Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neuroinflammation ; 20(1): 306, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38115011

RESUMEN

BACKGROUND: Excess tumor necrosis factor (TNF) is implicated in the pathogenesis of hyperinflammatory experimental cerebral malaria (eCM), including gliosis, increased levels of fibrin(ogen) in the brain, behavioral changes, and mortality. However, the role of TNF in eCM within the brain parenchyma, particularly directly on neurons, remains underdefined. Here, we investigate electrophysiological consequences of eCM on neuronal excitability and cell signaling mechanisms that contribute to observed phenotypes. METHODS: The split-luciferase complementation assay (LCA) was used to investigate cell signaling mechanisms downstream of tumor necrosis factor receptor 1 (TNFR1) that could contribute to changes in neuronal excitability in eCM. Whole-cell patch-clamp electrophysiology was performed in brain slices from eCM mice to elucidate consequences of infection on CA1 pyramidal neuron excitability and cell signaling mechanisms that contribute to observed phenotypes. Involvement of identified signaling molecules in mediating behavioral changes and sickness behavior observed in eCM were investigated in vivo using genetic silencing. RESULTS: Exploring signaling mechanisms that underlie TNF-induced effects on neuronal excitability, we found that the complex assembly of fibroblast growth factor 14 (FGF14) and the voltage-gated Na+ (Nav) channel 1.6 (Nav1.6) is increased upon tumor necrosis factor receptor 1 (TNFR1) stimulation via Janus Kinase 2 (JAK2). On account of the dependency of hyperinflammatory experimental cerebral malaria (eCM) on TNF, we performed patch-clamp studies in slices from eCM mice and showed that Plasmodium chabaudi infection augments Nav1.6 channel conductance of CA1 pyramidal neurons through the TNFR1-JAK2-FGF14-Nav1.6 signaling network, which leads to hyperexcitability. Hyperexcitability of CA1 pyramidal neurons caused by infection was mitigated via an anti-TNF antibody and genetic silencing of FGF14 in CA1. Furthermore, knockdown of FGF14 in CA1 reduced sickness behavior caused by infection. CONCLUSIONS: FGF14 may represent a therapeutic target for mitigating consequences of TNF-mediated neuroinflammation.


Asunto(s)
Conducta de Enfermedad , Malaria Cerebral , Ratones , Animales , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Inhibidores del Factor de Necrosis Tumoral , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Neuronas/metabolismo , Transducción de Señal
2.
Proc Natl Acad Sci U S A ; 117(14): 8143-8153, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32209671

RESUMEN

Although major depressive disorder (MDD) is highly prevalent, its pathophysiology is poorly understood. Recent evidence suggests that glycogen-synthase kinase 3ß (GSK3ß) plays a key role in memory formation, yet its role in mood regulation remains controversial. Here, we investigated whether GSK3ß activity in the nucleus accumbens (NAc) is associated with depression-like behaviors and synaptic plasticity. We performed whole-cell patch-clamp recordings of medium spiny neurons (MSNs) in the NAc and determined the role of GSK3ß in spike timing-dependent long-term potentiation (tLTP) in the chronic unpredictable mild stress (CUMS) mouse model of depression. To assess the specific role of GSK3ß in tLTP, we used in vivo genetic silencing by an adeno-associated viral vector (AAV2) short hairpin RNA against GSK3ß. In addition, we examined the role of the voltage-gated potassium Kv4.2 subunit, a molecular determinant of A-type K+ currents, as a potential downstream target of GSK3ß. We found increased levels of active GSK3ß and augmented tLTP in CUMS mice, a phenotype that was prevented by selective GSK3ß knockdown. Furthermore, knockdown of GSK3ß in the NAc ameliorated depressive-like behavior in CUMS mice. Electrophysiological, immunohistochemical, biochemical, and pharmacological experiments revealed that inhibition of the Kv4.2 channel through direct phosphorylation at Ser-616 mediated the GSK3ß-dependent tLTP changes in CUMS mice. Our results identify GSK3ß regulation of Kv4.2 channels as a molecular mechanism of MSN maladaptive plasticity underlying depression-like behaviors and suggest that the GSK3ß-Kv4.2 axis may be an attractive therapeutic target for MDD.


Asunto(s)
Trastorno Depresivo Mayor/patología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Plasticidad Neuronal , Núcleo Accumbens/patología , Canales de Potasio Shal/metabolismo , Potenciales de Acción , Animales , Conducta Animal , Trastorno Depresivo Mayor/etiología , Trastorno Depresivo Mayor/psicología , Modelos Animales de Enfermedad , Masculino , Ratones , Neuronas/patología , Núcleo Accumbens/citología , Técnicas de Placa-Clamp , Estrés Psicológico/complicaciones , Estrés Psicológico/psicología , Factores de Tiempo
3.
Int J Mol Sci ; 23(8)2022 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-35457230

RESUMEN

Glycogen synthase kinase 3ß (GSK3) is a multifaceted serine/threonine (S/T) kinase expressed in all eukaryotic cells. GSK3ß is highly enriched in neurons in the central nervous system where it acts as a central hub for intracellular signaling downstream of receptors critical for neuronal function. Unlike other kinases, GSK3ß is constitutively active, and its modulation mainly involves inhibition via upstream regulatory pathways rather than increased activation. Through an intricate converging signaling system, a fine-tuned balance of active and inactive GSK3ß acts as a central point for the phosphorylation of numerous primed and unprimed substrates. Although the full range of molecular targets is still unknown, recent results show that voltage-gated ion channels are among the downstream targets of GSK3ß. Here, we discuss the direct and indirect mechanisms by which GSK3ß phosphorylates voltage-gated Na+ channels (Nav1.2 and Nav1.6) and voltage-gated K+ channels (Kv4 and Kv7) and their physiological effects on intrinsic excitability, neuronal plasticity, and behavior. We also present evidence for how unbalanced GSK3ß activity can lead to maladaptive plasticity that ultimately renders neuronal circuitry more vulnerable, increasing the risk for developing neuropsychiatric disorders. In conclusion, GSK3ß-dependent modulation of voltage-gated ion channels may serve as an important pharmacological target for neurotherapeutic development.


Asunto(s)
Glucógeno Sintasa Quinasa 3 , Neuronas , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Canales Iónicos/metabolismo , Neuronas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas
4.
Int J Mol Sci ; 23(3)2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-35163623

RESUMEN

In neurons, changes in Akt activity have been detected in response to the stimulation of transmembrane receptors. However, the mechanisms that lead to changes in neuronal function upon Akt inhibition are still poorly understood. In the present study, we interrogate how Akt inhibition could affect the activity of the neuronal Nav channels with while impacting intrinsic excitability. To that end, we employed voltage-clamp electrophysiological recordings in heterologous cells expressing the Nav1.6 channel isoform and in hippocampal CA1 pyramidal neurons in the presence of triciribine, an inhibitor of Akt. We showed that in both systems, Akt inhibition resulted in a potentiation of peak transient Na+ current (INa) density. Akt inhibition correspondingly led to an increase in the action potential firing of the CA1 pyramidal neurons that was accompanied by a decrease in the action potential current threshold. Complementary confocal analysis in the CA1 pyramidal neurons showed that the inhibition of Akt is associated with the lengthening of Nav1.6 fluorescent intensity along the axonal initial segment (AIS), providing a mechanism for augmented neuronal excitability. Taken together, these findings provide evidence that Akt-mediated signal transduction might affect neuronal excitability in a Nav1.6-dependent manner.


Asunto(s)
Potenciales de Acción , Hipocampo/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Animales , Células HEK293 , Hipocampo/metabolismo , Hipocampo/fisiología , Humanos , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Células Piramidales/fisiología
5.
Int J Mol Sci ; 22(24)2021 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-34948337

RESUMEN

Voltage-gated Na+ (Nav) channels are the primary molecular determinant of the action potential. Among the nine isoforms of the Nav channel α subunit that have been described (Nav1.1-Nav1.9), Nav1.1, Nav1.2, and Nav1.6 are the primary isoforms expressed in the central nervous system (CNS). Crucially, these three CNS Nav channel isoforms display differential expression across neuronal cell types and diverge with respect to their subcellular distributions. Considering these differences in terms of their localization, the CNS Nav channel isoforms could represent promising targets for the development of targeted neuromodulators. However, current therapeutics that target Nav channels lack selectivity, which results in deleterious side effects due to modulation of off-target Nav channel isoforms. Among the structural components of the Nav channel α subunit that could be pharmacologically targeted to achieve isoform selectivity, the C-terminal domains (CTD) of Nav channels represent promising candidates on account of displaying appreciable amino acid sequence divergence that enables functionally unique protein-protein interactions (PPIs) with Nav channel auxiliary proteins. In medium spiny neurons (MSNs) of the nucleus accumbens (NAc), a critical brain region of the mesocorticolimbic circuit, the PPI between the CTD of the Nav1.6 channel and its auxiliary protein fibroblast growth factor 14 (FGF14) is central to the generation of electrical outputs, underscoring its potential value as a site for targeted neuromodulation. Focusing on this PPI, we previously developed a peptidomimetic derived from residues of FGF14 that have an interaction site on the CTD of the Nav1.6 channel. In this work, we show that whereas the compound displays dose-dependent effects on the activity of Nav1.6 channels in heterologous cells, the compound does not affect Nav1.1 or Nav1.2 channels at comparable concentrations. In addition, we show that the compound correspondingly modulates the action potential discharge and the transient Na+ of MSNs of the NAc. Overall, these results demonstrate that pharmacologically targeting the FGF14 interaction site on the CTD of the Nav1.6 channel is a strategy to achieve isoform-selective modulation, and, more broadly, that sites on the CTDs of Nav channels interacted with by auxiliary proteins could represent candidates for the development of targeted therapeutics.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.6/efectos de los fármacos , Neuronas/metabolismo , Peptidomiméticos/farmacología , Dominios Proteicos , Animales , Factores de Crecimiento de Fibroblastos/metabolismo , Células HEK293 , Humanos , Ratones , Simulación del Acoplamiento Molecular , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Canal de Sodio Activado por Voltaje NAV1.6/fisiología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiología , Unión Proteica
6.
Cereb Cortex ; 29(5): 1851-1865, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29790931

RESUMEN

Spike timing-dependent plasticity (STDP) is a form of activity-dependent remodeling of synaptic strength that underlies memory formation. Despite its key role in dictating learning rules in the brain circuits, the molecular mechanisms mediating STDP are still poorly understood. Here, we show that spike timing-dependent long-term depression (tLTD) and A-type K+ currents are modulated by pharmacological agents affecting the levels of active glycogen-synthase kinase 3 (GSK3) and by GSK3ß knockdown in layer 2/3 of the mouse somatosensory cortex. Moreover, the blockade of A-type K+ currents mimics the effects of GSK3 up-regulation on tLTD and occludes further changes in synaptic strength. Pharmacological, immunohistochemical and biochemical experiments revealed that GSK3ß influence over tLTD induction is mediated by direct phosphorylation at Ser-616 of the Kv4.2 subunit, a molecular determinant of A-type K+ currents. Collectively, these results identify the functional interaction between GSK3ß and Kv4.2 channel as a novel mechanism for tLTD modulation providing exciting insight into the understanding of GSK3ß role in synaptic plasticity.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta/metabolismo , Depresión Sináptica a Largo Plazo/fisiología , Neuronas/fisiología , Canales de Potasio Shal/metabolismo , Corteza Somatosensorial/fisiología , Animales , Potenciales Postsinápticos Excitadores , Ratones Endogámicos C57BL , Neuronas/metabolismo , Fosforilación , Corteza Somatosensorial/metabolismo
7.
Molecules ; 25(15)2020 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-32722255

RESUMEN

Disruption of protein:protein interactions (PPIs) that regulate the function of voltage-gated Na+ (Nav) channels leads to neural circuitry aberrations that have been implicated in numerous channelopathies. One example of this pathophysiology is mediated by dysfunction of the PPI between Nav1.6 and its regulatory protein fibroblast growth factor 14 (FGF14). Thus, peptides derived from FGF14 might exert modulatory actions on the FGF14:Nav1.6 complex that are functionally relevant. The tetrapeptide Glu-Tyr-Tyr-Val (EYYV) mimics surface residues of FGF14 at the ß8-ß9 loop, a structural region previously implicated in its binding to Nav1.6. Here, peptidomimetics derived from EYYV (6) were designed, synthesized, and pharmacologically evaluated to develop probes with improved potency. Addition of hydrophobic protective groups to 6 and truncation to a tripeptide (12) produced a potent inhibitor of FGF14:Nav1.6 complex assembly. Conversely, addition of hydrophobic protective groups to 6 followed by addition of an N-terminal benzoyl substituent (19) produced a potentiator of FGF14:Nav1.6 complex assembly. Subsequent functional evaluation using whole-cell patch-clamp electrophysiology confirmed their inverse activities, with 12 and 19 reducing and increasing Nav1.6-mediated transient current densities, respectively. Overall, we have identified a negative and positive allosteric modulator of Nav1.6, both of which could serve as scaffolds for the development of target-selective neurotherapeutics.


Asunto(s)
Factores de Crecimiento de Fibroblastos/química , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Peptidomiméticos/síntesis química , Peptidomiméticos/farmacología , Regulación Alostérica , Sitios de Unión , Diseño de Fármacos , Factores de Crecimiento de Fibroblastos/metabolismo , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Simulación del Acoplamiento Molecular , Estructura Molecular , Canal de Sodio Activado por Voltaje NAV1.6/química , Peptidomiméticos/química , Unión Proteica/efectos de los fármacos , Estructura Secundaria de Proteína
8.
Bioorg Med Chem Lett ; 29(3): 413-419, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30587448

RESUMEN

The voltage-gated sodium (Nav) channel is the molecular determinant of action potential in neurons. Protein-protein interactions (PPI) between the intracellular Nav1.6 C-tail and its regulatory protein fibroblast growth factor 14 (FGF14) provide an ideal and largely untapped opportunity for development of neurochemical probes. Based on a previously identified peptide FLPK, mapped to the FGF14:FGF14 PPI interface, we have designed and synthesized a series of peptidomimetics with the intent of increasing clogP values and improving cell permeability relative to the parental lead peptide. In-cell screening using the split-luciferase complementation (LCA) assay identified ZL0177 (13) as the most potent inhibitor of the FGF14:Nav1.6 channel complex assembly with an apparent IC50 of 11 µM. Whole-cell patch-clamp recordings demonstrated that ZL0177 significantly reduced Nav1.6-mediated transient current density and induced a depolarizing shift of the channel voltage-dependence of activation. Docking studies revealed strong interactions between ZL0177 and Nav1.6, mediated by hydrogen bonds, cation-π interactions and hydrophobic contacts. All together these results suggest that ZL0177 retains some key features of FGF14-dependent modulation of Nav1.6 currents. Overall, ZL0177 provides a chemical scaffold for developing Nav channel modulators as pharmacological probes with therapeutic potential of interest for a broad range of CNS and PNS disorders.


Asunto(s)
Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Sondas Moleculares/farmacología , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Oligopéptidos/farmacología , Peptidomiméticos/farmacología , Relación Dosis-Respuesta a Droga , Factores de Crecimiento de Fibroblastos/química , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Sondas Moleculares/síntesis química , Sondas Moleculares/química , Estructura Molecular , Canal de Sodio Activado por Voltaje NAV1.6/química , Oligopéptidos/síntesis química , Oligopéptidos/química , Peptidomiméticos/síntesis química , Peptidomiméticos/química , Unión Proteica/efectos de los fármacos , Relación Estructura-Actividad
9.
J Biol Chem ; 291(21): 11268-84, 2016 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-26994141

RESUMEN

The voltage-gated Na(+) (Nav) channel provides the basis for electrical excitability in the brain. This channel is regulated by a number of accessory proteins including fibroblast growth factor 14 (FGF14), a member of the intracellular FGF family. In addition to forming homodimers, FGF14 binds directly to the Nav1.6 channel C-tail, regulating channel gating and expression, properties that are required for intrinsic excitability in neurons. Seeking amino acid residues with unique roles at the protein-protein interaction interface (PPI) of FGF14·Nav1.6, we engineered model-guided mutations of FGF14 and validated their impact on the FGF14·Nav1.6 complex and the FGF14:FGF14 dimer formation using a luciferase assay. Divergence was found in the ß-9 sheet of FGF14 where an alanine (Ala) mutation of Val-160 impaired binding to Nav1.6 but had no effect on FGF14:FGF14 dimer formation. Additional analysis revealed also a key role of residues Lys-74/Ile-76 at the N-terminal of FGF14 in the FGF14·Nav1.6 complex and FGF14:FGF14 dimer formation. Using whole-cell patch clamp electrophysiology, we demonstrated that either the FGF14(V160A) or the FGF14(K74A/I76A) mutation was sufficient to abolish the FGF14-dependent regulation of peak transient Na(+) currents and the voltage-dependent activation and steady-state inactivation of Nav1.6; but only V160A with a concomitant alanine mutation at Tyr-158 could impede FGF14-dependent modulation of the channel fast inactivation. Intrinsic fluorescence spectroscopy of purified proteins confirmed a stronger binding reduction of FGF14(V160A) to the Nav1.6 C-tail compared with FGF14(K74A/I76A) Altogether these studies indicate that the ß-9 sheet and the N terminus of FGF14 are well positioned targets for drug development of PPI-based allosteric modulators of Nav channels.


Asunto(s)
Factores de Crecimiento de Fibroblastos/química , Factores de Crecimiento de Fibroblastos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.6/química , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Sustitución de Aminoácidos , Aminoácidos/química , Factores de Crecimiento de Fibroblastos/genética , Células HEK293 , Humanos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Canal de Sodio Activado por Voltaje NAV1.6/genética , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología Estructural de Proteína , Relación Estructura-Actividad
10.
FASEB J ; 30(6): 2171-86, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26917740

RESUMEN

Recent data shows that fibroblast growth factor 14 (FGF14) binds to and controls the function of the voltage-gated sodium (Nav) channel with phenotypic outcomes on neuronal excitability. Mutations in the FGF14 gene in humans have been associated with brain disorders that are partially recapitulated in Fgf14(-/-) mice. Thus, signaling pathways that modulate the FGF14:Nav channel interaction may be important therapeutic targets. Bioluminescence-based screening of small molecule modulators of the FGF14:Nav1.6 complex identified 4,5,6,7 -: tetrabromobenzotriazole (TBB), a potent casein kinase 2 (CK2) inhibitor, as a strong suppressor of FGF14:Nav1.6 interaction. Inhibition of CK2 through TBB reduces the interaction of FGF14 with Nav1.6 and Nav1.2 channels. Mass spectrometry confirmed direct phosphorylation of FGF14 by CK2 at S228 and S230, and mutation to alanine at these sites modified FGF14 modulation of Nav1.6-mediated currents. In 1 d in vitro hippocampal neurons, TBB induced a reduction in FGF14 expression, a decrease in transient Na(+) current amplitude, and a hyperpolarizing shift in the voltage dependence of Nav channel steady-state inactivation. In mature neurons, TBB reduces the axodendritic polarity of FGF14. In cornu ammonis area 1 hippocampal slices from wild-type mice, TBB impairs neuronal excitability by increasing action potential threshold and lowering firing frequency. Importantly, these changes in excitability are recapitulated in Fgf14(-/-) mice, and deletion of Fgf14 occludes TBB-dependent phenotypes observed in wild-type mice. These results suggest that a CK2-FGF14 axis may regulate Nav channels and neuronal excitability.-Hsu, W.-C. J., Scala, F., Nenov, M. N., Wildburger, N. C., Elferink, H., Singh, A. K., Chesson, C. B., Buzhdygan, T., Sohail, M., Shavkunov, A. S., Panova, N. I., Nilsson, C. L., Rudra, J. S., Lichti, C. F., Laezza, F. CK2 activity is required for the interaction of FGF14 with voltage-gated sodium channels and neuronal excitability.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Neuronas/fisiología , Canales de Sodio Activados por Voltaje/fisiología , Animales , Quinasa de la Caseína II/genética , Femenino , Factores de Crecimiento de Fibroblastos/genética , Regulación Enzimológica de la Expresión Génica , Células HEK293 , Hipocampo/citología , Hipocampo/fisiología , Humanos , Masculino , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp
11.
Mol Cell Proteomics ; 14(5): 1288-300, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25724910

RESUMEN

Voltage-gated sodium channels (Nav1.1-Nav1.9) are responsible for the initiation and propagation of action potentials in neurons, controlling firing patterns, synaptic transmission and plasticity of the brain circuit. Yet, it is the protein-protein interactions of the macromolecular complex that exert diverse modulatory actions on the channel, dictating its ultimate functional outcome. Despite the fundamental role of Nav channels in the brain, information on its proteome is still lacking. Here we used affinity purification from crude membrane extracts of whole brain followed by quantitative high-resolution mass spectrometry to resolve the identity of Nav1.2 protein interactors. Of the identified putative protein interactors, fibroblast growth factor 12 (FGF12), a member of the nonsecreted intracellular FGF family, exhibited 30-fold enrichment in Nav1.2 purifications compared with other identified proteins. Using confocal microscopy, we visualized native FGF12 in the brain tissue and confirmed that FGF12 forms a complex with Nav1.2 channels at the axonal initial segment, the subcellular specialized domain of neurons required for action potential initiation. Co-immunoprecipitation studies in a heterologous expression system validate Nav1.2 and FGF12 as interactors, whereas patch-clamp electrophysiology reveals that FGF12 acts synergistically with CaMKII, a known kinase regulator of Nav channels, to modulate Nav1.2-encoded currents. In the presence of CaMKII inhibitors we found that FGF12 produces a bidirectional shift in the voltage-dependence of activation (more depolarized) and the steady-state inactivation (more hyperpolarized) of Nav1.2, increasing the channel availability. Although providing the first characterization of the Nav1.2 CNS proteome, we identify FGF12 as a new functionally relevant interactor. Our studies will provide invaluable information to parse out the molecular determinant underlying neuronal excitability and plasticity, and extending the relevance of iFGFs signaling in the normal and diseased brain.


Asunto(s)
Encéfalo/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.2/metabolismo , Neuronas/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Encéfalo/citología , Encéfalo/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/farmacología , Membrana Celular , Factores de Crecimiento de Fibroblastos/química , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/farmacología , Expresión Génica , Células HEK293 , Humanos , Inmunoprecipitación , Anotación de Secuencia Molecular , Canal de Sodio Activado por Voltaje NAV1.2/química , Canal de Sodio Activado por Voltaje NAV1.2/genética , Plasticidad Neuronal , Neuronas/citología , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Unión Proteica , Proteoma/genética , Proteoma/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
12.
Biochim Biophys Acta ; 1850(4): 832-44, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25615535

RESUMEN

BACKGROUND: Phosphorylation plays an essential role in regulating voltage-gated sodium (Na(v)) channels and excitability. Yet, a surprisingly limited number of kinases have been identified as regulators of Na(v) channels. We posited that glycogen synthase kinase 3 (GSK3), a critical kinase found associated with numerous brain disorders, might directly regulate neuronal Na(v) channels. METHODS: We used patch-clamp electrophysiology to record sodium currents from Na(v)1.2 channels stably expressed in HEK-293 cells. mRNA and protein levels were quantified with RT-PCR, Western blot, or confocal microscopy, and in vitro phosphorylation and mass spectrometry to identify phosphorylated residues. RESULTS: We found that exposure of cells to GSK3 inhibitor XIII significantly potentiates the peak current density of Na(v)1.2, a phenotype reproduced by silencing GSK3 with siRNA. Contrarily, overexpression of GSK3ß suppressed Na(v)1.2-encoded currents. Neither mRNA nor total protein expression was changed upon GSK3 inhibition. Cell surface labeling of CD4-chimeric constructs expressing intracellular domains of the Na(v)1.2 channel indicates that cell surface expression of CD4-Na(v)1.2 C-tail was up-regulated upon pharmacological inhibition of GSK3, resulting in an increase of surface puncta at the plasma membrane. Finally, using in vitro phosphorylation in combination with high resolution mass spectrometry, we further demonstrate that GSK3ß phosphorylates T(1966) at the C-terminal tail of Na(v)1.2. CONCLUSION: These findings provide evidence for a new mechanism by which GSK3 modulates Na(v) channel function via its C-terminal tail. GENERAL SIGNIFICANCE: These findings provide fundamental knowledge in understanding signaling dysfunction common in several neuropsychiatric disorders.


Asunto(s)
Glucógeno Sintasa Quinasa 3/fisiología , Canal de Sodio Activado por Voltaje NAV1.2/fisiología , Secuencia de Aminoácidos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Células HEK293 , Humanos , Datos de Secuencia Molecular , Canal de Sodio Activado por Voltaje NAV1.2/química , Fosforilación
13.
J Neurosci ; 34(3): 1028-36, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24431460

RESUMEN

Hippocampal network hyperexcitability is considered an early indicator of Alzheimer's disease (AD) memory impairment. Some AD mouse models exhibit similar network phenotypes. In this study we focused on dentate gyrus (DG) granule cell spontaneous and evoked properties in 9-month-old Tg2576 mice that model AD amyloidosis and cognitive deficits. Using whole-cell patch-clamp recordings, we found that Tg2576 DG granule cells exhibited spontaneous EPSCs that were higher in frequency but not amplitude compared with wild-type mice, suggesting hyperactivity of DG granule cells via a presynaptic mechanism. Further support of a presynaptic mechanism was revealed by increased I-O relationships and probability of release in Tg2576 DG granule cells. Since we and others have shown that activation of the peroxisome proliferator-activated receptor gamma (PPARγ) axis improves hippocampal cognition in mouse models for AD as well as benefitting memory performance in some humans with early AD, we investigated how PPARγ agonism affected synaptic activity in Tg2576 DG. We found that PPARγ agonism normalized the I-O relationship of evoked EPSCs, frequency of spontaneous EPSCs, and probability of release that, in turn, correlated with selective expression of DG proteins essential for presynaptic SNARE function that are altered in patients with AD. These findings provide evidence that DG principal cells may contribute to early AD hippocampal network hyperexcitability via a presynaptic mechanism, and that hippocampal cognitive enhancement via PPARγ activation occurs through regulation of presynaptic vesicular proteins critical for proper glutamatergic neurotransmitter release, synaptic transmission, and short-term plasticity.


Asunto(s)
Giro Dentado/fisiología , Nootrópicos/farmacología , PPAR gamma/agonistas , PPAR gamma/fisiología , Terminales Presinápticos/fisiología , Tiazolidinedionas/farmacología , Precursor de Proteína beta-Amiloide/genética , Animales , Giro Dentado/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Terminales Presinápticos/efectos de los fármacos , Mapas de Interacción de Proteínas/efectos de los fármacos , Mapas de Interacción de Proteínas/fisiología , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Rosiglitazona
14.
J Neurophysiol ; 113(6): 1712-26, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25540218

RESUMEN

Early cognitive impairment in Alzheimer's disease (AD) correlates with medial temporal lobe dysfunction, including two areas essential for memory formation: the entorhinal cortex and dentate gyrus (DG). In the Tg2576 animal model for AD amyloidosis, activation of the peroxisome proliferator-activated receptor-gamma (PPARγ) with rosiglitazone (RSG) ameliorates hippocampus-dependent cognitive impairment and restores aberrant synaptic activity at the entorhinal cortex to DG granule neuron inputs. It is unknown, however, whether intrinsic firing properties of DG granule neurons in these animals are affected by amyloid-ß pathology and if they are sensitive to RSG treatment. Here, we report that granule neurons from 9-mo-old wild-type and Tg2576 animals can be segregated into two cell types with distinct firing properties and input resistance that correlate with less mature type I and more mature type II neurons. The DG type I cell population was greater than type II in wild-type littermates. In the Tg2576 animals, the type I and type II cell populations were nearly equal but could be restored to wild-type levels through cognitive enhancement with RSG. Furthermore, Tg2576 cell firing frequency and spike after depolarization were decreased in type I and increased in type II cells, both of which could also be restored to wild-type levels upon RSG treatment. That these parameters were restored by PPARγ activation emphasizes the therapeutic value of RSG against early AD cognitive impairment.


Asunto(s)
Potenciales de Acción , Enfermedad de Alzheimer/metabolismo , Neuronas/fisiología , PPAR gamma/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Cognición , Corteza Entorrinal/citología , Corteza Entorrinal/efectos de los fármacos , Corteza Entorrinal/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , PPAR gamma/agonistas , Rosiglitazona , Tiazolidinedionas/farmacología
15.
J Neurochem ; 133(1): 125-33, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25640170

RESUMEN

Compelling evidence indicates that type 2 diabetes mellitus, insulin resistance (IR), and metabolic syndrome are often accompanied by cognitive impairment. However, the mechanistic link between these metabolic abnormalities and CNS dysfunction requires further investigations. Here, we evaluated whether adipose tissue IR and related metabolic alterations resulted in CNS changes by studying synapse lipid composition and function in the adipocyte-specific ecto-nucleotide pyrophosphate phosphodiesterase over-expressing transgenic (AtENPP1-Tg) mouse, a model characterized by white adipocyte IR, systemic IR, and ectopic fat deposition. When fed a high-fat diet, AtENPP1-Tg mice recapitulate essential features of the human metabolic syndrome, making them an ideal model to characterize peripherally induced CNS deficits. Using a combination of gas chromatography and western blot analysis, we found evidence of altered lipid composition, including decreased phospholipids and increased triglycerides (TG) and free fatty acid in hippocampal synaptosomes isolated from high-fat diet-fed AtENPP1-Tg mice. These changes were associated with impaired basal synaptic transmission at the Schaffer collaterals to hippocampal cornu ammonis 1 (CA1) synapses, decreased phosphorylation of the GluN1 glutamate receptor subunit, down-regulation of insulin receptor expression, and up-regulation of the free fatty acid receptor 1.


Asunto(s)
Tejido Adiposo/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiología , Resistencia a la Insulina/fisiología , Metabolismo de los Lípidos/fisiología , Sinapsis/metabolismo , Sinapsis/fisiología , Animales , Química Encefálica/efectos de los fármacos , Hipoglucemiantes/farmacología , Insulina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor de Insulina/metabolismo , Sinaptosomas/metabolismo
16.
Anal Chem ; 87(19): 10006-14, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26356480

RESUMEN

Abnormal epigenetic reprogramming is one of the major causes leading to irregular gene expression and regulatory pathway perturbations, in the cells, resulting in unhealthy cell development or diseases. Accurate measurements of these changes of epigenetic modifications, especially the complex histone modifications, are very important, and the methods for these measurements are not trivial. By following our previous introduction of PRM to targeting histone modifications (Tang, H.; Fang, H.; Yin, E.; Brasier, A. R.; Sowers, L. C.; Zhang, K. Multiplexed parallel reaction monitoring targeting histone modifications on the QExactive mass spectrometer. Anal. Chem. 2014, 86 (11), 5526-34), herein we validated this method by varying the protein/trypsin ratios via serial dilutions. Our data demonstrated that PRM with SILAC histones as the internal standards allowed reproducible measurements of histone H3/H4 acetylation and methylation in the samples whose histone contents differ at least one-order of magnitude. The method was further validated by histones isolated from histone H3 K36 trimethyltransferase SETD2 knockout mouse embryonic fibroblasts (MEF) cells. Furthermore, histone acetylation and methylation in human neural stem cells (hNSC) treated with ascorbic acid phosphate (AAP) were measured by this method, revealing that H3 K36 trimethylation was significantly down-regulated by 6 days of treatment with vitamin C.


Asunto(s)
Histonas/análisis , Acetilación , Secuencia de Aminoácidos , Animales , Western Blotting/métodos , Línea Celular , Células Cultivadas , Epigénesis Genética , Código de Histonas , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Histonas/metabolismo , Humanos , Metilación , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Células-Madre Neurales/metabolismo , Espectrometría de Masas en Tándem/métodos , Tripsina/metabolismo
18.
J Neurosci ; 33(4): 1615-30, 2013 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-23345234

RESUMEN

Serotonin (5-hydroxytryptamine; 5-HT) signaling through the 5-HT(2C) receptor (5-HT(2C)R) is essential in normal physiology, whereas aberrant 5-HT(2C)R function is thought to contribute to the pathogenesis of multiple neural disorders. The 5-HT(2C)R interacts with specific protein partners, but the impact of such interactions on 5-HT(2C)R function is poorly understood. Here, we report convergent cellular and behavioral data that the interaction between the 5-HT(2C)R and protein phosphatase and tensin homolog (PTEN) serves as a regulatory mechanism to control 5-HT(2C)R-mediated biology but not that of the closely homologous 5-HT(2A)R. A peptide derived from the third intracellular loop of the human 5-HT(2C)R [3L4F (third loop, fourth fragment)] disrupted the association, allosterically augmented 5-HT(2C)R-mediated signaling in live cells, and acted as a positive allosteric modulator in rats in vivo. We identified the critical residues within an 8 aa fragment of the 3L4F peptide that maintained efficacy (within the picomolar range) in live cells similar to that of the 3L4F peptide. Last, molecular modeling identified key structural features and potential interaction sites of the active 3L4F peptides against PTEN. These compelling data demonstrate the specificity and importance of this protein assembly in cellular events and behaviors mediated by 5-HT(2C)R signaling and provide a chemical guidepost to the future development of drug-like peptide or small-molecule inhibitors as neuroprobes to study 5-HT(2C)R allostery and therapeutics for 5-HT(2C)R-mediated disorders.


Asunto(s)
Modelos Moleculares , Fosfohidrolasa PTEN/química , Fosfohidrolasa PTEN/metabolismo , Receptor de Serotonina 5-HT2C/química , Receptor de Serotonina 5-HT2C/metabolismo , Transducción de Señal/fisiología , Secuencia de Aminoácidos , Animales , Humanos , Immunoblotting , Inmunoprecipitación , Masculino , Datos de Secuencia Molecular , Actividad Motora/fisiología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Ratas , Ratas Sprague-Dawley , Serotonina/metabolismo , Transfección
19.
J Biol Chem ; 288(27): 19370-85, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23640885

RESUMEN

The FGF14 protein controls biophysical properties and subcellular distribution of neuronal voltage-gated Na(+) (Nav) channels through direct binding to the channel C terminus. To gain insights into the dynamic regulation of this protein/protein interaction complex, we employed the split luciferase complementation assay to screen a small molecule library of kinase inhibitors against the FGF14·Nav1.6 channel complex and identified inhibitors of GSK3 as hits. Through a combination of a luminescence-based counter-screening, co-immunoprecipitation, patch clamp electrophysiology, and quantitative confocal immunofluorescence, we demonstrate that inhibition of GSK3 reduces the assembly of the FGF14·Nav channel complex, modifies FGF14-dependent regulation of Na(+) currents, and induces dissociation and subcellular redistribution of the native FGF14·Nav channel complex in hippocampal neurons. These results further emphasize the role of FGF14 as a critical component of the Nav channel macromolecular complex, providing evidence for a novel GSK3-dependent signaling pathway that might control excitability through specific protein/protein interactions.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Hipocampo/metabolismo , Complejos Multiproteicos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Neuronas/metabolismo , Animales , Inhibidores Enzimáticos/farmacología , Factores de Crecimiento de Fibroblastos/genética , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Células HEK293 , Hipocampo/citología , Humanos , Ratones , Ratones Noqueados , Complejos Multiproteicos/genética , Canal de Sodio Activado por Voltaje NAV1.6/genética , Neuronas/citología , Ratas , Transducción de Señal/efectos de los fármacos
20.
Life (Basel) ; 13(8)2023 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-37629512

RESUMEN

Alzheimer's disease (AD) is the most common cause of dementia and is classically characterized by two major histopathological abnormalities: extracellular plaques composed of amyloid beta (Aß) and intracellular hyperphosphorylated tau. Due to the progressive nature of the disease, it is of the utmost importance to develop disease-modifying therapeutics that tackle AD pathology in its early stages. Attenuation of hippocampal hyperactivity, one of the earliest neuronal abnormalities observed in AD brains, has emerged as a promising strategy to ameliorate cognitive deficits and abate the spread of neurotoxic species. This aberrant hyperactivity has been attributed in part to the dysfunction of voltage-gated Na+ (Nav) channels, which are central mediators of neuronal excitability. Therefore, targeting Nav channels is a promising strategy for developing disease-modifying therapeutics that can correct aberrant neuronal phenotypes in early-stage AD. This review will explore the role of Nav channels in neuronal function, their connections to AD pathology, and their potential as therapeutic targets.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA