Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
1.
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
2.
Mater Horiz ; 11(10): 2406-2419, 2024 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-38440840

RESUMO

Enzymes provide a class of potential options to treat cancer, while the precise regulation of enzyme activities for effective and safe therapeutic actions has been poorly reported. Dual-enzyme decorated semiconducting polymer nanoagents for second near-infrared (NIR-II) photoactivatable ferroptosis-immunotherapy are reported in this study. Such nanoagents (termed SPHGA) consist of hemoglobin (Hb)-based semiconducting polymer (SP@Hb), adenosine deaminase (ADA) and glucose oxidase (GOx) with loadings in a thermal-responsive nanoparticle shell. NIR-II photoactivation of SPHGA results in the generation of heat to trigger on-demand releases of two enzymes (ADA and GOx) via destroying the thermal-responsive nanoparticle shells. In the tumor microenvironment, GOx oxidizes glucose to form hydrogen peroxide (H2O2), which promotes the Fenton reaction of iron in SP@Hb, resulting in an enhanced ferroptosis effect and immunogenic cell death (ICD). In addition, ADA degrades high-level adenosine to reverse the immunosuppressive microenvironment, thus amplifying antitumor immune responses. Via NIR-II photoactivatable ferroptosis-immunotherapy, SPHGA shows an improved effect to absolutely remove bilateral tumors and effectively suppress tumor metastases in subcutaneous 4T1 breast cancer models. This study presents a dual-enzyme-based nanoagent with controllable therapeutic actions for effective and precise cancer therapy.


Assuntos
Ferroptose , Imunoterapia , Raios Infravermelhos , Nanopartículas , Polímeros , Semicondutores , Ferroptose/efeitos dos fármacos , Animais , Imunoterapia/métodos , Camundongos , Polímeros/química , Polímeros/uso terapêutico , Feminino , Nanopartículas/uso terapêutico , Nanopartículas/química , Linhagem Celular Tumoral , Microambiente Tumoral/efeitos dos fármacos , Glucose Oxidase/metabolismo , Glucose Oxidase/farmacologia , Humanos , Camundongos Endogâmicos BALB C , Hemoglobinas/farmacologia , Hemoglobinas/metabolismo
3.
Colloids Surf B Biointerfaces ; 232: 113581, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37857184

RESUMO

In clinical practice, surgery is the preferred treatment for breast cancer; however, the high recurrence rate due to residual tumors after surgery remains a major issue. Hydrogels can reduce the side effects of residual tumors and exert strong anticancer effects, thereby showing potential as therapeutic agents for suppressing tumor recurrence after surgery. Glucose oxidase (GOD)-immobilized gelatin hydrogels (GOD-gelatin hydrogel) were prepared by bioorthogonal click chemistry. Then, the anticancer effect, tumor recurrence inhibition, and biodegradability of the resulting hydrogels were evaluated through cell and animal experiments. GOD-gelatin hydrogel showed cytotoxicity and anticancer effect via H2O2 generation. Unlike free GOD, GOD-gelatin hydrogel remained in the surgical site after implant and continued to suppress tumor recurrence over time. The proposed GOD-gelatin hydrogel system can be easily implanted at the surgical site after tumor surgery, representing a novel treatment to suppress tumor recurrence without any systemic toxicity.


Assuntos
Gelatina , Hidrogéis , Animais , Hidrogéis/farmacologia , Gelatina/farmacologia , Glucose Oxidase/farmacologia , Recidiva Local de Neoplasia/prevenção & controle , Peróxido de Hidrogênio/farmacologia , Neoplasia Residual
4.
Acta Biomater ; 172: 441-453, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37802309

RESUMO

Photothermal therapy (PTT) combined with chemodynamic therapy (CDT) presents an appealing complementary anti-tumor strategy, wherein PTT accelerates the production of reactive oxygen species (ROS) in CDT and CDT eliminates residual tumor tissues that survive from PTT treatment. However, nanomaterials utilized in PTT/CDT are limited by non-specific damage to the entire organism. Herein, a glucose-responsive enzymatic Fe@HRP-ABTS/GOx nanodot is judiciously designed for tumor-specific PTT/CDT via a simple and clean protein-templated biomimetic mineralization synthesis. By oxidizing glucose in tumor cells, glucose oxidase (GOx) activates glucose-responsive tumor therapy and increases the concentration of H2O2 at the tumor site. More importantly, the self-supplied peroxide hydrogen (H2O2) can convert ABTS (2,2'-Hydrazine-bis(3-ethylbenzothiazoline-6-sulfonic acid) diamine salt) into oxidized ABTS (oxABTS) through horseradish peroxidase (HRP) catalysis for PTT and photoacoustic (PA) imaging. Furthermore, the Fe2+ arising from the reduction of Fe3+ by overexpressed GSH reacts with H2O2 to generate intensely reactive •OH through the Fenton reaction, concurrently depleting GSH and inducing efficient tumor CDT. The in vitro and in vivo experiments demonstrate superior cancer cell killing and tumor eradication effect of Fe@HRP-ABTS/GOx nanodot under near-infrared (NIR) laser irradiation. Collectively, the nanodots provide mutually reinforcing catalytic PTT/CDT anti-tumor strategies for treating liver cancer and potentially other malignancies. STATEMENT OF SIGNIFICANCE: Combinatorial antitumor therapy with nanomedicines presents great prospects for development. However, the limitation of non-specific damage to normal tissues hinders its further clinical application. In this work, we fabricated tumor-selective biomimetic Fe@HRP-ABTS/GOx nanodots for H2O2 self-supplied catalytic photothermal/chemodynamic therapy of tumors. The biomimetic synthesis strategy provides the nanodots with enzymatic activity in response to glucose to produce H2O2. The self-supplied H2O2 initiates photothermal therapy with oxidized ABTS and enhances chemodynamic therapy through simultaneous •OH generation and GSH depletion. Our work provides a new paradigm for developing tumor-selective catalytic nanomedicines and will guide further clinical translation of the enzymatic biomimetic synthesis strategy.


Assuntos
Nanopartículas , Neoplasias , Humanos , Biomimética , Peróxido de Hidrogênio , Terapia Fototérmica , Catálise , Glucose , Glucose Oxidase/farmacologia , Peroxidase do Rábano Silvestre , Linhagem Celular Tumoral , Microambiente Tumoral , Nanopartículas/uso terapêutico
5.
Angew Chem Int Ed Engl ; 62(44): e202308761, 2023 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-37496129

RESUMO

Enzymatic reactions can consume endogenous nutrients of tumors and produce cytotoxic species and are therefore promising tools for treating malignant tumors. Inspired by nature where enzymes are compartmentalized in membranes to achieve high reaction efficiency and separate biological processes with the environment, we develop liposomal nanoreactors that can perform enzymatic cascade reactions in the aqueous nanoconfinement of liposomes. The nanoreactors effectively inhibited tumor growth in vivo by consuming tumor nutrients (glucose and oxygen) and producing highly cytotoxic hydroxyl radicals (⋅OH). Co-compartmentalization of glucose oxidase (GOx) and horseradish peroxidase (HRP) in liposomes could increase local concentration of the intermediate product hydrogen peroxide (H2 O2 ) as well as the acidity due to the generation of gluconic acid by GOx. Both H2 O2 and acidity accelerate the second-step reaction by HRP, hence improving the overall efficiency of the cascade reaction. The biomimetic compartmentalization of enzymatic tandem reactions in biocompatible liposomes provides a promising direction for developing catalytic nanomedicines in antitumor therapy.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Lipossomos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Glucose Oxidase/farmacologia , Peroxidase do Rábano Silvestre , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Nanotecnologia , Peróxido de Hidrogênio/uso terapêutico
6.
Adv Sci (Weinh) ; 10(21): e2301278, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37114827

RESUMO

Tumor starvation induced by intratumor glucose depletion emerges as a promising strategy for anticancer therapy. However, its antitumor potencies are severely compromised by intrinsic tumor hypoxia, low delivery efficiencies, and undesired off-target toxicity. Herein, a multifunctional cascade bioreactor (HCG), based on the self-assembly of pH-responsive hydroxyethyl starch prodrugs, copper ions, and glucose oxidase (GOD), is engineered, empowered by hyperbaric oxygen (HBO) for efficient cooperative therapy against aggressive breast cancers. Once internalized by tumor cells, HCG undergoes disassembly and releases cargoes in response to acidic tumor microenvironment. Subsequently, HBO activates GOD-catalyzed oxidation of glucose to H2 O2 and gluconic acid by ameliorating tumor hypoxia, fueling copper-catalyzed •OH generation and pH-responsive drug release. Meanwhile, HBO degrades dense tumor extracellular matrix, promoting tumor accumulation and penetration of HCG. Moreover, along with the consumption of glucose and the redox reaction of copper ions, the antioxidant capacity of tumor cells is markedly reduced, collectively boosting oxidative stress. As a result, the combination of HCG and HBO can not only remarkably suppress the growth of orthotopic breast tumors but also restrain pulmonary metastases by inhibiting cancer stem cells. Considering the clinical accessibility of HBO, this combined strategy holds significant translational potentials for GOD-based therapies.


Assuntos
Neoplasias da Mama , Oxigenoterapia Hiperbárica , Radiossensibilizantes , Humanos , Feminino , Cobre , Oxigênio , Neoplasias da Mama/terapia , Glucose Oxidase/farmacologia , Glucose/metabolismo , Microambiente Tumoral
7.
ACS Biomater Sci Eng ; 8(12): 5145-5154, 2022 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-36344935

RESUMO

The high systemic blood glucose concentration of hyperglycemic wound microenvironment (WME) severely impedes the disinfection and healing of infected skin wounds. Herein, a WME-activated smart natural product, integrated GOx-GA-Fe nanozyme (GGFzyme), is engineered, which combines a nanozyme and natural enzyme to promote reactive oxygen species (ROS) generation in situ for hyperglycemic wound disinfection. GGFzyme can consume a high concentration of glucose in hyperglycemia wounds and generate H2O2. The conversion of glucose into gluconic acid not avails starvation treatment but reduces the pH of WME to elevate the catalytic activities of both the nanozyme (GA-Fe) and natural enzyme (GOx). And H2O2 is then high efficiently catalyzed into •OH and O2•- in situ to combat pathogenic bacteria and promote wound disinfection. The high catalytic antibacterial capacity and superior biosafety, combined with beneficial WME modulation, demonstrate that GGFzyme is a promising therapeutic agent for hyperglycemic wounds.


Assuntos
Glucose Oxidase , Peróxido de Hidrogênio , Glucose Oxidase/farmacologia , Peróxido de Hidrogênio/farmacologia , Glucose , Cicatrização , Bactérias
8.
Nutrients ; 14(19)2022 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-36235593

RESUMO

Oxidative stress induces DNA damage which can be repaired by DNA repair proteins, such as Ku70/80. Excess reactive oxygen species (ROS) stimulate the activation of caspase-3, which degrades Ku 70/80. Cells with decreased Ku protein levels undergo apoptosis. Astaxanthin exerts antioxidant activity by inducing the expression of catalase, an antioxidant enzyme, in gastric epithelial cells. Therefore, astaxanthin may inhibit oxidative stress-induced DNA damage by preventing Ku protein degradation and thereby suppressing apoptosis. Ku proteins can be degraded via ubiquitination and neddylation which adds ubiquitin-like protein to substrate proteins. We aimed to determine whether oxidative stress decreases Ku70/80 expression through the ubiquitin-proteasome pathway to induce apoptosis and whether astaxanthin inhibits oxidative stress-induced changes in gastric epithelial AGS cells. We induced oxidative stress caused by the treatment of ß-D-glucose (G) and glucose oxidase (GO) in the cells. As a result, the G/GO treatment increased ROS levels, decreased nuclear Ku protein levels and Ku-DNA-binding activity, and induced the ubiquitination of Ku80. G/GO increased the DNA damage marker levels (γ-H2AX; DNA fragmentation) and apoptosis marker annexin V-positive cells and cell death. Astaxanthin inhibited G/GO-induced alterations, including Ku degradation in AGS cells. MLN4924, a neddylation inhibitor, and MG132, a proteasome inhibitor, suppressed G/GO-mediated DNA fragmentation and decreased cell viability. These results indicated that G/GO-induced oxidative stress causes Ku protein loss through the ubiquitin-proteasome pathway, resulting in DNA fragmentation and apoptotic cell death. Astaxanthin inhibited oxidative stress-mediated apoptosis via the reduction of ROS levels and inhibition of Ku protein degradation. In conclusion, dietary astaxanthin supplementation or astaxanthin-rich food consumption may be effective for preventing or delaying oxidative stress-mediated cell damage by suppressing Ku protein loss and apoptosis in gastric epithelial cells.


Assuntos
Antioxidantes , Complexo de Endopeptidases do Proteassoma , Anexina A5/metabolismo , Anexina A5/farmacologia , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Apoptose , Caspase 3/metabolismo , Catalase/metabolismo , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Células Epiteliais/metabolismo , Glucose/metabolismo , Glucose Oxidase/metabolismo , Glucose Oxidase/farmacologia , Autoantígeno Ku/metabolismo , Estresse Oxidativo , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Proteólise , Espécies Reativas de Oxigênio/metabolismo , Ubiquitinas/metabolismo , Ubiquitinas/farmacologia , Xantofilas
9.
ACS Nano ; 16(8): 12118-12133, 2022 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-35904186

RESUMO

To date, the construction of heterogeneous interfaces between sonosensitizers and other semiconductors or noble metals has aroused increasing attention, owing to an enhanced interface charge transfer, augmented spin-flip, and attenuated activation energy of oxygen. Here, a smart therapeutic nanoplatform is constructed by surface immobilization of glucose oxidase (GOx) onto a TiO2@Pt Schottky junction. The sonodynamic therapy (SDT) and starvation therapy (ST) mediated by TiO2@Pt/GOx (TPG) promote systemic tumor suppression upon hypoxia alleviation in tumor microenvironment. The band gap of TiO2@Pt is outstandingly decreased to 2.9 eV, in contrast to that of pristine TiO2. The energy structure optimization enables a more rapid generation of singlet oxygen (1O2) and hydroxyl radicals (•OH) by TiO2@Pt under ultrasound irradiation, resulting from an enhanced separation of hole-electron pair for redox utilization. The tumorous reactive oxygen species (ROS) accumulation and GOx-mediated glucose depletion facilitate oxidative damage and energy exhaustion of cancer cells, both of which can be tremendously amplified by Pt-catalyzed oxygen self-supply. Importantly, the combinatorial therapy triggers intense immunogenetic cell death, which favors a follow-up suppression of distant tumor and metastasis by evoking antitumor immunity. Collectively, this proof-of-concept paradigm provides an insightful strategy for highly efficient SDT/ST, which possesses good clinical potential for tackling cancer.


Assuntos
Neoplasias , Terapia por Ultrassom , Humanos , Platina , Microambiente Tumoral , Glucose , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Espécies Reativas de Oxigênio/metabolismo , Glucose Oxidase/farmacologia , Glucose Oxidase/metabolismo , Oxigênio , Linhagem Celular Tumoral
10.
Biomater Sci ; 10(15): 4170-4183, 2022 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-35726827

RESUMO

Although CuO-deposited bovine serum albumin (CuO-BSA) and glucose oxidase (GOx) were combined to achieve H2O2 self-supplied chemo-dynamic therapy (CDT) and glucose consumption-based starvation therapy, the uses of copper and GOx have not been optimized to enhance tumour-selective reactive oxygen species (ROS) generation and minimize toxicity to normal cells as well. Here, chemo-dynamic nanoparticles (CBGP NPs) were prepared through a facile biomineralization process and subsequent coatings with GOx and the cationic polymer PEG2k-PEI1.8k. Through optimizing the use of copper, GOx, and PEG2k-PEI1.8k, the CBGP NPs showed high cellular uptake efficiency, enhanced tumour-selective ROS generation, and minimal side effects toward normal cells. The CBGP NP-mediated glucose consumption, GSH-depletion, and ˙OH generation synergistically induced tumour cell apoptosis both in vitro and in vivo. It is believed that the optimized CBGP NPs can be a promising nanoplatform for effective tumour therapy with minimal side effects.


Assuntos
Glioma , Nanopartículas , Neoplasias , Animais , Linhagem Celular Tumoral , Cobre , Glioma/tratamento farmacológico , Glucose , Glucose Oxidase/farmacologia , Peróxido de Hidrogênio , Camundongos , Neoplasias/tratamento farmacológico , Espécies Reativas de Oxigênio
11.
Acta Biomater ; 145: 222-234, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35460908

RESUMO

Cancer cells rely on glycolysis to support a high proliferation rate. Metformin (Met) is a promising drug for tumor treatment that targets hexokinase 2 (HK2) to block the glycolytic process, thereby further disrupting the metabolism of cancer cells. Herein, an intelligent nanomedicine based on glucose deprivation and glycolysis inhibition is creatively constructed for enhanced cancer synergistic treatment. In brief, Met and glucose oxidase (GOx) was encapsulated into histidine/zeolitic imidazolate framework-8 (His/ZIF-8), which was followed by coating with Arg-Gly-Asp (RGD) peptides to obtain the desired nanomedicine (Met/GOx@His/ZIF-8∼RGD). This smart nanomedicine presents the controllable Met and GOx release behavior in an acidic responsive manner. The liberated Met blocks the glycolysis process via suppressing the activity of HK2 and impairing ATP production, which activates the AMP-activated protein kinase (AMPK) pathway and p53 pathway and damages the Warburg effect, eventually leading to cells apoptosis. And the GOx boosts the glucose shortage for starvation therapy by depleting accumulated glucose. According to in vitro and in vivo assays, the combination of glycolysis inhibition and starvation therapy demonstrates efficient cancer cells growth suppression and superior antitumor properties compared to the Met based or GOx-mediated monotherapy. This work provides an advanced therapeutic strategy via disrupting cellular metabolism against cancer. STATEMENT OF SIGNIFICANCE: The obtained nanomedicine (Met/GOx@His/ZIF-8∼RGD) presents the controllable Met and glucose oxidase (GOx) release behavior in an acidic responsive manner. The liberated Met blocks the glycolysis process via suppressing the activity of HK2 and impairing ATP production, which activates the AMP-activated protein kinase (AMPK) pathway and p53 pathway and damages the Warburg effect, eventually leading to cells apoptosis. And the GOx boosts the glucose shortage for starvation therapy by depleting accumulated glucose. The combination of glycolysis inhibition and starvation therapy demonstrate the efficient suppression of cancer cells growth and the superior antitumor properties when compared to the Met based or GOx-mediated monotherapy.


Assuntos
Glucose Oxidase , Metformina , Neoplasias , Proteínas Quinases Ativadas por AMP/metabolismo , Trifosfato de Adenosina/metabolismo , Linhagem Celular Tumoral , Tratamento Farmacológico/métodos , Glucose , Glucose Oxidase/farmacologia , Glucose Oxidase/uso terapêutico , Glicólise/efeitos dos fármacos , Humanos , Metformina/farmacologia , Metformina/uso terapêutico , Neoplasias/patologia , Proteína Supressora de Tumor p53/metabolismo
12.
Acta Biomater ; 141: 429-439, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35038584

RESUMO

Manganese has recently been exploited for cancer immunotherapy, fenton-like reaction-mediated chemo-dynamic therapy, and magnetic resonance imaging. The integration of multiple roles of manganese into one platform is of great significance for cancer theranostics and tumor inhibition. Here, we designed a multifunctional nanoplatform based on manganese, which consisted of a manganese-containing inner core and a phospholipid bilayer shell co-loaded with glucose oxidase (GOx), paclitaxel (PTX), and a NIR fluorescent dye (NanoMn-GOx-PTX). In a pH-dependent manner, the nanoplatform released manganese ions and payloads inside the tumor cells. In vitro characterization and cellular experiments indicated that NanoMn-GOx-PTX could catalyze the conversion of glucose into reactive oxygen species (ROS) through a cascade Fenton-like reaction as well as release free PTX. The consumption of glucose, ROS production, and the chemotherapeutic effect of PTX contributed to the superior cytotoxicity and apoptosis of 4T1 cancer cells. Moreover, NanoMn-GOx-PTX effectively induced the production of large amounts of type I interferon and pro-inflammatory cytokines in vivo, activating the innate immune response. Through the synergistic functions of the above components, NanoMn-GOx-PTX exerted the strongest anti-tumor effect in 4T1 tumor-bearing models. Therefore, the manganese-based nanoplatform could serve as a promising theranostic tool for breast cancer therapy. STATEMENT OF SIGNIFICANCE: 1) This nanoplatform can be used as a universal tool for delivering proteins and anticancer drugs into cells; 2) The PEG-modified phospholipid bilayer shell plays a significant role in retarding the release of overloaded manganese ions and drugs in a pH-sensitive manner; 3) The released Mn2+ has the ability to enhance T1 contrast in magnetic resonance imaging; 4) The released Mn2+ can function as nanoadjuvants to activate the cGAS-STING pathway and effectively induce the natural immune response;5) The overloaded manganese ions are combined with glucose oxidase to form a cascade reaction system, indirectly converting glucose into ROS to induce oxidative damage of tumor tissue.


Assuntos
Neoplasias da Mama , Nanopartículas , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Feminino , Glucose , Glucose Oxidase/farmacologia , Humanos , Íons , Manganês , Nanopartículas/uso terapêutico , Paclitaxel/farmacologia , Fosfolipídeos , Espécies Reativas de Oxigênio/metabolismo
13.
Theranostics ; 11(20): 10001-10011, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34815800

RESUMO

Rationale: Glucose oxidase (GOx)-based biocatalytic nanoreactors can cut off the energy supply of tumors for starvation therapy and deoxygenation-activated chemotherapy. However, these nanoreactors, including mesoporous silica, calcium phosphate, metal-organic framework, or polymer nanocarriers, cannot completely block the reaction of GOx with glucose in the blood, inducing systemic toxicity from hydrogen peroxide (H2O2) and anoxia. The low enzyme loading capacity can reduce systemic toxicity but limits its therapeutic effect. Here, we describe a real 'ON/OFF' intelligent nanoreactor with a core-shell structure (GOx + tirazapamine (TPZ))/ZIF-8@ZIF-8 modified with the red cell membrane (GTZ@Z-RBM) for cargo delivery. Methods: GTZ@Z-RBM nanoparticles (NPs) were prepared by the co-precipitation and epitaxial growth process under mild conditions. The core-shell structure loaded with GOx and TPZ was characterized for hydrate particle size and surface charge. The GTZ@Z-RBM NPs morphology, drug, and GOx loading/releasing abilities, system toxicity, multimodal synergistic therapy, and tumor metastasis suppression were investigated. The in vitro and in vivo outcomes of GTZ@Z-RBM NPs were assessed in 4T1 breast cancer cells. Results: GTZ@Z-RBM NPs could spatially isolate the enzyme from glucose in a physiological environment, reducing systemic toxicity. The fabricated nanoreactor with high enzyme loading capacity and good biocompatibility could deliver GOx and TPZ to the tumors, thereby exhausting glucose, generating H2O2, and aggravating hypoxic microenvironment for starvation therapy, DNA damage, and deoxygenation-activated chemotherapy. Significantly, the synergistic therapy effectively suppressed the breast cancer metastasis in mice and prolonged life without systemic toxicity. The in vitro and in vivo results provided evidence that our biomimetic nanoreactor had a powerful synergistic cascade effect in treating breast cancer. Conclusion: GTZ@Z-RBM NPs can be used as an 'ON/OFF' intelligent nanoreactor to deliver GOx and TPZ for multimodal synergistic therapy and tumor metastasis suppression.


Assuntos
Glucose Oxidase/farmacologia , Sistemas de Liberação de Fármacos por Nanopartículas/farmacologia , Nanotecnologia/métodos , Animais , Biomimética , Linhagem Celular Tumoral , China , Terapia Combinada , Feminino , Glucose Oxidase/administração & dosagem , Concentração de Íons de Hidrogênio , Camundongos , Nanopartículas/química , Neoplasias/tratamento farmacológico , Tirapazamina/administração & dosagem , Tirapazamina/farmacologia , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
14.
Molecules ; 26(18)2021 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-34577080

RESUMO

The selective disruption of nutritional supplements and the metabolic routes of cancer cells offer a promising opportunity for more efficient cancer therapeutics. Herein, a biomimetic cascade polymer nanoreactor (GOx/CAT-NC) was fabricated by encapsulating glucose oxidase (GOx) and catalase (CAT) in a porphyrin polymer nanocapsule for combined starvation and photodynamic anticancer therapy. Internalized by cancer cells, the GOx/CAT-NCs facilitate microenvironmental oxidation by catalyzing endogenous H2O2 to form O2, thereby accelerating intracellular glucose catabolism and enhancing cytotoxic singlet oxygen (1O2) production with infrared irradiation. The GOx/CAT-NCs have demonstrated synergistic advantages in long-term starvation therapy and powerful photodynamic therapy (PDT) in cancer treatment, which inhibits tumor cells at more than twice the rate of starvation therapy alone. The biomimetic polymer nanoreactor will further contribute to the advancement of complementary modes of spatiotemporal control of cancer therapy.


Assuntos
Nanopartículas/química , Neoplasias/terapia , Fotoquimioterapia/métodos , Polímeros/química , Animais , Biomimética , Catalase/química , Catalase/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Glucose Oxidase/química , Glucose Oxidase/farmacologia , Humanos , Peróxido de Hidrogênio/metabolismo , Raios Infravermelhos , Camundongos , Polímeros/síntese química , Porfirinas/síntese química , Porfirinas/química , Oxigênio Singlete/metabolismo , Oxigênio Singlete/farmacologia
15.
Nanotechnology ; 33(2)2021 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-34544066

RESUMO

Glucose oxidase-mediated starvation therapy that effectively cuts off energy supply holds great promise in cancer treatment. However, high glutathione (GSH) contents and anoxic conditions severely reduce therapy efficiency and cannot fully kill cancer cells. Herein, to resolve the above problem, this study constructed a biomimetic nanosystem based on nanreproo-MnO2with porous craspedia globose-like structure and high specific surface area, and it was further modified with dopamine and folic acid to guarantee good biocompatibility and selectivity toward cancer cells. This nanosystem responsively degraded and reacted with GSH and acid to regenerate O2, which significantly increased intracellular O2levels, accelerated glucose consumption, and improved starvation therapy efficiency. Moreover, anticancer drug of camptothecin was further loaded, and notably enhanced cancer growth inhibition was obtained at very low drug concentrations. Most importantly, this novel therapy could unprecedentedly inhibit cancer cell migration to a very low ratio of 19%, and detailed cell apoptosis analyses revealed late stage apoptosis contributed most to the good therapeutic effect. This work reported a new train of thought to improve starvation therapy in biomedicine, and provided a new strategy to design targeted nanocarrier to delivery mixed drugs to overcome the restriction of starvation therapy and develop new therapy patterns.


Assuntos
Antineoplásicos , Glucose Oxidase , Neoplasias/terapia , Oxigênio/metabolismo , Hipóxia Tumoral/efeitos dos fármacos , Células A549 , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Biomimética , Camptotecina/farmacocinética , Camptotecina/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Portadores de Fármacos , Glucose Oxidase/química , Glucose Oxidase/metabolismo , Glucose Oxidase/farmacologia , Células HeLa , Humanos , Indóis/química , Compostos de Manganês/química , Nanopartículas Metálicas/química , Nanomedicina , Óxidos/química , Polímeros/química , Propriedades de Superfície
16.
Theranostics ; 11(17): 8270-8282, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34373741

RESUMO

Rationale: Glucose oxidase (GOx) has gained tremendous research interest recently as a glucose-consuming enzyme for tumor starvation therapy, while its in vivo applications are strictly limited by rapid deactivation, as well as side effects of non-specific catalysis. Methods: To address these issues, here we report a protective nano-shell to encapsule GOx for localized melanoma therapy delivered by dissolving microneedles (MNs). Inspired by cell membrane that separates and protects cell organelles and components from outside environment while selectively ingesting nutrition sources, we designed polydopamine (PDA)-structured nano-shell to allow free transportation of glucose for catalytic reaction, while impede the penetration of GOx, proteinase, and other GOx-deactivating macromolecules across the shell membrane. Results: GOx was well protected in core layer with persistent catalytic activity for at least 6 d under various biological matrixes (e.g., PBS, serum, and cell lysate) and surviving different harsh conditions (e.g., acid/base treatments, and proteinase-induced degradation). Such long-acting nano-catalyst can be easily integrated into MNs as topical delivery carrier for effective glucose consumption in melanoma tissue, achieving significant tumor growth inhibition via starvation therapy with minimized side effects as compared to systemic administration. Conclusion: This work provides an elegant platform for in vivo delivery of GOx, and our cell-mimicking nano-system can also be applied for other enzyme-based therapeutics.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Glucose Oxidase/farmacologia , Melanoma/tratamento farmacológico , Nanoconchas/administração & dosagem , Administração Cutânea , Animais , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Glucose Oxidase/metabolismo , Indóis/química , Camundongos , Camundongos Endogâmicos C57BL , Nanoconchas/química , Polímeros/química , Neoplasias Cutâneas/tratamento farmacológico , Inanição
17.
Stem Cells Transl Med ; 10(10): 1446-1453, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34160898

RESUMO

Hematopoietic stem progenitor cells (HSPCs) mobilized to peripheral blood, rather than those remaining in the bone marrow (BM), are commonly used as stem cell source in the clinic. As reactive oxygen species (ROS) are suggested as mediator of HSPC mobilization, we examined the impacts of glucose oxidase (GO) on peripheral mobilization of BM HSPCs and the associated mechanisms. Intravenous injection of GO induced HSPC mobilization even by single treatment, and the GO-mobilized cells maintained their long-term reconstituting and differentiating potentials in conditioned recipients. GO-injected mice lived a normal life without adverse effects such as stem cell senescence, hematopoietic disorders, and blood parameter alteration. The mobilization effect of GO was even evident in animal models showing poor mobilization, such as old, 5-fluorouracil-treated, or alendronate-treated mice. Importantly, combined injection of GO with granulocyte colony-stimulating factor (G-CSF) and/or AMD3100 enhanced more greatly HSPC mobilization than did G-CSF, AMD3100, or both. The GO-stimulated HSPC mobilization was almost completely attenuated by N-acetyl-L-cysteine treatment. Collectively, our results not only highlight the potential role of GO in HSPC mobilization via ROS signaling, but also provide a GO-based new strategy to improve HSPC mobilization in poorly mobilizing allogeneic or autologous donors via combination with G-CSF and/or AMD3100.


Assuntos
Glucose Oxidase , Mobilização de Células-Tronco Hematopoéticas , Animais , Medula Óssea/metabolismo , Glucose Oxidase/metabolismo , Glucose Oxidase/farmacologia , Fator Estimulador de Colônias de Granulócitos/metabolismo , Fator Estimulador de Colônias de Granulócitos/farmacologia , Células-Tronco Hematopoéticas/metabolismo , Camundongos
18.
ACS Appl Mater Interfaces ; 13(11): 12960-12971, 2021 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-33720684

RESUMO

The optimal therapy effect of tumors is frequently restricted by the dense extracellular matrix (ECM) and anoxia. Herein, an intelligent BPNs-Arg-GOx@MnO2 (BAGM) nanozyme is innovatively designed as a multimodal synergistic therapeutic paradigm that possesses both nitric oxide (NO) self-supplying and ECM degradation properties to reinforce the therapy effect by a tumor microenvironment (TME)-activatable cyclic cascade catalytic reaction. This theranostic nanoplatform is constructed by using polyethyleneimine-modified black phosphorus nanosheets as a "fishnet" to attach l-Arginine (l-Arg) and glucose oxidase (GOx) and then depositing mini-sized MnO2 nanosheets (MNs) on the surface by a facile situ biomineralization method. As an intelligent "switch", the MNs can effectively trigger the cascade reaction by disintegrating intracellular H2O2 to release O2. Then, the conjugated GOx can utilize O2 production to catalyze intracellular glucose to generate H2O2, which not only starves the tumor cells but also promotes oxidation of l-Arg to NO. Thereafter, matrix metalloproteinases will be activated by NO production to degrade the dense ECM and transform matrix collagen into a loose state. In turn, a loose ECM can enhance the accumulation of the BAGM nanozyme and thereby reinforce synergistic photothermal therapy/starvation therapy/NO gas therapy. Both in vitro and in vivo results indicate that the TME-tunable BAGM therapeutic nanoplatform with cascade anticancer property and satisfactory biosecurity shows potential in nanomedicine.


Assuntos
Neoplasias da Mama/terapia , Glucose Oxidase/farmacologia , Compostos de Manganês/farmacologia , Nanoestruturas , Óxidos/farmacologia , Microambiente Tumoral , Animais , Neoplasias da Mama/metabolismo , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/patologia , Feminino , Glucose Oxidase/administração & dosagem , Humanos , Peróxido de Hidrogênio/metabolismo , Células MCF-7 , Compostos de Manganês/administração & dosagem , Camundongos , Nanomedicina , Nanoestruturas/administração & dosagem , Nanoestruturas/química , Óxido Nítrico/metabolismo , Óxidos/administração & dosagem , Oxigênio/metabolismo , Microambiente Tumoral/efeitos dos fármacos
19.
Poult Sci ; 99(12): 6549-6558, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33248570

RESUMO

The negative effects of dietary antibiotics have become a widespread concern. It is imperative to search for a new type of green, safe, and efficient feed additive that can replace antibiotics. This study was to investigate the effects of glucose oxidase (GOD) on growth performance, immune function, and intestinal barrier in ducks infected with Escherichia coli O88. First, we established the E. coli challenge model of ducks through a preliminary experiment and then carried out the formal experiment by using 144 1-day-old male lean Peking ducklings (50 ± 2.75 g). All ducks were randomly assigned to 1 of 3 dietary treatment groups of basal diet (control), 30 mg/kg virginiamycin (antibiotic), and 200 U/kg GOD (1,000 U/g). Each group consisted of 6 replications with 8 birds per replicate. At day 7, all ducks were orally administered 0.2 mL E coli O88 (3 × 109 cfu/mL) twice, 8 h apart based on the preliminary experiment. The experiment lasted for 28 d. Dietary supplementation with GOD improved growth performance of ducks infected with E. coli. The GOD increased contents of Ig in plasma and secreted Ig A in jejunal mucosa. The GOD group had lower concentrations of inflammatory cytokines (tumor necrosis factor-α, IL-1ß, and IL-6) and their upstream regulator Toll-like receptor 4 in the jejunum of ducks than the control group. Supplementation with GOD increased villus height and decreased crypt depth in the jejunum. The gene expression of tight junction proteins (zonula occludens-1, claudin-1 and claudin-2) was enhanced by adding GOD. The GOD decreased intestinal permeability by reducing the concentrations of diamine oxidase and D-lactic in plasma of ducks. There were no significant differences in almost all the indices tested between the GOD and the antibiotic groups. In conclusion, supplementation of GOD improved growth performance, immune function, and intestinal barrier of ducks infected with E. coli O88. Glucose oxidase may serve as a promising alternative therapy to antibiotics to relieve or prevent colibacillosis in ducks.


Assuntos
Suplementos Nutricionais , Patos , Infecções por Escherichia coli , Glucose Oxidase , Imunidade , Mucosa Intestinal , Doenças das Aves Domésticas , Animais , Dieta/veterinária , Patos/crescimento & desenvolvimento , Patos/imunologia , Escherichia coli , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/terapia , Infecções por Escherichia coli/veterinária , Glucose Oxidase/administração & dosagem , Glucose Oxidase/farmacologia , Imunidade/efeitos dos fármacos , Mucosa Intestinal/enzimologia , Masculino , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/terapia , Distribuição Aleatória
20.
J Mater Chem B ; 8(43): 9943-9950, 2020 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-33034314

RESUMO

The current trend of cancer therapy has changed from monotherapy to synergistic or combination therapies. Among the treatment strategies, photodynamic therapy (PDT) and starvation therapy are widely employed together. However, the therapeutic effect of these treatments could lead to strong resistance and poor prognosis due to tumor hypoxia. Therefore, a smart nanoplatform (MONs-GOx@MnO2-Ce6) has been constructed herein by the assembly of glucose oxidase (GOx)-coated mesoporous organosilica nanoparticles (MONs) and MnO2 nanosheets-chlorin e6 (Ce6), which form a nanosystem. Once MONs-GOx@MnO2-Ce6 enter tumor cells, it catalyzes the oxidation of glucose using oxygen (O2) and generates hydrogen peroxide (H2O2) and gluconic acid, the former of which may accelerate the decomposition of MnO2 nanosheets. The released MnO2 nanosheets would regenerate O2 in the presence of H2O2. In this case, MnO2 nanosheets serve as (i) a nanocarrier and fluorescence quencher for the photosensitizer Ce6, (ii) a degradable material that is activated by the tumor microenvironment (TME) for fluorescence recovery, and (iii) an O2-producing carrier that reacts with H2O2 for relieving hypoxia in the tumor, which contributes to the combined starvation/photodynamic cancer therapy since these treatment strategies need O2. MONs-GOx@MnO2-Ce6 could not only realize cancer cell imaging, but also reduce intracellular glucose uptake and Glut1 expression, inhibiting the metabolism of cancer cells. This strategy shows great potential for clinical applications.


Assuntos
Glucose Oxidase/farmacologia , Compostos de Manganês/farmacologia , Neoplasias/tratamento farmacológico , Óxidos/farmacologia , Oxigênio/metabolismo , Fármacos Fotossensibilizantes/farmacologia , Porfirinas/farmacologia , Clorofilídeos , Glucose Oxidase/química , Células HeLa , Humanos , Compostos de Manganês/química , Microscopia Confocal , Nanopartículas/química , Nanoestruturas/química , Neoplasias/diagnóstico por imagem , Neoplasias/metabolismo , Compostos de Organossilício/química , Compostos de Organossilício/farmacologia , Óxidos/química , Fármacos Fotossensibilizantes/química , Porfirinas/química , Hipóxia Tumoral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA