Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Pharmacol ; 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38821630

RESUMEN

Bipolar disorder impacts millions of patients in the United States but the mechanistic understanding of its pathophysiology and therapeutics is incomplete. Atypical antipsychotic serotonin2A (5-HT2A) receptor antagonists, such as quetiapine and olanzapine, and mood-stabilizing voltage-gated sodium channel (VGSC) blockers, such as lamotrigine, carbamazepine, and valproate, show therapeutic synergy and are often prescribed in combination for the treatment of bipolar disorder. Combination therapy is a complex task for clinicians and patients, often resulting in unexpected difficulties with dosing, drug tolerances, and decreased patient compliance. Thus, an unmet need for bipolar disorder treatment is to develop a therapeutic agent that targets both 5-HT2A receptors and VGSCs. Towards this goal, we developed a novel small molecule that simultaneously antagonizes 5-HT2A receptors and blocks sodium current. The new compound, N-(4-bromo-2,5-dimethoxyphenethyl)-6-(4-phenylbutoxy)hexan-1-amine (XOB) antagonizes 5-HT-stimulated, Gq-mediated, calcium flux at 5-HT2A receptors at low micromolar concentrations while displaying negligible affinity and activity at 5-HT1A, 5-HT2B, and 5-HT2C receptors. At similar concentrations, XOB administration inhibits sodium current in heterologous cells and results in reduced action potential (AP) firing and VGSC-related AP properties in mouse prefrontal cortex layer V pyramidal neurons. Thus, XOB represents a new, proof-of-principle tool that can be used for future preclinical investigations and therapeutic development. This polypharmacology approach of developing a single molecule to act upon two targets, which are currently independently targeted by combination therapies, may lead to safer alternatives for the treatment of psychiatric disorders that are increasingly being found to benefit from the simultaneous targeting of multiple receptors. Significance Statement We synthesized a novel small molecule (XOB) that simultaneously antagonizes two key therapeutic targets of bipolar disorder, 5-HT2A receptors and voltage-gated sodium channels (VGSCs), in heterologous cells, and inhibits the intrinsic excitability of mouse prefrontal cortex layer V pyramidal neurons in brain slices. XOB represents a valuable new proof-of-principle tool for future preclinical investigations and provides a novel molecular approach to the pharmacological treatment of complex neuropsychiatric disease, which often requires a combination of therapeutics for sufficient patient benefit.

2.
Sci Adv ; 9(23): eadf9524, 2023 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-37285436

RESUMEN

Perception, thoughts, and actions are encoded by the coordinated activity of large neuronal populations spread over large areas. However, existing electrophysiological devices are limited by their scalability in capturing this cortex-wide activity. Here, we developed an electrode connector based on an ultra-conformable thin-film electrode array that self-assembles onto silicon microelectrode arrays enabling multithousand channel counts at a millimeter scale. The interconnects are formed using microfabricated electrode pads suspended by thin support arms, termed Flex2Chip. Capillary-assisted assembly drives the pads to deform toward the chip surface, and van der Waals forces maintain this deformation, establishing Ohmic contact. Flex2Chip arrays successfully measured extracellular action potentials ex vivo and resolved micrometer scale seizure propagation trajectories in epileptic mice. We find that seizure dynamics in absence epilepsy in the Scn8a+/- model do not have constant propagation trajectories.


Asunto(s)
Corteza Cerebral , Epilepsia , Ratones , Animales , Microelectrodos , Fenómenos Electrofisiológicos , Convulsiones , Canal de Sodio Activado por Voltaje NAV1.6
3.
Sci Rep ; 13(1): 8887, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37264112

RESUMEN

Voltage gated sodium channels (VGSCs) are required for action potential initiation and propagation in mammalian neurons. As with other ion channel families, VGSC density varies between neurons. Importantly, sodium current (INa) density variability is reduced in pyramidal neurons of Scn1b null mice. Scn1b encodes the VGSC ß1/ ß1B subunits, which regulate channel expression, trafficking, and voltage dependent properties. Here, we investigate how variable INa density in cortical layer 6 and subicular pyramidal neurons affects spike patterning and network synchronization. Constitutive or inducible Scn1b deletion enhances spike timing correlations between pyramidal neurons in response to fluctuating stimuli and impairs spike-triggered average current pattern diversity while preserving spike reliability. Inhibiting INa with a low concentration of tetrodotoxin similarly alters patterning without impairing reliability, with modest effects on firing rate. Computational modeling shows that broad INa density ranges confer a similarly broad spectrum of spike patterning in response to fluctuating synaptic conductances. Network coupling of neurons with high INa density variability displaces the coupling requirements for synchronization and broadens the dynamic range of activity when varying synaptic strength and network topology. Our results show that INa heterogeneity between neurons potently regulates spike pattern diversity and network synchronization, expanding VGSC roles in the nervous system.


Asunto(s)
Neuronas , Sodio , Ratones , Animales , Sodio/metabolismo , Reproducibilidad de los Resultados , Tetrodotoxina/farmacología , Neuronas/metabolismo , Potenciales de Acción , Ratones Noqueados , Mamíferos/metabolismo , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/genética , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/metabolismo
4.
PLoS Biol ; 21(4): e3002078, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37079499

RESUMEN

Down syndrome (DS) is caused by the trisomy of human chromosome 21 (HSA21). A major challenge in DS research is to identify the HSA21 genes that cause specific symptoms. Down syndrome cell adhesion molecule (DSCAM) is encoded by a HSA21 gene. Previous studies have shown that the protein level of the Drosophila homolog of DSCAM determines the size of presynaptic terminals. However, whether the triplication of DSCAM contributes to presynaptic development in DS remains unknown. Here, we show that DSCAM levels regulate GABAergic synapses formed on neocortical pyramidal neurons (PyNs). In the Ts65Dn mouse model for DS, where DSCAM is overexpressed due to DSCAM triplication, GABAergic innervation of PyNs by basket and chandelier interneurons is increased. Genetic normalization of DSCAM expression rescues the excessive GABAergic innervations and the increased inhibition of PyNs. Conversely, loss of DSCAM impairs GABAergic synapse development and function. These findings demonstrate excessive GABAergic innervation and synaptic transmission in the neocortex of DS mouse models and identify DSCAM overexpression as the cause. They also implicate dysregulated DSCAM levels as a potential pathogenic driver in related neurological disorders.


Asunto(s)
Síndrome de Down , Neocórtex , Animales , Humanos , Ratones , Modelos Animales de Enfermedad , Síndrome de Down/genética , Síndrome de Down/metabolismo , Síndrome de Down/patología , Drosophila , Interneuronas/metabolismo , Terminales Presinápticos/metabolismo , Sinapsis/metabolismo
5.
Ann Clin Transl Neurol ; 7(11): 2137-2149, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32979291

RESUMEN

OBJECTIVE: Human variants in voltage-gated sodium channel (VGSC) α and ß subunit genes are linked to developmental and epileptic encephalopathies (DEEs). Inherited, biallelic, loss-of-function variants in SCN1B, encoding the ß1/ß1B subunits, are linked to early infantile DEE (EIEE52). De novo, monoallelic variants in SCN1A (Nav1.1), SCN2A (Nav1.2), SCN3A (Nav1.3), and SCN8A (Nav1.6) are also linked to DEEs. While these VGSC-linked DEEs have similar presentations, they have diverse mechanisms of altered neuronal excitability. Mouse models have suggested that Scn2a-, Scn3a-, and Scn8a-linked DEE variants are, in general, gain of function, resulting in increased persistent or resurgent sodium current (INa ) and pyramidal neuron hyperexcitability. In contrast, Scn1a-linked DEE variants, in general, are loss-of-function, resulting in decreased INa and hypoexcitability of fast-spiking interneurons. VGSC ß1 subunits associate with Nav1.1, Nav1.2, Nav1.3, and Nav1.6 and are expressed throughout the brain, raising the possibility that insults to both pyramidal and interneuron excitability may drive EIEE52 pathophysiology. METHODS: We investigated excitability defects in pyramidal and parvalbumin-positive (PV +) interneurons in the Scn1b-/- model of EIEE52. We also used Scn1bFL/FL mice to delete Scn1b in specific neuronal populations. RESULTS: Scn1b-/- cortical PV + interneurons were hypoexcitable, with reduced INa density. Scn1b-/- cortical pyramidal neurons had population-specific changes in excitability and impaired INa density. Scn1b deletion in PV + neurons resulted in 100% lethality, whereas deletion in Emx1 + or Camk2a + neurons did not affect survival. INTERPRETATION: This work suggests that SCN1B-linked DEE variants impact both excitatory and inhibitory neurons, leading to the increased severity of EIEE52 relative to other DEEs.


Asunto(s)
Corteza Cerebral/fisiopatología , Interneuronas/fisiología , Células Piramidales/fisiología , Espasmos Infantiles/genética , Espasmos Infantiles/fisiopatología , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/fisiología , Animales , Recuento de Células , Modelos Animales de Enfermedad , Humanos , Recién Nacido , Interneuronas/citología , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Parvalbúminas/metabolismo , Células Piramidales/citología , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/genética
6.
Neurosci Lett ; 724: 134853, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32114117

RESUMEN

Voltage-gated sodium channels (VGSCs) are responsible for the initiation and propagation of action potentials in neurons. The human genome includes ten human VGSC α-subunit genes, SCN(X)A, encoding Nav1.1-1.9 plus Nax. To understand the unique role that each VGSC plays in normal and pathophysiological function in neural networks, compounds with high affinity and selectivity for specific VGSC subtypes are required. Toward that goal, a structural analog of the VGSC pore blocker tetrodotoxin, 4,9-anhydrotetrodotoxin (4,9-ah-TTX), has been reported to be more selective in blocking Na+ current mediated by Nav1.6 than other TTX-sensitive VGSCs, including Nav1.2, Nav1.3, Nav1.4, and Nav1.7. While SCN1A, encoding Nav1.1, has been implicated in several neurological diseases, the effects of 4,9-ah-TTX on Nav1.1-mediated Na+ current have not been tested. Here, we compared the binding of 4,9-ah-TTX for human and mouse brain preparations, and the effects of 4,9-ah-TTX on human Nav1.1-, Nav1.3- and Nav1.6-mediated Na+ currents using the whole-cell patch clamp technique in heterologous cells. We show that, while 4,9-ah-TTX administration results in significant blockade of Nav1.6-mediated Na+ current in the nanomolar range, it also has significant effects on Nav1.1-mediated Na+ current. Thus, 4,9-ah-TTX is not a useful tool in identifying Nav1.6-specific effects in human brain networks.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.1/fisiología , Canal de Sodio Activado por Voltaje NAV1.6/fisiología , Tetrodotoxina/análogos & derivados , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Animales , Relación Dosis-Respuesta a Droga , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Transgénicos , Especificidad de la Especie , Tetrodotoxina/farmacología
7.
Pharmacol Rev ; 71(4): 450-466, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31471460

RESUMEN

Despite continuous clinical use for more than 170 years, the mechanism of general anesthetics has not been completely characterized. In this review, we focus on the role of voltage-gated sodium channels in the sedative-hypnotic actions of halogenated ethers, describing the history of anesthetic mechanisms research, the basic neurobiology and pharmacology of voltage-gated sodium channels, and the evidence for a mechanistic interaction between halogenated ethers and sodium channels in the induction of unconsciousness. We conclude with a more integrative perspective of how voltage-gated sodium channels might provide a critical link between molecular actions of the halogenated ethers and the more distributed network-level effects associated with the anesthetized state across species.


Asunto(s)
Éteres/farmacología , Inconsciencia/inducido químicamente , Inconsciencia/metabolismo , Canales de Sodio Activados por Voltaje/metabolismo , Animales , Éteres/química , Humanos , Hidrocarburos Halogenados/química , Hidrocarburos Halogenados/farmacología , Hipnóticos y Sedantes/química , Hipnóticos y Sedantes/farmacología
8.
Epilepsy Curr ; 19(4): 266-268, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31257984

RESUMEN

Not All SCN1A Epileptic Encephalopathies Are Dravet Syndrome: Early Profound Thr226Met Phenotype Sadleir LG, Mountier EI, Gill D, et al. Neurology. 2017;89(10):1035-1042. OBJECTIVE: To define a distinct SCN1A developmental and epileptic encephalopathy with early onset, profound impairment, and movement disorder. METHODS: A case series of 9 children were identified with a profound developmental and epileptic encephalopathy and SCN1A mutation. RESULTS: We identified 9 children 3 to 12 years of age; 7 were male. Seizure onset was at 6 to 12 weeks with hemiclonic seizures, bilateral tonic-clonic seizures, or spasms. All children had profound developmental impairment and were nonverbal and nonambulatory, and 7 of 9 required a gastrostomy. A hyperkinetic movement disorder occurred in all and was characterized by dystonia and choreoathetosis with prominent oral dyskinesia and onset from 2 to 20 months of age. Eight had a recurrent missense SCN1A mutation, p.Thr226Met. The remaining child had the missense mutation p.Pro1345Ser. The mutation arose de novo in 8 of 9; for the remaining case, the mother was negative and the father was unavailable. CONCLUSIONS: Here, we present a phenotype-genotype correlation for SCN1A. We describe a distinct SCN1A phenotype, early infantile SCN1A encephalopathy, which is readily distinguishable from the well-recognized entities of Dravet syndrome and genetic epilepsy with febrile seizures plus. This disorder has an earlier age at onset, profound developmental impairment, and a distinctive hyperkinetic movement disorder, setting it apart from Dravet syndrome. Remarkably, 8 of 9 children had the recurrent missense mutation p.Thr226Met. SCN1A Gain of Function in Early Infantile Encephalopathy Berecki G, Bryson A, Terhag J, et al. Ann Neurol. 2019; 85:514-525. OBJECTIVE: To elucidate the biophysical basis underlying the distinct and severe clinical presentation in patients with the recurrent missense SCN1A variant, p.Thr226Met. Patients with this variant show a well-defined genotype-phenotype correlation and present with developmental and early infantile epileptic encephalopathy that is far more severe than typical SCN1A Dravet syndrome. METHODS: Whole cell patch clamp and dynamic action potential clamp were used to study T226M Nav 1.1 channels expressed in mammalian cells. Computational modeling was used to explore the neuronal scale mechanisms that account for altered action potential firing. RESULTS: T226M channels exhibited hyperpolarizing shifts of the activation and inactivation curves and enhanced fast inactivation. Dynamic action potential clamp hybrid simulation showed that model neurons containing T226M conductance displayed a left shift in rheobase relative to control. At current stimulation levels that produced repetitive action potential firing in control model neurons, depolarization block and cessation of action potential firing occurred in T226M model neurons. Fully computationally simulated neuron models recapitulated the findings from dynamic action potential clamp and showed that heterozygous T226M models were also more susceptible to depolarization block. INTERPRETATION: From a biophysical perspective, the T226M mutation produces gain of function. Somewhat paradoxically, our data suggest that this gain of function would cause interneurons to more readily develop depolarization block. This "functional dominant negative" interaction would produce a more profound disinhibition than seen with haploinsufficiency that is typical of Dravet syndrome and could readily explain the more severe phenotype of patients with T226M mutation.

9.
Ann Clin Transl Neurol ; 6(6): 1121-1126, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31211177

RESUMEN

Pathogenic loss-of-function variants in SCN1B are linked to Dravet syndrome (DS). Previous work suggested that neuronal pathfinding defects underlie epileptogenesis and SUDEP in the Scn1b null mouse model of DS. We tested this hypothesis by inducing Scn1b deletion in adult mice that had developed normally. Epilepsy and SUDEP, which occur by postnatal day 21 in Scn1b null animals, were observed within 20 days of induced Scn1b deletion in adult mice, suggesting that epileptogenesis in SCN1B-DS does not result from defective brain development. Thus, the developmental brain defects observed previously in Scn1b null mice may model other co-morbidities of DS.


Asunto(s)
Convulsiones/genética , Convulsiones/fisiopatología , Muerte Súbita e Inesperada en la Epilepsia , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/deficiencia , Animales , Encéfalo/metabolismo , Epilepsia/genética , Epilepsia/fisiopatología , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
10.
Sci Rep ; 9(1): 6210, 2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30996233

RESUMEN

Dravet syndrome (DS) is a catastrophic developmental and epileptic encephalopathy characterized by severe, pharmacoresistant seizures and the highest risk of Sudden Unexpected Death in Epilepsy (SUDEP) of all epilepsy syndromes. Here, we investigated the time course of maturation of neuronal GABAergic signaling in the Scn1b-/- and Scn1a+/- mouse models of DS. We found that GABAergic signaling remains immature in both DS models, with a depolarized reversal potential for GABAA-evoked currents compared to wildtype in the third postnatal week. Treatment of Scn1b-/- mice with bumetanide resulted in a delay in SUDEP onset compared to controls in a subset of mice, without prevention of seizure activity or amelioration of failure to thrive. We propose that delayed maturation of GABAergic signaling may contribute to epileptogenesis in SCN1B- and SCN1A-linked DS. Thus, targeting the polarity of GABAergic signaling in brain may be an effective therapeutic strategy to reduce SUDEP risk in DS.


Asunto(s)
Epilepsias Mioclónicas/etiología , Canal de Sodio Activado por Voltaje NAV1.1/deficiencia , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/deficiencia , Ácido gamma-Aminobutírico/metabolismo , Animales , Bumetanida/uso terapéutico , Muerte Súbita , Modelos Animales de Enfermedad , Epilepsias Mioclónicas/tratamiento farmacológico , Epilepsias Mioclónicas/metabolismo , Epilepsias Mioclónicas/mortalidad , Epilepsia , Técnicas de Silenciamiento del Gen , Ratones , Convulsiones , Factores de Tiempo
11.
Neuropharmacology ; 132: 43-57, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28927993

RESUMEN

Voltage gated sodium channels (VGSCs) were first identified in terms of their role in the upstroke of the action potential. The underlying proteins were later identified as saxitoxin and scorpion toxin receptors consisting of α and ß subunits. We now know that VGSCs are heterotrimeric complexes consisting of a single pore forming α subunit joined by two ß subunits; a noncovalently linked ß1 or ß3 and a covalently linked ß2 or ß4 subunit. VGSC α subunits contain all the machinery necessary for channel cell surface expression, ion conduction, voltage sensing, gating, and inactivation, in one central, polytopic, transmembrane protein. VGSC ß subunits are more than simple accessories to α subunits. In the more than two decades since the original cloning of ß1, our knowledge of their roles in physiology and pathophysiology has expanded immensely. VGSC ß subunits are multifunctional. They confer unique gating mechanisms, regulate cellular excitability, affect brain development, confer distinct channel pharmacology, and have functions that are independent of the α subunits. The vast array of functions of these proteins stems from their special station in the channelome: being the only known constituents that are cell adhesion and intra/extracellular signaling molecules in addition to being part of channel complexes. This functional trifecta and how it goes awry demonstrates the power outside the pore in ion channel signaling complexes, broadening the term channelopathy beyond defects in ion conduction. This article is part of the Special Issue entitled 'Channelopathies.'


Asunto(s)
Encefalopatías/metabolismo , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Canalopatías/metabolismo , Subunidades beta de Canales de Sodio Activados por Voltaje/metabolismo , Animales , Humanos
12.
Proc Natl Acad Sci U S A ; 114(9): 2383-2388, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28193882

RESUMEN

Patients with early infantile epileptic encephalopathy (EIEE) experience severe seizures and cognitive impairment and are at increased risk for sudden unexpected death in epilepsy (SUDEP). EIEE13 [Online Mendelian Inheritance in Man (OMIM) # 614558] is caused by de novo missense mutations in the voltage-gated sodium channel gene SCN8A Here, we investigated the neuronal phenotype of a mouse model expressing the gain-of-function SCN8A patient mutation, p.Asn1768Asp (Nav1.6-N1768D). Our results revealed regional and neuronal subtype specificity in the effects of the N1768D mutation. Acutely dissociated hippocampal neurons from Scn8aN1768D/+ mice showed increases in persistent sodium current (INa) density in CA1 pyramidal but not bipolar neurons. In CA3, INa,P was increased in both bipolar and pyramidal neurons. Measurement of action potential (AP) firing in Scn8aN1768D/+ pyramidal neurons in brain slices revealed early afterdepolarization (EAD)-like AP waveforms in CA1 but not in CA3 hippocampal or layer II/III neocortical neurons. The maximum spike frequency evoked by depolarizing current injections in Scn8aN1768D/+ CA1, but not CA3 or neocortical, pyramidal cells was significantly reduced compared with WT. Spontaneous firing was observed in subsets of neurons in CA1 and CA3, but not in the neocortex. The EAD-like waveforms of Scn8aN1768D/+ CA1 hippocampal neurons were blocked by tetrodotoxin, riluzole, and SN-6, implicating elevated persistent INa and reverse mode Na/Ca exchange in the mechanism of hyperexcitability. Our results demonstrate that Scn8a plays a vital role in neuronal excitability and provide insight into the mechanism and future treatment of epileptogenesis in EIEE13.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Mutación , Canal de Sodio Activado por Voltaje NAV1.6/genética , Células Piramidales/metabolismo , Espasmos Infantiles/genética , Potenciales de Acción/efectos de los fármacos , Sustitución de Aminoácidos , Animales , Compuestos de Bencilo/farmacología , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/patología , Región CA3 Hipocampal/efectos de los fármacos , Región CA3 Hipocampal/metabolismo , Región CA3 Hipocampal/patología , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Neocórtex/efectos de los fármacos , Neocórtex/metabolismo , Neocórtex/patología , Especificidad de Órganos , Células Piramidales/efectos de los fármacos , Células Piramidales/patología , Riluzol/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Espasmos Infantiles/metabolismo , Espasmos Infantiles/fisiopatología , Tetrodotoxina/farmacología , Tiazolidinas/farmacología
14.
J Neurosci ; 36(23): 6213-24, 2016 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-27277800

RESUMEN

UNLABELLED: Voltage-gated sodium channel (VGSC) ß subunits signal through multiple pathways on multiple time scales. In addition to modulating sodium and potassium currents, ß subunits play nonconducting roles as cell adhesion molecules, which allow them to function in cell-cell communication, neuronal migration, neurite outgrowth, neuronal pathfinding, and axonal fasciculation. Mutations in SCN1B, encoding VGSC ß1 and ß1B, are associated with epilepsy. Autosomal-dominant SCN1B-C121W, the first epilepsy-associated VGSC mutation identified, results in genetic epilepsy with febrile seizures plus (GEFS+). This mutation has been shown to disrupt both the sodium-current-modulatory and cell-adhesive functions of ß1 subunits expressed in heterologous systems. The goal of this study was to compare mice heterozygous for Scn1b-C121W (Scn1b(+/W)) with mice heterozygous for the Scn1b-null allele (Scn1b(+/-)) to determine whether the C121W mutation results in loss-of-function in vivo We found that Scn1b(+/W) mice were more susceptible than Scn1b(+/-) and Scn1b(+/+) mice to hyperthermia-induced convulsions, a model of pediatric febrile seizures. ß1-C121W subunits are expressed at the neuronal cell surface in vivo However, despite this, ß1-C121W polypeptides are incompletely glycosylated and do not associate with VGSC α subunits in the brain. ß1-C121W subcellular localization is restricted to neuronal cell bodies and is not detected at axon initial segments in the cortex or cerebellum or at optic nerve nodes of Ranvier of Scn1b(W/W) mice. These data, together with our previous results showing that ß1-C121W cannot participate in trans-homophilic cell adhesion, lead to the hypothesis that SCN1B-C121W confers a deleterious gain-of-function in human GEFS+ patients. SIGNIFICANCE STATEMENT: The mechanisms underlying genetic epilepsy syndromes are poorly understood. Closing this gap in knowledge is essential to the development of new medicines to treat epilepsy. We have used mouse models to understand the mechanism of a mutation in the sodium channel gene SCN1B linked to genetic epilepsy with febrile seizures plus. We report that sodium channel ß1 subunit proteins encoded by this mutant gene are expressed at the surface of neuronal cell bodies; however, they do not associate with the ion channel complex nor are they transported to areas of the axon that are critical for proper neuronal firing. We conclude that this disease-causing mutation is not simply a loss-of-function, but instead results in a deleterious gain-of-function in the brain.


Asunto(s)
Epilepsia/genética , Neuronas/fisiología , Polimorfismo de Nucleótido Simple/genética , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/genética , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/metabolismo , Animales , Animales Recién Nacidos , Biotinilación , Células Cultivadas , Corteza Cerebral/citología , Cisteína/genética , Modelos Animales de Enfermedad , Epilepsia/etiología , Epilepsia/patología , Fiebre/complicaciones , Regulación del Desarrollo de la Expresión Génica/genética , Inmunoprecipitación , Ratones , Ratones Transgénicos , Estadísticas no Paramétricas , Triptófano/genética
15.
J Phys Chem A ; 116(22): 5445-52, 2012 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-22616955

RESUMEN

The citrate reduction method is one of the simplest and most common methods used in the synthesis of gold nanoparticles. It has been thought that citrate acts as both a reducing agent for the gold salt and as the capping agent. However, it has recently been reported using density functional theory (DFT) that electron density builds up on uncomplexed apex gold atoms and the binding of formate (the simplest carboxylate and a model for citrate) becomes unfavorable after two additions, limiting citrate's utility as a capping agent. In this study, Au(20)-formyloxyl radical interactions are investigated using DFT at the BP86/DZ level of theory to model neutral carboxylate-gold nanoparticle binding (corresponding to carboxylates interacting with a partially oxidized gold nanoparticle). Binding energies are refined using a TZP basis set. It is found that the incremental binding energies of formyloxyl radicals remain highly favorable through eight additions (the highest number tested). The addition of one formyloxyl radical is 56 kJ/mol less than the addition of one formate but becomes 210 kJ/mol more favorable for the second addition. The range of binding energies through the eight additions is 154-331 kJ/mol. Furthermore, after the third addition, the most favorable geometries feature distortion of the gold tetrahedron. These results suggest that oxidized species formed in the citrate reduction method are likely capping agents and that binding of these ligands may affect the properties of the nanoparticles through distortion of the gold structure.


Asunto(s)
Formiatos/química , Radicales Libres/química , Oro/química , Nanopartículas del Metal/química , Sitios de Unión , Simulación por Computador , Ligandos , Modelos Moleculares , Teoría Cuántica , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...