Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 222
Filtrar
1.
Mol Brain ; 14(1): 143, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34530877

RESUMEN

Vacuolar protein sorting 35 (VPS35) regulates neurotransmitter receptor recycling from endosomes. A missense mutation (D620N) in VPS35 leads to autosomal-dominant, late-onset Parkinson's disease. Here, we study the basic neurobiology of VPS35 and Parkinson's disease mutation effects in the D620N knock-in mouse and the effect of leucine-rich repeat kinase 2 (LRRK2) inhibition on synaptic phenotypes. The study was conducted using a VPS35 D620N knock-in mouse that expresses VPS35 at endogenous levels. Protein levels, phosphorylation states, and binding ratios in brain lysates from knock-in mice and wild-type littermates were assayed by co-immunoprecipitation and western blot. Dendritic protein co-localization, AMPA receptor surface expression, synapse density, and glutamatergic synapse activity in primary cortical cultures from knock-in and wild-type littermates were assayed using immunocytochemistry and whole-cell patch clamp electrophysiology. In brain tissue, we confirm VPS35 forms complexes with LRRK2 and AMPA-type glutamate receptor GluA1 subunits, in addition to NMDA-type glutamate receptor GluN1 subunits and D2-type dopamine receptors. Receptor and LRRK2 binding was unaltered in D620N knock-in mice, but we confirm the mutation results in reduced binding of VPS35 with WASH complex member FAM21, and increases phosphorylation of the LRRK2 kinase substrate Rab10, which is reversed by LRRK2 kinase inhibition in vivo. In cultured cortical neurons from knock-in mice, pRab10 is also increased, and reversed by LRRK2 inhibition. The mutation also results in increased endosomal recycling protein cluster density (VPS35-FAM21 co-clusters and Rab11 clusters), glutamate transmission, and GluA1 surface expression. LRRK2 kinase inhibition, which reversed Rab10 hyper-phosphorylation, did not rescue elevated glutamate release or surface GluA1 expression in knock-in neurons, but did alter AMPAR traffic in wild-type cells. The results improve our understanding of the cell biology of VPS35, and the consequences of the D620N mutation in developing neuronal networks. Together the data support a chronic synaptopathy model for latent neurodegeneration, providing phenotypes and candidate pathophysiological stresses that may drive eventual transition to late-stage parkinsonism in VPS35 PD. The study demonstrates the VPS35 mutation has effects that are independent of ongoing LRRK2 kinase activity, and that LRRK2 kinase inhibition alters basal physiology of glutamate synapses in vitro.


Asunto(s)
Endosomas/fisiología , Ácido Glutámico/fisiología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/antagonistas & inhibidores , Mutación Missense , Enfermedad de Parkinson/genética , Mutación Puntual , Proteínas de Transporte Vesicular/genética , Animales , Células Cultivadas , Dendritas/metabolismo , Mutación con Ganancia de Función , Técnicas de Sustitución del Gen , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/fisiología , Ratones , Ratones Endogámicos C57BL , Potenciales Postsinápticos Miniatura/fisiología , Proteínas del Tejido Nervioso/metabolismo , Técnicas de Placa-Clamp , Unión Proteica , Mapeo de Interacción de Proteínas , Receptores AMPA/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sinapsis/metabolismo , Proteínas de Transporte Vesicular/fisiología , Proteínas de Unión al GTP rab/metabolismo
2.
Neuropharmacology ; 196: 108714, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34271017

RESUMEN

Neurotensin (NT) serves as a neuromodulator in the brain where it regulates a variety of physiological functions. Whereas the central amygdala (CeA) expresses NT peptide and NTS1 receptors and application of NT has been shown to excite CeA neurons, the underlying cellular and molecular mechanisms have not been determined. We found that activation of NTS1 receptors increased the neuronal excitability of the lateral nucleus (CeL) of CeA. Both phospholipase Cß (PLCß) and phosphatidylinositol 4,5-bisphosphate (PIP2) depletion were required, whereas intracellular Ca2+ release and PKC were unnecessary for NT-elicited excitation of CeL neurons. NT increased the input resistance and time constants of CeL neurons, suggesting that NT excites CeL neurons by decreasing a membrane conductance. Depressions of the inwardly rectifying K+ (Kir) channels including both the Kir2 subfamily and the GIRK channels were required for NT-elicited excitation of CeL neurons. Activation of NTS1 receptors in the CeL led to GABAergic inhibition of medial nucleus of CeA neurons, suggesting that NT modulates the network activity in the amygdala. Our results may provide a cellular and molecular mechanism to explain the physiological functions of NT in vivo.


Asunto(s)
Potenciales de Acción/fisiología , Núcleo Amigdalino Central/metabolismo , Potenciales de la Membrana/fisiología , Neuronas/metabolismo , Neurotensina/metabolismo , Receptores de Neurotensina/metabolismo , Animales , Núcleo Amigdalino Central/fisiología , Proteínas de Unión al GTP/metabolismo , Potenciales Postsinápticos Inhibidores/fisiología , Potenciales Postsinápticos Miniatura/fisiología , Neuronas/fisiología , Técnicas de Placa-Clamp , Fosfolipasa C beta/metabolismo , Ratas , Transducción de Señal
3.
J Neurosci ; 41(35): 7329-7339, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34290081

RESUMEN

Post-tetanic potentiation (PTP) is a form of short-term plasticity that lasts for tens of seconds following a burst of presynaptic activity. It has been proposed that PTP arises from protein kinase C (PKC) phosphorylation of Munc18-1, an SM (Sec1/Munc-18 like) family protein that is essential for release. To test this model, we made a knock-in mouse in which all Munc18-1 PKC phosphorylation sites were eliminated through serine-to-alanine point mutations (Munc18-1SA mice), and we studied mice of either sex. The expression of Munc18-1 was not altered in Munc18-1SA mice, and there were no obvious behavioral phenotypes. At the hippocampal CA3-to-CA1 synapse and the granule cell parallel fiber (PF)-to-Purkinje cell (PC) synapse, basal transmission was largely normal except for small decreases in paired-pulse facilitation that are consistent with a slight elevation in release probability. Phorbol esters that mimic the activation of PKC by diacylglycerol still increased synaptic transmission in Munc18-1SA mice. In Munc18-1SA mice, 70% of PTP remained at CA3-to-CA1 synapses, and the amplitude of PTP was not reduced at PF-to-PC synapses. These findings indicate that at both CA3-to-CA1 and PF-to-PC synapses, phorbol esters and PTP enhance synaptic transmission primarily by mechanisms that are independent of PKC phosphorylation of Munc18-1.SIGNIFICANCE STATEMENT A leading mechanism for a prevalent form of short-term plasticity, post-tetanic potentiation (PTP), involves protein kinase C (PKC) phosphorylation of Munc18-1. This study tests this mechanism by creating a knock-in mouse in which Munc18-1 is replaced by a mutated form of Munc18-1 that cannot be phosphorylated. The main finding is that most PTP at hippocampal CA3-to-CA1 synapses or at cerebellar granule cell-to-Purkinje cell synapses does not rely on PKC phosphorylation of Munc18-1. Thus, mechanisms independent of PKC phosphorylation of Munc18-1 are important mediators of PTP.


Asunto(s)
Proteínas Munc18/metabolismo , Plasticidad Neuronal/fisiología , Proteína Quinasa C/metabolismo , Procesamiento Proteico-Postraduccional , Sustitución de Aminoácidos , Animales , Femenino , Técnicas de Sustitución del Gen , Hipocampo/fisiología , Masculino , Ratones , Ratones Noqueados , Potenciales Postsinápticos Miniatura/efectos de los fármacos , Potenciales Postsinápticos Miniatura/fisiología , Proteínas Munc18/deficiencia , Mutación Missense , Ésteres del Forbol/farmacología , Fosforilación , Mutación Puntual , Proteína Quinasa C/deficiencia , Células de Purkinje/fisiología , Proteínas Recombinantes/metabolismo , Transmisión Sináptica/efectos de los fármacos
4.
Elife ; 102021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34151775

RESUMEN

Sleep is important for brain plasticity, but its exact function remains mysterious. An influential but controversial idea is that a crucial function of sleep is to drive widespread downscaling of excitatory synaptic strengths. Here, we used real-time sleep classification, ex vivo measurements of postsynaptic strength, and in vivo optogenetic monitoring of thalamocortical synaptic efficacy to ask whether sleep and wake states can constitutively drive changes in synaptic strength within the neocortex of juvenile rats. We found that miniature excitatory postsynaptic current amplitudes onto L4 and L2/3 pyramidal neurons were stable across sleep- and wake-dense epochs in both primary visual (V1) and prefrontal cortex (PFC). Further, chronic monitoring of thalamocortical synaptic efficacy in V1 of freely behaving animals revealed stable responses across even prolonged periods of natural sleep and wake. Together, these data demonstrate that sleep does not drive widespread downscaling of synaptic strengths during the highly plastic critical period in juvenile animals. Whether this remarkable stability across sleep and wake generalizes to the fully mature nervous system remains to be seen.


Asunto(s)
Potenciales Postsinápticos Excitadores/fisiología , Neocórtex/fisiología , Sueño/fisiología , Sinapsis/fisiología , Vigilia/fisiología , Animales , Potenciales Evocados/fisiología , Potenciales Postsinápticos Miniatura/fisiología , Optogenética , Técnicas de Placa-Clamp , Células Piramidales , Ratas , Ratas Long-Evans , Corteza Visual/fisiología
5.
Nat Commun ; 12(1): 2849, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33990590

RESUMEN

Long-term depression (LTD) of synaptic strength can take multiple forms and contribute to circuit remodeling, memory encoding or erasure. The generic term LTD encompasses various induction pathways, including activation of NMDA, mGlu or P2X receptors. However, the associated specific molecular mechanisms and effects on synaptic physiology are still unclear. We here compare how NMDAR- or P2XR-dependent LTD affect synaptic nanoscale organization and function in rodents. While both LTDs are associated with a loss and reorganization of synaptic AMPARs, only NMDAR-dependent LTD induction triggers a profound reorganization of PSD-95. This modification, which requires the autophagy machinery to remove the T19-phosphorylated form of PSD-95 from synapses, leads to an increase in AMPAR surface mobility. We demonstrate that these post-synaptic changes that occur specifically during NMDAR-dependent LTD result in an increased short-term plasticity improving neuronal responsiveness of depressed synapses. Our results establish that P2XR- and NMDAR-mediated LTD are associated to functionally distinct forms of LTD.


Asunto(s)
Homólogo 4 de la Proteína Discs Large/fisiología , Depresión Sináptica a Largo Plazo/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Adenosina Trifosfato/administración & dosificación , Animales , Autofagia/fisiología , Células Cultivadas , Homólogo 4 de la Proteína Discs Large/deficiencia , Femenino , Hipocampo/citología , Hipocampo/fisiología , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Potenciales Postsinápticos Miniatura/fisiología , Modelos Neurológicos , N-Metilaspartato/administración & dosificación , Plasticidad Neuronal/fisiología , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley , Receptores AMPA/fisiología , Receptores Purinérgicos P2X/fisiología
6.
Mol Cell Neurosci ; 112: 103613, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33753311

RESUMEN

Presynaptic neurotransmitter release is strictly regulated by SNARE proteins, Ca2+ and a number of Ca2+ sensors including synaptotagmins (Syts) and Double C2 domain proteins (Doc2s). More than seventy years after the original description of spontaneous release, the mechanism that regulates this process is still poorly understood. Syt-1, Syt7 and Doc2 proteins contribute predominantly, but not exclusively, to synchronous, asynchronous and spontaneous phases of release. The proteins share a conserved tandem C2 domain architecture, but are functionally diverse in their subcellular location, Ca2+-binding properties and protein interactions. In absence of Syt-1, Doc2a and -b, neurons still exhibit spontaneous vesicle fusion which remains Ca2+-sensitive, suggesting the existence of additional sensors. Here, we selected Doc2c, rabphilin-3a and Syt-7 as three potential Ca2+ sensors for their sequence homology with Syt-1 and Doc2b. We genetically ablated each candidate gene in absence of Doc2a and -b and investigated spontaneous and evoked release in glutamatergic hippocampal neurons, cultured either in networks or on microglial islands (autapses). The removal of Doc2c had no effect on spontaneous or evoked release. Syt-7 removal also did not affect spontaneous release, although it altered short-term plasticity by accentuating short-term depression. The removal of rabphilin caused an increased spontaneous release frequency in network cultures, an effect that was not observed in autapses. Taken together, we conclude that Doc2c and Syt-7 do not affect spontaneous release of glutamate in hippocampal neurons, while our results suggest a possible regulatory role of rabphilin-3a in neuronal networks. These findings importantly narrow down the repertoire of synaptic Ca2+ sensors that may be implicated in the spontaneous release of glutamate.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas de Unión al Calcio/fisiología , Calcio/metabolismo , Hipocampo/metabolismo , Proteínas del Tejido Nervioso/fisiología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Sinaptotagmina I/fisiología , Proteínas de Transporte Vesicular/fisiología , Potenciales de Acción , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Animales , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/genética , Células Cultivadas , Secuencia Conservada , Ácido Glutámico/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Potenciales Postsinápticos Miniatura/fisiología , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Técnicas de Placa-Clamp , Dominios Proteicos , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Sinaptotagmina I/química , Sinaptotagmina I/deficiencia , Sinaptotagmina I/genética , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/deficiencia , Proteínas de Transporte Vesicular/genética , Rabfilina-3A
7.
Mol Cell Neurosci ; 109: 103563, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33039519

RESUMEN

Recent work demonstrated that sympathetic neurons innervate the skeletal muscle near the neuromuscular junction (NMJ), and muscle sympathectomy and sympathomimetic agents strongly influence motoneuron synaptic vesicle release ex vivo. Moreover, reports attest that the pontine nucleus locus coeruleus (LC) projects to preganglionic sympathetic neurons and regulates human mobility and skeletal muscle physiology. Thus, we hypothesized that peripheral and central sympathetic neurons projecting directly or indirectly to the skeletal muscle regulate NMJ transmission. The aim of this study was to define the specific neuronal groups in the peripheral and central nervous systems that account for such regulation in adult mice in vivo by using optogenetics and NMJ transmission recordings in 3-5-month-old, male and female ChR2(H134R/EYFP)/TH-Cre mice. After detecting ChR2(H134R)/EYFP fluorescence in the paravertebral ganglia and LC neurons, we tested whether optostimulating the plantar nerve near the lumbricalis muscle or LC neurons effectively modulates motor nerve terminal synaptic vesicle release in living mice. Nerve optostimulation increased motor synaptic vesicle release in vitro and in vivo, while the presynaptic adrenoceptor blockers propranolol (ß1/ß2) and atenolol (ß1) prevented this outcome. The effect is primarily presynaptic since miniature end-plate potential (MEPP) kinetics remained statistically unmodified after stimulation. In contrast, optostimulation of LC neurons did not regulate NMJ transmission. In summary, we conclude that postganglionic sympathetic neurons, but not LC neurons, increased NMJ transmission by acting on presynaptic ß1-adrenergic receptors in vivo.


Asunto(s)
Locus Coeruleus/fisiología , Neuronas Motoras/fisiología , Unión Neuromuscular/fisiología , Optogenética/métodos , Transmisión Sináptica/fisiología , Nervio Tibial/fisiología , Animales , Channelrhodopsins/análisis , Channelrhodopsins/genética , Dependovirus/fisiología , Femenino , Ganglios Simpáticos/fisiología , Genes Reporteros , Proteínas Fluorescentes Verdes/análisis , Rayos Láser , Luz , Masculino , Ratones , Ratones Transgénicos , Potenciales Postsinápticos Miniatura/fisiología , Neuronas Motoras/efectos de la radiación , Mutación Missense , Receptores Adrenérgicos beta 1/fisiología , Proteínas Recombinantes de Fusión/análisis , Fibras Simpáticas Posganglionares/fisiología , Transmisión Sináptica/efectos de la radiación , Nervio Tibial/efectos de la radiación
8.
Brain Res Bull ; 164: 307-313, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32937185

RESUMEN

High-frequency spinal cord stimulation (HFSCS) at 10 kHz provides paresthesia-free treatment for chronic pain. However, the underlying mechanisms of its action have not been fully elucidated. The aim of the present study was to investigate the effect of HFSCS treatment on spinal glutamate release and uptake in spared nerve injury (SNI) rats. HFSCS was applied to the T10/T11 spinal cord 3 days after SNI. The concentration of spinal glutamate, glutamate transporter activity and miniature excitatory postsynaptic currents (mEPSCs) from neurons in lamina II were evaluated. HFSCS treatment alleviated SNI pain induced by mechanical and cold allodynia. HFSCS treatment also partially restored altered spinal glutamate uptake activity, the levels of spinal glutamate, and the frequency of mEPSCs following SNI. In conclusion, HFSCS treatment attenuated SNI-induced neuropathic pain and partially restored the altered glutamate uptake after SNI.


Asunto(s)
Potenciales Postsinápticos Excitadores/fisiología , Ácido Glutámico/metabolismo , Potenciales Postsinápticos Miniatura/fisiología , Neuralgia/metabolismo , Estimulación de la Médula Espinal/métodos , Médula Espinal/metabolismo , Animales , Masculino , Actividad Motora/fisiología , Neuralgia/fisiopatología , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley
9.
J Neurosci ; 40(21): 4090-4102, 2020 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-32312887

RESUMEN

Neurons can respond to decreased network activity with a homeostatic increase in the amplitudes of miniature EPSCs (mEPSCs). The prevailing view is that mEPSC amplitudes are uniformly multiplied by a single factor, termed "synaptic scaling." Deviations from purely multiplicative scaling have been attributed to biological differences, or to a distortion imposed by a detection threshold limit. Here, we demonstrate in neurons dissociated from cortices of male and female mice that the shift in mEPSC amplitudes observed in the experimental data cannot be reproduced by simulation of uniform multiplicative scaling, with or without the distortion caused by applying a detection threshold. Furthermore, we demonstrate explicitly that the scaling factor is not uniform but is close to 1 for small mEPSCs, and increases with increasing mEPSC amplitude across a substantial portion of the data. This pattern was also observed for previously published data from dissociated mouse hippocampal neurons and dissociated rat cortical neurons. The finding of "divergent scaling" shifts the current view of homeostatic plasticity as a process that alters all synapses on a neuron equally to one that must accommodate the differential effect observed for small versus large mEPSCs. Divergent scaling still accomplishes the essential homeostatic task of modifying synaptic strengths in the opposite direction of the activity change, but the consequences are greatest for those synapses which individually are more likely to bring a neuron to threshold.SIGNIFICANCE STATEMENT In homeostatic plasticity, the responses to chronic increases or decreases in network activity act in the opposite direction to restore normal activity levels. Homeostatic plasticity is likely to play a role in diseases associated with long-term changes in brain function, such as epilepsy and neuropsychiatric illnesses. One homeostatic response is the increase in synaptic strength following a chronic block of activity. Research is focused on finding a globally expressed signaling pathway, because it has been proposed that the plasticity is uniformly expressed across all synapses. Here, we show that the plasticity is not uniform. Our work suggests that homeostatic signaling molecules are likely to be differentially expressed across synapses.


Asunto(s)
Corteza Cerebral/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Potenciales Postsinápticos Miniatura/fisiología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Animales , Células Cultivadas , Ratones , Técnicas de Placa-Clamp , Sinapsis/fisiología , Transmisión Sináptica/fisiología
10.
J Neurosci Res ; 98(6): 1213-1231, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32133675

RESUMEN

Tomosyn, a protein encoded by syntaxin-1-binding protein 5 (STXBP5) gene, has a well-established presynaptic role in the inhibition of neurotransmitter release and the reduction of synaptic transmission by its canonical interaction with the soluble N-ethylmaleimide-sensitive factor attachment protein receptor machinery. However, the postsynaptic role of tomosyn in dendritic arborization, spine stability, and trafficking of ionotropic glutamate receptors remains to be elucidated. We used short hairpin RNA to knock down tomosyn in mouse primary neurons to evaluate the postsynaptic cellular function and molecular signaling regulated by tomosyn. Knockdown of tomosyn led to an increase of RhoA GTPase activity accompanied by compromised dendritic arborization, loss of dendritic spines, decreased surface expression of AMPA receptors, and reduced miniature excitatory postsynaptic current frequency. Inhibiting RhoA signaling was sufficient to rescue the abnormal dendritic morphology and the surface expression of AMPA receptors. The function of tomosyn regulating RhoA is mediated through the N-terminal WD40 motif, where two variants each carrying a single nucleotide mutation in this region were found in individuals with autism spectrum disorder (ASD). We demonstrated that these variants displayed loss-of-function phenotypes. Unlike the wild-type tomosyn, these two variants failed to restore the reduced dendritic complexity, spine density, as well as decreased surface expression of AMPA receptors in tomosyn knockdown neurons. This study uncovers a novel role of tomosyn in maintaining neuronal function by inhibiting RhoA activity. Further analysis of tomosyn variants also provides a potential mechanism for explaining cellular pathology in ASD.


Asunto(s)
Dendritas/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas R-SNARE/metabolismo , Receptores AMPA/metabolismo , Animales , Línea Celular Tumoral , Potenciales Postsinápticos Excitadores/fisiología , Ratones , Potenciales Postsinápticos Miniatura/fisiología , Proteínas de Unión al GTP Monoméricas/genética , Proteínas del Tejido Nervioso/genética , Proteínas R-SNARE/genética , Receptores AMPA/genética
11.
Sci Rep ; 10(1): 962, 2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31969605

RESUMEN

Long-term memory of complex olfactory learning is expressed by wide spread enhancement in excitatory and inhibitory synaptic transmission onto piriform cortex pyramidal neurons. A particularly interesting modification in synaptic inhibition is the hyperpolarization of the reversal potential of the fast post synaptic inhibitory potential (fIPSP). Here we study the mechanism underlying the maintenance of such a shift in the fIPSP. Blocking of the neuronal specific K+-Cl- co-transporter (KCC2) in neurons of trained rats significantly depolarized the averaged fIPSP reversal potential of the spontaneous miniature inhibitory post synaptic currents (mIPSCs), to the averaged pre-training level. A similar effect was obtained by blocking PKC, which was previously shown to upregulate KCC2. Accordingly, the level of PKC-dependent phosphorylation of KCC2, at the serine 940 site, was significantly increased after learning. In contrast, blocking two other key second messenger systems CaMKII and PKA, which have no phosphorylation sites on KCC2, had no effect on the fIPSP reversal potential. Importantly, the PKC inhibitor also reduced the averaged amplitude of the spontaneous miniature excitatory synaptic currents (mEPSCs) in neurons of trained rats only, to the pre-training level. We conclude that learning-induced hyper-polarization of the fIPSP reversal potential is mediated by PKC-dependent increase of KCC2 phosphorylation.


Asunto(s)
Aprendizaje Discriminativo/fisiología , Inhibición Neural/fisiología , Proteína Quinasa C/metabolismo , Simportadores/metabolismo , Sinapsis/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Aprendizaje Discriminativo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Masculino , Potenciales Postsinápticos Miniatura/efectos de los fármacos , Potenciales Postsinápticos Miniatura/fisiología , Inhibición Neural/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Olfato/efectos de los fármacos , Olfato/fisiología , Simportadores/antagonistas & inhibidores , Sinapsis/efectos de los fármacos , Cotransportadores de K Cl
12.
Mol Brain ; 13(1): 6, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31941524

RESUMEN

Developing central synapses exhibit robust plasticity and undergo experience-dependent remodeling. Evidently, synapses in sensory systems such as auditory brainstem circuits mature rapidly to achieve high-fidelity neurotransmission for sound localization. This depends on a developmental switch in AMPAR composition from slow-gating GluA1-dominant to fast-gating GluA4-dominant, but the mechanisms underlying this switch remain unknown. We hypothesize that patterned stimuli mimicking spontaneous/sound evoked activity in the early postnatal stage drives this gating switch. We examined activity-dependent changes in evoked and miniature excitatory postsynaptic currents (eEPSCs and mEPSCs) at the calyx of Held synapse by breaking through the postsynaptic membrane at different time points following 2 min of theta burst stimulation (TBS) to afferents in mouse brainstem slices. We found the decay time course of eEPSCs accelerated, but this change was not apparent until > 30 min after TBS. Histogram analyses of the decay time constants of mEPSCs for naive and tetanized synapses revealed two populations centered around τfast ≈ 0.4 and 0.8 ms, but the relative weight of the τ0.4 population over the τ0.8 population increased significantly only in tetanized synapses. Such changes are blocked by NMDAR or mGluR1/5 antagonists or inhibitors of CaMKII, PKC and protein synthesis, and more importantly precluded in GluA4-/- synapses, suggesting GluA4 is the substrate underlying the acceleration. Our results demonstrate a novel form of plasticity working through NMDAR and mGluR activation to trigger a gating switch of AMPARs with a temporally delayed onset of expression, ultimately enhancing the development of high-fidelity synaptic transmission.


Asunto(s)
Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Potenciales Postsinápticos Miniatura/fisiología , Plasticidad Neuronal/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Sinapsis/metabolismo , Cuerpo Trapezoide/fisiología , Animales , Señalización del Calcio , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Ratones , Proteínas del Tejido Nervioso/biosíntesis , Proteína Quinasa C/metabolismo , Receptores AMPA/biosíntesis , Receptores AMPA/deficiencia , Receptores AMPA/genética , Receptores de Glutamato Metabotrópico/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Transmisión Sináptica/fisiología , Tetania/fisiopatología , Ritmo Teta , Factores de Tiempo , Cuerpo Trapezoide/ultraestructura
13.
Neuroscience ; 423: 162-171, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31698023

RESUMEN

Despite the long history of investigations of adrenergic compounds and their biological effects, specific mechanisms of their action in distinct compartments of the motor unit remain obscure. Recent results have suggested that not only skeletal muscles but also the neuromuscular junctions represent important targets for the action of catecholamines. In this paper, we describe the effects of adrenaline and noradrenaline on the frequency of miniature endplate potentials, the quantal content of the evoked endplate potentials and the kinetics of acetylcholine quantal release in the motor nerve endings of the mouse diaphragm. Noradrenaline and adrenaline decreased the frequency of the spontaneous release of acetylcholine quanta. The effect of noradrenaline was prevented by the ß adrenoreceptor blocker propranolol, whereas the action of adrenaline was abolished by the α adrenoreceptor antagonist phentolamine. Noradrenaline did not alter the quantal content of endplate potentials, while adrenaline suppressed the evoked release of acetylcholine. Blocking the α adrenoreceptors prevented the decrease in quantal secretion caused by adrenaline. Quantal release became more asynchronous under noradrenaline, as evidenced by a greater dispersion of real synaptic delays; in contrast, adrenaline synchronized the release process. Our data suggest an involvement of α and ß adrenoreceptors in the diverse modulation of the frequency of miniature endplate potentials, the quantal content of the evoked endplate potentials and the kinetics of acetylcholine quantal secretion in the mouse neuromuscular junction. Moreover, the adrenoblockers affected both the evoked and spontaneous quantal release of acetylcholine, suggesting the presence of endogenous catecholamines in the vicinity of cholinergic synapses.


Asunto(s)
Acetilcolina/metabolismo , Epinefrina/fisiología , Unión Neuromuscular/metabolismo , Norepinefrina/fisiología , Agonistas alfa-Adrenérgicos/farmacología , Agonistas Adrenérgicos beta/farmacología , Animales , Diafragma/fisiología , Epinefrina/antagonistas & inhibidores , Epinefrina/farmacología , Femenino , Cinética , Masculino , Ratones , Potenciales Postsinápticos Miniatura/fisiología , Norepinefrina/antagonistas & inhibidores , Norepinefrina/farmacología , Fentolamina/farmacología , Propranolol/farmacología , Receptores Adrenérgicos alfa/fisiología , Receptores Adrenérgicos beta/fisiología
14.
Neuron ; 103(4): 617-626.e6, 2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31257103

RESUMEN

The autism-associated synaptic-adhesion gene Neuroligin-4 (NLGN4) is poorly conserved evolutionarily, limiting conclusions from Nlgn4 mouse models for human cells. Here, we show that the cellular and subcellular expression of human and murine Neuroligin-4 differ, with human Neuroligin-4 primarily expressed in cerebral cortex and localized to excitatory synapses. Overexpression of NLGN4 in human embryonic stem cell-derived neurons resulted in an increase in excitatory synapse numbers but a remarkable decrease in synaptic strength. Human neurons carrying the syndromic autism mutation NLGN4-R704C also formed more excitatory synapses but with increased functional synaptic transmission due to a postsynaptic mechanism, while genetic loss of NLGN4 did not significantly affect synapses in the human neurons analyzed. Thus, the NLGN4-R704C mutation represents a change-of-function mutation. Our work reveals contrasting roles of NLGN4 in human and mouse neurons, suggesting that human evolution has impacted even fundamental cell biological processes generally assumed to be highly conserved.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Transmisión Sináptica/fisiología , Animales , Trastorno Autístico/genética , Moléculas de Adhesión Celular Neuronal/genética , Células Cultivadas , Corteza Cerebral/fisiología , Células Madre Embrionarias/citología , Potenciales Postsinápticos Excitadores/fisiología , Genes Reporteros , Ácido Glutámico/fisiología , Humanos , Ratones , Potenciales Postsinápticos Miniatura/fisiología , Mutación Missense , Neurogénesis , Neuronas/fisiología , Fenotipo , Receptores de Glutamato/fisiología , Especificidad de la Especie , Sinapsis/química
15.
Neuropharmacology ; 157: 107667, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31207251

RESUMEN

(2R,6R)-hydroxynorketamine (HNK), a metabolite of ketamine, has recently been suggested to be a potent antidepressant for treating animal depression and has rapid-onset and long-lasting action through potentiating glutamatergic transmission. However, its other effects are still unclear. In the present study, we tested the effects of (2R,6R)-HNK on offensive aggression. A resident-intruder (RI) test was used as the main model to test elements of offensive aggression, including threats and bites. Electrophysiological recordings in the ventrolateral periaqueductal gray (vlPAG) were used to measure the functions of glutamatergic synaptic transmission. A single systemic injection of (2R,6R)-HNK, but not (2S,6S)-HNK, increased elements of offensive aggression, including threats and bites, in a dose-dependent manner with long-lasting action. Moreover, (2R,6R)-HNK increased the input-output curve, the AMPA-mediated current, and the frequency and amplitude of miniature excitatory postsynaptic currents (mEPSCs) and decreased the paired-pulse ratio (PPR) in the vlPAG. Furthermore, intra-vlPAG application of (2R,6R)-HNK increased aggressive and biting behaviors, which were abolished by an intra-vlPAG pretreatment with the AMPA receptors antagonist, CNQX. Notably, the intra-vlPAG CNQX pretreatment eliminated systemic (2R,6R)-HNK-enhanced aggressive and biting behaviors. The results of this suggest that (2R,6R)-HNK evokes offensive aggression by increasing vlPAG glutamatergic transmission. Although (2R,6R)-HNK is currently suggested to be effective for treating depression, its side effect of increasing offensive aggression should be a subject of concern in future drug development and therapy.


Asunto(s)
Agresión/efectos de los fármacos , Agresión/fisiología , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Sustancia Gris Periacueductal/fisiología , Transmisión Sináptica/fisiología , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Animales , Conducta Animal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Ketamina/análogos & derivados , Masculino , Microinyecciones , Potenciales Postsinápticos Miniatura/fisiología , Ratas
16.
Neuron ; 103(4): 673-685.e5, 2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31230762

RESUMEN

Autism spectrum disorder (ASD) is strongly associated with de novo gene mutations. One of the most commonly affected genes is SCN2A. ASD-associated SCN2A mutations impair the encoded protein NaV1.2, a sodium channel important for action potential initiation and propagation in developing excitatory cortical neurons. The link between an axonal sodium channel and ASD, a disorder typically attributed to synaptic or transcriptional dysfunction, is unclear. Here we show that NaV1.2 is unexpectedly critical for dendritic excitability and synaptic function in mature pyramidal neurons in addition to regulating early developmental axonal excitability. NaV1.2 loss reduced action potential backpropagation into dendrites, impairing synaptic plasticity and synaptic strength, even when NaV1.2 expression was disrupted in a cell-autonomous fashion late in development. These results reveal a novel dendritic function for NaV1.2, providing insight into cellular mechanisms probably underlying circuit and behavioral dysfunction in ASD.


Asunto(s)
Trastorno del Espectro Autista/genética , Dendritas/fisiología , Canal de Sodio Activado por Voltaje NAV1.2/fisiología , Corteza Prefrontal/fisiología , Células Piramidales/fisiología , Potenciales de Acción , Animales , Señalización del Calcio , Femenino , Regulación del Desarrollo de la Expresión Génica , Heterocigoto , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Hipocampo/fisiología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Potenciales Postsinápticos Miniatura/fisiología , N-Metilaspartato/análisis , Canal de Sodio Activado por Voltaje NAV1.2/genética , Neocórtex/citología , Neocórtex/crecimiento & desarrollo , Neocórtex/fisiología , Corteza Prefrontal/citología , Corteza Prefrontal/crecimiento & desarrollo , Ingeniería de Proteínas , Conducta Social , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/análisis
17.
Glia ; 67(10): 1893-1909, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31246351

RESUMEN

Human astrocytes differ dramatically in cell morphology and gene expression from murine astrocytes. The latter are well known to be of major importance in the formation of neuronal networks by promoting synapse maturation. However, whether human astrocyte lineage cells have a similar role in network formation has not been firmly established. Here, we investigated the impact of human astrocyte lineage cells on the functional maturation of neural networks that were derived from human induced pluripotent stem cells (hiPSCs). Initial in vitro differentiation of hiPSC-derived neural progenitor cells and immature neurons (glia+ cultures) resulted in spontaneously active neural networks as indicated by synchronous neuronal Ca2+ transients. Depleting proliferating neural progenitors from these cultures by short-term antimitotic treatment resulted in strongly astrocyte lineage cell-depleted neuronal networks (glia- cultures). Strikingly, in contrast to glia+ cultures, glia- cultures did not exhibit spontaneous network activity. Detailed analysis of the morphological and electrophysiological properties of neurons by patch clamp recordings revealed reduced dendritic arborization in glia- cultures. In addition, a reduced action potential frequency upon current injection in pyramidal-like neurons was observed, whereas the electrical excitability of multipolar neurons was unaltered. Furthermore, we found a reduced dendritic density of PSD95-positive excitatory synapses, and more immature properties of AMPA (alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) miniature excitatory postsynaptic currents (mEPSCs) in glia- cultures, suggesting that the maturation of glutamatergic synapses depends on the presence of hiPSC-derived astrocyte lineage cells. Intriguingly, addition of the astrocyte-derived synapse maturation inducer cholesterol increased the dendritic density of PSD95-positive excitatory synapses in glia- cultures.


Asunto(s)
Astrocitos/fisiología , Linaje de la Célula , Células Madre Pluripotentes Inducidas/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Sinapsis/fisiología , Potenciales de Acción/fisiología , Células Cultivadas , Potenciales Postsinápticos Excitadores/fisiología , Ácido Glutámico/metabolismo , Humanos , Potenciales Postsinápticos Miniatura/fisiología , Vías Nerviosas/fisiología , Células-Madre Neurales/fisiología , Receptores AMPA/metabolismo
18.
Glia ; 67(9): 1667-1679, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31050055

RESUMEN

During postnatal neurodevelopment, excessive synapses must be eliminated by microglia to complete the establishment of neural circuits in the brain. The lack of synaptic regulation by microglia has been implicated in neurodevelopmental disorders such as autism, schizophrenia, and intellectual disability. Here we suggest that vaccinia-related kinase 2 (VRK2), which is expressed in microglia, may stimulate synaptic elimination by microglia. In VRK2-deficient mice (VRK2KO ), reduced numbers of presynaptic puncta within microglia were observed. Moreover, the numbers of presynaptic puncta and synapses were abnormally increased in VRK2KO mice by the second postnatal week. These differences did not persist into adulthood. Even though an increase in the number of synapses was normalized, adult VRK2KO mice showed behavioral defects in social behaviors, contextual fear memory, and spatial memory.


Asunto(s)
Encéfalo/enzimología , Encéfalo/crecimiento & desarrollo , Microglía/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Sinapsis/enzimología , Animales , Encéfalo/citología , Células Cultivadas , Potenciales Postsinápticos Excitadores/fisiología , Miedo/fisiología , Humanos , Masculino , Memoria/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/citología , Potenciales Postsinápticos Miniatura/fisiología , Proteínas Serina-Treonina Quinasas/genética , Conducta Social , Técnicas de Cultivo de Tejidos
19.
Brain ; 142(7): 1938-1954, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31056671

RESUMEN

We performed next generation sequencing on 1696 patients with epilepsy and intellectual disability using a gene panel with 480 epilepsy-related genes including all GABAA receptor subunit genes (GABRs), and we identified six de novo GABR mutations, two novel GABRA5 mutations (c.880G>T, p.V294F and c.1238C>T, p.S413F), two novel GABRA1 mutations (c.778C>T, p.P260S and c.887T>C, p.L296S/c.944G>T, p.W315L) and two known GABRA1 mutations (c.335G>A, p.R112Q and c.343A>G, p.N115D) in six patients with intractable early onset epileptic encephalopathy. The α5(V294F and S413F) and α1(P260S and L296S/W315L) subunit residue substitutions were all in transmembrane domains, while the α1(R112Q and N115R) subunit residue substitutions were in the N-terminal GABA binding domain. Using multidisciplinary approaches, we compared effects of mutant GABAA receptor α5 and α1 subunits on the properties of recombinant α5ß3γ2 and α1ß3γ2 GABAA receptors in both neuronal and non-neuronal cells and characterized their effects on receptor clustering, biogenesis and channel function. GABAA receptors containing mutant α5 and α1 subunits all had reduced cell surface and total cell expression with altered endoplasmic reticulum processing, impaired synaptic clustering, reduced GABAA receptor function and decreased GABA binding potency. Our study identified GABRA5 as a causative gene for early onset epileptic encephalopathy and expands the mutant GABRA1 phenotypic spectrum, supporting growing evidence that defects in GABAergic neurotransmission contribute to early onset epileptic encephalopathy phenotypes.


Asunto(s)
Epilepsia/genética , Discapacidad Intelectual/genética , Receptores de GABA-A/genética , Sinapsis/genética , Niño , Preescolar , Epilepsia/complicaciones , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Discapacidad Intelectual/complicaciones , Masculino , Potenciales de la Membrana/fisiología , Potenciales Postsinápticos Miniatura/fisiología , Mutación , Cultivo Primario de Células , Receptores de GABA-A/biosíntesis , Receptores de GABA-A/metabolismo , Receptores de GABA-A/fisiología , Sinapsis/fisiología , Adulto Joven , Ácido gamma-Aminobutírico/metabolismo
20.
J Neurosci ; 39(23): 4448-4460, 2019 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-30936241

RESUMEN

Striatal output pathways are known to play a crucial role in the control of movement. One possible component for shaping the synaptic output of striatal neuron is the glutamatergic input that originates from cortex and thalamus. Although reports focusing on quantifying glutamatergic-induced morphological changes in striatum exist, the role of glutamatergic input in regulating striatal function remains poorly understood. Using primary neurons from newborn mice of either sex in a reduced two-neuron microcircuit culture system, we examined whether glutamatergic input modulates the output of striatal neurons. We found that glutamatergic input enhanced striatal inhibition in vitro With a glutamatergic partner from either cortex or thalamus, we attributed this potentiation to an increase in the size of quantal IPSC, suggesting a strengthening of the postsynaptic response to GABAergic signaling. Additionally, a differential effect of cortical and thalamic innervation onto striatal GABAergic neurons output was revealed. We observed that cortical, but not thalamic input, enhanced the number of releasable GABAergic synaptic vesicles and morphological synapses. Importantly, these alterations were reverted by blockade of neuronal activity and glutamate receptors, as well as disruption of BDNF-TrkB signaling. Together, our data indicate, for first time, that GABAergic synapse formation in corticostriatal pairs depends on two parallel, but potentially intersecting, signaling pathways that involve glutamate receptor activation in striatal neurons, as well as BDNF signaling. Understanding how cortical and thalamic inputs refine striatal output will pave the way toward dissecting basal ganglia activity in both physiological and pathological conditions.SIGNIFICANCE STATEMENT Striatal GABAergic microcircuits are critical for motor function. However, the mechanisms controlling striatal output, particularly at the level of synaptic strength, are unclear. Using two-neuron culture system, we quantified the synaptic output of individual striatal GABAergic neurons paired with a glutamatergic partner and studied the influence of the excitatory connections that are known to be interregionally formed in vivo We found that glutamatergic input potentiated striatal inhibitory output, potentially involving an increased feedback and/or feedforward inhibition. Moreover, distinct components of glutamatergic innervation, such as firing activity or release of neurotrophic factors were shown to be required for the glutamatergic-induced phenotype. Investigation, therefore, of two-neuron in vitro microcircuits could be a powerful tool to explore synaptic mechanisms or disease pathophysiology.


Asunto(s)
Cuerpo Estriado/fisiología , Neuronas GABAérgicas/fisiología , Ácido Glutámico/fisiología , Sinapsis/fisiología , Ácido gamma-Aminobutírico/fisiología , 2-Amino-5-fosfonovalerato/farmacología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Anticuerpos Neutralizantes/farmacología , Factor Neurotrófico Derivado del Encéfalo/antagonistas & inhibidores , Factor Neurotrófico Derivado del Encéfalo/farmacología , Factor Neurotrófico Derivado del Encéfalo/fisiología , Células Cultivadas , Corteza Cerebral/citología , Cuerpo Estriado/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Neuronas GABAérgicas/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Masculino , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Potenciales Postsinápticos Miniatura/efectos de los fármacos , Potenciales Postsinápticos Miniatura/fisiología , Proteínas Tirosina Quinasas/fisiología , Quinoxalinas/farmacología , Proteínas Recombinantes/farmacología , Vesículas Sinápticas/fisiología , Tetrodotoxina/farmacología , Tálamo/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA