Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33593893

RESUMEN

Behaviors that rely on the hippocampus are particularly susceptible to chronological aging, with many aged animals (including humans) maintaining cognition at a young adult-like level, but many others the same age showing marked impairments. It is unclear whether the ability to maintain cognition over time is attributable to brain maintenance, sufficient cognitive reserve, compensatory changes in network function, or some combination thereof. While network dysfunction within the hippocampal circuit of aged, learning-impaired animals is well-documented, its neurobiological substrates remain elusive. Here we show that the synaptic architecture of hippocampal regions CA1 and CA3 is maintained in a young adult-like state in aged rats that performed comparably to their young adult counterparts in both trace eyeblink conditioning and Morris water maze learning. In contrast, among learning-impaired, but equally aged rats, we found that a redistribution of synaptic weights amplifies the influence of autoassociational connections among CA3 pyramidal neurons, yet reduces the synaptic input onto these same neurons from the dentate gyrus. Notably, synapses within hippocampal region CA1 showed no group differences regardless of cognitive ability. Taking the data together, we find the imbalanced synaptic weights within hippocampal CA3 provide a substrate that can explain the abnormal firing characteristics of both CA3 and CA1 pyramidal neurons in aged, learning-impaired rats. Furthermore, our work provides some clarity with regard to how some animals cognitively age successfully, while others' lifespans outlast their "mindspans."


Asunto(s)
Región CA1 Hipocampal/patología , Región CA3 Hipocampal/patología , Envejecimiento Cognitivo , Células Piramidales/patología , Sinapsis/patología , Animales , Masculino , Ratas , Ratas Endogámicas BN , Ratas Endogámicas F344
2.
J Biol Chem ; 298(7): 102069, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35623388

RESUMEN

Major depressive disorder is a critical public health problem with a lifetime prevalence of nearly 17% in the United States. One potential therapeutic target is the interaction between hyperpolarization-activated cyclic nucleotide-gated (HCN) channels and an auxiliary subunit of the channel named tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b). HCN channels regulate neuronal excitability in the mammalian hippocampus, and recent work has established that antagonizing HCN function rescues cognitive impairment caused by chronic stress. Here, we utilize a high-throughput virtual screen to find small molecules capable of disrupting the TRIP8b-HCN interaction. We found that the hit compound NUCC-0200590 disrupts the TRIP8b-HCN interaction in vitro and in vivo. These results provide a compelling strategy for developing new small molecules capable of disrupting the TRIP8b-HCN interaction.


Asunto(s)
Trastorno Depresivo Mayor , Animales , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Trastorno Depresivo Mayor/metabolismo , Hipocampo/metabolismo , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Mamíferos/metabolismo , Neuronas/metabolismo
3.
Biol Chem ; 404(4): 291-302, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36852869

RESUMEN

Tonic current through hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channels is influencing neuronal firing properties and channel function is strongly influenced by the brain-specific auxiliary subunit tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b). Since Kv1.2 channels and TRIP8b were also suggested to interact, we assessed brain Kv1.2 mRNA and protein expression as well as the reduction of K+ outward currents by Kv1.2-blocking compounds (Psora-4; tityustoxin-Kα, TsTX-Kα) in different brain areas of TRIP8b-deficient (TRIP8b -/- ) compared to wildtype (WT) mice. We found that transcription levels of Kv1.2 channels were not different between genotypes. Furthermore, Kv1.2 current amplitude was not affected upon co-expression with TRIP8b in oocytes. However, Kv1.2 immunofluorescence was stronger in dendritic areas of cortical and hippocampal neurons. Furthermore, the peak net outward current was increased and the inactivation of the Psora-4-sensitive current component was less pronounced in cortical neurons in TRIP8b -/- mice. In current clamp recordings, application of TsTX increased the excitability of thalamocortical (TC) neurons with increased number of elicited action potentials upon step depolarization. We conclude that TRIP8b may not preferentially influence the amplitude of current through Kv1.2 channels but seems to affect current inactivation and channel localization. In TRIP8b -/- a compensatory upregulation of other Kv channels was observed.


Asunto(s)
Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Neuronas , Ratones , Animales , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Neuronas/metabolismo , Hipocampo/metabolismo , Encéfalo/metabolismo , Oocitos
4.
Cereb Cortex ; 32(20): 4397-4421, 2022 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-35076711

RESUMEN

A consensus is yet to be reached regarding the exact prevalence of epileptic seizures or epilepsy in multiple sclerosis (MS). In addition, the underlying pathophysiological basis of the reciprocal interaction among neuroinflammation, demyelination, and epilepsy remains unclear. Therefore, a better understanding of cellular and network mechanisms linking these pathologies is needed. Cuprizone-induced general demyelination in rodents is a valuable model for studying MS pathologies. Here, we studied the relationship among epileptic activity, loss of myelin, and pro-inflammatory cytokines by inducing acute, generalized demyelination in a genetic mouse model of human absence epilepsy, C3H/HeJ mice. Both cellular and network mechanisms were studied using in vivo and in vitro electrophysiological techniques. We found that acute, generalized demyelination in C3H/HeJ mice resulted in a lower number of spike-wave discharges, increased cortical theta oscillations, and reduction of slow rhythmic intrathalamic burst activity. In addition, generalized demyelination resulted in a significant reduction in the amplitude of the hyperpolarization-activated inward current (Ih) in thalamic relay cells, which was accompanied by lower surface expression of hyperpolarization-activated, cyclic nucleotide-gated channels, and the phosphorylated form of TRIP8b (pS237-TRIP8b). We suggest that demyelination-related changes in thalamic Ih may be one of the factors defining the prevalence of seizures in MS.


Asunto(s)
Enfermedades Desmielinizantes , Epilepsia Tipo Ausencia , Animales , Corteza Cerebral/fisiología , Cuprizona/metabolismo , Cuprizona/toxicidad , Citocinas/metabolismo , Enfermedades Desmielinizantes/inducido químicamente , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Ratones , Ratones Endogámicos C3H , Neuronas/fisiología , Nucleótidos Cíclicos/metabolismo , Convulsiones , Tálamo/fisiología
5.
Int J Mol Sci ; 22(9)2021 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-33946275

RESUMEN

The tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b/PEX5R) is an interaction partner and auxiliary subunit of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, which are key for rhythm generation in the brain and in the heart. Since TRIP8b is expressed in central neurons but not in cardiomyocytes, the TRIP8b-HCN interaction has been studied intensely in the brain, but is deemed irrelevant in the cardiac conduction system. Still, to date, TRIP8b has not been studied in the intrinsic cardiac nervous system (ICNS), a neuronal network located within epicardial fat pads. In vitro electrophysiological studies revealed that TRIP8b-deficient mouse hearts exhibit increased atrial refractory and atrioventricular nodal refractory periods, compared to hearts of wild-type littermates. Meanwhile, heart rate, sino-nodal recovery time, and ventricular refractory period did not differ between genotypes. Trip8b mRNA was detected in the ICNS by quantitative polymerase chain reaction. RNAscope in situ hybridization confirmed Trip8b localization in neuronal somata and nerve fibers. Additionally, we found a very low amount of mRNAs in the sinus node and atrioventricular node, most likely attributable to the delicate fibers innervating the conduction system. In contrast, TRIP8b protein was not detectable. Our data suggest that TRIP8b in the ICNS may play a role in the modulation of atrial electrophysiology beyond HCN-mediated sino-nodal control of the heart.


Asunto(s)
Corazón/fisiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Proteínas de la Membrana/metabolismo , Peroxinas/metabolismo , Animales , Eliminación de Gen , Expresión Génica , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Peroxinas/genética , Mapas de Interacción de Proteínas , ARN Mensajero/genética
6.
J Biol Chem ; 294(43): 15743-15758, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31492750

RESUMEN

Temporal lobe epilepsy (TLE) is a prevalent neurological disorder with many patients experiencing poor seizure control with existing anti-epileptic drugs. Thus, novel insights into the mechanisms of epileptogenesis and identification of new drug targets can be transformative. Changes in ion channel function have been shown to play a role in generating the aberrant neuronal activity observed in TLE. Previous work demonstrates that hyperpolarization-activated cyclic nucleotide-gated (HCN) channels regulate neuronal excitability and are mislocalized within CA1 pyramidal cells in a rodent model of TLE. The subcellular distribution of HCN channels is regulated by an auxiliary subunit, tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b), and disruption of this interaction correlates with channel mislocalization. However, the molecular mechanisms responsible for HCN channel dysregulation in TLE are unclear. Here we investigated whether changes in TRIP8b phosphorylation are sufficient to alter HCN channel function. We identified a phosphorylation site at residue Ser237 of TRIP8b that enhances binding to HCN channels and influences channel gating by altering the affinity of TRIP8b for the HCN cytoplasmic domain. Using a phosphospecific antibody, we demonstrate that TRIP8b phosphorylated at Ser237 is enriched in CA1 distal dendrites and that phosphorylation is reduced in the kainic acid model of TLE. Overall, our findings indicate that the TRIP8b-HCN interaction can be modulated by changes in phosphorylation and suggest that loss of TRIP8b phosphorylation may affect HCN channel properties during epileptogenesis. These results highlight the potential of drugs targeting posttranslational modifications to restore TRIP8b phosphorylation to reduce excitability in TLE.


Asunto(s)
Epilepsia del Lóbulo Temporal/metabolismo , Proteínas de la Membrana/metabolismo , Peroxinas/metabolismo , Subunidades de Proteína/metabolismo , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Dendritas/metabolismo , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Activación del Canal Iónico , Ácido Kaínico , Proteínas de la Membrana/química , Ratones Endogámicos C57BL , Peroxinas/química , Fosforilación , Fosfoserina/metabolismo , Subunidades de Proteína/química , Ratas Sprague-Dawley , Reproducibilidad de los Resultados
7.
Cerebellum ; 18(6): 1036-1063, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31124049

RESUMEN

Tremor is the most common movement disorder; however, we are just beginning to understand the brain circuitry that generates tremor. Various neuroimaging, neuropathological, and physiological studies in human tremor disorders have been performed to further our knowledge of tremor. But, the causal relationship between these observations and tremor is usually difficult to establish and detailed mechanisms are not sufficiently studied. To overcome these obstacles, animal models can provide an important means to look into human tremor disorders. In this manuscript, we will discuss the use of different species of animals (mice, rats, fruit flies, pigs, and monkeys) to model human tremor disorders. Several ways to manipulate the brain circuitry and physiology in these animal models (pharmacology, genetics, and lesioning) will also be discussed. Finally, we will discuss how these animal models can help us to gain knowledge of the pathophysiology of human tremor disorders, which could serve as a platform towards developing novel therapies for tremor.


Asunto(s)
Encéfalo/diagnóstico por imagen , Consenso , Testimonio de Experto , Modelos Animales , Red Nerviosa/diagnóstico por imagen , Temblor/diagnóstico por imagen , Animales , Encéfalo/fisiopatología , Drosophila , Testimonio de Experto/normas , Haplorrinos , Ratones , Red Nerviosa/fisiopatología , Ratas , Porcinos , Temblor/fisiopatología
8.
J Biol Chem ; 292(43): 17718-17730, 2017 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-28887304

RESUMEN

Tetratricopeptide repeat (TPR) domains are ubiquitous structural motifs that mediate protein-protein interactions. For example, the TPR domains in the peroxisomal import receptor PEX5 enable binding to a range of type 1 peroxisomal targeting signal motifs. A homolog of PEX5, tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b), binds to and functions as an auxiliary subunit of hyperpolarization-activated cyclic nucleotide (HCN)-gated channels. Given the similarity between TRIP8b and PEX5, this difference in function raises the question of what mechanism accounts for their binding specificity. In this report, we found that the cyclic nucleotide-binding domain and the C terminus of the HCN channel are critical for conferring specificity to TRIP8b binding. We show that TRIP8b binds the HCN cyclic nucleotide-binding domain through a 37-residue domain and the HCN C terminus through the TPR domains. Using a combination of fluorescence polarization- and co-immunoprecipitation-based assays, we establish that binding at either site increases affinity at the other. Thus, allosteric coupling of the TRIP8b TPR domains both promotes binding to HCN channels and limits binding to type 1 peroxisomal targeting signal substrates. These results raise the possibility that other TPR domains may be similarly influenced by allosteric mechanisms as a general feature of protein-protein interactions.


Asunto(s)
Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Subunidades de Proteína/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Regulación Alostérica/fisiología , Sitios de Unión , Células HEK293 , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Subunidades de Proteína/genética , Receptores Citoplasmáticos y Nucleares/genética
9.
J Neurochem ; 146(6): 753-766, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29953635

RESUMEN

Active coping is an adaptive stress response that improves outcomes in medical and neuropsychiatric diseases. To date, most research into coping style has focused on neurotransmitter activity and little is known about the intrinsic excitability of neurons in the associated brain regions that facilitate coping. Previous studies have shown that HCN channels regulate neuronal excitability in pyramidal cells and that HCN channel current (Ih ) in the CA1 area increases with chronic mild stress. Reduction of Ih in the CA1 area leads to antidepressant-like behavior, and this region has been implicated in the regulation of coping style. We hypothesized that the antidepressant-like behavior achieved with CA1 knockdown of Ih is accompanied by increases in active coping. In this report, we found that global loss of TRIP8b, a necessary subunit for proper HCN channel localization in pyramidal cells, led to active coping behavior in numerous assays specific to coping style. We next employed a viral strategy using a dominant negative TRIP8b isoform to alter coping behavior by reducing HCN channel expression. This approach led to a robust reduction in Ih in CA1 pyramidal neurons and an increase in active coping. Together, these results establish that changes in HCN channel function in CA1 influences coping style.


Asunto(s)
Adaptación Psicológica/fisiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Proteínas de la Membrana/metabolismo , Peroxinas/metabolismo , Animales , Reacción de Prevención/fisiología , Depresión/fisiopatología , Modelos Animales de Enfermedad , Conducta Exploratoria , Hipocampo/citología , Hipocampo/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/ultraestructura , Masculino , Aprendizaje por Laberinto , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica , Peroxinas/genética , Células Piramidales/metabolismo , Natación/psicología
10.
Neurobiol Learn Mem ; 154: 141-157, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29906573

RESUMEN

Voltage-gated ion channels are critical for neuronal integration. Some of these channels, however, are misregulated in several neurological disorders, causing both gain- and loss-of-function channelopathies in neurons. Using several transgenic mouse models of Alzheimer's disease (AD), we find that sub-threshold voltage signals strongly influenced by hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels progressively deteriorate over chronological aging in hippocampal CA1 pyramidal neurons. The degraded signaling via HCN channels in the transgenic mice is accompanied by an age-related global loss of their non-uniform dendritic expression. Both the aberrant signaling via HCN channels and their mislocalization could be restored using a variety of pharmacological agents that target the endoplasmic reticulum (ER). Our rescue of the HCN channelopathy helps provide molecular details into the favorable outcomes of ER-targeting drugs on the pathogenesis and synaptic/cognitive deficits in AD mouse models, and implies that they might have beneficial effects on neurological disorders linked to HCN channelopathies.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Región CA1 Hipocampal/fisiología , Canalopatías/fisiopatología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/fisiología , Plasticidad Neuronal , Células Piramidales/fisiología , Potenciales de Acción , Envejecimiento , Animales , Región CA1 Hipocampal/ultraestructura , Modelos Animales de Enfermedad , Retículo Endoplásmico/fisiología , Femenino , Masculino , Ratones Transgénicos , Células Piramidales/ultraestructura
11.
Neurobiol Dis ; 85: 81-92, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26459112

RESUMEN

Absence seizures occur in several types of human epilepsy and result from widespread, synchronous feedback between the cortex and thalamus that produces brief episodes of loss of consciousness. Genetic rodent models have been invaluable for investigating the pathophysiological basis of these seizures. Here, we identify tetratricopeptide-containing Rab8b-interacting protein (TRIP8b) knockout mice as a new model of absence epilepsy, featuring spontaneous spike-wave discharges on electroencephalography (EEG) that are the electrographic hallmark of absence seizures. TRIP8b is an auxiliary subunit of the hyperpolarization-activated cyclic-nucleotide-gated (HCN) channels, which have previously been implicated in the pathogenesis of absence seizures. In contrast to mice lacking the pore-forming HCN channel subunit HCN2, TRIP8b knockout mice exhibited normal cardiac and motor function and a less severe seizure phenotype. Evaluating the circuit that underlies absence seizures, we found that TRIP8b knockout mice had significantly reduced HCN channel expression and function in thalamic-projecting cortical layer 5b neurons and thalamic relay neurons, but preserved function in inhibitory neurons of the reticular thalamic nucleus. Our results expand the known roles of TRIP8b and provide new insight into the region-specific functions of TRIP8b and HCN channels in constraining cortico-thalamo-cortical excitability.


Asunto(s)
Corteza Cerebral/fisiopatología , Epilepsia Tipo Ausencia/fisiopatología , Proteínas de la Membrana/deficiencia , Neuronas/fisiología , Tálamo/fisiopatología , Animales , Western Blotting , Modelos Animales de Enfermedad , Electrocardiografía , Electrocorticografía , Electrodos Implantados , Epilepsia Tipo Ausencia/genética , Inmunohistoquímica , Masculino , Potenciales de la Membrana/fisiología , Proteínas de la Membrana/genética , Ratones Noqueados , Actividad Motora/fisiología , Técnicas de Placa-Clamp , Peroxinas , Prueba de Desempeño de Rotación con Aceleración Constante , Eliminación de Secuencia , Técnicas de Cultivo de Tejidos
12.
FASEB J ; 28(5): 2177-90, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24451387

RESUMEN

HCN channels are important regulators of neuronal excitability. The proper function of these channels is governed by various mechanisms, including post-translational modifications of channel subunits. Here, we provide evidence that ubiquitination via a ubiquitin ligase, neuronal precursor cell expressed developmentally downregulated (Nedd)-4-2, is involved in the regulation of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. We identified a PY motif (L/PPxY), the characteristic binding motif for Nedd4-2 in the C terminus of the HCN1 subunit, and showed that HCN1 and Nedd4-2 interacted both in vivo (rat hippocampus, neocortex, and cerebellum) and in vitro [human embryonic kidney 293 (HEK293) cells], resulting in increased HCN1 ubiquitination. Elimination of the PY motif reduced, but did not abolish, Nedd4-2 binding, which further involved a stretch of ∼100 aa downstream in the HCN1 C terminus. Coexpression of Nedd4-2 and HCN1 drastically reduced the HCN1-mediated h-current amplitude (85-92%) in Xenopus laevis oocytes and reduced surface expression (34%) of HCN1 channels in HEK293 cells, thereby opposing effects of tetratricopeptide repeat-containing Rab8b interacting protein (TRIP8b)-(1a-4), an auxiliary subunit that promotes HCN1 surface expression. Regulation may further include N-glycosylation of HCN1 channels, which is significantly enhanced by TRIP8b(1a-4), but may be reduced by Nedd4-2. Taken together, our data indicate that Nedd4-2 plays an important role in the regulation of HCN1 trafficking and may compete with TRIP8b(1a-4) in this process.


Asunto(s)
Membrana Celular/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/fisiología , Regulación de la Expresión Génica , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Secuencias de Aminoácidos , Animales , Encéfalo/metabolismo , Regulación hacia Abajo , Electrofisiología , Femenino , Glicosilación , Células HEK293 , Humanos , Ubiquitina-Proteína Ligasas Nedd4 , Oocitos/citología , Estructura Terciaria de Proteína , Ratas , Ratas Wistar , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas de Xenopus , Xenopus laevis
13.
Proc Natl Acad Sci U S A ; 109(20): 7899-904, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22550182

RESUMEN

Ion channels operate in intact tissues as part of large macromolecular complexes that can include cytoskeletal proteins, scaffolding proteins, signaling molecules, and a litany of other molecules. The proteins that make up these complexes can influence the trafficking, localization, and biophysical properties of the channel. TRIP8b (tetratricopetide repeat-containing Rab8b-interacting protein) is a recently discovered accessory subunit of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels that contributes to the substantial dendritic localization of HCN channels in many types of neurons. TRIP8b interacts with the carboxyl-terminal region of HCN channels and regulates their cell-surface expression level and cyclic nucleotide dependence. Here we examine the molecular determinants of TRIP8b binding to HCN2 channels. Using a single-molecule fluorescence bleaching method, we found that TRIP8b and HCN2 form an obligate 4:4 complex in intact channels. Fluorescence-detection size-exclusion chromatography and fluorescence anisotropy allowed us to confirm that two different domains in the carboxyl-terminal portion of TRIP8b--the tetratricopepide repeat region and the TRIP8b conserved region--interact with two different regions of the HCN carboxyl-terminal region: the carboxyl-terminal three amino acids (SNL) and the cyclic nucleotide-binding domain, respectively. And finally, using X-ray crystallography, we determined the atomic structure of the tetratricopepide region of TRIP8b in complex with a peptide of the carboxy-terminus of HCN2. Together, these experiments begin to uncover the mechanism for TRIP8b binding and regulation of HCN channels.


Asunto(s)
Canales Iónicos/metabolismo , Modelos Moleculares , Complejos Multiproteicos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Cromatografía en Gel , Cristalografía , Polarización de Fluorescencia , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Canales Iónicos/genética , Ratones , Microscopía Fluorescente , Proteínas del Tejido Nervioso/genética , Oocitos , Técnicas de Placa-Clamp , Canales de Potasio , Unión Proteica , Difracción de Rayos X , Xenopus
14.
Front Cell Neurosci ; 18: 1321682, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38469353

RESUMEN

Mature oligodendrocytes (OLG) are the myelin-forming cells of the central nervous system. Recent work has shown a dynamic role for these cells in the plasticity of neural circuits, leading to a renewed interest in voltage-sensitive currents in OLG. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels and their respective current (Ih) were recently identified in mature OLG and shown to play a role in regulating myelin length. Here we provide a biochemical and electrophysiological characterization of HCN channels in cells of the oligodendrocyte lineage. We observed that mice with a nonsense mutation in the Hcn2 gene (Hcn2ap/ap) have less white matter than their wild type counterparts with fewer OLG and fewer oligodendrocyte progenitor cells (OPCs). Hcn2ap/ap mice have severe motor impairments, although these deficits were not observed in mice with HCN2 conditionally eliminated only in oligodendrocytes (Cnpcre/+; Hcn2F/F). However, Cnpcre/+; Hcn2F/F mice develop motor impairments more rapidly in response to experimental autoimmune encephalomyelitis (EAE). We conclude that HCN2 channels in OLG may play a role in regulating metabolism.

15.
J Neurosci ; 32(42): 14835-48, 2012 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-23077068

RESUMEN

Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are subthreshold activated voltage-gated ion channels. In the cortex, these channels are predominantly expressed in dendrites where they significantly modify dendritic intrinsic excitability as well synaptic potential shapes and integration. HCN channel trafficking to dendrites is regulated by the protein, TRIP8b. Additionally, altered TRIP8b expression may be one mechanism underlying seizure-induced dendritic HCN channel plasticity. HCN channels, though, are also located in certain mature cortical synaptic terminals, where they play a vital role in modulating synaptic transmission. In this study, using electrophysiological recordings as well as electron microscopy we show that presynaptic, but not dendritic, cortical HCN channel expression and function is comparable in adult TRIP8b-null mice and wild-type littermates. We further investigated whether presynaptic HCN channels undergo seizure-dependent plasticity. We found that, like dendritic channels, wild-type presynaptic HCN channel function was persistently decreased following induction of kainic acid-induced seizures. Since TRIP8b does not affect presynaptic HCN subunit trafficking, seizure-dependent plasticity of these cortical HCN channels is not conditional upon TRIP8b. Our results, thus, suggest that the molecular mechanisms underlying HCN subunit targeting, expression and plasticity in adult neurons is compartment selective, providing a means by which pre- and postsynaptic processes that are critically dependent upon HCN channel function may be distinctly influenced.


Asunto(s)
Corteza Cerebral/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/fisiología , Proteínas de la Membrana/metabolismo , Plasticidad Neuronal/fisiología , Canales de Potasio/metabolismo , Canales de Potasio/fisiología , Terminales Presinápticos/metabolismo , Animales , Corteza Cerebral/ultraestructura , Canales Catiónicos Regulados por Nucleótidos Cíclicos/deficiencia , Potenciales Postsinápticos Excitadores/fisiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Peroxinas , Canales de Potasio/deficiencia , Transporte de Proteínas/fisiología , Distribución Aleatoria
16.
J Neurosci ; 32(11): 3736-47, 2012 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-22423094

RESUMEN

The dorsoventral and developmental gradients of entorhinal layer II cell grid properties correlate with their resonance properties and with their hyperpolarization-activated cyclic nucleotide-gated (HCN) ion channel current characteristics. We investigated whether such correlation existed in rat hippocampal CA1 pyramidal cells, where place fields also show spatial and temporal gradients. Resonance was absent during the first postnatal week, and emerged during the second week. Resonance was stronger in dorsal than ventral cells, in accord with HCN current properties. Resonance responded to cAMP in ventral but not in dorsal cells. The dorsoventral distribution of HCN1 and HCN2 subunits and of the auxiliary protein tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b) could account for these differences between dorsal and ventral cells. The analogous distribution of the intrinsic properties of entorhinal stellate and hippocampal cells suggests the existence of general rules of organization among structures that process complementary features of the environment.


Asunto(s)
Región CA1 Hipocampal/citología , Región CA1 Hipocampal/crecimiento & desarrollo , Células Piramidales/citología , Células Piramidales/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Masculino , Técnicas de Cultivo de Órganos , Ratas , Ratas Wistar
17.
J Biol Chem ; 287(21): 17656-17661, 2012 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-22511771

RESUMEN

The dorsal and ventral regions of the hippocampus perform different functions. Whether the integrative properties of hippocampal cells reflect this heterogeneity is unknown. We focused on dendrites where most synaptic input integration takes place. We report enhanced backpropagation and theta resonance and decreased summation of synaptic inputs in ventral versus dorsal CA1 pyramidal cell distal dendrites. Transcriptional Kv4.2 down-regulation and post-transcriptional hyperpolarization-activated cyclic AMP-gated channel (HCN1/2) up-regulation may underlie these differences, respectively. Our results reveal differential dendritic integrative properties along the dorso-ventral axis, reflecting diverse computational needs.


Asunto(s)
Canales Catiónicos Regulados por Nucleótidos Cíclicos/biosíntesis , Dendritas/metabolismo , Regulación hacia Abajo/fisiología , Canales Iónicos/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Canales de Potasio/biosíntesis , Células Piramidales/metabolismo , Canales de Potasio Shal/biosíntesis , Regulación hacia Arriba/fisiología , Animales , Dendritas/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Especificidad de Órganos , Células Piramidales/citología , Ratas , Transcripción Genética/fisiología
18.
J Neurophysiol ; 110(10): 2350-7, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23966674

RESUMEN

Hyperpolarization-activated cyclic nucleotide-gated nonselective cation channels (HCN or h-channels) are important regulators of neuronal physiology contributing to passive membrane properties, such as resting membrane potential and input resistance (R(N)), and to intrinsic oscillatory activity and synaptic integration. The correct membrane targeting of h-channels is regulated in part by the auxiliary h-channel protein TRIP8b. The genetic deletion of TRIP8b results in a loss of functional h-channels, which affects the postsynaptic integrative properties of neurons. We investigated the impact of TRIP8b deletion on long-term potentiation (LTP) at the two major excitatory inputs to CA1 pyramidal neurons: Schaffer collateral (SC) and perforant path (PP). We found that SC LTP was not significantly different between neurons from wild-type and TRIP8b-knockout mice. There was, however, significantly more short-term potentiation in knockout neurons. We also found that the persistent increase in h-current (I(h)) that normally occurs after LTP induction was absent in knockout neurons. The lack of I(h) plasticity was not restricted to activity-dependent induction, because the depletion of intracellular calcium stores also failed to produce the expected increase in I(h). Interestingly, pairing of SC and PP inputs resulted in a form of LTP in knockout neurons that did not occur in wild-type neurons. These results suggest that the physiological impact of TRIP8b deletion is not restricted to the integrative properties of neurons but also includes both synaptic and intrinsic plasticity.


Asunto(s)
Región CA1 Hipocampal/fisiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Potenciación a Largo Plazo , Proteínas de la Membrana/metabolismo , Células Piramidales/fisiología , Animales , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vía Perforante/fisiología , Peroxinas , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo
19.
J Neurophysiol ; 109(7): 1940-53, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23324324

RESUMEN

The rodent hippocampus can be divided into dorsal (DHC) and ventral (VHC) domains on the basis of behavioral, anatomical, and biochemical differences. Recently, we reported that CA1 pyramidal neurons from the VHC were intrinsically more excitable than DHC neurons, but the specific ionic conductances contributing to this difference were not determined. Here we investigated the hyperpolarization-activated current (I(h)) and the expression of HCN1 and HCN2 channel subunits in CA1 pyramidal neurons from the DHC and VHC. Measurement of Ih with cell-attached patches revealed a significant depolarizing shift in the V(1/2) of activation for dendritic h-channels in VHC neurons (but not DHC neurons), and ultrastructural immunolocalization of HCN1 and HCN2 channels revealed a significantly larger HCN1-to-HCN2 ratio for VHC neurons (but not DHC neurons). These observations suggest that a shift in the expression of HCN1 and HCN2 channels drives functional changes in I(h) for VHC neurons (but not DHC neurons) and could thereby significantly alter the capacity for dendritic integration of these neurons.


Asunto(s)
Región CA1 Hipocampal/fisiología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Activación del Canal Iónico , Canales Iónicos/metabolismo , Canales de Potasio/metabolismo , Células Piramidales/fisiología , Potenciales de Acción , Animales , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Expresión Génica , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Canales Iónicos/genética , Especificidad de Órganos , Canales de Potasio/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Células Piramidales/metabolismo , Ratas , Ratas Sprague-Dawley
20.
J Neurosci ; 31(20): 7424-40, 2011 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-21593326

RESUMEN

Output properties of neurons are greatly shaped by voltage-gated ion channels, whose biophysical properties and localization within axodendritic compartments serve to significantly transform the original input. The hyperpolarization-activated current, I(h), is mediated by hyperpolarization-activated cyclic nucleotide-gated (HCN) channels and plays a fundamental role in influencing neuronal excitability by regulating both membrane potential and input resistance. In neurons such as cortical and hippocampal pyramidal neurons, the subcellular localization of HCN channels plays a critical functional role, yet mechanisms controlling HCN channel trafficking are not fully understood. Because ion channel function and localization are often influenced by interacting proteins, we generated a knock-out mouse lacking the HCN channel auxiliary subunit, tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b). Eliminating expression of TRIP8b dramatically reduced I(h) expression in hippocampal pyramidal neurons. Loss of I(h)-dependent membrane voltage properties was attributable to reduction of HCN channels on the neuronal surface, and there was a striking disruption of the normal expression pattern of HCN channels in pyramidal neuron dendrites. In heterologous cells and neurons, absence of TRIP8b increased HCN subunit targeting to and degradation by lysosomes. Mice lacking TRIP8b demonstrated motor learning deficits and enhanced resistance to multiple tasks of behavioral despair with high predictive validity for antidepressant efficacy. We observed similar resistance to behavioral despair in distinct mutant mice lacking HCN1 or HCN2. These data demonstrate that interaction with the auxiliary subunit TRIP8b is a major mechanism underlying proper expression of HCN channels and I(h) in vivo, and suggest that targeting I(h) may provide a novel approach to treatment of depression.


Asunto(s)
Canales Catiónicos Regulados por Nucleótidos Cíclicos/deficiencia , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Depresión/genética , Eliminación de Gen , Hipocampo/fisiología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Canales de Potasio/deficiencia , Canales de Potasio/metabolismo , Subunidades de Proteína/metabolismo , Animales , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Depresión/psicología , Depresión/terapia , Terapia Genética/métodos , Hipocampo/química , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peroxinas , Canales de Potasio/genética , Subunidades de Proteína/deficiencia , Subunidades de Proteína/fisiología , Transporte de Proteínas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA