Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
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.
J Mol Diagn ; 17(5): 616-22, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26134170

RESUMEN

Chromosomal instability is a well-described feature of malignant tumors. Melanomas have typical patterns of chromosomal instability compared with benign nevi, which have minimal DNA copy number change. A few malignant melanomas and their benign counterparts, nevi, prove difficult to diagnose on histopathologic analysis alone, which is currently the gold standard. Quantitative PCR-based assays called duplex ratio tests (DRTs) have been developed by our laboratory for application using DNA from FFPE samples of melanomas and nevi. The reproducibility and accuracy of the DRTs were demonstrated and appropriate correction factors for DNA quality calculated for each assay, based on the results of 108 diploid samples. As a panel, seven DRTs were able to differentiate unambiguous cases of melanoma and nevi with a sensitivity of 87% (95% CI, 83%-91%) and a specificity of 88% (95% CI, 84%-92%) in a series of 145 melanomas and 123 nevi. The DRT scores for 20 nonmetastasizing primary melanomas and 20 metastasizing primary melanomas revealed that DRTs had a marginal benefit as prognostic markers. DRTs have early potential to act as molecular biomarkers of melanoma on FFPE specimens pending validation, and DRTs may have applicability as prognostic markers in melanoma or other tumor types if new DRTs to relevant loci are developed.


Asunto(s)
Biomarcadores de Tumor/genética , Melanoma/diagnóstico , Neoplasias Cutáneas/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN/métodos , Femenino , Humanos , Masculino , Melanoma/genética , Persona de Mediana Edad , Nevo/diagnóstico , Nevo/genética , Sensibilidad y Especificidad , Neoplasias Cutáneas/genética , Adulto Joven
3.
J Exp Med ; 210(8): 1603-19, 2013 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-23878307

RESUMEN

During the initial hours after activation, CD4(+) T cells experience profound changes in gene expression. Co-stimulation via the CD28 receptor is required for efficient activation of naive T cells. However, the transcriptional consequences of CD28 co-stimulation are not completely understood. We performed expression microarray analysis to elucidate the effects of CD28 signals on the transcriptome of activated T cells. We show that the transcription factor DEC1 is highly induced in a CD28-dependent manner upon T cell activation, is involved in essential CD4(+) effector T cell functions, and participates in the transcriptional regulation of several T cell activation pathways, including a large group of CD28-regulated genes. Antigen-specific, DEC1-deficient CD4(+) T cells have cell-intrinsic defects in survival and proliferation. Furthermore, we found that DEC1 is required for the development of experimental autoimmune encephalomyelitis because of its critical role in the production of the proinflammatory cytokines GM-CSF, IFN-γ, and IL-2. Thus, we identify DEC1 as a critical transcriptional mediator in the activation of naive CD4(+) T cells that is required for the development of a T cell-mediated autoimmune disease.


Asunto(s)
Autoinmunidad , Antígenos CD28/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Antígenos CD28/genética , Antígenos CD28/inmunología , Línea Celular , Proliferación Celular , Citocinas/biosíntesis , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-2/metabolismo , Activación de Linfocitos/genética , Ratones , Ratones Noqueados , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Transcriptoma , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética
4.
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
5.
PLoS One ; 7(2): e32008, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22384126

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by a progressive loss of lower motor neurons in the spinal cord. The incretin hormone, glucagon-like peptide-1 (GLP-1), facilitates insulin signaling, and the long acting GLP-1 receptor agonist exendin-4 (Ex-4) is currently used as an anti-diabetic drug. GLP-1 receptors are widely expressed in the brain and spinal cord, and our prior studies have shown that Ex-4 is neuroprotective in several neurodegenerative disease rodent models, including stroke, Parkinson's disease and Alzheimer's disease. Here we hypothesized that Ex-4 may provide neuroprotective activity in ALS, and hence characterized Ex-4 actions in both cell culture (NSC-19 neuroblastoma cells) and in vivo (SOD1 G93A mutant mice) models of ALS. Ex-4 proved to be neurotrophic in NSC-19 cells, elevating choline acetyltransferase (ChAT) activity, as well as neuroprotective, protecting cells from hydrogen peroxide-induced oxidative stress and staurosporine-induced apoptosis. Additionally, in both wild-type SOD1 and mutant SOD1 (G37R) stably transfected NSC-19 cell lines, Ex-4 protected against trophic factor withdrawal-induced toxicity. To assess in vivo translation, SOD1 mutant mice were administered vehicle or Ex-4 at 6-weeks of age onwards to end-stage disease via subcutaneous osmotic pump to provide steady-state infusion. ALS mice treated with Ex-4 showed improved glucose tolerance and normalization of behavior, as assessed by running wheel, compared to control ALS mice. Furthermore, Ex-4 treatment attenuated neuronal cell death in the lumbar spinal cord; immunohistochemical analysis demonstrated the rescue of neuronal markers, such as ChAT, associated with motor neurons. Together, our results suggest that GLP-1 receptor agonists warrant further evaluation to assess whether their neuroprotective potential is of therapeutic relevance in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Neuronas Motoras/metabolismo , Péptidos/farmacología , Ponzoñas/farmacología , Animales , Apoptosis , Línea Celular , Colina O-Acetiltransferasa/metabolismo , Modelos Animales de Enfermedad , Exenatida , Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Péptido 1 Similar al Glucagón/metabolismo , Prueba de Tolerancia a la Glucosa/métodos , Peróxido de Hidrógeno/farmacología , Hipoglucemiantes/farmacología , Insulina/metabolismo , Masculino , Ratones , Estrés Oxidativo , Médula Espinal/metabolismo , Estaurosporina/farmacología , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1
6.
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
7.
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
8.
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
9.
Hum Mol Genet ; 20(4): 659-69, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21106706

RESUMEN

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by expanded polyglutamine repeats in the huntingtin (Htt) protein. Mutant Htt may damage and kill striatal neurons by a mechanism involving reduced production of brain-derived neurotrophic factor (BDNF) and increased oxidative and metabolic stress. Because electroconvulsive shock (ECS) can stimulate the production of BDNF and protect neurons against stress, we determined whether ECS treatment would modify the disease process and provide a therapeutic benefit in a mouse model of HD. ECS (50 mA for 0.2 s) or sham treatment was administered once weekly to male N171-82Q Htt mutant mice beginning at 2 months of age. Endpoints measured included motor function, striatal and cortical pathology, and levels of protein chaperones and BDNF. ECS treatment delayed the onset of motor symptoms and body weight loss and extended the survival of HD mice. Striatal neurodegeneration was attenuated and levels of protein chaperones (Hsp70 and Hsp40) and BDNF were elevated in striatal neurons of ECS-treated compared with sham-treated HD mice. Our findings demonstrate that ECS can increase the resistance of neurons to mutant Htt resulting in improved functional outcome and extended survival. The potential of ECS as an intervention in subjects that inherit the mutant Htt gene merits further consideration.


Asunto(s)
Progresión de la Enfermedad , Electrochoque , Enfermedad de Huntington/patología , Enfermedad de Huntington/terapia , Mutación/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Enfermedad de Huntington/genética , Masculino , Ratones , Ratones Transgénicos , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Transducción de Señal , Análisis de Supervivencia
10.
Curr Opin Endocrinol Diabetes Obes ; 18(1): 28-32, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21102321

RESUMEN

PURPOSE OF REVIEW: To assess factors that influence the tempo of bone mass accrual with emphasis on obesity, exercise, and nutritional factors. RECENT FINDINGS: The prevalence of childhood obesity has increased dramatically throughout the world. Recent studies suggest that adiposity may be detrimental to development of bone strength parameters, and bone mass accrual during growth. Weight-bearing exercise during prepubertal and peripubertal period appears to enhance bone strength parameters. Maternal ultraviolet B radiation exposure and vitamin D status has been shown to have a positive effect on neonatal bone status, which appears to track up to the prepubertal period. Administration of vitamin D with or without calcium, but not calcium alone, during the prepubertal period might be an important 'window' for improving skeletal mineralization. SUMMARY: Obesity in children appears to be detrimental to development of bone strength parameters and bone mass accrual. Weight-bearing exercise during prepubertal and peripubertal period and vitamin D supplementation during pregnancy, infancy, and peripubertal period might be important for bone mass accrual. However, adequately powered randomized controlled trials with follow-up into adulthood are needed to determine if these interventions improve the tempo of bone mass accrual.


Asunto(s)
Desarrollo Óseo/fisiología , Enfermedades del Desarrollo Óseo/complicaciones , Enfermedades del Desarrollo Óseo/etiología , Desarrollo Infantil/fisiología , Pubertad/fisiología , Adolescente , Enfermedades del Desarrollo Óseo/epidemiología , Huesos/anatomía & histología , Niño , Ejercicio Físico/fisiología , Femenino , Humanos , Recién Nacido , Obesidad/complicaciones , Obesidad/epidemiología , Tamaño de los Órganos/fisiología , Embarazo
11.
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
12.
J Comp Neurol ; 518(18): 3803-18, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20653035

RESUMEN

Clathrin-coated vesicles are known to play diverse and pivotal roles in cells. The proper formation of clathrin-coated vesicles is dependent on, and highly regulated by, a large number of clathrin assembly proteins. These assembly proteins likely determine the functional specificity of clathrin-coated vesicles, and together they control a multitude of intracellular trafficking pathways, including those involved in embryonic development. In this study, we focus on two closely related clathrin assembly proteins, AP180 and CALM (clathrin assembly lymphoid myeloid leukemia protein), in the developing embryonic rat brain. We find that AP180 begins to be expressed at embryonic day 14 (E14), but only in postmitotic cells that have acquired a neuronal fate. CALM, on the other hand, is expressed as early as E12, by both neural stem cells and postmitotic neurons. In vitro loss-of-function studies using RNA interference (RNAi) indicate that AP180 and CALM are dispensable for some aspects of embryonic neurogenesis but are required for the growth of postmitotic neurons. These results identify the developmental stage of AP180 and CALM expression and suggest that each protein has distinct functions in neural development.


Asunto(s)
Encéfalo , Embrión de Mamíferos , Proteínas de Ensamble de Clatrina Monoméricas/metabolismo , Animales , Encéfalo/citología , Encéfalo/embriología , Encéfalo/metabolismo , Línea Celular , Clatrina/metabolismo , Vesículas Cubiertas por Clatrina/metabolismo , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/metabolismo , Femenino , Neuronas/citología , Neuronas/metabolismo , Embarazo , Ratas , Células Madre/citología , Células Madre/metabolismo
13.
J Neurochem ; 115(6): 1337-49, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20456019

RESUMEN

Plumbagin (5-hydroxy-2-methyl-1,4 naphthoquinone) is a naturally occurring low molecular weight lipophilic phytochemical derived from roots of plants of the Plumbago genus. Plumbagin has been reported to have several clinically relevant biological activities in non-neural cells, including anti-atherosclerotic, anticoagulant, anticarcinogenic, antitumor, and bactericidal effects. In a recent screen of a panel of botanical pesticides, we identified plumbagin as having neuroprotective activity. In this study, we determined if plumbagin could modify the developmental fate of rat E14.5 embryonic neural progenitor cells (NPC). Plumbagin exhibited no cytotoxicity when applied to cultured NPC at concentrations below 1 µM. At a concentration of 0.1 µM, plumbagin significantly enhanced the proliferation of NPC as indicated by a 17% increase in the percentage of cells incorporating bromo-deoxyuridine. Plumbagin at a concentration of 0.1 pM (but not 0.1 µM), stimulated the production of astrocytes as indicated by increased GFAP expression. Plumbagin selectively induced the proliferation and differentiation of glial progenitor cells without affecting the proliferation or differentiation of neuron-restricted progenitors. Plumbagin (0.1 pM) rapidly activated the transcription factor signal transducer and activator of transcription 3 (Stat3) in NPC, and a Stat3 inhibitor peptide prevented both plumbagin-induced astrocyte formation and proliferation. These findings demonstrate the ability of a low molecular weight naturally occurring phytochemical to control the fate of glial progenitor cells by a mechanism involving the Stat3 signaling pathway.


Asunto(s)
Astrocitos/metabolismo , Células Madre Embrionarias/metabolismo , Naftoquinonas/farmacología , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Factor de Transcripción STAT3/metabolismo , Médula Espinal/metabolismo , Células Madre/metabolismo , Animales , Supervivencia Celular/fisiología , Femenino , Embarazo , Ratas , Ratas Sprague-Dawley , Médula Espinal/citología
14.
J Neurochem ; 112(5): 1316-26, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20028456

RESUMEN

Many phytochemicals function as noxious agents that protect plants against insects and other damaging organisms. However, at subtoxic doses, the same phytochemicals may activate adaptive cellular stress response pathways that can protect cells against a variety of adverse conditions. We screened a panel of botanical pesticides using cultured human and rodent neuronal cell models, and identified plumbagin as a novel potent activator of the nuclear factor E2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway. In vitro, plumbagin increases nuclear localization and transcriptional activity of Nrf2, and induces the expression of the Nrf2/ARE-dependent genes, such as heme oxygenase 1 in human neuroblastoma cells. Plumbagin specifically activates the Nrf2/ARE pathway in primary mixed cultures from ARE-human placental alkaline phosphatase reporter mice. Exposure of neuroblastoma cells and primary cortical neurons to plumbagin provides protection against subsequent oxidative and metabolic insults. The neuroprotective effects of plumbagin are abolished by RNA interference-mediated knockdown of Nrf2 expression. In vivo, administration of plumbagin significantly reduces the amount of brain damage and ameliorates-associated neurological deficits in a mouse model of focal ischemic stroke. Our findings establish precedence for the identification and characterization of neuroprotective phytochemicals based upon their ability to activate adaptive cellular stress response pathways.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Hipoxia/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Naftoquinonas/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Corteza Cerebral/citología , Infarto Cerebral/etiología , Infarto Cerebral/prevención & control , Modelos Animales de Enfermedad , Embrión de Mamíferos , Glucosa/deficiencia , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/genética , Naftoquinonas/metabolismo , Naftoquinonas/farmacología , Neuroblastoma , Examen Neurológico , Neuronas , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Transfección/métodos
15.
Neurobiol Aging ; 31(6): 917-25, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18687504

RESUMEN

Beta-secretase (BACE1), an enzyme responsible for the production of amyloid beta-peptide (Abeta), is increased by oxidative stress and is elevated in the brains of patients with sporadic Alzheimer's disease (AD). Here, we show that oxidative stress fails to induce BACE1 expression in presenilin-1 (gamma-secretase)-deficient cells and in normal cells treated with gamma-secretase inhibitors. Oxidative stress-induced beta-secretase activity and sAPPbeta levels were suppressed by gamma-secretase inhibitors. Levels of gamma- and beta-secretase activities were greater in brain tissue samples from AD patients compared to non-demented control subjects, and the elevated BACE1 level in the brains of 3xTgAD mice was reduced by treatment with a gamma-secretase inhibitor. Our findings suggest that gamma-secretase mediates oxidative stress-induced expression of BACE1 resulting in excessive Abeta production in AD.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Estrés Oxidativo/efectos de los fármacos , Aldehídos/farmacología , Secretasas de la Proteína Precursora del Amiloide/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Enfermedades de los Animales , Animales , Ácido Aspártico Endopeptidasas/genética , Encéfalo/enzimología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/farmacología , Ratones , Ratones Transgénicos , Mutación/genética , Neuroblastoma/patología , Oxidantes/farmacología , Estrés Oxidativo/genética , Fragmentos de Péptidos/metabolismo , Presenilina-1/genética , Presenilina-2/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Proteínas tau/genética
16.
Cancer Biol Ther ; 8(19): 1806-14, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19657234

RESUMEN

Claudin proteins are frequently overexpressed in various tumors such as breast, prostate and ovarian cancer. While their functions in cancer have not been completely elucidated, roles in survival, adhesion and invasion have been suggested. In order to clarify the roles of claudins in ovarian cancer, we have performed gene expression profiling of ovarian surface epithelial cells overexpressing claudin-4 and compared the expression patterns to the parental, non-expressing cells. Claudin-4 expression leads to the differential expression of several genes, including many that have previously been implicated in angiogenesis. In particular, angiogenic cytokines, such as IL-8, were found elevated while genes of the angiostatic interferon pathway were found downregulated. In vitro assays show that claudin-4-expressing cells produce factors that can stimulate angiogenesis as measured by tube formation and migration in HUVEC cells. In addition, an in vivo mouse dorsal skinfold assay confirms that cells expressing claudin-4 secrete factors that can mediate angiogenesis in the dorsal skin of mice. Our data suggest a novel function for claudin-4 in cancer and provide an additional rationale for its common overexpression in human tumors.


Asunto(s)
Proteínas de la Membrana/biosíntesis , Neoplasias Ováricas/irrigación sanguínea , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Claudina-4 , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de la Membrana/genética , Ratones , Análisis por Micromatrices , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Transfección
17.
Exp Neurol ; 215(2): 236-42, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19013457

RESUMEN

Brain iron insufficiency has been implicated in several neurological disorders. The dopamine system is consistently altered in studies of iron deficiency in rodent models. Changes in striatal dopamine D(2) receptors are directly proportional to the degree of iron deficiency. In light of the unknown mechanism for the iron deficiency-dopamine connection and because of the known interplay between adenosinergic and dopaminergic systems in the striatum we examined the effects of iron deficiency on the adenosine system. We first attempted to assess whether there is a functional change in the levels of adenosine receptors in response to this low iron. Mice made iron-deficient by diet had an increase in the density of striatal adenosine A(2A) (A(2A)R) but not A(1) receptor (A(1)R) compared to mice on a normal diet. Between two inbred murine strains, which had 2-fold differences in their striatal iron concentrations under normal dietary conditions, the strain with the lower striatal iron had the highest striatal A(2A)R density. Treatment of SH-SY5Y (human neuroblastoma) cells with an iron chelator resulted in increased density of A(2A)R. In these cells, A(2A)R agonist-induced cyclic AMP production was enhanced in response to iron chelation, also demonstrating a functional upregulation of A(2A)R. A significant correlation (r(2)=0.79) was found between a primary marker of cellular iron status (transferrin receptor (TfR)) and A(2A)R protein density. In conclusion, the A(2A)R is increased across different iron-insufficient conditions. The relation between A(2A)R and cellular iron status may be an important pathway by which adenosine may alter the function of the dopaminergic system.


Asunto(s)
Cuerpo Estriado/metabolismo , Deficiencias de Hierro , Neuroblastoma/metabolismo , Receptores de Adenosina A2/metabolismo , Regulación hacia Arriba/fisiología , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Línea Celular Tumoral , AMP Cíclico/metabolismo , Deferoxamina/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Hierro de la Dieta/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Fenetilaminas/farmacología , Receptores de Adenosina A2/genética , Receptores de Transferrina/agonistas , Receptores de Transferrina/genética , Receptores de Transferrina/metabolismo , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos
18.
PLoS One ; 2(11): e1167, 2007 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-18000539

RESUMEN

The Notch signaling pathway is critically involved in cell fate decisions during development of many tissues and organs. In the present study we employed in vivo and cell culture models to elucidate the role of Notch signaling in wound healing. The healing of full-thickness dermal wounds was significantly delayed in Notch antisense transgenic mice and in normal mice treated with gamma-secretase inhibitors that block proteolytic cleavage and activation of Notch. In contrast, mice treated with a Notch ligand Jagged peptide showed significantly enhanced wound healing compared to controls. Activation or inhibition of Notch signaling altered the behaviors of cultured vascular endothelial cells, keratinocytes and fibroblasts in a scratch wound healing model in ways consistent with roles for Notch signaling in wound healing functions all three cell types. These results suggest that Notch signaling plays important roles in wound healing and tissue repair, and that targeting the Notch pathway might provide a novel strategy for treatment of wounds and for modulation of angiogenesis in other pathological conditions.


Asunto(s)
Receptores Notch/metabolismo , Transducción de Señal , Cicatrización de Heridas , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Animales , Proteínas de Unión al Calcio/administración & dosificación , Péptidos y Proteínas de Señalización Intercelular/administración & dosificación , Proteína Jagged-1 , Proteínas de la Membrana/administración & dosificación , Ratones , Ratones Transgénicos , Proteínas Serrate-Jagged
19.
Nat Med ; 12(6): 621-3, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16680150

RESUMEN

Mice transgenic for antisense Notch and normal mice treated with inhibitors of the Notch-activating enzyme gamma-secretase showed reduced damage to brain cells and improved functional outcome in a model of focal ischemic stroke. Notch endangers neurons by modulating pathways that increase their vulnerability to apoptosis, and by activating microglial cells and stimulating the infiltration of proinflammatory leukocytes. These findings suggest that Notch signaling may be a therapeutic target for treatment of stroke and related neurodegenerative conditions.


Asunto(s)
Isquemia Encefálica/patología , Encéfalo/patología , Endopeptidasas/metabolismo , Receptor Notch1/metabolismo , Transducción de Señal/fisiología , Accidente Cerebrovascular/patología , Secretasas de la Proteína Precursora del Amiloide , Animales , Apoptosis , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/terapia , Células Cultivadas , Endopeptidasas/genética , Inhibidores Enzimáticos/metabolismo , Humanos , Leucocitos/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Microglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Péptidos/genética , Péptidos/metabolismo , Ratas , Receptor Notch1/genética , Daño por Reperfusión , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/terapia , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA