Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Struct Mol Biol ; 30(12): 1958-1969, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38049566

RESUMO

Transcription factors are among the most attractive therapeutic targets but are considered largely 'undruggable' in part due to the intrinsically disordered nature of their activation domains. Here we show that the aromatic character of the activation domain of the androgen receptor, a therapeutic target for castration-resistant prostate cancer, is key for its activity as transcription factor, allowing it to translocate to the nucleus and partition into transcriptional condensates upon activation by androgens. On the basis of our understanding of the interactions stabilizing such condensates and of the structure that the domain adopts upon condensation, we optimized the structure of a small-molecule inhibitor previously identified by phenotypic screening. The optimized compounds had more affinity for their target, inhibited androgen-receptor-dependent transcriptional programs, and had an antitumorigenic effect in models of castration-resistant prostate cancer in cells and in vivo. These results suggest that it is possible to rationally optimize, and potentially even to design, small molecules that target the activation domains of oncogenic transcription factors.


Assuntos
Neoplasias de Próstata Resistentes à Castração , Neoplasias da Próstata , Masculino , Humanos , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/genética , Receptores Androgênicos/genética , Receptores Androgênicos/química , Androgênios/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Domínios Proteicos , Fatores de Transcrição , Linhagem Celular Tumoral
2.
Methods Mol Biol ; 2406: 359-370, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35089568

RESUMO

Recombinant protein expression in E. coli often induces the expressed protein to accumulate in insoluble aggregates, named inclusion bodies (IBs), that represent easy to isolate, highly pure protein reservoirs. IBs can be solubilized by denaturing agents but this procedure requires, for complex globular proteins, a refolding step that can be challenging. However, the lack of cooperatively folded tertiary structure in intrinsically disordered proteins (IDP) makes them ideal candidates for this purification strategy. Given the wide abundance of IDPs, their relevance in many disease areas and the numerous IDP-associated biological functions, the interest in this class of proteins has increased substantially over the last decade. Here we present a broad and versatile method for the production and isolation of IDPs from inclusion bodies under denaturant conditions that overcomes the challenges associated with the propensity of these sequences to precipitate from solution and becoming proteolytically degraded.


Assuntos
Proteínas Intrinsicamente Desordenadas , Escherichia coli/genética , Escherichia coli/metabolismo , Corpos de Inclusão/metabolismo , Proteínas Intrinsicamente Desordenadas/química , Proteínas Recombinantes/química
3.
Protein Sci ; 30(7): 1427-1437, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33978290

RESUMO

The phase equilibria of intrinsically disordered proteins are exquisitely sensitive to changes in solution conditions and this can be used to investigate the driving forces of phase separation in vitro as well as the biological roles of phase transitions in live cells. Here we investigate how using D2 O as co-solvent in an aqueous buffer changes the phase equilibrium of a fragment of the activation domain of the androgen receptor, a transcription factor that plays a role in the development of the male phenotype and is a therapeutic target for castration resistant prostate cancer. We show how replacing even small fractions of H2 O with D2 O increases the propensity of this fragment to undergo liquid-liquid phase separation, likely reflecting a stabilization of the hydrophobic interactions that drive condensation. Our results indicate that it is necessary to take this effect into consideration when studying phase separation phenomena with biophysical methods that require using D2 O as a co-solvent. In addition, they suggest that additions of D2 O may be used to enhance phase separation phenomena in cells, facilitating their observation.


Assuntos
Óxido de Deutério/química , Proteínas Intrinsicamente Desordenadas/química , Receptores Androgênicos/química , Humanos , Domínios Proteicos
4.
Fish Shellfish Immunol ; 99: 578-586, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32105827

RESUMO

Nervous necrosis virus (NNV) reassortant strains RGNNV/SJNNV have emerged as a potent threat to the Mediterranean marine aquaculture industry, causing viral encephalopathy and retinopathy (VER) in Senegalese sole (Solea senegalensis). In this study, a cheap and practical vaccine strategy using bacterial inclusion bodies made of the coat protein of a virulent reassortant strain of this betanodavirus was devised. The nanostructured recombinant protein nanoparticles, VNNV-CNP, were administered without adjuvant to two groups of juvenile sole, one by intraperitoneal injection and the other by oral intubation. Specific antibodies were raised in vivo against the NNV coat protein via both routes, with a substantial specific antibody expansion in the injected group 30 days post homologous prime boost. Expression levels of five adaptive immune-related genes, cd8a, cd4, igm, igt and arg2, were also quantified in intestine, spleen and head kidney. Results showed cd4 and igm were upregulated in the head kidney of injected fish, indicating activation of an adaptive systemic response, while intubated fish exhibited a mucosal response in the intestine. Neither route showed significant differential expression of cd8a. The specific antibody response elicited in vivo and the lack of any signs of toxicity over the 6-week study period in young fish (n = 100), evidences the potential of the nanoparticle as a vaccine candidate.


Assuntos
Proteínas do Capsídeo/imunologia , Linguados/imunologia , Nanoestruturas/administração & dosagem , Infecções por Vírus de RNA/veterinária , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Aquicultura , Proteínas do Capsídeo/administração & dosagem , Feminino , Doenças dos Peixes/prevenção & controle , Rim Cefálico/imunologia , Imunidade nas Mucosas , Masculino , Nodaviridae , Infecções por Vírus de RNA/prevenção & controle , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Vacinas Virais/administração & dosagem
5.
Cell Rep ; 30(4): 1117-1128.e5, 2020 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-31995753

RESUMO

Prion-like proteins form multivalent assemblies and phase separate into membraneless organelles. Heterogeneous ribonucleoprotein D-like (hnRNPDL) is a RNA-processing prion-like protein with three alternative splicing (AS) isoforms, which lack none, one, or both of its two disordered domains. It has been suggested that AS might regulate the assembly properties of RNA-processing proteins by controlling the incorporation of multivalent disordered regions in the isoforms. This, in turn, would modulate their activity in the downstream splicing program. Here, we demonstrate that AS controls the phase separation of hnRNPDL, as well as the size and dynamics of its nuclear complexes, its nucleus-cytoplasm shuttling, and amyloidogenicity. Mutation of the highly conserved D378 in the disordered C-terminal prion-like domain of hnRNPDL causes limb-girdle muscular dystrophy 1G. We show that D378H/N disease mutations impact hnRNPDL assembly properties, accelerating aggregation and dramatically reducing the protein solubility in the muscle of Drosophila, suggesting a genetic loss-of-function mechanism for this muscular disorder.


Assuntos
Proteínas Amiloidogênicas/metabolismo , Núcleo Celular/metabolismo , Drosophila/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo D/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo D/metabolismo , Distrofia Muscular do Cíngulo dos Membros/genética , Agregação Patológica de Proteínas/metabolismo , Processamento Alternativo , Proteínas Amiloidogênicas/genética , Proteínas Amiloidogênicas/ultraestrutura , Animais , Núcleo Celular/efeitos dos fármacos , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Dactinomicina/farmacologia , Drosophila/metabolismo , Técnicas de Inativação de Genes , Células HeLa , Ribonucleoproteínas Nucleares Heterogêneas Grupo D/ultraestrutura , Humanos , Cinética , Microscopia Eletrônica de Transmissão , Células Musculares/metabolismo , Células Musculares/patologia , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Mutação , Agregação Patológica de Proteínas/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/ultraestrutura
6.
Acta Biomater ; 103: 272-280, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31812843

RESUMO

Fluorescent proteins are useful imaging and theranostic agents, but their potential superiority over alternative dyes is weakened by substantial photobleaching under irradiation. Enhancing protein photostability has been attempted through diverse strategies, with irregular results and limited applicability. In this context, we wondered if the controlled oligomerization of Green Fluorescent Protein (GFP) as nanoscale supramolecular complexes could stabilize the fluorophore through the newly formed protein-protein contacts, and thus, enhance its global photostability. For that, we have here analyzed the photobleaching profile of several GFP versions, engineered to self-assemble as tumour-homing nanoparticles with different targeting, size and structural stability. This has been done under prolonged irradiation in confocal laser scanning microscopy and by small-angle X-ray scattering. The results show that the oligomerization of GFP at the nanoscale enhances, by more than seven-fold, the stability of fluorescence emission. Interestingly, GFP nanoparticles are much more resistant to X-ray damage than the building block counterparts, indicating that the gained photostability is linked to enhanced structural resistance to radiation. Therefore, the controlled oligomerization of self-assembling fluorescent proteins as protein nanoparticles is a simple, versatile and powerful method to enhance their photostability for uses in precision imaging and therapy. STATEMENT OF SIGNIFICANCE: Fluorescent protein assembly into regular and highly symmetric nanoscale structures has been identified to confer enhanced structural stability against radiation stresses dramatically reducing their photobleaching. Being this the main bottleneck in the use of fluorescent proteins for imaging and theranostics, this protein architecture engineering principle appears as a powerful method to enhance their photostability for a broad applicability in precision imaging, drug delivery and theranostics.


Assuntos
Proteínas de Fluorescência Verde/metabolismo , Nanopartículas/química , Neoplasias/patologia , Fotodegradação , Fluorescência , Proteínas de Fluorescência Verde/química , Modelos Moleculares , Nanopartículas/ultraestrutura
7.
ACS Appl Mater Interfaces ; 12(5): 5381-5388, 2020 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-31840972

RESUMO

Five peptide ligands of four different cell surface receptors (nucleolin, CXCR1, CMKLR1, and CD44v6) have been evaluated as targeting moieties for triple-negative human breast cancers. Among them, the peptide F3, derived from phage display, promotes the fast and efficient internalization of a genetically fused green fluorescent protein (GFP) inside MDA-MB-231 cancer stem cells in a specific receptor-dependent fashion. The further engineering of this protein into the modular construct F3-RK-GFP-H6 and the subsequent construct F3-RK-PE24-H6 resulted in self-assembling polypeptides that organize as discrete and regular nanoparticles. These materials, 15-20 nm in size, show enhanced nucleolin-dependent cell penetrability. We show that the F3-RK-PE24-H6, based on the Pseudomonas aeruginosa exotoxin A (PE24) as a core functional domain, is highly cytotoxic over target cells. The combination of F3, the cationic peptide (RK)n, and the toxin domain PE24 in such unusual presentation appears as a promising approach to cell-targeted drug carriers in breast cancers and addresses selective drug delivery in otherwise difficult-to-treat triple-negative breast cancers.


Assuntos
Portadores de Fármacos/química , Nanoestruturas/química , Peptídeos/química , ADP Ribose Transferases/química , ADP Ribose Transferases/farmacologia , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Toxinas Bacterianas/química , Toxinas Bacterianas/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Exotoxinas/química , Exotoxinas/farmacologia , Feminino , Humanos , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Peptídeos/metabolismo , Peptídeos/farmacologia , Pseudomonas aeruginosa/metabolismo , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Fatores de Virulência/química , Fatores de Virulência/farmacologia
8.
Adv Sci (Weinh) ; 6(18): 1900849, 2019 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-31559131

RESUMO

Two structurally and functionally unrelated proteins, namely Omomyc and p31, are engineered as CD44-targeted inclusion bodies produced in recombinant bacteria. In this unusual particulate form, both types of protein materials selectively penetrate and kill CD44+ tumor cells in culture, and upon local administration, promote destruction of tumoral tissue in orthotropic mouse models of human breast cancer. These findings support the concept of bacterial inclusion bodies as versatile protein materials suitable for application in chronic diseases that, like cancer, can benefit from a local slow release of therapeutic proteins.

9.
Acta Biomater ; 83: 257-264, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30366134

RESUMO

Nanostructured protein materials show exciting biomedical applications, since both structure and function can be genetically programmed. In particular, self-assembling histidine-rich proteins benefit from functional plasticity that allows the generation of protein-only nanoparticles for cell targeted drug delivery. However, the rational development of constructs with improved functions is limited by a poor control of the oligomerization process. By exploring cross-interactions between histidine-tagged building blocks, we have identified a critical architectonic role of divalent cations. The obtained data instruct about how histidine-rich protein materials can be assembled, disassembled and reassembled within the nanoscale through the stoichiometric manipulation of divalent ions, in a biochemical approach to biomaterials design. STATEMENT OF SIGNIFICANCE: Divalent metal and non-metal cations such as Ni2+, Cu2+ Ca2+ and Zn2+ have been identified as unexpected molecular tools to control the assembling, disassembling and reassembling of histidine-rich protein materials at the nanoscale. Their stoichiometric manipulation allows generating defined protein-protein cross-molecular contacts between building blocks, for a powerful nano-biochemical manipulation of the material's architecture.


Assuntos
Cátions Bivalentes/química , Portadores de Fármacos/química , Metais/química , Nanopartículas/química , Proteínas/química , Proteínas Recombinantes/química
10.
Biomacromolecules ; 19(9): 3788-3797, 2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30052033

RESUMO

Protein materials are rapidly gaining interest in materials sciences and nanomedicine because of their intrinsic biocompatibility and full biodegradability. The controlled construction of supramolecular entities relies on the controlled oligomerization of individual polypeptides, achievable through different strategies. Because of the potential toxicity of amyloids, those based on alternative molecular organizations are particularly appealing, but the structural bases on nonamylogenic oligomerization remain poorly studied. We have applied spectrofluorimetry and spectropolarimetry to identify the conformational conversion during the oligomerization of His-tagged cationic stretches into regular nanoparticles ranging around 11 nm, useful for tumor-targeted drug delivery. We demonstrate that the novel conformation acquired by the proteins, as building blocks of these supramolecular assemblies, shows different extents of compactness and results in a beta structure enrichment that enhances their structural stability. The conformational profiling presented here offers clear clues for understanding and tailoring the process of nanoparticle formation through the use of cationic and histidine rich stretches in the context of protein materials usable in advanced nanomedical strategies.


Assuntos
Peptídeos Catiônicos Antimicrobianos/química , Nanopartículas/química , Multimerização Proteica , Peptídeos Catiônicos Antimicrobianos/genética , Antineoplásicos/administração & dosagem , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/genética , Conformação Proteica em Folha beta , Estabilidade Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
11.
Small ; 14(26): e1800665, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29845742

RESUMO

Under the unmet need of efficient tumor-targeting drugs for oncology, a recombinant version of the plant toxin ricin (the modular protein T22-mRTA-H6) is engineered to self-assemble as protein-only, CXCR4-targeted nanoparticles. The soluble version of the construct self-organizes as regular 11 nm planar entities that are highly cytotoxic in cultured CXCR4+ cancer cells upon short time exposure, with a determined IC50 in the nanomolar order of magnitude. The chemical inhibition of CXCR4 binding sites in exposed cells results in a dramatic reduction of the cytotoxic potency, proving the receptor-dependent mechanism of cytotoxicity. The insoluble version of T22-mRTA-H6 is, contrarily, moderately active, indicating that free, nanostructured protein is the optimal drug form. In animal models of acute myeloid leukemia, T22-mRTA-H6 nanoparticles show an impressive and highly selective therapeutic effect, dramatically reducing the leukemia cells affectation of clinically relevant organs. Functionalized T22-mRTA-H6 nanoparticles are then promising prototypes of chemically homogeneous, highly potent antitumor nanostructured toxins for precise oncotherapies based on self-mediated intracellular drug delivery.


Assuntos
Antineoplásicos/farmacologia , Nanoestruturas/química , Neoplasias/patologia , Receptores CXCR4/metabolismo , Proteínas Recombinantes/farmacologia , Ricina/farmacologia , Sequência de Aminoácidos , Animais , Permeabilidade da Membrana Celular/efeitos dos fármacos , Modelos Animais de Doenças , Células HeLa , Humanos , Leucemia Mieloide Aguda/patologia , Camundongos , Proteínas Recombinantes/química , Ricina/química
12.
J Control Release ; 279: 29-39, 2018 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-29641987

RESUMO

Sustained release of drug delivery systems (DDS) has the capacity to increase cancer treatment efficiency in terms of drug dosage reduction and subsequent decrease of deleterious side effects. In this regard, many biomaterials are being investigated but none offers morphometric and functional plasticity and versatility comparable to protein-based nanoparticles (pNPs). Here we describe a new DDS by which pNPs are fabricated as bacterial inclusion bodies (IB), that can be easily isolated, subcutaneously injected and used as reservoirs for the sustained release of targeted pNPs. Our approach combines the high performance of pNP, regarding specific cell targeting and biodistribution with the IB supramolecular organization, stability and cost effectiveness. This renders a platform able to provide a sustained source of CXCR4-targeted pNPs that selectively accumulate in tumor cells in a CXCR4+ colorectal cancer xenograft model. In addition, the proposed system could be potentially adapted to any other protein construct offering a plethora of possible new therapeutic applications in nanomedicine.


Assuntos
Neoplasias Colorretais/metabolismo , Sistemas de Liberação de Medicamentos , Nanopartículas , Proteínas/administração & dosagem , Amiloide/metabolismo , Animais , Bactérias/metabolismo , Preparações de Ação Retardada , Liberação Controlada de Fármacos , Feminino , Humanos , Corpos de Inclusão/metabolismo , Camundongos , Camundongos Nus , Proteínas/química , Receptores CXCR4/metabolismo , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Nanoscale ; 9(19): 6427-6435, 2017 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-28463351

RESUMO

Self-assembling proteins are gaining attention as building blocks for application-tailored nanoscale materials. This is mostly due to the biocompatibility, biodegradability, and functional versatility of peptide chains. Such a potential for adaptability is particularly high in the case of recombinant proteins, which are produced in living cells and are suitable for genetic engineering. However, how the cell factory itself and the particular protein folding machinery influence the architecture and function of the final material is still poorly explored. In this study we have used diverse analytical approaches, including small-angle X-ray scattering (SAXS) and field emission scanning electron microscopy (FESEM) to determine the fine architecture and geometry of recombinant, tumor-targeted protein nanoparticles of interest as drug carriers, constructed on a GFP-based modular scheme. A set of related oligomers were produced in alternative Escherichia coli strains with variant protein folding networks. This resulted in highly regular populations of morphometric types, ranging from 2.4 to 28 nm and from spherical- to rod-shaped materials. These differential geometric species, whose relative proportions were determined by the features of the producing strain, were found associated with particular fluorescence emission, cell penetrability and receptor specificity profiles. Then, nanoparticles with optimal properties could be analytically identified and further isolated from producing cells for use. The cell's protein folding machinery greatly modulates the final geometry reached by the constructs, which in turn defines the key parameters and biological performance of the material.


Assuntos
Portadores de Fármacos , Nanopartículas , Proteínas Recombinantes/química , Linhagem Celular Tumoral , Fluorescência , Proteínas de Fluorescência Verde/química , Células HeLa , Humanos , Microscopia Eletrônica de Varredura , Neoplasias/tratamento farmacológico , Espalhamento a Baixo Ângulo , Difração de Raios X
14.
Biofabrication ; 8(2): 025001, 2016 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-27078873

RESUMO

Biofabrication is attracting interest as a means to produce nanostructured functional materials because of its operational versatility and full scalability. Materials based on proteins are especially appealing, as the structure and functionality of proteins can be adapted by genetic engineering. Furthermore, strategies and tools for protein production have been developed and refined steadily for more than 30 years. However, protein conformation and therefore activity might be sensitive to production conditions. Here, we have explored whether the downstream strategy influences the structure and biological activities, in vitro and in vivo, of a self-assembling, CD44-targeted protein-only nanoparticle produced in Escherichia coli. This has been performed through the comparative analysis of particles built from soluble protein species or protein versions obtained by in vitro protein extraction from inclusion bodies, through mild, non-denaturing procedures. These methods have been developed recently as a convenient alternative to the use of toxic chaotropic agents for protein resolubilization from protein aggregates. The results indicate that the resulting material shows substantial differences in its physicochemical properties and its biological performance at the systems level, and that its building blocks are sensitive to the particular protein source.


Assuntos
Escherichia coli/metabolismo , Receptores de Hialuronatos/química , Receptores de Hialuronatos/metabolismo , Nanopartículas/química , Escherichia coli/genética , Engenharia Genética , Receptores de Hialuronatos/genética , Corpos de Inclusão/química , Corpos de Inclusão/genética , Corpos de Inclusão/metabolismo , Microbiologia Industrial , Nanopartículas/metabolismo , Conformação Proteica , Solubilidade
15.
Microb Cell Fact ; 15: 59, 2016 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-27059706

RESUMO

BACKGROUND: Production of recombinant drugs in process-friendly endotoxin-free bacterial factories targets to a lessened complexity of the purification process combined with minimized biological hazards during product application. The development of nanostructured recombinant materials in innovative nanomedical activities expands such a need beyond plain functional polypeptides to complex protein assemblies. While Escherichia coli has been recently modified for the production of endotoxin-free proteins, no data has been so far recorded regarding how the system performs in the fabrication of smart nanostructured materials. RESULTS: We have here explored the nanoarchitecture and in vitro and in vivo functionalities of CXCR4-targeted, self-assembling protein nanoparticles intended for intracellular delivery of drugs and imaging agents in colorectal cancer. Interestingly, endotoxin-free materials exhibit a distinguishable architecture and altered size and target cell penetrability than counterparts produced in conventional E. coli strains. These variant nanoparticles show an eventual proper biodistribution and highly specific and exclusive accumulation in tumor upon administration in colorectal cancer mice models, indicating a convenient display and function of the tumor homing peptides and high particle stability under physiological conditions. DISCUSSION: The observations made here support the emerging endotoxin-free E. coli system as a robust protein material producer but are also indicative of a particular conformational status and organization of either building blocks or oligomers. This appears to be promoted by multifactorial stress-inducing conditions upon engineering of the E. coli cell envelope, which impacts on the protein quality control of the cell factory.


Assuntos
Escherichia coli/metabolismo , Nanopartículas , Multimerização Proteica , Proteínas/química , Proteínas/metabolismo , Animais , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Células HeLa , Humanos , Camundongos , Camundongos Nus , Nanopartículas/química , Nanopartículas/metabolismo , Tamanho da Partícula , Polimerização , Engenharia de Proteínas/métodos , Proteínas/farmacologia , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Relação Estrutura-Atividade
16.
Nanomedicine ; 12(5): 1241-51, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26949165

RESUMO

A single chain polypeptide containing the low density lipoprotein receptor (LDLR) ligand Seq-1 with blood-brain barrier (BBB) crossing activity has been successfully modified by conventional genetic engineering to self-assemble into stable protein-only nanoparticles of 30nm. The nanoparticulate presentation dramatically enhances in vitro, LDLR-dependent cell penetrability compared to the parental monomeric version, but the assembled protein does not show any enhanced brain targeting upon systemic administration. While the presentation of protein drugs in form of nanoparticles is in general advantageous regarding correct biodistribution, this principle might not apply to brain targeting that is hampered by particular bio-physical barriers. Irrespective of this fact, which is highly relevant to the nanomedicine of central nervous system, engineering the cationic character of defined protein stretches is revealed here as a promising and generic approach to promote the controlled oligomerization of biologically active protein species as still functional, regular nanoparticles.


Assuntos
Barreira Hematoencefálica , Sistemas de Liberação de Medicamentos , Nanopartículas , Peptídeos , Humanos , Receptores de LDL , Distribuição Tecidual
17.
Adv Mater ; 27(47): 7816-22, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26509451

RESUMO

The impact of cell factory quality control on material properties is a neglected but critical issue in the fabrication of protein biomaterials, which are unique in merging structure and function. The molecular chaperoning of protein conformational status is revealed here as a potent molecular instructor of the macroscopic properties of self-assembling, cell-targeted protein nanoparticles, including biodistribution upon in vivo administration.


Assuntos
Nanotecnologia/métodos , Engenharia de Proteínas/métodos , Proteínas/química , Proteínas/genética , Escherichia coli K12/genética , Modelos Moleculares , Nanoestruturas/química , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Estrutura Secundária de Proteína , Proteínas/metabolismo , Controle de Qualidade
18.
Methods Mol Biol ; 1258: 321-30, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25447873

RESUMO

All researchers immersed in the world of recombinant protein production are in agreement that often the production and purification process of a protein can become a nightmare due to an unexpected behavior of the protein at different protocol stages. Once the protein is purified, scientists know that they still cannot relax. There is a decisive last step missing: performing a protein dialysis in a suitable buffer for subsequent experimental trials. Here is when we can find proteins that precipitate during dialysis by buffer-related factors (ionic strength, pH, etc.), which are intrinsic to each protein and are difficult to predict. How can we find the buffer in which a protein is more stable and with less tendency to precipitate? In this chapter we go over possible factors affecting the protein precipitation tendency during the dialysis process and describe a general dialysis protocol with tricks to reduce protein aggregation. Furthermore, we propose a fast method to detect the most appropriate buffer for the stability of a particular protein, performing microdialysis on a battery of different buffers to measure afterwards precipitation by a colorimetric method, and thus being able to choose the most suitable buffer for the dialysis of a given protein.


Assuntos
Agregados Proteicos/fisiologia , Proteínas Recombinantes/química , Animais , Soluções Tampão , Diálise/métodos , Humanos , Estabilidade Proteica
19.
Int J Pharm ; 473(1-2): 286-95, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25019161

RESUMO

CD44 is a multifunctional cell surface protein involved in proliferation and differentiation, angiogenesis and signaling. The expression of CD44 is up-regulated in several types of human tumors and particularly in cancer stem cells, representing an appealing target for drug delivery in the treatment of cancer. We have explored here several protein ligands of CD44 for the construction of self-assembling modular proteins designed to bind and internalize target cells. Among five tested ligands, two of them (A5G27 and FNI/II/V) drive the formation of protein-only, ring-shaped nanoparticles of about 14 nm that efficiently bind and penetrate CD44(+) cells by an endosomal route. The potential of these newly designed nanoparticles is evaluated regarding the need of biocompatible nanostructured materials for drug delivery in CD44-linked conditions.


Assuntos
Receptores de Hialuronatos/metabolismo , Nanopartículas/administração & dosagem , Peptídeos/química , Linhagem Celular Tumoral , Endocitose , Fibronectinas/química , Células HEK293 , Humanos , Laminina/química , Ligantes , Nanopartículas/química , Peptídeos/administração & dosagem
20.
ACS Nano ; 8(5): 4166-76, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24708510

RESUMO

The fully de novo design of protein building blocks for self-assembling as functional nanoparticles is a challenging task in emerging nanomedicines, which urgently demand novel, versatile, and biologically safe vehicles for imaging, drug delivery, and gene therapy. While the use of viruses and virus-like particles is limited by severe constraints, the generation of protein-only nanocarriers is progressively reachable by the engineering of protein-protein interactions, resulting in self-assembling functional building blocks. In particular, end-terminal cationic peptides drive the organization of structurally diverse protein species as regular nanosized oligomers, offering promise in the rational engineering of protein self-assembling. However, the in vivo stability of these constructs, being a critical issue for their medical applicability, needs to be assessed. We have explored here if the cross-molecular contacts between protein monomers, generated by end-terminal cationic peptides and oligohistidine tags, are stable enough for the resulting nanoparticles to overcome biological barriers in assembled form. The analyses of renal clearance and biodistribution of several tagged modular proteins reveal long-term architectonic stability, allowing systemic circulation and tissue targeting in form of nanoparticulate material. This observation fully supports the value of the engineered of protein building blocks addressed to the biofabrication of smart, robust, and multifunctional nanoparticles with medical applicability that mimic structure and functional capabilities of viral capsids.


Assuntos
Nanomedicina/métodos , Nanopartículas/química , Proteínas/química , Animais , Capsídeo/química , Cátions , Sistemas de Liberação de Medicamentos , Feminino , Engenharia Genética , Proteínas de Fluorescência Verde/química , Histidina/química , Humanos , Ligação de Hidrogênio , Rim/metabolismo , Ligantes , Camundongos , Camundongos Nus , Microscopia de Força Atômica , Microscopia Eletrônica de Transmissão , Peptídeos/química , Ligação Proteica , Mapeamento de Interação de Proteínas , Eletricidade Estática
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...