Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Nanobiotechnology ; 21(1): 200, 2023 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-37344839

RESUMEN

The emergence of human severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses significant challenges to global public health. Despite the extensive efforts of researchers worldwide, there remains considerable opportunities for improvement in timely diagnosis, specific treatment, and effective vaccines for SARS-CoV-2. This is due, in part, to the large number of asymptomatic carriers, rapid virus mutations, inconsistent confinement policies, untimely diagnosis and limited clear treatment plans. The emerging of nanozymes offers a promising approach for combating SARS-CoV-2 due to their stable physicochemical properties and high surface areas, which enable easier and multiple nano-bio interactions in vivo. Nanozymes inspire the development of sensitive and economic nanosensors for rapid detection, facilitate the development of specific medicines with minimal side effects for targeted therapy, trigger defensive mechanisms in the form of vaccines, and eliminate SARS-CoV-2 in the environment for prevention. In this review, we briefly present the limitations of existing countermeasures against coronavirus disease 2019 (COVID-19). We then reviewed the applications of nanozyme-based platforms in the fields of diagnostics, therapeutics and the prevention in COVID-19. Finally, we propose opportunities and challenges for the further development of nanozyme-based platforms for COVID-19. We expect that our review will provide valuable insights into the new emerging and re-emerging infectious pandemic from the perspective of nanozymes.


Asunto(s)
COVID-19 , Vacunas , Humanos , COVID-19/diagnóstico , COVID-19/prevención & control , SARS-CoV-2 , Vacunas contra la COVID-19 , Pandemias/prevención & control , Prueba de COVID-19
2.
Molecules ; 28(9)2023 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-37175248

RESUMEN

Low-density lipoprotein receptor-related protein 6 (LRP6), a member of the low-density lipoprotein receptor (LDLR) family, displays a unique structure and ligand-binding function. As a co-receptor of the Wnt/ß-catenin signaling pathway, LRP6 is a novel therapeutic target that plays an important role in the regulation of cardiovascular disease, lipid metabolism, tumorigenesis, and some classical signals. By using capillary electrophoresis-systematic evolution of ligands by exponential enrichment (CE-SELEX), with recombinant human LRP-6 as the target, four candidate aptamers with a stem-loop structure were selected from an ssDNA library-AptLRP6-A1, AptLRP6-A2, AptLRP6-A3, and AptLRP6-A4. The equilibrium dissociation constant KD values between these aptamers and the LRP6 protein were in the range of 0.105 to 1.279 µmol/L, as determined by CE-LIF analysis. Their affinities and specificities were further determined by the gold nanoparticle (AuNP) colorimetric method. Among them, AptLRP6-A3 showed the highest affinity with LRP6-overexpressed human breast cancer cells. Therefore, the LRP6 aptamer identified in this study constitutes a promising modality for the rapid diagnosis and treatment of LRP6-related diseases.


Asunto(s)
Aptámeros de Nucleótidos , Nanopartículas del Metal , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Lipoproteínas LDL , Oro , ADN de Cadena Simple , Aptámeros de Nucleótidos/química
3.
Small ; 18(33): e2202294, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35869033

RESUMEN

Artificial organelles are compartmentalized nanoreactors, in which enzymes or enzyme-mimic catalysts exhibit cascade catalytic activities to mimic the functions of natural organelles. Importantly, research on artificial organelles paves the way for the bottom-up design of synthetic cells. Due to the separation effect of microcompartments, the catalytic reactions of enzymes are performed without the influence of the surrounding medium. The current techniques for synthesizing artificial organelles rely on the strategies of encapsulating enzymes into vesicle-structured materials or reconstituting enzymes onto the microcompartment materials. However, there are still some problems including limited functions, unregulated activities, and difficulty in targeting delivery that hamper the applications of artificial organelles. The emergence of nanozymes (nanomaterials with enzyme-like activities) provides novel ideas for the fabrication of artificial organelles. Compared with natural enzymes, nanozymes are featured with multiple enzymatic activities, higher stability, easier to synthesize, lower cost, and excellent recyclability. Herein, the most recent advances in nanozyme-based artificial organelles are summarized. Moreover, the benefits of compartmental structures for the applications of nanozymes, as well as the functional requirements of microcompartment materials are also introduced. Finally, the potential applications of nanozyme-based artificial organelles in biomedicine and the related challenges are discussed.


Asunto(s)
Células Artificiales , Nanoestructuras , Catálisis , Nanoestructuras/química , Orgánulos
4.
Small ; 18(39): e2202145, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36026572

RESUMEN

Enzymes are an important component for bottom-up building of synthetic/artificial cells. Nanozymes are nanomaterials with intrinsic enzyme-like properties, however, the construction of synthetic cells using nanozymes is difficult owing to their high surface energy or large size. Herein, the authors show a protein-based general platform that biomimetically integrates various ultrasmall metal nanozymes into protein shells. Specifically, eight metal-based ultrasmall nano-particles/clusters are in situ incorporated into ferritin nanocages that are self-assembled by 24 subunits of ferritin heavy chain. As a nanozyme generator, such a platform is suitable for screening the desired enzyme-like activities, including peroxidase (POD), oxidase (OXD), catalase (CAT) and superoxide dismutase (SOD). After screening, it is found that Ru intrinsically possesses the highest POD-like and CAT-like activities, while Mn and Pt show the highest OXD-like and SOD-like activities, respectively. Additionally, the inducers/inhibitors of various nanozymes are screened from more than 50 compounds to improve or inhibit their enzyme-like activities. Based on the screened nanozymes and their inhibitors, a proof-of-conceptually constructs cell-mimicking catalytic vesicles to mimic or modulate the events of redox homeostasis in living cells. This study offers a type of artificial metalloenzyme based on nanotechnology and shows a choice for bottom-up enzyme-based synthetic cell systems in a fully synthetic manner.


Asunto(s)
Apoferritinas , Nanoestructuras , Catalasa , Catálisis , Ferritinas , Peroxidasa , Peroxidasas , Superóxido Dismutasa
5.
Small ; 17(25): e2101655, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34028968

RESUMEN

The detection of autoantibodies is critical for diagnosis of autoimmune diseases. However, the sensitivity is often limited by the properties of the antigens and the detection systems such as enzyme-linked immunosorbent assay (ELISA). Here, employing the multidisplay ability of ferritin, a highly sensitive nanocage-based capture-detection system is designed, of which the sensitivity is 100-1000-fold higher than that of conventional ELISA methods. The capture nanocages are constructed by displaying the primary Sjögren's syndrome (pSS)-related antigenic peptides on ferritin nanocage, which present epitopes effectively and high affinity, leading to tenfold higher capture capability for autoantibodies. Human IgG Fc-binding peptides are also engineered on ferritin nanocage, which enable high binding affinity and efficient horseradish peroxidase (HRP)-labeling. Compared with commercial HRP-conjugated anti-human IgG antibody, the nanocage-based detecting probe exhibited more than tenfold increased sensitivity. Autoantibodies are then examined in 91 sera from patients with pSS, 51 from rheumatoid arthritis, 54 from systemic lupus erythematosus, and 55 from healthy individuals by using the nanocage-based ELISA. The results indicate that the nanocage-based capture-detection system is an effective detection platform and provide a novel and more sensitive method for the diagnosis of autoimmune diseases.


Asunto(s)
Artritis Reumatoide , Enfermedades Autoinmunes , Lupus Eritematoso Sistémico , Síndrome de Sjögren , Autoanticuerpos , Enfermedades Autoinmunes/diagnóstico , Ensayo de Inmunoadsorción Enzimática , Humanos , Lupus Eritematoso Sistémico/diagnóstico
6.
J Am Chem Soc ; 142(3): 1636-1644, 2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31880441

RESUMEN

Recent advances in nanomedicine have facilitated the development of potent nanomaterials with intrinsic enzyme-like activities (nanozymes) for cancer therapy. However, it remains a great challenge to fabricate smart nanozymes that precisely perform enzymatic activity in tumor microenvironment without inducing off-target toxicity to surrounding normal tissues. Herein, we report on designed fabrication of biodegradation-medicated enzymatic activity-tunable molybdenum oxide nanourchins (MoO3-x NUs), which selectively perform therapeutic activity in tumor microenvironment via cascade catalytic reactions, while keeping normal tissues unharmed due to their responsive biodegradation in physiological environment. Specifically, the MoO3-x NUs first induce catalase (CAT)-like reactivity to decompose hydrogen peroxide (H2O2) in tumor microenvironment, producing a considerable amount of O2 for subsequent oxidase (OXD)-like reactivity of MoO3-x NUs; a substantial cytotoxic superoxide radical (·O2-) is thus generated for tumor cell apoptosis. Interestingly, once exposed to neutral blood or normal tissues, MoO3-x NUs rapidly lose the enzymatic activity via pH-responsive biodegradation and are excreted in urine, thus ultimately ensuring safety. The current study demonstrates a proof of concept of biodegradation-medicated in vivo catalytic activity-tunable nanozymes for tumor-specific cascade catalytic therapy with minimal off-target toxicity.


Asunto(s)
Catalasa/metabolismo , Molibdeno/química , Nanopartículas/química , Óxidos/química , Oxidorreductasas/metabolismo , Animales , Apoptosis , Catálisis , Humanos , Prueba de Estudio Conceptual , Microambiente Tumoral
7.
Nanomedicine ; 27: 102204, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32294568

RESUMEN

The vexing difficulty in distinguishing glioma from normal tissues is a major obstacle to prognosis. In an attempt to solve this problem, we used a joint strategy that combined targeted-cancer stem cells nanoparticles with precise photoacoustic and fluorescence navigation. We showed that traditional magnetic resonance imaging (MRI) did not represent the true morphology of tumors. Targeted nanoparticles specifically accumulated in the tumor area. Glioma was precisely revealed at the cellular level. Tumors could be non-invasively detected through the intact skull by fluorescence molecular imaging (FMI) and photoacoustic tomography (PAT). Moreover, PAT can be used to excise deep gliomas. Histological correlation confirmed that FMI imaging accurately delineated scattered tumor cells. The combination of optical PAT and FMI navigation fulfilled the promise of precise visual imaging in glioma detection and resection. This detection method was deeper and more intuitive than the current intraoperative pathology.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Glioma/tratamiento farmacológico , Nanopartículas/química , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Glioma/patología , Humanos , Imagen por Resonancia Magnética , Ratones , Imagen Molecular , Nanopartículas/uso terapéutico , Imagen Óptica , Técnicas Fotoacústicas/métodos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Nano Lett ; 19(12): 8887-8895, 2019 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-31671939

RESUMEN

Cerebral malaria is a lethal complication of malaria infection characterized by central nervous system dysfunction and is often not effectively treated by antimalarial combination therapies. It has been shown that the sequestration of the parasite-infected red blood cells that interact with cerebral vessel endothelial cells and the damage of the blood-brain barrier (BBB) play critical roles in the pathogenesis. In this study, we developed a ferritin nanozyme (Fenozyme) composed of recombinant human ferritin (HFn) protein shells that specifically target BBB endothelial cells (BBB ECs) and the inner Fe3O4 nanozyme core that exhibits reactive oxygen species-scavenging catalase-like activity. In the experimental cerebral malaria (ECM) mouse model, administration of the Fenozyme, but not HFn, markedly ameliorated the damage of BBB induced by the parasite and improved the survival rate of infected mice significantly. Further investigations found that Fenozyme, as well as HFn, was able to polarize the macrophages in the liver to the M1 phenotype and promote the elimination of malaria in the blood. Thus, the catalase-like activity of the Fenozyme is required for its therapeutic effect in the mouse model. Moreover, the Fenozyme significantly alleviated the brain inflammation and memory impairment in ECM mice that had been treated with artemether, indicating that combining Fenozyme with an antimalarial drug is a novel strategy for the treatment of cerebral malaria.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Ferritinas/farmacología , Malaria Cerebral/prevención & control , Plasmodium berghei/metabolismo , Animales , Barrera Hematoencefálica/parasitología , Barrera Hematoencefálica/patología , Modelos Animales de Enfermedad , Células Endoteliales/parasitología , Células Endoteliales/patología , Ferritinas/genética , Humanos , Inflamación/metabolismo , Inflamación/parasitología , Inflamación/patología , Inflamación/prevención & control , Hígado/metabolismo , Hígado/parasitología , Hígado/patología , Macrófagos/metabolismo , Macrófagos/parasitología , Macrófagos/patología , Malaria Cerebral/metabolismo , Malaria Cerebral/patología , Ratones , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
9.
Nano Lett ; 19(1): 203-209, 2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30539641

RESUMEN

Photoacoustic imaging (PAI) is an attractive imaging modality, which is promising for clinical cancer diagnosis due to its advantages on deep tissue penetration and fine spatial resolution. However, few tumor catalytic/responsive PAI strategies are developed. Here, we design an exosome-like nanozyme vesicle for in vivo H2O2-responsive PAI of nasopharyngeal carcinoma (NPC). The intrinsic peroxidase-like activity of graphene quantum dot nanozyme (GQDzyme) effectively converts the 2,2'-azino- bis (3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) into its oxidized form in the presence of H2O2. The oxidized ABTS exhibits strong near-infrared (NIR) absorbance, rendering it to be an ideal contrast agent for PAI. Thus, GQDzyme/ABTS nanoparticle is a novel type of catalytic PAI contrast agent, which is sensitive to H2O2 produced from NPC cells. Furthermore, we develop an approach to construct exosome-like nanozyme vesicle via biomimetic functionalization of GQDzyme/ABTS nanoparticle with natural erythrocyte membrane modified with folate acid. In vivo animal experiments demonstrated that this exosome-like nanozyme vesicle effectively accumulated in NPC and selectively triggered catalytic PAI for NPC. In addition, our nanozyme vesicle exhibits excellent biocompatibility and stealth ability for long blood circulation. Together, we demonstrate that GQDzyme/ABTS based exosome-like nanozyme vesicle is an ideal nanoplatform for developing deep-tissue tumor-targeted catalytic PAI in vivo.


Asunto(s)
Exosomas/química , Nanopartículas/administración & dosificación , Carcinoma Nasofaríngeo/tratamiento farmacológico , Técnicas Fotoacústicas , Animales , Benzotiazoles/química , Benzotiazoles/farmacología , Catálisis , Exosomas/efectos de los fármacos , Xenoinjertos , Humanos , Peróxido de Hidrógeno/química , Ratones , Nanopartículas/química , Carcinoma Nasofaríngeo/patología , Peroxidasa/química , Ácidos Sulfónicos/química , Ácidos Sulfónicos/farmacología
10.
Mikrochim Acta ; 186(12): 782, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31729634

RESUMEN

Nanomaterials with intrinsic enzymatic activity are often referred to as nanozymes. They exhibit many advantages over natural enzymes such as temporal and thermal stability, recyclability, controllable activity, and ease of large-scale preparation. Many efforts have been made in the past 5 years in order to improve their specificity for chiral substrates. This review (with 74 refs.) summarizes the state of the art in the design of nanozymes with chiral selectivity. Following an introduction into nanozymes and chiral selectivity in general, a first large section covers nanozymes based on the use of chiral chemicals. The next two sections describe nanozymes using amino acids and DNA as chiral ligands. A table summarizes the kinetic and selectivity parameters of the currently known chiral enzyme mimics. A concluding section addresses current challenges, and gives perspectives and an outlook on trends. Graphical abstract Chiral nanozymes exhibit the ability of asymmetric catalysis and enantioselective discrimination by modifying with chiral ligands.


Asunto(s)
Nanopartículas del Metal/química , Aminoácidos/química , Catálisis , ADN/química , Cinética , Ligandos , Estereoisomerismo
11.
Angew Chem Int Ed Engl ; 58(15): 4911-4916, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30697885

RESUMEN

Single-atom catalysts (SACs), as homogeneous catalysts, have been widely explored for chemical catalysis. However, few studies focus on the applications of SACs in enzymatic catalysis. Herein, we report that a zinc-based zeolitic-imidazolate-framework (ZIF-8)-derived carbon nanomaterial containing atomically dispersed zinc atoms can serve as a highly efficient single-atom peroxidase mimic. To reveal its structure-activity relationship, the structural evolution of the single-atom nanozyme (SAzyme) was systematically investigated. Furthermore, the coordinatively unsaturated active zinc sites and catalytic mechanism of the SAzyme are disclosed using density functional theory (DFT) calculations. The SAzyme, with high therapeutic effect and biosafety, shows great promises for wound antibacterial applications.


Asunto(s)
Antibacterianos/farmacología , Estructuras Metalorgánicas/farmacología , Nanopartículas/química , Infecciones por Pseudomonas/tratamiento farmacológico , Cicatrización de Heridas/efectos de los fármacos , Antibacterianos/química , Catálisis , Teoría Funcional de la Densidad , Desinfección , Imidazoles/química , Imidazoles/farmacología , Estructuras Metalorgánicas/química , Pruebas de Sensibilidad Microbiana , Tamaño de la Partícula , Infecciones por Pseudomonas/patología , Propiedades de Superficie , Zeolitas/química , Zeolitas/farmacología , Zinc/química , Zinc/farmacología
12.
Anal Chem ; 90(9): 5671-5677, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29634235

RESUMEN

Nanobodies consist of a single domain variable fragment of a camelid heavy-chain antibody. Nanobodies have potential applications in biomedical fields because of their simple production procedures and low cost. Occasionally, nanobody clones of interest exhibit low affinities for their target antigens, which, together with their short half-life limit bioanalytical or therapeutic applications. Here, we developed a novel platform we named fenobody, in which a nanobody developed against H5N1 virus is displayed on the surface of ferritin in the form of a 24mer. We constructed a fenobody by substituting the fifth helix of ferritin with the nanobody. TEM analysis showed that nanobodies were displayed on the surface of ferritin in the form of 6 × 4 bundles, and that these clustered nanobodies are flexible for antigen binding in spatial structure. Comparing fenobodies with conventional nanobodies currently used revealed that the antigen binding apparent affinity of anti-H5N1 fenobody was dramatically increased (∼360-fold). Crucially, their half-life extension in a murine model was 10-fold longer than anti-H5N1 nanobody. In addition, we found that our fenobodies are highly expressed in Escherichia coli, and are both soluble and thermo-stable nanocages that self-assemble as 24-polymers. In conclusion, our results demonstrate that fenobodies have unique advantages over currently available systems for apparent affinity enhancement and half-life extension of nanobodies. Our fenobody system presents a suitable platform for various large-scale biotechnological processes and should greatly facilitate the application of nanobody technology in these areas.


Asunto(s)
Antivirales/química , Ferritinas/química , Anticuerpos de Dominio Único/química , Animales , Antivirales/farmacología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Ferritinas/farmacología , Semivida , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Ratones , Microscopía Electrónica de Transmisión , Modelos Moleculares , Peso Molecular , Tamaño de la Partícula , Anticuerpos de Dominio Único/farmacología , Propiedades de Superficie
13.
Nanomedicine ; 14(7): 2259-2270, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30056091

RESUMEN

Optical imaging technologies improve clinical diagnostic accuracy of early gastric cancer (EGC). However, there was a lack of imaging agents exhibiting molecular specificity for EGCs. Here, we employed the dye labeled human heavy-chain ferritin (HFn) as imaging nanoprobe, which recognizes tumor biomarker transferrin receptor 1 (TfR1), to enable specific EGC imaging using confocal laser endomicroscopy (CLE). TfR1 expression was initially examined in vitro in gastric tumor cells and in vivo through whole-body fluorescence and CLE imaging in tumor-bearing mice. Subsequently, dye labeled HFn was topically applied to resected human tissues for EGC detection. CLE analysis of TfR1-targeted fluorescence imaging allowed distinction of neoplastic from non-neoplastic tissues (P < 0.0001), and TfR1 expression level was found to correlate with EGC differentiation degrees (P < 0.0001). Notably, the CLE evaluation correlated well with the immunohistochemical findings (κ-coefficient: 0.8023). Our HFn-nanoprobe-based CLE increases the accuracy of EGC detection and enables visualization of tumor margins and endoscopic resection.


Asunto(s)
Antígenos CD/metabolismo , Apoferritinas/metabolismo , Endoscopía/métodos , Colorantes Fluorescentes/química , Imagen Molecular/métodos , Nanopartículas/administración & dosificación , Receptores de Transferrina/metabolismo , Neoplasias Gástricas/diagnóstico por imagen , Anciano , Anciano de 80 o más Años , Animales , Apoferritinas/administración & dosificación , Apoferritinas/química , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Microscopía Fluorescente , Persona de Mediana Edad , Nanopartículas/química , Pronóstico , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Proc Natl Acad Sci U S A ; 111(41): 14900-5, 2014 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-25267615

RESUMEN

An ideal nanocarrier for efficient drug delivery must be able to target specific cells and carry high doses of therapeutic drugs and should also exhibit optimized physicochemical properties and biocompatibility. However, it is a tremendous challenge to engineer all of the above characteristics into a single carrier particle. Here, we show that natural H-ferritin (HFn) nanocages can carry high doses of doxorubicin (Dox) for tumor-specific targeting and killing without any targeting ligand functionalization or property modulation. Dox-loaded HFn (HFn-Dox) specifically bound and subsequently internalized into tumor cells via interaction with overexpressed transferrin receptor 1 and released Dox in the lysosomes. In vivo in the mouse, HFn-Dox exhibited more than 10-fold higher intratumoral drug concentration than free Dox and significantly inhibited tumor growth after a single-dose injection. Importantly, HFn-Dox displayed an excellent safety profile that significantly reduced healthy organ drug exposure and improved the maximum tolerated dose by fourfold compared with free Dox. Moreover, because the HFn nanocarrier has well-defined morphology and does not need any ligand modification or property modulation it can be easily produced with high purity and yield, which are requirements for drugs used in clinical trials. Thus, these unique properties make the HFn nanocage an ideal vehicle for efficient anticancer drug delivery.


Asunto(s)
Apoferritinas/uso terapéutico , Doxorrubicina/uso terapéutico , Nanopartículas/uso terapéutico , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoferritinas/farmacocinética , Apoferritinas/farmacología , Relación Dosis-Respuesta a Droga , Doxorrubicina/sangre , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Endocitosis/efectos de los fármacos , Femenino , Células HT29 , Humanos , Inyecciones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/ultraestructura , Neoplasias/sangre , Neoplasias/patología , Distribución Tisular/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Blood ; 120(11): 2330-9, 2012 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-22718841

RESUMEN

CD146 is a novel endothelial biomarker and plays an essential role in angiogenesis; however, its role in the molecular mechanism underlying angiogenesis remains poorly understood. In the present study, we show that CD146 interacts directly with VEGFR-2 on endothelial cells and at the molecular level and identify the structural basis of CD146 binding to VEGFR-2. In addition, we show that CD146 is required in VEGF-induced VEGFR-2 phosphorylation, AKT/p38 MAPKs/NF-κB activation, and thus promotion of endothelial cell migration and microvascular formation. Furthermore, we show that anti-CD146 AA98 or CD146 siRNA abrogates all VEGFR-2 activation induced by VEGF. An in vivo angiogenesis assay showed that VEGF-promoted microvascular formation was impaired in the endothelial conditional knockout of CD146 (CD146(EC-KO)). Our animal experiments demonstrated that anti-CD146 (AA98) and anti-VEGF (bevacizumab) have an additive inhibitory effect on xenografted human pancreatic and melanoma tumors. The results of the present study suggest that CD146 is a new coreceptor for VEGFR-2 and is therefore a promising target for blocking tumor-related angiogenesis.


Asunto(s)
Endotelio Vascular/metabolismo , Neovascularización Patológica/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antígeno CD146/química , Antígeno CD146/genética , Antígeno CD146/metabolismo , Línea Celular Tumoral , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Noqueados , Ratones Desnudos , Terapia Molecular Dirigida , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Organismos Libres de Patógenos Específicos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
16.
ACS Nano ; 18(4): 2533-2540, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38215476

RESUMEN

Nanozymes, nanomaterials exhibiting enzyme-like activities, have emerged as a prominent interdisciplinary field over the past decade. To date, over 1200 different nanomaterials have been identified as nanozymes, covering four catalytic categories: oxidoreductases, hydrolases, isomerases, and lyases. Catalytic activity and specificity are two pivotal benchmarks for evaluating enzymatic performance. Despite substantial progress being made in quantifying and optimizing the catalytic activity of nanozymes, there is still a lack of in-depth research on the catalytic specificity of nanozymes, preventing the formation of consensual knowledge and impeding a more refined and systematic classification of nanozymes. Recently, debates have emerged regarding whether nanozymes could possess catalytic specificity similar to that of enzymes. This Perspective discusses the specificity of nanozymes by referring to the catalytic specificity of enzymes, highlights the specificity gap between nanozymes and enzymes, and concludes by offering our perspective on future research on the specificity of nanozymes.


Asunto(s)
Nanoestructuras , Catálisis
17.
Adv Mater ; 36(10): e2210144, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36730098

RESUMEN

Ischemic stroke (IS) is one of the most common causes of disability and death. Thrombolysis and neuroprotection are two current major therapeutic strategies to overcome ischemic and reperfusion damage. In this work, a novel peptide-templated manganese dioxide nanozyme (PNzyme/MnO2 ) is designed that integrates the thrombolytic activity of functional peptides with the reactive oxygen species scavenging ability of nanozymes. Through self-assembled polypeptides that contain multiple functional motifs, the novel peptide-templated nanozyme is able to bind fibrin in the thrombus, cross the blood-brain barrier, and finally accumulate in the ischemic neuronal tissues, where the thrombolytic motif is "switched-on" by the action of thrombin. In mice and rat IS models, the PNzyme/MnO2 prolongs the blood-circulation time and exhibits strong thrombolytic action, and reduces the ischemic damages in brain tissues. Moreover, this peptide-templated nanozyme also effectively inhibits the activation of astrocytes and the secretion of proinflammatory cytokines. These data indicate that the rationally designed PNzyme/MnO2 nanozyme exerts both thrombolytic and neuroprotective actions. Giving its long half-life in the blood and ability to target brain thrombi, the biocompatible nanozyme may serve as a novel therapeutic agent to improve the efficacy and prevent secondary thrombosis during the treatment of IS.


Asunto(s)
Accidente Cerebrovascular Isquémico , Fármacos Neuroprotectores , Accidente Cerebrovascular , Ratas , Ratones , Animales , Compuestos de Manganeso/farmacología , Trombina , Neuroprotección , Óxidos , Fibrinolíticos/uso terapéutico , Isquemia , Péptidos/farmacología , Péptidos/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico
18.
Adv Mater ; 36(10): e2300387, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37086206

RESUMEN

Nanozymes have great potential to be used as an alternative to natural enzymes in a variety of fields. However, low catalytic activity compared with natural enzymes limits their practical use. It is still challenging to design nanozymes comparable to their natural counterparts in terms of the specific activity. In this study, a surface engineering strategy is employed to improve the specific activity of Ru nanozymes using charge-transferrable ligands such as polystyrene sulfonate (PSS). PSS-modified Ru nanozyme exhibits a peroxidase-like specific activity of up to 2820 U mg-1 , which is twice that of horseradish peroxidase (1305 U mg-1 ). Mechanism studies suggest that PSS readily accepts negative charge from Ru, thus reducing the affinity between Ru and ·OH. Importantly, the modified Ru-peroxidase nanozyme is successfully used to develop an immunoassay for human alpha-fetoprotein and achieves a 140-fold increase in detection sensitivity compared with traditional horseradish-peroxidase-based enzyme-linked immunosorbent assay. Therefore, this work provides a feasible route to design nanozymes with high specific activity that meets the practical use as an alternative to natural enzymes.


Asunto(s)
Rutenio , Humanos , Peroxidasa de Rábano Silvestre , Ligandos , Peroxidasa , Peroxidasas , Inmunoensayo
19.
Adv Mater ; 36(8): e2307337, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37724878

RESUMEN

Nanozymes, next-generation enzyme-mimicking nanomaterials, have entered an era of rational design; among them, Co-based nanozymes have emerged as captivating players over times. Co-based nanozymes have been developed and have garnered significant attention over the past five years. Their extraordinary properties, including regulatable enzymatic activity, stability, and multifunctionality stemming from magnetic properties, photothermal conversion effects, cavitation effects, and relaxation efficiency, have made Co-based nanozymes a rising star. This review presents the first comprehensive profiling of the Co-based nanozymes in the chemistry, biology, and environmental sciences. The review begins by scrutinizing the various synthetic methods employed for Co-based nanozyme fabrication, such as template and sol-gel methods, highlighting their distinctive merits from a chemical standpoint. Furthermore, a detailed exploration of their wide-ranging applications in biosensing and biomedical therapeutics, as well as their contributions to environmental monitoring and remediation is provided. Notably, drawing inspiration from state-of-the-art techniques such as omics, a comprehensive analysis of Co-based nanozymes is undertaken, employing analogous statistical methodologies to provide valuable guidance. To conclude, a comprehensive outlook on the challenges and prospects for Co-based nanozymes is presented, spanning from microscopic physicochemical mechanisms to macroscopic clinical translational applications.


Asunto(s)
Ciencia Ambiental , Nanoestructuras , Catálisis , Nanoestructuras/química
20.
Adv Sci (Weinh) ; 11(3): e2305217, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38029345

RESUMEN

Hydrogels are prevailing drug delivery depots to improve antitumor efficacy and reduce systemic toxicity. However, the application of conventional free drug-loaded hydrogel is hindered by poor drug penetration in solid tumors. Here, an injectable ferritin-based nanocomposite hydrogel is constructed to facilitate tumor penetration and improve cancer chemoimmunotherapy. Specifically, doxorubicin-loaded human ferritin (Dox@HFn) and oxidized dextran (Dex-CHO) are used to construct the injectable hydrogel (Dox@HFn Gel) through the formation of pH-sensitive Schiff-base bonds. After peritumoral injection, the Dox@HFn Gel is retained locally for up to three weeks, and released intact Dox@HFn gradually, which can not only facilitate tumor penetration through active transcytosis but also induce immunogenic cell death (ICD) to tumor cells to generate an antitumor immune response. Combining with anti-programmed death-1 antibody (αPD-1), Dox@HFn Gel induces remarkable regression of orthotopic 4T1 breast tumors, further elicits a strong systemic anti-tumor immune response to effectively suppress tumor recurrence and lung metastasis of 4T1 tumors after surgical resection. Besides, the combination of Dox@HFn GelL with anti-CD47 antibody (αCD47) inhibits postsurgical tumor recurrence of aggressive orthotopic glioblastoma tumor model and significantly extends mice survival. This work sheds light on the construction of local hydrogels to potentiate antitumor immune response for improved cancer therapy.


Asunto(s)
Ferritinas , Recurrencia Local de Neoplasia , Humanos , Ratones , Animales , Nanogeles , Recurrencia Local de Neoplasia/tratamiento farmacológico , Doxorrubicina/farmacología , Hidrogeles/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA