Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 8.818
Filtrer
1.
Methods Mol Biol ; 2835: 277-288, 2024.
Article de Anglais | MEDLINE | ID: mdl-39105923

RÉSUMÉ

Photodynamic therapy (PDT), a noninvasive cancer treatment, relies on three components: light source, oxygen, and photosensitizer (PS). When PS is excited by a specific wavelength of light in the presence of oxygen, it leads to the generation of reactive oxygen species (ROS), which results in targeted destruction of cancer cells. The success of PDT mainly depends on the properties of the chosen PS, emphasizing selectivity, high absorbance, drug conjugation, controlled biodistribution, and low toxicity. Nanomaterials not only play an important role in photochemical activity by maximizing the absorption of photons from the light source but can also adjust the pharmacokinetics and tumor selectivity of photoactive molecules. Therefore, they can be used as a PS on their own and conjugated with other PS molecules. When combined with selectivity, high targeting capacity, and finally, light of the appropriate wavelength, the scenario results in localized ROS formation and cell death. However, the signaling pathways of PDT-induced cell death may differ depending on the cell type or nanomaterial properties. For this reason, omics analyses are needed to clarify the mechanisms underlying photodynamic reactions. Proteomics, crucial in molecular sciences, sheds light on cancer mechanisms, identifying biomarkers and therapeutic targets. Examining nanoparticle-based PDT in cancer cell lines in vitro, this chapter aims to molecularly evaluate efficacy, utilizing proteomic analysis to understand the underlying mechanisms.


Sujet(s)
Nanostructures , Tumeurs , Photothérapie dynamique , Photosensibilisants , Espèces réactives de l'oxygène , Photothérapie dynamique/méthodes , Humains , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Photosensibilisants/usage thérapeutique , Photosensibilisants/pharmacologie , Espèces réactives de l'oxygène/métabolisme , Nanostructures/composition chimique , Lignée cellulaire tumorale , Protéomique/méthodes , Nanoparticules/composition chimique
2.
Lasers Med Sci ; 39(1): 204, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39088059

RÉSUMÉ

Due to antimicrobial drug resistance, there is a growing interest in the development of light based alternative antibacterial therapies. This research work is focused on the inactivation of Escherichia coli (E. coli) by exploiting the absorption bands 405, 505, 542, 580 and 631 nm of its indigenously produced Protoporphyrin IX (PpIX) excited by three LEDs with broad emission bands at 418, 522 and 630 nm and two laser diodes with narrow emission bands at 405 and 635 nm. Fluorescence spectroscopy and plate count method have been employed for studying the inactivation rate of E. coli strain in autoclaved water suspension. It has been found that LEDs at 418, 522 and 630 nm produced pronounced antimicrobial photodynamic effect on E. coli strain comparing laser diodes at 405 and 635 nm, which might be attributed to the overlapping of broad emission bands of LEDs with the absorption bands of PpIX than narrow emission bands of laser diodes. Particular effect of LED at 522 nm has been noticed because its broad emission band overlaps three absorption bands 505, 542 and 580 nm of PpIX. The gold standard plate count method strongly correlates with Fluorescence spectroscopy, making it an innovative tool to administer bacterial inactivation. The experimental results suggested the development of a light source that entirely overlap absorption bands of PpIx to produce a pronounced antimicrobial photodynamic effect, which might become an effective modality for in vivo disinfection of antibiotic resistant microbes in wounds and lesions.


Sujet(s)
Escherichia coli , Photothérapie dynamique , Photosensibilisants , Protoporphyrines , Spectrométrie de fluorescence , Escherichia coli/effets des médicaments et des substances chimiques , Photothérapie dynamique/méthodes , Photosensibilisants/pharmacologie , Lasers à semiconducteur/usage thérapeutique , Humains
3.
Bull Exp Biol Med ; 177(2): 271-273, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-39090464

RÉSUMÉ

In female Wistar rats with breast cancer, quantitative changes of pro-oncogenic miRNAs (miR-21, -27a, and -221) and tumor-suppressive miR-429 in the mesenteric lymph node were assessed after photodynamic therapy for breast cancer and after photodynamic therapy followed surgical treatment. The level of pro-oncogenic miR-221 in the mesenteric lymph node decreased, and the level of pro-oncogenic miR-21 increased after photodynamic therapy for breast cancer followed by surgical treatment in comparison with the corresponding parameters after photodynamic therapy alone. The content of tumor-suppressive miR-429 remained reduced, as in the group of animals receiving photodynamic therapy alone.


Sujet(s)
Noeuds lymphatiques , microARN , Photothérapie dynamique , Rat Wistar , Animaux , Femelle , microARN/génétique , microARN/métabolisme , Noeuds lymphatiques/anatomopathologie , Noeuds lymphatiques/effets des médicaments et des substances chimiques , Noeuds lymphatiques/métabolisme , Photothérapie dynamique/méthodes , Rats , Tumeurs expérimentales de la mamelle/traitement médicamenteux , Tumeurs expérimentales de la mamelle/anatomopathologie , Tumeurs expérimentales de la mamelle/génétique , Tumeurs expérimentales de la mamelle/métabolisme , Mésentère/anatomopathologie , Photosensibilisants/usage thérapeutique , Photosensibilisants/pharmacologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Tumeurs du sein/anatomopathologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Métastase lymphatique
4.
Commun Biol ; 7(1): 945, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39107369

RÉSUMÉ

Photosensitizing fluorescence protein is a promising tool for chromophore-assisted light inactivation (CALI) that enables specific oxidation and inactivation of intracellular molecules. However, a commonly used monomeric photosensitizing fluorescent protein, SuperNova, shows a low CALI efficiency due to its insufficient maturation at 37 °C, thereby limiting the application of CALI to various molecules, especially in mammalian cells. Here, we present a photosensitizing fluorescence protein, HyperNova, with markedly improved maturation at 37 °C, leading to greatly enhanced CALI efficiency. Exploiting this quality, HyperNova enables the application of CALI to variety of molecules such as a mitotic kinase and transcriptional factors that were highly challenging with conventional SuperNova. To further demonstrate the utility of HyperNova, we have also succeeded in developing novel CALI techniques for MAP kinases by HyperNova. Our findings suggest that HyperNova has the potential to expand the molecular toolbox for manipulating biological events in living cells, providing new avenues for investigating cellular signaling pathways.


Sujet(s)
Protéines luminescentes , Protéines luminescentes/métabolisme , Protéines luminescentes/génétique , Humains , Inactivation au moyen d'une lumière assistée par des chromophores , Photosensibilisants/pharmacologie , Cellules HeLa , Lumière , Animaux
5.
Gen Physiol Biophys ; 43(5): 469-484, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39140687

RÉSUMÉ

Ruthenium nitrosyl (Ru-NO) complexes are of interest as photoactive nitric oxide (NO) donor candidates for local therapeutic applications. NO plays a crucial regulatory role in skin homeostasis, concentration-dependently affecting processes like the proliferation, apoptosis, autophagy and redox balance. In this context, we investigated HE-10, a ruthenium-based photoinducible NO donor, for its pro-oxidant and cytotoxic effects under light and dark conditions in VH10 human foreskin fibroblast cells. We also tested its intracellular and extracellular NO-releasing function. Our study reveals a significant dose-dependent cytotoxic effect of HE-10, an increase in intracellular reactive oxygen and nitrogen species, and the occurrence of apoptosis in skin fibroblast cells. Furthermore, exposure to both increasing doses of HE-10 and white LED light led to substantial cellular events, including a significant induction of autophagy and G2/M phase cell cycle arrest. Paradoxically, these effects were not solely attributable to NO release based on DAF2-DA NO probe results, suggesting that intracellular photochemical reactions additional to NO photolysis contribute to HE-10's biological activity. This study shows that HE-10 exhibits both cytotoxic and potential therapeutic effects, depending on concentration and light exposure. These findings are crucial for developing targeted Ru-NO complex treatments for skin diseases and potentially certain types of skin cancer, where controlled NO release could be beneficial.


Sujet(s)
Fibroblastes , Monoxyde d'azote , Humains , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Monoxyde d'azote/métabolisme , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Ruthénium/composition chimique , Photosensibilisants/pharmacologie , Photosensibilisants/composition chimique , Espèces réactives de l'oxygène/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Donneur d'oxyde nitrique/pharmacologie , Donneur d'oxyde nitrique/composition chimique , Relation dose-effet des médicaments , Lumière
6.
Artif Cells Nanomed Biotechnol ; 52(1): 384-398, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39101753

RÉSUMÉ

Nanotechnology-based cancer treatment has received considerable attention, and these treatments generally use drug-loaded nanoparticles (NPs) to target and destroy cancer cells. Nanotechnology combined with photodynamic therapy (PDT) has demonstrated positive outcomes in cancer therapy. Combining nanotechnology and PDT is effective in targeting metastatic cancer cells. Nanotechnology can also increase the effectiveness of PDT by targeting cells at a molecular level. Dendrimer-based nanoconjugates (DBNs) are highly stable and biocompatible, making them suitable for drug delivery applications. Moreover, the hyperbranched structures in DBNs have the capacity to load hydrophobic compounds, such as photosensitizers (PSs) and chemotherapy drugs, and deliver them efficiently to tumour cells. This review primarily focuses on DBNs and their potential applications in cancer treatment. We discuss the chemical design, mechanism of action, and targeting efficiency of DBNs in tumour metastasis, intracellular trafficking in cancer treatment, and DBNs' biocompatibility, biodegradability and clearance properties. Overall, this study will provide the most recent insights into the application of DBNs and PDT in cancer therapy.


DBNs' intracellular journey in cancer-PDT refines targeted therapy, boosting efficacy.DBN in PDT for tumour metastasis: targeting and drug release mechanisms.DBNs' biocompatibility, biodegradability and clearance were explored thoroughly.


Sujet(s)
Dendrimères , Nanoconjugués , Tumeurs , Photothérapie dynamique , Humains , Dendrimères/composition chimique , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Nanoconjugués/composition chimique , Nanoconjugués/usage thérapeutique , Animaux , Photosensibilisants/composition chimique , Photosensibilisants/usage thérapeutique , Photosensibilisants/pharmacologie , Transport biologique , Espace intracellulaire/métabolisme , Espace intracellulaire/effets des médicaments et des substances chimiques , Vecteurs de médicaments/composition chimique
7.
Photochem Photobiol Sci ; 23(8): 1573-1586, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39103724

RÉSUMÉ

Since the molecular mechanisms behind adaptation and the bacterial stress response toward antimicrobial photodynamic therapy (aPDT) are not entirely clear yet, the aim of the present study was to investigate the transcriptomic stress response in Escherichia coli after sublethal treatment with aPDT using RNA sequencing (RNA-Seq). Planktonic cultures of stationary phase E. coli were treated with aPDT using a sublethal dose of the photosensitizer SAPYR. After treatment, RNA was extracted, and RNA-Seq was performed on the Illumina NextSeq 500. Differentially expressed genes were analyzed and validated by qRT-PCR. Furthermore, expression of specific stress response proteins was investigated using Western blot analysis.The analysis of the differential gene expression following pathway enrichment analysis revealed a considerable number of genes and pathways significantly up- or down-regulated in E. coli after sublethal treatment with aPDT. Expression of 1018 genes was up-regulated and of 648 genes was down-regulated after sublethal treatment with aPDT as compared to irradiated controls. Analysis of differentially expressed genes and significantly de-regulated pathways showed regulation of genes involved in oxidative stress response and bacterial membrane damage. In conclusion, the results show a transcriptomic stress response in E. coli upon exposure to aPDT using SAPYR and give an insight into potential molecular mechanisms that may result in development of adaptation.


Sujet(s)
Escherichia coli , Photothérapie dynamique , Photosensibilisants , Escherichia coli/effets des médicaments et des substances chimiques , Photosensibilisants/pharmacologie , RNA-Seq , Antibactériens/pharmacologie , Stress physiologique/effets des médicaments et des substances chimiques
8.
Anal Chim Acta ; 1320: 343035, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39142775

RÉSUMÉ

BACKGROUND: Photodynamic therapy (PDT) is a pioneering and effective anticancer modality with low adverse effects and high selectivity. Hypochlorous acid or hypochlorite (HClO/ClO-) is a type of inflammatory cytokine. The abnormal increase of ClO- in tumor cells is related to tumor pathogenesis and may be a "friend" for the design and synthesis of responsive phototherapy agents. However, preparing responsive phototherapy agents for all-in-one noninvasive diagnosis and simultaneous in situ therapy in a complex tumor environment is highly desirable but still remains an enormously demanding task. RESULTS: An acceptor-π bridge-donor-π bridge-acceptor (A-π-D-π-A) type photosensitizer TPTPy was designed and synthesized based on the phenothiazine structure which was used as the donor moiety as well as a ClO- responsive group. TPTPy was a multifunctional mitochondria targeted aggregation-induced emission (AIE) photosensitizer which could quickly and sensitively respond to ClO- with fluorescence "turn on" performance (19-fold fluorescence enhancement) and enhanced type I reactive oxygen species (ROS) generation to effectively ablate hypoxic tumor cells. The detection limit of TPTPy to ClO- was calculated to be 185.38 nM. The well-tailored TPTPy anchoring to mitochondria and producing ROS in situ could disrupt mitochondria and promote cell apoptosis. TPTPy was able to image inflammatory cells and tumor cells through ClO- response. In vivo results revealed that TPTPy was successfully utilized for PDT in tumor bearing nude mice and exhibited excellent biological safety for major organs. SIGNIFICANCE AND NOVELTY: A win-win integration strategy was proposed to design a tumor intracellular ClO- responsive photosensitizer TPTPy capable of both type I and type II ROS production to achieve photodynamic therapy of tumor. This work sheds light on the win-win integration design by taking full advantage of the characteristics of tumor microenvironment to build up responsive photosensitizer for in situ PDT of tumor.


Sujet(s)
Acide hypochloreux , Mitochondries , Photothérapie dynamique , Photosensibilisants , Photosensibilisants/composition chimique , Photosensibilisants/pharmacologie , Photosensibilisants/synthèse chimique , Photosensibilisants/usage thérapeutique , Acide hypochloreux/analyse , Acide hypochloreux/métabolisme , Animaux , Humains , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Souris , Espèces réactives de l'oxygène/métabolisme , Espèces réactives de l'oxygène/analyse , Souris de lignée BALB C , Phénothiazines/composition chimique , Phénothiazines/pharmacologie , Souris nude , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Imagerie optique , Survie cellulaire/effets des médicaments et des substances chimiques
9.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39125706

RÉSUMÉ

Oral candidiasis is a common problem among immunocompetent patients. The frequent resistance of Candida strains to popular antimycotics makes it necessary to look for alternative methods of treatment. The authors conducted a systematic review following the PRISMA 2020 guidelines. The objective of this review was to determine if curcumin-mediated blue light could be considered as an alternative treatment for oral candidiasis. PubMed, Google Scholar, and Cochrane Library databases were searched using a combination of the following keywords: (Candida OR candidiasis oral OR candidiasis oral OR denture stomatitis) AND (curcumin OR photodynamic therapy OR apt OR photodynamic antimicrobial chemotherapy OR PACT OR photodynamic inactivation OR PDI). The review included in vitro laboratory studies with Candida spp., in vivo animal studies, and randomized control trials (RCTs) involving patients with oral candidiasis or prosthetic stomatitis, published only in English. The method of elimination of Candida species in the studies was curcumin-mediated aPDT. A total of 757 studies were identified. Following the analysis of the titles and abstracts of the studies, only 42 studies were selected for in-depth screening, after which 26 were included in this study. All studies evaluated the antifungal efficacy of curcumin-mediated aPDT against C. albicans and non-albicans Candida. In studies conducted with planktonic cells solutions, seven studies demonstrated complete elimination of Candida spp. cells. The remaining studies demonstrated only partial elimination. In all cases, experiments on single-species yeast biofilms demonstrated partial, statistically significant inhibition of cell growth and reduction in biofilm mass. In vivo, curcumin-mediated aPDT has shown good antifungal activity against oral candidiasis also in an animal model. However, its clinical efficacy as a potent therapeutic strategy for oral candidiasis requires few further RCTs.


Sujet(s)
Candida , Candidose buccale , Curcumine , Photothérapie dynamique , Curcumine/pharmacologie , Photothérapie dynamique/méthodes , Humains , Candida/effets des médicaments et des substances chimiques , Animaux , Candidose buccale/traitement médicamenteux , Candidose buccale/microbiologie , Antifongiques/pharmacologie , Antifongiques/usage thérapeutique , Photosensibilisants/pharmacologie , Biofilms/effets des médicaments et des substances chimiques
10.
ACS Appl Mater Interfaces ; 16(32): 41843-41854, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39092532

RÉSUMÉ

Oxidative degradation of the pathogenic amyloid-ß-peptide (Aß) aggregation is an effective and promising method to treat Alzheimer's disease under light irradiation. However, the limited penetration of external light sources into deep tissues has hindered the development of this treatment. Therefore, we have designed an unprecedented chemiluminescence-initiated photodynamic therapy system to replace external laser irradiation, primarily composed of d-glucose-based polyoxalate (G-poly(oxalate)), the novel photosensitizer (BD-Se-QM), and bis [2,4,5-trichloro-6-(pentoxy-carbonyl) phenyl] ester. BD-Se-QM possesses excellent singlet oxygen (1O2) generation efficiency and the ability to photooxidize Aß1-42 aggregates under white light. G-poly(oxalate) not only helps the nanosystem to cross the blood-brain barrier but also has sufficient oxalate ester groups to significantly enhance the efficiency of chemiluminescence resonance energy transfer. The oxalate ester groups in BD-Se-QM/NPs can chemically react with H2O2 to produce high-energy intermediates that activate BD-Se-QM, which can generate 1O2 to inhibit Aß1-42 aggregates and also promote microglial uptake of Aß1-42, reducing the Aß1-42-induced neurotoxicity. The chemically stimulated nanoplatform not only solves the drug delivery problem but also eliminates the need for external light sources. We anticipate that this chemically excited nanosystem could also be used for targeted delivery of other small molecule drugs.


Sujet(s)
Peptides bêta-amyloïdes , Oxydoréduction , Fragments peptidiques , Photosensibilisants , Peptides bêta-amyloïdes/métabolisme , Peptides bêta-amyloïdes/composition chimique , Photosensibilisants/composition chimique , Photosensibilisants/pharmacologie , Fragments peptidiques/composition chimique , Fragments peptidiques/métabolisme , Photothérapie dynamique , Oxygène singulet/métabolisme , Oxygène singulet/composition chimique , Humains , Animaux , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Lumière , Peroxyde d'hydrogène/composition chimique , Agrégats de protéines/effets des médicaments et des substances chimiques , Souris
11.
Molecules ; 29(15)2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39125107

RÉSUMÉ

Photothermal, photodynamic and sonodynamic cancer therapies offer opportunities for precise tumor ablation and reduce side effects. The cyclic guanylate adenylate synthase-stimulator of interferon genes (cGAS-STING) pathway has been considered a potential target to stimulate the immune system in patients and achieve a sustained immune response. Combining photothermal, photodynamic and sonodynamic therapies with cGAS-STING agonists represents a newly developed cancer treatment demonstrating noticeable innovation in its impact on the immune system. Recent reviews have concentrated on diverse materials and their function in cancer therapy. In this review, we focus on the molecular mechanism of photothermal, photodynamic and sonodynamic cancer therapies and the connected role of cGAS-STING agonists in treating cancer.


Sujet(s)
Protéines membranaires , Tumeurs , Nucleotidyltransferases , Photothérapie dynamique , Humains , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Nucleotidyltransferases/métabolisme , Protéines membranaires/métabolisme , Photothérapie dynamique/méthodes , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Ultrasonothérapie/méthodes , Photosensibilisants/pharmacologie , Photosensibilisants/usage thérapeutique , Photosensibilisants/composition chimique
12.
Int J Mol Sci ; 25(15)2024 Aug 04.
Article de Anglais | MEDLINE | ID: mdl-39126073

RÉSUMÉ

The spread of multidrug-resistant mycobacterium strains requires the development of new approaches to combat diseases caused by these pathogens. For that, photodynamic inactivation (PDI) is a promising approach. In this study, a tricarbocyanine (TCC) is used for the first time as a near-infrared (740 nm) activatable PDI photosensitizer to kill mycobacteria with deep light penetration. For better targeting, a novel tricarbocyanine dye functionalized with two trehalose units (TCC2Tre) is developed. The photodynamic effect of the conjugates against mycobacteria, including Mycobacterium tuberculosis, is evaluated. Under irradiation, TCC2Tre causes more effective killing of mycobacteria compared to the photosensitizer without trehalose conjugation, with 99.99% dead vegetative cells of M. tuberculosis and M. smegmatis. In addition, effective photoinactivation of dormant forms of M. smegmatis is observed after incubation with TCC2Tre. Mycobacteria treated with TCC2Tre are more sensitive to 740 nm light than the Gram-positive Micrococcus luteus and the Gram-negative Escherichia coli. For the first time, this study demonstrates the proof of principle of in vitro PDI of mycobacteria including the fast-growing M. smegmatis and the slow-growing M. tuberculosis using near-infrared activatable photosensitizers conjugated with trehalose. These findings are useful for the development of new efficient alternatives to antibiotic therapy.


Sujet(s)
Rayons infrarouges , Mycobacterium smegmatis , Mycobacterium tuberculosis , Photosensibilisants , Tréhalose , Mycobacterium smegmatis/effets des médicaments et des substances chimiques , Mycobacterium smegmatis/effets des radiations , Tréhalose/pharmacologie , Tréhalose/composition chimique , Mycobacterium tuberculosis/effets des médicaments et des substances chimiques , Mycobacterium tuberculosis/effets des radiations , Photosensibilisants/pharmacologie , Photosensibilisants/composition chimique , Carbocyanines/composition chimique , Carbocyanines/pharmacologie , Photothérapie dynamique/méthodes
13.
ACS Nano ; 18(32): 21009-21023, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39087239

RÉSUMÉ

Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), induce high morbidity and mortality rates, which challenge the present approaches for the treatment of ALI/ARDS. The clinically used photosensitizer verteporfin (VER) exhibits great potential in the treatment of acute lung injury and acute respiratory distress syndrome (ALI/ARDS) by regulating macrophage polarization and reducing inflammation. Nevertheless, its hydrophobic characteristics, nonspecificity, and constrained bioavailability hinder its therapeutic efficacy. In this work, we developed a type of VER-cored artificial exosome (EVM), which was produced by using mesoporous silica nanoparticles (MSNs) to load VER, followed by the exocytosis of internalized VER-MSNs from mouse bone marrow-derived mesenchymal stem cells (mBMSCs) without further modification. Both in vitro and in vivo assessments confirmed the powerful anti-inflammation induced by EVM. EVM also showed significant higher accumulation to inflammatory lungs compared with healthy ones, which was beneficial to the treatment of ALI/ARDS. EVM improved pulmonary function, attenuated lung injury, and reduced mortality in ALI mice with high levels of biocompatibility, exhibiting a 5-fold higher survival rate than the control. This type of artificial exosome emitted near-infrared light in the presence of laser activation, which endowed EVM with trackable ability both in vitro and in vivo. Our work developed a type of clinically used photosensitizer-loaded artificial exosome with membrane integrity and traceability. To the best of our knowledge, this kind of intracellularly synthesized artificial exosome was developed and showed great potential in ALI/ARDS therapy.


Sujet(s)
Lésion pulmonaire aigüe , Exosomes , Silice , Animaux , Lésion pulmonaire aigüe/traitement médicamenteux , Lésion pulmonaire aigüe/anatomopathologie , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/thérapie , Souris , Exosomes/métabolisme , Exosomes/composition chimique , Silice/composition chimique , Vertéporfine/pharmacologie , Vertéporfine/composition chimique , Vertéporfine/usage thérapeutique , Nanoparticules/composition chimique , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/métabolisme , Souris de lignée C57BL , Mâle , Photosensibilisants/pharmacologie , Photosensibilisants/composition chimique , Porosité
14.
ACS Nano ; 18(26): 17086-17099, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38952327

RÉSUMÉ

Traditional external field-assisted therapies, e.g., microwave (MW) therapy and phototherapy, cannot effectively and minimally damage eliminate deep-seated infection, owing to the poor penetrability of light and low reactive oxygen species (ROS) stimulation capability of MW. Herein, an implantable and wireless-powered therapeutic platform (CNT-FeTHQ-TS), in which external MW can be converted into internal light via MW wireless-powered light-emitting chips, is designed to eradicate deep-seated tissue infections by MW-induced deep-seated photodynamic therapy. In application, CNT-FeTHQ-TS is implanted at internal lesions, and the chip emits light under external MW irradiation. Subsequently, CNT-FeTHQ coating in the platform can respond to both MW and light simultaneously to generate ROS and MW-hyperthermia for rapid and precise sterilization at focus. Importantly, MW also improves the photodynamic performance of CNT-FeTHQ by introducing vacancies in FeTHQ to facilitate the photoexcitation process and changing the spin state of electrons to inhibit the complexation of photogenerated electron-hole pairs, which were confirmed by simulation calculations and in situ MW-irradiated photoluminescence experiments. In vivo, CNT-FeTHQ-TS can effectively cure mice with Staphylococcus aureus infection in dorsal subcutaneous tissue. This work overcomes the key clinical limitations of safe energy transmission and conversion for treating deep-seated infections.


Sujet(s)
Micro-ondes , Photothérapie dynamique , Animaux , Souris , Espèces réactives de l'oxygène/métabolisme , Technologie sans fil , Photosensibilisants/pharmacologie , Photosensibilisants/composition chimique , Lumière , Staphylococcus aureus/effets des médicaments et des substances chimiques , Infections à staphylocoques/traitement médicamenteux , Souris de lignée BALB C , Antibactériens/pharmacologie , Antibactériens/composition chimique
15.
Methods Mol Biol ; 2833: 51-56, 2024.
Article de Anglais | MEDLINE | ID: mdl-38949700

RÉSUMÉ

Photodynamic therapy (PDT) is an established therapy used for the treatment of cutaneous skin cancers and other non-infective ailments. There has been recent interest in the opportunity to use aPDT (antimicrobial PDT) to treat skin and soft tissue infections. PDT utilizes photosensitizers that infiltrate all cells and "sensitize" them to a given wavelength of light. The photosensitizer is simply highly absorbent to a given wavelength of light and when excited will produce, in the presence of oxygen, damaging oxygen radicals and singlet oxygen. Bacterial cells are comparatively poor at combatting oxidative stress when compared with human cells therefore a degree of selective toxicity can be achieved with aPDT.In this chapter, we outline methodologies for testing aPDT in vitro using standard lab equipment.


Sujet(s)
Photothérapie dynamique , Photosensibilisants , Photosensibilisants/pharmacologie , Photosensibilisants/composition chimique , Photothérapie dynamique/méthodes , Humains , Oxygène singulet/métabolisme , Anti-infectieux/pharmacologie
16.
Eur J Med Chem ; 275: 116638, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-38950489

RÉSUMÉ

The cGAS (cyclic GMP-AMP synthase)-STING (stimulator of interferon genes) pathway promotes antitumor immune responses by sensing cytosolic DNA fragments leaked from nucleus and mitochondria. Herein, we designed a highly charged ruthenium photosensitizer (Ru1) with a ß-carboline alkaloid derivative as the ligand for photo-activating of the cGAS-STING pathway. Due to the formation of multiple non-covalent intermolecular interactions, Ru1 can self-assemble into carrier-free nanoparticles (NPs). By incorporating the triphenylphosphine substituents, Ru1 can target and photo-damage mitochondrial DNA (mtDNA) to cause the cytoplasmic DNA leakage to activate the cGAS-STING pathway. Finally, Ru1 NPs show potent antitumor effects and elicit intense immune responses in vivo. In conclusion, we report the first self-assembling mtDNA-targeted photosensitizer, which can effectively activate the cGAS-STING pathway, thus providing innovations for the design of new photo-immunotherapeutic agents.


Sujet(s)
Antinéoplasiques , Immunothérapie , Protéines membranaires , Nucleotidyltransferases , Photosensibilisants , Ruthénium , Photosensibilisants/composition chimique , Photosensibilisants/pharmacologie , Photosensibilisants/synthèse chimique , Humains , Nucleotidyltransferases/métabolisme , Protéines membranaires/métabolisme , Animaux , Ruthénium/composition chimique , Ruthénium/pharmacologie , Souris , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Structure moléculaire , Relation dose-effet des médicaments , Nanoparticules/composition chimique , Relation structure-activité , Tests de criblage d'agents antitumoraux , ADN mitochondrial/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie
17.
Dalton Trans ; 53(28): 11836-11849, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38949269

RÉSUMÉ

A novel lysosome-targeted photosensitizer/photoredox catalyst based on cyclometalated Ir(III) complex IrL has been designed and synthesized, which exhibited excellent phosphorescence properties and the ability to generate single oxygen (1O2) and photocatalytically oxidize 1,4-dihydronicotinamide adenine dinucleotide (NADH) under light irradiation. Most importantly, the aforementioned activities are significantly enhanced due to protonation under acidic conditions, which makes them highly attractive in light-activated tumor therapy, especially for acidic lysosomes and tumor microenvironments. The photocytotoxicity of IrL and the mechanism of cell death have been investigated. Additionally, the tumor-killing ability of IrL under light irradiation was evaluated using a 4T1 tumor-bearing mouse model. This work provides a strategy for the development of lysosome-targeted photosensitizers/photoredox catalysts to overcome hypoxic tumors.


Sujet(s)
Complexes de coordination , Iridium , Lysosomes , Oxydoréduction , Photosensibilisants , Lysosomes/métabolisme , Iridium/composition chimique , Iridium/pharmacologie , Photosensibilisants/composition chimique , Photosensibilisants/pharmacologie , Photosensibilisants/synthèse chimique , Animaux , Catalyse , Complexes de coordination/composition chimique , Complexes de coordination/pharmacologie , Complexes de coordination/synthèse chimique , Souris , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Humains , Photothérapie dynamique , Lignée cellulaire tumorale , Lumière , Souris de lignée BALB C
18.
ACS Nano ; 18(28): 18230-18245, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38950337

RÉSUMÉ

Therapy-induced modulation of the tumor microenvironment (TME) to overcome the immunosuppressive TME is considered to be an opportunity for cancer treatment. However, monitoring of TME modulation during the therapeutic process to accurately determine immune responses and adjust treatment plans in a timely manner remains to be challenging. Herein, we report a carrier-free nanotheranostic system (CANPs) assembled by two boron dipyrromethene (BODIPY) dyes, a sonophotosensitizer C-BDP, and a nitric oxide (NO) probe amino-BODIPY (A-BDP). CANPs can exert combined sonophototherapeutic effects of C-BDP under ultrasound and light irradiation and simultaneously induce inflammatory TME, as well as emit bright fluorescence via A-BDP by monitoring tumor-associated macrophages (TAMs) repolarization through the released NO in vitro and in vivo. Of note, transforming growth factor-ß (TGF-ß) could be the key cytokine involved in the sonophototherapy-induced TME reprogramming. By virtue of high physiological stability, good biocompatibility, and effective tumor targetability, CANPs could be a potential nanotheranostic system for the simultaneous induction and detection of TME reprogramming triggered by sonophototherapy.


Sujet(s)
Nanomédecine théranostique , Microenvironnement tumoral , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Animaux , Souris , Porphobilinogène/analogues et dérivés , Porphobilinogène/composition chimique , Porphobilinogène/pharmacologie , Humains , Photosensibilisants/composition chimique , Photosensibilisants/pharmacologie , Composés du bore/composition chimique , Composés du bore/pharmacologie , Monoxyde d'azote/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Femelle , Nanoparticules/composition chimique , Colorants fluorescents/composition chimique , Colorants fluorescents/pharmacologie , Cellules RAW 264.7
19.
J Photochem Photobiol B ; 257: 112971, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38955081

RÉSUMÉ

Bovine mastitis (BM) represents a significant challenge in the dairy industry. Limitations of conventional treatments have prompted the exploration of alternative approaches, such as photodynamic inactivation (PDI). In this study, we developed a PDI protocol to eliminate BM-associated pathogens using porphyrin-doped conjugated polymer nanoparticles (CPN). The PDI-CPN protocol was evaluated in four mastitis isolates of Staphylococcus and in a hyper-biofilm-forming reference strain. The results in planktonic cultures demonstrated that PDI-CPN exhibited a bactericidal profile upon relatively low light doses (∼9.6 J/cm2). Furthermore, following a seven-hour incubation period, no evidence of cellular reactivation was observed, indicating a highly efficient post-photodynamic inactivation effect. The successful elimination of bacterial suspensions encouraged us to test the PDI-CPN protocol on mature biofilms. Treatment using moderate light dose (∼64.8 J/cm2) reduced biofilm biomass and metabolic activity by up to 74% and 88%, respectively. The impact of PDI-CPN therapy on biofilms was investigated using scanning electron microscopy (SEM), which revealed nearly complete removal of the extracellular matrix and cocci. Moreover, ex vivo studies conducted on bovine udder skin demonstrated the efficacy of the therapy in eliminating bacteria from these scaffolds and its potential as a prophylactic method. Notably, the histological analysis of skin revealed no signs of cellular degeneration, suggesting that the protocol is safe and effective for BM treatment. Overall, this study demonstrates the potential of PDI-CPN in treating and preventing BM pathogens. It also provides insights into the effects of PDI-CPN on bacterial growth, metabolism, and survival over extended periods, aiding the development of effective control strategies and the optimization of future treatments.


Sujet(s)
Biofilms , Lumière , Mammite bovine , Nanoparticules , Polymères , Animaux , Bovins , Nanoparticules/composition chimique , Mammite bovine/microbiologie , Mammite bovine/traitement médicamenteux , Biofilms/effets des médicaments et des substances chimiques , Biofilms/effets des radiations , Femelle , Polymères/composition chimique , Polymères/pharmacologie , Photosensibilisants/pharmacologie , Photosensibilisants/composition chimique , Porphyrines/composition chimique , Porphyrines/pharmacologie , Staphylococcus/effets des médicaments et des substances chimiques , Staphylococcus/effets des radiations , Antibactériens/pharmacologie , Antibactériens/composition chimique , Microscopie électronique à balayage , Photothérapie dynamique
20.
J Photochem Photobiol B ; 257: 112974, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38964021

RÉSUMÉ

Pseudomonas aeruginosa, a gram-negative bacterium, accounts for 7% of all hospital-acquired infections. Despite advances in medicine and antibiotic therapy, P. aeruginosa infection still results in high mortality rates of up to 62% in certain patient groups. This bacteria is also known to form biofilms, that are 10 to 1000 times more resistant to antibiotics compared to their free-floating counterparts. Photodynamic Inactivation (PDI) has been proved to be an effective antimicrobial technique for microbial control. This method involves the incubation of the pathogen with a photosensitizer (PS), then, a light at appropriated wavelength is applied, leading to the production of reactive oxygen species that are toxic to the microbial cells. Studies have focused on strategies to enhance the PDI efficacy, such as a pre-treatment with enzymes to degrade the biofilm matrix and/or an addition of inorganic salts to the PS. The aim of the present study is to evaluate the effectiveness of PDI against P. aeruginosa biofilm in association with the application of the enzymes prior to PDI (enzymatic pre-treatment) or the addition of potassium iodide (KI) to the photosensitizer solution, to increase the inactivation effectiveness of the treatment. First, a range of enzymes and PSs were tested, and the best protocols for combined treatments were selected. The results showed that the use of enzymes as a pre-treatment was effective to reduce the total biomass, however, when associated with PDI, mild bacterial reductions were obtained. Then, the use of KI in association with the PS was evaluated and the results showed that, PDI mediated by methylene blue (MB) in the presence of KI was able to completely eradicate the biofilm. However, when the PDI was performed with curcumin and KI, no additive reduction was observed. In conclusion, out of all strategies evaluated in the present study, the most promising strategy to improve PDI against P. aeruginosa biofilm was the use of KI in association with MB, resulting in eradication with 108 log bacterial inactivation.


Sujet(s)
Biofilms , Photosensibilisants , Iodure de potassium , Pseudomonas aeruginosa , Pseudomonas aeruginosa/effets des médicaments et des substances chimiques , Pseudomonas aeruginosa/physiologie , Biofilms/effets des médicaments et des substances chimiques , Biofilms/effets des radiations , Iodure de potassium/pharmacologie , Iodure de potassium/composition chimique , Photosensibilisants/pharmacologie , Photosensibilisants/composition chimique , Lumière , Photothérapie dynamique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE