Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros

Banco de datos
Tipo del documento
Publication year range
1.
Small ; 20(14): e2306155, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37991257

RESUMEN

Helicobacter pylori (H. pylori) is the major etiological factor of a variety of gastric diseases. However, the treatment of H. pylori is challenged by the destruction of targeted drugs by gastric acid and pepsin. Herein, a dual-targeted cascade catalytic nanozyme PtCo@Graphene@Hemin-2(L-arginine) (PtCo@G@H2A) is designed for the treatment of H. pylori. The dual-targeting ability of PtCo@G@H2A is derived from directly targeting the receptor protein of H. pylori through hemin and responding to the acidic environment to cause charge reversal (protonation of L-arginine) to capture H. pylori, achieving efficient targeting effect. Compared with the single-targeting strategy relying on hemin, the dual-targeting strategy can greatly improve the targeting rate, achieving an increase of 850% targeting rate. At the concentration of NaHCO3 in intestinal fluid, the surface potential of PtCo@G@H2A can be quickly restored to avoid side effects. Meanwhile, PtCo@G@H2A has pH-responsive oxidase-like activity, which can generate nitric oxide (NO) through a cascade catalytic process that first generates reactive oxygen species (ROS) with oxygen, and further oxidizes L-arginine through ROS, realizing a superior acid-selective bactericidal effect. Overall, it proposes a promising strategy for the treatment of H. pylori that maintains high targeting and therapeutic effects in the environment of gastric acid and pepsin.


Asunto(s)
Grafito , Helicobacter pylori , Helicobacter pylori/metabolismo , Pepsina A/farmacología , Especies Reactivas de Oxígeno , Grafito/farmacología , Hemina , Arginina/metabolismo , Arginina/farmacología
2.
Small ; 19(5): e2206127, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36440672

RESUMEN

The pursuit of single-assembled molecular cage reactors for complex tandem reactions is a long-standing target in biomimetic catalysis but still a grand challenge. Herein, nanozyme-like organic cages are reported by engineering air-stable radicals into the skeleton upon photoinduced electron transfer. The generation of radicals is accompanied by single-crystal structural transformation and exhibits superior stability over six months in air. Impressively, the radicals throughout the cage skeleton can mimic the peroxidase of natural enzymes to decompose H2 O2 into OH· and facilitate oxidation reactions. Furthermore, an integrated catalyst by encapsulating Au clusters (glucose oxidase mimics) into the cage has been developed, in which the dual active sites (Au cluster and radical) are spatially isolated and can work as cascade nanozymes to prominently promote the enzyme-like tandem reaction via a substrate channeling effect.

3.
Int J Biol Macromol ; 270(Pt 1): 132277, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38735611

RESUMEN

The high-glycemic microenvironment of diabetic wounds promotes bacterial proliferation, leading to persistent infections and delayed wound healing. This poses a significant threat to human health, necessitating the development of new nanodrug visualization platforms. In this study, we designed and synthesized cascade nano-systems modified with targeted peptide and hyaluronic acid for diabetic infection therapy. The nano-systems were able to target the site of infection using LL-37, and in the microenvironment of wound infection, the hyaluronic acid shell of the nano-systems was degraded by endogenous hyaluronidase. This precise degradation released a cascade of nano-enzymes on the surface of the bacteria, effectively destroying their cytoskeleton. Additionally, the metals in the nano-enzymes provided a photo-thermal effect, accelerating wound healing. The cascade nano-visualization platform demonstrated excellent bactericidal efficacy in both in vitro antimicrobial assays and in vivo diabetic infection models. In conclusion, this nano-system employs multiple approaches including targeting, enzyme-catalyzed therapy, photothermal therapy, and chemodynamic therapy to kill bacteria and promote healing. The Ag@Pt-Au-LYZ/HA-LL-37 formulation shows great potential for the treatment of diabetic wounds.


Asunto(s)
Antibacterianos , Infecciones Bacterianas , Ácido Hialurónico , Cicatrización de Heridas , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Animales , Antibacterianos/farmacología , Antibacterianos/química , Antibacterianos/uso terapéutico , Cicatrización de Heridas/efectos de los fármacos , Infecciones Bacterianas/tratamiento farmacológico , Ratones , Diabetes Mellitus Experimental , Péptidos Catiónicos Antimicrobianos/farmacología , Péptidos Catiónicos Antimicrobianos/química , Hialuronoglucosaminidasa/metabolismo , Catelicidinas , Humanos , Complicaciones de la Diabetes/tratamiento farmacológico , Nanopartículas/química
4.
ACS Nano ; 18(19): 12295-12310, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38695532

RESUMEN

Immune checkpoint blockade (ICB) has brought tremendous clinical progress, but its therapeutic outcome can be limited due to insufficient activation of dendritic cells (DCs) and insufficient infiltration of cytotoxic T lymphocytes (CTLs). Evoking immunogenic cell death (ICD) is one promising strategy to promote DC maturation and elicit T-cell immunity, whereas low levels of ICD induction of solid tumors restrict durable antitumor efficacy. Herein, we report a genetically edited cell membrane-coated cascade nanozyme (gCM@MnAu) for enhanced cancer immunotherapy by inducing ICD and activating the stimulator of the interferon genes (STING) pathway. In the tumor microenvironment (TME), the gCM@MnAu initiates a cascade reaction and generates abundant cytotoxic hydroxyl (•OH), resulting in improved chemodynamic therapy (CDT) and boosted ICD activation. In addition, released Mn2+ during the cascade reaction activates the STING pathway and further promotes the DC maturation. More importantly, activated immunogenicity in the TME significantly improves gCM-mediated PD-1/PD-L1 checkpoint blockade therapy by eliciting systemic antitumor responses. In breast cancer subcutaneous and lung metastasis models, the gCM@MnAu showed synergistically enhanced therapeutic effects and significantly prolonged the survival of mice. This work develops a genetically edited nanozyme-based therapeutic strategy to improve DC-mediated cross-priming of T cells against poorly immunogenic solid tumors.


Asunto(s)
Inmunoterapia , Animales , Ratones , Femenino , Humanos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Ratones Endogámicos BALB C , Línea Celular Tumoral , Muerte Celular Inmunogénica/efectos de los fármacos , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Nanopartículas/química
5.
ACS Appl Mater Interfaces ; 15(39): 46213-46225, 2023 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-37740721

RESUMEN

Recently, nanozymes with peroxidase (POD)-like activity have shown great promise for ferroptosis-based tumor therapy, which are capable of transforming hydrogen peroxide (H2O2) to highly toxic hydroxyl radicals (•OH). However, the unsatisfactory therapeutic performance of nanozymes due to insufficient endogenous H2O2 and acidity at tumor sites has always been a conundrum. Herein, an ultrasmall gold (Au) @ ferrous sulfide (FeS) cascade nanozyme (AuNP@FeS) with H2S-releasing ability constructed with an Au nanoparticle (AuNP) and an FeS nanoparticle (FeSNP) is designed to increase the H2O2 level and acidity in tumor cells via the collaboration between cascade reactions of AuNP@FeS and the biological effects of released H2S, achieving enhanced •OH generation as well as effective ferroptosis for tumor therapy. The cascade reaction in tumor cells is activated by the glucose oxidase (GOD)-like activity of AuNP in AuNP@FeS to catalyze intratumoral glucose into H2O2 and gluconic acid; meanwhile, the released H2S from AuNP@FeS reduces H2O2 consumption by inhibiting intracellular catalase (CAT) activity and promotes lactic acid accumulation. The two pathways synergistically boost H2O2 and acidity in tumor cells, thus inducing a cascade to generate abundant •OH by catalyzing H2O2 through the POD-like activity of FeS in AuNP@FeS and ultimately causing amplified ferroptosis. In vitro and in vivo experiments demonstrated that AuNP@FeS presents a superior tumor therapeutic effect compared to that of AuNP or FeS alone. This strategy represents a simple but powerful method to amplify ferroptosis with H2S-releasing cascade nanozymes and will pave a new way for the development of tumor therapy.

6.
Adv Mater ; 35(44): e2304967, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37608768

RESUMEN

Inflammatory bowel disease (IBD) affects millions of individuals worldwide annually. Enteric reactive oxygen species (ROS) play critical roles in the physiology and pathology of IBD. Nanozymes hold great promise for the treatment of IBD because of their exceptional ability to regulate redox homeostasis during ROS-related inflammation. However, the rapid development of orally administered, acid-tolerant, antioxidant nanozymes for IBD therapy is challenging. Here, a nine-tier high-throughput screening strategy is established to address the multifaceted IBD treatment demands, including intrinsic stability, radioactivity, solubility, gut microbiome toxicity, biomimetic elements, intermediate frontier molecular orbitals, reaction energy barriers, negative charges, and acid tolerance. Ni3 S4 is selected as the best matching material from 146 323 candidates, which exhibits superoxide dismutase-catalase bienzyme-like activity and is 3.13- and 1.80-fold more active than natural enzymes. As demonstrated in a mouse model, Ni3 S4 is stable in the gastrointestinal tract without toxicity and specifically targets the diseased colon to alleviate oxidative stress. RNA and 16S rRNA sequencing analyses show that Ni3 S4 effectively inhibits the cellular pathways of pro-inflammatory factors and restores the gut microbiota. This study not develops a highly efficient orally administered cascade nanozyme for IBD therapy and offers a next-generation paradigm for the rational design of nanomedicine through data-driven approaches.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Ratones , Animales , Especies Reactivas de Oxígeno/metabolismo , ARN Ribosómico 16S/metabolismo , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Inflamación , Estrés Oxidativo
7.
Adv Mater ; 35(22): e2210464, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36964940

RESUMEN

Reactive oxygen species (ROS)-mediated tumor catalytic therapy is typically hindered by gap junction proteins that form cell-to-cell channels to remove cytotoxic ROS, thereby protecting tumor cells from oxidative damage. In this work, a multifunctional nanozyme, FePGOGA, is designed and prepared by Fe(III)-mediated oxidative polymerization (FeP), followed by glucose oxidase (GOx) and GAP19 peptides co-loading through electrostatic and π-π interactions. The FePGOGA nanozyme exhibits excellent cascade peroxidase- and glutathione-oxidase-like activities that efficiently catalyze hydrogen peroxide conversion to hydroxyl radicals and convert reduced glutathione to oxidized glutathione disulfide. The loaded GOx starves the tumors and aggravates tumor oxidative stress through glucose decomposition, while GAP19 peptides block the hemichannels by inducing degradation of Cx43, thus increasing the accumulation of intracellular ROS, and decreasing the transport of intracellular glucose. Furthermore, the ROS reacts with primary amines of heat shock proteins to destroy their structure and function, enabling tumor photothermal therapy at the widely sought-after mild temperature (mildPTT, ≤45 °C). In vivo experiments demonstrate the significant antitumor effectof FePGOGA on cal27 xenograft tumors under near-infrared light irradiation. This study demonstrates the successful ablation of gap junction proteins to overcome resistance to ROS-mediated therapy, providing a regulator to suppress tumor self-preservation during tumor starvation, catalytic therapy, and mildPTT.


Asunto(s)
Conexinas , Neoplasias , Humanos , Terapia Fototérmica , Compuestos Férricos , Especies Reactivas de Oxígeno , Temperatura , Neoplasias/terapia , Peróxido de Hidrógeno , Glucosa Oxidasa , Línea Celular Tumoral , Microambiente Tumoral
8.
Adv Mater ; 33(3): e2006363, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33283339

RESUMEN

Biocatalytic nanomaterials have been verified to modulate the immunosuppressive state of an extensive range of solid tumors and directly induce antitumor immune response, which effectively combats the holdbacks in cancer immunotherapy. Herein, biomimetic cascade enzyme-initiated toxic-radical-generating devices (GHZD NCs) are fabricated by enveloping glucose oxidase (GOx), artificial nanozyme hemin, and sesquiterpene lactone endoperoxide derived dihydroartemisinin (DHA) into zeolitic imidazolate framework (ZIF-8) for enhanced biocatalytic immunotherapy. The GHZD NCs exhibit amplified multienzyme-mimic (glucose oxidase, peroxidase, and glutathione peroxidase) cascade reactions in artificial nanoscale proximity. Concurrently, a glutathione (GSH)-stimulated labile iron-current amplifier boosts C-centered free radicals, which endows the GHZD NCs with tumor-specific and self-circulating generation ability of vicious C-centered free radicals. Irreversible free radicals (·C and ·OH) and sustainable H2 O2 from sequential catalytic processes logically and selectively elevate the oxidative stress in the tumor, which further triggers an efficient immunogenic cell death (ICD) progress. In addition, the in situ nanozyme-based immunotherapy employed for tumor suppression successfully elicits the long-lasting immunological memory effect, which hinders the growth of distant tumors and lung metastasis.


Asunto(s)
Biocatálisis , Biomimética/métodos , Inmunoterapia/métodos , Nanoestructuras , Oxidorreductasas/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda