Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.193
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Nano Lett ; 24(26): 8046-8054, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38912748

RESUMO

Bacteria invasion is the main factor hindering the wound-healing process. However, current antibacterial therapies inevitably face complex challenges, such as the abuse of antibiotics or severe inflammation during treatment. Here, a drug-free bioclay enzyme (Bio-Clayzyme) consisting of Fe2+-tannic acid (TA) network-coated kaolinite nanoclay and glucose oxidase (GOx) was reported to destroy harmful bacteria via bimetal antibacterial therapy. At the wound site, Bio-Clayzyme was found to enhance the generation of toxic hydroxyl radicals for sterilization via cascade catalysis of GOx and Fe2+-mediated peroxidase mimetic activity. Specifically, the acidic characteristics of the infection microenvironment accelerated the release of Al3+ from kaolinite, which further led to bacterial membrane damage and amplified the antibacterial toxicity of Fe2+. Besides, Bio-Clayzyme also performed hemostasis and anti-inflammatory functions inherited from Kaol and TA. By the combination of hemostasis and anti-inflammatory and bimetal synergistic sterilization, Bio-Clayzyme achieves efficient healing of infected wounds, providing a revolutionary approach for infectious wound regeneration.


Assuntos
Antibacterianos , Glucose Oxidase , Cicatrização , Glucose Oxidase/química , Glucose Oxidase/metabolismo , Glucose Oxidase/farmacologia , Cicatrização/efeitos dos fármacos , Antibacterianos/farmacologia , Antibacterianos/química , Animais , Esterilização/métodos , Argila/química , Infecção dos Ferimentos/tratamento farmacológico , Infecção dos Ferimentos/microbiologia , Ferro/química
2.
Anal Chem ; 96(8): 3672-3678, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38361229

RESUMO

Redox potentiometry has emerged as a new platform for in vivo sensing, with improved neuronal compatibility and strong tolerance against sensitivity variation caused by protein fouling. Although enzymes show great possibilities in the fabrication of selective redox potentiometry, the fabrication of an enzyme electrode to output open-circuit voltage (EOC) with fast response remains challenging. Herein, we report a concept of novel enzymatic galvanic redox potentiometry (GRP) with improved time response coupling the merits of the high selectivity of enzyme electrodes with the excellent biocompatibility and reliability of GRP sensors. With a glucose biosensor as an illustration, we use flavin adenine dinucleotide-dependent glucose dehydrogenase as the recognition element and carbon black as the potential relay station to improve the response time. We find that the enzymatic GRP biosensor rapidly responds to glucose with a good linear relationship between EOC and the logarithm of glucose concentration within a range from 100 µM to 2.65 mM. The GRP biosensor shows high selectivity over O2 and coexisting neurochemicals, good reversibility, and sensitivity and can in vivo monitor glucose dynamics in rat brain. We believe that this study will pave a new platform for the in vivo potentiometric biosensing of chemical events with high reliability.


Assuntos
Técnicas Biossensoriais , Glucose Oxidase , Potenciometria , Reprodutibilidade dos Testes , Glucose Oxidase/metabolismo , Eletrodos , Glucose , Oxirredução , Glucose 1-Desidrogenase/metabolismo
3.
Small ; 20(26): e2310283, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38227378

RESUMO

Conventional hydrogel microcapsules often suffer from inadequate mechanical stability, hindering their use. Here, water-cored double-network (DN) hydrogel shells are designed, formed by polyacrylamide and calcium alginate networks using triple-emulsion templates. These DN hydrogel shells offer robust mechanical stability, optical transparency, and a precisely-defined cut-off threshold. The feasibility of this platform is demonstrated through the development of a fluorometric glucose sensor. Glucose oxidase is enclosed within the water core, while a pH-responsive fluorescent dye is incorporated into the DN shells. Glucose diffuses into the core through the DN shells, where the glucose oxidase converts glucose into gluconic acid, leading to pH reduction and a subsequent decrease in fluorescence intensity of DN shells. Additionally, the pH-sensitive colorant dissolved in the medium enables visual pH assessment. Thus, glucose levels can be determined using both fluorometric and colorimetric methods. Notably, the DN shells exhibit exceptional stability, enduring intense mechanical stress and cycles of drying and rehydration without leakage. Moreover, the DN shells act as effective barriers, safeguarding glucose oxidase against proteolysis by large disruptive proteins, like pancreatin. This versatile DN shell platform extends beyond glucose oxidase encapsulation, serving as a foundation for various capsule sensors utilizing enzymes and heterogeneous catalysts.


Assuntos
Glucose Oxidase , Glucose , Hidrogéis , Glucose/análise , Glucose/química , Hidrogéis/química , Glucose Oxidase/química , Glucose Oxidase/metabolismo , Concentração de Íons de Hidrogênio , Técnicas Biossensoriais/métodos , Alginatos/química , Resinas Acrílicas/química
4.
Small ; 20(29): e2310247, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38368267

RESUMO

Chemodynamic therapy (CDT) has emerged as a promising approach for treating infected diabetic wounds, while reliable imaging technology for simultaneous monitoring of ROS and therapeutic processes is still a formidable challenge. Herein, smart covalent organic framework (COF) nanoreactors (COF NRs) are constructed by hyaluronic acid (HA) packaged glucose oxidase (GOx) covalently linked Fe-COF for diabetic wound healing. Upon the breakdown of the HA protective layer, GOx consumes glucose to produce gluconic acid and hydrogen peroxide (H2O2), resulting in decreased local pH and H2O2 supplementation. Density functional theory (DFT) calculations show that Fe-COF has high catalytic activity towards H2O2, leading to in situ generation of hydroxyl radicals (·OH) for sterilization, and the localized downregulation of glucose effectively improved the microenvironment of diabetic wounds. Meanwhile, based on the near-infrared photothermal imaging of oxidized 3,3',5,5'-tetramethylbenzidine (oxTMB), the authors showed that TMB can be applied for the point-of-care testing of ·OH and glucose, and assessing the sterilization progress in vivo. More significantly, the facile photothermal signaling strategy can be extended to monitor various ROS-mediated therapeutic systems, enabling accurate prediction of treatment outcomes.


Assuntos
Espécies Reativas de Oxigênio , Cicatrização , Cicatrização/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Animais , Glucose Oxidase/metabolismo , Glucose Oxidase/química , Peróxido de Hidrogênio/química , Esterilização/métodos , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Camundongos , Estruturas Metalorgânicas/química , Estruturas Metalorgânicas/farmacologia , Glucose
5.
Small ; 20(26): e2310248, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38234145

RESUMO

Interfering with intratumoral metabolic processes is proven to effectively sensitize different antitumor treatments. Here, a tumor-targeting catalytic nanoplatform (CQ@MIL-GOX@PB) loading with autophagy inhibitor (chloroquine, CQ) and glucose oxidase (GOX) is fabricated to interfere with the metabolisms of tumor cells and tumor-associated macrophages (TAMs), then realizing effective antitumor chemodynamic therapy (CDT). Once accumulating in the tumor site with the navigation of external biotin, CQ@MIL-GOX@PB will release Fe ions and CQ in the acid lysosomes of tumor cells, the latter can sensitize Fe ions-involved antitumor CDT by blocking the autophagy-dependent cell repair. Meanwhile, the GOX component will consume glucose, which not only generates many H2O2 for CDT but also once again decelerates the tumor repair process by reducing energy metabolism. What is more, the release of CQ can also drive the NO anabolism of TAMs to further sensitize CDT. This strategy of multiple metabolic regulations is evidenced to significantly improve the antitumor effect of traditional CDT nanoagents and might provide a new sight to overcome the bottlenecks of different antitumor treatments.


Assuntos
Glucose Oxidase , Animais , Glucose Oxidase/metabolismo , Humanos , Linhagem Celular Tumoral , Camundongos , Antineoplásicos/farmacologia , Antineoplásicos/química , Cloroquina/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Autofagia/efeitos dos fármacos , Nanopartículas/química
6.
Small ; 20(26): e2308861, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38372029

RESUMO

The anabolism of tumor cells can not only support their proliferation, but also endow them with a steady influx of exogenous nutrients. Therefore, consuming metabolic substrates or limiting access to energy supply can be an effective strategy to impede tumor growth. Herein, a novel treatment paradigm of starving-like therapy-triple energy-depleting therapy-is illustrated by glucose oxidase (GOx)/dc-IR825/sorafenib liposomes (termed GISLs), and such a triple energy-depleting therapy exhibits a more effective tumor-killing effect than conventional starvation therapy that only cuts off one of the energy supplies. Specifically, GOx can continuously consume glucose and generate toxic H2O2 in the tumor microenvironment (including tumor cells). After endocytosis, dc-IR825 (a near-infrared cyanine dye) can precisely target mitochondria and exert photodynamic and photothermal activities upon laser irradiation to destroy mitochondria. The anti-angiogenesis effect of sorafenib can further block energy and nutrition supply from blood. This work exemplifies a facile and safe method to exhaust the energy in a tumor from three aspects and starve the tumor to death and also highlights the importance of energy depletion in tumor treatment. It is hoped that this work will inspire the development of more advanced platforms that can combine multiple energy depletion therapies to realize more effective tumor treatment.


Assuntos
Glucose Oxidase , Lipossomos , Sorafenibe , Lipossomos/química , Humanos , Glucose Oxidase/metabolismo , Glucose Oxidase/química , Animais , Sorafenibe/farmacologia , Linhagem Celular Tumoral , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Metabolismo Energético , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/química , Indóis
7.
Chemistry ; 30(41): e202401256, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-38719746

RESUMO

Hydrogen-bonded organic frameworks (HOF) represent an emerging category of organic structures with high crystallinity and metal-free, which are not commonly observed in alternative porous organic frameworks. These needle-like porous structure can help in stabilizing enzymes and allow transfer of molecules between enzymes participating in cascade reactions for enhanced substrate channelling. Herein, we systematically synthesized and investigated the stability of HOF at extreme conditions followed by one-pot encapsulation of single and bi-enzyme systems. Firstly, we observed HOF to be stable at pH 1 to 14 and at high temperatures (up to 115 °C). Secondly, the encapsulated glucose oxidase enzyme (GOX) showed 80 % and 90 % of its original activity at 70 °C and pH 11, respectively. Thirdly, transient time close to 0 seconds was observed for HOF encapsulated bi-enzyme cascade reaction system demonstrating a 4.25-fold improvement in catalytic activity when compared to free enzymes with enhanced substrate channelling. Our findings showcase a facile system synthesized under ambient conditions to encapsulate and stabilize enzymes at extreme conditions.


Assuntos
Glucose Oxidase , Ligação de Hidrogênio , Estruturas Metalorgânicas , Glucose Oxidase/química , Glucose Oxidase/metabolismo , Estruturas Metalorgânicas/química , Porosidade , Concentração de Íons de Hidrogênio , Temperatura , Enzimas Imobilizadas/química , Enzimas Imobilizadas/metabolismo , Catálise
8.
Chemistry ; 30(31): e202400195, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38563653

RESUMO

Framework and polymeric nanoreactors (NRs) have distinct advantages in improving chemical reaction efficiency in the tumor microenvironment (TME). Nanoreactor-loaded oxidoreductase enzyme is activated by tumor acidity to produce H2O2 by increasing tumor oxidative stress. High levels of H2O2 induce self-destruction of the vesicles by releasing quinone methide to deplete glutathione and suppress the antioxidant potential of cancer cells. Therefore, the synergistic effect of the enzyme-loaded nanoreactors results in efficient tumor ablation via suppressing cancer-cell metabolism. The main driving force would be to take advantage of the distinct metabolic properties of cancer cells along with the high peroxidase-like activity of metalloenzyme/metalloprotein. A cascade strategy of dual enzymes such as glucose oxidase (GOx) and nitroreductase (NTR) wherein the former acts as an O2-consuming agent such as overexpression of NTR and further amplified NTR-catalyzed release for antitumor therapy. The design of cascade bioreductive hypoxia-responsive drug delivery via GOx regulates NTR upregulation and NTR-responsive nanoparticles. Herein, we discuss tumor hypoxia, reactive oxygen species (ROS) formation, and the effectiveness of these therapies. Nanoclusters in cascaded enzymes along with chemo-radiotherapy with synergistic therapy are illustrated. Finally, we outline the role of the nanoreactor strategy of cascading enzymes along with self-synergistic tumor therapy.


Assuntos
Glucose Oxidase , Neoplasias , Microambiente Tumoral , Humanos , Glucose Oxidase/metabolismo , Glucose Oxidase/química , Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Nitrorredutases/metabolismo , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/química , Espécies Reativas de Oxigênio/metabolismo , Nanopartículas/química , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Estresse Oxidativo/efeitos dos fármacos
9.
Chemphyschem ; 25(10): e202400460, 2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38778583

RESUMO

The front cover artwork is provided by Prof. Ron Naaman's group at the Weizmann Institute of Science. The image shows that direct electron transfer through GOx is governed by electron spins, which result from the chiral-induced spin selectivity (CISS) effect. Read the full text of the Research Article at 10.1002/cphc.202400033.


Assuntos
Glucose Oxidase , Glucose Oxidase/química , Glucose Oxidase/metabolismo , Transporte de Elétrons , Biocatálise , Elétrons
10.
Chemphyschem ; 25(10): e202400033, 2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38411033

RESUMO

The reaction of D-glucose oxidase (GOx) with D- and L-glucose was investigated using confocal fluorescence microscopy and Hall voltage measurements, after the enzyme was adsorbed as a monolayer. By adsorbing the enzyme on a ferromagnetic substrate, we verified that the reaction is spin dependent. This conclusion was supported by monitoring the reaction when the enzyme is adsorbed on a Hall device that does not contain any magnetic elements. The spin dependence is consistent with the chiral-induced spin selectivity (CISS) effect; it can be explained by the improved fidelity of the electron transfer process through the chiral enzyme due to the coupling of the linear momentum of the electrons and their spin. Since the reaction studied often serve as a model system for enzymatic activity, the results may suggest the general importance of the spin-dependent electron transfer in bio-chemical processes.


Assuntos
Glucose Oxidase , Glucose , Glucose Oxidase/química , Glucose Oxidase/metabolismo , Glucose/química , Glucose/metabolismo , Transporte de Elétrons , Biocatálise , Adsorção
11.
Biotechnol Bioeng ; 121(2): 735-748, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38037762

RESUMO

Glucose oxidase (EC 1.1.3.4, GOD) is a widely used industrial enzyme. To construct a GOD-hyperproducing Pichia pastoris strain, combinatorial strategies have been applied to improve GOD activity, synthesis, and secretion. First, wild-type GOD was subjected to saturation mutagenesis to obtain an improved variant, MGOD1 (V20W/T30S), with 1.7-fold higher kcat /KM . Subsequently, efficient signal peptides were screened, and the copy number of MGOD1 was optimized to generate a high-producing strain, 8GM1, containing eight copies of AOX1 promoter-GAS1 signal peptide-MGOD1 expression cassette. Finally, the vesicle trafficking of 8GM1 was engineered to obtain the hyperproducing strain G1EeSe co-expressing the trafficking components EES and SEC. 22, and the EES gene (PAS_chr3_0685) was found to facilitate both protein secretion and production for the first time. Using these strategies, GOD secretion was enhanced 65.2-fold. In the 5-L bioreactor, conventional fed-batch fermentation without any process optimization resulted in up to 7223.0 U/mL extracellular GOD activity (3.3-fold higher than the highest level reported to date), with almost only GOD in the fermentation supernatant at a protein concentration of 30.7 g/L. Therefore, a GOD hyperproducing strain for industrial applications was developed, and this successful case can provide a valuable reference for the construction of high-producing strains for other industrial enzymes.


Assuntos
Glucose Oxidase , Pichia , Saccharomycetales , Glucose Oxidase/genética , Glucose Oxidase/metabolismo , Pichia/metabolismo , Reatores Biológicos , Fermentação , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
12.
Langmuir ; 40(1): 170-178, 2024 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-38113389

RESUMO

Enzyme-based electrochemical biosensors play an important role in point-of-care diagnostics for personalized medicine. For such devices, lipid cubic phases (LCP) represent an attractive method to immobilize enzymes onto conductive surfaces with no need for chemical linking. However, research has been held back by the lack of effective strategies to stably co-immobilize enzymes with a redox shuttle that enhances the electrical connection between the enzyme redox center and the electrode. In this study, we show that a monoolein (MO) LCP system doped with an amphiphilic redox mediator (ferrocenylmethyl)dodecyldimethylammonium bromide (Fc12) can be used for enzyme immobilization to generate an effective biosensing platform. Small-angle X-ray scattering (SAXS) showed that MO LCP can incorporate Fc12 while maintaining the Pn3m symmetry morphology. Cyclic voltammograms of Fc12/MO showed quasi-reversible behavior, which implied that Fc12 was able to freely diffuse in the lipid membrane of LCP with a diffusion coefficient of 1.9 ± 0.2 × 10-8 cm2 s-1 at room temperature. Glucose oxidase (GOx) was then chosen as a model enzyme and incorporated into 0.2%Fc12/MO to evaluate the activity of the platform. GOx hosted in 0.2%Fc12/MO followed Michaelis-Menten kinetics toward glucose with a KM and Imax of 8.9 ± 0.5 mM and 1.4 ± 0.2 µA, respectively, and a linearity range of 2-17 mM glucose. Our results therefore demonstrate that GOx immobilized onto 0.2% Fc12/MO is a suitable platform for the electrochemical detection of glucose.


Assuntos
Técnicas Biossensoriais , Glucose , Espalhamento a Baixo Ângulo , Difração de Raios X , Oxirredução , Glucose Oxidase/metabolismo , Enzimas Imobilizadas/metabolismo , Técnicas Biossensoriais/métodos , Eletrodos
13.
Biomacromolecules ; 25(7): 4492-4509, 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38910355

RESUMO

A major shortcoming associated with the application of enzymes in drug synergism originates from the lack of site-specific, multifunctional nanomedicine. This study introduces catalytic nanocompartments (CNCs) made of a mixture of PDMS-b-PMOXA diblock copolymers, decorated with glycooligomer tethers comprising eight mannose-containing repeating units and coencapsulating two enzymes, providing multifunctionality by their in situ parallel reactions. Beta-glucuronidase (GUS) serves for local reactivation of the drug hymecromone, while glucose oxidase (GOx) induces cell starvation through glucose depletion and generation of the cytotoxic H2O2. The insertion of the pore-forming peptide, melittin, facilitates diffusion of substrates and products through the membranes. Increased cell-specific internalization of the CNCs results in a substantial decrease in HepG2 cell viability after 24 h, attributed to simultaneous production of hymecromone and H2O2. Such parallel enzymatic reactions taking place in nanocompartments pave the way to achieve efficient combinatorial cancer therapy by enabling localized drug production along with reactive oxygen species (ROS) elevation.


Assuntos
Glucose Oxidase , Peróxido de Hidrogênio , Humanos , Glucose Oxidase/química , Glucose Oxidase/metabolismo , Células Hep G2 , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/metabolismo , Glucuronidase/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Catálise , Espécies Reativas de Oxigênio/metabolismo , Oligossacarídeos/química , Oligossacarídeos/metabolismo
14.
Analyst ; 149(9): 2756-2761, 2024 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-38563766

RESUMO

New dynamic, wireless and cost-effective analytical devices are developing rapidly in biochemical analysis. Here, we report on a remotely-controlled rotating electrochemiluminescence (ECL) sensing system for enzymatic detection of a model analyte, glucose, on both polarized sides of an iron wire acting as a bipolar electrode. The iron wire is controlled by double contactless mode, involving remote electric field polarization, and magnetic field-induced rotational motion. The former triggers the interfacial polarization of both extremities of the wire by bipolar electrochemistry, which generates ECL emission of the luminol derivative (L-012) with the enzymatically produced hydrogen peroxide in presence of glucose, at both anodic and cathodic poles, simultaneously. The latter generates a convective flow, leading to an increase in mass transfer and amplifying the corresponding ECL signals. Quantitative glucose detection in human serum samples is achieved. The ECL signals were found to be a linear function of the glucose concentration within the range of 10-1000 µM and with a limit of detection of 10 µM. The dynamic bipolar ECL system simultaneously generates light emissions at both anodic and cathodic poles for glucose detection, which can be further applied to biosensing and imaging in autonomous devices.


Assuntos
Técnicas Eletroquímicas , Medições Luminescentes , Medições Luminescentes/métodos , Humanos , Técnicas Eletroquímicas/métodos , Técnicas Eletroquímicas/instrumentação , Eletrodos , Técnicas Biossensoriais/métodos , Técnicas Biossensoriais/instrumentação , Limite de Detecção , Glicemia/análise , Tecnologia sem Fio , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/análise , Glucose Oxidase/química , Glucose Oxidase/metabolismo , Luminol/química
15.
Analyst ; 149(15): 3920-3927, 2024 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-38912896

RESUMO

Anti-cancer therapy is crucial in cancer prevention and anti-cancer, and thus, highly sensitive methods for detecting cancer biomarkers are essential for cancer early diagnosis. Herein, an electrochemical aptamer biosensor based on the CRISPR-Cas12a system was constructed for the detection of cancer tumor biomarker MUC1 mucin. The sensitivity was significantly prompted by enzyme-catalyzed signal amplification, and the selectivity was improved by the dual recognition of the aptamer to MUC1 and crRNA-Cas12a system to the aptamer. Glucose oxidase (GOD) was loaded on the surface of magnetic Fe3O4@Au (MGNP) via probe single-stranded DNA (pDNA) with the terminal modification of mercapto (-SH) to form GOD-pDNA/MGNP. The corresponding aptamer of MUC1 (MUC1 Apt) binds to its complementary ssDNA (cDNA) to form the activator Apt/cDNA, which is specifically recognized by crRNA-Cas12a and excites the trans-cleavage function of Cas12a, thus in turn trans-cleaves pDNA and detaches GOD from the magnetic particles. The magnetic beads were separated and transferred into a glucose solution, and the oxidation current of H2O2 produced by the catalytic reaction of GOD was measured on a Pt-modified magnetically-controlled glassy carbon electrode, resulting in an indirect determination of MUC1. The current change was linear with the logarithm of MUC1 concentration in the range from 1.0 × 10-17 g mL-1 to 1.0 × 10-10 g mL-1. The detection limit was as low as 7.01 × 10-18 g mL-1. The method was applied for the detection of MUC1 in medical samples.


Assuntos
Aptâmeros de Nucleotídeos , Biomarcadores Tumorais , Técnicas Biossensoriais , Sistemas CRISPR-Cas , Técnicas Eletroquímicas , Glucose Oxidase , Limite de Detecção , Mucina-1 , Mucina-1/metabolismo , Humanos , Técnicas Eletroquímicas/métodos , Técnicas Biossensoriais/métodos , Aptâmeros de Nucleotídeos/química , Glucose Oxidase/química , Glucose Oxidase/metabolismo , Biomarcadores Tumorais/genética , Peróxido de Hidrogênio/química , DNA de Cadeia Simples/química , Ouro/química , Nanopartículas de Magnetita/química , Enzimas Imobilizadas/química , Proteínas Associadas a CRISPR/química , Proteínas Associadas a CRISPR/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Endodesoxirribonucleases
16.
Environ Res ; 247: 118257, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38262511

RESUMO

This study introduces the UV/glucose-oxidase@Kaolin (GOD@Kaolin) coupled organic green rust (OGR) system (UV/OGR/GOD@Kaolin) to investigate the promotion of glucose oxidase activity by UV light and its synergistic degradation mechanism for photosensitive pollutants, specifically targeting the efficient degradation of 4-chlorophenol (4-CP). The enzyme system demonstrates its ability to overcome drawbacks associated with traditional Fenton systems, including a narrow pH range and high localized concentration of H2O2, by gradually releasing hydrogen peroxide in situ within a neutral environment. In the presence of UV radiation under specific conditions, enhanced enzyme activity is observed, resulting in increased efficiency in pollutant removal. The gradual release of hydrogen peroxide plays a crucial role in preventing unwanted reactions among active substances. These unique features facilitate the generation of highly reactive species, such as Fe(IV)O, •OH, and •O2-, tailored to efficiently target the organic components of interest. Additionally, the system establishes a positive iron cycle, ensuring a sustained reactive capability throughout the degradation process. The results highlight the UV/OGR/GOD@Kaolin system as an effective and environmentally friendly approach for the degradation of 4-CP, and the resilience of the enzyme extends the system's applicability to a broader range of scenarios.


Assuntos
Poluentes Ambientais , Poluentes Químicos da Água , Raios Ultravioleta , Peróxido de Hidrogênio/química , Glucose Oxidase/metabolismo , Caulim , Glucose , Oxirredução , Poluentes Químicos da Água/química
17.
J Nanobiotechnology ; 22(1): 334, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38877463

RESUMO

Due to the limitations of single-model tumor therapeutic strategies, multimodal combination therapy have become a more favorable option to enhance efficacy by compensating for its deficiencies. However, in nanomaterial-based multimodal therapeutics for tumors, exploiting synergistic interactions and cascade relationships of materials to achieve more effective treatments is still a great challenge. Based on this, we constructed a nanoplatform with a "triple-linkage" effect by cleverly integrating polydopamine (PDA), silver nanoparticles (AgNPs), and glucose oxidase (GOx) to realize enhanced photothermal therapy (PTT) and activatable metal ion therapy (MIT) for hepatocellular carcinoma (HCC) treatment. First, the non-radiative conversion of PDA under light conditions was enhanced by AgNPs, which directly enhanced the photothermal conversion efficiency of PDA. In addition, GOx reduced the synthesis of cellular heat shock proteins by interfering with cellular energy metabolism, thereby enhancing cellular sensitivity to PTT. On the other hand, H2O2, a by-product of GOx-catalyzed glucose, could be used as an activation source to activate non-toxic AgNPs to release cytotoxic Ag+, achieving activatable Ag+-mediated MIT. In conclusion, this nanosystem achieved efficient PTT and MIT for HCC by exploiting the cascade effect among PDA, AgNPs, and GOx, providing a novel idea for the design of multimodal tumor therapeutic systems with cascade regulation.


Assuntos
Carcinoma Hepatocelular , Glucose Oxidase , Indóis , Neoplasias Hepáticas , Nanopartículas Metálicas , Terapia Fototérmica , Polímeros , Prata , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Prata/química , Prata/farmacologia , Prata/uso terapêutico , Nanopartículas Metálicas/química , Nanopartículas Metálicas/uso terapêutico , Humanos , Glucose Oxidase/metabolismo , Indóis/química , Indóis/farmacologia , Indóis/uso terapêutico , Animais , Terapia Fototérmica/métodos , Camundongos , Polímeros/química , Linhagem Celular Tumoral , Fototerapia/métodos , Camundongos Endogâmicos BALB C , Peróxido de Hidrogênio , Sobrevivência Celular/efeitos dos fármacos , Camundongos Nus
18.
J Nanobiotechnology ; 22(1): 234, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38724978

RESUMO

Radiotherapy-induced immune activation holds great promise for optimizing cancer treatment efficacy. Here, we describe a clinically used radiosensitizer hafnium oxide (HfO2) that was core coated with a MnO2 shell followed by a glucose oxidase (GOx) doping nanoplatform (HfO2@MnO2@GOx, HMG) to trigger ferroptosis adjuvant effects by glutathione depletion and reactive oxygen species production. This ferroptosis cascade potentiation further sensitized radiotherapy by enhancing DNA damage in 4T1 breast cancer tumor cells. The combination of HMG nanoparticles and radiotherapy effectively activated the damaged DNA and Mn2+-mediated cGAS-STING immune pathway in vitro and in vivo. This process had significant inhibitory effects on cancer progression and initiating an anticancer systemic immune response to prevent distant tumor recurrence and achieve long-lasting tumor suppression of both primary and distant tumors. Furthermore, the as-prepared HMG nanoparticles "turned on" spectral computed tomography (CT)/magnetic resonance dual-modality imaging signals, and demonstrated favorable contrast enhancement capabilities activated by under the GSH tumor microenvironment. This result highlighted the potential of nanoparticles as a theranostic nanoplatform for achieving molecular imaging guided tumor radiotherapy sensitization induced by synergistic immunotherapy.


Assuntos
Ferroptose , Imunoterapia , Compostos de Manganês , Proteínas de Membrana , Camundongos Endogâmicos BALB C , Nanopartículas , Nucleotidiltransferases , Óxidos , Radiossensibilizantes , Animais , Camundongos , Imunoterapia/métodos , Óxidos/química , Óxidos/farmacologia , Feminino , Nucleotidiltransferases/metabolismo , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Linhagem Celular Tumoral , Nanopartículas/química , Radiossensibilizantes/farmacologia , Radiossensibilizantes/química , Proteínas de Membrana/metabolismo , Ferroptose/efeitos dos fármacos , Glucose Oxidase/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Humanos , Dano ao DNA , Microambiente Tumoral/efeitos dos fármacos
19.
Mikrochim Acta ; 191(8): 447, 2024 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-38963544

RESUMO

An intelligent nanodrug delivery system (Cu/ZIF-8@GOx-DOX@HA, hereafter CZGDH) consisting of Cu-doped zeolite imidazolate framework-8 (Cu/ZIF-8, hereafter CZ), glucose oxidase (GOx), doxorubicin (DOX), and hyaluronic acid (HA) was established for targeted drug delivery and synergistic therapy of tumors. The CZGDH specifically entered tumor cells through the targeting effect of HA and exhibited acidity-triggered biodegradation for subsequent release of GOx, DOX, and Cu2+ in the tumor microenvironment (TME). The GOx oxidized the glucose (Glu) in tumor cells to produce H2O2 and gluconic acid for starvation therapy (ST). The DOX entered the intratumoral cell nucleus for chemotherapy (CT). The released Cu2+ consumed the overexpressed glutathione (GSH) in tumor cells to produce Cu+. The generated Cu+ and H2O2 triggered the Fenton-like reaction to generate toxic hydroxyl radicals (·OH), which disrupted the redox balance of tumor cells and effectively killed tumor cells for chemodynamic therapy (CDT). Therefore, synergistic multimodal tumor treatment via TME-activated cascade reaction was achieved. The nanodrug delivery system has a high drug loading rate (48.3 wt%), and the three-mode synergistic therapy has a strong killing effect on tumor cells (67.45%).


Assuntos
Cobre , Doxorrubicina , Glucose Oxidase , Ácido Hialurônico , Estruturas Metalorgânicas , Microambiente Tumoral , Zeolitas , Cobre/química , Doxorrubicina/farmacologia , Doxorrubicina/química , Microambiente Tumoral/efeitos dos fármacos , Glucose Oxidase/química , Glucose Oxidase/metabolismo , Humanos , Zeolitas/química , Animais , Estruturas Metalorgânicas/química , Ácido Hialurônico/química , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/metabolismo , Linhagem Celular Tumoral , Camundongos , Antibióticos Antineoplásicos/farmacologia , Antibióticos Antineoplásicos/química , Neoplasias/tratamento farmacológico , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Antineoplásicos/farmacologia , Antineoplásicos/química , Imidazóis
20.
Bioprocess Biosyst Eng ; 47(6): 919-929, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38644439

RESUMO

The growing need in the current market for innovative solutions to obtain lactose-free (L-F) milk is caused by the annual increase in the prevalence of lactose intolerance inside as well as the newborn, children, and adults. Various configurations of enzymes can yield two distinct L-F products: sweet (ß-galactosidase) and unsweet (ß-galactosidase and glucose oxidase) L-F milk. In addition, the reduction of sweetness through glucose decomposition should be performed in a one-pot mode with catalase to eliminate product inhibition caused by H2O2. Both L-F products enjoy popularity among a rapidly expanding group of consumers. Although enzyme immobilization techniques are well known in industrial processes, new carriers and economic strategies are still being searched. Polymeric carriers, due to the variety of functional groups and non-toxicity, are attractive propositions for individual and co-immobilization of food enzymes. In the presented work, two strategies (with free and immobilized enzymes; ß-galactosidase NOLA, glucose oxidase from Aspergillus niger, and catalase from Serratia sp.) for obtaining sweet and unsweet L-F milk under low-temperature conditions were proposed. For free enzymes, achieving the critical assumption, lactose hydrolysis and glucose decomposition occurred after 1 and 4.3 h, respectively. The tested catalytic membranes were created on regenerated cellulose and polyamide. In both cases, the time required for lactose and glucose bioconversion was extended compared to free enzymes. However, these preparations could be reused for up to five (ß-galactosidase) and ten cycles (glucose oxidase with catalase).


Assuntos
Enzimas Imobilizadas , Glucose Oxidase , Lactose , Leite , beta-Galactosidase , beta-Galactosidase/metabolismo , beta-Galactosidase/química , Leite/química , Lactose/metabolismo , Lactose/química , Glucose Oxidase/química , Glucose Oxidase/metabolismo , Enzimas Imobilizadas/química , Enzimas Imobilizadas/metabolismo , Animais , Aspergillus niger/enzimologia , Glucose/metabolismo , Glucose/química , Catalase/metabolismo , Catalase/química , Membranas Artificiais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA