Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Exp Neurol ; 256: 7-16, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24681156

RESUMEN

Neuroinflammation is a component of secondary injury following traumatic brain injury (TBI) that can persist beyond the acute phase. Leukotrienes are potent, pro-inflammatory lipid mediators generated from membrane phospholipids. In the absence of injury, leukotrienes are undetectable in the brain, but after trauma they are rapidly synthesized by a transcellular event involving infiltrating neutrophils and endogenous brain cells. Here, we investigate the efficacy of MK-886, an inhibitor of 5-lipoxygenase activating protein (FLAP), in blocking leukotriene synthesis, secondary brain damage, synaptic dysfunction, and cognitive impairments after TBI. Male Sprague Dawley rats (9-11weeks) received either MK-886 or vehicle after they were subjected to unilateral moderate fluid percussion injury (FPI) to assess the potential clinical use of FLAP inhibitors for TBI. MK-886 was also administered before FPI to determine the preventative potential of FLAP inhibitors. MK-886 given before or after injury significantly blocked the production of leukotrienes, measured by reverse-phase liquid chromatography coupled to tandem mass spectrometry (RP LC-MS/MS), and brain edema, measured by T2-weighted magnetic resonance imaging (MRI). MK-886 significantly attenuated blood-brain barrier disruption in the CA1 hippocampal region and deficits in long-term potentiation (LTP) at CA1 hippocampal synapses. The prevention of FPI-induced synaptic dysfunction by MK-886 was accompanied by fewer deficits in post-injury spatial learning and memory performance in the radial arm water maze (RAWM). These results indicate that leukotrienes contribute significantly to secondary brain injury and subsequent cognitive deficits. FLAP inhibitors represent a novel anti-inflammatory approach for treating human TBI that is feasible for both intervention and prevention of brain injury and neurologic deficits.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Trastornos del Conocimiento/tratamiento farmacológico , Indoles/uso terapéutico , Leucotrienos/biosíntesis , Inhibidores de la Lipooxigenasa/uso terapéutico , Animales , Encéfalo/efectos de los fármacos , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/psicología , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/psicología , Hipocampo/efectos de los fármacos , Indoles/farmacología , Inhibidores de la Lipooxigenasa/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem
2.
J Trauma ; 71(5): 1211-8, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21427623

RESUMEN

BACKGROUND: Enzymatic and nonenzymatic oxidation of polyunsaturated fatty acids leads to the formation of biologically active products known as lipid mediators. In the brain, lipid mediators play an important role in supporting homeostasis and normal function. Thus, levels of these metabolites in normal and pathologic conditions in the brain are particularly relevant in understanding the transition to disease. METHODS: In this study, liquid chromatography tandem mass spectrometry was used to analyze lipid mediators in cerebrospinal fluid (CSF) of controls and traumatic brain injured (TBI) patients. RESULTS: Our results showed that the levels of arachidonic acid (AA), docosahexaenoic acid (DHA), 5- and 12- eicosatetraenoic acid (HETE) were significantly increased in the CSF of TBI patients. The magnitude of increase was 10-fold for AA, DHA, and 5-HETE and 17-fold for 12-HETE. Prostaglandins and leukotrienes were not detected in CSF of either control or brain injured patients. Furthermore, this study found that isoprostanes and thromboxanes are present in CSF of brain injured patients. CONCLUSIONS: This study clearly shows that certain lipid mediators accumulate in the CSF of TBI patient. This study also suggests the potential use of DHA, AA, 5- and 12-HETE as biochemical markers of brain injury and to monitor the impact of interventions.


Asunto(s)
Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/líquido cefalorraquídeo , Ácido Araquidónico/líquido cefalorraquídeo , Lesiones Encefálicas/líquido cefalorraquídeo , Ácidos Docosahexaenoicos/líquido cefalorraquídeo , Ácidos Hidroxieicosatetraenoicos/líquido cefalorraquídeo , Adulto , Biomarcadores/líquido cefalorraquídeo , Estudios de Casos y Controles , Cromatografía Liquida , Femenino , Humanos , Masculino , Persona de Mediana Edad , Espectrometría de Masas en Tándem
3.
Neurosci Lett ; 488(2): 112-7, 2011 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-20849920

RESUMEN

Restoration of autophagy represents a potential therapeutic target for neurodegenerative disorders, but factors that regulate autophagic flux are largely unknown. When deprived of trophic factors, cultured Purkinje neurons die by an autophagy associated cell death mechanism. The accumulation of autophagic vesicles and cell death of Purkinje neurons is inhibited by insulin-like growth factor, by a mechanism that enhances autophagic vesicle turnover. In this report, we identify Rab7 as an IGF-I regulated target during neuronal autophagy. Purkinje neurons transfected with EGFP-Rab7-WT and constitutively active EGFP-Rab7-Q67L contained few RFP-LC3 positive autophagosomes and little co-localization with GFP-Rab7 under control conditions. Upon induction of autophagy, RFP-LC3 positive autophagosomes increased and co-localized with GFP-Rab7. Conversely, expression of the dominant negative mutant EGFP-Rab7-T22N increased the accumulation of autophagosomes under control conditions, which accumulated even further during trophic factor withdrawal. There was no vesicular co-localization between Rab7-T22N and RFP-LC3 under control or trophic factor withdrawal conditions. During prolonged trophic factor withdrawal, a condition that leads to the accumulation of autophagic vesicles and cell death, Rab7 activity decreased significantly. IGF-I, added at the time of trophic factor withdrawal, prevented the deactivation of Rab7 and increased the interaction of Rab7 with its interacting protein (RILP), restoring autophagic flux. These results provide a novel mechanism by which IGF-I regulates autophagic flux during neuronal stress.


Asunto(s)
Autofagia/fisiología , Proteínas Portadoras/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células de Purkinje/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Western Blotting , Microscopía Fluorescente , Fagosomas/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas de Unión a GTP rab7
4.
J Neurotrauma ; 26(11): 1977-86, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19886806

RESUMEN

The leukotrienes belong to a family of biologically active lipids derived from arachidonate that are often involved in inflammatory responses. In the central nervous system, a group of leukotrienes, known as the cysteinyl leukotrienes, is generated in brain tissue in response to a variety of acute brain injuries. Although the exact clinical significance of this excess production remains unclear, the cysteinyl leukotrienes may contribute to injury-related disruption of the brain-blood barrier and exacerbate secondary injury processes. In the present study, the formation and role of cysteinyl leukotrienes was explored in the fluid percussion injury model of traumatic brain injury in rats. The results showed that levels of the cysteinyl leukotrienes were elevated after fluid percussion injury with a maximal formation 1 hour after the injury. Neutrophils contributed to cysteinyl leukotriene formation in the injured brain hemisphere, potentially through a transcellular biosynthetic mechanism. Furthermore, pharmacological reduction of cysteinyl leukotriene formation after the injury, using MK-886, resulted in reduction of brain lesion volumes, suggesting that the cysteinyl leukotrienes play an important role in traumatic brain injury.


Asunto(s)
Lesiones Encefálicas/enzimología , Lesiones Encefálicas/patología , Cisteína/biosíntesis , Leucotrienos/biosíntesis , Animales , Cromatografía Liquida , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Indoles/farmacología , Leucotrieno B4/biosíntesis , Leucotrieno C4/biosíntesis , Leucotrieno D4/biosíntesis , Leucotrieno E4/biosíntesis , Masculino , Espectrometría de Masas , Neutrófilos/metabolismo , Ratas , Ratas Sprague-Dawley
5.
J Biol Chem ; 284(30): 20398-407, 2009 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-19509289

RESUMEN

Continuous macroautophagic activity is critical for the maintenance of neuronal homeostasis; however, unchecked or dysregulated autophagy can lead to cell death. Cultured Purkinje neurons die by an autophagy-associated cell death mechanism when deprived of trophic support. Here, we report that insulin-like growth factor-I (IGF-I) completely blocked the autophagy-associated cell death of Purkinje neurons. To examine the mechanism by which IGF-I influences autophagy, neurons were infected with adeno-RFP-LC3 and subjected to trophic factor withdrawal, and the size and number of autophagosomes were analyzed by live-cell fluorescence imaging. In control neurons, autophagy occurred at a constitutive low level with most autophagosomes measuring less than 0.75 microm. Trophic factor withdrawal increased the number and size of autophagosomes with most autophagosomes ranging between 0.75 and 1.5 microm and some reaching 1.5-2.25 microm. IGF-I added at the time of trophic factor withdrawal prevented the accumulation of the larger autophagosomes; however, it had no effect on the conversion of LC3, an indicator of autophagy induction. Instead, the rate of autophagosome-to-lysosome fusion measured by colocalization of RFP-LC3 and LysoSensor Green was accelerated by IGF-I. Treating the neurons with bafilomycin A(1) in the presence of IGF-I led to the accumulation of autophagosomes even larger than those induced by trophic factor withdrawal alone, indicating that IGF-I regulates autophagic vesicle turnover. Finally, the effect of IGF-I on autophagy was mediated by an Akt/mTOR-de pend ent and an ERK-independent pathway. These data suggest a novel role for IGF-I in protecting Purkinje neurons from autophagy-associated cell death by increasing autophagy efficiency downstream of autophagy induction.


Asunto(s)
Autofagia , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células de Purkinje/citología , Células de Purkinje/metabolismo , Animales , Muerte Celular , Regulación de la Expresión Génica , Lisosomas/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Serina-Treonina Quinasas TOR , Vacuolas/metabolismo
6.
Methods Enzymol ; 453: 145-58, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19216905

RESUMEN

The discovery that impaired autophagy is linked to a wide variety of prominent diseases including cancer and neurodegeneration has led to an explosion of research in this area. Methodologies that allow investigators to observe and quantify the autophagic process will clearly advance knowledge of how this process contributes to the pathophysiology of many clinical disorders. The recent identification of essential autophagy genes in higher eukaryotes has made it possible to analyze autophagy in mammalian cells that express autophagy proteins tagged with fluorescent markers. This chapter describes such methods using primary cultured neurons that undergo up-regulation of autophagy when trophic factors are removed from their medium. The prolonged up-regulated autophagy, in turn, contributes to the death of these neurons, thus providing a model to examine the relationship between enhanced autophagy and cell death. Neurons are isolated from the cerebellum of postnatal day 7 rat pups and cultured in the presence of trophic factors and depolarizing concentrations of potassium. Once established, the neurons are transfected with an adeno-viral vector expressing MAP1-LC3 with red fluorescent protein (RFP). MAP1-LC3 is the mammalian homolog of the yeast autophagosomal marker Atg8 and when tagged to GFP or RFP, it is the most widely used marker for autophagosomes. Once expression is stable, autophagy is induced by removing trophic factors. At various time points after inducing autophagy, the neurons are stained with LysoSensor Green (a pH-dependent lysosome marker) and Hoechst (a DNA marker) and subjected to live-cell imaging. In some cases, time-lapse imaging is used to examine the stepwise process of autophagy in live neurons.


Asunto(s)
Autofagia/fisiología , Lisosomas/metabolismo , Neuronas/metabolismo , Fagosomas/metabolismo , Animales , Células Cultivadas , Neuronas/citología , Ratas
7.
J Lipid Res ; 49(9): 1990-2000, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18503030

RESUMEN

Inflammatory lipid mediators derived from arachidonic acid (AA) and docosahexaenoic acid (DHA) modify the pathophysiology of brain ischemia. The goal of this work was to investigate the formation of eicosanoids and docosanoids generated from AA and DHA, respectively, during no-flow cerebral ischemia. Rats were subjected to head-focused microwave irradiation 5 min following decapitation (complete ischemia) or prior to decapitation (controls). Brain lipids were extracted and analyzed by reverse-phase liquid chromatography-tandem mass spectrometry. After complete ischemia, brain AA, DHA, and docosapentaenoic acid concentrations increased 18-, 5- and 4-fold compared with controls, respectively. Prostaglandin E(2) (PGE(2)) and PGD(2) could not be detected in control microwaved rat brain, suggesting little endogenous PGE(2)/D(2) production in the brain in the absence of experimental manipulation. Concentrations of thromboxane B(2), E(2)/D(2)-isoprostanes, 5-hydroxyeicosatetraenoic acid (5-HETE), 5-oxo-eicosatetraenoic acid, and 12-HETE were significantly elevated in ischemic brains. In addition, DHA products such as mono-, di- and trihydroxy-DHA were detected in control and ischemic brains. Monohydroxy-DHA, identified as 17-hydroxy-DHA and thought to be the immediate precursor of neuroprotectin D(1), was 6.5-fold higher in ischemic than in control brain. The present study demonstrated increased formation of eicosanoids, E(2)/D(2)-IsoPs, and docosanoids following cerebral ischemia. A balance of these lipid mediators may mediate immediate events of ischemic injury and recovery.


Asunto(s)
Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Encéfalo/efectos de la radiación , Dinoprostona/biosíntesis , Ácidos Docosahexaenoicos/metabolismo , Eicosanoides/biosíntesis , Isoprostanos/biosíntesis , Prostaglandina D2/biosíntesis , Animales , Química Encefálica , Decapitación/metabolismo , Masculino , Microondas , Ratas , Ratas Endogámicas F344
8.
J Neurochem ; 103(4): 1310-8, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17711426

RESUMEN

Leukotrienes are mediators of inflammation that belong to a family of lipids derived from arachidonic acid by the action of 5-lipoxygenase. Leukotrienes have been detected in the central nervous system in association with different pathological events, but little is known about their biosynthesis or function in the brain. When rat neurons and glial cells in primary culture were stimulated with the calcium ionophore, no significant biosynthesis of leukotrienes was detected using liquid chromatography/mass spectrometry (LC/MS) techniques. However, when exogenous LTA(4) was added to these cultured cells, both neurons and glia were able to synthesize LTC(4). Activated neutrophils are known to supply LTA(4) to other cells for transcellular biosynthesis of cysteinyl-leukotrienes. Since neutrophils can infiltrate brain tissue after stroke or traumatic brain injury, we examined whether neutrophils play a similar role in the central nervous system. When peripheral blood neutrophils were co-cultured with rat neurons, glia cells, and then stimulated with calcium ionophore, a robust production of LTC(4), LTD(4), and LTE(4) was observed, revealing that neurons and glia can participate in the transcellular mechanism of leukotriene biosynthesis. The formation of LTC(4) through this mechanism may be relevant in the genesis and progression of the inflammatory response as a result of brain injury.


Asunto(s)
Membrana Celular/metabolismo , Cisteína/biosíntesis , Leucotrienos/biosíntesis , Neuroglía/metabolismo , Neuronas/metabolismo , Animales , Membrana Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Humanos , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Leucotrieno C4/biosíntesis , Ratones , Ratones Endogámicos C57BL , Neuroglía/citología , Neuroglía/fisiología , Neuronas/citología , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley
9.
Endocrinology ; 148(6): 2806-14, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17332061

RESUMEN

GnRH neurons migrate into the hypothalamus during development. Although migratory defects may result in disordered activation of the reproductive axis and lead to delayed or absent sexual maturation, specific factors regulating GnRH neuronal migration remain largely unknown. The receptor tyrosine kinase, adhesion-related kinase (Ark) (also known as Axl, UFO, and Tyro7), has been implicated in the migration of GnRH neuronal cells. Binding of its ligand, growth arrest-specific gene 6 (Gas6), promotes cytoskeletal remodeling and migration of NLT GnRH neuronal cells via Rac and p38 MAPK. Here, we examined the Axl effectors proximal to Rac in the signaling pathway. Gas6/Axl-induced lamellipodia formation and migration were blocked after phosphatidylinositol-3-kinase (PI3K) inhibition in GnRH neuronal cells. The p85 subunit of PI3K coimmunoprecipitated with Axl and was phosphorylated in a Gas6-sensitive manner. In addition, PI3K inhibition in GnRH neuronal cells diminished Gas6-induced Rac activation. Exogenous expression of a dominant-negative form of Ras also decreased GnRH neuronal lamellipodia formation, migration, and Rac activation. PI3K inhibition blocked Ras in addition to Rac activation and migration. In contrast, pharmacological blockade of the phospholipase C gamma effectors, protein kinase C or calcium/calmodulin protein kinase II, had no effect on Gas6/Axl signaling to promote Rac activation or stimulate cytoskeletal reorganization and migration. Together, these data show that the PI3K-Ras pathway is a major mediator of Axl actions upstream of Rac to induce GnRH neuronal cell migration.


Asunto(s)
Movimiento Celular/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Neuronas/metabolismo , Proteína Oncogénica p21(ras)/fisiología , Proteínas Oncogénicas/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Tirosina Quinasas Receptoras/fisiología , Animales , Línea Celular Transformada , Citoesqueleto/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Ratones , Modelos Biológicos , Proteínas Oncogénicas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras/metabolismo , Tirosina Quinasa del Receptor Axl
10.
J Biol Chem ; 281(15): 10105-17, 2006 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-16492665

RESUMEN

FoxO transcription factors are important targets of insulin action. To better understand the role of FoxO proteins in the liver, we created transgenic mice expressing constitutively active FoxO1 in the liver using the alpha1-antitrypsin promoter. Fasting glucose levels are increased, and glucose tolerance is impaired in transgenic (TGN) versus wild type (WT) mice. Interestingly, fasting triglyceride and cholesterol levels are reduced despite hyperinsulinemia, and post-prandial changes in triglyceride levels are markedly suppressed in TGN versus WT mice. Activation of pro-lipogenic signaling pathways (atypical protein kinase C and protein kinase B) and the ability to suppress beta-hydroxybutyrate levels are not impaired in TGN. In contrast, de novo lipogenesis measured with (3)H(2)O is suppressed by approximately 70% in the liver of TGN versus WT mice after refeeding. Gene-array studies reveal that the expression of genes involved in gluconeogenesis, glycerol transport, and amino acid catabolism is increased, whereas genes involved in glucose utilization by glycolysis, the pentose phosphate shunt, lipogenesis, and sterol synthesis pathways are suppressed in TGN versus WT. Studies with adenoviral vectors in isolated hepatocytes confirm that FoxO1 stimulates expression of gluconeogenic genes and suppresses expression of genes involved in glycolysis, the shunt pathway, and lipogenesis, including glucokinase and SREBP-1c. Together, these results indicate that FoxO proteins promote hepatic glucose production through multiple mechanisms and contribute to the regulation of other metabolic pathways important in the adaptation to fasting and feeding in the liver, including glycolysis, the pentose phosphate shunt, and lipogenic and sterol synthetic pathways.


Asunto(s)
Factores de Transcripción Forkhead/fisiología , Regulación de la Expresión Génica , Hígado/enzimología , Adenoviridae/genética , Animales , Bioquímica/métodos , Glucemia/metabolismo , Cromatografía Líquida de Alta Presión , ADN Complementario/metabolismo , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Genoma , Gluconeogénesis , Glucosa/metabolismo , Glicerol/metabolismo , Glucólisis , Hepatocitos/metabolismo , Humanos , Inmunohistoquímica , Inmunoprecipitación , Insulina/metabolismo , Lípidos/química , Lipogénesis , Lipoproteína Lipasa/metabolismo , Hígado/metabolismo , Ratones , Ratones Transgénicos , Modelos Biológicos , Hibridación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transcripción Genética , Triglicéridos/metabolismo , alfa 1-Antitripsina/genética
11.
J Neurochem ; 94(4): 1025-39, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16092944

RESUMEN

Rho GTPases are key transducers of integrin/extracellular matrix and growth factor signaling. Although integrin-mediated adhesion and trophic support suppress neuronal apoptosis, the role of Rho GTPases in neuronal survival is unclear. Here, we have identified Rac as a critical pro-survival GTPase in cerebellar granule neurons (CGNs) and elucidated a death pathway triggered by its inactivation. GTP-loading of Rac1 was maintained in CGNs by integrin-mediated (RGD-dependent) cell attachment and trophic support. Clostridium difficile toxin B (ToxB), a specific Rho family inhibitor, induced a selective caspase-mediated degradation of Rac1 without affecting RhoA or Cdc42 protein levels. Both ToxB and dominant-negative N17Rac1 elicited CGN apoptosis, characterized by cytochrome c release and activation of caspase-9 and -3, whereas dominant-negative N19RhoA or N17Cdc42 did not cause significant cell death. ToxB stimulated mitochondrial translocation and conformational activation of Bax, c-Jun activation, and induction of the BH3-only protein Bim. Similarly, c-Jun activation and Bim induction were observed with N17Rac1. A c-jun N-terminal protein kinase (JNK)/p38 inhibitor, SB203580, and a JNK-specific inhibitor, SP600125, significantly decreased ToxB-induced Bim expression and blunted each subsequent step of the apoptotic cascade. These results indicate that Rac acts downstream of integrins and growth factors to promote neuronal survival by repressing c-Jun/Bim-mediated mitochondrial apoptosis.


Asunto(s)
Apoptosis/fisiología , Proteínas Portadoras/fisiología , Cerebelo/fisiología , Proteínas de la Membrana/fisiología , Mitocondrias/fisiología , Proteínas Proto-Oncogénicas c-jun/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteína de Unión al GTP rac1/fisiología , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis , Proteínas Bacterianas/farmacología , Toxinas Bacterianas/farmacología , Proteína 11 Similar a Bcl2 , Caspasas/metabolismo , Adhesión Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Citocromos c/metabolismo , Activación Enzimática/efectos de los fármacos , Genes Dominantes , Integrinas/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/farmacología , Proteínas de Unión al GTP rho/antagonistas & inhibidores
12.
J Neurochem ; 94(1): 22-36, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15953346

RESUMEN

Primary cerebellar granule neurons (CGNs) require depolarizing extracellular potassium for their survival. Removal of depolarizing potassium triggers CGN apoptosis that requires induction of Bim, a BH3-only Bcl-2 family member. Bim is classically thought to promote apoptosis by neutralizing pro-survival Bcl-2 proteins. To determine if this is the principal function of Bim in CGNs, we contrasted Bim-mediated apoptosis to neuronal death induced by HA14-1, a BH3-domain mimetic that antagonizes Bcl-2 and Bcl-x(L). HA14-1 elicited CGN apoptosis characterized by caspase 3 and 9 activation, cytochrome c release, conformational activation of Bax, and mitochondrial depolarization. HA14-1 provoked CGN apoptosis in the absence of Bim induction and negative regulators of Bim transcription did not prevent HA14-1-induced cell death. However, the antioxidant glutathione and its precursor, N-acetyl-l-cysteine, suppressed HA14-1-induced apoptosis. Similarly, apoptosis induced by either a structurally distinct Bcl-2/Bcl-x(L) inhibitor (compound 6) or Bcl-2 antisense oligonucleotides was diminished by glutathione. In contrast, antioxidants had no effect on CGN apoptosis provoked by either removal of depolarizing potassium or overexpression of a GFP-Bim fusion protein, two models of Bim-dependent death. These data show that antagonism of Bcl-2/Bcl-x(L) function elicits oxidative stress-dependent CGN apoptosis that is mechanistically distinct from Bim-mediated cell death. These results further indicate that, although Bcl-2/Bcl-x(L) antagonism is sufficient to induce neuronal apoptosis, Bim likely promotes neuronal death by interacting with additional proteins besides Bcl-2/Bcl-x(L).


Asunto(s)
Apoptosis/fisiología , Proteínas Portadoras/biosíntesis , Proteínas de la Membrana/biosíntesis , Neuronas/citología , Neuronas/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Proteínas Proto-Oncogénicas/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis , Proteína 11 Similar a Bcl2 , Benzopiranos/farmacología , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Recuento de Células , Células Cultivadas , Relación Dosis-Respuesta a Droga , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Neuronas/efectos de los fármacos , Nitrilos/farmacología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Ratas , Ratas Sprague-Dawley , Proteína bcl-X
13.
J Neurosci ; 24(44): 9993-10002, 2004 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-15525785

RESUMEN

Glycogen synthase kinase-3beta (GSK-3beta) is a critical activator of neuronal apoptosis induced by a diverse array of neurotoxic insults. However, the downstream substrates of GSK-3beta that ultimately induce neuronal death are unknown. Here, we show that GSK-3beta phosphorylates and regulates the activity of Bax, a pro-apoptotic Bcl-2 family member that stimulates the intrinsic (mitochondrial) death pathway by eliciting cytochrome c release from mitochondria. In cerebellar granule neurons undergoing apoptosis, inhibition of GSK-3beta suppressed both the mitochondrial translocation of an expressed green fluorescent protein (GFP)-Bax(alpha) fusion protein and the conformational activation of endogenous Bax. GSK-3beta directly phosphorylated Bax(alpha) on Ser163, a residue found within a species-conserved, putative GSK-3beta phosphorylation motif. Coexpression of GFP-Bax(alpha) with a constitutively active mutant of GSK-3beta, GSK-3beta(Ser9Ala), enhanced the in vivo phosphorylation of wild-type Bax(alpha), but not a Ser163Ala mutant of Bax(alpha), in transfected human embryonic kidney 293 (HEK293) cells. Moreover, cotransfection with constitutively active GSK-3beta promoted the localization of Bax(alpha) to mitochondria and induced apoptosis in both transfected HEK293 cells and cerebellar granule neurons. In contrast, neither a Ser163Ala point mutant of Bax(alpha) nor a naturally occurring splice variant that lacks 13 amino acids encompassing Ser163 (Bax(sigma)) were driven to mitochondria in HEK293 cells coexpressing constitutively active GSK-3beta. In a similar manner, either mutation or deletion of the identified GSK-3beta phosphorylation motif prevented the localization of Bax to mitochondria in cerebellar granule neurons undergoing apoptosis. Our results indicate that GSK-3beta exerts some of its pro-apoptotic effects in neurons by regulating the mitochondrial localization of Bax, a key component of the intrinsic apoptotic cascade.


Asunto(s)
Apoptosis/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Mitocondrias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Cerebelo/citología , Secuencia Conservada , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Datos de Secuencia Molecular , Neuronas/metabolismo , Neuronas/ultraestructura , Fosforilación , Conformación Proteica , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/metabolismo , Serina , Proteína X Asociada a bcl-2
14.
J Neurosci ; 24(19): 4498-509, 2004 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-15140920

RESUMEN

The cellular mechanisms underlying Purkinje neuron death in various neurodegenerative disorders of the cerebellum are poorly understood. Here we investigate an in vitro model of cerebellar neuronal death. We report that cerebellar Purkinje neurons, deprived of trophic factors, die by a form of programmed cell death distinct from the apoptotic death of neighboring granule neurons. Purkinje neuron death was characterized by excessive autophagic-lysosomal vacuolation. Autophagy and death of Purkinje neurons were inhibited by nerve growth factor (NGF) and were activated by NGF-neutralizing antibodies. Although treatment with antisense oligonucleotides to the p75 neurotrophin receptor (p75ntr) decreased basal survival of cultured cerebellar neurons, p75ntr-antisense decreased autophagy and completely inhibited death of Purkinje neurons induced by trophic factor withdrawal. Moreover, adenoviral expression of a p75ntr mutant lacking the ligand-binding domain induced vacuolation and death of Purkinje neurons. These results suggest that p75ntr is required for Purkinje neuron survival in the presence of trophic support; however, during trophic factor withdrawal, p75ntr contributes to Purkinje neuron autophagy and death. The autophagic morphology resembles that found in neurodegenerative disorders, suggesting a potential role for this pathway in neurological disease.


Asunto(s)
Adenina/análogos & derivados , Autofagia/fisiología , Cerebelo/citología , Células de Purkinje/metabolismo , Receptores de Factor de Crecimiento Nervioso/fisiología , Adenina/farmacología , Animales , Animales Recién Nacidos , Anticuerpos/farmacología , Autofagia/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Lisosomas/metabolismo , Lisosomas/patología , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Factor de Crecimiento Nervioso/farmacología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Fármacos Neuroprotectores/farmacología , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Células de Purkinje/efectos de los fármacos , Células de Purkinje/patología , Ratas , Ratas Sprague-Dawley , Receptor de Factor de Crecimiento Nervioso , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo , Vacuolas/efectos de los fármacos , Vacuolas/patología
15.
Mol Neurobiol ; 29(2): 155-66, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15126683

RESUMEN

Myocyte enhancer factor-2 (MEF2) transcription factors regulate genes that control critical cellular processes including proliferation, differentiation, and survival. Although MEF2 proteins were first identified as transcription factors that bound A/T rich DNA sequences and controlled muscle-specific genes during myogenic development, it is now apparent that MEF2 transcription factors are also highly expressed in neurons and are critical determinants of neuronal differentiation and fate. Here we focus our discussion on the role of MEF2 proteins in nervous tissue and the regulation of these transcription factors by calcium and phosphorylation signaling pathways.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas de Unión al ADN/fisiología , Neuronas/citología , Neuronas/metabolismo , Factores de Transcripción/fisiología , Animales , Supervivencia Celular/fisiología , Proteínas de Unión al ADN/genética , Humanos , Factores de Transcripción MEF2 , Células Musculares/fisiología , Factores Reguladores Miogénicos , Factores de Transcripción/genética
16.
J Neurochem ; 87(4): 914-21, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14622122

RESUMEN

In vivo, the pesticide rotenone induces degeneration of dopamine neurons and parkinsonian-like pathology in adult rats. In the current study, we utilized primary ventral mesencephalic (VM) cultures from E15 rats as an in vitro model to examine the mechanism underlying rotenone-induced death of dopamine neurons. After 11 h of exposure to 30 nm rotenone, the number of dopamine neurons identified by tyrosine hydroxylase (TH) immunostaining declined rapidly with only 23% of the neurons surviving. By contrast, 73% of total cells survived rotenone treatment, indicating that TH+ neurons are more sensitive to rotenone. Examination of the role of apoptosis in TH+ neuron death, revealed that 10 and 30 nm rotenone significantly increased the number of apoptotic TH+ neurons from 7% under control conditions to 38 and 55%, respectively. The increase in apoptotic TH+ neurons correlated with an increase in immunoreactivity for active caspase-3 in TH+ neurons. The caspase-3 inhibitor, DEVD, rescued a significant number of TH+ neurons from rotenone-induced death. Furthermore, this protective effect lasted for at least 32 h post-rotenone and DEVD exposure, indicating lasting neuroprotection achieved with an intervention prior to the death commitment point. Our results show for the first time in primary dopamine neurons that, at low nanomolar concentrations, rotenone induces caspase-3-mediated apoptosis. Understanding the mechanism of rotenone-induced apoptosis in dopamine neurons may contribute to the development of new neuroprotective strategies against Parkinson's disease.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Mesencéfalo , Neuronas/efectos de los fármacos , Plaguicidas/toxicidad , Rotenona/toxicidad , Animales , Apoptosis/efectos de los fármacos , Caspasa 3 , Inhibidores de Caspasas , Células Cultivadas , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Mesencéfalo/citología , Mesencéfalo/embriología , Neuronas/citología , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Oligopéptidos/farmacología , Ratas , Ratas Sprague-Dawley , Tirosina 3-Monooxigenasa/biosíntesis
17.
J Biol Chem ; 278(42): 41472-81, 2003 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-12896970

RESUMEN

Cerebellar granule neuron (CGN) survival depends on activity of the myocyte enhancer factor-2 (MEF2) transcription factors. Neuronal MEF2 activity is regulated by depolarization via a mechanism that is presently unclear. Here, we show that depolarization-mediated MEF2 activity and CGN survival are compromised by overexpression of the MEF2 repressor histone deacetylase-5 (HDAC5). Furthermore, removal of depolarization induced rapid cytoplasm-to-nuclear translocation of endogenous HDAC5. This effect was mimicked by addition of the calcium/calmodulin-dependent kinase (CaMK) inhibitor KN93 to depolarizing medium. Removal of depolarization or KN93 addition resulted in dephosphorylation of HDAC5 and its co-precipitation with MEF2D. HDAC5 nuclear translocation triggered by KN93 induced a marked loss of MEF2 activity and subsequent apoptosis. To selectively decrease CaMKII, CGNs were incubated with an antisense oligonucleotide to CaMKIIalpha. This antisense decreased CaMKIIalpha expression and induced nuclear shuttling of HDAC5 in CGNs maintained in depolarizing medium. Selectivity of the CaMKIIalpha antisense was demonstrated by its lack of effect on CaMKIV-mediated CREB phosphorylation. Finally, antisense to CaMKIIalpha induced caspase-3 activation and apoptosis, whereas a missense control oligonucleotide had no effect on CGN survival. These results indicate that depolarization-mediated calcium influx acts through CaMKII to inhibit HDAC5, thereby sustaining high MEF2 activity in CGNs maintained under depolarizing conditions.


Asunto(s)
Cerebelo/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas , Neuronas/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Adenoviridae/genética , Animales , Apoptosis , Western Blotting , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Caspasa 3 , Caspasas/metabolismo , Núcleo Celular/metabolismo , Medio de Cultivo Libre de Suero/farmacología , Citoplasma/metabolismo , Activación Enzimática , Epítopos , Genes Dominantes , Histona Desacetilasas , Inmunohistoquímica , Factores de Transcripción MEF2 , Mutación , Factores Reguladores Miogénicos , Oligonucleótidos Antisentido/farmacología , Fosforilación , Potasio/farmacología , Pruebas de Precipitina , Isoformas de Proteínas , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Transcripción Genética
18.
Ann N Y Acad Sci ; 991: 237-50, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12846991

RESUMEN

Chronic neurodegenerative diseases, including Parkinson's disease, are characterized by a selective loss of specific subsets of neuronal populations over a period of years or even decades. While the underlying causes of the various neurodegenerative diseases are not clear, the death of neurons and the loss of neuronal contacts are key pathological features. Pinpointing molecular events that control neuronal cell death is critical for the development of new strategies to prevent and treat neurodegenerative disorders.


Asunto(s)
Apoptosis/fisiología , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Animales , Muerte Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Transducción de Señal/fisiología
19.
J Neurochem ; 85(6): 1488-99, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12787068

RESUMEN

Depolarization promotes the survival of cerebellar granule neurons via activation of the transcription factor myocyte enhancer factor 2D (MEF2D). Removal of depolarization induces hyperphosphorylation of MEF2D on serine/threonine residues, resulting in its decreased DNA binding and susceptibility to caspases. The subsequent loss of MEF2-dependent gene transcription contributes to the apoptosis of granule neurons. The kinase(s) that phosphorylates MEF2D during apoptosis is currently unknown. The serine/threonine kinase, glycogen synthase kinase-3 beta (GSK-3 beta), plays a pro-apoptotic role in granule neurons. To investigate a potential role for GSK-3 beta in MEF2D phosphorylation, we examined the effects of lithium, a non-competitive inhibitor of GSK-3 beta, on MEF2D activity in cultured cerebellar granule neurons. Lithium inhibited caspase-3 activation and chromatin condensation in granule neurons induced to undergo apoptosis by removal of depolarizing potassium and serum. Concurrently, lithium suppressed the hyperphosphorylation and caspase-mediated degradation of MEF2D. Moreover, lithium sustained MEF2 DNA binding and transcriptional activity in the absence of depolarization. Lithium also attenuated MEF2D hyperphosphorylation and apoptosis induced by calcineurin inhibition under depolarizing conditions, a GSK-3 beta-independent model of neuronal death. In contrast to lithium, MEF2D hyperphosphorylation was not inhibited by forskolin, insulin-like growth factor-I, or valproate, three mechanistically distinct inhibitors of GSK-3 beta. These results demonstrate that the kinase that phosphorylates and inhibits the pro-survival function of MEF2D in cerebellar granule neurons is a novel lithium target distinct from GSK-3 beta.


Asunto(s)
Apoptosis/fisiología , Proteínas de Unión al ADN/metabolismo , Litio/farmacología , Neuronas/metabolismo , Fosfotransferasas/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas Sanguíneas/farmacología , Inhibidores de la Calcineurina , Caspasa 3 , Inhibidores de Caspasas , Caspasas/metabolismo , Células Cultivadas , Colforsina/farmacología , ADN/metabolismo , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Factores de Transcripción MEF2 , Factores Reguladores Miogénicos , Neuronas/citología , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosfotransferasas/metabolismo , Potasio/farmacología , Ratas , Ratas Sprague-Dawley , Transcripción Genética/efectos de los fármacos
20.
J Neurosci ; 22(21): 9287-97, 2002 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-12417654

RESUMEN

Cerebellar granule neurons depend on insulin-like growth factor-I (IGF-I) for their survival. However, the mechanism underlying the neuroprotective effects of IGF-I is presently unclear. Here we show that IGF-I protects granule neurons by suppressing key elements of the intrinsic (mitochondrial) death pathway. IGF-I blocked activation of the executioner caspase-3 and the intrinsic initiator caspase-9 in primary cerebellar granule neurons deprived of serum and depolarizing potassium. IGF-I inhibited cytochrome c release from mitochondria and prevented its redistribution to neuronal processes. The effects of IGF-I on cytochrome c release were not mediated by blockade of the mitochondrial permeability transition pore, because IGF-I failed to inhibit mitochondrial swelling or depolarization. In contrast, IGF-I blocked induction of the BH3-only Bcl-2 family member, Bim (Bcl-2 interacting mediator of cell death), a mediator of Bax-dependent cytochrome c release. The suppression of Bim expression by IGF-I did not involve inhibition of the c-Jun transcription factor. Instead, IGF-I prevented activation of the forkhead family member, FKHRL1, another transcriptional regulator of Bim. Finally, adenoviral-mediated expression of dominant-negative AKT activated FKHRL1 and induced expression of Bim. These data suggest that IGF-I signaling via AKT promotes survival of cerebellar granule neurons by blocking the FKHRL1-dependent transcription of Bim, a principal effector of the intrinsic death-signaling cascade.


Asunto(s)
Apoptosis/fisiología , Proteínas Portadoras/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/farmacología , Proteínas de la Membrana , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas , Transducción de Señal/fisiología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis , Proteína 11 Similar a Bcl2 , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 3 , Caspasa 9 , Caspasas/metabolismo , Células Cultivadas , Cerebelo/citología , Grupo Citocromo c/metabolismo , Proteínas de Unión al ADN/metabolismo , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Genes Dominantes , Humanos , Factor I del Crecimiento Similar a la Insulina/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas del Tejido Nervioso , Neuronas/citología , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/farmacología , Proteínas Proto-Oncogénicas c-akt , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA