Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 158
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35145028

RESUMEN

The cerebellum, the site where protein kinase C (PKC) was first discovered, contains the highest amount of PKC in the central nervous system, with PKCγ being the major isoform. Systemic PKCγ-knockout (KO) mice showed impaired motor coordination and deficient pruning of surplus climbing fibers (CFs) from developing cerebellar Purkinje cells (PCs). However, the physiological significance of PKCγ in the mature cerebellum and the cause of motor incoordination remain unknown. Using adeno-associated virus vectors targeting PCs, we showed that impaired motor coordination was restored by re-expression of PKCγ in mature PKCγ-KO mouse PCs in a kinase activity-dependent manner, while normal motor coordination in mature Prkcgfl/fl mice was impaired by the Cre-dependent removal of PKCγ from PCs. Notably, the rescue or removal of PKCγ from mature PKCγ-KO or Prkcgfl/fl mice, respectively, did not affect the CF innervation profile of PCs, suggesting the presence of a mechanism distinct from multiple CF innervation of PCs for the motor defects in PKCγ-deficient mice. We found marked potentiation of Ca2+-activated large-conductance K+ (BK) channel currents in PKCγ-deficient mice, as compared to wild-type mice, which decreased the membrane resistance, resulting in attenuation of the electrical signal during the propagation and significant alterations of the complex spike waveform. These changes in PKCγ-deficient mice were restored by the rescue of PKCγ or pharmacological suppression of BK channels. Our results suggest that PKCγ is a critical regulator that negatively modulates BK currents in PCs, which significantly influences PC output from the cerebellar cortex and, eventually, motor coordination.


Asunto(s)
Terapia Genética , Actividad Motora/genética , Canales de Potasio Calcio-Activados/metabolismo , Proteína Quinasa C/metabolismo , Células de Purkinje/enzimología , Animales , Señalización del Calcio , Eliminación de Gen , Ratones , Ratones Noqueados , Actividad Motora/fisiología , Canales de Potasio Calcio-Activados/genética , Proteína Quinasa C/genética , Potenciales Sinápticos
2.
Sci Rep ; 11(1): 22050, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34764308

RESUMEN

Gamma-aminobutyric acid (GABA)-releasing interneurons modulate neuronal network activity in the brain by inhibiting other neurons. The alteration or absence of these cells disrupts the balance between excitatory and inhibitory processes, leading to neurological disorders such as epilepsy. In this regard, cell-based therapy may be an alternative therapeutic approach. We generated light-sensitive human embryonic stem cell (hESC)-derived GABAergic interneurons (hdIN) and tested their functionality. After 35 days in vitro (DIV), hdINs showed electrophysiological properties and spontaneous synaptic currents comparable to mature neurons. In co-culture with human cortical neurons and after transplantation (AT) into human brain tissue resected from patients with drug-resistant epilepsy, light-activated channelrhodopsin-2 (ChR2) expressing hdINs induced postsynaptic currents in human neurons, strongly suggesting functional efferent synapse formation. These results provide a proof-of-concept that hESC-derived neurons can integrate and modulate the activity of a human host neuronal network. Therefore, this study supports the possibility of precise temporal control of network excitability by transplantation of light-sensitive interneurons.


Asunto(s)
Neuronas GABAérgicas/citología , Células Madre Embrionarias Humanas/citología , Red Nerviosa/citología , Animales , Línea Celular , Células Cultivadas , Técnicas de Cocultivo , Neuronas GABAérgicas/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Humanos , Ratones , Red Nerviosa/fisiología , Neurogénesis , Potenciales Sinápticos
3.
J Exp Med ; 218(7)2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-33978701

RESUMEN

Atypical antipsychotics such as risperidone cause drug-induced metabolic syndrome. However, the underlying mechanisms remain largely unknown. Here, we report a new mouse model that reliably reproduces risperidone-induced weight gain, adiposity, and glucose intolerance. We found that risperidone treatment acutely altered energy balance in C57BL/6 mice and that hyperphagia accounted for most of the weight gain. Transcriptomic analyses in the hypothalamus of risperidone-fed mice revealed that risperidone treatment reduced the expression of Mc4r. Furthermore, Mc4r in Sim1 neurons was necessary for risperidone-induced hyperphagia and weight gain. Moreover, we found that the same pathway underlies the obesogenic effect of olanzapine-another commonly prescribed antipsychotic drug. Remarkably, whole-cell patch-clamp recording demonstrated that risperidone acutely inhibited the activity of hypothalamic Mc4r neurons via the opening of a postsynaptic potassium conductance. Finally, we showed that treatment with setmelanotide, an MC4R-specific agonist, mitigated hyperphagia and obesity in both risperidone- and olanzapine-fed mice.


Asunto(s)
Antipsicóticos/farmacología , Receptor de Melanocortina Tipo 4/metabolismo , Risperidona/farmacología , Aumento de Peso/efectos de los fármacos , Animales , Femenino , Hiperfagia/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Síndrome Metabólico/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Obesidad/metabolismo , Olanzapina/farmacología , Potasio/metabolismo , Potenciales Sinápticos/efectos de los fármacos , Transcriptoma/efectos de los fármacos , alfa-MSH/análogos & derivados , alfa-MSH/farmacología
4.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33558223

RESUMEN

The perception of and response to danger is critical for an individual's survival and is encoded by subcortical neurocircuits. The amygdaloid complex is the primary neuronal site that initiates bodily reactions upon external threat with local-circuit interneurons scaling output to effector pathways. Here, we categorize central amygdala neurons that express secretagogin (Scgn), a Ca2+-sensor protein, as a subset of protein kinase Cδ (PKCδ)+ interneurons, likely "off cells." Chemogenetic inactivation of Scgn+/PKCδ+ cells augmented conditioned response to perceived danger in vivo. While Ca2+-sensor proteins are typically implicated in shaping neurotransmitter release presynaptically, Scgn instead localized to postsynaptic compartments. Characterizing its role in the postsynapse, we found that Scgn regulates the cell-surface availability of NMDA receptor 2B subunits (GluN2B) with its genetic deletion leading to reduced cell membrane delivery of GluN2B, at least in vitro. Conclusively, we describe a select cell population, which gates danger avoidance behavior with secretagogin being both a selective marker and regulatory protein in their excitatory postsynaptic machinery.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Interneuronas/metabolismo , Proteína Quinasa C-delta/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Secretagoginas/metabolismo , Amígdala del Cerebelo/citología , Amígdala del Cerebelo/fisiología , Animales , Reacción de Prevención , Línea Celular Tumoral , Células Cultivadas , Miedo , Femenino , Humanos , Interneuronas/fisiología , Masculino , Transporte de Proteínas , Ratas , Ratas Wistar , Secretagoginas/genética , Potenciales Sinápticos
5.
Clin Transl Sci ; 14(2): 568-581, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33340242

RESUMEN

Chemotherapy-induced peripheral neuropathy (CIPN) is a dose-limiting adverse event associated with treatment with paclitaxel and other chemotherapeutic agents. The prevention and treatment of CIPN are limited by a lack of understanding of the molecular mechanisms underlying this toxicity. In the current study, a human induced pluripotent stem cell-derived sensory neuron (iPSC-SN) model was developed for the study of chemotherapy-induced neurotoxicity. The iPSC-SNs express proteins characteristic of nociceptor, mechanoreceptor, and proprioceptor sensory neurons and show Ca2+ influx in response to capsaicin, α,ß-meATP, and glutamate. The iPSC-SNs are relatively resistant to the cytotoxic effects of paclitaxel, with half-maximal inhibitory concentration (IC50 ) values of 38.1 µM (95% confidence interval (CI) 22.9-70.9 µM) for 48-hour exposure and 9.3 µM (95% CI 5.7-16.5 µM) for 72-hour treatment. Paclitaxel causes dose-dependent and time-dependent changes in neurite network complexity detected by ßIII-tubulin staining and high content imaging. The IC50 for paclitaxel reduction of neurite area was 1.4 µM (95% CI 0.3-16.9 µM) for 48-hour exposure and 0.6 µM (95% CI 0.09-9.9 µM) for 72-hour exposure. Decreased mitochondrial membrane potential, slower movement of mitochondria down the neurites, and changes in glutamate-induced neuronal excitability were also observed with paclitaxel exposure. The iPSC-SNs were also sensitive to docetaxel, vincristine, and bortezomib. Collectively, these data support the use of iPSC-SNs for detailed mechanistic investigations of genes and pathways implicated in chemotherapy-induced neurotoxicity and the identification of novel therapeutic approaches for its prevention and treatment.


Asunto(s)
Antineoplásicos/efectos adversos , Paclitaxel/efectos adversos , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Células Receptoras Sensoriales/patología , Línea Celular , Humanos , Células Madre Pluripotentes Inducidas , Microscopía Intravital , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Imagen Óptica , Enfermedades del Sistema Nervioso Periférico/patología , Células Receptoras Sensoriales/citología , Células Receptoras Sensoriales/efectos de los fármacos , Potenciales Sinápticos/efectos de los fármacos
6.
Neuroendocrinology ; 111(12): 1219-1230, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33361699

RESUMEN

INTRODUCTION: Hypophysiotropic gonadotropin-releasing hormone (GnRH) neurons orchestrate various physiological events that control the onset of puberty. Previous studies showed that insulin-like growth factor 1 (IGF-1) induces the secretion of GnRH and accelerates the onset of puberty, suggesting a regulatory role of this hormone upon GnRH neurons. METHODS: To reveal responsiveness of GnRH neurons to IGF-1 and elucidate molecular pathways acting downstream to the IGF-1 receptor (IGF-1R), in vitro electrophysiological experiments were carried out on GnRH-GFP neurons in acute brain slices from prepubertal (23-29 days) and pubertal (50 days) male mice. RESULTS: Administration of IGF-1 (13 nM) significantly increased the firing rate and frequency of spontaneous postsynaptic currents and that of excitatory GABAergic miniature postsynaptic currents (mPSCs). No GABAergic mPSCs were induced by IGF-1 in the presence of the GABAA-R blocker picrotoxin. The increase in the mPSC frequency was prevented by the use of the IGF-1R antagonist, JB1 (1 µM), or the intracellularly applied PI3K blocker (LY294002, 50 µM), showing involvement of IGF-1R and PI3K in the mechanism. Blockade of the transient receptor potential vanilloid 1, an element of the tonic retrograde endocannabinoid machinery, by AMG9810 (10 µM) or antagonizing the cannabinoid receptor type-1 by AM251 (1 µM) abolished the effect. DISCUSSION/CONCLUSION: These findings indicate that IGF-1 arrests the tonic retrograde endocannabinoid pathway in GnRH neurons, and this disinhibition increases the release of GABA from presynaptic terminals that, in turn, activates GnRH neurons leading to the fine-tuning of the hypothalamo-pituitary-gonadal axis.


Asunto(s)
Endocannabinoides/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Neuronas/fisiología , Pubertad/metabolismo , Transducción de Señal/fisiología , Potenciales Sinápticos/fisiología , Ácido gamma-Aminobutírico/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Masculino , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Transducción de Señal/efectos de los fármacos , Potenciales Sinápticos/efectos de los fármacos
7.
Biol Open ; 9(12)2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33148607

RESUMEN

The amyotrophic lateral sclerosis (ALS) neurodegenerative disorder has been associated with multiple genetic lesions, including mutations in the gene for fused in sarcoma (FUS), a nuclear-localized RNA/DNA-binding protein. Neuronal expression of the pathological form of FUS proteins in Caenorhabditis elegans results in mislocalization and aggregation of FUS in the cytoplasm, and leads to impairment of motility. However, the mechanisms by which the mutant FUS disrupts neuronal health and function remain unclear. Here we investigated the impact of ALS-associated FUS on motor neuron health using correlative light and electron microscopy, electron tomography, and electrophysiology. We show that ectopic expression of wild-type or ALS-associated human FUS impairs synaptic vesicle docking at neuromuscular junctions. ALS-associated FUS led to the emergence of a population of large, electron-dense, and filament-filled endosomes. Electrophysiological recording revealed reduced transmission from motor neurons to muscles. Together, these results suggest a pathological effect of ALS-causing FUS at synaptic structure and function organization.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Esclerosis Amiotrófica Lateral/etiología , Expresión Génica , Mutación , Unión Neuromuscular/genética , Unión Neuromuscular/fisiopatología , Proteína FUS de Unión a ARN/genética , Transmisión Sináptica/genética , Animales , Caenorhabditis elegans , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Endosomas/metabolismo , Endosomas/ultraestructura , Humanos , Neuronas Motoras/metabolismo , Neuronas Motoras/ultraestructura , Unión Neuromuscular/patología , Unión Neuromuscular/ultraestructura , Potenciales Sinápticos
8.
J Neurosci ; 40(39): 7405-7420, 2020 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-32847966

RESUMEN

The AMPA subtype of synaptic glutamate receptors (AMPARs) plays an essential role in cognition. Their function, numbers, and change at synapses during synaptic plasticity are tightly regulated by neuronal activity. Although we know that long-distance transport of AMPARs is essential for this regulation, we do not understand the associated regulatory mechanisms of it. Neuronal transmission is a metabolically demanding process in which ATP consumption and production are tightly coupled and regulated. Aerobic ATP synthesis unavoidably produces reactive oxygen species (ROS), such as hydrogen peroxide, which are known modulators of calcium signaling. Although a role for calcium signaling in AMPAR transport has been described, there is little understanding of the mechanisms involved and no known link to physiological ROS signaling. Here, using real-time in vivo imaging of AMPAR transport in the intact C. elegans nervous system, we demonstrate that long-distance synaptic AMPAR transport is bidirectionally regulated by calcium influx and activation of calcium/calmodulin-dependent protein kinase II. Quantification of in vivo calcium dynamics revealed that modest, physiological increases in ROS decrease calcium transients in C. elegans glutamatergic neurons. By combining genetic and pharmacological manipulation of ROS levels and calcium influx, we reveal a mechanism in which physiological increases in ROS cause a decrease in synaptic AMPAR transport and delivery by modulating activity-dependent calcium signaling. Together, our results identify a novel role for oxidant signaling in the regulation of synaptic AMPAR transport and delivery, which in turn could be critical for coupling the metabolic demands of neuronal activity with excitatory neurotransmission.SIGNIFICANCE STATEMENT Synaptic AMPARs are critical for excitatory synaptic transmission. The disruption of their synaptic localization and numbers is associated with numerous psychiatric, neurologic, and neurodegenerative conditions. However, very little is known about the regulatory mechanisms controlling transport and delivery of AMPAR to synapses. Here, we describe a novel physiological signaling mechanism in which ROS, such as hydrogen peroxide, modulate AMPAR transport by modifying activity-dependent calcium signaling. Our findings provide the first evidence in support of a mechanistic link between physiological ROS signaling, AMPAR transport, localization, and excitatory transmission. This is of fundamental and clinical significance since dysregulation of intracellular calcium and ROS signaling is implicated in aging and the pathogenesis of several neurodegenerative disorders, including Alzheimer's and Parkinson's disease.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores AMPA/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Transporte Biológico , Caenorhabditis elegans , Señalización del Calcio , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Neuronas/metabolismo , Neuronas/fisiología , Potenciales Sinápticos
9.
J Neurophysiol ; 124(3): 703-714, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32727281

RESUMEN

The large-conductance calcium-activated potassium (BK) channel is a critical regulator of neuronal action potential firing and follows two distinct trends in early postnatal development: an increase in total expression and a shift from the faster activating STREX isoform to the slower ZERO isoform. We analyzed the functional consequences of developmental trends in BK channel expression in hippocampal neurons isolated from neonatal rats aged 1 to 7 days. Following overnight cultures, action potentials and currents were recorded using whole cell patch-clamp electrophysiology. These neurons undergo a steady increase in excitability during this time, and the effect of blockade of BK channel activity with 100 nM iberiotoxin changes as the neurons mature. BK currents contribute significantly more to total potassium current and single action potentials in neurons of 1-day old rats (with BK blockade extending action potential duration by 0.46 ± 0.12 ms) than in those of 7-day old rats (with BK blockade extending action potential duration by 0.17 ± 0.05 ms). BK currents contribute consistently to maintain firing rates in neurons of 1-day old rats throughout extended action potential firing; BK blockade evenly depresses firing frequency across action potential trains. In neurons from 7-day old rats, BK blockade initially increases firing frequency and then progressively decreases frequency as firing continues, ultimately depressing neuronal firing rates to a greater extent than in the neurons from 1-day-old animals. These results are consistent with a transition from low expression of a fast-activating BK isoform (STREX) to high expression of a slower activating isoform (ZERO).NEW & NOTEWORTHY This work describes the early developmental trends of large-conductance calcium-activated potassium (BK) channel activity. Early developmental trends in expression of BK channels, both total expression and relative isoform expression, have been previously reported, but little work describes the effect of these changes in expression patterns on excitability. Here, we show that early changes in BK channel expression patterns lead to changes in the role of BK channels in determining the action potential waveform and neuronal excitability.


Asunto(s)
Potenciales de Acción/fisiología , Hipocampo/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Neuronas/fisiología , Bloqueadores de los Canales de Potasio/farmacología , Potenciales Sinápticos/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Animales Recién Nacidos , Hipocampo/efectos de los fármacos , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Péptidos/farmacología , Ratas , Ratas Sprague-Dawley , Potenciales Sinápticos/efectos de los fármacos
10.
Front Neural Circuits ; 14: 24, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32581725

RESUMEN

cAMP is a positive regulator tightly involved in certain types of synaptic plasticity and related memory functions. However, its spatiotemporal roles at the synaptic and neural circuit levels remain elusive. Using a combination of a cAMP optogenetics approach and voltage-sensitive dye (VSD) imaging with electrophysiological recording, we define a novel capacity of postsynaptic cAMP in enabling dentate gyrus long-term potentiation (LTP) and depolarization in acutely prepared murine hippocampal slices. To manipulate cAMP levels at medial perforant path to granule neuron (MPP-DG) synapses by light, we generated transgenic (Tg) mice expressing photoactivatable adenylyl cyclase (PAC) in DG granule neurons. Using these Tg(CMV-Camk2a-RFP/bPAC)3Koka mice, we recorded field excitatory postsynaptic potentials (fEPSPs) from MPP-DG synapses and found that photoactivation of PAC during tetanic stimulation enabled synaptic potentiation that persisted for at least 30 min. This form of LTP was induced without the need for GABA receptor blockade that is typically required for inducing DG plasticity. The paired-pulse ratio (PPR) remained unchanged, indicating the cAMP-dependent LTP was likely postsynaptic. By employing fast fluorescent voltage-sensitive dye (VSD: di-4-ANEPPS) and fluorescence imaging, we found that photoactivation of the PAC actuator enhanced the intensity and extent of dentate gyrus depolarization triggered following tetanic stimulation. These results demonstrate that the elevation of cAMP in granule neurons is capable of rapidly enhancing synaptic strength and neuronal depolarization. The powerful actions of cAMP are consistent with this second messenger having a critical role in the regulation of synaptic function.


Asunto(s)
AMP Cíclico/fisiología , Giro Dentado/química , Giro Dentado/fisiología , Plasticidad Neuronal/fisiología , Optogenética/métodos , Potenciales Sinápticos/fisiología , Animales , AMP Cíclico/análisis , Hipocampo/química , Hipocampo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Periodo Refractario Electrofisiológico/fisiología , Transmisión Sináptica/fisiología
11.
FASEB J ; 34(4): 5563-5577, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32086857

RESUMEN

Transcriptome data revealed α1 adrenoceptors (ARs) expression in platelet-derived growth factor receptor α+ cells (PDGFRα+ cells) in murine colonic musculature. The role of PDGFRα+ cells in sympathetic neural regulation of murine colonic motility was investigated. Norepinephrine (NE), via α1A ARs, activated a small conductance Ca2+ -activated K+ (SK) conductance, evoked outward currents and hyperpolarized PDGFRα+ cells (the α1A AR-SK channel signal pathway). α1 AR agonists increased intracellular Ca2+ transients in PDGFRα+ cells and inhibited spontaneous phasic contractions (SPCs) of colonic muscle through activation of a SK conductance. Sympathetic nerve stimulation inhibited both contractions of distal colon and propulsive contractions represented by the colonic migrating motor complexes (CMMCs) via the α1A AR-SK channel signal pathway. Postsynaptic signaling through α1A ARs in PDGFRα+ cells is a novel mechanism that conveys part of stress responses in the colon. PDGFRα+ cells appear to be a primary effector of sympathetic neural regulation of murine colonic motility.


Asunto(s)
Colon/fisiología , Músculo Liso/fisiología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptores Adrenérgicos alfa 1/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Sistema Nervioso Simpático/fisiología , Potenciales Sinápticos , Adenosina Trifosfato , Animales , Calcio/metabolismo , Colon/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso/citología , Transducción de Señal , Sistema Nervioso Simpático/citología
12.
Sci Rep ; 10(1): 1240, 2020 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-31988377

RESUMEN

Facing on the complex electromagnetic environment of electrical equipment, based on the bio-anti-interference characteristics of neuron system, the bio-inspired electromagnetic protection is proposed in order to improve and assist the traditional electromagnetic protection method. In order to analyze the dynamical characteristics of electrical signal transfer process of neuron system, Hodgkin-Huxley (HH) model is adopted to calculate the action potential of single neuron. The initial value problem used in the parameters of Hodgkin-Huxley model is studied in order to satisfy the physiological phenomenon. The stability of HH model is analyzed to assess the dynamic stable performance of neuron. Based on the investigation of single neuron, a simple neuron system consisted of two neurons and one synapse is studied. The compassion between the action potential of posterior neuron and different synapse is performed, which explores how the mathematic models of different synapses influence the action potential. The relationship between action potential of posterior neuron and coupling strength of simplified synapse is calculated to explain the diversity of electrical signal output of neuron system. These numerical results enable to provide some datum for deeply developing the bio-inspired electromagnetic protection and well designing the bio-inspired circuit.


Asunto(s)
Sinapsis Eléctricas/fisiología , Campos Electromagnéticos/efectos adversos , Potenciales de Acción , Simulación por Computador , Radiación Electromagnética , Modelos Neurológicos , Modelos Estadísticos , Red Nerviosa/fisiología , Neuronas/fisiología , Potenciales Sinápticos/fisiología
13.
Front Neuroendocrinol ; 56: 100818, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31843506

RESUMEN

The consolidation of long-term memory is influenced by various neuromodulators. One of these is estradiol, a steroid hormone that is synthesized both in peripheral endocrine tissue and in the brain, including the hippocampus. Here, we examine the evidence regarding the role of estradiol in the hippocampus, specifically, in memory formation and its effects on the molecular mechanisms underlying synaptic plasticity. We conclude that estradiol improves memory consolidation and, thereby, long-term memory. Previous studies have shown that it does this in three, interconnected ways: (1) via functional changes in excitatory activity, (2) signaling changes in calcium dynamics, protein phosphorylation and protein expression, and (3) structural changes to synaptic morphology. Through a functional network analysis of proteins affected by estradiol, we identify potential protein-protein interactions that further support a role for estradiol in modulating synaptic plasticity as well as highlight signaling pathways that may be involved in these changes within the hippocampus.


Asunto(s)
Estradiol/fisiología , Hipocampo/fisiología , Memoria/fisiología , Plasticidad Neuronal/fisiología , Adulto , Animales , Estradiol/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Hipocampo/efectos de los fármacos , Humanos , Masculino , Ratones , Proteínas del Tejido Nervioso/metabolismo , Fosforilación/fisiología , Ratas , Receptores de Estrógenos/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Potenciales Sinápticos/efectos de los fármacos , Potenciales Sinápticos/fisiología
14.
Int J Mol Sci ; 20(17)2019 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-31450553

RESUMEN

The correct morphofunctional shaping of the cerebral cortex requires a continuous interaction between intrinsic (genes/molecules expressed within the tissue) and extrinsic (e.g., neural activity) factors at all developmental stages. Forkhead Box G1 (FOXG1) is an evolutionarily conserved transcription factor, essential for the cerebral cortex patterning and layering. FOXG1-related disorders, including the congenital form of Rett syndrome, can be caused by deletions, intragenic mutations or duplications. These genetic alterations are associated with a complex phenotypic spectrum, spanning from intellectual disability, microcephaly, to autistic features, and epilepsy. We investigated the functional correlates of dysregulated gene expression by performing electrophysiological assays on FoxG1+/- mice. Local Field Potential (LFP) recordings on freely moving animals detected cortical hyperexcitability. On the other hand, patch-clamp recordings showed a downregulation of spontaneous glutamatergic transmission. These findings were accompanied by overactivation of Akt/S6 signaling. Furthermore, the expression of vesicular glutamate transporter 2 (vGluT2) was increased, whereas the level of the potassium/chloride cotransporter KCC2 was reduced, thus indicating a higher excitation/inhibition ratio. Our findings provide evidence that altered expression of a key gene for cortical development can result in specific alterations in neural circuit function at the macro- and micro-scale, along with dysregulated intracellular signaling and expression of proteins controlling circuit excitability.


Asunto(s)
Corteza Cerebral/metabolismo , Corteza Cerebral/fisiopatología , Epilepsia/genética , Epilepsia/metabolismo , Factores de Transcripción Forkhead/genética , Proteínas del Tejido Nervioso/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transmisión Sináptica , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Epilepsia/fisiopatología , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Ratones , Ratones Noqueados , Fenotipo , Convulsiones , Transducción de Señal , Potenciales Sinápticos
15.
J Neurosci ; 39(36): 7195-7205, 2019 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-31320448

RESUMEN

Clinical and experimental data indicate striatal cholinergic dysfunction in dystonia, a movement disorder typically resulting in twisted postures via abnormal muscle contraction. Three forms of isolated human dystonia result from mutations in the TOR1A (DYT1), THAP1 (DYT6), and GNAL (DYT25) genes. Experimental models carrying these mutations facilitate identification of possible shared cellular mechanisms. Recently, we reported elevated extracellular striatal acetylcholine by in vivo microdialysis and paradoxical excitation of cholinergic interneurons (ChIs) by dopamine D2 receptor (D2R) agonism using ex vivo slice electrophysiology in Dyt1ΔGAG/+ mice. The paradoxical excitation was caused by overactive muscarinic receptors (mAChRs), leading to a switch in D2R coupling from canonical Gi/o to noncanonical ß-arrestin signaling. We sought to determine whether these mechanisms in Dyt1ΔGAG/+ mice are shared with Thap1C54Y/+ knock-in and Gnal+/- knock-out dystonia models and to determine the impact of sex. We found Thap1C54Y/+ mice of both sexes have elevated extracellular striatal acetylcholine and D2R-induced paradoxical ChI excitation, which was reversed by mAChR inhibition. Elevated extracellular acetylcholine was absent in male and female Gnal+/- mice, but the paradoxical D2R-mediated ChI excitation was retained and only reversed by inhibition of adenosine A2ARs. The Gi/o-preferring D2R agonist failed to increase ChI excitability, suggesting a possible switch in coupling of D2Rs to ß-arrestin, as seen previously in a DYT1 model. These data show that, whereas elevated extracellular acetylcholine levels are not always detected across these genetic models of human dystonia, the D2R-mediated paradoxical excitation of ChIs is shared and is caused by altered function of distinct G-protein-coupled receptors.SIGNIFICANCE STATEMENT Dystonia is a common and often disabling movement disorder. The usual medical treatment of dystonia is pharmacotherapy with nonselective antagonists of muscarinic acetylcholine receptors, which have many undesirable side effects. Development of new therapeutics is a top priority for dystonia research. The current findings, considered in context with our previous investigations, establish a role for cholinergic dysfunction across three mouse models of human genetic dystonia: DYT1, DYT6, and DYT25. The commonality of cholinergic dysfunction in these models arising from diverse molecular etiologies points the way to new approaches for cholinergic modulation that may be broadly applicable in dystonia.


Asunto(s)
Neuronas Colinérgicas/metabolismo , Cuerpo Estriado/metabolismo , Proteínas de Unión al ADN/genética , Distonía/genética , Glucosamina 6-Fosfato N-Acetiltransferasa/genética , Chaperonas Moleculares/genética , Acetilcolina/metabolismo , Animales , Neuronas Colinérgicas/fisiología , Cuerpo Estriado/fisiopatología , Distonía/metabolismo , Distonía/fisiopatología , Espacio Extracelular/metabolismo , Femenino , Interneuronas/metabolismo , Interneuronas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Dopamina D2/metabolismo , Receptores Muscarínicos/metabolismo , Potenciales Sinápticos , beta-Arrestinas/metabolismo
16.
Sci Rep ; 9(1): 9571, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31267026

RESUMEN

Adenosine A1 receptors (A1AR) are well characterized for their role in cytoprotection. Previous studies have demonstrated the presence of these receptors in the cochlea where their activation were shown to suppress cisplatin-induced inflammatory response and the resulting ototoxicity. Inhibition of A1AR by caffeine, a widely consumed psychoactive substance, could antagonize the endogenous protective role of these receptors in cochlea and potentiate cisplatin-induced hearing loss. This hypothesis was tested in a rat model of cisplatin ototoxicity following oral administration of caffeine. We report here that single-dose administration of caffeine exacerbates cisplatin-induced hearing loss without increasing the damage to outer hair cells (OHCs), but increased synaptopathy and inflammation in the cochlea. These effects of caffeine were mediated by its blockade of A1AR, as co-administration of R-PIA, an A1AR agonist, reversed the detrimental actions of caffeine and cisplatin on hearing loss. Multiple doses of caffeine exacerbated cisplatin ototoxicity which was associated with damage to OHCs and cochlear synaptopathy. These findings highlight a possible drug-drug interaction between caffeine and cisplatin for ototoxicity and suggest that caffeine consumption should be cautioned in cancer patients treated with a chemotherapeutic regimen containing cisplatin.


Asunto(s)
Antineoplásicos/efectos adversos , Cafeína/administración & dosificación , Estimulantes del Sistema Nervioso Central/administración & dosificación , Cisplatino/efectos adversos , Pérdida Auditiva/etiología , Administración Oral , Animales , Apoptosis/efectos de los fármacos , Biomarcadores , Cafeína/efectos adversos , Estimulantes del Sistema Nervioso Central/efectos adversos , Cóclea/efectos de los fármacos , Sinergismo Farmacológico , Técnica del Anticuerpo Fluorescente , Pérdida Auditiva/metabolismo , Pérdida Auditiva/patología , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Ratas , Potenciales Sinápticos/efectos de los fármacos
17.
Mol Cell Neurosci ; 96: 25-34, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30858140

RESUMEN

Synapsins are neuronal phosphoproteins that fine-tune synaptic transmission and suppress seizure activity. Synapsin II (SynII) deletion produces epileptic seizures and overexcitability in neuronal networks. Early studies in primary neuronal cultures have shown that SynII deletion results in a delay in synapse formation. More recent studies at hippocampal slices have revealed increased spontaneous activity in SynII knockout (SynII(-)) mice. To reconcile these observations, we systematically re-examined synaptic transmission, synapse formation, and neurite growth in primary hippocampal neuronal cultures. We find that spontaneous glutamatergic synaptic activity was suppressed in SynII(-) neurons during the initial developmental epoch (7 days in vitro, DIV) but was enhanced at later times (12 and18 DIV). The density of synapses, transmission between connected pairs of neurons, and the number of docked synaptic vesicles were not affected by SynII deletion. However, we found that neurite outgrowth in SynII(-) neurons was suppressed during the initial developmental epoch (7 DIV) but enhanced at subsequent developmental stages (12 and18 DIV). This finding can account for the observed effect of SynII deletion on synaptic activity. To test whether the observed phenotype resulted directly from the deletion of SynII we expressed SynII in SynII(-) cultures using an adeno-associated virus (AAV). Expression of SynII at 2 DIV rescued the SynII deletion-dependent alterations in both synaptic activity and neuronal growth. To test whether the increased neurite outgrowth in SynII(-) observed at DIV 12 and18 represents an overcompensation for the initial developmental delay or results directly from SynII deletion we performed "late expression" experiments, transfecting SynII(-) cultures with AAV at 7 DIV. The late SynII expression suppressed neurite outgrowth at 12 and 18 DIV to the levels observed in control neurons, suggesting that these phenotypes directly depend on SynII. These results reveal a novel developmentally regulated role for SynII function in the control of neurite growth.


Asunto(s)
Hipocampo/metabolismo , Proyección Neuronal , Neuronas/metabolismo , Sinapsinas/genética , Potenciales Sinápticos , Animales , Células Cultivadas , Ácido Glutámico/metabolismo , Hipocampo/citología , Hipocampo/fisiología , Ratones , Neuronas/citología , Neuronas/fisiología , Sinapsinas/deficiencia
18.
Elife ; 82019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30657452

RESUMEN

It is often assumed that highly-branched neuronal structures perform compartmentalized computations. However, previously we showed that the Gastric Mill (GM) neuron in the crustacean stomatogastric ganglion (STG) operates like a single electrotonic compartment, despite having thousands of branch points and total cable length >10 mm (Otopalik et al., 2017a; 2017b). Here we show that compact electrotonic architecture is generalizable to other STG neuron types, and that these neurons present direction-insensitive, linear voltage integration, suggesting they pool synaptic inputs across their neuronal structures. We also show, using simulations of 720 cable models spanning a broad range of geometries and passive properties, that compact electrotonus, linear integration, and directional insensitivity in STG neurons arise from their neurite geometries (diameters tapering from 10-20 µm to < 2 µm at their terminal tips). A broad parameter search reveals multiple morphological and biophysical solutions for achieving different degrees of passive electrotonic decrement and computational strategies in the absence of active properties.


Asunto(s)
Potenciales de Acción/fisiología , Braquiuros/fisiología , Molleja No Aviar/fisiología , Red Nerviosa/fisiología , Neuronas/fisiología , Animales , Braquiuros/citología , Ganglios de Invertebrados/citología , Ganglios de Invertebrados/fisiología , Molleja No Aviar/citología , Modelos Neurológicos , Neuritas/fisiología , Potenciales Sinápticos/fisiología
19.
Glia ; 67(5): 915-934, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30632636

RESUMEN

Optogenetics has been widely expanded to enhance or suppress neuronal activity and it has been recently applied to glial cells. Here, we have used a new approach based on selective expression of melanopsin, a G-protein-coupled photopigment, in astrocytes to trigger Ca2+ signaling. Using the genetically encoded Ca2+ indicator GCaMP6f and two-photon imaging, we show that melanopsin is both competent to stimulate robust IP3-dependent Ca2+ signals in astrocyte fine processes, and to evoke an ATP/Adenosine-dependent transient boost of hippocampal excitatory synaptic transmission. Additionally, under low-frequency light stimulation conditions, melanopsin-transfected astrocytes can trigger long-term synaptic changes. In vivo, melanopsin-astrocyte activation enhances episodic-like memory, suggesting melanopsin as an optical tool that could recapitulate the wide range of regulatory actions of astrocytes on neuronal networks in behaving animals. These results describe a novel approach using melanopsin as a precise trigger for astrocytes that mimics their endogenous G-protein signaling pathways, and present melanopsin as a valuable optical tool for neuron-glia studies.


Asunto(s)
Astrocitos/metabolismo , Red Nerviosa/metabolismo , Neuronas/metabolismo , Optogenética/métodos , Opsinas de Bastones/metabolismo , 2-Amino-5-fosfonovalerato/farmacología , Antagonistas del Receptor de Adenosina A2/farmacología , Alanina/análogos & derivados , Alanina/farmacología , Animales , Compuestos Azo/farmacología , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Quelantes/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Hipocampo/citología , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Luz , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacología , Pirimidinas/farmacología , Opsinas de Bastones/genética , Potenciales Sinápticos/fisiología , Triazoles/farmacología , Xantenos/farmacología
20.
Respir Physiol Neurobiol ; 265: 19-23, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30639504

RESUMEN

Acute intermittent hypoxia (AIH) elicits distinct mechanisms of phrenic motor plasticity initiated by brainstem neural network activation versus local (spinal) tissue hypoxia. With moderate AIH (mAIH), hypoxemia activates the carotid body chemoreceptors and (subsequently) brainstem neural networks associated with the peripheral chemoreflex, including medullary raphe serotonergic neurons. Serotonin release and receptor activation in the phrenic motor nucleus then elicits phrenic long-term facilitation (pLTF). This mechanism is independent of tissue hypoxia, since electrical carotid sinus nerve stimulation elicits similar serotonin-dependent pLTF. In striking contrast, severe AIH (sAIH) evokes a spinal adenosine-dependent, serotonin-independent mechanism of pLTF. Spinal tissue hypoxia per se is the likely cause of sAIH-induced pLTF, since local tissue hypoxia elicits extracellular adenosine accumulation. Thus, any physiological condition exacerbating spinal tissue hypoxia is expected to shift the balance towards adenosinergic pLTF. However, since these mechanisms compete for dominance due to mutual cross-talk inhibition, the transition from serotonin to adenosine dominant pLTF is rather abrupt. Any factor that compromises spinal cord circulation will limit oxygen availability in spinal cord tissue, favoring a shift in the balance towards adenosinergic mechanisms. Such shifts may arise experimentally from treatments such as carotid denervation, or spontaneous hypotension or anemia. Many neurological disorders, such as spinal cord injury or stroke compromise local circulatory control, potentially modulating tissue oxygen, adenosine levels and, thus, phrenic motor plasticity. In this brief review, we discuss the concept that local (spinal) circulatory control and/or oxygen delivery regulates the relative contributions of distinct pathways to phrenic motor plasticity.


Asunto(s)
Adenosina/metabolismo , Médula Cervical/fisiología , Hipoxia , Plasticidad Neuronal/fisiología , Oxígeno/metabolismo , Nervio Frénico/fisiología , Fenómenos Fisiológicos Respiratorios , Serotonina/metabolismo , Potenciales Sinápticos/fisiología , Animales , Humanos , Hipoxia/metabolismo , Hipoxia/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA