Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Neurosci Res ; 94(6): 579-89, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26707710

RESUMEN

Traumatic brain injury (TBI) is a leading cause of death and disability among young adults and is highly prevalent among recently deployed military personnel. Survivors of TBI often experience cognitive and emotional deficits, suggesting that long-term effects of injury may disrupt neuronal function in critical brain regions, including the amygdala, which is involved in emotion and fear memory. Amygdala hyperexcitability has been reported in both TBI and posttraumatic stress disorder patients, yet little is known regarding the effects of combined stress and TBI on amygdala structure and function at the neuronal level. The present study seeks to determine how the long-term effects of preinjury foot-shock stress and TBI interact to influence synaptic plasticity in the lateral amygdala (LA) of adult male C57BL/6J mice by using whole-cell patch clamp electrophysiology 2-3 months postinjury. In the absence of stress, TBI resulted in a significant increase in membrane excitability and spontaneous excitatory postsynaptic currents (sEPSCs) in LA pyramidal-like neurons. Foot-shock stress in the absence of TBI also resulted in increased sEPSC activity. In contrast, when preinjury stress and TBI occurred in combination, sEPSC activity was significantly decreased compared with either condition alone. There were no significant differences in inhibitory activity or total dendritic length among any of the treatment groups. These results demonstrate that stress and TBI may be contributing to amygdala hyperexcitability via different mechanisms and that these pathways may counterbalance each other with respect to long-term pathophysiology in the LA.


Asunto(s)
Amígdala del Cerebelo/patología , Lesiones Traumáticas del Encéfalo/patología , Potenciales Postsinápticos Excitadores/fisiología , Neuronas/fisiología , Estrés Psicológico/patología , Amígdala del Cerebelo/fisiopatología , Análisis de Varianza , Animales , Biofisica , Dendritas/patología , Modelos Animales de Enfermedad , Estimulación Eléctrica , Electrochoque/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/patología , Técnicas de Placa-Clamp , Estrés Psicológico/etiología
2.
J Neuroinflammation ; 10: 103, 2013 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-23962089

RESUMEN

BACKGROUND: Intracerebral hemorrhage (ICH) is a devastating stroke subtype characterized by a prominent neuroinflammatory response. Antagonism of pro-inflammatory cytokines by specific antibodies represents a compelling therapeutic strategy to improve neurological outcome in patients after ICH. To test this hypothesis, the tumor necrosis factor alpha (TNF-α) antibody CNTO5048 was administered to mice after ICH induction, and histological and functional endpoints were assessed. METHODS: Using 10 to 12-week-old C57BL/6J male mice, ICH was induced by collagenase injection into the left basal ganglia. Brain TNF-α concentration, microglia activation/macrophage recruitment, hematoma volume, cerebral edema, and rotorod latency were assessed in mice treated with the TNF-α antibody, CNTO5048, or vehicle. RESULTS: After ICH induction, mice treated with CNTO5048 demonstrated reduction in microglial activation/macrophage recruitment compared to vehicle-treated animals, as assessed by unbiased stereology (P = 0.049). This reduction in F4/80-positive cells was associated with a reduction in cleaved caspase-3 (P = 0.046) and cerebral edema (P = 0.026) despite similar hematoma volumes, when compared to mice treated with vehicle control. Treatment with CNTO5048 after ICH induction was associated with a reduction in functional deficit when compared to mice treated with vehicle control, as assessed by rotorod latencies (P = 0.024). CONCLUSIONS: Post-injury treatment with the TNF-α antibody CNTO5048 results in less neuroinflammation and improved functional outcomes in a murine model of ICH.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Hemorragia Cerebral/terapia , Recuperación de la Función/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/fisiología , Animales , Hemorragia Cerebral/inmunología , Hemorragia Cerebral/patología , Modelos Animales de Enfermedad , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/terapia , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedades del Sistema Nervioso/inmunología , Enfermedades del Sistema Nervioso/patología , Enfermedades del Sistema Nervioso/terapia , Distribución Aleatoria
3.
J Neurosci ; 31(5): 1688-92, 2011 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-21289177

RESUMEN

Amyloid ß (Aß) and tau protein are both implicated in memory impairment, mild cognitive impairment (MCI), and early Alzheimer's disease (AD), but whether and how they interact is unknown. Consequently, we asked whether tau protein is required for the robust phenomenon of Aß-induced impairment of hippocampal long-term potentiation (LTP), a widely accepted cellular model of memory. We used wild-type mice and mice with a genetic knock-out of tau protein and recorded field potentials in an acute slice preparation. We demonstrate that the absence of tau protein prevents Aß-induced impairment of LTP. Moreover, we show that Aß increases tau phosphorylation and that a specific inhibitor of the tau kinase glycogen synthase kinase 3 blocks the increased tau phosphorylation induced by Aß and prevents Aß-induced impairment of LTP in wild-type mice. Together, these findings show that tau protein is required for Aß to impair synaptic plasticity in the hippocampus and suggest that the Aß-induced impairment of LTP is mediated by tau phosphorylation. We conclude that preventing the interaction between Aß and tau could be a promising strategy for treating cognitive impairment in MCI and early AD.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Glucógeno Sintasa Quinasa 3/farmacología , Hipocampo/fisiopatología , Potenciación a Largo Plazo , Plasticidad Neuronal , Neuronas , Fragmentos de Péptidos/farmacología , Proteínas tau/metabolismo , Animales , Western Blotting , Electrofisiología , Hipocampo/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Fosforilación/efectos de los fármacos , Reacción en Cadena de la Polimerasa , Proteínas tau/deficiencia , Proteínas tau/genética
4.
Anesthesiology ; 116(6): 1299-311, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22487803

RESUMEN

BACKGROUND: Microglial inhibition may reduce secondary tissue injury and improve functional outcome following acute brain injury. Utilizing clinically relevant murine models of traumatic brain injury and intracerebral hemorrhage, neuroinflammatory responses and functional outcome were examined in the presence of a potential microglial inhibitor, TT-301. METHODS: TT-301 or saline was administered following traumatic brain injury or intracerebral hemorrhage, and then for four subsequent days. The effect of TT-301 on neuroinflammatory responses and neuronal viability was assessed, as well as short-term vestibulomotor deficit (Rotorod) and long-term neurocognitive impairment (Morris water maze). Finally differential gene expression profiles of mice treated with TT-301 were compared with those of vehicle. RESULTS: Reduction in F4/80+ staining was demonstrated at 1 and 10 days, but not 28 days, after injury in mice treated with TT-301 (n = 6). These histologic findings were associated with improved neurologic function as assessed by Rotorod, which improved by 52.7% in the treated group by day 7, and Morris water maze latencies, which improved by 232.5% as a function of treatment (n = 12; P < 0.05). Similar benefit was demonstrated following intracerebral hemorrhage, in which treatment with TT-301 was associated with functional neurologic improvement of 39.6% improvement in Rotorod and a reduction in cerebral edema that was independent of hematoma volume (n = 12; P < 0.05). Differential gene expression was evaluated following treatment with TT-301, and hierarchical cluster analysis implicated involvement of the Janus kinase-Signal Transducer and Activator of Transcription pathway after administration of TT-301 (n = 3/group). CONCLUSIONS: Modulation of neuroinflammatory responses through TT-301 administration improved histologic and functional parameters in murine models of acute neurologic injury.


Asunto(s)
Sistema Nervioso Central/lesiones , Activación de Macrófagos/efectos de los fármacos , Microglía/efectos de los fármacos , Piperazinas/farmacología , Piridazinas/farmacología , Piridinas/farmacología , Animales , Análisis de los Gases de la Sangre , Glucemia/metabolismo , Agua Corporal/metabolismo , Química Encefálica , Lesiones Encefálicas/tratamiento farmacológico , Recuento de Células , Línea Celular , Hemorragia Cerebral/inducido químicamente , Hemorragia Cerebral/tratamiento farmacológico , Citocinas/biosíntesis , Expresión Génica/efectos de los fármacos , Inmunohistoquímica , Inflamación/tratamiento farmacológico , Inflamación/patología , Lipopolisacáridos/farmacología , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Equilibrio Postural/efectos de los fármacos , Desempeño Psicomotor/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Resultado del Tratamiento
5.
Neurocrit Care ; 16(2): 316-26, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21989844

RESUMEN

BACKGROUND: Apolipoprotein E has previously been demonstrated to modulate acute brain injury responses, and administration of COG1410, an apoE-mimetic peptide derived from the receptor-binding region of apoE, improves outcome in preclinical models of acute neurological injury. In the current study, we sought to establish the optimal dose and timing of peptide administration associated with improved functional outcome in a murine model of intracerebral hemorrhage (ICH). METHODS: Ten to twelve-week-old C57/BL6 male mice were injured by collagenase-induced ICH and randomly selected to receive either vehicle or one of four doses of COG1410 (0.5, 1, 2, or 4 mg/kg) via tail vein injection at 30 min after injury and then daily for 5 days. The injured mice were euthanized at various time points to assess inflammatory mediators, cerebral edema, and hematoma volume. Over the first 5 days following injury, vestibulomotor function was tested via Rotorod (RR) latency. After an optimal dose was demonstrated, a final cohort of animals was injured with ICH and randomly assigned to receive the first dose of COG1410 or vehicle at increasingly longer treatment initiation times after injury. The mice were then assessed for functional deficit via RR testing over the first 5 days following injury. RESULTS: The mice receiving 2 mg/kg of COG1410 after injury demonstrated reduced functional deficit, decreased brain concentrations of inflammatory proteins, and less cerebral edema, although hematoma volume did not vary. The improved RR performance was maintained when peptide administration was delayed for up to 2 h after ICH. CONCLUSIONS: COG1410 administered at a dose of 2 mg/kg within 2 h after injury improves functional recovery in a murine model of ICH.


Asunto(s)
Apolipoproteínas E/uso terapéutico , Hemorragia Cerebral/tratamiento farmacológico , Péptidos/uso terapéutico , Animales , Apolipoproteínas E/fisiología , Edema Encefálico/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Inflamación/tratamiento farmacológico , Masculino , Ratones , Modelos Animales , Recuperación de la Función/efectos de los fármacos , Resultado del Tratamiento
6.
Neurobiol Dis ; 37(3): 622-9, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20004245

RESUMEN

It has been proposed that deregulation of neuronal glycogen synthase kinase 3 (GSK3) activity may be a key feature in Alzheimer disease pathogenesis. We have previously generated transgenic mice that overexpress GSK3beta in forebrain regions including dentate gyrus (DG), a region involved in learning and memory acquisition. We have found that GSK3 overexpression results in DG degeneration. To test whether tau protein modified by GSK3 plays a role in that neurodegeneration, we have brought GSK3 overexpressing mice to a tau knockout background. Our results indicate that the toxic effect of GSK3 overexpression is milder and slower in the absence of tau. Thus, we suggest that the hyperphosphorylated tau mediates, at least in part, the pathology observed in the brain of GSK3 overexpressing mice.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Hipocampo/metabolismo , Discapacidades para el Aprendizaje/metabolismo , Degeneración Nerviosa/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Animales , Atrofia/genética , Atrofia/metabolismo , Atrofia/patología , Biomarcadores/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/patología , Giro Dentado/metabolismo , Giro Dentado/patología , Giro Dentado/fisiopatología , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Regulación Enzimológica de la Expresión Génica/genética , Gliosis/genética , Gliosis/metabolismo , Gliosis/patología , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Hipocampo/patología , Hipocampo/fisiopatología , Discapacidades para el Aprendizaje/genética , Discapacidades para el Aprendizaje/fisiopatología , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Ovillos Neurofibrilares/genética , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Neuronas/metabolismo , Neuronas/patología , Fosforilación , beta Catenina/metabolismo , Proteínas tau/genética
8.
J Neurochem ; 109(6): 1756-66, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19457097

RESUMEN

Analysis of brain microtubule protein from patients with Alzheimer's disease showed decreased alpha tubulin levels along with increased acetylation of the alpha tubulin subunit, mainly in those microtubules from neurons containing neurofibrillary tau pathology. To determine the relationship of tau protein and increased tubulin acetylation, we studied the effect of tau on the acetylation-deacetylation of tubulin. Our results indicate that tau binds to the tubulin-deacetylase, histone deacetylase 6 (HDAC6), decreasing its activity with a consequent increase in tubulin acetylation. As expected, increased acetylation was also found in tubulin from wild-type mice compared with tubulin from mice lacking tau because of the tau-mediated inhibition of the deacetylase. In addition, we found that an excess of tau protein, as a HDAC6 inhibitor, prevents induction of autophagy by inhibiting proteasome function.


Asunto(s)
Encéfalo/metabolismo , Histona Desacetilasas/metabolismo , Proteínas tau/metabolismo , Acetilación/efectos de los fármacos , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Animales , Ácidos Borónicos/farmacología , Encéfalo/citología , Células Cultivadas , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Femenino , Regulación de la Expresión Génica/fisiología , Histona Desacetilasa 6 , Histona Desacetilasas/genética , Humanos , Inmunoprecipitación/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Cambios Post Mortem , Transfección/métodos , Tubulina (Proteína)/metabolismo , Proteínas tau/deficiencia
9.
J Neurosci ; 27(34): 9155-68, 2007 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-17715352

RESUMEN

Intracellular tau deposits are characteristic of several neurodegenerative disorders called tauopathies. The tau protein regulates the stability and assembly of microtubules by binding to microtubules through three or four microtubule-binding repeats (3R and 4R). The number of microtubule-binding repeats is determined by the inclusion or exclusion of the second microtubule-binding repeat encoded by exon 10 of the TAU gene. TAU gene mutations that alter the inclusion of exon 10, and hence the 4R:3R ratio, are causal in the tauopathy frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17). A mutation located in exon 10 has been identified in several FTDP-17 families that present with increased exon 10 inclusion in both mRNA and protein, parkinsonism, movement disorders, and dementia. We have engineered a human tau minigene construct that was designed to allow alternative splicing of the tau exon 10. Here we demonstrate that transgenic mice expressing human tau protein with this mutation develop neurodegeneration as result of aberrant splicing. The mice recapitulate many of the disease hallmarks that are seen in patients with this mutation, including increased tau exon 10 inclusion in both mRNA and protein, motor and behavioral deficits, and tau protein accumulation in neurons and tufted astrocytes. Furthermore, these mice present with degeneration of the nigrostriatal dopaminergic pathway, suggesting a possible mechanism for parkinsonism in FTDP-17. Additionally, activated caspase-3 immunoreactivity in both neurons and astrocytes implicates the involvement of the apoptotic pathway in the pathology of these mice.


Asunto(s)
Asparagina/genética , Cromosomas Humanos Par 17/genética , Demencia/genética , Lisina/genética , Trastornos Parkinsonianos/genética , Empalme del ARN/genética , Proteínas tau/genética , Factores de Edad , Análisis de Varianza , Animales , Conducta Animal , Sistema Nervioso Central/patología , Embrión de Mamíferos , Exones/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Modelos Moleculares , Actividad Motora/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo
10.
Biochem J ; 397(1): 53-9, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16536727

RESUMEN

The MAPs (microtubule-associated proteins) MAP1B and tau are well known for binding to microtubules and stabilizing these structures. An additional role for MAPs has emerged recently where they appear to participate in the regulation of transport of cargos on the microtubules found in axons. In this role, tau has been associated with the regulation of anterograde axonal transport. We now report that MAP1B is associated with the regulation of retrograde axonal transport of mitochondria. This finding potentially provides precise control of axonal transport by MAPs at several levels: controlling the anterograde or retrograde direction of transport depending on the type of MAP involved, controlling the speed of transport and controlling the stability of the microtubule tracks upon which transport occurs.


Asunto(s)
Transporte Axonal/fisiología , Proteínas Asociadas a Microtúbulos/fisiología , Mitocondrias/metabolismo , Animales , Células Cultivadas , Hipocampo/citología , Ratones , Ratones Endogámicos C57BL
11.
Sci Rep ; 7: 46461, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28429734

RESUMEN

At present, there are no proven pharmacological treatments demonstrated to improve long term functional outcomes following traumatic brain injury(TBI). In the setting of non-penetrating TBI, sterile brain inflammatory responses are associated with the development of cerebral edema, intracranial hypertension, and secondary neuronal injury. There is increasing evidence that endogenous apolipoprotein E(apoE) modifies the neuroinflammatory response through its role in downregulating glial activation, however, the intact apoE holoprotein does not cross the blood-brain barrier due to its size. To address this limitation, we developed a small 5 amino acid apoE mimetic peptide(CN-105) that mimics the polar face of the apoE helical domain involved in receptor interactions. The goal of this study was to investigate the therapeutic potential of CN-105 in a murine model of closed head injury. Treatment with CN-105 was associated with a durable improvement in functional outcomes as assessed by Rotarod and Morris Water Maze and a reduction in positive Fluoro-Jade B stained injured neurons and microglial activation. Administration of CN-105 was also associated with reduction in mRNA expression of a subset of inflammatory and immune-related genes.


Asunto(s)
Apolipoproteínas E/uso terapéutico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Fragmentos de Péptidos/uso terapéutico , Recuperación de la Función/efectos de los fármacos , Animales , Apolipoproteínas E/farmacología , Hipocampo/efectos de los fármacos , Ratones , Modelos Animales , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/farmacología
12.
Ann Clin Transl Neurol ; 4(4): 246-265, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28382306

RESUMEN

OBJECTIVE: At present, the absence of a pharmacological neuroprotectant represents an important unmet clinical need in the treatment of ischemic and traumatic brain injury. Recent evidence suggests that administration of apolipoprotein E mimetic therapies represent a viable therapeutic strategy in this setting. We investigate the neuroprotective and anti-inflammatory properties of the apolipoprotein E mimetic pentapeptide, CN-105, in a microglial cell line and murine model of ischemic stroke. METHODS: Ten to 13-week-old male C57/BL6 mice underwent transient middle cerebral artery occlusion and were randomly selected to receive CN-105 (0.1 mg/kg) in 100 µL volume or vehicle via tail vein injection at various time points. Survival, motor-sensory functional outcomes using rotarod test and 4-limb wire hanging test, infarct volume assessment using 2,3,5-Triphenyltetrazolium chloride staining method, and microglial activation in the contralateral hippocampus using F4/80 immunostaining were assessed at various time points. In vitro assessment of tumor necrosis factor-alpha secretion in a microglial cell line was performed, and phosphoproteomic analysis conducted to explore early mechanistic pathways of CN-105 in ischemic stroke. RESULTS: Mice receiving CN-105 demonstrated improved survival, improved functional outcomes, reduced infarct volume, and reduced microglial activation. CN-105 also suppressed inflammatory cytokines secretion in microglial cells in vitro. Phosphoproteomic signals suggest that CN-105 reduces proinflammatory pathways and lower oxidative stress. INTERPRETATION: CN-105 improves functional and histological outcomes in a murine model of ischemic stroke via modulation of neuroinflammatory pathways. Recent clinical trial of this compound has demonstrated favorable pharmacokinetic and safety profile, suggesting that CN-105 represents an attractive candidate for clinical translation.

13.
PLoS One ; 11(7): e0159435, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27459671

RESUMEN

Amyloid precursor protein (APP) is cleaved by gamma-secretase to simultaneously generate amyloid beta (Aß) and APP Intracellular Domain (AICD) peptides. Aß plays a pivotal role in Alzheimer's disease (AD) pathogenesis but recent studies suggest that amyloid-independent mechanisms also contribute to the disease. We previously showed that AICD transgenic mice (AICD-Tg) exhibit AD-like features such as tau pathology, aberrant neuronal activity, memory deficits and neurodegeneration in an age-dependent manner. Since AD is a tauopathy and tau has been shown to mediate Aß-induced toxicity, we examined the role of tau in AICD-induced pathological features. We report that ablating endogenous tau protects AICD-Tg mice from deficits in adult neurogenesis, seizure severity, short-term memory deficits and neurodegeneration. Deletion of tau restored abnormal phosphorylation of NMDA receptors, which is likely to underlie hyperexcitability and associated excitotoxicity in AICD-Tg mice. Conversely, overexpression of wild-type human tau aggravated receptor phosphorylation, impaired adult neurogenesis, memory deficits and neurodegeneration. Our findings show that tau is essential for mediating the deleterious effects of AICD. Since tau also mediates Aß-induced toxic effects, our findings suggest that tau is a common downstream factor in both amyloid-dependent and-independent pathogenic mechanisms and therefore could be a more effective drug target for therapeutic intervention in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Dominios y Motivos de Interacción de Proteínas , Proteínas tau/metabolismo , Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Animales , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Ácido Kaínico/efectos adversos , Litio/farmacología , Masculino , Aprendizaje por Laberinto , Memoria a Corto Plazo , Ratones , Ratones Transgénicos , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Neuronas/metabolismo , Neuronas/patología , Fenotipo , Fosforilación/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsiones/inducido químicamente , Convulsiones/metabolismo , Proteínas tau/genética
14.
Neuron ; 90(5): 941-7, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27210553

RESUMEN

Pathological evidence for selective four-repeat (4R) tau deposition in certain dementias and exon 10-positioned MAPT mutations together suggest a 4R-specific role in causing disease. However, direct assessments of 4R toxicity have not yet been accomplished in vivo. Increasing 4R-tau expression without change to total tau in human tau-expressing mice induced more severe seizures and nesting behavior abnormality, increased tau phosphorylation, and produced a shift toward oligomeric tau. Exon 10 skipping could also be accomplished in vivo, providing support for a 4R-tau targeted approach to target 4R-tau toxicity and, in cases of primary MAPT mutation, eliminate the disease-causing mutation.


Asunto(s)
Modelos Biológicos , Comportamiento de Nidificación , Convulsiones/metabolismo , Proteínas tau/química , Proteínas tau/metabolismo , Animales , Encéfalo/metabolismo , Exones/genética , Humanos , Infusiones Intraventriculares , Ratones , Mutación/efectos de los fármacos , Comportamiento de Nidificación/efectos de los fármacos , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos Antisentido/farmacología , Fosforilación/efectos de los fármacos , Isoformas de Proteínas/metabolismo , Empalme del ARN/efectos de los fármacos , Empalme del ARN/genética , Convulsiones/inducido químicamente , Convulsiones/genética , Solubilidad , Expansión de Repetición de Trinucleótido/efectos de los fármacos , Proteínas tau/genética , Proteínas tau/toxicidad
15.
PLoS One ; 9(7): e103969, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25080336

RESUMEN

Female sex is associated with improved outcome in experimental brain injury models, such as traumatic brain injury, ischemic stroke, and intracerebral hemorrhage. This implies female gonadal steroids may be neuroprotective. A mechanism for this may involve modulation of post-injury neuroinflammation. As the resident immunomodulatory cells in central nervous system, microglia are activated during acute brain injury and produce inflammatory mediators which contribute to secondary injury including proinflammatory cytokines, and nitric oxide (NO) and prostaglandin E2 (PGE2), mediated by inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2), respectively. We hypothesized that female gonadal steroids reduce microglia mediated neuroinflammation. In this study, the progesterone's effects on tumor necrosis factor alpha (TNF-α), iNOS, and COX-2 expression were investigated in lipopolysaccharide (LPS)-stimulated BV-2 microglia. Further, investigation included nuclear factor kappa B (NF-κB) and mitogen activated protein kinase (MAPK) pathways. LPS (30 ng/ml) upregulated TNF-α, iNOS, and COX-2 protein expression in BV-2 cells. Progesterone pretreatment attenuated LPS-stimulated TNF-α, iNOS, and COX-2 expression in a dose-dependent fashion. Progesterone suppressed LPS-induced NF-κB activation by decreasing inhibitory κBα and NF-κB p65 phosphorylation and p65 nuclear translocation. Progesterone decreased LPS-mediated phosphorylation of p38, c-Jun N-terminal kinase and extracellular regulated kinase MAPKs. These progesterone effects were inhibited by its antagonist mifepristone. In conclusion, progesterone exhibits pleiotropic anti-inflammatory effects in LPS-stimulated BV-2 microglia by down-regulating proinflammatory mediators corresponding to suppression of NF-κB and MAPK activation. This suggests progesterone may be used as a potential neurotherapeutic to treat inflammatory components of acute brain injury.


Asunto(s)
Antiinflamatorios/farmacología , Lipopolisacáridos/farmacología , Microglía/efectos de los fármacos , Progesterona/farmacología , Animales , Línea Celular , Ciclooxigenasa 2/metabolismo , Evaluación Preclínica de Medicamentos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Ratones , Microglía/inmunología , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Receptores de Progesterona/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis
16.
J Neuroimmunol ; 276(1-2): 112-8, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25241288

RESUMEN

Intravenous immunoglobulin (IVIG) may improve neuroinflammation after traumatic brain injury (TBI). IVIG administration after TBI improved rotarod latencies over the first 7 days (p=0.039) and water maze latencies over 29-32 days (p=0.027), decreased F4/80-positive cells at 2 (p=0.001) and 7 days (p<0.001), decreased Fluoro-Jade B-positive cells (p=0.020), increased NeuN-positive cells (p=0.014), decreased IL-6 production at 4 (p=0.032) and 24h (p=0.023), and decreased blood-brain barrier breakdown by IgG extravasation (p=0.001) and brain edema (p=0.006); however, TNF-α concentration was unchanged. IVIG administration was associated with long-term neurobehavioral and histological improvement through modulation of neuroinflammation and blood-brain barrier permeability in a murine TBI model.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/patología , Encéfalo/metabolismo , Inmunoglobulinas Intravenosas/uso terapéutico , Aprendizaje por Laberinto/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Animales , Antígenos de Diferenciación/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiopatología , Encéfalo/efectos de los fármacos , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/etiología , Lesiones Encefálicas/complicaciones , Recuento de Células , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Fluoresceínas , Inmunoglobulinas Intravenosas/farmacología , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Tiempo de Reacción/efectos de los fármacos , Factores de Tiempo
17.
J Neurotrauma ; 29(7): 1388-400, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22233347

RESUMEN

Traumatic brain injury (TBI) and intracerebral hemorrhage (ICH) are leading causes of neurological mortality and disability in the U.S. However, therapeutic options are limited and clinical management remains largely supportive. HMG-CoA reductase inhibitors (statins) have pleiotropic mechanisms of action in the setting of acute brain injury, and have been demonstrated to improve outcomes in preclinical models of ICH and TBI. To facilitate translation to clinical practice, we now characterize the optimal statin and dosing paradigm in murine models of ICH and TBI. In a preclinical model of TBI, mice received vehicle, simvastatin, and rosuvastatin at doses of 1 mg/kg and 5 mg/kg for 5 days after the impact. Immunohistochemistry, differential gene expression, and functional outcomes (rotarod and Morris water maze testing) were assessed to gauge treatment response. Following TBI, administration of rosuvastatin 1 mg/kg was associated with the greatest improvement in functional outcomes. Rosuvastatin treatment was associated with histological evidence of reduced neuronal degeneration at 24 h post-TBI, reduced microgliosis at day 7 post-TBI, and preserved neuronal density in the CA3 region at 35 days post-injury. Administration of rosuvastatin following TBI was also associated with downregulation of inflammatory gene expression in the brain. Following ICH, treatment with simvastatin 1 mg/kg was associated with the greatest improvement in functional outcomes, an effect that was independent of hemorrhage volume. Clinically relevant models of acute brain injury may be used to define variables such as optimal statin and dosing paradigms to facilitate the rational design of pilot clinical trials.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hemorragias Intracraneales/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Investigación Biomédica Traslacional/tendencias , Animales , Daño Encefálico Crónico/tratamiento farmacológico , Daño Encefálico Crónico/fisiopatología , Daño Encefálico Crónico/prevención & control , Lesiones Encefálicas/fisiopatología , Modelos Animales de Enfermedad , Fluorobencenos/farmacología , Fluorobencenos/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hemorragias Intracraneales/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/uso terapéutico , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Rosuvastatina Cálcica , Simvastatina/farmacología , Simvastatina/uso terapéutico , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Investigación Biomédica Traslacional/métodos , Resultado del Tratamiento
18.
FEBS Lett ; 584(11): 2265-70, 2010 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-20338169

RESUMEN

Although tau is mainly located in the cell cytoplasm, mostly bound to tubulin, it may also be found in the nucleus of neurons. Hence, we tested whether tau might play a role in regulating the expression of certain genes by comparing gene expression in mice containing or lacking the tau protein. Our results identified a significant difference in the expression of the smarce1 gene, which codes for the BAF-57 protein, a protein involved in the repression of neuron specific genes. These data suggest a role for tau in neuron maturation.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Expresión Génica , Neuronas/metabolismo , Regulación hacia Arriba , Proteínas tau/metabolismo , Animales , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas del Citoesqueleto/metabolismo , Ratones , Ratones Noqueados , Tubulina (Proteína)/metabolismo
19.
J Neurotrauma ; 27(11): 1983-95, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20812776

RESUMEN

Cognitive impairment is common following traumatic brain injury (TBI), and neuroinflammatory mechanisms may predispose to the development of neurodegenerative disease. Apolipoprotein E (apoE) polymorphisms modify neuroinflammatory responses, and influence both outcome from acute brain injury and the risk of developing neurodegenerative disease. We demonstrate that TBI accelerates neurodegenerative pathology in double-transgenic animals expressing the common human apoE alleles and mutated amyloid precursor protein, and that pathology is exacerbated in the presence of the apoE4 allele. The administration of an apoE-mimetic peptide markedly reduced the development of neurodegenerative pathology in mice homozygous for apoE3 as well as apoE3/E4 heterozygotes. These results demonstrate that TBI accelerates the cardinal neuropathological features of neurodegenerative disease, and establishes the potential for apoE mimetic therapies in reducing pathology associated with neurodegeneration.


Asunto(s)
Apolipoproteínas E/genética , Lesiones Encefálicas/patología , Lesiones Encefálicas/terapia , Terapia Genética , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/terapia , Péptidos beta-Amiloides/metabolismo , Animales , Western Blotting , Encéfalo/patología , Citocinas/biosíntesis , Ensayo de Inmunoadsorción Enzimática , Gliosis/patología , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Actividad Motora/fisiología , Factor de Crecimiento Derivado de Plaquetas/genética , Polimorfismo Genético/genética , Desempeño Psicomotor/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas tau/metabolismo
20.
FEBS Lett ; 583(18): 3063-8, 2009 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-19695252

RESUMEN

Levels of tau phosphorylation are high during the developmental period of intense neurite outgrowth, but decrease later. We here investigated whether tau protein plays a role in adult neurogenesis. First we demonstrate that new neurons generated in the subgranular zone express tau in a hyperphosphorylated form. Phospho-tau expression colocalized with doublecortin but not with glial fibrillary acidic protein, Ki67 or calbindin. The same was observed in the subventricular zone. Tau knockout mice did not show a significant decrease in the number of doublecortin-positive cells, although a deficit in migration was observed. These findings suggest that basal tau phosphorylation present in adult animals is in part due to neurogenesis, and from Tau knockout mice it seems that tau is involved in normal migration of new neurons.


Asunto(s)
Neurogénesis , Neuronas/fisiología , Proteínas tau/fisiología , Animales , Movimiento Celular , Ventrículos Cerebrales , Proteínas de Dominio Doblecortina , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/citología , Neuropéptidos/metabolismo , Fosforilación/fisiología , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA