Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 221
Filtrar
1.
Photochem Photobiol ; 97(5): 1101-1103, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33884636

RESUMO

This report describes studies involving ER vs. lysosomal targeting and is designed to assess the initiation of different death pathways as a function of subcellular targeting and PDT dose. Photodamage directed at mitochondria or lysosomes initiates apoptosis, a death pathway generally considered to be irreversible. Photodamage that involves the ER can lead to another death pathway termed paraptosis. This does not involve caspase activation, can eradicate cell types with impaired apoptosis; at high levels of irradiation, apoptosis and necrosis were observed. Autophagy has a cytoprotective function unless lysosomes are targeted; loss of lysosomal integrity can interfere with the autophagic recycling processes.


Assuntos
Fotoquimioterapia , Apoptose , Autofagia , Linhagem Celular Tumoral , Lisossomos/efeitos da radiação , Fármacos Fotossensibilizantes/metabolismo , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico
2.
Chem Commun (Camb) ; 57(14): 1790-1793, 2021 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-33475648

RESUMO

A Ru(ii)-BODIPY conjugate has been rationally designed and exhibits an intense absorption in the NIR region to boost lysosome-targeted PDT in vitro and in vivo. The advantages of Ru(ii) and BODIPY were successfully instilled into the conjugate to yield highly effective PDT efficacy against malignant melanoma A375 cells (PI = 3448) and A375 mice xenografts.


Assuntos
Compostos de Boro/química , Lisossomos/efeitos da radiação , Fotoquimioterapia , Compostos de Rutênio/química , Animais , Linhagem Celular Tumoral , Humanos , Raios Infravermelhos , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/terapia , Fármacos Fotossensibilizantes , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Cell Death Dis ; 11(12): 1070, 2020 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-33318476

RESUMO

Lack of effective treatments for aggressive breast cancer is still a major global health problem. We have previously reported that photodynamic therapy using methylene blue as photosensitizer (MB-PDT) massively kills metastatic human breast cancer, marginally affecting healthy cells. In this study, we aimed to unveil the molecular mechanisms behind MB-PDT effectiveness and specificity towards tumor cells. Through lipidomics and biochemical approaches, we demonstrated that MB-PDT efficiency and specificity rely on polyunsaturated fatty acid-enriched membranes and on the better capacity to deal with photo-oxidative damage displayed by non-tumorigenic cells. We found out that, in tumorigenic cells, lysosome membrane permeabilization is accompanied by ferroptosis and/or necroptosis. Our results also pointed at a cross-talk between lysosome-dependent cell death (LDCD) and necroptosis induction after photo-oxidation, and contributed to broaden the understanding of MB-PDT-induced mechanisms and specificity in breast cancer cells. Therefore, we demonstrated that efficient approaches could be designed on the basis of lipid composition and metabolic features for hard-to-treat cancers. The results further reinforce MB-PDT as a therapeutic strategy for highly aggressive human breast cancer cells.


Assuntos
Neoplasias da Mama/patologia , Luz , Antioxidantes/farmacologia , Neoplasias da Mama/tratamento farmacológico , Carcinogênese/efeitos dos fármacos , Carcinogênese/patologia , Carcinogênese/efeitos da radiação , Morte Celular/efeitos dos fármacos , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Feminino , Ferroptose/efeitos dos fármacos , Ferroptose/efeitos da radiação , Humanos , Lipídeos/química , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Lisossomos/efeitos da radiação , Azul de Metileno/farmacologia , Azul de Metileno/uso terapêutico , Modelos Biológicos , Necroptose/efeitos dos fármacos , Necroptose/efeitos da radiação , Oxirredução , Fotoquimioterapia , Neoplasias de Mama Triplo Negativas/patologia
4.
Sci Rep ; 10(1): 6881, 2020 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-32327691

RESUMO

Triphenylamines (TPAs) were previously shown to trigger cell death under prolonged one- or two-photon illumination. Their initial subcellular localization, before prolonged illumination, is exclusively cytoplasmic and they translocate to the nucleus upon photoactivation. However, depending on their structure, they display significant differences in terms of precise initial localization and subsequent photoinduced cell death mechanism. Here, we investigated the structural features of TPAs that influence cell death by studying a series of molecules differing by the number and chemical nature of vinyl branches. All compounds triggered cell death upon one-photon excitation, however to different extents, the nature of the electron acceptor group being determinant for the overall cell death efficiency. Photobleaching susceptibility was also an important parameter for discriminating efficient/inefficient compounds in two-photon experiments. Furthermore, the number of branches, but not their chemical nature, was crucial for determining the cellular uptake mechanism of TPAs and their intracellular fate. The uptake of all TPAs is an active endocytic process but two- and three-branch compounds are taken up via distinct endocytosis pathways, clathrin-dependent or -independent (predominantly caveolae-dependent), respectively. Two-branch TPAs preferentially target mitochondria and photoinduce both apoptosis and a proper necrotic process, whereas three-branch TPAs preferentially target late endosomes and photoinduce apoptosis only.


Assuntos
Aminas/toxicidade , Endocitose/efeitos dos fármacos , Endocitose/efeitos da radiação , Espaço Intracelular/metabolismo , Luz , Aminas/química , Morte Celular/efeitos dos fármacos , Morte Celular/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Células HeLa , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Lisossomos/efeitos da radiação , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Espécies Reativas de Oxigênio/metabolismo , Espectrometria de Fluorescência
5.
Biochem Biophys Res Commun ; 526(2): 479-484, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32234235

RESUMO

Exposure to blue light from light-emitting diodes (LEDs) is a source of damage for human eyes in today's modern life. Although it is well known that blue light can cause cellular damage and death, the molecular mechanism underlying this is still not fully understood. Here, we demonstrated that exposure to blue LED light increased lysosome levels and perinuclear cluster formation in 661W murine photoreceptor-derived cells. Irradiation with blue LED light promoted the nuclear transport of transcription factor EB (TFEB) and a subsequent increase in lysosomal-related gene expression. Moreover, blue LED light induced morphological changes in lysosomal structure and lysosomal membrane permeabilization (LMP). These effects were suppressed by an antioxidant, N-acetylcysteine (NAC). Finally, a calcium ion chelator, BAPTA-AM, attenuated blue LED light-induced lysosomal biogenesis and cell death. Taken together, these findings suggest that oxidative stress under blue LED light increases lysosome levels via the TFEB pathway in a calcium-dependent manner, resulting in the accumulation of damaged lysosomes and subsequently lysosomal cell death. Our results imply that lysosomal homeostasis plays a key role in the maintenance of eye function and the progression of retinal diseases.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Morte Celular/efeitos da radiação , Luz/efeitos adversos , Lisossomos/efeitos da radiação , Células Fotorreceptoras de Vertebrados/efeitos da radiação , Transporte Ativo do Núcleo Celular/efeitos da radiação , Animais , Linhagem Celular , Lisossomos/metabolismo , Camundongos , Estresse Oxidativo/efeitos da radiação , Células Fotorreceptoras de Vertebrados/metabolismo
6.
Sci Rep ; 10(1): 2015, 2020 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-32029749

RESUMO

Although metallothionein-3 (MT3), a brain-enriched form of metallothioneins, has been linked to Alzheimer's disease, little is known regarding the role of MT3 in glioma. As MT3 plays a role in autophagy in astrocytes, here, we investigated its role in irradiated glioma cells. Irradiation increased autophagy flux in GL261 glioma cells as evidenced by increased levels of LC3-II but decreased levels of p62 (SQSTM1). Indicating that autophagy plays a cytoprotective role in glioma cell survival following irradiation, measures inhibiting autophagy flux at various steps decreased their clonogenic survival of irradiated GL261 as well as SF295 and U251 glioma cells. Knockdown of MT3 with siRNA in irradiated glioma cells induced arrested autophagy, and decreased cell survival. At the same time, the accumulation of labile zinc in lysosomes was markedly attenuated by MT3 knockdown. Indicating that such zinc accumulation was important in autophagy flux, chelation of zinc with tetrakis-(2-pyridylmethyl)ethylenediamine (TPEN), induced arrested autophagy in and reduced survival of GL261 cells following irradiation. Suggesting a possible mechanism for arrested autophagy, MT3 knockdown and zinc chelation were found to impair lysosomal acidification. Since autophagy flux plays a cytoprotective role in irradiated glioma cells, present results suggest that MT3 and zinc may be regarded as possible therapeutic targets to sensitize glioma cells to ionizing radiation therapy.


Assuntos
Autofagia/efeitos da radiação , Neoplasias Encefálicas/radioterapia , Glioma/radioterapia , Metalotioneína/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fótons/uso terapêutico , Animais , Autofagia/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Quelantes/farmacologia , Etilenodiaminas/farmacologia , Técnicas de Silenciamento de Genes , Glioma/patologia , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Lisossomos/efeitos da radiação , Metalotioneína/genética , Metalotioneína 3 , Camundongos , Proteínas do Tecido Nervoso/genética , RNA Interferente Pequeno/metabolismo , Tolerância a Radiação , Zinco/metabolismo
7.
ACS Appl Mater Interfaces ; 12(2): 1988-1996, 2020 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-31771326

RESUMO

Photodynamic therapy (PDT) takes advantage of reactive oxygen species (ROS) to trigger the apoptosis for cancer therapy. Given that cell apoptosis is a form of programmed cell death involved with multiple suborganelles and cancer cells are more sensitive to ROS than normal cells, early confirmation of the apoptosis induced by ROS would effectively avoid overtreatment. Herein, we highlight an aggregation-induced emission (AIE)-based theranostic agent (TPA3) to in situ dynamically track mitophagy prior to late apoptosis. TPA3 showed high specificity to autophagy vacuoles (AVs), of which appearance is the signature event of mitophagy during early apoptosis and delivered photocytotoxicity to cancer cells and skin cancer tumors in nude mice under irradiation of white light. Furthermore, in situ monitoring of the dynamical mitophagy process involved with mitochondria, AVs, and lysosomes was performed for the first time under confocal microscopy, providing a real-time self-monitoring system for assessing the curative effect prior to late apoptosis. This fluorescence imaging guided PDT witness great advances for applying in the clinical application.


Assuntos
Apoptose , Mitofagia , Fotoquimioterapia , Nanomedicina Teranóstica , Animais , Apoptose/efeitos da radiação , Autofagossomos/metabolismo , Autofagossomos/efeitos da radiação , Fluorescência , Células HeLa , Humanos , Lisossomos/metabolismo , Lisossomos/efeitos da radiação , Fusão de Membrana/efeitos da radiação , Camundongos Nus , Mitofagia/efeitos da radiação , Imagem Óptica , Espécies Reativas de Oxigênio/metabolismo , Tela Subcutânea/patologia
8.
Biochem Biophys Res Commun ; 518(4): 611-618, 2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31445710

RESUMO

Autophagy has been associated with a variety of diseases especially aging. Human dermal fibroblasts (HDFs) can internalize and then degrade elastin, collagen and advanced glycation end products (AGEs) in lysosomes, which plays prominent roles in extracellular matrix homeostasis and AGEs removal in the dermis. Although autophagy has been reported to be decreased in photoaged fibroblasts, the underlying mechanism and its relevance to photoaging remain elusive. Here, we showed that GFP-LC3 puncta per cell, LC3Ⅰ/Ⅱ conversion and p62 expression were significantly increased, whereas beclin1 expression was not altered in UVA-induced photoaged fibroblasts compared with non-photoaged control. Moreover, autophagic flux was not significantly affected by chloroquine treatment, but was remarkably induced by rapamycin treatment in photoaged fibroblasts, suggesting that UVA-induced photoaging might inhibit autophagy at the degradation stage. Further lysosomal function studies demonstrated that degradation of formed autophagosomes, LC3Ⅱprotein and DQ-Green BSA was all dramatically decreased in photoaged fibroblasts. LysoSensor yellow/blue DND 160 staining and flow cytometry assays demonstrated that photoaging obviously attenuated lysosomal acidification. Also, decreased expression of cathepsin B, L and D was found in photoaged fibroblasts. These data suggest that lowered lysosomal acidity and decreased cathepsins expression might contribute to the inhibition of autophagic degradation, which might be crucial in the development of photoaging through impairing intracellular degradation.


Assuntos
Autofagia/efeitos da radiação , Fibroblastos/efeitos da radiação , Lisossomos/efeitos da radiação , Envelhecimento da Pele/efeitos da radiação , Raios Ultravioleta/efeitos adversos , Autofagossomos/metabolismo , Autofagossomos/efeitos da radiação , Células Cultivadas , Criança , Pré-Escolar , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Lisossomos/metabolismo , Pele/citologia , Pele/metabolismo , Pele/efeitos da radiação
9.
Sci Total Environ ; 662: 990-1002, 2019 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-30795485

RESUMO

Tritium entering the aquatic environment can confer a whole body internal radiological dose to aquatic organisms. Multiple stressors inherent in natural environments, however, confound estimates for observable radiation specific responses. To disentangle differences between field and laboratory outcomes to tritium exposures, a multivariate analysis comparing biomarkers for radiation exposure at the cellular level with changes in biological processes within tissues is described for fathead minnows (Pimephales promelas). Over tritium activity concentrations up to 180,000 Bq/L, DNA damage in the field were lower than DNA damage in the laboratory. This finding does not support an increase in morbidity of biota in field exposures. Energy deposited by tritium decay produces oxidised free radicals, yet the biological responses in brain, muscle and liver to oxidative stress differed between the studies and were not related to the tritium. For both studies, DNA damage in gonad and blood increased with increased tritium as did the fluorescence associated with lysosomal function in spleen. The studies differed in spleen phagocytosis activity were, in the laboratory but not the field, activity increased with increased tritium-and was correlatd with lysosomal function (Spearman coefficient of 0.98 (p = 0.001). The higher phagocytosis activity in the field reflects exposures to unmeasured factors that were not present within the laboratory. In the laboratory, DNA damage and lysosomal function were correlated: Spearman coefficients of 0.9 (Comet, p = 0.03) and 0.9 (micronuclei, p = 0.08). In the field, DNA damage by the Comet assay, but not by micronucleus frequency, correlated with lysosomal function: Spearman coefficients of 0.91 (Comet, p < 0.001) and 0.47 (micronuclei, p = 0.21). These observations highlight a need for better physiologic understanding of linkages between radiation-induced damage within cells and responses at higher levels of biological organization.


Assuntos
Cyprinidae/fisiologia , Dano ao DNA , Lisossomos/efeitos da radiação , Fagocitose/efeitos da radiação , Exposição à Radiação/análise , Trítio/toxicidade , Poluentes Radioativos da Água/toxicidade , Ração Animal , Animais , Cyprinidae/genética , Relação Dose-Resposta à Radiação , Feminino , Laboratórios , Masculino , Exposição à Radiação/efeitos adversos , Reprodutibilidade dos Testes
10.
Exp Eye Res ; 181: 325-345, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30296412

RESUMO

Oxidative cleavage of docosahexaenoate (DHA) in retinal pigmented epithelial (RPE) cells produces 4-hydroxy-7-oxohept-5-enoic acid (HOHA) esters of 2-lysophosphatidylcholine (PC). HOHA-PC spontaneously releases a membrane-permeant HOHA lactone that modifies primary amino groups of proteins and ethanolamine phospholipids to produce 2-(ω-carboxyethyl)pyrrole (CEP) derivatives. CEPs have significant pathological relevance to age-related macular degeneration (AMD) including activation of CEP-specific T-cells leading to inflammatory M1 polarization of macrophages in the retina involved in "dry AMD" and TLR2-dependent induction of angiogenesis that characterizes "wet AMD". RPE cells accumulate DHA from shed rod photoreceptor outer segments through phagocytosis and from plasma lipoproteins secreted by the liver through active uptake from the choriocapillaris. As a cell model of light-induced oxidative damage of DHA phospholipids in RPE cells, ARPE-19 cells were supplemented with DHA, with or without the lipofuscin fluorophore A2E. In this model, light exposure, in the absence of A2E, promoted the generation HOHA lactone-glutathione (GSH) adducts, depletion of intracellular GSH and a competing generation of CEPs. While DHA-rich RPE cells exhibit an inherent proclivity toward light-induced oxidative damage, photosensitization by A2E nearly doubled the amount of lipid oxidation and expanded the spectral range of photosensitivity to longer wavelengths. Exposure of ARPE-19 cells to 1 µM HOHA lactone for 24 h induced massive (50%) loss of lysosomal membrane integrity and caused loss of mitochondrial membrane potential. Using senescence-associated ß-galactosidase (SA ß-gal) staining that detects lysosomal ß-galactosidase, we determined that exposure to HOHA lactone induces senescence in ARPE-19 cells. The present study shows that products of light-induced oxidative damage of DHA phospholipids in the absence of A2E can lead to RPE cell dysfunction. Therefore, their toxicity may be especially important in the early stages of AMD before RPE cells accumulate lipofuscin fluorophores.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Luz/efeitos adversos , Degeneração Macular/metabolismo , Estresse Oxidativo/efeitos da radiação , Epitélio Pigmentado da Retina/metabolismo , Células Cultivadas , Humanos , Peroxidação de Lipídeos , Lisossomos/metabolismo , Lisossomos/efeitos da radiação , Degeneração Macular/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos da radiação , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Epitélio Pigmentado da Retina/efeitos dos fármacos
11.
Autophagy ; 15(2): 259-279, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30176156

RESUMO

Cells challenged by photosensitized oxidations face strong redox stresses and rely on autophagy to either survive or die. However, the use of macroautophagy/autophagy to improve the efficiency of photosensitizers, in terms of inducing cell death, remains unexplored. Here, we addressed the concept that a parallel damage in the membranes of mitochondria and lysosomes leads to a scenario of autophagy malfunction that can greatly improve the efficiency of the photosensitizer to cause cell death. Specific damage to these organelles was induced by irradiation of cells pretreated with 2 phenothiazinium salts, methylene blue (MB) and 1,9-dimethyl methylene blue (DMMB). At a low concentration level (10 nM), only DMMB could induce mitochondrial damage, leading to mitophagy activation, which did not progress to completion because of the parallel damage in lysosome, triggering cell death. MB-induced photodamage was perceived almost instantaneously after irradiation, in response to a massive and nonspecific oxidative stress at a higher concentration range (2 µM). We showed that the parallel damage in mitochondria and lysosomes activates and inhibits mitophagy, leading to a late and more efficient cell death, offering significant advantage (2 orders of magnitude) over photosensitizers that cause unspecific oxidative stress. We are confident that this concept can be used to develop better light-activated drugs. Abbreviations: ΔΨm: mitochondrial transmembrane inner potential; AAU: autophagy arbitrary units; ATG5, autophagy related 5; ATG7: autophagy related 7; BAF: bafilomycin A1; BSA: bovine serum albumin; CASP3: caspase 3; CF: carboxyfluorescein; CTSB: cathepsin B; CVS: crystal violet staining; DCF: dichlorofluorescein; DCFH2: 2',7'-dichlorodihydrofluorescein; DMMB: 1,9-dimethyl methylene blue; ER: endoplasmic reticulum; HaCaT: non-malignant immortal keratinocyte cell line from adult human skin; HP: hydrogen peroxide; LC3B-II: microtubule associated protein 1 light chain 3 beta-II; LMP: lysosomal membrane permeabilization; LTG: LysoTracker™ Green DND-26; LTR: LysoTracker™ Red DND-99; 3-MA: 3-methyladenine; MB: methylene blue; mtDNA: mitochondrial DNA; MitoSOX™: red mitochondrial superoxide probe; MTDR: MitoTracker™ Deep Red FM; MTO: MitoTracker™ Orange CMTMRos; MT-ND1: mitochondrially encoded NADH:ubiquinone oxidoreductase core subunit 1; MTT: methylthiazolyldiphenyl-tetrazolium bromide; 1O2: singlet oxygen; OH. hydroxil radical; PRKN/parkin: parkin RBR E3 ubiquitin protein ligase; PBS: phosphate-buffered saline; PI: propidium iodide; PDT: photodynamic therapy; PS: photosensitizer; QPCR: gene-specific quantitative PCR-based; Rh123: rhodamine 123; ROS: reactive oxygen species RTN: rotenone; SQSTM1/p62: sequestosome 1; SUVs: small unilamellar vesicles; TBS: Tris-buffered saline.


Assuntos
Luz , Lisossomos/patologia , Mitocôndrias/patologia , Autofagia/efeitos dos fármacos , Autofagia/efeitos da radiação , Morte Celular/efeitos da radiação , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Lisossomos/efeitos da radiação , Azul de Metileno/análogos & derivados , Azul de Metileno/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Modelos Biológicos
12.
Sci Transl Med ; 10(456)2018 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-30158153

RESUMO

Cockayne syndrome (CS), a hereditary form of premature aging predominantly caused by mutations in the csb gene, affects multiple organs including skin where it manifests with hypersensitivity toward ultraviolet (UV) radiation and loss of subcutaneous fat. There is no curative treatment for CS, and its pathogenesis is only partially understood. Originally considered for its role in DNA repair, Cockayne syndrome group B (CSB) protein most likely serves additional functions. Using CSB-deficient human fibroblasts, Caenorhabditiselegans, and mice, we show that CSB promotes acetylation of α-tubulin and thereby regulates autophagy. At the organ level, chronic exposure of csbm/m mice to UVA radiation caused a severe skin phenotype with loss of subcutaneous fat, inflammation, and fibrosis. These changes in skin tissue were associated with an accumulation of autophagic/lysosomal proteins and reduced amounts of acetylated α-tubulin. At the cellular level, we found that CSB directly interacts with the histone deacetylase 6 (HDAC6) and the α-tubulin acetyltransferase MEC-17. Upon UVA irradiation, CSB is recruited to the centrosome where it colocalizes with dynein and HDAC6. Administration of the pan-HDAC inhibitor SAHA (suberoylanilide hydroxamic acid) enhanced α-tubulin acetylation, improved autophagic function in CSB-deficient models from all three species, and rescued the skin phenotype in csbm/m mice. HDAC inhibition may thus represent a therapeutic option for CS.


Assuntos
Autofagia/efeitos dos fármacos , Síndrome de Cockayne/patologia , Inibidores de Histona Desacetilases/farmacologia , Lisossomos/metabolismo , Gordura Subcutânea/patologia , Acetilação , Animais , Autofagia/efeitos da radiação , Proteínas Relacionadas à Autofagia/metabolismo , Caenorhabditis elegans/efeitos dos fármacos , Centrossomo/efeitos dos fármacos , Centrossomo/metabolismo , Centrossomo/efeitos da radiação , DNA Helicases/deficiência , DNA Helicases/metabolismo , Enzimas Reparadoras do DNA/deficiência , Enzimas Reparadoras do DNA/metabolismo , Modelos Animais de Doenças , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibroblastos/efeitos da radiação , Lisossomos/efeitos dos fármacos , Lisossomos/efeitos da radiação , Camundongos , Fenótipo , Proteínas de Ligação a Poli-ADP-Ribose/deficiência , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Pele/patologia , Pele/efeitos da radiação , Gordura Subcutânea/efeitos dos fármacos , Gordura Subcutânea/efeitos da radiação , Tubulina (Proteína)/metabolismo , Proteínas Ubiquitinadas/metabolismo , Raios Ultravioleta , Vorinostat/farmacologia
13.
Int J Radiat Biol ; 94(5): 443-453, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29611442

RESUMO

PURPOSE: Lysosomes may have an important role in response to ionizing radiation. Moreover, radiation could affect autophagy, which process involves the activity of lysosomal enzymes. In the present study, the effect of ionizing radiation on the lysosomal compartment of mouse liver was investigated after in vivo exposure. MATERIALS AND METHODS: Morphology and ultrastructure of hepatocytes were assessed by light and electron microscopy, and activities of selected lysosomal enzymes were assessed in 12, 36 and 120 h after exposure to the mean dose of 1 Gy. The levels of autophagy-related proteins LC3-II and p62 were compared by Western blotting between untreated and irradiated animals (120 h after exposure). RESULTS: Increased number of autophagic vacuoles in hepatocytes from exposed animals was documented in the ultrastructural study; destroyed mitochondria were the dominant component of such vacuoles. Moreover, an increased activity of lysosomal hydrolases was observed after exposure. However, levels of autophagy substrates LC3-II and p62 were barely affected in exposed animals 120 h after irradiation when the accumulation of autophagic vacuoles was observed. CONCLUSION: Effects of irradiation included an increased number of autophagic vacuoles, especially of autophagosomes, and increased activity of lysosomal enzymes. However, putative markers of autophagic flux were not observed, which suggested suppression of the completion of the radiation-mediated autophagy pathway.


Assuntos
Fígado/efeitos da radiação , Lisossomos/efeitos da radiação , Animais , Autofagia , Relação Dose-Resposta à Radiação , Coração/efeitos da radiação , Hepatócitos/efeitos da radiação , Fígado/diagnóstico por imagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Frações Subcelulares , Fatores de Tempo , Vacúolos
14.
J Dermatol Sci ; 90(3): 263-275, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29501392

RESUMO

BACKGROUND: The deposition of advanced glycation end products (AGEs) is accelerated in photoaged skin, but the underlying mechanisms remain elusive. Intracellular degradation has been recently considered to play an important role in AGEs removal. Although lysosomal cathepsin D (CatD), B (CatB), L(CatL) and proteasomes are found to degrade internalized AGEs, it remains unknown which protease degrades internalized AGEs in human dermal fibroblasts (HDFs), and whether a decrease in intracellular degradation contributes to enhanced AGEs deposition in photoaged skin. OBJECTIVE: This study aims to investigate the specific proteases that contribute to intracellular AGEs degradation in HDFs and regulate AGEs accumulation in photoaged skin. METHODS: Repetitive UVA irradiation was used to induce primary HDF photoaging in vitro. Uptake and degradation of AGE-BSA were verified and compared between photoaged and non-photoaged fibroblasts with flow cytometry, ELISA and confocal microscopy. Proteasomal and lysosomal activity, expression of CatD, CatB and CatL were also investigated between photoaged and non-photoaged fibroblasts. Further, the effect of protease inhibitors and CatD overexpression via lentiviral transduction on AGE-BSA degradation was analyzed. Finally, the correlation between CatD expression and AGEs accumulation in sun-exposed and sun-protected skin of people from different age was studied with immunohistochemistry. RESULTS: Fibroblasts underwent photoaging in vitro after repetitive UVA irradiation. AGE-BSA was taken up by both photoaged and non-photoaged fibroblasts, but its degradation was significantly decreased in photoaged cells than that of non-photoaged cells. Although the activity of proteasome, CatB, Cat L and Cat D was significantly reduced in photoaged fibroblasts compared to that of non-photoaged cells, and the expression of CatB, CatL and CatD was profoundly attenuated in photoaged fibroblasts, inhibiting proteasome, CatB and CatL did not affect AGE-BSA degradation in HDFs. In contrast, inhibiting CatD activity dose-dependently decreased AGE-BSA degradation; whereas CatD overexpression significantly increased AGE-BSA degradation. Importantly, AGEs accumulation in photo-damaged skin in vivo was inversely correlated with CatD expression. CONCLUSION: CatD plays a major role in intracellular AGEs degradation. Decreased CatD expression and activity impairs intracellular AGEs degradation in photoaged fibroblasts, which may contribute to accelerated AGEs deposition in photoaged skin. The present study provides a potentially novel molecular basis for antiphotoaging therapy.


Assuntos
Catepsina D/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Envelhecimento da Pele/patologia , Pele/patologia , Raios Ultravioleta/efeitos adversos , Adulto , Células Cultivadas , Criança , Fibroblastos/citologia , Fibroblastos/patologia , Fibroblastos/efeitos da radiação , Humanos , Lisossomos/patologia , Lisossomos/efeitos da radiação , Masculino , Microscopia Confocal , Pessoa de Meia-Idade , Cultura Primária de Células , Pele/citologia , Pele/efeitos da radiação , Envelhecimento da Pele/efeitos da radiação , Adulto Jovem
15.
Toxicol In Vitro ; 47: 238-248, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29223573

RESUMO

Recently, designed platelet shaped micron particles that are composed of nano primary particles, called MicNo (=Micron+naNo) particles, have been developed to exploit the benefits of nano size, while removing the adverse effects of nanoparticles. It has been shown that MicNo-ZnO particles exhibit both micron and nanosized particle characteristics. Although physical and chemical properties of MicNo-ZnO particles have been studied, their biocompatibility has not yet been evaluated. Accordingly, the research objective of this study was to evaluate in vitro cytotoxicity, genotoxicity and phototoxicity behaviors of designed MicNo-ZnO particles over human epidermal keratinocyte (HaCaT) cells. MicNo-ZnO particles exhibit much less cytotoxicity with IC50 concentrations between 40 and 50µg/ml, genotoxicity above 40µg/ml and lower photo genotoxicity under UVA on HaCaT than the ZnO nanoparticles. Although their chemistries are the same, the source of this difference in toxicity values may be attributed to size differences between the particles that are probably due to their ability to penetrate into the cells. In the present study, the expansive and detailed in vitro toxicity tests show that the biocompatibility of MicNo-ZnO particles is much better than that of the ZnO nanoparticles. Consequently, MicNo-ZnO particles can be considered an important active ingredient alternative for sunscreen applications due to their safer characteristics with respect to ZnO nanoparticles.


Assuntos
Queratinócitos/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Nanopartículas Metálicas/toxicidade , Modelos Biológicos , Oxidantes Fotoquímicos/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Óxido de Zinco/toxicidade , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Ensaio Cometa , Humanos , Concentração Inibidora 50 , Queratinócitos/citologia , Queratinócitos/metabolismo , Queratinócitos/efeitos da radiação , Lisossomos/metabolismo , Lisossomos/efeitos da radiação , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos da radiação , Nanopartículas Metálicas/química , Nanopartículas Metálicas/efeitos da radiação , Nanopartículas Metálicas/ultraestrutura , Microscopia Eletrônica de Varredura , Microscopia de Fluorescência , Testes de Mutagenicidade , Oxidantes Fotoquímicos/química , Estresse Oxidativo/efeitos da radiação , Tamanho da Partícula , Espécies Reativas de Oxigênio/metabolismo , Propriedades de Superfície , Raios Ultravioleta/efeitos adversos , Óxido de Zinco/química , Óxido de Zinco/efeitos da radiação
16.
Sci Rep ; 7(1): 12843, 2017 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-28993630

RESUMO

Retinal degeneration is prominent in Parkinson's disease (PD), a neuromotor disorder associated with aggregation of α-synuclein (α-syn) in the substantia-nigra (SN). Although α-syn is expressed in the neuroretina, absence of prominent aggregates suggests altered function as the likely cause of retinal pathology. We demonstrate that α-syn impairs ferritinophagy, resulting in the accumulation of iron-rich ferritin in the outer retina in-vivo and retinal-pigment-epithelial (RPE) cells in-vitro. Over-expression of Rab1a restores ferritinophagy, suggesting that α-syn impairs lysosomal function by disrupting the trafficking of lysosomal hydrolases. Surprisingly, upregulation of ferritin in RPE cells by exogenous iron in-vitro stimulated the release of ferritin and α-syn in exosomes, suggesting that iron overload due to impaired ferritinophagy or other cause(s) is likely to initiate prion-like spread of α-syn and ferritin, creating retinal iron dyshomeostasis and associated cytotoxicity. Since over-expression of α-syn is a known cause of PD, these results explain the likely cause of PD-associated retinal degeneration.


Assuntos
Autofagia , Ferritinas/metabolismo , Homeostase , Ferro/metabolismo , Doença de Parkinson/metabolismo , Epitélio Pigmentado da Retina/metabolismo , alfa-Sinucleína/metabolismo , Animais , Autofagossomos/metabolismo , Autofagossomos/efeitos da radiação , Autofagia/efeitos da radiação , Exossomos/metabolismo , Humanos , Luz , Lisossomos/metabolismo , Lisossomos/efeitos da radiação , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Biológicos , Doença de Parkinson/patologia , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/efeitos da radiação , Proteólise/efeitos da radiação , RNA Interferente Pequeno/metabolismo , Epitélio Pigmentado da Retina/patologia , Epitélio Pigmentado da Retina/efeitos da radiação , Proteínas rab1 de Ligação ao GTP/metabolismo
17.
Dalton Trans ; 46(34): 11395-11407, 2017 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-28813052

RESUMO

Many phosphorescent iridium complexes are potent candidates as photodynamic therapeutic agents. In this work, a series of mixed-ligand phosphorescent iridium complexes (Ir1: [Ir(L1)(bpy)Cl](PF6)2; Ir2: [Ir(L1)(ppy)Cl](PF6); Ir3: [Ir(L2)(bpy)Cl](PF6)2; Ir4: [Ir(L2)(ppy)Cl](PF6). L1 = 2,6-bis(2-benzimidazolyl)pyridine; bpy = 2,2'-bipyridine; L2 = 2,6-bis(1-methyl-benzimidazol-2-yl)pyridine; ppy = 2-phenylpyridine) have been synthesized and characterized. These complexes display high luminescence quantum yields and long phosphorescence lifetimes. All the complexes are resistant to hydrolysis in aqueous solutions, and can produce singlet oxygen (1O2) effectively upon irradiation. Ir1 and Ir2 show pH-sensitive emission properties. Interestingly, higher cellular uptake efficiency is observed for Ir2 and Ir4 with the cyclometalated ppy ligand in human lung adenocarcinoma A549 cells. Ir2 with pH-sensitive emission properties can selectively image lysosomes, and Ir4 can specifically target mitochondria. Both Ir2 and Ir4 exhibit potent photodynamic therapy (PDT) effects, with Ir2 displaying a higher phototoxicity index (PI) especially in A549 cells (PI > 54). Mechanism studies indicate that Ir2 and Ir4 can induce apoptosis through reactive oxygen species (ROS) generation and caspase activation upon visible light (425 nm) irradiation. As expected, Ir2 can damage lysosomes more effectively with a pH-sensitive singlet oxygen (1O2) yield, while Ir4 tends to impair mitochondrial function. Nevertheless, the practical application of Ir2 and Ir4 for PDT may be limited to superficial tumors due to the short excitation wavelength (425 nm). Our study gives insights into the design and anticancer mechanisms of new metal-based PDT anticancer agents.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Irídio/química , Compostos Organometálicos/química , Compostos Organometálicos/farmacologia , Fotoquimioterapia , Antineoplásicos/síntese química , Antineoplásicos/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Transporte Biológico , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Estabilidade de Medicamentos , Humanos , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Espaço Intracelular/efeitos da radiação , Ligantes , Lisossomos/efeitos dos fármacos , Lisossomos/efeitos da radiação , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/efeitos da radiação , Compostos Organometálicos/síntese química , Compostos Organometálicos/metabolismo , Oxigênio Singlete/metabolismo , Água/química
18.
Sci Rep ; 7(1): 6734, 2017 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-28751688

RESUMO

Mobilization of specific mechanisms of regulated cell death is a promising alternative to treat challenging illness such as neurodegenerative disease and cancer. The use of light to activate these mechanisms may provide a route for target-specific therapies. Two asymmetric porphyrins with opposite charges, the negatively charged TPPS2a and the positively charged CisDiMPyP were compared in terms of their properties in membrane mimics and in cells. CisDiMPyP interacts to a larger extent with model membranes and with cells than TPPS2a, due to a favorable electrostatic interaction. CisDiMPyP is also more effective than TPPS2a in damaging membranes. Surprisingly, TPPS2a is more efficient in causing photoinduced cell death. The lethal concentration on cell viability of 50% (LC50) found for TPPS2a was ~3.5 (raw data) and ~5 (considering photosensitizer incorporation) times smaller than for CisDiMPyP. CisDiMPyP damaged mainly mitochondria and triggered short-term phototoxicity by necro-apoptotic cell death. Photoexcitation of TPPS2a promotes mainly lysosomal damage leading to autophagy-associated cell death. Our data shows that an exact damage in lysosome is more effective to diminish proliferation of HeLa cells than a similar damage in mitochondria. Precisely targeting organelles and specifically triggering regulated cell death mechanisms shall help in the development of new organelle-target therapies.


Assuntos
Apoptose/efeitos dos fármacos , Benzenossulfonatos/farmacologia , Lisossomos/efeitos dos fármacos , Fármacos Fotossensibilizantes/farmacologia , Porfirinas/farmacologia , Compostos de Piridínio/farmacologia , Apoptose/efeitos da radiação , Benzenossulfonatos/química , Membrana Celular/efeitos dos fármacos , Membrana Celular/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Células HeLa , Humanos , Cinética , Luz , Lisossomos/metabolismo , Lisossomos/efeitos da radiação , Membranas Artificiais , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Fotoquimioterapia , Fármacos Fotossensibilizantes/química , Porfirinas/química , Compostos de Piridínio/química , Eletricidade Estática , Relação Estrutura-Atividade
19.
Photochem Photobiol ; 93(6): 1502-1508, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28696570

RESUMO

We previously reported that a low level of lysosomal photodamage potentiated the phototoxic effect of subsequent mitochondrial photodamage mediated by the benzoporphyrin derivative (BPD) in murine hepatoma 1c1c7 cells. This was attributed to release of Ca2+ from damaged lysosomes and a calpain-mediated conversion of the autophagy-related protein ATG5 to a pro-apoptotic fragment. We now report a comparison of these results with those obtained with the human non-small-cell lung cancer A549 cell line. A549 cells contained lower levels of ATG5 and were less responsive than 1c1c7 cultures to the PDT combination. A rapid appearance of caspase 3/7 activation together with formation of condensed chromatin indicated initiation of apoptosis in both cell lines, but to a lesser extent in A549 cultures. Both cell lines became highly vacuolated within 16 h of combination PDT or an equivalent phototoxic dose from BPD alone. The vacuole periphery was labeled with a fluorescent probe for the endoplasmic reticulum (ER), and vacuole formation was prevented by presence of the protein synthesis inhibitor cycloheximide. These effects are characteristics of a caspase-independent death mode termed paraptosis previously associated with ER stress. These studies suggest that paraptosis may be a more frequent outcome of PDT than has hitherto been realized.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Lisossomos/efeitos da radiação , Mitocôndrias/efeitos da radiação , Fotólise , Apoptose , Western Blotting , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/patologia
20.
Nanotechnology ; 28(5): 055101, 2017 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-28029105

RESUMO

Stimuli-responsive anticancer formulations can promote drug release and activation within the target tumour, facilitate cellular uptake, as well as improve the therapeutic efficacy of drugs and reduce off-target effects. In the present work, indocyanine green (ICG)-containing polyglutamate (PGA) nanoparticles were developed and characterized. Digestion of nanoparticles with cathepsin B, a matrix metalloproteinase overexpressed in the microenvironment of advanced tumours, decreased particle size and increased ICG cellular uptake. Incorporation of ICG in PGA nanoparticles provided the NIR-absorbing agent with time-dependent altered optical properties in the presence of cathepsin B. Having minimal dark toxicity, the formulation exhibited significant cytotoxicity upon NIR exposure. Combined use of the formulation with saporin, a ribosome-inactivating protein, resulted in synergistically enhanced cytotoxicity attributed to the photo-induced release of saporin from endo/lysosomes. The results suggest that this therapeutic approach can offer significant therapeutic benefit in the treatment of superficial malignancies, such as head and neck tumours.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Catepsina B/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Células Epiteliais/efeitos dos fármacos , Terapia de Alvo Molecular/métodos , Nanopartículas/química , Proteínas Inativadoras de Ribossomos Tipo 1/farmacologia , Antineoplásicos Fitogênicos/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Corantes/química , Composição de Medicamentos/métodos , Liberação Controlada de Fármacos , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Endossomos/efeitos da radiação , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Células Epiteliais/efeitos da radiação , Humanos , Verde de Indocianina/química , Raios Infravermelhos , Cinética , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Lisossomos/efeitos da radiação , Nanopartículas/ultraestrutura , Tamanho da Partícula , Ácido Poliglutâmico/química , Proteólise , Proteínas Inativadoras de Ribossomos Tipo 1/química , Saporinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA