RESUMO
While numerous methods exist for diagnosing tumors through the detection of miRNA within tumor cells, few can simultaneously achieve both tumor diagnosis and treatment. In this study, a novel graphene oxide (GO)-based DNA nanodevice (DND), initiated by miRNA, was developed for fluorescence signal amplification imaging and photodynamic therapy in tumor cells. After entering the cells, tumor-associated miRNA drives DND to Catalyzed hairpin self-assembly (CHA). The CHA reaction generated a multitude of DNA Y-type structures, resulting in a substantial amplification of Ce6 fluorescence release and the generation of numerous singlet oxygen (1O2) species induced by laser irradiation, consequently inducing cell apoptosis. In solution, DND exhibited high selectivity and sensitivity to miRNA-21, with a detection limit of 11.47 pM. Furthermore, DND discriminated between normal and tumor cells via fluorescence imaging and specifically generated O21 species in tumor cells upon laser irradiation, resulting in tumor cells apoptosis. The DND offer a new approach for the early diagnosis and timely treatment of malignant tumors.
Assuntos
DNA , Grafite , MicroRNAs , Fotoquimioterapia , Nanomedicina Teranóstica , Fotoquimioterapia/métodos , Humanos , MicroRNAs/análise , Grafite/química , Nanomedicina Teranóstica/métodos , DNA/química , Apoptose/efeitos dos fármacos , Imagem Óptica , Linhagem Celular Tumoral , Oxigênio Singlete/metabolismo , Oxigênio Singlete/química , Neoplasias/tratamento farmacológico , Neoplasias/diagnóstico por imagemRESUMO
Ferroptosis, a recently identified form of cell death, holds promise for cancer therapy, but concerns persist regarding its uncontrolled actions and potential side effects. Here, we present a semiconducting polymer nanoprodrug (SPNpro) featuring an innovative ferroptosis prodrug (DHU-CBA7) to induce sono-activatable ferroptosis for tumor-specific therapy. DHU-CBA7 prodrug incorporate methylene blue, ferrocene and urea bond, which can selectively and specifically respond to singlet oxygen (1O2) to turn on ferroptosis action via rapidly cleaving the urea bonds. DHU-CBA7 prodrug and a semiconducting polymer are self-assembled with an amphiphilic polymer to construct SPNpro. Ultrasound irradiation of SPNpro leads to the production of 1O2 via sonodynamic therapy (SDT) of the semiconducting polymer, and the generated 1O2 activated DHU-CBA7 prodrug to achieve sono-activatable ferroptosis. Consequently, SPNpro combine SDT with the controlled ferroptosis to effectively cure 4T1 tumors covered by 2-cm tissue with a tumor inhibition efficacy as high as 100 %, and also completely restrain tumor metastases. This study introduces a novel sono-activatable prodrug strategy for regulating ferroptosis, allowing for precise cancer therapy.
Assuntos
Ferroptose , Camundongos Endogâmicos BALB C , Polímeros , Pró-Fármacos , Semicondutores , Ferroptose/efeitos dos fármacos , Pró-Fármacos/farmacologia , Pró-Fármacos/química , Pró-Fármacos/uso terapêutico , Animais , Polímeros/química , Feminino , Linhagem Celular Tumoral , Camundongos , Terapia por Ultrassom/métodos , Nanopartículas/química , Humanos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos/química , Oxigênio Singlete/metabolismoRESUMO
We made structural modifications on the A-ring of camptothecin (CPT) by incorporating methyl substituents on positions 9 and 12. This allows conversion of the camptothecin-derivative to an endoperoxide (ENDO-CPT). The endoperoxide obtained this way thermally releases singlet oxygen, reverting back to the original 9,12-dimethylcamptothecin (DM-CPT) with a half-life of 1.4 hours at 37 °C. Endoperoxide modification yields a significant improvement in cytotoxicity against MDR-cell lines, compared to both CPT and DM-CPT. It appears that the simultaneous action of singlet oxygen and CPT is highly effective due to the targeting of P-glycoprotein by singlet oxygen.
Assuntos
Camptotecina , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Oxigênio Singlete , Camptotecina/química , Camptotecina/farmacologia , Oxigênio Singlete/química , Oxigênio Singlete/metabolismo , Humanos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Linhagem Celular Tumoral , Peróxidos/química , Peróxidos/farmacologia , Estrutura Molecular , Ensaios de Seleção de Medicamentos Antitumorais , Sobrevivência Celular/efeitos dos fármacosRESUMO
Over the past few years, numerous bacterial strains have become resistant to selected drugs from various therapeutic groups. A potential tool in the fight against these strains is antimicrobial photodynamic therapy (APDT). APDT acts in a non-specific manner by generating reactive oxygen species and radicals, thereby inducing multidimensional intracellular effects. Importantly, the chance that bacteria will develop defense mechanisms against APDT is considered to be low. In our research, we performed the synthesis and physicochemical characterization of curcumin derivatives enriched with morpholine motifs. The obtained compounds were assessed regarding photostability, singlet oxygen generation, aggregation, and acute toxicity toward prokaryotic Aliivibrio fischeri cells in the Microtox® test. The impact of the compounds on the survival of eukaryotic cells in the MTT assay was also tested (WM266-4, WM115-melanoma, MRC-5-lung fibroblasts, and PHDF-primary human dermal fibroblasts). Initial studies determining the photocytotoxicity, and thus the potential APDT usability, were conducted with the following microbial strains: Candida albicans, Escherichia coli, Staphylococcus aureus, Streptococcus pneumoniae, and Pseudomonas aeruginosa. It was noted that the exposure of bacteria to LED light at 470 nm (fluence: 30 J/cm2) in the presence of quaternized curcumin derivatives at the conc. of 10 µM led to a reduction in Staphylococcus aureus survival of over 5.4 log.
Assuntos
Antibacterianos , Curcumina , Luz , Curcumina/farmacologia , Curcumina/química , Humanos , Antibacterianos/farmacologia , Antibacterianos/química , Antibacterianos/síntese química , Testes de Sensibilidade Microbiana , Aliivibrio fischeri/efeitos dos fármacos , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/síntese química , Sobrevivência Celular/efeitos dos fármacos , Oxigênio Singlete/metabolismo , Pseudomonas aeruginosa/efeitos dos fármacos , Fotoquimioterapia , Bactérias/efeitos dos fármacos , Luz AzulRESUMO
Phototheranostics with near-infrared fluorescence and reactive oxygen species generation ability and high photothermal conversion efficiency (PCE) plays a significant role in fluorescence imaging-guided synergetic photodynamic and photothermal therapy of tumors. Here, a star molecule in organic photovoltaic materials, NCBDT-4C with an A-D-A conjugated structure, was assembled with DSPE-PEG-NH2 to prepare water dispersive nanoparticles (NPs). The prepared NCBDT-4Cl NPs exhibited a maximum NIR absorption peak at 764 nm and a maximum fluorescence peak at 798 nm. These NPs could generate superoxide anion, singlet oxygen (1O2), and heat under 808 nm laser irradiation. The 1O2 generation quantum yield and PCE of the NPs were 37.5% and 53.6%, respectively. The combination of photodynamic and photothermal therapy of cancer was demonstrated in vitro and in vivo. This work presents the advanced application of organic photovoltaic materials in cancer phototherapy.
Assuntos
Raios Infravermelhos , Imagem Óptica , Fotoquimioterapia , Terapia Fototérmica , Animais , Humanos , Camundongos , Nanopartículas/química , Nanomedicina Teranóstica , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Estrutura Molecular , Camundongos Endogâmicos BALB C , Neoplasias/diagnóstico por imagem , Neoplasias/terapia , Sobrevivência Celular/efeitos dos fármacos , Oxigênio Singlete/química , Oxigênio Singlete/metabolismoRESUMO
Clearing of toxic polyglutamine aggregates from neuronal cells is crucial for ameliorating Huntington's disease. However, such clearance is challenging, requiring the targeting of affected neuron cells in the brain, followed by the removal of polyglutamine from cells. Here we report a designed nanodrug that can be used for the ultrasound-based removal of toxic polyglutamine aggregates from neuron cells. The nanodrug is composed of a sonosensitizer molecule, chlorin e6- or protoporphyrin IX-loaded polymer micelle of 20-30 nm in size that rapidly delivers the sonosensitizer into the cell nucleus. Ultrasound exposure of these cells generates singlet oxygen in the nucleus/perinuclear region that induces strong autophagic flux and clears toxic polyglutamine aggregates from cells. It has been demonstrated that the nanodrug and ultrasound treatment can enhance the cell survival against polyglutamine aggregates by 4 times. This result suggests that the nanodrug can be designed for focused ultrasound-based wireless treatment of various neurodegenerative diseases.
Assuntos
Clorofilídeos , Neurônios , Peptídeos , Porfirinas , Protoporfirinas , Peptídeos/química , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Protoporfirinas/química , Porfirinas/química , Porfirinas/farmacologia , Animais , Humanos , Micelas , Sobrevivência Celular/efeitos dos fármacos , Oxigênio Singlete/metabolismo , Oxigênio Singlete/química , Ondas UltrassônicasRESUMO
Upconversion nanoparticles (UCNPs) have been used as a potential nanocarrier for photosensitizers (PSs), which have demonstrated a great deal of promise in achieving an effective photodynamic therapy (PDT) for deep-seated tumors. However, overcoming biological barriers to achieve mitochondria-targeted PDT remains a major challenge. Herein, CD44- and mitochondria-targeted photodynamic nanosystems were fabricated through the self-assembly of hyaluronic acid-conjugated-methoxy poly(ethylene glycol)-diethylenetriamine-grafted-(chlorin e6-dihydrolipoic acid-(3-carboxypropyl)triphenylphosphine bromide) polymeric ligands (HA-c-mPEG-Deta-g-(Ce6-DHLA-TPP)) and NaErF4:Tm@NaYF4 core-shell UCNPs (termed CMPNs). The CMPNs presented ideal physiological stability, a good drug loading capacity and an improved capacity for the generation of singlet oxygen (1O2) based on the FRET mechanism. Significantly, confocal images revealed that CMPNs not only facilitated cellular uptake through CD44-receptor-targeted endocytosis, subsequently enabling rapid evasion from endo-lysosomal sequestration, but also specifically targeted mitochondria, ultimately inducing a profound disruption of mitochondrial membrane potential, which triggered apoptosis upon laser irradiation, thereby significantly enhancing the therapeutic effect. Furthermore, in vitro antitumor experiments further confirmed the substantial enhancement in cancer cell killing efficiency achieved by treating with CMPNs upon near-infrared (NIR) laser irradiation. This innovative approach holds promise for the development of NIR-laser-activated photodynamic nanoagents specifically designed for mitochondria-targeted PDT, thus addressing the limitations of the current PDT treatments.
Assuntos
Receptores de Hialuronatos , Ácido Hialurônico , Mitocôndrias , Nanopartículas , Fotoquimioterapia , Fármacos Fotossensibilizantes , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Receptores de Hialuronatos/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Nanopartículas/química , Humanos , Ligantes , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Oxigênio Singlete/metabolismo , Oxigênio Singlete/química , Apoptose/efeitos dos fármacos , Polietilenoglicóis/química , Sobrevivência Celular/efeitos dos fármacos , Clorofilídeos , Linhagem Celular Tumoral , Porfirinas/química , Porfirinas/farmacologia , Fluoretos/química , ÍtrioRESUMO
Despite the promising advances in photodynamic therapy (PDT), it remains challenging to target and treat deep-seated solid tumors effectively. Herein, we developed an organoplatinum(II) complex (Pt-TPE) with self-assembly properties for sonodynamic therapy (SDT). Pt-TPE forms a nanofiber network structure through Pt-Pt and π-π stacking interactions. Notably, under ultrasound (US), Pt-TPE demonstrates unique self-assembly-induced singlet oxygen (1O2) generation due to a significantly enhanced singlet-triplet intersystem crossing (ISC). This generation of 1O2 occurs exclusively in the self-assembled state of Pt-TPE. Additionally, Pt-TPE exhibits sono-cytotoxicity against cancer cells by impairing mitochondrial membrane potential (MMP), inhibiting glucose uptake, and aerobic glycolysis. Furthermore, US-activated Pt-TPE significantly inhibits deep solid tumors in mice, achieving remarkable therapeutic efficacy even at penetration depths greater than 10 cm. This study highlights the potential of self-assembled metal complexes to enhance the efficacy of SDT for treating deep tumors.
Assuntos
Antineoplásicos , Animais , Humanos , Camundongos , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Antineoplásicos/síntese química , Compostos Organoplatínicos/farmacologia , Compostos Organoplatínicos/química , Compostos Organoplatínicos/uso terapêutico , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/uso terapêutico , Fármacos Fotossensibilizantes/síntese química , Terapia por Ultrassom/métodos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Linhagem Celular Tumoral , Oxigênio Singlete/metabolismo , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Camundongos Endogâmicos BALB C , Feminino , Camundongos NusRESUMO
The worldwide transmission of infectious respiratory pathogens has caused innumerable deaths and suffering, while wearing a face mask is still the most effective way to terminate the respiratory infections spread. However, the frequent mask replacement as a result of the lack of pathogen sterilization ability not only increases the cross-contamination risk but also, even worse, produces a large amount of medical waste. In this work, we report on a ketonized carbonitride functionalized bioprotective face mask with pathogen sterilization activity that can effectively produce biocidal singlet oxygen triggered by light irradiation. Ketonized carbonitride loading on the outer layer of the mask is found to be capable of generating singlet oxygen, enabling the mask with antibacterial ability. Thanks to its high pathogen inactivation activity, the as-prepared mask exhibits long-term light triggered health protection performance, which, in return, reduces medical waste production as a result of the decreased mask replacement frequency. The synthesis of a fascinating bioprotective mask provides a new viewpoint into the development of personal bioprotective devices for health protection.
Assuntos
Luz , Máscaras , Antibacterianos/química , Antibacterianos/farmacologia , Humanos , Oxigênio Singlete/química , Oxigênio Singlete/metabolismo , Escherichia coli/efeitos dos fármacosRESUMO
The development of new and improved mitochondria-targeting photosensitisers (PSs) for photodynamic therapy (PDT) remains highly desirable, due to the critical role the mitochondria play in maintaining healthy cellular function. Here, we report the design, synthesis, photophysical properties and biological characterisation of a series of di-iodinated BODIPY-based PSs, BODIPY-Mito-I-n, for mitochondria-targeted PDT applications. Six BODIPY-Mito-I-n analogues were synthesised in good yields, with fast reaction times of between 30 and 60 min under mild conditions. The di-iodination of the BODIPY scaffold enabled highly efficient population of the triplet state, leading to high singlet oxygen (1O2) photosensitisation efficiencies (ΦΔ = 0.55-0.65). All BODIPY-Mito-I-n compounds exhibited very high photocytotoxic activity towards HeLa cells, with IC50,light values of between 1.30 and 6.93 nM, due to photoinduced 1O2 generation. Notably, the poly(ethylene glycol) (PEG)-modified BODIPY-Mito-I-6 showed remarkably lower dark cytotoxicity (IC50,dark = 6.68-7.25 µM) than the non-PEGylated analogues BODIPY-Mito-I-1 to BODIPY-Mito-I-5 (IC50,dark = 0.58-1.09 µM), resulting in photocytotoxicity indices up to 2120. Mechanistic studies revealed that BODIPY-Mito-I-6 induced reactive oxygen species overproduction and mitochondrial dysfunction in cells upon irradiation, leading to significant cell death through a combination of apoptosis and necrosis. It is anticipated that our design will contribute to the development of more effective mitochondria-targeting PSs for cancer therapy.
Assuntos
Compostos de Boro , Mitocôndrias , Fotoquimioterapia , Fármacos Fotossensibilizantes , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/síntese química , Humanos , Compostos de Boro/química , Compostos de Boro/farmacologia , Compostos de Boro/síntese química , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Células HeLa , Sobrevivência Celular/efeitos dos fármacos , Oxigênio Singlete/metabolismo , Estrutura Molecular , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/síntese química , Espécies Reativas de Oxigênio/metabolismoRESUMO
Controlling the succession of chemical processes with high specificity in complex systems is advantageous for widespread applications, from biomedical research to drug manufacturing. Despite synthetic advances in bioorthogonal and photochemical methodologies, there is a need for generic chemical approaches that can universally modulate photodynamic reactivity in organic photosensitizers. Herein we present a strategy to fine-tune the production of singlet oxygen in multiple photosensitive scaffolds under the activation of bioresponsive and bioorthogonal stimuli. We demonstrate that the photocatalytic activity of nitrobenzoselenadiazoles can be fully blocked by site-selective incorporation of electron-withdrawing carbamate moieties and restored on demand upon uncaging with a wide range of molecular triggers, including abiotic transition-metal catalysts. We also prove that this strategy can be expanded to most photosensitizers, including diverse structures and spectral properties. Finally, we show that such advanced control of singlet oxygen generation can be broadly applied to the photodynamic ablation of human cells as well as to regulate the release of singlet oxygen in the semi-synthesis of natural product drugs.
Assuntos
Fármacos Fotossensibilizantes , Oxigênio Singlete , Oxigênio Singlete/química , Oxigênio Singlete/metabolismo , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Humanos , Catálise/efeitos da radiação , Fotoquimioterapia/métodos , Carbamatos/química , Células HeLaRESUMO
Photodynamic therapy (PDT) is a promising alternative treatment for localized lesions and infections, utilizing reactive oxygen species (ROS) generated by photosensitizers (PS) upon light activation. Singlet oxygen (1O2) is a key ROS responsible for photodynamic damage. However, the effectiveness of PS in biological systems may not correlate with the efficiency of singlet oxygen generation in homogeneous solutions. This study investigated singlet oxygen generation and its decay in various cellular microenvironments using liposome and ARPE-19 cell models. Rose Bengal (RB), methylene blue (MB), and protoporphyrin IX (PpIX) were employed as selected PS. Lifetimes of singlet oxygen generated by the selected photosensitizers in different cellular compartments varied, indicating different quenching rates with singlet oxygen. RB, located near cell membranes, exhibited the highest phototoxicity and lipid/protein peroxidation, followed by PpIX, while MB showed minimal cytotoxicity in similar conditions. Singlet oxygen decay lifetimes provide insights into PS localization and potential phototoxicity, highlighting the importance of the lipid microenvironment in PDT efficacy, providing useful screening method prior to in vivo applications.
Assuntos
Lipossomos , Azul de Metileno , Fotoquimioterapia , Fármacos Fotossensibilizantes , Protoporfirinas , Rosa Bengala , Oxigênio Singlete , Oxigênio Singlete/metabolismo , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/química , Humanos , Rosa Bengala/farmacologia , Rosa Bengala/química , Protoporfirinas/química , Protoporfirinas/farmacologia , Protoporfirinas/metabolismo , Lipossomos/química , Azul de Metileno/química , Azul de Metileno/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Linhagem Celular , LuzRESUMO
Porphyrazines (Pzs) are porphyrin derivatives that show potential application as photosensitizers for photodynamic therapy (PDT), but are still far less explored in the literature. In this work, we evaluate how the photophysics and phototoxicity of the octakis(trifluoromethylphenyl)porphyrazine (H2Pz) against tumor cells can be modulated by coordination with Mg(II), Zn(II), Cu(II) and Co(II) ions. Fluorescence and singlet oxygen quantum yields for the Pzs were measured in organic solvents and in soy phosphatidylcholine (PC) liposomes suspended in water. While H2Pz and the respective complexes with Cu(II) and Co(II) showed very low efficiency to fluoresce and to produce 1O2, the Mg(II) and Zn(II) complexes showed significantly higher quantum yields in organic solvents. The fluorescence of these two Pzs in the liposomes was sensitive to the fluidity of the membrane, showing potential use as viscosity markers. The cytotoxicity of the compounds was tested in HaCaT (normal) and A431 (tumor) cells using soy PC liposomes as drug carriers. Despite the low 1O2 quantum yields in water, the Mg(II) and Zn(II) complexes showed IC50 values against A431 cells in the nanomolar range when activated with low doses of red LED light. Their phototoxicity was ca. three times higher for the tumor cells compared to the normal ones, showing promising application as photosensitizers for PDT protocols. Considering that H2Pz and the respective Co(II) and Cu(II) complexes were practically non-phototoxic to the cells, we demonstrate the importance of the central metal ion in the modulation of the photodynamic activity of porphyrazines.
Assuntos
Lipossomos , Fármacos Fotossensibilizantes , Porfirinas , Humanos , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/química , Porfirinas/química , Porfirinas/farmacologia , Lipossomos/química , Fotoquimioterapia , Oxigênio Singlete/metabolismo , Oxigênio Singlete/química , Complexos de Coordenação/farmacologia , Complexos de Coordenação/química , Complexos de Coordenação/síntese química , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/síntese química , Zinco/química , Zinco/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Íons/químicaRESUMO
Two phthalocyanine derivatives tetra-peripherally substituted with tert-butylsulfonyl groups and coordinating either zinc(II) or platinum(II) ions have been synthesized and subsequently investigated in terms of their optical and photochemical properties, as well as biological activity in cellular, tissue-engineered, and animal models. Our research has revealed that both synthesized phthalocyanines are effective generators of reactive oxygen species (ROS). PtSO2tBu demonstrated an outstanding ability to generate singlet oxygen (ΦΔ = 0.87-0.99), while ZnSO2tBu in addition to 1O2 (ΦΔ = 0.45-0.48) generated efficiently other ROS, in particular ·OH. Considering future biomedical applications, the affinity of the tested phthalocyanines for biological membranes (partition coefficient; log Pow) and their primary interaction with serum albumin were also determined. To facilitate their biological administration, a water-dispersible formulation of these phthalocyanines was developed using Pluronic triblock copolymers to prevent self-aggregation and improve their delivery to cancer cells and tissues. The results showed a significant increase in cellular uptake and phototoxicity when phthalocyanines were incorporated into the customizable polymeric micelles. Moreover, the improved distribution in the body and photodynamic efficacy of the encapsulated phthalocyanines were investigated in hiPSC-delivered organoids and BALB/c mice bearing CT26 tumors. Both photosensitizers exhibit strong antitumor activity. Notably, vascular-targeted photodynamic therapy (V-PDT) led to complete tumor eradication in 84% of ZnSO2tBu and 100% of PtSO2tBu-treated mice, and no recurrence has so far been observed for up to five months after treatment. In the case of PtSO2tBu, the effect was significantly stronger, offering a wider range of light doses suitable for achieving effective PDT.
Assuntos
Indóis , Isoindóis , Fotoquimioterapia , Fármacos Fotossensibilizantes , Zinco , Animais , Indóis/química , Indóis/farmacologia , Humanos , Camundongos , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/síntese química , Zinco/química , Zinco/farmacologia , Platina/química , Platina/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Camundongos Endogâmicos BALB C , Linhagem Celular Tumoral , Oxigênio Singlete/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/síntese químicaRESUMO
Meta-tetra(hydroxyphenyl)chlorin (m-THPC) is among the most potent photosensitizers, known for its high singlet oxygen generation efficiency. However, its clinical effectiveness in photodynamic therapy (PDT) is compromised by its propensity to aggregate in aqueous solutions, adversely affecting its photophysical properties and therapeutic potential. A series of spectroscopic techniques, including UV-Vis absorption, fluorescence spectroscopy, and laser flash photolysis, revealed that m-THPC exhibits significant aggregation, particularly in MeOH-PBS mixtures with MeOH content below 30%. This aggregation adversely affects its photophysical properties leading to reduced fluorescence quantum yield and most importantly reducing its singlet oxygen quantum yield. This study introduces the use of bovine serum albumin (BSA) to counteract the aggregation of m-THPC, aiming to enhance its solubility, stability, and efficacy in physiological settings. Through advanced spectroscopic analyses we demonstrated that the m-THPC@BSA complex exhibits restored photophysical properties characteristic for monomeric form. Notably, the complex showed a significant restoration of the singlet oxygen quantum yield (ΦΔ = 0.21) compared to aggregated m-THPC. These results underscore the potential of BSA to preserve the monomeric form of m-THPC, mitigating aggregation-induced losses in singlet oxygen production. Our findings suggest that BSA-mediated delivery systems could play a crucial role in optimizing the clinical utility of hydrophobic photosensitizers like m-THPC.
Assuntos
Fármacos Fotossensibilizantes , Soroalbumina Bovina , Oxigênio Singlete , Espectrometria de Fluorescência , Soroalbumina Bovina/química , Fármacos Fotossensibilizantes/química , Oxigênio Singlete/química , Oxigênio Singlete/metabolismo , Animais , Bovinos , Porfirinas/química , Fotoquimioterapia/métodos , Análise EspectralRESUMO
Cefadroxil is a widely used antibiotic with a low elimination efficiency in wastewater treatments plants, so it represents a contaminants of emerging concern that should be removed. The photosensitization process that involves natural pigments and visible sunlight can be offered as an environmentally friendly alternative to be considered for Cefadroxil degradation. In this investigation, we carried out a mechanistic and kinetic approach to Cefadroxil photodegradation sensitized by Riboflavin and Humic Acid, in individual and combined processes. Our experiments indicate that Cefadroxil is able to interact with the excited states of Riboflavin as well as with the photogenerated reactive oxygen species, with an important contribution of singlet oxygen. The antibiotic was less sensitive to the photodegradation in the presence of Humic Acids and in the mixture of Riboflavin and Humic Acids. Self-sensitization processes and internal filter effects are proposed as possible explanations for the observed phenomenon. The reaction between Cefadroxil and singlet oxygen showed a dependence with the pH of the medium, the photodegradation kinetic constants are greater at alkaline pH compared to neutral pH. The reaction is favored when the anionic species of the antibiotic is present. Microbiological tests on S. aureus indicated that the antibiotic reduce its antimicrobial activity as a consequence of the photooxidative process mediated by singlet oxygen. We believe that the results are relevant since, the sensitized photodegradation process could lead to the oxidation of Cefadroxil and to the progressive loss of its antimicrobial function, this fact could contribute to the decrease in the generation of bacterial multi-resistance to antibiotics in the environment.
Assuntos
Antibacterianos , Cefadroxila , Fotólise , Antibacterianos/química , Antibacterianos/farmacologia , Cefadroxila/química , Cefadroxila/farmacologia , Oxigênio Singlete/química , Oxigênio Singlete/metabolismo , Substâncias Húmicas , Pigmentos Biológicos/química , Pigmentos Biológicos/farmacologia , Cinética , Riboflavina/química , Riboflavina/farmacologia , Concentração de Íons de HidrogênioRESUMO
Singlet oxygen is considered an important cell damaging agent due to its propensity to react with organic compounds. This drives the interest in developing methods for determination of 1O2. Simplicity of application and high sensitivity makes fluorescent probes a popular choice for in vivo 1O2 detection. Despite its proclaimed cell-impermeability, the commercially available Singlet Oxygen Sensor Green (SOSG) is widely applied to support assertions of 1O2 involvement in cell and tissue damage. Our investigation, however, demonstrate that different microbial species and cancer cells become fluorescent when exposed to SOSG under conditions which exclude generation of 1O2. Cells, permeabilized with chlorhexidine or by heat exposure under anaerobic conditions, exhibited SOSG fluorescence. Permeabilized cells could be stained with SOSG even 24 h post-permeabilization. Since SOSG is cell impermeable, the main factor that led to fluorescent staining was plasma membrane damage. Spectral analyses of different batches of SOSG revealed that SOSG endoperoxide (SOSG-EP) did not increase even after prolonged storage under the recommended conditions. The commercial preparations of SOSG, however, were not SOSG-EP free, which can produce erroneous results when SOSG staining is used as a proof of singlet oxygen production in vivo.
Assuntos
Corantes Fluorescentes , Oxigênio Singlete , Oxigênio Singlete/metabolismo , Humanos , Corantes Fluorescentes/química , Coloração e Rotulagem/métodos , Membrana Celular/metabolismoRESUMO
Rationale: One of the main challenges in chemotherapy is achieving high treatment efficacy while minimizing adverse events. Fully exploiting the therapeutic potential of an old drug and monitoring its effects in vivo is highly valuable, but often difficult to achieve. Methods: In this study, by encapsulating disulfiram (DSF) approved by US Food and Drug Administration, semiconducting polymer nanocomplex (MEHPPV), and Chlorin e6 into a polymeric matrix F127 via nanoprecipitation method, a nanosystem (FCMC) was developed for afterglow imaging guided cancer treatment. The characteristics, stability as well as the ability of singlet oxygen (1O2) production of FCMC were first carefully examined. Then, we studied the mechanism for enhanced anti-cancer efficiency and afterglow luminescence in vitro. For experiments in vivo, 4T1 subcutaneous xenograft tumor mice were injected with FCMC via the tail vein every three days and the antitumor effect of FCMC was evaluated by monitoring tumor volume and body weight every three day. Results: The nanosystem, which combines DSF with Ce6, can generates abundant 1O2 that enhances the antitumor activity of DSF. The in vivo results show that FCMC-treated group exhibits an obviously higher tumor-growth inhibition rate of 67.89% after 15 days of treatment, compared to the control group of F127@MEHPPV-CuET. Moreover, Ce6 also greatly enhances the afterglow luminescence intensity of MEHPPV and promotes the redshift of the afterglow emission towards the ideal near-infrared imaging window, thereby enabling efficient afterglow tumor imaging in vivo. Conclusions: This multifunctional nanoplatform not only improves the anticancer efficacy of DSF, but also enables monitoring tumor via robust afterglow imaging, thus exhibiting great potential for cancer therapy and early therapeutic outcome prediction.
Assuntos
Clorofilídeos , Dissulfiram , Polímeros , Porfirinas , Animais , Dissulfiram/farmacologia , Dissulfiram/química , Porfirinas/farmacologia , Porfirinas/química , Camundongos , Polímeros/química , Linhagem Celular Tumoral , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos/administração & dosagem , Feminino , Camundongos Endogâmicos BALB C , Oxigênio Singlete/metabolismo , Nanopartículas/química , Semicondutores , Humanos , Ensaios Antitumorais Modelo de Xenoenxerto , Imagem Óptica/métodosRESUMO
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.
Assuntos
Peptídeos beta-Amiloides , Oxirredução , Fragmentos de Peptídeos , Fármacos Fotossensibilizantes , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/química , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fotoquimioterapia , Oxigênio Singlete/metabolismo , Oxigênio Singlete/química , Humanos , Animais , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Luz , Peróxido de Hidrogênio/química , Agregados Proteicos/efeitos dos fármacos , CamundongosRESUMO
The rise of antimicrobial resistance poses a critical public health threat worldwide. While antimicrobial photodynamic therapy (aPDT) has demonstrated efficacy against multidrug-resistant (MDR) bacteria, its effectiveness can be limited by several factors, including the delivery of the photosensitizer (PS) to the site of interest and the development of bacterial resistance to PS uptake. There is a need for alternative methods, one of which is superhydrophobic antimicrobial photodynamic therapy (SH-aPDT), which we report here. SH-aPDT is a technique that isolates the PS on a superhydrophobic (SH) membrane, generating airborne singlet oxygen (1O2) that can diffuse up to 1 mm away from the membrane. In this study, we developed a SH polydimethylsiloxane dressing coated with PS verteporfin. These dressings contain air channels called a plastron for supplying oxygen for aPDT and are designed so that there is no direct contact of the PS with the tissue. Our investigation focuses on the efficacy of SH-aPDT on biofilms formed by drug-sensitive and MDR strains of Gram-positive (Staphylococcus aureus and S. aureus methicillin-resistant) and Gram-negative bacteria (Pseudomonas aeruginosa and P. aeruginosa carbapenem-resistant). SH-aPDT reduces bacterial biofilms by approximately 3 log with a concomitant decrease in their metabolism as measured by MTT. Additionally, the treatment disrupted extracellular polymeric substances, leading to a decrease in biomass and biofilm thickness. This innovative SH-aPDT approach holds great potential for combating antimicrobial resistance, offering an effective strategy to address the challenges posed by drug-resistant wound infections.