Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Biochim Biophys Acta Biomembr ; 1859(9 Pt B): 1536-1547, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28235469

RESUMEN

Heterotrimeric G proteins are peripheral membrane proteins that frequently localize to the plasma membrane where their presence in molar excess over G protein coupled receptors permits signal amplification. Their distribution is regulated by protein-lipid interactions, which has a clear influence on their activity. Gßγ dimer drives the interaction between G protein heterotrimers with cell membranes. We focused our study on the role of the C-terminal region of the Gγ2 protein in G protein interactions with cell membranes. The Gγ2 subunit is modified at cysteine (Cys) 68 by the addition of an isoprenyl lipid, which is followed by the proteolytic removal of the last three residues that leaves an isoprenylated and carboxyl methylated Cys-68 as the terminal amino acid. The role of Cys isoprenylation of the CAAX box has been defined for other proteins, yet the importance of proteolysis and carboxyl methylation of isoprenylated proteins is less clear. Here, we showed that not only geranylgeranylation but also proteolysis and carboxyl methylation are essential for the correct localization of Gγ2 in the plasma membrane. Moreover, we showed the importance of electrostatic interactions between the inner leaflet of the plasma membrane and the positively charged C-terminal domain of the Gγ2 subunit (amino acids Arg-62, Lys-64 and Lys-65) as a second signal to reach the plasma membrane. Indeed, single or multiple point mutations at Gγ2 C-terminal amino acids have a significant effect on Gγ2 protein-plasma membrane interactions and its localization to charged Ld (liquid disordered) membrane microdomains. This article is part of a Special Issue entitled: Membrane Lipid Therapy: Drugs Targeting Biomembranes edited by Pablo V. Escribá.


Asunto(s)
Membrana Celular/química , Subunidades gamma de la Proteína de Unión al GTP/química , Lípidos de la Membrana/química , Secuencia de Aminoácidos , Línea Celular Tumoral , Diterpenos/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/análisis , Humanos , Unión Proteica , Prenilación de Proteína
2.
Biochim Biophys Acta ; 1838(6): 1680-92, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24374316

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative pathology with relevant unmet therapeutic needs. Both natural aging and AD have been associated with a significant decline in the omega-3 polyunsaturated fatty acid docosahexaenoic acid (DHA), and accordingly, administration of DHA has been proposed as a possible treatment for this pathology. However, recent clinical trials in mild-to-moderately affected patients have been inconclusive regarding the real efficacy of DHA in halting this disease. Here, we show that the novel hydroxyl-derivative of DHA (2-hydroxydocosahexaenoic acid - OHDHA) has a strong therapeutic potential to treat AD. We demonstrate that OHDHA administration increases DHA levels in the brain of a transgenic mouse model of AD (5xFAD), as well as those of phosphatidylethanolamine (PE) species that carry long polyunsaturated fatty acids (PUFAs). In 5xFAD mice, administration of OHDHA induced lipid modifications that were paralleled with a reduction in amyloid-ß (Αß) accumulation and full recovery of cognitive scores. OHDHA administration also reduced Aß levels in cellular models of AD, in association with alterations in the subcellular distribution of secretases and reduced Aß-induced tau protein phosphorylation as well. Furthermore, OHDHA enhanced the survival of neuron-like differentiated cells exposed to different insults, such as oligomeric Aß and NMDA-mediated neurotoxicity. These results were supported by model membrane studies in which incorporation of OHDHA into lipid-raft-like vesicles was shown to reduce the binding affinity of oligomeric and fibrillar Aß to membranes. Finally, the OHDHA concentrations used here did not produce relevant toxicity in zebrafish embryos in vivo. In conclusion, we demonstrate the pleitropic effects of OHDHA that might prove beneficial to treat AD, which suggests that an upstream event, probably the modulation of the membrane lipid composition and structure, influences cellular homeostasis reversing the neurodegenerative process. This Article is Part of a Special Issue Entitled: Membrane Structure and Function: Relevance in the Cell's Physiology, Pathology and Therapy.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Ácidos Docosahexaenoicos/farmacología , Lípidos de la Membrana/química , Neuroblastoma/tratamiento farmacológico , Fosfolípidos/metabolismo , Esfingolípidos/metabolismo , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/fisiología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Colesterol/metabolismo , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/química , Embrión no Mamífero/citología , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Humanos , Masculino , Lípidos de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuroblastoma/metabolismo , Fosforilación/efectos de los fármacos , Presenilina-1/fisiología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Liposomas Unilamelares/metabolismo , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas tau/genética , Proteínas tau/metabolismo
3.
Apoptosis ; 20(5): 712-24, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25663172

RESUMEN

The unfolded protein response (UPR) and autophagy are two cellular processes involved in the clearing of intracellular misfolded proteins. Both pathways are targets for molecules that may serve as treatments for several diseases, including neurodegenerative disorders like Alzheimer's disease (AD). In the present work, we show that 2-hydroxy-DHA (HDHA), a docosahexaenoic acid (DHA) derivate that restores cognitive function in a transgenic mouse model of AD, modulates UPR and autophagy in differentiated neuron-like SH-SY5Y cells. Mild therapeutic HDHA exposure induced UPR activation, characterized by the up-regulation of the molecular chaperone Bip as well as PERK-mediated stimulation of eIF2α phosphorylation. Key proteins involved in initiating autophagy, such as beclin-1, and several Atg proteins involved in autophagosome maturation (Atg3, Atg5, Atg12 and Atg7), were also up-regulated on exposure to HDHA. Moreover, when HDHA-mediated autophagy was studied after amyloid-ß peptide (Aß) stimulation to mimic the neurotoxic environment of AD, it was associated with increased cell survival, suggesting that HDHA driven modulation of this process at least in part mediates the neuroprotective effects of this new anti-neurodegenerative drug. The present results in part explain the pharmacological effects of HDHA inducing full recovery of the cognitive scores in murine models of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Hidroxiácidos/farmacología , Fármacos Neuroprotectores/farmacología , Respuesta de Proteína Desplegada , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Apoptosis , Autofagia , Línea Celular Tumoral , Supervivencia Celular , Evaluación Preclínica de Medicamentos , Humanos , Hidroxiácidos/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico
4.
Proc Natl Acad Sci U S A ; 109(22): 8489-94, 2012 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-22586083

RESUMEN

Despite recent advances in the development of new cancer therapies, the treatment options for glioma remain limited, and the survival rate of patients has changed little over the past three decades. Here, we show that 2-hydroxyoleic acid (2OHOA) induces differentiation and autophagy of human glioma cells. Compared to the current reference drug for this condition, temozolomide (TMZ), 2OHOA combated glioma more efficiently and, unlike TMZ, tumor relapse was not observed following 2OHOA treatment. The novel mechanism of action of 2OHOA is associated with important changes in membrane-lipid composition, primarily a recovery of sphingomyelin (SM) levels, which is markedly low in glioma cells before treatment. Parallel to membrane-lipid regulation, treatment with 2OHOA induced a dramatic translocation of Ras from the membrane to the cytoplasm, which inhibited the MAP kinase pathway, reduced activity of the PI3K/Akt pathway, and downregulated Cyclin D-CDK4/6 proteins followed by hypophosphorylation of the retinoblastoma protein (RB). These regulatory effects were associated with induction of glioma cell differentiation into mature glial cells followed by autophagic cell death. Given its high efficacy, low toxicity, ease of oral administration, and good distribution to the brain, 2OHOA constitutes a new and potentially valuable therapeutic tool for glioma patients.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Glioma/tratamiento farmacológico , Ácidos Oléicos/farmacología , Animales , Antineoplásicos/metabolismo , Línea Celular Tumoral , Membrana Celular/química , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Relación Dosis-Respuesta a Droga , Glioma/metabolismo , Glioma/patología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Lípidos de la Membrana/química , Lípidos de la Membrana/metabolismo , Ratones , Ratones Desnudos , Microscopía Confocal , Ácidos Oléicos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Temozolomida , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas ras/metabolismo
5.
Proc Natl Acad Sci U S A ; 108(49): 19569-74, 2011 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-22106271

RESUMEN

The mechanism of action of 2-hydroxyoleic acid (2OHOA), a potent antitumor compound, has not yet been fully elucidated. Here, we show that human cancer cells have markedly lower levels of sphingomyelin (SM) than nontumor (MRC-5) cells. In this context, 2OHOA treatment strongly augments SM mass (4.6-fold), restoring the levels found in MRC-5 cells, while a loss of phosphatidylethanolamine and phosphatidylcholine is observed (57 and 30%, respectively). The increased SM mass was due to a rapid and highly specific activation of SM synthases (SMS). This effect appeared to be specific against cancer cells as it did not affect nontumor MRC-5 cells. Therefore, low SM levels are associated with the tumorigenic transformation that produces cancer cells. SM accumulation occurred at the plasma membrane and caused an increase in membrane global order and lipid raft packing in model membranes. These modifications would account for the observed alteration by 2OHOA in the localization of proteins involved in cell apoptosis (Fas receptor) or differentiation (Ras). Importantly, SMS inhibition by D609 diminished 2OHOA effect on cell cycle. Therefore, we propose that the regulation of SMS activity in tumor cells is a critical upstream event in 2OHOA antitumor mechanism, which also explains its specificity for cancer cells, its potency, and the lack of undesired side effects. Finally, the specific activation of SMS explains the ability of this compound to trigger cell cycle arrest, cell differentiation, and autophagy or apoptosis in cancer cells.


Asunto(s)
Transformación Celular Neoplásica , Ácidos Oléicos/farmacología , Esfingomielinas/metabolismo , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo , Apoptosis/efectos de los fármacos , Hidrocarburos Aromáticos con Puentes/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Células , Regulación Enzimológica de la Expresión Génica , Glioma/genética , Glioma/metabolismo , Glioma/patología , Humanos , Immunoblotting , Células Jurkat , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Norbornanos , Inhibidores de Fosfodiesterasa/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tiocarbamatos , Tionas/farmacología , Transferasas (Grupos de Otros Fosfatos Sustitutos)/antagonistas & inhibidores , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética , Receptor fas/metabolismo , Proteínas ras/metabolismo
6.
PLoS One ; 7(10): e48235, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23133576

RESUMEN

BACKGROUND: 2-Hydroxyoleic acid is a synthetic fatty acid with potent anti-cancer activity which does not induce undesired side effects. However, the molecular and cellular mechanisms by which this compound selectively kills human glioma cancer cells without killing normal cells is not fully understood. The present study was designed to determine the molecular bases underlying the potency against 1321N1, SF-767 and U118 human glioma cell lines growth without affecting non cancer MRC-5 cells. METHODOLOGY/PRINCIPAL FINDINGS: The cellular levels of endoplasmic reticulum (ER) stress, unfolded protein response (UPR) and autophagy markers were determined by quantitative RT-PCR and immunoblotting on 1321N1, SF-767 and U118 human glioma cells and non-tumor MRC-5 cells incubated in the presence or absence of 2OHOA or the ER stress/autophagy inducer, palmitate. The cellular response to these agents was evaluated by fluorescence microscopy, electron microscopy and flow cytometry. We have observed that 2OHOA treatments induced augments in the expression of important ER stress/UPR markers, such as phosphorylated eIF2α, IRE1α, CHOP, ATF4 and the spliced form of XBP1 in human glioma cells. Concomitantly, 2OHOA led to the arrest of 1321N1 cells in the G(2)/M phase of the cell cycle, with down-regulation of cyclin B1 and Cdk1/Cdc2 proteins in the three glioma cell lines studied. Finally, 2OHOA induced autophagy in 1321N1, SF-767 and U118 cells, with the appearance of autophagic vesicles and the up-regulation of LC3BI, LC3BII and ATG7 in 1321N1 cells, increases of LC3BI, LC3BII and ATG5 in SF-767 cells and up-regulation of LC3BI and LC3BII in U118 cells. Importantly, 2OHOA failed to induce such changes in non-tumor MRC-5 cells. CONCLUSION/SIGNIFICANCE: The present results demonstrate that 2OHOA induces ER stress/UPR and autophagy in human glioma (1321N1, SF-767 and U118 cell lines) but not normal (MRC-5) cells, unraveling the molecular bases underlying the efficacy and lack of toxicity of this compound.


Asunto(s)
Neoplasias Encefálicas/patología , Retículo Endoplásmico/metabolismo , Glioma/patología , Ácidos Oléicos/metabolismo , Antineoplásicos/farmacología , Autofagia , Neoplasias Encefálicas/metabolismo , Línea Celular , Línea Celular Tumoral , Supervivencia Celular , Ácidos Grasos/química , Fibroblastos/metabolismo , Glioma/metabolismo , Humanos , Pulmón/patología , Transducción de Señal , Sales de Tetrazolio/farmacología , Tiazoles/farmacología
7.
Autophagy ; 8(10): 1542-4, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22892762

RESUMEN

The very high mortality rate of gliomas reflects the unmet therapeutic need associated with this type of brain tumor. We have discovered that the plasma membrane fulfills a critical role in the propagation of tumorigenic signals, whereby changes in membrane lipid content can either activate or silence relevant pathways. We have designed a synthetic fatty acid, 2-hydroxyoleic acid (2OHOA), that specifically activates sphingomyelin synthase (SGMS), thereby modifying the lipid content of cancer cell membranes and restoring lipid levels to those found in normal cells. In reverting, the structure of the membrane by activating SGMS, 2OHOA inhibits the RAS-MAPK pathway, which in turn fails to activate the CCND (Cyclin D)-CDK4/CDK6 and PI3K-AKT1 pathways. The overall result in SF767 cancer cells, a line that is resistant to apoptosis, is the sequential induction of cell cycle arrest, cell differentiation and autophagy. Such effects are not observed in normal cells (MRC-5) and thus, this specific activation of programmed cell death infers greater efficacy and lower toxicity to 2OHOA than that associated with temozolomide (TMZ), the reference drug for the treatment of glioma.


Asunto(s)
Autofagia/efectos de los fármacos , Glioma/patología , Ácidos Oléicos/farmacología , Esfingomielinas/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Modelos Biológicos , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA