Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 38(9)2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30886048

RESUMEN

Neurodegenerative diseases are characterized by the accumulation of misfolded proteins in the brain. Insights into protein quality control mechanisms to prevent neuronal dysfunction and cell death are crucial in developing causal therapies. Here, we report that various disease-associated protein aggregates are modified by the linear ubiquitin chain assembly complex (LUBAC). HOIP, the catalytic component of LUBAC, is recruited to misfolded Huntingtin in a p97/VCP-dependent manner, resulting in the assembly of linear polyubiquitin. As a consequence, the interactive surface of misfolded Huntingtin species is shielded from unwanted interactions, for example with the low complexity sequence domain-containing transcription factor Sp1, and proteasomal degradation of misfolded Huntingtin is facilitated. Notably, all three core LUBAC components are transcriptionally regulated by Sp1, linking defective LUBAC expression to Huntington's disease. In support of a protective activity of linear ubiquitination, silencing of OTULIN, a deubiquitinase with unique specificity for linear polyubiquitin, decreases proteotoxicity, whereas silencing of HOIP has the opposite effect. These findings identify linear ubiquitination as a protein quality control mechanism and hence a novel target for disease-modifying strategies in proteinopathies.


Asunto(s)
Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Poliubiquitina/metabolismo , Procesamiento Proteico-Postraduccional , Factor de Transcripción Sp1/metabolismo , Proteína que Contiene Valosina/metabolismo , Adulto , Anciano , Animales , Encéfalo/metabolismo , Encéfalo/patología , Estudios de Casos y Controles , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , FN-kappa B/genética , FN-kappa B/metabolismo , Neuronas/metabolismo , Neuronas/patología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal , Factor de Transcripción Sp1/genética , Ubiquitinación , Proteína que Contiene Valosina/genética
2.
Hum Mol Genet ; 29(15): 2551-2567, 2020 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-32761094

RESUMEN

The expanded HTT CAG repeat causing Huntington's disease (HD) exhibits somatic expansion proposed to drive the rate of disease onset by eliciting a pathological process that ultimately claims vulnerable cells. To gain insight into somatic expansion in humans, we performed comprehensive quantitative analyses of CAG expansion in ~50 central nervous system (CNS) and peripheral postmortem tissues from seven adult-onset and one juvenile-onset HD individual. We also assessed ATXN1 CAG repeat expansion in brain regions of an individual with a neurologically and pathologically distinct repeat expansion disorder, spinocerebellar ataxia type 1 (SCA1). Our findings reveal similar profiles of tissue instability in all HD individuals, which, notably, were also apparent in the SCA1 individual. CAG expansion was observed in all tissues, but to different degrees, with multiple cortical regions and neostriatum tending to have the greatest instability in the CNS, and liver in the periphery. These patterns indicate different propensities for CAG expansion contributed by disease locus-independent trans-factors and demonstrate that expansion per se is not sufficient to cause cell type or disease-specific pathology. Rather, pathology may reflect distinct toxic processes triggered by different repeat lengths across cell types and diseases. We also find that the HTT CAG length-dependent expansion propensity of an individual is reflected in all tissues and in cerebrospinal fluid. Our data indicate that peripheral cells may be a useful source to measure CAG expansion in biomarker assays for therapeutic efforts, prompting efforts to dissect underlying mechanisms of expansion that may differ between the brain and periphery.


Asunto(s)
Enfermedad de Huntington/genética , Ataxias Espinocerebelosas/genética , Expansión de Repetición de Trinucleótido/genética , Repeticiones de Trinucleótidos/genética , Adulto , Anciano , Autopsia , Sistema Nervioso Central/patología , Niño , Femenino , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/diagnóstico por imagen , Enfermedad de Huntington/patología , Masculino , Persona de Mediana Edad , Neostriado/diagnóstico por imagen , Neostriado/metabolismo , Neostriado/patología , Ataxias Espinocerebelosas/diagnóstico por imagen , Ataxias Espinocerebelosas/patología
3.
J Neurochem ; 156(5): 589-603, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32083308

RESUMEN

Reelin is a protein that is best known for its role in controlling neuronal layer formation in the developing cortex. Here, we studied its role for post-natal cortical network function, which is poorly explored. To preclude early cortical migration defects caused by Reelin deficiency, we used a conditional Reelin knock-out (RelncKO ) mouse, and induced Reelin deficiency post-natally. Induced Reelin deficiency caused hyperexcitability of the neocortical network in vitro and ex vivo. Blocking Reelin binding to its receptors ApoER2 and VLDLR resulted in a similar effect. Hyperexcitability in RelncKO organotypic slice cultures could be rescued by co-culture with wild-type organotypic slice cultures. Moreover, the GABAB receptor (GABAB R) agonist baclofen failed to activate and the antagonist CGP35348 failed to block GABAB Rs in RelncKO mice. Immunolabeling of RelncKO cortical slices revealed a reduction in GABAB R1 and GABAB R2 surface expression at the plasma membrane and western blot of RelncKO cortical tissue revealed decreased phosphorylation of the GABAB R2 subunit at serine 892 and increased phosphorylation at serine 783, reflecting receptor deactivation and proteolysis. These data show a role of Reelin in controlling early network activity, by modulating GABAB R function. Cover Image for this issue: https://doi.org/10.1111/jnc.15054.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/deficiencia , Proteínas de la Matriz Extracelular/deficiencia , Neocórtex/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Receptores de GABA-B/fisiología , Serina Endopeptidasas/deficiencia , Transducción de Señal/fisiología , Animales , Animales Recién Nacidos , Moléculas de Adhesión Celular Neuronal/genética , Proteínas de la Matriz Extracelular/genética , Femenino , Agonistas de Receptores GABA-B/farmacología , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Técnicas de Cultivo de Órganos , Proteína Reelina , Serina Endopeptidasas/genética , Transducción de Señal/efectos de los fármacos
4.
Eur Arch Psychiatry Clin Neurosci ; 270(7): 819-828, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32062729

RESUMEN

There is increasing evidence from genetic, biochemical, pharmacological, neuroimaging and post-mortem studies that immunological dysregulation plays a crucial role in the pathogenesis of psychoses. The involvement of microglia in schizophrenia and bipolar disorder (BD) has remained controversial, however, since results from various post-mortem studies are still inconclusive. Here, we analyzed the estimated density of microglia of age-matched individuals with schizophrenia (n = 17), BD (n = 13), and non-psychiatric control subjects (n = 17) in the anterior midcingulate cortex (aMCC), a brain area putatively involved in the pathogenesis of psychoses, using ionized calcium binding adaptor molecule 1 (Iba1)-immunohistochemistry. The microglial cells displayed a homogenously distributed Iba1-staining pattern in the aMCC with slightly varying activation states in all three groups. The estimated microglial densities did not differ significantly between individuals with schizophrenia, BD and control subjects. Remarkably, when both hemispheres were investigated separately within the three groups, the density was significantly lateralized towards the right aMCC in schizophrenia (p = 0.01) and-even more evident-in BD subjects (p = 0.008). This left-right lateralization was not observed in the control group (p = 0.52). Of note, microglial density was significantly lower in BD individuals who did not commit suicide compared with BD individuals who died from suicide (p = 0.002). This difference was not observed between individuals with BD who committed suicide and controls. The results, tentatively interpreted, suggest a hitherto unknown increased lateralization of microglial density to the right hemisphere in both psychiatric groups. If confirmed in independent samples, lateralization should be considered in all post-mortem studies on microglia. Density differences between suicide and non-suicide individuals needs further elucidation.


Asunto(s)
Trastorno Bipolar/inmunología , Proteínas de Unión al Calcio/inmunología , Giro del Cíngulo/inmunología , Proteínas de Microfilamentos/inmunología , Microglía/inmunología , Esquizofrenia/inmunología , Adulto , Diagnóstico , Femenino , Lateralidad Funcional/fisiología , Humanos , Masculino , Persona de Mediana Edad , Suicidio Completo
6.
Cereb Cortex ; 24(7): 1923-36, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23448871

RESUMEN

The nitric oxide (NO)/cyclic guanosine monophosphate (cGMP) signaling cascade participates in the modulation of synaptic transmission. The effects of NO are mediated by the NO-sensitive cGMP-forming guanylyl cyclases (NO-GCs), which exist in 2 isoforms with indistinguishable regulatory properties. The lack of long-term potentiation (LTP) in knock-out (KO) mice deficient in either one of the NO-GC isoforms indicates the contribution of both NO-GCs to LTP. Recently, we showed that the NO-GC1 isoform is located presynaptically in glutamatergic neurons and increases the glutamate release via hyperpolarization-activated cyclic nucleotide (HCN)-gated channels in the hippocampus. Electrophysiological analysis of hippocampal CA1 neurons in whole-cell recordings revealed a reduction of HCN currents and a hyperpolarizing shift of the activation curve in the NO-GC2 KOs associated with reduced resting membrane potentials. These features were mimicked in wild-type (WT) neurons with an NO-GC inhibitor. Analysis of glutamate receptors revealed a cGMP-dependent reduction of NMDA receptor currents in the NO-GC2 KO mice, which was mimicked in WT by HCN channel inhibition. Lowering extracellular Mg(2+) increased NMDA receptor currents in the NO-GC2 KO and allowed the induction of LTP that was absent at physiological Mg(2+). In sum, our data indicate that postsynaptic cGMP increases the N-methyl-D-aspartate (NMDA) receptor current by gating HCN channels and thereby is required for LTP.


Asunto(s)
Región CA1 Hipocampal/citología , GMP Cíclico/metabolismo , Potenciación a Largo Plazo/fisiología , Neuronas/fisiología , Óxido Nítrico/deficiencia , Receptores de N-Metil-D-Aspartato/metabolismo , Anestésicos Locales/farmacología , Animales , Animales Recién Nacidos , GMP Cíclico/farmacología , Estimulación Eléctrica , Antagonistas de Aminoácidos Excitadores/farmacología , Guanilato Ciclasa/genética , Guanilato Ciclasa/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Técnicas In Vitro , Lidocaína/análogos & derivados , Lidocaína/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Óxido Nítrico/genética , Técnicas de Placa-Clamp , Pirimidinas/farmacología , Tetraetilamonio/farmacología
7.
J Neurosci ; 33(21): 9068-81, 2013 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-23699518

RESUMEN

Spinocerebellar ataxia 17 (SCA17) is an autosomal-dominant, late-onset neurodegenerative disorder caused by an expanded polyglutamine (polyQ) repeat in the TATA-box-binding protein (TBP). To further investigate this devastating disease, we sought to create a first transgenic rat model for SCA17 that carries a full human cDNA fragment of the TBP gene with 64 CAA/CAG repeats (TBPQ64). In line with previous observations in mouse models for SCA17, TBPQ64 rats show a severe neurological phenotype including ataxia, impairment of postural reflexes, and hyperactivity in early stages followed by reduced activity, loss of body weight, and early death. Neuropathologically, the severe phenotype of SCA17 rats was associated with neuronal loss, particularly in the cerebellum. Degeneration of Purkinje, basket, and stellate cells, changes in the morphology of the dendrites, nuclear TBP-positive immunoreactivity, and axonal torpedos were readily found by light and electron microscopy. While some of these changes are well recapitulated in existing mouse models for SCA17, we provide evidence that some crucial characteristics of SCA17 are better mirrored in TBPQ64 rats. Thus, this SCA17 model represents a valuable tool to pursue experimentation and therapeutic approaches that may be difficult or impossible to perform with SCA17 transgenic mice. We show for the first time positron emission tomography (PET) and diffusion tensor imaging (DTI) data of a SCA animal model that replicate recent PET studies in human SCA17 patients. Our results also confirm that DTI are potentially useful correlates of neuropathological changes in TBPQ64 rats and raise hope that DTI imaging could provide a biomarker for SCA17 patients.


Asunto(s)
Imagen de Difusión Tensora , Modelos Animales de Enfermedad , Ataxias Espinocerebelosas , Proteína de Unión a TATA-Box/genética , Expansión de Repetición de Trinucleótido/genética , Animales , Ansiedad/etiología , Ansiedad/genética , Peso Corporal/genética , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/ultraestructura , Procesamiento Automatizado de Datos , Femenino , Genotipo , Humanos , Masculino , Aprendizaje por Laberinto , Actividad Motora , Examen Neurológico , Tomografía de Emisión de Positrones , Desempeño Psicomotor/fisiología , Racloprida/farmacocinética , Ratas , Ratas Transgénicas , Prueba de Desempeño de Rotación con Aceleración Constante , Índice de Severidad de la Enfermedad , Ataxias Espinocerebelosas/diagnóstico , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/fisiopatología , Tubulina (Proteína)/metabolismo
8.
J Biol Chem ; 288(29): 21433-21447, 2013 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-23760501

RESUMEN

The N-acyl chain length of ceramides is determined by the specificity of different ceramide synthases (CerS). The CerS family in mammals consists of six members with different substrate specificities and expression patterns. We have generated and characterized a mouse line harboring an enzymatically inactive ceramide synthase 6 (CerS6KO) gene and lacz reporter cDNA coding for ß-galactosidase directed by the CerS6 promoter. These mice display a decrease in C16:0 containing sphingolipids. Relative to wild type tissues the amount of C16:0 containing sphingomyelin in kidney is ∼35%, whereas we find a reduction of C16:0 ceramide content in the small intestine to about 25%. The CerS6KO mice show behavioral abnormalities including a clasping abnormality of their hind limbs and a habituation deficit. LacZ reporter expression in the brain reveals CerS6 expression in hippocampus, cortex, and the Purkinje cell layer of the cerebellum. Using newly developed antibodies that specifically recognize the CerS6 protein we show that the endogenous CerS6 protein is N-glycosylated and expressed in several tissues of mice, mainly kidney, small and large intestine, and brain.


Asunto(s)
Conducta Animal , Esfingolípidos/metabolismo , Esfingosina N-Aciltransferasa/metabolismo , Animales , Ansiedad/patología , Ansiedad/fisiopatología , Encéfalo/metabolismo , Encéfalo/patología , Activación Enzimática , Pruebas de Enzimas , Conducta Exploratoria , Técnica del Anticuerpo Fluorescente , Glicosilación , Células HEK293 , Habituación Psicofisiológica , Humanos , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Glomérulos Renales/ultraestructura , Espectrometría de Masas , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de Órganos , Fenotipo , Esfingolípidos/química , Esfingosina N-Aciltransferasa/deficiencia , beta-Galactosidasa/metabolismo
9.
BMC Dermatol ; 14: 17, 2014 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-25363238

RESUMEN

BACKGROUND: Atopic dermatitis (AD) is a chronic inflammatory skin disorder caused by complex interaction of genetic and environmental factors. Besides mutations in the filaggrin gene, leading to impaired skin barrier function, variation in genes encoding additional skin proteins has been suggested to contribute to disease risk. Laminin 5, playing an important role in skin integrity, is composed of three subunits encoded by the LAMA3, LAMB3 and LAMC2 genes in which biallelic mutations cause epidermolysis bullosa junctionalis. We aimed at evaluating the role of variation in the LAMA3, LAMB3 and LAMC2 genes for AD pathogenesis. METHODS: 29 single nucleotide polymorphisms (SNPs) were genotyped in the three genes in a German AD case-control cohort comprising 470 unrelated AD patients and 320 non-atopic controls by means of restriction enzyme digestion. Allele, genotype and haplotype frequencies were compared between cases and controls using chi-square testing and the Haploview software. RESULTS: Several SNPs in the LAMA3 gene showed significant association with AD in our cohort (p <0.01), while we did not detect association with variations in the LAMB3 and LAMC2 genes. Haplotype analysis additionally revealed several significantly associated haplotypes in the LAMA3 gene. Due to extensive linkage disequilibrium, though, we were not able to further differentiate the specific disease causing variation(s) in this region. CONCLUSIONS: We established the LAMA3 gene as novel potential susceptibility gene for AD. Additional studies in independent cohorts are needed to replicate these results.


Asunto(s)
Moléculas de Adhesión Celular/genética , Dermatitis Atópica/genética , Predisposición Genética a la Enfermedad , Laminina/genética , Polimorfismo de Nucleótido Simple , Adolescente , Adulto , Alelos , Estudios de Casos y Controles , Niño , Preescolar , Proteínas Filagrina , Haplotipos , Humanos , Desequilibrio de Ligamiento , Adulto Joven , Kalinina
10.
J Neurosci ; 32(44): 15426-38, 2012 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-23115180

RESUMEN

Huntington disease (HD) is an inherited progressive neurodegenerative disorder, characterized by motor, cognitive, and psychiatric deficits as well as neurodegeneration and brain atrophy beginning in the striatum and the cortex and extending to other subcortical brain regions. The genetic cause is an expansion of the CAG repeat stretch in the HTT gene encoding huntingtin protein (htt). Here, we generated an HD transgenic rat model using a human bacterial artificial chromosome (BAC), which contains the full-length HTT genomic sequence with 97 CAG/CAA repeats and all regulatory elements. BACHD transgenic rats display a robust, early onset and progressive HD-like phenotype including motor deficits and anxiety-related symptoms. In contrast to BAC and yeast artificial chromosome HD mouse models that express full-length mutant huntingtin, BACHD rats do not exhibit an increased body weight. Neuropathologically, the distribution of neuropil aggregates and nuclear accumulation of N-terminal mutant huntingtin in BACHD rats is similar to the observations in human HD brains. Aggregates occur more frequently in the cortex than in the striatum and neuropil aggregates appear earlier than mutant htt accumulation in the nucleus. Furthermore, we found an imbalance in the striatal striosome and matrix compartments in early stages of the disease. In addition, reduced dopamine receptor binding was detectable by in vivo imaging. Our data demonstrate that this transgenic BACHD rat line may be a valuable model for further understanding the disease mechanisms and for preclinical pharmacological studies.


Asunto(s)
Cromosomas Artificiales Bacterianos/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Proteínas del Tejido Nervioso/genética , Empalme Alternativo , Animales , Ansiedad/genética , Ansiedad/psicología , Conducta Animal/fisiología , Western Blotting , Peso Corporal/fisiología , Ingestión de Alimentos/fisiología , Trastornos Neurológicos de la Marcha/psicología , Dosificación de Gen , Humanos , Proteína Huntingtina , Enfermedad de Huntington/psicología , Inmunohistoquímica , Actividad Motora/fisiología , Tomografía de Emisión de Positrones , Equilibrio Postural/fisiología , Regiones Promotoras Genéticas , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
Hum Mol Genet ; 20(18): 3620-31, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21680557

RESUMEN

Retinitis pigmentosa (RP) is a group of human retinal disorders, with more than 100 genes involved in retinal degeneration. Canine and murine models are useful for investigating human RP based on known, naturally occurring mutations. In Schapendoes dogs, for example, a mutation in the CCDC66 gene has been shown to cause autosomal recessively inherited, generalized progressive retinal atrophy (gPRA), the canine counterpart to RP. Here, a novel mouse model with a disrupted Ccdc66 gene was investigated to reveal the function of protein CCDC66 and the pathogenesis of this form of gPRA. Homozygous Ccdc66 mutant mice lack retinal Ccdc66 RNA and protein expression. Light and electron microscopy reveal an initial degeneration of photoreceptors already at 13 days of age, followed by a slow, progressive retinal degeneration over months. Retinal dysfunction causes reduced scotopic a-wave amplitudes, declining from 1 to 7 months of age as well as an early reduction of the photopic b-wave at 1 month, improving slightly at 7 months, as evidenced by electroretinography. In the retina of the wild-type (WT) mouse, protein CCDC66 is present at highest levels after birth, followed by a decline until adulthood, suggesting a crucial role in early development. Protein CCDC66 is expressed predominantly in the developing rod outer segments as confirmed by subcellular analyses. These findings illustrate that the lack of protein CCDC66 causes early, slow progressive rod-cone dysplasia in the novel Ccdc66 mutant mouse model, thus providing a sound foundation for the development of therapeutic strategies.


Asunto(s)
Proteínas del Ojo/genética , Degeneración Retiniana/genética , Degeneración Retiniana/fisiopatología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/fisiopatología , Eliminación de Secuencia , Animales , Modelos Animales de Enfermedad , Femenino , Silenciador del Gen , Humanos , Masculino , Ratones , Ratones Noqueados , Retina/metabolismo , Retina/patología , Retina/fisiopatología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología
12.
J Neurosci ; 31(4): 1193-203, 2011 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-21273404

RESUMEN

Human cortical excitability can be modified by repetitive transcranial magnetic stimulation (rTMS), but the cellular mechanisms are largely unknown. Here, we show that the pattern of delivery of theta-burst stimulation (TBS) (continuous versus intermittent) differently modifies electric activity and protein expression in the rat neocortex. Intermittent TBS (iTBS), but not continuous TBS (cTBS), enhanced spontaneous neuronal firing and EEG gamma band power. Sensory evoked cortical inhibition increased only after iTBS, although both TBS protocols increased the first sensory response arising from the resting cortical state. Changes in the cortical expression of the calcium-binding proteins parvalbumin (PV) and calbindin D-28k (CB) indicate that changes in spontaneous and evoked cortical activity following rTMS are in part related to altered activity of inhibitory systems. By reducing PV expression in the fast-spiking interneurons, iTBS primarily affected the inhibitory control of pyramidal cell output activity, while cTBS, by reducing CB expression, more likely affected the dendritic integration of synaptic inputs controlled by other classes of inhibitory interneurons. Calretinin, the third major calcium-binding protein expressed by another class of interneurons was not affected at all. We conclude that different patterns of TBS modulate the activity of inhibitory cell classes differently, probably depending on the synaptic connectivity and the preferred discharge pattern of these inhibitory neurons.


Asunto(s)
Corteza Cerebral/fisiología , Potenciales de Acción , Animales , Calbindina 2 , Calbindinas , Electroencefalografía , Potenciales Evocados Somatosensoriales , Interneuronas/fisiología , Masculino , Inhibición Neural , Parvalbúminas/biosíntesis , Células Piramidales/fisiología , Ratas , Proteína G de Unión al Calcio S100/biosíntesis , Estimulación Magnética Transcraneal
13.
J Neurosci ; 31(36): 12790-801, 2011 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-21900558

RESUMEN

Posttranslational amyloid-ß (Aß) modification is considered to play an important role in Alzheimer's disease (AD) etiology. An N-terminally modified Aß species, pyroglutamate-amyloid-ß (pE3-Aß), has been described as a major constituent of Aß deposits specific to human AD but absent in normal aging. Formed via cyclization of truncated Aß species by glutaminyl cyclase (QC; QPCT) and/or its isoenzyme (isoQC; QPCTL), pE3-Aß aggregates rapidly and is known to seed additional Aß aggregation. To directly investigate pE3-Aß toxicity in vivo, we generated and characterized transgenic TBA2.1 and TBA2.2 mice, which express truncated mutant human Aß. Along with a rapidly developing behavioral phenotype, these mice showed progressively accumulating Aß and pE3-Aß deposits in brain regions of neuronal loss, impaired long-term potentiation, microglial activation, and astrocytosis. Illustrating a threshold for pE3-Aß neurotoxicity, this phenotype was not found in heterozygous animals but in homozygous TBA2.1 or double-heterozygous TBA2.1/2.2 animals only. A significant amount of pE3-Aß formation was shown to be QC-dependent, because crossbreeding of TBA2.1 with QC knock-out, but not isoQC knock-out, mice significantly reduced pE3-Aß levels. Hence, lowering the rate of QC-dependent posttranslational pE3-Aß formation can, in turn, lower the amount of neurotoxic Aß species in AD.


Asunto(s)
Precursor de Proteína beta-Amiloide/biosíntesis , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Trastornos Heredodegenerativos del Sistema Nervioso/patología , Hipocampo/patología , Ácido Pirrolidona Carboxílico/metabolismo , Envejecimiento/patología , Envejecimiento/psicología , Enfermedad de Alzheimer/patología , Animales , Conducta Animal , Encéfalo/patología , Ensayo de Inmunoadsorción Enzimática , Gliosis/patología , Trastornos Heredodegenerativos del Sistema Nervioso/psicología , Humanos , Inmunohistoquímica , Cinética , Potenciación a Largo Plazo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Microscopía Electrónica , Plasticidad Neuronal/genética , Plasticidad Neuronal/fisiología , Fenotipo , Equilibrio Postural/fisiología , Procesamiento Proteico-Postraduccional , Reflejo de Sobresalto/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Eur Heart J ; 32(10): 1287-97, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20965886

RESUMEN

AIMS: Mice with a cardiomyocyte (CM)-restricted knockout of signal transducer and activator of transcription 3 (STAT3-KO) develop spontaneous heart failure. We investigated the impact of STAT3-mediated regulation of microRNAs for pathophysiological alterations in the heart. METHODS AND RESULTS: MicroRNAchip and qRT-PCR analysis revealed elevated cardiac expression of miR-199a in STAT3-KO mice. Lentiviral shRNA-mediated STAT3-knock-down in neonatal rat CMs markedly increased miR-199a promoter activity and miR-199a levels indicative of a suppressive effect of STAT3 on miR-199a transcription. Up-regulated miR-199a in CM by pre-miR-199a transfection (pre-miR-199a-CM) reduced expression of components of the ubiquitin-proteasome system (UPS), i.e. the ubiquitin-conjugating enzymes Ube2g1 (mRNA and protein) and Ube2i (protein). Pre-miR-199a-CM or CM with siRNA-mediated down-regulation of Ube2i and Ube2g1 (siRNA-Ube2i/2g1-CM) displayed massive down-regulation of α- and ß-myosin heavy chain expression associated with disrupted sarcomere structures. In addition, protein arginine methyltransferase I (PRMT-I) expression and asymmetric dimethylarginine (ADMA) synthesis were increased in pre-miR-199a-CM or in siRNA-Ube2i/2g1-CM. Increased ADMA in cell culture supernatant (SN) from pre-miR-199a-CM or siRNA-Ube2i/2g1-CM lowered nitric oxide (NO) bioavailability of rat cardiac endothelial cells while lowering ADMA concentration in CM SNs by the PRMT inhibitor arginine methyltransferase inhibitor 1 (AMI-1) (100 µM) improved NO bioavailability. In STAT3-KO hearts Ube2i and Ube2g1 expression were markedly reduced. Human terminal failing hearts harbouring low STAT3 protein levels displayed increased miR-199a levels and decreased Ube2g1 expression. CONCLUSION: This study identifies a novel pathophysiological circuit in the heart between reduced STAT3 protein levels, increased miR-199a expression, and subsequent impairment of the UPS that disrupts CM sarcomere structure and impairs via the release of ADMA endothelial cell function.


Asunto(s)
Células Endoteliales/fisiología , Insuficiencia Cardíaca/enzimología , MicroARNs/metabolismo , Miocitos Cardíacos/fisiología , Factor de Transcripción STAT3/fisiología , Enzimas Ubiquitina-Conjugadoras/fisiología , Animales , Arginina/análogos & derivados , Arginina/metabolismo , Regulación hacia Abajo , Endotelio Vascular/fisiología , Insuficiencia Cardíaca/fisiopatología , Humanos , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/fisiología , Ratas , Factor de Transcripción STAT3/metabolismo , Regulación hacia Arriba
16.
Neurobiol Dis ; 44(2): 192-204, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21767644

RESUMEN

Mutations in the N-terminus of the gene encoding α-synuclein (α-syn) are linked to autosomal dominantly inherited Parkinson's disease (PD). The vast majority of PD patients develop neuropsychiatric symptoms preceding motor impairments. During this premotor stage, synucleinopathy is first detectable in the olfactory bulb (OB) and brain stem nuclei; however its impact on interconnected brain regions and related symptoms is still less far understood. Using a novel conditional transgenic mouse model, displaying region-specific expression of human mutant α-syn, we evaluated effect and reversibility of olfactory synucleinopathy. Our data showed that induction of mutant A30P α-syn expression increased transgenic deposition into somatodendritic compartment of dopaminergic neurons, without generating fibrillar inclusions. We found reversibly reduced levels of dopamine and metabolites in the OB, suggesting an impact of A30P α-syn on olfactory neurotransmitter content. We further showed that mutant A30P expression led to neurodegenerative changes on an ultrastructural level and a behaviorally hyperactive response correlated with novelty, odor processing and stress associated with an increased dopaminergic tone in midbrain regions. Our present data indicate that mutant (A30P) α-syn is directly implicated in reduction of dopamine signaling in OB interneurons, which mediates further alterations in brain regions without transgenic expression leading functionally to a hyperactive response. These modulations of neurotransmission may underlie in part some of the early neuropsychiatric symptoms in PD preceding dysfunction of the nigrostriatal dopaminergic system.


Asunto(s)
Dopamina/deficiencia , Neuronas/metabolismo , Bulbo Olfatorio/metabolismo , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/fisiopatología , alfa-Sinucleína/genética , Sustitución de Aminoácidos/genética , Animales , Cricetinae , Modelos Animales de Enfermedad , Dopamina/biosíntesis , Femenino , Humanos , Hipercinesia/genética , Hipercinesia/metabolismo , Hipercinesia/fisiopatología , Masculino , Ratones , Ratones Transgénicos , Mutación/genética , Neuronas/patología , Bulbo Olfatorio/patología , Bulbo Olfatorio/fisiopatología , Trastornos Parkinsonianos/genética , alfa-Sinucleína/biosíntesis , alfa-Sinucleína/fisiología
17.
Ann Neurol ; 67(5): 684-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20437567

RESUMEN

Familial Parkinson disease (PD) due to the A30P mutation in the SNCA gene encoding alpha-synuclein is clinically associated with PD symptoms. In this first pathoanatomical study of the brain of an A30P mutation carrier, we observed neuronal loss in the substantia nigra, locus coeruleus, and dorsal motor vagal nucleus, as well as widespread occurrence of alpha-synuclein immunopositive Lewy bodies, Lewy neurites, and glial aggregates. Alpha-synuclein aggregates ultrastructurally resembled Lewy bodies, and biochemical analyses disclosed a significant load of insoluble alpha-synuclein, indicating neuropathological similarities between A30P disease patients and idiopathic PD, with a more severe neuropathology in A30P carriers.


Asunto(s)
Encefalopatías/genética , Encefalopatías/patología , Encéfalo/patología , Mutación/genética , alfa-Sinucleína/genética , Anciano , Alanina/genética , Encéfalo/ultraestructura , Salud de la Familia , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Cuerpos de Inclusión/patología , Masculino , Prolina/genética
18.
Neurogenetics ; 11(2): 163-74, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19777273

RESUMEN

Canine generalized progressive retinal atrophy (gPRA) is characterized by continuous degeneration of photoreceptor cells leading to night blindness and progressive vision loss. Until now, mutations in 11 genes have been described that account for gPRA in dogs, mostly following an autosomal recessive inheritance mode. Here, we describe a gPRA locus comprising the newly identified gene coiled-coil domain containing 66 (CCDC66) on canine chromosome 20, as identified via linkage analysis in the Schapendoes breed. Mutation screening of the CCDC66 gene revealed a 1-bp insertion in exon 6 leading to a stop codon as the underlying cause of disease. The insertion is present in all affected dogs in the homozygous state as well as in all obligatory mutation carriers in the heterozygous state. The CCDC66 gene is evolutionarily conserved in different vertebrate species and exhibits a complex pattern of differential RNA splicing resulting in various isoforms in the retina. Immunohistochemically, CCDC66 protein is detected mainly in the inner segments of photoreceptors in mouse, dog, and man. The affected Schapendoes retina lacks CCDC66 protein. Thus this natural canine model for gPRA yields superior potential to understand functional implications of this newly identified protein including its physiology, and it opens new perspectives for analyzing different aspects of the general pathophysiology of gPRA.


Asunto(s)
Perros , Mutación , Degeneración Retiniana , Secuencia de Aminoácidos , Animales , Análisis Mutacional de ADN , Ligamiento Genético , Humanos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Polimorfismo Genético , Estructura Terciaria de Proteína , Retina/patología , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Degeneración Retiniana/veterinaria , Alineación de Secuencia
19.
Acta Neuropathol ; 119(6): 771-8, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20309567

RESUMEN

The anterior cingulate cortex (ACC) represents a phylogenetically ancient region of the mammalian brain that has undergone recent adaptive changes in humans. It contains a large spindle-shaped cell type, referred to as von Economo neuron (VEN) that has been shown to be involved in the pathophysiology of various neuropsychiatric disorders. Schizophrenia is a group of disorders that is, in part, characterised by a disruption of neuronal migration in early ontogeny and presumably secondary degeneration after the first psychotic episode in some patients. Accordingly, we tested the hypothesis that the density of VENs is reduced in a neurodevelopmental subtype of schizophrenia, which we defined by an early onset of the disorder. The density of VENs was estimated in layer Vb of Brodmann's area 24 in 20 subjects diagnosed with schizophrenia. The results were compared with 19 specimens from patients with bipolar disorder as a clinical control and 22 non-psychiatric samples. The density of VENs did not differ between the three groups. However, the VEN density in the right ACC correlated with the age at onset, and inversely with the duration of the illness in schizophrenia, but not in bipolar disorder. Thus, patients with early onset schizophrenia (and longer duration of illness) had a reduced VEN density. Age, sex, postmortem interval, brain weight, and cortical thickness had no significant impact on the results. These findings suggest that VENs in the ACC are involved in neurodevelopmental and perhaps neurodegenerative processes specific to schizophrenia.


Asunto(s)
Giro del Cíngulo/patología , Neuronas/patología , Esquizofrenia/patología , Adulto , Factores de Edad , Edad de Inicio , Trastorno Bipolar/patología , Encéfalo/patología , Estudios de Casos y Controles , Recuento de Células , Femenino , Humanos , Masculino , Persona de Mediana Edad , Tamaño de los Órganos , Factores Sexuales , Factores de Tiempo , Adulto Joven
20.
Mol Cell Probes ; 24(6): 357-63, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20691256

RESUMEN

Generalized progressive retinal atrophy (gPRA) belongs to a group of inherited retinal diseases which are associated with gradual vision loss in various dog breeds, including the Irish Glen of Imaal Terrier (GIT). By genome-wide homozygosity mapping using SNP arrays and fine mapping of candidate regions, we assigned the gPRA candidate locus in this breed to canine chromosome 16. The respective region is syntenic with human chromosome 8 comprising the ADAM metallopeptidase domain 9 (ADAM9) gene. ADAM9 represents a strong candidate gene for canine retinal disease because mutations have previously been shown to cause autosomal recessively inherited human cone-rod dystrophy, a retinal disorder affecting photoreceptor function. Sequence analysis of ADAM9 in affected and carrier GITs revealed a deletion of exons 15 and 16 which alters the reading frame leading to a premature stop codon. This mutation was absent from 34 other dog breeds. A variable and, at times, very late onset of gPRA was confirmed in GITs by a relatively mild retinal degeneration at an advanced age. Hence, the identification of the genetic defect underlying gPRA in the GIT represents a suitable model for cone-rod dystrophy of humans, with superior potential to elucidate functional consequences of the recently described null mutations in the human ADAM9 gene.


Asunto(s)
Proteínas ADAM/genética , Enfermedades de los Perros/enzimología , Enfermedades de los Perros/genética , Eliminación de Gen , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Degeneración Retiniana/veterinaria , Animales , Secuencia de Bases , ADN Complementario/análisis , ADN Complementario/genética , Enfermedades de los Perros/patología , Perros , Regulación de la Expresión Génica , Sitios Genéticos/genética , Haplotipos/genética , Humanos , Inmunohistoquímica , Irlanda , Datos de Secuencia Molecular , Mapeo Físico de Cromosoma , Reacción en Cadena de la Polimerasa , Degeneración Retiniana/enzimología , Degeneración Retiniana/genética , Degeneración Retiniana/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA