Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Brain ; 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38478593

RESUMEN

Full-length RIM1 and 2 are key components of the presynaptic active zone that ubiquitously control excitatory and inhibitory neurotransmitter release. Here, we report that the function of the small RIM isoform RIM4, consisting of a single C2 domain, is strikingly different from that of the long isoforms. RIM4 is dispensable for neurotransmitter release but plays a postsynaptic, cell-type specific role in cerebellar Purkinje cells that is essential for normal motor function. In the absence of RIM4, Purkinje cell intrinsic firing is reduced and caffeine-sensitive, and dendritic integration of climbing fibre input is disturbed. Mice lacking RIM4, but not mice lacking RIM1/2, selectively in Purkinje cells exhibit a severe, hours-long paroxysmal dystonia. These episodes can also be induced by caffeine, ethanol or stress and closely resemble the deficits seen with mutations of the PNKD (paroxysmal non-kinesigenic dystonia) gene. Our data reveal essential postsynaptic functions of RIM proteins and show non-overlapping specialized functions of a small isoform despite high homology to a single domain in the full-length proteins.

2.
Neurobiol Dis ; 190: 106364, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38008342

RESUMEN

Gangliogliomas (GGs) represent the most frequent glioneuronal tumor entity associated with chronic recurrent seizures; rare anaplastic GGs variants retain the glioneuronal character. So far, key mechanisms triggering chronic hyperexcitability in the peritumoral area are unresolved. Based on a recent mouse model for anaplastic GG (BRAFV600E, mTOR activation and Trp53KO) we here assessed the influence of GG-secreted factors on non-neoplastic cells in-vitro. We generated conditioned medium (CM) from primary GG cell cultures to developing primary cortical neurons cultured on multielectrode-arrays and assessed their electrical activity in comparison to neurons incubated with naïve and neuronal CMs. Our results showed that the GG CM, while not affecting the mean firing rates of networks, strongly accelerated the formation of functional networks as indicated increased synchrony of firing and burst activity. Washing out the GG CM did not reverse these effects indicating an irreversible effect on the neuronal network. Mass spectrometry analysis of GG CM detected several enriched proteins associated with neurogenesis as well as gliogenesis, including Gap43, App, Apoe, S100a8, Tnc and Sod1. Concomitantly, immunocytochemical analysis of the neuronal cultures exposed to GG CM revealed abundant astrocytes suggesting that the GG-secreted factors induce astroglial proliferation. Pharmacological inhibition of astrocyte proliferation only partially reversed the accelerated network maturation in neuronal cultures exposed to GG CM indicating that the GG CM exerts a direct effect on the neuronal component. Taken together, we demonstrate that GG-derived paracrine signaling alone is sufficient to induce accelerated neuronal network development accompanied by astrocytic proliferation. Perspectively, a deeper understanding of factors involved may serve as the basis for future therapeutic approaches.


Asunto(s)
Neoplasias Encefálicas , Ganglioglioma , Humanos , Animales , Ratones , Ganglioglioma/complicaciones , Ganglioglioma/metabolismo , Ganglioglioma/patología , Neoplasias Encefálicas/metabolismo , Alta del Paciente , Convulsiones/complicaciones , Neuronas/metabolismo
3.
Glia ; 71(9): 2210-2233, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37226895

RESUMEN

Oligodendrocyte precursor cells (OPCs) generate oligodendrocytes, a process that may be tuned by neuronal activity, possibly via synaptic connections to OPCs. However, a developmental role of synaptic signaling to OPCs has so far not been shown unequivocally. To address this question, we comparatively analyzed functional and molecular characteristics of highly proliferative and migratory OPCs in the embryonic brain. Embryonic OPCs in mice (E18.5) shared the expression of voltage-gated ion channels and their dendritic morphology with postnatal OPCs, but almost completely lacked functional synaptic currents. Transcriptomic profiling of PDGFRα+ OPCs revealed a limited abundance of genes coding for postsynaptic signaling and synaptogenic cell adhesion molecules in the embryonic versus the postnatal period. RNA sequencing of single OPCs showed that embryonic synapse-lacking OPCs are found in clusters distinct from postnatal OPCs and with similarities to early progenitors. Furthermore, single-cell transcriptomics demonstrated that synaptic genes are transiently expressed only by postnatal OPCs until they start to differentiate. Taken together, our results indicate that embryonic OPCs represent a unique developmental stage biologically resembling postnatal OPCs but without synaptic input and a transcriptional signature in the continuum between OPCs and neural precursors.


Asunto(s)
Células Precursoras de Oligodendrocitos , Ratones , Animales , Células Precursoras de Oligodendrocitos/metabolismo , Ratones Transgénicos , Oligodendroglía/metabolismo , Neuronas/fisiología , Neurogénesis/fisiología , Diferenciación Celular/fisiología
4.
Dev Neurosci ; 45(2): 53-65, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36538906

RESUMEN

Gangliogliomas (GGs), composed of dysmorphic neurons and neoplastic astroglia, represent the most frequent tumor entity associated with chronic recurrent epileptic seizures. So far, a systematic analysis of potential differences in neurochemical profiles of dysmorphic tumoral neurons as well as neurons of the peritumoral microenvironment (PTME) was hampered by the inability to unequivocally differentiate between the distinct neuronal components in human GG biopsies. Here, we have applied a novel GG mouse model that allows to clearly resolve the neurochemical profiles of GG-intrinsic versus PTME neurons. For this purpose, glioneuronal tumors in mice were induced by intraventricular in utero electroporation (IUE) of piggyBac-based plasmids for BRAFV600E and activated Akt (AktT308D/S473D, further referred to as AktDD) and analyzed neurochemically by immunocytochemistry against specific marker proteins. IUE of BRAFV600E/AktDD in mice resulted in tumors with the morphological features of human GGs. Our immunocytochemical analysis revealed a strong reduction of GABAARα1 immunoreactivity in the tumor compared to the PTME. In contrast, the extent of NMDAR1 immunoreactivity in the tumor appeared comparable to the PTME. Interestingly, tumor cells maintained the potential to express both receptors. Fittingly, the abundance of the presynaptic vesicular neurotransmitter transporters VGLUT1 and VGAT was also decreased in the tumor. Additionally, the fraction of parvalbumin and somatostatin nonneoplastic interneurons was reduced. In conclusion, changes in the levels of key proteins in neurotransmitter signaling suggest a loss of synapses and may thereby lead to neuronal network alterations in mouse GGs.


Asunto(s)
Neoplasias Encefálicas , Epilepsia , Ganglioglioma , Humanos , Ratones , Animales , Ganglioglioma/complicaciones , Ganglioglioma/metabolismo , Ganglioglioma/patología , Convulsiones , Neuronas/metabolismo , Epilepsia/complicaciones , Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Microambiente Tumoral
5.
J Neurosci ; 41(39): 8111-8125, 2021 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-34400520

RESUMEN

The size and structure of the dendritic arbor play important roles in determining how synaptic inputs of neurons are converted to action potential output. The regulatory mechanisms governing the development of dendrites, however, are insufficiently understood. The evolutionary conserved Ste20/Hippo kinase pathway has been proposed to play an important role in regulating the formation and maintenance of dendritic architecture. A key element of this pathway, Ste20-like kinase (SLK), regulates cytoskeletal dynamics in non-neuronal cells and is strongly expressed throughout neuronal development. However, its function in neurons is unknown. We show that, during development of mouse cortical neurons, SLK has a surprisingly specific role for proper elaboration of higher, ≥ third-order dendrites both in male and in female mice. Moreover, we demonstrate that SLK is required to maintain excitation-inhibition balance. Specifically, SLK knockdown caused a selective loss of inhibitory synapses and functional inhibition after postnatal day 15, whereas excitatory neurotransmission was unaffected. Finally, we show that this mechanism may be relevant for human disease, as dysmorphic neurons within human cortical malformations revealed significant loss of SLK expression. Overall, the present data identify SLK as a key regulator of both dendritic complexity during development and inhibitory synapse maintenance.SIGNIFICANCE STATEMENT We show that dysmorphic neurons of human epileptogenic brain lesions have decreased levels of the Ste20-like kinase (SLK). Decreasing SLK expression in mouse neurons revealed that SLK has essential functions in forming the neuronal dendritic tree and in maintaining inhibitory connections with neighboring neurons.


Asunto(s)
Corteza Cerebral/metabolismo , Dendritas/genética , Inhibición Neural/genética , Proteínas Serina-Treonina Quinasas/genética , Sinapsis/genética , Transmisión Sináptica/fisiología , Adolescente , Adulto , Anciano , Animales , Corteza Cerebral/patología , Niño , Preescolar , Dendritas/metabolismo , Dendritas/patología , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Persona de Mediana Edad , Proteínas Serina-Treonina Quinasas/metabolismo , Sinapsis/metabolismo , Sinapsis/patología , Adulto Joven
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.
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.

8.
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
9.
Acta Neuropathol ; 144(1): 107-127, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35551471

RESUMEN

Mesial temporal lobe epilepsy with hippocampal sclerosis and a history of febrile seizures is associated with common variation at rs7587026, located in the promoter region of SCN1A. We sought to explore possible underlying mechanisms. SCN1A expression was analysed in hippocampal biopsy specimens of individuals with mesial temporal lobe epilepsy with hippocampal sclerosis who underwent surgical treatment, and hippocampal neuronal cell loss was quantitatively assessed using immunohistochemistry. In healthy individuals, hippocampal volume was measured using MRI. Analyses were performed stratified by rs7587026 type. To study the functional consequences of increased SCN1A expression, we generated, using transposon-mediated bacterial artificial chromosome transgenesis, a zebrafish line expressing exogenous scn1a, and performed EEG analysis on larval optic tecta at 4 day post-fertilization. Finally, we used an in vitro promoter analysis to study whether the genetic motif containing rs7587026 influences promoter activity. Hippocampal SCN1A expression differed by rs7587026 genotype (Kruskal-Wallis test P = 0.004). Individuals homozygous for the minor allele showed significantly increased expression compared to those homozygous for the major allele (Dunn's test P = 0.003), and to heterozygotes (Dunn's test P = 0.035). No statistically significant differences in hippocampal neuronal cell loss were observed between the three genotypes. Among 597 healthy participants, individuals homozygous for the minor allele at rs7587026 displayed significantly reduced mean hippocampal volume compared to major allele homozygotes (Cohen's D = - 0.28, P = 0.02), and to heterozygotes (Cohen's D = - 0.36, P = 0.009). Compared to wild type, scn1lab-overexpressing zebrafish larvae exhibited more frequent spontaneous seizures [one-way ANOVA F(4,54) = 6.95 (P < 0.001)]. The number of EEG discharges correlated with the level of scn1lab overexpression [one-way ANOVA F(4,15) = 10.75 (P < 0.001]. Finally, we showed that a 50 bp promoter motif containing rs7587026 exerts a strong regulatory role on SCN1A expression, though we could not directly link this to rs7587026 itself. Our results develop the mechanistic link between rs7587026 and mesial temporal lobe epilepsy with hippocampal sclerosis and a history of febrile seizures. Furthermore, we propose that quantitative precision may be important when increasing SCN1A expression in current strategies aiming to treat seizures in conditions involving SCN1A haploinsufficiency, such as Dravet syndrome.


Asunto(s)
Epilepsia del Lóbulo Temporal , Epilepsia , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Convulsiones Febriles , Proteínas de Pez Cebra/metabolismo , Animales , Epilepsia/genética , Epilepsia del Lóbulo Temporal/genética , Genómica , Gliosis/patología , Hipocampo/patología , Humanos , Canal de Sodio Activado por Voltaje NAV1.1/genética , Esclerosis/patología , Convulsiones Febriles/complicaciones , Convulsiones Febriles/genética , Pez Cebra
10.
PLoS Biol ; 17(3): e3000170, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30822303

RESUMEN

Depolarization of presynaptic terminals stimulates calcium influx, which evokes neurotransmitter release and activates phosphorylation-based signalling. Here, we present the first global temporal profile of presynaptic activity-dependent phospho-signalling, which includes two KCl stimulation levels and analysis of the poststimulus period. We profiled 1,917 regulated phosphopeptides and bioinformatically identified six temporal patterns of co-regulated proteins. The presynaptic proteins with large changes in phospho-status were again prominently regulated in the analysis of 7,070 activity-dependent phosphopeptides from KCl-stimulated cultured hippocampal neurons. Active zone scaffold proteins showed a high level of activity-dependent phospho-regulation that far exceeded the response from postsynaptic density scaffold proteins. Accordingly, bassoon was identified as the major target of neuronal phospho-signalling. We developed a probabilistic computational method, KinSwing, which matched protein kinase substrate motifs to regulated phosphorylation sites to reveal underlying protein kinase activity. This approach allowed us to link protein kinases to profiles of co-regulated presynaptic protein networks. Ca2+- and calmodulin-dependent protein kinase IIα (CaMKIIα) responded rapidly, scaled with stimulus strength, and had long-lasting activity. Mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) was the main protein kinase predicted to control a distinct and significant pattern of poststimulus up-regulation of phosphorylation. This work provides a unique resource of activity-dependent phosphorylation sites of synaptosomes and neurons, the vast majority of which have not been investigated with regard to their functional impact. This resource will enable detailed characterization of the phospho-regulated mechanisms impacting the plasticity of neurotransmitter release.


Asunto(s)
Terminales Presinápticos/metabolismo , Sinaptosomas/metabolismo , Animales , Calcio/metabolismo , Calmodulina/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Masculino , Espectrometría de Masas , Fosfoproteínas/metabolismo , Fosforilación , Cloruro de Potasio/farmacología , Terminales Presinápticos/fisiología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Sinaptosomas/fisiología
11.
J Cell Sci ; 132(12)2019 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-31138677

RESUMEN

Mechanisms that regulate the formation of membrane-less cellular organelles, such as neuronal RNA granules and stress granules, have gained increasing attention over the past years. These granules consist of RNA and a plethora of RNA-binding proteins. Mutations in RNA-binding proteins have been found in neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). By performing pulldown experiments and subsequent mass spectrometry on mouse brain lysates, we discovered that the de-ubiquitylating enzyme OTU domain-containing protein 4 (OTUD4) unexpectedly is part of a complex network of multiple RNA-binding proteins, including core stress granule factors, such as FMRP (also known as FMR1), SMN1, G3BP1 and TIA1. We show that OTUD4 binds RNA, and that several of its interactions with RNA-binding proteins are RNA dependent. OTUD4 is part of neuronal RNA transport granules in rat hippocampal neurons under physiological conditions, whereas upon cellular stress, OTUD4 is recruited to cytoplasmic stress granules. Knockdown of OTUD4 in HeLa cells resulted in defects in stress granule formation and led to apoptotic cell death. Together, we characterize OTUD4 as a new RNA-binding protein with a suggested function in regulation of translation.


Asunto(s)
ADN Helicasas/genética , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Proteasas Ubiquitina-Específicas/genética , Proteasas Ubiquitina-Específicas/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Gránulos Citoplasmáticos/metabolismo , Proteínas de Unión al ADN/metabolismo , Células HeLa , Humanos , Ratones Endogámicos C57BL , Mutación/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo
12.
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
13.
Ann Neurol ; 87(6): 869-884, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32196746

RESUMEN

OBJECTIVE: Assess occurrence of the dendritic spine scaffolding protein Drebrin as a pathophysiologically relevant autoantibody target in patients with recurrent seizures and suspected encephalitis as leading symptoms. METHODS: Sera of 4 patients with adult onset epilepsy and suspected encephalitis of unresolved etiology and equivalent results in autoantibody screening were subjected to epitope identification. We combined a wide array of approaches, ranging from immunoblotting, immunoprecipitation, mass spectrometry, subcellular binding pattern analyses in primary neuronal cultures, and immunohistochemistry in brains of wild-type and Drebrin knockout mice to in vitro analyses of impaired synapse formation, morphology, and aberrant neuronal excitability by antibody exposure. RESULTS: In the serum of a patient with adult onset epilepsy and suspected encephalitis, a strong signal at ∼70kDa was detected by immunoblotting, for which mass spectrometry revealed Drebrin as the putative antigen. Three other patients whose sera also showed strong immunoreactivity around 70kDa on Western blotting were also anti-Drebrin-positive. Seizures, memory impairment, and increased protein content in cerebrospinal fluid occurred in anti-Drebrin-seropositive patients. Alterations in cerebral magnetic resonance imaging comprised amygdalohippocampal T2-signal increase and hippocampal sclerosis. Diagnostic biopsy revealed T-lymphocytic encephalitis in an anti-Drebrin-seropositive patient. Exposure of primary hippocampal neurons to anti-Drebrin autoantibodies resulted in aberrant synapse composition and Drebrin distribution as well as increased spike rates and the emergence of burst discharges reflecting network hyperexcitability. INTERPRETATION: Anti-Drebrin autoantibodies define a chronic syndrome of recurrent seizures and neuropsychiatric impairment as well as inflammation of limbic and occasionally cortical structures. Immunosuppressant therapies should be considered in this disorder. ANN NEUROL 2020;87:869-884.


Asunto(s)
Autoanticuerpos/inmunología , Encefalitis/inmunología , Neuropéptidos/inmunología , Convulsiones/inmunología , Adulto , Anciano , Animales , Encefalitis/diagnóstico por imagen , Epítopos/inmunología , Femenino , Hipocampo/inmunología , Hipocampo/patología , Humanos , Imagen por Resonancia Magnética , Masculino , Trastornos Mentales/inmunología , Trastornos Mentales/psicología , Ratones Noqueados , Persona de Mediana Edad , Neuroimagen , Convulsiones/diagnóstico por imagen , Sinapsis/inmunología , Adulto Joven
14.
Epilepsia ; 62(1): e29-e34, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33319393

RESUMEN

Increasing evidence indicates the pathogenetic relevance of regulatory genomic motifs for variability in the manifestation of brain disorders. In this context, cis-regulatory effects of single nucleotide polymorphisms (SNPs) on gene expression can contribute to changing transcript levels of excitability-relevant molecules and episodic seizure manifestation in epilepsy. Biopsy specimens of patients undergoing epilepsy surgery for seizure relief provide unique insights into the impact of promoter SNPs on corresponding mRNA expression. Here, we have scrutinized whether two linked regulatory SNPs (rs2744575; 4779C > G and rs4646830; 4854C > G) located in the aldehyde dehydrogenase 5a1 (succinic semialdehyde dehydrogenase; ALDH5A1) gene promoter are associated with expression of corresponding mRNAs in epileptic hippocampi (n = 43). The minor ALDH5A1-GG haplotype associates with significantly lower ALDH5A1 transcript abundance. Complementary in vitro analyses in neural cell cultures confirm this difference and further reveal a significantly constricted range for the minor ALDH5A1 haplotype of promoter activity regulation through the key epileptogenesis transcription factor Egr1 (early growth response 1). The present data suggest systematic analyses in human hippocampal tissue as a useful approach to unravel the impact of epilepsy candidate SNPs on associated gene expression. Aberrant ALDH5A1 promoter regulation in functional terms can contribute to impaired γ-aminobutyric acid homeostasis and thereby network excitability and seizure propensity.


Asunto(s)
Epilepsia del Lóbulo Temporal/genética , Hipocampo/metabolismo , Neuronas/metabolismo , ARN Mensajero/metabolismo , Succionato-Semialdehído Deshidrogenasa/genética , Animales , Línea Celular , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Epilepsia del Lóbulo Temporal/patología , Epilepsia del Lóbulo Temporal/cirugía , Perfilación de la Expresión Génica , Haplotipos , Hipocampo/patología , Humanos , Técnicas In Vitro , Ratones , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas/genética , Ratas , Esclerosis
15.
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
16.
Nat Chem Biol ; 14(9): 861-869, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30061718

RESUMEN

Fluorescent sensors are an essential part of the experimental toolbox of the life sciences, where they are used ubiquitously to visualize intra- and extracellular signaling. In the brain, optical neurotransmitter sensors can shed light on temporal and spatial aspects of signal transmission by directly observing, for instance, neurotransmitter release and spread. Here we report the development and application of the first optical sensor for the amino acid glycine, which is both an inhibitory neurotransmitter and a co-agonist of the N-methyl-D-aspartate receptors (NMDARs) involved in synaptic plasticity. Computational design of a glycine-specific binding protein allowed us to produce the optical glycine FRET sensor (GlyFS), which can be used with single and two-photon excitation fluorescence microscopy. We took advantage of this newly developed sensor to test predictions about the uneven spatial distribution of glycine in extracellular space and to demonstrate that extracellular glycine levels are controlled by plasticity-inducing stimuli.


Asunto(s)
Colorantes Fluorescentes/química , Glicina/análisis , Hipocampo/química , Animales , Células Cultivadas , Transferencia Resonante de Energía de Fluorescencia , Colorantes Fluorescentes/síntesis química , Células HEK293 , Humanos , Masculino , Imagen Óptica , Ratas , Ratas Wistar
17.
Am J Hum Genet ; 99(6): 1292-1304, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27866708

RESUMEN

Uncombable hair syndrome (UHS), also known as "spun glass hair syndrome," "pili trianguli et canaliculi," or "cheveux incoiffables" is a rare anomaly of the hair shaft that occurs in children and improves with age. UHS is characterized by dry, frizzy, spangly, and often fair hair that is resistant to being combed flat. Until now, both simplex and familial UHS-affected case subjects with autosomal-dominant as well as -recessive inheritance have been reported. However, none of these case subjects were linked to a molecular genetic cause. Here, we report the identification of UHS-causative mutations located in the three genes PADI3 (peptidylarginine deiminase 3), TGM3 (transglutaminase 3), and TCHH (trichohyalin) in a total of 11 children. All of these individuals carry homozygous or compound heterozygous mutations in one of these three genes, indicating an autosomal-recessive inheritance pattern in the majority of UHS case subjects. The two enzymes PADI3 and TGM3, responsible for posttranslational protein modifications, and their target structural protein TCHH are all involved in hair shaft formation. Elucidation of the molecular outcomes of the disease-causing mutations by cell culture experiments and tridimensional protein models demonstrated clear differences in the structural organization and activity of mutant and wild-type proteins. Scanning electron microscopy observations revealed morphological alterations in hair coat of Padi3 knockout mice. All together, these findings elucidate the molecular genetic causes of UHS and shed light on its pathophysiology and hair physiology in general.


Asunto(s)
Antígenos/genética , Enfermedades del Cabello/genética , Cabello/crecimiento & desarrollo , Hidrolasas/genética , Proteínas de Filamentos Intermediarios/genética , Mutación , Transglutaminasas/genética , Adolescente , Animales , Secuencia de Bases , Línea Celular , Codón sin Sentido , Femenino , Cabello/anomalías , Cabello/anatomía & histología , Cabello/metabolismo , Humanos , Hidrolasas/deficiencia , Hidrolasas/metabolismo , Masculino , Ratones , Ratones Noqueados , Modelos Moleculares , Mutación Missense/genética , Conformación Proteica , Arginina Deiminasa Proteína-Tipo 3 , Desiminasas de la Arginina Proteica , Transglutaminasas/deficiencia , Transglutaminasas/metabolismo , Vibrisas/anomalías
18.
Brain ; 141(5): 1350-1374, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29538625

RESUMEN

De novo heterozygous mutations in STXBP1/Munc18-1 cause early infantile epileptic encephalopathies (EIEE4, OMIM #612164) characterized by infantile epilepsy, developmental delay, intellectual disability, and can include autistic features. We characterized the cellular deficits for an allelic series of seven STXBP1 mutations and developed four mouse models that recapitulate the abnormal EEG activity and cognitive aspects of human STXBP1-encephalopathy. Disease-causing STXBP1 variants supported synaptic transmission to a variable extent on a null background, but had no effect when overexpressed on a heterozygous background. All disease variants had severely decreased protein levels. Together, these cellular studies suggest that impaired protein stability and STXBP1 haploinsufficiency explain STXBP1-encephalopathy and that, therefore, Stxbp1+/- mice provide a valid mouse model. Simultaneous video and EEG recordings revealed that Stxbp1+/- mice with different genomic backgrounds recapitulate the seizure/spasm phenotype observed in humans, characterized by myoclonic jerks and spike-wave discharges that were suppressed by the antiepileptic drug levetiracetam. Mice heterozygous for Stxbp1 in GABAergic neurons only, showed impaired viability, 50% died within 2-3 weeks, and the rest showed stronger epileptic activity. c-Fos staining implicated neocortical areas, but not other brain regions, as the seizure foci. Stxbp1+/- mice showed impaired cognitive performance, hyperactivity and anxiety-like behaviour, without altered social behaviour. Taken together, these data demonstrate the construct, face and predictive validity of Stxbp1+/- mice and point to protein instability, haploinsufficiency and imbalanced excitation in neocortex, as the underlying mechanism of STXBP1-encephalopathy. The mouse models reported here are valid models for development of therapeutic interventions targeting STXBP1-encephalopathy.


Asunto(s)
Encefalopatías/complicaciones , Encefalopatías/genética , Epilepsia/fisiopatología , Haploinsuficiencia/genética , Discapacidad Intelectual/genética , Proteínas Munc18/genética , Animales , Anticonvulsivantes/uso terapéutico , Encefalopatías/tratamiento farmacológico , Células Cultivadas , Corteza Cerebral/citología , Embrión de Mamíferos , Epilepsia/tratamiento farmacológico , Epilepsia/genética , Conducta Exploratoria/efectos de los fármacos , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Discapacidad Intelectual/complicaciones , Levetiracetam/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Munc18/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Sinapsinas/genética , Sinapsinas/metabolismo , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/genética
19.
J Neurosci ; 37(33): 7848-7863, 2017 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-28701482

RESUMEN

RAB3A-interacting molecule (RIM) proteins are important regulators of transmitter release from active zones. At conventional chemical synapses, RIMs contribute substantially to vesicle priming and docking and their loss reduces the readily releasable pool of synaptic vesicles by up to 75%. The priming function of RIMs is mediated via the formation of a tripartite complex with Munc13 and RAB3A, which brings synaptic vesicles in close proximity to Ca2+ channels and the fusion site and activates Munc13. We reported previously that, at mouse photoreceptor ribbon synapses, vesicle priming is Munc13 independent. In this study, we examined RIM expression, distribution, and function at male and female mouse photoreceptor ribbon synapses. We provide evidence that RIM1α and RIM1ß are highly likely absent from mouse photoreceptors and that RIM2α is the major large RIM isoform present at photoreceptor ribbon synapses. We show that mouse photoreceptors predominantly express RIM2 variants that lack the interaction domain for Munc13. Loss of full-length RIM2α in a RIM2α mutant mouse only marginally perturbs photoreceptor synaptic transmission. Our findings therefore strongly argue for a priming mechanism at the photoreceptor ribbon synapse that is independent of the formation of a RIM-Munc13-RAB3A complex and thus provide further evidence for a fundamental difference between photoreceptor ribbon synapses and conventional chemical synapses in synaptic vesicle exocytosis.SIGNIFICANCE STATEMENT RAB3A-interacting molecules 1 and 2 (RIM1/2) are essential regulators of exocytosis. At conventional chemical synapses, their function involves Ca2+ channel clustering and synaptic vesicle priming and docking through interactions with Munc13 and RAB3A, respectively. Examining wild-type and RIM2 mutant mice, we show here that the sensory photoreceptor ribbon synapses most likely lack RIM1 and predominantly express RIM2 variants that lack the interaction domain for Munc13. Our findings demonstrate that the photoreceptor-specific RIM variants are not essential for synaptic vesicle priming at photoreceptor ribbon synapses, which represents a fundamental difference between photoreceptor ribbon synapses and conventional chemical synapses with respect to synaptic vesicle priming mechanisms.


Asunto(s)
Proteínas de Unión al GTP/biosíntesis , Células Fotorreceptoras de Vertebrados/metabolismo , Sinapsis/metabolismo , Animales , Células Cultivadas , Femenino , Proteínas de Unión al GTP/análisis , Proteínas de Unión al GTP/genética , Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células 3T3 NIH , Células Fotorreceptoras de Vertebrados/química , Sinapsis/química , Sinapsis/genética
20.
Proc Natl Acad Sci U S A ; 112(24): E3141-9, 2015 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-26034270

RESUMEN

Ca(2+) influx triggers the fusion of synaptic vesicles at the presynaptic active zone (AZ). Here we demonstrate a role of Ras-related in brain 3 (Rab3)-interacting molecules 2α and ß (RIM2α and RIM2ß) in clustering voltage-gated CaV1.3 Ca(2+) channels at the AZs of sensory inner hair cells (IHCs). We show that IHCs of hearing mice express mainly RIM2α, but also RIM2ß and RIM3γ, which all localize to the AZs, as shown by immunofluorescence microscopy. Immunohistochemistry, patch-clamp, fluctuation analysis, and confocal Ca(2+) imaging demonstrate that AZs of RIM2α-deficient IHCs cluster fewer synaptic CaV1.3 Ca(2+) channels, resulting in reduced synaptic Ca(2+) influx. Using superresolution microscopy, we found that Ca(2+) channels remained clustered in stripes underneath anchored ribbons. Electron tomography of high-pressure frozen synapses revealed a reduced fraction of membrane-tethered vesicles, whereas the total number of membrane-proximal vesicles was unaltered. Membrane capacitance measurements revealed a reduction of exocytosis largely in proportion with the Ca(2+) current, whereas the apparent Ca(2+) dependence of exocytosis was unchanged. Hair cell-specific deletion of all RIM2 isoforms caused a stronger reduction of Ca(2+) influx and exocytosis and significantly impaired the encoding of sound onset in the postsynaptic spiral ganglion neurons. Auditory brainstem responses indicated a mild hearing impairment on hair cell-specific deletion of all RIM2 isoforms or global inactivation of RIM2α. We conclude that RIM2α and RIM2ß promote a large complement of synaptic Ca(2+) channels at IHC AZs and are required for normal hearing.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Células Ciliadas Auditivas Internas/metabolismo , Proteínas de Unión al GTP rab3/metabolismo , Animales , Señalización del Calcio , Tomografía con Microscopio Electrónico , Potenciales Evocados Auditivos del Tronco Encefálico , Exocitosis , Células Ciliadas Auditivas Internas/ultraestructura , Audición/fisiología , Activación del Canal Iónico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Emisiones Otoacústicas Espontáneas , Técnicas de Placa-Clamp , Ganglio Espiral de la Cóclea/metabolismo , Sinapsis/metabolismo , Sinapsis/ultraestructura , Vesículas Sinápticas/metabolismo , Proteínas de Unión al GTP rab3/deficiencia , Proteínas de Unión al GTP rab3/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA