Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Sci Rep ; 7: 42270, 2017 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-28205585

RESUMEN

Alzheimer's disease (AD) is characterized by extensive neuron loss that accompanies profound impairments in memory and cognition. We examined the neuronally directed effects of the retinoid X receptor agonist bexarotene in an aggressive model of AD. We report that a two week treatment of 3.5 month old 5XFAD mice with bexarotene resulted in the clearance of intraneuronal amyloid deposits. Importantly, neuronal loss was attenuated by 44% in the subiculum in mice 4 months of age and 18% in layer V of the cortex in mice 8 months of age. Moreover, bexarotene treatment improved remote memory stabilization in fear conditioned mice and improved olfactory cross habituation. These improvements in neuron viability and function were correlated with significant increases in the levels of post-synaptic marker PSD95 and the pre-synaptic marker synaptophysin. Moreover, bexarotene pretreatment improved neuron survival in primary 5XFAD neurons in vitro in response to glutamate-induced excitotoxicity. The salutary effects of bexarotene were accompanied by reduced plaque burden, decreased astrogliosis, and suppression of inflammatory gene expression. Collectively, these data provide evidence that bexarotene treatment reduced neuron loss, elevated levels of markers of synaptic integrity that was linked to improved cognition and in an aggressive model of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Neuronas/metabolismo , Receptores X Retinoide/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Bexaroteno , Biomarcadores/metabolismo , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Gliosis/complicaciones , Gliosis/tratamiento farmacológico , Gliosis/patología , Ácido Glutámico/toxicidad , Habituación Psicofisiológica/efectos de los fármacos , Hipocampo/patología , Inflamación/patología , Masculino , Proteínas de Transporte de Membrana/metabolismo , Memoria/efectos de los fármacos , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/patología , Neurotoxinas/toxicidad , Bulbo Olfatorio/efectos de los fármacos , Bulbo Olfatorio/patología , Placa Amiloide/metabolismo , Placa Amiloide/patología , Receptores de LDL/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Tetrahidronaftalenos/farmacología , Tetrahidronaftalenos/uso terapéutico
2.
Neuroscience ; 170(3): 846-57, 2010 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-20691766

RESUMEN

Stroke is a leading cause of death and disability but has limited therapeutic options. Thiazolidinediones (TZDs), agonists for the nuclear receptor, peroxisome proliferator-activated receptor (PPAR)γ, reduce infarct volume and improve neurologic function following transient middle cerebral artery occlusion (MCAO) in rats. Translation of these findings into clinical therapy will require careful assessment of dosing paradigms and effective time windows for treatment. Understanding the mechanisms by which TZDs protect the brain provides insight into how time windows for neuroprotection might be extended. We find that two TZDs, pioglitazone and rosiglitazone, significantly reduce infarct volume at doses similar to those used clinically (1 mg/kg for pioglitazone and 0.1 mg/kg for rosiglitazone). We also find that pioglitazone reduces infarction volume in a transient, but not a permanent MCAO model suggesting that reperfusion plays an important role in TZD mediated neuroprotection. Since PPARγ agonists reduce inflammation and oxidative stress, both of which are exacerbated by reperfusion, we hypothesized that TZDs would be most effective if administered prior to reperfusion. We administered TZDs 3 h after MCAO and found that infarction volume and neurologic function are significantly improved in animals reperfused at 3 h and 15 min (after TZD treatment), but not in animals reperfused at 2 h (before TZD treatment) when assessed either 24 h or 3 weeks after MCAO. While TZDs reduce intercellular adhesion molecule (ICAM) expression to a similar extent regardless of the time of reperfusion, leukocyte entry into brain parenchyma is more dramatically reduced when reperfusion is delayed until after drug treatment. The finding that delaying reperfusion until after TZD treatment is beneficial despite a longer period of ischemia, is dramatic given the widely held view that duration of ischemia is the most important determinate of injury.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Fármacos Neuroprotectores/administración & dosificación , Reperfusión/métodos , Accidente Cerebrovascular/tratamiento farmacológico , Tiazolidinedionas/administración & dosificación , Animales , Conducta Animal/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/patología , Moléculas de Adhesión Celular/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Leucocitos/efectos de los fármacos , Masculino , Pioglitazona , Ratas , Ratas Wistar , Flujo Sanguíneo Regional/efectos de los fármacos , Rosiglitazona
3.
Mol Psychiatry ; 14(4): 448-61, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18227838

RESUMEN

The extracellular signal-regulated kinase (ERK) pathway mediates neuronal plasticity in the CNS. The mood stabilizers lithium and valproate activate the ERK pathway in prefrontal cortex and hippocampus and potentiate ERK pathway-mediated neurite growth, neuronal survival and hippocampal neurogenesis. Here, we examined the role of the ERK pathway in behavioral plasticity related to facets of bipolar disorder. Mice with ERK1 ablation acquired reduced phosphorylation of RSK1, an ERK substrate, in prefrontal cortex and striatum, but not in hippocampus or cerebellum, indicating the ablation-induced brain region-specific ERK signaling deficits. ERK1 ablation produced a behavioral excitement profile similar to that induced by psychostimulants. The profile is characterized by hyperactivity, enhanced goal-directed activity and increased pleasure-related activity with potential harmful consequence. ERK1-ablated mice were hyperactive in multiple tests and resistant to behavioral despair in the forced swim test. These mice displayed more home-cage voluntary wheel running activities, rearings in a large arena and open-arm visits in an elevated plus maze. Treatments with valproate and olanzapine, but not lithium reduced baseline activities in ERK1-ablated mice. All three treatments attenuated amphetamine-induced hyperactivity in ablated mice. These data indicate a profound involvement of ERK1 signaling in behavioral excitement and in the behavioral action of antimanic agents. The extent to which ERK pathway perturbation contributes to the susceptibility, mood switch mechanism(s) and symptom pathophysiology of bipolar disorder requires further investigation. Whether there is a shared mechanism through which mood stabilizers produce their clinical actions on mood, thought and behavioral symptoms of mania also requires further investigation.


Asunto(s)
Conducta Animal/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Transducción de Señal/fisiología , Adyuvantes Inmunológicos , Administración Oral , Anfetamina/farmacología , Análisis de Varianza , Animales , Antipsicóticos/farmacología , Conducta Animal/efectos de los fármacos , Benzodiazepinas/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Inhibidores Enzimáticos/farmacología , Cloruro de Litio/administración & dosificación , Locomoción/efectos de los fármacos , Locomoción/genética , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Noqueados , Proteína Quinasa 3 Activada por Mitógenos/deficiencia , Olanzapina , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Transducción de Señal/genética , Natación , Ácido Valproico/farmacología
4.
Eur J Neurosci ; 24(6): 1653-63, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17004929

RESUMEN

Stroke is a devastating disease with limited treatment options. Recently, we found that the peroxisome proliferator-activated receptor-gamma (PPARgamma) agonists troglitazone and pioglitazone reduce injury and inflammation in a rat model of transient cerebral ischemia. The mechanism of this protection is unclear, as these agents can act through PPAR-gamma activation or through PPAR-gamma-independent mechanisms. Therefore, we examined PPAR-gamma expression, DNA binding and transcriptional activity following stroke. In addition, we used a PPAR-gamma antagonist, T0070907, to determine the role of PPAR-gamma during ischemia. Using immunohistochemical techniques and real-time PCR, we found low levels of PPAR-gamma mRNA and PPAR-gamma immunoreactivity in nonischemic brain; however, PPAR-gamma expression dramatically increased in ischemic neurons, peaking 24 h following middle cerebral artery occlusion. Interestingly, we found that in both vehicle- and agonist-treated brains, DNA binding was reduced in the ischemic hemisphere relative to the contralateral hemisphere. Expression of a PPAR-gamma target gene, lipoprotein lipase, was also reduced in ischemic relative to nonischemic brain. Both DNA binding and lipoprotein lipase expression were increased by the addition of the PPAR-gamma agonist rosiglitazone. Finally, we found that rosiglitazone-mediated protection after stroke was reversed by the PPAR-gamma antagonist T0070907. Interestingly, infarction size was also increased by T0070907 in the absence of PPAR-gamma agonist, suggesting that endogenous PPAR-gamma ligands may mitigate the effects of cerebral ischemia.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Ataque Isquémico Transitorio/metabolismo , PPAR gamma/metabolismo , Animales , Benzamidas/farmacología , Activación Enzimática/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Inmunohistoquímica/métodos , Ataque Isquémico Transitorio/tratamiento farmacológico , Ataque Isquémico Transitorio/patología , Masculino , PPAR gamma/agonistas , PPAR gamma/antagonistas & inhibidores , PPAR gamma/genética , Unión Proteica/fisiología , Piridinas/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Rosiglitazona , Tiazolidinedionas/uso terapéutico , Factores de Tiempo
5.
Neuroscience ; 130(3): 685-96, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15590152

RESUMEN

Newly developed insulin-sensitizing agents, which target the nuclear receptor peroxisome proliferator-activated receptor-gamma have recently been appreciated to exhibit potent anti-inflammatory actions. Since stroke is associated with an intense inflammatory response, we reasoned that these agents may ameliorate injury from stroke. We report that administration of troglitazone or pioglitazone 24 h before and at the time of cerebral infarction dramatically reduced infarction volume and improved neurological function following middle cerebral artery occlusion in rats. Furthermore, we find that delayed therapy also significantly reduced infarct volume. The brains of the drug-treated animals displayed reduced inflammation as evidenced by decreased immunoreactivity for microglial/macrophage markers and reduced protein and mRNA for interleukin-1beta, cyclooxygenase-2 and inducible nitric oxide synthase. We argue that the beneficial effects of these drugs are likely due to reduced expression of these inflammatory mediators, which are known to exacerbate ischemic injury following stroke. These results are of particular relevance to diabetic patients chronically treated with these agents who may benefit from the neuroprotective actions of these drugs.


Asunto(s)
Cromanos/uso terapéutico , Encefalitis/tratamiento farmacológico , Encefalitis/patología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Ataque Isquémico Transitorio/tratamiento farmacológico , Ataque Isquémico Transitorio/patología , PPAR gamma/efectos de los fármacos , Tiazolidinedionas/uso terapéutico , Animales , Glucemia/metabolismo , Química Encefálica/efectos de los fármacos , Química Encefálica/genética , Recuento de Células , Circulación Cerebrovascular/fisiología , Relación Dosis-Respuesta a Droga , Encefalitis/etiología , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/etiología , Ataque Isquémico Transitorio/metabolismo , Ligandos , Macrófagos/efectos de los fármacos , Masculino , Microglía/efectos de los fármacos , Arteria Cerebral Media/fisiología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Pioglitazona , Desempeño Psicomotor/efectos de los fármacos , Desempeño Psicomotor/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Troglitazona
6.
Neurochem Int ; 39(5-6): 449-57, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11578780

RESUMEN

Amyloid deposition within the brains of Alzheimer's Disease patients results in the activation of microglial cells and the induction of a local inflammatory response. The interaction of microglia or monocytes with beta-amyloid (A beta) fibrils elicits the activation a complex tyrosine kinase-based signal transduction cascade leading to stimulation of multiple independent signaling pathways and ultimately to changes in proinflammatory gene expression. The A beta-stimulated expression of proinflammatory genes in myeloid lineage cells is antagonized by the action of a family of ligand-activated nuclear hormone receptors, the peroxisome proliferator-activated receptors (PPARs). We report that THP-1 monocytes express predominantly PPAR gamma isoform and lower levels of PPAR alpha and PPAR delta isoforms. PPAR mRNA levels are not affected by differentiation of the cells into a macrophage phenotype, nor are they altered following exposure to the classical immune stimulus, lipopolysaccharide. Previous studies have found that PPAR gamma agonists act broadly to inhibit inflammatory responses. The present study explored the action of the PPAR alpha isoform and found that PPAR alpha agonists inhibited the A beta-stimulated expression of TNFalpha and IL-6 reporter genes in a dose-dependent manner. Moreover, the PPAR alpha agonist WY14643 inhibited macrophage differentiation and COX-2 gene expression. However, the PPAR alpha agonists failed to inhibit A beta-stimulated elaboration of neurotoxic factors by THP-1 cells. These findings demonstrate that PPAR alpha acts to suppress a diverse array of inflammatory responses in monocytes.


Asunto(s)
Péptidos beta-Amiloides/fisiología , Inflamación/etiología , Proteínas de la Membrana , Receptores Citoplasmáticos y Nucleares/fisiología , Receptores de Lipoproteína , Factores de Transcripción/fisiología , Amiloide/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Ciclooxigenasa 2 , Humanos , Interleucina-6/antagonistas & inhibidores , Isoenzimas/metabolismo , Macrófagos/citología , Microglía/fisiología , Monocitos/citología , Monocitos/fisiología , Neurotoxinas/antagonistas & inhibidores , Prostaglandina-Endoperóxido Sintasas/metabolismo , Proteínas Tirosina Quinasas/fisiología , Receptor para Productos Finales de Glicación Avanzada , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Inmunológicos/fisiología , Receptores Depuradores , Receptores Depuradores de Clase B , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factores de Transcripción/agonistas , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
7.
Microsc Res Tech ; 54(2): 59-70, 2001 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-11455613

RESUMEN

The etiology of Alzheimer's disease (AD) involves a significant inflammatory component as evidenced by the presence of elevated levels of a diverse range of proinflammatory molecules in the AD brain. These inflammatory molecules are produced principally by activated microglia, which are found to be clustered within and adjacent to the senile plaque. Moreover, long-term treatment of patients with non-steroidal anti-inflammatory drugs has been shown to reduce risk and incidence of AD and delay disease progression. The microglia respond to beta-amyloid (Abeta) deposition in the brain through the interaction of fibrillar forms of amyloid with cell surface receptors, leading to the activation of intracellular signal transduction cascades. The activation of multiple independent signaling pathways ultimately leads to the induction of proinflammatory gene expression and production of reactive oxygen and nitrogen species. These microglial inflammatory products act in concert to produce neuronal toxicity and death. Therapeutic approaches focused on inhibition of the microglial-mediated local inflammatory response in the AD brain offer new opportunities to intervene in the disease.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/metabolismo , Inflamación , Microglía/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Humanos , Transducción de Señal
8.
J Biol Chem ; 276(21): 18169-77, 2001 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-11278445

RESUMEN

In PC12 cells, epidermal growth factor (EGF) transiently stimulates the mitogen-activated protein (MAP) kinases, ERK1 and ERK2, and provokes cellular proliferation. In contrast, nerve growth factor (NGF) stimulation leads to the sustained activation of the MAPKs and subsequently to neuronal differentiation. It has been shown that both the magnitude and longevity of MAPK activation governs the nature of the cellular response. The activations of MAPKs are dependent upon two distinct small G-proteins, Ras and Rap1, that link the growth factor receptors to the MAPK cascade by activating c-Raf and B-Raf, respectively. We found that Ras was transiently stimulated upon both EGF and NGF treatment of PC12 cells. However, EGF transiently activated Rap1, whereas NGF stimulated prolonged Rap1 activation. The activation of the ERKs was due almost exclusively (>90%) to the action of B-Raf. The transient activation of the MAPKs by EGF was a consequence of the formation of a short lived complex assembling on the EGF receptor itself, composed of Crk, C3G, Rap1, and B-Raf. In contrast, NGF stimulation of the cells resulted in the phosphorylation of FRS2. FRS2 scaffolded the assembly of a stable complex of Crk, C3G, Rap1, and B-Raf resulting in the prolonged activation of the MAPKs. Together, these data provide a signaling link between growth factor receptors and MAPK activation and a mechanistic explanation of the differential MAPK kinetics exhibited by these growth factors.


Asunto(s)
Factor de Crecimiento Epidérmico/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Factor de Crecimiento Nervioso/farmacología , Animales , Células PC12 , Ratas , Transducción de Señal/efectos de los fármacos
9.
Learn Mem ; 8(1): 11-9, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11160759

RESUMEN

The extracellular signal-regulated kinases (ERKs) are members of the mitogen-activated protein kinase (MAPK) superfamily of enzymes and have recently garnered considerable attention in the field of learning and memory. ERK activation has been shown to be required for the induction of long-term potentiation (LTP) in the rat hippocampus and for the formation of associative and spatial memories in both the rat and the mouse. However, the individual roles for the two isoforms of ERK have yet to be deciphered. To investigate the specific contribution of the ERK1 (p44) isoform of MAPK to mammalian learning, we performed a general behavioral and physiological characterization of mice lacking the ERK1 gene. The ERK1-null animals demonstrated significantly higher levels of activity in the open field test. However, we observed no other discernible deficits in the ERK1 knockout mice in our behavioral testing. Specifically, no differences were observed in the acquisition or retention (24 h and 2 wk after training) of either contextual or cue fear conditioning between the ERK1(-/-) and their wild-type littermate controls. In addition, no learning phenotype was observed in the passive avoidance test. When hippocampal slices were analyzed, we found no deficits in baseline synaptic transmission or in tetanus-induced LTP in hippocampal area CA1. We found no apparent compensatory changes in the expression of ERK2 (p42 MAPK). We conclude that hippocampus- and amygdala-dependent emotional learning does not depend critically on the activity of ERK1.


Asunto(s)
Emociones/fisiología , Aprendizaje/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Animales , Hipocampo/fisiología , Ratones , Ratones Noqueados/genética , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/genética , Actividad Motora/fisiología , Valores de Referencia , Sensación/fisiología
11.
J Neurosci ; 21(4): 1179-88, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11160388

RESUMEN

Reactive microglia associated with the beta-amyloid plaques in Alzheimer's disease (AD) brains initiate a sequence of inflammatory events integral to the disease process. We have observed that fibrillar beta-amyloid peptides activate a tyrosine kinase-based signaling response in primary mouse microglia and the human monocytic cell line, THP-1, resulting in production of neurotoxic secretory products, proinflammatory cytokines, and reactive oxygen species. We report that most of the amyloid-induced tyrosine kinase activity was stimulated after activation of Src family members such as Lyn. However, transduction of the signaling response required for increased production of the cytokines TNFalpha and IL1-beta was mediated by the nonreceptor tyrosine kinase, Syk. Additionally, beta-amyloid stimulated an NFkappaB-dependent pathway in parallel that was required for cytokine production. Importantly, TNFalpha generated by the monocytes and microglia was responsible for the majority of the neuorotoxic activity secreted by these cells after beta-amyloid stimulation but must act in concert with other factors elaborated by microglia to elicit neuronal death. Moreover, we observed that the neuronal loss was apoptotic in nature and involved increased neuronal expression of inducible nitric oxide synthase and subsequent peroxynitrite production. Selective inhibitors of inducible nitric oxide synthase effectively protected cells from toxicity associated with the microglial and monocytic secretory products. This study demonstrates a functional linkage between beta-amyloid-dependent activation of microglia and several characteristic markers of neuronal death occurring in Alzheimer's disease brains.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Microglía/metabolismo , Monocitos/metabolismo , Neuronas/metabolismo , Óxido Nítrico Sintasa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/farmacología , Animales , Apoptosis , Células Cultivadas , Contraindicaciones , Precursores Enzimáticos/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Microglía/efectos de los fármacos , Monocitos/citología , Monocitos/efectos de los fármacos , FN-kappa B/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal/fisiología , Quinasa Syk , Factores de Transcripción/metabolismo , Familia-src Quinasas/metabolismo
12.
Neurobiol Aging ; 22(6): 937-44, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11755002

RESUMEN

The role of inflammatory processes in the brains of Alzheimer's Disease (AD) patients has recently attracted considerable interest. Indeed, the only demonstrated effective therapy for AD patients is long-term treatment with non-steroidal anti-inflammatory drugs (NSAIDs). The mechanistic basis of the efficacy of NSAIDs in AD remains unclear. However, the recent recognition that NSAIDs can bind to and activate the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma), has offered an explanation for the action of these drugs in AD. PPARgamma activation leads to the inhibition of microglial activation and the expression of a broad range of proinflammatory molecules. The newly appreciated anti-inflammatory actions of PPARgamma agonists may allow novel therapies for AD and other CNS indications with an inflammatory component.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/prevención & control , Antiinflamatorios no Esteroideos/farmacología , Inflamación/prevención & control , Receptores Citoplasmáticos y Nucleares/agonistas , Factores de Transcripción/agonistas , Animales , Humanos , Receptores Citoplasmáticos y Nucleares/biosíntesis , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción/biosíntesis , Factores de Transcripción/fisiología
13.
J Cell Sci ; 113 ( Pt 18): 3241-53, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10954422

RESUMEN

CLK1 was one of the first identified dual specificity kinases and is the founding member of the 'LAMMER' family of kinases. We have established the substrate site specificity of CLK1. We report here that truncation of the N terminus of CLK1 resulted in a dramatic increase in CLK1 enzymatic activity, indicating that the N terminus acts as a negative regulatory domain. The N-terminal truncation resulted in a 45-fold increase in V(max), suggesting that this domain does not contain a pseudo-substrate motif, but may act to conformationally constrain the catalytic activity of CLK1. Tyrosine phosphorylation has been proposed to be critical for CLK1 activity, however, CLK1 activity was unaffected by exposure to tyrosine phosphatases. Treatment of CLK1 with the serine/threonine specific phosphatase PP2A, resulted in a 2- to 6-fold increase in enzymatic activity. Incubation of CLK1 with tyrosine phosphatases in combination with PP2A abolished CLK1 activity. These data suggest that CLK1 is regulated by three distinct mechanisms that serve to both positively and negatively regulate CLK1 activity. CLK1 activity is positively regulated by phosphorylation on either tyrosine residues or serine/threonine residues, and is negatively regulated by steric constraints mediated by the N-terminal domain, as well as, by phosphorylation on a subset of serine/threonine residues within the catalytic domain. CLK1 mRNA is expressed at low levels in all tissues and cell lines examined. The full-length and truncated splice forms are expressed at roughly equivalent levels in most tissues. The ratio of the two splice variants of CLK1 can be altered by treatment with cycloheximide. CLK1 protein expression is limited to a small subset of highly localized neuronal populations in the rat brain. Contrary to previous studies using overexpression systems, we show that CLK1 protein is primarily found in the cytoplasm of these cells, with only a small fraction localized to the nucleus.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Sitios de Unión , Western Blotting/métodos , Encéfalo/enzimología , Encéfalo/patología , Secuencia de Consenso , Ratones , Datos de Secuencia Molecular , Proteína Básica de Mielina/metabolismo , Células PC12 , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Empalme del ARN , ARN Mensajero , Conejos , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Coloración y Etiquetado/métodos , Especificidad por Sustrato , Transfección
14.
J Neurosci ; 20(2): 558-67, 2000 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-10632585

RESUMEN

Alzheimer's disease (AD) is characterized by the extracellular deposition of beta-amyloid fibrils within the brain and the subsequent association and phenotypic activation of microglial cells associated with the amyloid plaque. The activated microglia mount a complex local proinflammatory response with the secretion of a diverse range of inflammatory products. Nonsteroidal anti-inflammatory drugs (NSAIDs) are efficacious in reducing the incidence and risk of AD and significantly delaying disease progression. A recently appreciated target of NSAIDs is the ligand-activated nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma). PPARgamma is a DNA-binding transcription factor whose transcriptional regulatory actions are activated after agonist binding. We report that NSAIDs, drugs of the thiazolidinedione class, and the natural ligand prostaglandin J2 act as agonists for PPARgamma and inhibit the beta-amyloid-stimulated secretion of proinflammatory products by microglia and monocytes responsible for neurotoxicity and astrocyte activation. The activation of PPARgamma also arrested the differentiation of monocytes into activated macrophages. PPARgamma agonists were shown to inhibit the beta-amyloid-stimulated expression of the cytokine genes interleukin-6 and tumor necrosis factor alpha. Furthermore, PPARgamma agonists inhibited the expression of cyclooxygenase-2. These data provide direct evidence that PPARgamma plays a critical role in regulating the inflammatory responses of microglia and monocytes to beta-amyloid. We argue that the efficacy of NSAIDs in the treatment of AD may be a consequence of their actions on PPARgamma rather than on their canonical targets the cyclooxygenases. Importantly, the efficacy of these agents in inhibiting a broad range of inflammatory responses suggests PPARgamma agonists may provide a novel therapeutic approach to AD.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/farmacología , Antiinflamatorios no Esteroideos/farmacología , Astrocitos/fisiología , Microglía/fisiología , Fragmentos de Péptidos/farmacología , Receptores Citoplasmáticos y Nucleares/agonistas , Tiazolidinedionas , Factores de Transcripción/agonistas , Animales , Animales Recién Nacidos , Astrocitos/citología , Encéfalo/citología , Encéfalo/fisiología , Diferenciación Celular , Cromanos/farmacología , Ciclooxigenasa 2 , Dinoprost/farmacología , Genes Reporteros , Humanos , Inflamación , Interleucina-6/genética , Isoenzimas/metabolismo , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Microcuerpos/fisiología , Microglía/citología , Microglía/efectos de los fármacos , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/fisiología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Proteínas Recombinantes/biosíntesis , Acetato de Tetradecanoilforbol/farmacología , Tiazoles/farmacología , Transfección , Troglitazona , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/genética
15.
J Neurosci ; 19(19): 8182-98, 1999 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-10493720

RESUMEN

Post-traumatic cystic cavitation, in which the size and severity of a CNS injury progress from a small area of direct trauma to a greatly enlarged secondary injury surrounded by glial scar tissue, is a poorly understood complication of damage to the brain and spinal cord. Using minimally invasive techniques to avoid primary physical injury, this study demonstrates in vivo that inflammatory processes alone initiate a cascade of secondary tissue damage, progressive cavitation, and glial scarring in the CNS. An in vitro model allowed us to test the hypothesis that specific molecules that stimulate macrophage inflammatory activation are an important step in initiating secondary neuropathology. Time-lapse video analyses of inflammation-induced cavitation in our in vitro model revealed that this process occurs primarily via a previously undescribed cellular mechanism involving dramatic astrocyte morphological changes and rapid migration. The physical process of cavitation leads to astrocyte abandonment of neuronal processes, neurite stretching, and secondary injury. The macrophage mannose receptor and the complement receptor type 3 beta2-integrin are implicated in the cascade that induces cavity and scar formation. We also demonstrate that anti-inflammatory agents modulating transcription via the nuclear hormone receptor peroxisome proliferator-activated receptor-gamma may be therapeutic in preventing progressive cavitation by limiting inflammation and subsequent secondary damage after CNS injury.


Asunto(s)
Astrocitos/patología , Lesiones Encefálicas/patología , Corteza Cerebral/patología , Ganglios Espinales/patología , Inflamación , Neuroglía/patología , Neuronas/patología , Tiazolidinedionas , Animales , Animales Recién Nacidos , Astrocitos/fisiología , Astrocitos/ultraestructura , Axones/patología , Axones/ultraestructura , Lesiones Encefálicas/inducido químicamente , Lesiones Encefálicas/fisiopatología , Movimiento Celular , Células Cultivadas , Ganglios Espinales/lesiones , Ganglios Espinales/fisiopatología , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/análisis , Indometacina/farmacología , Inflamación/inducido químicamente , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/toxicidad , Activación de Macrófagos , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/fisiología , Microscopía por Video , Neuritas/patología , Neuritas/fisiología , Neuritas/ultraestructura , Neuroglía/fisiología , Neuroglía/ultraestructura , Neuronas/fisiología , Neuronas/ultraestructura , Proteoglicanos/biosíntesis , Proteoglicanos/genética , Ratas , Ratas Sprague-Dawley , Tiazoles/farmacología , Zimosan/administración & dosificación , Zimosan/toxicidad
16.
J Biol Chem ; 274(38): 26697-704, 1999 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-10480872

RESUMEN

The protein-tyrosine phosphatase PTP-1B is an important regulator of intracellular protein tyrosine phosphorylation, and is itself regulated by phosphorylation. We report that PTP-1B and its yeast analog, YPTP, are phosphorylated and activated by members of the CLK family of dual specificity kinases. CLK1 and CLK2 phosphorylation of PTP-1B in vitro activated the phosphatase activity approximately 3-5-fold using either p-nitrophenol phosphate, or tyrosine-phosphorylated myelin basic protein as substrates. Co-expression of CLK1 or CLK2 with PTP-1B in HEK 293 cells led to a 2-fold stimulation of phosphatase activity in vivo. Phosphorylation of PTP-1B at Ser(50) by CLK1 or CLK2 is responsible for its enzymatic activation. These findings suggest that phosphorylation at Ser(50) by serine threonine kinases may regulate the activation of PTP-1B in vivo. We also show that CLK1 and CLK2 phosphorylate and activate the S. cerevisiae PTP-1B family member, YPTP1. CLK1 phosphorylation of YPTP1 led to a 3-fold stimulation of phosphatase activity in vitro. We demonstrate that CLK phosphorylation of Ser(83) on YPTP1 is responsible for the activation of this enzyme. These findings demonstrate that the CLK kinases activate PTP-1B family members, and this phosphatase may be an important cellular target for CLK action.


Asunto(s)
Proteínas de Caenorhabditis elegans , Proteínas Portadoras/metabolismo , Proteínas del Helminto/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Fosfatasas , Línea Celular , Electroforesis en Gel de Poliacrilamida , Activación Enzimática , Humanos , Fosforilación , Pliegue de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteínas Tirosina Quinasas , Serina/metabolismo
17.
J Neurosci ; 19(3): 928-39, 1999 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-9920656

RESUMEN

Microglial interaction with amyloid fibrils in the brains of Alzheimer's and prion disease patients results in the inflammatory activation of these cells. We observed that primary microglial cultures and the THP-1 monocytic cell line are stimulated by fibrillar beta-amyloid and prion peptides to activate identical tyrosine kinase-dependent inflammatory signal transduction cascades. The tyrosine kinases Lyn and Syk are activated by the fibrillar peptides and initiate a signaling cascade resulting in a transient release of intracellular calcium that results in the activation of classical PKC and the recently described calcium-sensitive tyrosine kinase PYK2. Activation of the MAP kinases ERK1 and ERK2 follows as a subsequent downstream signaling event. We demonstrate that PYK2 is positioned downstream of Lyn, Syk, and PKC. PKC is a necessary intermediate required for ERK activation. Importantly, the signaling response elicited by beta-amyloid and prion fibrils leads to the production of neurotoxic products. We have demonstrated in a tissue culture model that conditioned media from beta-amyloid- and prion-stimulated microglia or from THP-1 monocytes are neurotoxic to mouse cortical neurons. This toxicity can be ameliorated by treating THP-1 cells with specific enzyme inhibitors that target various components of the signal transduction pathway linked to the inflammatory responses.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Amiloide/biosíntesis , Microglía/fisiología , Neurotoxinas/metabolismo , Fragmentos de Péptidos/farmacología , Priones/farmacología , Transducción de Señal/fisiología , Péptidos beta-Amiloides/química , Animales , Calcio/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Células Cultivadas , Activación Enzimática/fisiología , Precursores Enzimáticos/fisiología , Quinasa 2 de Adhesión Focal , Membranas Intracelulares/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Fosforilación , Priones/química , Proteína Quinasa C/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas/fisiología , Quinasa Syk , Tirosina/metabolismo , Familia-src Quinasas/fisiología
18.
J Biol Chem ; 274(6): 3385-95, 1999 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-9920881

RESUMEN

The in vitro phosphorylation of transcription factors by growth factor-activated protein kinases has resulted in the discovery of a number of activities whose identities and relationships to one another are unclear. Fos kinase is a growth factor-stimulated serine/threonine protein kinase that phosphorylates c-Fos at serine 362 within the carboxyl-terminal regulatory domain. Fos kinase activation is dependent on p21(ras) and mitogen-activated protein kinase/ERK kinase kinase (MEK) activity and is independent of phosphatidylinositol 3-kinase activity. We have purified Fos kinase by affinity chromatography using the Sepharose-linked protein kinase inhibitor, bisindolylmaleimide (BIM). Fos kinase has an apparent molecular mass of 88 kDa, and mass spectrophotometric analysis of the isolated protein showed that it produced tryptic fragments identical to those predicted for pp90(rsk2). Fos kinase isolated from nerve growth factor-stimulated PC12 cells is indistinguishable from NGFI-B kinase I, based on their chromatographic behavior, substrate specificities, and relative sensitivity to BIM. Furthermore, we have distinguished Fos kinase from calcium/cAMP response element-binding protein (CREB) kinase. Therefore, Fos kinase and NGFI-B kinase I and pp90(rsk2) represent the same protein kinase species. Moreover, we report that pp90(rsk2) exists within nerve growth factor-stimulated PC12 cells as two chromatographically and immunologically distinct species. Finally, we demonstrate that CREB kinase is distinct from pp90(rsk2).


Asunto(s)
Proteínas Quinasas S6 Ribosómicas/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Datos de Secuencia Molecular , Células PC12 , Fosforilación , Ratas , Proteínas Quinasas S6 Ribosómicas/aislamiento & purificación , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Especificidad por Sustrato
19.
J Neurobiol ; 36(4): 537-49, 1998 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-9740025

RESUMEN

The stress-activated protein kinases (SAPKs) are differentially activated by a variety of cellular stressors in PC12 cells. SAPK activation has been linked to the induction of apoptotic cell death upon serum withdrawal from undifferentiated cells or following nerve growth factor (NGF) withdrawal of neuronally differentiated PC12 cells. However, withdrawal of trophic support from differentiated cells led to only a very modest elevation of SAPK activity and led us to investigate the basis of the relative insensitivity of these enzymes to stressors. NGF-stimulated differentiation of the cells resulted in the elevation of basal SAPK activity to levels four- to sevenfold greater than in untreated cells, which was correlated with an approximate fivefold increase in SAPK protein levels. Paradoxically, in NGF-differentiated PC12 cells, exposure to cellular stressors provoked a proportionately smaller stimulation of SAPK activity than that observed in naive cells, despite the presence of much higher levels of SAPK protein. The insensitivity of SAPK to activation by stressors was reflective of the activity of the SAPK activator SEK, whose activation was also diminished following NGF differentiation of the cells. The data demonstrate that SAPKs are subject to complex controls through both induction of SAPK expression and the regulation mediated by upstream elements within this pathway.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/biosíntesis , Proteínas Quinasas Activadas por Mitógenos , Factores de Crecimiento Nervioso/farmacología , Estrés Fisiológico/enzimología , Animales , Western Blotting , Diferenciación Celular/fisiología , Medio de Cultivo Libre de Suero , Calor , Proteínas Quinasas JNK Activadas por Mitógenos , Células PC12 , Proteínas Proto-Oncogénicas c-jun/biosíntesis , Ratas , Estimulación Química
20.
J Biol Chem ; 273(32): 19965-71, 1998 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-9685331

RESUMEN

Extracellular purine nucleotides elicit a diverse range of biological responses through binding to specific cell surface receptors. The ionotrophic P2X subclass of purinoreceptors respond to ATP by stimulation of calcium ion permeability; however, it is unknown how P2X purinoreceptor activation is linked to intracellular signaling pathways. We report that stimulation of PC12 cells with ATP results in the activation of the mitogen-activated protein (MAP) kinases ERK1 and ERK2 and was wholly dependent upon extracellular calcium ions. Treatment of the cells with adenosine, AMP, ADP, UTP, or alpha,beta-methylene ATP was without effect; however, MAP kinase activation was abolished by pretreatment with suramin and reactive blue 2. The calcium-activated tyrosine kinase, Pyk2, acts as an upstream regulator of the MAP kinases and became tyrosine phosphorylated following treatment of the cells with ATP. We have ruled out the involvement of depolarization-mediated calcium influx because specific blockers of voltage-gated calcium channels did not affect MAP kinase activation. These data provide direct evidence that calcium influx through P2X2 receptors results in the activation of the MAP kinase cascade. Finally, we demonstrate that a different line of PC12 cells respond to ATP through P2Y2 purinoreceptors, providing an explanation for the conflicting findings of purine nucleotide responsiveness in PC12 cells.


Asunto(s)
Adenosina Trifosfato/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas Quinasas Activadas por Mitógenos , Animales , Calcio/fisiología , Bloqueadores de los Canales de Calcio/farmacología , Activación Enzimática/fisiología , Quinasa 2 de Adhesión Focal , Proteína Quinasa 1 Activada por Mitógenos , Proteína Quinasa 3 Activada por Mitógenos , Factores de Crecimiento Nervioso/farmacología , Células PC12 , Fosfotirosina/análisis , Proteínas Tirosina Quinasas/fisiología , Ratas , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2X2 , Receptores Purinérgicos P2Y2 , Transducción de Señal/fisiología , Suramina/farmacología , Triazinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...