Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 41(4): e109108, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35019161

RESUMEN

Haploinsufficiency of the progranulin (PGRN)-encoding gene (GRN) causes frontotemporal lobar degeneration (GRN-FTLD) and results in microglial hyperactivation, TREM2 activation, lysosomal dysfunction, and TDP-43 deposition. To understand the contribution of microglial hyperactivation to pathology, we used genetic and pharmacological approaches to suppress TREM2-dependent transition of microglia from a homeostatic to a disease-associated state. Trem2 deficiency in Grn KO mice reduced microglia hyperactivation. To explore antibody-mediated pharmacological modulation of TREM2-dependent microglial states, we identified antagonistic TREM2 antibodies. Treatment of macrophages from GRN-FTLD patients with these antibodies led to reduced TREM2 signaling due to its enhanced shedding. Furthermore, TREM2 antibody-treated PGRN-deficient microglia derived from human-induced pluripotent stem cells showed reduced microglial hyperactivation, TREM2 signaling, and phagocytic activity, but lysosomal dysfunction was not rescued. Similarly, lysosomal dysfunction, lipid dysregulation, and glucose hypometabolism of Grn KO mice were not rescued by TREM2 ablation. Synaptic loss and neurofilament light-chain (NfL) levels, a biomarker for neurodegeneration, were further elevated in the Grn/Trem2 KO cerebrospinal fluid (CSF). These findings suggest that TREM2-dependent microglia hyperactivation in models of GRN deficiency does not promote neurotoxicity, but rather neuroprotection.


Asunto(s)
Degeneración Lobar Frontotemporal/patología , Glicoproteínas de Membrana/metabolismo , Microglía/fisiología , Monocitos/metabolismo , Progranulinas/deficiencia , Receptores Inmunológicos/metabolismo , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Encéfalo/diagnóstico por imagen , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Femenino , Degeneración Lobar Frontotemporal/metabolismo , Humanos , Lisosomas/metabolismo , Lisosomas/patología , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/efectos de los fármacos , Monocitos/efectos de los fármacos , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Quinasa Syk/metabolismo
2.
Proc Natl Acad Sci U S A ; 117(3): 1753-1761, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31896584

RESUMEN

Carbon dioxide (CO2), the major product of metabolism, has a strong impact on cerebral blood vessels, a phenomenon known as cerebrovascular reactivity. Several vascular risk factors such as hypertension or diabetes dampen this response, making cerebrovascular reactivity a useful diagnostic marker for incipient vascular pathology, but its functional relevance, if any, is still unclear. Here, we found that GPR4, an endothelial H+ receptor, and endothelial Gαq/11 proteins mediate the CO2/H+ effect on cerebrovascular reactivity in mice. CO2/H+ leads to constriction of vessels in the brainstem area that controls respiration. The consequential washout of CO2, if cerebrovascular reactivity is impaired, reduces respiration. In contrast, CO2 dilates vessels in other brain areas such as the amygdala. Hence, an impaired cerebrovascular reactivity amplifies the CO2 effect on anxiety. Even at atmospheric CO2 concentrations, impaired cerebrovascular reactivity caused longer apneic episodes and more anxiety, indicating that cerebrovascular reactivity is essential for normal brain function. The site-specific reactivity of vessels to CO2 is reflected by regional differences in their gene expression and the release of vasoactive factors from endothelial cells. Our data suggest the central nervous system (CNS) endothelium as a target to treat respiratory and affective disorders associated with vascular diseases.


Asunto(s)
Ansiedad/metabolismo , Sistema Cardiovascular/metabolismo , Endotelio/metabolismo , Trastornos Respiratorios/metabolismo , Amígdala del Cerebelo , Animales , Arteriolas/patología , Encéfalo/fisiología , Tronco Encefálico/metabolismo , Dióxido de Carbono/metabolismo , Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Endotelio/patología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Expresión Génica , Humanos , Hipercapnia/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Respiración , Factores de Riesgo , Transducción de Señal
3.
Basic Res Cardiol ; 117(1): 44, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-36068417

RESUMEN

Myocardial infarction (MI) with subsequent depression is associated with increased cardiac mortality. Impaired central mineralocorticoid (MR) and glucocorticoid receptor (GR) equilibrium has been suggested as a key mechanism in the pathogenesis of human depression. Here, we investigate if deficient central MR/GR signaling is causative for a poor outcome after MI in mice. Mice with an inducible forebrain-specific MR/GR knockout (MR/GR-KO) underwent baseline and follow-up echocardiography every 2 weeks after MI or sham operation. Behavioral testing at 4 weeks confirmed significant depressive-like behavior and, strikingly, a higher mortality after MI, while cardiac function and myocardial damage remained unaffected. Telemetry revealed cardiac autonomic imbalance with marked bradycardia and ventricular tachycardia (VT) upon MI in MR/GR-KO. Mechanistically, we found a higher responsiveness to atropine, pointing to impaired parasympathetic tone of 'depressive' mice after MI. Serum corticosterone levels were increased but-in line with the higher vagal tone-plasma and cardiac catecholamines were decreased. MR/GR deficiency in the forebrain led to significant depressive-like behavior and a higher mortality after MI. This was accompanied by increased vagal tone, depleted catecholaminergic compensatory capacity and VTs. Thus, limbic MR/GR disequilibrium may contribute to the impaired outcome of depressive patients after MI and possibly explain the lack of anti-depressive treatment benefit.


Asunto(s)
Depresión , Infarto del Miocardio , Animales , Humanos , Ratones , Infarto del Miocardio/patología , Miocardio/patología , Prosencéfalo/metabolismo , Receptores de Glucocorticoides/metabolismo
4.
FASEB J ; 32(6): 3005-3019, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29401591

RESUMEN

Systemic corticosteroids have been the mainstay of treatment for various hearing disorders for more than 30 yr. Accordingly, numerous studies have described glucocorticoids (GCs) and stressors to be protective in the auditory organ against damage associated with a variety of health conditions, including noise exposure. Conversely, stressors are also predictive risk factors for hearing disorders. How both of these contrasting stress actions are linked has remained elusive. Here, we demonstrate that higher corticosterone levels during acoustic trauma in female rats is highly correlated with a decline of auditory fiber responses in high-frequency cochlear regions, and that hearing thresholds and the outer hair cell functions (distortion products of otoacoustic emissions) are left unaffected. Moreover, when GC receptor (GR) or mineralocorticoid receptor (MR) activation was antagonized by mifepristone or spironolactone, respectively, GR, but not MR, inhibition significantly and permanently attenuated trauma-induced effects on auditory fiber responses, including inner hair cell ribbon loss and related reductions of early and late auditory brainstem responses. These findings strongly imply that higher corticosterone stress levels profoundly impair auditory nerve processing, which may influence central auditory acuity. These changes are likely GR mediated as they are prevented by mifepristone.-Singer, W., Kasini, K., Manthey, M., Eckert, P., Armbruster, P., Vogt, M. A., Jaumann, M., Dotta, M., Yamahara, K., Harasztosi, C., Zimmermann, U., Knipper, M., Rüttiger, L. The glucocorticoid antagonist mifepristone attenuates sound-induced long-term deficits in auditory nerve response and central auditory processing in female rats.


Asunto(s)
Nervio Coclear/fisiopatología , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Glucocorticoides/antagonistas & inhibidores , Trastornos de la Audición/fisiopatología , Pérdida Auditiva Provocada por Ruido/fisiopatología , Mifepristona/farmacología , Animales , Cóclea/metabolismo , Cóclea/patología , Cóclea/fisiopatología , Nervio Coclear/metabolismo , Nervio Coclear/patología , Femenino , Glucocorticoides/efectos adversos , Glucocorticoides/farmacología , Trastornos de la Audición/inducido químicamente , Trastornos de la Audición/tratamiento farmacológico , Trastornos de la Audición/metabolismo , Pérdida Auditiva Provocada por Ruido/inducido químicamente , Pérdida Auditiva Provocada por Ruido/tratamiento farmacológico , Pérdida Auditiva Provocada por Ruido/metabolismo , Ratas , Ratas Wistar , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo
5.
Biochim Biophys Acta Biomembr ; 1860(4): 887-894, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29069569

RESUMEN

The vestibule loop regions of aquaglyceroporins are involved in accumulation of glycerol inside the channel pore. Even though most loop regions do not show high sequence similarity among aquaglyceroporins, loop E is highly conserved in aquaglyceroporins, but not in members of the homologous aquaporins. Specifically, a tryptophan residue is extremely conserved within this loop. We have investigated the role of this residue (Trp219) that deeply protrudes into the protein and potentially interacts with adjacent loops, using the E. coli aqualgyeroporin GlpF as a model. Replacement of Trp219 affects the activity of GlpF and impairs the stability of the tetrameric protein. Furthermore, we have identified an amino acid cluster involving Trp219 that stabilizes the GlpF tetramer. Based on our results we propose that Trp219 is key for formation of a defined vestibule structure, which is crucial for glycerol accumulation as well as for the stability of the active GlpF tetramer.


Asunto(s)
Aminoácidos/metabolismo , Acuagliceroporinas/metabolismo , Acuaporinas/metabolismo , Proteínas de Escherichia coli/metabolismo , Triptófano/metabolismo , Aminoácidos/genética , Acuagliceroporinas/química , Acuagliceroporinas/genética , Acuaporinas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Glicerol/metabolismo , Modelos Moleculares , Mutación , Conformación Proteica , Multimerización de Proteína , Estabilidad Proteica , Triptófano/genética
6.
J Neurochem ; 141(3): 461-471, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28266720

RESUMEN

The p75 neurotrophin receptor (p75NTR) is a low-affinity receptor that is capable of binding neurotrophins. Two different p75NTR knockout mouse lines are available either with a deletion in Exon III (p75NTRExIII-/- ) or in Exon IV (p75NTRExIV-/- ). In p75NTRExIII knockout mice, only the full-length p75NTR is deleted, whereas in p75NTRExIV knockout mice, the full-length as well as the truncated isoform of the receptor is deleted. Deletion of p75NTR has been shown to affect, among others, the septohippocampal cholinergic innervation pattern and neuronal plasticity within the hippocampus. We hypothesize that deletion of p75NTR also alters the morphology and physiology of a further key structure of the limbic system, the amygdala. Our results indicate that deletion of p75NTR also increases cholinergic innervation in the basolateral amygdala in adult as well as aged p75NTRExIII-/- and p75NTRExIV-/- mice. The p75NTRExIV-/- mice did not display altered long-term potentiation (LTP) in the basolateral amygdala as compared to age-matched control littermates. However, p75NTRExIII-/- mice display stronger LTP in the basolateral amygdala compared to age-matched controls. Bath-application of K252a (a trk antagonist) did not inhibit the induction of LTP in the basolateral amygdala, but reduced the level of LTP in p75NTRExIII-/- mice to levels seen in respective controls. Moreover, p75NTRExIII-/- mice display altered behavior in the dark/light box. Thus, deletion of p75NTR in mice leads to physiological and morphological changes in the amygdala and altered behavior that is linked to the limbic system.


Asunto(s)
Amígdala del Cerebelo , Ansiedad/psicología , Sistema Nervioso Parasimpático , Receptores de Factor de Crecimiento Nervioso/deficiencia , Amígdala del Cerebelo/química , Animales , Conducta Animal , Química Encefálica/genética , Fibras Colinérgicas , Condicionamiento Psicológico , Fenómenos Electrofisiológicos , Exones , Miedo , Inmunohistoquímica , Potenciación a Largo Plazo , Ratones , Ratones Noqueados , Sistema Nervioso Parasimpático/química , Receptores de Factor de Crecimiento Nervioso/genética
7.
Cereb Cortex ; 26(10): 3991-4003, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27507786

RESUMEN

A chromosomal microdeletion at the 22q11.2 locus is associated with extensive cognitive impairments, schizophrenia and other psychopathology in humans. Previous reports indicate that mouse models of the 22q11.2 microdeletion syndrome (22q11.2DS) may model the genetic basis of cognitive deficits relevant for neuropsychiatric disorders such as schizophrenia. To assess the models usefulness for drug discovery, a novel mouse (Df(h22q11)/+) was assessed in an extensive battery of cognitive assays by partners within the NEWMEDS collaboration (Innovative Medicines Initiative Grant Agreement No. 115008). This battery included classic and touchscreen-based paradigms with recognized sensitivity and multiple attempts at reproducing previously published findings in 22q11.2DS mouse models. This work represents one of the most comprehensive reports of cognitive functioning in a transgenic animal model. In accordance with previous reports, there were non-significant trends or marginal impairment in some tasks. However, the Df(h22q11)/+ mouse did not show comprehensive deficits; no robust impairment was observed following more than 17 experiments and 14 behavioral paradigms. Thus - within the current protocols - the 22q11.2DS mouse model fails to mimic the cognitive alterations observed in human 22q11.2 deletion carriers. We suggest that the 22q11.2DS model may induce liability for cognitive dysfunction with additional "hits" being required for phenotypic expression.


Asunto(s)
Cognición , Síndrome de DiGeorge/psicología , Modelos Animales de Enfermedad , Ratones Transgénicos , Animales , Atención , Estudios de Cohortes , Discriminación en Psicología , Descubrimiento de Drogas , Función Ejecutiva , Femenino , Inhibición Psicológica , Aprendizaje , Masculino , Memoria , Ratones Endogámicos C57BL , Pruebas Neuropsicológicas , Fenotipo , Investigación Biomédica Traslacional
8.
Cell Mol Neurobiol ; 36(7): 1215-8, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26645823

RESUMEN

The recently identified Cystine-knot containing AMPAR-associated protein (Ckamp44) represents a novel AMPAR-related protein that critically controls AMPAR-mediated currents and short-term plasticity. However, the effects of the lack of this protein at network level are not entirely understood. Here we used c-Fos brain mapping to analyse whether the excitatory/inhibitory balance is altered in the absence of the Ckamp44. We found that Ckamp44(-/-) mice treated with an NMDAR antagonist exhibited a very robust c-Fos expression pattern, similar with that seen in mice lacking the GluN2A subunit of NMDAR treated with the same compound. This finding is unexpected, in particular, since Ckamp44 expression is strongest in dentate gyrus granule cells and less abundant in the rest of the brain.


Asunto(s)
Hipocampo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal/fisiología , Receptores AMPA/metabolismo , Sinapsis/metabolismo , Animales , Genes fos/fisiología , Ratones , Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica/fisiología
9.
Horm Behav ; 81: 97-105, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27059527

RESUMEN

The translational assessment of mechanisms underlying cognitive functions using touchscreen-based approaches for rodents is growing in popularity. In these paradigms, daily training is usually accompanied by extended food restriction to maintain animals' motivation to respond for rewards. Here, we show a transient elevation in stress hormone levels due to food restriction and touchscreen training, with subsequent adaptation effects, in fecal corticosterone metabolite concentrations, indicating effective coping in response to physical and psychological stressors. Corticosterone concentrations of experienced but training-deprived mice revealed a potential anticipation of task exposure, indicating a possible temporary environmental enrichment-like effect caused by cognitive challenge. Furthermore, the analyses of immediate early gene (IEG) immunoreactivity in the hippocampus revealed alterations in Arc, c-Fos and zif268 expression immediately following training. In addition, BDNF expression was altered as a function of satiation state during food restriction. These findings suggest that standard protocols for touchscreen-based training induce changes in hippocampal neuronal activity related to satiation and learning that should be considered when using this paradigm.


Asunto(s)
Glándulas Suprarrenales/metabolismo , Restricción Calórica/psicología , Condicionamiento Psicológico/fisiología , Neuronas/metabolismo , Recompensa , Tacto , Adaptación Psicológica/fisiología , Animales , Restricción Calórica/veterinaria , Corticosterona/metabolismo , Exposición a Riesgos Ambientales , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Condicionamiento Físico Animal/fisiología , Condicionamiento Físico Animal/psicología , ARN Mensajero/metabolismo
10.
Eur Arch Psychiatry Clin Neurosci ; 266(7): 673-7, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26482736

RESUMEN

NMDA receptor (NMDAR) antagonists induce in perinatal rodent cortical apoptosis and protracted schizophrenia-like alterations ameliorated by antipsychotic treatment. The broad-spectrum antibiotic minocycline elicits antipsychotic and neuroprotective effects. Here we tested, if minocycline protects also against apoptosis triggered by the NMDAR antagonist MK-801 at postnatal day 7. Surprisingly, minocycline induced widespread cortical apoptosis and exacerbated MK-801-triggered cell death. In some areas such as the subiculum, the pro-apoptotic effect of minocycline was even more pronounced than that elicited by MK-801. These data reveal among antipsychotics unique pro-apoptotic properties of minocycline, raising concerns regarding consequences for brain development and the use in children.


Asunto(s)
Antibacterianos/farmacología , Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Maleato de Dizocilpina/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Minociclina/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Animales , Antibacterianos/administración & dosificación , Encéfalo/patología , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Minociclina/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación
11.
EMBO J ; 30(11): 2266-80, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21522131

RESUMEN

Despite its key role in Alzheimer pathogenesis, the physiological function(s) of the amyloid precursor protein (APP) and its proteolytic fragments are still poorly understood. Previously, we generated APPsα knock-in (KI) mice expressing solely the secreted ectodomain APPsα. Here, we generated double mutants (APPsα-DM) by crossing APPsα-KI mice onto an APLP2-deficient background and show that APPsα rescues the postnatal lethality of the majority of APP/APLP2 double knockout mice. Surviving APPsα-DM mice exhibited impaired neuromuscular transmission, with reductions in quantal content, readily releasable pool, and ability to sustain vesicle release that resulted in muscular weakness. We show that these defects may be due to loss of an APP/Mint2/Munc18 complex. Moreover, APPsα-DM muscle showed fragmented post-synaptic specializations, suggesting impaired postnatal synaptic maturation and/or maintenance. Despite normal CNS morphology and unaltered basal synaptic transmission, young APPsα-DM mice already showed pronounced hippocampal dysfunction, impaired spatial learning and a deficit in LTP that could be rescued by GABA(A) receptor inhibition. Collectively, our data show that APLP2 and APP are synergistically required to mediate neuromuscular transmission, spatial learning and synaptic plasticity.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/deficiencia , Animales , Cruzamientos Genéticos , Aprendizaje , Ratones , Ratones Noqueados , Unión Neuromuscular/fisiología , Plasticidad Neuronal , Transmisión Sináptica
12.
Hippocampus ; 24(4): 424-35, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24339333

RESUMEN

Adolescence is characterized by important molecular and anatomical changes with relevance for the maturation of brain circuitry and cognitive function. This time period is of critical importance in the emergence of several neuropsychiatric disorders accompanied by cognitive impairment, such as affective disorders and schizophrenia. The molecular mechanisms underlying these changes at neuronal level during this specific developmental stage remains however poorly understood. GluA1-containing AMPA receptors, which are located predominantly on hippocampal neurons, are the primary molecular determinants of synaptic plasticity. We investigated here the consequences of the inducible deletion of GluA1 AMPA receptors in glutamatergic neurons during late adolescence. We generated mutant mice with a tamoxifen-inducible deletion of GluA1 under the control of the CamKII promoter for temporally and spatially restricted gene manipulation. GluA1 ablation during late adolescence induced cognitive impairments, but also marked hyperlocomotion and sensorimotor gating deficits. Unlike the global genetic deletion of GluA1, inducible GluA1 ablation during late adolescence resulted in normal sociability. Deletion of GluA1 induced redistribution of GluA2 subunits, suggesting AMPA receptor trafficking deficits. Mutant animals showed increased hippocampal NMDA receptor expression and no change in striatal dopamine concentration. Our data provide new insight into the role of deficient AMPA receptors specifically during late adolescence in inducing several cognitive and behavioral alterations with possible relevance for neuropsychiatric disorders.


Asunto(s)
Trastornos del Conocimiento/metabolismo , Cuerpo Estriado/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Receptores AMPA/metabolismo , Conducta Social , Animales , Cuerpo Estriado/crecimiento & desarrollo , Dopamina/metabolismo , Hipocampo/crecimiento & desarrollo , Aprendizaje por Laberinto/fisiología , Memoria a Corto Plazo , Trastornos Mentales , Ratones , Ratones Transgénicos , Actividad Motora/fisiología , Fenotipo , Receptores AMPA/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Filtrado Sensorial/fisiología
13.
Hum Mol Genet ; 21(2): 344-57, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21994763

RESUMEN

Mutations in the postsynaptic scaffolding gene SHANK2 have recently been identified in individuals with autism spectrum disorder (ASD) and intellectual disability. However, the cellular and physiological consequences of these mutations in neurons remain unknown. We have analyzed the functional impact caused by two inherited and one de novo SHANK2 mutations from ASD individuals (L1008_P1009dup, T1127M, R462X). Although all three variants affect spine volume and have smaller SHANK2 cluster sizes, T1127M additionally fails to rescue spine volume in Shank2 knock-down neurons. R462X is not able to rescue spine volume and dendritic branching and lacks postsynaptic clustering, indicating the most severe dysfunction. To demonstrate that R462X when expressed in mouse can be linked to physiological effects, we analyzed synaptic transmission and behavior. Principal neurons of mice expressing rAAV-transduced SHANK2-R462X present a specific, long-lasting reduction in miniature postsynaptic AMPA receptor currents. This dominant negative effect translates into dose-dependent altered cognitive behavior of SHANK2-R462X-expressing mice, with an impact on the penetrance of ASD.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Morfogénesis , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Animales , Niño , Trastornos Generalizados del Desarrollo Infantil/patología , Técnicas de Silenciamiento del Gen , Hipocampo/patología , Humanos , Ratones , Mutación , Neuronas/fisiología
14.
Eur Arch Psychiatry Clin Neurosci ; 264(7): 625-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24895223

RESUMEN

Glutamatergic dysfunctions have recently been postulated to play a considerable role in mood disorders. However, molecular mechanisms underlying these effects have been poorly deciphered. Previous work demonstrated the contribution of GluA1-containing AMPA receptors (AMPAR) to a depression-like and anxiety-like phenotype. Here we investigated the effect of temporally and spatially restricted gene manipulation of GluA1 on behavioural correlates of mood disorders in mice. Here we show that tamoxifen-induced GluA1 deletion restricted to forebrain glutamatergic neurons of post-adolescent mice does not induce depression- and anxiety-like changes. This differs from the phenotype of mice with global AMPAR deletion suggesting that for mood regulation AMPAR may be particularly important on inhibitory interneurons or already early in development.


Asunto(s)
Regulación de la Expresión Génica/genética , Desamparo Adquirido , Trastornos del Humor/patología , Neuronas/metabolismo , Prosencéfalo/metabolismo , Receptores AMPA/deficiencia , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Conducta Exploratoria/fisiología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Trastornos del Humor/genética , Prosencéfalo/patología , Tiempo de Reacción/fisiología , Receptores AMPA/genética , Tamoxifeno/farmacología
15.
Transl Neurosci ; 15(1): 20220330, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38283997

RESUMEN

Objective: Heterozygous mutations within the voltage-gated sodium channel α subunit (SCN1A) are responsible for the majority of cases of Dravet syndrome (DS), a severe developmental and epileptic encephalopathy. Development of novel therapeutic approaches is mandatory in order to directly target the molecular consequences of the genetic defect. The aim of the present study was to investigate whether cis-acting long non-coding RNAs (lncRNAs) of SCN1A are expressed in brain specimens of children and adolescent with epilepsy as these molecules comprise possible targets for precision-based therapy approaches. Methods: We investigated SCN1A mRNA expression and expression of two SCN1A related antisense RNAs in brain tissues in different age groups of pediatric non-Dravet patients who underwent surgery for drug resistant epilepsy. The effect of different antisense oligonucleotides (ASOs) directed against SCN1A specific antisense RNAs on SCN1A expression was tested. Results: The SCN1A related antisense RNAs SCN1A-dsAS (downstream antisense, RefSeq identifier: NR_110598) and SCN1A-usAS (upstream AS, SCN1A-AS, RefSeq identifier: NR_110260) were widely expressed in the brain of pediatric patients. Expression patterns revealed a negative correlation of SCN1A-dsAS and a positive correlation of lncRNA SCN1A-usAS with SCN1A mRNA expression. Transfection of SK-N-AS cells with an ASO targeted against SCN1A-dsAS was associated with a significant enhancement of SCN1A mRNA expression and reduction in SCN1A-dsAS transcripts. Conclusion: These findings support the role of SCN1A-dsAS in the suppression of SCN1A mRNA generation. Considering the haploinsufficiency in genetic SCN1A related DS, SCN1A-dsAS is an interesting target candidate for the development of ASOs (AntagoNATs) based precision medicine therapeutic approaches aiming to enhance SCN1A expression in DS.

16.
Synapse ; 67(10): 648-55, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23505009

RESUMEN

We have recently demonstrated that, in C57/Bl6 mice, long-term voluntary wheel running is anxiogenic, and focal hippocampal irradiation prevents the increase in anxiety-like behaviors and neurobiological changes in the hippocampus induced by wheel running. Evidence supports a role of hippocampal 5-HT1A receptors in anxiety. Therefore, we investigated hippocampal binding and function of 5-HT1A receptors in this mouse model of anxiety. Four weeks of voluntary wheel running resulted in hippocampal subregion-specific changes in 5-HT1A receptor binding sites and function, as measured by autoradiography of [(3) H] 8-hydroxy-2-(di-n-propylamino)tetralin binding and agonist-stimulated binding of [(35) S]GTPγS to G proteins, respectively. In the dorsal CA1 region, 5-HT1A receptor binding and function were not altered by wheel running or irradiation. In the dorsal dentate gyrus and CA2/3 region, 5-HT1A receptor function was decreased by not only running but also irradiation. In the ventral pyramidal layer, wheel running resulted in a decrease of 5-HT1A receptor function, which was prevented by irradiation. Neither irradiation nor wheel running affected 5-HT1A receptors in medial prefrontal cortex or in the dorsal or median raphe nuclei. Our data indicate that downregulation of 5-HT1A receptor function in ventral pyramidal layer may play a role in anxiety-like behavior induced by wheel running.


Asunto(s)
Ansiedad/metabolismo , Hipocampo/metabolismo , Esfuerzo Físico , Receptor de Serotonina 5-HT1A/metabolismo , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Animales , Ansiedad/diagnóstico por imagen , Ansiedad/etiología , Modelos Animales de Enfermedad , Hipocampo/diagnóstico por imagen , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Radiografía , Ensayo de Unión Radioligante , Carrera , Agonistas de Receptores de Serotonina/farmacología , Tritio
17.
FASEB J ; 25(9): 2898-910, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21593433

RESUMEN

Parkinson's disease (PD) is a progressive age-related movement disorder that results primarily from the selective loss of midbrain dopaminergic (DA) neurons. Symptoms of PD can be induced by genetic mutations or by DA neuron-specific toxins. A specific ablation of an essential factor controlling ribosomal RNA transcription, TifIa, in adult mouse DA neurons represses mTOR signaling and leads to progressive neurodegeneration and PD-like phenotype. Using an inducible Cre system in adult mice, we show here that the specific ablation of Pten in adult mouse DA neurons leads to activation of mTOR pathway and is neuroprotective in genetic (TifIa deletion) and neurotoxin-induced (MPTP or 6OHDA) mouse models of PD. Adult mice with DA neuron-specific Pten deletion exhibit elevated expression of tyrosine hydroxylase, a rate-limiting enzyme in the dopamine biosynthesis pathway, associated with increased striatal dopamine content, and increased mRNA levels of Foxa2, Pitx3, En1, Nurr1, and Lmx1b-the essential factors for maintaining physiological functions of adult DA neurons. Pten deletion attenuates the loss of tyrosine hydroxylase-positive cells after 6OHDA treatment, restores striatal dopamine in TifIa-knockout and MPTP-treated mice, and rescues locomotor impairments caused by TifIa loss. Inhibition of Pten-dependent functions in adult DA neurons may represent a promising PD therapy.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Neuronas/metabolismo , Fosfohidrolasa PTEN/metabolismo , Enfermedad de Parkinson/prevención & control , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/efectos adversos , Animales , Cuerpo Estriado/metabolismo , Dihidroxifenilalanina/análogos & derivados , Dihidroxifenilalanina/toxicidad , Modelos Animales de Enfermedad , Dopamina/metabolismo , Dopaminérgicos/toxicidad , Eliminación de Gen , Ratones , Ratones Noqueados , Fosfohidrolasa PTEN/genética , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/genética , Proteínas del Complejo de Iniciación de Transcripción Pol1/genética , Proteínas del Complejo de Iniciación de Transcripción Pol1/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
18.
Hippocampus ; 21(9): 1028-35, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20572199

RESUMEN

Because stress represents a major precipitating event for psychiatric disorders, it is important to identify molecular mechanisms that may be altered in vulnerable individuals when exposed to stress. Here, we studied GluR-A(-/-) mice, animals with compromised AMPA receptor signaling, and characterized by a schizophrenic as well as depressive phenotype to investigate changes occurring in response to an acute stress. Wild-type and GluR-A(-/-) mice were exposed to a single immobilization stress and sacrificed immediately after the end of the stress for the analysis of activity regulated genes and of glutamatergic synapse responsiveness. The acute stress produced a marked increase in the hippocampal expression of Arc (activity-regulated cytoskeletal-associated protein) in GluR-A(-/-) , but not in wild-type mice, which was associated with a similar increase of phospho-CaMKII, a partner in the action of Arc. When looking at the glutamatergic response to stress in wild-type animals, we found that stress increased GluR-A phosphorylation on serine831, an effect that was paralleled by a significant increase of the phosphorylation of the main NMDA receptor subunits, that is, NR-1 and NR-2B. Conversely, the stress-induced modulation of NMDA receptor subunits was not observed in GluR-A(-/-) mice. We suggest that enhanced stress responsiveness in GluR-A(-/-) mice may be due, at least in part, to their inability to activate NMDA-mediated glutamatergic neurotransmission, suggesting that the integrity of AMPA/NMDA receptor function may be important for successful coping under stressful conditions.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteínas del Citoesqueleto/metabolismo , Ácido Glutámico/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Estrés Fisiológico , Animales , Glucocorticoides/metabolismo , Hipocampo/metabolismo , Ratones , Ratones Noqueados , Fosforilación , Serina/metabolismo , Sinapsinas/metabolismo , Transmisión Sináptica/fisiología
19.
Animals (Basel) ; 11(5)2021 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-34066005

RESUMEN

Ensuring optimal housing conditions for laboratory animals is a crucial prerequisite for high-quality and ethically justifiable in vivo science. In addition to guaranteeing animal welfare and promoting scientific validity, environmental sustainability is also increasingly gaining attention in laboratory animal facilities. Consequently, comprehensive management of such aspects is one of the core tasks of any research vivarium. Hygienic monitoring and adhering to standardized experimental protocols have been highlighted in the past; nevertheless, various environmental aspects of housing animals still need to be evaluated in greater depth. In this pilot study, we aimed at assessing the suitability of spelt and corncob as economical and ecologically friendly bedding substrates as compared with commonly used aspen wood chips. Therefore, following a descriptive study design, we examined the preferences of male and female Wistar rats for corncob and spelt under specific conditions. In addition, we evaluated potential effects on behavior, metabolism, and stress physiology. The type of bedding did not seem to influence behavior in the observed parameters but did have time- and sex-dependent effects on blood glucose. Furthermore, housing animals on spelt led to a significant reduction in food consumption, probably compensated for by the intake of spelt, and although it did not influence glucose levels, it may have certainly impacted the nutrient supply. Our descriptive pilot study, therefore, highlights the importance of a thorough condition-associated evaluation of even seemingly marginal environmental factors, when balancing potential cost-benefit advances in sustainability and questions of standardization and reproducibility of experimental protocols.

20.
Front Psychiatry ; 12: 750106, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34899420

RESUMEN

Extensive evidence suggests a dysfunction of the glutamate NMDA receptor (NMDAR) in schizophrenia, a severe psychiatric disorder with putative early neurodevelopmental origins, but clinical onset mainly during late adolescence. On the other hand, pharmacological models using NMDAR antagonists and the clinical manifestation of anti-NMDAR encephalitis indicate that NMDAR blockade/hypofunction can trigger psychosis also at adult stages, without any early developmental dysfunction. Previous genetic models of NMDAR hypofunction restricted to parvalbumin-positive interneurons indicate the necessity of an early postnatal impairment to trigger schizophrenia-like abnormalities, whereas the cellular substrates of NMDAR-mediated psychosis at adolescent/adult stages are unknown. Neuregulin 1 (NRG1) and its receptor ErbB4 represent schizophrenia-associated susceptibility factors that closely interact with NMDAR. To determine the neuronal populations implicated in "late" NMDAR-driven psychosis, we analyzed the effect of the inducible ablation of NMDARs in ErbB4-expressing cells in mice during late adolescence using a pharmacogenetic approach. Interestingly, the tamoxifen-inducible NMDAR deletion during this late developmental stage did not induce behavioral alterations resembling depression, schizophrenia or anxiety. Our data indicate that post-adolescent NMDAR deletion, even in a wider cell population than parvalbumin-positive interneurons, is also not sufficient to generate behavioral abnormalities resembling psychiatric disorders. Other neuronal substrates that have to be revealed by future studies, may underlie post-adolescent NMDAR-driven psychosis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA