Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 602
1.
J Nanobiotechnology ; 22(1): 294, 2024 May 28.
Article En | MEDLINE | ID: mdl-38807127

BACKGROUND: Ulcerative colitis (UC) is one chronic and relapsing inflammatory bowel disease. Macrophage has been reputed as one trigger for UC. Recently, phosphodiesterase 4 (PDE4) inhibitors, for instance roflumilast, have been regarded as one latent approach to modulating macrophage in UC treatment. Roflumilast can decelerate cyclic adenosine monophosphate (cAMP) degradation, which impedes TNF-α synthesis in macrophage. However, roflumilast is devoid of macrophage-target and consequently causes some unavoidable adverse reactions, which restrict the utilization in UC. RESULTS: Membrane vesicles (MVs) from probiotic Escherichia coli Nissle 1917 (EcN 1917) served as a drug delivery platform for targeting macrophage. As model drugs, roflumilast and MnO2 were encapsulated in MVs (Rof&MnO2@MVs). Roflumilast inhibited cAMP degradation via PDE4 deactivation and MnO2 boosted cAMP generation by activating adenylate cyclase (AC). Compared with roflumilast, co-delivery of roflumilast and MnO2 apparently produced more cAMP and less TNF-α in macrophage. Besides, Rof&MnO2@MVs could ameliorate colitis in mouse model and regulate gut microbe such as mitigating pathogenic Escherichia-Shigella and elevating probiotic Akkermansia. CONCLUSIONS: A probiotic-based nanoparticle was prepared for precise codelivery of roflumilast and MnO2 into macrophage. This biomimetic nanoparticle could synergistically modulate cAMP in macrophage and ameliorate experimental colitis.


Aminopyridines , Benzamides , Cyclic AMP , Cyclopropanes , Macrophages , Manganese Compounds , Oxides , Probiotics , Animals , Aminopyridines/pharmacology , Mice , Cyclic AMP/metabolism , Probiotics/pharmacology , Cyclopropanes/pharmacology , Cyclopropanes/chemistry , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Benzamides/pharmacology , Benzamides/chemistry , Oxides/pharmacology , Oxides/chemistry , Macrophages/drug effects , Macrophages/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/chemistry , Colitis/drug therapy , Colitis/chemically induced , RAW 264.7 Cells , Escherichia coli/drug effects , Tumor Necrosis Factor-alpha/metabolism , Mice, Inbred C57BL , Male , Disease Models, Animal
2.
J Nanobiotechnology ; 22(1): 234, 2024 May 09.
Article En | MEDLINE | ID: mdl-38724978

Radiotherapy-induced immune activation holds great promise for optimizing cancer treatment efficacy. Here, we describe a clinically used radiosensitizer hafnium oxide (HfO2) that was core coated with a MnO2 shell followed by a glucose oxidase (GOx) doping nanoplatform (HfO2@MnO2@GOx, HMG) to trigger ferroptosis adjuvant effects by glutathione depletion and reactive oxygen species production. This ferroptosis cascade potentiation further sensitized radiotherapy by enhancing DNA damage in 4T1 breast cancer tumor cells. The combination of HMG nanoparticles and radiotherapy effectively activated the damaged DNA and Mn2+-mediated cGAS-STING immune pathway in vitro and in vivo. This process had significant inhibitory effects on cancer progression and initiating an anticancer systemic immune response to prevent distant tumor recurrence and achieve long-lasting tumor suppression of both primary and distant tumors. Furthermore, the as-prepared HMG nanoparticles "turned on" spectral computed tomography (CT)/magnetic resonance dual-modality imaging signals, and demonstrated favorable contrast enhancement capabilities activated by under the GSH tumor microenvironment. This result highlighted the potential of nanoparticles as a theranostic nanoplatform for achieving molecular imaging guided tumor radiotherapy sensitization induced by synergistic immunotherapy.


Ferroptosis , Immunotherapy , Manganese Compounds , Membrane Proteins , Mice, Inbred BALB C , Nanoparticles , Nucleotidyltransferases , Oxides , Radiation-Sensitizing Agents , Animals , Mice , Immunotherapy/methods , Oxides/chemistry , Oxides/pharmacology , Female , Nucleotidyltransferases/metabolism , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Cell Line, Tumor , Nanoparticles/chemistry , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/chemistry , Membrane Proteins/metabolism , Ferroptosis/drug effects , Glucose Oxidase/metabolism , Reactive Oxygen Species/metabolism , Humans , DNA Damage , Tumor Microenvironment/drug effects
3.
J Nanobiotechnology ; 22(1): 264, 2024 May 17.
Article En | MEDLINE | ID: mdl-38760771

Glioblastoma (GBM) is the most aggressive primary brain tumor with low survival rate. Currently, temozolomide (TMZ) is the first-line drug for GBM treatment of which efficacy is unfortunately hindered by short circulation time and drug resistance associated to hypoxia and redox tumor microenvironment. Herein, a dual-targeted and multi-responsive nanoplatform is developed by loading TMZ in hollow manganese dioxide nanoparticles functionalized by polydopamine and targeting ligands RAP12 for photothermal and receptor-mediated dual-targeted delivery, respectively. After accumulated in GBM tumor site, the nanoplatform could respond to tumor microenvironment and simultaneously release manganese ion (Mn2+), oxygen (O2) and TMZ. The hypoxia alleviation via O2 production, the redox balance disruption via glutathione consumption and the reactive oxygen species generation, together would down-regulate the expression of O6-methylguanine-DNA methyltransferase under TMZ medication, which is considered as the key to drug resistance. These strategies could synergistically alleviate hypoxia microenvironment and overcome TMZ resistance, further enhancing the anti-tumor effect of chemotherapy/chemodynamic therapy against GBM. Additionally, the released Mn2+ could also be utilized as a magnetic resonance imaging contrast agent for monitoring treatment efficiency. Our study demonstrated that this nanoplatform provides an alternative approach to the challenges including low delivery efficiency and drug resistance of chemotherapeutics, which eventually appears to be a potential avenue in GBM treatment.


Brain Neoplasms , Drug Resistance, Neoplasm , Glioblastoma , Manganese Compounds , Nanoparticles , Oxides , Temozolomide , Tumor Microenvironment , Glioblastoma/drug therapy , Glioblastoma/metabolism , Temozolomide/pharmacology , Temozolomide/therapeutic use , Tumor Microenvironment/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Cell Line, Tumor , Animals , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Nanoparticles/chemistry , Brain Neoplasms/drug therapy , Oxides/chemistry , Oxides/pharmacology , Mice , Drug Delivery Systems/methods , Indoles/chemistry , Indoles/pharmacology , Polymers/chemistry , Mice, Nude , Mice, Inbred BALB C , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Reactive Oxygen Species/metabolism
4.
J Colloid Interface Sci ; 670: 499-508, 2024 Sep 15.
Article En | MEDLINE | ID: mdl-38776685

Manganese oxide nanoparticles (MONs)-based contrast agents have attracted increasing attention for magnetic resonance imaging (MRI), attributed to their good biocompatibility and advantageous paramagnetism. However, conventional MONs have poor imaging performance due to low T1 relaxivity. Additionally, their lack of tumor-targeting theranostics capabilities and complex synthesis pathways have impeded clinical applications. Rutin (Ru) is an ideal tumor-targeted ligand that targets glucose transporters (GLUTs) overexpressed in various malignant tumors, and exhibits photothermal effects upon chelation with metal ions. Herein, a series of Ru-coated MONs (Ru/MnO2) were synthesized using a straightforward, rapid one-step process. Specifically, Ru/MnO2-5, with the smallest crystal size of approximately 4 nm, exhibits the highest T1 relaxivity (33.3 mM-1s-1 at 1.5 T, surpassing prior MONs) along with notable stability, photothermal efficacy, and tumor-targeting ability. Furthermore, Ru/MnO2-5 shows promise in MRI and photothermal therapy of H22 tumors owing to its superior GLUTs-mediated tumor-targeting capability.


Magnetic Resonance Imaging , Manganese Compounds , Nanoparticles , Oxides , Photothermal Therapy , Rutin , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Oxides/chemistry , Oxides/pharmacology , Animals , Nanoparticles/chemistry , Rutin/chemistry , Rutin/pharmacology , Mice , Humans , Particle Size , Surface Properties , Contrast Media/chemistry , Cell Survival/drug effects , Cell Line, Tumor , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Neoplasms/diagnostic imaging , Neoplasms/therapy , Neoplasms/drug therapy
5.
Acta Biomater ; 181: 402-414, 2024 Jun.
Article En | MEDLINE | ID: mdl-38734282

Tumor hypoxia, high oxidative stress, and low immunogenic create a deep-rooted immunosuppressive microenvironment, posing a major challenge to the therapeutic efficiency of cancer immunotherapy for solid tumor. Herein, an intelligent nanoplatform responsive to the tumor microenvironment (TME) capable of hypoxia relief and immune stimulation has been engineered for efficient solid tumor immunotherapy. The MnO2@OxA@OMV nanoreactor, enclosing bacterial-derived outer membrane vesicles (OMVs)-wrapped MnO2 nanoenzyme and the immunogenic cell death inducer oxaliplatin (OxA), demonstrated intrinsic catalase-like activity within the TME, which effectively catalyzed the endogenous H2O2 into O2 to enable a prolonged oxygen supply, thereby alleviating the tumor's oxidative stress and hypoxic TME, and expediting OxA release. The combinational action of OxA-caused ICD effect and Mn2+ from nanoreactor enabled the motivation of the cGAS-STING pathway to significantly improve the activation of STING and dendritic cells (DCs) maturation, resulting in metalloimmunotherapy. Furthermore, the immunostimulant OMVs played a crucial role in promoting the infiltration of activated CD8+T cells into the solid tumor. Overall, the nanoreactor offers a robust platform for solid tumor treatment, highlighting the significant potential of combining relief from tumor hypoxia and immune stimulation for metalloimmunotherapy. STATEMENT OF SIGNIFICANCE: A tailor-made nanoreactor was fabricated by enclosing bacterial-derived outer membrane vesicles (OMVs) onto MnO2 nanoenzyme and loading with immunogenic cell death inducer oxaliplatin (OxA) for tumor metalloimmunotherapy. The nanoreactor possesses intrinsic catalase-like activity within the tumor microenvironment, which effectively enabled a prolonged oxygen supply by catalyzing the conversion of endogenous H2O2 into O2, thereby alleviating tumor hypoxia and expediting OxA release. Furthermore, the TME-responsive release of nutritional Mn2+ sensitized the cGAS-STING pathway and collaborated with OxA-induced immunogenic cell death (ICD). Combing with immunostimulatory OMVs enhances the uptake of nanoreactors by DCs and promotes the infiltration of activated CD8+T cells. This nanoreactor offers a robust platform for solid tumor treatment, highlighting the significant potential of combining relief from tumor hypoxia and immune stimulation for metalloimmunotherapy.


Immunotherapy , Tumor Microenvironment , Animals , Immunotherapy/methods , Mice , Tumor Microenvironment/drug effects , Cell Line, Tumor , Tumor Hypoxia/drug effects , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Oxaliplatin/pharmacology , Oxaliplatin/chemistry , Oxides/chemistry , Oxides/pharmacology , Manganese/chemistry , Manganese/pharmacology , Humans , Female , Neoplasms/therapy , Neoplasms/pathology , Neoplasms/immunology , Neoplasms/drug therapy , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dendritic Cells/immunology , Mice, Inbred C57BL
6.
ACS Appl Mater Interfaces ; 16(21): 27187-27201, 2024 May 29.
Article En | MEDLINE | ID: mdl-38747985

Development of theranostic nanomedicines to tackle glioma remains to be challenging. Here, we present an advanced blood-brain barrier (BBB)-crossing nanovaccine based on cancer cell membrane-camouflaged poly(N-vinylcaprolactam) (PVCL) nanogels (NGs) incorporated with MnO2 and doxorubicin (DOX). We show that the disulfide bond-cross-linked redox-responsive PVCL NGs can be functionalized with dermorphin and imiquimod R837 through cell membrane functionalization. The formed functionalized PVCL NGs having a size of 220 nm are stable, can deplete glutathione, and responsively release both Mn2+ and DOX under the simulated tumor microenvironment to exert the chemo/chemodynamic therapy mediated by DOX and Mn2+, respectively. The combined therapy induces tumor immunogenic cell death to maturate dendritic cells (DCs) and activate tumor-killing T cells. Further, the nanovaccine composed of cancer cell membranes as tumor antigens, R837 as an adjuvant with abilities of DC maturation and macrophages M1 repolarization, and MnO2 with Mn2+-mediated stimulator of interferon gene activation of tumor cells can effectively act on both targets of tumor cells and immune cells. With the dermorphin-mediated BBB crossing, cell membrane-mediated homologous tumor targeting, and Mn2+-facilitated magnetic resonance (MR) imaging property, the designed NG-based theranostic nanovaccine enables MR imaging and combination chemo-, chemodynamic-, and imnune therapy of orthotopic glioma with a significantly decreased recurrence rate.


Glioma , Magnetic Resonance Imaging , Manganese Compounds , Theranostic Nanomedicine , Glioma/diagnostic imaging , Glioma/drug therapy , Glioma/therapy , Glioma/pathology , Animals , Mice , Humans , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Cancer Vaccines/chemistry , Immunotherapy , Oxides/chemistry , Oxides/pharmacology , Cell Line, Tumor , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/drug therapy , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Blood-Brain Barrier/metabolism , Nanogels/chemistry , Imiquimod/chemistry , Imiquimod/pharmacology , Nanovaccines
7.
J Colloid Interface Sci ; 666: 244-258, 2024 Jul 15.
Article En | MEDLINE | ID: mdl-38598997

Starvation therapy has shown promise as a cancer treatment, but its efficacy is often limited when used alone. In this work, a multifunctional nanoscale cascade enzyme system, named CaCO3@MnO2-NH2@GOx@PVP (CMGP), was fabricated for enhanced starvation/chemodynamic combination cancer therapy. CMGP is composed of CaCO3 nanoparticles wrapped in a MnO2 shell, with glucose oxidase (GOx) adsorbed and modified with polyvinylpyrrolidone (PVP). MnO2 decomposes H2O2 in cancer cells into O2, which enhances the efficiency of GOx-mediated starvation therapy. CaCO3 can be decomposed in the acidic cancer cell environment, causing Ca2+ overload in cancer cells and inhibiting mitochondrial metabolism. This synergizes with GOx to achieve more efficient starvation therapy. Additionally, the H2O2 and gluconic acid produced during glucose consumption by GOx are utilized by MnO2 with catalase-like activity to enhance O2 production and Mn2+ release. This process accelerates glucose consumption, reactive oxygen species (ROS) generation, and CaCO3 decomposition, promoting the Ca2+ release. CMGP can alleviate tumor hypoxia by cycling the enzymatic cascade reaction, which increases enzyme activity and combines with Ca2+ overload to achieve enhanced combined starvation/chemodynamic therapy. In vitro and in vivo studies demonstrate that CMGP has effective anticancer abilities and good biosafety. It represents a new strategy with great potential for combined cancer therapy.


Calcium Carbonate , Glucose Oxidase , Manganese Compounds , Oxides , Glucose Oxidase/metabolism , Glucose Oxidase/chemistry , Glucose Oxidase/pharmacology , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Oxides/chemistry , Oxides/pharmacology , Humans , Animals , Calcium Carbonate/chemistry , Calcium Carbonate/pharmacology , Calcium Carbonate/metabolism , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Nanoparticles/chemistry , Povidone/chemistry , Povidone/pharmacology , Tumor Hypoxia/drug effects , Reactive Oxygen Species/metabolism , Cell Survival/drug effects , Particle Size , Cell Line, Tumor , Hydrogen Peroxide/metabolism , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Surface Properties , Mice, Inbred BALB C
8.
Int J Biol Macromol ; 268(Pt 2): 131871, 2024 May.
Article En | MEDLINE | ID: mdl-38677691

Multifunctional hydrogels have been developed to meet the various requirements of wound healing. Herein, an innovative hydrogel (QCMC-HA-PEG) was formed through the Schiff base reaction, composed of quaternary ammonium-modified carboxymethyl chitosan (QCMC), hyaluronic acid (HA), and 8-arms Polyethylene Glycol aldehyde (8-ARM-PEG-CHO). The resulting hydrogels exhibited good mechanical and adhesive properties with improved antibacterial efficacy against both Gram-positive and Gram-negative bacteria compared to CMC hydrogels. QCMC-HA-PEG hydrogels demonstrated remarkable adhesive ability in lap-shear test. Furthermore, the incorporation of MnO2 nanosheets into the hydrogel significantly enhanced its reactive oxygen species (ROS) scavenging and oxygen generation capabilities. Finally, experimental results from a full-thickness skin wound model revealed that the QCMC-HA-PEG@MnO2 hydrogel promoted skin epithelization, collagen deposition, and inflammatory regulation significantly accelerated the wound healing process. Therefore, QCMC-HA-PEG@MnO2 hydrogel could be a promising wound dressing to promote wound healing.


Anti-Bacterial Agents , Antioxidants , Chitosan , Hydrogels , Quaternary Ammonium Compounds , Wound Healing , Chitosan/chemistry , Chitosan/analogs & derivatives , Chitosan/pharmacology , Wound Healing/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Animals , Quaternary Ammonium Compounds/chemistry , Quaternary Ammonium Compounds/pharmacology , Antioxidants/pharmacology , Antioxidants/chemistry , Reactive Oxygen Species/metabolism , Mice , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Oxides/chemistry , Oxides/pharmacology , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Skin/drug effects
9.
Water Res ; 256: 121608, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38657310

The application of ozone (O3) disinfection has been hindered by its low solubility in water and the formation of disinfection by-products (DBPs). In this study, capacitive disinfection is applied as a pre-treatment for O3 oxidation, in which manganese dioxide with a rambutan-like hollow spherical structure is used as the electrode to increase the charge density on the electrode surface. When a voltage is applied, the negative-charged microbes are attracted to the electrodes and killed by electrical interactions. The contact between microbes and capacitive electrodes leads to changes in cell permeability and burst of reactive oxygen species, thereby promoting the diffusion of O3 into the cells. After O3 penetrates the cell membrane, it can directly attack the cytoplasmic constituents, accelerating fatal and irreversible damage to pathogens. As a result, the performance of the capacitance-O3 process is proved better than the direct sum of the two individual process efficiencies. The design of capacitance-O3 system is beneficial to reduce the ozone dosage and DBPs with a broader inactivation spectrum, which is conducive to the application of ozone in primary water disinfection.


Disinfection , Manganese Compounds , Oxides , Ozone , Ozone/pharmacology , Ozone/chemistry , Oxides/pharmacology , Oxides/chemistry , Disinfection/methods , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Cell Membrane/drug effects , Water Purification/methods , Electrodes , Bacteria/drug effects
10.
Colloids Surf B Biointerfaces ; 238: 113921, 2024 Jun.
Article En | MEDLINE | ID: mdl-38631280

Tumor microenvironment (TME)-responsive size-changeable and biodegradable nanoplatforms for multimodal therapy possess huge advantages in anti-tumor therapy. Hence, we developed a hyaluronic acid (HA) modified CuS/MnO2 nanosheets (HCMNs) as a multifunctional nanoplatform for synergistic chemodynamic therapy (CDT)/photothermal therapy (PTT)/photodynamic therapy (PDT). The prepared HCMNs exhibited significant NIR light absorption and photothermal conversion efficiency because of the densely deposited ultra-small sized CuS nanoparticles on the surface of MnO2 nanosheet. They could precisely target the tumor cells and rapidly decomposed into small sized nanostructures in the TME, and then efficiently promote intracellular ROS generation through a series of cascade reactions. Moreover, the local temperature elevation induced by photothermal effect also promote the PDT based on CuS nanoparticles and the Fenton-like reaction of Mn2+, thereby enhancing the therapeutic efficiency. Furthermore, the T1-weighted magnetic resonance (MR) imaging was significantly enhanced by the abundant Mn2+ ions from the decomposition process of HCMNs. In addition, the CDT/PTT/PDT synergistic therapy using a single NIR light source exhibited considerable anti-tumor effect via in vitro cell test. Therefore, the developed HCMNs will provide great potential for MR imaging and multimodal synergistic cancer therapy.


Copper , Hyaluronic Acid , Magnetic Resonance Imaging , Manganese Compounds , Oxides , Photochemotherapy , Tumor Microenvironment , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Tumor Microenvironment/drug effects , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Oxides/chemistry , Oxides/pharmacology , Humans , Copper/chemistry , Copper/pharmacology , Particle Size , Nanostructures/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Phototherapy , Nanoparticles/chemistry , Cell Survival/drug effects , Surface Properties , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Drug Screening Assays, Antitumor , Animals
11.
ACS Nano ; 18(19): 12453-12467, 2024 May 14.
Article En | MEDLINE | ID: mdl-38686995

Traditional magnetic resonance imaging (MRI) contrast agents (CAs) are a type of "always on" system that accelerates proton relaxation regardless of their enrichment region. This "always on" feature leads to a decrease in signal differences between lesions and normal tissues, hampering their applications in accurate and early diagnosis. Herein, we report a strategy to fabricate glutathione (GSH)-responsive one-dimensional (1-D) manganese oxide nanoparticles (MONPs) with improved T2 relaxivities and achieve effective T2/T1 switchable MRI imaging of tumors. Compared to traditional contrast agents with high saturation magnetization to enhance T2 relaxivities, 1-D MONPs with weak Ms effectively increase the inhomogeneity of the local magnetic field and exhibit obvious T2 contrast. The inhomogeneity of the local magnetic field of 1-D MONPs is highly dependent on their number of primary particles and surface roughness according to Landau-Lifshitz-Gilbert simulations and thus eventually determines their T2 relaxivities. Furthermore, the GSH responsiveness ensures 1-D MONPs with sensitive switching from the T2 to T1 mode in vitro and subcutaneous tumors to clearly delineate the boundary of glioma and metastasis margins, achieving precise histopathological-level MRI. This study provides a strategy to improve T2 relaxivity of magnetic nanoparticles and construct switchable MRI CAs, offering high tumor-to-normal tissue contrast signal for early and accurate diagnosis.


Contrast Media , Magnetic Resonance Imaging , Manganese Compounds , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Animals , Mice , Contrast Media/chemistry , Humans , Magnetic Fields , Glutathione/chemistry , Oxides/chemistry , Cell Line, Tumor , Glioma/diagnostic imaging , Glioma/pathology , Particle Size , Magnetite Nanoparticles/chemistry
12.
ACS Biomater Sci Eng ; 10(5): 3188-3202, 2024 May 13.
Article En | MEDLINE | ID: mdl-38592024

Chronic wound repair is a clinical treatment challenge. The development of multifunctional hydrogels is of great significance in the key aspects of treating chronic wounds, including reducing oxidative stress, promoting angiogenesis, and improving the natural remodeling of extracellular matrix and immune regulation. In this study, we prepared a composite hydrogel, sodium alginate (SA)@MnO2/recombinant humanized collagen III (RHC)/mesenchymal stem cells (MSCs), composed of SA, MnO2 nanoparticles, RHC, and MSCs. The hydrogel has high mechanical properties and good biocompatibility. In vitro, SA@MnO2/RHC/MSCs hydrogel effectively enhanced the formation of intricate tubular structures and angiogenesis and showed synergistic effects on cell proliferation and migration. In vivo, the SA@MnO2/RHC/MSCs hydrogel enhanced diabetes wound healing, rapid re-epithelization, favorable collagen deposition, and abundant wound angiogenesis. These findings demonstrated that the combined effects of SA, MnO2, RHC, and MSCs synergistically accelerate healing, resulting in a reduced healing time. These observed healing effects demonstrated the potential of this multifunctional hydrogel to transform chronic wound care and improve patient outcomes.


Hydrogels , Manganese Compounds , Mesenchymal Stem Cells , Oxides , Wound Healing , Wound Healing/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Animals , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Humans , Oxides/chemistry , Oxides/pharmacology , Diabetes Mellitus, Experimental , Cell Proliferation/drug effects , Collagen/chemistry , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Alginates/chemistry , Alginates/pharmacology , Male , Mice
13.
Molecules ; 29(6)2024 Mar 21.
Article En | MEDLINE | ID: mdl-38543041

Design of amyloid ß-protein (Aß) inhibitors is considered an effective strategy for the prevention and treatment of Alzheimer's disease (AD). However, the limited blood-brain barrier (BBB) penetration and poor Aß-targeting capability restricts the therapeutic efficiency of candidate drugs. Herein, we have proposed to engineer transthyretin (TTR) by fusion of the Aß-targeting peptide KLVFF and cell-penetrating peptide Penetratin to TTR, and derived a fusion protein, KLVFF-TTR-Penetratin (KTP). Moreover, to introduce the scavenging activity for reactive oxygen species (ROS), a nanocomposite of KTP and manganese dioxide nanoclusters (KTP@MnO2) was fabricated by biomineralization. Results revealed that KTP@MnO2 demonstrated significantly enhanced inhibition on Aß aggregation as compared to TTR. The inhibitory effect was increased from 18%, 33%, and 49% (10, 25, and 50 µg/mL TTR, respectively) to 52%, 81%, and 100% (10, 25, and 50 µg/mL KTP@MnO2). In addition, KTP@MnO2 could penetrate the BBB and target amyloid plaques. Moreover, multiple ROS, including hydroxyl radicals, superoxide radicals, hydrogen peroxide, and Aß-induced-ROS, which cannot be scavenged by TTR, were scavenged by KTP@MnO2, thus resulting in the mitigation of cellular oxidative damages. More importantly, cell culture and in vivo experiments with AD nematodes indicated that KTP@MnO2 at 50 µg/mL increased the viability of Aß-treated cells from 66% to more than 95%, and completely cleared amyloid plaques in AD nematodes and extended their lifespan by 7 d. Overall, despite critical aspects such as the stability, metabolic distribution, long-term biotoxicity, and immunogenicity of the nanocomposites in mammalian models remaining to be investigated, this work has demonstrated the multifunctionality of KTP@MnO2 for targeting Aß in vivo, and provided new insights into the design of multifunctional nanocomposites of protein-metal clusters against AD.


Alzheimer Disease , Cell-Penetrating Peptides , Peptide Fragments , Animals , Humans , Amyloid beta-Peptides/metabolism , Alzheimer Disease/metabolism , Manganese Compounds/pharmacology , Oxides/pharmacology , Prealbumin/metabolism , Reactive Oxygen Species/metabolism , Plaque, Amyloid/metabolism , Mammals/metabolism
14.
J Colloid Interface Sci ; 665: 188-203, 2024 Jul.
Article En | MEDLINE | ID: mdl-38522159

Anti-tumor therapies reliant on reactive oxygen species (ROS) as primary therapeutic agents face challenges due to a limited oxygen substrate. Photodynamic therapy (PDT) is particularly hindered by inherent hypoxia, while chemodynamic therapy (CDT) encounters obstacles from insufficient endogenous hydrogen peroxide (H2O2) levels. In this study, we engineered biodegradable tumor microenvironment (TME)-activated hollow mesoporous MnO2-based nanotheranostic agents, designated as HAMnO2A. This construct entails loading artemisinin (ART) into the cavity and surface modification with a mussel-inspired polymer ligand, namely hyaluronic acid-linked poly(ethylene glycol)-diethylenetriamine-conjugated (3,4-dihydroxyphenyl) acetic acid, and the photosensitizer Chlorin e6 (mPEG-HA-Dien-(Dhpa/Ce6)), facilitating dual-modal imaging-guided PDT/CDT synergistic therapy. In vitro experimentation revealed that HAMnO2A exhibited ideal physiological stability and enhanced cellular uptake capability via CD44-mediated endocytosis. Additionally, it was demonstrated that accelerated endo-lysosomal escape through the pH-dependent protonation of Dien. Within the acidic and highly glutathione (GSH)-rich TME, the active component of HAMnO2A, MnO2, underwent decomposition, liberating oxygen and releasing both Mn2+ and ART. This process alleviates hypoxia within the tumor region and initiates a Fenton-like reaction through the combination of ART and Mn2+, thereby enhancing the effectiveness of PDT and CDT by generating increased singlet oxygen (1O2) and hydroxyl radicals (•OH). Moreover, the presence of Mn2+ ions enabled the activation of T1-weighted magnetic resonance imaging. In vivo findings further validated that HAMnO2A displayed meaningful tumor-targeting capabilities, prolonged circulation time in the bloodstream, and outstanding efficacy in restraining tumor growth while inducing minimal damage to normal tissues. Hence, this nanoplatform serves as an efficient all-in-one solution by facilitating the integration of multiple functions, ultimately enhancing the effectiveness of tumor theranostics.


Nanoparticles , Neoplasms , Photochemotherapy , Humans , Photochemotherapy/methods , Manganese Compounds/pharmacology , Manganese Compounds/chemistry , Tumor Microenvironment , Theranostic Nanomedicine/methods , Hydrogen Peroxide/chemistry , Oxides/chemistry , Photosensitizing Agents/chemistry , Neoplasms/drug therapy , Oxygen , Hypoxia/drug therapy , Cell Line, Tumor , Nanoparticles/chemistry
15.
Nanoscale ; 16(12): 6095-6108, 2024 Mar 21.
Article En | MEDLINE | ID: mdl-38444228

In photothermal therapy (PTT), the photothermal conversion of the second near-infrared (NIR-II) window allows deeper penetration and higher laser irradiance and is considered a promising therapeutic strategy for deep tissues. Since cancer remains a leading cause of deaths worldwide, despite the numerous treatment options, we aimed to develop an improved bionic nanotheranostic for combined imaging and photothermal cancer therapy. We combined a gold nanobipyramid (Au NBP) as a photothermal agent and MnO2 as a magnetic resonance enhancer to produce core/shell structures (Au@MnO2; AM) and modified their surfaces with homologous cancer cell plasma membranes (PM) to enable tumour targeting. The performance of the resulting Au@MnO2@PM (AMP) nanotheranostic was evaluated in vitro and in vivo. AMP exhibits photothermal properties under NIR-II laser irradiation and has multimodal in vitro imaging functions. AMP enables the computed tomography (CT), photothermal imaging (PTI), and magnetic resonance imaging (MRI) of tumours. In particular, AMP exhibited a remarkable PTT effect on cancer cells in vitro and inhibited tumour cell growth under 1064 nm laser irradiation in vivo, with no significant systemic toxicity. This study achieved tumour therapy guided by multimodal imaging, thereby demonstrating a novel strategy for the use of bionic gold nanoparticles for tumour PTT under NIR-II laser irradiation.


Metal Nanoparticles , Nanoparticles , Neoplasms , Humans , Phototherapy/methods , Photothermal Therapy , Theranostic Nanomedicine/methods , Gold/pharmacology , Manganese Compounds/pharmacology , Manganese Compounds/chemistry , Bionics , Metal Nanoparticles/therapeutic use , Oxides , Neoplasms/diagnostic imaging , Neoplasms/therapy , Multimodal Imaging/methods , Cell Line, Tumor
16.
ACS Appl Mater Interfaces ; 16(11): 13543-13562, 2024 Mar 20.
Article En | MEDLINE | ID: mdl-38452225

We use low-molecular-weight branched polyethylenimine (PEI) to produce cytocompatible reduced graphene oxide quantum dots (rGOQD) as a photothermal agent and covalently bind it with the photosensitizer IR-820. The rGOQD/IR820 shows high photothermal conversion efficiency and produces reactive oxygen species (ROS) after irradiation with near-infrared (NIR) light for photothermal/photodynamic therapy (PTT/PDT). To improve suspension stability, rGOQD/IR820 was PEGylated by anchoring with the DSPE hydrophobic tails in DSPE-PEG-Mal, leaving the maleimide (Mal) end group for covalent binding with manganese dioxide/bovine serum albumin (MnO2/BSA) and targeting ligand cell-penetrating peptide (CPP) to synthesize rGOQD/IR820/MnO2/CPP. As MnO2 can react with intracellular hydrogen peroxide to produce oxygen for alleviating the hypoxia condition in the acidic tumor microenvironment, the efficacy of PDT could be enhanced by generating more cytotoxic ROS with NIR light. Furthermore, quercetin (Q) was loaded to rGOQD through π-π interaction, which can be released in the endosomes and act as an inhibitor of heat shock protein 70 (HSP70). This sensitizes tumor cells to thermal stress and increases the efficacy of mild-temperature PTT with NIR irradiation. By simultaneously incorporating the HSP70 inhibitor (Q) and the in situ hypoxia alleviating agent (MnO2), the rGOQD/IR820/MnO2/Q/CPP can overcome the limitation of PTT/PDT and enhance the efficacy of targeted phototherapy in vitro. From in vivo study with an orthotopic brain tumor model, rGOQD/IR820/MnO2/Q/CPP administered through tail vein injection can cross the blood-brain barrier and accumulate in the intracranial tumor, after which NIR laser light irradiation can shrink the tumor and prolong the survival times of animals by simultaneously enhancing the efficacy of PTT/PDT to treat glioblastoma.


Antineoplastic Agents , Glioblastoma , Graphite , Photochemotherapy , Quantum Dots , Animals , Manganese Compounds/pharmacology , Manganese Compounds/chemistry , Glioblastoma/drug therapy , Quantum Dots/therapeutic use , Heat-Shock Proteins , Reactive Oxygen Species , Tumor Hypoxia , Oxides/pharmacology , Oxides/chemistry , Phototherapy , Hypoxia , Cell Line, Tumor , Tumor Microenvironment
17.
ACS Appl Mater Interfaces ; 16(14): 17120-17128, 2024 Apr 10.
Article En | MEDLINE | ID: mdl-38554083

Cell-based therapies offer tremendous potential for skin flap regeneration. However, the hostile microenvironment of the injured tissue adversely affects the longevity and paracrine effects of the implanted cells, severely reducing their therapeutic effectiveness. Here, an injectable hydrogel (nGk) with reactive oxygen species (ROS) scavenging capability, which can amplify the cell viability and functions of encapsulated mesenchymal stem cells (MSCs), is employed to promote skin flap repair. nGk is formulated by dispersing manganese dioxide nanoparticles (MnO2 NPs) in a gelatin/κ-carrageenan hydrogel, which exhibits satisfactory injectable properties and undergoes a sol-gel phase transition at around 40 °C, leading to the formation of a solid gel at physiological temperature. MnO2 NPs enhance the mechanical properties of the hydrogel and give it the ability to scavenge ROS, thus providing a cell-protective system for MSCs. Cell culture studies show that nGk can mitigate the oxidative stress, improve cell viability, and boost stem cell paracrine function to promote angiogenesis. Furthermore, MSC-loaded nGk (nGk@MSCs) can improve the survival of skin flaps by promoting angiogenesis, reducing inflammatory reactions, and attenuating necrosis, providing an effective approach for tissue regeneration. Collectively, injectable nGk has substantial potential to enhance the therapeutic benefits of MSCs, making it a valuable delivery system for cell-based therapies.


Hydrogels , Mesenchymal Stem Cells , Hydrogels/pharmacology , Reactive Oxygen Species/pharmacology , Manganese Compounds/pharmacology , Oxides/pharmacology
18.
J Nanobiotechnology ; 22(1): 98, 2024 Mar 09.
Article En | MEDLINE | ID: mdl-38461231

Chemodynamic therapy (CDT) based on intracellular Fenton reaction to produce highly cytotoxic reactive oxygen species (ROS) has played an essential role in tumor therapy. However, this therapy still needs to be improved by weakly acidic pH and over-expression of glutathione (GSH) in tumor microenvironment (TEM), which hinders its future application. Herein, we reported a multifunctional bimetallic composite nanoparticle MnO2@GA-Fe@CAI based on a metal polyphenol network (MPN) structure, which could reduce intracellular pH and endogenous GSH by remodeling tumor microenvironment to improve Fenton activity. MnO2 nanoparticles were prepared first and MnO2@GA-Fe nanoparticles with Fe3+ as central ion and gallic acid (GA) as surface ligands were prepared by the chelation reaction. Then, carbonic anhydrase inhibitor (CAI) was coupled with GA to form MnO2@GA-Fe@CAI. The properties of the bimetallic composite nanoparticles were studied, and the results showed that CAI could reduce intracellular pH. At the same time, MnO2 could deplete intracellular GSH and produce Mn2+ via redox reactions, which re-established the TME with low pH and GSH. In addition, GA reduced Fe3+ to Fe2+. Mn2+ and Fe2+ catalyzed the endogenous H2O2 to produce high-lever ROS to kill tumor cells. Compared with MnO2, MnO2@GA-Fe@CAI could reduce the tumor weight and volume for the xenograft MDA-MB-231 tumor-bearing mice and the final tumor inhibition rate of 58.09 ± 5.77%, showing the improved therapeutic effect as well as the biological safety. Therefore, this study achieved the high-efficiency CDT effect catalyzed by bimetallic through reshaping the tumor microenvironment.


Nanoparticles , Neoplasms , Triple Negative Breast Neoplasms , Humans , Animals , Mice , Hydrogen Peroxide , Manganese Compounds/pharmacology , Reactive Oxygen Species , Oxides , Gallic Acid , Glutathione , Hydrogen-Ion Concentration , Cell Line, Tumor , Tumor Microenvironment
19.
ACS Appl Bio Mater ; 7(3): 1790-1800, 2024 Mar 18.
Article En | MEDLINE | ID: mdl-38424007

A sustainable approach for pharmaceutically important pyrimidine derivatives is achieved by using biogenically produced single-phase δ-MnO2 NPs under external ligand-free conditions. The phytochemicals that comprise the extract of Areca Nut Husk (ANH) have been discovered to serve as reducing agents. The role of phytochemicals is not only to aid in the reduction of Mn(VII) into Mn(IV), but they also have an important role in stabilizing the catalyst. The establishment of δ-MnO2 NPs was confirmed inveterate by FE-SEM, p-XRD, ICP-OES (Mn content = 43.17% w/w), EDX, and with an active Mn content of 43.17% w/w. A series of pyrimidine derivatives were prepared in good yields using a one-pot multicomponent synthesis approach under atmospheric conditions. In addition, hot filtration tests, control experiments, gram-scale synthesis, and mechanistic investigations were demonstrated. Additionally, antimicrobial activity studies of δ-MnO2 NPs and pyrimidine derivatives against the Gram-negative bacteria E. coli, growth curve and minimum inhibitory concentration were studied.


Anti-Infective Agents , Nanoparticles , Escherichia coli , Manganese Compounds/pharmacology , Oxides , Nanoparticles/therapeutic use , Pyrimidines/pharmacology , Anti-Infective Agents/pharmacology
20.
Plant Physiol Biochem ; 207: 108428, 2024 Feb.
Article En | MEDLINE | ID: mdl-38364633

Manganese (Mn) is one of the essential elements for plant growth and is involved in the process of photosynthesis and seed germination. Herein, we applied two Mn-based nanoparticles, MnO2 and Mn3O4, to the soil to investigate their effects on radish growth, antioxidant system, and nutrients. The radish plant height after treatment with 10 mg/kg of MnO2 and Mn3O4 NPs were increased, compare to the control. In radish's shoot, MnO2 NPs at high concentrations (100 mg/kg) increased MDA activity by 58 % compared to the control group, while exposure to Mn3O4 NPs at the same concentration decreased MDA activity by 14 %. The nutrient content of radishes, such as soluble sugar and vitamin C, was improved. Moreover, single particle inductively coupled plasma mass spectrometry (SP ICP-MS) was used to understand the patterns of migration of Mn-based NPs in radish and subsequent impact on nutrients. We found that Mn-based NPs accumulated mainly in the roots of radish. Interestingly, the accumulation characteristics of MnO2 NPs and Mn3O4 NPs were different. MnO2 NPs accumulated more in radish leaves than in fruits, while the accumulation of Mn3O4 NPs gradually decreased from roots to leaves. Finally, we determined the mineral element content of the leaves, fruits, and roots of radish, and found that the uptake of main metallic mineral elements (e.g. Cu, Fe, Mg, Zn, Na, K) was inhibited by the application of Mn-based NPs. These findings underscore the importance of considering species and multifaceted impacts of Mn-based NPs as nanofertilizers for their wide application in agriculture.


Nanoparticles , Raphanus , Raphanus/chemistry , Manganese/pharmacology , Manganese Compounds/pharmacology , Oxides/pharmacology , Minerals/pharmacology
...