery nanoplatform in the present study. The periostin peptide was genetically inserted into lumazine synthase (isolated from Aquifex aeolicus) consisting of 60 identical subunits with an icosahedral capsid architecture. The periostin peptide-bearing lumazine synthase protein cage nanoparticle with 60 periostin peptides multivalently displayed was expressed in Escherichia coli and purified to homogeneity. Next, we examined angiogenic activities of this periostin peptide-bearing lumazine synthase protein cage nanoparticle. AaLS-periostin peptide (AaLS-PP), but not AaLS, promoted migration, proliferation, and tube formation of human endothelial colony-forming cells in vitro. Intramuscular injection of PP and AaLS-PP increased blood perfusion and attenuated severe limb loss in the ischemic hindlimb. However, AaLS did not increase blood perfusion or alleviate tissue necrosis. Moreover, in vivo administration of AaLS-PP, but not AaLS, stimulated angiogenesis in the ischemic hindlimb. These results suggest that AaLS is a highly useful nanoplatform for delivering pro-angiogenic peptides such as PP. [BMB Reports 2022; 55(4): 175-180].


Asunto(s)
Nanopartículas , Neovascularización Patológica , Animales , Miembro Posterior , Humanos , Isquemia/tratamiento farmacológico , Isquemia/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Fisiológica , Péptidos/farmacología
12.
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
13.
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
14.
J Control Release ; 335: 269-280, 2021 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-34044091

RESUMEN

Magnetic resonance imaging (MRI) is a non-invasive in vivo imaging tool, providing high enough spatial resolution to obtain both the anatomical and the physiological information of patients. However, MRI generally suffers from relatively low sensitivity often requiring the aid of contrast agents (CA) to enhance the contrast of vessels and/or the tissues of interest from the background. The targeted delivery of diagnostic probes to the specific lesion is a powerful approach for early diagnosis and signal enhancement leading to the effective treatment of various diseases. Here, we established targeting ligand switchable nanoplatforms using lumazine synthase protein cage nanoparticles derived from Aquifex aeolicus (AaLS) by genetically introducing the SpyTag peptide (ST) to the C-terminus of the AaLS subunits to form an ST-displaying AaLS (AaLS-ST). Conversely, multiple targeting ligands were constructed by genetically fusing SpyCatcher protein (SC) to either HER2 or EGFR targeting affibody molecules (SC-HER2Afb or SC-EGFRAfb). Gd(III)-DOTA complexes were chemically attached to the AaLS-ST and the external surface of the Gd(III)-DOTA conjugated AaLS-ST (Gd(III)-DOTA-AaLS-ST) were successfully decorated with either the HER2Afb or the EGFRAfb. The resulting Gd(III)-DOTA-AaLS/HER2Afb and Gd(III)-DOTA-AaLS/EGFR2Afb exhibited high r1 relaxivity values of 57 and 25 mM-1 s-1 at 1.4 and 7 T, respectively, which were 10-fold or higher than those of the clinically used Dotarem. Their target-selective contrast enhancements were confirmed with in vitro cell-based MRI scans and the in vivo MR imaging of tumor-bearing mouse models at 7 T. A target-switchable AaLS-based nanoplatform that was developed in this study might serve as a promising T1 CA developing platform at a high magnetic field to detect various tumor sites in a target-specific manner in future clinical applications.


Asunto(s)
Nanopartículas , Neoplasias , Animales , Medios de Contraste , Humanos , Ligandos , Imagen por Resonancia Magnética , Ratones , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico
15.
Int J Biol Macromol ; 175: 171-178, 2021 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-33549659

RESUMEN

Covalent protein-ligation methods were used not only to visualize the localization of proteins of interest in cells, but also to study the topology of plasma and subcellular organelle membrane proteins using fluorescent cell imaging. A 13-amino-acid SpyTag (ST) peptide was genetically introduced either into a variety of subcellular proteins of interest or into different positions of plasma or subcellular organelle membrane proteins individually. Conversely, a 15 kDa SpyCatcher (SC) protein was chemically conjugated with either fluorescent dyes or horseradish peroxidase (HRP) via a thiol-maleimide reaction. The extracellular ST-fused plasma membrane proteins were efficiently labeled with the fluorescent-dye-conjugated SC in both live and permeabilized cells, whereas the intracellularly localized ST-fused subcellular proteins were only labeled in permeabilized cells because of the limited accessibility of the fluorescent-dye-conjugated SC to the membrane. The fluorescent-dye-labeled SC together with selective membrane-permeabilizing agents successfully labeled the plasma or the subcellular organelle membrane proteins in a topology-dependent manner. Moreover, the HRP-conjugated SC not only successfully labeled the ST-fused plasma membrane proteins, thus significantly enhancing fluorescent signals in combination with the tyramide signal amplification agents, but also ligated with an external ST-fused target ligand, thus selectively binding to the endogenously expressed cellular receptors of the target cancer cells.


Asunto(s)
Proteínas de la Membrana/química , Péptidos/química , Membrana Celular/química , Membrana Celular/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana/metabolismo , Péptidos/metabolismo , Ingeniería de Proteínas/métodos
16.
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
17.
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
18.
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
19.
Nat Commun ; 9(1): 4548, 2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30382085

RESUMEN

Targeted drug delivery using nanoparticles can minimize the side effects of conventional pharmaceutical agents and enhance their efficacy. However, translating nanoparticle-based agents into clinical applications still remains a challenge due to the difficulty in regulating interactions on the interfaces between nanoparticles and biological systems. Here, we present a targeting strategy for nanoparticles incorporated with a supramolecularly pre-coated recombinant fusion protein in which HER2-binding affibody combines with glutathione-S-transferase. Once thermodynamically stabilized in preferred orientations on the nanoparticles, the adsorbed fusion proteins as a corona minimize interactions with serum proteins to prevent the clearance of nanoparticles by macrophages, while ensuring systematic targeting functions in vitro and in vivo. This study provides insight into the use of the supramolecularly built protein corona shield as a targeting agent through regulating the interfaces between nanoparticles and biological systems.


Asunto(s)
Sistemas de Liberación de Medicamentos , Nanopartículas/química , Corona de Proteínas/química , Animales , Antineoplásicos/farmacología , Proteínas Sanguíneas/química , Línea Celular Tumoral , Femenino , Células HEK293 , Humanos , Ratones , Ratones Desnudos , Unión Proteica , Proteómica , Células RAW 264.7
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...