Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Intervalo de año de publicación
2.
Semin Cancer Biol ; 80: 58-72, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-32070764

RESUMEN

The recent advances in cancer immunotherapy confirm the crucial role of the immune system in cancer progression and treatment. Chronic inflammation and reduced immune surveillance are both features of the tumor microenvironment. Strategies aimed at reverting pro-tumor inflammation and stimulating the antitumor immune components are being actively searched, and the anticancer effects of many candidate drugs have been linked to their ability to modulate the immune system. Marine organisms constitute a rich reservoir of new bioactive molecules; some of them have already been exploited for pharmaceutical use, whereas many others are undergoing clinical or preclinical investigations for the treatment of different diseases, including cancer. In this review, we will discuss the immune-modulatory properties of marine compounds for their potential use in cancer prevention and treatment and as possible tools in the context of cancer immunotherapy.


Asunto(s)
Neoplasias , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Organismos Acuáticos , Humanos , Inmunoterapia , Inflamación/tratamiento farmacológico , Inflamación/prevención & control , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
3.
Clin Epigenetics ; 11(1): 68, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-31060628

RESUMEN

BACKGROUND: DNA methyltransferases (DNMTs) are epigenetic enzymes involved in embryonic development, cell differentiation, epithelial to mesenchymal transition, and control of gene expression, whose overexpression or enhanced catalytic activity has been widely reported in cancer initiation and progression. To date, two DNMT inhibitors (DNMTi), 5-azacytidine (5-AZA) and 5-aza-2'-deoxycytidine (DAC), are approved for the treatment of myelodysplastic syndromes and acute myeloid leukemia. Nevertheless, they are chemically instable and quite toxic for healthy cells; thus, the discovery of novel DNMTi is urgent. RESULTS: Here, we report the identification of a new quinoline-based molecule, MC3353, as a non-nucleoside inhibitor and downregulator of DNMT. This compound was able, in promoter demethylating assays, to induce enhanced green fluorescence protein (EGFP) gene expression in HCT116 cells and transcription in a cytomegalovirus (CMV) promoter-driven luciferase reporter system in KG-1 cells. Moreover, MC3353 displayed a strong antiproliferative activity when tested on HCT116 colon cancer cells after 48 h of treatment at 0.5 µM. At higher doses, this compound provided a cytotoxic effect in double DNMT knockout HCT116 cells. MC3353 was also screened on a different panel of cancer cells (KG-1 and U-937 acute myeloid leukemia, RAJI Burkitt's lymphoma, PC-3 prostate cancer, and MDA-MB-231 breast cancer), where it arrested cell proliferation and reduced viability after 48 h of treatment with IC50 values ranging from 0.3 to 0.9 µM. Compared to healthy cell models, MC3353 induced apoptosis (e.g., U-937 and KG-1 cells) or necrosis (e.g., RAJI cells) at lower concentrations. Importantly, together with the main DNMT3A enzyme inhibition, MC3353 was also able to downregulate the DNMT3A protein level in selected HCT116 and PC-3 cell lines. Additionally, this compound provided impairment of the epithelial-to-mesenchymal transition (EMT) by inducing E-cadherin while reducing matrix metalloproteinase (MMP2) mRNA and protein levels in PC-3 and HCT116 cells. Last, tested on a panel of primary osteosarcoma cell lines, MC3353 markedly inhibited cell growth with low single-digit micromolar IC50 ranging from 1.1 to 2.4 µM. Interestingly, in Saos-2 osteosarcoma cells, MC3353 induced both expression of genes and mineralized the matrix as evidence of osteosarcoma to osteoblast differentiation. CONCLUSIONS: The present work describes MC3353 as a novel DNMTi displaying a stronger in cell demethylating ability than both 5-AZA and DAC, providing re-activation of the silenced ubiquitin C-terminal hydrolase L1 (UCHL1) gene. MC3353 displayed dose- and time-dependent antiproliferative activity in several cancer cell types, inducing cell death and affecting EMT through E-cadherin and MMP2 modulation. In addition, this compound proved efficacy even in primary osteosarcoma cell models, through the modulation of genes involved in osteoblast differentiation.


Asunto(s)
Aminoquinolinas/síntesis química , Aminoquinolinas/farmacología , ADN-Citosina Metilasas/antagonistas & inhibidores , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Neoplasias/metabolismo , Aminoquinolinas/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Metilación de ADN , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/química , Epigénesis Genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Humanos , Neoplasias/tratamiento farmacológico , Pirimidinas/química
4.
Free Radic Biol Med ; 134: 177-189, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30639617

RESUMEN

Redox changes and generation of reactive oxygen species (ROS) are part of normal cell metabolism. While low ROS levels are implicated in cellular signaling pathways necessary for survival, higher levels play major roles in cancer development as well as cell death signaling and execution. A role for redox changes in apoptosis has been long established; however, several new modalities of regulated cell death have been brought to light, for which the importance of ROS production as well as ROS source and targets are being actively investigated. In this review, we summarize recent findings on the role of ROS and redox changes in the activation and execution of two major forms of regulated cell death, necroptosis and ferroptosis. We also discuss the potential of using modulators of these two forms of cell death to exacerbate ROS as a promising anticancer therapy.


Asunto(s)
Ferroptosis , Necroptosis , Neoplasias/patología , Animales , Humanos , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
5.
Mar Drugs ; 16(12)2018 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-30572618

RESUMEN

Treatment of acute myeloid leukemia (AML) patients is still hindered by resistance and relapse, resulting in an overall poor survival rate. Recently, combining specific B-cell lymphoma (Bcl)-2 inhibitors with compounds downregulating myeloid cell leukemia (Mcl)-1 has been proposed as a new effective strategy to eradicate resistant AML cells. We show here that 1(R), 6(S), 1'(R), 6'(S), 11(R), 17(S)-fistularin-3, a bromotyrosine compound of the fistularin family, isolated from the marine sponge Suberea clavata, synergizes with Bcl-2 inhibitor ABT-199 to efficiently kill Mcl-1/Bcl-2-positive AML cell lines, associated with Mcl-1 downregulation and endoplasmic reticulum stress induction. The absolute configuration of carbons 11 and 17 of the fistularin-3 stereoisomer was fully resolved in this study for the first time, showing that the fistularin we isolated from the marine sponge Subarea clavata is in fact the (+)-11(R), 17(S)-fistularin-3 stereoisomer keeping the known configuration 1(R), 6(S), 1'(R), and 6'(S) for the verongidoic acid part. Docking studies and in vitro assays confirm the potential of this family of molecules to inhibit DNA methyltransferase 1 activity.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Isoxazoles/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Sulfonamidas/farmacología , Tirosina/análogos & derivados , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células HL-60 , Humanos , Isoxazoles/administración & dosificación , Isoxazoles/química , Isoxazoles/aislamiento & purificación , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Simulación del Acoplamiento Molecular , Poríferos/química , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sulfonamidas/administración & dosificación , Tirosina/administración & dosificación , Tirosina/química , Tirosina/aislamiento & purificación , Tirosina/farmacología , Células U937
6.
Oncotarget ; 7(17): 24027-49, 2016 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-27006469

RESUMEN

We characterized the brominated alkaloid Isofistularin-3 (Iso-3), from the marine sponge Aplysina aerophoba, as a new DNA methyltransferase (DNMT)1 inhibitor. Docking analysis confirmed our in vitro DNMT inhibition data and revealed binding of Iso-3 within the DNA binding site of DNMT1. Subsequent increased expression of tumor suppressor gene aryl hydrocarbon receptor (AHR) could be correlated to decreased methylation of CpG sites within the essential Sp1 regulatory region of its promoter. Iso-3 induced growth arrest of cancer cells in G0/G1 concomitant with increased p21 and p27 expression and reduced cyclin E1, PCNA and c-myc levels. Reduced proliferation was accompanied by morphological changes typical of autophagy revealed by fluorescent and transmission electron microscopy and validated by LC3I-II conversion. Furthermore, Iso-3 strongly synergized with tumor-necrosis-factor related apoptosis inducing ligand (TRAIL) in RAJI [combination index (CI) = 0.22] and U-937 cells (CI = 0.21) and increased TRAIL-induced apoptosis via a mechanism involving reduction of survivin expression but not of Bcl-2 family proteins nor X-linked inhibitor of apoptosis protein (XIAP). Iso-3 treatment decreased FLIPL expression and triggered activation of endoplasmatic reticulum (ER) stress with increased GRP78 expression, eventually inducing TRAIL receptor death receptor (DR)5 surface expression. Importantly, as a potential candidate for further anticancer drug development, Iso-3 reduced the viability, colony and in vivo tumor forming potential without affecting the viability of PBMCs from healthy donors or zebrafish development.


Asunto(s)
Alcaloides/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Pez Cebra/crecimiento & desarrollo , Alcaloides/química , Animales , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor , Proliferación Celular/efectos de los fármacos , Descubrimiento de Drogas , Chaperón BiP del Retículo Endoplásmico , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Poríferos/química , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Células Tumorales Cultivadas , Pez Cebra/metabolismo
7.
Curr Top Med Chem ; 16(7): 745-76, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26303418

RESUMEN

Despite considerable scientific progress, the burden of cancer in our society remains a major public health problem. Tumorigenesis is recognized as a complex and multistep process that involves the accumulation of successive transformational events with multi-factorial etiology. Nevertheless, such events result in the acquisition of key hallmark characteristics that are shared by all cancer cells. Accumulating evidence indicates that, besides genetic alterations, epigenetic mechanisms (heritable changes in gene expression caused by modifications in chromatin structure without alterations of DNA sequence) are implicated in the acquisition of malignant phenotype. The potential reversibility of epigenetic alterations linked to tumorigenesis offers a promising avenue for therapeutic intervention. This review focuses on the epigenetic regulation of the cancer hallmarks and the foreseeable use of epigenetic drugs to target these features as a promising strategy for anti-cancer therapy. Based on this body of evidence, we believe that epigenetic deregulations can affect virtually all cell functions and therefore therapeutic approaches with epigenetic drugs could allow multi-target approach against the hallmarks of cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Epigénesis Genética/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Medicina de Precisión/tendencias , Animales , Metilación de ADN , Inhibidores de Histona Desacetilasas/uso terapéutico , Histonas/efectos de los fármacos , Histonas/genética , Humanos
8.
J Med Chem ; 57(3): 701-13, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24387159

RESUMEN

DNA methyltransferases (DNMTs) are important enzymes involved in epigenetic control of gene expression and represent valuable targets in cancer chemotherapy. A number of nucleoside DNMT inhibitors (DNMTi) have been studied in cancer, including in cancer stem cells, and two of them (azacytidine and decitabine) have been approved for treatment of myelodysplastic syndromes. However, only a few non-nucleoside DNMTi have been identified so far, and even fewer have been validated in cancer. Through a process of hit-to-lead optimization, we report here the discovery of compound 5 as a potent non-nucleoside DNMTi that is also selective toward other AdoMet-dependent protein methyltransferases. Compound 5 was potent at single-digit micromolar concentrations against a panel of cancer cells and was less toxic in peripheral blood mononuclear cells than two other compounds tested. In mouse medulloblastoma stem cells, 5 inhibited cell growth, whereas related compound 2 showed high cell differentiation. To the best of our knowledge, 2 and 5 are the first non-nucleoside DNMTi tested in a cancer stem cell line.


Asunto(s)
Aminoquinolinas/síntesis química , Antineoplásicos/síntesis química , Benzamidas/síntesis química , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Células Madre Neoplásicas/efectos de los fármacos , Pirimidinas/síntesis química , Quinolinas/síntesis química , Aminoquinolinas/química , Aminoquinolinas/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Benzamidas/química , Benzamidas/farmacología , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Pirimidinas/química , Pirimidinas/farmacología , Quinolinas/química , Quinolinas/farmacología , Relación Estructura-Actividad
10.
Biochimie ; 94(11): 2264-79, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22627380

RESUMEN

Epigenetic alterations are involved in every step of carcinogenesis. The development of chromatin-modifying agents (CMAs) has provided the ability to fight cancer by reversing these alterations. Currently, four CMAs have been approved for cancer treatment; two DNA demethylating agents and two deacetylase inhibitors. A number of promising CMAs are undergoing clinical trials in several cancer types. Moreover, already approved CMAs are still under clinical investigation to improve their efficacy and to extend their use to a broader panel of cancers. Combinatorial treatments with CMAs are already considered a promising strategy to improve clinical benefits and to limit side effects. The real mechanisms by which these CMAs allow the improvement and remission of patients are still obscure. A deeper analysis of the molecular features expressed by responding patients should be performed to reveal this information. In this review, we focus on clinical trials with CMAs, discussing the success and the pitfalls of this new class of anti-cancer drugs.


Asunto(s)
Antineoplásicos/farmacología , Cromatina/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Animales , Antineoplásicos/uso terapéutico , Cromatina/genética , Cromatina/metabolismo , Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Neoplasias/enzimología
11.
Epigenomics ; 3(5): 581-609, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22126248

RESUMEN

Leukemogenesis is a multistep process in which successive transformational events enhance the ability of a clonal population arising from hematopoietic progenitor cells to proliferate, differentiate and survive. Clinically and pathologically, leukemia is subdivided into four main categories: chronic lymphocytic leukemia, chronic myeloid leukemia, acute lymphocytic leukemia and acute myeloid leukemia. Leukemia has been previously considered only as a genetic disease. However, in recent years, significant advances have been made in the elucidation of the leukemogenesis-associated processes. Thus, we have come to understand that epigenetic alterations including DNA methylation, histone modifications and miRNA are involved in the permanent changes of gene expression controlling the leukemia phenotype. In this article, we will focus on the epigenetic defects associated with leukemia and their implications as biomarkers for diagnostic, prognostic and therapeutic applications.


Asunto(s)
Metilación de ADN/fisiología , Epigénesis Genética/fisiología , Código de Histonas/fisiología , Leucemia/fisiopatología , MicroARNs/metabolismo , Modelos Biológicos , Fenotipo , Genes Supresores de Tumor/fisiología , Humanos , Leucemia/diagnóstico , Leucemia/terapia
12.
J Cell Mol Med ; 15(9): 1833-46, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21435179

RESUMEN

Histone deacetylases (HDACs) are important regulators of gene expression. Specific structural features and distinct regulative mechanisms rationalize the separation of the 18 different human HDACs into four classes. The class II comprises a heterogeneous group of nuclear and cytosolic HDACs involved in the regulation of several cellular functions, not just limited to transcriptional repression. In particular, HDAC4, 5, 7 and 9 belong to the subclass IIa and share many transcriptional partners, including members of the MEF2 family. Genetic studies in mice have disclosed the fundamental contribution of class IIa HDACs to specific developmental/differentiation pathways. In this review, we discuss about the recent literature, which hints a role of class IIa HDACs in the development, growth and aggressiveness of cancer cells.


Asunto(s)
Diferenciación Celular , Transformación Celular Neoplásica/patología , Histona Desacetilasas/metabolismo , Animales , Histona Desacetilasas/química , Humanos , Modelos Biológicos
13.
Mol Biol Cell ; 22(2): 278-89, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21118993

RESUMEN

HDAC4 (histone deacetylase 4) belongs to class IIa of histone deacetylases, which groups important regulators of gene expression, controlling pleiotropic cellular functions. Here we show that, in addition to the well-defined nuclear/cytoplasmic shuttling, HDAC4 activity is modulated by the ubiquitin-proteasome system. Serum starvation elicits the poly-ubiquitination and degradation of HDAC4 in nontransformed cells. Phosphorylation of serine 298 within the PEST1 sequence plays an important role in the control of HDAC4 stability. Serine 298 lies within a glycogen synthase kinase 3ß consensus sequence, and removal of growth factors fails to trigger HDAC4 degradation in cells deficient in this kinase. GSK3ß can phosphorylate HDAC4 in vitro, and phosphorylation of serine 302 seems to play the role of priming phosphate. We have also found that HDAC4 modulates random cell motility possibly through the regulation of KLF2 transcription. Apoptosis, autophagy, cell proliferation, and growth arrest were unaffected by HDAC4. Our data suggest a link between regulation of HDAC4 degradation and the control of cell motility as operated by growth factors.


Asunto(s)
Movimiento Celular , Glucógeno Sintasa Quinasa 3/metabolismo , Histona Desacetilasas/metabolismo , Proteínas Represoras/metabolismo , Ubiquitina/metabolismo , Animales , Línea Celular , Medio de Cultivo Libre de Suero , Inhibidores de Cisteína Proteinasa/farmacología , Glucógeno Sintasa Quinasa 3 beta , Histona Desacetilasas/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Leupeptinas/farmacología , Ratones , Mutagénesis Sitio-Dirigida , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/genética , Ubiquitinación
14.
J Cell Mol Med ; 14(4): 970-81, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20569277

RESUMEN

Curcumin is a non-toxic polyphenol with pleiotropic activities and limited bioavailability. We investigated whether a brief exposure to low doses of curcumin would induce in the myogenic C2C12 cell line an endoplasmic reticulum (ER) stress response and protect against oxidative stress. A 3-hr curcumin administration (5-10 microM) increased protein levels of the ER chaperone Grp94, without affecting those of Grp78, calreticulin and haeme-oxygenase-1 (HO-1). Exposure of cells to hydrogen peroxide 24 hrs after the curcumin treatment decreased caspase-12 activation, total protein oxidation and translocation of NF-kappaB to the nucleus, compared with untreated cells. Grp94 overexpression, achieved by means of either stable or transient trasfection, induced comparable cytoprotective effects to hydrogen peroxide. The delayed cytoprotection induced by curcumin acted through Grp94, because the curcumin-induced increase in Grp94 expression was hampered by either stable or transient transfection with antisense cDNA; in these latter cells, the extent of total protein oxidation, as well as the translocation of NF-kappaB to the nucleus, and the percentage of apoptotic cells were comparable to those observed in both curcumin-untreated wild-type and empty vector transfected cells. Defining the mechanism(s) by which Grp94 exerts its antioxidant defence, the determination of cytosolic calcium levels in C2C12 cells by fura-2 showed a significantly reduced amount of releasable calcium from intracellular stores, both in conditions of Grp94 overexpression and after curcumin pre-treatment. Therefore, a brief exposure to curcumin induces a delayed cytoprotection against oxidative stress in myogenic cells by increasing Grp94 protein level, which acts as a regulator of calcium homeostasis.


Asunto(s)
Antioxidantes/metabolismo , Curcumina/farmacología , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas de la Membrana/metabolismo , Mioblastos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Caspasa 12/metabolismo , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citoprotección/efectos de los fármacos , ADN Complementario/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Chaperón BiP del Retículo Endoplásmico , Peróxido de Hidrógeno/farmacología , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Ratones , Mioblastos/citología , Mioblastos/efectos de los fármacos , FN-kappa B/metabolismo , Oxidación-Reducción/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Transfección
15.
Mol Cancer Ther ; 8(11): 3140-50, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19887551

RESUMEN

The regulation of the necrotic death and its relevance in anticancer therapy are largely unknown. Here, we have investigated the proapoptotic and pronecrotic activities of two ubiquitin-proteasome system inhibitors: bortezomib and G5. The present study points out that the glioblastoma cell lines U87MG and T98G are useful models to study the susceptibility to apoptosis and necrosis in response to ubiquitin-proteasome system inhibitors. U87MG cells show resistance to apoptosis induced by bortezomib and G5, but they are more susceptible to necrosis induced by G5. Conversely, T98G cells are more susceptible to apoptosis induced by both inhibitors but show some resistance to G5-induced necrosis. No overt differences in the induction of Noxa and Mcl-1 or in the expression levels of other components of the apoptotic machinery were observed between U87MG and T98G cells. Instead, by comparing the transcriptional profiles of the two cell lines, we have found that the resistance to G5-induced necrosis could arise from differences in glutathione synthesis/utilization and in the microenvironment. In particular, collagen IV, which is highly expressed in T98G cells, and fibronectin, whose adhesive function is counteracted by tenascin-C in U87MG cells, can restrain the necrotic response to G5. Collectively, our results provide an initial characterization of the molecular signals governing cell death by necrosis in glioblastoma cell lines.


Asunto(s)
Ácidos Borónicos/farmacología , Caspasas/metabolismo , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasoma , Piranos/farmacología , Pirazinas/farmacología , Compuestos de Sulfhidrilo/farmacología , Ubiquitina/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Bortezomib , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Perfilación de la Expresión Génica , Glioblastoma/enzimología , Glioblastoma/genética , Glutatión/deficiencia , Glutatión/metabolismo , Humanos , Análisis por Micromatrices , Necrosis , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Ubiquitina/metabolismo
16.
Anal Chem ; 81(23): 9590-8, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19873978

RESUMEN

Recombinant fluorescent probes allow the detection of molecular events inside living cells. Many of them exploit the intracellular space to provide positional signals and, thus, require detection by single cell imaging. We describe here a novel strategy based on probes capable of encoding the spatial dimension of intracellular signals into "all-or-none" fluorescence intensity changes (differential anchorage probes, DAPs). The resulting signals can be acquired in single cells at high throughput by automated flow cytometry, (i) bypassing image acquisition and analysis, (ii) providing a direct quantitative readout, and (iii) allowing the exploration of large experimental series. We illustrate our purpose with DAPs for Bax and the effector caspases 3 and 7, which are keys players in apoptotic cell death, and show applications in basic research, high content multiplexed library screening, compound characterization, and drug profiling.


Asunto(s)
Colorantes Fluorescentes/metabolismo , Espacio Intracelular/metabolismo , Proteínas Recombinantes/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Ciclo Celular , Muerte Celular , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Proliferación Celular , Descubrimiento de Drogas , Citometría de Flujo , Ensayos Analíticos de Alto Rendimiento , Humanos , Imagen Molecular , Datos de Secuencia Molecular , Péptido Hidrolasas/metabolismo , Proteínas Recombinantes/química , Proteína X Asociada a bcl-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA