Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(3)2021 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-33530349

RESUMEN

Tauopathies are a group of more than twenty known disorders that involve progressive neurodegeneration, cognitive decline and pathological tau accumulation. Current therapeutic strategies provide only limited, late-stage symptomatic treatment. This is partly due to lack of understanding of the molecular mechanisms linking tau and cellular dysfunction, especially during the early stages of disease progression. In this study, we treated early stage tau transgenic mice with a multi-target kinase inhibitor to identify novel substrates that contribute to cognitive impairment and exhibit therapeutic potential. Drug treatment significantly ameliorated brain atrophy and cognitive function as determined by behavioral testing and a sensitive imaging technique called manganese-enhanced magnetic resonance imaging (MEMRI) with quantitative R1 mapping. Surprisingly, these benefits occurred despite unchanged hyperphosphorylated tau levels. To elucidate the mechanism behind these improved cognitive outcomes, we performed quantitative proteomics to determine the altered protein network during this early stage in tauopathy and compare this model with the human Alzheimer's disease (AD) proteome. We identified a cluster of preserved pathways shared with human tauopathy with striking potential for broad multi-target kinase intervention. We further report high confidence candidate proteins as novel therapeutically relevant targets for the treatment of tauopathy. Proteomics data are available via ProteomeXchange with identifier PXD023562.


Asunto(s)
Neuronas/efectos de los fármacos , Neuronas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Tauopatías/etiología , Tauopatías/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Ratones , Ratones Transgénicos , Enfermedades Neurodegenerativas/diagnóstico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteoma , Proteómica/métodos , Índice de Severidad de la Enfermedad , Tauopatías/diagnóstico , Tauopatías/tratamiento farmacológico , Respuesta de Proteína Desplegada , eIF-2 Quinasa/metabolismo , Proteínas tau/metabolismo
2.
J Neurosci ; 36(3): 1001-7, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26791227

RESUMEN

One of the most common symptoms of Alzheimer's disease (AD) and related tauopathies is memory loss. The exact mechanisms leading to memory loss in tauopathies are not yet known; however, decreased translation due to ribosomal dysfunction has been implicated as a part of this process. Here we use a proteomics approach that incorporates subcellular fractionation and coimmunoprecipitation of tau from human AD and non-demented control brains to identify novel interactions between tau and the endoplasmic reticulum (ER). We show that ribosomes associate more closely with tau in AD than with tau in control brains, and that this abnormal association leads to a decrease in RNA translation. The aberrant tau-ribosome association also impaired synthesis of the synaptic protein PSD-95, suggesting that this phenomenon contributes to synaptic dysfunction. These findings provide novel information about tau-protein interactions in human brains, and they describe, for the first time, a dysfunctional consequence of tau-ribosome associations that directly alters protein synthesis. Significance statement: Despite the identification of abnormal tau-ribosomal interactions in tauopathies >25 years ago, the consequences of this association remained elusive until now. Here, we show that pathological tau associates closely with ribosomes in AD brains, and that this interaction impairs protein synthesis. The overall result is a stark reduction of nascent proteins, including those that participate in synaptic plasticity, which is crucial for learning and memory. These data mechanistically link a common pathologic sign, such as the appearance of pathological tau inside brain cells, with cognitive impairments evident in virtually all tauopathies.


Asunto(s)
Neuronas/metabolismo , Neuronas/patología , Biosíntesis de Proteínas/fisiología , Ribosomas/fisiología , Proteínas tau/biosíntesis , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Femenino , Humanos , Masculino , Microsomas/metabolismo , Microsomas/patología , Tauopatías/metabolismo , Tauopatías/patología
3.
Neuron ; 85(1): 101-115, 2015 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-25533482

RESUMEN

Abnormal NFκB activation has been implicated in Alzheimer's disease (AD). However, the signaling pathways governing NFκB regulation and function in the brain are poorly understood. We identify complement protein C3 as an astroglial target of NFκB and show that C3 release acts through neuronal C3aR to disrupt dendritic morphology and network function. Exposure to Aß activates astroglial NFκB and C3 release, consistent with the high levels of C3 expression in brain tissue from AD patients and APP transgenic mice, where C3aR antagonist treatment rescues cognitive impairment. Therefore, dysregulation of neuron-glia interaction through NFκB/C3/C3aR signaling may contribute to synaptic dysfunction in AD, and C3aR antagonists may be therapeutically beneficial.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Astrocitos/metabolismo , Complemento C3/metabolismo , FN-kappa B/metabolismo , Neuronas/metabolismo , Receptores de Complemento/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/metabolismo , Humanos , Ratones , Ratones Transgénicos , Microscopía Confocal , Neuronas/patología , Receptores de Complemento/antagonistas & inhibidores , Transducción de Señal
4.
Curr Alzheimer Res ; 11(10): 928-40, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25387341

RESUMEN

Amyloid formation is the pathological hallmark of type 2 diabetes (T2D) and Alzheimer's disease (AD). These diseases are marked by extracellular amyloid deposits of islet amyloid polypeptide (IAPP) in the pancreas and amyloid ß (Aß) in the brain. Since IAPP may enter the brain and disparate amyloids can cross-seed each other to augment amyloid formation, we hypothesized that pancreatic derived IAPP may enter the brain to augment misfolding of Aß in AD. The corollaries for validity of this hypothesis are that IAPP [1] enters the brain, [2] augments Aß misfolding, [3] associates with Aß plaques, and most importantly [4] plasma levels correlate with AD diagnosis. We demonstrate the first 3 corollaries that: (1) IAPP is present in the brain in human cerebrospinal fluid (CSF), (2) synthetic IAPP promoted oligomerization of Aß in vitro, and (3) endogenous IAPP localized to Aß oligomers and plaques. For the 4th corollary, we did not observe correlation of peripheral IAPP levels with AD pathology in either an African American cohort or AD transgenic mice. In the African American cohort, with increased risk for both T2D and AD, peripheral IAPP levels were not significantly different in samples with no disease, T2D, AD, or both T2D and AD. In the Tg2576 AD mouse model, IAPP plasma levels were not significantly elevated at an age where the mice exhibit the glucose intolerance of pre-diabetes. Based on this negative data, it appears unlikely that peripheral IAPP cross-seeds or "infects" Aß pathology in AD brain. However, we provide novel and additional data which demonstrate that IAPP protein is present in astrocytes in murine brain and secreted from primary cultured astrocytes. This preliminary report suggests a potential and novel association between brain derived IAPP and AD, however whether astrocytic derived IAPP cross-seeds Aß in the brain requires further research.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Negro o Afroamericano , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Animales Recién Nacidos , Encéfalo/citología , Células Cultivadas , Estudios de Cohortes , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Placa Amiloide/metabolismo , Placa Amiloide/patología , Ratas , Ratas Sprague-Dawley , Distribución Tisular/efectos de los fármacos
5.
J Neurosci ; 34(40): 13314-25, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25274811

RESUMEN

Impaired neurogenesis in the adult hippocampus has been implicated in AD pathogenesis. Here we reveal that the APP plays an important role in the neural progenitor proliferation and newborn neuron maturation in the mouse dentate gyrus. APP controls adult neurogenesis through a non cell-autonomous mechanism by GABAergic neurons, as selective deletion of GABAergic, but not glutamatergic, APP disrupts adult hippocampal neurogenesis. APP, highly expressed in the majority of GABAergic neurons in the dentate gyrus, enhances the inhibitory tone to granule cells. By regulating both tonic and phasic GABAergic inputs to dentate granule cells, APP maintains excitatory-inhibitory balance and preserves cognitive functions. Our studies uncover an indispensable role of APP in the GABAergic system for controlling adult hippocampal neurogenesis, and our findings indicate that APP dysfunction may contribute to impaired neurogenesis and cognitive decline associated with AD.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Neuronas GABAérgicas/fisiología , Hipocampo/citología , Interneuronas/fisiología , Neurogénesis/fisiología , Factores de Edad , Precursor de Proteína beta-Amiloide/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Células Cultivadas , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/deficiencia , Nestina/genética , Nestina/metabolismo , Neurogénesis/genética , Bloqueadores de los Canales de Sodio/farmacología , Transmisión Sináptica/genética , Tetrodotoxina/farmacología , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/deficiencia
6.
ACS Chem Neurosci ; 4(3): 379-84, 2013 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-23509974

RESUMEN

Alzheimer's disease (AD) is a devastating neurodegenerative disease most notably characterized by the misfolding of amyloid-ß (Aß) into fibrils and its accumulation into plaques. In this Article, we utilize the affinity of Aß fibrils to bind metal cations and subsequently imprint their chirality to bound molecules to develop novel imaging compounds for staining Aß aggregates. Here, we investigate the cationic dye ruthenium red (ammoniated ruthenium oxychloride) that binds calcium-binding proteins, as a labeling agent for Aß deposits. Ruthenium red stained amyloid plaques red under light microscopy, and exhibited birefringence under crossed polarizers when bound to Aß plaques in brain tissue sections from the Tg2576 mouse model of AD. Staining of Aß plaques was confirmed via staining of the same sections with the fluorescent amyloid binding dye Thioflavin S. In addition, it was confirmed that divalent cations such as calcium displace ruthenium red, consistent with a mechanism of binding by electrostatic interaction. We further characterized the interaction of ruthenium red with synthetic Aß fibrils using independent biophysical techniques. Ruthenium red exhibited birefringence and induced circular dichroic bands at 540 nm upon binding to Aß fibrils due to induced chirality. Thus, the chirality and cation binding properties of Aß aggregates could be capitalized for the development of novel amyloid labeling methods, adding to the arsenal of AD imaging techniques and diagnostic tools.


Asunto(s)
Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Rojo de Rutenio/química , Coloración y Etiquetado/métodos , Animales , Birrefringencia , Química Encefálica/efectos de los fármacos , Química Encefálica/genética , Colorimetría/métodos , Humanos , Ratones , Ratones Transgénicos , Impresión Molecular/métodos , Placa Amiloide/química , Placa Amiloide/genética , Unión Proteica/fisiología , Rojo de Rutenio/metabolismo
7.
J Neurosci ; 32(47): 16725-35a, 2012 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-23175826

RESUMEN

We previously reported that the peroxisome proliferator-activated receptor γ (PPARγ) agonist rosiglitazone (RSG) improved hippocampus-dependent cognition in the Alzheimer's disease (AD) mouse model, Tg2576. RSG had no effect on wild-type littermate cognitive performance. Since extracellular signal-regulated protein kinase mitogen-activated protein kinase (ERK MAPK) is required for many forms of learning and memory that are affected in AD, and since both PPARγ and ERK MAPK are key mediators of insulin signaling, the current study tested the hypothesis that RSG-mediated cognitive improvement induces a hippocampal PPARγ pattern of gene and protein expression that converges with the ERK MAPK signaling axis in Tg2576 AD mice. In the hippocampal PPARγ transcriptome, we found significant overlap between peroxisome proliferator response element-containing PPARγ target genes and ERK-regulated, cAMP response element-containing target genes. Within the Tg2576 dentate gyrus proteome, RSG induced proteins with structural, energy, biosynthesis and plasticity functions. Several of these proteins are known to be important for cognitive function and are also regulated by ERK MAPK. In addition, we found the RSG-mediated augmentation of PPARγ and ERK2 activity during Tg2576 cognitive enhancement was reversed when hippocampal PPARγ was pharmacologically antagonized, revealing a coordinate relationship between PPARγ transcriptional competency and phosphorylated ERK that is reciprocally affected in response to chronic activation, compared with acute inhibition, of PPARγ. We conclude that the hippocampal transcriptome and proteome induced by cognitive enhancement with RSG harnesses a dysregulated ERK MAPK signal transduction pathway to overcome AD-like cognitive deficits in Tg2576 mice. Thus, PPARγ represents a signaling system that is not crucial for normal cognition yet can intercede to restore neural networks compromised by AD.


Asunto(s)
Hipocampo/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Nootrópicos/farmacología , PPAR gamma/fisiología , Transducción de Señal/fisiología , Tiazolidinedionas/farmacología , Péptidos beta-Amiloides/metabolismo , Animales , Western Blotting , Núcleo Celular/fisiología , Condicionamiento Psicológico , Electrochoque , Miedo , Femenino , Inyecciones Intraventriculares , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR gamma/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa , Rosiglitazona , Espectrometría de Masas en Tándem , Transcriptoma/fisiología
8.
Mol Neurodegener ; 7: 47, 2012 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-22992283

RESUMEN

BACKGROUND: The transcription factor NFκB is an important mediator of cell survival and inflammation in the immune system. In the central nervous system (CNS), NFκB signaling has been implicated in regulating neuronal survival following acute pathologic damage such as traumatic brain injury (TBI) and stroke. NFκB is normally bound by the principal inhibitory protein, IκBα, and sequestered in the cytoplasm. Activation of NFκB requires the degradation of IκBα, thereby freeing NFκB to translocate to the nucleus and activate the target genes. Mice deficient in IκBα display deregulated and sustained NFκB activation and early postnatal lethality, highlighting a critical role of IκBα in NFκB regulation. RESULTS: We investigated the role of IκBα in regulating NFκB activity in the brain and the effects of the NFκB/IκBα pathway in mediating neuroinflammation under both physiological and brain injury conditions. We report that astrocytes, but not neurons, exhibit prominent NFκB activity, and that basal NFκB activity in astrocytes is elevated in the absence of IκBα. By generating mice with brain-specific deletion of IκBα, we show that IκBα deficiency does not compromise normal brain development. However, basal neuroinflammation detected by GFAP and Iba1 immunoreactivity is elevated. This leads to impaired inflammatory responses following TBI and worsened brain damage including higher blood brain barrier permeability, increased injury volumes and enlarged ventricle volumes. CONCLUSIONS: We conclude that, in the CNS, astrocyte is the primary cell type subject to NFκB regulation. We further demonstrate that IκBα plays an important role in regulating NFκB activity in the brain and a robust NFκB/IκBα-mediated neuroinflammatory response immediately following TBI is beneficial.


Asunto(s)
Astrocitos/metabolismo , Lesiones Encefálicas/metabolismo , Encéfalo/metabolismo , Proteínas I-kappa B/deficiencia , Recuperación de la Función/fisiología , Animales , Barrera Hematoencefálica/fisiología , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Lesiones Encefálicas/patología , Proteína Ácida Fibrilar de la Glía , Inflamación/metabolismo , Ratones , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/patología , Fosforilación , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
9.
Behav Brain Res ; 216(1): 255-61, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20709114

RESUMEN

Converging lines of evidence associate gluco-regulatory abnormalities and peroxisome-proliferator-activated receptor (PPAR) gamma function with increased risk for Alzheimer's disease (AD). In this study, we used the Tg2576 AD mouse model to test the hypothesis that cognitive improvement following 1 month of PPAR gamma agonism with rosiglitazone (RTZ) correlates with peripheral gluco-regulatory status. We assessed cognition and peripheral gluco-regulatory status of Tg2576 mice following 1 month treatment with RTZ initiated prior to, coincident with, or after, the onset of peripheral gluco-regulatory abnormalities (4, 8, and 12 months of age, respectively). Whereas 5 months old (MO) and 13 MO Tg2576 did not gain cognitive improvement after 1 month treatment with RTZ, 9 MO Tg2576 mice exhibited reversal of associative learning and memory deficits. Peripheral gluco-regulatory abnormalities were improved in 9 and 13 MO Tg2576 with RTZ treatment; RTZ treatment had no effect on the normal glucose status of 5 MO Tg2576 mice. These findings suggest that RTZ-mediated cognitive improvement does not correlate with peripheral gluco-regulatory abnormalities per se, but reflects the age-dependent mechanistic differences that underlie cognitive decline in this mouse model.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Aprendizaje por Asociación/efectos de los fármacos , Glucemia/metabolismo , Cognición/efectos de los fármacos , Memoria/efectos de los fármacos , Tiazolidinedionas/uso terapéutico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Análisis de Varianza , Animales , Área Bajo la Curva , Modelos Animales de Enfermedad , Hiperinsulinismo/genética , Hiperinsulinismo/metabolismo , Hipoglucemiantes/uso terapéutico , Insulina/sangre , Ratones , Ratones Transgénicos , Rosiglitazona
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA