Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 16(6)2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38932260

RESUMO

Soon after its birth in 1985, following a short lag period [...].


Assuntos
Neoplasias , Humanos , Neoplasias/terapia , Neoplasias/genética , Técnicas de Visualização da Superfície Celular/métodos , Biblioteca de Peptídeos , Animais
2.
Viruses ; 16(2)2024 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-38400052

RESUMO

Prostate cancer (PC) is the second most diagnosed cancer among men. It was observed that early diagnosis of disease is highly beneficial for the survival of cancer patients. Therefore, the extension and increasing quality of life of PC patients can be achieved by broadening the cancer screening programs that are aimed at the identification of cancer manifestation in patients at earlier stages, before they demonstrate well-understood signs of the disease. Therefore, there is an urgent need for standard, sensitive, robust, and commonly available screening and diagnosis tools for the identification of early signs of cancer pathologies. In this respect, the "Holy Grail" of cancer researchers and bioengineers for decades has been molecular sensing probes that would allow for the diagnosis, prognosis, and monitoring of cancer diseases via their interaction with cell-secreted and cell-associated PC biomarkers, e.g., PSA and PSMA, respectively. At present, most PSA tests are performed at centralized laboratories using high-throughput total PSA immune analyzers, which are suitable for dedicated laboratories and are not readily available for broad health screenings. Therefore, the current trend in the detection of PC is the development of portable biosensors for mobile laboratories and individual use. Phage display, since its conception by George Smith in 1985, has emerged as a premier tool in molecular biology with widespread application. This review describes the role of the molecular evolution and phage display paradigm in revolutionizing the methods for the early diagnosis and monitoring of PC.


Assuntos
Bacteriófagos , Neoplasias da Próstata , Masculino , Humanos , Antígeno Prostático Específico , Qualidade de Vida , Neoplasias da Próstata/diagnóstico , Detecção Precoce de Câncer
3.
Viruses ; 14(2)2022 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-35215976

RESUMO

The evolution of the SARS-CoV-2 virus during the COVID-19 pandemic was accompanied by the emergence of new heavily mutated viral variants with increased infectivity and/or resistance to detection by the human immune system. To respond to the urgent need for advanced methods and materials to empower a better understanding of the mechanisms of virus's adaptation to human host cells and to the immuno-resistant human population, we suggested using recombinant filamentous bacteriophages, displaying on their surface foreign peptides termed "mimotopes", which mimic the structure of viral receptor-binding sites on the viral spike protein and can serve as molecular probes in the evaluation of molecular mechanisms of virus infectivity. In opposition to spike-binding antibodies that are commonly used in studying the interaction of the ACE2 receptor with SARS-CoV-2 variants in vitro, phage spike mimotopes targeted to other cellular receptors would allow discovery of their role in viral infection in vivo using cell culture, tissue, organs, or the whole organism. Phage mimotopes of the SARS-CoV-2 Spike S1 protein have been developed using a combination of phage display and molecular mimicry concepts, termed here "phage mimicry", supported by bioinformatics methods. The key elements of the phage mimicry concept include: (1) preparation of a collection of p8-type (landscape) phages, which interact with authentic active receptors of live human cells, presumably mimicking the binding interactions of human coronaviruses such as SARS-CoV-2 and its variants; (2) discovery of closely related amino acid clusters with similar 3D structural motifs on the surface of natural ligands (FGF1 and NRP1), of the model receptor of interest FGFR and the S1 spike protein; and (3) an ELISA analysis of the interaction between candidate phage mimotopes with FGFR3 (a potential alternative receptor) in comparison with ACE2 (the authentic receptor).


Assuntos
Bacteriófagos/genética , Técnicas de Visualização da Superfície Celular/métodos , Mimetismo Molecular , Receptores de Superfície Celular/genética , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/genética , Bacteriófagos/metabolismo , Sítios de Ligação , Humanos , Ligação Proteica , Receptores de Superfície Celular/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Ligação Viral
4.
ACS Appl Mater Interfaces ; 12(8): 9090-9097, 2020 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-32023032

RESUMO

It is important to detect pathogens rapidly, sensitively, and selectively for clinical medicine, homeland security, food safety, and environmental control. We report here a specific and sensitive colorimetric assay that incorporated a bovine serum albumin-templated Co3O4 magnetic nanozyme (Co3O4 MNE) with a novel specific fusion phage protein and magnetophoretic chromatography to detect Staphylococcus aureus. The Co3O4 MNE was conjugated to S. aureus-specific fusion-pVIII (Co3O4 MNE@fusion-pVIII), screened from the S. aureus-specific phage AQTFLGEQD (the phage monoclone is denoted by the peptide sequence). The as-prepared triple-functional Co3O4 MNE@fusion-pVIII particles were capable of capturing S. aureus in sterile milk, which were then isolated from milk magnetically. Assisted by polyethylene glycol, the Co3O4 MNE@fusion-pVIII@S. aureus complex was separated from the free Co3O4 MNE@fusion-pVIII by magnetophoretic chromatography in an external magnetic field. After transferring the isolated Co3O4 MNE@fusion-pVIII@S. aureus complexes into a 96-well plate, diammonium salt of 2,2'-azino-bis(3-ethylbenzo-thiazoline-6-sulfonic acid) and H2O2 were added to develop color because of the peroxidase mimetics activity of the Co3O4 MNE. A S. aureus concentration within 10-10,000 cfu/mL in milk can be detected (detection limit: 8 cfu/mL). The as-developed method is simple, cost-efficient, and sensitive, which is useful for rapidly diagnosing pathogenic bacteria and helpful to prevent disease outbreaks induced by pathogens in developing countries.


Assuntos
Cobalto/química , Microbiologia de Alimentos , Leite/microbiologia , Nanopartículas/química , Óxidos/química , Peroxidase/química , Fagos de Staphylococcus/química , Staphylococcus aureus/metabolismo , Proteínas Virais de Fusão/química , Animais , Colorimetria , Campos Magnéticos , Staphylococcus aureus/virologia
5.
Viruses ; 11(11)2019 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-31717800

RESUMO

Peptide-displayed phage libraries are billion-clone collections of diverse chimeric bacteriophage particles, decorated by genetically fused peptides built from a random combination of natural amino acids. Studying the molecular evolution of peptide-displayed libraries in mammalian model systems, using in vivo phage display techniques, can provide invaluable knowledge about the underlying physiology of the vasculature system, allow recognition of organ- and tissue-specific networks of protein-protein interactions, and provide ligands for targeted diagnostics and therapeutics. Recently, we discovered that landscape phage libraries, a specific type of multivalent peptide phage display library, expose on their surface comprehensive collections of elementary binding units (EBUs), which can form short linear motifs (SLiMs) that interact with functional domains of physiologically relevant proteins. Because of their unique structural and functional features, landscape phages can use an alternative mechanism of directed molecular evolution, i.e., combinatorial avidity selection. These discoveries fueled our interest in revisiting the in vivo evolution of phage displayed libraries using another format of display, i.e., landscape phages. In this study, we monitored the evolution of a landscape phage library in a mouse model with and without an implanted human breast cancer tumor xenograft. As expected, the multivalent architecture of landscape phage displayed proteins provided strong tissue selectivity and resulted in a huge diversity of tissue penetrating, chimeric phage particles. We identified several types of EBU interactions that evolved during the course of tissue distribution, which included interactions of EBUs with all tissue types, those EBUs that interacted selectively with specific organs or tissues with shared gene expression profiles or functionalities, and other EBUs that interacted in a tissue-selective manner. We demonstrated that landscape phage libraries are a rich collection of unique nanobioparticles that can be used to identify functional organ and tissue-binding elements after the evolution of a phage display library in vivo.


Assuntos
Bacteriófagos/genética , Neoplasias da Mama/genética , Técnicas de Visualização da Superfície Celular/métodos , Biblioteca Gênica , Animais , Evolução Molecular , Feminino , Xenoenxertos , Humanos , Camundongos , Camundongos Nus
6.
Viruses ; 11(9)2019 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-31454976

RESUMO

Low performance of actively targeted nanomedicines required revision of the traditional drug targeting paradigm and stimulated the development of novel phage-programmed, self-navigating drug delivery vehicles. In the proposed smart vehicles, targeting peptides, selected from phage libraries using traditional principles of affinity selection, are substituted for phage proteins discovered through combinatorial avidity selection. Here, we substantiate the potential of combinatorial avidity selection using landscape phage in the discovery of Short Linear Motifs (SLiMs) and their partner domains. We proved an algorithm for analysis of phage populations evolved through multistage screening of landscape phage libraries against the MDA-MB-231 breast cancer cell line. The suggested combinatorial avidity selection model proposes a multistage accumulation of Elementary Binding Units (EBU), or Core Motifs (CorMs), in landscape phage fusion peptides, serving as evolutionary initiators for formation of SLiMs. Combinatorial selection has the potential to harness directed molecular evolution to create novel smart materials with diverse novel, emergent properties.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Técnicas de Química Combinatória/métodos , Evolução Molecular Direcionada , Sistemas de Liberação de Medicamentos/métodos , Antineoplásicos/análise , Bacteriófagos/metabolismo , Linhagem Celular Tumoral , Humanos , Nanomedicina , Biblioteca de Peptídeos , Peptídeos/química
7.
Viruses ; 10(6)2018 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-29880747

RESUMO

The development of phage engineering technology has led to the construction of a novel type of phage display library-a collection of nanofiber materials with diverse molecular landscapes accommodated on the surface of phage particles. These new nanomaterials, called the "landscape phage", serve as a huge resource of diagnostic/detection probes and versatile construction materials for the preparation of phage-functionalized biosensors and phage-targeted nanomedicines. Landscape-phage-derived probes interact with biological threat agents and generate detectable signals as a part of robust and inexpensive molecular recognition interfaces introduced in mobile detection devices. The use of landscape-phage-based interfaces may greatly improve the sensitivity, selectivity, robustness, and longevity of these devices. In another area of bioengineering, landscape-phage technology has facilitated the development and testing of targeted nanomedicines. The development of high-throughput phage selection methods resulted in the discovery of a variety of cancer cell-associated phages and phage proteins demonstrating natural proficiency to self-assemble into various drug- and gene-targeting nanovehicles. The application of this new "phage-programmed-nanomedicines" concept led to the development of a number of cancer cell-targeting nanomedicine platforms, which demonstrated anticancer efficacy in both in vitro and in vivo experiments. This review was prepared to attract the attention of chemical scientists and bioengineers seeking to develop functionalized nanomaterials and use them in different areas of bioscience, medicine, and engineering.


Assuntos
Biotecnologia/métodos , Técnicas de Visualização da Superfície Celular/métodos , Nanomedicina/métodos , Biotecnologia/tendências , Humanos , Nanomedicina/tendências
8.
Biosens Bioelectron ; 106: 1-6, 2018 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-29414074

RESUMO

The level of total prostate-specific antigen (t-PSA) is generally known as the key index of prostate cancer. Here, phage probes against t-PSA were selected from f8/8 landscape phage library. After three rounds of biopanning, four t-PSA-binding phage clones were isolated and identified by the DNA sequencing. Based on the phage capture assay, the phage clone displaying the fusion peptide ATRSANGM showed highest affinity and specificity against t-PSA. Subsequently, the t-PSA-specific phage was used as t-PSA capture probe in a sandwich enzyme-linked immunosorbent assay (ELISA) and differential pulse voltammetry (DPV) assay systems. Both assay methods showed high specificity and acceptable reliability for real serum samples analysis. By comparison, DPV method showed wider linear range (0.01-100 ng mL-1) and lower limit of detection (3 pg mL-1) than those (3.3-330 ng mL-1 and 1.6 ng mL-1) of ELISA. Moreover, DPV system showed smaller distinction to the authoritative method in real samples assay. Excitingly, the phage probe based DPV immunosensor showed high sensitivity for the detection of t-PSA and LOD achieved the pg mL-1 level, which was far lower than those values (usually above 0.1 ng mL-1) for reported immunosensors based on antibodies. Due to the biocompatibility, multivalency, stability, and high structural homogeneity, the t-PSA-specific landscape phage demonstrates as a novel specific interface in biosensors.


Assuntos
Técnicas Biossensoriais , Ensaio de Imunoadsorção Enzimática , Antígeno Prostático Específico/isolamento & purificação , Neoplasias da Próstata/sangue , Bacteriófagos/genética , Ouro/química , Humanos , Masculino , Nanopartículas Metálicas/química , Antígeno Prostático Específico/sangue
9.
Nanoscale ; 10(6): 2825-2833, 2018 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-29362753

RESUMO

Pathogen detection continues to receive significant attention due to the harmful effects of pathogens on public health. Herein, specific nonapeptide-fusion proteins pVIII (pVIII fusion) were isolated from phage VQTVQIGSD (designated by the sequence of a fused foreign peptide), which was specifically screened from the f8/9 landscape phage library against Vibrio parahaemolyticus (V. parahaemolyticus) in a high-throughput way. The as-prepared V. parahaemolyticus-specific recognition element is cheaper and more available than antibodies. Further, a highly sensitive colorimetric immunoassay for V. parahaemolyticus was established using pVIII fusion as capture probes coupled with protein-templated MnO2 nanosheets (NSs) as signal probes. In the presence of a target bacterium, V. parahaemolyticus, a sandwich-type complex of pVIII fusion-V. parahaemolyticus-MnO2 NS@pVIII fusion was formed through specific recognition of pVIII fusion and V. parahaemolyticus. The signal probes (MnO2 NSs) could catalyze the reaction of 3,3',5,5'-tetramethylbenzidine and H2O2 to generate a colorimetric change. The proposed V. parahaemolyticus detection method demonstrated a wide detection range (20-104 colony-forming units (CFU) mL-1), low limit of detection (15 CFU mL-1), excellent selectivity, and high reliability for real marine samples, showing potential application in marine microbiological detection and control.


Assuntos
Colorimetria , Imunoensaio , Compostos de Manganês , Nanoestruturas , Óxidos , Vibrio parahaemolyticus/isolamento & purificação , Peróxido de Hidrogênio , Biblioteca de Peptídeos , Peroxidases , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
10.
J Drug Target ; 26(5-6): 505-515, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29132246

RESUMO

Pancreatic cancer has been a life-threatening illness associated with high incidence and mortality rates. Paclitaxel (PCT) that causes mitotic arrest in cancer cells disrupting microtubule function is used for pancreatic cancer treatment. Nausea, anorexia and abdominal pain are some of the typical dose-limiting toxicity associated gastrointestinal side effects of the drug. Here, we present the use of polymeric mixed micelles to enable a targeted delivery of PCT and to provide additional advantages such as enhanced drug solubility, bioavailability and minimal dose-limiting toxicity. Also, these micelles self-assemble with pancreatic cancer cells-specific phage proteins P38, L1 and with the hydrophobic drug PCT resolving the issue of complex chemistry efforts normally needed for any conjugation. Our cytotoxicity and binding experiment results in vitro in 2 D and 3 D models suggested that the phage protein-targeted drug-loaded micelles bind and exhibit higher cell killing over the non-targeted ones.


Assuntos
Bacteriófagos/química , Sistemas de Liberação de Medicamentos , Paclitaxel/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/farmacologia , Linhagem Celular Tumoral , Portadores de Fármacos/química , Humanos , Micelas , Paclitaxel/farmacologia , Neoplasias Pancreáticas/patologia , Polímeros/química
11.
Ther Deliv ; 8(12): 1063-1075, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29125066

RESUMO

Low efficacy of targeted nanomedicines in biological experiments enforced us to challenge the traditional concept of drug targeting and suggest a paradigm of 'addressed self-navigating drug-delivery vehicles,' in which affinity selection of targeting peptides and vasculature-directed in vivo phage screening is replaced by the migration selection, which explores ability of 'promiscuous' phages and their proteins to migrate through the tumor-surrounding cellular barriers, using a 'hub and spoke' delivery strategy, and penetrate into the tumor affecting the diverse tumor cell population. The 'self-navigating' drug-delivery paradigm can be used as a theoretical and technical platform in design of a novel generation of molecular medications and imaging probes for precise and personal medicine. [Formula: see text].


Assuntos
Portadores de Fármacos/química , Bacteriófagos/fisiologia , Técnicas de Visualização da Superfície Celular , Humanos , Nanomedicina , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Peptídeos/química , Peptídeos/metabolismo , Preparações Farmacêuticas/química , Preparações Farmacêuticas/metabolismo , Microambiente Tumoral
12.
J Mater Chem B ; 5(41): 8209-8218, 2017 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-32264464

RESUMO

Magnetic nanoparticles with superparamagnetic properties have provided a versatile platform for constructing multifunctional nanostructures, which show great promise in tumor-targeted multimodal imaging and non-invasive therapy. Herein, we first systematically investigated the effect of crystalline water in the reactants on the assembly of primary Fe3O4 nanocrystals prepared by a solvothermal method. The presence of water would hinder the formation of monodisperse Fe3O4 nanocrystals. The regular spheric Fe3O4 nanoclusters with high saturated magnetization values and superparamagnetism can be synthesized with anhydrous reactants and sodium citrate as a stabilizer. Furthermore, the monodisperse Fe3O4 nanoclusters were used as cores and coated with fluorescent dye molecule covalently-doped silica layers, on which carbohydrate-stabilized gold nanoparticles could be assembled. Fe3O4 core@dye/SiO2@Au shell nanocomposites were gradually formed by several cycles of a reduction process in the growth solution. The resultant ferroferric oxide@dye/silica@Au nanoshells exhibited good biocompatibility, an excellent T2-weighted relaxation rate, a strong fluorescence signal and tunable near IR surface plasmon resonance (SPR) spectra. Finally, colorectal cancer cell SW620-specific phage fusion proteins (fusion-pVIII) were conjugated onto the surface of gold nanoshells, which exhibited a maximal SPR peak of 774 nm and effectively achieved the photothermal ablation of tumor cells selectively with 808 nm laser irradiation for 10 min in a light intensity of 3 W cm-2. Additionally, the prepared bio-nanocomposite showed good T2-weighted magnetic resonance imaging (MRI). Therefore, the Fe3O4@dye/SiO2@Au@fusion-pVIII nanocomposites were successfully prepared and applied for targeted optical imaging and the targeted photothermal therapy of cancer cells. The prepared bio-nanocomposites can be potentially applied as ideal contrast agents for tumors in MRI.

13.
Expert Opin Drug Deliv ; 14(3): 373-384, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27466706

RESUMO

INTRODUCTION: New phage-directed nanomedicines have emerged recently as a result of the in-depth study of the genetics and structure of filamentous phage and evolution of phage display and phage nanobiotechnology. This review focuses on the progress made in the development of the cancer-targeted nanomaterials and discusses the trends in using phage as a bioselectable molecular navigation system. Areas covered: The merging of phage display technologies with nanotechnology in recent years has proved promising in different areas of medicine and technology, such as medical diagnostics, molecular imaging, vaccine development and targeted drug/gene delivery, which is the focus of this review. The authors used data obtained from their research group and sourced using Science Citation Index (Web of Science) and NCBI PubMed search resources. Expert opinion: First attempts of adapting traditional concepts of direct targeting of tumor using phage-targeted nanomedicines has shown minimal improvements. With discovery and study of biological and technical barriers that prevent anti-tumor drug delivery, a paradigm shift from traditional drug targeting to nanomedicine navigation systems is required. The advanced bacteriophage-driven self-navigation systems are thought to overcome those barriers using more precise, localized phage selection methods, multi-targeting 'promiscuous' ligands and advanced multifunctional nanomedicine platforms.


Assuntos
Antineoplásicos/administração & dosagem , Bacteriófagos , Neoplasias/tratamento farmacológico , Antineoplásicos/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Técnicas de Transferência de Genes , Humanos , Ligantes , Nanomedicina/métodos , Nanoestruturas
14.
Biosens Bioelectron ; 82: 195-203, 2016 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-27085951

RESUMO

Staphylococcus aureus (S. aureus) is one of the most ubiquitous pathogens in public healthcare worldwide. It holds great insterest in establishing robust analytical method for S. aureus. Herein, we report a S. aureus-specific recognition element, isolated from phage monoclone GQTTLTTS, which was selected from f8/8 landscape phage library against S. aureus in a high-throughput way. By functionalizing cysteamine (CS)-stabilized gold nanoparticles (CS-AuNPs) with S. aureus-specific pVIII fusion protein (fusion-pVIII), a bifunctional nanoprobe (CS-AuNPs@fusion-pVIII) for S. aureus was developed. In this strategy, the CS-AuNPs@fusion-pVIII could be induced to aggregate quickly in the presence of target S. aureus, resulting in a rapid colorimetric response of gold nanoparticles. More importantly, the as-designed probe exhibited excellent selectivity over other bacteria. Thus, the CS-AuNPs@fusion-pVIII could be used as the indicator of target S. aureus. This assay can detect as low as 19CFUmL(-1)S. aureus within 30min. Further, this approach can be applicable to detect S. aureus in real water samples. Due to its sensitivity, specificity and rapidness, this proposed method is promising for on-site testing of S. aureus without using any costly instruments.


Assuntos
Bacteriófagos/química , Técnicas Biossensoriais/métodos , Proteínas do Capsídeo/química , Colorimetria/métodos , Staphylococcus aureus/isolamento & purificação , Microbiologia da Água , Técnicas Biossensoriais/economia , Colorimetria/economia , Ouro/química , Humanos , Nanopartículas Metálicas/química , Biblioteca de Peptídeos , Proteínas Recombinantes de Fusão/química , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/virologia
15.
Comb Chem High Throughput Screen ; 19(5): 412-22, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27095536

RESUMO

Cancer cell-specific diagnostic or therapeutic tools are commonly believed to significantly increase the success rate of cancer diagnosis and targeted therapies. To extend the repertoire of available cancer cell-specific phage fusion proteins and study their efficacy as navigating moieties, we used two landscape phage display libraries f8/8 and f8/9 displaying an 8- or 9-mer random peptide fusion to identify a panel of novel peptide families that are specific to Calu-3 cells. Using a phage capture assay, we showed that two of the selected phage clones, ANGRPSMT and VNGRAEAP (phage and their recombinant proteins are named by the sequence of the fusion peptide), are selective for the Calu-3 cell line in comparison to phenotypically normal lung epithelial cells and distribute into unique subcellular fractions.


Assuntos
Neoplasias Pulmonares/tratamento farmacológico , Terapia de Alvo Molecular/métodos , Biblioteca de Peptídeos , Sequência de Aminoácidos , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Humanos , Neoplasias Pulmonares/patologia , Proteínas de Fusão de Membrana/metabolismo , Peptídeos , Frações Subcelulares/metabolismo
16.
Sci Rep ; 6: 22199, 2016 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-26908277

RESUMO

One of the major challenges in the design of biosensors for cancer diagnosis is to introduce a low-cost and selective probe that can recognize cancer cells. In this paper, we combined the phage display technology and electrochemical impedance spectroscopy (EIS) to develop a label-free cytosensor for the detection of cancer cells, without complicated purification of recognition elements. Fabrication steps of the cytosensing interface were monitored by EIS. Due to the high specificity of the displayed octapeptides and avidity effect of their multicopy display on the phage scaffold, good biocompatibility of recombinant phage, the fibrous nanostructure of phage, and the inherent merits of EIS technology, the proposed cytosensor demonstrated a wide linear range (2.0 × 10(2) - 2.0 × 10(8) cells mL(-1)), a low limit of detection (79 cells mL(-1), S/N = 3), high specificity, good inter-and intra-assay reproducibility and satisfactory storage stability. This novel cytosensor designing strategy will open a new prospect for rapid and label-free electrochemical platform for tumor diagnosis.


Assuntos
Bacteriófagos/genética , Técnicas Biossensoriais/métodos , Espectroscopia Dielétrica/métodos , Biblioteca de Peptídeos , Espectroscopia Dielétrica/instrumentação , Células HEK293 , Células Hep G2 , Humanos , Proteínas Virais/química , Proteínas Virais/genética
17.
Protein Eng Des Sel ; 29(3): 93-103, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26764410

RESUMO

Cancer cell-specific targeting ligands against numerous cancer cell lines have been selected previously and used as ligands for cell-specific delivery of chemotherapies and various nanomedicines. However, tumor heterogeneity is one recognized problem hampering clinical translation of targeted anti-cancer medicines. Therefore, a novel class of targeting ligands is required that recognize receptors expressed between a variety of cancer phenotypes, identified here as 'promiscuous' ligands. In this work, promiscuous phage fusion proteins were first identified by a novel selection scheme to enrich for pan-cancer cell binding abilities, as indicated by conserved structural motifs identified previously in other cancer types. Additionally, peptide sequences containing a combination of motifs were identified to modulate binding. A panel of phage fusion proteins was studied for their specificity and selectivity for lung and pancreatic cancer cells. Phage displaying the fusion peptides GSLEEVSTL or GEFDELMTM, the two predominate clones with greatest binding ability, were used to modify preformed, doxorubicin-loaded, liposomes. These modified liposomes increased cytotoxicity up to 8.1-fold in several cancer cell lines when compared with unmodified liposomal doxorubicin. Taken together, these data indicate that promiscuous phage proteins, selected against different cancer cell lines, can be used as targeting ligands for treatment of heterogeneous tumor populations.


Assuntos
Terapia de Alvo Molecular , Biblioteca de Peptídeos , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , Motivos de Aminoácidos , Sequência de Aminoácidos , Linhagem Celular Tumoral , Clonagem Molecular , Doxorrubicina/administração & dosagem , Doxorrubicina/uso terapêutico , Humanos , Espaço Intracelular/metabolismo , Ligantes , Lipossomos , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Especificidade por Substrato
18.
Front Microbiol ; 6: 628, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26157433

RESUMO

Active tumor targeting of nanomedicines has recently shown significant improvements in the therapeutic activity of currently existing drug delivery systems, such as liposomal doxorubicin (Doxil/Caelyx/Lipodox). Previously, we have shown that isolated pVIII major coat proteins of the fd-tet filamentous phage vector, containing cancer cell-specific peptide fusions at their N-terminus, can be used as active targeting ligands in a liposomal doxorubicin delivery system in vitro and in vivo. Here, we show a novel major coat protein isolation procedure in 2-propanol that allows spontaneous incorporation of the hydrophobic protein core into preformed liposomal doxorubicin with minimal damage or drug loss while still retaining the targeting ligand exposed for cell-specific targeting. Using a panel of 12 structurally unique ligands with specificity toward breast, lung, and/or pancreatic cancer, we showed the feasibility of pVIII major coat proteins to significantly increase the throughput of targeting ligand screening in a common nanomedicine core. Phage protein-modified Lipodox samples showed an average doxorubicin recovery of 82.8% across all samples with 100% of protein incorporation in the correct orientation (N-terminus exposed). Following cytotoxicity screening in a doxorubicin-sensitive breast cancer line (MCF-7), three major groups of ligands were identified. Ligands showing the most improved cytotoxicity included: DMPGTVLP, ANGRPSMT, VNGRAEAP, and ANDVYLD showing a 25-fold improvement (p < 0.05) in toxicity. Similarly DGQYLGSQ, ETYNQPYL, and GSSEQLYL ligands with specificity toward a doxorubicin-insensitive pancreatic cancer line (PANC-1) showed significant increases in toxicity (2-fold; p < 0.05). Thus, we demonstrated proof-of-concept that pVIII major coat proteins can be screened in significantly higher throughput to identify novel ligands displaying improved therapeutic activity in a desired cancer phenotype.

19.
Sci Rep ; 4: 6808, 2014 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-25348392

RESUMO

Nanomaterials with near-infrared (NIR) absorption have been widely studied in cancer detection and photothermal therapy (PTT), while it remains a great challenge in targeting tumor efficiently with minimal side effects. Herein we report a novel multifunctional phage-mimetic nanostructure, which was prepared by layer-by-layer self-assembly of Au@Ag heterogenous nanorods (NRs) with rhodamine 6G, and specific pVIII fusion proteins. Au@Ag NRs, first being applied for PTT, exhibited excellent stability, cost-effectivity, biocompatibility and tunable NIR absorption. The fusion proteins were isolated from phage DDAGNRQP specifically selected from f8/8 landscape phage library against colorectal cancer cells in a high-throughput way. Considering the definite charge distribution and low molecular weight, phage fusion proteins were assembled on the negatively charged NR core by electrostatic interactions, exposing the N-terminus fused with DDAGNRQP peptide on the surface. The fluorescent images showed that assembled phage fusion proteins can direct the nanostructure into cancer cells. The nanostructure was more efficient than gold nanorods and silver nanotriangle-based photothermal agents and was capable of specifically ablating SW620 cells after 10 min illumination with an 808 nm laser in the light intensity of 4 W/cm(2). The prepared nanostructure would become an ideal reagent for simutaneously targeted optical imaging and PTT of tumor.


Assuntos
Biomimética , Proteínas do Capsídeo/química , Neoplasias Colorretais/terapia , Nanoestruturas/química , Fototerapia , Proteínas do Capsídeo/uso terapêutico , Linhagem Celular Tumoral , Neoplasias Colorretais/ultraestrutura , Neoplasias Colorretais/virologia , Ouro/química , Humanos , Nanoestruturas/uso terapêutico , Nanotubos/química , Rodaminas/química , Rodaminas/uso terapêutico , Prata/química , Espectroscopia de Luz Próxima ao Infravermelho
20.
Mol Cancer Ther ; 13(12): 2864-75, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25239936

RESUMO

In an effort to improve the therapeutic index of cancer chemotherapy, we developed an advanced nanopreparation based on the combination of landscape phage display to obtain new targeting ligands with micellar nanoparticles for tumor targeting of water-insoluble neoplastic agents. With paclitaxel as a drug, this self-assembled nanopreparation composed of MCF-7-specific phage protein and polyethylene glycol-phosphatidylethanolamine (PEG-PE) micelles showed selective toxicity to target cancer cells rather than nontarget, non cancer cells in vitro. In vivo, the targeted phage micelles triggered a dramatic tumor reduction and extensive necrosis as a result of improved tumor delivery of paclitaxel. The enhanced anticancer effect was also verified by an enhanced apoptosis and reduced tumor cell proliferation following the treatment with the targeted micellar paclitaxel both in vitro and in vivo. The absence of hepatotoxicity and pathologic changes in tissue sections of vital organs, together with maintenance of overall health of mice following the treatment, further support its translational potential as an effective and safe chemotherapy for improved breast cancer treatment.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Micelas , Nanopartículas , Neoplasias/metabolismo , Neoplasias/patologia , Paclitaxel/administração & dosagem , Fosfatidiletanolaminas , Polietilenoglicóis , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Camundongos , Neoplasias/tratamento farmacológico , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...