Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 532
1.
Int J Nanomedicine ; 19: 3847-3859, 2024.
Article En | MEDLINE | ID: mdl-38708182

Background: Dihydroartemisinin (DHA) has emerged as a promising candidate for anticancer therapy. However, the application of DHA in clinics has been hampered by several limitations including poor bioavailability, short circulation life, and low solubility, significantly restricting its therapeutic efficacy and leading to notable side effects during the treatment. Purpose: We present DHA-loaded zeolitic imidazolate framework-8 (D-ZIF) with controllable and targeted DHA release properties, leading to enhanced antitumor effects while reducing potential side effects. Methods: D-ZIF was prepared by one-pot synthesis method using methylimidazole (MIM), Zn(NO3)2•6H2O and DHA. We characterized the physical and chemical properties of D-ZIF by TEM, DLS, XRD, FT-IR, and TG. We measured the drug loading efficiency and the cumulative release of DHA in different pH conditions. We evaluated the cytotoxicity of D-ZIF on renal cell carcinoma (RCC786-O), glioma cells (U251), TAX-resistant human lung adenocarcinoma (A549-TAX) cells by CCK8 in vitro. We explored the possible antitumor mechanism of D-ZIF by Western blot. We evaluated the biocompatibility and hemolysis of D-ZIF and explored the in vivo antitumor efficiency in mice model by TUNEL testing and blood biomarker evaluations. Results: D-ZIF showed rhombic dodecahedral morphology with size of 129±7.2 nm and possessed a noticeable DHA encapsulation efficiency (72.9%). After 48 hours, D-ZIF released a cumulative 70.0% of the loaded DHA at pH 6.5, and only 42.1% at pH 7.4. The pH-triggered programmed release behavior of D-ZIF could enhance anticancer effect of DHA while minimizing side effects under normal physiological conditions. Compared with the free DHA group with 31.75% of A549-TAX cell apoptosis, the percentage of apoptotic cells was approximately 76.67% in the D-ZIF group. D-ZIF inhibited tumor growth by inducing tumor cell apoptosis through the mechanism of ROS production and regulation of Nrf2/HO-1 and P38 MAPK signaling pathways. D-ZIF showed potent effects in treating tumors with high safety in vivo. Conclusion: This pH-responsive release mechanism enhanced the targeting efficiency of DHA towards tumor cells, thereby increasing drug concentration in tumor sites with negligible side effects. Herein, D-ZIF holds great promise for curing cancers with minimal adverse effects.


Antineoplastic Agents , Artemisinins , Drug Resistance, Neoplasm , Imidazoles , Lung Neoplasms , Metal-Organic Frameworks , Reactive Oxygen Species , Artemisinins/chemistry , Artemisinins/pharmacology , Artemisinins/pharmacokinetics , Animals , Humans , Reactive Oxygen Species/metabolism , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacokinetics , Metal-Organic Frameworks/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Hydrogen-Ion Concentration , A549 Cells , Drug Liberation , Mice, Nude , Apoptosis/drug effects , Mice, Inbred BALB C , Xenograft Model Antitumor Assays , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Hemolysis/drug effects
2.
Int J Nanomedicine ; 19: 3957-3972, 2024.
Article En | MEDLINE | ID: mdl-38711614

Purpose: Current treatment approaches for Prostate cancer (PCa) often come with debilitating side effects and limited therapeutic outcomes. There is urgent need for an alternative effective and safe treatment for PCa. Methods: We developed a nanoplatform to target prostate cancer cells based on graphdiyne (GDY) and a copper-based metal-organic framework (GDY-CuMOF), that carries the chemotherapy drug doxorubicin (DOX) for cancer treatment. Moreover, to provide GDY-CuMOF@DOX with homotypic targeting capability, we coated the PCa cell membrane (DU145 cell membrane, DCM) onto the surface of GDY-CuMOF@DOX, thus obtaining a biomimetic nanoplatform (DCM@GDY-CuMOF@DOX). The nanoplatform was characterized by using transmission electron microscope, atomic force microscope, X-ray diffraction, etc. Drug release behavior, antitumor effects in vivo and in vitro, and biosafety of the nanoplatform were evaluated. Results: We found that GDY-CuMOF exhibited a remarkable capability to load DOX mainly through π-conjugation and pore adsorption, and it responsively released DOX and generated Cu+ in the presence of glutathione (GSH). In vivo experiments demonstrated that this nanoplatform exhibits remarkable cell-killing efficiency by generating lethal reactive oxygen species (ROS) and mediating cuproptosis. In addition, DCM@GDY-CuMOF@DOX effectively suppresses tumor growth in vivo without causing any apparent side effects. Conclusion: The constructed DCM@GDY-CuMOF@DOX nanoplatform integrates tumor targeting, drug-responsive release and combination with cuproptosis and chemodynamic therapy, offering insights for further biomedical research on efficient PCa treatment.


Copper , Doxorubicin , Graphite , Metal-Organic Frameworks , Prostatic Neoplasms , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Doxorubicin/pharmacology , Doxorubicin/chemistry , Animals , Humans , Cell Line, Tumor , Copper/chemistry , Copper/pharmacology , Graphite/chemistry , Graphite/pharmacology , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Mice , Drug Liberation , Reactive Oxygen Species/metabolism , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Mice, Nude , Nanoparticles/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Carriers/chemistry , Xenograft Model Antitumor Assays
3.
Sci Rep ; 14(1): 10508, 2024 05 07.
Article En | MEDLINE | ID: mdl-38714808

In this study, a novel nanobiocomposite consisting of agar (Ag), tragacanth gum (TG), silk fibroin (SF), and MOF-5 was synthesized and extensively investigated by various analytical techniques and basic biological assays for potential biomedical applications. The performed Trypan blue dye exclusion assay indicated that the proliferation percentage of HEK293T cells was 71.19%, while the proliferation of cancer cells (K-562 and MCF-7) was significantly lower, at 10.74% and 3.33%. Furthermore, the Ag-TG hydrogel/SF/MOF-5 nanobiocomposite exhibited significant antimicrobial activity against both E. coli and S. aureus strains, with growth inhibition rates of 76.08% and 69.19% respectively. Additionally, the hemolytic index of fabricated nanobiocomposite was found approximately 19%. These findings suggest that the nanobiocomposite exhibits significant potential for application in cancer therapy and wound healing.


Agar , Fibroins , Hydrogels , Nanocomposites , Tragacanth , Fibroins/chemistry , Humans , Hydrogels/chemistry , Agar/chemistry , Nanocomposites/chemistry , Tragacanth/chemistry , Escherichia coli/drug effects , Escherichia coli/growth & development , Staphylococcus aureus/drug effects , HEK293 Cells , Zinc/chemistry , Cell Proliferation/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Microbial Sensitivity Tests , MCF-7 Cells , Cell Line, Tumor
4.
J Nanobiotechnology ; 22(1): 228, 2024 May 08.
Article En | MEDLINE | ID: mdl-38715049

Development of ferroptosis-inducible nanoplatforms with high efficiency and specificity is highly needed and challenging in tumor ferrotherapy. Here, we demonstrate highly effective tumor ferrotherapy using iron (II)-based metal-organic framework (FessMOF) nanoparticles, assembled from disulfide bonds and ferrous ions. The as-prepared FessMOF nanoparticles exhibit peroxidase-like activity and pH/glutathione-dependent degradability, which enables tumor-responsive catalytic therapy and glutathione depletion by the thiol/disulfide exchange to suppress glutathione peroxidase 4, respectively. Upon PEGylation and Actinomycin D (ActD) loading, the resulting FessMOF/ActD-PEG nanoplatform induces marked DNA damage and lipid peroxidation. Concurrently, we found that ActD can inhibit Xc- system and elicit ferritinophagy, which further boosts the ferrotherapeutic efficacy of the FessMOF/ActD-PEG. In vivo experiments demonstrate that our fabricated nanoplatform presents excellent biocompatibility and a high tumor inhibition rate of 91.89%.


DNA Damage , Ferroptosis , Iron , Metal-Organic Frameworks , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Ferroptosis/drug effects , Animals , Humans , Mice , DNA Damage/drug effects , Iron/chemistry , Cell Line, Tumor , DNA Repair/drug effects , Nanoparticles/chemistry , Neoplasms/drug therapy , Mice, Inbred BALB C , Female
5.
Int J Nanomedicine ; 19: 4163-4180, 2024.
Article En | MEDLINE | ID: mdl-38751660

Purpose: The study aimed to address the non-specific toxicity of cytotoxins (CTX) in liver cancer treatment and explore their combined application with the photosensitizer Ce6, co-loaded into carbonized Zn/Co bimetallic organic frameworks. The goal was to achieve controlled CTX release and synergistic photodynamic therapy, with a focus on evaluating anti-tumor activity against human liver cancer cell lines (Hep G2). Methods: Purified cobra cytotoxin (CTX) and photosensitizer Ce6 were co-loaded into carbonized Zn/Co bimetallic organic frameworks, resulting in RGD-PDA@C-ZIF@(CTX+Ce6). The formulation was designed with surface-functionalization using polydopamine and tumor-penetrating peptide RGD. This approach aimed to facilitate controlled CTX release and enhance the synergistic effect of photodynamic therapy. The accumulation of RGD-PDA@C-ZIF@(CTX+Ce6) at tumor sites was achieved through RGD's active targeting and the enhanced permeability and retention (EPR) effect. In the acidic tumor microenvironment, the porous structure of the metal-organic framework disintegrated, releasing CTX and Ce6 into tumor cells. Results: Experiments demonstrated that RGD-PDA@C-ZIF@(CTX+Ce6) nanoparticles, combined with near-infrared laser irradiation, exhibited optimal anti-tumor effects against human liver cancer cells. The formulation showcased heightened anti-tumor activity without discernible systemic toxicity. Conclusion: The study underscores the potential of utilizing metal-organic frameworks as an efficient nanoplatform for co-loading cytotoxins and photodynamic therapy in liver cancer treatment. The developed formulation, RGD-PDA@C-ZIF@(CTX+Ce6), offers a promising avenue for advancing the clinical application of cytotoxins in oncology, providing a solid theoretical foundation for future research and development.


Indoles , Liver Neoplasms , Metal-Organic Frameworks , Photochemotherapy , Photosensitizing Agents , Zinc , Humans , Photochemotherapy/methods , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Liver Neoplasms/drug therapy , Zinc/chemistry , Zinc/pharmacology , Indoles/chemistry , Indoles/pharmacology , Indoles/administration & dosage , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/administration & dosage , Animals , Hep G2 Cells , Cobalt/chemistry , Cobalt/pharmacology , Oligopeptides/chemistry , Oligopeptides/pharmacology , Oligopeptides/pharmacokinetics , Polymers/chemistry , Mice , Cytotoxins/chemistry , Cytotoxins/pharmacology , Cytotoxins/pharmacokinetics , Mice, Nude , Mice, Inbred BALB C , Cell Survival/drug effects
6.
ACS Nano ; 18(19): 12386-12400, 2024 May 14.
Article En | MEDLINE | ID: mdl-38699808

Current cancer vaccines face challenges due to an immunosuppressive tumor microenvironment and their limited ability to produce an effective immune response. To address the above limitations, we develop a 3-(2-spiroadamantyl)-4-methoxy-4-(3-phosphoryloxy)-phenyl-1,2-dioxetane (alkaline phosphatase substrate) and XMD8-92 (extracellular signal-regulated kinase 5 inhibitor)-codelivered copper-tetrahydroxybenzoquinone (Cu-THBQ/AX) nanosized metal-organic framework to in situ-generate therapeutic vaccination. Once inside the early endosome, the alkaline phosphatase overexpressed in the tumor cells' membrane activates the in situ type I photodynamic effect of Cu-THBQ/AX for generating •O2-, and the Cu-THBQ/AX catalyzes O2 and H2O2 to •O2- and •OH via semiquinone radical catalysis and Fenton-like reactions. This surge of ROS in early endosomes triggers caspase-3-mediated proinflammatory pyroptosis via activating phospholipase C. Meanwhile, Cu-THBQ/AX can also induce the oligomerization of dihydrolipoamide S-acetyltransferase to trigger tumor cell cuproptosis. The production of •OH could also trigger the release of XMD8-92 for effectively inhibiting the efferocytosis of macrophages to convert immunosuppressive apoptosis of cancer cells into proinflammatory secondary necrosis. The simultaneous induction of pyroptosis, cuproptosis, and secondary necrosis effectively converts the tumor microenvironment from "cold" to "hot" conditions, making it an effective antigen pool. This transformation successfully activates the antitumor immune response, inhibiting tumor growth and metastasis.


Cancer Vaccines , Copper , Macrophages , Metal-Organic Frameworks , Pyroptosis , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Animals , Mice , Pyroptosis/drug effects , Macrophages/drug effects , Macrophages/metabolism , Humans , Copper/chemistry , Copper/pharmacology , Cancer Vaccines/chemistry , Tumor Microenvironment/drug effects , Nanoparticles/chemistry , Phagocytosis/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism , Mice, Inbred BALB C , Efferocytosis , Nanovaccines
7.
J Nanobiotechnology ; 22(1): 246, 2024 May 13.
Article En | MEDLINE | ID: mdl-38735970

Excessive production of reactive oxygen species (ROS) and inflammation are the key problems that impede diabetic wound healing. In particular, dressings with ROS scavenging capacity play a crucial role in the process of chronic wound healing. Herein, Zr-based large-pore mesoporous metal-organic frameworks (mesoMOFs) were successfully developed for the construction of spatially organized cascade bioreactors. Natural superoxide dismutase (SOD) and an artificial enzyme were spatially organized in these hierarchical mesoMOFs, forming a cascade antioxidant defense system, and presenting efficient intracellular and extracellular ROS scavenging performance. In vivo experiments demonstrated that the SOD@HMUiO-MnTCPP nanoparticles (S@M@H NPs) significantly accelerated diabetic wound healing. Transcriptomic and western blot results further indicated that the nanocomposite could inhibit fibroblast senescence and ferroptosis as well as the stimulator of interferon genes (STING) signaling pathway activation in macrophages mediated by mitochondrial oxidative stress through ROS elimination. Thus, the biomimetic multi-enzyme cascade catalytic system with spatial ordering demonstrated a high potential for diabetic wound healing, where senescence, ferroptosis, and STING signaling pathways may be potential targets.


Inflammation , Metal-Organic Frameworks , Reactive Oxygen Species , Wound Healing , Wound Healing/drug effects , Reactive Oxygen Species/metabolism , Animals , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Mice , Superoxide Dismutase/metabolism , Porosity , Oxidative Stress/drug effects , Signal Transduction/drug effects , RAW 264.7 Cells , Male , Ferroptosis/drug effects , Macrophages/drug effects , Macrophages/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Diabetes Mellitus, Experimental , Nanoparticles/chemistry , Humans , Antioxidants/pharmacology , Nanocomposites/chemistry , Membrane Proteins
8.
Food Chem ; 451: 139451, 2024 Sep 01.
Article En | MEDLINE | ID: mdl-38703724

Active antibacterial materials play an important role in solving food safety problems caused by pathogen contamination. In this study, a composite active antibacterial material with the synergistic antibacterial effectiveness of photothermal, photodynamic and the surface charge of polyphenols was developed, where the multi-porous polyphenol functionalized metal-organic frameworks (ZIF-8-TA) were used as the framework carrier, and black phosphorus quantum dots (BPQDs) were used as the photosensitive source. The resulted ZIF-8-TA/PBQDs possesses excellent photothermal conversion efficiency (27.92%), photodynamic performance and surface charge, and these factors ensure the outstanding broad-spectrum antibacterial performance (100%). Multifunctional characteristics and excellent biocompatibility endow the materials with vast potential for foodstuff packaging. The results showed that the composite antibacterial film produced by doping ZIF-8-TA/PBQDs into chitosan could effectively prolong the shelf life of foodstuff compared with commercial membrane. The successful implementation of this research provides a new idea for controlling microbial contamination and developing multifunctional antibacterial materials.


Anti-Bacterial Agents , Food Preservation , Metal-Organic Frameworks , Polyphenols , Quantum Dots , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Polyphenols/chemistry , Polyphenols/pharmacology , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Quantum Dots/chemistry , Food Preservation/methods , Food Preservation/instrumentation , Phosphorus/chemistry , Phosphorus/pharmacology , Food Packaging/instrumentation , Microbial Sensitivity Tests
9.
Food Chem ; 451: 139464, 2024 Sep 01.
Article En | MEDLINE | ID: mdl-38704990

Silver-metal organic framework (Ag@MOF) has exhibited outstanding antimicrobial activity in antimicrobial applications, and reducing the biotoxicity associated with silver has become a research priority. In this study, Ag@MOF was initially modified with sodium alginate (SA) to form SA-Ag@MOF. The results showed that SA could control the release of Ag+, reducing the release by about 8% at 24 h, and the biotoxicity was significantly reduced. Finally, SA-Ag@MOF was applied as an antimicrobial agent in citric acid-modified PVA film to develop a novel composite antimicrobial film. When added at 2 MIC, the CA3-M2 film can effectively inhibit the growth of E. coli and S. aureus, and the inhibition rate has reached 98%. For white radish packaging applications, CA3-M2 film inhibited the growth of surface microorganisms, while ensuring moisture and tissue hardness to extend shelf-life up to 7 days. Overall, the strategy conceived here can be a theoretical basis for novel antimicrobial packaging.


Alginates , Citric Acid , Escherichia coli , Food Packaging , Metal-Organic Frameworks , Silver , Staphylococcus aureus , Alginates/chemistry , Alginates/pharmacology , Food Packaging/instrumentation , Citric Acid/chemistry , Citric Acid/pharmacology , Silver/chemistry , Silver/pharmacology , Escherichia coli/drug effects , Escherichia coli/growth & development , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Microbial Sensitivity Tests , Raphanus/chemistry , Raphanus/growth & development , Raphanus/drug effects , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry
10.
J Nanobiotechnology ; 22(1): 287, 2024 May 26.
Article En | MEDLINE | ID: mdl-38797862

Periodontitis is a prevalent chronic inflammatory disease, which leads to gradual degradation of alveolar bone. The challenges persist in achieving effective alveolar bone repair due to the unique bacterial microenvironment's impact on immune responses. This study explores a novel approach utilizing Metal-Organic Frameworks (MOFs) (comprising magnesium and gallic acid) for promoting bone regeneration in periodontitis, which focuses on the physiological roles of magnesium ions in bone repair and gallic acid's antioxidant and immunomodulatory properties. However, the dynamic oral environment and irregular periodontal pockets pose challenges for sustained drug delivery. A smart responsive hydrogel system, integrating Carboxymethyl Chitosan (CMCS), Dextran (DEX) and 4-formylphenylboronic acid (4-FPBA) was designed to address this problem. The injectable self-healing hydrogel forms a dual-crosslinked network, incorporating the MOF and rendering its on-demand release sensitive to reactive oxygen species (ROS) levels and pH levels of periodontitis. We seek to analyze the hydrogel's synergistic effects with MOFs in antibacterial functions, immunomodulation and promotion of bone regeneration in periodontitis. In vivo and in vitro experiment validated the system's efficacy in inhibiting inflammation-related genes and proteins expression to foster periodontal bone regeneration. This dynamic hydrogel system with MOFs, shows promise as a potential therapeutic avenue for addressing the challenges in bone regeneration in periodontitis.


Bone Regeneration , Chitosan , Drug Delivery Systems , Hydrogels , Metal-Organic Frameworks , Periodontitis , Periodontitis/drug therapy , Hydrogels/chemistry , Bone Regeneration/drug effects , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Animals , Chitosan/chemistry , Chitosan/analogs & derivatives , Mice , Drug Delivery Systems/methods , Dextrans/chemistry , Male , Reactive Oxygen Species/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Delayed-Action Preparations/chemistry , Humans
11.
Int J Nanomedicine ; 19: 4569-4588, 2024.
Article En | MEDLINE | ID: mdl-38799697

Purpose: The primary objective of this study was to develop an innovative nanomedicine-based therapeutic strategy to alleviate Postoperative Neurocognitive Disorder (PND) in patients undergoing surgery. Patients and Methods: To achieve this goal, polydopamine-coated Kaempferol-loaded Metal-Organic Framework nanoparticles (pDA/KAE@ZIF-8) were synthesized and evaluated. The study involved encapsulating Kaempferol (KAE) within ZIF-8 nanoparticles, followed by coating with polydopamine (PDA) to enhance biocompatibility and targeted delivery. The characterization of these nanoparticles (NPs) was conducted using various techniques including Scanning Electron Microscopy, Fourier-Transform Infrared Spectroscopy, X-ray Diffraction, and Ultraviolet-Visible spectroscopy. The efficacy of pDA/KAE@ZIF-8 NPs was tested in both in vitro and in vivo models, specifically focusing on their ability to penetrate the blood-brain barrier and protect neuronal cells against oxidative stress. Results: The study found that pDA/KAE@ZIF-8 NPs efficiently penetrated the blood-brain barrier and were significantly taken up by neuronal cells. These nanoparticles demonstrated remarkable Reactive Oxygen Species (ROS) scavenging capabilities and stability under physiological conditions. In vitro studies showed that pDA/KAE@ZIF-8 NPs provided protection to HT-22 neuronal cells against H2O2-induced oxidative stress, reduced the levels of pro-inflammatory cytokines, and decreased apoptosis rates. In a PND mouse model, the treatment with pDA/KAE@ZIF-8 NPs significantly improved cognitive functions, surpassing the effects of KAE alone. This improvement was substantiated through behavioral tests and a noted reduction in hippocampal inflammation. Conclusion: The findings from this study underscore the potential of pDA/KAE@ZIF-8 NPs as an effective nanotherapeutic agent for PND. This approach offers a novel direction in the postoperative care of elderly patients, with the potential to transform the therapeutic landscape for neurocognitive disorders following surgery. The application of nanotechnology in this context opens new avenues for more effective and targeted treatments, thereby improving the quality of life for patients suffering from PND.


Indoles , Kaempferols , Metal-Organic Frameworks , Nanoparticles , Oxidative Stress , Polymers , Animals , Indoles/chemistry , Indoles/pharmacology , Polymers/chemistry , Kaempferols/chemistry , Kaempferols/pharmacology , Kaempferols/pharmacokinetics , Kaempferols/administration & dosage , Mice , Nanoparticles/chemistry , Oxidative Stress/drug effects , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cell Line , Reactive Oxygen Species/metabolism , Postoperative Cognitive Complications , Humans , Male , Neurons/drug effects , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/pharmacology
12.
Int J Biol Macromol ; 269(Pt 2): 132207, 2024 Jun.
Article En | MEDLINE | ID: mdl-38723823

To overcome the low efficacy of sonodynamic therapy (SDT) caused by hypoxia in the tumor microenvironment, we developed a multiple anti-tumor nanoplatform with synergistic SDT, photothermal therapy (PTT), and ferroptosis effects. PCN-224@FcCaO2/Mn/dihydroartemisinin/imiquimod/PDA (PFC) was prepared by modified with dihydroartemisinin (DHA), imiquimod (R837), CaO2, ferrocene (Fc) and Mn2+ on the PCN-224 (Cu) to achieve self-replenishment of H2O2/O2 and GSH consumption. FcCaO2 decomposed into H2O2 in the tumor microenvironment, triggering the Fenton effect to produce OH, and Cu2+ reduced the potential loss of OH by the depletion of GSH. Under ultrasonic (US) and laser irradiation, PFC exhibits exciting PTT and SDT effects from polydopamine (PDA) and PCN-224. Mn2+ not only promoted the reaction of H2O2 to produce O2 to effectively enhance SDT but also induced tumor cell apoptosis by Mn2+ combined with DHA. PFC induced ferroptosis via Fe interaction with DHA to produce ROS and reduce the expression of GPX4. The released R837 and tumor-associated antigens from SDT/PTT can produce damage associated molecular patterns (DAMPs), which can initiate adaptive immune responses to kill cancer cells, and released again to promote the tumor immune cycle. What's more, SDT/PTT and ferroptosis combined with aPD-L1 can effectively suppress both primary and distant tumor growth.


Indoles , Metal-Organic Frameworks , Photothermal Therapy , Polymers , Indoles/chemistry , Indoles/pharmacology , Polymers/chemistry , Polymers/pharmacology , Humans , Animals , Mice , Photothermal Therapy/methods , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Cell Line, Tumor , Nanoparticles/chemistry , Apoptosis/drug effects , Ferroptosis/drug effects , Tumor Microenvironment/drug effects , Combined Modality Therapy , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/chemistry , Hydrogen Peroxide/pharmacology , Imiquimod/pharmacology , Metallocenes/chemistry , Metallocenes/pharmacology
13.
J Mater Chem B ; 12(21): 5220-5237, 2024 May 29.
Article En | MEDLINE | ID: mdl-38695162

The HIV attacks the immune system provoking an infection that is considered a global health challenge. Despite antiretroviral treatments being effective in reducing the plasma viral load in the blood to undetectable levels in people living with HIV (PLWH), the disease is not cured and has become chronic. This happens because of the existence of anatomical and cellular viral reservoirs, mainly located in the lymph nodes and gastrointestinal tract, which are composed of infected CD4+ T cells with a resting memory phenotype and inaccessible to antiretroviral therapy. Herein, a new therapeutic strategy based on nanotechnology is presented. Different combinations of antiretroviral drugs (bictegravir/tenofovir/emtricitabine and nevirapine/tenofovir/emtricitabine) and toll-like receptor agonists were encapsulated into metal-organic frameworks (MOFs) PCN-224 and ZIF-8. The encapsulation efficiencies of all the drugs, as well as their release rate from the carriers, were measured. In vitro studies about the cell viability, the hemocompatibility, and the platelet aggregation of the MOFs were carried out. Epifluorescence microscopy assays confirmed the ability of ZIF-8 to target a carboxyfluorescein probe inside HeLa cell lines and PBMCs. These results pave the way for the use of these structures to eliminate latent HIV reservoirs from anatomical compartments through the activation of innate immune cells, and a higher efficacy of the triplet combinations of antiretroviral drugs.


Anti-HIV Agents , Biocompatible Materials , HIV Infections , Metal-Organic Frameworks , Humans , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , HIV Infections/drug therapy , Anti-HIV Agents/pharmacology , Anti-HIV Agents/chemistry , HeLa Cells , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , HIV-1/drug effects , Particle Size , Cell Survival/drug effects , Surface Properties
14.
Int J Nanomedicine ; 19: 4253-4261, 2024.
Article En | MEDLINE | ID: mdl-38766662

Purpose: Recently, Single-atom-loaded carbon-based material is a new environmentally friendly and stable photothermal antibacterial nanomaterial. It is still a great challenge to achieve single-atom loading on carbon materials. Materials and Methods: Herein, We doped single-atom Ag into ZIF-8-derived porous carbon to obtain Ag-doped ZIF-8-derived porous carbon(AgSA-ZDPC). The as-prepared samples were characterized by XRD, XPS, FESEM, EDX, TEM, and HAADF-STEM which confirmed that the single-atom Ag successfully doped into the porous carbon. Further, the photothermal properties and antimicrobial activity of AgSA-ZDPC have been tested. Results: The results showed that the temperature increased by 30 °C after near-infrared light irradiation(1 W/cm2) for 5 min which was better than ZIF-8-derived porous carbon(ZDPC). It also exhibits excellent photothermal stability after the laser was switched on and off 5 times. When the AgSA-ZDPC concentration was greater than 50 µg/mL and the near-infrared irradiation was performed for 5 min, the growth inhibition of S. aureus and E. coli was almost 100%. Conclusion: This work provides a simple method for the preparation of single-atom Ag-doped microporous carbon which has potential antibacterial application.


Anti-Bacterial Agents , Carbon , Escherichia coli , Silver , Staphylococcus aureus , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Silver/chemistry , Silver/pharmacology , Porosity , Escherichia coli/drug effects , Staphylococcus aureus/drug effects , Carbon/chemistry , Carbon/pharmacology , Infrared Rays , Microbial Sensitivity Tests , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Zeolites/chemistry , Zeolites/pharmacology , Imidazoles
15.
Acta Biomater ; 180: 183-196, 2024 May.
Article En | MEDLINE | ID: mdl-38604465

The utilization of biodegradable magnesium (Mg) alloys in the fabrication of temporary non-vascular stents is an innovative trend in biomedical engineering. However, the heterogeneous degradation profiles of these biomaterials, together with potential bacterial colonization that could precipitate infectious or stenotic complications, are critical obstacles precluding their widespread clinical application. In pursuit of overcoming these limitations, this study applies the principles of biomimicry, particularly the hydrophobic and anti-fouling characteristics of lotus leaves, to pioneer the creation of nanocomposite coatings. These coatings integrate poly-trimethylene carbonate (PTMC) with covalent organic frameworks (COFs), to modify the stent's surface property. The strategic design of the coating's topography, porosity, and self-polishing capabilities collectively aims to decelerate degradation processes and minimize biological adhesion. The protective qualities of the coatings were substantiated through rigorous testing in both in vitro dynamic bile tests and in vivo New Zealand rabbit choledochal models. Empirical findings from these trials confirmed that the implementation of COF-based nanocomposite coatings robustly fortifies Mg implantations, conferring heightened resistance to both biocorrosion and biofouling as well as improved biocompatibility within bodily environments. The outcomes of this research elucidate a comprehensive framework for the multifaceted strategies against stent corrosion and fouling, thereby charting a visionary pathway toward the systematic conception of a new class of reliable COF-derived surface modifications poised to amplify the efficacy of Mg-based stents. STATEMENT OF SIGNIFICANCE: Biodegradable magnesium (Mg) alloys are widely utilized in temporary stents, though their rapid degradation and susceptibility to bacterial infection pose significant challenges. Our research has developed a nanocomposite coating inspired by the lotus, integrating poly-trimethylene carbonate with covalent organic frameworks (COF). The coating achieved self-polishing property and optimal surface energy on the Mg substrate, which decelerates stent degradation and reduces biofilm formation. Comprehensive evaluations utilizing dynamic bile simulations and implantation in New Zealand rabbit choledochal models reveal that the coating improves the durability and longevity of the stent. The implications of these findings suggest the potential COF-based Mg alloy stent surface treatments and a leap forward in advancing stent performance and endurance in clinical applications.


Absorbable Implants , Coated Materials, Biocompatible , Magnesium , Nanocomposites , Stents , Animals , Rabbits , Magnesium/chemistry , Magnesium/pharmacology , Nanocomposites/chemistry , Corrosion , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Biofouling/prevention & control , Dioxanes/chemistry , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Polymers/chemistry , Polymers/pharmacology , Alloys/chemistry , Alloys/pharmacology
16.
Nanoscale ; 16(19): 9406-9411, 2024 May 16.
Article En | MEDLINE | ID: mdl-38629905

In the field of contemporary medicine, inflammation has emerged as a significant concern in global public health. Among the current anti-inflammatory strategies, nanozymes possess distinctive advantages and demonstrate unexpected efficacy in combating inflammation. However, the indeterminate structures and limited enzyme-like activity exhibited by most developed nanozymes impede their clinical translation and therapeutic effectiveness. In this paper, we developed a nanozyme derived from a well-defined metal-organic cage (MOC). The oxidized MOC (MOC-O), containing pyridine nitrogen oxide moieties, exhibited effective cascade superoxide dismutase (SOD) and catalase (CAT)-like activities for scavenging reactive oxygen species (ROS). This ROS scavenging ability was confirmed through flow cytometry analysis using DCFH-DA in a hypoxia/reoxygenation (H/R) model, where MOC-O significantly alleviated oxidative stress. Furthermore, the administration of MOC-O resulted in preserved renal function during renal ischemia-reperfusion (I/R) injury due to downregulated oxidative stress levels and reduced cell apoptosis.


Antioxidants , Kidney , Oxidative Stress , Reactive Oxygen Species , Reperfusion Injury , Superoxide Dismutase , Reperfusion Injury/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Animals , Antioxidants/chemistry , Antioxidants/pharmacology , Reactive Oxygen Species/metabolism , Kidney/metabolism , Oxidative Stress/drug effects , Superoxide Dismutase/metabolism , Mice , Apoptosis/drug effects , Catalase/metabolism , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Male
17.
Nanoscale ; 16(19): 9496-9508, 2024 May 16.
Article En | MEDLINE | ID: mdl-38651386

"Transition" metal-coordinated plant polyphenols are a type of promising antitumor nanodrugs owing to their high biosafety and catalytic therapy potency; however, the major obstacle restricting their clinical application is their poor tumor accumulation. Herein, Fe-doped ZIF-8 was tailored using tannic acid (TA) into a hollow mesoporous nanocarrier for gallic acid (GA) loading. After hyaluronic acid (HA) modification, the developed nanosystem of HFZIF-8/GA@HA was used for the targeted delivery of Fe ions and GA, thereby intratumorally achieving the synthesis of an Fe-GA coordinated complex. The TA-etching strategy facilitated the development of a cavitary structure and abundant coordination sites of ZIF-8, thus ensuring an ideal loading efficacy of GA (23.4 wt%). When HFZIF-8/GA@HA accumulates in the tumor microenvironment (TME), the framework is broken due to the competitive protonation ability of overexpressed protons in the TME. Interestingly, the intratumoral degradation of HFZIF-8/GA@HA provides the opportunity for the in situ "meeting" of GA and Fe ions, and through the coordination of polyhydroxyls assisted by conjugated electrons on the benzene ring, highly stable Fe-GA nanochelates are formed. Significantly, owing to the electron delocalization effect of GA, intratumorally coordinated Fe-GA could efficiently absorb second near-infrared (NIR-II, 1064 nm) laser irradiation and transfer it into thermal energy with a conversion efficiency of 36.7%. The photothermal performance could speed up the Fenton reaction rate of Fe-GA with endogenous H2O2 for generating more hydroxyl radicals, thus realizing thermally enhanced chemodynamic therapy. Overall, our research findings demonstrate that HFZIF-8/GA@HA has potential as a safe and efficient anticancer nanodrug.


Gallic Acid , Gallic Acid/chemistry , Gallic Acid/pharmacology , Mice , Animals , Humans , Cell Line, Tumor , Tannins/chemistry , Hyaluronic Acid/chemistry , Iron/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Tumor Microenvironment/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Catalysis , Drug Carriers/chemistry , Nanoparticles/chemistry , Imidazoles
18.
J Mater Chem B ; 12(19): 4717-4723, 2024 May 15.
Article En | MEDLINE | ID: mdl-38655651

Metal-organic frameworks (MOFs) possess a variety of interesting features related to their composition and structure that make them excellent candidates to be used in agriculture. However, few studies have reported their use as delivery agents of agrochemicals. In this work, the natural polyphenol chlorogenic acid (CGA) was entrapped via simple impregnation in the titanium aminoterephthalate MOF, MIL-125-NH2. A combination of experimental and computational techniques was used to understand and quantify the encapsulated CGA in MIL-125-NH2. Subsequently, CGA delivery studies were carried out in water at different pHs, showing a fast release of CGA during the first 2 h (17.3 ± 0.3% at pH = 6.5). In vivo studies were also performed against larvae of mealworm (Tenebrio molitor), evidencing the long-lasting insecticidal activity of CGA@MIL-125-NH2. This report demonstrates the potential of MOFs in the efficient release of agrochemicals, and paves the way to their study against in vivo models.


Chlorogenic Acid , Insecticides , Metal-Organic Frameworks , Chlorogenic Acid/chemistry , Chlorogenic Acid/pharmacology , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Insecticides/chemistry , Insecticides/pharmacology , Animals , Tenebrio/chemistry , Tenebrio/drug effects , Larva/drug effects
19.
ACS Appl Mater Interfaces ; 16(19): 24235-24247, 2024 May 15.
Article En | MEDLINE | ID: mdl-38688002

The spread of upper respiratory tract (URT) infections harms people's health and causes social burdens. Developing targeted treatment strategies for URT infections that exhibit good biocompatibility, stability, and strong antimicrobial effects remains challenging. The dual antimicrobial and antiviral effects of iodine (I2) in combination with the cooling sensation of l-menthol in the respiratory tract can simultaneously alleviate URT inflammation symptoms. However, as both I2 and l-menthol are volatile, addressing stability issues is crucial. In this study, a potassium iodide ß-cyclodextrin metal-organic framework [ß-CD-POF(I)] with appropriate particle size was used to coload and deliver I2 and l-menthol. Primarily, ß-CD-POF(I) was employed as the most efficient carrier to significantly enhance the stability of I2, surpassing any other known protection strategies in the pharmaceutical field (CD complexations, PVP conjugations, and cadexomer iodine). The mechanism underlying the improvement in stability of I2 by ß-CD-POF(I) was investigated through scanning electron microscopy with energy-dispersive X-ray spectroscopy, X-ray photoelectron spectroscopy, Raman spectroscopy, and molecular docking. The results revealed that the key processes involved in improving stability were the inclusion of I2 by ß-CD cavities in ß-CD-POF(I) and the formation of polyiodide anion between iodine ions and I2. Furthermore, the potential of ß-CD-POF(I) to load and deliver drugs was validated, and coloading of l-menthol and I2 demonstrated reliable stability. ß-CD-POF(I) achieved a rate of URT deposition ≥95% in vitro, and the combined antibacterial effects of coloaded I2 and l-menthol was better than I2 or PVP-I alone, with no irritation noted following URT administration in rabbits. Therefore, the stable coloading of drugs by ß-CD-POF(I), leading to enhanced antimicrobial effects, provides a new strategy for treating URT infections.


Anti-Bacterial Agents , Iodine , Metal-Organic Frameworks , beta-Cyclodextrins , Iodine/chemistry , Iodine/pharmacology , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , beta-Cyclodextrins/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Animals , Microbial Sensitivity Tests , Rabbits , Drug Carriers/chemistry , Staphylococcus aureus/drug effects , Molecular Docking Simulation
20.
ACS Nano ; 18(19): 12261-12275, 2024 May 14.
Article En | MEDLINE | ID: mdl-38683132

Cancer immunotherapy holds significant promise for addressing diverse malignancies. Nevertheless, its efficacy remains constrained by the intricate tumor immunosuppressive microenvironment. Herein, a light-triggered nanozyme Fe-TCPP-R848-PEG (Fe-MOF-RP) was designed for remodeling the immunosuppressive microenvironment. The Fe-TCPP-MOFs were utilized not only as a core catalysis component against tumor destruction but also as a biocompatible delivery vector of an immunologic agonist, improving its long circulation and tumor enrichment. Concurrently, it catalyzes the decomposition of H2O2 within the tumor, yielding oxygen to augment photodynamic therapy. The induced ferroptosis, in synergy with photodynamic therapy, prompts the liberation of tumor-associated antigens from tumor cells inducing immunogenic cell death. Phototriggered on-demand release of R848 agonists stimulated the maturation of dendritic cells and reverted the tumor-promoting M2 phenotypes into adoptive M1 macrophages, which further reshaped the tumor immunosuppressive microenvironment. Notably, the nanozyme effectively restrains well-established tumors, such as B16F10 melanoma. Moreover, it demonstrates a distal tumor-inhibiting effect upon in situ light treatment. What is more, in a lung metastasis model, it elicits robust immune memory, conferring enduring protection against tumor rechallenge. Our study presents a straightforward and broadly applicable strategy for crafting nanozymes with the potential to effectively thwart cancer recurrence and metastasis.


Ferroptosis , Light , Tumor Microenvironment , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Animals , Ferroptosis/drug effects , Mice , Mice, Inbred C57BL , Photochemotherapy , Tumor Hypoxia/drug effects , Nanoparticles/chemistry , Immunotherapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Humans , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Melanoma, Experimental/pathology , Cell Line, Tumor
...