Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Photochem Photobiol B ; 234: 112500, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35816857

RESUMO

BACKGROUND AND AIM: A photosensitizer (PS) delivery and comprehensive tumor targeting platform was developed that is centered on the photosensitization of key pharmacological targets in solid tumors (cancer cells, tumor vascular endothelium, and cellular and non-cellular components of the tumor microenvironment) before photodynamic therapy (PDT). Interstitially targeted liposomes (ITLs) encapsulating zinc phthalocyanine (ZnPC) and aluminum phthalocyanine (AlPC) were formulated for passive targeting of the tumor microenvironment. In previous work it was established that the PEGylated ITLs were taken up by cultured cholangiocarcinoma cells. The aim of this study was to verify previous results in cancer cells and to determine whether the ITLs can also be used to photosensitize cells in the tumor microenvironment and vasculature. Following positive results, rudimentary in vitro and in vivo experiments were performed with ZnPC-ITLs and AlPC-ITLs as well as their water-soluble tetrasulfonated derivatives (ZnPCS4 and AlPCS4) to assemble a research dossier and bring this platform closer to clinical transition. METHODS: Flow cytometry and confocal microscopy were employed to determine ITL uptake and PS distribution in cholangiocarcinoma (SK-ChA-1) cells, endothelial cells (HUVECs), fibroblasts (NIH-3T3), and macrophages (RAW 264.7). Uptake of ITLs by endothelial cells was verified under flow conditions in a flow chamber. Dark toxicity and PDT efficacy were determined by cell viability assays, while the mode of cell death and cell cycle arrest were assayed by flow cytometry. In vivo systemic toxicity was assessed in zebrafish and chicken embryos, whereas skin phototoxicity was determined in BALB/c nude mice. A PDT efficacy pilot was conducted in BALB/c nude mice bearing human triple-negative breast cancer (MDA-MB-231) xenografts. RESULTS: The key findings were that (1) photodynamically active PSs (i.e., all except ZnPCS4) were able to effectively photosensitize cancer cells and non-cancerous cells; (2) following PDT, photodynamically active PSs were highly toxic-to-potent as per anti-cancer compound classification; (3) the photodynamically active PSs did not elicit notable systemic toxicity in zebrafish and chicken embryos; (4) ITL-delivered ZnPC and ZnPCS4 were associated with skin phototoxicity, while the aluminum-containing PSs did not exert detectable skin phototoxicity; and (5) ITL-delivered ZnPC and AlPC were equally effective in their tumor-killing capacity in human tumor breast cancer xenografts and superior to other non-phthalocyanine PSs when appraised on a per mole administered dose basis. CONCLUSIONS: AlPC(S4) are the safest and most effective PSs to integrate into the comprehensive tumor targeting and PS delivery platform. Pending further in vivo validation, these third-generation PSs may be used for multi-compartmental tumor photosensitization.


Assuntos
Colangiocarcinoma , Compostos Organometálicos , Fotoquimioterapia , Animais , Linhagem Celular Tumoral , Embrião de Galinha , Células Endoteliais , Humanos , Lipossomos , Camundongos , Camundongos Nus , Compostos Organometálicos/farmacologia , Compostos Organometálicos/uso terapêutico , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Microambiente Tumoral , Peixe-Zebra
2.
Methods Mol Biol ; 2451: 405-480, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35505025

RESUMO

Photodynamic therapy (PDT) is a minimally to noninvasive treatment modality that has emerged as a promising alternative to conventional cancer treatments. PDT induces hyperoxidative stress and disrupts cellular homeostasis in photosensitized cancer cells, resulting in cell death and ultimately removal of the tumor. However, various survival pathways can be activated in sublethally afflicted cancer cells following PDT. The acute stress response is one of the known survival pathways in PDT, which is activated by reactive oxygen species and signals via ASK-1 (directly) or via TNFR (indirectly). The acute stress response can activate various other survival pathways that may entail antioxidant, pro-inflammatory, angiogenic, and proteotoxic stress responses that culminate in the cancer cell's ability to cope with redox stress and oxidative damage. This review provides an overview of the immediate early stress response in the context of PDT, mechanisms of activation by PDT, and molecular intervention strategies aimed at inhibiting survival signaling and improving PDT outcome.


Assuntos
Neoplasias , Fotoquimioterapia , Morte Celular , Humanos , Neoplasias/patologia , Fotoquimioterapia/métodos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
3.
Methods Mol Biol ; 2451: 285-403, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35505024

RESUMO

Photodynamic therapy (PDT) is a non-to-minimally invasive treatment modality that utilizes photoactivatable drugs called photosensitizers to disrupt tumors with locally photoproduced reactive oxygen species (ROS). Photosensitizer activation by light results in hyperoxidative stress and subsequent tumor cell death, vascular shutdown and hypoxia, and an antitumor immune response. However, sublethally afflicted tumor cells initiate several survival mechanisms that account for decreased PDT efficacy. The hypoxia inducible factor 1 (HIF-1) pathway is one of the most effective cell survival pathways that contributes to cell recovery from PDT-induced damage. Several hundred target genes of the HIF-1 heterodimeric complex collectively mediate processes that are involved in tumor cell survival directly and indirectly (e.g., vascularization, glucose metabolism, proliferation, and metastasis). The broad spectrum of biological ramifications culminating from the activation of HIF-1 target genes reflects the importance of HIF-1 in the context of therapeutic recalcitrance. This chapter elaborates on the involvement of HIF-1 in cancer biology, the hypoxic response mechanisms, and the role of HIF-1 in PDT. An overview of inhibitors that either directly or indirectly impede HIF-1-mediated survival signaling is provided. The inhibitors may be used as pharmacological adjuvants in combination with PDT to augment therapeutic efficacy.


Assuntos
Neoplasias , Fotoquimioterapia , Sobrevivência Celular , Humanos , Fator 1 Induzível por Hipóxia/genética , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo
4.
Mater Sci Eng C Mater Biol Appl ; 123: 111984, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33812612

RESUMO

We have prepared and characterized a cholesterol-rich nanoemulsion called LDE, a mimic of classic lipoprotein macromolecules, that can be applied as a new drug delivery system for aluminum phthalocyanine chloride (PcAlCl). The LDE containing PcAlCl system prepared herein had mean size and zeta potential of 127 nm and -29 mV, respectively, and encapsulation rate efficiency was 81%, and stability of 17 months. Compared to classical liposomes, LDE was more efficient, especially in brain diseases like glioblastoma (GBM), as revealed by tests on the U-87 MG cell line. The LDEPc formulation did not display dark cytotoxicity, as expected. The best light dose for LDEPc was 1.0 J·cm-2: its activity was 55% higher than PcAlCl in a compatible organic medium. In the U-87 MG cells, apoptosis was the preferential pathway activated by PDT. These results strongly support the use of LDE as a new theranostic system.


Assuntos
Glioblastoma , Colesterol , Sistemas de Liberação de Medicamentos , Emulsões , Glioblastoma/tratamento farmacológico , Humanos
5.
J Photochem Photobiol B ; 216: 112146, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33601256

RESUMO

BACKGROUND AND AIM: Oncological photodynamic therapy (PDT) relies on photosensitizers (PSs) to photo-oxidatively destroy tumor cells. Currently approved PSs yield satisfactory results in superficial and easy-to-access tumors but are less suited for solid cancers in internal organs such as the biliary system and the pancreas. For these malignancies, second-generation PSs such as metallated phthalocyanines are more appropriate. Presently it is not known which of the commonly employed metallated phtahlocyanines, namely aluminum phthalocyanine (AlPC) and zinc phthalocyanine (ZnPC) as well as their tetrasulfonated derivatives AlPCS4 and ZnPCS4, is most cytotoxic to tumor cells. This study therefore employed an attritional approach to ascertain the best metallated phthalocyanine for oncological PDT in a head-to-head comparative analysis and standardized experimental design. METHODS: ZnPC and AlPC were encapsulated in PEGylated liposomes. Analyses were performed in cultured A431 cells as a template for tumor cells with a dysfunctional P53 tumor suppressor gene and EGFR overexpression. First, dark toxicity was assessed as a function of PS concentration using the WST-1 and sulforhodamine B assay. Second, time-dependent uptake and intracellular distribution were determined by flow cytometry and confocal microscopy, respectively, using the intrinsic fluorescence of the PSs. Third, the LC50 values were established for each PS at 671 nm and a radiant exposure of 15 J/cm2 following 1-h PS exposure. Finally, the mode of cell death as a function of post-PDT time and cell cycle arrest at 24 h after PDT were analyzed. RESULTS: In the absence of illumination, AlPC and ZnPC were not toxic to cells up to a 1.5-µM PS concentration and exposure for up to 72 h. Dark toxicity was noted for AlPCS4 at 5 µM and ZnPCS4 at 2.5 µM. Uptake of all PSs was observed as early as 1 min after PS addition to cells and increased in amplitude during a 2-h incubation period. After 60 min, the entire non-nuclear space of the cell was photosensitized, with PS accumulation in multiple subcellular structures, especially in case of AlPC and AlPCS4. PDT of cells photosensitized with ZnPC, AlPC, and AlPCS4 yielded LC50 values of 0.13 µM, 0.04 µM, and 0.81 µM, respectively, 24 h post-PDT (based on sulforhodamine B assay). ZnPCS4 did not induce notable phototoxicity, which was echoed in the mode of cell death and cell cycle arrest data. At 4 h post-PDT, the mode of cell death comprised mainly apoptosis for ZnPC and AlPC, the extent of which was gradually exacerbated in AlPC-photosensitized cells during 8 h. ZnPC-treated cells seemed to recover at 8 h post-PDT compared to 4 h post-PDT, which had been observed before in another cell line. AlPCS4 induced considerable necrosis in addition to apoptosis, whereby most of the cell death had already manifested at 2 h after PDT. During the course of 8 h, necrotic cell death transitioned into mainly late apoptotic cell death. Cell death signaling coincided with a reduction in cells in the G0/G1 phase (ZnPC, AlPC, AlPCS4) and cell cycle arrest in the S-phase (ZnPC, AlPC, AlPCS4) and G2 phase (ZnPC and AlPC). Cell cycle arrest was most profound in cells that had been photosensitized with AlPC and subjected to PDT. CONCLUSIONS: Liposomal AlPC is the most potent PS for oncological PDT, whereas ZnPCS4 was photodynamically inert in A431 cells. AlPC did not induce dark toxicity at PS concentrations of up to 1.5 µM, i.e., > 37 times the LC50 value, which is favorable in terms of clinical phototoxicity issues. AlPC photosensitized multiple intracellular loci, which was associated with extensive, irreversible cell death signaling that is expected to benefit treatment efficacy and possibly immunological long-term tumor control, granted that sufficient AlPC will reach the tumor in vivo. Given the differential pharmacokinetics, intracellular distribution, and cell death dynamics, liposomal AlPC may be combined with AlPCS4 in a PS cocktail to further improve PDT efficacy.


Assuntos
Antineoplásicos/química , Portadores de Fármacos/química , Indóis/química , Lipossomos/química , Fármacos Fotossensibilizantes/química , Antineoplásicos/farmacologia , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Relação Dose-Resposta à Radiação , Liberação Controlada de Fármacos , Humanos , Indóis/farmacologia , Isoindóis , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Relação Estrutura-Atividade , Fatores de Tempo
6.
Artigo em Inglês | MEDLINE | ID: mdl-26774669

RESUMO

Nanoparticles (NPs) have been used in a range of products due to their unique properties. Nevertheless, these NPs can cause adverse biological effects and because of that, there is a great concern about the health and environmental risks related to their use. Recently, silver nanoparticles (Ag NPs) have been used in a variety of cytotoxicity and genotoxicity studies, but there are still controversies regarding the association between the size and the toxicity of these particles. Therefore, in this study, we aimed to evaluate the cytotoxicity and genotoxicity of Ag NPs (10 and 100 nm) in two different cell lines, CHO-K1 and CHO-XRS5, by performing cell viability assay (XTT), clonogenic assay, micronucleus test, comet assay, as well as by investigating the cell cycle kinetics using the flow cytometry. Cell cultures were exposed to different concentrations of AgNPs (0.025-5.0 µg/ml) for 24 h. Our results indicated that cytotoxicity and genotoxicity induced by the 100 nm-Ag NPs were greater than those induced by the 10 nm-Ag NPs for both cell lines, which suggests that the exposure to greater size particles (100 nm) can cause more adverse biological effects than the exposure to the smaller ones (10 nm).


Assuntos
Dano ao DNA/efeitos dos fármacos , Nanopartículas Metálicas/toxicidade , Prata/toxicidade , Animais , Células CHO , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaio Cometa , Cricetulus , Nanopartículas Metálicas/química , Testes para Micronúcleos , Testes de Mutagenicidade/métodos , Tamanho da Partícula , Prata/química
7.
Carbohydr Polym ; 136: 892-8, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26572426

RESUMO

Regenerated cellulose scaffolds (RCS) may be used as alloplastic materials for tissue repair. In this work, the RCS were obtained by viscose process and characterized by scanning electron microscopy (SEM), wide angle X-ray diffraction (WAXD), Fourier transform infrared spectroscopy (FTIR) and thermogravimetry analysis (TG). In vitro enzymatic degradation assay and toxicological assays were also evaluated. The physicochemical characterizations revealed the formation of a porous material with distinct thermal profile and crystallinity compared to pristine cellulose pulp. Enzymatic degradation assay revealed that lysozyme showed a mildest catalytic action when compared to cellulase, Tricoderma reesei (Tr). Nevertheless, both enzymes were efficient for degrading the RCS. RCS did not show cytotoxicity, mutagenic or genotoxic effects. The systematically characterization of this work suggests that RCS presented distinct features that make it a viable material for future studies related to the development of scaffolds for biological applications.


Assuntos
Celulose/análogos & derivados , Alicerces Teciduais/efeitos adversos , Animais , Células CHO , Linhagem Celular Tumoral , Sobrevivência Celular , Celulase/química , Celulose/química , Celulose/toxicidade , Cricetinae , Cricetulus , Dano ao DNA , Muramidase/química , Ratos , Alicerces Teciduais/química
8.
Toxicol In Vitro ; 29(7): 1319-31, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26028148

RESUMO

Metallic nanoparticles such as silver (Ag), cerium dioxide (CeO2) and titanium dioxide (TiO2) are produced at a large scale and included in many consumer products. It is well known that most metallic NPs are toxic to humans which raise concerns about these engineered particles. Various studies have already been published on the subject, however, almost all of these studies have been conducted in cancer or transformed cell lines. In this work we performed a comparative evaluation of these metallic NPs on normal untransformed human fibroblasts (GM07492) detecting cyto- and geno-toxic responses after exposure to these NPs. Our results showed that all three metallic NPs were able to cross the plasma membrane and were mainly found in endocytic vesicles. The Ag and TiO2 NPs affected mitochondrial enzymatic activity (XTT), increased DNA fragmentation, oxidative damage (Comet assay) and induced cell death mainly by the apoptotic pathway. Ag NPs increased GADD45α transcript levels and the phosphorylation of proteins γH2AX. Transient genotoxicity was also observed from exposure to CeO2 NPs while TiO2 NPs showed no increase in DNA damage at sub-cytotoxic concentrations. In comparison, Ag NPs were found to be the most cyto-genotoxic NPs to fibroblasts. Thus, these results support the use of normal fibroblast as a more informative tool to detect the mechanisms of action induced by metallic NPs.


Assuntos
Cério/toxicidade , Fibroblastos/efeitos dos fármacos , Nanopartículas Metálicas/toxicidade , Prata/toxicidade , Titânio/toxicidade , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Morte Celular/efeitos dos fármacos , Linhagem Celular , Ensaio Cometa , Dano ao DNA , Fibroblastos/metabolismo , Histonas/metabolismo , Humanos , Proteínas Nucleares/genética
9.
An Acad Bras Cienc ; 85(2): 585-94, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23828338

RESUMO

Noni, a Hawaiian name for the fruit of Morinda citrifolia L., is a traditional medicinal plant from Polynesia widely used for the treatment of many diseases including arthritis, diabetes, asthma, hypertension and cancer. Here, a commercial noni juice (TNJ) was evaluated for its protective activities against the lesions induced by mitomycin C (MMC) and doxorrubicin (DXR) using the Somatic Mutation and Recombination Test (SMART) in Drosophila melanogaster. Three-day-old larvae, trans-heterozygous for two genetic markers (mwh and flr3 ), were co-treated with TNJ plus MMC or DXR. We have observed a reduction in genotoxic effects of MMC and DXR caused by the juice. TNJ provoked a marked decrease in all kinds of MMC- and DXR-induced mutant spots, mainly due to its antirecombinagenic activity. The TNJ protective effects were concentration-dependent, indicating a dose-response correlation, that can be attributed to a powerful antioxidant and/or free radical scavenger ability of TNJ.


Assuntos
Antimutagênicos/farmacologia , Bebidas , Frutas/química , Morinda/química , Testes de Mutagenicidade/métodos , Animais , Dano ao DNA/efeitos dos fármacos , Drosophila melanogaster/citologia , Drosophila melanogaster/efeitos dos fármacos , Mitomicina/farmacologia , Teofilina/análogos & derivados , Teofilina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...