Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 516
Filter
1.
Sci Rep ; 14(1): 10508, 2024 05 07.
Article in English | MEDLINE | ID: mdl-38714808

ABSTRACT

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.


Subject(s)
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
2.
J Nanobiotechnology ; 22(1): 246, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38735970

ABSTRACT

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.


Subject(s)
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
3.
Int J Nanomedicine ; 19: 3847-3859, 2024.
Article in English | MEDLINE | ID: mdl-38708182

ABSTRACT

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.


Subject(s)
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
4.
J Nanobiotechnology ; 22(1): 228, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38715049

ABSTRACT

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%.


Subject(s)
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: 3957-3972, 2024.
Article in English | MEDLINE | ID: mdl-38711614

ABSTRACT

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.


Subject(s)
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
6.
ACS Nano ; 18(19): 12386-12400, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38699808

ABSTRACT

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.


Subject(s)
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 Colloid Interface Sci ; 667: 91-100, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38621335

ABSTRACT

The development of efficient and multifunctional sonosensitizers is crucial for enhancing the efficacy of sonodynamic therapy (SDT). Herein, we have successfully constructed a CoOx-loaded amorphous metal-organic framework (MOF) UIO-66 (A-UIO-66-CoOx) sonosensitizer with excellent catalase (CAT)- and glutathione-oxidase (GSH-OXD)-like activities. The A-UIO-66-CoOx exhibits a 2.6-fold increase in singlet oxygen (1O2) generation under ultrasound (US) exposure compared to crystalline UIO-66 sonosensitizer, which is attributed to its superior charge transfer efficiency and consistent oxygen (O2) supply. Additionally, the A-UIO-66-CoOx composite reduces the expression of glutathione peroxidase (GPX4) by depleting glutathione (GSH) through Co3+ and Co2+ valence changes. The high levels of highly cytotoxic 1O2 and deactivation of GPX4 can lead to lethal lipid peroxidation, resulting in concurrent apoptosis and ferroptosis. Both in vitro and vivo tumor models comprehensively confirmed the enhanced SDT antitumor effect using A-UIO-66-CoOx sonosensitizer. Overall, this study emphasizes the possibility of utilizing amorphization engineering to improve the effectiveness of MOFs-based sonosensitizers for combined cancer therapies.


Subject(s)
Apoptosis , Ferroptosis , Metal-Organic Frameworks , Ultrasonic Therapy , Ferroptosis/drug effects , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Humans , Apoptosis/drug effects , Animals , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mice, Inbred BALB C , Drug Screening Assays, Antitumor , Cell Survival/drug effects , Cell Proliferation/drug effects , Particle Size , Cobalt/chemistry , Cobalt/pharmacology , Surface Properties , Singlet Oxygen/metabolism , Singlet Oxygen/chemistry , Cell Line, Tumor
8.
ACS Nano ; 18(19): 12261-12275, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38683132

ABSTRACT

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.


Subject(s)
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
9.
J Mater Chem B ; 12(19): 4717-4723, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38655651

ABSTRACT

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.


Subject(s)
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
10.
J Dent Res ; 103(5): 516-525, 2024 May.
Article in English | MEDLINE | ID: mdl-38581213

ABSTRACT

Titanium (Ti)-based biomaterials lack inherent antimicrobial activities, and the dental plaque formed on the implant surface is one of the main risk factors for implant infections. Construction of an antibacterial surface can effectively prevent implant infections and enhance implant success. Silver nanoparticles (AgNPs) exhibit broad antibacterial activity and a low tendency to induce drug resistance, but AgNPs easily self-aggregate in the aqueous environment, which significantly impairs their antibacterial activity. In this study, UiO-66/AgNP (U/A) nanocomposite was prepared, where zirconium metal-organic frameworks (UiO-66) were employed as the confinement matrix to control the particle size and prevent aggregation of AgNPs. The bactericidal activity of U/A against methicillin-resistant Staphylococcus aureus and Escherichia coli increased nearly 75.51 and 484.50 times compared with individually synthesized Ag. The antibacterial mechanism can be attributed to the enhanced membrane rupture caused by the ultrafine AgNPs on UiO-66, leading to protein leakage and generation of intracellular reactive oxygen species. Then, U/A was loaded onto Ti substrates (Ti-U/A) by using self-assembly deposition methods to construct an antibacterial surface coating. Ti-U/A exhibited excellent antibacterial activities and desired biocompatibility both in vitro and in vivo. The U/A nanocomposite coating technique is thus expected to be used as a promising surface modification strategy for Ti-based dental implants for preventing dental implant infections.


Subject(s)
Anti-Bacterial Agents , Coated Materials, Biocompatible , Dental Implants , Escherichia coli , Metal Nanoparticles , Methicillin-Resistant Staphylococcus aureus , Silver , Zirconium , Silver/pharmacology , Dental Implants/microbiology , Anti-Bacterial Agents/pharmacology , Metal Nanoparticles/therapeutic use , Escherichia coli/drug effects , Zirconium/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Coated Materials, Biocompatible/pharmacology , Metal-Organic Frameworks/pharmacology , Metal-Organic Frameworks/chemistry , Animals , Titanium/chemistry , Nanocomposites/chemistry , Surface Properties , Mice , Reactive Oxygen Species
11.
J Mater Chem B ; 12(16): 4018-4028, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38578014

ABSTRACT

On-demand controlled drug release holds great promise for cancer therapy. Light-degradable nanocarriers have gained increasing attention for designing controllable drug delivery systems owing to their spatiotemporally controllable properties. Herein, a highly luminescent and light-degradable nanocarrier is constructed by intercalating glutathione-capped gold nanoclusters (AuNCs) into zeolitic imidazolate framework-8 (ZIF-8) via competitive coordination assembly, named AuNC@ZIF-8, for light-triggered drug release. Glutathione-capped AuNCs and 2-methylimidazole (MIm) competitively coordinated with Zn2+ to form AuNC@ZIF-8 using a one step process in an aqueous solution. Specifically, the obtained AuNC@ZIF-8 has a high quantum yield of 52.96% and displays a distinctive property of photolysis. Competitive coordination interactions within AuNC@ZIF-8 were evidenced by X-ray diffraction and X-ray photoelectron spectroscopy, in which Zn2+ strongly coordinated with the N of MIm and weakly coordinated with the carboxyl/amino groups in the glutathione of AuNCs. Under light irradiation, the Au-S bond in AuNCs breaks, enhancing the coordination ability between carboxyl/amino groups and Zn2+. This collapses the crystal structure of AuNC@ZIF-8 and causes subsequent fluorescence quenching. Additionally, AuNC@ZIF-8 is successfully employed as a luminescent nanocarrier of anticancer drugs to form drug-AuNC@ZIF-8, in which three typical anticancer drugs are selected due to different coordination interactions. The obtained smart drug-AuNC@ZIF-8 can be effectively internalized into HeLa cells and degraded in response to blue light, with negligible dark cytotoxicity and high light cytotoxicity. This study highlights the crucial role of competitive coordination interactions in synthesizing functional materials with fluorescence efficiency and photolytic properties.


Subject(s)
Drug Liberation , Gold , Light , Metal Nanoparticles , Metal-Organic Frameworks , Gold/chemistry , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Humans , Metal Nanoparticles/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , HeLa Cells , Cell Survival/drug effects , Drug Carriers/chemistry , Particle Size , Surface Properties , Doxorubicin/chemistry , Doxorubicin/pharmacology
12.
J Nanobiotechnology ; 22(1): 206, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658950

ABSTRACT

The insufficient abundance and weak activity of tumour-infiltrating lymphocytes (TILs) are two important reasons for the poor efficacy of PD-1 inhibitors in hepatocellular carcinoma (HCC) treatment. The combined administration of tanshinone IIA (TSA) and astragaloside IV (As) can up-regulate the abundance and activity of TILs by normalising tumour blood vessels and reducing the levels of immunosuppressive factors respectively. For enhancing the efficacy of PD-1 antibody, a magnetic metal-organic framework (MOF) with a homologous tumour cell membrane (Hm) coating (Hm@TSA/As-MOF) is established to co-deliver TSA&As into the HCC microenvironment. Hm@TSA/As-MOF is a spherical nanoparticle and has a high total drug-loading capacity of 16.13 wt%. The Hm coating and magnetic responsiveness of Hm@TSA/As-MOF provide a homologous-magnetic dual-targeting, which enable Hm@TSA/As-MOF to counteract the interference posed by ascites tumour cells and enhance the precision of targeting solid tumours. Hm coating also enable Hm@TSA/As-MOF to evade immune clearance by macrophages. The release of TSA&As from Hm@TSA/As-MOF can be accelerated by HCC microenvironment, thereby up-regulating the abundance and activity of TILs to synergistic PD-1 antibody against HCC. This study presents a nanoplatform to improve the efficacy of PD-1 inhibitors in HCC, providing a novel approach for anti-tumour immunotherapy in clinical practice.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Metal-Organic Frameworks , Programmed Cell Death 1 Receptor , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Liver Neoplasms/drug therapy , Carcinoma, Hepatocellular/drug therapy , Animals , Mice , Humans , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Cell Line, Tumor , Immune Checkpoint Inhibitors/pharmacology , Tumor Microenvironment/drug effects , Mice, Inbred BALB C , Saponins/pharmacology , Saponins/chemistry , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology
13.
J Colloid Interface Sci ; 667: 491-502, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38653070

ABSTRACT

An injectable hydrogel dressing, Zr/Fc-MOF@CuO2@FH, was constructed by combing acid-triggered chemodynamic treatment (CDT) with low-temperature photothermal treatment (LT-PTT) to effectively eliminate bacteria without harming the surrounding normal tissues. The Zr/Fc-MOF acts as both photothermal reagent and nanozyme to generate reactive oxygen species (ROS). The CuO2 nanolayer can be decomposed by the acidic microenvironment of the bacterial infection to release Cu2+ and H2O2, which not only induces Fenton-like reaction but also enhances the catalytic capability of the Zr/Fc-MOF. The generated heat augments ROS production, resulting in highly efficient bacterial elimination at low temperature. Precisely, injectable hydrogel dressing can match irregular wound sites, which shortens the distance of heat dissipation and ROS diffusion to bacteria, thus improving sterilization efficacy and decreasing non-specific systemic toxicity. Both in vitro and in vivo experiments validated the predominant sterilization efficiency of drug-resistant methicillin-resistant Staphylococcus aureus (MRSA) and kanamycin-resistant Escherichia coli (KREC), presenting great potential for application in clinical therapy.


Subject(s)
Anti-Bacterial Agents , Copper , Photothermal Therapy , Reactive Oxygen Species , Catalysis , Copper/chemistry , Copper/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Reactive Oxygen Species/metabolism , Methicillin-Resistant Staphylococcus aureus/drug effects , Escherichia coli/drug effects , Animals , Mice , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Zirconium/chemistry , Zirconium/pharmacology , Cold Temperature , Microbial Sensitivity Tests , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/chemistry , Particle Size , Surface Properties , Hydrogels/chemistry , Hydrogels/pharmacology
14.
Acta Biomater ; 180: 183-196, 2024 May.
Article in English | MEDLINE | ID: mdl-38604465

ABSTRACT

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.


Subject(s)
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
15.
Int J Biol Macromol ; 267(Pt 2): 131606, 2024 May.
Article in English | MEDLINE | ID: mdl-38631566

ABSTRACT

This study aimed to investigate the effect of cinnamon essential oil (CEO)-loaded metal-organic frameworks (CEO@MOF) on the properties of gelatin/pullulan (Gel/Pull)-based composite films (Gel/Pull-based films). The incorporation of CEO@MOF into Gel/Pull-based films demonstrated significant antimicrobial activity against S. aureus, S. enterica, E. coli, and L. monocytogenes. Additionally, CEO@MOF integrated film exhibited a 98.16 % ABTS radical scavenging, with no significant change in the mechanical properties of the neat Gel/Pull film. The UV blocking efficiency of the composite films increased significantly from 81.38 to 99.56 % at 280 nm with the addition of 3 wt% CEO@MOF. Additionally, Gel/Pull/CEO@MOF films effectively extended the shelf life of meat preserved at 4 °C by reducing moisture loss by 3.35 %, maintaining the pH within the threshold limit (6.2), and inhibiting bacterial growth by 99.9 %. These results propose that CEO@MOF has significant potential as an effective additive in active packaging to improve shelf life and food safety.


Subject(s)
Cinnamomum zeylanicum , Food Packaging , Gelatin , Glucans , Metal-Organic Frameworks , Oils, Volatile , Gelatin/chemistry , Oils, Volatile/chemistry , Oils, Volatile/pharmacology , Cinnamomum zeylanicum/chemistry , Food Packaging/methods , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Glucans/chemistry , Glucans/pharmacology , Food Preservation/methods , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Meat/microbiology , Animals , Microbial Sensitivity Tests
16.
Int J Biol Macromol ; 266(Pt 2): 131330, 2024 May.
Article in English | MEDLINE | ID: mdl-38570003

ABSTRACT

The challenge of drug resistance in bacteria caused by the over use of biotics is increasing during the therapy process, which has attracted great attentions of the clinicians and scientists around the world. Recently, photodynamic therapy (PDT) triggered by photosensitizer (PS) has become a promising treatment method because of its high efficacy, easy operation, and low side effect. Herein, the poly-l-lysine (PLL) modified metal-organic framework (MOF) nanoparticles, ZIF/PLL-CIP/CUR, were synthesized to allow both reactive oxygen species (ROS) responsive drug release and photodynamic effect for synergistic therapy against drug resistant bacterial infections. The PLL was modified on the shell of the zeolite imidazole framework (ZIF) by the ROS-responsive thioketal linker for controllable CIP release. CUR were encapsulated in ZIF as the photosensitizer for blue light mediated photodynamic effect to produce singlet oxygen (1O2) and superoxide anion radical (O2-) for efficient inhibition towards methicillin-resistant Staphylococcus aureus (MRSA). The charge conversion from negative charge (-4.6 mV) to positive charge (2.6 mV) was observed at pH 7.4 and pH 5.5, and 70.9 % CIP was found released at pH 5.5 in the presence of H2O2, which suggests the good biosafety at physiological pH and ROS-responsive drug release of the as-prepared nanoparticle in the bacterial microenvironment. The as-prepared nanoparticles could effectively kill MRSA and disrupt bacterial biofilm by combination of chemo- and photodynamic therapy. In mice model, the as-prepared nanoparticles exhibited excellent biosafety and synergistic effect with 98.81 % healing rate in treatment of MRSA infection, which is considered as a promising candidate in combating drug resistant bacterial infection.


Subject(s)
Metal-Organic Frameworks , Methicillin-Resistant Staphylococcus aureus , Nanoparticles , Photochemotherapy , Photosensitizing Agents , Polylysine , Reactive Oxygen Species , Polylysine/chemistry , Polylysine/pharmacology , Photochemotherapy/methods , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Nanoparticles/chemistry , Animals , Mice , Reactive Oxygen Species/metabolism , Hydrogen-Ion Concentration , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Drug Liberation , Curcumin/pharmacology , Curcumin/chemistry , Staphylococcal Infections/drug therapy
17.
ACS Nano ; 18(12): 9100-9113, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38478044

ABSTRACT

Reactive oxygen species (ROS) mediated tumor cell death is a powerful anticancer strategy. Cuproptosis is a copper-dependent and ROS-mediated prospective tumor therapy strategy. However, the complex tumor microenvironment (TME), low tumor specificity, poor therapy efficiency, and lack of imaging capability impair the therapy output of current cuproptosis drugs. Herein, we designed a dual-responsive two-dimensional metal-organic framework (2D MOF) nanotheranostic via a coordination self-assembly strategy using Au(III) tetra-(4-pyridyl) porphine (AuTPyP) as the ligand and copper ions (Cu2+) as nodes. The dual-stimulus combined with the protonation of the pyridyl group in AuTPyP and deep-penetration ultrasound (US) together triggered the controlled release in an acidic TME. The ultrathin structure (3.0 nm) of nanotheranostics promoted the release process. The released Cu2+ was reduced to Cu+ by depleting the overexpressed glutathione (GSH) in the tumor, which not only activated the Ferredoxin 1 (FDX1)-mediated cuproptosis but also catalyzed the overexpressed hydrogen peroxide (H2O2) in the tumor into reactive oxygen species via Fenton-like reaction. Simultaneously, the released AuTPyP could specifically bind with thioredoxin reductase and activate the redox imbalance of tumor cells. These together selectively induced significant mitochondrial vacuoles and prominent tumor cell death but did not damage the normal cells. The fluorescence and magnetic resonance imaging (MRI) results verified this nanotheranostic could target the HeLa tumor to greatly promote the self-enhanced effect of chemotherapy/cuproptosis and tumor inhibition efficiency. The work helped to elucidate the controlled assembly of multiresponsive nanotheranostics and the high-specificity ROS regulation for application in anticancer therapy.


Subject(s)
Metal-Organic Frameworks , Nanoparticles , Neoplasms , Humans , Copper , Metal-Organic Frameworks/pharmacology , Reactive Oxygen Species , Hydrogen Peroxide , Prospective Studies , Glutathione , Hydrogen-Ion Concentration , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Cell Line, Tumor , Tumor Microenvironment
18.
Redox Biol ; 71: 103106, 2024 May.
Article in English | MEDLINE | ID: mdl-38442647

ABSTRACT

Cytoprotection has emerged as an effective therapeutic strategy for mitigating brain injury following acute ischemic stroke (AIS). The sulfonylurea receptor 1-transient receptor potential M4 (SUR1-TRPM4) channel plays a pivotal role in brain edema and neuroinflammation. However, the practical use of the inhibitor glyburide (GLB) is hindered by its low bioavailability. Additionally, the elevated reactive oxygen species (ROS) after AIS exacerbate SUR1-TRPM4 activation, contributing to irreversible brain damage. To overcome these challenges, GLB and superoxide dismutase (SOD) were embedded in a covalent organic framework (COF) with a porous structure and great stability. The resulting S/G@COF demonstrated significant improvements in survival and neurological functions. This was achieved by eliminating ROS, preventing neuronal loss and apoptosis, suppressing neuroinflammation, modulating microglia activation, and ameliorating blood-brain barrier (BBB) disruption. Mechanistic investigations revealed that S/G@COF concurrently activated the Wnt/ß-catenin signaling pathway while suppressing the upregulation of SUR1-TRPM4. This study underscores the potential of employing multi-target therapy and drug modification in cytoprotective strategies for ischemic stroke.


Subject(s)
Ischemic Stroke , Metal-Organic Frameworks , Stroke , Humans , Ischemic Stroke/drug therapy , Metal-Organic Frameworks/metabolism , Metal-Organic Frameworks/pharmacology , Neuroinflammatory Diseases , Reactive Oxygen Species/metabolism , Blood-Brain Barrier , Glyburide/metabolism , Glyburide/pharmacology , Glyburide/therapeutic use , Stroke/drug therapy , Stroke/metabolism
19.
Int J Biol Macromol ; 265(Pt 1): 130797, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38479662

ABSTRACT

In recent years, photocatalytic technology has been introduced to develop a new kind antimicrobial agents fighting antibiotic abusing and related drug resistance. The efforts have focused on non-precious metal photocatalysts along with green additives. In the present work, a novel bis-S heterojunctions based on the coupling of polysaccharide (CS) and bismuth-based MOF (CAU-17) s synthesized through a two-step method involving amidation reaction under mild conditions. The as prepared photocatalyst literally extended the light response to the near-infrared region. Owing to its double S-type heterostructure, the lifetime of the photocarriers is significantly prolonged and the redox capacity are enhanced. As a result, the as prepared photocatalyst indicated inhibition up to 99.9 % under 20 min of light exposure against Gram-positive (S. aureus) and Gram-negative (E. coli) bacteria as well as drug-resistant bacteria (MRSA). The outstanding photocatalytic performance is attributed to the effective charge separation and migration due to the unique double S heterostructure. Such a double S heterostructure was confirmed through transient photocurrent response, electrochemical impedance spectroscopy tests and electron spin resonance measurements. The present work provides a basis for the simple synthesis of high-performance heterojunction photocatalytic inhibitors, which extends the application of CAU-17 in environmental disinfection and wastewater purification.


Subject(s)
Chitosan , Metal-Organic Frameworks , Bismuth/chemistry , Escherichia coli , Chitosan/pharmacology , Metal-Organic Frameworks/pharmacology , Staphylococcus aureus , Catalysis
20.
Sci Bull (Beijing) ; 69(9): 1286-1301, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38519399

ABSTRACT

Adavosertib (ADA) is a WEE1 inhibitor that exhibits a synthetic lethal effect on p53-mutated gallbladder cancer (GBC). However, drug resistance due to DNA damage response compensation pathways and high toxicity limits further applications. Herein, estrone-targeted ADA-encapsulated metal-organic frameworks (ADA@MOF-EPL) for GBC synthetic lethal treatment by inducing conditional factors are developed. The high expression of estrogen receptors in GBC enables ADA@MOF-EPL to quickly enter and accumulate near the cell nucleus through estrone-mediated endocytosis and release ADA to inhibit WEE1 upon entering the acidic tumor microenvironment. Ultrasound irradiation induces ADA@MOF-EPL to generate reactive oxygen species (ROS), which leads to a further increase in DNA damage, resulting in a higher sensitivity of p53-mutated cancer cells to WEE1 inhibitor and promoting cell death via conditional synthetic lethality. The conditional factor induced by ADA@MOF-EPL further enhances the antitumor efficacy while significantly reducing systemic toxicity. Moreover, ADA@MOF-EPL demonstrates similar antitumor abilities in other p53-mutated solid tumors, revealing its potential as a broad-spectrum antitumor drug.


Subject(s)
Antineoplastic Agents , Gallbladder Neoplasms , Metal-Organic Frameworks , Protein-Tyrosine Kinases , Pyrimidinones , Tumor Suppressor Protein p53 , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Gallbladder Neoplasms/drug therapy , Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/pathology , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Protein-Tyrosine Kinases/antagonists & inhibitors , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Synthetic Lethal Mutations , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays , Mutation , Mice, Nude , DNA Damage/drug effects , Female
SELECTION OF CITATIONS
SEARCH DETAIL
...