Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Small ; : e2310706, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38446096

ABSTRACT

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.

2.
Angew Chem Int Ed Engl ; 63(14): e202319690, 2024 04 02.
Article in English | MEDLINE | ID: mdl-38320965

ABSTRACT

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.


Subject(s)
Bacterial Infections , Gallium , Porphyrins , Humans , Iron/metabolism , Hemin/metabolism , Bacteria/metabolism , Anti-Bacterial Agents/metabolism , Biofilms , Gallium/pharmacology , Porphyrins/pharmacology , Porphyrins/metabolism , Bacterial Infections/drug therapy , Homeostasis , Ions/metabolism , Polymers/metabolism
3.
Small ; : e2309664, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38057126

ABSTRACT

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.

4.
Small ; 19(43): e2302547, 2023 10.
Article in English | MEDLINE | ID: mdl-37376834

ABSTRACT

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.


Subject(s)
Nanocomposites , Periodontitis , Photochemotherapy , Animals , Reactive Oxygen Species , Photochemotherapy/methods , Periodontitis/drug therapy , Periodontitis/microbiology , Biofilms , Inflammation/drug therapy , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Ions
5.
Int J Mol Sci ; 24(3)2023 Feb 03.
Article in English | MEDLINE | ID: mdl-36769328

ABSTRACT

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.


Subject(s)
COVID-19 , Periodontitis , Humans , SARS-CoV-2 , Pandemics , Periodontitis/epidemiology , NF-kappa B
6.
Mikrochim Acta ; 189(10): 382, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36103003

ABSTRACT

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.


Subject(s)
Graphite , Quantum Dots , Animals , Copper , Disease Models, Animal , Dopamine/chemistry , Graphite/chemistry , Humans , Mice
7.
Small ; 17(41): e2101505, 2021 10.
Article in English | MEDLINE | ID: mdl-34499411

ABSTRACT

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.


Subject(s)
Nanocomposites , Quercetin , Animals , Inflammation/drug therapy , Macrophage Activation , Macrophages
8.
Int J Mol Sci ; 20(2)2019 Jan 11.
Article in English | MEDLINE | ID: mdl-30641958

ABSTRACT

Periodontitis is a common infectious disease characterized by loss of tooth-supporting structures, which eventually leads to tooth loss. The heavy burden of periodontal disease and its negative consequence on the patient's quality of life indicate a strong need for developing effective therapies. According to the World Health Organization, 10⁻15% of the global population suffers from severe periodontitis. Advances in understanding the etiology, epidemiology and microbiology of periodontal pocket flora have called for antibacterial therapeutic strategies for periodontitis treatment. Currently, antimicrobial strategies combining with polymer science have attracted tremendous interest in the last decade. This review focuses on the state of the art of antibacterial polymer application against periodontal pathogens and biofilms. The first part focuses on the different polymeric materials serving as antibacterial agents, drug carriers and periodontal barrier membranes to inhibit periodontal pathogens. The second part reviews cutting-edge research on the synthesis and evaluation of a new generation of bioactive dental polymers for Class-V restorations with therapeutic effects. They possess antibacterial, acid-reduction, protein-repellent, and remineralization capabilities. In addition, the antibacterial photodynamic therapy with polymeric materials against periodontal pathogens and biofilms is also briefly described in the third part. These novel bioactive and therapeutic polymeric materials and treatment methods have great potential to inhibit periodontitis and protect tooth structures.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/growth & development , Periodontitis/therapy , Polymers/chemical synthesis , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Bacteria/drug effects , Biofilms/drug effects , Dental Materials/chemical synthesis , Dental Materials/chemistry , Drug Delivery Systems , Humans , Periodontitis/microbiology , Photochemotherapy , Polymers/chemistry , Polymers/pharmacology , Quality of Life
9.
Molecules ; 24(15)2019 Jul 24.
Article in English | MEDLINE | ID: mdl-31344909

ABSTRACT

Photodynamic therapy (PDT) based periodontal disease treatment has received extensive attention. However, the deep tissue location of periodontal plaque makes the conventional PDT encounter a bottleneck. Herein, upconversion fluorescent nanomaterial with near-infrared light excitation was introduced into the treatment of periodontal disease, overcoming the limited tissue penetration depth of visible light in PDT. Photosensitizer Ce6 molecules were combined with upconversion nanoparticles (UCNPs) NaYF4:Yb,Er with a novel strategy. The hydrophobic UCNPs were modified with amphiphilic silane, utilizing the hydrophobic chain of the silane to bind to the hydrophobic groups of the UCNPs through a hydrophobic-hydrophobic interaction, and the Ce6 molecules were loaded in this hydrophobic layer. This achieves both the conversion of the hydrophobic to the hydrophilic surface and the loading of the oily photosensitizer molecules. Because the excitation position of the Ce6 molecule is in the red region, Mn ions were doped to enhance red light, and thus the improved PDT function. This Ce6 loaded UCNPs composites with efficient red upconversion luminescence show remarkable bacteriological therapeutic effect on Porphyromonas gingivalis, Prevotella intermedia and Fusobacterium nucleatum and the corresponding biofilms under 980 nm irradiation, indicating a high application prospect in the treatment of periodontal diseases.


Subject(s)
Anti-Infective Agents/pharmacology , Biofilms/drug effects , Nanocomposites , Porphyrins/pharmacology , Anti-Infective Agents/chemistry , Chlorophyllides , Humans , Hydrophobic and Hydrophilic Interactions , Light , Microbial Sensitivity Tests , Nanocomposites/chemistry , Nanoparticles/chemistry , Periodontitis/drug therapy , Periodontitis/microbiology , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Porphyrins/chemistry , Silanes/chemistry
10.
Biomed Eng Online ; 17(1): 88, 2018 Jun 20.
Article in English | MEDLINE | ID: mdl-29925387

ABSTRACT

BACKGROUND: Minocycline has been widely used in central nervous system disease. However, the effect of minocycline on the repairing of nerve fibers around dental implants had not been previously investigated. The aim of the present study was to evaluate the possibility of using minocycline for the repairing of nerve fibers around dental implants by investigating the effect of minocycline on the proliferation of Schwann cells and secretion of neurotrophic factors nerve growth factor and glial cell line-derived neurotrophic factor in vitro. METHODS: TiO2 nanotubes were fabricated on the surface of pure titanium via anodization at the voltage of 20, 30, 40 and 50 V. The nanotubes structure were characterized by scanning electron microscopy and examined with an optical contact angle. Then drug loading capability and release behavior were detected in vitro. The TiO2 nanotubes loaded with different concentration of minocycline were used to produce conditioned media with which to treat the Schwann cells. A cell counting kit-8 assay and cell viability were both selected to study the proliferative effect of the specimens on Schwann cell. Reverse transcription-quantitative PCR and western blot analyses were used to detect the related gene/protein expression of Schwann cells. RESULTS: The results showed that the diameter of TiO2 nanotubes at different voltage varied from 100 to 200 nm. The results of optical contact angle and releasing profile showed the nanotubes fabricated at the voltage of 30 V met the needs of the carrier of minocycline. In addition, the TiO2 nanotubes loaded with the concentration of 20 µg/mL minocycline increased Schwann cells proliferation and secretion of neurotrophic factors in vitro. CONCLUSIONS: The results suggested that the surface functionalization of TiO2 nanotubes with minocycline was a promising candidate biomaterial for the peripheral nerve regeneration around dental implants and has potential to be applied in improving the osseoperception of dental implant.


Subject(s)
Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Minocycline/chemistry , Nanotubes/chemistry , Schwann Cells/drug effects , Titanium/chemistry , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Schwann Cells/cytology , Schwann Cells/metabolism , Surface Properties
11.
Biosens Bioelectron ; 252: 116135, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38387230

ABSTRACT

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.


Subject(s)
Biosensing Techniques , Metal Nanoparticles , MicroRNAs , Nanoparticles , Humans , Biosensing Techniques/methods , Silver , Fluorescence Resonance Energy Transfer , Biomarkers
12.
J Colloid Interface Sci ; 663: 834-846, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38447398

ABSTRACT

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.


Subject(s)
Anti-Infective Agents , Nanocomposites , Nanoparticles , Photochemotherapy , Humans , Hydrogen Peroxide , Manganese , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Oxygen , Inflammation/drug therapy , Nanocomposites/chemistry , Nanoparticles/chemistry , Photosensitizing Agents/chemistry , Cell Line, Tumor
13.
Natl Sci Rev ; 11(8): nwae225, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39071842

ABSTRACT

Periodontitis involves hyperactivated stromal cells that recruit immune cells, exacerbating inflammation. This study presents an ATP-responsive metal-organic framework (Mg/Zn-MOF) designed for periodontitis treatment, utilizing ion interference to modulate immune responses and prevent tissue destruction. Addressing the challenges of synergistic ion effects and targeted delivery faced by traditional immunomodulatory nanomaterials, the Mg/Zn-MOF system is activated by extracellular ATP-a pivotal molecule in periodontitis pathology-ensuring targeted ion release. Magnesium and zinc ions released from the framework synergistically inhibit membrane pore formation by attenuating Gasdermin D (GSDMD) expression and activation. This action curtails pyroptosis, lactate dehydrogenase and IL-1ß release, thwarting the onset of inflammatory cascades. Mechanistically, Mg/Zn-MOF intervenes in both the NLRP3/Caspase-1/GSDMD and Caspase-11/GSDMD pathways to mitigate pyroptosis. In vivo assessments confirm its effectiveness in diminishing inflammatory cell infiltration and preserving collagen integrity, thereby safeguarding against periodontal tissue damage and bone loss. This investigation highlights the promise of ion-interference strategies in periodontitis immunotherapy, representing a significant stride in developing targeted therapeutic approaches.

14.
J Control Release ; 370: 600-613, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38735394

ABSTRACT

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.


Subject(s)
Cerium , Gold , Nanocomposites , Periodontitis , Sulfates , Cerium/chemistry , Cerium/pharmacology , Animals , Periodontitis/drug therapy , Nanocomposites/chemistry , Gold/chemistry , Sulfates/chemistry , Reactive Oxygen Species/metabolism , Catalysis , Nanotubes/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Male , Mice , Biofilms/drug effects , Porphyromonas gingivalis/drug effects
15.
Adv Mater ; 36(13): e2310529, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38145555

ABSTRACT

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.


Subject(s)
Metal Nanoparticles , Metal Nanoparticles/chemistry , Metals , Nanomedicine , Proteins , Precision Medicine
16.
Front Microbiol ; 14: 948092, 2023.
Article in English | MEDLINE | ID: mdl-36846804

ABSTRACT

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.

17.
Lab Chip ; 23(6): 1493-1523, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36776104

ABSTRACT

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.


Subject(s)
Nanostructures , Neoplastic Cells, Circulating , Humans , Neoplastic Cells, Circulating/pathology , Microfluidics/methods , Flow Cytometry , Cell Separation/methods , Cell Line, Tumor
18.
Adv Healthc Mater ; 12(6): e2202652, 2023 01.
Article in English | MEDLINE | ID: mdl-36373219

ABSTRACT

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.


Subject(s)
Nanocomposites , Photochemotherapy , Bacteria , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Nanocomposites/therapeutic use , Nanocomposites/chemistry , Wound Healing
19.
Theranostics ; 13(7): 2350-2367, 2023.
Article in English | MEDLINE | ID: mdl-37153739

ABSTRACT

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.


Subject(s)
Complementary Therapies , Nanocomposites , Photochemotherapy , Animals , Nitric Oxide , Reactive Oxygen Species , Photochemotherapy/methods , Biofilms , Anti-Bacterial Agents/pharmacology , Models, Animal
20.
Adv Healthc Mater ; 12(19): e2300313, 2023 07.
Article in English | MEDLINE | ID: mdl-36933236

ABSTRACT

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.


Subject(s)
Nanoparticles , Periodontitis , Humans , Manganese Compounds/pharmacology , Manganese Compounds/chemistry , Oxides/pharmacology , Oxides/chemistry , Periodontitis/drug therapy , Bacteria , Biofilms , Nanoparticles/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL