Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 138(6): 1222-35, 2009 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-19766573

RESUMEN

Plasticity related gene-1 (PRG-1) is a brain-specific membrane protein related to lipid phosphate phosphatases, which acts in the hippocampus specifically at the excitatory synapse terminating on glutamatergic neurons. Deletion of prg-1 in mice leads to epileptic seizures and augmentation of EPSCs, but not IPSCs. In utero electroporation of PRG-1 into deficient animals revealed that PRG-1 modulates excitation at the synaptic junction. Mutation of the extracellular domain of PRG-1 crucial for its interaction with lysophosphatidic acid (LPA) abolished the ability to prevent hyperexcitability. As LPA application in vitro induced hyperexcitability in wild-type but not in LPA(2) receptor-deficient animals, and uptake of phospholipids is reduced in PRG-1-deficient neurons, we assessed PRG-1/LPA(2) receptor-deficient animals, and found that the pathophysiology observed in the PRG-1-deficient mice was fully reverted. Thus, we propose PRG-1 as an important player in the modulatory control of hippocampal excitability dependent on presynaptic LPA(2) receptor signaling.


Asunto(s)
Proteoglicanos/metabolismo , Sinapsis/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Electroencefalografía , Hipocampo/química , Hipocampo/citología , Hipocampo/metabolismo , Lisofosfolípidos/metabolismo , Ratones , Ratones Noqueados , Proteoglicanos/análisis , Proteoglicanos/genética , Receptores AMPA/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Transducción de Señal , Proteínas de Transporte Vesicular/análisis , Proteínas de Transporte Vesicular/genética
2.
Brain Behav Immun ; 101: 153-164, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34998939

RESUMEN

Interferon-γ (IFN-γ), an important mediator of the antiviral immune response, can also act as a neuromodulator. CNS IFN-γ levels rise acutely in response to infection and therapeutically applied IFN-γ provokes CNS related side effects. Moreover, IFN-γ plays a key role in neurophysiological processes and a variety of chronic neurological and neuropsychiatric conditions. To close the gap between basic research, behavioral implications and clinical applicability, knowledge of the mechanism behind IFN-γ related changes in brain function is crucial. Here, we studied the underlying mechanism of acutely augmented neocortical inhibition by IFN-γ (1.000 IU ml-1) in layer 5 pyramidal neurons of male Wistar rats. We demonstrate postsynaptic mediation of IFN-γ augmented inhibition by pressure application of GABA and analysis of paired pulse ratios. IFN-γ increases membrane presence of GABAAR γ2, as quantified by cell surface biotinylation and functional synaptic GABAAR number, as determined by peak-scaled non-stationary noise analysis. The increase in functional receptor number was comparable to the increase in underlying miniature inhibitory postsynaptic current (mIPSC) amplitudes. Blockage of putative intracellular mediators, namely phosphoinositide 3-kinase and protein kinase C (PKC) by Wortmannin and Calphostin C, respectively, revealed PKC-dependency of the pro-inhibitory IFN-γ effect. This was corroborated by increased serine phosphorylation of P-serine PKC motifs on GABAAR γ2 upon IFN-γ application. GABAAR single channel conductance, intracellular chloride levels and GABAAR driving force are unlikely to contribute to the effect, as shown by single channel recordings and chloride imaging. The effect of IFN-γ on mIPSC amplitudes was similar in female and male rats, suggesting a gender-independent mechanism of action. Collectively, these results indicate a novel mechanism for the regulation of inhibition by IFN-γ, which could impact on neocortical function and therewith behavior.


Asunto(s)
Neocórtex , Receptores de GABA-A , Animales , Cloruros/metabolismo , Femenino , Interferón gamma/metabolismo , Interferón gamma/farmacología , Masculino , Neocórtex/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteína Quinasa C/metabolismo , Ratas , Ratas Wistar , Receptores de GABA-A/metabolismo , Serina/metabolismo , Ácido gamma-Aminobutírico/metabolismo
3.
J Neuroinflammation ; 17(1): 69, 2020 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-32087716

RESUMEN

BACKGROUND: Interferon-γ (IFN-γ, a type II IFN) is present in the central nervous system (CNS) under various conditions. Evidence is emerging that, in addition to its immunological role, IFN-γ modulates neuronal morphology, function, and development in several brain regions. Previously, we have shown that raising levels of IFN-ß (a type I IFN) lead to increased neuronal excitability of neocortical layer 5 pyramidal neurons. Because of shared non-canonical signaling pathways of both cytokines, we hypothesized a similar neocortical role of acutely applied IFN-γ. METHODS: We used semi-quantitative RT-PCR, immunoblotting, and immunohistochemistry to analyze neuronal expression of IFN-γ receptors and performed whole-cell patch-clamp recordings in layer 5 pyramidal neurons to investigate sub- and suprathreshold excitability, properties of hyperpolarization-activated cyclic nucleotide-gated current (Ih), and inhibitory neurotransmission under the influence of acutely applied IFN-γ. RESULTS: We show that IFN-γ receptors are present in the membrane of rat's neocortical layer 5 pyramidal neurons. As expected from this and the putative overlap in IFN type I and II alternative signaling pathways, IFN-γ diminished Ih, mirroring the effect of type I IFNs, suggesting a likewise activation of protein kinase C (PKC). In contrast, IFN-γ did neither alter subthreshold nor suprathreshold neuronal excitability, pointing to augmented inhibitory transmission by IFN-γ. Indeed, IFN-γ increased electrically evoked inhibitory postsynaptic currents (IPSCs) on neocortical layer 5 pyramidal neurons. Furthermore, amplitudes of spontaneous IPSCs and miniature IPSCs were elevated by IFN-γ, whereas their frequency remained unchanged. CONCLUSIONS: The expression of IFN-γ receptors on layer 5 neocortical pyramidal neurons together with the acute augmentation of inhibition in the neocortex by direct application of IFN-γ highlights an additional interaction between the CNS and immune system. Our results strengthen our understanding of the role of IFN-γ in neocortical neurotransmission and emphasize its impact beyond its immunological properties, particularly in the pathogenesis of neuropsychiatric disorders.


Asunto(s)
Interferón gamma/metabolismo , Neocórtex/metabolismo , Neuroinmunomodulación/fisiología , Células Piramidales/metabolismo , Receptores de Interferón/metabolismo , Animales , Interferón gamma/farmacología , Masculino , Neocórtex/efectos de los fármacos , Neocórtex/inmunología , Células Piramidales/efectos de los fármacos , Células Piramidales/inmunología , Ratas , Ratas Wistar
4.
Neurobiol Dis ; 129: 130-143, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31102767

RESUMEN

Congenital microcephaly is highly associated with intellectual disability. Features of autosomal recessive primary microcephaly subtype 3 (MCPH3) also include hyperactivity and seizures. The disease is caused by biallelic mutations in the Cyclin-dependent kinase 5 regulatory subunit-associated protein 2 gene CDK5RAP2. In the mouse, Cdk5rap2 mutations similar to the human condition result in reduced brain size and a strikingly thin neocortex already at early stages of neurogenesis that persists through adulthood. The microcephaly phenotype in MCPH arises from a neural stem cell proliferation defect. Here, we report a novel role for Cdk5rap2 in the regulation of dendritic development and synaptogenesis of neocortical layer 2/3 pyramidal neurons. Cdk5rap2-deficient murine neurons show poorly branched dendritic arbors and an increased density of immature thin spines and glutamatergic synapses in vivo. Moreover, the excitatory drive is enhanced in ex vivo brain slice preparations of Cdk5rap2 mutant mice. Concurrently, we show that pyramidal neurons receive fewer inhibitory inputs. Together, these findings point towards a shift in the excitation - inhibition balance towards excitation in Cdk5rap2 mutant mice. Thus, MCPH3 is associated not only with a neural progenitor proliferation defect but also with altered function of postmitotic neurons and hence with altered connectivity.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Microcefalia/fisiopatología , Neocórtex/fisiopatología , Vías Nerviosas/fisiopatología , Neurogénesis/fisiología , Animales , Proteínas de Ciclo Celular/genética , Diferenciación Celular/fisiología , Ratones , Ratones Mutantes , Microcefalia/genética , Microcefalia/metabolismo , Mutación , Neocórtex/metabolismo , Vías Nerviosas/metabolismo , Células Piramidales/metabolismo , Células Piramidales/patología , Transmisión Sináptica/fisiología
5.
Cereb Cortex ; 27(1): 131-145, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27909001

RESUMEN

Altered synaptic bioactive lipid signaling has been recently shown to augment neuronal excitation in the hippocampus of adult animals by activation of presynaptic LPA2-receptors leading to increased presynaptic glutamate release. Here, we show that this results in higher postsynaptic Ca2+ levels and in premature onset of spontaneous neuronal activity in the developing entorhinal cortex. Interestingly, increased synchronized neuronal activity led to reduced axon growth velocity of entorhinal neurons which project via the perforant path to the hippocampus. This was due to Ca2+-dependent molecular signaling to the axon affecting stabilization of the actin cytoskeleton. The spontaneous activity affected the entire entorhinal cortical network and thus led to reduced overall axon fiber numbers in the mature perforant path that is known to be important for specific memory functions. Our data show that precise regulation of early cortical activity by bioactive lipids is of critical importance for proper circuit formation.


Asunto(s)
Axones/fisiología , Señalización del Calcio/fisiología , Ácido Glutámico/metabolismo , Redes y Vías Metabólicas/fisiología , Proyección Neuronal/fisiología , Fosfolípidos/metabolismo , Transmisión Sináptica/fisiología , Animales , Axones/ultraestructura , Calcio/metabolismo , Células Cultivadas , Ratones
6.
Cell Physiol Biochem ; 38(6): 2079-93, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27184742

RESUMEN

BACKGROUND/AIMS: Cationic currents (Ih) through the fast activating and relatively cAMP insensitive subtype of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, HCN1, are limited by cytosolic factors in mammalian cells. This cytosolic HCN1 break is boosted by changes in membrane voltage that are not characterized on a biophysical level, yet. METHODS: We overexpressed rat (r)HCN1 in human embryonic kidney cells (HEK293) and recorded pharmacologically isolated Ih in cell-attached or whole-cell mode of the patch-clamp technique. RESULTS: Recurring activation of rHCN1 reduced and slowed Ih in intact HEK293 cells (cell-attached mode). On the contrary, sustained disruption of the intracellular content (whole-cell mode) ceased activity dependence and partially enables voltage dependent hysteresis. The activity induced Ih attenuation in intact cells was independent of the main external cation, depended on the number of previous forced activations and was - at least in part - due to a shift in the voltage dependence of activation towards hyperpolarization as estimated by an adapted tail current analysis. Intracellular elevation of cAMP could not reverse the changes in Ih. CONCLUSION: Reduction of rHCN1 mediated Ih is use dependent and may involve the coupling of voltage sensor and pore.


Asunto(s)
Células HEK293/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Canales de Potasio/metabolismo , Animales , Cationes/metabolismo , AMP Cíclico/metabolismo , Células HEK293/citología , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Técnicas de Placa-Clamp , Canales de Potasio/genética , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sodio/metabolismo , Regulación hacia Arriba
7.
J Biol Chem ; 289(36): 24956-70, 2014 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-25074937

RESUMEN

The transmembrane protein plasticity-related genes 3 and 5 (PRG3 and PRG5) increase filopodial formation in various cell lines, independently of Cdc42. However, information on the effects of PRG5 during neuronal development is sparse. Here, we present several lines of evidence for the involvement of PRG5 in the genesis and stabilization of dendritic spines. First, PRG5 was strongly expressed during mouse brain development from embryonic day 14 (E14), peaked around the time of birth, and remained stable at least until early adult stages (i.e. P30). Second, on a subcellular level, PRG5 expression shifted from an equal distribution along all neurites toward accumulation only along dendrites during hippocampal development in vitro. Third, overexpression of PRG5 in immature hippocampal neurons induced formation of spine-like structures ahead of time. Proper amino acid sequences in the extracellular domains (D1 to D3) of PRG5 were a prerequisite for trafficking and induction of spine-like structures, as shown by mutation analysis. Fourth, at stages when spines are present, knockdown of PRG5 reduced the number but not the length of protrusions. This was accompanied by a decrease in the number of excitatory synapses and, consequently, by a reduction of miniature excitatory postsynaptic current frequencies, although miniature excitatory postsynaptic current amplitudes remained similar. In turn, overexpressing PRG5 in mature neurons not only increased Homer-positive spine numbers but also augmented spine head diameters. Mechanistically, PRG5 interacts with phosphorylated phosphatidylinositols, phospholipids involved in dendritic spine formation by different lipid-protein assays. Taken together, our data propose that PRG5 promotes spine formation.


Asunto(s)
Espinas Dendríticas/genética , Hipocampo/metabolismo , Proteínas de la Membrana/genética , Neuronas/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Animales , Astrocitos/metabolismo , Western Blotting , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Células Cultivadas , Espinas Dendríticas/metabolismo , Potenciales Postsinápticos Excitadores/genética , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Hipocampo/embriología , Hipocampo/crecimiento & desarrollo , Humanos , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Microglía/metabolismo , Microscopía Confocal , Neuronas/fisiología , Monoéster Fosfórico Hidrolasas/metabolismo , Embarazo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transmisión Sináptica/genética , Transmisión Sináptica/fisiología
8.
Cereb Cortex ; 24(1): 199-210, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23042740

RESUMEN

Central nervous system (CNS) inflammation involves the generation of inducible cytokines such as interferons (IFNs) and alterations in brain activity, yet the interplay of both is not well understood. Here, we show that in vivo elevation of IFNs by viral brain infection reduced hyperpolarization-activated currents (Ih) in cortical pyramidal neurons. In rodent brain slices directly exposed to type I IFNs, the hyperpolarization-activated cyclic nucleotide (HCN)-gated channel subunit HCN1 was specifically affected. The effect required an intact type I receptor (IFNAR) signaling cascade. Consistent with Ih inhibition, IFNs hyperpolarized the resting membrane potential, shifted the resonance frequency, and increased the membrane impedance. In vivo application of IFN-ß to the rat and to the mouse cerebral cortex reduced the power of higher frequencies in the cortical electroencephalographic activity only in the presence of HCN1. In summary, these findings identify HCN1 channels as a novel neural target for type I IFNs providing the possibility to tune neural responses during the complex event of a CNS inflammation.


Asunto(s)
Corteza Cerebral/fisiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/fisiología , Interferón Tipo I/fisiología , Neuronas/fisiología , Canales de Potasio/fisiología , Animales , Western Blotting , Corteza Cerebral/citología , Simulación por Computador , Citocinas/fisiología , Electroencefalografía , Fenómenos Electrofisiológicos/fisiología , Células HEK293 , Humanos , Inmunohistoquímica , Interferón Tipo I/biosíntesis , Interferón beta/farmacología , Masculino , Ratones Endogámicos C57BL , Neocórtex/citología , Neocórtex/metabolismo , Neocórtex/fisiología , Red Nerviosa/citología , Red Nerviosa/fisiología , Técnicas de Placa-Clamp , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Interferón/fisiología , Transducción de Señal/fisiología , Transfección
9.
Biochim Biophys Acta ; 1831(1): 133-8, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23388400

RESUMEN

Plasticity-related genes (PRGs, Lipid phosphate phosphatase-related proteins LPPRs) are a defined as a subclass of the lipid phosphate phosphatase (LPP) superfamily, comprising so far five brain- and vertebrate-specific membrane-spanning proteins. LPPs interfere with lipid phosphate signaling and are thereby involved in mediating the extracellular concentration and signal transduction of lipid phosphate esters such as lysophosphatidate (LPA) and spingosine-1 phosphate (S1P). LPPs dephosphorylate their substrates through extracellular catalytic domains, thus making them ecto-phosphatases. PRGs/LPPRs are structurally similar to the other LPP family members in general. They are predominantly expressed in the CNS in a subtype specific pattern rather than having a wide tissue distribution. In contrast to LPPs, PRGs/LPPRs may act by modifying bioactive lipids and their signaling pathways, rather than possessing an ecto-phosphatase activity. However, the exact functional roles of PRGs/LPPRs have just begun to be explored. Here, we discuss new findings on the neuron-specific transcriptional regulation of PRG1/LPPR4 and new insights into protein-protein interaction and signaling pathway regulation. Further, we start to shed light on the subcellular localization and the resulting functional modulatory influence of PRG1/LPPR4 expression in excitatory synaptic transmission to the established neural effects such as promotion of filopodia formation, neurite extension, axonal sprouting and reorganization after lesion. This range of effects suggests an involvement in the pathogenesis and/or reparation attempts in disease. Therefore, we summarize available data on the association of PRGs/LPPRs with several neurological and other diseases in humans and experimental animals. Finally we highlight important open questions and emerging future directions of research. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.


Asunto(s)
Encéfalo/metabolismo , Proteínas del Tejido Nervioso/genética , Plasticidad Neuronal/genética , Monoéster Fosfórico Hidrolasas/genética , Animales , Encéfalo/patología , Humanos , Neoplasias/genética , Neoplasias/patología , Proteínas del Tejido Nervioso/metabolismo , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/patología , Enfermedades del Sistema Nervioso/fisiopatología , Monoéster Fosfórico Hidrolasas/metabolismo
10.
J Neuroinflammation ; 11: 185, 2014 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-25359459

RESUMEN

BACKGROUND: Cytokines are key players in the interactions of the immune and nervous systems. Recently, we showed that such interplay is mediated by type I interferons (IFNs), which elevate the excitability of neocortical pyramidal neurons. A line of indirect evidence suggested that modulation of multiple ion channels underlies the effect. However, which currents are principally involved and how the IFN signaling cascade is linked to the respective ion channels remains elusive. METHODS: We tested several single and combined ionic current modulations using an in silico model of a neocortical layer 5 neuron. Subsequently we investigated resulting predictions by whole-cell patch-clamp recordings in layer 5 neurons of ex vivo neocortical rat brain slices pharmacologically reproducing or prohibiting neuronal IFN effects. RESULTS: The amount and type of modulation necessary to replicate IFN effects in silico suggested protein kinase C (PKC) activation as link between the type I IFN signaling and ion channel modulations. In line with this, PKC activation with 4ß-phorbol 12-myristate 13-acetate (4ß-PMA) or Bryostatin1 augmented the excitability of neocortical layer 5 neurons comparable to IFN-ß in our ex vivo recordings. In detail, both PKC activators attenuated the rheobase and increased the input-output gain as well as the input resistance, thereby augmenting the neuronal excitability. Similar to IFN-ß they also left the threshold of action potential generation unaffected. In further support of PKC mediating type I IFN effects, IFN-ß, 4ß-PMA and Bryostatin1 reduced the amplitude of post-train after-hyperpolarizations in a similar manner. In conjunction with this finding, IFN-ß reduced M-currents, which contribute to after-hyperpolarizations and are modulated by PKC. Finally, blocking PKC activation with GF109203X at the catalytic site or calphostin C at the regulatory site prevented the main excitatory effects of IFN-ß. CONCLUSION: Multiple ion channel modulations underlie the neuromodulatory effect of type I IFNs. PKC activation is both sufficient and necessary for mediating the effect, and links the IFN signaling cascade to the intrinsic ion channels. Therefore, we regard PKC activation as unitary mechanism for the neuromodulatory potential of type I IFNs in neocortical neurons.


Asunto(s)
Interferón beta/metabolismo , Neocórtex/metabolismo , Neuroinmunomodulación/fisiología , Proteína Quinasa C/metabolismo , Células Piramidales/metabolismo , Transducción de Señal/fisiología , Animales , Activación Enzimática/fisiología , Canales Iónicos/fisiología , Masculino , Modelos Neurológicos , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Ratas , Ratas Wistar
11.
Sci Adv ; 10(25): eadj0720, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38896627

RESUMEN

Mutations in the transcription factors encoded by PHOX2B or LBX1 correlate with congenital central hypoventilation disorders. These conditions are typically characterized by pronounced hypoventilation, central apnea, and diminished chemoreflexes, particularly to abnormally high levels of arterial PCO2. The dysfunctional neurons causing these respiratory disorders are largely unknown. Here, we show that distinct, and previously undescribed, sets of medullary neurons coexpressing both transcription factors (dB2 neurons) account for specific respiratory functions and phenotypes seen in congenital hypoventilation. By combining intersectional chemogenetics, intersectional labeling, lineage tracing, and conditional mutagenesis, we uncovered subgroups of dB2 neurons with key functions in (i) respiratory tidal volumes, (ii) the hypercarbic reflex, (iii) neonatal respiratory stability, and (iv) neonatal survival. These data provide functional evidence for the critical role of distinct medullary dB2 neurons in neonatal respiratory physiology. In summary, our work identifies distinct subgroups of dB2 neurons regulating breathing homeostasis, dysfunction of which causes respiratory phenotypes associated with congenital hypoventilation.


Asunto(s)
Proteínas de Homeodominio , Hipoventilación , Bulbo Raquídeo , Neuronas , Factores de Transcripción , Hipoventilación/congénito , Hipoventilación/genética , Animales , Neuronas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Bulbo Raquídeo/metabolismo , Apnea Central del Sueño/genética , Fenotipo , Humanos
12.
Cell Physiol Biochem ; 31(4-5): 532-41, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23571312

RESUMEN

BACKGROUND/AIMS: Hyperpolarization activated cyclic nucleotide gated 1 (HCN1) channels determine neuronal excitability in several brain regions. In contrast to HCN2 and HCN4, HCN1 is less sensitive to cAMP and the number of other known modulators is limited. One of those, the protein kinase C (PKC), showed opposing effects on mouse HCN1 channels expressed in Xenopus oocytes. METHODS: In order to study PKC effects on HCN1 mediated currents in a mammalian environment we expressed rat HCN1 or human HCN1 in human embryonic kidney (HEK293) cells and rat HCN1 in murine neuroblastoma (N1E-115) cells. We recorded the resulting Ih before and during the application of the membrane permeable non-metabolizable PKC-activator 4ßPMA in cell-attached mode of the patch-clamp technique, leaving the intracellular environment intact. RESULTS: 4ßPMA reduced maximal HCN1 mediated currents to about 60-70 % and slowed its activation, but left its voltage sensitivity unchanged. The effect was neither due to species-related differences nor restricted to HEK293 cells, because it was comparable for human and rat HCN1 in HEK293 and for rat HCN1 in N1E-115 cells. However, pre-treatment with the PKC blocker GF109203X abolished 4ßPMA induced Ih changes. Disrupting the intracellular environment by recording in whole-cell mode drastically reduced the 4ßPMA effect. CONCLUSION: PKC activation reduces and slows Ih in non-neuronal and neuronal mammalian cells transfected with rat or human HCN1 if the intracellular content remains intact.


Asunto(s)
Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Proteína Quinasa C/metabolismo , Animales , Línea Celular , Células HEK293 , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/antagonistas & inhibidores , Indoles/farmacología , Maleimidas/farmacología , Potenciales de la Membrana/efectos de los fármacos , Ratones , Técnicas de Placa-Clamp , Proteína Quinasa C/química , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacología
13.
BMC Neurosci ; 14: 36, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-23506325

RESUMEN

BACKGROUND: Recently, we and others proposed plasticity-related gene 3 (PRG3) as a novel molecule in neuritogenesis based on PRG3 overexpression experiments in neuronal and non-neuronal cell lines. However, direct information on PRG3 effects in neuronal development and, in particular, its putative spatio-temporal distribution and conditions of action, is sparse. RESULTS: We demonstrate here that PRG3 induces filopodia formation in HEK293 cells depending on its N-glycosylation status. The PRG3 protein was strongly expressed during mouse brain development in vivo from embryonic day 16 to postnatal day 5 (E16 - P5). From P5 on, expression declined. Furthermore, in early, not yet polarized hippocampal cultured neurons, PRG3 was expressed along the neurite shaft. Knock-down of PRG3 in these neurons led to a decreased number of neurites. This phenotype is rescued by expression of an shRNA-resistant PRG3 construct in PRG3 knock-down neurons. After polarization, endogenous PRG3 expression shifted mainly to axons, specifically to the plasma membrane along the neurite shaft. These PRG3 pattern changes appeared temporally and spatially related to ongoing synaptogenesis. Therefore we tested (i) whether dendritic PRG3 re-enhancement influences synaptic currents and (ii) whether synaptic inputs contribute to the PRG3 shift. Our results rendered both scenarios unlikely: (i) PRG3 over-expression had no influence on miniature excitatory postsynaptic currents (mEPSC) and (ii) blocking of incoming signals did not alter PRG3 distribution dynamics. In addition, PRG3 levels did not interfere with intrinsic neuronal properties. CONCLUSION: Taken together, our data indicate that endogenous PRG3 promotes neurite shaft protrusion and therefore contributes to regulating filopodia formation in immature neurons. PRG3 expression in more mature neurons, however, is predominantly localized in the axon. Changes in PRG3 levels did not influence intrinsic or synaptic neuronal properties.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Neuritas/metabolismo , Neuronas/citología , Análisis de Varianza , Animales , Animales Recién Nacidos , Asparagina/genética , Asparagina/metabolismo , Células Cultivadas , Proteína Mayor Básica del Eosinófilo/genética , Potenciales Postsinápticos Excitadores/genética , Femenino , Glicosilación , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Hipocampo/citología , Humanos , Masculino , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Neuroglía/citología , Técnicas de Placa-Clamp , Mutación Puntual/genética , Embarazo , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Seudópodos/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transfección
14.
Cell Rep ; 42(8): 112934, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37537840

RESUMEN

Extracellular potassium [K+]o elevation during synaptic activity retrogradely modifies presynaptic release and astrocytic uptake of glutamate. Hence, local K+ clearance and replenishment mechanisms are crucial regulators of glutamatergic transmission and plasticity. Based on recordings of astrocytic inward rectifier potassium current IKir and K+-sensitive electrodes as sensors of [K+]o as well as on in silico modeling, we demonstrate that the neuronal K+-Cl- co-transporter KCC2 clears local perisynaptic [K+]o during synaptic excitation by operating in an activity-dependent reversed mode. In reverse mode, KCC2 replenishes K+ in dendritic spines and complements clearance of [K+]o, therewith attenuating presynaptic glutamate release and shortening LTP. We thus demonstrate a physiological role of KCC2 in neuron-glial interactions and regulation of synaptic signaling and plasticity through the uptake of postsynaptically released K+.


Asunto(s)
Potasio , Simportadores , Animales , Glutamatos , Potasio/metabolismo , Sinapsis/metabolismo , Cotransportadores de K Cl
15.
Front Cell Neurosci ; 16: 913299, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36035261

RESUMEN

Interferon-γ (IFN-γ), a cytokine with neuromodulatory properties, has been shown to enhance inhibitory transmission. Because early inhibitory neurotransmission sculpts functional neuronal circuits, its developmental alteration may have grave consequences. Here, we investigated the acute effects of IFN-γ on γ-amino-butyric acid (GABA)ergic currents in layer 5 pyramidal neurons of the somatosensory cortex of rats at the end of the first postnatal week, a period of GABA-dependent cortical maturation. IFN-γ acutely increased the frequency and amplitude of spontaneous/miniature inhibitory postsynaptic currents (s/mIPSC), and this could not be reversed within 30 min. Neither the increase in amplitude nor frequency of IPSCs was due to upregulated interneuron excitability as revealed by current clamp recordings of layer 5 interneurons labeled with VGAT-Venus in transgenic rats. As we previously reported in more mature animals, IPSC amplitude increase upon IFN-γ activity was dependent on postsynaptic protein kinase C (PKC), indicating a similar activating mechanism. Unlike augmented IPSC amplitude, however, we did not consistently observe an increased IPSC frequency in our previous studies on more mature animals. Focusing on increased IPSC frequency, we have now identified a different activating mechanism-one that is independent of postsynaptic PKC but is dependent on inducible nitric oxide synthase (iNOS) and soluble guanylate cyclase (sGC). In addition, IFN-γ shifted short-term synaptic plasticity toward facilitation as revealed by a paired-pulse paradigm. The latter change in presynaptic function was not reproduced by the application of a nitric oxide donor. Functionally, IFN-γ-mediated alterations in GABAergic transmission overall constrained early neocortical activity in a partly nitric oxide-dependent manner as revealed by microelectrode array field recordings in brain slices analyzed with a spike-sorting algorithm. In summary, with IFN-γ-induced, NO-dependent augmentation of spontaneous GABA release, we have here identified a mechanism by which inflammation in the central nervous system (CNS) plausibly modulates neuronal development.

16.
J Neurosci Res ; 88(14): 3067-78, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20806410

RESUMEN

Hyperpolarization-activated cyclic nucleotide-gated ion channels (HCN) are key determinants of CNS functions. Here we describe an increase in hyperpolarization-activated current (I(h)) at the beginning of whole-cell recordings in rat layer 5 cortical neurons. For a closer investigation of this I(h) increase, we overexpressed the predominant layer 5 rat subunit HCN1 in HEK293 cells. We characterized the resulting I(h) in the cell-attached and whole-cell configurations. Breaking into whole-cell configuration led to about a 30% enhancement of rat HCN1-mediated I(h) accompanied by a depolarizing shift in voltage dependence and an accelerated time course of activation. This current enhancement is not species specific; for human HCN1, the current similarly increases in amount and kinetics. Although the changes were bound to cytosolic solution exchange, they were independent of cAMP, ATP, GTP, and the phosphate group donor phosphocreatine. Together, these data provide a characterization of heterologous expression of rat HCN1 and suggest that cytosolic contents suppress I(h). Such a mechanism might constitute a reserve in h-channel function in vivo.


Asunto(s)
Canales Catiónicos Regulados por Nucleótidos Cíclicos/fisiología , Potenciales de la Membrana/fisiología , Neocórtex/metabolismo , Neuronas/metabolismo , Canales de Potasio/fisiología , Animales , Polaridad Celular/fisiología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Citosol/metabolismo , Citosol/fisiología , Femenino , Células HEK293 , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Activación del Canal Iónico/fisiología , Neocórtex/citología , Neuronas/citología , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Fosforilación/fisiología , Canales de Potasio/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo
17.
Sci Rep ; 10(1): 13488, 2020 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-32778682

RESUMEN

We explored the non-thermal effects of radiofrequency (RF) electromagnetic fields and established a theoretical framework to elucidate their electrophysiological mechanisms. In experiments, we used a preclinical treatment device to treat the human colon cancer cell lines HT-29 and SW480 with either water bath heating (WB-HT) or 13.56 MHz RF hyperthermia (RF-HT) at 42 °C for 60 min and analyzed the proliferation and clonogenicity. We elaborated an electrical model for cell membranes and ion channels and estimated the resulting ion fluxes. The results showed that, for both cell lines, using RF-HT significantly reduced proliferation and clonogenicity compared to WB-HT. According to our model, the RF electric field component was rectified and smoothed in the direction of the channel, which resulted in a DC voltage of ~ 1 µV. This may induce ion fluxes that can potentially cause relevant disequilibrium of most ions. Therefore, RF-HT creates additional non-thermal effects in association with significant ion fluxes. Increasing the understanding of these effects can help improve cancer therapy.

18.
Neuropharmacology ; 144: 58-69, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30315843

RESUMEN

Alterations of the hyperpolarization activated nonselective cation current (Ih) are associated with epileptogenesis. Accordingly, the second-generation antiepileptic drug lamotrigine (LTG) enhances Ih in rodent hippocampus. We directly evaluated here whether LTG fails to enhance Ih in neocortical slices from patients with pharmacoresistant epilepsy. With somatic current clamp recordings we observed that LTG depolarized the membrane potential, decreased the input resistance and increased the "sag" in human layer 2/3 neocortical pyramidal neurons when confounding IKir was blocked. In subsequent voltage clamp recordings we confirmed a LTG induced increase of Ih that was qualitatively similar to the one we found in rat neocortical and hippocampal pyramidal neurons. This increase is sufficient to curtail single excitatory postsynaptic potentials (EPSPs) and reduces their temporal summation in human neocortical pyramidal neurons under physiological conditions, i.e. without blocking any other currents, as estimated by sharp microelectrode recordings. Taken together LTG increases Ih and thereby alters neuronal excitability, even in neurons of pharmacoresistant patients. However, whether this increase fully countervails the deficits of Ih in epileptic patients, remains elusive.


Asunto(s)
Anticonvulsivantes/farmacología , Epilepsia Refractaria/tratamiento farmacológico , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Lamotrigina/farmacología , Neocórtex/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Adulto , Animales , Epilepsia Refractaria/fisiopatología , Epilepsia Refractaria/cirugía , Epilepsia del Lóbulo Temporal/fisiopatología , Epilepsia del Lóbulo Temporal/cirugía , Femenino , Humanos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Neocórtex/fisiopatología , Células Piramidales/fisiología , Ratas Wistar , Técnicas de Cultivo de Tejidos
19.
Eur J Neurosci ; 28(6): 1068-79, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18783365

RESUMEN

This study investigates the impact of intrinsic currents on early neural development. A rat striatal ST14A cell line immortalized by SV40 large T antigen was employed as a model system because these cells act as multipotent neural progenitors when maintained at a permissive temperature of 33 degrees C. The whole-cell patch-clamp, molecular and immunocytochemical experiments point to a unique role of sodium currents in the multipotential stage of neural development. In initial experiments, action potential-like responses were only present when multipotential ST14A cells were substantially hyperpolarized. This led us to presume that sodium channels were only recruited during deep hyperpolarization. Subsequent voltage-clamp studies confirmed a remarkably hyperpolarized steady-state inactivation of the sodium currents and also showed that the underlying channels were tetrodotoxin resistant. Direct comparison with cells whose neuronal fate was already determined, i.e. short-term cultured striatal cells isolated at embryonic day 14 and after birth (post-natal day 0), showed that both traits are unique to ST14A cells. However, sodium currents in all three groups had a fast time- and voltage-dependent activation, as well as full inactivation with roughly similar kinetics. The peculiarity in ST14A might be explained by a relative excess of heart-type Na(V)1.5 and particularly its splice variant Na(V)1.5a, as suggested by reverse transcription-polymerase chain reaction results. We conclude that multipotent neural progenitor cells express Na(+) channels in their membrane irrespective of their fate but these channels have little effect due to their subunit composition, which is regulated by alternative splicing.


Asunto(s)
Células Madre Multipotentes/metabolismo , Neuronas/metabolismo , Sodio/metabolismo , Empalme Alternativo , Animales , Línea Celular , Activación del Canal Iónico/fisiología , Células Madre Multipotentes/citología , Miocardio/metabolismo , Neuronas/citología , Técnicas de Placa-Clamp , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ratas , Bloqueadores de los Canales de Sodio/metabolismo , Canales de Sodio/genética , Canales de Sodio/metabolismo , Tetrodotoxina/metabolismo
20.
Neurosci Res ; 62(4): 246-53, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18824050

RESUMEN

In rat models of Parkinson's disease, injections of 6-hydroxydopamine (6-OHDA) into different areas of the basal ganglia result in dopaminergic neurodegeneration in the substantia nigra. The extent and time course of the dopaminergic lesions varies between the models. While the effects on neurons have been extensively studied, little is known about the effects on astrocytes. We compared astrocytic activation (i.e. increase in number and staining intensity of glial fibrillary acidic protein immunoreactive cells) at the injection site and in downstream structures of the motor loop, i.e. the globus pallidus (GP) and the subthalamic nucleus (STN) following 6-OHDA lesion of the medial forebrain bundle (MFB) or the striatum. Lesions in both regions resulted in astrocytic activation at the lesion site, but their remote effects varied. MFB injections caused astrocytic activation in the ipsi- and contralateral striatum, whereas striatal injections resulted in astrocytic activation in the GP and STN. Since 6-OHDA injections into the MFB and the striatum result in complete and partial SNc lesions, respectively, we hypothesize that communication links exist between astrocytes, or between neurons and astrocytes, along neuronal pathways that transmit activating signals in response to neuronal damage-but only if the neuronal pathways are at least partially intact.


Asunto(s)
Adrenérgicos/toxicidad , Neuroglía/patología , Oxidopamina/toxicidad , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/patología , Núcleo Subtalámico/patología , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Lateralidad Funcional , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Haz Prosencefálico Medial/patología , Neuroglía/metabolismo , Enfermedad de Parkinson Secundaria/fisiopatología , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA