Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
Add more filters











Publication year range
1.
Adv Mater ; : e2403921, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39101290

ABSTRACT

Radiotherapy (RT), essential for treating various cancers, faces challenges from tumor hypoxia, which induces radioresistance. A tumor-targeted "prosthetic-Arginine" coassembled nanozyme system, engineered to catalytically generate nitric oxide (NO) and oxygen (O2) in the tumor microenvironment (TME), overcoming hypoxia and enhancing radiosensitivity is presented. This system integrates the prosthetic heme of nitric oxide synthase (NOS) and catalase (CAT) with NO-donating Fmoc-protected Arginine and Ru3+ ions, creating HRRu nanozymes that merge NOS and CAT functionalities. Surface modification with human heavy chain ferritin (HFn) improves the targeting ability of nanozymes (HRRu-HFn) to tumor tissues. In the TME, strategic arginine incorporation within the nanozyme allows autonomous O2 and NO release, triggered by endogenous hydrogen peroxide, elevating NO and O2 levels to normalize vasculature and improve blood perfusion, thus mitigating hypoxia. Employing the intrinsic O2-transporting ability of heme, HRRu-HFn nanozymes also deliver O2 directly to the tumor site. Utilizing esophageal squamous cell carcinoma as a tumor model, the studies reveal that the synergistic functions of NO and O2 production, alongside targeted delivery, enable the HRRu-HFn nanozymes to combat tumor hypoxia and potentiate radiotherapy. This HRRu-HFn nanozyme based approach holds the potential to reduce the radiation dose required and minimize side effects associated with conventional radiotherapy.

2.
J Agric Food Chem ; 72(26): 14967-14974, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38957086

ABSTRACT

Nanobodies (Nbs) serve as powerful tools in immunoassays. However, their small size and monovalent properties pose challenges for practical application. Multimerization emerges as a significant strategy to address these limitations, enhancing the utilization of nanobodies in immunoassays. Herein, we report the construction of a Salmonella-specific fenobody (Fb) through the fusion of a nanobody to ferritin, resulting in a self-assembled 24-valent nanocage-like structure. The fenobody exhibits a 35-fold increase in avidity compared to the conventional nanobody while retaining good thermostability and specificity. Leveraging this advancement, three ELISA modes were designed using Fb as the capture antibody, along with unmodified Nb422 (FbNb-ELISA), biotinylated Nb422 (FbBio-ELISA), and phage-displayed Nb422 (FbP-ELISA) as the detection antibody, respectively. Notably, the FbNb-ELISA demonstrates a detection limit (LOD) of 3.56 × 104 CFU/mL, which is 16-fold lower than that of FbBio-ELISA and similar to FbP-ELISA. Moreover, a fenobody and nanobody sandwich chemiluminescent enzyme immunoassay (FbNb-CLISA) was developed by replacing the TMB chromogenic substrate with luminal, resulting in a 12-fold reduction in the LOD. Overall, the ferritin-displayed technology represents a promising methodology for enhancing the detection performance of nanobody-based sandwich ELISAs, thereby expanding the applicability of Nbs in food detection and other fields requiring multivalent modification.


Subject(s)
Enzyme-Linked Immunosorbent Assay , Ferritins , Salmonella , Single-Domain Antibodies , Ferritins/immunology , Ferritins/chemistry , Ferritins/genetics , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology , Salmonella/immunology , Salmonella/genetics , Enzyme-Linked Immunosorbent Assay/methods , Limit of Detection , Antibody Affinity , Antibodies, Bacterial/immunology , Immunoassay/methods
3.
Int J Nanomedicine ; 19: 5793-5812, 2024.
Article in English | MEDLINE | ID: mdl-38882535

ABSTRACT

This review article discusses the potential of nanomaterials in targeted therapy and immunomodulation for stroke-induced immunosuppression. Although nanomaterials have been extensively studied in various biomedical applications, their specific use in studying and addressing immunosuppression after stroke remains limited. Stroke-induced neuroinflammation is characterized by T-cell-mediated immunodepression, which leads to increased morbidity and mortality. Key observations related to immunodepression after stroke, including lymphopenia, T-cell dysfunction, regulatory T-cell imbalance, and cytokine dysregulation, are discussed. Nanomaterials, such as liposomes, micelles, polymeric nanoparticles, and dendrimers, offer advantages in the precise delivery of drugs to T cells, enabling enhanced targeting and controlled release of immunomodulatory agents. These nanomaterials have the potential to modulate T-cell function, promote neuroregeneration, and restore immune responses, providing new avenues for stroke treatment. However, challenges related to biocompatibility, stability, scalability, and clinical translation need to be addressed. Future research efforts should focus on comprehensive studies to validate the efficacy and safety of nanomaterial-based interventions targeting T cells in stroke-induced immunosuppression. Collaborative interdisciplinary approaches are necessary to advance the field and translate these innovative strategies into clinical practice, ultimately improving stroke outcomes and patient care.


Subject(s)
Nanostructures , Stroke , T-Lymphocytes , Animals , Humans , Cytokines/metabolism , Cytokines/immunology , Nanomedicine , Nanoparticles/chemistry , Nanostructures/chemistry , Stroke/immunology , T-Lymphocytes/immunology , T-Lymphocytes/drug effects
4.
J Colloid Interface Sci ; 667: 529-542, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38653074

ABSTRACT

Diabetic wounds are characterized by chronic trauma, with long-term non-healing attributed to persistent inflammation and recurrent bacterial infections. Exacerbation of the inflammatory response is largely due to increased levels of reactive oxygen species (ROS). In this study, catalase (CAT) was used as a biological template to synthesize nanozyme-supported natural enzymes (CAT-Mn(SH)x) using a biomimetic mineralization method. Subsequently, polymyxin B (CAT-Mn(SH)x@PMB) was immobilized on its surface through electrostatic assembly. CAT-Mn(SH)x@PMB demonstrates the ability for slow and sustained release of hydrogen sulfide (H2S). Finally, CAT-Mn(SH)x@PMB loaded microneedles (MNs) substrate were synthesized using polyvinyl alcohol (PVA) and hydroxyethyl methacrylate (HEMA), and named CAT-(MnSH)x@PMB-MNs. It exhibited enhanced enzyme and antioxidant activities, along with effective antibacterial properties. Validation findings indicate that it can up-regulate the level of M2 macrophages and reduce the level of pro-inflammatory cytokine tumor necrosis factor-α (TNF-α). Additionally, it promotes angiogenesis and rapid nerve regeneration, thereby facilitating wound healing through its dual anti-inflammatory and antibacterial effects. Hence,this study introduces a time-space tissue-penetrating and soluble microneedle patch with dual anti-inflammatory and antibacterial effects for the treatment of diabetic wounds.


Subject(s)
Anti-Bacterial Agents , Catalase , Needles , Polymyxin B , Wound Healing , Polymyxin B/pharmacology , Polymyxin B/chemistry , Polymyxin B/administration & dosage , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/administration & dosage , Animals , Catalase/metabolism , Catalase/chemistry , Wound Healing/drug effects , Mice , Escherichia coli/drug effects , Diabetes Mellitus, Experimental/drug therapy , Rats , RAW 264.7 Cells , Microbial Sensitivity Tests , Particle Size
5.
ACS Nano ; 18(17): 11217-11233, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38627234

ABSTRACT

Due to its intrinsic tumor-targeting attribute, limited immunogenicity, and cage architecture, ferritin emerges as a highly promising nanocarrier for targeted drug delivery. In the effort to develop ferritin cage-encapsulated cisplatin (CDDP) as a therapeutic agent, we found unexpectedly that the encapsulation led to inactivation of the drug. Guided by the structural information, we deciphered the interactions between ferritin cages and CDDP, and we proposed a potential mechanism responsible for attenuating the antitumor efficacy of CDDP encapsulated within the cage. Six platinum prodrugs were then designed to avoid the inactivation. The antitumor activities of these ferritin-platinum prodrug complexes were then evaluated in cells of esophageal squamous cell carcinoma (ESCC). Compared with free CDDP, the complexes were more effective in delivering and retaining platinum in the cells, leading to increased DNA damage and enhanced cytotoxic action. They also exhibited improved pharmacokinetics and stronger antitumor activities in mice bearing ESCC cell-derived xenografts as well as patient-derived xenografts. The successful encapsulation also illustrates the critical significance of comprehending the interactions between small molecular drugs and ferritin cages for the development of precision-engineered nanocarriers.


Subject(s)
Antineoplastic Agents , Cisplatin , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Ferritins , Prodrugs , Prodrugs/chemistry , Prodrugs/pharmacology , Humans , Ferritins/chemistry , Ferritins/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Esophageal Neoplasms/metabolism , Mice , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Cisplatin/pharmacology , Cisplatin/chemistry , Drug Design , Platinum/chemistry , Platinum/pharmacology , Mice, Nude , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Drug Delivery Systems
6.
Adv Healthc Mater ; 13(16): e2303548, 2024 06.
Article in English | MEDLINE | ID: mdl-38507709

ABSTRACT

Diabetic wounds are susceptible to bacterial infections, largely linked to high blood glucose levels (hyperglycemia). To treat such wounds, enzymes like glucose oxidase (GOx) can be combined with nanozymes (nanomaterials mimic enzymes) to use glucose effectively for purposes. However, there is still room for improvement in these systems, particularly in terms of process simplification, enzyme activity regulation, and treatment effects. Herein, the approach utilizes GOx to directly facilitate the biomineralized growth of osmium (Os) nanozyme (GOx-OsNCs), leading to dual-active centers and remarkable triple enzyme activities. Initially, GOx-OsNCs use vicinal dual-active centers, enabling a self-cascaded mechanism that significantly enhances glucose sensing performance compared to step-by-step reactions, surpassing the capabilities of other metal sources such as gold and platinum. In addition, GOx-OsNCs are integrated into a glucose-sensing gel, enabling instantaneous visual feedback. In the treatment of infected diabetic wounds, GOx-OsNCs exhibit multifaceted benefits by lowering blood glucose levels and exhibiting antibacterial properties through the generation of hydroxyl free radicals, thereby expediting healing by fostering a favorable microenvironment. Furthermore, the catalase-like activity of GOx-OsNCs aids in reducing oxidative stress, inflammation, and hypoxia, culminating in improved healing outcomes. Overall, this synergistic enzyme-nanozyme blend is user-friendly and holds considerable promise for diverse applications.


Subject(s)
Glucose Oxidase , Osmium , Glucose Oxidase/chemistry , Glucose Oxidase/metabolism , Animals , Osmium/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Wound Healing/drug effects , Mice , Blood Glucose/metabolism , Diabetes Mellitus, Experimental , Humans , Glucose/metabolism , Wound Infection/drug therapy , Wound Infection/metabolism
7.
Adv Sci (Weinh) ; 11(18): e2303901, 2024 May.
Article in English | MEDLINE | ID: mdl-38445847

ABSTRACT

Oxidative stress induced by excess reactive oxygen species (ROS) is a primary pathogenic cause of acute kidney injury (AKI). Development of an effective antioxidation system to mitigate oxidative stress for alleviating AKI remains to be investigated. This study presents the synthesis of an ultra-small Platinum (Pt) sulfur cluster (Pt5.65S), which functions as a pH-activatable prefabricated nanozyme (pre-nanozyme). This pre-nanozyme releases hydrogen sulfide (H2S) and transforms into a nanozyme (Ptzyme) that mimics various antioxidant enzymes, including superoxide dismutase and catalase, within the inflammatory microenvironment. Notably, the Pt5.65S pre-nanozyme exhibits an endo-exogenous synergy-enhanced antioxidant therapeutic mechanism. The Ptzyme reduces oxidative damage and inflammation, while the released H2S gas promotes proneurogenesis by activating Nrf2 and upregulating the expression of antioxidant molecules and enzymes. Consequently, the Pt5.65S pre-nanozyme shows cytoprotective effects against ROS/reactive nitrogen species (RNS)-mediated damage at remarkably low doses, significantly improving treatment efficacy in mouse models of kidney ischemia-reperfusion injury and cisplatin-induced AKI. Based on these findings, the H2S-generating pre-nanozyme may represent a promising therapeutic strategy for mitigating inflammatory diseases such as AKI and others.


Subject(s)
Acute Kidney Injury , Disease Models, Animal , Hydrogen Sulfide , Oxidative Stress , Acute Kidney Injury/metabolism , Acute Kidney Injury/drug therapy , Animals , Oxidative Stress/drug effects , Mice , Hydrogen Sulfide/metabolism , Hydrogen-Ion Concentration , Antioxidants/metabolism , Antioxidants/pharmacology , Reactive Oxygen Species/metabolism , Male , Mice, Inbred C57BL
8.
Nat Commun ; 15(1): 1626, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38388471

ABSTRACT

Developing strategies that emulate the killing mechanism of neutrophils, which involves the enzymatic cascade of superoxide dismutase (SOD) and myeloperoxidase (MPO), shows potential as a viable approach for cancer therapy. Nonetheless, utilizing natural enzymes as therapeutics is hindered by various challenges. While nanozymes have emerged for cancer treatment, developing SOD-MPO cascade in one nanozyme remains a challenge. Here, we develop nanozymes possessing both SOD- and MPO-like activities through alloying Au and Pd, which exhibits the highest cascade activity when the ratio of Au and Pd is 1:3, attributing to the high d-band center and adsorption energy for superoxide anions, as determined through theoretical calculations. The Au1Pd3 alloy nanozymes exhibit excellent tumor therapeutic performance and safety in female tumor-bearing mice, with safety attributed to their tumor-specific killing ability and renal clearance ability caused by ultrasmall size. Together, this work develops ultrasmall AuPd alloy nanozymes that mimic neutrophil enzymatic cascades for catalytic treatment of tumors.


Subject(s)
Nanostructures , Neoplasms , Female , Animals , Mice , Neutrophils , Catalysis , Superoxide Dismutase , Neoplasms/drug therapy
9.
ACS Nano ; 18(4): 2533-2540, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38215476

ABSTRACT

Nanozymes, nanomaterials exhibiting enzyme-like activities, have emerged as a prominent interdisciplinary field over the past decade. To date, over 1200 different nanomaterials have been identified as nanozymes, covering four catalytic categories: oxidoreductases, hydrolases, isomerases, and lyases. Catalytic activity and specificity are two pivotal benchmarks for evaluating enzymatic performance. Despite substantial progress being made in quantifying and optimizing the catalytic activity of nanozymes, there is still a lack of in-depth research on the catalytic specificity of nanozymes, preventing the formation of consensual knowledge and impeding a more refined and systematic classification of nanozymes. Recently, debates have emerged regarding whether nanozymes could possess catalytic specificity similar to that of enzymes. This Perspective discusses the specificity of nanozymes by referring to the catalytic specificity of enzymes, highlights the specificity gap between nanozymes and enzymes, and concludes by offering our perspective on future research on the specificity of nanozymes.


Subject(s)
Nanostructures , Catalysis
10.
Nat Commun ; 15(1): 233, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38172125

ABSTRACT

Biominerals, the inorganic minerals of organisms, are known mainly for their physical property-related functions in modern living organisms. Our recent discovery of the enzyme-like activities of nanomaterials, coined as nanozyme, inspires the hypothesis that nano-biominerals might function as enzyme-like catalyzers in cells. Here we report that the iron cores of biogenic ferritins act as natural nanozymes to scavenge superoxide radicals. Through analyzing eighteen representative ferritins from three living kingdoms, we find that the iron core of prokaryote ferritin possesses higher superoxide-diminishing activity than that of eukaryotes. Further investigation reveals that the differences in catalytic capability result from the iron/phosphate ratio changes in the iron core, which is mainly determined by the structures of ferritins. The phosphate in the iron core switches the iron core from single crystalline to amorphous iron phosphate-like structure, resulting in decreased affinity to the hydrogen proton of the ferrihydrite-like core that facilitates its reaction with superoxide in a manner different from that of ferric ions. Furthermore, overexpression of ferritins with high superoxide-diminishing activities in E. coli increases the resistance to superoxide, whereas bacterioferritin knockout or human ferritin knock-in diminishes free radical tolerance, highlighting the physiological antioxidant role of this type of nanozymes.


Subject(s)
Escherichia coli , Superoxides , Humans , Escherichia coli/genetics , Escherichia coli/metabolism , Ferritins/chemistry , Iron/metabolism , Phosphates
11.
Adv Mater ; 36(10): e2210144, 2024 Mar.
Article in English | MEDLINE | ID: mdl-36730098

ABSTRACT

Ischemic stroke (IS) is one of the most common causes of disability and death. Thrombolysis and neuroprotection are two current major therapeutic strategies to overcome ischemic and reperfusion damage. In this work, a novel peptide-templated manganese dioxide nanozyme (PNzyme/MnO2 ) is designed that integrates the thrombolytic activity of functional peptides with the reactive oxygen species scavenging ability of nanozymes. Through self-assembled polypeptides that contain multiple functional motifs, the novel peptide-templated nanozyme is able to bind fibrin in the thrombus, cross the blood-brain barrier, and finally accumulate in the ischemic neuronal tissues, where the thrombolytic motif is "switched-on" by the action of thrombin. In mice and rat IS models, the PNzyme/MnO2 prolongs the blood-circulation time and exhibits strong thrombolytic action, and reduces the ischemic damages in brain tissues. Moreover, this peptide-templated nanozyme also effectively inhibits the activation of astrocytes and the secretion of proinflammatory cytokines. These data indicate that the rationally designed PNzyme/MnO2 nanozyme exerts both thrombolytic and neuroprotective actions. Giving its long half-life in the blood and ability to target brain thrombi, the biocompatible nanozyme may serve as a novel therapeutic agent to improve the efficacy and prevent secondary thrombosis during the treatment of IS.


Subject(s)
Ischemic Stroke , Neuroprotective Agents , Stroke , Rats , Mice , Animals , Manganese Compounds/pharmacology , Thrombin , Neuroprotection , Oxides , Fibrinolytic Agents/therapeutic use , Ischemia , Peptides/pharmacology , Peptides/therapeutic use , Stroke/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
12.
Adv Mater ; 36(8): e2307337, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37724878

ABSTRACT

Nanozymes, next-generation enzyme-mimicking nanomaterials, have entered an era of rational design; among them, Co-based nanozymes have emerged as captivating players over times. Co-based nanozymes have been developed and have garnered significant attention over the past five years. Their extraordinary properties, including regulatable enzymatic activity, stability, and multifunctionality stemming from magnetic properties, photothermal conversion effects, cavitation effects, and relaxation efficiency, have made Co-based nanozymes a rising star. This review presents the first comprehensive profiling of the Co-based nanozymes in the chemistry, biology, and environmental sciences. The review begins by scrutinizing the various synthetic methods employed for Co-based nanozyme fabrication, such as template and sol-gel methods, highlighting their distinctive merits from a chemical standpoint. Furthermore, a detailed exploration of their wide-ranging applications in biosensing and biomedical therapeutics, as well as their contributions to environmental monitoring and remediation is provided. Notably, drawing inspiration from state-of-the-art techniques such as omics, a comprehensive analysis of Co-based nanozymes is undertaken, employing analogous statistical methodologies to provide valuable guidance. To conclude, a comprehensive outlook on the challenges and prospects for Co-based nanozymes is presented, spanning from microscopic physicochemical mechanisms to macroscopic clinical translational applications.


Subject(s)
Environmental Science , Nanostructures , Catalysis , Nanostructures/chemistry
13.
Adv Sci (Weinh) ; 11(3): e2305217, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38029345

ABSTRACT

Hydrogels are prevailing drug delivery depots to improve antitumor efficacy and reduce systemic toxicity. However, the application of conventional free drug-loaded hydrogel is hindered by poor drug penetration in solid tumors. Here, an injectable ferritin-based nanocomposite hydrogel is constructed to facilitate tumor penetration and improve cancer chemoimmunotherapy. Specifically, doxorubicin-loaded human ferritin (Dox@HFn) and oxidized dextran (Dex-CHO) are used to construct the injectable hydrogel (Dox@HFn Gel) through the formation of pH-sensitive Schiff-base bonds. After peritumoral injection, the Dox@HFn Gel is retained locally for up to three weeks, and released intact Dox@HFn gradually, which can not only facilitate tumor penetration through active transcytosis but also induce immunogenic cell death (ICD) to tumor cells to generate an antitumor immune response. Combining with anti-programmed death-1 antibody (αPD-1), Dox@HFn Gel induces remarkable regression of orthotopic 4T1 breast tumors, further elicits a strong systemic anti-tumor immune response to effectively suppress tumor recurrence and lung metastasis of 4T1 tumors after surgical resection. Besides, the combination of Dox@HFn GelL with anti-CD47 antibody (αCD47) inhibits postsurgical tumor recurrence of aggressive orthotopic glioblastoma tumor model and significantly extends mice survival. This work sheds light on the construction of local hydrogels to potentiate antitumor immune response for improved cancer therapy.


Subject(s)
Ferritins , Neoplasm Recurrence, Local , Humans , Mice , Animals , Nanogels , Neoplasm Recurrence, Local/drug therapy , Doxorubicin/pharmacology , Hydrogels/chemistry
14.
Adv Mater ; 36(7): e2310033, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37994246

ABSTRACT

Single-atom nanozymes (SANzymes) emerge as promising alternatives to conventional enzymes. However, chemical instability limits their application. Here, a systematic synthesis of highly active and stable SANzymes is presented by leveraging noble metal-porphyrins. Four noble metal-porphyrins are successfully synthesized to mimic the active site of natural peroxidases through atomic metal-N coordination anchored to the porphyrin center. These noble metal-porphyrins are integrated into a stable and biocompatible Zr-based metal-organic framework (MxP, x denoting Ir, Ru, Pt, and Pd). Among these, MIrP demonstrates superior peroxidase-like activity (685.61 U mg-1 ), catalytic efficiency, and selectivity compared to horseradish peroxidase (267.71 U mg-1 ). Mechanistic investigations unveil heightened catalytic activity of MIrP arises from its robust H2 O2 adsorption capacity, unique rate-determining step, and low energy threshold. Crucially, MIrP exhibits remarkable chemical stability under both room temperature and high H2 O2 concentrations. Further, through modification with (-)-Epigallocatechin-3-Gallate, a natural ligand for Epstein-Barr virus (EBV)-encoded latent membrane protein 1, targeted SANzyme (MIrPHE) tailored for EBV-associated nasopharyngeal carcinoma is engineered. This study not only presents an innovative strategy for augmenting the catalytic activity and chemical stability of SANzymes but also highlights the substantial potential of MIrP as a potent nanomedicine for targeted catalytic tumor therapy.


Subject(s)
Epstein-Barr Virus Infections , Nasopharyngeal Neoplasms , Humans , Nasopharyngeal Carcinoma , Herpesvirus 4, Human , Engineering , Catalysis , Metals
15.
Adv Healthc Mater ; 13(10): e2303454, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38031989

ABSTRACT

Neuroinflammation is associated with a series of pathological symptoms in Parkinson's disease (PD), including α-synuclein aggregation and dopaminergic neuronal death. The NOD-like receptor protein 3 (NLRP3) inflammasome plays a crucial role in neuroinflammation at the lesion site and is a promising target for PD treatment. In this study, a nanoscale metal-organic framework (Zr-FeP MOF) based nanozyme is fabricated using Fe-5,10,15,20-tetra (4-carboxyphenyl) porphyrin (Fe-TCPP) and Zr6 cluster as ligands. The Zr-FeP MOF is subsequently encapsulated with mannitol (Man)-liposome, resulting in the formation of Zr-FeP MOF@Man liposome (MOF@Man Liposome) nanozyme system. The in vitro studies show that this nanozyme system is effective in relieving the formation of NLRP3 inflammasome and mitochondrial dysfunction. In mouse models of PD, the nanozyme system demonstrates a significant blood-brain barrier-crossing capability attributed to the Man-mediated brain targeting. Additionally, transcriptomic and biochemical studies show that the nanozyme system effectively inhibits the formation and assembly of inflammasome components, mitigating the activation of glial cells and neuroinflammatory response, and ultimately regulating the pathological symptoms of PD effectively.


Subject(s)
Metal-Organic Frameworks , Parkinson Disease , Humans , Mice , Animals , Inflammasomes/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Proteins/metabolism , Neuroinflammatory Diseases , Liposomes , Microglia , Mice, Inbred C57BL
16.
Adv Mater ; 36(10): e2300387, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37086206

ABSTRACT

Nanozymes have great potential to be used as an alternative to natural enzymes in a variety of fields. However, low catalytic activity compared with natural enzymes limits their practical use. It is still challenging to design nanozymes comparable to their natural counterparts in terms of the specific activity. In this study, a surface engineering strategy is employed to improve the specific activity of Ru nanozymes using charge-transferrable ligands such as polystyrene sulfonate (PSS). PSS-modified Ru nanozyme exhibits a peroxidase-like specific activity of up to 2820 U mg-1 , which is twice that of horseradish peroxidase (1305 U mg-1 ). Mechanism studies suggest that PSS readily accepts negative charge from Ru, thus reducing the affinity between Ru and ·OH. Importantly, the modified Ru-peroxidase nanozyme is successfully used to develop an immunoassay for human alpha-fetoprotein and achieves a 140-fold increase in detection sensitivity compared with traditional horseradish-peroxidase-based enzyme-linked immunosorbent assay. Therefore, this work provides a feasible route to design nanozymes with high specific activity that meets the practical use as an alternative to natural enzymes.


Subject(s)
Ruthenium , Humans , Horseradish Peroxidase , Ligands , Peroxidase , Peroxidases , Immunoassay
17.
Adv Sci (Weinh) ; 11(6): e2307844, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38054654

ABSTRACT

Iron single-atom nanozymes represent a promising artificial enzyme with superior activity owing to uniform active sites that can precisely mimic active center of nature enzymes. However, current synthetic strategies are hard to guarantee each active site at single-atom state. In this work, an erythrocyte-templated strategy by utilizing intrinsic hemin active center of hemoglobin as sing-atom source for nanozyme formation is developed. By combining cell fixation, porous salinization, and high-temperature carbonization, erythrocytes are successfully served as uniform templates to synthesize nanozymes with fully single-atom FeN4 active sites which derived from hemin of hemoglobin, resulting in an enhanced peroxidase (POD)-like activity. Interestingly, the catalytic activity of erythrocyte-templated nanozyme (ETN) shows dependence on animal species, among which murine ETN performed superior catalytic efficiency. In addition, the as-prepared ETNs display a honeycomb-like network structure, serving as a sponge to accelerate hemostasis based on the interactions with prothrombin and fibrinogen. These features enable ETN to effectively kill methicillin-resistant Staphylococcus aureus (MRSA) by combining POD-like catalysis with near-infrared (NIR) induced photothermal effect, and subsequently suitable to promote wound healing. This study provides a proof-of-concept for facile fabrication of multifunctional nanozymes with uniform single-atom active sites by utilizing intrinsic iron structure characteristics of biogenic source like erythrocytes.


Subject(s)
Hemin , Methicillin-Resistant Staphylococcus aureus , Animals , Mice , Erythrocytes , Wound Healing , Iron
18.
Adv Healthc Mater ; 13(11): e2303623, 2024 04.
Article in English | MEDLINE | ID: mdl-38142309

ABSTRACT

PD-1/PD-L1 blockade immunotherapy has gained approval for the treatment of a diverse range of tumors; however, its efficacy is constrained by the insufficient infiltration of T lymphocytes into the tumor microenvironment, resulting in suboptimal patient responses. Here, a pioneering immunotherapy ferritin nanodrug delivery system denoted as ITFn-Pt(IV) is introduced. This system orchestrates a synergistic fusion of PD-L1 blockade, chemotherapy, and T-cell activation, aiming to augment the efficacy of tumor immunotherapy. Leveraging genetic engineering approach and temperature-regulated channel-based drug loading techniques, the architecture of this intelligent responsive system is refined. It is adept at facilitating the precise release of T-cell activating peptide Tα1 in the tumor milieu, leading to an elevation in T-cell proliferation and activation. The integration of PD-L1 nanobody KN035 ensures targeted engagement with tumor cells and mediates the intracellular delivery of the encapsulated Pt(IV) drugs, culminating in immunogenic cell death and the subsequent dendritic cell maturation. Employing esophageal squamous cell carcinoma (ESCC) as tumor model, the potent antitumor efficacy of ITFn-Pt(IV) is elucidated, underscored by augmented T-cell infiltration devoid of systemic adverse effects. These findings accentuate the potential of ITFn-Pt(IV) for ESCC treatment and its applicability to other malignancies resistant to established PD-1/PD-L1 blockade therapies.


Subject(s)
B7-H1 Antigen , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , T-Lymphocytes , Animals , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Esophageal Neoplasms/immunology , Humans , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Mice , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/therapy , Cell Line, Tumor , Lymphocyte Activation/drug effects , Ferritins/chemistry , Tumor Microenvironment/drug effects , Immunotherapy/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Nanoparticles/chemistry , Drug Delivery Systems/methods
19.
Nat Commun ; 14(1): 8137, 2023 Dec 08.
Article in English | MEDLINE | ID: mdl-38065945

ABSTRACT

Nanomedicine-based anti-neuroinflammation strategy has become a promising dawn of Parkinson's disease (PD) treatment. However, there are significant gaps in our understanding of the therapeutic mechanisms of antioxidant nanomedicines concerning the pathways traversing the blood-brain barrier (BBB) and subsequent inflammation mitigation. Here, we report nanozyme-integrated metal-organic frameworks with excellent antioxidant activity and chiral-dependent BBB transendocytosis as anti-neuroinflammatory agents for the treatment of PD. These chiral nanozymes are synthesized by embedding ultra-small platinum nanozymes (Ptzymes) into L-chiral and D-chiral imidazolate zeolite frameworks (Ptzyme@L-ZIF and Ptzyme@D-ZIF). Compared to Ptzyme@L-ZIF, Ptzyme@D-ZIF shows higher accumulation in the brains of male PD mouse models due to longer plasma residence time and more pathways to traverse BBB, including clathrin-mediated and caveolae-mediated endocytosis. These factors contribute to the superior therapeutic efficacy of Ptzyme@D-ZIF in reducing behavioral disorders and pathological changes. Bioinformatics and biochemical analyses suggest that Ptzyme@D-ZIF inhibits neuroinflammation-induced apoptosis and ferroptosis in damaged neurons. The research uncovers the biodistribution, metabolic variances, and therapeutic outcomes of nanozymes-integrated chiral ZIF platforms, providing possibilities for devising anti-PD drugs.


Subject(s)
Metal-Organic Frameworks , Parkinson Disease , Animals , Mice , Male , Metal-Organic Frameworks/pharmacology , Parkinson Disease/drug therapy , Neuroinflammatory Diseases , Tissue Distribution , Antioxidants
20.
Adv Sci (Weinh) ; 10(35): e2300698, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37888866

ABSTRACT

The success of arsenic in degrading PML-RARα oncoprotein illustrates the great anti-leukemia value of inorganics. Inspired by this, the therapeutic effect of inorganic selenium on t(8; 21) leukemia is studied, which has shown promising anti-cancer effects on solid tumors. A leukemia-targeting selenium nanomedicine is rationally built with bioengineered protein nanocage and is demonstrated to be an effective epigenetic drug for inducing the differentiation of t(8;21) leukemia. The selenium drug significantly induces the differentiation of t(8;21) leukemia cells into more mature myeloid cells. Mechanistic analysis shows that the selenium is metabolized into bioactive forms in cells, which drives the degradation of the AML1-ETO oncoprotein by inhibiting histone deacetylases activity, resulting in the regulation of AML1-ETO target genes. The regulation results in a significant increase in the expression levels of myeloid differentiation transcription factors PU.1 and C/EBPα, and a significant decrease in the expression level of C-KIT protein, a member of the type III receptor tyrosine kinase family. This study demonstrates that this protein-nanocaged selenium is a potential therapeutic drug against t(8;21) leukemia through epigenetic regulation.


Subject(s)
Leukemia, Myeloid, Acute , Selenium , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Selenium/pharmacology , Selenium/metabolism , Epigenesis, Genetic , RUNX1 Translocation Partner 1 Protein/genetics , RUNX1 Translocation Partner 1 Protein/metabolism , Cell Differentiation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL