Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Neurosci ; 43(32): 5741-5752, 2023 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-37474311

RESUMEN

Neurotransmission is shaped by extracellular pH. Alkalization enhances pH-sensitive transmitter release and receptor activation, whereas acidification inhibits these processes and can activate acid-sensitive conductances in the synaptic cleft. Previous work has shown that the synaptic cleft can either acidify because of synaptic vesicular release and/or alkalize because of Ca2+ extrusion by the plasma membrane ATPase (PMCA). The direction of change differs across synapse types. At the mammalian neuromuscular junction (NMJ), the direction and magnitude of pH transients in the synaptic cleft during transmission remain ambiguous. We set out to elucidate the extracellular pH transients that occur at this cholinergic synapse under near-physiological conditions and identify their sources. We monitored pH-dependent changes in the synaptic cleft of the mouse levator auris longus using viral expression of the pseudoratiometric probe pHusion-Ex in the muscle. Using mice from both sexes, a significant and prolonged alkalization occurred when stimulating the connected nerve for 5 s at 50 Hz, which was dependent on postsynaptic intracellular Ca2+ release. Sustained stimulation for a longer duration (20 s at 50 Hz) caused additional prolonged net acidification at the cleft. To investigate the mechanism underlying cleft alkalization, we used muscle-expressed GCaMP3 to monitor the contribution of postsynaptic Ca2+ Activity-induced liberation of intracellular Ca2+ in muscle positively correlated with alkalization of the synaptic cleft, whereas inhibiting PMCA significantly decreased the extent of cleft alkalization. Thus, cholinergic synapses of the mouse NMJ typically alkalize because of cytosolic Ca2+ liberated in muscle during activity, unless under highly strenuous conditions where acidification predominates.SIGNIFICANCE STATEMENT Changes in synaptic cleft pH alter neurotransmission, acting on receptors and channels on both sides of the synapse. Synaptic acidification has been associated with a myriad of diseases in the central and peripheral nervous system. Here, we report that in near-physiological recording conditions the cholinergic neuromuscular junction shows use-dependent bidirectional changes in synaptic cleft pH-immediate alkalinization and a long-lasting acidification under prolonged stimulation. These results provide further insight into physiologically relevant changes at cholinergic synapses that have not been defined previously. Understanding and identifying synaptic pH transients during and after neuronal activity provides insight into short-term synaptic plasticity synapses and may identify therapeutic targets for diseases.


Asunto(s)
Calcio , Sinapsis , Femenino , Masculino , Animales , Ratones , Calcio/metabolismo , Sinapsis/fisiología , Unión Neuromuscular/metabolismo , Transmisión Sináptica/fisiología , Colinérgicos , Mamíferos
2.
J Physiol ; 601(24): 5705-5732, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37942946

RESUMEN

Motor neurons are the longest neurons in the body, with axon terminals separated from the soma by as much as a meter. These terminals are largely autonomous with regard to their bioenergetic metabolism and must burn energy at a high rate to sustain muscle contraction. Here, through computer simulation and drawing on previously published empirical data, we determined that motor neuron terminals in Drosophila larvae experience highly volatile power demands. It might not be surprising then, that we discovered the mitochondria in the motor neuron terminals of both Drosophila and mice to be heavily decorated with phosphagen kinases - a key element in an energy storage and buffering system well-characterized in fast-twitch muscle fibres. Knockdown of arginine kinase 1 (ArgK1) in Drosophila larval motor neurons led to several bioenergetic deficits, including mitochondrial matrix acidification and a faster decline in the cytosol ATP to ADP ratio during axon burst firing. KEY POINTS: Neurons commonly fire in bursts imposing highly volatile demands on the bioenergetic machinery that generates ATP. Using a computational approach, we built profiles of presynaptic power demand at the level of single action potentials, as well as the transition from rest to sustained activity. Phosphagen systems are known to buffer ATP levels in muscles and we demonstrate that phosphagen kinases, which support such phosphagen systems, also localize to mitochondria in motor nerve terminals of fruit flies and mice. By knocking down phosphagen kinases in fruit fly motor nerve terminals, and using fluorescent reporters of the ATP:ADP ratio, lactate, pH and Ca2+ , we demonstrate a role for phosphagen kinases in stabilizing presynaptic ATP levels. These data indicate that the maintenance of phosphagen systems in motor neurons, and not just muscle, could be a beneficial initiative in sustaining musculoskeletal health and performance.


Asunto(s)
Mitocondrias , Terminales Presinápticos , Animales , Ratones , Simulación por Computador , Mitocondrias/metabolismo , Terminales Presinápticos/fisiología , Neuronas Motoras/fisiología , Drosophila/metabolismo , Adenosina Trifosfato/metabolismo
3.
J Neurosci ; 40(8): 1611-1624, 2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-31964719

RESUMEN

The dogma that the synaptic cleft acidifies during neurotransmission is based on the corelease of neurotransmitters and protons from synaptic vesicles, and is supported by direct data from sensory ribbon-type synapses. However, it is unclear whether acidification occurs at non-ribbon-type synapses. Here we used genetically encoded fluorescent pH indicators to examine cleft pH at conventional neuronal synapses. At the neuromuscular junction of female Drosophila larvae, we observed alkaline spikes of over 1 log unit during fictive locomotion in vivo. Ex vivo, single action potentials evoked alkalinizing pH transients of only ∼0.01 log unit, but these transients summated rapidly during burst firing. A chemical pH indicator targeted to the cleft corroborated these findings. Cleft pH transients were dependent on Ca2+ movement across the postsynaptic membrane, rather than neurotransmitter release per se, a result consistent with cleft alkalinization being driven by the Ca2+/H+ antiporting activity of the plasma membrane Ca2+-ATPase at the postsynaptic membrane. Targeting the pH indicators to the microenvironment of the presynaptic voltage gated Ca2+ channels revealed that alkalinization also occurred within the cleft proper at the active zone and not just within extrasynaptic regions. Application of the pH indicators at the mouse calyx of Held, a mammalian central synapse, similarly revealed cleft alkalinization during burst firing in both males and females. These findings, made at two quite different non-ribbon type synapses, suggest that cleft alkalinization during neurotransmission, rather than acidification, is a generalizable phenomenon across conventional neuronal synapses.SIGNIFICANCE STATEMENT Neurotransmission is highly sensitive to the pH of the extracellular milieu. This is readily evident in the neurological symptoms that accompany systemic acid/base imbalances. Imaging data from sensory ribbon-type synapses show that neurotransmission itself can acidify the synaptic cleft, likely due to the corelease of protons and glutamate. It is not clear whether the same phenomenon occurs at conventional neuronal synapses due to the difficulties in collecting such data. If it does occur, it would provide for an additional layer of activity-dependent modulation of neurotransmission. Our findings of alkalinization, rather than acidification, within the cleft of two different neuronal synapses encourages a reassessment of the scope of activity-dependent pH influences on neurotransmission and short-term synaptic plasticity.


Asunto(s)
Ácido Glutámico/metabolismo , Unión Neuromuscular/metabolismo , Neuronas/metabolismo , Transmisión Sináptica/fisiología , Animales , Drosophila , Femenino , Concentración de Iones de Hidrógeno , Plasticidad Neuronal/fisiología , Vesículas Sinápticas/metabolismo
4.
J Neurophysiol ; 126(4): 976-996, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34432991

RESUMEN

A considerable amount of energy is expended following presynaptic activity to regenerate electrical polarization and maintain efficient release and recycling of neurotransmitter. Mitochondria are the major suppliers of neuronal energy, generating ATP via oxidative phosphorylation. However, the specific utilization of energy from cytosolic glycolysis rather than mitochondrial respiration at the presynaptic terminal during synaptic activity remains unclear and controversial. We use a synapse specialized for high-frequency transmission in mice, the calyx of Held, to test the sources of energy used to maintain energy during short activity bursts (<1 s) and sustained neurotransmission (30-150 s). We dissect the role of presynaptic glycolysis versus mitochondrial respiration by acutely and selectively blocking these ATP-generating pathways in a synaptic preparation where mitochondria and synaptic vesicles are prolific, under near-physiological conditions. Surprisingly, if either glycolysis or mitochondrial ATP production is intact, transmission during repetitive short bursts of activity is not affected. In slices from young animals before the onset of hearing, where the synapse is not yet fully specialized, both glycolytic and mitochondrial ATP production are required to support sustained, high-frequency neurotransmission. In mature synapses, sustained transmission relies exclusively on mitochondrial ATP production supported by bath lactate, but not glycolysis. At both ages, we observe that action potential propagation begins to fail before defects in synaptic vesicle recycling. Our data describe a specific metabolic profile to support high-frequency information transmission at the mature calyx of Held, shifting during postnatal synaptic maturation from glycolysis to rely on monocarboxylates as a fuel source.NEW & NOTEWORTHY We dissect the role of presynaptic glycolysis versus mitochondrial respiration in supporting high-frequency neurotransmission, by acutely blocking these ATP-generating pathways at a synapse tuned for high-frequency transmission. We find that massive energy expenditure is required to generate failure when only one pathway is inhibited. Action potential propagation is lost before impaired synaptic vesicle recycling. Synaptic transmission is exclusively dependent on oxidative phosphorylation in mature synapses, indicating presynaptic glycolysis may be dispensable for ATP maintenance.


Asunto(s)
Núcleo Coclear/metabolismo , Glucólisis/fisiología , Mitocondrias/metabolismo , Respiración , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL
5.
J Physiol ; 596(24): 6263-6287, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30285293

RESUMEN

KEY POINTS: This study characterizes the mechanisms underlying defects in synaptic transmission when dynamin-related protein 1 (DRP1) is genetically eliminated. Viral-mediated knockout of DRP1 from the presynaptic terminal at the mouse calyx of Held increased initial release probability, reduced the size of the synaptic vesicle recycling pool and impaired synaptic vesicle recycling. Transmission defects could be partially restored by increasing the intracellular calcium buffering capacity with EGTA-AM, implying close coupling of Ca2+ channels to synaptic vesicles was compromised. Acute restoration of ATP to physiological levels in the presynaptic terminal did not reverse the synaptic defects. Loss of DRP1 impairs mitochondrial morphology in the presynaptic terminal, which in turn seems to arrest synaptic maturation. ABSTRACT: Impaired mitochondrial biogenesis and function is implicated in many neurodegenerative diseases, and likely affects synaptic neurotransmission prior to cellular loss. Dynamin-related protein 1 (DRP1) is essential for mitochondrial fission and is disrupted in neurodegenerative disease. In this study, we used the mouse calyx of Held synapse as a model to investigate the impact of presynaptic DRP1 loss on synaptic vesicle (SV) recycling and sustained neurotransmission. In vivo viral expression of Cre recombinase in ventral cochlear neurons of floxed-DRP1 mice generated a presynaptic-specific DRP1 knockout (DRP1-preKO), where the innervated postsynaptic cell was unperturbed. Confocal reconstruction of the calyx terminal suggested SV clusters and mitochondrial content were disrupted, and presynaptic terminal volume was decreased. Using postsynaptic voltage-clamp recordings, we found that DRP1-preKO synapses had larger evoked responses at low frequency stimulation. DRP1-preKO synapses also had profoundly altered short-term plasticity, due to defects in SV recycling. Readily releasable pool size, estimated with high-frequency trains, was dramatically reduced in DRP1-preKO synapses, suggesting an important role for DRP1 in maintenance of release-competent SVs at the presynaptic terminal. Presynaptic Ca2+ accumulation in the terminal was also enhanced in DRP1-preKO synapses. Synaptic transmission defects could be partially rescued with EGTA-AM, indicating close coupling of Ca2+ channels to SV distance normally found in mature terminals may be compromised by DRP1-preKO. Using paired recordings of the presynaptic and postsynaptic compartments, recycling defects could not be reversed by acute dialysis of ATP into the calyx terminals. Taken together, our results implicate a requirement for mitochondrial fission to coordinate postnatal synapse maturation.


Asunto(s)
Dinaminas/metabolismo , Neuronas/fisiología , Terminales Presinápticos/metabolismo , Vesículas Sinápticas/fisiología , Adenoviridae , Animales , Calcio/metabolismo , Células Cultivadas , Dinaminas/genética , Femenino , Colorantes Fluorescentes , Masculino , Ratones , Ratones Noqueados , Mitocondrias
6.
Synapse ; 72(12): e22040, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29935099

RESUMEN

Synaptic vesicle (SV) exocytosis is intimately dependent on free local Ca2+ near active zones. Genetically encoded calcium indicators (GECIs) have become an indispensable tool to monitor calcium dynamics during physiological responses, and they are widely used as a proxy to monitor activity in neuronal ensembles and at synaptic terminals. However, GECIs' ability to bind Ca2+ at physiologically relevant concentration makes them strong candidates to affect calcium homeostasis and alter synaptic transmission by exogenously increasing Ca2+ buffering. In the present study, we show that genetically expressed GCaMP6m modulates SV release probability at the mouse calyx of Held synapse. GCaMP6m expression for approximately three weeks decreased initial SV release for both low-frequency stimulation and high-frequency stimulation trains, and slowed presynaptic short-term depression. However, GCaMP6m does not affect quantal events during spontaneous activity at this synapse. This study emphasizes the careful use of GECIs as monitors of neuronal activity and inspects the role of these transgenic indicators which may alter calcium-dependent physiological responses.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Exocitosis , Vesículas Sinápticas/metabolismo , Animales , Calcio/metabolismo , Proteínas de Unión al Calcio/genética , Femenino , Genes Reporteros , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Terminales Presinápticos/metabolismo
7.
PLoS Comput Biol ; 12(4): e1004855, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27035349

RESUMEN

The readily releasable pool (RRP) of vesicles is a core concept in studies of presynaptic function. However, operating principles lack consensus definition and the utility for quantitative analysis has been questioned. Here we confirm that RRPs at calyces of Held from 14 to 21 day old mice have a fixed capacity for storing vesicles that is not modulated by Ca2+. Discrepancies with previous studies are explained by a dynamic flow-through pool, established during heavy use, containing vesicles that are released with low probability despite being immediately releasable. Quantitative analysis ruled out a posteriori explanations for the vesicles with low release probability, such as Ca2+-channel inactivation, and established unexpected boundary conditions for remaining alternatives. Vesicles in the flow-through pool could be incompletely primed, in which case the full sequence of priming steps downstream of recruitment to the RRP would have an average unitary rate of at least 9/s during heavy use. Alternatively, vesicles with low and high release probability could be recruited to distinct types of release sites; in this case the timing of recruitment would be similar at the two types, and the downstream transition from recruited to fully primed would be much faster. In either case, further analysis showed that activity accelerates the upstream step where vesicles are initially recruited to the RRP. Overall, our results show that the RRP can be well defined in the mathematical sense, and support the concept that the defining mechanism is a stable group of autonomous release sites.


Asunto(s)
Vías Auditivas/fisiología , Modelos Neurológicos , Cuerpo Trapezoide/fisiología , Animales , Calcio/metabolismo , Núcleo Coclear/fisiología , Biología Computacional , Simulación por Computador , Estimulación Eléctrica , Potenciales Postsinápticos Excitadores , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal , Terminales Presinápticos/fisiología , Vesículas Sinápticas/fisiología
8.
J Neurophysiol ; 116(6): 2523-2540, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27605535

RESUMEN

Mitochondria are major suppliers of cellular energy in neurons; however, utilization of energy from glycolysis vs. mitochondrial oxidative phosphorylation (OxPhos) in the presynaptic compartment during neurotransmission is largely unknown. Using presynaptic and postsynaptic recordings from the mouse calyx of Held, we examined the effect of acute selective pharmacological inhibition of glycolysis or mitochondrial OxPhos on multiple mechanisms regulating presynaptic function. Inhibition of glycolysis via glucose depletion and iodoacetic acid (1 mM) treatment, but not mitochondrial OxPhos, rapidly altered transmission, resulting in highly variable, oscillating responses. At reduced temperature, this same treatment attenuated synaptic transmission because of a smaller and broader presynaptic action potential (AP) waveform. We show via experimental manipulation and ion channel modeling that the altered AP waveform results in smaller Ca2+ influx, resulting in attenuated excitatory postsynaptic currents (EPSCs). In contrast, inhibition of mitochondria-derived ATP production via extracellular pyruvate depletion and bath-applied oligomycin (1 µM) had no significant effect on Ca2+ influx and did not alter the AP waveform within the same time frame (up to 30 min), and the resultant EPSC remained unaffected. Glycolysis, but not mitochondrial OxPhos, is thus required to maintain basal synaptic transmission at the presynaptic terminal. We propose that glycolytic enzymes are closely apposed to ATP-dependent ion pumps on the presynaptic membrane. Our results indicate a novel mechanism for the effect of hypoglycemia on neurotransmission. Attenuated transmission likely results from a single presynaptic mechanism at reduced temperature: a slower, smaller AP, before and independent of any effect on synaptic vesicle release or receptor activity.


Asunto(s)
Potenciales de Acción/fisiología , Glucólisis/fisiología , Terminales Presinápticos/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Animales Recién Nacidos , Antimetabolitos/farmacología , Tronco Encefálico/citología , Células Cultivadas , Corteza Cerebral/citología , Desoxiglucosa/farmacología , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Glucosa/farmacología , Glucólisis/efectos de los fármacos , Ácidos Indolacéticos/farmacología , Ácido Yodoacético/farmacología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Neurológicos , Neuronas/efectos de los fármacos , Oligomicinas/farmacología , Terminales Presinápticos/efectos de los fármacos
9.
J Neurosci ; 33(22): 9402-7, 2013 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-23719808

RESUMEN

Auditory neuropathies are linked to loss of temporal acuity of sound-evoked signals, which may be related to myelin loss. However, it is not known how myelin loss affects the waveform and temporal precision of action potentials (APs) in auditory CNS nerve terminals. Here we investigated the excitability of the calyx of Held nerve terminal in dysmyelinated auditory brainstems using the Long-Evans Shaker (LES) rat, a spontaneous mutant where compact myelin wrapping does not occur due to a genetic deletion of myelin basic protein. We found at relatively mature postnatal ages (15-17 d after birth) LES rat calyces showed prolonged spike latencies, indicative of a threefold reduction in the AP propagation velocity. Furthermore, LES rat afferent fiber-evoked APs showed a pronounced loss of temporal precision, even at low stimulation frequencies (10 Hz). While normal calyces were able to fire APs without failures at impressive rates of up to 1 kHz, LES calyces were unable to do so. Direct recordings of the presynaptic calyx terminal AP waveform revealed that myelin loss does not affect the AP spike upstroke and downstroke kinetics, but dysmyelination reduces the after-depolarization and enhances the fast after-hyperpolarization peak following the AP spike in the LES rat. Together these findings show that proper myelination is essential not only for fast AP propagation, but also for precise presynaptic AP firing that minimizes both spike jitter and failures, two characteristics critically important for the accurate processing of sound signals in the auditory brainstem.


Asunto(s)
Potenciales de Acción/fisiología , Axones/patología , Enfermedades Desmielinizantes/patología , Neuronas Aferentes/patología , Animales , Tronco Encefálico/fisiología , Fenómenos Electrofisiológicos , Femenino , Inmunohistoquímica , Técnicas In Vitro , Masculino , Vaina de Mielina/fisiología , Fibras Nerviosas Mielínicas/fisiología , Técnicas de Placa-Clamp , Terminales Presinápticos/fisiología , Ratas , Ratas Long-Evans
10.
Sci Prog ; 107(1): 368504231225066, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38196184

RESUMEN

Proton concentration can change within the cleft during synaptic activity due to vesicular release and Ca2+ extrusion from cellular compartments. These changes within the synaptic cleft can impact neural activity by proton-dependent modulation of ion channel function. The pH transient differs in magnitude and direction between synapses, requiring different synapse types to be measured to generate a complete understanding of this mechanism and its impacts on physiology. With a focus on the mouse neuromuscular junction (NMJ), the recently published "Postsynaptic Calcium Extrusion at the Mouse Neuromuscular Junction Alkalinizes the Synaptic Cleft" measured synaptic cleft pH at a cholinergic synapse and found a biphasic pH transient. The study demonstrated that the changes in proton concentration found were due to postsynaptic signaling when measuring pH at the muscle membrane, despite the expectation of a presynaptic contribution. This result suggests a diffusional barrier within the NMJ isolates pH transients to presynaptic versus postsynaptic compartments. Generating a Donnan equilibrium that impacts protons, evidence suggests the basal lamina may be a key regulator of pH at the NMJ. Exploring synaptic pH, proton regulating factors, and downstream pH transient effects at presynaptic versus postsynaptic membranes may lead to new insight for a variety of diseases.


Asunto(s)
Unión Neuromuscular , Protones , Animales , Ratones , Membrana Basal , Concentración de Iones de Hidrógeno , Transducción de Señal
11.
Commun Biol ; 7(1): 287, 2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38459113

RESUMEN

Dynamic changes in astrocyte Ca2+ are recognized as contributors to functional hyperemia, a critical response to increased neuronal activity mediated by a process known as neurovascular coupling (NVC). Although the critical role of glutamatergic signaling in this process has been extensively investigated, the impact of behavioral state, and the release of behavior-associated neurotransmitters, such as norepinephrine and serotonin, on astrocyte Ca2+ dynamics and functional hyperemia have received less attention. We used two-photon imaging of the barrel cortex in awake mice to examine the role of noradrenergic and serotonergic projections in NVC. We found that both neurotransmitters facilitated sensory stimulation-induced increases in astrocyte Ca2+. Interestingly, while ablation of serotonergic neurons reduced sensory stimulation-induced functional hyperemia, ablation of noradrenergic neurons caused both attenuation and potentiation of functional hyperemia. Our study demonstrates that norepinephrine and serotonin are involved in modulating sensory stimulation-induced astrocyte Ca2+ elevations and identifies their differential effects in regulating functional hyperemia.


Asunto(s)
Neuronas Adrenérgicas , Hiperemia , Acoplamiento Neurovascular , Ratones , Animales , Acoplamiento Neurovascular/fisiología , Serotonina , Neurotransmisores , Norepinefrina , Transducción de Señal
12.
Br J Pharmacol ; 181(18): 3503-3526, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38779706

RESUMEN

BACKGROUND AND PURPOSE: Excitotoxicity due to mitochondrial calcium (Ca2+) overloading can trigger neuronal cell death in a variety of pathologies. Inhibiting the mitochondrial calcium uniporter (MCU) has been proposed as a therapeutic avenue to prevent calcium overloading. Ru265 (ClRu(NH3)4(µ-N)Ru(NH3)4Cl]Cl3) is a cell-permeable inhibitor of the mitochondrial calcium uniporter (MCU) with nanomolar affinity. Ru265 reduces sensorimotor deficits and neuronal death in models of ischemic stroke. However, the therapeutic use of Ru265 is limited by the induction of seizure-like behaviours. EXPERIMENTAL APPROACH: We examined the effect of Ru265 on synaptic and neuronal function in acute brain slices and hippocampal neuron cultures derived from mice, in control and where MCU expression was genetically abrogated. KEY RESULTS: Ru265 decreased evoked responses from calyx terminals and induced spontaneous action potential firing of both the terminal and postsynaptic principal cell. Recordings of presynaptic Ca2+ currents suggested that Ru265 blocks the P/Q type channel, confirmed by the inhibition of currents in cells exogenously expressing the P/Q type channel. Measurements of presynaptic K+ currents further revealed that Ru265 blocked a KCNQ current, leading to increased membrane excitability, underlying spontaneous spiking. Ca2+ imaging of hippocampal neurons showed that Ru265 increased synchronized, high-amplitude events, recapitulating seizure-like activity seen in vivo. Importantly, MCU ablation did not suppress Ru265-induced increases in neuronal activity and seizures. CONCLUSIONS AND IMPLICATIONS: Our findings provide a mechanistic explanation for the pro-convulsant effects of Ru265 and suggest counter screening assays based on the measurement of P/Q and KCNQ channel currents to identify safe MCU inhibitors.


Asunto(s)
Canales de Calcio , Neuronas , Compuestos de Rutenio , Transmisión Sináptica , Animales , Canales de Calcio/metabolismo , Canales de Calcio/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Compuestos de Rutenio/farmacología , Ratones , Transmisión Sináptica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ratones Endogámicos C57BL , Masculino , Células Cultivadas , Calcio/metabolismo
13.
Res Sq ; 2023 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-37502946

RESUMEN

Dynamic changes in astrocyte Ca2+ are recognized as contributors to functional hyperemia, a critical response to increased neuronal activity mediated by a process known as neurovascular coupling (NVC). Although the critical role of glutamatergic signaling in this process has been extensively investigated, the impact of behavioral state, and the release of behavior-associated neurotransmitters, such as norepinephrine and serotonin, on astrocyte Ca2+ dynamics and functional hyperemia have received less attention. We used two-photon imaging of the barrel cortex in awake mice to examine the role of noradrenergic and serotonergic projections in NVC. We found that both neurotransmitters facilitated sensory-induced increases in astrocyte Ca2+. Interestingly, while ablation of serotonergic neurons reduced sensory-induced functional hyperemia, ablation of noradrenergic neurons caused both attenuation and potentiation of functional hyperemia. Our study demonstrates that norepinephrine and serotonin are involved in modulating sensory-induced astrocyte Ca2+ elevations and identifies their differential effects in regulating functional hyperemia.

14.
Front Cell Neurosci ; 17: 1226630, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37484823

RESUMEN

The neurovascular unit (NVU) is composed of vascular cells, glia, and neurons that form the basic component of the blood brain barrier. This intricate structure rapidly adjusts cerebral blood flow to match the metabolic needs of brain activity. However, the NVU is exquisitely sensitive to damage and displays limited repair after a stroke. To effectively treat stroke, it is therefore considered crucial to both protect and repair the NVU. Mitochondrial calcium (Ca2+) uptake supports NVU function by buffering Ca2+ and stimulating energy production. However, excessive mitochondrial Ca2+ uptake causes toxic mitochondrial Ca2+ overloading that triggers numerous cell death pathways which destroy the NVU. Mitochondrial damage is one of the earliest pathological events in stroke. Drugs that preserve mitochondrial integrity and function should therefore confer profound NVU protection by blocking the initiation of numerous injury events. We have shown that mitochondrial Ca2+ uptake and efflux in the brain are mediated by the mitochondrial Ca2+ uniporter complex (MCUcx) and sodium/Ca2+/lithium exchanger (NCLX), respectively. Moreover, our recent pharmacological studies have demonstrated that MCUcx inhibition and NCLX activation suppress ischemic and excitotoxic neuronal cell death by blocking mitochondrial Ca2+ overloading. These findings suggest that combining MCUcx inhibition with NCLX activation should markedly protect the NVU. In terms of promoting NVU repair, nuclear hormone receptor activation is a promising approach. Retinoid X receptor (RXR) and thyroid hormone receptor (TR) agonists activate complementary transcriptional programs that stimulate mitochondrial biogenesis, suppress inflammation, and enhance the production of new vascular cells, glia, and neurons. RXR and TR agonism should thus further improve the clinical benefits of MCUcx inhibition and NCLX activation by increasing NVU repair. However, drugs that either inhibit the MCUcx, or stimulate the NCLX, or activate the RXR or TR, suffer from adverse effects caused by undesired actions on healthy tissues. To overcome this problem, we describe the use of nanoparticle drug formulations that preferentially target metabolically compromised and damaged NVUs after an ischemic or hemorrhagic stroke. These nanoparticle-based approaches have the potential to improve clinical safety and efficacy by maximizing drug delivery to diseased NVUs and minimizing drug exposure in healthy brain and peripheral tissues.

15.
Trends Neurosci ; 31(11): 559-68, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18817990

RESUMEN

Several modes of synaptic vesicle release, retrieval and recycling have been identified. In a well-established mode of exocytosis, termed 'full-collapse fusion', vesicles empty their neurotransmitter content fully into the synaptic cleft by flattening out and becoming part of the presynaptic membrane. The fused vesicle membrane is then reinternalized via a slow and clathrin-dependent mode of compensatory endocytosis that takes several seconds. A more fleeting mode of vesicle fusion, termed 'kiss-and-run' exocytosis or 'flicker-fusion', indicates that during synaptic transmission some vesicles are only briefly connected to the presynaptic membrane by a transient fusion pore. Finally, a mode that retrieves a large amount of membrane, equivalent to that of several fused vesicles, termed 'bulk endocytosis', has been found after prolonged exocytosis. We are of the opinion that both fast and slow modes of endocytosis co-exist at central nervous system nerve terminals and that one mode can predominate depending on stimulus strength, temperature and synaptic maturation.


Asunto(s)
Endocitosis/fisiología , Modelos Neurológicos , Vesículas Sinápticas/fisiología , Animales , Neuronas/citología , Neuronas/fisiología
16.
Neurobiol Aging ; 67: 108-119, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29656010

RESUMEN

Hearing acuity and sound localization are affected by aging and may contribute to cognitive dementias. Although loss of sensorineural conduction is well documented to occur with age, little is known regarding short-term synaptic plasticity in central auditory nuclei. Age-related changes in synaptic transmission properties were evaluated at the mouse calyx of Held, a sign-inverting relay synapse in the circuit for sound localization, in juvenile adults (1 month old) and late middle-aged (18-21 months old) mice. Synaptic timing and short-term plasticity were severely disrupted in older mice. Surprisingly, acetyl-l-carnitine (ALCAR), an anti-inflammatory agent that facilitates mitochondrial function, fully reversed synaptic transmission delays and defects in short-term plasticity in aged mice to reflect transmission similar to that seen in juvenile adults. These findings support ALCAR supplementation as an adjuvant to improve short-term plasticity and potentially central nervous system performance in animals compromised by age and/or neurodegenerative disease.


Asunto(s)
Acetilcarnitina/farmacología , Envejecimiento , Antiinflamatorios/farmacología , Vías Auditivas/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Sinapsis/fisiología , Transmisión Sináptica/efectos de los fármacos , Acetilcarnitina/uso terapéutico , Animales , Antiinflamatorios/uso terapéutico , Percepción Auditiva/efectos de los fármacos , Percepción Auditiva/fisiología , Femenino , Audición/fisiología , Pérdida Auditiva Sensorineural/tratamiento farmacológico , Pérdida Auditiva Sensorineural/fisiopatología , Pérdida Auditiva Sensorineural/psicología , Masculino , Ratones Endogámicos C57BL , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/fisiopatología , Enfermedades Neurodegenerativas/psicología , Transmisión Sináptica/fisiología
17.
Elife ; 72018 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-29384476

RESUMEN

Perisynaptic glial cells respond to neural activity by increasing cytosolic calcium, but the significance of this pathway is unclear. Terminal/perisynaptic Schwann cells (TPSCs) are a perisynaptic glial cell at the neuromuscular junction that respond to nerve-derived substances such as acetylcholine and purines. Here, we provide genetic evidence that activity-induced calcium accumulation in neonatal TPSCs is mediated exclusively by one subtype of metabotropic purinergic receptor. In P2ry1 mutant mice lacking these responses, postsynaptic, rather than presynaptic, function was altered in response to nerve stimulation. This impairment was correlated with a greater susceptibility to activity-induced muscle fatigue. Interestingly, fatigue in P2ry1 mutants was more greatly exacerbated by exposure to high potassium than in control mice. High potassium itself increased cytosolic levels of calcium in TPSCs, a response which was also reduced P2ry1 mutants. These results suggest that activity-induced calcium responses in TPSCs regulate postsynaptic function and muscle fatigue by regulating perisynaptic potassium.


Asunto(s)
Señalización del Calcio , Fatiga Muscular , Receptores Purinérgicos P2Y1/metabolismo , Células de Schwann/fisiología , Animales , Ratones , Ratones Transgénicos , Receptores Purinérgicos P2Y1/deficiencia
18.
J Neurosci ; 26(5): 1366-77, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16452660

RESUMEN

The timing and strength of synaptic transmission is profoundly dependent on temperature. However, the temperature dependence of the multiple mechanisms that contribute to short-term synaptic plasticity is poorly understood. Here, we use voltage-clamp recordings to quantify the temperature dependence of exocytosis at the calyx of Held synapse. EPSC and miniature EPSC amplitudes were larger at physiological temperature, but quantal content during low-frequency (0.05 Hz) stimulation was constant after temperature jumps from 22-24 degrees C to 35-37 degrees C. The initial degree of EPSC depression during 100 Hz stimuli trains was unchanged with temperature, as were estimates of release probability and vesicle pool size. In contrast, physiological temperatures dramatically relieved depression measured after 40 stimuli at 100 Hz by increasing twofold the rate of recovery from depression. Presynaptic calyx recordings revealed that physiological temperature increased capacitance jumps resulting from 0.5 and 1 ms depolarizations by increasing Ca2+ influx. When Ca2+ entry was equalized at the two temperatures, exocytosis exhibited little temperature dependence for brief depolarizations. However, in response to longer depolarizations, raising temperature increased a slow phase of exocytosis, without affecting overall Ca2+ entry or the size of the readily releasable pool of vesicles. Higher temperatures also increased the rate of presynaptic Ca2+ current inactivation; nevertheless, the degree of steady-state EPSC depression was greatly reduced. Our results thus suggest that changes in steady-state EPSCs during stimulus trains at physiological temperature reflect larger quantal amplitudes and faster refilling of synaptic vesicle pools, leading to reduced short-term depression during prolonged high-frequency firing.


Asunto(s)
Potenciales Postsinápticos Excitadores , Plasticidad Neuronal , Transmisión Sináptica , Vesículas Sinápticas/metabolismo , Temperatura , Animales , Tronco Encefálico/crecimiento & desarrollo , Tronco Encefálico/fisiología , Potenciales Evocados Auditivos del Tronco Encefálico , Exocitosis , Cinética , Inhibición Neural , Terminales Presinápticos/fisiología , Ratas , Ratas Sprague-Dawley , Receptores AMPA/metabolismo , Sinapsis/metabolismo
19.
J Neurosci ; 25(14): 3724-38, 2005 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-15814803

RESUMEN

Na+ channel recovery from inactivation limits the maximal rate of neuronal firing. However, the properties of presynaptic Na+ channels are not well established because of the small size of most CNS boutons. Here we study the Na+ currents of the rat calyx of Held terminal and compare them with those of postsynaptic cells. We find that presynaptic Na+ currents recover from inactivation with a fast, single-exponential time constant (24 degrees C, tau of 1.4-1.8 ms; 35 degrees C, tau of 0.5 ms), and their inactivation rate accelerates twofold during development, which may contribute to the shortening of the action potential as the terminal matures. In contrast, recordings from postsynaptic cells in brainstem slices, and acutely dissociated, reveal that their Na+ currents recover from inactivation with a double-exponential time course (tau(fast) of 1.2-1.6 ms; tau(slow) of 80-125 ms; 24 degrees C). Surprisingly, confocal immunofluorescence revealed that Na+ channels are mostly absent from the calyx terminal but are instead highly concentrated in an unusually long (approximately 20-40 microm) unmyelinated axonal heminode. Outside-out patch recordings confirmed this segregation. Expression of Na(v)1.6 alpha-subunit increased during development, whereas the Na(v)1.2alpha-subunit was not present. Serial EM reconstructions also revealed a long pre-calyx heminode, and biophysical modeling showed that exclusion of Na+ channels from the calyx terminal produces an action potential waveform with a shorter half-width. We propose that the high density and polarized locus of Na+ channels on a long heminode are critical design features that allow the mature calyx of Held terminal to fire reliably at frequencies near 1 kHz.


Asunto(s)
Neuronas/citología , Terminales Presinápticos/metabolismo , Canales de Sodio/fisiología , Sinapsis/fisiología , Potenciales de Acción/fisiología , Potenciales de Acción/efectos de la radiación , Vías Aferentes/fisiología , Vías Aferentes/efectos de la radiación , Factores de Edad , Animales , Animales Recién Nacidos , Tronco Encefálico/citología , Tronco Encefálico/crecimiento & desarrollo , Cloruro de Cadmio/farmacología , Relación Dosis-Respuesta en la Radiación , Estimulación Eléctrica/métodos , Técnica del Anticuerpo Fluorescente/métodos , Técnicas In Vitro , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Potenciales de la Membrana/efectos de la radiación , Microscopía Confocal/métodos , Microscopía Electrónica de Transmisión/métodos , Modelos Neurológicos , Canal de Sodio Activado por Voltaje NAV1.6 , Neuronas/efectos de los fármacos , Neuronas/fisiología , Neuronas/ultraestructura , Técnicas de Placa-Clamp/métodos , Bloqueadores de los Canales de Potasio/farmacología , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/ultraestructura , Subunidades de Proteína/metabolismo , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/fisiología , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/ultraestructura , Tetraetilamonio/farmacología , Tetrodotoxina/farmacología
20.
J Neurosci ; 25(37): 8482-97, 2005 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-16162930

RESUMEN

We examined the effect of glutamate transporter blockade at the calyx of Held synapse. In immature synapses [defined as postnatal day 8 (P8) to P10 rats], transporter blockade causes tonic activation of NMDA receptors and strong inhibition of the AMPA receptor-mediated EPSC amplitude. EPSC inhibition was blocked with a metabotropic glutamate receptor (mGluR) antagonist [1 microm LY341495 (2S-2-amino-2-(1S,2S-2-carboxycycloprop-1-yl)-3-(xanth-9-yl)propanoic acid)], suggesting that elevated resting glutamate concentration specifically activates group II and group III mGluRs. Using mGluR subtype-specific agonists and antagonists, we determined that increased glutamate activates presynaptic mGluR2/3 and mGluR8 receptors but not mGluR4, although this receptor is present. Surprisingly, in older animals (P16-P18), transporter blockade had no effect on EPSC amplitude because of a developmental downregulation of group II/III mGluR activation in rats and mice. In contrast to other CNS synapses, we observed no effect of transporter blockade on EPSC decay kinetics, although expression of glutamate transporters was strong in nearby glial processes at both P9 and P17. Finally, using a low-affinity AMPA receptor antagonist (gamma-D-glutamylglycine), we show that desensitization occurs at P8-P10 but is absent at P16-P18, even during trains of high-frequency (100-300 Hz) stimulation. We suggest that diffusion and transporter activation are insufficient to clear synaptically released glutamate at immature calyces, resulting in significant desensitization. Thus, mGluRs may be expressed in the immature calyx to help limit glutamate release. In the more mature calyx, there is a far smaller diffusional barrier attributable to the highly fenestrated synaptic terminal morphology, so AMPA receptor desensitization is avoided and mGluR-mediated inhibition is not necessary.


Asunto(s)
Receptores AMPA/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Sinapsis/fisiología , Animales , Técnicas In Vitro , Ratones , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA