Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
J Neurosci ; 41(44): 9257-9273, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34544834

RESUMEN

SCN8A epileptic encephalopathy is a devastating epilepsy syndrome caused by mutant SCN8A, which encodes the voltage-gated sodium channel NaV1.6. To date, it is unclear if and how inhibitory interneurons, which express NaV1.6, influence disease pathology. Using both sexes of a transgenic mouse model of SCN8A epileptic encephalopathy, we found that selective expression of the R1872W SCN8A mutation in somatostatin (SST) interneurons was sufficient to convey susceptibility to audiogenic seizures. Patch-clamp electrophysiology experiments revealed that SST interneurons from mutant mice were hyperexcitable but hypersensitive to action potential failure via depolarization block under normal and seizure-like conditions. Remarkably, GqDREADD-mediated activation of WT SST interneurons resulted in prolonged electrographic seizures and was accompanied by SST hyperexcitability and depolarization block. Aberrantly large persistent sodium currents, a hallmark of SCN8A mutations, were observed and were found to contribute directly to aberrant SST physiology in computational modeling and pharmacological experiments. These novel findings demonstrate a critical and previously unidentified contribution of SST interneurons to seizure generation not only in SCN8A epileptic encephalopathy, but epilepsy in general.SIGNIFICANCE STATEMENTSCN8A epileptic encephalopathy is a devastating neurological disorder that results from de novo mutations in the sodium channel isoform Nav1.6. Inhibitory neurons express NaV1.6, yet their contribution to seizure generation in SCN8A epileptic encephalopathy has not been determined. We show that mice expressing a human-derived SCN8A variant (R1872W) selectively in somatostatin (SST) interneurons have audiogenic seizures. Physiological recordings from SST interneurons show that SCN8A mutations lead to an elevated persistent sodium current which drives initial hyperexcitability, followed by premature action potential failure because of depolarization block. Furthermore, chemogenetic activation of WT SST interneurons leads to audiogenic seizure activity. These findings provide new insight into the importance of SST inhibitory interneurons in seizure initiation, not only in SCN8A epileptic encephalopathy, but for epilepsy broadly.


Asunto(s)
Interneuronas/fisiología , Convulsiones/fisiopatología , Somatostatina/metabolismo , Potenciales de Acción , Animales , Ondas Encefálicas , Interneuronas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación Missense , Canal de Sodio Activado por Voltaje NAV1.6/genética , Convulsiones/genética , Convulsiones/metabolismo , Somatostatina/genética
2.
Ann Neurol ; 89(5): 1023-1035, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33604927

RESUMEN

OBJECTIVE: Sudden unexpected death in epilepsy (SUDEP) is an unpredictable and devastating comorbidity of epilepsy that is believed to be due to cardiorespiratory failure immediately after generalized convulsive seizures. METHODS: We performed cardiorespiratory monitoring of seizure-induced death in mice carrying either a p.Arg1872Trp or p.Asn1768Asp mutation in a single Scn8a allele-mutations identified from patients who died from SUDEP-and of seizure-induced death in pentylenetetrazole-treated wild-type mice. RESULTS: The primary cause of seizure-induced death for all mice was apnea, as (1) apnea began during a seizure and continued for tens of minutes until terminal asystole, and (2) death was prevented by mechanical ventilation. Fatal seizures always included a tonic phase that was coincident with apnea. This tonic phase apnea was not sufficient to produce death, as it also occurred during many nonfatal seizures; however, all seizures that were fatal had tonic phase apnea. We also made the novel observation that continuous tonic diaphragm contraction occurred during tonic phase apnea, which likely contributes to apnea by preventing exhalation, and this was only fatal when breathing did not resume after the tonic phase ended. Finally, recorded seizures from a patient with developmental epileptic encephalopathy with a previously undocumented SCN8A likely pathogenic variant (p.Leu257Val) revealed similarities to those of the mice, namely, an extended tonic phase that was accompanied by apnea. INTERPRETATION: We conclude that apnea coincident with the tonic phase of a seizure, and subsequent failure to resume breathing, are the determining events that cause seizure-induced death in Scn8a mutant mice. ANN NEUROL 2021;89:1023-1035.


Asunto(s)
Apnea/complicaciones , Epilepsia/complicaciones , Muerte Súbita e Inesperada en la Epilepsia , Animales , Convulsivantes , Diafragma/fisiopatología , Electroencefalografía , Electromiografía , Femenino , Humanos , Lactante , Masculino , Ratones , Canal de Sodio Activado por Voltaje NAV1.6/genética , Pentilenotetrazol , Embarazo , Respiración Artificial , Mecánica Respiratoria
3.
J Neurosci ; 37(17): 4565-4583, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28363984

RESUMEN

Current understanding of the contribution of C1 neurons to blood pressure (BP) regulation derives predominantly from experiments performed in anesthetized animals or reduced ex vivo preparations. Here, we use ArchaerhodopsinT3.0 (ArchT) loss-of-function optogenetics to explore BP regulation by C1 neurons in intact, unanesthetized rats. Using a lentivirus that expresses ArchT under the Phox2b-activated promoter PRSx8 (PRSx8-ArchT), ∼65% of transduced neurons were C1 (balance retrotrapezoid nucleus, RTN). Other rats received CaMKII-ArchT3.0 AAV2 (CaMKII-ArchT), which transduced C1 neurons and larger numbers of unidentified glutamatergic and GABAergic cells. Under anesthesia, ArchT photoactivation reduced sympathetic nerve activity and BP and silenced/strongly inhibited most (7/12) putative C1 neurons. In unanesthetized PRSx8-ArchT-treated rats breathing room air, bilateral ArchT photoactivation caused a very small BP reduction that was only slightly larger under hypercapnia (6% FiCO2), but was greatly enhanced during hypoxia (10 and 12% FiO2), after sino-aortic denervation, or during isoflurane anesthesia. The degree of hypotension correlated with percentage of ArchT-transduced C1 neurons. ArchT photoactivation produced similar BP changes in CaMKII-ArchT-treated rats. Photoactivation in PRSX8-ArchT rats reduced breathing frequency (FR), whereas FR increased in CaMKII-ArchT rats. We conclude that the BP drop elicited by ArchT activation resulted from C1 neuron inhibition and was unrelated to breathing changes. C1 neurons have low activity under normoxia, but their activation is important to BP stability during hypoxia or anesthesia and contributes greatly to the hypertension caused by baroreceptor deafferentation. Finally, C1 neurons are marginally activated by hypercapnia and the large breathing stimulation caused by this stimulus has very little impact on resting BP.SIGNIFICANCE STATEMENT C1 neurons are glutamatergic/peptidergic/catecholaminergic neurons located in the medulla oblongata, which may operate as a switchboard for differential, behavior-appropriate activation of selected sympathetic efferents. Based largely on experimentation in anesthetized or reduced preparations, a rostrally located subset of C1 neurons may contribute to both BP stabilization and dysregulation (hypertension). Here, we used Archaerhodopsin-based loss-of-function optogenetics to explore the contribution of these neurons to BP in conscious rats. The results suggest that C1 neurons contribute little to resting BP under normoxia or hypercapnia, C1 neuron discharge is restrained continuously by arterial baroreceptors, and C1 neuron activation is critical to stabilize BP under hypoxia or anesthesia. This optogenetic approach could also be useful to explore the role of C1 neurons during specific behaviors or in hypertensive models.


Asunto(s)
Anestesia , Presión Sanguínea , Hipercapnia/fisiopatología , Hipoxia/fisiopatología , Bulbo Raquídeo/fisiopatología , Presorreceptores , Anestésicos por Inhalación/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Células Quimiorreceptoras , Hipercapnia/genética , Hipertensión/fisiopatología , Isoflurano/farmacología , Masculino , Neuronas , Optogenética , Ratas , Ratas Sprague-Dawley , Transducción Genética
4.
J Physiol ; 596(15): 3029-3042, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29168167

RESUMEN

The retrotrapezoid nucleus (RTN) regulates breathing in a CO2 - and state-dependent manner. RTN neurons are glutamatergic and innervate principally the respiratory pattern generator; they regulate multiple aspects of breathing, including active expiration, and maintain breathing automaticity during non-REM sleep. RTN neurons encode arterial PCO2 /pH via cell-autonomous and paracrine mechanisms, and via input from other CO2 -responsive neurons. In short, RTN neurons are a pivotal structure for breathing automaticity and arterial PCO2 homeostasis. The carotid bodies stimulate the respiratory pattern generator directly and indirectly by activating RTN via a neuronal projection originating within the solitary tract nucleus. The indirect pathway operates under normo- or hypercapnic conditions; under respiratory alkalosis (e.g. hypoxia) RTN neurons are silent and the excitatory input from the carotid bodies is suppressed. Also, silencing RTN neurons optogenetically quickly triggers a compensatory increase in carotid body activity. Thus, in conscious mammals, breathing is subject to a dual and interdependent feedback regulation by chemoreceptors. Depending on the circumstance, the activity of the carotid bodies and that of RTN vary in the same or the opposite directions, producing additive or countervailing effects on breathing. These interactions are mediated either via changes in blood gases or by brainstem neuronal connections, but their ultimate effect is invariably to minimize arterial PCO2 fluctuations. We discuss the potential relevance of this dual chemoreceptor feedback to cardiorespiratory abnormalities present in diseases in which the carotid bodies are hyperactive at rest, e.g. essential hypertension, obstructive sleep apnoea and heart failure.


Asunto(s)
Tronco Encefálico/fisiología , Neuronas/fisiología , Respiración , Animales , Retroalimentación Fisiológica , Humanos
5.
J Physiol ; 594(6): 1529-51, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26748771

RESUMEN

We discuss recent evidence which suggests that the principal central respiratory chemoreceptors are located within the retrotrapezoid nucleus (RTN) and that RTN neurons are directly sensitive to [H(+) ]. RTN neurons are glutamatergic. In vitro, their activation by [H(+) ] requires expression of a proton-activated G protein-coupled receptor (GPR4) and a proton-modulated potassium channel (TASK-2) whose transcripts are undetectable in astrocytes and the rest of the lower brainstem respiratory network. The pH response of RTN neurons is modulated by surrounding astrocytes but genetic deletion of RTN neurons or deletion of both GPR4 and TASK-2 virtually eliminates the central respiratory chemoreflex. Thus, although this reflex is regulated by innumerable brain pathways, it seems to operate predominantly by modulating the discharge rate of RTN neurons, and the activation of RTN neurons by hypercapnia may ultimately derive from their intrinsic pH sensitivity. RTN neurons increase lung ventilation by stimulating multiple aspects of breathing simultaneously. They stimulate breathing about equally during quiet wake and non-rapid eye movement (REM) sleep, and to a lesser degree during REM sleep. The activity of RTN neurons is regulated by inhibitory feedback and by excitatory inputs, notably from the carotid bodies. The latter input operates during normo- or hypercapnia but fails to activate RTN neurons under hypocapnic conditions. RTN inhibition probably limits the degree of hyperventilation produced by hypocapnic hypoxia. RTN neurons are also activated by inputs from serotonergic neurons and hypothalamic neurons. The absence of RTN neurons probably underlies the sleep apnoea and lack of chemoreflex that characterize congenital central hypoventilation syndrome.


Asunto(s)
Células Quimiorreceptoras/metabolismo , Bulbo Raquídeo/fisiología , Protones , Respiración , Animales , Humanos , Bulbo Raquídeo/citología , Bulbo Raquídeo/metabolismo , Canales de Potasio de Dominio Poro en Tándem/genética , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Reflejo , Sueño REM
6.
J Physiol ; 593(5): 1067-74, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25524282

RESUMEN

The rostral ventrolateral medulla oblongata (RVLM) contains two functionally distinct types of neurons that control and orchestrate cardiovascular and respiratory responses to hypoxia and hypercapnia. One group is composed of the central chemoreceptor neurons of the retrotrapezoid nucleus, which provides a CO2/H(+) -dependent drive to breathe and serves as an integration centre and a point of convergence of chemosensory information from other central and peripheral sites, including the carotid bodies. The second cluster of RVLM cells forms a population of neurons belonging to the C1 catecholaminergic group that controls sympathetic vasomotor tone in resting conditions and in conditions of hypoxia and hypercapnia. Recent evidence suggests that ATP-mediated purinergic signalling at the level of the RVLM co-ordinates cardiovascular and respiratory responses triggered by hypoxia and hypercapnia by activating retrotrapezoid nucleus and C1 neurons, respectively. The role of ATP-mediated signalling in the RVLM mechanisms of cardiovascular and respiratory activities is the main subject of this short review.


Asunto(s)
Células Quimiorreceptoras/metabolismo , Bulbo Raquídeo/metabolismo , Receptores Purinérgicos/metabolismo , Sistema Nervioso Simpático/metabolismo , Animales , Dióxido de Carbono/sangre , Humanos , Bulbo Raquídeo/fisiología , Sistema Nervioso Simpático/fisiología
7.
J Physiol ; 592(6): 1309-23, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24445316

RESUMEN

Several brain regions are thought to function as important sites of chemoreception including the nucleus of the solitary tract (NTS), medullary raphe and retrotrapezoid nucleus (RTN). In the RTN, mechanisms of chemoreception involve direct H(+)-mediated activation of chemosensitive neurons and indirect modulation of chemosensitive neurons by purinergic signalling. Evidence suggests that RTN astrocytes are the source of CO2-evoked ATP release. However, it is not clear whether purinergic signalling also influences CO2/H(+) responsiveness of other putative chemoreceptors. The goals of this study are to determine if CO2/H(+)-sensitive neurons in the NTS and medullary raphe respond to ATP, and whether purinergic signalling in these regions influences CO2 responsiveness in vitro and in vivo. In brain slices, cell-attached recordings of membrane potential show that CO2/H(+)-sensitive NTS neurons are activated by focal ATP application; however, purinergic P2-receptor blockade did not affect their CO2/H(+) responsiveness. CO2/H(+)-sensitive raphe neurons were unaffected by ATP or P2-receptor blockade. In vivo, ATP injection into the NTS increased cardiorespiratory activity; however, injection of a P2-receptor blocker into this region had no effect on baseline breathing or CO2/H(+) responsiveness. Injections of ATP or a P2-receptor blocker into the medullary raphe had no effect on cardiorespiratory activity or the chemoreflex. As a positive control we confirmed that ATP injection into the RTN increased breathing and blood pressure by a P2-receptor-dependent mechanism. These results suggest that purinergic signalling is a unique feature of RTN chemoreception.


Asunto(s)
Células Quimiorreceptoras/fisiología , Núcleos del Rafe/fisiología , Receptores Purinérgicos P2/fisiología , Núcleo Solitario/fisiología , Adenosina Trifosfato/fisiología , Animales , Hipercapnia/fisiopatología , Masculino , Ratas , Ratas Wistar , Centro Respiratorio/fisiología , Fenómenos Fisiológicos Respiratorios , Transducción de Señal
8.
J Neurosci ; 32(47): 16943-52, 2012 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-23175845

RESUMEN

Chemosensitive neurons in the retrotrapezoid nucleus (RTN) regulate breathing in response to CO(2)/H(+) changes. Their activity is also sensitive to neuromodulatory inputs from multiple respiratory centers, and thus they serve as a key nexus of respiratory control. However, molecular mechanisms that control their activity and susceptibility to neuromodulation are unknown. Here, we show in vitro and in vivo that KCNQ channels are critical determinants of RTN neural activity. In particular, we find that pharmacological block of KCNQ channels (XE991, 10 µm) increased basal activity and CO(2) responsiveness of RTN neurons in rat brain slices, whereas KCNQ channel activation (retigabine, 2-40 µm) silenced these neurons. Interestingly, we also find that KCNQ and apamin-sensitive SK channels act synergistically to regulate firing rate of RTN chemoreceptors; simultaneous blockade of both channels led to a increase in CO(2) responsiveness. Furthermore, we also show that KCNQ channels but not SK channels are downstream effectors of serotonin modulation of RTN activity in vitro. In contrast, inhibition of KCNQ channel did not prevent modulation of RTN activity by Substance P or thyrotropin-releasing hormone, previously identified neuromodulators of RTN chemoreception. Importantly, we also show that KCNQ channels are critical for RTN activity in vivo. Inhibition of KCNQ channels lowered the CO(2) threshold for phrenic nerve discharge in anesthetized rats and decreased the ventilatory response to serotonin in awake and anesthetized animals. Given that serotonergic dysfunction may contribute to respiratory failure, our findings suggest KCNQ channels as a new therapeutic avenue for respiratory complications associated with multiple neurological disorders.


Asunto(s)
Tronco Encefálico/fisiología , Células Quimiorreceptoras/fisiología , Impulso (Psicología) , Canales de Potasio KCNQ/fisiología , Mecánica Respiratoria/fisiología , Serotonina/fisiología , Anestesia , Animales , Animales Recién Nacidos , Carbamatos/farmacología , Dióxido de Carbono/fisiología , Fenómenos Electrofisiológicos , Técnicas In Vitro , Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/antagonistas & inhibidores , Masculino , Fenilendiaminas/farmacología , Nervio Frénico/efectos de los fármacos , Nervio Frénico/fisiología , Ratas , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Sustancia P/farmacología , Hormona Liberadora de Tirotropina/farmacología
9.
Prog Neurobiol ; 231: 102531, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37778436

RESUMEN

Repeated generalized tonic-clonic seizures (GTCSs) are the most critical risk factor for sudden unexpected death in epilepsy (SUDEP). GTCSs can cause fatal apnea. We investigated neuronal plasticity mechanisms that precipitate postictal apnea and seizure-induced death. Repeated seizures worsened behavior, precipitated apnea, and enlarged active neuronal circuits, recruiting more neurons in such brainstem nuclei as periaqueductal gray (PAG) and dorsal raphe, indicative of brainstem plasticity. Seizure-activated neurons are more excitable and have enhanced AMPA-mediated excitatory transmission after a seizure. Global deletion of the GluA1 subunit of AMPA receptors abolishes postictal apnea and seizure-induced death. Treatment with a drug that blocks Ca2+-permeable AMPA receptors also renders mice apnea-free with five-fold better survival than untreated mice. Repeated seizures traffic the GluA1 subunit-containing AMPA receptors to synapses, and blocking this mechanism decreases the probability of postictal apnea and seizure-induced death.


Asunto(s)
Apnea , Receptores AMPA , Ratones , Animales , Receptores AMPA/uso terapéutico , Convulsiones/tratamiento farmacológico , Tronco Encefálico , Factores de Riesgo
10.
J Physiol ; 590(9): 2137-50, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22411009

RESUMEN

Central chemoreception is the mechanism by which the brain regulates breathing in response to changes in tissue CO(2)H+. A brainstem region called the retrotrapezoid nucleus (RTN) contains a population of CO2/H+-sensitive neurons that appears to function as an important chemoreceptor. Evidence also indicates that CO2-evoked ATP release from RTN astrocytes modulates activity of CO2/H+-sensitive neurons; however, the extent to which purinergic signalling contributes to chemoreception by RTN neurons is not clear and the mechanism(s) underlying CO2/H+-evoked ATP release is not fully elucidated. The goals of this study are to determine the extent to which ATP contributes to RTN chemoreception both in vivo and in vitro, and whether purinergic drive to chemoreceptors relies on extracellular Ca(2+) or gap junction hemichannels. We also examine the possible contribution of P2Y1 receptors expressed in the RTN to the purinergic drive to breathe.We show that purinergic signalling contributes, in part, to the CO(2)/H+ sensitivity of RTN neurons. In vivo, phrenic nerve recordings of respiratory activity in adult rats show that bilateral injections of pyridoxal-phosphate-6-azophenyl-2',4'-disulfonate (PPADS, a P2 receptor blocker) decreased the ventilatory response to CO2 by 30%. In vitro, loose-patch recordings from RTN neurons show that P2 receptor blockers decreased responsiveness to both 10% and 15% CO2 also by 30%. In the slice, the contribution of purinergic signalling to RTN chemoreception did not increase with temperature (22­35◦C) and was retained in low extracellular Ca2+ medium. Conversely, the gap junction blockers carbenoxolone and cobalt decreased neuronal CO2/H+ sensitivity by an amount similar to P2 receptor antagonists. Inhibition of the P2Y1 receptor in the RTN had no effect on CO2 responsivness in vitro or in vivo; thus, the identity of P2 receptors underlying the purinergic component of RTN chemoreception remains unknown. These results support the possibility that CO2/H+-evoked ATP release is mediated by a mechanism involving gap junction hemichannels.


Asunto(s)
Adenosina Trifosfato/metabolismo , Tronco Encefálico/metabolismo , Dióxido de Carbono/metabolismo , Células Quimiorreceptoras/metabolismo , Hipercapnia/metabolismo , Receptores Purinérgicos/metabolismo , Transducción de Señal , Potenciales de Acción , Animales , Presión Sanguínea , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/fisiopatología , Calcio/metabolismo , Carbenoxolona/farmacología , Células Quimiorreceptoras/efectos de los fármacos , Cobalto/farmacología , Conexinas/metabolismo , Desnervación , Modelos Animales de Enfermedad , Uniones Comunicantes/metabolismo , Concentración de Iones de Hidrógeno , Hipercapnia/fisiopatología , Masculino , Técnicas de Placa-Clamp , Nervio Frénico/metabolismo , Nervio Frénico/fisiopatología , Antagonistas del Receptor Purinérgico P2/farmacología , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacología , Ratas , Ratas Wistar , Receptores Purinérgicos/efectos de los fármacos , Receptores Purinérgicos P2Y1/metabolismo , Centro Respiratorio/metabolismo , Centro Respiratorio/fisiopatología , Mecánica Respiratoria , Transducción de Señal/efectos de los fármacos , Suramina/farmacología , Temperatura , Factores de Tiempo
11.
J Neurophysiol ; 107(5): 1489-99, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22131386

RESUMEN

Nitric oxide (NO) is an important signaling molecule that regulates numerous physiological processes, including activity of respiratory motoneurons. However, molecular mechanism(s) underlying NO modulation of motoneurons remain obscure. Here, we used a combination of in vivo and in vitro recording techniques to examine NO modulation of motoneurons in the hypoglossal motor nucleus (HMN). Microperfusion of diethylamine (DEA; an NO donor) into the HMN of anesthetized adult rats increased genioglossus muscle activity. In the brain slice, whole cell current-clamp recordings from hypoglossal motoneurons showed that exposure to DEA depolarized membrane potential and increased responsiveness to depolarizing current injections. Under voltage-clamp conditions, we found that NO inhibited a Ba(2+)-sensitive background K(+) conductance and activated a Cs(+)-sensitive hyperpolarization-activated inward current (I(h)). When I(h) was blocked with Cs(+) or ZD-7288, the NO-sensitive K(+) conductance exhibited properties similar to TWIK-related acid-sensitive K(+) (TASK) channels, i.e., voltage independent, resistant to tetraethylammonium and 4-aminopyridine but inhibited by methanandamide. The soluble guanylyl cyclase blocker 1H-(1,2,4)oxadiazole(4,3-a)quinoxaline-1-one (ODQ) and the PKG blocker KT-5823 both decreased NO modulation of this TASK-like conductance. To characterize modulation of I(h) in relative isolation, we tested effects of NO in the presence of Ba(2+) to block TASK channels. Under these conditions, NO activated both the instantaneous (I(inst)) and time-dependent (I(ss)) components of I(h). Interestingly, at more hyperpolarized potentials NO preferentially increased I(inst). The effects of NO on I(h) were retained in the presence of ODQ and blocked by the cysteine-specific oxidant N-ethylmaleimide. These results suggest that NO activates hypoglossal motoneurons by cGMP-dependent inhibition of a TASK-like current and S-nitrosylation-dependent activation of I(h).


Asunto(s)
GMP Cíclico/fisiología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Nervio Hipogloso/fisiología , Neuronas Motoras/fisiología , Óxido Nítrico/fisiología , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores , Canales de Potasio de Dominio Poro en Tándem/fisiología , Animales , Masculino , Proteínas del Tejido Nervioso , Ratas , Ratas Wistar
12.
Epilepsia Open ; 7(2): 280-292, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34826216

RESUMEN

OBJECTIVE: SCN8A epileptic encephalopathy is caused predominantly by de novo gain-of-function mutations in the voltage-gated sodium channel Nav 1.6. The disorder is characterized by early onset of seizures and developmental delay. Most patients with SCN8A epileptic encephalopathy are refractory to current anti-seizure medications. Previous studies determining the mechanisms of this disease have focused on neuronal dysfunction as Nav 1.6 is expressed by neurons and plays a critical role in controlling neuronal excitability. However, glial dysfunction has been implicated in epilepsy and alterations in glial physiology could contribute to the pathology of SCN8A encephalopathy. In the current study, we examined alterations in astrocyte and microglia physiology in the development of seizures in a mouse model of SCN8A epileptic encephalopathy. METHODS: Using immunohistochemistry, we assessed microglia and astrocyte reactivity before and after the onset of spontaneous seizures. Expression of glutamine synthetase and Nav 1.6, and Kir 4.1 channel currents were assessed in astrocytes in wild-type (WT) mice and mice carrying the N1768D SCN8A mutation (D/+). RESULTS: Astrocytes in spontaneously seizing D/+ mice become reactive and increase expression of glial fibrillary acidic protein (GFAP), a marker of astrocyte reactivity. These same astrocytes exhibited reduced barium-sensitive Kir 4.1 currents compared to age-matched WT mice and decreased expression of glutamine synthetase. These alterations were only observed in spontaneously seizing mice and not before the onset of seizures. In contrast, microglial morphology remained unchanged before and after the onset of seizures. SIGNIFICANCE: Astrocytes, but not microglia, become reactive only after the onset of spontaneous seizures in a mouse model of SCN8A encephalopathy. Reactive astrocytes have reduced Kir 4.1-mediated currents, which would impair their ability to buffer potassium. Reduced expression of glutamine synthetase would modulate the availability of neurotransmitters to excitatory and inhibitory neurons. These deficits in potassium and glutamate handling by astrocytes could exacerbate seizures in SCN8A epileptic encephalopathy. Targeting astrocytes may provide a new therapeutic approach to seizure suppression.


Asunto(s)
Epilepsia Generalizada , Epilepsia , Animales , Astrocitos/metabolismo , Modelos Animales de Enfermedad , Epilepsia/tratamiento farmacológico , Epilepsia/genética , Glutamato-Amoníaco Ligasa/metabolismo , Humanos , Ratones , Canal de Sodio Activado por Voltaje NAV1.6/genética , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Potasio/metabolismo , Potasio/uso terapéutico
13.
Front Neural Circuits ; 16: 1002013, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36160949

RESUMEN

Sudden unexpected death in epilepsy (SUDEP) accounts for the deaths of 8-17% of patients with epilepsy. Although the mechanisms of SUDEP are essentially unknown, one proposed mechanism is respiratory arrest initiated by a convulsive seizure. In mice, we have previously observed that extended apnea occurs during the tonic phase of seizures. Although often survived, tonic seizures became fatal when breathing did not immediately recover postictally. We also found that respiratory muscles were tonically contracted during the apnea, suggesting that muscle contraction could be the cause of apnea. In the present study, we tested the hypothesis that pyramidal neurons of the motor cortex drive motor units during the tonic phase, which produces apnea. Mice harboring the patient-derived N1768D point mutation of an Scn8a allele were crossed with transgenic mice such that inhibitory Designer Receptors Exclusively Activated by Designer Drugs (DREADD) receptors were selectively expressed in excitatory forebrain neurons. We then triggered audiogenic and hippocampal (HC) stimulated seizures under control conditions and when excitatory forebrain neurons were inhibited with the synthetic ligand Clozapine-N-Oxide (CNO). We found that inhibition with CNO was sufficient to increase seizure threshold of HC stimulated, but not audiogenic, seizures. In addition, regardless of seizure type, CNO nearly eliminated epileptiform activity that occurred proximal to the tonic phase; however, the seizure behaviors, notably the tonic phase and concomitant apnea, were unchanged. We interpret these results to indicate that while cortical neurons are likely critical for epileptogenesis and seizure initiation, the behavioral manifestations of tonic seizures are generated by neural circuitry in the mid- and/or hindbrain.


Asunto(s)
Clozapina , Drogas de Diseño , Epilepsia , Muerte Súbita e Inesperada en la Epilepsia , Animales , Apnea/genética , Modelos Animales de Enfermedad , Epilepsia/genética , Ligandos , Ratones , Ratones Transgénicos , Canal de Sodio Activado por Voltaje NAV1.6 , Óxidos , Prosencéfalo , Convulsiones/genética
14.
Brain Res ; 1775: 147743, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34843701

RESUMEN

Dravet Syndrome (DS) is a severe developmental and epileptic encephalopathy typically caused by loss-of-function de novo mutations in the SCN1A gene which encodes the voltage-gated sodium channel isoform NaV1.1. Decreased NaV1.1 expression results in impaired excitability of inhibitory interneurons and seizure onset. To date, there are no clinically available treatments for DS that directly address the core mechanism of disease; reduced NaV1.1 expression levels in interneurons. Recently, Targeted Augmentation of Nuclear Gene Output (TANGO) of SCN1A by the antisense oligonucleotide (ASO) STK-001, was shown to increase Scn1a mRNA levels, increase NaV1.1 protein expression, reduce seizures, and improve survival in the Scn1a+/- mouse model of DS. However, it remains unknown whether STK-001 treatment rescues the reduced intrinsic excitability of parvalbumin-positive (PV) inhibitory interneurons associated with DS. In this study, we demonstrate that STK-001 treatment reduces seizures, prolongs survival, and rescues PV interneuron excitability in Scn1a+/- mice to levels observed in WT littermates. Together, these results support the notion that TANGO-mediated augmentation of NaV1.1 levels directly targets and rescues one of the core disease mechanisms of DS.


Asunto(s)
Potenciales de Acción/fisiología , Epilepsias Mioclónicas/genética , Interneuronas/metabolismo , Canal de Sodio Activado por Voltaje NAV1.1/genética , Parvalbúminas/metabolismo , Convulsiones/genética , Animales , Modelos Animales de Enfermedad , Epilepsias Mioclónicas/fisiopatología , Ratones , Oligonucleótidos Antisentido , Convulsiones/fisiopatología
15.
Exp Physiol ; 96(4): 400-6, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21169332

RESUMEN

Central chemoreception is the mechanism by which CO(2)/pH-sensitive neurons (i.e. chemoreceptors) regulate breathing, presumably in response to changes in tissue pH. A region of the brainstem called the retrotrapezoid nucleus (RTN) is thought to be an important site of chemoreception; select neurons (i.e. chemoreceptors) in this region sense changes in CO(2)/H(+) and send excitatory glutamatergic drive to respiratory centres to modulate the depth and frequency of breathing. Purinergic signalling may also contribute to chemoreception; for instance, it was shown in vivo that CO(2)/H(+) facilitates ATP release within the RTN to stimulate breathing, and recent evidence suggests that CO(2)/H(+)-sensitive RTN astrocytes are the source of this purinergic drive to breathe. In this review, we summarize evidence that RTN astrocytes sense changes in CO(2)/H(+), identify mechanisms that are likely to confer CO(2)/H(+) sensitivity to RTN astrocytes, including inhibition of heteromeric Kir4.1-Kir5.1 channels and activation of a depolarizing inward current generated by the sodium bicarbonate cotransporter, and discuss the extent to which astrocytes contribute to respiratory drive.


Asunto(s)
Astrocitos/fisiología , Células Quimiorreceptoras/fisiología , Centro Respiratorio/fisiología , Mecánica Respiratoria/fisiología , Animales , Dióxido de Carbono/fisiología , Humanos , Concentración de Iones de Hidrógeno
16.
Front Neurosci ; 15: 795145, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35126041

RESUMEN

Sudden unexpected death in epilepsy (SUDEP) accounts for the deaths of 8-17% of patients with epilepsy. Although the mechanisms of SUDEP are unknown, one proposed mechanism is abnormal control of the heart by the autonomic nervous system (ANS). Our objective was to determine whether the broad changes in ictal heart rate experienced by mouse models of SUDEP are (1) due to the ANS and (2) contribute to seizure-induced death. Seizures were induced by electrical stimulation of the hippocampus of a mouse carrying the human SCN8A encephalopathy mutation p.Asn1768Asp (N1768D; "D/+ mice"). Using standard autonomic pharmacology, the relative roles of the parasympathetic and sympathetic nervous systems on heart rate changes associated with seizures were determined. All induced seizures had pronounced ictal bradycardia and postictal tachycardia. Seizure susceptibility or severity were unchanged by the pharmacological agents. Administration of Atropine, a muscarinic antagonist, eliminated ictal bradycardia, while carbachol, a muscarinic agonist, had no effect on ictal bradycardia, but reduced postictal tachycardia. Sotalol, an adrenergic ß-receptor antagonist, had no effect on ictal bradycardia, but did suppress postictal tachycardia. Isoproterenol, a ß-receptor agonist, had no effect on either ictal bradycardia or postictal tachycardia. Administration of the α1-receptor antagonist prazosin increases the incidence of seizure-induced death in D/+ mice. Although postictal heart rate was lower for these fatal seizures in the presence of prazosin, rates were not as low as that recorded for carbachol treated mice, which all survived. Both ictal bradycardia and postictal tachycardia are manifestations of the ANS. Bradycardia is mediated by a maximal activation of the parasympathetic arm of the ANS, and tachycardia is mediated by parasympathetic inactivation and sympathetic activation. While the changes in heart rate during seizures are profound, suppression of postictal heart rate did not increase seizure mortality.

17.
Front Neurosci ; 15: 581048, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33762902

RESUMEN

Sudden unexpected death in epilepsy (SUDEP) is the leading cause of death amongst patients whose seizures are not adequately controlled by current therapies. Patients with SCN8A encephalopathy have an elevated risk for SUDEP. While transgenic mouse models have provided insight into the molecular mechanisms of SCN8A encephalopathy etiology, our understanding of seizure-induced death has been hampered by the inability to reliably trigger both seizures and seizure-induced death in these mice. Here, we demonstrate that mice harboring an Scn8a allele with the patient-derived mutation N1768D (D/+) are susceptible to audiogenic seizures and seizure-induced death. In adult D/+ mice, audiogenic seizures are non-fatal and have nearly identical behavioral, electrographical, and cardiorespiratory characteristics as spontaneous seizures. In contrast, at postnatal days 20-21, D/+ mice exhibit the same seizure behavior, but have a significantly higher incidence of seizure-induced death following an audiogenic seizure. Seizure-induced death was prevented by either stimulating breathing via mechanical ventilation or by acute activation of adrenergic receptors. Conversely, in adult D/+ mice inhibition of adrenergic receptors converted normally non-fatal audiogenic seizures into fatal seizures. Taken together, our studies show that in our novel audiogenic seizure-induced death model adrenergic receptor activation is necessary and sufficient for recovery of breathing and prevention of seizure-induced death.

18.
J Neurophysiol ; 104(6): 3042-52, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20926613

RESUMEN

Central chemoreception is the mechanism by which CO(2)/pH sensors regulate breathing in response to tissue pH changes. There is compelling evidence that pH-sensitive neurons in the retrotrapezoid nucleus (RTN) are important chemoreceptors. Evidence also indicates that CO(2)/H(+)-evoked adenosine 5'-triphosphate (ATP) release in the RTN, from pH-sensitive astrocytes, contributes to chemoreception. However, mechanism(s) by which RTN astrocytes sense pH is unknown and their contribution to chemoreception remains controversial. Here, we use the brain slice preparation and a combination of patch-clamp electrophysiology and immunohistochemistry to confirm that RTN astrocytes are pH sensitive and to determine mechanisms by which they sense pH. We show that pH-sensitive RTN glia are immunoreactive for aldehyde dehydrogenase 1L1, a marker of astrocytes. In HEPES buffer the pH-sensitive current expressed by RTN astrocytes reversed near E(K(+)) (the equilibrium potential for K(+)) and was inhibited by Ba(2+) and desipramine (blocker of Kir4.1-containing channels), characteristics most consistent with heteromeric Kir4.1-Kir5.1 channels. In bicarbonate buffer, the sodium/bicarbonate cotransporter also contributed to the CO(2)/H(+)-sensitive current in RTN astrocytes. To test the hypothesis that RTN astrocytes contribute to chemoreception by a purinergic mechanism, we used fluorocitrate to selectively depolarize astrocytes while measuring neuronal activity. We found that fluorocitrate increased baseline activity and pH sensitivity of RTN neurons by a P2-receptor-dependent mechanism, suggesting that astrocytes may release ATP to activate RTN chemoreceptors. We also found in bicarbonate but not HEPES buffer that P2-receptor antagonists decreased CO(2) sensitivity of RTN neurons. We conclude that RTN astrocytes sense CO(2)/H(+) in part by inhibition of a Kir4.1-Kir5.1-like current and may provide an excitatory purinergic drive to pH-sensitive neurons.


Asunto(s)
Astrocitos/fisiología , Células Quimiorreceptoras/fisiología , Concentración de Iones de Hidrógeno , Canales de Potasio de Rectificación Interna/fisiología , Receptores Purinérgicos P2/fisiología , Centro Respiratorio/fisiología , Adenosina Trifosfato/fisiología , Animales , Astrocitos/efectos de los fármacos , Bario/farmacología , Dióxido de Carbono/farmacología , Células Quimiorreceptoras/efectos de los fármacos , Citratos/farmacología , Desipramina/farmacología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Técnicas de Placa-Clamp , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Antagonistas del Receptor Purinérgico P2/farmacología , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacología , Ratas , Simportadores de Sodio-Bicarbonato/fisiología , Suramina/farmacología , Canal Kir5.1
19.
Epilepsy Curr ; 23(3): 185-187, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37334424
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA