Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Anal Biochem ; 405(2): 246-54, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20561505

RESUMEN

Glycogen synthase kinase-3beta (GSK3beta) and casein kinase-1alpha (CK1alpha) are multifunctional kinases that play critical roles in the regulation of a number of cellular processes. In spite of their importance, molecular imaging tools for noninvasive and real-time monitoring of their kinase activities have not been devised. Here we report development of the bioluminescent GSK3beta and CK1alpha reporter (BGCR) based on firefly luciferase complementation. Treatment of SW620 cells stably expressing the reporter with inhibitors of GSK3beta (SB415286 and LiCl) or CK1alpha (CKI-7) resulted in dose- and time-dependent increases in BGCR activity that were validated using Western blotting. No increase in bioluminescence was observed in the case of S37A mutant (GSK3beta inhibitors) or S45A mutant (CKI-7), demonstrating the specificity of the reporter. Imaging of mice tumor xenograft generated with BGCR-expressing SW620 cells following treatment with LiCl showed unique oscillations in GSK3beta activity that were corroborated by phosphorylated GSK3beta immunoblotting. Taken together, the BGCR is a novel molecular imaging tool that reveals unique insight into GSK3beta and CK1alpha kinase activities and may provide a powerful tool in experimental therapeutics for rapid optimization of dose and schedule of targeted therapies and for monitoring therapeutic response.


Asunto(s)
Caseína Quinasa Ialfa/química , Glucógeno Sintasa Quinasa 3/química , Imagen Molecular/métodos , Animales , Caseína Quinasa Ialfa/metabolismo , Células Cultivadas , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Proteínas Luminiscentes/química , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Trasplante Heterólogo , Proteínas Wnt/química , Proteínas Wnt/metabolismo , Proteína Wnt3
2.
Mol Pharm ; 7(6): 1921-9, 2010 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-20964352

RESUMEN

The past decade has seen momentous development in brain cancer research in terms of novel imaging-assisted surgeries, molecularly targeted drug-based treatment regimens or adjuvant therapies and in our understanding of molecular footprints of initiation and progression of malignancy. However, mortality due to brain cancer has essentially remained unchanged in the last three decades. Thus, paradigm-changing diagnostic and therapeutic reagents are urgently needed. Nanotheranostic platforms are powerful tools for imaging and treatment of cancer. Multifunctionality of these nanovehicles offers a number of advantages over conventional agents. These include targeting to a diseased site thereby minimizing systemic toxicity, the ability to solubilize hydrophobic or labile drugs leading to improved pharmacokinetics and their potential to image, treat and predict therapeutic response. In this article, we will discuss the application of newer theranostic nanoparticles in targeted brain cancer imaging and treatment.


Asunto(s)
Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/tratamiento farmacológico , Diagnóstico por Imagen/métodos , Nanopartículas/uso terapéutico , Animales , Humanos , Nanopartículas/efectos adversos
3.
Cancer Biol Ther ; 2(4 Suppl 1): S71-8, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14508083

RESUMEN

The therapeutic use of TRAIL as an inducer of tumor specific cell death prove to be an useful strategy to overcome resistant of cancer cells to conventional chemotherapeutic agents. Although five death receptors have been identified which bind TRAIL and a number of checkpoint involved in TRAIL pathway are known, the molecular mechanism underlying the resistant or sensitivity of normal/transformed cells remains poorly understood. The key to the clinical use of TRAIL is that normal cells are resistant to its cytotoxic activity and that it specifically targets cancer cells thus resulting in large therapeutic index. In spite of this observation, TRAIL will probably not be viable as single agent as there are numerous reports that have demonstrated the resistance of various tumor cells to TRAIL. The concept of combination therapy is therefore key for the use of TRAIL against tumors that are refractory to treatment with either agent alone. Results from preclinical studies on TRAIL have been very promising for cancer treatment; however, outcome from phase I/II clinical trials is eagerly awaited to assess its safety. Only time will tell whether the search for the Holy Grail for treatment of cancer will end with TRAIL?


Asunto(s)
Antineoplásicos/farmacología , Glicoproteínas de Membrana/fisiología , Neoplasias/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/fisiología , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis , Caspasas/metabolismo , Muerte Celular , Línea Celular Tumoral , Daño del ADN , Humanos , Modelos Biológicos , Neoplasias/metabolismo , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF
4.
Mol Imaging ; 5(2): 129-37, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16954027

RESUMEN

Programmed cell death (apoptosis) is a ubiquitous means utilized by multicellular organisms for elimination of unwanted cells during development and homeostasis. Dysregulated apoptosis is implicated in an array of clinical disorders including cancer, autoimmune diseases, neurodegenerative disorders, and ischemia. During programmed cell death, a series of proteases, known as caspases, with different specificities play crucial roles in the apoptotic process. Caspase-3, a group II cysteine aspartate protease, recognizes and cleaves substrates harboring the amino acid sequence aspartic acid-glutamic acid-valine-aspartic acid (DEVD), and it plays an important role in the terminal phase of apoptosis. Here we report the development of a novel imaging platform for sensing the activation of cellular proteases. A recombinant chimeric protein was constructed, composed of a cell-surface-targeted single-chain antibody (sFv) fused to a Golgi retention signal. The DEVD tetrapeptide sequence was included between the single-chain antibody and the Golgi retention signal as a caspase-3 protease cleavage site. When expressed in cultured cells this fusion protein was localized to Golgi bodies and was not detected on the cell surface. Induction of apoptosis resulted in cleavage of the fusion protein releasing the single-chain antibody from the Golgi retention signal in a caspase-dependent manner. As a result, in cells undergoing apoptosis the single-chain antibody was visualized at the cell surface by immunofluorescence microscopy. The expression of sFv on the surface of cells in a protease-dependent manner provides a unique opportunity for real-time imaging through the use of targeted nanoparticles. This methodology may provide for a multimodal noninvasive real-time imaging of apoptosis and a new opportunity for high-throughput screening of cell-death-modulating therapeutic agents.


Asunto(s)
Apoptosis/fisiología , Péptido Hidrolasas/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Técnica del Anticuerpo Fluorescente , Vectores Genéticos , Humanos , Hidrólisis , Microscopía Fluorescente
5.
Proc Natl Acad Sci U S A ; 99(26): 16551-5, 2002 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-12475931

RESUMEN

Strict coordination of proliferation and programmed cell death (apoptosis) is essential for normal physiology. An imbalance in these two opposing processes results in various diseases including AIDS, neurodegenerative disorders, myelodysplastic syndromes, ischemiareperfusion injury, cancer, autoimmune disease, among others. Objective and quantitative noninvasive imaging of apoptosis would be a significant advance for rapid and dynamic screening as well as validation of experimental therapeutic agents. Here, we report the development of a recombinant luciferase reporter molecule that when expressed in mammalian cells has attenuated levels of reporter activity. In cells undergoing apoptosis, a caspase-3-specific cleavage of the recombinant product occurs, resulting in the restoration of luciferase activity that can be detected in living animals with bioluminescence imaging. The ability to image apoptosis noninvasively and dynamically over time provides an opportunity for high-throughput screening of proapoptotic and antiapoptotic compounds and for target validation in vivo in both cell lines and transgenic animals.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis , Caspasa 3 , Activación Enzimática , Glioma/tratamiento farmacológico , Glioma/patología , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes , Glicoproteínas de Membrana/uso terapéutico , Ratones , Receptores de Estrógenos/química , Receptores de Estrógenos/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/uso terapéutico
6.
Mol Imaging ; 1(1): 43-55, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12920860

RESUMEN

Cancer gene therapy is an active area of research relying upon the transfer and subsequent expression of a therapeutic transgene into tumor cells in order to provide for therapeutic selectivity. Noninvasive assessment of therapeutic response and correlation of the location, magnitude, and duration of transgene expression in vivo would be particularly useful in the development of cancer gene therapy protocols by facilitating optimization of gene transfer protocols, vector development, and prodrug dosing schedules. In this study, we developed an adenoviral vector containing both the therapeutic transgene yeast cytosine deaminase (yCD) along with an optical reporter gene (luciferase). Following intratumoral injection of the vector into orthotopic 9 L gliomas, anatomical and diffusion-weighted MR images were obtained over time in order to provide for quantitative assessment of overall therapeutic efficacy and spatial heterogeneity of cell kill, respectively. In addition, bioluminescence images were acquired to assess the duration and magnitude of gene expression. MR images revealed significant reduction in tumor growth rates associated with yCD/5-fluorocytosine (5FC) gene therapy. Significant increases in mean tumor diffusion values were also observed during treatment with 5FC. Moreover, spatial heterogeneity in tumor diffusion changes were also observed revealing that diffusion magnetic resonance imaging could detect regional therapeutic effects due to the nonuniform delivery and/or expression of the therapeutic yCD transgene within the tumor mass. In addition, in vivo bioluminescence imaging detected luciferase gene expression, which was found to decrease over time during administration of the prodrug providing a noninvasive surrogate marker for monitoring gene expression. These results demonstrate the efficacy of the yCD/5FC strategy for the treatment of brain tumors and reveal the feasibility of using multimodality molecular and functional imaging for assessment of gene expression and therapeutic efficacy.


Asunto(s)
Adenoviridae/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioma/genética , Nucleósido Desaminasas/genética , Animales , Neoplasias Encefálicas/patología , Citosina Desaminasa , Expresión Génica , Genes Reporteros , Vectores Genéticos , Glioma/patología , Glioma/terapia , Luciferasas/genética , Imagen por Resonancia Magnética/métodos , Masculino , Ratas , Ratas Endogámicas F344 , Proteínas Recombinantes de Fusión/genética , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA