Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
J Neurosci ; 40(20): 3969-3980, 2020 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-32277045

RESUMEN

The amygdala is a brain area critical for the formation of fear memories. However, the nature of the teaching signal(s) that drive plasticity in the amygdala are still under debate. Here, we use optogenetic methods to investigate the contribution of ventral tegmental area (VTA) dopamine neurons to auditory-cued fear learning in male mice. Using anterograde and retrograde labeling, we found that a sparse and relatively evenly distributed population of VTA neurons projects to the basal amygdala (BA). In vivo optrode recordings in behaving mice showed that many VTA neurons, among them putative dopamine neurons, are excited by footshocks, and acquire a response to auditory stimuli during fear learning. Combined cfos imaging and retrograde labeling in dopamine transporter (DAT) Cre mice revealed that a large majority of BA projectors (>95%) are dopamine neurons, and that BA projectors become activated by the tone-footshock pairing of fear learning protocols. Finally, silencing VTA dopamine neurons, or their axon terminals in the BA during the footshock, reduced the strength of fear memory as tested 1 d later, whereas silencing the VTA-central amygdala (CeA) projection had no effect. Thus, VTA dopamine neurons projecting to the BA contribute to fear memory formation, by coding for the saliency of the footshock event and by signaling such events to the basal amygdala.SIGNIFICANCE STATEMENT Powerful mechanisms of fear learning have evolved in animals and humans to enable survival. During fear conditioning, a sensory cue, such as a tone (the conditioned stimulus), comes to predict an innately aversive stimulus, such as a mild footshock (the unconditioned stimulus). A brain representation of the unconditioned stimulus must act as a teaching signal to instruct plasticity of the conditioned stimulus representation in fear-related brain areas. Here we show that dopamine neurons in the VTA that project to the basal amygdala contribute to such a teaching signal for plasticity, thereby facilitating the formation of fear memories. Knowledge about the role of dopamine in aversively motivated plasticity might allow further insights into maladaptive plasticities that underlie anxiety and post-traumatic stress disorders in humans.


Asunto(s)
Amígdala del Cerebelo/fisiología , Neuronas Dopaminérgicas/fisiología , Potenciales Evocados Somatosensoriales/fisiología , Miedo/fisiología , Miedo/psicología , Aprendizaje/fisiología , Área Tegmental Ventral/fisiología , Estimulación Acústica , Animales , Señales (Psicología) , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática , Fenómenos Electrofisiológicos/fisiología , Electrochoque , Masculino , Ratones , Neuroimagen
2.
J Neurosci ; 37(17): 4604-4617, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28363983

RESUMEN

Parvalbumin-expressing inhibitory neurons in the mammalian CNS are specialized for fast transmitter release at their output synapses. However, the Ca2+ sensor(s) used by identified inhibitory synapses, including the output synapses of parvalbumin-expressing inhibitory neurons, have only recently started to be addressed. Here, we investigated the roles of Syt1 and Syt2 at two types of fast-releasing inhibitory connections in the mammalian CNS: the medial nucleus of the trapezoid body to lateral superior olive glycinergic synapse, and the basket/stellate cell-Purkinje GABAergic synapse in the cerebellum. We used conditional and conventional knock-out (KO) mouse lines, with viral expression of Cre-recombinase and a light-activated ion channel for optical stimulation of the transduced fibers, to produce Syt1-Syt2 double KO synapses in vivo Surprisingly, we found that KO of Syt2 alone had only minor effects on evoked transmitter release, despite the clear presence of the protein in inhibitory nerve terminals revealed by immunohistochemistry. We show that Syt1 is weakly coexpressed at these inhibitory synapses and must be genetically inactivated together with Syt2 to achieve a significant reduction and desynchronization of fast release. Thus, our work identifies the functionally relevant Ca2+ sensor(s) at fast-releasing inhibitory synapses and shows that two major Syt isoforms can cooperate to mediate release at a given synaptic connection.SIGNIFICANCE STATEMENT During synaptic transmission, the influx of Ca2+ into the presynaptic nerve terminal activates a Ca2+ sensor for vesicle fusion, a crucial step in the activity-dependent release of neurotransmitter. Synaptotagmin (Syt) proteins, and especially Syt1 and Syt2, have been identified as the Ca2+ sensor at excitatory synapses, but the Ca2+ sensor(s) at inhibitory synapses in native brain tissue are not well known. We found that both Syt1 and Syt2 need to be genetically inactivated to cause a significant reduction of activity-evoked release at two types of fast inhibitory synapses in mouse brain. Thus, we identify Syt2 as a functionally important Ca2+ sensor at fast-releasing inhibitory synapses, and show that Syt1 and Syt2 can redundantly control transmitter release at specific brain synapses.


Asunto(s)
Neuronas/fisiología , Parvalbúminas/metabolismo , Transmisión Sináptica/fisiología , Sinaptotagmina II/fisiología , Sinaptotagmina I/fisiología , Animales , Cerebelo/metabolismo , Glicina/metabolismo , Ratones , Ratones Noqueados , Fibras Nerviosas/fisiología , Neuronas/metabolismo , Neurotransmisores/metabolismo , Estimulación Luminosa , Ácido gamma-Aminobutírico/fisiología
3.
J Physiol ; 596(20): 4945-4967, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30051910

RESUMEN

KEY POINTS: During the computation of sound localization, neurons of the lateral superior olive (LSO) integrate synaptic excitation arising from the ipsilateral ear with inhibition from the contralateral ear. We characterized the functional connectivity of the inhibitory and excitatory inputs onto LSO neurons in terms of unitary synaptic strength and convergence. Unitary IPSCs can generate large conductances, although their strength varies over a 10-fold range in a given recording. By contrast, excitatory inputs are relatively weak. The conductance associated with IPSPs needs to be at least 2-fold stronger than the excitatory one to guarantee effective inhibition of action potential (AP) firing. Computational modelling showed that strong unitary inhibition ensures an appropriate slope and midpoint of the tuning curve of LSO neurons. Conversely, weak but numerous excitatory inputs filter out spontaneous AP firing from upstream auditory neurons. ABSTRACT: The lateral superior olive (LSO) is a binaural nucleus in the auditory brainstem in which excitation from the ipsilateral ear is integrated with inhibition from the contralateral ear. It is unknown whether the strength of the unitary inhibitory and excitatory inputs is adapted to allow for optimal tuning curves of LSO neuron action potential (AP) firing. Using electrical and optogenetic stimulation of afferent synapses, we found that the strength of unitary inhibitory inputs to a given LSO neuron can vary over a ∼10-fold range, follows a roughly log-normal distribution, and, on average, causes a large conductance (9 nS). Conversely, unitary excitatory inputs, stimulated optogenetically under the bushy-cell specific promoter Math5, were numerous, and each caused a small conductance change (0.7 nS). Approximately five to seven bushy cell inputs had to be active simultaneously to bring an LSO neuron to fire. In double stimulation experiments, the effective inhibition window caused by IPSPs was short (1-3 ms) and its length depended on the inhibitory conductance; an ∼2-fold stronger inhibition than excitation was needed to suppress AP firing. Computational modelling suggests that few, but strong, unitary IPSPs create a tuning curve of LSO neuron firing with an appropriate slope and midpoint. Furthermore, weak but numerous excitatory inputs reduce the spontaneous AP firing that LSO neurons would otherwise inherit from their upstream auditory neurons. Thus, the specific connectivity and strength of unitary excitatory and inhibitory inputs to LSO neurons is optimized for the computations performed by these binaural neurons.


Asunto(s)
Potenciales Postsinápticos Excitadores , Potenciales Postsinápticos Inhibidores , Localización de Sonidos , Complejo Olivar Superior/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/fisiología , Complejo Olivar Superior/citología
4.
J Physiol ; 596(20): 4969-4982, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30054922

RESUMEN

KEY POINTS: Neurons of the lateral superior olive (LSO) in the brainstem receive powerful glycinergic inhibition that originates from the contralateral ear, and that plays an important role in sound localization. We investigated the ultrastructural basis for strong inhibition of LSO neurons using serial block face scanning electron microscopy. The soma and the proximal dendrite of an LSO neuron are surrounded by a high density of inhibitory axons, whereas excitatory axons are much sparser. A given inhibitory axon establishes contacts via several large axonal thickenings, called varicosities, which typically elaborate several active zones (range 1-11). The number of active zones across inhibitory axon segments is variable. These data thus provide an ultrastructural correlate for the strong and multiquantal, but overall variable, unitary IPSC amplitude observed for inhibitory inputs to LSO neuron. ABSTRACT: Binaural neurons in the lateral superior olive (LSO) integrate sound information arriving from each ear, and powerful glycinergic inhibition of these neurons plays an important role in this process. In the present study, we investigated the ultrastructural basis for strong inhibitory inputs onto LSO neurons using serial block face scanning electron microscopy. We reconstructed axon segments that make contact with the partially reconstructed soma and proximal dendrite of a mouse LSO neuron at postnatal day 18. Using functional measurements and the Sr2+ method, we find a constant quantal size but a variable quantal content between 'weak' and 'strong' unitary IPSCs. A 3-D reconstruction of a LSO neuron and its somatic synaptic afferents reveals how a large number of inhibitory axons intermingle in a complex fashion on the soma and proximal dendrite of an LSO neuron; a smaller number of excitatory axons was also observed. A given inhibitory axon typically contacts an LSO neuron via several large varicosities (average diameter 3.7 µm), which contain several active zones (range 1-11). The number of active zones across individual axon segments was highly variable. These data suggest that the variable unitary IPSC amplitude is caused by a variable number of active zones between inhibitory axons that innervate a given LSO neuron. The results of the present study show that relatively large multi-active zone varicosities, which can be repeated many times in a given presynaptic axon, provide the ultrastructural basis for the strong multiquantal inhibition received by LSO neurons.


Asunto(s)
Potenciales Postsinápticos Inhibidores , Terminales Presinápticos/ultraestructura , Complejo Olivar Superior/fisiología , Animales , Dendritas/fisiología , Dendritas/ultraestructura , Ratones , Ratones Endogámicos C57BL , Terminales Presinápticos/fisiología , Complejo Olivar Superior/ultraestructura
5.
PLoS Comput Biol ; 11(5): e1004253, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25951120

RESUMEN

The spatial arrangement of Ca2+ channels and vesicles remains unknown for most CNS synapses, despite of the crucial importance of this geometrical parameter for the Ca2+ control of transmitter release. At a large model synapse, the calyx of Held, transmitter release is controlled by several Ca2+ channels in a "domain overlap" mode, at least in young animals. To study the geometrical constraints of Ca2+ channel placement in domain overlap control of release, we used stochastic MCell modelling, at active zones for which the position of docked vesicles was derived from electron microscopy (EM). We found that random placement of Ca2+ channels was unable to produce high slope values between release and presynaptic Ca2+ entry, a hallmark of domain overlap, and yielded excessively large release probabilities. The simple assumption that Ca2+ channels can be located anywhere at active zones, except below a critical distance of ~ 30 nm away from docked vesicles ("exclusion zone"), rescued high slope values and low release probabilities. Alternatively, high slope values can also be obtained by placing all Ca2+ channels into a single supercluster, which however results in significantly higher heterogeneity of release probabilities. We also show experimentally that high slope values, and the sensitivity to the slow Ca2+ chelator EGTA-AM, are maintained with developmental maturation of the calyx synapse. Taken together, domain overlap control of release represents a highly organized active zone architecture in which Ca2+ channels must obey a certain distance to docked vesicles. Furthermore, domain overlap can be employed by near-mature, fast-releasing synapses.


Asunto(s)
Canales de Calcio/metabolismo , Modelos Neurológicos , Sinapsis/metabolismo , Animales , Quelantes del Calcio , Biología Computacional , Simulación por Computador , Ácido Egtácico/análogos & derivados , Técnicas In Vitro , Cinética , Ratones , Ratones Endogámicos C57BL , Procesos Estocásticos , Vesículas Sinápticas/metabolismo , Cuerpo Trapezoide/metabolismo
6.
J Neurosci ; 34(38): 12622-35, 2014 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-25232102

RESUMEN

Ca(2+)-dependent transmitter release occurs in a fast and in a slow phase, but the differential roles of Ca(2+) buffers and Ca(2+) sensors in shaping release kinetics are still controversial. Replacing extracellular Ca(2+) by Sr(2+) causes decreased fast release but enhanced slow release at many synapses. Here, we established presynaptic Sr(2+) uncaging and made quantitative Sr(2+)- and Ca(2+)-imaging experiments at the mouse calyx of Held synapse, to reveal the interplay between Ca(2+) sensors and Ca(2+) buffers in the control of fast and slow release. We show that Sr(2+) activates the fast, Synaptotagmin-2 (Syt2) sensor for vesicle fusion with sixfold lower affinity but unchanged high cooperativity. Surprisingly, Sr(2+) also activates the slow sensor that remains in Syt2 knock-out synapses with a lower efficiency, and Sr(2+) was less efficient than Ca(2+) in the limit of low concentrations in wild-type synapses. Quantitative imaging experiments show that the buffering capacity of the nerve terminal is markedly lower for Sr(2+) than for Ca(2+) (~5-fold). This, together with an enhanced Sr(2+) permeation through presynaptic Ca(2+) channels (~2-fold), admits a drastically higher spatially averaged Sr(2+) transient compared with Ca(2+). Together, despite the lower affinity of Sr(2+) at the fast and slow sensors, the massively higher amplitudes of spatially averaged Sr(2+) transients explain the enhanced late release. This also allows us to conclude that Ca(2+) buffering normally controls late release and prevents the activation of the fast release sensor by residual Ca(2+).


Asunto(s)
Calcio/metabolismo , Terminaciones Nerviosas/fisiología , Estroncio/metabolismo , Transmisión Sináptica/fisiología , Animales , Quelantes/farmacología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Noqueados , Terminaciones Nerviosas/efectos de los fármacos , Terminaciones Nerviosas/metabolismo , Imagen Óptica , Transmisión Sináptica/efectos de los fármacos , Sinaptotagmina II/genética , Sinaptotagmina II/metabolismo
7.
J Neurophysiol ; 113(1): 255-63, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25343783

RESUMEN

The localization and density of voltage-gated Ca(2+) channels at active zones are essential for the amount and kinetics of transmitter release at synapses. RIM proteins are scaffolding proteins at the active zone that bind to several other presynaptic proteins, including voltage-gated Ca(2+) channel α-subunits. The long isoforms of RIM proteins, which contain NH2-terminal Rab3- and Munc13-interacting domains, as well as a central PDZ domain and two COOH-terminal C2 domains, are encoded by two genes, Rim1 and Rim2. Here, we used the ideal accessibility of the large calyx of Held synapse for direct presynaptic electrophysiology to investigate whether the two Rim genes have redundant, or separate, functions in determining the presynaptic Ca(2+) channel density, and the size of a readily releasable vesicle pool (RRP). Quantitative PCR showed that cochlear nucleus neurons, which include calyx of Held generating neurons, express both RIM1 and RIM2. Conditional genetic inactivation of RIM2 at the calyx of Held led to a subtle reduction in presynaptic Ca(2+) current density, whereas deletion of RIM1 was ineffective. The release efficiency of brief presynaptic Ca(2+) "tail" currents and the RRP were unaffected in conditional single RIM1 and RIM2 knockout (KO) mice, whereas both parameters were strongly reduced in RIM1/2 double KO mice. Thus, despite a somewhat more decisive role for RIM2 in determining presynaptic Ca(2+) channel density, RIM1 and RIM2 can overall replace each other's presynaptic functions at a large relay synapse in the hindbrain, the calyx of Held.


Asunto(s)
Canales de Calcio/metabolismo , Núcleo Coclear/fisiología , Proteínas de Unión al GTP/metabolismo , Sinapsis/fisiología , Vesículas Sinápticas/fisiología , Proteínas de Unión al GTP rab3/metabolismo , Animales , Calcio/metabolismo , Núcleo Coclear/efectos de los fármacos , Proteínas de Unión al GTP/genética , Ácido Glutámico/metabolismo , Ratones Noqueados , Técnicas de Placa-Clamp , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/fisiología , Sinapsis/efectos de los fármacos , Vesículas Sinápticas/efectos de los fármacos , Técnicas de Cultivo de Tejidos , Proteínas de Unión al GTP rab3/genética
8.
Elife ; 122023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36655978

RESUMEN

During fear learning, defensive behaviors like freezing need to be finely balanced in the presence or absence of threat-predicting cues (conditioned stimulus, CS). Nevertheless, the circuits underlying such balancing are largely unknown. Here, we investigate the role of the ventral tail striatum (vTS) in auditory-cued fear learning of male mice. In vivo Ca2+ imaging showed that sizable sub-populations of direct (D1R+) and indirect pathway neurons (Adora+) in the vTS responded to footshocks, and to the initiation of movements after freezing; moreover, a sub-population of D1R+ neurons increased its responsiveness to an auditory CS during fear learning. In-vivo optogenetic silencing shows that footshock-driven activity of D1R+ neurons contributes to fear memory formation, whereas Adora+ neurons modulate freezing in the absence of a learned CS. Circuit tracing identified the posterior insular cortex (pInsCx) as an important cortical input to the vTS, and recording of optogenetically evoked EPSCs revealed long-term plasticity with opposite outcomes at the pInsCx synapses onto D1R+ - and Adora+ neurons. Thus, direct- and indirect pathways neurons of the vTS show differential signs of plasticity after fear learning, and balance defensive behaviors in the presence and absence of learned sensory cues.


Asunto(s)
Señales (Psicología) , Aprendizaje , Masculino , Ratones , Animales , Aprendizaje/fisiología , Condicionamiento Clásico/fisiología , Neuronas/fisiología , Miedo/fisiología
9.
Acta Neuropathol ; 123(5): 653-69, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22361813

RESUMEN

α-Synuclein (α-syn) is a presynaptic protein present at most nerve terminals, but its function remains largely unknown. The familial forms of Parkinson's disease associated with multiplications of the α-syn gene locus indicate that overabundance of this protein might have a detrimental effect on dopaminergic transmission. To investigate this hypothesis, we use adeno-associated viral (AAV) vectors to overexpress human α-syn in the rat substantia nigra. Moderate overexpression of either wild-type (WT) or A30P α-syn differs in the motor phenotypes induced, with only the WT form generating hemiparkinsonian impairments. Wild-type α-syn causes a reduction of dopamine release in the striatum that exceeds the loss of dopaminergic neurons, axonal fibers, and the reduction in total dopamine. At the ultrastructural level, the reduced dopamine release corresponds to a decreased density of dopaminergic vesicles and synaptic contacts in striatal terminals. Interestingly, the membrane-binding-deficient A30P mutant does neither notably reduce dopamine release nor it cause ultrastructural changes in dopaminergic axons, showing that α-syn's membrane-binding properties are critically involved in the presynaptic defects. To further determine if the affinity of the protein for membranes determines the extent of motor defects, we compare three forms of α-syn in conditions leading to pronounced degeneration. While membrane-binding α-syns (wild-type and A53T) induce severe motor impairments, an N-terminal deleted form with attenuated affinity for membranes is inefficient in inducing motor defects. Overall, these results demonstrate that α-syn overabundance is detrimental to dopamine neurotransmission at early stages of the degeneration of nigrostriatal dopaminergic axons.


Asunto(s)
Cuerpo Estriado/metabolismo , Dopamina/deficiencia , Proteínas de Filamentos Intermediarios/metabolismo , Actividad Motora/fisiología , Sustancia Negra/metabolismo , Vesículas Sinápticas/metabolismo , Ácido 3,4-Dihidroxifenilacético/metabolismo , Anfetamina/farmacología , Análisis de Varianza , Animales , Apomorfina/farmacología , Cromatografía Líquida de Alta Presión/métodos , Cuerpo Estriado/efectos de los fármacos , Agonistas de Dopamina/farmacología , Inhibidores de Captación de Dopamina/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/ultraestructura , Electroquímica , Ensayo de Inmunoadsorción Enzimática/métodos , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Femenino , Lateralidad Funcional , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Vectores Genéticos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ácido Homovanílico/metabolismo , Humanos , Técnicas In Vitro , Proteínas de Filamentos Intermediarios/genética , Proteínas de Filamentos Intermediarios/ultraestructura , Microscopía Electrónica de Transmisión , Actividad Motora/efectos de los fármacos , Mutación/genética , Potasio/farmacología , Ratas , Ratas Sprague-Dawley , Sustancia Negra/efectos de los fármacos , Sustancia Negra/ultraestructura , Vesículas Sinápticas/ultraestructura , Factores de Tiempo , Transducción Genética , Proteínas de Transporte Vesicular de Monoaminas/metabolismo
10.
Cell Rep ; 39(8): 110850, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35613578

RESUMEN

The medial amygdala (MeA) receives pheromone information about conspecifics and has crucial functions in social behaviors. A previous study showed that activation of GABA neurons in the postero-dorsal MeA (MeApd) with channelrhodopsin-2H134R (ChR2) stimulates inter-male aggression. When performing these experiments using the faster channelrhodopsinH134R,E123T (ChETA), we find the opposite behavioral outcome. A systematic comparison between the two channelrhodopsin variants reveals that optogenetic activation of MeApd GABA neurons with ChETA suppresses aggression, whereas activation under ChR2 increases aggression. Although the mechanism for this paradoxical difference is not understood, we observe that activation of MeApd GABA neurons with ChR2 causes larger plateau depolarizations, smaller action potentials, and larger local inhibition than with ChETA. Thus, the channelrhodopsin variant used for in vivo optogenetic experiments can radically influence the behavioral outcome. Future work should continue to study the role of specific sub-populations of MeApd GABA neurons in aggression control.


Asunto(s)
Neuronas GABAérgicas , Optogenética , Potenciales de Acción/fisiología , Amígdala del Cerebelo , Channelrhodopsins/genética , Humanos , Masculino
11.
Neuroscientist ; : 10738584221108083, 2022 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-35822657

RESUMEN

Unraveling the neuronal mechanisms of fear learning might allow neuroscientists to make links between a learned behavior and the underlying plasticity at specific synaptic connections. In fear learning, an innocuous sensory event such as a tone (called the conditioned stimulus, CS) acquires an emotional value when paired with an aversive outcome (unconditioned stimulus, US). Here, we review earlier studies that have shown that synaptic plasticity at thalamic and cortical afferents to the lateral amygdala (LA) is critical for the formation of auditory-cued fear memories. Despite the early progress, it has remained unclear whether there are separate synaptic inputs that carry US information to the LA to act as a teaching signal for plasticity at CS-coding synapses. Recent findings have begun to fill this gap by showing, first, that thalamic and cortical auditory afferents can also carry US information; second, that the release of neuromodulators contributes to US-driven teaching signals; and third, that synaptic plasticity additionally happens at connections up- and downstream of the LA. Together, a picture emerges in which coordinated synaptic plasticity in serial and parallel circuits enables the formation of a finely regulated fear memory.

12.
J Neurosci ; 30(6): 2007-16, 2010 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-20147529

RESUMEN

The two fundamental forms of short-term plasticity, short-term depression and facilitation, coexist at most synapses, but little is known about their interaction. Here, we studied the interplay between short-term depression and facilitation at calyx of Held synapses. Stimulation at a "low" frequency of 10 or 20 Hz, which is in the range of the spontaneous activity of these auditory neurons in vivo, induced synaptic depression. Surprisingly, an instantaneous increase of the stimulation frequency to 100 or 200 Hz following the low-frequency train uncovered a robust facilitation of EPSCs relative to the predepressed amplitude level. This facilitation decayed rapidly ( approximately 30 ms) and depended on presynaptic residual Ca(2+), but it was not caused by Ca(2+) current facilitation. To probe the release probability of the remaining readily releasable vesicles following the low-frequency train we made presynaptic Ca(2+) uncaging experiments in the predepressed state of the synapse. We found that low-frequency stimulation depletes the fast-releasable vesicle pool (FRP) down to approximately 40% of control and that the remaining FRP vesicles are released with approximately 2-fold slower release kinetics, indicating a hitherto unknown intrinsic heterogeneity among FRP vesicles. Thus, vesicles with an intrinsically lower release probability predominate after low frequency stimulation and undergo facilitation during the onset of subsequent high-frequency trains. Facilitation in the predepressed state of the synapse might help to stabilize the amount of transmitter release at the onset of high-frequency firing at these auditory synapses.


Asunto(s)
Vías Auditivas/fisiología , Tronco Encefálico/fisiología , Sinapsis/fisiología , Estimulación Acústica , Potenciales de Acción , Animales , Calcio/fisiología , Potenciales Postsinápticos Excitadores , Técnicas In Vitro , Plasticidad Neuronal , Neurotransmisores/metabolismo , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Vesículas Sinápticas/fisiología
13.
J Neurosci ; 30(40): 13281-90, 2010 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-20926654

RESUMEN

In pre-hearing mice, vesicle exocytosis at cochlear inner hair cell (IHC) ribbon synapses is triggered by spontaneous Ca(2+) spikes. At the onset of hearing, IHC exocytosis is then exclusively driven by graded potentials, and is characterized by higher Ca(2+) efficiency and improved synchronization of vesicular release. The molecular players involved in this transition are still unknown. Here we addressed the involvement of synaptotagmins and otoferlin as putative Ca(2+) sensors in IHC exocytosis during postnatal maturation of the cochlea. Using cell capacitance measurements, we showed that Ca(2+)-evoked exocytosis in mouse IHCs switches from an otoferlin-independent to an otoferlin-dependent mechanism at postnatal day 4. During this early exocytotic period, several synaptotagmins (Syts), including Syt1, Syt2 and Syt7, were detected in IHCs. The exocytotic response as well as the release of the readily releasable vesicle pool (RRP) was, however, unchanged in newborn mutant mice lacking Syt1, Syt2 or Syt7 (Syt1(-/-), Syt2(-/-) and Syt7(-/-) mice). We only found a defect in RRP recovery in Syt1(-/-) mice which was apparent as a strongly reduced response to repetitive stimulations. In post-hearing Syt2(-/-) and Syt7(-/-) mutant mice, IHC synaptic exocytosis was unaffected. The transient expression of Syt1 and Syt2, which were no longer detected in IHCs after the onset of hearing, indicates that these two most common Ca(2+)-sensors in CNS synapses are not involved in mature IHCs. We suggest that otoferlin underlies highly efficient Ca(2+)-dependent membrane-membrane fusion, a process likely essential to increase the probability and synchrony of vesicle fusion events at the mature IHC ribbon synapse.


Asunto(s)
Cóclea/crecimiento & desarrollo , Exocitosis , Células Ciliadas Auditivas Internas/fisiología , Proteínas de la Membrana/fisiología , Sinaptotagmina II/fisiología , Sinaptotagmina I/fisiología , Animales , Calcio/fisiología , Señalización del Calcio/genética , Senescencia Celular/genética , Senescencia Celular/fisiología , Cóclea/citología , Capacidad Eléctrica , Exocitosis/genética , Femenino , Células Ciliadas Auditivas Internas/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Sinapsis/genética , Sinapsis/fisiología , Transmisión Sináptica/genética , Sinaptotagmina I/genética , Sinaptotagmina II/genética
14.
Nature ; 435(7041): 497-501, 2005 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-15917809

RESUMEN

Neurotransmitter release is triggered by an increase in the cytosolic Ca2+ concentration ([Ca2+]i), but it is unknown whether the Ca2+-sensitivity of vesicle fusion is modulated during synaptic plasticity. We investigated whether the potentiation of neurotransmitter release by phorbol esters, which target presynaptic protein kinase C (PKC)/munc-13 signalling cascades, exerts a direct effect on the Ca2+-sensitivity of vesicle fusion. Using direct presynaptic Ca2+-manipulation and Ca2+ uncaging at a giant presynaptic terminal, the calyx of Held, we show that phorbol esters potentiate transmitter release by increasing the apparent Ca2+-sensitivity of vesicle fusion. Phorbol esters potentiate Ca2+-evoked release as well as the spontaneous release rate. We explain both effects by an increased fusion 'willingness' in a new allosteric model of Ca2+-activation of vesicle fusion. In agreement with an allosteric mechanism, we observe that the classically high Ca2+ cooperativity in triggering vesicle fusion (approximately 4) is gradually reduced below 3 microM [Ca2+]i, reaching a value of <1 at basal [Ca2+]i. Our data indicate that spontaneous transmitter release close to resting [Ca2+]i is a consequence of an intrinsic property of the molecular machinery that mediates synaptic vesicle fusion.


Asunto(s)
Calcio/metabolismo , Fusión de Membrana , Neurotransmisores/metabolismo , Terminales Presinápticos/metabolismo , Sinapsis/metabolismo , Regulación Alostérica/efectos de los fármacos , Animales , Tronco Encefálico/citología , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/fisiología , Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Células Cromafines/citología , Células Cromafines/efectos de los fármacos , Células Cromafines/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Técnicas In Vitro , Fusión de Membrana/efectos de los fármacos , Modelos Biológicos , Forbol 12,13-Dibutirato/farmacología , Terminales Presinápticos/efectos de los fármacos , Ratas , Sinapsis/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos
15.
Mol Cell Neurosci ; 44(4): 374-85, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20470891

RESUMEN

The large glutamatergic calyx of Held synapse in the auditory brainstem has become a powerful model for studying transmitter release mechanisms, but the molecular bases of presynaptic function at this synapse are not well known. Here, we have used single-cell quantitative PCR (qPCR) to study the developmental expression of all major Synaptotagmin (Syt) isoforms in putative calyx of Held-generating neurons (globular bushy cells) of the ventral cochlear nucleus. Using electrophysiological criteria and the expression of marker genes including VGluTs (vesicular glutamate transporters), Ca(2+) binding proteins, and the transcription factor Math5, we identified a subset of the recorded neurons as putative calyx of Held-generating bushy cells. At postnatal days 12-15 these neurons expressed Syt-2 and Syt-11, and also Syt-3, -4, -7 and -13 at lower levels, whereas Syt-1 and -9 were absent. Interestingly, early in development (at P3-P6), immature bushy cells expressed a larger number of Syt-isoforms, with Syt-1, Syt-5, Syt-9 and Syt-13 detected in a significantly higher percentage of neurons. Our study sheds light on the molecular properties of putative calyx of Held-generating neurons and shows the developmental regulation of the Syt-isoform expression profile in a single neuron type.


Asunto(s)
Tronco Encefálico/crecimiento & desarrollo , Tronco Encefálico/metabolismo , Neuronas/metabolismo , Sinapsis/metabolismo , Sinaptotagminas/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Vías Auditivas/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/análisis , Tronco Encefálico/citología , Proteínas de Unión al Calcio/análisis , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Neuronas/citología , Terminales Presinápticos/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Ratas Wistar , Sinaptotagminas/genética , Proteínas de Transporte Vesicular de Glutamato/análisis
16.
Sci Rep ; 10(1): 10047, 2020 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-32572071

RESUMEN

Parvalbumin (PV)-expressing interneurons (PV-INs) mediate well-timed inhibition of cortical principal neurons, and plasticity of these interneurons is involved in map remodeling of primary sensory cortices during critical periods of development. To assess whether bone morphogenetic protein (BMP) signaling contributes to the developmental acquisition of the synapse- and plasticity properties of PV-INs, we investigated conditional/conventional double KO mice of BMP-receptor 1a (BMPR1a; targeted to PV-INs) and 1b (BMPR1a/1b (c)DKO mice). We report that spike-timing dependent LTP at the synapse between PV-INs and principal neurons of layer 4 in the auditory cortex was absent, concomitant with a decreased paired-pulse ratio (PPR). On the other hand, baseline synaptic transmission at this connection, and action potential (AP) firing rates of PV-INs were unchanged. To explore possible gene expression targets of BMP signaling, we measured the mRNA levels of the BDNF receptor TrkB and of P/Q-type Ca2+ channel α-subunits, but did not detect expression changes of the corresponding genes in PV-INs of BMPR1a/1b (c)DKO mice. Our study suggests that BMP-signaling in PV-INs during and shortly after the critical period is necessary for the expression of LTP at PV-IN output synapses, involving gene expression programs that need to be addressed in future work.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Interneuronas/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Parvalbúminas/metabolismo , Animales , Corteza Auditiva/metabolismo , Femenino , Técnicas de Inactivación de Genes , Potenciación a Largo Plazo , Masculino , Ratones , Transducción de Señal
17.
Cell Rep ; 33(6): 108359, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33176133

RESUMEN

Activation of the basal forebrain (BF) has been associated with increased attention, arousal, and a heightened cortical representation of the external world. In addition, BF has been implicated in the regulation of the default mode network (DMN) and associated behaviors. Here, we provide causal evidence for a role of BF in DMN regulation, highlighting a prominent role of parvalbumin (PV) GABAergic neurons. The optogenetic activation of BF PV neurons reliably drives animals toward DMN-like behaviors, with no effect on memory encoding. In contrast, BF electrical stimulation enhances memory performance and increases DMN-like behaviors. BF stimulation has a correlated impact on peptide regulation in the BF and ACC, enhancing peptides linked to grooming behavior and memory functions, supporting a crucial role of the BF in DMN regulation. We suggest that in addition to enhancing attentional functions, the BF harbors a network encompassing PV GABAergic neurons that promotes self-directed behaviors associated with the DMN.


Asunto(s)
Prosencéfalo Basal/metabolismo , Red en Modo Predeterminado/fisiopatología , Optogenética/métodos , Parvalbúminas/metabolismo , Animales , Modelos Animales de Enfermedad , Ratas
18.
J Neurosci ; 28(33): 8257-67, 2008 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-18701688

RESUMEN

Diacylglycerol (DAG) and phorbol esters strongly potentiate transmitter release at synapses by activating protein kinase C (PKC) and members of the Munc13 family of presynaptic vesicle priming proteins. This PKC/Munc13 pathway has emerged as a crucial regulator of release probability during various forms of activity-dependent enhancement of release. Here, we investigated the relative roles of PKC and Munc13-1 in the phorbol ester potentiation of evoked and spontaneous transmitter release at the calyx of Held synapse. The phorbol ester phorbol 12,13-dibutyrate (1 microM) potentiated the frequency of miniature EPSCs, and the amplitudes of evoked EPSCs with a similar time course. Preincubating slices with the PKC blocker Ro31-82200 reduced the potentiation, mainly by affecting a late phase of the phorbol ester potentiation. The Ro31-8220-insensitive potentiation was most likely mediated by Munc13-1, because in organotypic slices of Munc13-1(H567K) knock-in mice, in which DAG binding to Munc13-1 is abolished, the potentiation of spontaneous release by phorbol ester was strongly suppressed. Using direct presynaptic depolarizations in paired recordings, we show that the phorbol ester potentiation does not go along with an increase in the number of readily releasable vesicles, despite an increase in the cumulative EPSC amplitude during 100 Hz stimulation trains. Our data indicate that activation of Munc13 and PKC both contribute to an enhancement of the fusion probability of readily releasable vesicles. Thus, docked and readily releasable vesicles are a substrate for modulation via intracellular second-messenger pathways that act via Munc13 and PKC.


Asunto(s)
Calcio/metabolismo , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Neurotransmisores/metabolismo , Ésteres del Forbol/farmacología , Proteína Quinasa C/metabolismo , Animales , Sinergismo Farmacológico , Potenciales Postsinápticos Excitadores/genética , Indoles/farmacología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Neurotransmisores/deficiencia , Neurotransmisores/genética , Técnicas de Cultivo de Órganos , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/enzimología , Terminales Presinápticos/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Ratas , Ratas Wistar
19.
J Physiol ; 587(Pt 12): 3009-23, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19403608

RESUMEN

Developmental refinement of synaptic transmission can occur via changes in several pre- and postsynaptic factors, but it has been unknown whether the intrinsic Ca2+ sensitivity of vesicle fusion in the nerve terminal can be regulated during development. Using the calyx of Held, a giant synapse in the auditory pathway, we studied the presynaptic mechanisms underlying the developmental regulation of Ca2+-secretion coupling, comparing a time period before, and shortly after the onset of hearing in rats. We found an approximately 2-fold leftward shift in the relationship between EPSC amplitude and presynaptic Ca2+ current charge (QCa), indicating that brief presynaptic Ca2+ currents become significantly more efficient in driving release. Using a Ca2+ tail current protocol, we also found that the high cooperativity between EPSC amplitude and QCa was slightly reduced with development. In contrast, in presynaptic Ca2+ uncaging experiments, the intrinsic Ca2+ cooperativity of vesicle fusion was identical, and the intrinsic Ca2+ sensitivity was slightly reduced with development. This indicates that the significantly enhanced release efficiency of brief Ca2+ currents must be caused by a tighter co-localization of Ca2+ channels and readily releasable vesicles, but not by changes in the intrinsic properties of Ca2+-dependent release. Using the parameters of the intrinsic Ca2+ sensitivity measured at each developmental stage, we estimate that during a presynaptic action potential (AP), a given readily releasable vesicle experiences an about 1.3-fold higher 'local' intracellular Ca2+ concentration ([Ca2+]i) signal with development. Thus, the data indicate a tightening in the Ca2+ channel-vesicle co-localization during development, without a major change in the intrinsic Ca2+ sensitivity of vesicle fusion.


Asunto(s)
Vías Auditivas/fisiología , Tronco Encefálico/fisiología , Señalización del Calcio/fisiología , Calcio/fisiología , Vesículas Sinápticas/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Envejecimiento/fisiología , Animales , Vías Auditivas/crecimiento & desarrollo , Tronco Encefálico/crecimiento & desarrollo , Canales de Calcio/fisiología , Interpretación Estadística de Datos , Electrofisiología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Técnicas In Vitro , Neurotransmisores/metabolismo , Neurotransmisores/fisiología , Técnicas de Placa-Clamp , Ratas , Ratas Wistar
20.
Dev Neurobiol ; 79(2): 155-174, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30548566

RESUMEN

Large excitatory synapses are found at specific points in the neuronal circuits of the auditory brainstem, to enable fast information transfer and the preservation of acoustic timing information. The extracellular cues and signaling mechanisms that lead to the development of these specialized synaptic connections, exemplified by the calyx of Held in the medial nucleus of the trapezoid body (MNTB), are still largely unknown. Here, we investigate the role of BMP signaling for the early development of the ventral cochlear nucleus (VCN) and MNTB, and for the initial formation of the calyx of Held synaptic connection. We used conditional alleles of two BMP type-1 receptors in the background of a constitutive BMPR1b knock-out (KO), or else a conditional allele of SMAD4. The conditional alleles were recombined by the Krox20Cre mouse line that is active around mid-gestation in rhombomeres (r) 3 and 5 from which the VCN and MNTB are derived; alternatively, virus-mediated Cre-expression was performed early postnatally in the VCN. The data show that embryonic SMAD-dependent BMP-signaling in r3 and r5 contributes to the histogenesis of auditory brainstem nuclei. On the other hand, BMP-receptor signaling early postnatally in presynaptic neurons of the calyx of Held projection is necessary for correct axon branch retraction, which suggests a cell-autonomous role of presynaptic BMP-receptors in synapse elimination at the developing calyx of Held. Thus, our work dissects developmentally early and late roles of BMP-signaling for the formation of auditory brainstem nuclei, and the highly specialized synaptic connectivity in these structures.


Asunto(s)
Vías Auditivas/fisiología , Tronco Encefálico/fisiología , Neuronas/fisiología , Transducción de Señal , Sinapsis/fisiología , Animales , Axones/fisiología , Tronco Encefálico/crecimiento & desarrollo , Ratones Transgénicos , Transducción de Señal/fisiología , Transmisión Sináptica/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA