Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Control Release ; 370: 600-613, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38735394

RESUMEN

The sulfate radical (SO4•-), known for its high reactivity and long lifespan, has emerged as a potent antimicrobial agent. Its exceptional energy allows for the disruption of vital structures and metabolic pathways in bacteria that are usually inaccessible to common radicals. Despite its promising potential, the efficient generation of this radical, particularly through methods involving enzymes and photocatalysis, remains a substantial challenge. Here, we capitalized on the peroxidase (POD)-mimicking activity and photocatalytic properties of cerium oxide (CeO2) nanozymes, integrating these properties with the enhanced concept of plasma gold nanorod (GNR) to develop a half-encapsulated core@shell GNRs@CeO2 Janus heterostructure impregnated with persulfate. Under near-infrared irradiation, the GNRs generate hot electrons, thereby boosting the CeO2's enzyme-like activity and initiating a potent reactive oxygen species (ROS) storm. This distinct nanoarchitecture facilitates functional specialization, wherein the heterostructure and efficient light absorption ensured continuous hot electron flow, not only enhancing the POD-like activity of CeO2 for the production of SO4•- effectively, but also contributing a significant photothermal effect, disrupting periodontal plaque biofilm and effectively eradicating pathogens. Furthermore, the local temperature elevation synergistically enhances the POD-like activity of CeO2. Transcriptomics analysis, as well as animal experiments of the periodontitis model, have revealed that pathogens undergo genetic information destruction, metabolic disorders, and pathogenicity changes in the powerful ROS system, and profound therapeutic outcomes in vivo, including anti-inflammation and bone preservation. This study demonstrated that energy transfer to augment nanozyme activity, specifically targeting ROS generation, constitutes a significant advancement in antibacterial treatment.

2.
J Colloid Interface Sci ; 663: 834-846, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38447398

RESUMEN

Antibacterial photodynamic therapy (aPDT) is highly effective in killing bacteria, while the problem of hypoxia and limited light penetration in deep tissue has not been properly solved. In addition, few aPDT works take into account the regulation of inflammation, which is an important regulatory process after antimicrobial therapy and the final purpose of treatment. In this work, to address the above isssues, we have designed a multi-functional composite UCNPs-Ce6-Mn(CO)5Br@Silane (referred to as UCM@Si), which consists of several key components: Up-conversion nanoparticles (UCNPs: NaErF4:Tm3+@NaYF4:Yb3+), Chlorin e6 (Ce6) and Manganese pentacarbonyl bromide (Mn(CO)5Br). When exposed to near-infrared (NIR) light (980 nm), the UCNPs can emit strong red light at 655 nm which further trigger the aPDT of Ce6. The generated reactive oxygen (ROS) subsequently break the metal carbonyl bond of Mn(CO)5Br, leading to the production of carbon monoxide (CO) molecules as well as manganese ions (Mn2+), which further decomposes hydrogen peroxide (H2O2) in the microenvironment to oxygen (O2). Therefore, this simple nanocomposite not only provides substantial self-oxygen replenishment for enhanced aPDT, but also facilitates effective inflammation regulation via CO across a wide range of deep infections. This approach leverages the unique properties of these materials to combat bacterial infections by simultaneously killing bacteria, regulating inflammation, and enhancing the oxygen levels in the affected microenvironment. This O2 and CO gas based aPDT treatment system offers a promising approach to comprehensively address microbial-induced infectious diseases, particularly deep infections, holding the potential clinical applications.


Asunto(s)
Antiinfecciosos , Nanocompuestos , Nanopartículas , Fotoquimioterapia , Humanos , Peróxido de Hidrógeno , Manganeso , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Oxígeno , Inflamación/tratamiento farmacológico , Nanocompuestos/química , Nanopartículas/química , Fármacos Fotosensibilizantes/química , Línea Celular Tumoral
3.
Small ; : e2310706, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38446096

RESUMEN

Photothermal treatment (PTT) has emerged as a promising avenue for biofilm elimination, yet its potential drawbacks, such as local hyperpyrexia and bacterial heat resistance, have posed challenges. To address these concerns, an innovative nanoplatform (Au@mSiO2 -arg/ICG) is devised that integrates phototherapeutic and gas therapeutic functionalities. This multifaceted nanoplatform is composed of mesoporous silica-coated Au nanorods (Au@mSiO2 ), supplemented with l-arginine (l-arg) and indocyanine green (ICG), and is engineered for mild temperature PTT aimed at biofilm eradication. Au@mSiO2 -arg/ICG nanoparticles (NPs) show excellent antibacterial effects through the generation of nitric oxide (NO) gas, heat, and reactive oxygen species (ROS) under 808 nm light irradiation. The ROS generated by ICG initiates a cascade reaction with l-arg, ultimately yielding NO gas molecules. This localized release of NO not only effectively curbs the expression of heat shock proteins 70 mitigating bacterial thermoresistance, but also reduces extracellular polymeric substance allowing better penetration of the therapeutic agents. Furthermore, this nanoplatform achieves an outstanding biofilm elimination rate of over 99% in an abscess model under 808 nm light irradiation (0.8 W·cm-2 ), thereby establishing its potential as a dependable strategy for NO-enhanced mild PTT and antibacterial photodynamic therapy (aPDT) in clinical settings.

4.
Angew Chem Int Ed Engl ; 63(14): e202319690, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38320965

RESUMEN

Given the scarcity of novel antibiotics, the eradication of bacterial biofilm infections poses formidable challenges. Upon bacterial infection, the host restricts Fe ions, which are crucial for bacterial growth and maintenance. Having coevolved with the host, bacteria developed adaptive pathways like the hemin-uptake system to avoid iron deficiency. Inspired by this, we propose a novel strategy, termed iron nutritional immunity therapy (INIT), utilizing Ga-CT@P nanocomposites constructed with gallium, copper-doped tetrakis (4-carboxyphenyl) porphyrin (TCPP) metal-organic framework, and polyamine-amine polymer dots, to target bacterial iron intakes and starve them. Owing to the similarity between iron/hemin and gallium/TCPP, gallium-incorporated porphyrin potentially deceives bacteria into uptaking gallium ions and concurrently extracts iron ions from the surrounding bacteria milieu through the porphyrin ring. This strategy orchestrates a "give and take" approach for Ga3+/Fe3+ exchange. Simultaneously, polymer dots can impede bacterial iron metabolism and serve as real-time fluorescent iron-sensing probes to continuously monitor dynamic iron restriction status. INIT based on Ga-CT@P nanocomposites induced long-term iron starvation, which affected iron-sulfur cluster biogenesis and carbohydrate metabolism, ultimately facilitating biofilm eradication and tissue regeneration. Therefore, this study presents an innovative antibacterial strategy from a nutritional perspective that sheds light on refractory bacterial infection treatment and its future clinical application.


Asunto(s)
Infecciones Bacterianas , Galio , Porfirinas , Humanos , Hierro/metabolismo , Hemina/metabolismo , Bacterias/metabolismo , Antibacterianos/metabolismo , Biopelículas , Galio/farmacología , Porfirinas/farmacología , Porfirinas/metabolismo , Infecciones Bacterianas/tratamiento farmacológico , Homeostasis , Iones/metabolismo , Polímeros/metabolismo
5.
Biosens Bioelectron ; 252: 116135, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38387230

RESUMEN

MicroRNAs are small single-stranded RNA molecules associated with gene expression and immune response, suggesting their potential as biomarkers for health monitoring. Herein, we designed a novel upconversion-based multimode lateral flow assay (LFA) system to detect microRNAs in body fluids by simultaneously producing three unique signals within a detection strip. The core-shell Au-DTNB@Ag nanoparticles act as both the Raman reporters and acceptors, quenching fluorescence from upconversion nanoparticles (UCNPs, NaYF4: Yb3+, Er3+) via the Förster resonance energy transfer mechanism. Using microRNA-21 as a representative analyte, the LFA system offers remarkable detection range from 2 nM to 1 fM, comparable to outcomes from signal amplification methods, due to the successful single-layer self-assembly of UCNPs on the NC membrane, which greatly enhances both the convenience and sensitivity of the LFA technique. Additionally, our proprietary fluorescence-Raman detection platform simplifies result acquisition by reducing procedural intricacies. The biosensor, when evaluated with diverse bodily fluids, showed remarkable selectivity and sustained stability. Importantly, our LFA biosensor effectively identified periodontitis and lung cancer patients from healthy subjects in genuine samples, indicating significant potential for disease prediction, early diagnosis, and progression tracking. This system holds promise as a multifunctional tool for various biomarker assays.


Asunto(s)
Técnicas Biosensibles , Nanopartículas del Metal , MicroARNs , Nanopartículas , Humanos , Técnicas Biosensibles/métodos , Plata , Transferencia Resonante de Energía de Fluorescencia , Biomarcadores
6.
Adv Mater ; 36(13): e2310529, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38145555

RESUMEN

Metal nanoclusters (NCs) are well-recognized novel nano-agents that hold great promise for applications in nanomedicine because of their ultrafine size, low toxicity, and high renal clearance. As foreign substances, however, an in-depth understanding of the bioresponses to metal NCs is necessary but is still far from being realized. Herein, this review is deployed to summarize the biofates of metal NCs at various biological levels, emphasizing their multiscale bioresponses at the molecular, cellular, and organismal levels. In the parts-to-whole schema, the interactions between biomolecules and metal NCs are discussed, presenting typical protein-dictated nano-bio interfaces, hierarchical structures, and in vivo trajectories. Then, the accumulation, internalization, and metabolic evolution of metal NCs in the cellular environment and as-imparted theranostic functionalization are demonstrated. The organismal metabolism and transportation processes of the metal NCs are subsequently distilled. Finally, this review ends with the conclusions and perspectives on the outstanding issues of metal NC-mediated bioresponses in the near future. This review is expected to provide inspiration for tailoring the customization of metal NC-based nano-agents to meet practical requirements in different sectors of nanomedicine.


Asunto(s)
Nanopartículas del Metal , Nanopartículas del Metal/química , Metales , Nanomedicina , Proteínas , Medicina de Precisión
7.
Small ; : e2309664, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-38057126

RESUMEN

Nanozymes with peroxidase-mimic activity have recently emerged as effective strategies for eliminating infections. However, challenges in enhancing catalytic activities and the ability to target bacteria have hindered the broader application of nanozymes in bacterial infections. Herein, a novel nanozyme based on mesoporous CeO2 nanosphere and meso-tetra(4-carboxyphenyl)porphine (TCPP) encapsulated within pathogen-activated macrophage membranes, demonstrates photodynamic capability coupled with photo-enhanced chemodynamic therapy for selective and efficient antibacterial application against infected wounds. Interestingly, the expression of Toll-like receptors accordingly upregulates when macrophages are co-cultured with specific bacteria, thereby facilitating to recognition of the pathogen-associated molecular patterns originating from bacteria. The CeO2 not only serve as carriers for TCPP, but also exhibit intrinsic peroxidase-like catalytic activity. Consequently, Staphylococcus aureus (S. aureus)-activated macrophage membrane-coated CeO2 -TCPP (S-MM@CeO2 -TCPP) generated singlet oxygen, and simultaneously promoted photo-enhanced chemodynamic therapy, significantly boosting reactive oxygen species (ROS) to effectively eliminate bacteria. S-MM@CeO2 -TCPP specifically targeted S. aureus via Toll-like receptor, thereby directly disrupting bacterial structural integrity to eradicate S. aureus in vitro and relieve bacteria-induced inflammation to accelerate infected wound healing in vivo. By selectively targeting specific bacteria and effectively killing pathogens, such strategy provides a more efficient and reliable alternative for precise elimination of pathogens and inflammation alleviation in microorganism-infected wounds.

8.
Nanoscale ; 15(33): 13617-13627, 2023 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-37575088

RESUMEN

The emergence of multidrug-resistant (MDR) bacterial infections calls for novel strategies for effective bacterial inhibition and wound healing. Phototherapeutic approaches are promising in treating bacterial infection because of their high efficiency, noninvasiveness, and few side effects; however, their antibacterial effect is limited by the formation of biofilms in wounds. Herein, we report novel composite nanoparticles (mPt/ICG-αA NPs) combining mesoporous platinum (mPt) nanoparticles, indocyanine green (ICG) and α-amylase (αA) for combating MDR bacteria and treating wound infection, which integrates a triple bacterial inhibition mechanism arising from the combination of photodynamic therapy (PDT), photothermal therapy (PTT) and α-amylase enzymatic activities. The combination of mPt and ICG significantly enhances the effect of PTT and the temperature can be increased up to 80.8 °C to induce efficacious bacterial degeneration. Meanwhile, mPt/ICG-αA (mPIA) NPs with a low concentration of 25 µg mL-1 exhibited a remarkable catalase activity (CAT) and could continuously decompose endogenous H2O2 into O2 in a hypoxic microenvironment, thereby enhancing the PDT effect to achieve broad-spectrum bactericidal activity. mPIA NPs showed excellent MDR antibacterial efficiency against both Gram-positive Staphylococcus aureus (S. aureus) and Gram-negative Escherichia coli (E. coli), and the bactericidal rate reached up to 99.0% and 97.2% with single 808 nm near-infrared light irradiation, respectively. mPIA NPs also exhibited an excellent ability to destroy biofilms and biocompatibility. Animal experiments further suggested that mPIA NPs could achieve the successful repairment of wounds infected with S. aureus in living systems, while this platform demonstrated negligible toxicity towards mice. Considering the superior performances of mPIA NPs, the synergistic αA-CAT-PDT-PTT boosted therapeutic activity presented in the current work provides a promising method to effectively fight against biofilm-related infectious diseases and wound healing.


Asunto(s)
Verde de Indocianina , Nanopartículas , Ratones , Animales , Verde de Indocianina/farmacología , Platino (Metal)/farmacología , Escherichia coli , Staphylococcus aureus , alfa-Amilasas , Peróxido de Hidrógeno , Nanopartículas/uso terapéutico , Cicatrización de Heridas , Antibacterianos/farmacología
9.
Small ; 19(43): e2302547, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37376834

RESUMEN

Porphyrin-based antibacterial photodynamic therapy (aPDT) has found widespread applications in treating periodontitis. However, its clinical use is limited by poor energy absorption, resulting in limited reactive oxygen species (ROS) generation. To overcome this challenge, a novel Z-scheme heterostructured nanocomposite of Bi2 S3 /Cu-TCPP is developed. This nanocomposite exhibits highly efficient light absorption and effective electron-hole separation, thanks to the presence of heterostructures. The enhanced photocatalytic properties of the nanocomposite facilitate effective biofilm removal. Theoretical calculations confirm that the interface of the Bi2 S3 /Cu-TCPP nanocomposite readily adsorbs oxygen molecules and hydroxyl radicals, thereby improving ROS production rates. Additionally, the photothermal treatment (PTT) using Bi2 S3 nanoparticles promotes the release of Cu2+ ions, enhancing the chemodynamic therapy (CDT) effect and facilitating the eradication of dense biofilms. Furthermore, the released Cu2+ ions deplete glutathione in bacterial cells, weakening their antioxidant defense mechanisms. The synergistic effect of aPDT/PTT/CDT demonstrates potent antibacterial activity against periodontal pathogens, particularly in animal models of periodontitis, resulting in significant therapeutic effects, including inflammation alleviation and bone preservation. Therefore, this design of semiconductor-sensitized energy transfer represents an important advancement in improving aPDT efficacy and the treatment of periodontal inflammation.


Asunto(s)
Nanocompuestos , Periodontitis , Fotoquimioterapia , Animales , Especies Reactivas de Oxígeno , Fotoquimioterapia/métodos , Periodontitis/tratamiento farmacológico , Periodontitis/microbiología , Biopelículas , Inflamación/tratamiento farmacológico , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Iones
10.
Theranostics ; 13(7): 2350-2367, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37153739

RESUMEN

Background: Periodontal disease, an oral disease that initiates with plaque biofilm infection, affects 10% of the global population. Due to the complexity of tooth root anatomy, biofilm resistance and antibiotic resistance, traditional mechanical debridement and antibiotic removal of biofilms are not ideal. Nitric oxide (NO) gas therapy and its multifunctional therapy are effective methods to clear biofilms. However, large and controlled delivery of NO gas molecules is currently a great challenge. Methods: The core-shell structure of Ag2S@ZIF-90/Arg/ICG was developed and characterized in detail. The ability of Ag2S@ZIF-90/Arg/ICG to produce heat, ROS and NO under 808 nm NIR excitation was detected by an infrared thermal camera, probes and Griess assay. In vitro anti-biofilm effects were evaluated by CFU, Dead/Live staining and MTT assays. Hematoxylin-eosin staining, Masson staining and immunofluorescence staining were used to analyze the therapeutic effects in vivo. Results: Antibacterial photothermal therapy (aPTT) and antibacterial photodynamic therapy (aPDT) could be excited by 808 nm NIR light, and the produced heat and ROS further triggered the release of NO gas molecules simultaneously. The antibiofilm effect had a 4-log reduction in vitro. The produced NO caused biofilm dispersion through the degradation of the c-di-AMP pathway and improved biofilm eradication performance. In addition, Ag2S@ZIF-90/Arg/ICG had the best therapeutic effect on periodontitis and NIR II imaging ability in vivo. Conclusions: We successfully prepared a novel nanocomposite with NO synergistic aPTT and aPDT. It had an outstanding therapeutic effect in treating deep tissue biofilm infection. This study not only enriches the research on compound therapy with NO gas therapy but also provides a new solution for other biofilm infection diseases.


Asunto(s)
Terapias Complementarias , Nanocompuestos , Fotoquimioterapia , Animales , Óxido Nítrico , Especies Reactivas de Oxígeno , Fotoquimioterapia/métodos , Biopelículas , Antibacterianos/farmacología , Modelos Animales
11.
Adv Healthc Mater ; 12(19): e2300313, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36933236

RESUMEN

Nowadays the multifunctional approaches to kill oral bacteria based on various nanocomposites have made great progress against periodontal infections, while the material structure and its functional integration are still insufficient. Herein, this work proposes a therapeutic strategy of chemodynamical therapy (CDT) and photothermal therapy (PTT) in monocrystals to effectively enhance the synergistic treatment. The CuS/MnS@MnO2 consisting of hexagonal CuS/MnS nano-twin-crystal with a shell layer of MnO2 is developed. In this nanosystem, the purpose of synergistic treatment of periodontitis by combining PTT/CDT is achieved within a CuS/MnS monocrystal, where CuS serves to achieve photothermal conversion, dissipate the biofilm and transfer the heat in situ to the integrated MnS, thus promoting the Mn2+ -mediated CDT process. Meanwhile, the CDT process can generate the highly toxic hydroxyl radical to destroy extracellular DNA by utilization of endogenous H2 O2 produced by Streptococci in the oral biofilm, cooperating with PTT to dissipate the bacterial biofilm. With the design of the outer shell of MnO2 , the selective bacteria-killing can be realized by producing oxygen which can protect the periodontal non-pathogenic aerobic bacteria and threaten the survival of anaerobic pathogens. Therefore, such design via multipattern strategies to combat microorganisms would provide a bright prospect for the clinical treatment of bacterial infections.


Asunto(s)
Nanopartículas , Periodontitis , Humanos , Compuestos de Manganeso/farmacología , Compuestos de Manganeso/química , Óxidos/farmacología , Óxidos/química , Periodontitis/tratamiento farmacológico , Bacterias , Biopelículas , Nanopartículas/química
12.
Int J Mol Sci ; 24(3)2023 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-36769328

RESUMEN

Both periodontitis and Coronavirus disease 2019 (COVID-19) pose grave threats to public health and social order, endanger human life, and place a significant financial strain on the global healthcare system. Since the COVID-19 pandemic, mounting research has revealed a link between COVID-19 and periodontitis. It is critical to comprehend the immunological mechanisms of the two illnesses as well as their immunological interaction. Much evidence showed that there are many similar inflammatory pathways between periodontitis and COVID-19, such as NF-κB pathway, NLRP3/IL-1ß pathway, and IL-6 signaling pathway. Common risk factors such as gender, lifestyle, and comorbidities contribute to the severity of both diseases. Revealing the internal relationship between the two diseases is conducive to the treatment of the two diseases in an emergency period. It is also critical to maintain good oral hygiene and a positive attitude during treatment. This review covers four main areas: immunological mechanisms, common risk factors, evidence of the association between the two diseases, and possible interventions and potential targets. These will provide potential ideas for drug development and clinical treatment of the two diseases.


Asunto(s)
COVID-19 , Periodontitis , Humanos , SARS-CoV-2 , Pandemias , Periodontitis/epidemiología , FN-kappa B
13.
Lab Chip ; 23(6): 1493-1523, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36776104

RESUMEN

The isolation and detection of circulating tumor cells (CTCs) play an important role in early cancer diagnosis and prognosis, providing easy access to identify metastatic cells before clinically detectable metastases. In the past 20 years, according to the heterogeneous expression of CTCs on the surface and their special physical properties (size, morphology, electricity, etc.), a series of in vitro enrichment methods of CTCs have been developed based on microfluidic chip technology, nanomaterials and various nanostructures. In recent years, the in vivo detection of CTCs has attracted considerable attention. Photoacoustic flow cytometry and fluorescence flow cytometry were used to detect CTCs in a noninvasive manner. In addition, flexible magnetic wire and indwelling intravascular non-circulating CTCs isolation system were developed for in vivo CTCs study. In the aspect of downstream analysis, gene analysis and drug sensitivity tests of enriched CTCs were developed based on various existing molecular analysis techniques. All of these studies constitute a complete study of CTCs. Although the existing reviews mainly focus on one aspect of capturing CTCs study, a review that includes the in vivo and in vitro capture and downstream analysis study of CTCs is highly needed. This review focuses on not only the classic work and latest research progress in in vitro capture but also includes the in vivo capture and downstream analysis, discussing the advantages and significance of the different research methods and providing new ideas for solving the heterogeneity and rarity of CTCs.


Asunto(s)
Nanoestructuras , Células Neoplásicas Circulantes , Humanos , Células Neoplásicas Circulantes/patología , Microfluídica/métodos , Citometría de Flujo , Separación Celular/métodos , Línea Celular Tumoral
14.
Front Microbiol ; 14: 948092, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36846804

RESUMEN

Infectious diseases remain a serious global challenge threatening human health. Oral infectious diseases, a major neglected global problem, not only affect people's lifestyles but also have an intimate association with systemic diseases. Antibiotic therapy is a common treatment. However, the emergence of new resistance problems hindered and enhanced the complication of the treatment. Currently, antimicrobial photodynamic therapy (aPDT) has long been the topic of intense interest due to the advantage of being minimally invasive, low toxicity, and high selectivity. aPDT is also becoming increasingly popular and applied in treating oral diseases such as tooth caries, pulpitis, periodontal diseases, peri-implantitis, and oral candidiasis. Photothermal therapy (PTT), another phototherapy, also plays an important role in resisting resistant bacterial and biofilm infections. In this mini-review, we summarize the latest advances in photonics-based treatments of oral infectious diseases. The whole review is divided into three main parts. The first part focuses on photonics-based antibacterial strategies and mechanisms. The second part presents applications for photonics-based treatments of oral infectious diseases. The last part discusses present problems in current materials and future perspectives.

15.
Adv Healthc Mater ; 12(6): e2202652, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36373219

RESUMEN

The rising dangers of bacterial infections have created an urgent need for the development of a new generation of antibacterial technologies and therapeutics. Antibacterial photodynamic therapy (PDT), considered as a noninvasive treatment with no drug resistance, has become a new promising photochemistry-involved treatment strategy. Titanium oxide (TiO2 ) is proved to be a very efficient PDT agent among the photosensitive materials, while the property of a large bandgap of TiO2 makes it only be excited by ultraviolet light, which is harmful to organisms. In this work, a novel ligand-to-metal charge transfer (LMCT) mediated TiO2 PDT strategy is proposed via the harmless near-infrared light irradiation. By choosing a mussel-inspired material, polydopamine (PDA) is involved in forming mesoporous TiO2 @PDA nanoparticles (mTiO2 @PDA NPs). The catechol groups of PDA can attach the TiO2 tightly even in colloidal environments, and can also form the LMCT bridge, exciting TiO2 to exert PDT function via 808 nm irradiation. Combining the sonodynamic therapy (SDT) of TiO2 and the photothermal therapy properties of PDA, this simple structure mTiO2 @PDA enables synergistic antibacterial applications with multiple functions under the dual excitation of NIR and ultrasound. This reliable all-in-one NPs can achieve great antibacterial effect and a rapid repair of infected wounds.


Asunto(s)
Nanocompuestos , Fotoquimioterapia , Bacterias , Antibacterianos/farmacología , Antibacterianos/química , Nanocompuestos/uso terapéutico , Nanocompuestos/química , Cicatrización de Heridas
16.
J Funct Biomater ; 13(4)2022 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-36412867

RESUMEN

Sulfate radicals (SO4-·) play important biological roles in biomedical and environmental engineering, such as antimicrobial, antitumor, and disinfection. Compared with other common free radicals, it has the advantages of a longer half-life and higher oxidation potential, which could bring unexpected effects. These properties have prompted researchers to make great contributions to biology and environmental engineering by exploiting their properties. Peroxymonosulfate (PMS) and peroxydisulfate (PDS) are the main raw materials for SO4-· formation. Due to the remarkable progress in nanotechnology, a large number of nanomaterials have been explored that can efficiently activate PMS/PDS, which have been used to generate SO4-· for biological applications. Based on the superior properties and application potential of SO4-·, it is of great significance to review its chemical mechanism, biological effect, and application field. Therefore, in this review, we summarize the latest design of nanomaterials that can effectually activate PMS/PDS to create SO4-·, including metal-based nanomaterials, metal-free nanomaterials, and nanocomposites. Furthermore, we discuss the underlying mechanism of the activation of PMS/PDS using these nanomaterials and the application of SO4-· in the fields of environmental remediation and biomedicine, liberating the application potential of SO4-·. Finally, this review provides the existing problems and prospects of nanomaterials being used to generate SO4-· in the future, providing new ideas and possibilities for the development of biomedicine and environmental remediation.

17.
Mikrochim Acta ; 189(10): 382, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-36103003

RESUMEN

A series of three-dimensional copper oxide (CuO) inverse opals anchored with carboxylated graphene quantum dots (CuO/cGQDs) have been fabricated for non-enzymatic tracking of dopamine (DA). Heterostructures composed of various building blocks are promising to construct versatile biosensing platforms. The optimal CuO/cGQDs modified electrode demonstrates sensitivities of 243.45 µA mM-1 cm-2 (50 nM-1888.5 µM) with the practical detection limit as low as 0.5 nM in mimic physiological environment (at + 0.45 V vs. Ag/AgCl). The extraordinary tolerance to various interferents enables the practical detection of intracellular DA amount in human neural cells. On this basis, the proposed biosensor attains precise evaluation of antipsychotic drug effects on stimulated DA release. Particularly, it successfully spots fluctuation of DA in plasma and cerebrospinal fluid in murine model of Parkinson's disease, which serves as a crucial tool to understand neuropathology and symptomatology of DA-related diseases. This study developed a reliable sensing platform and is expected to be applied to physiological and pathological studies.


Asunto(s)
Grafito , Puntos Cuánticos , Animales , Cobre , Modelos Animales de Enfermedad , Dopamina/química , Grafito/química , Humanos , Ratones
18.
Small ; 17(41): e2101505, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34499411

RESUMEN

Macrophage polarization toward M1 phenotype (pro-inflammation) is closely associated with the destructive phase of periodontal inflammation. Nanoceria is verified to inhibit M1 polarization of macrophages by the favorable ability of reactive oxygen species (ROS) scavenging. However, the function of nanoceria on macrophage polarization toward M2 phenotype (anti-inflammation) in reparative phase of periodontal inflammation is quite limited. In this work, by introducing an antioxidant drug quercetin onto nano-octahedral ceria, synergistic and intense regulation of host immunity against periodontal disease is realized. Such nanocomposite can control the phenotypic switch of macrophages by not only inhibition of M1 polarization for suppressing the damage in the destructive phase but also promotion of M2 polarization for regenerating the surrounding tissues in reparative phase of periodontal disease. As-prepared nanocomposite can effectively increase the M2/M1 ratio of macrophage polarization in inflammatory cellular models by lipopolysaccharide stimulation. More importantly, the nanocomposite also exerts an improved therapeutic potential against local inflammation by significant downregulation of pro-inflammatory cytokines and upregulation of anti-inflammatory cytokines in an animal model with periodontal inflammation. Therefore, this newly developed nanomedicine is efficient in ROS scavenging and driving pro-inflammatory macrophages to the anti-inflammatory phenotype to eliminate inflammation, thereby providing a promising candidate for treating periodontal inflammation.


Asunto(s)
Nanocompuestos , Quercetina , Animales , Inflamación/tratamiento farmacológico , Activación de Macrófagos , Macrófagos
19.
Adv Healthc Mater ; 10(22): e2101515, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34558227

RESUMEN

Numerous nanomedicines currently emerge to reduce the dramatic threat in antibiotics resistance for antibacterial application against severe bacterial infections, while it is restricted by over-reacted immune response to pathogenic bacteria. Herein, enzymatic activity is introduced into the zeolitic imidazolate framework-8 (ZIF-8) to achieve sterilization by releasing Zn ions, as well as inflammation regulation through the variable valence of Mn ions that are uniformly doped into its framework. Within this simple metal organic framework (MOF) structure design, Mn-ZIF-8 possesses the co-existence of Mn2+ /Mn4+ to endow the nanocomposite with the anti-inflammatory capabilities, which can be adjusted through the redox environment. The enzymatic activity of Mn ions and superiority of pore structure of ZIF-8 are effectively combined to realize the substrate selection via reactant molecular size and high-efficiency internal catalytic performance. By such design, this nanocomposite would not only exhibit an excellent antibacterial performance against pathogenic bacteria, but also reshape the inflammatory immunity by regulating macrophage polarization to suppress over-reacted inflammation, leading to a favorably therapeutic efficiency on bacteria-infected wound healing in animal models. Taken together, this nanoplatform provides effective approach for accelerating infected wound healing via bacteria killing and inflammation modulation, and may be extended for the therapy of other severe bacteria-induced infections.


Asunto(s)
Zeolitas , Animales , Antibacterianos/farmacología , Imidazoles , Inmunidad , Inmunomodulación , Iones , Manganeso , Cicatrización de Heridas
20.
J Diabetes Res ; 2019: 6757428, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31886284

RESUMEN

Osteocyte plays an essential role in bone metabolism by regulating osteoblast and osteoclast activities. Dysfunction or apoptosis of osteocyte will severely endanger the bone homeostasis and result in bone diseases such as osteoporosis. Osteoporosis has been considered as one of the diabetes complications; however, the mechanism is still to be discovered. Advanced glycation end products (AGEs), as the main pathogenic factor of diabetes mellitus, have the capacity to induce osteocyte apoptosis thus sabotaging bone homeostasis. Here, we examined the role of AGE during osteocyte apoptosis and how this effect would affect osteocyte's regulation of osteoblast and osteoclast. Mouse osteocyte-like MLO-Y4 cells were used to study the properties of osteocyte and to examine its biological and pathological function. MTT assay and Annexin V assay showed that AGE significantly induce MLO-Y4 cell apoptosis. qPCR and Western blot results have shown that AGE upregulates proapoptotic gene p53 and its downstream target gene Bax, which leads to enhanced activation of caspase-3, thus inducing apoptosis in MLO-Y4 cells. Increased expression of sclerostin and RANKL in osteocytes has shown that AGE induces osteocyte dysfunction thus severely damaging the bone homeostasis by decreasing osteoblast and increasing osteoclast activities. Furthermore, the role of the transcription factor FOXO1, which is intensely associated with apoptosis, has been determined. Western blot has shown that AGE significantly decreases Akt activities. Immunofluorescence has shown that AGE promotes FOXO1 nuclei localization and enhances FOXO1 expression. Silencing of FOXO1 suppressed AGE-enhanced apoptosis; mRNA and protein expressions of cleaved caspase-3, sclerostin, and RANKL were downregulated as well. Moreover, exogenous FOXO1 increased caspase-3 mRNA levels and caspase-3 transcriptional activity. Lastly, ChIP assay has established the capacity of FOXO1 binding directly on the caspase-3, sclerostin, and RANKL promoter region in AGE environment, providing the mechanism of the AGE-induced osteocyte apoptosis and dysfunction. Our results have shown that FOXO1 plays a crucial role in AGE-induced osteocyte dysfunction and apoptosis through its regulation of caspase-3, sclerostin, and RANKL. This study provides new insight into diabetes-enhanced risk of osteoporosis given the critical role of AGE in the pathogenesis of diabetes and the essential part of osteocyte in bone metabolism.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo , Productos Finales de Glicación Avanzada/toxicidad , Osteocitos/efectos de los fármacos , Albúmina Sérica Bovina/toxicidad , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Sitios de Unión , Caspasa 3/genética , Caspasa 3/metabolismo , Línea Celular , Proteína Forkhead Box O1/genética , Regulación de la Expresión Génica , Ratones , Osteocitos/metabolismo , Osteocitos/patología , Regiones Promotoras Genéticas , Ligando RANK/genética , Ligando RANK/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...