Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 402
Filtrar
1.
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
2.
J Colloid Interface Sci ; 666: 244-258, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38598997

RESUMO

Starvation therapy has shown promise as a cancer treatment, but its efficacy is often limited when used alone. In this work, a multifunctional nanoscale cascade enzyme system, named CaCO3@MnO2-NH2@GOx@PVP (CMGP), was fabricated for enhanced starvation/chemodynamic combination cancer therapy. CMGP is composed of CaCO3 nanoparticles wrapped in a MnO2 shell, with glucose oxidase (GOx) adsorbed and modified with polyvinylpyrrolidone (PVP). MnO2 decomposes H2O2 in cancer cells into O2, which enhances the efficiency of GOx-mediated starvation therapy. CaCO3 can be decomposed in the acidic cancer cell environment, causing Ca2+ overload in cancer cells and inhibiting mitochondrial metabolism. This synergizes with GOx to achieve more efficient starvation therapy. Additionally, the H2O2 and gluconic acid produced during glucose consumption by GOx are utilized by MnO2 with catalase-like activity to enhance O2 production and Mn2+ release. This process accelerates glucose consumption, reactive oxygen species (ROS) generation, and CaCO3 decomposition, promoting the Ca2+ release. CMGP can alleviate tumor hypoxia by cycling the enzymatic cascade reaction, which increases enzyme activity and combines with Ca2+ overload to achieve enhanced combined starvation/chemodynamic therapy. In vitro and in vivo studies demonstrate that CMGP has effective anticancer abilities and good biosafety. It represents a new strategy with great potential for combined cancer therapy.


Assuntos
Carbonato de Cálcio , Glucose Oxidase , Compostos de Manganês , Óxidos , Glucose Oxidase/metabolismo , Glucose Oxidase/química , Glucose Oxidase/farmacologia , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Óxidos/química , Óxidos/farmacologia , Humanos , Animais , Carbonato de Cálcio/química , Carbonato de Cálcio/farmacologia , Carbonato de Cálcio/metabolismo , Camundongos , Antineoplásicos/farmacologia , Antineoplásicos/química , Nanopartículas/química , Povidona/química , Povidona/farmacologia , Hipóxia Tumoral/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Tamanho da Partícula , Linhagem Celular Tumoral , Peróxido de Hidrogênio/metabolismo , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Propriedades de Superfície , Camundongos Endogâmicos BALB C
3.
Nat Commun ; 15(1): 3440, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38653977

RESUMO

Oxidative stress from excess H2O2 activates transcription factors that restore redox balance and repair oxidative damage. Although many transcription factors are activated by H2O2, it is unclear whether they are activated at the same H2O2 concentration, or time. Dose-dependent activation is likely as oxidative stress is not a singular state and exhibits dose-dependent outcomes including cell-cycle arrest and cell death. Here, we show that transcription factor activation is both dose-dependent and coordinated over time. Low levels of H2O2 activate p53, NRF2 and JUN. Yet under high H2O2, these transcription factors are repressed, and FOXO1, NF-κB, and NFAT1 are activated. Time-lapse imaging revealed that the order in which these two groups of transcription factors are activated depends on whether H2O2 is administered acutely by bolus addition, or continuously through the glucose oxidase enzyme. Finally, we provide evidence that 2-Cys peroxiredoxins control which group of transcription factors are activated.


Assuntos
Peróxido de Hidrogênio , Estresse Oxidativo , Fatores de Transcrição , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Humanos , Peroxirredoxinas/metabolismo , Peroxirredoxinas/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/genética , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , NF-kappa B/metabolismo , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Fatores de Transcrição NFATC/metabolismo , Glucose Oxidase/metabolismo , Animais
4.
Spectrochim Acta A Mol Biomol Spectrosc ; 315: 124269, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38608561

RESUMO

A colorimetric immunoassay was built for determination of carcinoembryonic antigen (CEA) based on papain-based colorimetric catalytic sensing system through the use of glucose oxidase (GOx). In the presence of GOx, glucose was catalytically oxidized to produce H2O2. Through the assistance of papain (as a peroxide mimetic enzyme), the signal came from the oxidative color development of 3,3',5,5'-tetramethylbenzidine (TMB, from colorless to blue) catalyzed by the generated H2O2. Herein, a sandwich-type immunoassay was built based on GOx as labels. As the concentration of CEA increased, more GOx-labeled antibodies specifically associate with target, which leaded to more H2O2 generation. Immediately following this, more TMB were oxidized with the addition of papain. Accordingly, the absorbance increased further. As a result, the concentration of CEA is positively correlated with the change in absorbance of the solution. Under optimal conditions, the CEA concentration was linear in the range of 0.05-20.0 ng/mL, and the limit of detection (LOD) reached 37 pg/mL. The papain-based colorimetric immunoassay also exhibited satisfactory repeatability, stability, and selectivity.


Assuntos
Antígeno Carcinoembrionário , Colorimetria , Limite de Detecção , Papaína , Antígeno Carcinoembrionário/análise , Colorimetria/métodos , Papaína/metabolismo , Imunoensaio/métodos , Humanos , Glucose Oxidase/química , Glucose Oxidase/metabolismo , Peróxido de Hidrogênio/química , Catálise , Benzidinas/química , Técnicas Biossensoriais/métodos , Reprodutibilidade dos Testes
5.
Adv Sci (Weinh) ; 11(18): e2308251, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38447152

RESUMO

Nanomedicine has reshaped the landscape of cancer treatment. However, its efficacy is still hampered by innate tumor defense systems that rely on adenosine triphosphate (ATP) for fuel, including damage repair, apoptosis resistance, and immune evasion. Inspired by the naturally enzymatic reaction of glucose oxidase (GOx) with glucose, here a novel "two birds with one stone" technique for amplifying enzyme-mediated tumor apoptosis and enzyme-promoted metabolic clearance is proposed and achieved using GOx-functionalized rhenium nanoclusters-doped polypyrrole (Re@ReP-G). Re@ReP-G reduces ATP production while increasing H2O2 concentrations in the tumor microenvironment through GOx-induced enzymatic oxidation, which in turn results in the downregulation of defense (HSP70 and HSP90) and anti-apoptotic Bcl-2 proteins, the upregulation of pro-apoptotic Bax, and the release of cytochrome c. These processes are further facilitated by laser-induced hyperthermia effect, ultimately leading to severe tumor apoptosis. As an enzymatic byproduct, H2O2 catalyzes the conversion of rhenium nanoclusters in Re@ReP-G nanostructures into rhenate from the outside in, which accelerates their metabolic clearance in vivo. This Re@ReP-G-based "two birds with one stone" therapeutic strategy provides an effective tool for amplifying tumor apoptosis and safe metabolic mechanisms.


Assuntos
Apoptose , Animais , Camundongos , Glucose Oxidase/metabolismo , Neoplasias/metabolismo , Humanos , Modelos Animais de Doenças , Linhagem Celular Tumoral , Nanomedicina/métodos , Microambiente Tumoral , Peróxido de Hidrogênio/metabolismo , Polímeros/química , Polímeros/metabolismo
6.
ACS Appl Bio Mater ; 7(3): 1862-1877, 2024 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-38450575

RESUMO

Elevated levels of reactive oxygen species (ROS) have demonstrated efficacy in eliminating tumor cells by modifying the tumor microenvironment and inducing the polarization of tumor-associated macrophages (TAMs). Nevertheless, the transient nature and limited diffusion distance inherent in ROS present significant challenges in cancer treatment. In response to these limitations, we have developed a nanoparticle (MnClPc-HSA@GOx) that not only inhibits tumor energy metabolism but also facilitates the transition of TAMs from the M2 type (anti-inflammatory type) to the M1 type (proinflammatory type). MnClPc-HSA@GOx comprises a manganese phthalocyanine complex (MnClPc) enveloped in human serum albumin (HSA), with glucose oxidase (GOx) loaded onto MnClPc@HSA nanoparticles. GOx was employed to catalyze the decomposition of glucose to produce H2O2 and gluconic acid. Additionally, in the presence of MnClPc, it catalyzes the conversion of H2O2 into •O2- and 1O2. Results indicate that the nanoparticle effectively impedes the glucose supply to tumor cells and suppresses their energy metabolism. Simultaneously, the ROS-mediated polarization of TAMs induces a shift from M2 to M1 macrophages, resulting in a potent inhibitory effect on tumors. This dual-action strategy holds promising clinical inhibition applications in the treatment of cancer.


Assuntos
Isoindóis , Nanopartículas , Neoplasias , Humanos , Manganês/farmacologia , Glucose Oxidase/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Peróxido de Hidrogênio/metabolismo , Neoplasias/metabolismo , Macrófagos , Oxigênio/metabolismo , Metabolismo Energético , Glucose , Microambiente Tumoral
7.
Chem Asian J ; 19(1): e202300879, 2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-37930193

RESUMO

We propose a glucose-powered Janus nanomotor where two faces are functionalized with glucose oxidase (GOx) and polydopamine-Fe3+ chelates (PDF), respectively. In the glucose fuel solution, the GOx on the one side of these Janus nanomotors catalytically decomposes glucose fuels into gluconic acid and hydrogen peroxide (H2 O2 ) to drive them at a speed of 2.67 µm/s. The underlying propulsion mechanism is the glucose-based self-diffusiophoresis owing to the generated local glucose concentration gradient by the enzymatic reaction. Based on the enhanced diffusion motion, such nanomotors with catalytic activity increase the uptake towards cells and subsequently exhibit excellent capabilities for Fe3+ ions delivery and H2 O2 generation for enhancing ferroptosis efficiency for inducing cancer cell death. In particular, the Fe3+ ions are released from nanomotors in a slightly acidic environment, and subsequently generate toxic hydroxyl radicals via Fenton reactions, which accumulation reactive oxygen species (ROS) level (~300 %) and further lipid peroxidation (LPO) strengthened ferroptosis therapy for cancer treatment. The as-developed glucose-powered Janus nanomotor with efficient diffusion and Fe ions delivery capabilities show great promise as a potential in biomedical applications.


Assuntos
Ferroptose , Neoplasias , Humanos , Glucose , Sistemas de Liberação de Medicamentos , Glucose Oxidase/metabolismo , Concentração de Íons de Hidrogênio , Íons , Peróxido de Hidrogênio/metabolismo , Linhagem Celular Tumoral
8.
Angew Chem Int Ed Engl ; 62(47): e202308827, 2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-37802975

RESUMO

Enzymatic catalysis with high efficiency allows them a great prospect in metabolite monitoring in living cells. However, complex tumor microenvironments, such as acidity, H2 O2 , and hypoxia, are bound to disturb catalytic reactions for misleading results. Here, we report a spatially compartmentalized artificial organelle to correct intratumoral glucose analysis, where the zeolitic imidazolate framework-8 immobilized glucose oxidase-horseradish peroxidase cascade core and catalase-directed shell act as signal transduction and guarding rooms respectively. The acid-digested core and stable shell provide appropriate spaces to boost biocatalytic efficiency with good tolerability. Notably, the endogenous H2 O2 is in situ decomposed to O2 by catalase, which not only overcomes the interference in signal output but also alleviates the hypoxic states to maximize glucose oxidation. The marked protective effect and biocompatibility render artificial organelles to correct the signal transduction for dynamic monitoring glucose in vitro and in vivo, achieving our goal of accurate intratumoral metabolite analysis.


Assuntos
Células Artificiais , Estruturas Metalorgânicas , Estruturas Metalorgânicas/metabolismo , Glucose/análise , Catalase/metabolismo , Oxirredução , Glucose Oxidase/metabolismo
9.
J Mater Chem B ; 11(41): 9798-9839, 2023 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-37842806

RESUMO

Nanomaterial-based cancer therapy faces significant limitations due to the complex nature of the tumor microenvironment (TME). Starvation therapy is an emerging therapeutic approach that targets tumor cell metabolism using glucose oxidase (GOx). Importantly, it can provide a material or environmental foundation for other diverse therapeutic methods by manipulating the properties of the TME, such as acidity, hydrogen peroxide (H2O2) levels, and hypoxia degree. In recent years, this cascade strategy has been extensively applied in nanoplatforms for ongoing synergetic therapy and still holds undeniable potential. However, only a few review articles comprehensively elucidate the rational designs of nanoplatforms for synergetic therapeutic regimens revolving around the conception of the cascade strategy. Therefore, this review focuses on innovative cascade strategies for GOx-based synergetic therapy from representative paradigms to state-of-the-art reports to provide an instructive, comprehensive, and insightful reference for readers. Thereafter, we discuss the remaining challenges and offer a critical perspective on the further advancement of GOx-facilitated cancer treatment toward clinical translation.


Assuntos
Nanopartículas , Nanoestruturas , Neoplasias , Humanos , Glucose Oxidase/uso terapêutico , Glucose Oxidase/metabolismo , Peróxido de Hidrogênio/uso terapêutico , Neoplasias/terapia , Nanoestruturas/uso terapêutico , Microambiente Tumoral
10.
J Colloid Interface Sci ; 651: 76-92, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37540932

RESUMO

One of the major challenges in effective cancer therapy arises because of the hypoxic microenvironment in the tumor. This compromises the efficacy of both chemo- and radiotherapy, and thus hinders patient outcomes. To solve this problem, we constructed polydopamine (PDA)-cloaked Fe-based metal organic frameworks (MOFs) loaded with d-arginine (d-Arg), glucose oxidase (GOX), and the chemotherapeutic drug tirapazamine (TPZ). These offer simultaneous multifaceted therapy combining chemodynamic therapy (CDT)/radiotherapy (RT)/starvation therapy (ST)/gas therapy (GT) and chemotherapy. The particles further can act as contrast agents in magnetic resonance imaging. GOX catalyses the conversion of endogenous glucose and O2 to hydrogen peroxide and gluconic acid, blocking the cells' energy supply and providing ST. With the resultant acidification of the local environment, the breakdown of the MOF releases TPZ (for chemotherapy) and Fe3+, which reacts with H2O2 to produce reactive oxygen species and thus stimulates the conversion of d-Arg to NO for GT and RT sensitization. The PDA coating not only seals the pores and chelates Fe3+ to enhance the T1-weighted magnetic resonance imaging (MRI) properties, but also is used to graft folate bovine serum albumin (FA-BSA) and thereby target the tumor site. The combined administration of low doses of X-ray irradiation and nanoparticles reduces the side effects on healthy tissue and can prevent lung metastases in mice. This work highlights the synergistic treatment of osteosarcoma via ST/GT/CDT/RT/MRI/ chemotherapy using a PDA-cloaked MOF system.


Assuntos
Neoplasias Ósseas , Estruturas Metalorgânicas , Nanopartículas , Neoplasias , Osteossarcoma , Camundongos , Animais , Peróxido de Hidrogênio/metabolismo , Neoplasias/tratamento farmacológico , Osteossarcoma/tratamento farmacológico , Linhagem Celular Tumoral , Glucose Oxidase/metabolismo , Microambiente Tumoral
11.
Adv Healthc Mater ; 12(26): e2300968, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37543843

RESUMO

The recurrence and metastasis of colorectal cancer (CRC) have been considered as a severe challenge in clinical treatment. Recent studies have demonstrated that matrix metalloproteinases (MMPs) and lactate can promote local tumor angiogenesis, recurrence, and metastasis. The expression of MMPs is highly dependent on energy metabolism, and lactate is considered an alternative energy source for tumor proliferation and metastasis. Therefore, using a rational approach, a photothermal-starvation therapy nanomodulator that can reduce energy metabolism to suppress CRC recurrence and metastasis is designed. To design a suitable nanomodulator, glucose oxidase (GOX), indocyanine green (IR820), and α-cyano-4-hydroxycinnamic acid (CHC) into nanoparticles by a coassembly method are combined. The photothermal properties of IR820 provide the appropriate temperature and oxygen supply for the enzymatic reaction of GOX to promote intracellular glucose consumption. CHC inhibits the expression of monocarboxylate transporter 1 (MCT1), the transporter of lactic acid into cells, and also reduces oxygen consumption and promotes the GOX reaction. Additionally, altering adenosine triphosphate synthesis to block heat shock proteins expression can be an effective means to prevent IR820-mediated photothermal therapy resistance. Thus, this dual photothermal-starvation therapy nanomodulator efficiently suppresses the recurrence and metastasis of CRC by depleting intracellular nutrients.


Assuntos
Neoplasias Colorretais , Nanopartículas , Neoplasias , Humanos , Fototerapia/métodos , Terapia Fototérmica , Neoplasias/patologia , Metabolismo Energético , Lactatos , Metaloproteinases da Matriz/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Linhagem Celular Tumoral , Glucose Oxidase/metabolismo
12.
Small ; 19(42): e2303253, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37330663

RESUMO

Tumor-dependent glucose and glutamine metabolisms are essential for maintaining survival, while the accordingly metabolic suppressive therapy is limited by the compensatory metabolism and inefficient delivery efficiency. Herein, a functional metal-organic framework (MOF)-based nanosystem composed of the weakly acidic tumor microenvironment-activated detachable shell and reactive oxygen species (ROS)-responsive disassembled MOF nanoreactor core is designed to co-load glycolysis and glutamine metabolism inhibitors glucose oxidase (GOD) and bis-2-(5-phenylacetmido-1,2,4-thiadiazol-2-yl) ethyl sulfide (BPTES) for tumor dual-starvation therapy. The nanosystem excitingly improves tumor penetration and cellular uptake efficiency via integrating the pH-responsive size reduction and charge reversal and ROS-sensitive MOF disintegration and drug release strategy. Furthermore, the degradation of MOF and cargoes release can be self-amplified via additional self-generation H2 O2 mediated by GOD. Last, the released GOD and BPTES collaboratively cut off the energy supply of tumors and induce significant mitochondrial damage and cell cycle arrest via simultaneous restriction of glycolysis and compensatory glutamine metabolism pathways, consequently realizing the remarkable triple negative breast cancer killing effect in vivo with good biosafety via the dual starvation therapy.


Assuntos
Estruturas Metalorgânicas , Neoplasias , Humanos , Estruturas Metalorgânicas/farmacologia , Glutamina/metabolismo , Glutamina/uso terapêutico , Espécies Reativas de Oxigênio , Glucose , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Nanotecnologia , Glucose Oxidase/metabolismo , Linhagem Celular Tumoral , Microambiente Tumoral
13.
Small ; 19(35): e2301402, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37162448

RESUMO

Cascade hydroxyl radical generating hydrogel reactor structures including a chemotherapeutic agent are invented for multiple treatment of breast cancer. Glucose oxidase (GOx) and cupric sulfate (Cu) are introduced for transforming accumulated glucose (in cancer cells) to hydroxyl radicals for starvation/chemodynamic therapy. Cu may also suppress cancer cell growth via cuproptosis-mediated cell death. Berberine hydrochloride (BER) is engaged as a chemotherapeutic agent in the hydrogel reactor for combining with starvation/chemodynamic/cuproptosis therapeutic modalities. Moreover, Cu is participated as a gel crosslinker by coordinating with catechol groups in hyaluronic acid-dopamine (HD) polymer. Controlling viscoelasticity of hydrogel reactor can extend the retention time following local injection and provide sustained drug release patterns. Low biodegradation rate of designed HD/BER/GOx/Cu hydrogel can reduce dosing frequency in local cancer therapy and avoid invasiveness-related inconveniences. Especially, it is anticipated that HD/BER/GOx/Cu hydrogel system can be applied for reducing size of breast cancer prior to surgery as well as tumor growth suppression in clinical application.


Assuntos
Apoptose , Neoplasias da Mama , Neoplasias , Feminino , Humanos , Neoplasias da Mama/tratamento farmacológico , Catálise , Linhagem Celular Tumoral , Glucose Oxidase/metabolismo , Hidrogéis , Peróxido de Hidrogênio/química , Radical Hidroxila/química , Neoplasias/terapia , Cobre
14.
Colloids Surf B Biointerfaces ; 227: 113347, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37196465

RESUMO

Radiotherapy (RT) efficacy can be promoted with the help of nanoenzyme that can "re-programing" the tumour's micro-environment by changing the expression level of special bio-molecules. However, problems such as low reaction efficiency, limited endogenous H2O2, and/or unsatisfactory results of a single catalysis mode in treatment limit the application in the RT field. Herein, a novel Au nanoparticles (AuNPs) decorated iron SAE (FeSAE@Au) was formulated for self-cascade catalytic RT. In this dual-nanozyme system, embedded AuNPs can sever as GOx and endow FeSAE@Au with self-H2O2 supplying ability, which can elevate the H2O2 level in tumors by catalyzing cellular glucose in situ, further improving the catalytic performance of FeSAE with peroxidase-like activity. The self-cascade catalytic reaction can significantly increase cellular hydroxyl radicals (•OH) level, further promoting RT's effect. Furthermore, in vivo findings demonstrated that FeSAE can effectively limit tumor growth while causing low damage in important organs. According to our understanding, FeSAE@Au is the first description of a hybrid SAE-based nanomaterial employed in cascade catalytic RT. The research yields new and interesting insights for developing various SAE systems for anticancer therapy.


Assuntos
Neoplasias da Mama , Nanopartículas Metálicas , Neoplasias , Humanos , Feminino , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/radioterapia , Ouro/uso terapêutico , Peróxido de Hidrogênio , Neoplasias/patologia , Catálise , Glucose Oxidase/metabolismo , Microambiente Tumoral
15.
Adv Healthc Mater ; 12(21): e2300118, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37094801

RESUMO

Radiotherapy is a common cancer treatment approach in clinical practice, yet its efficacy has been restricted by tumor hypoxia. Nanomaterials-mediated systemic delivery of glucose oxidase (GOx) and catalase (CAT) or CAT-like nanoenzymes holds the potential to enhance tumor oxygenation. However, they face the challenge of intermediate (hydrogen peroxide [H2 O2 ]) escape during systemic circulation if the enzyme pair is not closely placed to largely decompose H2 O2 , leading to oxidative stress on normal tissues. In the present study, a oxygen-generating nanocascade, n(GOx-CAT)C7A , constructed by strategically placing an enzymatic cascade (GOx and CAT) within a polymeric coating rich in hexamethyleneimine (C7A) moieties, is reported. During blood circulation, C7A remains predominantly non-protonated , achieving prolonged blood circulation due to its low-fouling surface. Once n(GOx-CAT)C7A reaches the tumor site, the acidic tumor microenvironment (TME) induces protonation of C7A moieties, resulting in a positively charged surface for enhanced tumor transcytosis. Moreover, GOx and CAT are covalently conjugated into close spatial proximity (<10 nm) for effective H2 O2  elimination. As demonstrated by the in vivo results, n(GOx-CAT)C7A achieves effective tumor retention and oxygenation, potent radiosensitization and antitumor effects. Such a dual-enzyme nanocascade for smart O2  delivery holds great potential for enhancing the hypoxia-compromised cancer therapies.


Assuntos
Nanopartículas , Nanoestruturas , Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Neoplasias/patologia , Peróxido de Hidrogênio , Estresse Oxidativo , Hipóxia Tumoral , Oxigênio , Glucose Oxidase/metabolismo , Linhagem Celular Tumoral , Microambiente Tumoral
16.
Int J Biol Macromol ; 239: 124363, 2023 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-37031790

RESUMO

Strategies which are used to address the low levels of intracellular hydrogen peroxide and the development of biocompatible catalysts still need to be fulfilled in tumor chemodynamic therapy. Therefore, a novel tumor-targeted glycogen-based nanoparticle system (GN/He/GOx/HA) was developed to co-deliver hemin (He) and GOx, which can self-supply glucose formed upon degradation of glycogen by α-glycosidase in the lysosome environment, in order to achieve synergistic antitumor therapy. Hyaluronic acid (HA) was selected as the outer shell to protect the activity of GOx, and to increase the uptake by tumor cells via CD44 receptor-mediated endocytosis. GN/He/GOx/HA NPs had a good stability in the blood circulation, but fast release of the therapeutic cargos upon intracellular uptake. Hemin had a cascade catalytic reaction with GOx. Furthermore, GN/He/GOx/HA NPs had the strongest cytotoxicity in Hela cells in a glucose concentration dependent manner. The NPs could efficiently produce reactive oxygen species in tumor cells, resulting in a decrease in the mitochondrial membrane potential and apoptosis of tumor cells. The in vivo results showed that the drug-loaded nanoparticles had good safety, biocompatibility, and efficacious antitumor effect. Therefore, the glycogen-based nanoparticle delivery system provides potential application for self-enhancing CDT, which can be used for effective antitumor therapy.


Assuntos
Antineoplásicos , Nanopartículas , Neoplasias , Humanos , Antineoplásicos/farmacologia , Células HeLa , Glucose Oxidase/metabolismo , Hemina , Glicogênio , Neoplasias/metabolismo , Glucose , Peróxido de Hidrogênio/metabolismo , Linhagem Celular Tumoral
17.
J Nanobiotechnology ; 21(1): 59, 2023 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-36810074

RESUMO

Chemodynamic therapy of cancer is limited by insufficient endogenous H2O2 generation and acidity in the tumor microenvironment (TME). Herein, we developed a biodegradable theranostic platform (pLMOFePt-TGO) involving composite of dendritic organosilica and FePt alloy, loaded with tamoxifen (TAM) and glucose oxidase (GOx), and encapsulated by platelet-derived growth factor-B (PDGFB)-labeled liposomes, that effectively uses the synergy among chemotherapy, enhanced chemodynamic therapy (CDT), and anti-angiogenesis. The increased concentration of glutathione (GSH) present in the cancer cells induces the disintegration of pLMOFePt-TGO, releasing FePt, GOx, and TAM. The synergistic action of GOx and TAM significantly enhanced the acidity and H2O2 level in the TME by aerobiotic glucose consumption and hypoxic glycolysis pathways, respectively. The combined effect of GSH depletion, acidity enhancement, and H2O2 supplementation dramatically promotes the Fenton-catalytic behavior of FePt alloys, which, in combination with tumor starvation caused by GOx and TAM-mediated chemotherapy, significantly increases the anticancer efficacy of this treatment. In addition, T2-shortening caused by FePt alloys released in TME significantly enhances contrast in the MRI signal of tumor, enabling a more accurate diagnosis. Results of in vitro and in vivo experiments suggest that pLMOFePt-TGO can effectively suppress tumor growth and angiogenesis, thus providing an exciting potential strategy for developing satisfactory tumor theranostics.


Assuntos
Ferroptose , Neoplasias , Humanos , Linhagem Celular Tumoral , Microambiente Tumoral , Peróxido de Hidrogênio/metabolismo , Neoplasias/tratamento farmacológico , Apoptose , Glucose Oxidase/metabolismo
18.
Adv Healthc Mater ; 12(12): e2203035, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36661124

RESUMO

Biocatalytic systems based on enzyme cascade reactions have attracted growing interest in the field of biocatalytic medicine. However, it is a major challenge to reasonably construct enzyme cascade reactions with high stability, selectivity, and catalytic efficiency for the in vivo biocatalytic application. Herein, two-in-one engineered glucose oxidase (GOx-Fe0 ) is fabricated by a biomineralization strategy, through which a nanozyme (Fe0 NP) is anchored within the inner cavity of GOx. Then, GOx-Fe0 is immobilized in a pH-sensitive metal-organic framework (MOF) zeolitic imidazolate framework-8 (ZIF-8) to establish a stable and effective MOF-immobilized two-in-one engineered enzyme, GOx-Fe0 @ZIF-8. In vitro studies show that GOx-Fe0 @ZIF-8 exhibits excellent stability and high pH/glucose selectivity, and the shorter spacing between cascade enzymes can increase the cascade throughput and effectively improve the reaction efficiency of the enzyme cascade. In vivo experiments exhibit that GOx-Fe0 @ZIF-8 solves the instability and systemic toxicity of free enzymes, and achieves deep tumor penetration and significant chemodynamic therapeutic efficacy through a pH/glucose-selective enzyme cascade reaction in tumor site. Taken together, such an orchestrated enzyme engineering strategy can effectively improve enzyme stability, selectivity, and enzyme cascade reaction efficiency via chemical transformations, and also provide a promising strategy for the application of biocatalytic cascade reactions in vivo.


Assuntos
Estruturas Metalorgânicas , Zeolitas , Enzimas Imobilizadas/uso terapêutico , Enzimas Imobilizadas/metabolismo , Glucose , Biocatálise , Estabilidade Enzimática , Glucose Oxidase/metabolismo
19.
Small ; 19(5): e2206592, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36437115

RESUMO

Starvation therapy kills tumor cells via consuming glucose to cut off their energy supply. However, since glucose oxidase (GOx)-mediated glycolysis is oxygen-dependent, the cascade reaction based on GOx faces the challenge of a hypoxic tumor microenvironment. By decomposition of glycolysis production of H2 O2 into O2 , starvation therapy can be enhanced, but chemodynamic therapy is limited. Here, a close-loop strategy for on demand H2 O2 and O2 delivery, release, and recycling is proposed. The nanoreactor (metal-protein-polyphenol capsule) is designed by incorporating two native proteins, GOx and hemoglobin (Hb), in polyphenol networks with zeolitic imidazolate framework as sacrificial templates. Glycolysis occurs in the presence of GOx with O2 consumption and the produced H2 O2 reacts with Hb to produce highly cytotoxic hydroxyl radicals (•OH) and methemoglobin (MHb) (Fenton reaction). Benefiting from the different oxygen carrying capacities of Hb and MHb, oxygen on Hb is rapidly released to supplement its consumption during glycolysis. Glycolysis and Fenton reactions are mutually reinforced by oxygen supply, consuming more glucose and producing more hydroxyl radicals and ultimately enhancing both starvation therapy and chemodynamic therapy. This cascade nanoreactor exhibits high efficiency for tumor suppression and provides an effective strategy for oxygen-mediated synergistic starvation therapy and chemodynamic therapy.


Assuntos
Nanopartículas , Neoplasias , Humanos , Oxigênio , Polifenóis , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Metais , Radical Hidroxila , Glucose Oxidase/metabolismo , Glucose , Nanotecnologia , Peróxido de Hidrogênio , Linhagem Celular Tumoral , Microambiente Tumoral
20.
Small ; 19(10): e2206257, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36549673

RESUMO

This work focusses on developing a hybrid enzyme biofuel cell-based self-powered biosensor with appreciable stability and durability using murine leukemia fusion gene fragments (tDNA) as a model analyte. The cell consists of a Ti3 C2 Tx /multiwalled carbon nanotube/gold nanoparticle/glucose oxidase bioanode and a Zn/Co-modified carbon nanotube cathode. The bioanode uniquely exhibits strong electron transfer ability and a high surface area for the loading of 1.14 × 10-9  mol cm-2 glucose oxidase to catalyze glucose oxidation. Meanwhile, the abiotic cathode with a high oxygen reduction reaction activity negates the use of conventional bioenzymes as catalysts, which aids in extending the stability and durability of the sensing system. The biosensor offers a 0.1 fm-1 nm linear range and a detection limit of 0.022 fm tDNA. Additionally, the biosensor demonstrates a reproducibility of ≈4.85% and retains ≈87.42% of the initial maximal power density after a 4-week storage at 4 °C, verifying a significantly improved long-term stability.


Assuntos
Fontes de Energia Bioelétrica , Técnicas Biossensoriais , Nanopartículas Metálicas , Nanotubos de Carbono , Animais , Camundongos , Glucose Oxidase/metabolismo , Biocombustíveis , Ouro , Reprodutibilidade dos Testes , Titânio , Eletrodos , Glucose
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA