Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Aging (Albany NY) ; 8(10): 2449-2462, 2016 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-27744420

RESUMEN

Cellular senescence, a form of cell cycle arrest, is one of the cellular responses to different types of exogenous and endogenous damage. The senescence phenotype can be induced in vitro by oncogene overexpression and/or DNA damage. Recently, we have reported a novel mechanism of cellular senescence induction by mild genotoxic stress. Specifically, we have shown that the formation of a small number of DNA lesions in normal and cancer cells during S phase leads to cellular senescence-like arrest within the same cell cycle. Here, based on this mechanism, we suggest an approach to remotely induce premature senescence in human cell cultures using short-term light irradiation. We used the genetically encoded photosensitizers, tandem KillerRed and miniSOG, targeted to chromatin by fusion to core histone H2B to induce moderate levels of DNA damage by light in S phase cells. We showed that the cells that express the H2B-fused photosensitizers acquire a senescence phenotype upon illumination with the appropriate light source. Furthermore, we demonstrated that both chromatin-targeted tandem KillerRed (produces O2¯) and miniSOG (produces 1O2) induce single-stranded DNA breaks upon light illumination. Interestingly, miniSOG was also able to induce double-stranded DNA breaks.


Asunto(s)
Senescencia Celular/genética , Roturas del ADN de Doble Cadena , Roturas del ADN de Cadena Simple , Luz , Fármacos Fotosensibilizantes/farmacología , Humanos , Fase S/genética
2.
Biotechniques ; 61(2): 92-4, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27528074

RESUMEN

Genetically encoded photosensitizers represent a promising optogenetic tool for the induction of light-controlled oxidative stress strictly localized to a selected intracellular compartment. Here we tested the phototoxic effects of the flavin-containing phototoxic protein miniSOG targeted to the cytoplasmic surfaces of late endosomes and lysosomes by fusion with Rab7. In HeLa Kyoto cells stably expressing miniSOG-Rab7, we demonstrated a high level of cell death upon blue-light illumination. Pepstatin A completely abolished phototoxicity of miniSOG-Rab7, showing a key role for cathepsin D in this model. Using a far-red fluorescence sensor for caspase-3, we observed caspase-3 activation during miniSOG-Rab7-mediated cell death. We conclude that upon illumination, miniSOG-Rab7 induces lysosomal membrane permeabilization (LMP) and leakage of cathepsins into the cytosol, resulting in caspase-dependent apoptosis.


Asunto(s)
Muerte Celular , Lisosomas , Microscopía Fluorescente/métodos , Optogenética/métodos , Fármacos Fotosensibilizantes/metabolismo , Oxígeno Singlete/farmacología , Caspasa 3/análisis , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Células HeLa , Humanos , Luz , Proteínas Luminiscentes/química , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Lisosomas/genética , Lisosomas/metabolismo , Fármacos Fotosensibilizantes/química , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Oxígeno Singlete/metabolismo , Proteínas de Unión al GTP rab/química , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
3.
Cytokine ; 84: 10-6, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27203665

RESUMEN

OX40 receptor-expressing regulatory T cells (Tregs) populate tumors and suppress a variety of immune cells, posing a major obstacle for cancer immunotherapy. Different ways to functionally inactivate Tregs by triggering OX40 receptor have been suggested, including anti-OX40 antibodies and Fc:OX40L fusion proteins. To investigate whether the soluble extracellular domain of OX40L (OX40Lexo) is sufficient to enhance antitumor immune response, we generated an OX40Lexo-expressing CT26 colon carcinoma cell line and studied its tumorigenicity in immunocompetent BALB/c and T cell deficient nu/nu mice. We found that soluble OX40L expressed in CT26 colon carcinoma favors the induction of an antitumor response which is not limited just to cells co-expressing EGFP as an antigenic determinant, but also eliminates CT26 cells expressing another fluorescent protein, KillerRed. Tumor rejection required the presence of T lymphocytes, as indicated by the unhampered tumor growth in nu/nu mice. Subsequent re-challenge of tumor-free BALB/c mice with CT26 EGFP cells resulted in no tumor growth, which is indicative of the formation of immunological memory. Adoptive transfer of splenocytes from mice that successfully rejected CT26 OX40Lexo EGFP tumors to naïve mice conferred 100% resistance to subsequent challenge with the CT26 EGFP tumor.


Asunto(s)
Carcinoma/metabolismo , Neoplasias del Colon/metabolismo , Ligando OX40/metabolismo , Traslado Adoptivo/métodos , Animales , Carcinoma/inmunología , Carcinoma/terapia , Línea Celular , Neoplasias del Colon/inmunología , Neoplasias del Colon/terapia , Femenino , Proteínas Fluorescentes Verdes/inmunología , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Memoria Inmunológica/inmunología , Memoria Inmunológica/fisiología , Inmunoterapia/métodos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ligando OX40/inmunología , Receptores OX40/inmunología , Receptores OX40/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
4.
Biophys J ; 109(2): 380-9, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26200874

RESUMEN

Spectral diversity of fluorescent proteins, crucial for multiparameter imaging, is based mainly on chemical diversity of their chromophores. Recently we have reported, to our knowledge, a new green fluorescent protein WasCFP-the first fluorescent protein with a tryptophan-based chromophore in the anionic state. However, only a small portion of WasCFP molecules exists in the anionic state at physiological conditions. In this study we report on an improved variant of WasCFP, named NowGFP, with the anionic form dominating at 37°C and neutral pH. It is 30% brighter than enhanced green fluorescent protein (EGFP) and exhibits a fluorescence lifetime of 5.1 ns. We demonstrated that signals of NowGFP and EGFP can be clearly distinguished by fluorescence lifetime in various models, including mammalian cells, mouse tumor xenograft, and Drosophila larvae. NowGFP thus provides an additional channel for multiparameter fluorescence lifetime imaging microscopy of green fluorescent proteins.


Asunto(s)
Proteínas Fluorescentes Verdes/química , Animales , Animales Modificados Genéticamente , Aniones/química , Drosophila , Escherichia coli , Fluorescencia , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Microscopía Confocal , Mutación , Procesos Fotoquímicos , Temperatura , Triptófano/química , Triptófano/metabolismo
5.
J Biomed Opt ; 19(7): 071403, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24365992

RESUMEN

KillerRed is a unique phototoxic red fluorescent protein that can be used to induce local oxidative stress by green-orange light illumination. Here we studied phototoxicity of KillerRed targeted to cytoplasmic surface of lysosomes via fusion with Rab7, a small GTPase that is known to be attached to membranes of late endosomes and lysosomes. It was found that lysosome-associated KillerRed ensures efficient light-induced cell death similar to previously reported mitochondria- and plasma membrane-localized KillerRed. Inhibitory analysis demonstrated that lysosomal cathepsins play an important role in the manifestation of KillerRed-Rab7 phototoxicity. Time-lapse monitoring of cell morphology, membrane integrity, and nuclei shape allowed us to conclude that KillerRed-Rab7-mediated cell death occurs via necrosis at high light intensity or via apoptosis at lower light intensity. Potentially, KillerRed-Rab7 can be used as an optogenetic tool to direct target cell populations to either apoptosis or necrosis.


Asunto(s)
Apoptosis , Proteínas Fluorescentes Verdes/química , Lisosomas/química , Necrosis , Fármacos Fotosensibilizantes/química , Animales , Línea Celular , Membrana Celular/metabolismo , Citoplasma/metabolismo , Fibroblastos/metabolismo , GTP Fosfohidrolasas/metabolismo , Células HeLa , Humanos , Proteínas Luminiscentes/química , Lisosomas/metabolismo , Estrés Oxidativo , Fotoquimioterapia/instrumentación , Fotoquimioterapia/métodos , Ratas , Proteínas de Unión al GTP rab/química , Proteínas de Unión a GTP rab7 , Proteína Fluorescente Roja
6.
Biochim Biophys Acta ; 1830(11): 5059-67, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23876295

RESUMEN

BACKGROUND: Genetically encoded photosensitizers are a promising optogenetic instrument for light-induced production of reactive oxygen species in desired locations within cells in vitro or whole body in vivo. Only two such photosensitizers are currently known, GFP-like protein KillerRed and FMN-binding protein miniSOG. In this work we studied phototoxic effects of miniSOG in cancer cells. METHODS: HeLa Kyoto cell lines stably expressing miniSOG in different localizations, namely, plasma membrane, mitochondria or chromatin (fused with histone H2B) were created. Phototoxicity of miniSOG was tested on the cells in vitro and tumor xenografts in vivo. RESULTS: Blue light induced pronounced cell death in all three cell lines in a dose-dependent manner. Caspase 3 activation was characteristic of illuminated cells with mitochondria- and chromatin-localized miniSOG, but not with miniSOG in the plasma membrane. In addition, H2B-miniSOG-expressing cells demonstrated light-induced activation of DNA repair machinery, which indicates massive damage of genomic DNA. In contrast to these in vitro data, no detectable phototoxicity was observed on tumor xenografts with HeLa Kyoto cell lines expressing mitochondria- or chromatin-localized miniSOG. CONCLUSIONS: miniSOG is an excellent genetically encoded photosensitizer for mammalian cells in vitro, but it is inferior to KillerRed in the HeLa tumor. GENERAL SIGNIFICANCE: This is the first study to assess phototoxicity of miniSOG in cancer cells. The results suggest an effective ontogenetic tool and may be of interest for molecular and cell biology and biomedical applications.


Asunto(s)
Flavoproteínas/genética , Terapia Genética/métodos , Oxígeno/metabolismo , Fármacos Fotosensibilizantes/metabolismo , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Muerte Celular/genética , Línea Celular , Línea Celular Tumoral , Membrana Celular/genética , Membrana Celular/metabolismo , Cromatina/genética , Cromatina/metabolismo , Daño del ADN , Reparación del ADN , Dermatitis Fototóxica/etiología , Dermatitis Fototóxica/genética , Dermatitis Fototóxica/metabolismo , Femenino , Flavoproteínas/metabolismo , Células HEK293 , Células HeLa , Humanos , Luz/efectos adversos , Ratones , Ratones Desnudos , Mitocondrias/genética , Mitocondrias/metabolismo , Riboflavina/genética , Riboflavina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA