Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 6(1): 1053, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37853211

RESUMEN

Genetic variation accounts for much of the risk for developing a substance use disorder, but the underlying genetic factors and their genetic effector mechanisms are mostly unknown. Inbred mouse strains exhibit substantial and heritable differences in the extent of voluntary cocaine self-administration. Computational genetic analysis of cocaine self-administration data obtained from twenty-one inbred strains identified Nav1, a member of the neuron navigator family that regulates dendrite formation and axonal guidance, as a candidate gene. To test this genetic hypothesis, we generated and characterized Nav1 knockout mice. Consistent with the genetic prediction, Nav1 knockout mice exhibited increased voluntary cocaine intake and had increased motivation for cocaine consumption. Immunohistochemistry, electrophysiology, and transcriptomic studies were performed as a starting point for investigating the mechanism for the Nav1 knockout effect. Nav1 knockout mice had a reduced inhibitory synapse density in their cortex, increased excitatory synaptic transmission in their cortex and hippocampus, and increased excitatory neurons in a deep cortical layer. Collectively, our results indicate that Nav1 regulates the response to cocaine, and we identified Nav1 knockout induced changes in the excitatory and inhibitory synaptic balance in the cortex and hippocampus that could contribute to this effect.


Asunto(s)
Cocaína , Ratones , Animales , Cocaína/farmacología , Transmisión Sináptica , Neuronas , Ratones Noqueados , Hipocampo
2.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35165147

RESUMEN

Dravet syndrome (DS) is one of the most severe childhood epilepsies, characterized by intractable seizures and comorbidities including cognitive and social dysfunction and high premature mortality. DS is mainly caused by loss-of-function mutations in the Scn1a gene encoding Nav1.1 that is predominantly expressed in inhibitory parvalbumin-containing (PV) interneurons. Decreased Nav1.1 impairs PV cell function, contributing to DS phenotypes. Effective pharmacological therapy that targets defective PV interneurons is not available. The known role of brain-derived neurotrophic factor (BDNF) in the development and maintenance of interneurons, together with our previous results showing improved PV interneuronal function and antiepileptogenic effects of a TrkB receptor agonist in a posttraumatic epilepsy model, led to the hypothesis that early treatment with a TrkB receptor agonist might prevent or reduce seizure activity in DS mice. To test this hypothesis, we treated DS mice with LM22A-4 (LM), a partial agonist at the BDNF TrkB receptor, for 7 d starting at postnatal day 13 (P13), before the onset of spontaneous seizures. Results from immunohistochemistry, Western blot, whole-cell patch-clamp recording, and in vivo seizure monitoring showed that LM treatment increased the number of perisomatic PV interneuronal synapses around cortical pyramidal cells in layer V, upregulated Nav1.1 in PV neurons, increased inhibitory synaptic transmission, and decreased seizures and the mortality rate in DS mice. The results suggest that early treatment with a partial TrkB receptor agonist may be a promising therapeutic approach to enhance PV interneuron function and reduce epileptogenesis and premature death in DS.


Asunto(s)
Benzamidas/uso terapéutico , Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas/mortalidad , Receptor trkB/agonistas , Receptor trkB/metabolismo , Convulsiones/etiología , Convulsiones/genética , Animales , Epilepsias Mioclónicas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Canal de Sodio Activado por Voltaje NAV1.1/genética , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Neocórtex/citología , Células Piramidales/metabolismo , Receptor trkB/genética
3.
Cereb Cortex ; 30(9): 5180-5189, 2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32488246

RESUMEN

Decreased GABAergic inhibition due to dysfunction of inhibitory interneurons plays an important role in post-traumatic epileptogenesis. Reduced N-current Ca2+ channel function in GABAergic terminals contributes to interneuronal abnormalities and neural circuit hyperexcitability in the partial neocortical isolation (undercut, UC) model of post-traumatic epileptogenesis. Because brain-derived neurotrophic factor (BDNF) supports the development and maintenance of interneurons, we hypothesized that the activation of BDNF tropomyosin kinase B (TrkB) receptors by a small molecule, TrkB partial agonist, PTX BD4-3 (BD), would correct N channel abnormalities and enhance inhibitory synaptic transmission in UC cortex. Immunocytochemistry (ICC) and western blots were used to quantify N- and P/Q-type channels. We recorded evoked (e)IPSCs and responses to N and P/Q channel blockers to determine the effects of BD on channel function. Field potential recordings were used to determine the effects of BD on circuit hyperexcitability. Chronic BD treatment 1) upregulated N and P/Q channel immunoreactivity in GABAergic terminals; 2) increased the effects of N or P/Q channel blockade on evoked inhibitory postsynaptic currents (eIPSCs); 3) increased GABA release probability and the frequency of sIPSCs; and 4) reduced the incidence of epileptiform discharges in UC cortex. The results suggest that chronic TrkB activation is a promising approach for rescuing injury-induced calcium channel abnormalities in inhibitory terminals, thereby improving interneuronal function and suppressing circuit hyperexcitability.


Asunto(s)
Interneuronas/metabolismo , Neocórtex/metabolismo , Receptor trkB/metabolismo , Transmisión Sináptica/fisiología , Animales , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/metabolismo , Canales de Calcio/metabolismo , Epilepsia/etiología , Epilepsia/metabolismo , Masculino , Neocórtex/lesiones , Ratas , Ratas Sprague-Dawley
4.
Neurobiol Dis ; 113: 45-58, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29408225

RESUMEN

Post-traumatic epilepsy is one of the most common and difficult to treat forms of acquired epilepsy worldwide. Currently, there is no effective way to prevent post-traumatic epileptogenesis. It is known that abnormalities of interneurons, particularly parvalbumin-containing interneurons, play a critical role in epileptogenesis following traumatic brain injury. Thus, enhancing the function of existing parvalbumin interneurons might provide a logical therapeutic approach to prevention of post-traumatic epilepsy. The known positive effects of brain-derived neurotrophic factor on interneuronal growth and function through activation of its receptor tropomyosin receptor kinase B, and its decrease after traumatic brain injury, led us to hypothesize that enhancing trophic support might improve parvalbumin interneuronal function and decrease epileptogenesis. To test this hypothesis, we used the partial neocortical isolation ('undercut', UC) model of posttraumatic epileptogenesis in mature rats that were treated for 2 weeks, beginning on the day of injury, with LM22A-4, a newly designed partial agonist at the tropomyosin receptor kinase B. Effects of treatment were assessed with Western blots to measure pAKT/AKT; immunocytochemistry and whole cell patch clamp recordings to examine functional and structural properties of GABAergic interneurons; field potential recordings of epileptiform discharges in vitro; and video-EEG recordings of PTZ-induced seizures in vivo. Results showed that LM22A-4 treatment 1) increased pyramidal cell perisomatic immunoreactivity for VGAT, GAD65 and parvalbumin; 2) increased the density of close appositions of VGAT/gephyrin immunoreactive puncta (putative inhibitory synapses) on pyramidal cell somata; 3) increased the frequency of mIPSCs in pyramidal cells; and 4) decreased the incidence of spontaneous and evoked epileptiform discharges in vitro. 5) Treatment of rats with PTX BD4-3, another partial TrkB receptor agonist, reduced the incidence of bicuculline-induced ictal episodes in vitro and PTZ induced electrographic and behavioral ictal episodes in vivo. 6) Inactivation of TrkB receptors in undercut TrkBF616A mice with 1NMPP1 abolished both LM22A-4-induced effects on mIPSCs and on increased perisomatic VGAT-IR. Results indicate that chronic activation of the tropomyosin receptor kinase B by a partial agonist after cortical injury can enhance structural and functional measures of GABAergic inhibition and suppress posttraumatic epileptogenesis. Although the full agonist effects of brain-derived neurotrophic factor and tropomyosin receptor kinase B activation in epilepsy models have been controversial, the present results indicate that such trophic activation by a partial agonist may potentially serve as an effective therapeutic option for prophylactic treatment of posttraumatic epileptogenesis, and treatment of other neurological and psychiatric disorders whose pathogenesis involves impaired parvalbumin interneuronal function.


Asunto(s)
Epilepsia/metabolismo , Interneuronas/metabolismo , Glicoproteínas de Membrana/metabolismo , Parvalbúminas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Corteza Somatosensorial/metabolismo , Animales , Epilepsia/fisiopatología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Interneuronas/efectos de los fármacos , Masculino , Glicoproteínas de Membrana/agonistas , Ratones , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Corteza Somatosensorial/efectos de los fármacos , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/farmacología
5.
Neurobiol Dis ; 108: 100-114, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28823934

RESUMEN

Electrophysiological experiments in the partial cortical isolation ("undercut" or "UC") model of injury-induced neocortical epileptogenesis have shown alterations in GABAergic synaptic transmission attributable to abnormalities in presynaptic terminals. To determine whether the decreased inhibition was associated with structural abnormalities in GABAergic interneurons, we used immunocytochemical techniques, confocal microscopy and EM in UC and control sensorimotor rat cortex to analyze structural alterations in fast-spiking parvalbumin-containing interneurons and pyramidal (Pyr) cells of layer V. Principle findings were: 1) there were no decreases in counts of parvalbumin (PV)- or GABA-immunoreactive interneurons in UC cortex, however there were significant reductions in expression of VGAT and GAD-65 and -67 in halos of GABAergic terminals around Pyr somata in layer V. 2) Consistent with previous results, somatic size and density of Pyr cells was decreased in infragranular layers of UC cortex. 3) Dendrites of biocytin-filled FS interneurons were significantly decreased in volume. 4) There were decreases in the size and VGAT content of GABAergic boutons in axons of biocytin-filled FS cells in the UC, together with a decrease in colocalization with postsynaptic gephyrin, suggesting a reduction in GABAergic synapses. Quantitative EM of layer V Pyr somata confirmed the reduction in inhibitory synapses. 5) There were marked and lasting reductions in brain derived neurotrophic factor (BDNF)-IR and -mRNA in Pyr cells and decreased TrkB-IR on PV cells in UC cortex. 6) Results lead to the hypothesis that reduction in trophic support by BDNF derived from Pyr cells may contribute to the regressive changes in axonal terminals and dendrites of FS cells in the UC cortex and decreased GABAergic inhibition. SIGNIFICANCE: Injury to cortical structures is a major cause of epilepsy, accounting for about 20% of cases in the general population, with an incidence as high as ~50% among brain-injured personnel in wartime. Loss of GABAergic inhibitory interneurons is a significant pathophysiological factor associated with epileptogenesis following brain trauma and other etiologies. Results of these experiments show that the largest population of cortical interneurons, the parvalbumin-containing fast-spiking (FS) interneurons, are preserved in the partial neocortical isolation model of partial epilepsy. However, axonal terminals of these cells are structurally abnormal, have decreased content of GABA synthetic enzymes and vesicular GABA transporter and make fewer synapses onto pyramidal neurons. These structural abnormalities underlie defects in GABAergic neurotransmission that are a key pathophysiological factor in epileptogenesis found in electrophysiological experiments. BDNF, and its TrkB receptor, key factors for maintenance of interneurons and pyramidal neurons, are decreased in the injured cortex. Results suggest that supplying BDNF to the injured epileptogenic brain may reverse the structural and functional abnormalities in the parvalbumin FS interneurons and provide an antiepileptogenic therapy.


Asunto(s)
Epilepsia Postraumática/patología , Neuronas GABAérgicas/patología , Interneuronas/patología , Potenciales de Acción , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Epilepsia Postraumática/fisiopatología , Neuronas GABAérgicas/fisiología , Glutamato Descarboxilasa/metabolismo , Inmunohistoquímica , Interneuronas/fisiología , Masculino , Microscopía Confocal , Microscopía Electrónica , Neocórtex/patología , Neocórtex/fisiopatología , Parvalbúminas/metabolismo , Células Piramidales/patología , Células Piramidales/fisiología , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Receptor trkB/metabolismo , Sinaptofisina/metabolismo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
6.
Sci Rep ; 7(1): 7711, 2017 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-28794441

RESUMEN

Brain damage due to stroke or traumatic brain injury (TBI), both leading causes of serious long-term disability, often leads to the development of epilepsy. Patients who develop post-injury epilepsy tend to have poor functional outcomes. Emerging evidence highlights a potential role for blood-brain barrier (BBB) dysfunction in the development of post-injury epilepsy. However, common mechanisms underlying the pathological hyperexcitability are largely unknown. Here, we show that comparative transcriptome analyses predict remodeling of extracellular matrix (ECM) as a common response to different types of injuries. ECM-related transcriptional changes were induced by the serum protein albumin via TGFß signaling in primary astrocytes. In accordance with transcriptional responses, we found persistent degradation of protective ECM structures called perineuronal nets (PNNs) around fast-spiking inhibitory interneurons, in a rat model of TBI as well as in brains of human epileptic patients. Exposure of a naïve brain to albumin was sufficient to induce the transcriptional and translational upregulation of molecules related to ECM remodeling and the persistent breakdown of PNNs around fast-spiking inhibitory interneurons, which was contingent on TGFß signaling activation. Our findings provide insights on how albumin extravasation that occurs upon BBB dysfunction in various brain injuries can predispose neural circuitry to the development of chronic inhibition deficits.


Asunto(s)
Matriz Extracelular/metabolismo , Expresión Génica , Neuronas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Astrocitos/metabolismo , Barrera Hematoencefálica/metabolismo , Lesiones Traumáticas del Encéfalo/etiología , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Biología Computacional/métodos , Matriz Extracelular/genética , Perfilación de la Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Interneuronas/metabolismo , Losartán/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Activación Transcripcional , Transcriptoma
7.
Neurobiol Dis ; 102: 70-80, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28193459

RESUMEN

The alpha2delta-1 subunit (α2δ-1) of voltage-gated calcium channels is a receptor for astrocyte-secreted thrombospondins that promote developmental synaptogenesis. Alpha2delta-1 receptors are upregulated in models of injury-induced peripheral pain and epileptogenic neocortical trauma associated with an enhancement of excitatory synaptic connectivity. These results lead to the hypothesis that overexpression of α2δ-1 alone in neocortex of uninjured transgenic (TG) mice might result in increased excitatory connectivity and consequent cortical hyperexcitability and epileptiform activity. Whole cell recordings from layer V pyramidal neurons in somatosensory cortical slices of TG mice showed increased frequency and amplitude of miniature and spontaneous EPSCs and prolonged bursts of polysynaptic EPSCs. Epileptiform field potentials were evoked in layers II/III and V of brain slices from TG mice, but not controls. Dual immunoreactivity for Vglut-2 and PSD95 showed increased density of close appositions in TG mice compared to controls, suggesting an increased number of excitatory synapses. Video-EEG monitoring showed that 13/13 implanted TG mice aged >P21, but not controls, had frequent abnormal spontaneous epileptiform events, consisting of variable duration, high amplitude bi-hemispheric irregular bursts of delta activity, spikes and sharp waves lasting many seconds, with a variable peak frequency of ~1-3Hz, associated with behavioral arrest. The epileptiform EEG abnormalities and behavioral arrests were reversibly eliminated by treatment with i.p. ethosuximide. Behavioral seizures, consisting of ~15-30s duration episodes of rigid arched tail and head and body extension, followed by loss of balance and falling, frequently occurred in adult TG mice during recovery from isoflurane-induced anesthesia, but were rare in WT mice. Results show that over-expression of α2δ-1 subunits increases cortical excitatory connectivity and leads to neocortical hyperexcitability and epileptiform activity associated with behavioral arrests in adult TG mice. Similar increases in expression of α2δ-1 in models of cortical injury may play an important role in epileptogenesis. SIGNIFICANCE: Binding of astrocytic-secreted thrombospondins to their α2δ-1 receptor facilitates excitatory synapse formation and excitatory transmission during cortical development and after injury. Upregulation of α2δ-1 is present in models of injury-induced pain and epileptogenic cortical trauma, along with many other molecular alterations. Here we show that overexpression of α2δ-1 alone in TG mice can enhance excitatory connectivity in neocortex and lead to neural circuit hyperexcitability and episodes of electrographic epileptiform activity, associated with behavioral arrests in transgenic mice. α2δ-1 is the high-affinity receptor for gabapentinoids and a potential target for prophylactic treatment of posttraumatic epilepsy and other disorders in which excessive aberrant excitatory connectivity is a pathophysiological feature.


Asunto(s)
Canales de Calcio/metabolismo , Epilepsia/metabolismo , Corteza Somatosensorial/metabolismo , Animales , Anticonvulsivantes/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Canales de Calcio/genética , Epilepsia/tratamiento farmacológico , Epilepsia/patología , Etosuximida/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Isoflurano/toxicidad , Masculino , Ratones Transgénicos , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Células Piramidales/patología , Corteza Somatosensorial/efectos de los fármacos , Corteza Somatosensorial/patología , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Sinapsis/patología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Técnicas de Cultivo de Tejidos
8.
Neurobiol Dis ; 91: 166-81, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26956396

RESUMEN

Lesioned neuronal circuits form new functional connections after a traumatic brain injury (TBI). In humans and animal models, aberrant excitatory connections that form after TBI may contribute to the pathogenesis of post-traumatic epilepsy. Partial neocortical isolation ("undercut" or "UC") leads to altered neuronal circuitry and network hyperexcitability recorded in vivo and in brain slices from chronically lesioned neocortex. Recent data suggest a critical period for maladaptive excitatory circuit formation within the first 3days post UC injury (Graber and Prince 1999, 2004; Li et al. 2011, 2012b). The present study focuses on alterations in excitatory connectivity within this critical period. Immunoreactivity (IR) for growth-associated protein (GAP)-43 was increased in the UC cortex 3days after injury. Some GAP-43-expressing excitatory terminals targeted the somata of layer V pyramidal (Pyr) neurons, a domain usually innervated predominantly by inhibitory terminals. Immunocytochemical analysis of pre- and postsynaptic markers showed that putative excitatory synapses were present on somata of these neurons in UC neocortex. Excitatory postsynaptic currents from UC layer V Pyr cells displayed properties consistent with perisomatic inputs and also reflected an increase in the number of synaptic contacts. Laser scanning photostimulation (LSPS) experiments demonstrated reorganized excitatory connectivity after injury within the UC. Concurrent with these changes, spontaneous epileptiform bursts developed in UC slices. Results suggest that aberrant reorganization of excitatory connectivity contributes to early neocortical hyperexcitability in this model. The findings are relevant for understanding the pathophysiology of neocortical post-traumatic epileptogenesis and are important in terms of the timing of potential prophylactic treatments.


Asunto(s)
Potenciales Postsinápticos Excitadores/fisiología , Neocórtex/fisiopatología , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Traumatismos del Sistema Nervioso/fisiopatología , Animales , Proteína GAP-43/metabolismo , Masculino , Inhibición Neural/fisiología , Técnicas de Placa-Clamp/métodos , Células Piramidales/fisiología , Ratas
9.
J Neurophysiol ; 115(3): 1183-95, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26631150

RESUMEN

The GABAergic neurons of the thalamic reticular nucleus (nRt) provide the primary source of inhibition within the thalamus. Using physiology, pharmacology, and immunohistochemistry in mice, we characterized postsynaptic developmental changes in these inhibitory projection neurons. First, at postnatal days 3-5 (P3-5), inhibitory postsynaptic currents (IPSCs) decayed very slowly, followed by a biphasic developmental progression, becoming faster at P6-8 and then slower again at P9-11 before stabilizing in a mature form around P12. Second, the pharmacological profile of GABA(A) receptor (GABA(A)R)-mediated IPSCs differed between neonatal and mature nRt neurons, and this was accompanied by reciprocal changes in α3 (late) and α5 (early) subunit expression in nRt. Zolpidem, selective for α1- and α3-containing GABA(A)Rs, augmented only mature IPSCs, whereas clonazepam enhanced IPSCs at all stages. This effect was blocked by the α5-specific inverse agonist L-655,708, but only in immature neurons. In α3(H126R) mice, in which α3-subunits were mutated to become benzodiazepine insensitive, IPSCs were enhanced compared with those in wild-type animals in early development. Third, tonic GABA(A)R activation in nRt is age dependent and more prominent in immature neurons, which correlates with early expression of α5-containing GABA(A)Rs. Thus neonatal nRt neurons show relatively high expression of α5-subunits, which contributes to both slow synaptic and tonic extrasynaptic inhibition. The postnatal switch in GABA(A)R subunits from α5 to α3 could facilitate spontaneous network activity in nRt that occurs at this developmental time point and which is proposed to play a role in early circuit development.


Asunto(s)
Núcleos Talámicos Intralaminares/metabolismo , Receptores de GABA-A/metabolismo , Animales , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Potenciales Postsinápticos Inhibidores , Núcleos Talámicos Intralaminares/citología , Núcleos Talámicos Intralaminares/crecimiento & desarrollo , Núcleos Talámicos Intralaminares/fisiología , Ratones , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Receptores de GABA-A/genética
10.
Epilepsia ; 54(7): 1232-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23621154

RESUMEN

PURPOSE: To determine whether developmental synaptic pruning defects in epileptic C1q-knockout (KO) mice are accompanied by postsynaptic abnormalities in dendrites and/or spines. METHODS: Immunofluorescence staining was performed on biocytin-filled layer Vb pyramidal neurons in sensorimotor cortex. Basal dendritic arbors and their spines were reconstructed with NEUROLUCIDA software, and their morphologic characteristics were quantitated in Neuroexplorer. KEY FINDINGS: Seven to nine completely filled pyramidal neurons were analyzed from the wild-type (WT) and C1q KO groups. Compared to WT controls, KO mice showed significant structural modifications in their basal dendrites including (1) higher density of dendritic spines (0.60 ± 0.03/µm vs. 0.49 ± 0.03/µm dendritic length in WT, p < 0.05); (2) remarkably increased occurrence of thin spines (0.26 ± 0.02/µm vs. 0.14 ± 0.02/µm dendritic length in control, p < 0.01); (3) longer dendritic length (2,680 ± 159 µm vs. 2,119 ± 108 µm in control); and (4) increased branching (22.6 ± 1.9 vs. 16.2 ± 1.3 in WT at 80 µm from soma center, p < 0.05; 12.4 ± 1.4 vs. 8.2 ± 0.6 in WT at 120 µm from soma center, respectively, p < 0.05). Dual immunolabeling demonstrated the expression of putative glutamate receptor 2 (GluR2) on some thin spines. These dendritic alterations are likely postsynaptic structural consequences of failure of synaptic pruning in the C1q KO mice. SIGNIFICANCE: Failure to prune excessive excitatory synapses in C1q KO mice is a likely mechanism underlying abnormalities in postsynaptic dendrites, including increased branching and alterations in spine type and density. It is also possible that seizure activity contributes to these abnormalities. These structural abnormalities, together with increased numbers of excitatory synapses, likely contribute to epileptogenesis in C1q KO mice.


Asunto(s)
Complemento C1q/deficiencia , Dendritas/patología , Espinas Dendríticas/patología , Epilepsia/genética , Epilepsia/patología , Células Piramidales/patología , Animales , Complemento C1q/genética , Dendritas/genética , Dendritas/ultraestructura , Espinas Dendríticas/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Imagenología Tridimensional , Masculino , Ratones , Ratones Noqueados , Neocórtex/patología , Receptores AMPA/metabolismo
11.
Nat Neurosci ; 16(1): 64-70, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23143518

RESUMEN

Cerebrocortical injuries such as stroke are a major source of disability. Maladaptive consequences can result from post-injury local reorganization of cortical circuits. For example, epilepsy is a common sequela of cortical stroke, but the mechanisms responsible for seizures following cortical injuries remain unknown. In addition to local reorganization, long-range, extra-cortical connections might be critical for seizure maintenance. In rats, we found that the thalamus, a structure that is remote from, but connected to, the injured cortex, was required to maintain cortical seizures. Thalamocortical neurons connected to the injured epileptic cortex underwent changes in HCN channel expression and became hyperexcitable. Targeting these neurons with a closed-loop optogenetic strategy revealed that reducing their activity in real-time was sufficient to immediately interrupt electrographic and behavioral seizures. This approach is of therapeutic interest for intractable epilepsy, as it spares cortical function between seizures, in contrast with existing treatments, such as surgical lesioning or drugs.


Asunto(s)
Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Corteza Cerebral/fisiopatología , Vías Nerviosas/fisiología , Optogenética , Convulsiones/etiología , Tálamo/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Fenómenos Biofísicos/fisiología , Biofisica , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Modelos Animales de Enfermedad , Capacidad Eléctrica , Estimulación Eléctrica , Electroencefalografía , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Técnicas In Vitro , Canales Iónicos/genética , Canales Iónicos/metabolismo , Luz , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Lisina/análogos & derivados , Lisina/metabolismo , Potenciales de la Membrana/genética , Microscopía Confocal , Modelos Neurológicos , Inhibición Neural/genética , Neuronas/efectos de los fármacos , Neuronas/fisiología , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Análisis Espectral , Vigilia/genética
12.
J Perinat Med ; 40(4): 427-31, 2012 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-22752775

RESUMEN

OBJECTIVE: The objective of this study was to determine whether acquisition of normal stooling pattern is associated with full enteral feeding and growth. STUDY DESIGN: This is a prospective observational study of infants with a gestational age of <28 weeks (n=121). All infants admitted to our unit during the study period were managed using the same protocol for rectal stimulation and enemas: we used rectal stimulation if abdominal distension and no defecation occurred in the previous 24 h; enema administration was practiced if abdominal distension persists and no defecation had occurred after rectal stimulation. Age of normal bowel habit (T-NBH) is defined as days when two stools passed each day without enemas or stimulation for three consecutive days. Full enteral feeding is measured by time to receive 120 mL/kg/day (T-120). Multivariable linear regression estimated the contribution of T-NBH on T-120 and the effect of tolerance on growth. RESULTS: T-NBH was 16 days (12, 24 days) and T-120 was 16 days (12, 24 days). T-120 decreased with earlier normal bowel habit (r=0.625, P<0.0001). Change in weight z-score between 36 weeks postmenstrual age and birth was less pronounced with earlier full enteral feeding (r=-0.446, P<0.0001). CONCLUSION: Normalization of bowel habit is an important determinant of full enteral feeding of very immature infants. Earlier full enteral feeding is associated with less postnatal growth failure.


Asunto(s)
Defecación/fisiología , Nutrición Enteral , Recien Nacido Prematuro/fisiología , Femenino , Tracto Gastrointestinal/crecimiento & desarrollo , Tracto Gastrointestinal/fisiología , Tránsito Gastrointestinal/fisiología , Edad Gestacional , Humanos , Recién Nacido , Recien Nacido Prematuro/crecimiento & desarrollo , Masculino , Estimulación Física , Estudios Prospectivos , Recto
13.
Neurobiol Dis ; 45(2): 821-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22172650

RESUMEN

Decreased release probability (Pr) and increased failure rate for monosynaptic inhibitory postsynaptic currents (IPSCs) indicate abnormalities in presynaptic inhibitory terminals on pyramidal (Pyr) neurons of the undercut (UC) model of posttraumatic epileptogenesis. These indices of inhibition are normalized in high [Ca++] ACSF, suggesting dysfunction of Ca2+ channels in GABAergic terminals. We tested this hypothesis using selective blockers of P/Q and N-type Ca2+ channels whose activation underlies transmitter release in cortical inhibitory terminals. Pharmacologically isolated monosynaptic IPSCs were evoked in layer V Pyr cells by extracellular stimuli in adult rat sensorimotor cortical slices. Local perfusion of 0.2/1 µM ω-agatoxin IVa and/or 1 µM ω-conotoxin GVIA was used to block P/Q and N-type calcium channels, respectively. In control layer V Pyr cells, peak amplitude of eIPSCs was decreased by ~50% after treatment with either 1 µM ω-conotoxin GVIA or 1 µM ω-agatoxin IVa. In contrast, there was a lack of sensitivity to 1 µM ω-conotoxin GVIA in UCs. Immunocytochemical results confirmed decreased perisomatic density of N-channels on Pyr cells in UCs. We suggest that decreased calcium influx via N-type channels in presynaptic GABAergic terminals is a mechanism contributing to decreased inhibitory input onto layer V Pyr cells in this model of cortical posttraumatic epileptogenesis.


Asunto(s)
Canales de Calcio/metabolismo , Epilepsia/metabolismo , Neocórtex/metabolismo , Células Piramidales/metabolismo , Animales , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/metabolismo , Epilepsia/etiología , Inmunohistoquímica , Potenciales Postsinápticos Inhibidores/fisiología , Masculino , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley
14.
Neurosci Lett ; 497(3): 172-6, 2011 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-21354270

RESUMEN

Prophylaxis of posttraumatic epilepsy will require a detailed knowledge of the epileptogenic pathophysiological processes that follow brain injury. Results from studies of experimental models and human epilepsy highlight alterations in GABAergic interneurons and formation of excessive new excitatory synaptic connectivity as prominent targets for prophylactic therapies. Promising laboratory results suggest that it will be possible to experimentally modify these aberrant processes and interfere with epileptogenesis. However, a number of key issues must be addressed before these results can be used to frame clinical antiepileptogenic therapy.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Lesiones Encefálicas/fisiopatología , Lesiones Encefálicas/terapia , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Epilepsia/prevención & control , Epilepsia/fisiopatología , Animales , Encéfalo/efectos de los fármacos , Lesiones Encefálicas/complicaciones , Epilepsia/etiología , Humanos , Ratas
15.
Epilepsia ; 51 Suppl s5: 30, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21158780

RESUMEN

Development of new excitatory connectivity and decreases in γ-aminobutyric acid (GABA)ergic inhibition are mechanisms underlying posttraumatic epileptogenesis in animal models. Experimental strategies that interfere with these processes, applied between the trauma and seizure onset, are antiepileptogenic in the laboratory, and have promise for prophylaxis of epileptogenesis after cortical injury in humans. For an expanded treatment of this topic see Jasper's Basic Mechanisms of the Epilepsies, Fourth Edition (Noebels JL, Avoli M, Rogawski MA, Olsen RW, Delgado-Escueta AV, eds) National Library of Medicine Bookshelf [NCBI] at http://www.ncbi.nlm.nih.gov/books).

16.
J Neurophysiol ; 104(4): 2214-23, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20631216

RESUMEN

The density of somatostatin (SOM)-containing GABAergic interneurons in the hilus of the dentate gyrus is significantly decreased in both human and experimental temporal lobe epilepsy. We used the pilocarpine model of status epilepticus and temporal lobe epilepsy in mice to study anatomical and electrophysiological properties of surviving somatostatin interneurons and determine whether compensatory functional changes occur that might offset loss of other inhibitory neurons. Using standard patch-clamp techniques and pipettes containing biocytin, whole cell recordings were obtained in hippocampal slices maintained in vitro. Hilar SOM cells containing enhanced green fluorescent protein (EGFP) were identified with fluorescent and infrared differential interference contrast video microscopy in epileptic and control GIN (EGFP-expressing Inhibitory Neurons) mice. Results showed that SOM cells from epileptic mice had 1) significant increases in somatic area and dendritic length; 2) changes in membrane properties, including a small but significant decrease in resting membrane potential, and increases in time constant and whole cell capacitance; 3) increased frequency of slowly rising spontaneous excitatory postsynaptic currents (sEPSCs) due primarily to increased mEPSC frequency, without changes in the probability of release; 4) increased evoked EPSC amplitude; and 5) increased spontaneous action potential generation in cell-attached recordings. Results suggest an increase in excitatory innervation, perhaps on distal dendrites, considering the slower rising EPSCs and increased output of hilar SOM cells in this model of epilepsy. In sum, these changes would be expected to increase the inhibitory output of surviving SOM interneurons and in part compensate for interneuronal loss in the epileptogenic hippocampus.


Asunto(s)
Giro Dentado/metabolismo , Modelos Animales de Enfermedad , Epilepsia/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Interneuronas/metabolismo , Somatostatina/biosíntesis , Animales , Enfermedad Crónica , Giro Dentado/química , Giro Dentado/patología , Epilepsia/patología , Femenino , Interneuronas/química , Interneuronas/patología , Masculino , Ratones , Técnicas de Cultivo de Órganos
17.
J Neurosci ; 30(15): 5465-79, 2010 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-20392967

RESUMEN

Focal cortical injuries result in death of cortical neurons and their efferents and ultimately in death or damage of thalamocortical relay (TCR) neurons that project to the affected cortical area. Neurons of the inhibitory reticular thalamic nucleus (nRT) receive excitatory inputs from corticothalamic and thalamocortical axons and are thus denervated by such injuries, yet nRT cells generally survive these insults to a greater degree than TCR cells. nRT cells inhibit TCR cells, regulate thalamocortical transmission, and generate cerebral rhythms including those involved in thalamocortical epilepsies. The survival and reorganization of nRT after cortical injury would determine recovery of thalamocortical circuits after injury. However, the physiological properties and connectivity of the survivors remain unknown. To study possible alterations in nRT neurons, we used the rat photothrombosis model of cortical stroke. Using in vitro patch-clamp recordings at various times after the photothrombotic injury, we show that localized strokes in the somatosensory cortex induce long-term reductions in intrinsic excitability and evoked synaptic excitation of nRT cells by the end of the first week after the injury. We find that nRT neurons in injured rats show (1) decreased membrane input resistance, (2) reduced low-threshold calcium burst responses, and (3) weaker evoked excitatory synaptic responses. Such alterations in nRT cellular excitability could lead to loss of nRT-mediated inhibition in relay nuclei, increased output of surviving TCR cells, and enhanced thalamocortical excitation, which may facilitate recovery of thalamic and cortical sensory circuits. In addition, such changes could be maladaptive, leading to injury-induced epilepsy.


Asunto(s)
Infarto Encefálico/fisiopatología , Corteza Cerebral/fisiopatología , Neuronas/fisiología , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Núcleos Talámicos/fisiopatología , Animales , Infarto Encefálico/patología , Calcio/metabolismo , Canales de Calcio Tipo T/metabolismo , Muerte Celular , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Impedancia Eléctrica , Potenciales Evocados , Técnicas In Vitro , Potenciales de la Membrana/fisiología , Vías Nerviosas/fisiopatología , Plasticidad Neuronal/fisiología , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Accidente Cerebrovascular/fisiopatología , Sinapsis/patología , Núcleos Talámicos/patología , Factores de Tiempo
18.
Proc Natl Acad Sci U S A ; 107(17): 7975-80, 2010 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-20375278

RESUMEN

Excessive CNS synapses are eliminated during development to establish mature patterns of neuronal connectivity. A complement cascade protein, C1q, is involved in this process. Mice deficient in C1q fail to refine retinogeniculate connections resulting in excessive retinal innervation of lateral geniculate neurons. We hypothesized that C1q knockout (KO) mice would exhibit defects in neocortical synapse elimination resulting in enhanced excitatory synaptic connectivity and epileptiform activity. We recorded spontaneous and evoked field potential activity in neocortical slices and obtained video-EEG recordings from implanted C1q KO and wild-type (WT) mice. We also used laser scanning photostimulation of caged glutamate and whole cell recordings to map excitatory and inhibitory synaptic connectivity. Spontaneous and evoked epileptiform field potentials occurred at multiple sites in neocortical slices from C1q KO, but not WT mice. Laser mapping experiments in C1q KO slices showed that the proportion of glutamate uncaging sites from which excitatory postsynaptic currents (EPSCs) could be evoked ("hotspot ratio") increased significantly in layer IV and layer V, although EPSC amplitudes were unaltered. Density of axonal boutons was significantly increased in layer V pyramidal neurons of C1q KO mice. Implanted KO mice had frequent behavioral seizures consisting of behavioral arrest associated with bihemispheric spikes and slow wave activity lasting from 5 to 30 s. Results indicate that epileptogenesis in C1q KO mice is related to a genetically determined failure to prune excessive excitatory synapses during development.


Asunto(s)
Complemento C1q/deficiencia , Epilepsia/fisiopatología , Neocórtex/fisiopatología , Sinapsis/fisiología , Análisis de Varianza , Animales , Complemento C1q/genética , Complemento C1q/metabolismo , Electroencefalografía , Potenciales Evocados , Glutamatos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía por Video , Técnicas de Placa-Clamp , Estimulación Luminosa , Sinapsis/metabolismo
19.
Epilepsia ; 51 Suppl 5: 30, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22056919

RESUMEN

Development of new excitatory connectivity and decreases in GABAergic inhibition are mechanisms underlying posttraumatic epileptogenesis in animal models. Experimental strategies that interfere with these processes, applied between the trauma andseizure onset, are antiepileptogenic in the laboratory, and have promise for prophylaxis of epileptogenesis after cortical injury in man. For an expanded treatment of this topic see Jasper's Basic Mechanisms of the Epilepsies, Fourth Edition (Noebels JL, Avoli M, Rogawski MA, Olsen RW, Delgado-Escueta AV, eds) published by Oxford University Press. Available on NCBI Bookshelf.

20.
Epilepsia ; 50 Suppl 2: 30-40, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19187292

RESUMEN

The sequelae of traumatic brain injury, including posttraumatic epilepsy, represent a major societal problem. Significant resources are required to develop a better understanding of the underlying pathophysiologic mechanisms as targets for potential prophylactic therapies. Posttraumatic epilepsy undoubtedly involves numerous pathogenic factors that develop more or less in parallel. We have highlighted two potential "prime movers": disinhibition and development of new functional excitatory connectivity, which occur in a number of animal models and some forms of epilepsy in humans. Previous experiments have shown that tetrodotoxin (TTX) applied to injured cortex during a critical period early after lesion placement can prevent epileptogenesis in the partial cortical ("undercut") model of posttraumatic epilepsy. Here we show that such treatment markedly attenuates histologic indices of axonal and terminal sprouting and presumably associated aberrant excitatory connectivity. A second finding in the undercut model is a decrease in spontaneous inhibitory events. Current experiments show that this is accompanied by regressive alterations in fast-spiking gamma-aminobutyric acid (GABA)ergic interneurons, including shrinkage of dendrites, marked decreases in axonal length, structural changes in inhibitory boutons, and loss of inhibitory synapses on pyramidal cells. Other data support the hypothesis that these anatomic abnormalities may result from loss of trophic support normally provided to interneurons by brain-derived neurotrophic factor (BDNF). Approaches that prevent these two pathophysiologic mechanisms may offer avenues for prophylaxis for posttraumatic epilepsy. However, major issues such as the role of these processes in functional recovery from injury and the timing of the critical period(s) for application of potential therapies in humans need to be resolved.


Asunto(s)
Lesiones Encefálicas/complicaciones , Corteza Cerebral/lesiones , Epilepsia Postraumática/fisiopatología , Animales , Anticonvulsivantes/farmacología , Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Epilepsia Postraumática/patología , Epilepsia Postraumática/prevención & control , Humanos , Interneuronas/efectos de los fármacos , Interneuronas/patología , Interneuronas/fisiología , Red Nerviosa/efectos de los fármacos , Red Nerviosa/patología , Red Nerviosa/fisiopatología , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/fisiología , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Células Piramidales/efectos de los fármacos , Células Piramidales/patología , Células Piramidales/fisiología , Tetrodotoxina/farmacología , Ácido gamma-Aminobutírico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...