Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Neurosci ; 2021 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-34099506

RESUMEN

Cortical pyramidal neurons possess a persistent Na+ current (INaP) which, in contrast to the larger transient current, does not undergo rapid inactivation. Although relatively quite small, INaP is active at subthreshold voltages and therefore plays an important role in neuronal input-output processing. The subcellular distribution of channels responsible for INaP and the mechanisms which render them persistent are not known. Using high-speed fluorescence Na+ imaging and whole-cell recordings in brain slices obtained from mice of either sex, we reconstructed the INaP elicited by slow voltage ramps in soma and processes of cortical pyramidal neurons. We found that in all neuronal compartments, the relationship between persistent Na+ conductance and membrane voltage has the shape of a Boltzmann function. Although the density of channels underlying INaP was about twofold lower in the axon initial segment (AIS) than in the soma, the axonal channels were activated by about 10 mV less depolarization than were somatic channels. This difference in voltage dependence explains why, at functionally critical subthreshold voltages, most INaP originates in the AIS. Finally, we show that endogenous polyamines constrain INaP availability in both somato-dendritic and axonal compartments of non-dialyzed cortical neurons.SIGNIFICANCE STATEMENT:The most salient characteristic of neuronal sodium channels is fast inactivation. However, a fraction of the sodium current does not inactivate. In cortical neurons, persistent current (INaP) plays a prominent role in many important functions. Its subcellular distribution and generation mechanisms are, however, elusive. Using high-speed fluorescence Na+ imaging and electrical recordings, we reconstructed the INaP in soma and processes of cortical pyramidal neurons. We found that at near-threshold voltages INaP originates predominately from the axon, due to the distinctive voltage dependence of the underlying channels and not because of their high density. Finally, we show that the presence of endogenous polyamines significantly constrains INaP availability in all compartments of non-dialyzed cortical neurons.

2.
Int J Mol Sci ; 23(17)2022 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-36077024

RESUMEN

Traumatic brain injury (TBI) is associated with significant cognitive and psychiatric conditions. Neuropsychiatric symptoms can persist for years following brain injury, causing major disruptions in patients' lives. In this review, we examine the role of glutamate as an aftereffect of TBI that contributes to the development of neuropsychiatric conditions. We hypothesize that TBI causes long-term blood-brain barrier (BBB) dysfunction lasting many years and even decades. We propose that dysfunction in the BBB is the central factor that modulates increased glutamate after TBI and ultimately leads to neurodegenerative processes and subsequent manifestation of neuropsychiatric conditions. Here, we have identified factors that determine the upper and lower levels of glutamate concentration in the brain after TBI. Furthermore, we consider treatments of disruptions to BBB integrity, including repairing the BBB and controlling excess glutamate, as potential therapeutic modalities for the treatment of acute and chronic neuropsychiatric conditions and symptoms. By specifically focusing on the BBB, we hypothesize that restoring BBB integrity will alleviate neurotoxicity and related neurological sequelae.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Síndromes de Neurotoxicidad , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/terapia , Ácido Glutámico/metabolismo , Humanos , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/metabolismo
3.
Proc Natl Acad Sci U S A ; 115(30): E7184-E7192, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-29991598

RESUMEN

Neocortical pyramidal neurons express several distinct subtypes of voltage-gated Na+ channels. In mature cells, Nav1.6 is the dominant channel subtype in the axon initial segment (AIS) as well as in the nodes of Ranvier. Action potentials (APs) are initiated in the AIS, and it has been proposed that the high excitability of this region is related to the unique characteristics of the Nav1.6 channel. Knockout or loss-of-function mutation of the Scn8a gene is generally lethal early in life because of the importance of this subtype in noncortical regions of the nervous system. Using the Cre/loxP system, we selectively deleted Nav1.6 in excitatory neurons of the forebrain and characterized the excitability of Nav1.6-deficient layer 5 pyramidal neurons by patch-clamp and Na+ and Ca2+ imaging recordings. We now report that, in the absence of Nav1.6 expression, the AIS is occupied by Nav1.2 channels. However, APs are generated in the AIS, and differences in AP propagation to soma and dendrites are minimal. Moreover, the channels that are expressed in the AIS still show a clear hyperpolarizing shift in voltage dependence of activation, compared with somatic channels. The only major difference between Nav1.6-null and wild-type neurons was a strong reduction in persistent sodium current. We propose that the molecular environment of the AIS confers properties on whatever Na channel subtype is present and that some other benefit must be conferred by the selective axonal presence of the Nav1.6 channel.


Asunto(s)
Potenciales de Acción/fisiología , Axones/metabolismo , Canal de Sodio Activado por Voltaje NAV1.2/metabolismo , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Neocórtex/metabolismo , Células Piramidales/metabolismo , Animales , Eliminación de Gen , Ratones , Ratones Transgénicos , Canal de Sodio Activado por Voltaje NAV1.2/genética , Canal de Sodio Activado por Voltaje NAV1.6/genética , Neocórtex/citología , Células Piramidales/citología
4.
J Neurosci ; 39(39): 7790-7800, 2019 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-31399533

RESUMEN

Cortical regions that are damaged by insults, such as ischemia, hypoxia, and trauma, frequently generate spreading depolarization (SD). At the neuronal level, SDs entail complete breakdown of ionic gradients, persisting for seconds to minutes. It is unclear whether these transient events have a more lasting influence on neuronal function. Here, we describe electrophysiological changes in cortical neurons after recovery from hypoxia-induced SD. When examined with standard measures of neuronal excitability several hours after recovery from SD, layer 5 pyramidal neurons in brain slices from mice of either sex appear surprisingly normal. However, we here introduce an additional parameter, dynamic gain, which characterizes the bandwidth of action potential encoding by a neuron, and thereby reflects its potential efficiency in a multineuronal circuit. We find that the ability of neurons that recover from SD to track high-frequency inputs is markedly curtailed; exposure to hypoxia did not have this effect when SD was prevented pharmacologically. Staining for Ankyrin G revealed at least a fourfold decrease in the number of intact axon initial segments in post-SD slices. Since this effect, along with the effect on encoding, was blocked by an inhibitor of the Ca2+-dependent enzyme, calpain, we conclude that both effects were mediated by the SD-induced rise in intracellular Ca2+ Although effects of calpain activation were detected in the axon initial segment, changes in soma-dendritic compartments may also be involved. Whatever the precise molecular mechanism, our findings indicate that in the context of cortical circuit function, effectiveness of neurons that survive SD may be limited.SIGNIFICANCE STATEMENT Spreading depolarization, which commonly accompanies cortical injury, entails transient massive breakdown of neuronal ionic gradients. The function of cortical neurons that recover from hypoxia-induced spreading depolarization is not obviously abnormal when tested for usual measures of neuronal excitability. However, we now demonstrate that they have a reduced bandwidth, reflecting a significant impairment of their ability to precisely encode high-frequency components of their synaptic input in output spike trains. Thus, neurons that recover from spreading depolarizations are less able to function normally as elements in the multineuronal cortical circuitry. These changes are correlated with activation of the calcium-dependent enzyme, calpain.


Asunto(s)
Calpaína/metabolismo , Depresión de Propagación Cortical/fisiología , Hipoxia Encefálica/fisiopatología , Modelos Neurológicos , Neuronas/fisiología , Potenciales de Acción/fisiología , Animales , Femenino , Hipoxia Encefálica/metabolismo , Masculino , Ratones
6.
Proc Natl Acad Sci U S A ; 114(47): E10234-E10243, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29109270

RESUMEN

Alterations in synaptic input, persisting for hours to days, elicit homeostatic plastic changes in the axon initial segment (AIS), which is pivotal for spike generation. Here, in hippocampal pyramidal neurons of both primary cultures and slices, we triggered a unique form of AIS plasticity by selectively targeting M-type K+ channels, which predominantly localize to the AIS and are essential for tuning neuronal excitability. While acute M-current inhibition via cholinergic activation or direct channel block made neurons more excitable, minutes to hours of sustained M-current depression resulted in a gradual reduction in intrinsic excitability. Dual soma-axon patch-clamp recordings combined with axonal Na+ imaging and immunocytochemistry revealed that these compensatory alterations were associated with a distal shift of the spike trigger zone and distal relocation of FGF14, Na+, and Kv7 channels but not ankyrin G. The concomitant distal redistribution of FGF14 together with Nav and Kv7 segments along the AIS suggests that these channels relocate as a structural and functional unit. These fast homeostatic changes were independent of l-type Ca2+ channel activity but were contingent on the crucial AIS protein, protein kinase CK2. Using compartmental simulations, we examined the effects of varying the AIS position relative to the soma and found that AIS distal relocation of both Nav and Kv7 channels elicited a decrease in neuronal excitability. Thus, alterations in M-channel activity rapidly trigger unique AIS plasticity to stabilize network excitability.


Asunto(s)
Segmento Inicial del Axón/fisiología , Quinasa de la Caseína II/metabolismo , Canal de Potasio KCNQ1/fisiología , Plasticidad Neuronal , Células Piramidales/fisiología , Potenciales de Acción , Animales , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/fisiología , Células Cultivadas , Ratones , Ratones Endogámicos BALB C , Modelos Neurológicos , Técnicas de Placa-Clamp , Cultivo Primario de Células , Imagen de Colorante Sensible al Voltaje
7.
Proc Natl Acad Sci U S A ; 114(26): E5167-E5176, 2017 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-28611221

RESUMEN

Key mitochondrial functions such as ATP production, Ca2+ uptake and release, and substrate accumulation depend on the proton electrochemical gradient (ΔµH+) across the inner membrane. Although several drugs can modulate ΔµH+, their effects are hardly reversible, and lack cellular specificity and spatial resolution. Although channelrhodopsins are widely used to modulate the plasma membrane potential of excitable cells, mitochondria have thus far eluded optogenetic control. Here we describe a toolkit of optometabolic constructs based on selective targeting of channelrhodopsins with distinct functional properties to the inner mitochondrial membrane of intact cells. We show that our strategy enables a light-dependent control of the mitochondrial membrane potential (Δψm) and coupled mitochondrial functions such as ATP synthesis by oxidative phosphorylation, Ca2+ dynamics, and respiratory metabolism. By directly modulating Δψm, the mitochondria-targeted opsins were used to control complex physiological processes such as spontaneous beats in cardiac myocytes and glucose-dependent ATP increase in pancreatic ß-cells. Furthermore, our optometabolic tools allow modulation of mitochondrial functions in single cells and defined cell regions.


Asunto(s)
Señalización del Calcio/fisiología , Channelrhodopsins/metabolismo , Células Secretoras de Insulina/metabolismo , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/metabolismo , Optogenética , Animales , Células HEK293 , Células HeLa , Humanos , Células Secretoras de Insulina/citología , Consumo de Oxígeno/fisiología , Ratas , Ratas Sprague-Dawley
8.
J Immunol ; 195(4): 1713-22, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26136430

RESUMEN

TGF-ß1 is a master cytokine in immune regulation, orchestrating both pro- and anti-inflammatory reactions. Recent studies show that whereas TGF-ß1 induces a quiescent microglia phenotype, it plays a pathogenic role in the neurovascular unit and triggers neuronal hyperexcitability and epileptogenesis. In this study, we show that, in primary glial cultures, TGF-ß signaling induces rapid upregulation of the cytokine IL-6 in astrocytes, but not in microglia, via enhanced expression, phosphorylation, and nuclear translocation of SMAD2/3. Electrophysiological recordings show that administration of IL-6 increases cortical excitability, culminating in epileptiform discharges in vitro and spontaneous seizures in C57BL/6 mice. Intracellular recordings from layer V pyramidal cells in neocortical slices obtained from IL-6 -: treated mice show that during epileptogenesis, the cells respond to repetitive orthodromic activation with prolonged after-depolarization with no apparent changes in intrinsic membrane properties. Notably, TGF-ß1 -: induced IL-6 upregulation occurs in brains of FVB/N but not in brains of C57BL/6 mice. Overall, our data suggest that TGF-ß signaling in the brain can cause astrocyte activation whereby IL-6 upregulation results in dysregulation of astrocyte -: neuronal interactions and neuronal hyperexcitability. Whereas IL-6 is epileptogenic in C57BL/6 mice, its upregulation by TGF-ß1 is more profound in FVB/N mice characterized as a relatively more susceptible strain to seizure-induced cell death.


Asunto(s)
Epilepsia/metabolismo , Interleucina-6/metabolismo , Neuroglía/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Electroencefalografía , Epilepsia/genética , Epilepsia/fisiopatología , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-6/genética , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Especificidad de Órganos/genética , Fosforilación/efectos de los fármacos , Transporte de Proteínas , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/administración & dosificación , Factor de Crecimiento Transformador beta/farmacología
9.
Neurobiol Dis ; 95: 158-67, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27443966

RESUMEN

Soon after exposure to hypoxia or ischemia, neurons in cortical tissues undergo massive anoxic depolarization (AD). This precipitous event is preceded by more subtle neuronal changes, including enhanced excitatory and inhibitory synaptic transmitter release. Here, we have used patch-in-slice techniques to identify the earliest effects of acute hypoxia on the synaptic and intrinsic properties of Layer 5 neurons, to determine their time course and to evaluate the role of glutamate receptors in their generation. Coronal slices of mouse somatosensory cortex were maintained at 36°C in an interface chamber and challenged with episodes of hypoxia. In recordings with cell-attached electrodes, the open probability of Ca(2+)-dependent BK channels began to increase within seconds of hypoxia onset, indicating a sharp rise in [Ca(2+)]i just beneath the membrane. By using a high concentration of K(+) in the pipette, we simultaneously monitored the membrane potential and showed that the [Ca(2+)]i rise was not associated with membrane depolarization. The earliest hypoxia-induced synaptic disturbance was a marked increase in the frequency of sPSCs, which also began soon after the removal of oxygen and long before AD. This synaptic effect was accompanied by depletion of the readily releasable transmitter pools, as demonstrated by a decreased response to hyperosmotic solutions. The early [Ca(2+)]i rise, the early increase in transmitter release and the subsequent AD itself were all prevented by bathing in a cocktail containing blockers of ionotropic glutamate receptors. We found no evidence for involvement of pannexin hemichannels or TRPM7 channels in the early responses to hypoxia in this experimental preparation. Our data indicate that the earliest cellular consequences of cortical hypoxia are triggered by activation of glutamate-gated channels.


Asunto(s)
Ácido Glutámico/farmacología , Hipoxia/fisiopatología , Canales de Potasio de Gran Conductancia Activados por el Calcio/efectos de los fármacos , Neocórtex/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Calcio/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Ratones , Neocórtex/metabolismo , Técnicas de Placa-Clamp/métodos , Receptores de Glutamato/metabolismo
10.
Acta Pharmacol Sin ; 37(1): 75-81, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26687934

RESUMEN

Axon initial segment (AIS) is the proximal part of the axon, which is not covered with a myelin sheath and possesses a distinctive, specialized assembly of voltage-gated ion channels and associated proteins. AIS plays critical roles in synaptic integration and action potential generation in central neurons. Recent evidence shows that stroke causes rapid, irreversible calpain-mediated proteolysis of the AIS cytoskeleton of neurons surrounding the ischemic necrotic core. A better understanding of the molecular mechanisms underlying this "non-lethal" neuronal damage might provide new therapeutic strategies for improving stroke outcome. Here, we present a brief overview of the structure and function of the AIS. We then discuss possible mechanisms underlying stroke-induced AIS damage, including the roles of calpains and possible sources of Ca(2+) ions, which are necessary for the activation of calpains. Finally, we discuss the potential functional implications of the loss of the AIS cytoskeleton and ion channel clusters for neuronal excitability.


Asunto(s)
Axones/patología , Citoesqueleto/patología , Accidente Cerebrovascular/patología , Animales , Axones/ultraestructura , Citoesqueleto/ultraestructura , Humanos , Canales Iónicos/fisiología , Plasticidad Neuronal , Accidente Cerebrovascular/fisiopatología
11.
Proc Natl Acad Sci U S A ; 110(10): 4051-6, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23341597

RESUMEN

It is widely believed that, in cortical pyramidal cells, action potentials (APs) initiate in the distal portion of axon initial segment (AIS) because that is where Na(+) channel density is highest. To investigate the relationship between the density of Na(+) channels and the spatiotemporal pattern of AP initiation, we simultaneously recorded Na(+) flux and action currents along the proximal axonal length. We found that functional Na(+) channel density is approximately four times lower in the AP trigger zone than in the middle of the AIS, where it is highest. Computational analysis of AP initiation revealed a paradoxical mismatch between the AP threshold and Na(+) channel density, which could be explained by the lopsided capacitive load imposed on the proximal end of the AIS by the somatodendritic compartment. Favorable conditions for AP initiation are therefore achieved in the distal AIS portion, close to the edge of myelin, where the current source-load ratio is highest. Our findings suggest that cable properties play a central role in determining where the AP starts, such that small plastic changes in the local AIS Na(+) channel density could have a large influence on neuronal excitability as a whole.


Asunto(s)
Axones/metabolismo , Sodio/metabolismo , Potenciales de Acción/fisiología , Animales , Axones/ultraestructura , Benzofuranos , Éteres Cíclicos , Colorantes Fluorescentes , Ratones , Modelos Neurológicos , Células Piramidales/metabolismo , Células Piramidales/ultraestructura , Canales de Sodio/metabolismo , Corteza Somatosensorial/metabolismo , Corteza Somatosensorial/ultraestructura
12.
Gen Comp Endocrinol ; 207: 21-7, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24859253

RESUMEN

Organotypic cultures, prepared from hypothalamo-pituitary slices of tilapia, were developed to enable long-term study of secretory cells in the pituitary of a teleost. Values of membrane potential at rest were similar to those recorded from acute slices, and cells presented similar spontaneous spikes and spikelets. Some cells also exhibited slow spontaneous oscillations in membrane potential, which may be network-driven. Long-term (6days) continuous exposure to GnRH induced increases in LH and FSH secretion. FSH levels reached the highest levels after 24h of exposure to GnRH, and the highest secretion of LH was observed in days 4 and 5 of the experiment. Since slices were viable for several weeks in culture, maintaining the original cytoarchitecture, electrical membrane properties and the ability to secrete hormones in response to exogenous GnRH, this technique is ideal for studying the mechanisms regulating cell-to-cell communication under conditions resembling the in vivo tissue organization.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Sistema Hipotálamo-Hipofisario/metabolismo , Hormona Luteinizante/metabolismo , Hipófisis/metabolismo , Tilapia/metabolismo , Animales , Células Cultivadas , Electrofisiología , Sistema Hipotálamo-Hipofisario/citología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Técnicas para Inmunoenzimas , Técnicas de Cultivo de Órganos , Hipófisis/citología , Hipófisis/efectos de los fármacos , Tilapia/crecimiento & desarrollo
13.
Bio Protoc ; 13(21): e4876, 2023 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-37969753

RESUMEN

Measuring the action potential (AP) propagation velocity in axons is critical for understanding neuronal computation. This protocol describes the measurement of propagation velocity using a combination of somatic whole cell and axonal loose patch recordings in brain slice preparations. The axons of neurons filled with fluorescent dye via somatic whole-cell pipette can be targeted under direct optical control using the fluorophore-filled pipette. The propagation delays between the soma and 5-7 axonal locations can be obtained by analyzing the ensemble averages of 500-600 sweeps of somatic APs aligned at times of maximal rate-of-rise (dV/dtmax) and axonal action currents from these locations. By plotting the propagation delays against the distance, the location of the AP initiation zone becomes evident as the site exhibiting the greatest delay relative to the soma. Performing linear fitting of the delays obtained from sites both proximal and distal from the trigger zone allows the determination of the velocities of AP backward and forward propagation, respectively. Key features • Ultra-thin axons in cortical slices are targeted under direct optical control using the SBFI-filled pipette. • Dual somatic whole cell and axonal loose patch recordings from 5-7 axonal locations. • Ensemble averaging of 500-600 sweeps of somatic APs and axonal action currents. • Plotting the propagation delays against the distance enables the determination of the trigger zone's position and velocities of AP backward and forward propagation.

14.
Transl Psychiatry ; 13(1): 41, 2023 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-36739271

RESUMEN

Traumatic brain injury (TBI) is a serious condition that is associated with an increased risk of severe, long-term psychiatric consequences. Drugs that target the glutamatergic system have proven successful in treating both TBI and many of its psychiatric sequelae. Blood glutamate scavengers (BGS) cause a decrease in blood glutamate levels, leading to a reduction in glutamate's concentration gradient from the brain to the blood and decreased levels of brain glutamate. This study evaluated the BGS pyruvate as a treatment for TBI-related neuropsychiatric conditions in a rat model. 213 rats were divided into four groups in a 2 × 2 design: Sham or TBI rats treated with pyruvate or control treatment. Magnetic resonance imaging, neurological status, brain glutamate and blood glutamate levels were assessed following the injury. Four weeks after the start of treatment, all rats underwent behavioral tests to assess anxious behavior and social impairment (aggressive and hierarchical behavior). Rats responded positively to pyruvate in several tasks, lowering brain glutamate levels and reducing anxiety and depression, as well as modulating TBI-related changes in social behavior. Glutamate scavenging with pyruvate may be an effective therapeutic option for post-TBI behavioral changes by reducing associated elevations in brain glutamate levels.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Ácido Glutámico , Ratas , Animales , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Encéfalo , Ansiedad/tratamiento farmacológico , Piruvatos
15.
Elife ; 122023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36794908

RESUMEN

Voltage-gated sodium channels located in axon initial segments (AIS) trigger action potentials (AP) and play pivotal roles in the excitability of cortical pyramidal neurons. The differential electrophysiological properties and distributions of NaV1.2 and NaV1.6 channels lead to distinct contributions to AP initiation and propagation. While NaV1.6 at the distal AIS promotes AP initiation and forward propagation, NaV1.2 at the proximal AIS promotes the backpropagation of APs to the soma. Here, we show the small ubiquitin-like modifier (SUMO) pathway modulates Na+ channels at the AIS to increase neuronal gain and the speed of backpropagation. Since SUMO does not affect NaV1.6, these effects were attributed to SUMOylation of NaV1.2. Moreover, SUMO effects were absent in a mouse engineered to express NaV1.2-Lys38Gln channels that lack the site for SUMO linkage. Thus, SUMOylation of NaV1.2 exclusively controls INaP generation and AP backpropagation, thereby playing a prominent role in synaptic integration and plasticity.


Asunto(s)
Segmento Inicial del Axón , Sumoilación , Ratones , Animales , Potenciales de Acción/fisiología , Células Piramidales/fisiología , Neuronas , Segmento Inicial del Axón/metabolismo
16.
J Neurosci ; 31(36): 12916-26, 2011 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-21900570

RESUMEN

Vesicular Zn(2+) regulates postsynaptic neuronal excitability upon its corelease with glutamate. We previously demonstrated that synaptic Zn(2+) acts via a distinct metabotropic zinc-sensing receptor (mZnR) in neurons to trigger Ca(2+) responses in the hippocampus. Here, we show that physiological activation of mZnR signaling induces enhanced K(+)/Cl(-) cotransporter 2 (KCC2) activity and surface expression. As KCC2 is the major Cl(-) outward transporter in neurons, Zn(2+) also triggers a pronounced hyperpolarizing shift in the GABA(A) reversal potential. Mossy fiber stimulation-dependent upregulation of KCC2 activity is eliminated in slices from Zn(2+) transporter 3-deficient animals, which lack synaptic Zn(2+). Importantly, activity-dependent ZnR signaling and subsequent enhancement of KCC2 activity are also absent in slices from mice lacking the G-protein-coupled receptor GPR39, identifying this protein as the functional neuronal mZnR. Our work elucidates a fundamentally important role for synaptically released Zn(2+) acting as a neurotransmitter signal via activation of a mZnR to increase Cl(-) transport, thereby enhancing inhibitory tone in postsynaptic cells.


Asunto(s)
Receptores Acoplados a Proteínas G/efectos de los fármacos , Simportadores/biosíntesis , Transmisión Sináptica/efectos de los fármacos , Zinc/farmacología , Animales , Western Blotting , Región CA3 Hipocampal/citología , Región CA3 Hipocampal/fisiología , Fenómenos Electrofisiológicos , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Genotipo , Técnicas In Vitro , Masculino , Ratones , Ratones Noqueados , Microscopía Fluorescente , Fibras Musgosas del Hipocampo/fisiología , Técnicas de Placa-Clamp , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores de GABA-A/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Simportadores/fisiología , Sinapsis/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Zinc/metabolismo , Cotransportadores de K Cl
17.
Metabolites ; 12(5)2022 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-35629963

RESUMEN

Depression is a significant cause of disability and affects millions worldwide; however, antidepressant therapies often fail or are inadequate. Current medications for treating major depressive disorder can take weeks or months to reach efficacy, have troubling side effects, and are limited in their long-term capabilities. Recent studies have identified a new set of glutamate-based approaches, such as blood glutamate scavengers, which have the potential to provide alternatives to traditional antidepressants. In this review, we hypothesize as to the involvement of the glutamate system in the development of depression. We identify the mechanisms underlying glutamate dysregulation, offering new perspectives on the therapeutic modalities of depression with a focus on its relationship to blood-brain barrier (BBB) permeability. Ultimately, we conclude that in diseases with impaired BBB permeability, such as depression following stroke or traumatic brain injury, or in neurogenerative diseases, the glutamate system should be considered as a pathway to treatment. We propose that drugs such as blood glutamate scavengers should be further studied for treatment of these conditions.

18.
Elife ; 112022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35192454

RESUMEN

Mitochondrial activity is crucial for the plasticity of central synapses, but how the firing pattern of pre- and postsynaptic neurons affects the mitochondria remains elusive. We recorded changes in the fluorescence of cytosolic and mitochondrial Ca2+ indicators in cell bodies, axons, and dendrites of cortical pyramidal neurons in mouse brain slices while evoking pre- and postsynaptic spikes. Postsynaptic spike firing elicited fast mitochondrial Ca2+ responses that were about threefold larger in the somas and apical dendrites than in basal dendrites and axons. The amplitude of these responses and metabolic activity were extremely sensitive to the firing frequency. Furthermore, while an EPSP alone caused no detectable Ca2+ elevation in the dendritic mitochondria, the coincidence of EPSP with a backpropagating spike produced prominent, highly localized mitochondrial Ca2+ hotspots. Our results indicate that mitochondria decode the spike firing frequency and the Hebbian temporal coincidences into the Ca2+ signals, which are further translated into the metabolic output and most probably lead to long-term changes in synaptic efficacy.


Asunto(s)
Dendritas , Células Piramidales , Potenciales de Acción/fisiología , Animales , Dendritas/fisiología , Ratones , Mitocondrias , Neuronas/fisiología , Células Piramidales/fisiología , Sinapsis/fisiología
19.
Proc Natl Acad Sci U S A ; 105(48): 18994-9, 2008 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-19020082

RESUMEN

Because the excitable properties of neurons in the neocortex depend on the characteristics of voltage-gated Na(+) channels, factors which regulate those characteristics can fundamentally modify the dynamics of cortical circuits. Here, we report on a novel neuromodulatory mechanism that links the availability of Na(+) channels to metabolism of polyamines (PAs) in the cerebral cortex. Using single channel and whole-cell recordings, we found that products of PA metabolism, the ubiquitous aliphatic polycations spermine and spermidine, are endogenous blockers of Na(+) channels in layer 5 pyramidal cells. Because the blockade is activity-dependent, it is particularly effective against Na(+) channels which fail to inactivate rapidly and thus underlie the persistent Na(+) current. At the level of the local cortical circuit, pharmacological depletion of PAs led to increased spontaneous spiking and periods of hypersynchronous discharge. Our data suggest that changes in PA levels, whether associated with normal brain states or pathological conditions, profoundly modify Na(+) channel availability and thereby shape the integrative behavior of single neurons and neocortical circuits.


Asunto(s)
Corteza Cerebral/citología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Poliaminas/farmacología , Canales de Sodio/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Corteza Cerebral/fisiología , Activación del Canal Iónico , Ratones , Neuronas/citología , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Sodio/metabolismo , Espermidina/farmacología , Espermina/farmacología , Tetrodotoxina/metabolismo
20.
Commun Biol ; 4(1): 666, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34079053

RESUMEN

Calcium dynamics control synaptic transmission. Calcium triggers synaptic vesicle fusion, determines release probability, modulates vesicle recycling, participates in long-term plasticity and regulates cellular metabolism. Mitochondria, the main source of cellular energy, serve as calcium signaling hubs. Mitochondrial calcium transients are primarily determined by the balance between calcium influx, mediated by the mitochondrial calcium uniporter (MCU), and calcium efflux through the sodium/lithium/calcium exchanger (NCLX). We identified a human recessive missense SLC8B1 variant that impairs NCLX activity and is associated with severe mental retardation. On this basis, we examined the effect of deleting NCLX in mice on mitochondrial and synaptic calcium homeostasis, synaptic activity, and plasticity. Neuronal mitochondria exhibited basal calcium overload, membrane depolarization, and a reduction in the amplitude and rate of calcium influx and efflux. We observed smaller cytoplasmic calcium transients in the presynaptic terminals of NCLX-KO neurons, leading to a lower probability of release and weaker transmission. In agreement, synaptic facilitation in NCLX-KO hippocampal slices was enhanced. Importantly, deletion of NCLX abolished long term potentiation of Schaffer collateral synapses. Our results show that NCLX controls presynaptic calcium transients that are crucial for defining synaptic strength as well as short- and long-term plasticity, key elements of learning and memory processes.


Asunto(s)
Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Intercambiador de Sodio-Calcio/genética , Intercambiador de Sodio-Calcio/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Señalización del Calcio , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Femenino , Hipocampo/metabolismo , Humanos , Técnicas In Vitro , Potenciación a Largo Plazo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/química , Proteínas Mitocondriales/deficiencia , Plasticidad Neuronal , Neuronas/metabolismo , Linaje , Mutación Puntual , Terminales Presinápticos/metabolismo , Intercambiador de Sodio-Calcio/química , Transmisión Sináptica/genética , Transmisión Sináptica/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA