Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-37962617

RESUMEN

PURPOSE: Staphylococcus aureus is the most common and impactful multi-drug resistant pathogen implicated in (periprosthetic) joint infections (PJI) and fracture-related infections (FRI). Therefore, the present proof-of-principle study was aimed at the rapid detection of S. aureus in synovial fluids and biofilms on extracted osteosynthesis materials through bacteria-targeted fluorescence imaging with the 'smart-activatable' DNA-based AttoPolyT probe. This fluorogenic oligonucleotide probe yields large fluorescence increases upon cleavage by micrococcal nuclease, an enzyme secreted by S. aureus. METHODS: Synovial fluids from patients with suspected PJI and extracted osteosynthesis materials from trauma patients with suspected FRI were inspected for S. aureus nuclease activity with the AttoPolyT probe. Biofilms on osteosynthesis materials were imaged with the AttoPolyT probe and a vancomycin-IRDye800CW conjugate (vanco-800CW) specific for Gram-positive bacteria. RESULTS: 38 synovial fluid samples were collected and analyzed. Significantly higher fluorescence levels were measured for S. aureus-positive samples compared to, respectively, other Gram-positive bacterial pathogens (p < 0.0001), Gram-negative bacterial pathogens (p = 0.0038) and non-infected samples (p = 0.0030), allowing a diagnosis of S. aureus-associated PJI within 2 h. Importantly, S. aureus-associated biofilms on extracted osteosynthesis materials from patients with FRI were accurately imaged with the AttoPolyT probe, allowing their correct distinction from biofilms formed by other Gram-positive bacteria detected with vanco-800CW within 15 min. CONCLUSION: The present study highlights the potential clinical value of the AttoPolyT probe for fast and accurate detection of S. aureus infection in synovial fluids and biofilms on extracted osteosynthesis materials.

2.
J Neurosci ; 41(46): 9633-9649, 2021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34580165

RESUMEN

Epilepsy Aphasia Syndromes (EAS) are a spectrum of childhood epileptic, cognitive, and language disorders of unknown etiology. CNKSR2 is a strong X-linked candidate gene implicated in EAS; however, there have been no studies of genetic models to dissect how its absence may lead to EAS. Here we develop a novel Cnksr2 KO mouse line and show that male mice exhibit increased neural activity and have spontaneous electrographic seizures. Cnksr2 KO mice also display significantly increased anxiety, impaired learning and memory, and a progressive and dramatic loss of ultrasonic vocalizations. We find that Cnksr2 is expressed in cortical, striatal, and cerebellar regions and is localized at both excitatory and inhibitory postsynapses. Proteomics analysis reveals Cnksr2 anchors key binding partners at synapses, and its loss results in significant alterations of the synaptic proteome, including proteins implicated in epilepsy disorders. Our results validate that loss of CNKSR2 leads to EAS and highlights the roles of Cnksr2 in synaptic organization and neuronal network activity.SIGNIFICANCE STATEMENT Epilepsy Aphasia Syndromes (EAS) are at the severe end of a spectrum of cognitive-behavioral symptoms seen in childhood epilepsies, and they remain an inadequately understood disorder. The prognosis of EAS is frequently poor, and patients have life-long language and cognitive disturbances. Here we describe a genetic mouse model of EAS, based on the KO of the EAS risk gene Cnksr2 We show that these mice exhibit electrophysiological and behavioral phenotypes similar to those of patients, providing an important new model for future studies of EAS. We also provide insights into the molecular disturbances downstream of Cnksr2 loss by using in vivo quantitative proteomics tools.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Modelos Animales de Enfermedad , Síndrome de Landau-Kleffner , Proteínas del Tejido Nervioso/deficiencia , Animales , Conducta Animal , Ratones , Ratones Noqueados , Fenotipo , Síndrome
3.
Nature ; 538(7623): 104-108, 2016 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-27680697

RESUMEN

The Rho GTPase proteins Rac1, RhoA and Cdc42 have a central role in regulating the actin cytoskeleton in dendritic spines, thereby exerting control over the structural and functional plasticity of spines and, ultimately, learning and memory. Although previous work has shown that precise spatiotemporal coordination of these GTPases is crucial for some forms of cell morphogenesis, the nature of such coordination during structural spine plasticity is unclear. Here we describe a three-molecule model of structural long-term potentiation (sLTP) of murine dendritic spines, implicating the localized, coincident activation of Rac1, RhoA and Cdc42 as a causal signal of sLTP. This model posits that complete tripartite signal overlap in spines confers sLTP, but that partial overlap primes spines for structural plasticity. By monitoring the spatiotemporal activation patterns of these GTPases during sLTP, we find that such spatiotemporal signal complementation simultaneously explains three integral features of plasticity: the facilitation of plasticity by brain-derived neurotrophic factor (BDNF), the postsynaptic source of which activates Cdc42 and Rac1, but not RhoA; heterosynaptic facilitation of sLTP, which is conveyed by diffusive Rac1 and RhoA activity; and input specificity, which is afforded by spine-restricted Cdc42 activity. Thus, we present a form of biochemical computation in dendrites involving the controlled complementation of three molecules that simultaneously ensures signal specificity and primes the system for plasticity.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Espinas Dendríticas/metabolismo , Potenciación a Largo Plazo , Neuropéptidos/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Activación Enzimática , Femenino , Humanos , Masculino , Ratones , Inhibición Neural , Neuropéptidos/antagonistas & inhibidores , Densidad Postsináptica/metabolismo , Ratas , Transducción de Señal , Análisis Espacio-Temporal , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rhoA
4.
Nature ; 538(7623): 99-103, 2016 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-27680698

RESUMEN

Brain-derived neurotrophic factor (BDNF) and its receptor TrkB are crucial for many forms of neuronal plasticity, including structural long-term potentiation (sLTP), which is a correlate of an animal's learning. However, it is unknown whether BDNF release and TrkB activation occur during sLTP, and if so, when and where. Here, using a fluorescence resonance energy transfer-based sensor for TrkB and two-photon fluorescence lifetime imaging microscopy, we monitor TrkB activity in single dendritic spines of CA1 pyramidal neurons in cultured murine hippocampal slices. In response to sLTP induction, we find fast (onset < 1 min) and sustained (>20 min) activation of TrkB in the stimulated spine that depends on NMDAR (N-methyl-d-aspartate receptor) and CaMKII signalling and on postsynaptically synthesized BDNF. We confirm the presence of postsynaptic BDNF using electron microscopy to localize endogenous BDNF to dendrites and spines of hippocampal CA1 pyramidal neurons. Consistent with these findings, we also show rapid, glutamate-uncaging-evoked, time-locked BDNF release from single dendritic spines using BDNF fused to superecliptic pHluorin. We demonstrate that this postsynaptic BDNF-TrkB signalling pathway is necessary for both structural and functional LTP. Together, these findings reveal a spine-autonomous, autocrine signalling mechanism involving NMDAR-CaMKII-dependent BDNF release from stimulated dendritic spines and subsequent TrkB activation on these same spines that is crucial for structural and functional plasticity.


Asunto(s)
Comunicación Autocrina , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Espinas Dendríticas/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Espinas Dendríticas/ultraestructura , Activación Enzimática , Femenino , Transferencia Resonante de Energía de Fluorescencia , Ácido Glutámico/metabolismo , Proteínas Fluorescentes Verdes , Células HeLa , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/ultraestructura , Humanos , Potenciación a Largo Plazo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Microscopía de Fluorescencia por Excitación Multifotónica , Densidad Postsináptica/metabolismo , Células Piramidales/metabolismo , Células Piramidales/ultraestructura , Ratas , Receptores de N-Metil-D-Aspartato/metabolismo , Técnicas de Cultivo de Tejidos
5.
J Neurosci ; 39(23): 4624-4630, 2019 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-30926745

RESUMEN

Temporal lobe epilepsy (TLE) is a common and commonly devastating form of human epilepsy for which only symptomatic therapy is available. One cause of TLE is an episode of de novo prolonged seizures [status epilepticus (SE)]. Understanding the molecular signaling mechanisms by which SE transforms a brain from normal to epileptic may reveal novel targets for preventive and disease-modifying therapies. SE-induced activation of the BDNF receptor tyrosine kinase, TrkB, is one signaling pathway by which SE induces TLE. Although activation of TrkB signaling promotes development of epilepsy in this context, it also reduces SE-induced neuronal death. This led us to hypothesize that distinct signaling pathways downstream of TrkB mediate the desirable (neuroprotective) and undesirable (epileptogenesis) consequences. We subsequently demonstrated that TrkB-mediated activation of phospholipase Cγ1 is required for epileptogenesis. Here we tested the hypothesis that the TrkB-Shc-Akt signaling pathway mediates the neuroprotective consequences of TrkB activation following SE. We studied measures of molecular signaling and cell death in a model of SE in mice of both sexes, including wild-type and TrkBShc/Shc mutant mice in which a point mutation (Y515F) of TrkB prevents the binding of Shc to activated TrkB kinase. Genetic disruption of TrkB-Shc signaling had no effect on severity of SE yet partially inhibited activation of the prosurvival adaptor protein Akt. Importantly, genetic disruption of TrkB-Shc signaling exacerbated hippocampal neuronal death induced by SE. We conclude that therapies targeting TrkB signaling for preventing epilepsy should spare TrkB-Shc-Akt signaling and thereby preserve the neuroprotective benefits.SIGNIFICANCE STATEMENT Temporal lobe epilepsy (TLE) is a common and devastating form of human epilepsy that lacks preventive therapies. Understanding the molecular signaling mechanisms underlying the development of TLE may identify novel therapeutic targets. BDNF signaling thru TrkB receptor tyrosine kinase is one molecular mechanism promoting TLE. We previously discovered that TrkB-mediated activation of phospholipase Cγ1 promotes epileptogenesis. Here we reveal that TrkB-mediated activation of Akt protects against hippocampal neuronal death in vivo following status epilepticus. These findings strengthen the evidence that desirable and undesirable consequences of status epilepticus-induced TrkB activation are mediated by distinct signaling pathways downstream of this receptor. These results provide a strong rationale for a novel therapeutic strategy selectively targeting individual signaling pathways downstream of TrkB for preventing epilepsy.


Asunto(s)
Proteínas Hedgehog/metabolismo , Hipocampo/metabolismo , Glicoproteínas de Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Proteínas Tirosina Quinasas/fisiología , Células Receptoras Sensoriales/fisiología , Estado Epiléptico/metabolismo , Animales , Sitios de Unión , Factor Neurotrófico Derivado del Encéfalo/farmacología , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Hipocampo/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mecanotransducción Celular , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Mutagénesis Sitio-Dirigida , Factores de Crecimiento Nervioso/farmacología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Fosforilación , Mutación Puntual , Unión Proteica/genética , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Recombinantes/metabolismo , Células Receptoras Sensoriales/efectos de los fármacos , Estado Epiléptico/genética , Tacto/fisiología
6.
Ann Neurol ; 86(6): 939-950, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31525273

RESUMEN

OBJECTIVE: Temporal lobe epilepsy (TLE) is a devastating disease in which seizures persist in 35% of patients despite optimal use of antiseizure drugs. Clinical and preclinical evidence implicates seizures themselves as one factor promoting epilepsy progression. What is the molecular consequence of a seizure that promotes progression? Evidence from preclinical studies led us to hypothesize that activation of tropomyosin kinase B (TrkB)-phospholipase-C-gamma-1 (PLCγ1) signaling induced by a seizure promotes epileptogenesis. METHODS: To examine the effects of inhibiting TrkB signaling on epileptogenesis following an isolated seizure, we implemented a modified kindling model in which we induced a seizure through amygdala stimulation and then used either a chemical-genetic strategy or pharmacologic methods to disrupt signaling for 2 days following the seizure. The severity of a subsequent seizure was assessed by behavioral and electrographic measures. RESULTS: Transient inhibition of TrkB-PLCγ1 signaling initiated after an isolated seizure limited progression of epileptogenesis, evidenced by the reduced severity and duration of subsequent seizures. Unexpectedly, transient inhibition of TrkB-PLCγ1 signaling initiated following a seizure also reverted a subset of animals to an earlier state of epileptogenesis. Remarkably, inhibition of TrkB-PLCγ1 signaling in the absence of a recent seizure did not reduce severity of subsequent seizures. INTERPRETATION: These results suggest a novel strategy for limiting progression or potentially ameliorating severity of TLE whereby transient inhibition of TrkB-PLCγ1 signaling is initiated following a seizure. ANN NEUROL 2019;86:939-950.


Asunto(s)
Excitación Neurológica/fisiología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Quinasas/metabolismo , Convulsiones/tratamiento farmacológico , Convulsiones/enzimología , Transducción de Señal/fisiología , Animales , Electroencefalografía/efectos de los fármacos , Electroencefalografía/métodos , Femenino , Excitación Neurológica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos
8.
J Neurophysiol ; 121(2): 609-619, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30517040

RESUMEN

Hippocampal mossy fiber axons simultaneously activate CA3 pyramidal cells and stratum lucidum interneurons (SLINs), the latter providing feedforward inhibition to control CA3 pyramidal cell excitability. Filopodial extensions of giant boutons of mossy fibers provide excitatory synaptic input to the SLIN. These filopodia undergo extraordinary structural plasticity causally linked to execution of memory tasks, leading us to seek the mechanisms by which activity regulates these synapses. High-frequency stimulation of the mossy fibers induces long-term depression (LTD) of their calcium-permeable AMPA receptor synapses with SLINs; previous work localized the site of induction to be postsynaptic and the site of expression to be presynaptic. Yet, the underlying signaling events and the identity of the retrograde signal are incompletely understood. We used whole cell recordings of SLINs in hippocampal slices from wild-type and mutant mice to explore the mechanisms. Genetic and pharmacologic perturbations revealed a requirement for both the receptor tyrosine kinase TrkB and its agonist, brain-derived neurotrophic factor (BDNF), for induction of LTD. Inclusion of inhibitors of Trk receptor kinase and PLC in the patch pipette prevented LTD. Endocannabinoid receptor antagonists and genetic deletion of the CB1 receptor prevented LTD. We propose a model whereby release of BDNF from mossy fiber filopodia activates TrkB and PLCγ1 signaling postsynaptically within SLINs, triggering synthesis and release of an endocannabinoid that serves as a retrograde signal, culminating in reduced glutamate release. Insights into the signaling pathways by which activity modifies function of these synapses will facilitate an understanding of their contribution to the local circuit and behavioral consequences of hippocampal granule cell activity. NEW & NOTEWORTHY We investigated signaling mechanisms underlying plasticity of the hippocampal mossy fiber filopodial synapse with interneurons in stratum lucidum. High-frequency stimulation of the mossy fibers induces long-term depression of this synapse. Our findings are consistent with a model in which brain-derived neurotrophic factor released from filopodia activates TrkB of a stratum lucidum interneuron; the ensuing activation of PLCγ1 induces synthesis of an endocannabinoid, which provides a retrograde signal leading to reduced release of glutamate presynaptically.


Asunto(s)
Región CA3 Hipocampal/metabolismo , Interneuronas/metabolismo , Depresión Sináptica a Largo Plazo , Fibras Musgosas del Hipocampo/metabolismo , Receptor Cannabinoide CB1/metabolismo , Receptor trkB/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Región CA3 Hipocampal/citología , Región CA3 Hipocampal/fisiología , Endocannabinoides/metabolismo , Femenino , Ácido Glutámico/metabolismo , Interneuronas/fisiología , Masculino , Ratones , Fibras Musgosas del Hipocampo/fisiología , Fosfolipasa C gamma/metabolismo , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/genética , Transducción de Señal
10.
Mol Ther ; 25(6): 1353-1362, 2017 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-28391960

RESUMEN

Rapid and accurate bacterial detection methods are needed for clinical diagnostic, water, and food testing applications. The wide diversity of bacterial nucleases provides a rich source of enzymes that could be exploited as signal amplifying biomarkers to enable rapid, selective detection of bacterial species. With the exception of the use of micrococcal nuclease activity to detect Staphylococcus aureus, rapid methods that detect bacterial pathogens via their nuclease activities have not been developed. Here, we identify endonuclease I as a robust biomarker for E. coli and develop a rapid ultrasensitive assay that detects its activity. Comparison of nuclease activities of wild-type and nuclease-knockout E. coli clones revealed that endonuclease I is the predominant DNase in E. coli lysates. Endonuclease I is detectable by immunoblot and activity assays in uropathogenic E. coli strains. A rapid assay that detects endonuclease I activity in patient urine with an oligonucleotide probe exhibited substantially higher sensitivity for urinary tract infections than that reported for rapid urinalysis methods. The 3 hr turnaround time is much shorter than that of culture-based methods, thereby providing a means for expedited administration of appropriate antimicrobial therapy. We suggest this approach could address various unmet needs for rapid detection of E. coli.


Asunto(s)
Bacterias/enzimología , Endodesoxirribonucleasas/metabolismo , Infecciones Urinarias/diagnóstico , Infecciones Urinarias/microbiología , Biomarcadores , Desoxirribonucleasa I/metabolismo , Activación Enzimática , Pruebas de Enzimas/métodos , Escherichia coli/enzimología , Humanos , Nucleasa Microcócica/metabolismo , Oportunidad Relativa , Curva ROC , Reproducibilidad de los Resultados , Staphylococcus aureus/enzimología , Infecciones Urinarias/orina
11.
J Neurochem ; 140(4): 629-644, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27973753

RESUMEN

Vagal Nerve Stimulation (VNS) Therapy® is a United States Food and Drug Administration approved neurotherapeutic for medically refractory partial epilepsy and treatment-resistant depression. The molecular mechanisms underlying its beneficial effects are unclear. We hypothesized that one mechanism involves neuronal activity-dependent modifications of central nervous system excitatory synapses. To begin to test this hypothesis, we asked whether VNS modifies the activity of neurons in amygdala and hippocampus. Neuronal recordings from adult, freely moving rats revealed that activity in both amygdala and hippocampus was modified by VNS immediately after its application, and changes were detected following 1 week of stimulation. To investigate whether VNS modifies the proteome of excitatory synapses, we established a label-free, quantitative liquid chromatography-tandem mass spectrometry workflow that enables global analysis of the constituents of the postsynaptic density (PSD) proteome. PSD proteins were biochemically purified from amygdala/piriform cortex of VNS- or dummy-treated rats following 1-week stimulation, and individual PSD protein levels were quantified by liquid chromatography-tandem mass spectrometry analysis. We identified 1899 unique peptides corresponding to 425 proteins in PSD fractions, of which expression levels of 22 proteins were differentially regulated by VNS with changes greater than 150%. Changes in a subset of these proteins, including significantly increased expression of neurexin-1α, cadherin 13 and voltage-dependent calcium channel α2δ1, the primary target of the antiepileptic drug gabapentin, and decreased expression of voltage-dependent calcium channel γ3, were confirmed by western blot analysis of PSD samples. These results demonstrate that VNS modulates excitatory synapses through regulating a subset of the PSD proteome. Our study reveals molecular targets of VNS and point to possible mechanisms underlying its beneficial effects, including activity-dependent formation of excitatory synapses.


Asunto(s)
Amígdala del Cerebelo/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Corteza Piriforme/fisiología , Proteoma/metabolismo , Sinapsis/metabolismo , Estimulación del Nervio Vago/métodos , Animales , Masculino , Neuronas/fisiología , Proteoma/genética , Ratas , Ratas Sprague-Dawley , Sinapsis/genética
12.
Bioconjug Chem ; 28(1): 183-193, 2017 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-28095683

RESUMEN

Current water quality monitoring methods rely on growth-based measurements to detect fecal indicator bacteria, such as Escherichia coli and enterococci, and Staphylococcus aureus (S. aureus). These growth-based measurements, however, can take days to complete. This is a significant limitation in the evaluation of contaminated food and water sources. Various methods for selective in vitro detection of S. aureus have also been reported; however, these strategies, such as ELISA, agar-diffusion, PCR, or liquid chromatography-tandem mass spectrometry, all require overnight culturing or sophisticated instrumentation. There is a pressing need for a portable, simple diagnostic for S. aureus. Here, we demonstrate that oligonucleotide-functionalized gold nanoparticles (Oligo-AuNPs) can be designed to rapidly and selectively detect S. aureus with a colorimetric readout. We have functionalized a chemically modified 11-mer sequence onto AuNPs and have found that aggregation occurs in the presence of S. aureus supernantants. The particles can be stored as a lyophilized powder and reconstituted at time of use, and this has been tested in biologically relevant samples such as creek and ocean water. This approach requires minimal sample preparation and requires no extraneous instrumentation, leading to a rapid and simple diagnostic read-out that could be used in field tests to monitor food and water sources.


Asunto(s)
Colorimetría/métodos , Staphylococcus/aislamiento & purificación , Colorantes Fluorescentes , Liofilización , Oro/química , Concentración de Iones de Hidrógeno , Nanopartículas del Metal/química , Pruebas de Sensibilidad Microbiana , Soluciones , Staphylococcus/clasificación , Staphylococcus/efectos de los fármacos
13.
J Neurosci ; 35(23): 8866-81, 2015 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-26063919

RESUMEN

We identified a family in which a translocation between chromosomes X and 14 was associated with cognitive impairment and a complex genetic disorder termed "Genetic Epilepsy and Febrile Seizures Plus" (GEFS(+)). We demonstrate that the breakpoint on the X chromosome disrupted a gene that encodes an auxiliary protein of voltage-gated Na(+) channels, fibroblast growth factor 13 (Fgf13). Female mice in which one Fgf13 allele was deleted exhibited hyperthermia-induced seizures and epilepsy. Anatomic studies revealed expression of Fgf13 mRNA in both excitatory and inhibitory neurons of hippocampus. Electrophysiological recordings revealed decreased inhibitory and increased excitatory synaptic inputs in hippocampal neurons of Fgf13 mutants. We speculate that reduced expression of Fgf13 impairs excitability of inhibitory interneurons, resulting in enhanced excitability within local circuits of hippocampus and the clinical phenotype of epilepsy. These findings reveal a novel cause of this syndrome and underscore the powerful role of FGF13 in control of neuronal excitability.


Asunto(s)
Epilepsia , Factores de Crecimiento de Fibroblastos/genética , Mutación/genética , Sinapsis/genética , Potenciales Sinápticos/genética , Factores de Edad , Animales , Animales Recién Nacidos , Línea Celular , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/genética , Modelos Animales de Enfermedad , Embrión de Mamíferos , Epilepsia/genética , Epilepsia/patología , Epilepsia/fisiopatología , Salud de la Familia , Femenino , Fiebre/complicaciones , Hipocampo/patología , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Canal de Sodio Activado por Voltaje NAV1.1/genética , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Convulsiones Febriles/etiología , Convulsiones Febriles/genética , Factores Sexuales , Translocación Genética/genética , Cromosoma X/genética , Adulto Joven
14.
Am J Hum Genet ; 91(2): 293-302, 2012 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22863189

RESUMEN

Idiopathic generalized epilepsy (IGE) is a complex disease with high heritability, but little is known about its genetic architecture. Rare copy-number variants have been found to explain nearly 3% of individuals with IGE; however, it remains unclear whether variants with moderate effect size and frequencies below what are reliably detected with genome-wide association studies contribute significantly to disease risk. In this study, we compare the exome sequences of 118 individuals with IGE and 242 controls of European ancestry by using next-generation sequencing. The exome-sequenced epilepsy cases include study subjects with two forms of IGE, including juvenile myoclonic epilepsy (n = 93) and absence epilepsy (n = 25). However, our discovery strategy did not assume common genetic control between the subtypes of IGE considered. In the sequence data, as expected, no variants were significantly associated with the IGE phenotype or more specific IGE diagnoses. We then selected 3,897 candidate epilepsy-susceptibility variants from the sequence data and genotyped them in a larger set of 878 individuals with IGE and 1,830 controls. Again, no variant achieved statistical significance. However, 1,935 variants were observed exclusively in cases either as heterozygous or homozygous genotypes. It is likely that this set of variants includes real risk factors. The lack of significant association evidence of single variants with disease in this two-stage approach emphasizes the high genetic heterogeneity of epilepsy disorders, suggests that the impact of any individual single-nucleotide variant in this disease is small, and indicates that gene-based approaches might be more successful for future sequencing studies of epilepsy predisposition.


Asunto(s)
Epilepsia Generalizada/genética , Exoma/genética , Predisposición Genética a la Enfermedad/genética , Secuencia de Bases , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Datos de Secuencia Molecular , Alineación de Secuencia , Análisis de Secuencia de ADN , Población Blanca/genética
15.
J Biol Chem ; 288(19): 13258-68, 2013 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-23536187

RESUMEN

BACKGROUND: The necessity for, or redundancy of, distinctive KChIP proteins is not known. RESULTS: Deletion of KChIP2 leads to increased susceptibility to epilepsy and to a reduction in IA and increased excitability in pyramidal hippocampal neurons. CONCLUSION: KChIP2 is essential for homeostasis in hippocampal neurons. SIGNIFICANCE: Mutations in K(A) channel auxiliary subunits may be loci for epilepsy. The somatodendritic IA (A-type) K(+) current underlies neuronal excitability, and loss of IA has been associated with the development of epilepsy. Whether any one of the four auxiliary potassium channel interacting proteins (KChIPs), KChIP1-KChIP4, in specific neuronal populations is critical for IA is not known. Here we show that KChIP2, which is abundantly expressed in hippocampal pyramidal cells, is essential for IA regulation in hippocampal neurons and that deletion of Kchip2 affects susceptibility to limbic seizures. The specific effects of Kchip2 deletion on IA recorded from isolated hippocampal pyramidal neurons were a reduction in amplitude and shift in the V½ for steady-state inactivation to hyperpolarized potentials when compared with WT neurons. Consistent with the relative loss of IA, hippocampal neurons from Kchip2(-/-) mice showed increased excitability. WT cultured neurons fired only occasional single action potentials, but the average spontaneous firing rate (spikes/s) was almost 10-fold greater in Kchip2(-/-) neurons. In slice preparations, spontaneous firing was detected in CA1 pyramidal neurons from Kchip2(-/-) mice but not from WT. Additionally, when seizures were induced by kindling, the number of stimulations required to evoke an initial class 4 or 5 seizure was decreased, and the average duration of electrographic seizures was longer in Kchip2(-/-) mice compared with WT controls. Together, these data demonstrate that the KChIP2 is essential for physiologic IA modulation and homeostatic stability and that there is a lack of functional redundancy among the different KChIPs in hippocampal neurons.


Asunto(s)
Potenciales de Acción , Homeostasis , Proteínas de Interacción con los Canales Kv/fisiología , Subunidades de Proteína/fisiología , Amígdala del Cerebelo/patología , Amígdala del Cerebelo/fisiopatología , Animales , Región CA1 Hipocampal/patología , Región CA1 Hipocampal/fisiopatología , Células Cultivadas , Estimulación Eléctrica , Potenciales Postsinápticos Excitadores , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores , Excitación Neurológica , Ratones , Ratones de la Cepa 129 , Ratones Transgénicos , Células Piramidales/metabolismo , Células Piramidales/fisiología , Convulsiones
16.
Epilepsia ; 55(8): 1264-73, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24903749

RESUMEN

OBJECTIVE: The principal molecular targets of conventional antiseizure drugs consist of ligand-gated and voltage-gated ion channels and proteins subserving synaptic function. Inhibition of the receptor tyrosine kinase TrkB limits epileptogenesis, but its effect on individual seizures is unknown. We sought to determine whether inhibition of TrkB kinase exerts an antiseizure effect. METHODS: We utilized the kindling model in combination with an inducible conditional knockout of the TrkB gene (Act-CreER TrkB(flox/flox) mice treated with tamoxifen), and also with a chemical-genetic approach in which mice carry a TrkB kinase with a phenylalanine to alanine substitution of residue 616 (TrkB(F) (616A) ), which allows inhibition of the kinase by a blood-brain barrier permeable small molecule, 1'-naphthylmethyl-4-amino-1-tert-butyl-3-(p-methylphenyl)pyrazolo[3,4-d]pyrimidine (1NMPP1). RESULTS: Following induction of kindling, reduction of TrkB protein levels in Act-CreER TrkB(flox/flox) mice treated with tamoxifen was associated with reduced severity of behavioral seizures evoked by stimulation. Treatment with 1NMPP1 for 2 weeks following induction of kindling reversibly elevated both focal electrographic and generalized seizure thresholds in TrkB(F) (616A) , but not wild-type (WT), mice. In contrast to kindled animals, treatment of naive TrkB(F) (616A) mice for 2 weeks had no detectable effect on electrographic seizure threshold (EST). SIGNIFICANCE: This study provides proof of concept of a novel molecular target for antiseizure drugs, namely the receptor tyrosine kinase TrkB.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Quinasas/metabolismo , Convulsiones/tratamiento farmacológico , Convulsiones/enzimología , Animales , Glicoproteínas de Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Tirosina Quinasas , Receptor trkB , Resultado del Tratamiento
17.
Epilepsia ; 55(3): 456-63, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24502564

RESUMEN

OBJECTIVE: Elucidating molecular mechanisms underlying limbic epileptogenesis may reveal novel targets for preventive therapy. Studies of TrkB mutant mice led us to hypothesize that signaling through a specific phospholipase (PLC), PLCγ1, promoted development of kindling. METHODS: To test this hypothesis, we examined the development of kindling in PLCγ1 heterozygous mice. We also examined the cellular and subcellular location of PLCγ1 in adult wild-type mice. RESULTS: The development of kindling was impaired in PLCγ1 heterozygous mice compared to wild-type controls. PLCγ1 immunoreactivity was localized to the soma and dendrites of both excitatory and inhibitory neurons in the hippocampus of adult mice. SIGNIFICANCE: This study implicates PLCγ1 signaling as the dominant pathway by which TrkB activation promotes limbic epileptogenesis. Its cellular localization places PLCγ1 in a position to modify the efficacy of both excitatory and inhibitory synaptic transmission. These findings advance PLCγ1 as a novel target for therapies aimed at preventing temporal lobe epilepsy induced by status epilepticus.


Asunto(s)
Hipocampo/química , Hipocampo/enzimología , Excitación Neurológica/genética , Fosfolipasa C gamma/análisis , Fosfolipasa C gamma/genética , Animales , Heterocigoto , Hipocampo/patología , Excitación Neurológica/patología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Convulsiones/genética , Convulsiones/patología , Transducción de Señal/fisiología
18.
Nucleic Acids Res ; 40(13): 6319-37, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22467215

RESUMEN

Human epidermal growth factor receptor 2 (HER2) expression in breast cancer is associated with an aggressive phenotype and poor prognosis, making it an appealing therapeutic target. Trastuzumab, an HER2 antibody-based inhibitor, is currently the leading targeted treatment for HER2(+)-breast cancers. Unfortunately, many patients inevitably develop resistance to the therapy, highlighting the need for alternative targeted therapeutic options. In this study, we used a novel, cell-based selection approach for isolating 'cell-type specific', 'cell-internalizing RNA ligands (aptamers)' capable of delivering therapeutic small interfering RNAs (siRNAs) to HER2-expressing breast cancer cells. RNA aptamers with the greatest specificity and internalization potential were covalently linked to siRNAs targeting the anti-apoptotic gene, Bcl-2. We demonstrate that, when applied to cells, the HER2 aptamer-Bcl-2 siRNA conjugates selectively internalize into HER2(+)-cells and silence Bcl-2 gene expression. Importantly, Bcl-2 silencing sensitizes these cells to chemotherapy (cisplatin) suggesting a potential new therapeutic approach for treating breast cancers with HER2(+)-status. In summary, we describe a novel cell-based selection methodology that enables the identification of cell-internalizing RNA aptamers for targeting therapeutic siRNAs to HER2-expressing breast cancer cells. The future refinement of this technology may promote the widespread use of RNA-based reagents for targeted therapeutic applications.


Asunto(s)
Aptámeros de Nucleótidos/química , Neoplasias Mamarias Experimentales/genética , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , Receptor ErbB-2/metabolismo , Animales , Antineoplásicos/farmacología , Aptámeros de Nucleótidos/análisis , Línea Celular Tumoral , Cisplatino/farmacología , Femenino , Humanos , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Técnica SELEX de Producción de Aptámeros
19.
Adv Exp Med Biol ; 813: 243-51, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25012381

RESUMEN

Treatment of the epilepsies have benefitted immensely from study of animal models, most notably in the development of diverse anti-seizure medications in current clinical use. However, available drugs provide only symptomatic relief from seizures and are often ineffective. As a result, a critical need remains for developing improved symptomatic or disease-modifying therapies - or ideally, preventive therapies. Animal models will undoubtedly play a central role in such efforts. To ensure success moving forward, a critical question arises, namely "How does one make laboratory models relevant to our clinical understanding and treatment?" Our answer to this question: It all begins with a detailed understanding of the clinical phenotype one seeks to model. To make our case, we point to two examples - Fragile X syndrome and status epilepticus-induced mesial temporal lobe epilepsy - and examine how development of animal models for these distinct syndromes is based upon observations by astute clinicians and systematic study of the disorder. We conclude that the continuous and effective interaction of skilled clinicians and bench scientists is critical to the optimal design and study of animal models to facilitate insight into the nature of human disorders and enhance likelihood of improved therapies.


Asunto(s)
Epilepsia/fisiopatología , Modelos Animales , Animales , Epilepsia/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Humanos , Ratones , Ratones Noqueados
20.
J Neurosci ; 32(44): 15521-32, 2012 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-23115189

RESUMEN

Reactive oxygen species (ROS) have diverse biological consequences in the mammalian CNS, but the molecular targets mediating these pleiotropic effects are incompletely understood. Like ROS, the neurotrophin receptor, TrkB receptor tyrosine kinase, has diverse effects in the developing and mature mammalian brain. Our discovery that zinc can transactivate TrkB, together with the finding that ROS can trigger zinc release from cytosolic zinc binding proteins, led us to hypothesize that ROS can transactivate TrkB in CNS neurons by a zinc-dependent mechanism. We found that both exogenous H(2)O(2) and endogenous ROS activate TrkB signaling by a Src family kinase-dependent but brain-derived neurotrophic factor-independent mechanism in cultured rat cortical neurons. Exogenous H(2)O(2) enhances cytosolic zinc content in a metallothionein-3 (MT-3)-requiring manner. Both exogenous H(2)O(2) and endogenous ROS mediated transactivation of TrkB requires intracellular zinc and MT-3. The ROS-triggered transactivation of TrkB exerts neuroprotective effects, because inhibition of TrkB kinase activity or uncoupling Shc signaling from TrkB exacerbates neuronal cell death induced by H(2)O(2). Thus, we propose a molecular signaling event whereby ROS induce release of zinc from cytosolic MT-3, the increased cytosolic zinc transactivates TrkB, and the enhanced Shc signaling downstream from TrkB promotes prosurvival effects. We suggest that such neuroprotective effects mediated by ROS are operative in diverse acute and chronic neurological disorders.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/fisiología , Fármacos Neuroprotectores , Especies Reactivas de Oxígeno/farmacología , Receptor trkB/fisiología , Animales , Western Blotting , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células HEK293 , Hipocampo/citología , Hipocampo/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/farmacología , Inmunoprecipitación , Indicadores y Reactivos , Metalotioneína 3 , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/fisiología , Neuronas/efectos de los fármacos , Receptor trkB/agonistas , Activación Transcripcional/efectos de los fármacos , Transfección , Zinc/fisiología , Familia-src Quinasas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA