Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Auton Neurosci ; 216: 25-32, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30206032

RESUMEN

Sympathetic neurons of SCG are dependent on availability of nerve growth factor (NGF) for their survival. SCG neurons express nicotinic receptors (nAChR) whose expression levels are modulated by nicotine. Nicotine exerts multiple effects on neurons, including neuroprotection, through nAChR binding. Although sympathetic neurons express robust levels of nAChR, a possible neuroprotective role for nicotine in these neurons is not well-understood. Therefore we determined the effect of nicotine exposure on survival of SCG neurons during NGF withdrawal in a well-established cell culture system. NGF was withdrawn in rat neonatal SCG neuron cultures which were then treated with either 10 µM nicotine alone or with nAChR antagonists 0.1 µM α-bungarotoxin (antagonist for α7 subunit bearing nAChR) and 10 µM mecamylamine (non-specific antagonist for ganglionic nAChR) for 48 h. Apoptotic death was determined by TUNEL staining. Cell survival was also determined by MTS assay. Western blot analysis of ERK1/2 was also performed. Our results showed that exposure to 10 µM nicotine significantly reduced apoptotic cell death in SCG neurons resulting from NGF withdrawal as shown by fewer TUNEL positive cells. The MTS assay results also revealed that 10 µM nicotine concentration significantly increased cell survival thus indicating neuroprotective effect of nicotine against cell death resulting from NGF withdrawal. Nicotinic receptor antagonists (bungarotoxin & mecamylamine) attenuated the effect of nicotine's action of neuroprotection. Western blot analysis showed an increased expression of ERK1/2 in nicotine treated cultures suggesting nicotine provided neuroprotection in SCG neurons by increasing the expression of ERK1/2 through nicotinic receptor dependent mechanisms.


Asunto(s)
Neuronas/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Nicotina/farmacología , Ganglio Cervical Superior/citología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Bungarotoxinas/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Mecamilamina/farmacología , Proteína Quinasa 1 Activada por Mitógenos/biosíntesis , Proteína Quinasa 3 Activada por Mitógenos/biosíntesis , Factor de Crecimiento Nervioso/deficiencia , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Ratas , Ganglio Cervical Superior/efectos de los fármacos
2.
Mol Neurobiol ; 55(1): 382-389, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-27957682

RESUMEN

Withdrawal of nerve growth factor (NGF) from sympathetic neurons causes their apoptotic death. Activation of c-Jun NH2-terminal kinase (JNK) may contribute to this death by the induction and phosphorylation of pro-apoptotic Bcl-2 proteins, such as Bax, that are involved in cytochrome c release from mitochondria and reactive oxygen species (ROS) production. Induction of either JNK or ROS may stimulate the other, and both may regulate release of apoptogenic factors from the mitochondria. In order to discern the relationship between JNK and ROS in apoptosis, we treated NGF-deprived, mouse sympathetic neurons with a JNK inhibitor and examined the effect on several important apoptotic events. Block of JNK activation prevented induction of c-Jun expression and resulted in a dose-dependent, yet surprisingly modest, increase in cell survival after 48 h of NGF deprivation. JNK suppression was also not sufficient to prevent the elevation in ROS or the release of cytochrome c from the mitochondria in NGF-deprived sympathetic neurons. Bax deletion prevents apoptotic death of NGF-deprived neurons by preventing release of cytochrome c from their mitochondria. It also prevents increased ROS on NGF deprivation. However, we found that induction of c-Jun in cells lacking Bax was equivalent to that in wild-type neurons. Our results suggest that while JNK activation plays an important role in many forms of apoptosis, it may not be a crucial regulator of Bax-dependent events involved in the apoptotic death of mouse sympathetic neurons deprived of NGF and that ROS is not involved in its activation in these cells.


Asunto(s)
Citocromos c/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Factor de Crecimiento Nervioso/deficiencia , Neuronas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ganglio Cervical Superior/metabolismo , Fibras Adrenérgicas/metabolismo , Fibras Adrenérgicas/patología , Animales , Muerte Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Activación Enzimática/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/patología , Ganglio Cervical Superior/patología
3.
PLoS One ; 11(10): e0165586, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27792755

RESUMEN

Perineural invasion (PNI) is thought to be one of the factors responsible for the high rate of tumor recurrence after surgery and the pain generation associated with pancreatic cancer. Signaling via the nerve growth factor (NGF) pathway between pancreatic cancer cells and the surrounding nerves has been implicated in PNI, and increased levels of these proteins have been correlated to poor prognosis. In this study, we examine the molecular mechanism of the NGF signaling pathway in PNI in pancreatic cancer. We show that knocking down NGF or its receptors, TRKA and p75NTR, or treatment with GW441756, a TRKA kinase inhibitor, reduces the proliferation and migration of pancreatic cancer cells in vitro. Furthermore, pancreatic cancer cells migrate towards dorsal root ganglia (DRG) in a co-culture assay, indicating a paracrine NGF signaling between the DRGs and pancreatic cancer cells. Knocking down the expression of NGF pathway proteins or inhibiting the activity of TRKA by GW441756 reduced the migratory ability of Mia PaCa2 towards the DRGs. Finally, blocking NGF signaling by NGF neutralizing antibodies or GW441756 inhibited the neurite formation in PC-12 cells in response to conditioned media from pancreatic cancer cells, indicating a reciprocal signaling pathway between the pancreatic cancer cells and nerves. Our results indicate that NGF signaling pathway provides a potential target for developing molecularly targeted therapies to decrease PNI and reduce pain generation. Since there are several TRKA antagonists currently in early clinical trials they could now be tested in the clinical situation of pancreatic cancer induced pain.


Asunto(s)
Factor de Crecimiento Nervioso/metabolismo , Sistema Nervioso/patología , Neoplasias Pancreáticas/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Ganglios Espinales/patología , Técnicas de Inactivación de Genes , Humanos , Indoles/farmacología , Invasividad Neoplásica , Factor de Crecimiento Nervioso/deficiencia , Factor de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Sistema Nervioso/efectos de los fármacos , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Receptor trkA/antagonistas & inhibidores , Receptor trkA/deficiencia , Receptor trkA/genética , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/genética
4.
Oxid Med Cell Longev ; 2015: 217258, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26236423

RESUMEN

Dietary antioxidants may be useful in counteracting the chronic inflammatory status in neurodegenerative diseases by reducing oxidative stress due to accumulation of reactive oxygen species (ROS). In this study, we newly described the efficacy of a number of dietary antioxidants (polyphenols, carotenoids, thiolic compounds, and oligoelements) on viability of neuronal PC12 cells following Nerve Growth Factor (NGF) deprivation, a model of age-related decrease of neurotrophic support that triggers neuronal loss. Neuroprotection by antioxidants during NGF deprivation for 24 h was largely dependent on their concentrations: all dietary antioxidants were able to efficiently support cell viability by reducing ROS levels and restoring mitochondrial function, while preserving the neuronal morphology. Moreover, ROS reduction and neuroprotection during NGF withdrawal were also achieved with defined cocktails of 3-6 different antioxidants at concentrations 5-60 times lower than those used in single treatments, suggesting that their antioxidant activity was preserved also at very low concentrations. Overall, these data indicate the beneficial effects of antioxidants against oxidative stress induced by decreased NGF availability and suggest that defined cocktails of dietary factors at low concentrations might be a suitable strategy to reduce oxidative damage in neurodegenerative diseases, while limiting possible side effects.


Asunto(s)
Antioxidantes/farmacología , Factor de Crecimiento Nervioso/deficiencia , Neuroprotección/efectos de los fármacos , Animales , Supervivencia Celular/efectos de los fármacos , Suplementos Dietéticos , Inmunohistoquímica , Microscopía Fluorescente , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Células PC12 , Ratas , Especies Reactivas de Oxígeno/metabolismo
5.
Nat Neurosci ; 17(1): 36-45, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24270184

RESUMEN

Retrograde communication from axonal targets to neuronal cell bodies is critical for both the development and function of the nervous system. Much progress has been made in recent years linking long-distance, retrograde signaling to a signaling endosome, yet the mechanisms governing the trafficking and signaling of these endosomes remain mostly uncharacterized. Here we report that in mouse sympathetic neurons, the target-derived nerve growth factor (NGF)-tropomyosin-related kinase type 1 (TrkA, also called Ntrk1) signaling endosome, on arrival at the cell body, induces the expression and recruitment of a new effector protein known as Coronin-1 (also called Coro1a). In the absence of Coronin-1, the NGF-TrkA signaling endosome fuses to lysosomes sixfold to tenfold faster than when Coronin-1 is intact. We also define a new Coronin-1-dependent trafficking event in which signaling endosomes recycle and re-internalize on arrival at the cell body. Beyond influencing endosomal trafficking, Coronin-1 is also required for several NGF-TrkA-dependent signaling events, including calcium release, calcineurin activation and phosphorylation of cAMP responsive element binding protein (CREB). These results establish Coronin-1 as an essential component of a feedback loop that mediates NGF-TrkA endosome stability, recycling and signaling as a critical mechanism governing developmental competition for survival.


Asunto(s)
Endosomas/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Microfilamentos/metabolismo , Neuronas/fisiología , Transducción de Señal/fisiología , Animales , Animales Recién Nacidos , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Células Cultivadas , Electroporación , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Inmunoprecipitación , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/deficiencia , Factor de Crecimiento Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor trkA/deficiencia , Transducción de Señal/genética , Médula Espinal/citología , Médula Espinal/crecimiento & desarrollo , Médula Espinal/metabolismo , Ganglio Cervical Superior/citología , Transfección , Proteína X Asociada a bcl-2/deficiencia
6.
Cell Death Differ ; 20(12): 1719-30, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24212932

RESUMEN

The mechanisms governing neuron death following NGF deprivation are incompletely understood. Here, we show that Trib3, a protein induced by NGF withdrawal, has a key role in such death via a loop involving the survival kinase Akt and FoxO transcription factors. Trib3 overexpression is sufficient to induce neuron death, and silencing of endogenous Trib3 strongly protects from death when NGF is withdrawn. Mechanism studies reveal that Trib3 interferes with phosphorylation/activity of Akt and contributes to Akt inactivation after NGF deprivation. FoxO1a, a direct Akt substrate, is dephosphorylated upon NGF withdrawal and consequently undergoes nuclear translocation and activates pro-apoptotic genes. We find that Trib3 is required for FoxO1a dephosphorylation and nuclear translocation after NGF deprivation. Conversely, Trib3 induction requires FoxO transcription factors, which show enhanced occupancy of the Trib3 promoter region following NGF withdrawal. Collectively, these findings support a mechanism in which NGF deprivation, Akt dephosphorylation/inactivation, FoxO dephosphorylation/activation and Trib3 induction are linked in a self-amplifying feed-forward loop that culminates in neuron death.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Factor de Crecimiento Nervioso/deficiencia , Neuronas/enzimología , Neuronas/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Muerte Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Factor de Crecimiento Nervioso/farmacología , Proteínas del Tejido Nervioso , Neuronas/efectos de los fármacos , Células PC12 , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , ARN Interferente Pequeño/metabolismo , Ratas , Proteínas Represoras/metabolismo , Especificidad por Sustrato/efectos de los fármacos
7.
Biochem J ; 455(1): 15-25, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23815625

RESUMEN

Neuronal apoptotic death generally requires de novo transcription, and activation of the transcription factor c-Jun has been shown to be necessary in multiple neuronal death paradigms. Caspase-2 has been implicated in death of neuronal and non-neuronal cells, but its relationship to transcriptional activation has not been clearly elucidated. In the present study, using two different neuronal apoptotic paradigms, ß-amyloid treatment and NGF (nerve growth factor) withdrawal, we examined the hierarchical role of caspase-2 activation in the transcriptional control of neuron death. Both paradigms induce rapid activation of caspase-2 as well as activation of the transcription factor c-Jun and subsequent induction of the pro-apoptotic BH3 (Bcl-homology domain 3)-only protein Bim (Bcl-2-interacting mediator of cell death). Caspase-2 activation is dependent on the adaptor protein RAIDD {RIP (receptor-interacting protein)-associated ICH-1 [ICE (interleukin-1ß-converting enzyme)/CED-3 (cell-death determining 3) homologue 1] protein with a death domain}, and both caspase-2 and RAIDD are required for c-Jun activation and Bim induction. The present study thus shows that rapid caspase-2 activation is essential for c-Jun activation and Bim induction in neurons subjected to apoptotic stimuli. This places caspase-2 at an apical position in the apoptotic cascade and demonstrates for the first time that caspase-2 can regulate transcription.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Proteína Adaptadora de Señalización CRADD/genética , Caspasa 2/genética , Proteínas de la Membrana/genética , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas/genética , Activación Transcripcional/efectos de los fármacos , Péptidos beta-Amiloides/farmacología , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Proteína Adaptadora de Señalización CRADD/metabolismo , Caspasa 2/metabolismo , Feto , Proteínas de la Membrana/metabolismo , Factor de Crecimiento Nervioso/deficiencia , Neuronas/citología , Neuronas/efectos de los fármacos , Cultivo Primario de Células , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
8.
Cell Death Differ ; 20(8): 1017-30, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23538417

RESUMEN

ProNGF, the precursor of mature nerve growth factor (NGF), is the most abundant form of NGF in the brain. ProNGF and mature NGF differ significantly in their receptor interaction properties and in their bioactivity. ProNGF increases markedly in the cortex of Alzheimer's disease (AD) brains and proNGF\NGF imbalance has been postulated to play a role in neurodegeneration. However, a direct proof for a causal link between increased proNGF and AD neurodegeneration is lacking. In order to evaluate the consequences of increased levels of proNGF in the postnatal brain, transgenic mice expressing a furin cleavage-resistant form of proNGF, under the control of the neuron-specific mouse Thy1.2 promoter, were derived and characterized. Different transgenic lines displayed a phenotypic gradient of neurodegenerative severity features. We focused the analysis on the two lines TgproNGF#3 and TgproNGF#72, which shared learning and memory impairments in behavioral tests, cholinergic deficit and increased Aß-peptide immunoreactivity. In addition, TgproNGF#3 mice developed Aß oligomer immunoreactivity, as well as late diffuse astrocytosis. Both TgproNGF lines also display electrophysiological alterations related to spontaneous epileptic-like events. The results provide direct evidence that alterations in the proNGF/NGF balance in the adult brain can be an upstream driver of neurodegeneration, contributing to a circular loop linking alterations of proNGF/NGF equilibrium to excitatory/inhibitory synaptic imbalance and amyloid precursor protein (APP) dysmetabolism.


Asunto(s)
Epilepsia/fisiopatología , Homeostasis/fisiología , Discapacidades para el Aprendizaje/fisiopatología , Trastornos de la Memoria/fisiopatología , Factor de Crecimiento Nervioso/fisiología , Enfermedades Neurodegenerativas/fisiopatología , Precursores de Proteínas/fisiología , Envejecimiento/fisiología , Animales , Conducta Animal/fisiología , Modelos Animales de Enfermedad , Hipocampo/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Crecimiento Nervioso/deficiencia , Factor de Crecimiento Nervioso/genética , Fenotipo , Precursores de Proteínas/deficiencia , Precursores de Proteínas/genética
9.
Biochem Biophys Res Commun ; 431(3): 579-85, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23313508

RESUMEN

Sortilin is a member of the family of vacuolar protein sorting 10 protein domain receptors which has emerged as a co-receptor in cell death and neurodegeneration processes mediated by proneurotrophins. Here we tested the possibility that sortilin deficiency interferes with behavioral and neuropathological endpoints in a chronic Nerve Growth factor (NGF)-deprivation model of Alzheimer's disease (AD), the AD10 anti-NGF mouse. AD10 mice show cholinergic deficit, increased APP processing and tau hyper-phosphorylation, resulting in behavioral deficits in learning and memory paradigms assessed by novel object recognition and Morris water maze tests. Sort1(-/-) mice were crossed with AD10 anti-NGF mice and the neurodegenerative phenotype was studied. We found that the loss of sortilin partially protected AD10 anti-NGF mice from neurodegeneration. A protective effect was observed on non-spatial memory as assessed by novel object recognition, and histopathologically at the level of Aß and BFCNs, while the phosphotau increase was unaltered by knocking out sortilin. We suggest that sortilin might be involved in different aspects of neurodegeneration in a complex way, supporting the view that sortilin functions in the CNS are broader than being a co-receptor in proneurotrophin and neurotrophin signaling.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/fisiología , Enfermedad de Alzheimer/metabolismo , Factor de Crecimiento Nervioso/deficiencia , Proteínas Adaptadoras del Transporte Vesicular/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Colina/metabolismo , Neuronas Colinérgicas/metabolismo , Neuronas Colinérgicas/patología , Modelos Animales de Enfermedad , Trastornos de la Memoria/genética , Ratones , Ratones Mutantes , Neuritis/genética , Fosforilación , Modificación Traduccional de las Proteínas , Receptor trkA/biosíntesis , Transducción de Señal , Proteínas tau/metabolismo
10.
J Neurosci ; 32(43): 14885-98, 2012 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-23100411

RESUMEN

Many molecules expressed in the CNS contribute to cognitive functions either by modulating neuronal activity or by mediating neuronal trophic support and/or connectivity. An ongoing discussion is whether signaling of nerve growth factor (NGF) through its high-affinity receptor TrkA contributes to attention behavior and/or learning and memory, based on its expression in relevant regions of the CNS such as the hippocampus, cerebral cortex, amygdala and basal forebrain. Previous animal models carrying either a null allele or transgenic manipulation of Ngf or Trka have proved difficult in addressing this question. To overcome this problem, we conditionally deleted Ngf or Trka from the CNS. Our findings confirm that NGF-TrkA signaling supports survival of only a small proportion of cholinergic neurons during development; however, this signaling is not required for trophic support or connectivity of the remaining basal forebrain cholinergic neurons. Moreover, comprehensive behavioral analysis of young adult and intermediate-aged mice lacking NGF-TrkA signaling demonstrates that this signaling is dispensable for both attention behavior and various aspects of learning and memory.


Asunto(s)
Envejecimiento , Sistema Nervioso Central/metabolismo , Trastornos del Conocimiento/patología , Factor de Crecimiento Nervioso/metabolismo , Receptor trkA/metabolismo , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Atención/fisiología , Reacción de Prevención/fisiología , Recuento de Células/métodos , Sistema Nervioso Central/patología , Conducta de Elección/fisiología , Colina O-Acetiltransferasa/metabolismo , Neuronas Colinérgicas/patología , Trastornos del Conocimiento/fisiopatología , Condicionamiento Psicológico/fisiología , Señales (Psicología) , Modelos Animales de Enfermedad , Conducta Exploratoria/fisiología , Miedo , Etiquetado Corte-Fin in Situ , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Crecimiento Nervioso/deficiencia , Receptor trkA/deficiencia , Receptores de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal/genética
11.
J Neurosci ; 32(39): 13439-53, 2012 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-23015435

RESUMEN

In addition to being a hallmark of neurodegenerative disease, axon degeneration is used during development of the nervous system to prune unwanted connections. In development, axon degeneration is tightly regulated both temporally and spatially. Here, we provide evidence that degeneration cues are transduced through various kinase pathways functioning in spatially distinct compartments to regulate axon degeneration. Intriguingly, glycogen synthase kinase-3 (GSK3) acts centrally, likely modulating gene expression in the cell body to regulate distally restricted axon degeneration. Through a combination of genetic and pharmacological manipulations, including the generation of an analog-sensitive kinase allele mutant mouse for GSK3ß, we show that the ß isoform of GSK3, not the α isoform, is essential for developmental axon pruning in vitro and in vivo. Additionally, we identify the dleu2/mir15a/16-1 cluster, previously characterized as a regulator of B-cell proliferation, and the transcription factor tbx6, as likely downstream effectors of GSK3ß in axon degeneration.


Asunto(s)
Axones/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Degeneración Nerviosa/enzimología , Degeneración Nerviosa/patología , Neuronas/patología , Fosfotransferasas/metabolismo , Transducción de Señal/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Electroporación , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Femenino , Ganglios Espinales/citología , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Genotipo , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Proteínas Fluorescentes Verdes/genética , Hipocampo/citología , Humanos , Inmunoprecipitación , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/prevención & control , Factor de Crecimiento Nervioso/deficiencia , Proteínas del Tejido Nervioso/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Técnicas de Cultivo de Órganos , Fosforilación/fisiología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Células Ganglionares de la Retina/metabolismo , Transducción de Señal/efectos de los fármacos , Transfección , Proteína Fluorescente Roja
12.
J Alzheimers Dis ; 31(1): 1-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22504318

RESUMEN

Several studies suggest that systemic infection occurring during aging and chronic neurodegenerative diseases can evoke an exaggerated immune response that contributes to the progression of neurodegeneration and cognitive decline. However, studies directly addressing the relationship between microbial environment and the onset of neurodegeneration in Alzheimer's disease animal models are lacking. Here we show that the onset of neurodegeneration that transgenic mice develop when raised in conventional husbandry slows down when raising anti-nerve growth factor transgenic mice in a murine pathogen free condition.


Asunto(s)
Factor de Crecimiento Nervioso/deficiencia , Enfermedades Neurodegenerativas/complicaciones , Enfermedades Neurodegenerativas/etiología , Animales , Infecciones por Caliciviridae/complicaciones , Colina O-Acetiltransferasa/metabolismo , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Interleucina-6/sangre , Lipopolisacáridos/toxicidad , Ratones , Ratones Transgénicos , Enfermedades Neurodegenerativas/sangre , Enfermedades Neurodegenerativas/patología , Norovirus/patogenicidad , Tricomoniasis/complicaciones , Proteínas tau/metabolismo
13.
Biochem J ; 444(3): 591-9, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22515271

RESUMEN

Caspase 2 was initially identified as a neuronally expressed developmentally down-regulated gene (HUGO gene nomenclature CASP2) and has been shown to be required for neuronal death induced by several stimuli, including NGF (nerve growth factor) deprivation and Aß (ß-amyloid). In non-neuronal cells the PIDDosome, composed of caspase 2 and two death adaptor proteins, PIDD (p53-inducible protein with a death domain) and RAIDD {RIP (receptor-interacting protein)-associated ICH-1 [ICE (interleukin-1ß-converting enzyme)/CED-3 (cell-death determining 3) homologue 1] protein with a death domain}, has been proposed as the caspase 2 activation complex, although the absolute requirement for the PIDDosome is not clear. To investigate the requirement for the PIDDosome in caspase-2-dependent neuronal death, we have examined the necessity for each component in induction of active caspase 2 and in execution of caspase-2-dependent neuronal death. We find that both NGF deprivation and Aß treatment of neurons induce active caspase 2 and that induction of this activity depends on expression of RAIDD, but is independent of PIDD expression. We show that treatment of wild-type or PIDD-null neurons with Aß or NGF deprivation induces formation of a complex of caspase 2 and RAIDD. We also show that caspase-2-dependent execution of neurons requires RAIDD, not PIDD. Caspase 2 activity can be induced in neurons from PIDD-null mice, and NGF deprivation or Aß use caspase 2 and RAIDD to execute death of these neurons.


Asunto(s)
Proteína Adaptadora de Señalización CRADD/biosíntesis , Caspasa 2/metabolismo , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/biosíntesis , Neuronas/enzimología , Animales , Animales Recién Nacidos , Células Cultivadas , Activación Enzimática/fisiología , Ratones , Ratones Noqueados , Factor de Crecimiento Nervioso/deficiencia , Factor de Crecimiento Nervioso/farmacología , Células PC12 , Ratas , Ratas Sprague-Dawley
14.
Int J Dev Neurosci ; 30(2): 99-103, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22245319

RESUMEN

Nerve growth factor (NGF) is a neurotrophin, which exerts an important role in the development and function of the central and peripheral nervous system. There is limited information regarding the levels of NGF during pregnancy and its role in fetal development. We have earlier reported increased oxidative stress in pregnancy complications. The present study examines the levels of NGF in maternal and cord samples in preterm deliveries and its association with oxidative stress marker. A total number of 96 women delivering preterm (<37 weeks gestation) and 94 women delivering at term (control group) (≥37 weeks gestation) were recruited. Plasma NGF levels were measured in both mother and cord plasma using the Emax Immuno Assay System Promega kit. Maternal and cord plasma NGF levels were significantly reduced (p<0.05 for both) in women delivering preterm as compared to term. There was a positive association between maternal and cord plasma NGF levels (p=0.022). Maternal NGF levels were negatively (p=0.017) associated with maternal malondialdehyde (MDA) levels. Reduced cord NGF levels may affect fetal growth in preterm deliveries which may have implications for the neurodevelopmental pathologies in later life. Circulating maternal NGF levels in preterm pregnancies may be a useful marker to predict NGF levels in the neonate.


Asunto(s)
Discapacidades del Desarrollo/sangre , Recien Nacido Prematuro/crecimiento & desarrollo , Factor de Crecimiento Nervioso/sangre , Factor de Crecimiento Nervioso/deficiencia , Nacimiento Prematuro/sangre , Adulto , Discapacidades del Desarrollo/etiología , Discapacidades del Desarrollo/fisiopatología , Femenino , Humanos , Recién Nacido , Recien Nacido Prematuro/sangre , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Estrés Oxidativo/fisiología , Embarazo , Nacimiento Prematuro/fisiopatología , Estudios Prospectivos , Adulto Joven
15.
J Cell Biol ; 194(5): 751-64, 2011 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-21893599

RESUMEN

The c-Jun N-terminal kinase (JNK) signaling pathway is essential for neuronal degeneration in multiple contexts but also regulates neuronal homeostasis. It remains unclear how neurons are able to dissociate proapoptotic JNK signaling from physiological JNK activity. In this paper, we show that the mixed lineage kinase dual leucine zipper kinase (DLK) selectively regulates the JNK-based stress response pathway to mediate axon degeneration and neuronal apoptosis without influencing other aspects of JNK signaling. This specificity is dependent on interaction of DLK with the scaffolding protein JIP3 to form a specialized JNK signaling complex. Local activation of DLK-based signaling in the axon results in phosphorylation of c-Jun and apoptosis after redistribution of JNK to the cell body. In contrast, regulation of axon degeneration by DLK is c-Jun independent and mediated by distinct JNK substrates. DLK-null mice displayed reduced apoptosis in multiple neuronal populations during development, demonstrating that prodegenerative DLK signaling is required in vivo.


Asunto(s)
Apoptosis/fisiología , Sistema Nervioso Central/embriología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis/efectos de los fármacos , Axones/metabolismo , Axones/patología , Axones/fisiología , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Células Cultivadas , Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ganglios Espinales/citología , Ganglios Espinales/embriología , Ganglios Espinales/patología , Proteínas de Homeodominio/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/deficiencia , Quinasas Quinasa Quinasa PAM/genética , Ratones , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Factor de Crecimiento Nervioso/deficiencia , Factor de Crecimiento Nervioso/farmacología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fosforilación , Unión Proteica/fisiología , Inhibidores de Proteínas Quinasas/farmacocinética , Transporte de Proteínas/fisiología , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , ARN Interferente Pequeño/genética , Médula Espinal/embriología , Médula Espinal/metabolismo , Médula Espinal/patología , Factores de Transcripción/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
J Neural Transm (Vienna) ; 118(8): 1215-25, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21301897

RESUMEN

There is considerable evidence showing that the neurodegenerative processes that lead to sporadic Parkinson's disease (PD) begin many years before the appearance of the characteristic motor symptoms and that impairments in olfactory, cognitive and motor functions are associated with time-dependent disruption of dopaminergic neurotransmission in different brain areas. Midkine is a 13-kDa retinoic acid-induced heparin-binding growth factor involved in many biological processes in the central nervous system such as cell migration, neurogenesis and tissue repair. The abnormal midkine expression may be associated with neurochemical dysfunction in the dopaminergic system and cognitive impairments in rodents. Here, we employed adult midkine knockout mice (Mdk(-/-)) to further investigate the relevance of midkine in dopaminergic neurotransmission and in olfactory, cognitive and motor functions. Mdk(/-) mice displayed pronounced impairments in their olfactory discrimination ability and short-term social recognition memory with no gross motor alterations. Moreover, the genetic deletion of midkine decreased the expression of the enzyme tyrosine hydroxylase in the substantia nigra reducing partially the levels of dopamine and its metabolites in the olfactory bulb and striatum of mice. These findings indicate that the genetic deletion of midkine causes a partial loss of dopaminergic neurons and depletion of dopamine, resulting in olfactory and memory deficits with no major motor impairments. Therefore, Mdk(-/-) mice may represent a promising animal model for the study of the early stages of PD and for testing new therapeutic strategies to restore sensorial and cognitive processes in PD.


Asunto(s)
Citocinas/deficiencia , Modelos Animales de Enfermedad , Eliminación de Gen , Factor de Crecimiento Nervioso/deficiencia , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Animales , Encéfalo/patología , Encéfalo/fisiología , Citocinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Midkina , Factor de Crecimiento Nervioso/genética , Enfermedad de Parkinson/metabolismo , Reconocimiento en Psicología/fisiología , Olfato/genética
17.
J Neurosci Res ; 89(5): 689-99, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21337369

RESUMEN

A significant loss of neurons in the dorsal root ganglia (DRG) has been reported in animal models of peripheral nerve injury. Neonatal sensory neurons are more susceptible than adult neurons to axotomy- or nerve growth factor (NGF) withdrawal-induced cell death. To develop therapies for preventing irreversible sensory cell loss, it is essential to understand the molecular mechanisms responsible for DRG cell death and survival. Here we describe how the expression of the growth arrest- and DNA damage-inducible gene 45α (GADD45A) is correlated with neuronal survival after axotomy in vivo and after NGF withdrawal in vitro. GADD45A expression is low at birth and does not change significantly after spinal nerve ligation (SNL). In contrast, GADD45A is robustly up-regulated in the adult rat DRG 24 hr after SNL, and this up-regulation persists as long as the injured fibers are prevented from regenerating. In vitro delivery of GADD45A protects neonatal rat DRG neurons from NGF withdrawal-induced cytochrome c release and cell death. In addition, in vivo knockdown of GADD45A expression in adult injured DRG by small hairpin RNA increased cell death. Our results indicate that GADD45A protects neuronal cells from SNL-induced cell death.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ganglios Espinales/metabolismo , Degeneración Nerviosa/patología , Degeneración Nerviosa/prevención & control , Proteínas Nucleares/metabolismo , Enfermedades del Sistema Nervioso Periférico/patología , Enfermedades del Sistema Nervioso Periférico/prevención & control , Células Receptoras Sensoriales/efectos de los fármacos , Animales , Animales Recién Nacidos , Axotomía/métodos , Proteínas de Ciclo Celular/genética , Muerte Celular/genética , Modelos Animales de Enfermedad , Ganglios Espinales/patología , Ligandos , Masculino , Degeneración Nerviosa/metabolismo , Factor de Crecimiento Nervioso/deficiencia , Proteínas Nucleares/genética , Enfermedades del Sistema Nervioso Periférico/metabolismo , Ratas , Ratas Sprague-Dawley , Neuropatía Ciática/genética , Neuropatía Ciática/metabolismo , Neuropatía Ciática/prevención & control , Células Receptoras Sensoriales/metabolismo , Regulación hacia Arriba/genética
18.
Neurobiol Aging ; 32(6): 969-90, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19628305

RESUMEN

Alzheimer's disease (AD) is characterized by Aß overproduction and tau hyperphosphorylation. We report that an early, transient and site-specific AD-like tau hyperphosphorylation at Ser262 and Thr231 epitopes is temporally and causally related with an activation of the endogenous amyloidogenic pathway that we previously reported in hippocampal neurons undergoing cell death upon NGF withdrawal [Matrone, C., Ciotti, M.T., Mercanti, D., Marolda, R., Calissano, P., 2008b. NGF and BDNF signaling control amyloidogenic route and Ab production in hippocampal neurons. Proc. Natl. Acad. Sci. 105, 13138-13143]. Such tau hyperphosphorylation, as well as apoptotic death, is (i) blocked by 4G8 and 6E10 Aß antibodies or by specific ß and/or γ-secretases inhibitors; (ii) temporally precedes tau cleavage mediated by a delayed (6-12h after NGF withdrawal) activation of caspase-3 and calpain-I; (iii) under control of Akt-GSK3ß-mediated signaling. Finally, we show that such site-specific tau hyperphosphorylation causes tau detachment from microtubules and an impairment of mitochondrial trafficking. These results depict, for the first time, a rapid interplay between endogenous Aß and tau post-translational modifications which act co-ordinately to compromise neuronal functions in the same neuronal system, under physiological conditions as seen in AD brain.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Neuronas/fisiología , Proteínas tau/metabolismo , Péptidos beta-Amiloides/inmunología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Anticuerpos/farmacología , Transporte Axonal/efectos de los fármacos , Caspasa 3/metabolismo , Muerte Celular/genética , Células Cultivadas , Medio de Cultivo Libre de Suero/farmacología , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Hipocampo/citología , Humanos , Microtúbulos/metabolismo , Factor de Crecimiento Nervioso/deficiencia , Factor de Crecimiento Nervioso/inmunología , Factor de Crecimiento Nervioso/farmacología , Neuronas/efectos de los fármacos , Fosforilación/genética , Embarazo , Unión Proteica/efectos de los fármacos , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Sales de Tetrazolio , Tiazoles , Factores de Tiempo , Proteínas tau/genética
19.
J Neurosci ; 30(39): 13089-94, 2010 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-20881126

RESUMEN

The etiology of Alzheimer's disease (AD) remains elusive. The "amyloid" hypothesis states that toxic action of accumulated ß-amyloid peptide (Aß) on synaptic function causes AD cognitive decline. This hypothesis is supported by analysis of familial AD (FAD)-based transgenic mouse models, where altered amyloid precursor protein (APP) processing leads to Aß accumulation correlating with hippocampal-dependent memory deficits. Some studies report prominent dentate gyrus (DG) glutamatergic plasticity alterations in these mice, while CA1 plasticity remains relatively unaffected. The "neurotrophic unbalance" hypothesis, on the other hand, states that AD-related loss of cholinergic signaling and altered APP processing are due to alterations in nerve growth factor (NGF) trophic support. This hypothesis is supported by analysis of the AD11 mouse, which exhibits chronic NGF deprivation during adulthood and displays AD-like pathology, including Aß accumulation and hippocampal-dependent memory deficits. In this study, we analyzed CA1 and DG glutamatergic plasticity in AD11 mice to evaluate whether these mice also share with FAD models a common phenotype in hippocampal synaptic dysfunction. We report that AD11 mice display age-dependent short- and long-term DG plasticity deficits, while CA1 plasticity remains relatively spared. We also report that both structures exhibit enhanced glutamatergic transmission under lower, yet physiological, neurotransmitter release conditions, a defect that should be considered when further evaluating hippocampal synaptic deficits underlying AD pathology. We conclude that severe deficits in DG plasticity represent another common denominator between these two etiologically different types of AD mouse models, independent of the initial insult (overexpression of FAD mutation or NGF deprivation).


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Hipocampo/metabolismo , Hipocampo/fisiopatología , Factor de Crecimiento Nervioso/deficiencia , Plasticidad Neuronal/genética , Enfermedad de Alzheimer/fisiopatología , Animales , Modelos Animales de Enfermedad , Ácido Glutámico/fisiología , Hipocampo/patología , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Mesencéfalo/metabolismo , Mesencéfalo/patología , Mesencéfalo/fisiopatología , Ratones , Ratones Transgénicos , Factor de Crecimiento Nervioso/genética , Técnicas de Cultivo de Órganos , Vía Perforante/metabolismo , Vía Perforante/patología , Vía Perforante/fisiopatología , Transmisión Sináptica/genética
20.
Neuron ; 67(3): 422-34, 2010 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-20696380

RESUMEN

We report a role for long-distance retrograde neurotrophin signaling in the establishment of synapses in the sympathetic nervous system. Target-derived NGF is both necessary and sufficient for formation of postsynaptic specializations on dendrites of sympathetic neurons. This, in turn, is a prerequisite for formation of presynaptic specializations, but not preganglionic axonal ingrowth from the spinal cord into sympathetic ganglia. We also find that NGF-TrkA signaling endosomes travel from distal axons to cell bodies and dendrites where they promote PSD clustering. Furthermore, the p75 neurotrophin receptor restricts PSD formation, suggesting an important role for antagonistic NGF-TrkA and p75 signaling pathways during retrograde control of synapse establishment. Thus, in addition to defining the appropriate number of sympathetic neurons that survive the period of developmental cell death, target-derived NGF also exerts control over the degree of connectivity between the spinal cord and sympathetic ganglia through retrograde control of synapse assembly.


Asunto(s)
Potenciales Postsinápticos Inhibidores/fisiología , Factor de Crecimiento Nervioso/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factor de Crecimiento Nervioso/deficiencia , Factor de Crecimiento Nervioso/genética , Neuronas/fisiología , Receptor de Factor de Crecimiento Nervioso/deficiencia , Receptor de Factor de Crecimiento Nervioso/genética , Receptor de Factor de Crecimiento Nervioso/fisiología , Receptor trkA/fisiología , Transducción de Señal/fisiología , Médula Espinal/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA