Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 268
Filter
1.
J Colloid Interface Sci ; 666: 244-258, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38598997

ABSTRACT

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.


Subject(s)
Calcium Carbonate , Glucose Oxidase , Manganese Compounds , Oxides , Glucose Oxidase/metabolism , Glucose Oxidase/chemistry , Glucose Oxidase/pharmacology , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Oxides/chemistry , Oxides/pharmacology , Humans , Animals , Calcium Carbonate/chemistry , Calcium Carbonate/pharmacology , Calcium Carbonate/metabolism , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Nanoparticles/chemistry , Povidone/chemistry , Povidone/pharmacology , Tumor Hypoxia/drug effects , Reactive Oxygen Species/metabolism , Cell Survival/drug effects , Particle Size , Cell Line, Tumor , Hydrogen Peroxide/metabolism , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Surface Properties , Mice, Inbred BALB C
2.
Mater Horiz ; 11(10): 2406-2419, 2024 05 20.
Article in English | MEDLINE | ID: mdl-38440840

ABSTRACT

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.


Subject(s)
Ferroptosis , Immunotherapy , Infrared Rays , Nanoparticles , Polymers , Semiconductors , Ferroptosis/drug effects , Animals , Immunotherapy/methods , Mice , Polymers/chemistry , Polymers/therapeutic use , Female , Nanoparticles/therapeutic use , Nanoparticles/chemistry , Cell Line, Tumor , Tumor Microenvironment/drug effects , Glucose Oxidase/metabolism , Glucose Oxidase/pharmacology , Humans , Mice, Inbred BALB C , Hemoglobins/pharmacology , Hemoglobins/metabolism
3.
Colloids Surf B Biointerfaces ; 232: 113581, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37857184

ABSTRACT

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.


Subject(s)
Gelatin , Hydrogels , Animals , Hydrogels/pharmacology , Gelatin/pharmacology , Glucose Oxidase/pharmacology , Neoplasm Recurrence, Local/prevention & control , Hydrogen Peroxide/pharmacology , Neoplasm, Residual
4.
Acta Biomater ; 172: 441-453, 2023 12.
Article in English | MEDLINE | ID: mdl-37802309

ABSTRACT

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.


Subject(s)
Nanoparticles , Neoplasms , Humans , Biomimetics , Hydrogen Peroxide , Photothermal Therapy , Catalysis , Glucose , Glucose Oxidase/pharmacology , Horseradish Peroxidase , Cell Line, Tumor , Tumor Microenvironment , Nanoparticles/therapeutic use
5.
Acta Biomater ; 168: 580-592, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37451659

ABSTRACT

Healing bacterial chronic wounds caused by hyperglycemia is of great significance to protect the physical and mental health of diabetic patients. In this context, emerging chemodynamic therapy (CDT) and photothermal therapy (PTT) with broad antibacterial spectra and high spatiotemporal controllability have flourished. However, CDT was challenged by the near-neutral pH and inadequate H2O2 surrounding the chronic wound site, while PTT showed overheating-triggered side effects (e.g., damaging the normal tissue) and poor effects on thermotolerant bacterial biofilms. Therefore, we engineered an all-in-one glucose-responsive photothermal nanozyme, GOX/MPDA/Fe@CDs, consisting of glucose oxidase (GOX), Fe-doped carbon dots (Fe@CDs), and mesoporous polydopamine (MPDA), to efficiently treat chronic diabetic wound bacterial infections and eradicate biofilms without impacting the surrounding normal tissues. Specifically, GOX/MPDA/Fe@CDs produced a local temperature (∼ 45.0°C) to enhance the permeability of the pathogenic bacterium and its biofilm upon near-infrared (NIR) 808 nm laser irradiation, which was seized to initiate endogenous high blood glucose to activate the catalytic activity of GOX on the GOX/MPDA/Fe@CD surface to achieve the simultaneous self-supplying of H2O2 and H+, cascade catalyzing •OH production via a subsequent peroxidase-mimetic activity-induced Fenton/Fenton-like reaction. As such, the in vivo diabetic wound infected with methicillin-resistant Staphylococcus aureus was effectively healed after 12.0 days of treatment. This work was expected to provide an innovative approach to the clinical treatment of bacterially infected diabetic chronic wounds. STATEMENT OF SIGNIFICANCE: An all-in-one glucose-responsive photothermal nanozyme GOX/MPDA/Fe@CDs was constructed. Cascade nanozyme GOX/MPDA/Fe@CDs self-supply H2O2 and H+ to break H2O2 and pH limits to fight bacterial infections. Synergistic chemotherapy and photothermal therapy with nanozyme GOX/MPDA/Fe@CDs accelerates healing of biofilm-infected diabetic wounds.


Subject(s)
Diabetes Mellitus , Hyperglycemia , Methicillin-Resistant Staphylococcus aureus , Humans , Hydrogen Peroxide/pharmacology , Photothermal Therapy , Anti-Bacterial Agents/pharmacology , Carbon/pharmacology , Glucose , Glucose Oxidase/pharmacology , Nanotechnology
6.
Angew Chem Int Ed Engl ; 62(44): e202308761, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37496129

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Liposomes , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Glucose Oxidase/pharmacology , Horseradish Peroxidase , Neoplasms/drug therapy , Neoplasms/pathology , Nanotechnology , Hydrogen Peroxide/therapeutic use
7.
Adv Sci (Weinh) ; 10(21): e2301278, 2023 07.
Article in English | MEDLINE | ID: mdl-37114827

ABSTRACT

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.


Subject(s)
Breast Neoplasms , Hyperbaric Oxygenation , Radiation-Sensitizing Agents , Humans , Female , Copper , Oxygen , Breast Neoplasms/therapy , Glucose Oxidase/pharmacology , Glucose/metabolism , Tumor Microenvironment
8.
Acta Biomater ; 158: 252-265, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36584802

ABSTRACT

Antibiotic resistance of bacteria and persistent inflammation are critical challenges in treating bacteria infected wounds. Thus, it is urgent to develop versatile wound dressings that possess high-efficiency antibacterial performance and inflammation regulation. Herein, we have successfully constructed a hydrogel wound dressing consisting of the bimetallic metal-organic framework (MOF) loaded with glucose oxidase (GOx), termed as MOF(Fe-Cu)/GOx-polyacrylamide (PAM) gel. Hydrogel dressings can provide an efficient cascade-catalyzed system to accelerate wound healing via synergistic antibacterial and inflammatory modulation. Importantly, the catalytic property of the bimetallic MOF(Fe-Cu) is about five times that of the monometallic MOF(Fe). Based on such a cascade-catalyzed system, the abundant gluconic acid and H2O2 can be continuously produced by decomposing glucose via GOx. Such gluconic acid can notably improve the peroxidase performance of MOF(Fe-Cu), which can further efficiently decompose H2O2 to achieve the antibacterial. Meanwhile, MOF (Fe Cu)/GOx PAM gel can induce macrophages to change into an M2 phenotype, which can accelerate the transformation of the wound microenvironment to a remodeling state and then accelerate angiogenesis and neurogenesis. This work provides multifunctional bioactive materials for accelerating wound healing and will have great potential in clinical applications. STATEMENT OF SIGNIFICANCE: Antibiotic resistance and persistent inflammation are still the critical reasons for the slow healing of bacteria infected wounds. Herein, we prepared a hydrogel wound dressing composed of bimetallic metal organic framework (MOF) loaded with glucose oxidase (GOx). The catalytic activity of the bimetallic MOF(Fe-Cu) is significantly enhanced due to doping of copper, which makes it possess outstanding antibacterial ability based on cascade catalysis. Such dressing can promote the remodeling of inflammatory immunity by regulating macrophage polarization to suppress over-reactive inflammation, further accelerating the healing of bacteria-infected wounds. This study provides an innovative and effective way to accelerate the healing of bacteria infected wound by combining bacteria killing and inflammation modulation.


Subject(s)
Glucose Oxidase , Hydrogels , Humans , Glucose Oxidase/pharmacology , Hydrogels/pharmacology , Hydrogen Peroxide/pharmacology , Anti-Bacterial Agents/pharmacology , Bandages , Inflammation/drug therapy
9.
ACS Biomater Sci Eng ; 8(12): 5145-5154, 2022 Dec 12.
Article in English | MEDLINE | ID: mdl-36344935

ABSTRACT

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.


Subject(s)
Glucose Oxidase , Hydrogen Peroxide , Glucose Oxidase/pharmacology , Hydrogen Peroxide/pharmacology , Glucose , Wound Healing , Bacteria
10.
Nutrients ; 14(19)2022 Sep 22.
Article in English | MEDLINE | ID: mdl-36235593

ABSTRACT

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.


Subject(s)
Antioxidants , Proteasome Endopeptidase Complex , Annexin A5/metabolism , Annexin A5/pharmacology , Antioxidants/metabolism , Antioxidants/pharmacology , Apoptosis , Caspase 3/metabolism , Catalase/metabolism , DNA/metabolism , DNA-Binding Proteins/genetics , Epithelial Cells/metabolism , Glucose/metabolism , Glucose Oxidase/metabolism , Glucose Oxidase/pharmacology , Ku Autoantigen/metabolism , Oxidative Stress , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Proteolysis , Reactive Oxygen Species/metabolism , Ubiquitins/metabolism , Ubiquitins/pharmacology , Xanthophylls
11.
ACS Nano ; 16(8): 12118-12133, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35904186

ABSTRACT

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.


Subject(s)
Neoplasms , Ultrasonic Therapy , Humans , Platinum , Tumor Microenvironment , Glucose , Neoplasms/drug therapy , Neoplasms/pathology , Reactive Oxygen Species/metabolism , Glucose Oxidase/pharmacology , Glucose Oxidase/metabolism , Oxygen , Cell Line, Tumor
12.
Biomater Sci ; 10(15): 4170-4183, 2022 Jul 26.
Article in English | MEDLINE | ID: mdl-35726827

ABSTRACT

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.


Subject(s)
Glioma , Nanoparticles , Neoplasms , Animals , Cell Line, Tumor , Copper , Glioma/drug therapy , Glucose , Glucose Oxidase/pharmacology , Hydrogen Peroxide , Mice , Neoplasms/drug therapy , Reactive Oxygen Species
13.
Acta Biomater ; 145: 222-234, 2022 06.
Article in English | MEDLINE | ID: mdl-35460908

ABSTRACT

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.


Subject(s)
Glucose Oxidase , Metformin , Neoplasms , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Cell Line, Tumor , Drug Therapy/methods , Glucose , Glucose Oxidase/pharmacology , Glucose Oxidase/therapeutic use , Glycolysis/drug effects , Humans , Metformin/pharmacology , Metformin/therapeutic use , Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism
14.
ACS Biomater Sci Eng ; 8(4): 1522-1531, 2022 04 11.
Article in English | MEDLINE | ID: mdl-35274927

ABSTRACT

The emergence of peroxidase (POD)-like nanozyme-derived catalytic therapy has provided a promising choice for reactive oxygen species (ROS)-mediated broad-spectrum antibacterials to replace antibiotics, but it still suffers from limitations of low therapeutic efficiency and unusual addition of unstable H2O2. Considering that the higher blood glucose in diabetic wounds provides much more numerous nutrients for bacterial growth, a cascade nanoenzymatic active material was developed by coating glucose oxidase (GOx) onto POD-like Fe2(MoO4)3 [Fe2(MoO4)3@GOx]. GOx could consume the nutrient of glucose to produce gluconic acid (weakly acidic) and H2O2, which could be subsequently converted into highly oxidative •OH via the catalysis of POD-like Fe2(MoO4)3. Accordingly, the synergistic effect of starvation and ROS-mediated therapy showed significantly efficient antibacterial effect while avoiding the external addition of H2O2 that affects the stability and efficacy of the therapy system. Compared with the bactericidal rates of 46.2-59.404% of GOx or Fe2(MoO4)3 alone on extended-spectrum ß-lactamases producing Escherichia coli and methicillin-resistant Staphylococcus aureus, those of the Fe2(MoO4)3@GOx group are 98.396 and 98.776%, respectively. Animal experiments showed that the as-synthesized Fe2(MoO4)3@GOx could much efficiently promote the recovery of infected wounds in type 2 diabetic mice while showing low cytotoxicity in vivo.


Subject(s)
Diabetes Mellitus, Experimental , Methicillin-Resistant Staphylococcus aureus , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Escherichia coli , Glucose Oxidase/pharmacology , Glucose Oxidase/therapeutic use , Hydrogen Peroxide/pharmacology , Mice , Reactive Oxygen Species/pharmacology
15.
Biomed Res Int ; 2022: 4133562, 2022.
Article in English | MEDLINE | ID: mdl-35342759

ABSTRACT

Mandibular bone regeneration is still a big challenge in those diabetic patients with poorly controlled blood glucose. In this study, we prepared a novel glucose-sensitive controlled-release fiber scaffold (PVA-HTCC/PEO-rhBMP2-glucose oxidase (PHPB-G)), which contained the recombinant human bone morphogenetic protein 2 (rhBMP2) by coaxial cospinning and grafted with glucose oxidase (GOD). We presented evidence that PHPB-G could undergo a series of structural changes with the blood glucose and promoted bone regeneration in diabetic rat. PHPB-G expanded the voids in nanofibers when blood glucose levels elevated. More importantly, its slow-release rhBMP2 effectively promoted the healing of bone defects. These data suggested that the PHPB-G delivery system may provide a potential treatment strategy for patients with severe diabetic alveolar bone defects.


Subject(s)
Diabetes Mellitus , Osteogenesis , Animals , Blood Glucose , Bone Morphogenetic Protein 2/chemistry , Bone Morphogenetic Protein 2/pharmacology , Bone Regeneration , Glucose/pharmacology , Glucose Oxidase/pharmacology , Humans , Rats , Recombinant Proteins/pharmacology , Tissue Scaffolds/chemistry
16.
Biomolecules ; 12(3)2022 03 19.
Article in English | MEDLINE | ID: mdl-35327664

ABSTRACT

Glucose oxidase (GOx) is an important oxidoreductase enzyme with many important roles in biological processes. It is considered an "ideal enzyme" and is often called an oxidase "Ferrari" because of its fast mechanism of action, high stability and specificity. Glucose oxidase catalyzes the oxidation of ß-d-glucose to d-glucono-δ-lactone and hydrogen peroxide in the presence of molecular oxygen. d-glucono-δ-lactone is sequentially hydrolyzed by lactonase to d-gluconic acid, and the resulting hydrogen peroxide is hydrolyzed by catalase to oxygen and water. GOx is presently known to be produced only by fungi and insects. The current main industrial producers of glucose oxidase are Aspergillus and Penicillium. An important property of GOx is its antimicrobial effect against various pathogens and its use in many industrial and medical areas. The aim of this review is to summarize the structure, function, production strains and biophysical and biochemical properties of GOx in light of its various industrial, biotechnological and medical applications.


Subject(s)
Glucose Oxidase , Hydrogen Peroxide , Biotechnology , Glucose , Glucose Oxidase/chemistry , Glucose Oxidase/pharmacology , Hydrogen Peroxide/chemistry , Oxygen
17.
Animal ; 16(3): 100473, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35218993

ABSTRACT

Glucose oxidase (GOD) is an aerobic dehydrogenase, which catalyses the oxidation of ß-D-glucose to gluconic acid and hydrogen peroxide. This study aimed to investigate the effects of dietary glucose oxidase and its combined effects with Bacillus amyloliquefaciens SC06 (BaSC06) on the intestinal microbiota, immune function and antioxidant capacity of broilers. One-day-old male Lingnan yellow-feathered broilers (n = 720) were randomly assigned to four treatment groups: Control group (basal diet), Anti group (basal diet supplemented with 200 mg/kg enramycin), GOD group (basal diet supplemented with 75 U/kg GOD), and combination of GOD and BaSC06 (GB) group (GOD diet (75 U/kg) supplemented with 1 × 108 colony-forming units BaSC06/kg feed), with six replicates per group and 30 birds per replicate. The experiment was conducted over 52 days. The results indicated a significant decrease in α-diversity (Observed species, Chao1, PD_whole_tree and Shannon) with GOD treatment, compared with the control group. GB treatment also significantly decreased the Shannon index of cecal microbiota. GOD treatment significantly decreased the α-diversity, whereas GB treatment significantly increased these indices except for the Chao1 index, compared with the Anti group. Compared with the control group, the relative abundance of Bacteroides in the GOD and GB groups was significantly increased, whereas a decrease in Firmicutes was observed. Compared with the Anti group, GOD treatment significantly increased the relative abundances of Bacteroides and Lactobacillales, while GB treatment significantly increased Lactobacillales and decreased Proteobacteria levels. In addition, GOD treatment significantly decreased interleukin-10 and interferon-γ levels, compared with the control group. In contrast, GB treatment significantly downregulated interferon-γ levels and upregulated secretory immunoglobulin A, transforming growth factor-ß and interleukin-2 expression in the jejunal mucosa. GOD treatment significantly decreased transforming growth factor-ß and interleukin-10 levels, whereas GB treatment markedly increased interferon-γ expression in the jejunal mucosa compared with the Anti group. Furthermore, GB treatment significantly increased the total antioxidant capability levels and the total superoxide dismutase (T-SOD) and catalase (CAT) activities compared with the control group. Meanwhile, GOD treatment significantly increased glutathione peroxidase (GSH-Px) activity in the jejunal mucosa. Total superoxide dismutase, GSH-Px and CAT activities in the Anti group were higher than in the GOD and GB groups. The malondialdehyde levels in the control group were the highest among all groups. In conclusion, our results indicated that supplementation with GOD alone and its combination with BaSC06 in diet could increase antioxidant capacity, immune function and improve the intestinal microbiota composition of broilers. Combination treatment with GOD with BaSC06 exerted stronger effects than GOD treatment only.


Subject(s)
Gastrointestinal Microbiome , Animal Feed/analysis , Animals , Antioxidants/pharmacology , Chickens/physiology , Diet/veterinary , Dietary Supplements/analysis , Glucose Oxidase/metabolism , Glucose Oxidase/pharmacology , Immunity , Male
18.
Adv Sci (Weinh) ; 9(6): e2104576, 2022 02.
Article in English | MEDLINE | ID: mdl-34989177

ABSTRACT

Increasing antibiotic resistance becomes a serious threat to public health. Photothermal therapy (PTT) and antibacterial enzyme-based therapy are promising nonresistant strategies for efficiently killing drug-resistant bacteria. However, the poor thermostability of enzymes in PTT hinders their synergistic therapy. Herein, antibacterial glucose oxidase (GOx) is embedded in a Ag graphitic nanocapsule (Ag@G) arrayed silk film to fabricate a GOx-synergistic PTT system (named silk-GOx-Ag@G, SGA). The SGA system can stabilize GOx by a vitrification process through the restriction of hydrogen bond and rigid ß-sheet, and keep the antibacterial activity in the hyperthermal PTT environment. Moreover, the arrayed Ag@G possesses excellent chemical stability due to the protection of graphitic shell, providing stable plasmonic effect for integrating PTT and surface enhanced Raman scattering (SERS) analysis even in the GOx-produced H2 O2 environment. With in situ SERS identification of bacterial intrinsic signals in the mouse wound model, such SGA realizes superior synergistic antibacterial effect on the infected Escherichia coli, Staphylococcus aureus, and methicillin-resistant Staphylococcus aureus in vivo, while without causing significant biotoxicity. This system provides a therapeutic method with low resistance and in situ diagnosis capability for efficiently eliminating bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Glucose Oxidase/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Silk/pharmacology , Silver/pharmacology , Wound Infection/drug therapy , Animals , Disease Models, Animal , Female , Metal Nanoparticles/chemistry , Mice , Mice, Inbred BALB C , Spectrum Analysis, Raman/methods
19.
Acta Biomater ; 141: 429-439, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35038584

ABSTRACT

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.


Subject(s)
Breast Neoplasms , Nanoparticles , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Cell Line, Tumor , Female , Glucose , Glucose Oxidase/pharmacology , Humans , Ions , Manganese , Nanoparticles/therapeutic use , Paclitaxel/pharmacology , Phospholipids , Reactive Oxygen Species/metabolism
20.
Microbiol Spectr ; 10(1): e0175721, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35019769

ABSTRACT

Mouthwash is a commonly used product and has been proposed as an alternative intervention to prevent gonorrhea transmission. However, the long-term effects of mouthwash on the oral microbiota are largely unknown. We investigated the impact of 12 weeks of daily mouthwash use on the oropharyngeal microbiota in a subset of men who have sex with men who participated in a randomized trial comparing the efficacy of two alcohol-free mouthwashes for the prevention of gonorrhea. We characterized the oropharyngeal microbiota using 16S rRNA gene sequencing of tonsillar fossae samples collected before and after 12 weeks of daily use of Listerine mouthwash or Biotène dry mouth oral rinse. Permutational multivariate analysis of variance (PERMANOVA) was used to assess differences in oropharyngeal microbiota composition following mouthwash use. Differential abundance testing was performed using ALDEx2, with false-discovery rate correction. A total of 306 samples from 153 men were analyzed (Listerine, n = 78 and Biotène, n = 75). There was no difference in the overall structure of the oropharyngeal microbiota following Listerine or Biotène use (PERMANOVA P = 0.413 and P = 0.331, respectively). Although no bacterial taxa were significantly differentially abundant following Listerine use, we observed a small but significant decrease in the abundance of both Streptococcus and Leptotrichia following Biotène use. Overall, our findings suggest that daily use of antiseptic mouthwash has minimal long-term effects on the composition of the oropharyngeal microbiota. IMPORTANCE Given the role of the oral microbiota in human health, it is important to understand if and how external factors influence its composition. Mouthwash use is common in some populations, and the use of antiseptic mouthwash has been proposed as an alternative intervention to prevent gonorrhea transmission. However, the long-term effect of mouthwash use on the oral microbiota composition is largely unknown. We found that daily use of two different commercially available mouthwashes had limited long-term effects on the composition of the oropharyngeal microbiota over a 12-week period. The results from our study and prior studies highlight that different mouthwashes may differentially affect the oral microbiome composition and that further studies are needed to determine if mouthwash use induces short-term changes to the oral microbiota that may have detrimental effects.


Subject(s)
Homosexuality, Male , Microbiota/drug effects , Mouthwashes/pharmacology , Sexual and Gender Minorities , Adult , Double-Blind Method , Drug Combinations , Glucose Oxidase/pharmacology , Gonorrhea , Humans , Lactoperoxidase/pharmacology , Male , Microbiota/genetics , Muramidase/pharmacology , Oropharynx/microbiology , RNA, Ribosomal, 16S , Salicylates , Terpenes , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...