Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Gerontol A Biol Sci Med Sci ; 76(1): 23-31, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32154567

RESUMEN

The accumulation of amyloid-ß (Aß) is a characteristic event in the pathogenesis of Alzheimer's disease (AD). Aquaporin 1 (AQP1) is a membrane water channel protein belonging to the AQP family. AQP1 levels are elevated in the cerebral cortex during the early stages of AD, but the role of AQP1 in AD pathogenesis is unclear. We first determined the expression and distribution of AQP1 in brain tissue samples of AD patients and two AD mouse models (3xTg-AD and 5xFAD). AQP1 accumulation was observed in vulnerable neurons in the cerebral cortex of AD patients, and in neurons affected by the Aß or tau pathology in the 3xTg-AD and 5xFAD mice. AQP1 levels increased in neurons as aging progressed in the AD mouse models. Stress stimuli increased AQP1 in primary cortical neurons. In response to cellular stress, AQP1 appeared to translocate to endocytic compartments of ß- and γ-secretase activities. Ectopic expression of AQP1 in human neuroblastoma cells overexpressing amyloid precussir protein (APP) with the Swedish mutations reduced ß-secretase (BACE1)-mediated cleavage of APP and reduced Aß production without altering the nonamyloidogenic pathway. Conversely, knockdown of AQP1 enhanced BACE1 activity and Aß production. Immunoprecipitation experiments showed that AQP1 decreased the association of BACE1 with APP. Analysis of a human database showed that the amount of Aß decreases as the expression of AQP1 increases. These results suggest that the upregulation of AQP1 is an adaptive response of neurons to stress that reduces Aß production by inhibiting the binding between BACE1 and APP.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/fisiología , Precursor de Proteína beta-Amiloide/fisiología , Amiloide/biosíntesis , Acuaporina 1/fisiología , Enfermedad de Alzheimer/metabolismo , Animales , Acuaporina 1/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Neuronas/metabolismo
2.
Brain Behav Immun ; 48: 301-12, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26021559

RESUMEN

Neuroinflammation in the central nervous system is detrimental for learning and memory, as evident form epidemiological studies linking developmental defects and maternal exposure to harmful pathogens. Postnatal infections can also induce neuroinflammatory responses with long-term consequences. These inflammatory responses can lead to motor deficits and/or behavioral disabilities. Toll like receptors (TLRs) are a family of innate immune receptors best known as sensors of microbial-associated molecular patterns, and are the first responders to infection. TLR2 forms heterodimers with either TLR1 or TLR6, is activated in response to gram-positive bacterial infections, and is expressed in the brain during embryonic development. We hypothesized that early postnatal TLR2-mediated neuroinflammation would adversely affect cognitive behavior in the adult. Our data indicate that postnatal TLR2 activation affects learning and memory in adult mice in a heterodimer-dependent manner. TLR2/6 activation improved motor function and fear learning, while TLR2/1 activation impaired spatial learning and enhanced fear learning. Moreover, developmental TLR2 deficiency significantly impairs spatial learning and enhances fear learning, stressing the involvement of the TLR2 pathway in learning and memory. Analysis of the transcriptional effects of TLR2 activation reveals both common and unique transcriptional programs following heterodimer-specific TLR2 activation. These results imply that adult cognitive behavior could be influenced in part, by activation or alterations in the TLR2 pathway at birth.


Asunto(s)
Aprendizaje/fisiología , Memoria/fisiología , Destreza Motora/fisiología , Neuronas/metabolismo , Receptor Toll-Like 2/metabolismo , Animales , Condicionamiento Psicológico/fisiología , Conducta Exploratoria/fisiología , Miedo/fisiología , Ratones , Ratones Noqueados , Prueba de Desempeño de Rotación con Aceleración Constante , Aprendizaje Espacial/fisiología , Receptor Toll-Like 2/genética
4.
Neuromolecular Med ; 16(2): 292-307, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24272426

RESUMEN

Multiple sclerosis (MS) is an inflammatory autoimmune disease of the central nervous system (CNS) involving demyelinating and neurodegenerative processes. Several of the major pathological CNS alterations and behavioral deficits of MS are recapitulated in the experimental autoimmune encephalitis (EAE) mouse model in which the disease process is induced by administration of myelin peptides. Development of EAE requires infiltration of inflammatory cytokine-generating monocytes and macrophages, and auto-reactive T cells, into the CNS. Very late antigen-4 (VLA-4, α4ß1) is an integrin molecule that plays a role in inflammatory responses by facilitating the migration of leukocytes across the blood-brain barrier during inflammatory disease, and antibodies against VLA-4 exhibit therapeutic efficacy in mouse and monkey MS models. Here, we report that the tellurium compound AS101 (ammonium trichloro (dioxoethylene-o,o') tellurate) ameliorates EAE by inhibiting monocyte and T cell infiltration into the CNS. CD49d is an alpha subunit of the VLA-4 (α4ß1) integrin. During the peak stage of EAE, AS101 treatment effectively ameliorated the disease process by reducing the number of CD49d(+) inflammatory monocyte/macrophage cells in the spinal cord. AS101 treatment markedly reduced the pro-inflammatory cytokine levels, while increasing anti-inflammatory cytokine levels. In contrast, AS101 treatment did not affect the peripheral populations of CD11b(+) monocytes and macrophages. AS101 treatment reduced the infiltration of CD4(+) and CD49(+)/VLA4 T cells. In addition, treatment of T cells from MS patients with AS101 resulted in apoptosis, while such treatment did not affect T cells from healthy donors. These results suggest that AS101 reduces accumulation of leukocytes in the CNS by inhibiting the activity of the VLA-4 integrin and provide a rationale for the potential use of Tellurium IV compounds for the treatment of MS.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Etilenos/uso terapéutico , Factores Inmunológicos/uso terapéutico , Integrina alfa4beta1/antagonistas & inhibidores , Monocitos/efectos de los fármacos , Médula Espinal/inmunología , Subgrupos de Linfocitos T/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Barrera Hematoencefálica/inmunología , Encéfalo/inmunología , Encéfalo/patología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Quimiotaxis de Leucocito/efectos de los fármacos , Citocinas/biosíntesis , Citocinas/genética , Encefalomielitis Autoinmune Experimental/inmunología , Etilenos/farmacología , Femenino , Humanos , Factores Inmunológicos/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Médula Espinal/metabolismo , Médula Espinal/patología , Bazo/metabolismo , Subgrupos de Linfocitos T/inmunología
5.
Brain Res ; 1529: 200-8, 2013 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-23856323

RESUMEN

Age-associated dysregulation of sleep can be worsened by Alzheimer's disease (AD). AD and sleep restriction both impair cognition, yet it is unknown if mild chronic sleep restriction modifies the proteopathic processes involved in AD. The goal of this work was to test the hypothesis that sleep restriction worsens memory impairments, and amyloid ß-peptide (Aß) and pTau accumulations in the brain in a mouse model of AD, with a focus on a role for circulating glucocorticoids (GC). Male 3xTgAD mice were subjected to sleep restriction (SR) for 6h/day for 6 weeks using the modified multiple platform technique, and behavioral (Morris water maze, fear conditioning, open field) and biochemical (immunoblot) outcomes were compared to mice undergoing daily cage transfers (large cage control; LCC) as well as control mice that remained in their home cage (control; CTL). At one week, both LCC and SR mice displayed significant elevations in plasma corticosterone compared to CTL (p<0.002). By four weeks, SR mice displayed a two-fold increase in circulating corticosterone levels compared to CTL. Behavioral data indicated deficits in contextual and cued memory in SR mice that were not present for LCC or CTL (p<0.04). Both Aß and pTau levels increased in the cortex of SR mice compared to CTL and LCC; however these changes were not noted in the hippocampus. Significant positive correlations between cortical Aß and pTau levels and circulating corticosterone indicate a potential role for GCs in mediating behavioral and biochemical changes observed after sleep restriction in a mouse model of AD.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Corteza Cerebral/metabolismo , Trastornos de la Memoria/etiología , Privación de Sueño/fisiopatología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Condicionamiento Psicológico/fisiología , Corticosterona/sangre , Modelos Animales de Enfermedad , Conducta Exploratoria , Miedo/psicología , Humanos , Masculino , Aprendizaje por Laberinto , Trastornos de la Memoria/sangre , Ratones , Ratones Transgénicos , Mutación/genética , Presenilina-1/genética , Proteínas tau/genética
6.
Neuromolecular Med ; 15(2): 351-63, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23519441

RESUMEN

Tomosyn, a syntaxin-binding protein, is known to inhibit vesicle priming and synaptic transmission via interference with the formation of SNARE complexes. Using a lentiviral vector, we specifically overexpressed tomosyn1 in hippocampal dentate gyrus neurons in adult mice. Mice were then subjected to spatial learning and memory tasks and electrophysiological measurements from hippocampal slices. Tomosyn1-overexpression significantly impaired hippocampus-dependent spatial memory while tested in the Morris water maze. Further, tomosyn1-overexpressing mice utilize swimming strategies of lesser cognitive ability in the Morris water maze compared with control mice. Electrophysiological measurements at mossy fiber-CA3 synapses revealed impaired paired-pulse facilitation in the mossy fiber of tomosyn1-overexpressing mice. This study provides evidence for novel roles for tomosyn1 in hippocampus-dependent spatial learning and memory, potentially via decreased synaptic transmission in mossy fiber-CA3 synapses. Moreover, it provides new insight regarding the role of the hippocampal dentate gyrus and mossy fiber-CA3 synapses in swimming strategy preference, and in learning and memory.


Asunto(s)
Región CA3 Hipocampal/fisiopatología , Giro Dentado/fisiopatología , Discapacidades para el Aprendizaje/genética , Trastornos de la Memoria/genética , Proteínas del Tejido Nervioso/fisiología , Proteínas R-SNARE/fisiología , Animales , Proteínas Bacterianas/genética , Región CA3 Hipocampal/metabolismo , Giro Dentado/metabolismo , Conducta Exploratoria/fisiología , Genes Reporteros , Vectores Genéticos , Discapacidades para el Aprendizaje/fisiopatología , Lentivirus , Proteínas Luminiscentes/genética , Masculino , Aprendizaje por Laberinto , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Fibras Musgosas del Hipocampo/fisiopatología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Plasticidad Neuronal/fisiología , Desempeño Psicomotor/fisiología , Proteínas R-SNARE/biosíntesis , Proteínas R-SNARE/genética , Proteínas Recombinantes de Fusión/metabolismo , Natación , Regulación hacia Arriba
7.
J Neurosci Res ; 91(5): 671-80, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23404341

RESUMEN

Tumor necrosis factor-α (TNF) plays a prominent role in the brain damage and functional deficits that result from ischemic stroke. It was recently reported that the thalidomide analog 3,6'-dithiothalidomide (3,6'-DT) can selectively inhibit the synthesis of TNF in cultured cells. We therefore tested the therapeutic potential of 3,6'-DT in a mouse model of focal ischemic stroke. Administration of 3,6'-DT immediately prior to a stroke or within 3 hr after the stroke reduced infarct volume, neuronal death, and neurological deficits, whereas thalidomide was effective only when administered prior to stroke. Neuroprotection was accompanied by decreased inflammation; 3,6'-DT-treated mice exhibited reduced expression of TNF, interleukin-1ß, and inducible nitric oxide synthase; reduced numbers of activated microglia/macrophages, astrocytes, and neutrophils; and reduced expression of intercellular adhesion molecule-1 in the ischemic brain tissue. 3,6'-DT treatment attenuated stroke-induced disruption of the blood-brain barrier by a mechanism that appears to involve suppression of matrix metalloproteinase-9 and preservation of occludin. Treatment with 3,6'-DT did not reduce ischemic brain damage in mice lacking TNF receptors, consistent with a critical role for suppression of TNF production and TNF signaling in the therapeutic action of 3,6'-DT. These findings suggest that anti-inflammatory mechanisms underlie the therapeutic actions of 3,6-DT in an animal model of stroke.


Asunto(s)
Antiinflamatorios/uso terapéutico , Encefalitis/tratamiento farmacológico , Encefalitis/etiología , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/tratamiento farmacológico , Talidomida/análogos & derivados , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiopatología , Infarto Encefálico/etiología , Infarto Encefálico/prevención & control , Muerte Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Factor Estimulante de Colonias de Granulocitos/metabolismo , Etiquetado Corte-Fin in Situ , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-3/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/genética , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Talidomida/uso terapéutico , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
8.
Neurobiol Aging ; 34(6): 1530-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23276384

RESUMEN

Alzheimer's disease (AD) involves progressive accumulation of amyloid ß-peptide (Aß) and neurofibrillary pathologies, and glucose hypometabolism in brain regions critical for memory. The 3xTgAD mouse model was used to test the hypothesis that a ketone ester-based diet can ameliorate AD pathogenesis. Beginning at a presymptomatic age, 2 groups of male 3xTgAD mice were fed a diet containing a physiological enantiomeric precursor of ketone bodies (KET) or an isocaloric carbohydrate diet. The results of behavioral tests performed at 4 and 7 months after diet initiation revealed that KET-fed mice exhibited significantly less anxiety in 2 different tests. 3xTgAD mice on the KET diet also exhibited significant, albeit relatively subtle, improvements in performance on learning and memory tests. Immunohistochemical analyses revealed that KET-fed mice exhibited decreased Aß deposition in the subiculum, CA1 and CA3 regions of the hippocampus, and the amygdala. KET-fed mice exhibited reduced levels of hyperphosphorylated tau deposition in the same regions of the hippocampus, amygdala, and cortex. Thus, a novel ketone ester can ameliorate proteopathic and behavioral deficits in a mouse AD model.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Ansiedad/metabolismo , Trastornos del Conocimiento/metabolismo , Dieta Cetogénica/métodos , Proteínas tau/metabolismo , Enfermedad de Alzheimer/dietoterapia , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/efectos adversos , Animales , Ansiedad/dietoterapia , Ansiedad/patología , Trastornos del Conocimiento/dietoterapia , Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Distribución Aleatoria , Proteínas tau/efectos adversos
9.
Biomaterials ; 34(9): 2194-201, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23266256

RESUMEN

Rapid and effective wound healing requires a coordinated cellular response involving fibroblasts, keratinocytes and vascular endothelial cells (VECs). Impaired wound healing can result in multiple adverse health outcomes and, although antibiotics can forestall infection, treatments that accelerate wound healing are lacking. We now report that topical application of water soluble cerium oxide nanoparticles (Nanoceria) accelerates the healing of full-thickness dermal wounds in mice by a mechanism that involves enhancement of the proliferation and migration of fibroblasts, keratinocytes and VECs. The Nanoceria penetrated into the wound tissue and reduced oxidative damage to cellular membranes and proteins, suggesting a therapeutic potential for topical treatment of wounds with antioxidant nanoparticles.


Asunto(s)
Antioxidantes/farmacología , Cerio/farmacología , Células Endoteliales/efectos de los fármacos , Queratinocitos/efectos de los fármacos , Nanopartículas/química , Cicatrización de Heridas/efectos de los fármacos , Animales , Antioxidantes/química , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cerio/química , Células Endoteliales/citología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Humanos , Queratinocitos/citología , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Piel/efectos de los fármacos , Piel/lesiones
10.
PLoS One ; 7(10): e47522, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23071817

RESUMEN

Toll-like receptors (TLRs) play essential roles in innate immunity and increasing evidence indicates that these receptors are expressed in neurons, astrocytes and microglia in the brain where they mediate responses to infection, stress and injury. Very little is known about the roles of TLRs in cognition. To test the hypothesis that TLR4 has a role in hippocampus-dependent spatial learning and memory, we used mice deficient for TLR4 and mice receiving chronic TLR4 antagonist infusion to the lateral ventricles in the brain. We found that developmental TLR4 deficiency enhances spatial reference memory acquisition and memory retention, impairs contextual fear-learning and enhances motor functions, traits that were correlated with CREB up-regulation in the hippocampus. TLR4 antagonist infusion into the cerebral ventricles of adult mice did not affect cognitive behavior, but instead affected anxiety responses. Our findings indicate a developmental role for TLR4 in shaping spatial reference memory, and fear learning and memory. Moreover, we show that central TLR4 inhibition using a TLR4 antagonist has no discernible physiological role in regulating spatial and contextual hippocampus-dependent cognitive behavior.


Asunto(s)
Ansiedad/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación de la Expresión Génica/fisiología , Hipocampo/fisiología , Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Receptor Toll-Like 4/fisiología , Análisis de Varianza , Animales , Condicionamiento Psicológico/fisiología , Miedo/fisiología , Hipocampo/metabolismo , Immunoblotting , Infusiones Intraventriculares , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Noqueados , Prueba de Desempeño de Rotación con Aceleración Constante , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/genética
11.
Neurobiol Aging ; 33(7): 1481.e1-5, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22209255

RESUMEN

Huntington's disease (HD) is associated with profound autonomic dysfunction including dysregulation of cardiovascular control often preceding cognitive or motor symptoms. Brain-derived neurotrophic factor (BDNF) levels are decreased in the brains of HD patients and HD mouse models, and restoring BDNF levels prevents neuronal loss and extends survival in HD mice. We reasoned that heart rate changes in HD may be associated with altered BDNF signaling in cardiovascular control nuclei in the brainstem. Here we show that heart rate is elevated in HD (N171-82Q) mice at presymptomatic and early disease stages, and heart rate responses to restraint stress are attenuated. BDNF levels were significantly reduced in brainstem regions containing cardiovascular nuclei in HD mice and human HD patients. Central administration of BDNF restored the heart rate to control levels. Our findings establish a link between diminished BDNF expression in brainstem cardiovascular nuclei and abnormal heart rates in HD mice, and suggest a novel therapeutic target for correcting cardiovascular dysfunction in HD.


Asunto(s)
Tronco Encefálico/metabolismo , Factor Neurotrófico Derivado del Encéfalo/fisiología , Modelos Animales de Enfermedad , Frecuencia Cardíaca/fisiología , Enfermedad de Huntington/metabolismo , Transducción de Señal/fisiología , Animales , Tronco Encefálico/fisiopatología , Humanos , Enfermedad de Huntington/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
12.
Neurobiol Aging ; 33(4): 830.e1-12, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21855175

RESUMEN

Chronic stress may be a risk factor for developing Alzheimer's disease (AD), but most studies of the effects of stress in models of AD utilize acute adverse stressors of questionable clinical relevance. The goal of this work was to determine how chronic psychosocial stress affects behavioral and pathological outcomes in an animal model of AD, and to elucidate underlying mechanisms. A triple-transgenic mouse model of AD (3xTgAD mice) and nontransgenic control mice were used to test for an affect of chronic mild social stress on blood glucose, plasma glucocorticoids, plasma insulin, anxiety, and hippocampal amyloid ß-particle (Aß), phosphorylated tau (ptau), and brain-derived neurotrophic factor (BDNF) levels. Despite the fact that both control and 3xTgAD mice experienced rises in corticosterone during episodes of mild social stress, at the end of the 6-week stress period 3xTgAD mice displayed increased anxiety, elevated levels of Aß oligomers and intraneuronal Aß, and decreased brain-derived neurotrophic factor levels, whereas control mice did not. Findings suggest 3xTgAD mice are more vulnerable than control mice to chronic psychosocial stress, and that such chronic stress exacerbates Aß accumulation and impairs neurotrophic signaling.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Péptidos beta-Amiloides/metabolismo , Ansiedad/etiología , Conducta Animal/fisiología , Conducta Social , Estrés Psicológico/fisiopatología , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Glucemia/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Ayuno , Glucocorticoides/sangre , Hipocampo/patología , Humanos , Insulina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Presenilina-1/genética , Factores de Tiempo , Proteínas tau/genética , Proteínas tau/metabolismo
13.
Neurobiol Aging ; 33(1): 205.e1-18, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20947216

RESUMEN

Activity-dependent modulation of neuronal gene expression promotes neuronal survival and plasticity, and neuronal network activity is perturbed in aging and Alzheimer's disease (AD). Here we show that cerebral cortical neurons respond to chronic suppression of excitability by downregulating the expression of genes and their encoded proteins involved in inhibitory transmission (GABAergic and somatostatin) and Ca(2+) signaling; alterations in pathways involved in lipid metabolism and energy management are also features of silenced neuronal networks. A molecular fingerprint strikingly similar to that of diminished network activity occurs in the human brain during aging and in AD, and opposite changes occur in response to activation of N-methyl-D-aspartate (NMDA) and brain-derived neurotrophic factor (BDNF) receptors in cultured cortical neurons and in mice in response to an enriched environment or electroconvulsive shock. Our findings suggest that reduced inhibitory neurotransmission during aging and in AD may be the result of compensatory responses that, paradoxically, render the neurons vulnerable to Ca(2+)-mediated degeneration.


Asunto(s)
Envejecimiento/genética , Envejecimiento/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Corteza Cerebral/fisiopatología , Expresión Génica , Interneuronas/fisiología , Red Nerviosa/fisiopatología , Animales , Señalización del Calcio/genética , Supervivencia Celular/genética , Células Cultivadas , Corteza Cerebral/citología , Electrochoque , Metabolismo Energético/genética , Ambiente , Humanos , Metabolismo de los Lípidos/genética , Ratones , Ratones Endogámicos C57BL , N-Metilaspartato/fisiología , Plasticidad Neuronal/genética , Receptor trkB/fisiología , Transmisión Sináptica
14.
Hum Mol Genet ; 21(5): 963-77, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22045699

RESUMEN

Endoplasmic reticulum (ER) stress has been implicated as an initiator or contributing factor in neurodegenerative diseases. The mechanisms that lead to ER stress and whereby ER stress contributes to the degenerative cascades remain unclear but their understanding is critical to devising effective therapies. Here we show that knockdown of Herp (Homocysteine-inducible ER stress protein), an ER stress-inducible protein with an ubiquitin-like (UBL) domain, aggravates ER stress-mediated cell death induced by mutant α-synuclein (αSyn) that causes an inherited form of Parkinson's disease (PD). Functionally, Herp plays a role in maintaining ER homeostasis by facilitating proteasome-mediated degradation of ER-resident Ca(2+) release channels. Deletion of the UBL domain or pharmacological inhibition of proteasomes abolishes the Herp-mediated stabilization of ER Ca(2+) homeostasis. Furthermore, knockdown or pharmacological inhibition of ER Ca(2+) release channels ameliorates ER stress, suggesting that impaired homeostatic regulation of Ca(2+) channels promotes a protracted ER stress with the consequent activation of ER stress-associated apoptotic pathways. Interestingly, sustained upregulation of ER stress markers and aberrant accumulation of ER Ca(2+) release channels were detected in transgenic mutant A53T-αSyn mice. Collectively, these data establish a causative link between impaired ER Ca(2+) homeostasis and chronic ER stress in the degenerative cascades induced by mutant αSyn and suggest that Herp is essential for the resolution of ER stress through maintenance of ER Ca(2+) homeostasis. Our findings suggest a therapeutic potential in PD for agents that increase Herp levels or its ER Ca(2+)-stabilizing action.


Asunto(s)
Calcio/metabolismo , Retículo Endoplásmico/fisiología , Proteínas de la Membrana/metabolismo , Estrés Fisiológico , alfa-Sinucleína/metabolismo , Animales , Canales de Calcio/metabolismo , Muerte Celular , Degradación Asociada con el Retículo Endoplásmico , Células HEK293 , Homeostasis , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Proteínas Mutantes/metabolismo , Células PC12 , Interferencia de ARN , Ratas , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , alfa-Sinucleína/genética
15.
J Neurochem ; 120(1): 125-34, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22035068

RESUMEN

Ceruloplasmin (Cp) is a ferroxidase involved in iron metabolism by converting Fe(2+) to Fe(3+), and by regulating cellular iron efflux. In the ceruloplasmin knockout (CpKO) mouse, the deregulation of iron metabolism results in moderate liver and spleen hemosiderosis, but the impact of Cp deficiency on brain neurochemistry and behavior in this animal model is unknown. We found that in contrast to peripheral tissues, iron levels in the hippocampus are significantly reduced in CpKO mice. Although it does not cause any discernable deficits in motor function or learning and memory, Cp deficiency results in heightened anxiety-like behavior in the open field and elevated plus maze tests. This anxiety phenotype is associated with elevated levels of plasma corticosterone. Previous studies provided evidence that anxiety disorders and long-standing stress are associated with reductions in levels of serotonin (5HT) and brain-derived neurotrophic factor (BDNF) in the hippocampus. We found that levels of 5HT and norepinephrine (NE), and the expression of BDNF and its receptor trkB, are significantly reduced in the hippocampus of CpKO mice. Thus, Cp deficiency causes an anxiety phenotype by a mechanism that involves decreased levels of iron, 5HT, NE, and BDNF in the hippocampus.


Asunto(s)
Ansiedad/metabolismo , Ansiedad/psicología , Factor Neurotrófico Derivado del Encéfalo/deficiencia , Ceruloplasmina/deficiencia , Hipocampo/metabolismo , Deficiencias de Hierro , Serotonina/deficiencia , Animales , Química Encefálica/genética , Ceruloplasmina/genética , Corticosterona/sangre , Miedo/fisiología , Suspensión Trasera , Aprendizaje/fisiología , Masculino , Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/fisiología , Equilibrio Postural/fisiología , Desempeño Psicomotor/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Reconocimiento en Psicología/fisiología , Transcripción Genética
16.
PLoS One ; 6(9): e24515, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21931736

RESUMEN

CHD5 is frequently deleted in neuroblastoma and is a tumor suppressor gene. However, little is known about the role of CHD5 other than it is homologous to chromatin remodeling ATPases. We found CHD5 mRNA was restricted to the brain; by contrast, most remodeling ATPases were broadly expressed. CHD5 protein isolated from mouse brain was associated with HDAC2, p66ß, MTA3 and RbAp46 in a megadalton complex. CHD5 protein was detected in several rat brain regions and appeared to be enriched in neurons. CHD5 protein was predominantly nuclear in primary rat neurons and brain sections. Microarray analysis revealed genes that were upregulated and downregulated when CHD5 was depleted from primary neurons. CHD5 depletion altered expression of neuronal genes, transcription factors, and brain-specific subunits of the SWI/SNF remodeling enzyme. Expression of gene sets linked to aging and Alzheimer's disease were strongly altered by CHD5 depletion from primary neurons. Chromatin immunoprecipitation revealed CHD5 bound to these genes, suggesting the regulation was direct. Together, these results indicate that CHD5 protein is found in a NuRD-like multi-protein complex. CHD5 expression is restricted to the brain, unlike the closely related family members CHD3 and CHD4. CHD5 regulates expression of neuronal genes, cell cycle genes and remodeling genes. CHD5 is linked to regulation of genes implicated in aging and Alzheimer's disease.


Asunto(s)
Cromatina/química , ADN Helicasas/biosíntesis , Regulación Enzimológica de la Expresión Génica , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/biosíntesis , Neuronas/metabolismo , Transactivadores/biosíntesis , Envejecimiento , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/enzimología , Encéfalo/fisiología , Inmunoprecipitación de Cromatina , Perfilación de la Expresión Génica , Humanos , Ratones , Complejos Multiproteicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas
17.
Neuromolecular Med ; 13(3): 175-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21660587

RESUMEN

Preclinical evaluation of drugs for neurological disorders is usually performed on overfed rodents, without consideration of how metabolic state might affect drug efficacy. Using a widely employed mouse model of focal ischemic stroke, we found that that the NMDA receptor antagonist dizocilpine (MK-801) reduces brain damage and improves functional outcome in mice on the usual ad libitum diet, but exhibits little or no therapeutic efficacy in mice maintained on an energy-restricted diet. Thus, NMDA receptor activation plays a central role in the mechanism by which a high dietary energy intake exacerbates ischemic brain injury. These findings suggest that inclusion of subjects with a wide range of energy intakes in clinical trials for stroke may mask a drug benefit in the overfed/obese subpopulation of subjects.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Restricción Calórica , Ingestión de Energía , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Isquemia Encefálica/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Accidente Cerebrovascular/patología
18.
J Neurosci ; 31(9): 3500-7, 2011 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-21368062

RESUMEN

Several mouse models of Alzheimer's disease (AD) with abundant ß-amyloid and/or aberrantly phosphorylated tau develop memory impairments. However, multiple non-mnemonic cognitive domains such as attention and executive control are also compromised early in AD individuals. Currently, it is unclear whether mutations in the ß-amyloid precursor protein (APP) and tau are sufficient to cause similar, AD-like attention deficits in mouse models of the disease. To address this question, we tested 3xTgAD mice (which express APPswe, PS1M146V, and tauP301L mutations) and wild-type control mice on a newly developed touchscreen-based 5-choice serial reaction time test of attention and response control. The 3xTgAD mice attended less accurately to short, spatially unpredictable stimuli when the attentional demand of the task was high, and also showed a general tendency to make more perseverative responses than wild-type mice. The attentional impairment of 3xTgAD mice was comparable to that of AD patients in two aspects: first, although 3xTgAD mice initially responded as accurately as wild-type mice, they subsequently failed to sustain their attention over the duration of the task; second, the ability to sustain attention was enhanced by the cholinesterase inhibitor donepezil (Aricept). These findings demonstrate that familial AD mutations not only affect memory, but also cause significant impairments in attention, a cognitive domain supported by the prefrontal cortex and its afferents. Because attention deficits are likely to affect memory encoding and other cognitive abilities, our findings have important consequences for the assessment of disease mechanisms and therapeutics in animal models of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Atención/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Modelos Animales de Enfermedad , Indanos/uso terapéutico , Piperidinas/uso terapéutico , Enfermedad de Alzheimer/genética , Animales , Atención/fisiología , Condicionamiento Operante/fisiología , Donepezilo , Humanos , Indanos/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Piperidinas/administración & dosificación
19.
J Alzheimers Dis ; 24(2): 349-61, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21258150

RESUMEN

The cell fate determinant Numb exists in four alternatively spliced variants that differ in the length of their PTB (phosphotyrosine-binding domain, either lacking or containing an 11 amino acid insertion) and PRR (proline-rich region, either lacking or containing a 48 amino acid insertion). We previously reported that Numb switches from isoforms containing the PTB insertion to isoforms lacking this insertion in neural cultures subjected to stress induced by trophic factor withdrawal. The switch in Numb isoforms enhances the generation of amyloid-ß peptide (Aß), the principle component of senile plaques in Alzheimer's disease (AD). Here we examine the expression of the Numb isoforms in brains from AD patients and triple transgenic (3xTg) AD mice. We found that levels of the Numb isoforms lacking the PTB insertion are significantly elevated in the parietal cortex but not in the cerebellum of AD patients when compared to control subjects. Levels of Numb isoforms lacking the PTB insertion were also elevated in the cortex but not cerebellum of 12 month-old 3xTg AD mice with Aß deposits compared to younger 3xTg-AD mice and to non-transgenic mice. Exposure of cultured neurons to Aß resulted in an increase in the levels of Numb isoforms lacking the PTB domain, consistent with a role for Aß in the aberrant expression of Numb in vulnerable brain regions of AD patients and mice. Collectively, the data show that altered expression of Numb isoforms in vulnerable neurons occurs during AD pathogenesis and suggest a role for Numb in the disease process.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Regulación de la Expresión Génica/genética , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Isoformas de Proteínas/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Corteza Cerebral/citología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Inmunoprecipitación/métodos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Fragmentos de Péptidos/farmacología , Fosfopiruvato Hidratasa/metabolismo , Presenilina-1/genética , Isoformas de Proteínas/genética , Factores de Tiempo , Transfección/métodos , Proteínas de Unión al GTP rab5/metabolismo , Proteínas tau/genética
20.
Cardiovasc Res ; 89(1): 72-8, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20736238

RESUMEN

AIMS: glucagon-like peptide 1 (GLP-1) is an incretin hormone released from the gut in response to food intake. Whereas GLP-1 acts in the periphery to inhibit glucagon secretion and stimulate insulin release, it also acts in the central nervous system to mediate autonomic control of feeding, body temperature, and cardiovascular function. Because of its role as an incretin hormone, GLP-1 receptor analogs are used as a treatment for type 2 diabetes. Central or peripheral administration of GLP-1 increases blood pressure and heart rate, possibly by activating brainstem autonomic nuclei and increasing vagus nerve activity. However, the mechanism(s) by which GLP-1 receptor stimulation affects cardiovascular function are unknown. We used the long-lasting GLP-1 receptor agonist Exendin-4 (Ex-4) to test the hypothesis that GLP-1 signalling modulates central parasympathetic control of heart rate. METHODS AND RESULTS: using a telemetry system, we assessed heart rate in mice during central Ex-4 administration. Heart rate was increased by both acute and chronic central Ex-4 administration. Spectral analysis indicated that the high frequency and low frequency powers of heart rate variability were diminished by Ex-4 treatment. Finally, Ex-4 decreased both excitatory glutamatergic and inhibitory glycinergic neurotransmission to preganglionic parasympathetic cardiac vagal neurons. CONCLUSION: these data suggest that central GLP-1 receptor stimulation diminishes parasympathetic modulation of the heart thereby increasing heart rate.


Asunto(s)
Frecuencia Cardíaca/fisiología , Corazón/inervación , Receptores de Glucagón/fisiología , Transmisión Sináptica/fisiología , Nervio Vago/fisiología , Animales , Exenatida , Receptor del Péptido 1 Similar al Glucagón , Corazón/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Ratones , Sistema Nervioso Parasimpático/efectos de los fármacos , Sistema Nervioso Parasimpático/fisiología , Péptidos/administración & dosificación , Péptidos/farmacología , Receptores de Glucagón/agonistas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Transmisión Sináptica/efectos de los fármacos , Nervio Vago/efectos de los fármacos , Ponzoñas/administración & dosificación , Ponzoñas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...