Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 39(3): 110696, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35443170

RESUMEN

Stable function of networks requires that synapses adapt their strength to levels of neuronal activity, and failure to do so results in cognitive disorders. How such homeostatic regulation may be implemented in mammalian synapses remains poorly understood. Here we show that the phosphorylation status of several positions of the active-zone (AZ) protein RIM1 are relevant for synaptic glutamate release. Position RIMS1045 is necessary and sufficient for expression of silencing-induced homeostatic plasticity and is kept phosphorylated by serine arginine protein kinase 2 (SRPK2). SRPK2-induced upscaling of synaptic release leads to additional RIM1 nanoclusters and docked vesicles at the AZ and is not observed in the absence of RIM1 and occluded by RIMS1045E. Our data suggest that SRPK2 and RIM1 represent a presynaptic phosphosignaling hub that is involved in the homeostatic balance of synaptic coupling of neuronal networks.


Asunto(s)
Transmisión Sináptica , Vesículas Sinápticas , Animales , Proteínas de Unión al GTP/metabolismo , Homeostasis/fisiología , Mamíferos/metabolismo , Plasticidad Neuronal/fisiología , Terminales Presinápticos/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Vesículas Sinápticas/metabolismo
2.
Neuron ; 110(1): 51-69.e7, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-34706221

RESUMEN

Axons in the adult mammalian central nervous system fail to regenerate after spinal cord injury. Neurons lose their capacity to regenerate during development, but the intracellular processes underlying this loss are unclear. We found that critical components of the presynaptic active zone prevent axon regeneration in adult mice. Transcriptomic analysis combined with live-cell imaging revealed that adult primary sensory neurons downregulate molecular constituents of the synapse as they acquire the ability to rapidly grow their axons. Pharmacogenetic reduction of neuronal excitability stimulated axon regeneration after adult spinal cord injury. Genetic gain- and loss-of-function experiments uncovered that essential synaptic vesicle priming proteins of the presynaptic active zone, but not clostridial-toxin-sensitive VAMP-family SNARE proteins, inhibit axon regeneration. Systemic administration of Baclofen reduced voltage-dependent Ca2+ influx in primary sensory neurons and promoted their regeneration after spinal cord injury. These findings indicate that functional presynaptic active zones constitute a major barrier to axon regeneration.


Asunto(s)
Axones , Traumatismos de la Médula Espinal , Animales , Axones/metabolismo , Sistema Nervioso Central/metabolismo , Mamíferos , Ratones , Regeneración Nerviosa/fisiología , Neuronas/metabolismo , Traumatismos de la Médula Espinal/metabolismo
3.
Ann Neurol ; 89(4): 666-685, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33368582

RESUMEN

OBJECTIVE: Limbic encephalitis (LE) comprises a spectrum of inflammatory changes in affected brain structures including the presence of autoantibodies and lymphoid cells. However, the potential of distinct lymphocyte subsets alone to elicit key clinicopathological sequelae of LE potentially inducing temporal lobe epilepsy (TLE) with chronic spontaneous seizures and hippocampal sclerosis (HS) is unresolved. METHODS: Here, we scrutinized pathogenic consequences emerging from CD8+ T cells targeting hippocampal neurons by recombinant adeno-associated virus-mediated expression of the model-autoantigen ovalbumin (OVA) in CA1 neurons of OT-I/RAG1-/- mice (termed "OVA-CD8+ LE model"). RESULTS: Viral-mediated antigen transfer caused dense CD8+ T cell infiltrates confined to the hippocampal formation starting on day 5 after virus transduction. Flow cytometry indicated priming of CD8+ T cells in brain-draining lymph nodes preceding hippocampal invasion. At the acute model stage, the inflammatory process was accompanied by frequent seizure activity and impairment of hippocampal memory skills. Magnetic resonance imaging scans at day 7 of the OVA-CD8+ LE model revealed hippocampal edema and blood-brain barrier disruption that converted into atrophy until day 40. CD8+ T cells specifically targeted OVA-expressing, SIINFEKL-H-2Kb -positive CA1 neurons and caused segmental apoptotic neurodegeneration, astrogliosis, and microglial activation. At the chronic model stage, mice exhibited spontaneous recurrent seizures and persisting memory deficits, and the sclerotic hippocampus was populated with CD8+ T cells escorted by NK cells. INTERPRETATION: These data indicate that a CD8+ T-cell-initiated attack of distinct hippocampal neurons is sufficient to induce LE converting into TLE-HS. Intriguingly, the role of CD8+ T cells exceeds neurotoxic effects and points to their major pathogenic role in TLE following LE. ANN NEUROL 2021;89:666-685.


Asunto(s)
Linfocitos T CD8-positivos/patología , Epilepsia del Lóbulo Temporal/etiología , Epilepsia del Lóbulo Temporal/patología , Encefalitis Límbica/complicaciones , Encefalitis Límbica/patología , Animales , Barrera Hematoencefálica/patología , Región CA1 Hipocampal/patología , Epilepsia del Lóbulo Temporal/psicología , Hipocampo/patología , Proteínas de Homeodominio/genética , Encefalitis Límbica/psicología , Ganglios Linfáticos/patología , Imagen por Resonancia Magnética , Trastornos de la Memoria/etiología , Trastornos de la Memoria/psicología , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/patología , Ovalbúmina/genética , Ovalbúmina/inmunología , Fragmentos de Péptidos/genética , Convulsiones/genética , Convulsiones/patología
4.
Nat Methods ; 16(4): 351, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30820033

RESUMEN

The version of this paper originally published cited a preprint version of ref. 12 instead of the published version (Proc. Natl. Acad. Sci. USA 115, 5594-5599; 2018), which was available before this Nature Methods paper went to press. The reference information has been updated in the PDF and HTML versions of the article.

5.
J Neurosci ; 39(17): 3175-3187, 2019 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-30792272

RESUMEN

Transient brain insults, including status epilepticus (SE), can trigger a period of epileptogenesis during which functional and structural reorganization of neuronal networks occurs resulting in the onset of focal epileptic seizures. In recent years, mechanisms that regulate the dynamic transcription of individual genes during epileptogenesis and thereby contribute to the development of a hyperexcitable neuronal network have been elucidated. Our own results have shown early growth response 1 (Egr1) to transiently increase expression of the T-type voltage-dependent Ca2+ channel (VDCC) subunit CaV3.2, a key proepileptogenic protein. However, epileptogenesis involves complex and dynamic transcriptomic alterations; and so far, our understanding of the transcriptional control mechanism of gene regulatory networks that act in the same processes is limited. Here, we have analyzed whether Egr1 acts as a key transcriptional regulator for genes contributing to the development of hyperexcitability during epileptogenesis. We found Egr1 to drive the expression of the VDCC subunit α2δ4, which was augmented early and persistently after pilocarpine-induced SE. Furthermore, we show that increasing levels of α2δ4 in the CA1 region of the hippocampus elevate seizure susceptibility of mice by slightly decreasing local network activity. Interestingly, we also detected increased expression levels of Egr1 and α2δ4 in human hippocampal biopsies obtained from epilepsy surgery. In conclusion, Egr1 controls the abundance of the VDCC subunits CaV3.2 and α2δ4, which act synergistically in epileptogenesis, and thereby contributes to a seizure-induced "transcriptional Ca2+ channelopathy."SIGNIFICANCE STATEMENT The onset of focal recurrent seizures often occurs after an epileptogenic process induced by transient insults to the brain. Recently, transcriptional control mechanisms for individual genes involved in converting neurons hyperexcitable have been identified, including early growth response 1 (Egr1), which activates transcription of the T-type Ca2+ channel subunit CaV3.2. Here, we find Egr1 to regulate also the expression of the voltage-dependent Ca2+ channel subunit α2δ4, which was augmented after pilocarpine- and kainic acid-induced status epilepticus. In addition, we observed that α2δ4 affected spontaneous network activity and the susceptibility for seizure induction. Furthermore, we detected corresponding dynamics in human biopsies from epilepsy patients. In conclusion, Egr1 orchestrates a seizure-induced "transcriptional Ca2+ channelopathy" consisting of CaV3.2 and α2δ4, which act synergistically in epileptogenesis.


Asunto(s)
Canales de Calcio/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Epilepsia del Lóbulo Temporal/metabolismo , Hipocampo/metabolismo , Convulsiones/metabolismo , Estado Epiléptico/metabolismo , Animales , Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/fisiopatología , Hipocampo/fisiopatología , Humanos , Ácido Kaínico , Masculino , Ratones , Red Nerviosa/metabolismo , Red Nerviosa/fisiopatología , Pilocarpina , Convulsiones/inducido químicamente , Convulsiones/fisiopatología , Estado Epiléptico/inducido químicamente , Estado Epiléptico/fisiopatología
6.
Nat Methods ; 16(2): 206, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30602783

RESUMEN

In the version of this paper originally published, important figure labels in Fig. 3d were not visible. An image layer present in the authors' original figure that included two small dashed outlines and text labels indicating ROI 1 and ROI 2, as well as a scale bar and the name of the cell label, was erroneously altered during image processing. The figure has been corrected in the HTML and PDF versions of the paper.

7.
Mol Neurobiol ; 56(3): 1825-1840, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29934763

RESUMEN

Temporal lobe epilepsy (TLE) represents a devastating neurological condition, in which approximately 4/5 of patients remain refractory for anti-convulsive drugs. Epilepsy surgery biopsies often reveal the damage pattern of "hippocampal sclerosis" (HS) characterized not only by neuronal loss but also pronounced astrogliosis and inflammatory changes. Since TLE shares distinct pathogenetic aspects with multiple sclerosis (MS), we have here scrutinized therapeutic effects in experimental TLE of the immunmodulator fingolimod, which is established in MS therapy. Fingolimod targets sphingosine-phosphate receptors (S1PRs). mRNAs of fingolimod target S1PRs were augmented in two experimental post status epilepticus (SE) TLE mouse models (suprahippocampal kainate/pilocarpine). SE frequently induces chronic recurrent seizures after an extended latency referred to as epileptogenesis. Transient fingolimod treatment of mice during epileptogenesis after suprahippocampal kainate-induced SE revealed substantial reduction of chronic seizure activity despite lacking acute attenuation of SE itself. Intriguingly, fingolimod exerted robust anti-convulsive activity in kainate-induced SE mice treated in the chronic TLE stage and had neuroprotective and anti-gliotic effects and reduced cytotoxic T cell infiltrates. Finally, the expression profile of fingolimod target-S1PRs in human hippocampal biopsy tissue of pharmacoresistant TLE patients undergoing epilepsy surgery for seizure relief suggests repurposing of fingolimod as novel therapeutic perspective in focal epilepsies.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Clorhidrato de Fingolimod/uso terapéutico , Convulsiones/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/inducido químicamente , Ácido Kaínico , Masculino , Ratones , Pilocarpina , Convulsiones/inducido químicamente
8.
Nat Methods ; 15(11): 936-939, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30377363

RESUMEN

Single-wavelength fluorescent reporters allow visualization of specific neurotransmitters with high spatial and temporal resolution. We report variants of intensity-based glutamate-sensing fluorescent reporter (iGluSnFR) that are functionally brighter; detect submicromolar to millimolar amounts of glutamate; and have blue, cyan, green, or yellow emission profiles. These variants could be imaged in vivo in cases where original iGluSnFR was too dim, resolved glutamate transients in dendritic spines and axonal boutons, and allowed imaging at kilohertz rates.


Asunto(s)
Ácido Glutámico/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Microscopía Fluorescente/métodos , Neuronas/citología , Retina/citología , Corteza Visual/citología , Animales , Color , Femenino , Hurones , Colorantes Fluorescentes , Ácido Glutámico/análisis , Masculino , Ratones Endogámicos C57BL , Neuronas/metabolismo , Retina/metabolismo , Corteza Visual/metabolismo
9.
Epilepsia ; 58(7): 1159-1171, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28542864

RESUMEN

OBJECTIVE: Seizures in mesial temporal lobe epilepsy (MTLE) associated with hippocampal sclerosis are thought to develop with various latency intervals after an initial transient brain insult. To study seizure dynamics after an initial transient precipitating insult in a systematic fashion, we utilized continuous video-electroencephalography (EEG) monitoring after the induction of status epilepticus (SE) in a mouse MTLE model. METHODS: Continuous 24/7 video/telemetric hippocampal EEG recordings in the systemic pilocarpine MTLE mouse model. RESULTS: After SE, we observed emerging seizures interfering with the circadian EEG rhythms. The physiologic circadian EEG pattern of mice was transiently suppressed for 2.9 (mean) ± (SEM) 0.5 days after SE. This period was accompanied predominately by nonconvulsive seizure activity, followed by convulsive seizures at later stages. After the circadian rhythm was restored, spontaneous generalized seizures occurred mainly in a clustered manner in a narrow time window between 4 and 7 p.m. (light cycle 7 a.m./7 p.m.). Moreover, we demonstrate that depth-electrode implantation surgery transiently disturbs the physiologic EEG circadian cycle; variation of the time point of SE induction after electrode insertion surgery revealed a substantial impact on the epilepsy phenotype, which was more severe when SE occurred after postsurgical reappearance of EEG circadian cycling. SIGNIFICANCE: These data have several experimental and pathophysiologic implications. The impact of depth-electrode surgery on the phenotype has to be tightly controlled. In mice monitored after pilocarpine-induced SE, the "epileptogenesis" period is characterized by the dynamics of epileptiform activity toward behavioral recurrent seizure patterns. The striking clustering of spontaneous seizures at the transition from sleep to activity stages of mice has to be taken into account for future studies on the model. Improving our understanding of the molecular mechanisms that determine the circadian dynamics of seizure threshold remains an intriguing task for the future.


Asunto(s)
Ritmo Circadiano/efectos de los fármacos , Electroencefalografía/efectos de los fármacos , Epilepsia del Lóbulo Temporal/inducido químicamente , Epilepsia del Lóbulo Temporal/fisiopatología , Hipocampo/efectos de los fármacos , Hipocampo/fisiopatología , Pilocarpina , Procesamiento de Señales Asistido por Computador , Estado Epiléptico/inducido químicamente , Estado Epiléptico/fisiopatología , Grabación en Video , Animales , Enfermedad Crónica , Análisis por Conglomerados , Modelos Animales de Enfermedad , Hipocampo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Esclerosis , Sueño/efectos de los fármacos , Sueño/fisiología , Telemetría , Vigilia/efectos de los fármacos , Vigilia/fisiología
10.
J Neurosci ; 36(9): 2561-70, 2016 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-26936998

RESUMEN

Neuronal degeneration represents a pathogenetic hallmark after different brain insults, such as ischemia and status epilepticus (SE). Excessive release of glutamate triggered by pathophysiologic synaptic activity has been put forward as key mechanism in this context. In response to pathophysiologic synaptic activity, multiple signaling cascades are activated that ultimately initiate expression of specific sets of genes, which may decide between neuronal survival versus death. Recently, a core set of genes ["activity-regulated inhibitor of death" (AID) genes] including the transcription factor (TF) NPAS4 (neuronal PAS domain protein 4) has been found to provide activity-induced protection against neuronal death caused by excitotoxic stimulation. However, the downstream targets of AID action mediating neuroprotection remained so far unknown. Here, we have identified synaptotagmin 10 (Syt10), a vesicular Ca(2+) sensor, as the first neuroprotective effector protein downstream of the TF NPAS4. The expression of Syt10 is strongly upregulated by pathophysiologic synaptic activity after kainic acid (KA) exposure and its absence renders mouse hippocampal neurons highly susceptible to excitotoxic insults. We found NPAS4 as critical for the increase in Syt10 levels and in turn the ability of NPAS4 to confer neuroprotection against KA-induced excitotoxicity to be severely diminished in Syt10 knock-out neurons. In summary, our results point to an important role for signaling of the NPAS4-Syt10 pathway in the neuronal response to strong synaptic activity as a consequence of excitotoxic insults. SIGNIFICANCE STATEMENT: Aberrant synaptic activity is observed in many neurological disorders and has been suggested as an important factor contributing to the pathophysiology. Intriguingly, pathophysiologic activity can also trigger signaling cascades mediating potentially compensatory neuroprotection against excitotoxic insult. Here, we identify a new neuroprotective signaling cascade involving the activity-induced transcriptional regulator NPAS4 and the vesicular Ca(2+)-sensor protein synaptotagmin 10 (Syt10). Syt10 is required for NPAS4 to protect hippocampal neurons against excitotoxic cell death. NPAS4 in turn controls the activity of the Syt10 gene, which is strongly induced by pathophysiologic activity. Our results uncover an entirely unexpected, novel function of Syt10 underlying the response of neurons to pathophysiologic activity and provide new therapeutic perspectives for neurological disorders.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Degeneración Nerviosa/tratamiento farmacológico , Neuronas/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Sinaptotagminas/metabolismo , Animales , Apoptosis , Supervivencia Celular/fisiología , Células Cultivadas , Femenino , Hipocampo/citología , Humanos , Ácido Kaínico/toxicidad , Masculino , Ratones , Ratones Transgénicos , Degeneración Nerviosa/etiología , Neuronas/efectos de los fármacos , Cloruro de Potasio/farmacología , Embarazo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Wistar , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Sinaptotagminas/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
11.
Exp Cell Res ; 335(2): 157-64, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25720549

RESUMEN

Before fusing with the presynaptic plasma membrane to release neurotransmitter into the synaptic cleft synaptic vesicles have to be recruited to and docked at a specialized area of the presynaptic nerve terminal, the active zone. Exocytosis of synaptic vesicles is restricted to the presynaptic active zone, which is characterized by a unique and highly interconnected set of proteins. The protein network at the active zone is integrally involved in this process and also mediates changes in release properties, for example in response to alterations in the level of neuronal network activity. In recent years the development of novel techniques has greatly advanced our understanding of the molecular identity of respective active zone components as well as of the ultrastructure of this membranous subcompartment and of the SV release machinery. Furthermore, active zones are now viewed as dynamic structures whose composition and size are correlated with synaptic efficacy. Therefore, the dynamic remodeling of the protein network at the active zone has emerged as one potential mechanism underlying acute and long-term synaptic plasticity. Here, we will discuss this recent progress and its implications for our view of the role of the AZ in synaptic function.


Asunto(s)
Terminales Presinápticos/metabolismo , Vesículas Sinápticas/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/fisiología , Animales , Humanos , Neurotransmisores/metabolismo , Terminales Presinápticos/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA