Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 175
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
Acc Chem Res ; 52(8): 2190-2200, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31276379

RESUMEN

Nanozymes are nanomaterials with intrinsic enzyme-like characteristics that have been booming over the past decade because of their capability to address the limitations of natural enzymes such as low stability, high cost, and difficult storage. Along with the rapid development and ever-deepening understanding of nanoscience and nanotechnology, nanozymes hold promise to serve as direct surrogates of traditional enzymes by mimicking and further engineering the active centers of natural enzymes. In 2007, we reported the first evidence that Fe3O4 nanoparticles (NPs) have intrinsic peroxidase-mimicking activity, and since that time, hundreds of nanomaterials have been found to mimic the catalytic activity of peroxidase, oxidase, catalase, haloperoxidase, glutathione peroxidase, uricase, methane monooxygenase, hydrolase, and superoxide dismutase. Uniquely, a broad variety of nanomaterials have been reported to simultaneously exhibit dual- or multienzyme mimetic activity. For example, Fe3O4 NPs show pH-dependent peroxidase-like and catalase-like activities; Prussian blue NPs simultaneously possess peroxidase-, catalase-, and superoxide dismutase-like activity; and Mn3O4 NPs mimic all three cellular antioxidant enzymes including superoxide dismutase, catalase, and glutathione peroxidase. Taking advantage of the physiochemical properties of nanomaterials, nanozymes have shown a broad range of applications from in vitro detection to replacing specific enzymes in living systems. With the emergence of the new concept of "nanozymology", nanozymes have now become an emerging new field connecting nanotechnology and biology. Since the landmark paper on nanozymes was published in 2007, we have extensively explored their catalytic mechanism, established the corresponding standards to quantitatively determine their catalytic activities, and opened up a broad range of applications from biological detection and environmental monitoring to disease diagnosis and biomedicine development. Here we mainly focus on our progress in the systematic design and construction of functionally specific nanozymes, the standardization of nanozyme research, and the exploration of their applications for replacing natural enzymes in living systems. We also show that, by combining the unique physicochemical properties and enzyme-like catalytic activities, nanozymes can offer a variety of multifunctional platforms with a broad of applications from in vitro detection to in vivo monitoring and therapy. For instance, targeting antibody-conjugated ferromagnetic nanozymes simultaneously provide three functions: target capture, magnetic separation, and nanozyme color development for target detection. We finally will address the prospect of nanozyme research to become "nanozymology". We expect that nanozymes with unique physicochemical properties and intrinsic enzyme-mimicking catalytic properties will attract broad interest in both fundamental research and practical applications and offer new opportunities for traditional enzymology.


Asunto(s)
Nanopartículas de Magnetita/química , Animales , Carbono/química , Catálisis , Colorimetría/métodos , Colorantes/química , Humanos , Inmunoensayo/métodos , Inmunohistoquímica/métodos , Cinética , Neoplasias/diagnóstico , Nitrógeno/química , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo
6.
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
7.
Proc Natl Acad Sci U S A ; 114(36): E7622-E7631, 2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28827364

RESUMEN

The blood-brain barrier (BBB) establishes a protective interface between the central neuronal system and peripheral blood circulation and is crucial for homeostasis of the CNS. BBB formation starts when the endothelial cells (ECs) invade the CNS and pericytes are recruited to the nascent vessels during embryogenesis. Despite the essential function of pericyte-EC interaction during BBB development, the molecular mechanisms coordinating the pericyte-EC behavior and communication remain incompletely understood. Here, we report a single cell receptor, CD146, that presents dynamic expression patterns in the cerebrovasculature at the stages of BBB induction and maturation, coordinates the interplay of ECs and pericytes, and orchestrates BBB development spatiotemporally. In mouse brain, CD146 is first expressed in the cerebrovascular ECs of immature capillaries without pericyte coverage; with increased coverage of pericytes, CD146 could only be detected in pericytes, but not in cerebrovascular ECs. Specific deletion of Cd146 in mice ECs resulted in reduced brain endothelial claudin-5 expression and BBB breakdown. By analyzing mice with specific deletion of Cd146 in pericytes, which have defects in pericyte coverage and BBB integrity, we demonstrate that CD146 functions as a coreceptor of PDGF receptor-ß to mediate pericyte recruitment to cerebrovascular ECs. Moreover, we found that the attached pericytes in turn down-regulate endothelial CD146 by secreting TGF-ß1 to promote further BBB maturation. These results reveal that the dynamic expression of CD146 controls the behavior of ECs and pericytes, thereby coordinating the formation of a mature and stable BBB.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Comunicación Celular/fisiología , Células Endoteliales/metabolismo , Pericitos/metabolismo , Animales , Barrera Hematoencefálica/fisiología , Encéfalo/fisiología , Antígeno CD146/metabolismo , Regulación hacia Abajo/fisiología , Células Endoteliales/fisiología , Ratones , Ratones Noqueados , Pericitos/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
8.
Chem Soc Rev ; 48(14): 3683-3704, 2019 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-31119258

RESUMEN

Nanozymes are nanomaterial-based artificial enzymes. By effectively mimicking catalytic sites of natural enzymes or harboring multivalent elements for reactions, nanozyme systems have successfully served as direct surrogates of traditional enzymes for catalysis. With the rapid development and ever-deepening understanding of nanotechnology, nanozymes offer higher catalytic stability, ease of modification and lower manufacturing cost than protein enzymes. Additionally, nanozymes possess inherent nanomaterial properties, providing not only a simple substitute of enzymes but also a multimodal platform interfacing complex biologic environments. Recent extensive research has focused on designing various nanozyme systems that are responsive to one or multiple substrates by tailored means. Catalytic activities of nanozymes can be regulated by pH, H2O2 and glutathione concentrations and levels of oxygenation in different microenvironments. Moreover, nanozymes can be remotely-controlled via different stimuli, including a magnetic field, light, ultrasound, and heat. Collectively, these factors can be adjusted to maximize the diagnostic and therapeutic efficacies of different diseases in biomedical settings. Therefore, by integrating the catalytic property and inherent nanomaterial nature of nanozyme systems, we anticipate that stimuli-responsive nanozymes will open up new horizons for diagnosis, treatment, and theranostics.


Asunto(s)
Tecnología Biomédica , Nanoestructuras/química , Animales , Humanos
9.
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
10.
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
11.
Adv Exp Med Biol ; 1174: 291-312, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31713203

RESUMEN

Fe3O4, also called magnetite, is a naturally occurring mineral and has been widely used in biomedical applications. However, in the past, all the applications were based on its excellent magnetic properties and neglected its catalytic properties. In 2007, we found that Fe3O4 nanoparticles are able to perform intrinsic enzyme-like activities. A specific term, "nanozyme", is used to describe the new property of intrinsic enzymatic activity of nanomaterials. Since then, Fe3O4 nanoparticles have been used as enzyme mimics, which broadens their applications beyond simply their magnetic properties, with applications in biomedical diagnosis and therapy, environmental monitoring and treatment, the food industry and chemical synthesis. In this chapter, we will summarize the basic features of Fe3O4 as an enzyme mimetic and its applications in biomedicine.


Asunto(s)
Biomimética , Nanopartículas de Magnetita , Tecnología Biomédica/tendencias , Catálisis , Enzimas/metabolismo , Nanopartículas de Magnetita/química
12.
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
13.
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
14.
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
15.
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
16.
Nat Mater ; 15(2): 217-26, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26569474

RESUMEN

The notion that animals can detect the Earth's magnetic field was once ridiculed, but is now well established. Yet the biological nature of such magnetosensing phenomenon remains unknown. Here, we report a putative magnetic receptor (Drosophila CG8198, here named MagR) and a multimeric magnetosensing rod-like protein complex, identified by theoretical postulation and genome-wide screening, and validated with cellular, biochemical, structural and biophysical methods. The magnetosensing complex consists of the identified putative magnetoreceptor and known magnetoreception-related photoreceptor cryptochromes (Cry), has the attributes of both Cry- and iron-based systems, and exhibits spontaneous alignment in magnetic fields, including that of the Earth. Such a protein complex may form the basis of magnetoreception in animals, and may lead to applications across multiple fields.


Asunto(s)
Proteínas Hierro-Azufre/metabolismo , Magnetismo , Animales , Anticuerpos , Materiales Biocompatibles , Biofisica , Columbidae/metabolismo , Simulación por Computador , Drosophila melanogaster/metabolismo , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Proteínas Hierro-Azufre/genética , Microscopía Electrónica , Modelos Moleculares , Mutagénesis , Conformación Proteica , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Retina/metabolismo
17.
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
18.
Am J Pathol ; 184(5): 1604-16, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24767106

RESUMEN

Recently, enhanced CD146 expression was reported on endothelial cells in intestinal biopsies from patients with inflammatory bowel disease. However, the underlying mechanism remains unknown. Here, we found that overexpressed endothelial CD146 promoted the inflammatory responses in inflammatory bowel disease, which further potentiated the occurrence of colitis-associated colorectal carcinogenesis. Eliminating endothelial CD146 by conditional knockout significantly ameliorated the severity of inflammation in two different murine models of colitis, and decreased tumor incidence and tumor progression in a murine model of colitis-associated colorectal carcinogenesis. Mechanistic study showed that cytokine tumor necrosis factor-α (TNF-α) up-regulated the expression of endothelial CD146 through NF-κB transactivation. In turn, the enhanced endothelial CD146 expression promoted both angiogenesis and proinflammatory leukocyte extravasations, contributing to inflammation. Using an anti-CD146 antibody, AA98, alone or together with an anti-TNF-α antibody significantly attenuated colitis and prevented colitis-associated colorectal carcinogenesis in mice. Our study provides the first evidence that CD146 plays a dual role on endothelium, facilitating leukocyte extravasations and angiogenesis, thus promoting inflammation. This finding not only reveals the function and regulating mechanism of CD146 in inflammatory bowel disease, but also provides a promising therapeutic strategy for treating inflammatory bowel disease and preventing colitis-associated colorectal carcinogenesis.


Asunto(s)
Antígeno CD146/metabolismo , Carcinogénesis/patología , Colitis/patología , Colitis/prevención & control , Animales , Anticuerpos/farmacología , Comunicación Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/prevención & control , Sulfato de Dextran , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/patología , Interleucina-1beta/metabolismo , Leucocitos/efectos de los fármacos , Leucocitos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Neovascularización Patológica/patología , Activación Transcripcional/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
19.
Am J Pathol ; 184(12): 3308-20, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25307346

RESUMEN

The function of blood-brain barrier is often disrupted during the progression of multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE). However, the molecular mechanism of blood-brain barrier modulation during neuroinflammation remains unclear. Herein, we show that the expression of interferon-γ (IFNγ) receptor on endothelial cells (ECs) protected mice from the brain inflammation during EAE. IFNγ stabilized the integrity of the cerebral endothelium and prevented the infiltration of leukocytes into the brain. Further analysis revealed that IFNγ increased the expression of tight junction proteins zonula occludens protein 1 and occludin, as well as membranous distribution of claudin-5, in brain ECs. Silencing claudin-5 abolished the IFNγ-mediated improvement of EC integrity. Taken together, our results show that IFNγ, a pleiotropic proinflammatory cytokine, stabilizes blood-brain barrier integrity and, therefore, prevents brain inflammation during EAE.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Interferón gamma/inmunología , Animales , Encéfalo/metabolismo , Movimiento Celular , Separación Celular , Células Cultivadas , Claudina-5/metabolismo , Células Endoteliales/metabolismo , Femenino , Citometría de Flujo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Inflamación , Leucocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor TIE-2/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
20.
J Pathol ; 233(2): 170-82, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24615277

RESUMEN

Although IFNγ is regarded as a key cytokine in angiostatic response, our poor understanding of its effective cellular target drastically limits its clinical trials against angiogenesis-related disorders. Here, we investigated the effect of IFNγ on endothelial cells (ECs) and possible molecular mechanisms in angiostasis. By employing Tie2(IFNγR) mice, in which IFNγR expression was reconstituted under the control of Tie2 promoter in IFNγR-deficient mice, we found that the response of ECs to IFNγ was highly effective in inhibiting blood supply and retarding tumour growth. Interestingly, the expression of IFNγR on Tie2(-) cells did not inhibit, but promoted tumour growth in control wild-type mice. Mechanism studies showed that IFNγ reacting on ECs down-regulated the delta-like ligand 4 (Dll4)/Notch signalling pathway. Accordingly, overexpression of Dll4 in human ECs diminished the effect of IFNγ on ECs. This study demonstrates that the action of IFNγ on ECs, but not other cells, is highly effective for tumour angiostasis, which involves down-regulating Dll4. It provides insights for EC-targeted angiostatic therapy in treating angiogenesis-associated disorders in the clinic.


Asunto(s)
Células Endoteliales/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interferón gamma/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neovascularización Patológica , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Unión al Calcio , Línea Celular Tumoral , Regulación hacia Abajo , Células Endoteliales/patología , Regulación Neoplásica de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Neoplasias/genética , Neoplasias/patología , Pericitos/metabolismo , Pericitos/patología , Regiones Promotoras Genéticas , Receptor TIE-2/genética , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Transducción de Señal , Factores de Tiempo , Transfección , Carga Tumoral , Receptor de Interferón gamma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA