Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Psychiatry ; 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38486049

RESUMEN

Combinatorial expression of postsynaptic proteins underlies synapse diversity within and between neuron types. Thus, characterization of neuron-type-specific postsynaptic proteomes is key to obtaining a deeper understanding of discrete synaptic properties and how selective dysfunction manifests in synaptopathies. To overcome the limitations associated with bulk measures of synaptic protein abundance, we developed a biotin proximity protein tagging probe to characterize neuron-type-specific postsynaptic proteomes in vivo. We found Shank3 protein isoforms are differentially expressed by direct and indirect pathway spiny projection neurons (dSPNs and iSPNs). Investigation of Shank3B-/- mice lacking exons 13-16 within the Shank3 gene, reveal distinct Shank3 protein isoform expression in iSPNs and dSPNs. In Shank3B-/- striatum, Shank3E and Shank3NT are expressed by dSPNs but are undetectable in iSPNs. Proteomic analysis indicates significant and selective alterations in the postsynaptic proteome of Shank3B-/- iSPNs. Correspondingly, the deletion of exons 13-16 diminishes dendritic spine density, reduces spine head diameter, and hampers corticostriatal synaptic transmission in iSPNs. Remarkably, reintroducing Shank3E in adult Shank3B-/- iSPNs significantly rectifies the observed dendritic spine morphological and corticostriatal synaptic transmission deficits. We report unexpected cell-type specific synaptic protein isoform expression which could play a key causal role in specifying synapse diversity and selective synapse dysfunction in synaptopathies.

4.
ACS Chem Biol ; 15(9): 2539-2550, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32881483

RESUMEN

Ca2+ channels with a CaV1.3 pore-forming α1 subunit have been implicated in both neurodegenerative and neuropsychiatric disorders, motivating the development of selective and potent inhibitors of CaV1.3 versus CaV1.2 channels, the calcium channels implicated in hypertensive disorders. We have previously identified pyrimidine-2,4,6-triones (PYTs) that preferentially inhibit CaV1.3 channels, but the structural determinants of their interaction with the channel have not been identified, impeding their development into drugs. By a combination of biochemical, computational, and molecular biological approaches, it was found that PYTs bind to the dihydropyridine (DHP) binding pocket of the CaV1.3 subunit, establishing them as negative allosteric modulators of channel gating. Site-directed mutagenesis, based on homology models of CaV1.3 and CaV1.2 channels, revealed that a single amino acid residue within the DHP binding pocket (M1078) is responsible for the selectivity of PYTs for CaV1.3 over CaV1.2. In addition to providing direction for chemical optimization, these results suggest that, like dihydropyridines, PYTs have pharmacological features that could make them of broad clinical utility.


Asunto(s)
Bloqueadores de los Canales de Calcio/metabolismo , Canales de Calcio Tipo L/metabolismo , Pirimidinonas/metabolismo , Regulación Alostérica , Sitio Alostérico , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/genética , Neuronas Dopaminérgicas/efectos de los fármacos , Células HEK293 , Humanos , Ratones , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Mutación , Unión Proteica , Conejos , Ratas
5.
Cell Stem Cell ; 27(4): 633-645.e7, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32846140

RESUMEN

The G2019S mutation in leucine-rich repeat kinase 2 (LRRK2) is a common cause of familial Parkinson's disease (PD). This mutation results in dopaminergic neurodegeneration via dysregulated protein translation, although how alterations in protein synthesis contribute to neurodegeneration in human neurons is not known. Here we define the translational landscape in LRRK2-mutant dopaminergic neurons derived from human induced pluripotent stem cells (hiPSCs) via ribosome profiling. We found that mRNAs that have complex secondary structure in the 5' untranslated region (UTR) are translated more efficiently in G2019S LRRK2 neurons. This leads to the enhanced translation of multiple genes involved in Ca2+ regulation and to increased Ca2+ influx and elevated intracellular Ca2+ levels, a major contributor to PD pathogenesis. This study reveals a link between dysregulated translation control and Ca2+ homeostasis in G2019S LRRK2 human dopamine neurons, which potentially contributes to the progressive and selective dopaminergic neurotoxicity in PD.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad de Parkinson , Calcio , Neuronas Dopaminérgicas/metabolismo , Homeostasis , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Mutación/genética , Enfermedad de Parkinson/genética , Biosíntesis de Proteínas
6.
J Neurosci ; 39(29): 5760-5772, 2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31097622

RESUMEN

The accumulation of misfolded proteins is a common pathological feature of many neurodegenerative disorders, including synucleinopathies such as Parkinson's disease (PD), which is characterized by the presence of α-synuclein (α-syn)-containing Lewy bodies. However, although recent studies have investigated α-syn accumulation and propagation in neurons, the molecular mechanisms underlying α-syn transmission have been largely unexplored. Here, we examined a monogenic form of synucleinopathy caused by loss-of-function mutations in lysosomal ATP13A2/PARK9. These studies revealed that lysosomal exocytosis regulates intracellular levels of α-syn in human neurons. Loss of PARK9 function in patient-derived dopaminergic neurons disrupted lysosomal Ca2+ homeostasis, reduced lysosomal Ca2+ storage, increased cytosolic Ca2+, and impaired lysosomal exocytosis. Importantly, this dysfunction in lysosomal exocytosis impaired α-syn secretion from both axons and soma, promoting α-syn accumulation. However, activation of the lysosomal Ca2+ channel transient receptor potential mucolipin 1 (TRPML1) was sufficient to upregulate lysosomal exocytosis, rescue defective α-syn secretion, and prevent α-syn accumulation. Together, these results suggest that intracellular α-syn levels are regulated by lysosomal exocytosis in human dopaminergic neurons and may represent a potential therapeutic target for PD and other synucleinopathies.SIGNIFICANCE STATEMENT Parkinson's disease (PD) is the second most common neurodegenerative disease linked to the accumulation of α-synuclein (α-syn) in patient neurons. However, it is unclear what the mechanism might be. Here, we demonstrate a novel role for lysosomal exocytosis in clearing intracellular α-syn and show that impairment of this pathway by mutations in the PD-linked gene ATP13A2/PARK9 contributes to α-syn accumulation in human dopaminergic neurons. Importantly, upregulating lysosomal exocytosis by increasing lysosomal Ca2+ levels was sufficient to rescue defective α-syn secretion and accumulation in patient neurons. These studies identify lysosomal exocytosis as a potential therapeutic target in diseases characterized by the accumulation of α-syn, including PD.


Asunto(s)
Agonistas de los Canales de Calcio/farmacología , Neuronas Dopaminérgicas/metabolismo , Exocitosis/fisiología , Células Madre Pluripotentes Inducidas/metabolismo , Lisosomas/metabolismo , alfa-Sinucleína/toxicidad , Línea Celular Tumoral , Células Cultivadas , Neuronas Dopaminérgicas/efectos de los fármacos , Exocitosis/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Lisosomas/efectos de los fármacos , Lisosomas/genética , ATPasas de Translocación de Protón/genética , ATPasas de Translocación de Protón/metabolismo
7.
Mov Disord ; 34(5): 684-696, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30726572

RESUMEN

BACKGROUND: Huntington's disease (HD) is caused by a CAG repeat expansion in the huntingtin gene. This mutation leads to progressive dysfunction that is largely attributable to dysfunction of the striatum. The earliest signs of striatal pathology in HD are found in indirect pathway gamma-Aminobutyric acid (GABA)-ergic spiny projection neurons that innervate the external segment of the globus pallidus (GPe). What is less clear is whether the synaptic coupling of spiny projection neurons with GPe neurons changes in HD. OBJECTIVES: The principal goal of this study was to determine whether striatopallidal synaptic transmission was altered in 2 mouse models of HD. METHODS: Striatopallidal synaptic transmission was studied using electrophysiological and optogenetic approaches in ex vivo brain slices from 2 HD models: Q175 heterozygous (het) and R6/2 mice. RESULTS: Striatopallidal synaptic transmission increased in strength with the progression of behavioral deficits in Q175 and R6/2 mice. The alteration in synaptic transmission was evident in both prototypical and arkypallidal GPe neurons. This change did not appear attributable to an increase in the probability of GABA release but, rather, to an enhancement in the postsynaptic response to GABA released at synaptic sites. This alteration significantly increased the ability of striatopallidal axon terminals to pause ongoing GPe activity. CONCLUSIONS: In 2 mouse models of HD, striatopallidal synaptic transmission increased in parallel with the progression of behavioral deficits. This adaptation could compensate in part for the concomitant deficit in the ability of corticostriatal signals to activate spiny projection neurons and pause GPe activity. © 2019 International Parkinson and Movement Disorder Society.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Globo Pálido/metabolismo , Enfermedad de Huntington/metabolismo , Potenciales Postsinápticos Inhibidores/fisiología , Neostriado/metabolismo , Animales , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Estimulación Eléctrica , Fenómenos Electrofisiológicos , Técnicas de Sustitución del Gen , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Ratones , Vías Nerviosas/metabolismo , Neuronas/metabolismo , Optogenética , Técnicas de Placa-Clamp , Transmisión Sináptica/fisiología , Ácido gamma-Aminobutírico/metabolismo
8.
Stem Cell Reports ; 9(1): 149-161, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28579395

RESUMEN

A major challenge for clinical application of pluripotent stem cell therapy for Parkinson's disease (PD) is large-scale manufacturing and cryopreservation of neurons that can be efficiently prepared with minimal manipulation. To address this obstacle, midbrain dopamine neurons were derived from human induced pluripotent stem cells (iPSC-mDA) and cryopreserved in large production lots for biochemical and transplantation studies. Cryopreserved, post-mitotic iPSC-mDA neurons retained high viability with gene, protein, and electrophysiological signatures consistent with midbrain floor-plate lineage. To test therapeutic efficacy, cryopreserved iPSC-mDA neurons were transplanted without subculturing into the 6-OHDA-lesioned rat and MPTP-lesioned non-human-primate models of PD. Grafted neurons retained midbrain lineage with extensive fiber innervation in both rodents and monkeys. Behavioral assessment in 6-OHDA-lesioned rats demonstrated significant reversal in functional deficits up to 6 months post transplantation with reinnervation of the host striatum and no aberrant growth, supporting the translational development of pluripotent cell-based therapies in PD.


Asunto(s)
Criopreservación , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/trasplante , Células Madre Pluripotentes Inducidas/citología , Enfermedad de Parkinson/terapia , Animales , Línea Celular , Cuerpo Estriado/citología , Cuerpo Estriado/patología , Criopreservación/métodos , Modelos Animales de Enfermedad , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Haplorrinos , Humanos , Mesencéfalo/citología , Mesencéfalo/patología , Neurogénesis , Enfermedad de Parkinson/patología , Ratas , Ratas Sprague-Dawley
9.
Neurobiol Dis ; 81: 14-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25796568

RESUMEN

In many synapses of the CNS, mobile zinc is packaged into glutamatergic vesicles and co-released with glutamate during neurotransmission. Following synaptic release, the mobilized zinc modulates ligand- and voltage-gated channels and receptors, functioning as an inhibitory neuromodulator. However, the origin and role of tonic, as opposed to phasically released, zinc are less well understood. We investigated tonic zinc in the dorsal cochlear nucleus (DCN), a zinc-rich, auditory brainstem nucleus. Our results show that application of a high-affinity, extracellular zinc chelator (ZX1) enhances spontaneous firing in DCN principal neurons (fusiform cells), consistent with inhibition of this neuronal property by tonic zinc. The enhancing effect was prevented by prior application of strychnine, a glycine receptor antagonist, suggesting that ZX1 interferes with zinc-mediated modulation of spontaneous glycinergic inhibition. In particular, ZX1 decreased the amplitude and the frequency of glycinergic miniature inhibitory postsynaptic currents in fusiform cells, from which we conclude that tonic zinc enhances glycinergic inhibitory neurotransmission. The observed zinc-mediated inhibition in spontaneous firing is present in mice lacking the vesicular zinc transporter (ZnT3), indicating that non-vesicular zinc inhibits spontaneous firing. Noise-induced increase in the spontaneous firing of fusiform cells is crucial for the induction of tinnitus. In this context, tonic zinc provides a powerful break of spontaneous firing that may protect against pathological run-up of spontaneous activity in the DCN.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Núcleo Coclear/citología , Glicina/metabolismo , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Neuronas/efectos de los fármacos , Zinc/farmacología , Potenciales de Acción/genética , Animales , Animales Recién Nacidos , Proteínas Portadoras/genética , Proteínas de Transporte de Catión , Quelantes/farmacología , Interacciones Farmacológicas , Glicinérgicos/farmacología , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Técnicas de Placa-Clamp , Piridinas/farmacología , Estricnina/farmacología , Ácidos Sulfanílicos/farmacología
10.
J Neurosci ; 33(22): 9259-72, 2013 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-23719795

RESUMEN

Although it is well established that many glutamatergic neurons sequester Zn(2+) within their synaptic vesicles, the physiological significance of synaptic Zn(2+) remains poorly understood. In experiments performed in a Zn(2+)-enriched auditory brainstem nucleus--the dorsal cochlear nucleus--we discovered that synaptic Zn(2+) and GPR39, a putative metabotropic Zn(2+)-sensing receptor (mZnR), are necessary for triggering the synthesis of the endocannabinoid 2-arachidonoylglycerol (2-AG). The postsynaptic production of 2-AG, in turn, inhibits presynaptic probability of neurotransmitter release, thus shaping synaptic strength and short-term synaptic plasticity. Zn(2+)-induced inhibition of transmitter release is absent in mutant mice that lack either vesicular Zn(2+) or the mZnR. Moreover, mass spectrometry measurements of 2-AG levels reveal that Zn(2+)-mediated initiation of 2-AG synthesis is absent in mice lacking the mZnR. We reveal a previously unknown action of synaptic Zn(2+): synaptic Zn(2+) inhibits glutamate release by promoting 2-AG synthesis.


Asunto(s)
Endocannabinoides/biosíntesis , Neurotransmisores/metabolismo , Sinapsis/fisiología , Zinc/fisiología , Animales , Ácidos Araquidónicos/metabolismo , Cromatografía Liquida , Dendritas/fisiología , Endocannabinoides/metabolismo , Femenino , Ácido Glutámico/metabolismo , Glicéridos/metabolismo , Masculino , Espectrometría de Masas , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Microscopía Fluorescente , Fibras Nerviosas/fisiología , Técnicas de Placa-Clamp , Transducción de Señal/fisiología , Transmisión Sináptica/fisiología
11.
J Comput Neurosci ; 31(2): 159-82, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21207127

RESUMEN

Although associational/commissural (A/C) and perforant path (PP) inputs to CA3b pyramidal cells play a central role in hippocampal mnemonic functions, the active and passive processes that shape A/C and PP AMPA and NMDA receptor-mediated unitary EPSP/EPSC (AMPA and NMDA uEPSP/uEPSC) have not been fully characterized yet. Here we find no differences in somatic amplitude between A/C and PP for either AMPA or NMDA uEPSPs. However, larger AMPA uEPSCs were evoked from proximal than from distal A/C or PP. Given the space-clamp constraints in CA3 pyramidal cells, these voltage clamp data suggest that the location-independence of A/C and PP AMPA uEPSP amplitudes is achieved in part through the activation of voltage dependent conductances at or near the soma. Moreover, similarity in uEPSC amplitudes for distal A/C and PP points to the additional participation of unclamped active conductances. Indeed, the pharmacological blockade of voltage-dependent conductances eliminates the location-independence of these inputs. In contrast, the location-independence of A/C and PP NMDA uEPSP/uEPSC amplitudes is maintained across all conditions indicating that propagation is not affected by active membrane processes. The location-independence for A/C uEPSP amplitudes may be relevant in the recruitment of CA3 pyramidal cells by other CA3 pyramidal cells. These data also suggest that PP excitation represents a significant input to CA3 pyramidal cells. Implication of the passive data on local synaptic properties is further investigated in the companion paper with a detailed computational model.


Asunto(s)
Región CA3 Hipocampal/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Vía Perforante/fisiología , Células Piramidales/fisiología , Sinapsis/fisiología , Animales , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Masculino , Técnicas de Cultivo de Órganos , Células Piramidales/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Sinapsis/efectos de los fármacos
12.
J Comput Neurosci ; 31(1): 137-58, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21191641

RESUMEN

Despite the central position of CA3 pyramidal cells in the hippocampal circuit, the experimental investigation of their synaptic properties has been limited. Recent slice experiments from adult rats characterized AMPA and NMDA receptor unitary synaptic responses in CA3b pyramidal cells. Here, excitatory synaptic activation is modeled to infer biophysical parameters, aid analysis interpretation, explore mechanisms, and formulate predictions by contrasting simulated somatic recordings with experimental data. Reconstructed CA3b pyramidal cells from the public repository NeuroMorpho.Org were used to allow for cell-specific morphological variation. For each cell, synaptic responses were simulated for perforant pathway and associational/commissural synapses. Means and variability for peak amplitude, time-to-peak, and half-height width in these responses were compared with equivalent statistics from experimental recordings. Synaptic responses mediated by AMPA receptors are best fit with properties typical of previously characterized glutamatergic receptors where perforant path synapses have conductances twice that of associational/commissural synapses (0.9 vs. 0.5 nS) and more rapid peak times (1.0 vs. 3.3 ms). Reanalysis of passive-cell experimental traces using the model shows no evidence of a CA1-like increase of associational/commissural AMPA receptor conductance with increasing distance from the soma. Synaptic responses mediated by NMDA receptors are best fit with rapid kinetics, suggestive of NR2A subunits as expected in mature animals. Predictions were made for passive-cell current clamp recordings, combined AMPA and NMDA receptor responses, and local dendritic depolarization in response to unitary stimulations. Models of synaptic responses in active cells suggest altered axial resistivity and the presence of synaptically activated potassium channels in spines.


Asunto(s)
Región CA3 Hipocampal/fisiología , Células Piramidales/fisiología , Sinapsis/fisiología , Animales , Simulación por Computador , Modelos Neurológicos , Ratas , Receptores AMPA/fisiología , Receptores de N-Metil-D-Aspartato
13.
J Neurophysiol ; 94(6): 3771-87, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16120665

RESUMEN

The modulatory effect of D(2) dopamine receptor activation on calcium currents was studied in neostriatal projection neurons at two stages of rat development: postnatal day (PD)14 and PD40. D(2)-class receptor agonists reduced whole cell calcium currents by about 35% at both stages, and this effect was blocked by the D(2) receptor antagonist sulpiride. Nitrendipine partially occluded this modulation at both stages, indicating that modulation of Ca(V)1 channels was present throughout this developmental interval. Nevertheless, modulation of Ca(V)1 channels was significantly larger in PD40 neurons. omega-Conotoxin GVIA occluded most of the Ca(2+) current modulation in PD14 neurons. However, this occlusion was greatly decreased in PD40 neurons. omega-Agatoxin TK occluded a great part of the modulation in PD40 neurons but had a negligible effect in PD14 neurons. The data indicate that dopaminergic D(2)-mediated modulation undergoes a change in target during development: from Ca(V)2.2 to Ca(V)2.1 Ca(2+) channels. This change occurred while Ca(V)2.2 channels were being down-regulated and Ca(V)2.1 channels were being up-regulated. Presynaptic modulation mediated by D(2) receptors reflected these changes; Ca(V)2.2 type channels were used for release in young animals but very little in mature animals, suggesting that changes took place simultaneously at the somatodendritic and the synaptic membranes.


Asunto(s)
Caveolina 2/fisiología , Neostriado/citología , Neuronas/fisiología , Receptores de Dopamina D2/fisiología , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Quelantes/farmacología , Agonistas de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Estimulación Eléctrica/métodos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Potenciales de la Membrana/efectos de la radiación , Neostriado/crecimiento & desarrollo , Técnicas de Placa-Clamp/métodos , Quinolinas/farmacología , Ratas , Ratas Wistar , Factores de Tiempo
14.
J Neurophysiol ; 93(5): 2507-19, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15615835

RESUMEN

Besides a reduction of L-type Ca2+-currents (Ca(V)1), muscarine and the peptidic M1-selective agonist, MT-1, reduced currents through Ca(V)2.1 (P/Q) and Ca(V)2.2 (N) Ca2+ channel types. This modulation was strongly blocked by the peptide MT-7, a specific muscarinic M1-type receptor antagonist but not significantly reduced by the peptide MT-3, a specific muscarinic M4-type receptor antagonist. Accordingly, MT-7, but not MT-3, blocked a muscarinic reduction of the afterhyperpolarizing potential (AHP) and decreased the GABAergic inhibitory postsynaptic currents (IPSCs) produced by axon collaterals that interconnect spiny neurons. Both these functions are known to be dependent on P/Q and N types Ca2+ channels. The action on the AHP had an important effect in increasing firing frequency. The action on the IPSCs was shown to be caused presynaptically as it coursed with an increase in the paired-pulse ratio. These results show: first, that muscarinic M1-type receptor activation is the main cholinergic mechanism that modulates Ca2+ entry through voltage-dependent Ca2+ channels in spiny neurons. Second, this muscarinic modulation produces a postsynaptic facilitation of discharge together with a presynaptic inhibition of the GABAergic control mediated by axon collaterals. Together, both effects would tend to recruit more spiny neurons for the same task.


Asunto(s)
Acetilcolina/metabolismo , Caveolinas/fisiología , Neostriado/citología , Neuronas/fisiología , Transmisión Sináptica/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Bloqueadores de los Canales de Calcio/farmacología , Caveolina 2 , Caveolinas/clasificación , Caveolinas/efectos de los fármacos , Células Cultivadas , Interacciones Farmacológicas , Estimulación Eléctrica/métodos , Técnicas In Vitro , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Potenciales de la Membrana/efectos de la radiación , Muscarina/farmacología , Agonistas Muscarínicos/clasificación , Agonistas Muscarínicos/farmacología , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp/métodos , Ratas , Ratas Wistar , Receptores Muscarínicos/clasificación , Receptores Muscarínicos/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA