Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.571
Filtrar
1.
J Nanobiotechnology ; 22(1): 230, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38720322

RESUMEN

Tumor vaccines, a crucial immunotherapy, have gained growing interest because of their unique capability to initiate precise anti-tumor immune responses and establish enduring immune memory. Injected tumor vaccines passively diffuse to the adjacent draining lymph nodes, where the residing antigen-presenting cells capture and present tumor antigens to T cells. This process represents the initial phase of the immune response to the tumor vaccines and constitutes a pivotal determinant of their effectiveness. Nevertheless, the granularity paradox, arising from the different requirements between the passive targeting delivery of tumor vaccines to lymph nodes and the uptake by antigen-presenting cells, diminishes the efficacy of lymph node-targeting tumor vaccines. This study addressed this challenge by employing a vaccine formulation with a tunable, controlled particle size. Manganese dioxide (MnO2) nanoparticles were synthesized, loaded with ovalbumin (OVA), and modified with A50 or T20 DNA single strands to obtain MnO2/OVA/A50 and MnO2/OVA/T20, respectively. Administering the vaccines sequentially, upon reaching the lymph nodes, the two vaccines converge and simultaneously aggregate into MnO2/OVA/A50-T20 particles through base pairing. This process enhances both vaccine uptake and antigen delivery. In vitro and in vivo studies demonstrated that, the combined vaccine, comprising MnO2/OVA/A50 and MnO2/OVA/T20, exhibited robust immunization effects and remarkable anti-tumor efficacy in the melanoma animal models. The strategy of controlling tumor vaccine size and consequently improving tumor antigen presentation efficiency and vaccine efficacy via the DNA base-pairing principle, provides novel concepts for the development of efficient tumor vaccines.


Asunto(s)
Vacunas contra el Cáncer , Ganglios Linfáticos , Compuestos de Manganeso , Ratones Endogámicos C57BL , Nanopartículas , Ovalbúmina , Óxidos , Animales , Vacunas contra el Cáncer/inmunología , Ganglios Linfáticos/inmunología , Ratones , Ovalbúmina/inmunología , Ovalbúmina/química , Óxidos/química , Nanopartículas/química , Compuestos de Manganeso/química , Inmunidad Celular , Femenino , Línea Celular Tumoral , ADN/química , ADN/inmunología , Inmunoterapia/métodos , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Tamaño de la Partícula , Antígenos de Neoplasias/inmunología
2.
Biomed Mater ; 19(4)2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38697132

RESUMEN

During the process of malignant tumor treatment, photodynamic therapy (PDT) exerts poor efficacy due to the hypoxic environment of the tumor cells, and long-time chemotherapy reduces the sensitivity of tumor cells to chemotherapy drugs due to the presence of drug-resistant proteins on the cell membranes for drug outward transportation. Therefore, we reported a nano platform based on mesoporous silica coated with polydopamine (MSN@PDA) loading PDT enhancer MnO2, photosensitizer indocyanine green (ICG) and chemotherapeutic drug doxorubicin (DOX) (designated as DMPIM) to achieve a sequential release of different drugs to enhance treatment of malignant tumors. MSN was first synthesized by a template method, then DOX was loaded into the mesoporous channels of MSN, and locked by the PDA coating. Next, ICG was modified by π-π stacking on PDA, and finally, MnO2layer was accumulated on the surface of DOX@MSN@PDA- ICG@MnO2, achieving orthogonal loading and sequential release of different drugs. DMPIM first generated oxygen (O2) through the reaction between MnO2and H2O2after entering tumor cells, alleviating the hypoxic environment of tumors and enhancing the PDT effect of sequentially released ICG. Afterwards, ICG reacted with O2in tumor tissue to produce reactive oxygen species, promoting lysosomal escape of drugs and inactivation of p-glycoprotein (p-gp) on tumor cell membranes. DOX loaded in the MSN channels exhibited a delay of approximately 8 h after ICG release to exert the enhanced chemotherapy effect. The drug delivery system achieved effective sequential release and multimodal combination therapy, which achieved ideal therapeutic effects on malignant tumors. This work offers a route to a sequential drug release for advancing the treatment of malignant tumors.


Asunto(s)
Doxorrubicina , Liberación de Fármacos , Verde de Indocianina , Indoles , Compuestos de Manganeso , Óxidos , Fotoquimioterapia , Fármacos Fotosensibilizantes , Polímeros , Fotoquimioterapia/métodos , Doxorrubicina/química , Doxorrubicina/farmacología , Doxorrubicina/administración & dosificación , Verde de Indocianina/química , Indoles/química , Animales , Compuestos de Manganeso/química , Humanos , Polímeros/química , Línea Celular Tumoral , Óxidos/química , Fármacos Fotosensibilizantes/química , Dióxido de Silicio/química , Ratones , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/administración & dosificación , Neoplasias/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Sistemas de Liberación de Medicamentos , Nanopartículas/química , Portadores de Fármacos/química , Porosidad
3.
J Nanobiotechnology ; 22(1): 294, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38807127

RESUMEN

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.


Asunto(s)
Aminopiridinas , Benzamidas , AMP Cíclico , Ciclopropanos , Macrófagos , Compuestos de Manganeso , Óxidos , Probióticos , Animales , Aminopiridinas/farmacología , Ratones , AMP Cíclico/metabolismo , Probióticos/farmacología , Ciclopropanos/farmacología , Ciclopropanos/química , Compuestos de Manganeso/química , Compuestos de Manganeso/farmacología , Benzamidas/farmacología , Benzamidas/química , Óxidos/farmacología , Óxidos/química , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Inhibidores de Fosfodiesterasa 4/farmacología , Inhibidores de Fosfodiesterasa 4/química , Colitis/tratamiento farmacológico , Colitis/inducido químicamente , Células RAW 264.7 , Escherichia coli/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Ratones Endogámicos C57BL , Masculino , Modelos Animales de Enfermedad
4.
J Nanobiotechnology ; 22(1): 264, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760771

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas , Resistencia a Antineoplásicos , Glioblastoma , Compuestos de Manganeso , Nanopartículas , Óxidos , Temozolomida , Microambiente Tumoral , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Temozolomida/farmacología , Temozolomida/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Línea Celular Tumoral , Animales , Compuestos de Manganeso/química , Compuestos de Manganeso/farmacología , Nanopartículas/química , Neoplasias Encefálicas/tratamiento farmacológico , Óxidos/química , Óxidos/farmacología , Ratones , Sistemas de Liberación de Medicamentos/métodos , Indoles/química , Indoles/farmacología , Polímeros/química , Ratones Desnudos , Ratones Endogámicos BALB C , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo
5.
J Nanobiotechnology ; 22(1): 234, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724978

RESUMEN

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.


Asunto(s)
Ferroptosis , Inmunoterapia , Compuestos de Manganeso , Proteínas de la Membrana , Ratones Endogámicos BALB C , Nanopartículas , Nucleotidiltransferasas , Óxidos , Fármacos Sensibilizantes a Radiaciones , Animales , Ratones , Inmunoterapia/métodos , Óxidos/química , Óxidos/farmacología , Femenino , Nucleotidiltransferasas/metabolismo , Compuestos de Manganeso/química , Compuestos de Manganeso/farmacología , Línea Celular Tumoral , Nanopartículas/química , Fármacos Sensibilizantes a Radiaciones/farmacología , Fármacos Sensibilizantes a Radiaciones/química , Proteínas de la Membrana/metabolismo , Ferroptosis/efectos de los fármacos , Glucosa Oxidasa/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Humanos , Daño del ADN , Microambiente Tumoral/efectos de los fármacos
6.
Anal Chim Acta ; 1308: 342664, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38740454

RESUMEN

Nanozymes is a kind of nanomaterials with enzyme catalytic properties. Compared with natural enzymes, nanozymes merge the advantages of both nanomaterials and natural enzymes, which is highly important in applications such as biosensing, clinical diagnosis, and food inspection. In this study, we prepared ß-MnOOH hexagonal nanoflakes with a high oxygen vacancy ratio by utilizing SeO2 as a sacrificial agent. The defect-rich MnOOH hexagonal nanoflakes demonstrated excellent oxidase-like activity, catalyzing the oxidation substrate in the presence of O2, thereby rapidly triggering a color reaction. Consequently, a colorimetric sensing platform was constructed to assess the total antioxidant capacity in commercial beverages. The strategy of introducing defects in situ holds great significance for the synthesis of a series of high-performance metal oxide nanozymes, driving the development of faster and more efficient biosensing and analysis methods.


Asunto(s)
Antioxidantes , Compuestos de Manganeso , Óxidos , Óxidos/química , Antioxidantes/química , Antioxidantes/metabolismo , Antioxidantes/análisis , Compuestos de Manganeso/química , Colorimetría , Oxidorreductasas/química , Oxidorreductasas/metabolismo , Oxidación-Reducción , Nanoestructuras/química , Catálisis
7.
Environ Sci Pollut Res Int ; 31(24): 35678-35687, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38740682

RESUMEN

This study effectively addresses the rapid deactivation of manganese-based catalysts in humid environments during ozone decomposition by introducing iron-doped manganese oxide octahedral molecular sieve (Fe-OMS-2) catalysts supported on activated carbon (AC). By optimizing the doping ratio of Fe-OMS-2, the Fe-OMS-20.5/AC catalyst achieves nearly 100% ozone decomposition efficiency across a wide range of relative humidity levels (0 to 60%), even at elevated air flow rates of 800 L·g-1·h-1, outperforming standalone AC, Fe-OMS-2, or a simple mixture of OMS-2 and AC. The Fe-OMS-20.5/AC catalyst features a porous surface and a mesoporous structure, providing a substantial specific surface area that facilitates the uniform distribution of the Fe-OMS-2 active phase on the AC surface. The incorporation of Fe3+ ions enhances electron transfer between valence state transitions of Mn, thereby improving the catalyst's efficiency in ozone decomposition. Additionally, the AC component protects catalytic sites and enhances the catalyst's humidity resistance. In conclusion, this research presents a novel strategy for developing highly efficient and cost-effective ozone decomposition catalysts that enhance dehumidification, significantly contributing to industrial ozone treatment technologies and advancing environmental protection.


Asunto(s)
Carbón Orgánico , Humedad , Hierro , Ozono , Ozono/química , Carbón Orgánico/química , Hierro/química , Catálisis , Óxidos/química , Carbono/química , Compuestos de Manganeso/química
8.
ACS Appl Mater Interfaces ; 16(21): 27187-27201, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38747985

RESUMEN

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.


Asunto(s)
Glioma , Imagen por Resonancia Magnética , Compuestos de Manganeso , Nanomedicina Teranóstica , Glioma/diagnóstico por imagen , Glioma/tratamiento farmacológico , Glioma/terapia , Glioma/patología , Animales , Ratones , Humanos , Compuestos de Manganeso/química , Compuestos de Manganeso/farmacología , Doxorrubicina/química , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Vacunas contra el Cáncer/química , Inmunoterapia , Óxidos/química , Óxidos/farmacología , Línea Celular Tumoral , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/patología , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Barrera Hematoencefálica/metabolismo , Nanogeles/química , Imiquimod/química , Imiquimod/farmacología , Nanovacunas
9.
Anal Chem ; 96(21): 8814-8821, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38751335

RESUMEN

Highly responsive interface of semiconductor nanophotoelectrochemical materials provides a broad development prospect for the identification of low-abundance cancer marker molecules. This work innovatively proposes an efficient blank WO3/SnIn4S8 heterojunction interface formed by self-assembly on the working electrode for interface regulation and photoregulation. Different from the traditional biomolecular layered interface, a hydrogel layer containing manganese dioxide with a wide light absorption range is formed at the interface after an accurate response to external immune recognition. The formation of the hydrogel layer hinders the effective contact between the heterojunction interface and the electrolyte solution, and manganese dioxide in the hydrogel layer forms a strong competition between the light source and the substrate photoelectric material. The process effectively improves the carrier recombination efficiency at the interface, reduces the interface reaction kinetics and photoelectric conversion efficiency, and thus provides strong support for target identification. Taking advantage of the process, the resulting biosensors are being explored for sensitive detection of human epidermal growth factor receptor 2, with a limit of detection as low as 0.037 pg/mL. Also, this study contributes to the advancement of photoelectrochemical biosensing technology and opens up new avenues for the development of sensitive and accurate analytical tools in the field of bioanalysis.


Asunto(s)
Técnicas Biosensibles , Técnicas Electroquímicas , Compuestos de Manganeso , Óxidos , Receptor ErbB-2 , Humanos , Técnicas Electroquímicas/métodos , Óxidos/química , Compuestos de Manganeso/química , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo , Hidrogeles/química , Procesos Fotoquímicos , Límite de Detección , Electrodos , Inmunoensayo/métodos , Tungsteno/química
10.
Mikrochim Acta ; 191(5): 239, 2024 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-38570399

RESUMEN

To accurately detect tumor marker carbohydrate antigen 72-4 (CA72-4) of serum samples is of great significance for the early diagnosis of malignant tumors. In the present study, MnO2/hollow nanobox metal-organic framework (HNM)-AuPtPd nanocomposites were prepared via multi-step synthesis and superposition method and a series of characterizations were carried out. A highly sensitive immunosensor Ab/MnO2/HNM-AuPtPd/GCE based on the composite nanomaterial was further prepared and used to detect the tumor marker CA72-4. The constructed immunosensor achieved signal amplification by increasing the electrocatalytic activity to H2O2 by means of the synergistic effect of MnO2 ultra-thin nanosheets (MnO2 UNs) and HNM-AuPtPd. At the same time, the electrochemical properties of the immunosensor were analyzed using cyclic voltammetry, electrochemical impedance, amperometry (with the test voltage of -0.4 V), and differential pulse voltammetry. The experimental results showed that the MnO2/HNM-AuPtPd nanocomposites were successfully prepared, and the immunosensor Ab/MnO2/HNM-AuPtPd/GCE demonstrated an excellent electrochemical performance. The electrochemical immunosensor had the highest detection sensitivity under the optimal experimental conditions, such as incubation pH of 7.0, incubation time of 60 min, with the addition of 15 µL of H2O2, and in the concentration range 0.001-500 U/mL. It had a low detection limit of 1.78×10-5 U/mL (S/N = 3). Moreover, the serum sample recovery were in the range from 99.38 to 100.52%. This study provides a new method and experimental basis for the detection of tumor markers in clinical practice.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores , Técnicas Biosensibles , Nanocompuestos , Biomarcadores de Tumor , Técnicas Biosensibles/métodos , Peróxido de Hidrógeno/química , Compuestos de Manganeso/química , Óxidos/química , Inmunoensayo , Nanocompuestos/química
12.
Free Radic Res ; 58(3): 194-216, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38563404

RESUMEN

Microwave (MW) radiations are widely used in communications, radar and medical treatment and thus human exposure to MW radiations have increased tremendously, raising health concerns as MW has been implicated in induction of oxidative stress condition in our body. Few metallic nanoparticles (NPs) have been shown to mimic the activity of antioxidant enzymes and hence can be applied for the modulation of adverse effects caused by MW. Present study aimed to assess the biocompatibility of Bovine serum albumin (BSA) conjugated manganese dioxide nanoparticles (MNP*) and to counteract the impact of MW on the haematological system of male Wistar rats. Experiments were conducted in two sets. Set I involved biodistribution and antioxidant activity evaluation of MNP* at different doses. Results showed a dose-dependent increase in antioxidant potential and significant biodistribution in the liver, spleen, kidney, and testis, with no organ damage, indicating its biocompatibility. Experiment set II constituted the study of separate and combined effects of MW and MNP* on haematological parameters, oxidative status, and genotoxic study in the blood of rats. MW exposure significantly altered red blood cell count, hemoglobin, packed cell volume percentage, monocyte percentage, aspartate aminotransferase, Alanine aminotransferase and uric acid. MW also induced significant DNA damage in the blood. A significant increase in lipid peroxidation and a decrease in antioxidant enzyme superoxide dismutase was also observed in MW exposed group. However, these alterations were reduced significantly when MNP* was administered. Thus, MNP* showed biocompatibility and modulatory effects against MW-induced alterations in the haematological system of rats.


Asunto(s)
Compuestos de Manganeso , Microondas , Nanopartículas , Óxidos , Ratas Wistar , Albúmina Sérica Bovina , Animales , Masculino , Compuestos de Manganeso/química , Ratas , Albúmina Sérica Bovina/química , Óxidos/química , Nanopartículas/química , Estrés Oxidativo/efectos de los fármacos , Bovinos , Antioxidantes/farmacología , Nanopartículas del Metal/química
13.
Mikrochim Acta ; 191(5): 282, 2024 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-38652326

RESUMEN

A novel dual-mode fluorometric and colorimetric sensing platform is reported for determining glutathione S-transferase (GST) by utilizing polyethyleneimine-capped silver nanoclusters (PEI-AgNCs) and cobalt-manganese oxide nanosheets (CoMn-ONSs) with oxidase-like activity. Abundant active oxygen species (O2•-) can be produced through the CoMn-ONSs interacting with dissolved oxygen. Afterward, the pink oxDPD was generated through the oxidation of colorless N,N-diethyl-p-phenylenediamine (DPD) by O2•-, and two absorption peaks at 510 and 551 nm could be observed. Simultaneously, oxDPD could quench the fluorescence of PEI-AgNCs at 504 nm via the inner filter effect (IFE). However, in the presence of glutathione (GSH), GSH prevents the oxidation of DPD due to the reducibility of GSH, leading to the absorbance decrease at 510 and 551 nm. Furthermore, the fluorescence at 504 nm was restored due to the quenching effect of oxDPD on decreased PEI-AgNCs. Under the catalysis of GST, GSH and1-chloro-2,4-dinitrobenzo (CDNB) conjugate to generate an adduct, initiating the occurrence of the oxidation of the chromogenic substrate DPD, thereby inducing a distinct colorimetric response again and the significant quenching of PEI-AgNCs. The detection limits for GST determination were 0.04 and 0.21 U/L for fluorometric and colorimetric modes, respectively. The sensing platform illustrated reliable applicability in detecting GST in real samples.


Asunto(s)
Cobalto , Colorimetría , Glutatión Transferasa , Compuestos de Manganeso , Nanopartículas del Metal , Óxidos , Polietileneimina , Plata , Polietileneimina/química , Plata/química , Cobalto/química , Óxidos/química , Compuestos de Manganeso/química , Nanopartículas del Metal/química , Colorimetría/métodos , Glutatión Transferasa/metabolismo , Glutatión Transferasa/química , Límite de Detección , Oxidorreductasas/química , Oxidorreductasas/metabolismo , Humanos , Glutatión/química , Oxidación-Reducción , Técnicas Biosensibles/métodos , Fenilendiaminas/química , Nanoestructuras/química
14.
Int J Pharm ; 656: 124093, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38583822

RESUMEN

A multifunctional nanoplatform was constructed in this work, with the goal of ameliorating the challenges faced with traditional cancer chemotherapy. Cisplatin (CP) was loaded into mesoporous polydopamine (mPDA) nanoparticles (NPs) with a drug loading of 15.8 ± 0.1 %, and MnO2 used as pore sealing agent. Finally, the NPs were wrapped with platelet membrane (PLTM). P-selectin on the PLTM can bind to CD44, which is highly expressed on the tumor cell membrane, so as to improve the targeting performance of the NPs. In addition, the CD47 on the PLTM can prevent the NPs from being phagocytosed by macrophages, which is conducive to immune escape. The final PLTM-CP@mPDA/MnO2 NPs were found to have a particle size of approximately 198 nm. MnO2 is degraded into Mn2+ in the tumor microenvironment, leading to CP release from the pores in the mPDA. CP both acts as a chemotherapy agent and can also increase the concentration of H2O2 in cells. Mn2+ can catalyze the conversion of H2O2 to OH, resulting in oxidative damage and chemodynamic therapy. In addition, Mn2+ can be used as a contrast agent in magnetic resonance imaging (MRI). In vitro and in vivo experiments were performed to explore the therapeutic effect of the NPs. When the concentration of CP is 30 µg/mL, the NPs cause approximately 50 % cell death. It was found that the PLTM-CP@mPDA/MnO2 NPs are targeted to cancerous cells, and in the tumor site cause extensive apoptosis. Tumor growth is thereby repressed. No negative off-target side effects were noted. MRI could be used to confirm the presence of the NPs in the tumor site. Overall, the nano-platform developed here provides cooperative chemotherapy and chemodynamic therapy, and can potentially be used for effective cancer treatment which could be monitored by MRI.


Asunto(s)
Antineoplásicos , Plaquetas , Cisplatino , Indoles , Compuestos de Manganeso , Nanopartículas , Óxidos , Polímeros , Compuestos de Manganeso/química , Cisplatino/administración & dosificación , Cisplatino/farmacología , Cisplatino/química , Polímeros/química , Indoles/química , Indoles/administración & dosificación , Animales , Óxidos/química , Nanopartículas/química , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Humanos , Ratones , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Liberación de Fármacos , Porosidad , Ratones Endogámicos BALB C , Imagen por Resonancia Magnética , Portadores de Fármacos/química , Femenino , Peróxido de Hidrógeno , Tamaño de la Partícula , Ratones Desnudos
15.
J Mater Chem B ; 12(19): 4724-4735, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38655674

RESUMEN

We have developed a highly sensitive and reliable fluorescence resonance energy transfer (FRET) probe using nitro-dopamine (ND) and dopamine (DA) coated MnO2 nanosheet (ND@MnO2 NS and DA@MnO2 NS) as an energy acceptor and MoS2 quantum dots (QDs) as an energy donor. By employing surface-modified MnO2 NS, we can effectively reduce the fluorescence intensity of MoS2 QDs through FRET. It can reduce MnO2 NS to Mn2+ and facilitate the fluorescence recovery of the MoS2 QDs. This ND@MnO2 NS@MoS2 QD-based nanoprobe demonstrates excellent sensitivity to GSH, achieving an LOD of 22.7 nM in an aqueous medium while exhibiting minimal cytotoxicity and good biocompatibility. Moreover, our sensing platform shows high selectivity to GSH towards various common biomolecules and electrolytes. Confocal fluorescence imaging revealed that the nanoprobe can image GSH in A549 cells. Interestingly, the ND@MnO2 NS nanoprobe demonstrates no cytotoxicity in living cancer cells, even at concentrations up to 100 µg mL-1. Moreover, the easy fabrication and eco-friendliness of ND@MnO2 NS make it a rapid and simple method for detecting GSH. We envision the developed nanoprobe as an incredible platform for real-time monitoring of GSH levels in both extracellular and intracellular mediums, proving valuable for biomedical research and clinical diagnostics.


Asunto(s)
Disulfuros , Dopamina , Glutatión , Compuestos de Manganeso , Molibdeno , Nanocompuestos , Óxidos , Puntos Cuánticos , Humanos , Compuestos de Manganeso/química , Disulfuros/química , Óxidos/química , Puntos Cuánticos/química , Molibdeno/química , Glutatión/análisis , Glutatión/química , Dopamina/análisis , Nanocompuestos/química , Transferencia Resonante de Energía de Fluorescencia , Células A549 , Tamaño de la Partícula , Colorantes Fluorescentes/química , Colorantes Fluorescentes/síntesis química
16.
Spectrochim Acta A Mol Biomol Spectrosc ; 316: 124352, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-38678841

RESUMEN

Mucin 1 is an essential tumor biomarker, and developing cost-effective and portable methods for mucin 1 detection is crucial in resource-limited settings. Herein, the pH-regulated dual-enzyme mimic activities of manganese dioxide nanosheets were demonstrated, which were integrated into an aptasensor for dual-mode detection of mucin 1. Under acidic conditions, manganese dioxide nanosheets with oxidase mimic activities catalyzed the oxidation of 3,3',5,5'-tetramethylbenzidine sulfate, producing visible multicolor signals; while under basic conditions, manganese dioxide nanosheets with catalase mimic activities were used as catalyst for the decomposition of hydrogen peroxide, generating gas pressure signals. The proposed method allows the naked eye detection of mucin 1 through multicolor signal readout and the quantitative detection of mucin 1 with a handheld pressure meter or a UV-vis spectrophotometer. The study demonstrates that manganese dioxide nanosheets with pH-regulated dual-enzyme mimic activities can facilitate multidimensional transducing signals. The use of manganese dioxide nanosheets for the transduction of different signals avoids extra labels and simplifies the operation procedures. Besides, the signal readout mode can be selected according to the available detection instruments. Therefore, the use of manganese dioxide nanosheets with pH-regulated dual-enzyme mimic activities for dual-signal readout provides a new way for mucin 1 detection.


Asunto(s)
Compuestos de Manganeso , Mucina-1 , Nanoestructuras , Óxidos , Compuestos de Manganeso/química , Concentración de Iones de Hidrógeno , Mucina-1/análisis , Óxidos/química , Nanoestructuras/química , Humanos , Colorimetría/métodos , Bencidinas/química , Presión , Técnicas Biosensibles/métodos , Peróxido de Hidrógeno/análisis , Peróxido de Hidrógeno/química , Aptámeros de Nucleótidos/química
17.
ACS Biomater Sci Eng ; 10(5): 3188-3202, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38592024

RESUMEN

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.


Asunto(s)
Hidrogeles , Compuestos de Manganeso , Células Madre Mesenquimatosas , Óxidos , Cicatrización de Heridas , Cicatrización de Heridas/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Animales , Compuestos de Manganeso/química , Compuestos de Manganeso/farmacología , Hidrogeles/química , Hidrogeles/farmacología , Humanos , Óxidos/química , Óxidos/farmacología , Diabetes Mellitus Experimental , Proliferación Celular/efectos de los fármacos , Colágeno/química , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Alginatos/química , Alginatos/farmacología , Masculino , Ratones
18.
Anal Chim Acta ; 1303: 342520, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38609255

RESUMEN

BACKGROUND: Cluster of Differentiation 44 (CD44) is considered an important biomarker for various cancers, and achieving highly sensitive detection of CD44 is crucial, which plays a significant role in tumor invasion and metastasis, providing essential information for clinical tumor diagnosis. Commonly used methods for analysis include fluorescence spectroscopy (FL), photoelectrochemical analysis (PEC), electrochemical analysis (EC), and commercial ELISA kits. Although these methods offer high sensitivity, they can be relatively complex to perform experimentally. Electrochemiluminescence (ECL) has gained widespread research attention due to its high sensitivity, ease of operation, effective spatiotemporal control, and close to zero background signal. RESULTS: In this work, a sandwich-type ECL immunosensor for detecting CD44 was constructed using luminol as a luminophore. In this sensing platform, bimetallic MOFs (Pd@FeNi-MIL-88B) loaded with palladium nanoparticles (Pd NPs) were used as a novel enzyme mimic, exhibiting excellent catalytic performance towards the electroreduction of H2O2. The hybrids provided a strong support platform for luminol and antibodies, significantly enhancing the initial ECL signal of luminol. Subsequently, core-shell Au@MnO2 nanocomposites were synthesised by gold nanoparticles (Au NPs) encapsulated in manganese dioxide (MnO2) thin layers, as labels. In the luminol/H2O2 system, Au@MnO2 exhibited strong light absorption in the broad UV-vis spectrum, similar to the black body effect, and the scavenging effect of Mn2+ on O2•-, which achieved the dual-quenching of ECL signal. Under the optimal experimental conditions, the immunosensor demonstrated a detection range of 0.1 pg mL-1 - 100 ng mL-1, with a detection limit of 0.069 pg mL-1. SIGNIFICANCE: Based on Pd@FeNi-MIL-88B nanoenzymes and Au@MnO2 nanocomposites, a dual-quenching sandwich-type ECL immunosensor for the detection of CD44 was constructed. The proposed immunosensor exhibited excellent reproducibility, stability, selectivity, and sensitivity, and provided a valuable analytical strategy and technical platform for the accurate detection of disease biomarkers, and opened up potential application prospects for early clinical treatment.


Asunto(s)
Técnicas Biosensibles , Nanopartículas del Metal , Neoplasias , Humanos , Compuestos de Manganeso , Oro , Peróxido de Hidrógeno , Luminol , Reproducibilidad de los Resultados , Inmunoensayo , Óxidos , Paladio , Receptores de Hialuranos
19.
Anal Chim Acta ; 1303: 342521, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38609263

RESUMEN

BACKGROUND: Theranostic nanoplatforms with integrated diagnostic imaging and multiple therapeutic functions play a vital role in precise diagnosis and efficient treatment for breast cancer, but unfortunately, these nanoplatforms are usually stuck in single-site imaging and single mode of treatment, causing unsatisfactory diagnostic and therapeutic efficiency. Herein, a dual biomarkers-activatable facile hollow mesoporous MnO2 (H-MnO2)-based theranostic nanoplatform, DNAzyme@H-MnO2-MUC1 aptamer (DHMM), was constructed for the simultaneous multi-site diagnosis and multiple treatment of breast cancer. RESULTS: The DHMM acted as an integrated diagnostic and therapeutic nanoplatform that realizes multi-site fluorescence imaging-guided high-efficient photothermal/chemodynamic/gene synergistic therapy (PTT/CDT/GT) for breast cancer. The H-MnO2 exhibits high loading capacity for Cy5-MUC1 aptamer (3.05 pmoL µg-1) and FAM-DNAzyme (3.37 pmoL µg-1), and excellent quenching for the probes. In the presence of MUC1 on the cell membrane and GSH in the cytoplasm, Cy5-MUC1 aptamer and FAM-DNAzyme was activated triggering dual-channel fluorescence imaging at different sites. Moreover, the self-supplied Mn2+ was further supplied as DNAzyme cofactors to catalytic cleavage intracellular EGR-1 mRNA for high-efficient GT and stimulated the Fenton-like reaction for CDT. The H-MnO2 also showcases a favorable photothermal performance with a photothermal conversion efficiency of 44.16%, which ultimately contributes to multi-site fluorescence imaging-guided synergistic treatment with an apoptosis rate of 71.82%. SIGNIFICANCE: This dual biomarker-activatable multiple therapeutic nanoplatform was realized multi-site fluorescence imaging-guided PTT/CDT/GT combination therapy for breast cancer with higher specificity and efficiency, which provides a promising theranostic nanoplatform for the precision and efficiency of breast cancer treatment.


Asunto(s)
Carbocianinas , ADN Catalítico , Neoplasias , Medicina de Precisión , Compuestos de Manganeso , Óxidos , Imagen Óptica , Biomarcadores
20.
Colloids Surf B Biointerfaces ; 238: 113921, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38631280

RESUMEN

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.


Asunto(s)
Cobre , Ácido Hialurónico , Imagen por Resonancia Magnética , Compuestos de Manganeso , Óxidos , Fotoquimioterapia , Microambiente Tumoral , Compuestos de Manganeso/química , Compuestos de Manganeso/farmacología , Microambiente Tumoral/efectos de los fármacos , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Óxidos/química , Óxidos/farmacología , Humanos , Cobre/química , Cobre/farmacología , Tamaño de la Partícula , Nanoestructuras/química , Antineoplásicos/farmacología , Antineoplásicos/química , Fototerapia , Nanopartículas/química , Supervivencia Celular/efectos de los fármacos , Propiedades de Superficie , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA