Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.528
Filter
1.
J Environ Sci (China) ; 149: 164-176, 2025 Mar.
Article in English | MEDLINE | ID: mdl-39181631

ABSTRACT

Cerium and cobalt loaded Co-Ce/TiO2 catalyst prepared by impregnation method was investigated for photothermal catalytic toluene oxidation. Based on catalyst characterizations (XPS, EPR and H2-TPR), redox cycle between Co and TiO2 (Co2+ + Ti4+ ↔ Co3+ + Ti3+) results in the formation of Co3+, Ti3+ and oxygen vacancies, which play important roles in toluene catalytic oxidation reaction. The introduction of Ce brings in the dual redox cycles (Co2+ + Ti4+ ↔ Co3+ + Ti3+, Co2+ + Ce4+ ↔ Co3+ + Ce3+), further promoting the elevation of reaction sites amount. Under full spectrum irradiation with light intensity of 580 mW/cm2, Co-Ce/TiO2 catalyst achieved 96% of toluene conversion and 73% of CO2 yield, obviously higher than Co/P25 and Co/TiO2. Co-Ce/TiO2 efficiently maintains 10-hour stability test under water vapor conditions and exhibits better photothermal catalytic performance than counterparts under different wavelengths illumination. Photothermal catalytic reaction displays improved activities compared with thermal catalysis, which is attributed to the promotional effect of light including photocatalysis and light activation of reactive oxygen species.


Subject(s)
Cerium , Cobalt , Oxidation-Reduction , Titanium , Toluene , Titanium/chemistry , Cobalt/chemistry , Catalysis , Toluene/chemistry , Cerium/chemistry , Models, Chemical , Photochemical Processes
2.
Biomaterials ; 312: 122724, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39106818

ABSTRACT

The residual bone tumor and defects which is caused by surgical therapy of bone tumor is a major and important problem in clinicals. And the sequential treatment for irradiating residual tumor and repairing bone defects has wildly prospects. In this study, we developed a general modification strategy by gallic acid (GA)-assisted coordination chemistry to prepare black calcium-based materials, which combines the sequential photothermal therapy of bone tumor and bone defects. The GA modification endows the materials remarkable photothermal properties. Under the near-infrared (NIR) irradiation with different power densities, the black GA-modified bone matrix (GBM) did not merely display an excellent performance in eliminating bone tumor with high temperature, but showed a facile effect of the mild-heat stimulation to accelerate bone regeneration. GBM can efficiently regulate the microenvironments of bone regeneration in a spatial-temporal manner, including inflammation/immune response, vascularization and osteogenic differentiation. Meanwhile, the integrin/PI3K/Akt signaling pathway of bone marrow mesenchymal stem cells (BMSCs) was revealed to be involved in the effect of osteogenesis induced by the mild-heat stimulation. The outcome of this study not only provides a serial of new multifunctional biomaterials, but also demonstrates a general strategy for designing novel blacked calcium-based biomaterials with great potential for clinical use.


Subject(s)
Bone Neoplasms , Bone Regeneration , Calcium , Gallic Acid , Mesenchymal Stem Cells , Gallic Acid/chemistry , Bone Regeneration/drug effects , Animals , Calcium/metabolism , Bone Neoplasms/therapy , Bone Neoplasms/drug therapy , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Photothermal Therapy/methods , Osteogenesis/drug effects , Mice , Humans , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Line, Tumor
3.
Biomaterials ; 312: 122751, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39121726

ABSTRACT

Tumor immunotherapies have emerged as a promising frontier in the realm of cancer treatment. However, challenges persist in achieving localized, durable immunostimulation while counteracting the tumor's immunosuppressive environment. Here, we develop a natural mussel foot protein-based nanomedicine with spatiotemporal control for tumor immunotherapy. In this nanomedicine, an immunoadjuvant prodrug and a photosensitizer are integrated, which is driven by their dynamic bonding and non-covalent assembling with the protein carrier. Harnessing the protein carrier's bioadhesion, this nanomedicine achieves a drug co-delivery with spatiotemporal precision, by which it not only promotes tumor photothermal ablation but also broadens tumor antigen repertoire, facilitating in situ immunotherapy with durability and maintenance. This nanomedicine also modulates the tumor microenvironment to overcome immunosuppression, thereby amplifying antitumor responses against tumor progression. Our strategy underscores a mussel foot protein-derived design philosophy of drug delivery aimed at refining combinatorial immunotherapy, offering insights into leveraging natural proteins for cancer treatment.


Subject(s)
Immunotherapy , Nanomedicine , Animals , Immunotherapy/methods , Nanomedicine/methods , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/pharmacology , Photothermal Therapy/methods , Mice , Humans , Tumor Microenvironment/drug effects , Cell Line, Tumor , Proteins/chemistry , Female , Neoplasms/therapy , Neoplasms/immunology , Adhesives/chemistry , Mice, Inbred C57BL , Adjuvants, Immunologic/pharmacology
4.
Carbohydr Polym ; 345: 122550, 2024 Dec 01.
Article in English | MEDLINE | ID: mdl-39227117

ABSTRACT

In the realm of natural polysaccharides, hydrogen bonding is a prevalent feature, yet its role in enhancing photocatalytic antimicrobial properties has been underexplored. In this paper, heterojunctions formed by graphene oxide (GO) and ZIF-8 were locked in sodium alginate/ carboxylated cellulose nanocrystals via hydrogen bonding networks, designated as SCGZ. The SCGZ films exhibit superior photocatalytic performance compared to either ZIF-8 or heterojunctions. This enhancement is primarily due to two key factors: firstly, the hydrogen bonding network significantly enhances the transfer of protons and holes, thereby improving the separation efficiency of photo-generated carriers; secondly, the hydrogen bonding between the layers facilitates a more efficient charge transfer, which expedites the movement of electrons from ZIF-8 to GO upon illumination. In vitro studies demonstrated that the SCGZ films possess remarkable antibacterial capabilities, achieving 99.75 % and 99.61 % inhibition rates against S. aureus and E. coli, respectively. In vivo animal experiments have shown that SCGZ films can significantly accelerate the healing process of damaged tissues, with a healing efficiency of up to 90.5 %. This research provides additional insights into the development of natural polysaccharide-based multi­hydrogen bonded macromolecules with enhanced photocatalytic properties.


Subject(s)
Alginates , Anti-Bacterial Agents , Cellulose , Escherichia coli , Graphite , Nanoparticles , Staphylococcus aureus , Wound Healing , Alginates/chemistry , Alginates/pharmacology , Cellulose/chemistry , Cellulose/pharmacology , Nanoparticles/chemistry , Wound Healing/drug effects , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Escherichia coli/drug effects , Animals , Graphite/chemistry , Graphite/pharmacology , Sterilization/methods , Hydrogen Bonding , Mice , Microbial Sensitivity Tests , Catalysis
5.
Carbohydr Polym ; 345: 122568, 2024 Dec 01.
Article in English | MEDLINE | ID: mdl-39227122

ABSTRACT

Bleeding and bacterial infection are common problems associated with wound treatment, while effective blood clotting and vessel regeneration promotion are the primary considerations to design the wound dressing materials. This research presents a chitosan-based hydrogel with grafted quaternary ammonium and polyphosphate (QCSP hydrogel) as the antibacterial hemostatic dressing to achieve burn wound treatment. The tissue adhesion of the hydrogel sealed the blood flow and the polyphosphate grafted to the chitosan promoted the activation of coagulation factor V to enhance the hemostasis. At the same time, the grafted quaternary ammonium enhanced the antibacterial ability of the biodegradable hydrogel wound dressing. In addition, the polydopamine as a photothermal agent was composited into the hydrogel to enhance the antibacterial and reactive oxygen scavenging performance. The in vivo hemostasis experiment proved the polyphosphate enhanced the coagulation property. Moreover, this photothermal property of the composite hydrogel enhanced the burn wound repairing rate combined with the NIR stimulus. As a result, this hydrogel could have potential application in clinic as dressing material for hemostasis and infection prone would repairing.


Subject(s)
Anti-Bacterial Agents , Burns , Chitosan , Hemostasis , Hydrogels , Indoles , Polymers , Wound Healing , Chitosan/chemistry , Chitosan/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Burns/drug therapy , Burns/therapy , Polymers/chemistry , Polymers/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Animals , Indoles/chemistry , Indoles/pharmacology , Wound Healing/drug effects , Hemostasis/drug effects , Mice , Hemostatics/chemistry , Hemostatics/pharmacology , Bandages , Male , Rats , Staphylococcus aureus/drug effects , Escherichia coli/drug effects , Rats, Sprague-Dawley , Microbial Sensitivity Tests , Photothermal Therapy/methods
6.
Article in English | MEDLINE | ID: mdl-39228328

ABSTRACT

Photothermal therapy (PTT) and photodynamic therapy (PDT) provide targeted approaches to cancer treatment, but each therapy has inherent limitations such as insufficient tissue penetration, uneven heat distribution, extreme hypoxia, and overexpressed HSP90 in tumor cells. To address these issues, herein, by encapsulating the IR780 dye and glucose oxidase (GOx) enzyme within ZIF-8 nanoparticles, we created a versatile system capable of combining photodynamic and enhanced photothermal therapy. The integration of the IR780 dye facilitated the generation of reactive oxygen species and hyperthermia upon light activation, enabling dual-mode cancer cell ablation. Moreover, GOx catalyzes the decomposition of glucose into gluconic acid and hydrogen peroxide, leading to the inhibition of ATP production and downregulation of heat shock protein 90 (HSP90) expression, sensitizing cancer cells to heat-induced cytotoxicity. This synergistic combination resulted in significantly improved therapeutic outcomes. Both in vitro and in vivo results validated that the nanoplatform demonstrated superior specificity and favorable therapeutic responses. Our innovative approach represents a promising strategy for overcoming current limitations in cancer treatments and offers the potential for clinical translation in the future.

7.
Front Ophthalmol (Lausanne) ; 4: 1408869, 2024.
Article in English | MEDLINE | ID: mdl-39224466

ABSTRACT

Correlating damage outcomes to a retinal laser exposure is critical for diagnosis and choosing appropriate treatment modalities. Therefore, it is important to understand the causal relationships between laser parameters, such as wavelength, power density, and length of exposure, and any resulting injury. Differentiating photothermal from photochemical processes in an in vitro retinal model using cultured retinal pigment epithelial cells would be a first step in achieving this goal. The first-order rate constant of Arrhenius has been used for decades to approximate cellular thermal damage. A modification of this equation, called the damage integral (Ω), has been used extensively to predict the accumulation of laser damage from photothermal inactivation of critical cellular proteins. Damage from photochemical processes is less well studied and most models have not been verified because they require quantification of one or more uncharacterized chemical species. Additionally, few reports on photochemical damage report temperature history, measured or simulated. We used simulated threshold temperatures from a previous in vitro study to distinguish between photothermal and photochemical processes. Assuming purely photochemical processes also inactivate critical cellular proteins, we report the use of a photothermal Ω and a photochemical Ω that work in tandem to indicate overall damage accumulation. The combined damage integral (ΩCDI) applies a mathematical switch designed to describe photochemical damage relative to wavelength and rate of photon delivery. Although only tested in an in vitro model, this approach may transition to predict damage at the mammalian retina.

8.
Adv Healthc Mater ; : e2402505, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39233538

ABSTRACT

Achieving the clinically staged treatment of osteosarcoma-associated bone defects encounters the multiple challenges of promptly removing postoperative residual tumor cells and bacterial infection, followed by bone reconstruction. Herein, a core/shell hydrogel with multiple-effect combination is designed to first exert antitumor and antibacterial activities and then promote osteogenesis. Specifically, doxorubicin (DOX) is loaded by magnesium-iron-based layered double hydroxide (LDH) to prepare LDOX, which is introduced into a thermo-sensitive hydrogel to serve as an outer shell of the core/shell hydrogel, meanwhile, LDH-contained liquid crystal hydrogel, abbreviated as LCgel-L, is served as an inner core. At the early stage of treatment, the dissociation of the outer shell triggered by moderate hyperthermia led to the thermo-sensitive release of LDOX, which can be targeted for the release of DOX within tumor cells, thereby promptly removing postoperative residual tumor cells based on the synergistic effect of photothermal therapy (PTT) and DOX, and postoperative bacterial infection can also be effectively prevented by PTT simultaneously. More importantly, the dissociation of the outer shell prompted the full exposure of the inner core, which will exert osteogenic activity based on the synergy of liquid crystal hydrogel as well as LDH-induced mild hyperthermia and ion effects, thereby enabling "temporal regulation" treatment of osteosarcoma-associated bone defects. This study provides a valuable insight for the development of osteosarcoma-associated bone repair materials.

9.
Int J Nanomedicine ; 19: 8901-8927, 2024.
Article in English | MEDLINE | ID: mdl-39233743

ABSTRACT

Introduction: Graphene oxide (GO) nanoparticles have emerged as a compelling photothermal agent (PHTA) in the realm of photothermal antibacterial therapy, owing to their cost-effectiveness, facile synthesis, and remarkable photostability. Nevertheless, the therapeutic efficacy of GO nanoparticles is commonly hindered by their inherent drawback of low photothermal conversion efficiency (PCE). Methods: Herein, we engineer the Ag/GO-GelMA platform by growing the Ag on the surface of GO and encapsulating the Ag/GO nanoparticles into the GelMA hydrogels. Results: The resulting Ag/GO-GelMA platform demonstrates a significantly enhanced PCE (47.6%), surpassing that of pure GO (11.8%) by more than fourfold. As expected, the Ag/GO-GelMA platform, which was designed to integrate the benefits of Ag/GO nanoparticles (high PCE) and hydrogel (slowly releasing Ag+ to exert an inherent antibacterial effect), has been shown to exhibit exceptional antibacterial efficacy. Furthermore, transcriptome analyses demonstrated that the Ag/GO-GelMA platform could significantly down-regulate pathways linked to inflammation (the MAPK and PI3K-Akt pathways) and had the ability to promote cell migration. Discussion: Taken together, this study presents the design of a potent photothermal antibacterial platform (Ag/GO-GelMA) aimed at enhancing the healing of infectious wounds. The platform utilizes a handy method to enhance the PCE of GO, thereby making notable progress in the utilization of GO nano-PHTAs.


Subject(s)
Anti-Bacterial Agents , Graphite , Hydrogels , Silver , Wound Healing , Graphite/chemistry , Graphite/pharmacology , Wound Healing/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Silver/chemistry , Silver/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Animals , Humans , Mice , Photothermal Therapy/methods , Nanoparticles/chemistry , Wound Infection/drug therapy , Wound Infection/microbiology , Escherichia coli/drug effects , Staphylococcus aureus/drug effects , Cell Movement/drug effects
10.
Mater Today Bio ; 28: 101209, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39221205

ABSTRACT

The development of novel phototheranostic agents with significant potential in bioimaging-guided therapy is highly desirable for precise tumor therapy. Herein, NIR laser-activated ruthenium phthalocyanine (PcRu) loaded sub-30 nm targeting lipid nanoparticles (α-PcRu-NPs) were fabricated for photoacoustic imaging (PAI)-guided photothermal therapy (PTT). Due to the formation of J-type aggregation of PcRu in the core of the nanostructure, the α-PcRu-NPs exhibited high stability, efficient NIR absorption, reduced singlet oxygen generation, high photothermal activity, and intense photoacoustic signal. With the M2 macrophage target peptide (M2pep) modification and small size of α-PcRu-NPs, in vivo evaluations reveal that α-PcRu-NPs can specifically target and deeply penetrate the tumor foci. Under a high contrast PAI guidance with α-PcRu-NPs (744 nm, 0.35 µW), it also realizes superior photothermal therapy (PTT) for breast cancer under 670 nm laser irradiation (0.5 W/cm2). The prominent therapeutic efficacy of α-PcRu-NP-based PTT not only directly kills tumor cells, but also enhances the immune response by promoting dendritic cell maturation and increasing cytotoxic T cell infiltration. Thus, this work broadens the applications of phthalocyanine derivatives as phototheranostics in the PAI-guided PTT field.

11.
Mater Today Bio ; 28: 101200, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39221207

ABSTRACT

Triple-negative breast cancer (TNBC) is characterized by higher recurrence rate and mortality. Thermally-mediated ablation via photothermal therapy (PTT) demonstrates considerable promise for the eradication of breast cancer. Nonetheless, the efficacy of PTT is impeded by the thermal tolerance of tumor cells, which is attributed to the augmented expression of heat shock proteins (HSPs). These proteins, which function as ATP-dependent molecular chaperones, confer protection to cancer cells against the cytotoxic heat generated during PTT. Glycolysis is an important way for breast cancer cells to produce ATP, which can promote the occurrence and development of lung metastasis of breast cancer. Therefore, inhibiting glycolysis may diminish the expression of HSPs, curtail the growth of breast cancer, and prevent its metastasis. Glycolytic metabolism plays a pivotal role in the ATP biosynthesis within breast cancer cells, facilitating the progression and dissemination of pulmonary metastases. Consequently, targeting glycolysis presents a strategic approach to HSP expression, the proliferation of breast cancer, and impede its metastatic spread. Herein, we designed an indocyanine green (ICG) and cryptotanshinone (CTS) loaded hyaluronic acid (HA) coated Zeolitic Imidazolate Framework-8 (ZIF-8) drug delivery system. The drug delivery system had excellent photothermal properties, which could reach temperature sufficient for photothermal ablation of tumor cells. (ICG + CTS)@HA-ZIF-8 also showed pH-responsive drug release, enhancing the sustained release of ICG and CTS to extend their systemic circulation duration. Moreover, the HA modification of ZIF-8 served to augment its targeting capabilities both in vitro and in vivo, leveraging the enhanced permeation and retention (EPR) effect, as well as active tumor targeting via the CD44 receptor pathway, resulting in a higher drug concentration and a better therapeutic effect in tumor. (ICG + CTS)@HA-ZIF-8 could downregulate the expression of glycolysis-related protein pyruvate kinase-M2 (PKM2), thereby inhibiting the glycolysis process, further suppressing tumor cell energy metabolism, downregulating the expression of HSPs, overcoming tumor cell heat resistance, and improving PTT effect. It exhibited a notable suppressive impact on both the proliferation and migration of breast cancer cells, potentially offering innovative insights for the visualized PTT in breast cancer treatment.

12.
Int J Pharm ; : 124665, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39236772

ABSTRACT

Stimuli-responsive nanoplatforms have been popular in controlled drug delivery research because of their ability to differentiate the tumor microenvironment from the normal tissue environment in a spatiotemporally controllable manner. The synergistic therapeutic approach of combining cancer chemotherapy with photothermal tumor ablation has improved the therapeutic efficacy of cancer therapeutics. In this study, a UiO-66 metal organic framework (MOF)-based system loaded with doxorubicin (DOX), surface decorated with the photothermal agents indocyanine green (ICG) and polydopamine (PDA), and conjugated with transferrin (TF) was successfully designed to operate as a responsive system to pH changes, featuring photothermal capabilities and target specificity for the purpose of treating breast cancer. The synthesized nanoplatform benefits from its uniform size, excellent DOX encapsulation efficiency (91.66 %), and efficient pH/NIR-mediated controlled release of the drug. In vitro photothermal studies indicate excellent photothermal stability of the formulation even after 6 on-off cycles of NIR irradiation. The in vitro cytotoxicity assessment using an NIR laser (808 nm) revealed that the DOX-loaded functionalized UiO-66 nanocarriers had outstanding inhibitory effects on 4T1 cells because of synergistic chemo-photo therapies, with no substantial toxicity by the carriers. In addition, cellular uptake evaluations revealed that UiO-DOX-ICG@PDA-TF could specifically target 4T1 cells on the basis of receptor-mediated internalization of transferrin receptors. Additionally, in vivo toxicity studies in Wistar rats indicated no signs of significant toxicity. The UiO-based nanoformulations effectively inhibited and destroyed cancer cells under 808 nm laser irradiation because of their minimal toxicity, strong biocompatibility, and outstanding synergistic chemo/photothermal/photodynamic treatment.

13.
Urolithiasis ; 52(1): 125, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39237676

ABSTRACT

Laser lithotripsy mechanisms can cause the chemical decomposition of stone components and the emergence of different end products. However, the potentially toxic end products formed during thulium fiber laser (TFL) lithotripsy of cystine stones have not been sufficiently investigated. The aim of our in vitro study is to analyze the chemical content of the gas products formed during the fragmentation of cystine stone with TFL. Human renal calculi consisting of 100% pure cystine, calcium oxalate monohydrate, or uric acid were fragmented separately with TFL in experimental setups and observed for gas release. After the lithotripsy, only the cystine stones showed gas formation. Gas chromatography-mass spectrometry was used to analyze the gas qualitatively, and scanning electron microscopy with energy-dispersive X-ray spectroscopy (SEM-EDX) and X-ray diffraction was used to examine the dried cystine stone fragments. Fragmentation of the cystine stones released free cystine, sulfur, hydrogen sulfide, and carbon disulfide gas. The SEM-EDX and X-ray diffraction analyses revealed that the free cystine in the dried fragments contained 43.1% oxygen, 28.7% sulfur, 16.1% nitrogen, and 12.1% carbon atoms according to atomic weight. The detection of potentially toxic gases after lithotripsy of cystine stones with TFL indicates a risk of in vivo production. Awareness needs to be increased among healthcare professionals to prevent potential inhalation and systemic toxicity for patients and operating room personnel during TFL lithotripsy of cystine stones.


Subject(s)
Calcium Oxalate , Cystine , Lithotripsy, Laser , Microscopy, Electron, Scanning , Thulium , Uric Acid , Cystine/analysis , Cystine/chemistry , Humans , Calcium Oxalate/analysis , Calcium Oxalate/chemistry , Lithotripsy, Laser/methods , Uric Acid/analysis , Thulium/chemistry , Kidney Calculi/chemistry , Kidney Calculi/therapy , Gases/analysis , Gas Chromatography-Mass Spectrometry , X-Ray Diffraction
14.
Adv Mater ; : e2408450, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39240024

ABSTRACT

Tailored photophysical properties and chemical activity is the ultimate pursuit of functional dyes for in vivo biomedical theranostics. In this work, the independent regulation of the absorption and fluorescence emission wavelengths of heptamethine cyanines is reported. These dyes retain near-infrared fluorescence emission (except a nitro-modified dye) while feature variable absorption wavelengths ranging from 590 to 860 nm. This enables to obtain customized functional dyes that meet the excitation and fluorescence wavelength requirements defined by the optical properties of tissues for in vivo biomedical applications. Typically, a nitro-modified photothermal active derivative Cy-Mu-7-9 is used, which features strong absorption at 810 nm in PBS, a wavelength that balanced the tissue penetration depth and non-specific photothermal effect, to realize non-destructive inflammatory bowel disease (IBD) therapy via photothermal induced up-regulation of heat shock protein 70 in the intestinal epithelial cells. The corresponding amino-modified dye Cy-Mu-7-9-NH2, which can be formed in health enteric cavity by Cy-Mu-7-9 after oral administration, is a fluorescence compound with the emission of 800 nm in PBS. Based on the IBD sensitive transformation of Cy-Mu-7-9 and Cy-Mu-7-9-NH2, in vivo IBD theranostic and therapeutic effect evaluation is realized via the synergy of fluorescence imaging and photothermal therapy for the first time.

15.
Article in English | MEDLINE | ID: mdl-39234752

ABSTRACT

Photothermal therapy (PTT) shows promise in cancer treatments due to its good spatiotemporal selectivity and minimal invasiveness. However, PTT has some problems such as excessive heat damage to normal tissues, tumor thermo-resistance caused by heat shock proteins (HSPs), and limited efficacy of monotherapy. Here, we construct a patch named "partitioned microneedles" (PMN-SNAP/CuS), which separates the "catalyst" bovine serum albumin-based copper sulfide nanoparticles (CuS@BSA NPs) and the "reactant" S-nitroso-N-acetylpenicillamine (SNAP) into different regions of microneedles, for enhancing mild PTT (mPTT) of melanoma. PMN-SNAP/CuS showed an excellent photothermal effect, Fenton-like catalytic activity, and nitric oxide (NO) generation ability. The combination of NO and reactive oxygen species (ROS) produced by PMN-SNAP/CuS effectively blocked the synthesis of HSPs at the source and enhanced the efficacy of mPTT. Both in vitro and in vivo results proved that PMN-SNAP/CuS significantly enhanced the inhibition of melanoma under 808 nm laser irradiation. In conclusion, our partitioned microneedle strategy based on the combination of enhanced mPTT and gas therapy (GT) provides a promising approach to enhance the therapeutic effect on melanoma.

16.
Adv Sci (Weinh) ; : e2406800, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39234816

ABSTRACT

Open-shell organic radical semiconductor materials have received increasing attention in recent years due to their distinctive properties compared to the traditional materials with closed-shell singlet ground state. However, their poor chemical and photothermal stability in ambient conditions remains a significant challenge, primarily owing to their high reactivity with oxygen. Herein, a novel open-shell poly(3,4-dioxythiophene) radical PTTO2 is designed and readily synthesized for the first time using low-cost raw material via a straightforward BBr3-demethylation of the copolymer PTTOMe2 precursor. The open-shell character of PTTO2 is carefully studied and confirmed via the signal-silent 1H nuclear magnetic resonance spectrum, highly enhanced electron spin resonance signal compared with PTTOMe2, as well as the ultra-wide ultraviolet-visible-near nfraredUV-vis-NIR absorption and other technologies. Interestingly, the powder of PTTO2 exhibits an extraordinary absorption range spanning from 300 to 2500 nm and can reach 274 °C under the irradiation of 1.2 W cm-2, substantially higher than the 108 °C achieved by PTTOMe2. The low-cost PTTO2 stands as one of the best photothermal conversion materials among the pure organic photothermal materials and provides a new scaffold for the design of stable non-doped open-shell polymers.

17.
BMC Chem ; 18(1): 163, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227976

ABSTRACT

PURPOSE: The antibacterial properties of silver nanoparticles (AgNPs) are extensively identified. In large quantities, they might be harmful. So many fields of nanotechnology have shown a great deal of interest in the development of an environmentally friendly, efficient method for synthesizing metal nanoparticles. Because of its antibacterial and antifungal properties toward a wide range of microbes, chitosan silver nanoparticles (AgNPs@Cs) constitute a newly developing class of bio-nanostructured hybrid materials. Furthermore, the use of photothermal therapy (PTT) has been suggested as a means of elimination of germs. These light-stimulated treatments are minimally invasive and have a few side effects. In the present work, the antibacterial effect of AgNPs at low concentrations; prepared by chemical and green methods as antimicrobial and photothermal agents in photothermal therapy; with laser irradiation were explored as combined treatment against MRSA, Pseudomonas aeruginosa, and Klebsiella pneumoniae. METHODS: Silver nanoparticles were produced in two ways. First, by sodium borohydrides, second, by chitosan (as a natural eco-friendly reducing, and capping agent). The nanostructure of AgNPs and AgNPs@Cs was confirmed by UV-visible spectrometer, transmission electron microscopy (TEM), Fourier transform infrared spectroscopy (FTIRs), and direct light scattering (DLS). The antibacterial activity of the prepared nanoparticles and the laser irradiation was tested against three bacterial species of zoonotic importance; MRSA, Pseudomonas aeruginosa, and Klebsiella pneumoniae; and was evaluated by measuring their minimum inhibitory concentrations (MIC). RESULTS: Silver nanoparticles produced by the two methods had spherical shapes with nearly the same particle size. The analysis of DLS showed that AgNPs were very stable with zeta potential - 28.8 mv, and 47.7 mv by chemical and chitosan synthesis, respectively. Furthermore, AgNPs@Cs showed higher antibacterial activity toward the tested bacterial species than AgNPs by chemical method. Additionally, the bacterial viability using photothermal laser therapy was reduced compared to laser and AgNPs alone. The bactericidal activities were higher when laser diode was coupled with AgNPs@Cs than by chemical reduction. CONCLUSION: The laser combined treatment had a higher antimicrobial effect than AgNPs alone or laser irradiation alone.

18.
Article in English | MEDLINE | ID: mdl-39221601

ABSTRACT

One-dimensional nanomaterials have become one of the most available nanoreinforcing agents for developing next-generation high-performance functional self-healing composites owing to their unique structural characteristics and surface electron structure. However, nanoscale control, structural regulation, and crystal growth are still enormous challenges in the synthesis of specific one-dimensional nanomaterials. Here, oxygen-defective MoO3-x nanowires with abundant surface dynamic bonding were successfully synthesized as novel nanofillers and photothermal response agents combined with a polyurethane matrix to construct composite elastomers, thus achieving mechanically enhanced and self-healing properties. Benefiting from the surface plasmon resonance of the MoO3-x nanowires and interfacial multiple dynamic bonding interactions, the composite elastomers demonstrated strong mechanical performance (with a strength of 31.45 MPa and elongation of 1167.73%) and ultrafast photothermal toughness self-healing performance (20 s and an efficiency of 94.34%). The introduction of MoO3-x nanowires allows the construction of unique three-dimensional cross-linked nanonetworks that can move and regulate interfacial dynamic interactions under 808 nm infrared laser stimulation, resulting in controlled mechanical and healing performance. Therefore, such special elastomers with strong photothermal responses and mechanical properties are expected to be useful in next-generation biological antibacterial materials, wearable devices, and artificial muscles.

19.
Small ; : e2404097, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39221696

ABSTRACT

Anisotropic gold nanoparticles (AuNPs) are renowned for their unique properties - including localized surface plasmon resonance (LSPR) and adjustable optical responses to light exposure - that enable the conversion of light into heat and make them a promising tool in cancer therapy. Nonetheless, their tendency to aggregate and consequently lose their photothermal conversion capacity during prolonged irradiation periods represents a central challenge in developing anisotropic AuNPs for clinical use. To overcome this issue, an innovative approach that facilitates the encapsulation of individual anisotropic AuNPs within thin nanogels, forming hybrid nanomaterials that mirror the inorganic core's morphology while introducing a negligible (2-8 nm) increase in overall diameter is proposed. The encapsulation of rod- and star-shaped anisotropic AuNPs within poly-acrylamide (pAA) or poly-(N-isopropylacrylamide) (pNIPAM) nanogels is successfully demonstrated. The ultrathin polymeric layers display remarkable durability, significantly enhancing the photothermal stability of anisotropic AuNPs during their interaction with near-infrared light and effectively boosting their photothermal capacities for extended irradiation periods. The outcomes of the research thus support the development of more stable and reliable AuNPs as hybrid nanomaterials, positioning them as promising nanomedicinal platforms.

20.
Colloids Surf B Biointerfaces ; 245: 114188, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39226744

ABSTRACT

To maximize the therapeutic effects and minimize the adverse effects of synergistic tumor therapies, a multifunctional nanozyme Au-Bi/ZIF-8@DOX@HA (ABZ@DOX@HA) was designed and synthesized through the Au and Bi bimetallic doping of ZIF-8, loading of the DOX, and modifying with hyaluronic acid (HA). The ABZ@DOX@HA nanoparticles (NPs) could simulate the enzymatic activities of glucose oxidase (GOx) and peroxidase (POD). Upon irradiated by near-infrared region (NIR-II) laser, the strong synergism of the photothermal abilities of the loaded Au and Bi nanodots accelerated the collapse of the ABZ structure at the tumor site considerably and released Au, Bi nanodots and DOX. The results in vitro and in vivo proved that ABZ@DOX@HA nanozyme could effectively exert the combined tumor therapy of starvation treatment, photothermal therapy (PTT), chemodynamic therapy (CDT) and chemotherapy. The current research provides a new strategy to address the inherent challenges of easy clearance and short blood circulation of small-sized NPs during the treatment of tumors with nanomedicine, as well as the aggregation and oxidation of inorganic nanodots.

SELECTION OF CITATIONS
SEARCH DETAIL