Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Biol Pharm Bull ; 47(2): 509-517, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38403661

RESUMEN

(-)-Epigallocatechin-3-gallate (EGCg), a major constituent of green tea extract, is well-known to exhibit many beneficial actions for human health by interacting with numerous proteins. In this study we identified synaptic vesicle membrane protein VAT-1 homolog (VAT1) as a novel EGCg-binding protein in human neuroglioma cell extracts using a magnetic pull-down assay and LC-tandem mass spectrometry. We prepared recombinant human VAT1 and analyzed its direct binding to EGCg and its alkylated derivatives using surface plasmon resonance. For EGCg and the derivative NUP-15, we measured an association constant of 0.02-0.85 ×103 M-1s-1 and a dissociation constant of nearly 8 × 10-4 s-1. The affinity Km(affinity) of their binding to VAT1 was in the 10-20 µM range and comparable with that of other EGCg-binding proteins reported previously. Based on the common structure of the compounds, VAT1 appeared to recognize a catechol or pyrogallol moiety around the B-, C- and G-rings of EGCg. Next, we examined whether VAT1 mediates the effects of EGCg and NUP-15 on expression of neprilysin (NEP). Treatments of mock cells with these compounds upregulated NEP, as observed previously, whereas no effect was observed in the VAT1-overexpressing cells, indicating that VAT1 prevented the effects of EGCg or NUP-15 by binding to and inactivating them in the cells overexpressing VAT1. Further investigation is required to determine the biological significance of the VAT1-EGCg interaction.


Asunto(s)
Catequina , Proteínas de Transporte Vesicular , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Vesículas Sinápticas/metabolismo , Té/química , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
2.
Biol Pharm Bull ; 45(9): 1385-1388, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36047209

RESUMEN

Docosahexaenoic acid (DHA; 22:6n-3), which is enriched in the neuronal membrane, plays a variety of roles in the brain. Vesicular glutamate transporters (VGLUTs) are responsible for incorporating glutamine into synaptic vesicles. We investigated the influence of DHA on the fatty acid profile and the levels of VGLUT1 and VGLUT2 proteins in differentiated NG108-15 cells, a neuroblastoma-glioma hybrid cell line. NG108-15 cells were plated and 24 h later the medium was replaced with Dulbecco's modified Eagle's medium supplemented with 1% fetal bovine serum, 0.2 mM dibutyryl cAMP, and 100 nM dexamethasone, which was added to induce differentiation. After 6 d, the amount of DHA in the cells was increased by addition of DHA to the medium. VGLUT2 levels were increased by the addition of DHA. These data indicate that DHA affected the levels of VGLUT2 in NG108-15 cells under differentiation-promoting conditions, suggesting that DHA affects brain functions involving VGLUT2.


Asunto(s)
Ácidos Docosahexaenoicos , Vesículas Sinápticas , Ácidos Docosahexaenoicos/farmacología , Ácido Glutámico/metabolismo , Neuronas/metabolismo , Vesículas Sinápticas/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo
3.
Molecules ; 25(10)2020 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-32422902

RESUMEN

Many neurological disorders are related to synaptic loss or pathologies. Before the boom of positrons emission tomography (PET) imaging of synapses, synaptic quantification could only be achieved in vitro on brain samples after autopsy or surgical resections. Until the mid-2010s, electron microscopy and immunohistochemical labelling of synaptic proteins were the gold-standard methods for such analyses. Over the last decade, several PET radiotracers for the synaptic vesicle 2A protein have been developed to achieve in vivo synapses visualization and quantification. Different strategies were used, namely radiolabelling with either 11C or 18F, preclinical development in rodent and non-human primates, and binding quantification with different kinetic modelling methods. This review provides an overview of these PET tracers and underlines their perspectives and limitations by focusing on radiochemical aspects, as well as preclinical proof-of-concept and the main clinical outcomes described so far.


Asunto(s)
Encéfalo/diagnóstico por imagen , Enfermedades Neurodegenerativas/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Piridinas/farmacocinética , Pirrolidinas/farmacocinética , Pirrolidinonas/farmacocinética , Vesículas Sinápticas/patología , Animales , Encéfalo/metabolismo , Radioisótopos de Carbono , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Radioisótopos de Flúor , Humanos , Macaca mulatta , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Neuronas/patología , Tomografía de Emisión de Positrones/normas , Prueba de Estudio Conceptual , Piridinas/síntesis química , Pirrolidinas/síntesis química , Pirrolidinonas/síntesis química , Radiofármacos/síntesis química , Radiofármacos/farmacocinética , Roedores , Vesículas Sinápticas/metabolismo
4.
PLoS Genet ; 16(2): e1008609, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32097408

RESUMEN

Neuromodulators such as monoamines are often expressed in neurons that also release at least one fast-acting neurotransmitter. The release of a combination of transmitters provides both "classical" and "modulatory" signals that could produce diverse and/or complementary effects in associated circuits. Here, we establish that the majority of Drosophila octopamine (OA) neurons are also glutamatergic and identify the individual contributions of each neurotransmitter on sex-specific behaviors. Males without OA display low levels of aggression and high levels of inter-male courtship. Males deficient for dVGLUT solely in OA-glutamate neurons (OGNs) also exhibit a reduction in aggression, but without a concurrent increase in inter-male courtship. Within OGNs, a portion of VMAT and dVGLUT puncta differ in localization suggesting spatial differences in OA signaling. Our findings establish a previously undetermined role for dVGLUT in OA neurons and suggests that glutamate uncouples aggression from OA-dependent courtship-related behavior. These results indicate that dual neurotransmission can increase the efficacy of individual neurotransmitters while maintaining unique functions within a multi-functional social behavior neuronal network.


Asunto(s)
Agresión , Proteínas de Drosophila/genética , Drosophila melanogaster/fisiología , Neuronas/metabolismo , Transmisión Sináptica/genética , Proteínas de Transporte Vesicular de Glutamato/genética , Animales , Animales Modificados Genéticamente , Conducta Animal , Cortejo , Proteínas de Drosophila/metabolismo , Femenino , Ácido Glutámico/metabolismo , Masculino , Octopamina/metabolismo , Factores Sexuales , Transducción de Señal/genética , Vesículas Sinápticas/metabolismo , Proteínas de Transporte Vesicular de Glutamato/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/metabolismo
5.
Gen Physiol Biophys ; 38(5): 379-388, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31411574

RESUMEN

With the rapidly increasing use of mobile phones and their close-contact usage to the brain, there are some concerns about the possible neuronal effects induced by exposure to excessive electromagnetic radiation. Exposure to a radiofrequency electromagnetic field (RF-EMF) of 835 MHz (4.0 W/kg specific absorption rate (SAR) 5 h/day for 12 weeks) may affect hypothalamic presynaptic neurons in C57BL/6 mice. The number and size of the synaptic vesicles (SVs) in the hypothalamic presynaptic terminals were significantly decreased after RF-EMF exposure. Further, the density (SVs numbers/µm) of docking and fusing SVs in the active zones of the presynaptic terminal membrane was significantly decreased in hypothalamic neurons. The expression levels of synapsin I/II and synaptotagmin 1, two regulators of SV trafficking in neurons, were also significantly decreased in the hypothalamus. In parallel, the expression of calcium channel was significantly decreased. These changes in SVs in the active zones may directly decrease the release of neurotransmitters in hypothalamic presynaptic terminals. Therefore, we further studied the possible changes in hypothalamic function by testing the core body temperature and body weight and performed the buried pellet test. The trafficking of SVs was changed by RF-EMF; however, we could not find any significant phenotypical changes in our experimental condition.


Asunto(s)
Hipotálamo/metabolismo , Hipotálamo/efectos de la radiación , Ondas de Radio , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/efectos de la radiación , Animales , Transporte Biológico/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL
6.
Neurochem Int ; 129: 104507, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31325476

RESUMEN

Heightened activity of glycogen synthase kinase-3ß (GSK-3ß) is linked to the degeneration of dopaminergic neurons in Parkinson's disease (PD). Phytic acid (PA), a naturally occurring compound with potent antioxidant property, has been shown to confer neuroprotection on dopaminergic neurons in PD. However, the underlying mechanism remains unclear. In the present study, MPTP and MPP+ treatments were used to model PD in mice and SH-SY5Y cells, respectively. We observed reduced tissue dopamine, disrupted synaptic vesicle recycling, and defective neurotransmitter exocytosis. Furthermore, expression of GSK-3ß was upregulated while that of ß-catenin was downregulated, concentration of cytosolic calcium was increased, and expressions of two dopamine carriers, dopamine transporter (DAT) and vesicular monoamine transporter 2 (VMAT2) were decreased. PA treatment attenuated the MPTP-induced upregulation of GSK-3ß, increase in cytosolic calcium concentration, decreases in the levels of DAT, VMAT2, tissue dopamine, and synaptic vesicle recycling. Importantly, disturbances in synaptic vesicle recycling are thought to be early events in PD pathology. These findings suggest that PA is a promising therapeutic agent to treat early events in PD.


Asunto(s)
Antiparkinsonianos/uso terapéutico , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Intoxicación por MPTP/tratamiento farmacológico , Ácido Fítico/uso terapéutico , Vesículas Sinápticas/efectos de los fármacos , Animales , Antiparkinsonianos/farmacología , Calcio/metabolismo , Línea Celular Tumoral , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/biosíntesis , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Inducción Enzimática/efectos de los fármacos , Exocitosis/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/biosíntesis , Glucógeno Sintasa Quinasa 3 beta/genética , Humanos , Intoxicación por MPTP/metabolismo , Ratones Endogámicos C57BL , Neuroblastoma/patología , Ácido Fítico/farmacología , Prueba de Desempeño de Rotación con Aceleración Constante , Vesículas Sinápticas/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/biosíntesis , Proteínas de Transporte Vesicular de Monoaminas/genética , Vía de Señalización Wnt/efectos de los fármacos
7.
J Neurochem ; 150(3): 264-281, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31148170

RESUMEN

Leucine-rich repeat kinase 2 (LRRK2) is a large multidomain scaffolding protein with kinase and GTPase activities involved in synaptic vesicle (SV) dynamics. While its role in Parkinson's disease has been largely investigated, little is known about LRRK2 physiological role and until now few proteins have been described as substrates. We have previously demonstrated that LRRK2 through its WD40 domain interacts with synapsin I, an important SV-associated phosphoprotein involved in neuronal development and in the regulation of neurotransmitter release. To test whether synapsin I is substrate for LRRK2 and characterize the properties of its phosphorylation, we used in vitro kinase and binding assays as well as cellular model and site-direct mutagenesis. Using synaptosomes in superfusion, patch-clamp recordings in autaptic WT and synapsin I KO cortical neurons and SypHy assay on primary cortical culture from wild-type and BAC human LRRK2 G2019S mice we characterized the role of LRRK2 kinase activity on glutamate release and SV trafficking. Here we reported that synapsin I is phosphorylated by LRRK2 and demonstrated that the interaction between LRRK2 WD40 domain and synapsin I is crucial for this phosphorylation. Moreover, we showed that LRRK2 phosphorylation of synapsin I at threonine 337 and 339 significantly reduces synapsin I-SV/actin interactions. Using complementary experimental approaches, we demonstrated that LRRK2 controls glutamate release and SV dynamics in a kinase activity and synapsin I-dependent manner. Our findings show that synapsin I is a LRRK2 substrate and describe a novel mechanisms of regulation of glutamate release by LRRK2 kinase activity.


Asunto(s)
Ácido Glutámico/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Sinapsinas/metabolismo , Transmisión Sináptica/fisiología , Animales , Encéfalo/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Fosforilación , Vesículas Sinápticas/metabolismo
8.
Cell Rep ; 21(13): 3794-3806, 2017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29281828

RESUMEN

Neurotransmission is a tightly regulated Ca2+-dependent process. Upon Ca2+ influx, Synaptotagmin1 (Syt1) promotes fusion of synaptic vesicles (SVs) with the plasma membrane. This requires regulation at multiple levels, but the role of metabolites in SV release is unclear. Here, we uncover a role for isocitrate dehydrogenase 3a (idh3a), a Krebs cycle enzyme, in neurotransmission. Loss of idh3a leads to a reduction of the metabolite, alpha-ketoglutarate (αKG), causing defects in synaptic transmission similar to the loss of syt1. Supplementing idh3a flies with αKG suppresses these defects through an ATP or neurotransmitter-independent mechanism. Indeed, αKG, but not glutamate, enhances Syt1-dependent fusion in a reconstitution assay. αKG promotes interaction between the C2-domains of Syt1 and phospholipids. The data reveal conserved metabolic regulation of synaptic transmission via αKG. Our studies provide a synaptic role for αKG, a metabolite that has been proposed as a treatment for aging and neurodegenerative disorders.


Asunto(s)
Ciclo del Ácido Cítrico , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Drosophila melanogaster/fisiología , Isocitrato Deshidrogenasa/metabolismo , Mitocondrias/metabolismo , Transmisión Sináptica , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Drosophila melanogaster/ultraestructura , Ácidos Cetoglutáricos/metabolismo , Larva/metabolismo , Mitocondrias/ultraestructura , Unión Neuromuscular/metabolismo , Unión Neuromuscular/ultraestructura , Terminales Presinápticos/metabolismo , Terminales Presinápticos/ultraestructura , Unión Proteica , Dominios Proteicos , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestructura , Sinaptotagminas/química , Sinaptotagminas/metabolismo
9.
J Neurosci ; 37(9): 2435-2448, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28137974

RESUMEN

The traditional classification of primary motor cortex (M1) as an agranular area has been challenged recently when a functional layer 4 (L4) was reported in M1. L4 is the principal target for thalamic input in sensory areas, which raises the question of how thalamocortical synapses formed in M1 in the mouse compare with those in neighboring sensory cortex (S1). We identified thalamic boutons by their immunoreactivity for the vesicular glutamate transporter 2 (VGluT2) and performed unbiased disector counts from electron micrographs. We discovered that the thalamus contributed proportionately only half as many synapses to the local circuitry of L4 in M1 compared with S1. Furthermore, thalamic boutons in M1 targeted spiny dendrites exclusively, whereas ∼9% of synapses were formed with dendrites of smooth neurons in S1. VGluT2+ boutons in M1 were smaller and formed fewer synapses per bouton on average (1.3 vs 2.1) than those in S1, but VGluT2+ synapses in M1 were larger than in S1 (median postsynaptic density areas of 0.064 µm2 vs 0.042 µm2). In M1 and S1, thalamic synapses formed only a small fraction (12.1% and 17.2%, respectively) of all of the asymmetric synapses in L4. The functional role of the thalamic input to L4 in M1 has largely been neglected, but our data suggest that, as in S1, the thalamic input is amplified by the recurrent excitatory connections of the L4 circuits. The lack of direct thalamic input to inhibitory neurons in M1 may indicate temporal differences in the inhibitory gating in L4 of M1 versus S1.SIGNIFICANCE STATEMENT Classical interpretations of the function of primary motor cortex (M1) emphasize its lack of the granular layer 4 (L4) typical of sensory cortices. However, we show here that, like sensory cortex (S1), mouse M1 also has the canonical circuit motif of a core thalamic input to the middle cortical layer and that thalamocortical synapses form a small fraction (M1: 12%; S1: 17%) of all asymmetric synapses in L4 of both areas. Amplification of thalamic input by recurrent local circuits is thus likely to be a significant mechanism in both areas. Unlike M1, where thalamocortical boutons typically form a single synapse, thalamocortical boutons in S1 usually formed multiple synapses, which means they can be identified with high probability in the electron microscope without specific labeling.


Asunto(s)
Corteza Motora/ultraestructura , Corteza Somatosensorial/ultraestructura , Tálamo/ultraestructura , Animales , Imagenología Tridimensional , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Modelos Anatómicos , Corteza Motora/metabolismo , Vías Nerviosas/fisiología , Vías Nerviosas/ultraestructura , Fosfopiruvato Hidratasa/metabolismo , Terminales Presinápticos/metabolismo , Terminales Presinápticos/ultraestructura , Corteza Somatosensorial/metabolismo , Sinapsis/metabolismo , Sinapsis/ultraestructura , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestructura , Tálamo/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/ultraestructura
10.
Br J Pharmacol ; 174(2): 119-125, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27771943

RESUMEN

Zn2+ , the second most prevalent trace element in the body, is essential for supporting a wide range of biological functions. While the majority of Zn2+ in the brain is protein-bound, a significant proportion of free Zn2+ is found co-localized with glutamate in synaptic vesicles and is released in an activity-dependent manner. Clinical studies have shown Zn2+ levels are significantly lower in blood and cerebrospinal fluid of children that suffer febrile seizures. Likewise, investigations in multiple animal models demonstrate that low levels of brain Zn2+ increase seizure susceptibility. Recent work provides human genetic evidence that disruption of brain Zn2+ homeostasis at the level of the synapse is associated with increased seizure susceptibility. In this review, we have explored the clinical, functional and genetic data supporting the view that low synaptic Zn2+ increases cellular excitability and febrile seizure susceptibility. Finally, the review focuses on the potential of therapeutic Zn2+ supplementation for at risk patients.


Asunto(s)
Convulsiones Febriles/metabolismo , Vesículas Sinápticas/metabolismo , Zinc/metabolismo , Animales , Humanos
11.
J Biol Chem ; 290(37): 22325-36, 2015 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-26126824

RESUMEN

Synaptic mitochondria are thought to be critical in supporting neuronal energy requirements at the synapse, and bioenergetic failure at the synapse may impair neural transmission and contribute to neurodegeneration. However, little is known about the energy requirements of synaptic vesicle release or whether these energy requirements go unmet in disease, primarily due to a lack of appropriate tools and sensitive assays. To determine the dependence of synaptic vesicle cycling on mitochondrially derived ATP levels, we developed two complementary assays sensitive to mitochondrially derived ATP in individual, living hippocampal boutons. The first is a functional assay for mitochondrially derived ATP that uses the extent of synaptic vesicle cycling as a surrogate for ATP level. The second uses ATP FRET sensors to directly measure ATP at the synapse. Using these assays, we show that endocytosis has high ATP requirements and that vesicle reacidification and exocytosis require comparatively little energy. We then show that to meet these energy needs, mitochondrially derived ATP is rapidly dispersed in axons, thereby maintaining near normal levels of ATP even in boutons lacking mitochondria. As a result, the capacity for synaptic vesicle cycling is similar in boutons without mitochondria as in those with mitochondria. Finally, we show that loss of a key respiratory subunit implicated in Leigh disease markedly decreases mitochondrially derived ATP levels in axons, thus inhibiting synaptic vesicle cycling. This proves that mitochondria-based energy failure can occur and be detected in individual neurons that have a genetic mitochondrial defect.


Asunto(s)
Adenosina Trifosfato/metabolismo , Metabolismo Energético/fisiología , Hipocampo/metabolismo , Mitocondrias/metabolismo , Neuronas/metabolismo , Vesículas Sinápticas/metabolismo , Adenosina Trifosfato/genética , Animales , Células Cultivadas , Endocitosis/fisiología , Exocitosis/fisiología , Hipocampo/citología , Mitocondrias/genética , Neuronas/citología , Ratas , Vesículas Sinápticas/genética
12.
J Cell Sci ; 128(4): 638-44, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25609709

RESUMEN

Ribbon synapses of cochlear inner hair cells (IHCs) employ efficient vesicle replenishment to indefatigably encode sound. In neurons, neuroendocrine and immune cells, vesicle replenishment depends on proteins of the mammalian uncoordinated 13 (Munc13, also known as Unc13) and Ca(2+)-dependent activator proteins for secretion (CAPS) families, which prime vesicles for exocytosis. Here, we tested whether Munc13 and CAPS proteins also regulate exocytosis in mouse IHCs by combining immunohistochemistry with auditory systems physiology and IHC patch-clamp recordings of exocytosis in mice lacking Munc13 and CAPS isoforms. Surprisingly, we did not detect Munc13 or CAPS proteins at IHC presynaptic active zones and found normal IHC exocytosis as well as auditory brainstem responses (ABRs) in Munc13 and CAPS deletion mutants. Instead, we show that otoferlin, a C2-domain protein that is crucial for vesicular fusion and replenishment in IHCs, clusters at the plasma membrane of the presynaptic active zone. Electron tomography of otoferlin-deficient IHC synapses revealed a reduction of short tethers holding vesicles at the active zone, which might be a structural correlate of impaired vesicle priming in otoferlin-deficient IHCs. We conclude that IHCs use an unconventional priming machinery that involves otoferlin.


Asunto(s)
Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Células Ciliadas Auditivas Internas/metabolismo , Proteínas de la Membrana/genética , Transmisión Sináptica/fisiología , Vesículas Sinápticas/metabolismo , Animales , Proteínas de Unión al Calcio/genética , Tomografía con Microscopio Electrónico , Exocitosis/fisiología , Femenino , Células Ciliadas Auditivas Internas/citología , Audición/genética , Audición/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp
13.
Brain Struct Funct ; 220(5): 3011-22, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25012114

RESUMEN

The expression of a vesicular glutamate transporter (VGLUT) suffices to assign a glutamatergic phenotype to neurons and other secretory cells. For example, intestinal L cells express VGLUT2 and secrete glutamate along with glucagon-like peptide 1 (GLP1). We hypothesized that GLP1-positive neurons within the caudal (visceral) nucleus of the solitary tract (cNST) also are glutamatergic. To test this, the axonal projections of GLP1 and other neurons within the cNST were labeled in rats via iontophoretic delivery of anterograde tracer. Dual immunofluorescence and confocal microscopy was used to visualize tracer-, GLP1-, and VGLUT2-positive fibers within brainstem, hypothalamic, and limbic forebrain nuclei that receive input from the cNST. Electron microscopy was used to confirm GLP1 and VGLUT2 immunolabeling within the same axon varicosities, and fluorescent in situ hybridization was used to examine VGLUT2 mRNA expression by GLP1-positive neurons. Most anterograde tracer-labeled fibers displayed VGLUT2-positive varicosities, providing new evidence that ascending axonal projections from the cNST are primarily glutamatergic. Virtually all GLP1-positive varicosities also were VGLUT2-positive. Electron microscopy confirmed the colocalization of GLP1 and VGLUT2 immunolabeling in axon terminals that formed asymmetric (excitatory-type) synapses with unlabeled dendrites in the hypothalamus. Finally, in situ hybridization confirmed that GLP1-positive cNST neurons express VGLUT2 mRNA. Thus, hindbrain GLP1 neurons in rats are equipped to store glutamate in synaptic vesicles, and likely co-release both glutamate and GLP1 from axon varicosities and terminals in the hypothalamus and other brain regions.


Asunto(s)
Péptido 1 Similar al Glucagón/metabolismo , Neuronas/metabolismo , Núcleo Solitario/metabolismo , Sinapsis/metabolismo , Animales , Ácido Glutámico/metabolismo , Hipotálamo/metabolismo , Masculino , Vías Nerviosas/metabolismo , Fenotipo , Terminales Presinápticos/metabolismo , Ratas Sprague-Dawley , Vesículas Sinápticas/metabolismo
14.
J Neurosci ; 34(32): 10554-63, 2014 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-25100589

RESUMEN

Patterned depolarization of Drosophila motor neurons can rapidly induce the outgrowth of new synaptic boutons at the larval neuromuscular junction (NMJ), providing a model system to investigate mechanisms underlying acute structural plasticity. Correlative light and electron microscopy analysis revealed that new boutons typically form near the edge of postsynaptic reticulums of presynaptic boutons. Unlike mature boutons, new varicosities have synaptic vesicles which are distributed uniformly throughout the bouton and undeveloped postsynaptic specializations. To characterize the presynaptic mechanisms mediating new synaptic growth induced by patterned activity, we investigated the formation of new boutons in NMJs lacking synapsin [Syn(-)], a synaptic protein important for vesicle clustering, neurodevelopment, and plasticity. We found that budding of new boutons at Syn(-) NMJs was significantly diminished, and that new boutons in Syn(-) preparations were smaller and had reduced synaptic vesicle density. Since synapsin is a target of protein kinase A (PKA), we assayed whether activity-dependent synaptic growth is regulated via a cAMP/PKA/synapsin pathway. We pretreated preparations with forskolin to raise cAMP levels and found this manipulation significantly enhanced activity-dependent synaptic growth in control but not Syn(-) preparations. To examine the trafficking of synapsin during synaptic growth, we generated transgenic animals expressing fluorescently tagged synapsin. Fluorescence recovery after photobleaching analysis revealed that patterned depolarization promoted synapsin movement between boutons. During new synaptic bouton formation, synapsin redistributed upon stimulation toward the sites of varicosity outgrowth. These findings support a model whereby synapsin accumulates at sites of synaptic growth and facilitates budding of new boutons via a cAMP/PKA-dependent pathway.


Asunto(s)
Unión Neuromuscular/citología , Terminales Presinápticos/fisiología , Sinapsinas/metabolismo , Adyuvantes Inmunológicos/farmacología , Análisis de Varianza , Animales , Animales Modificados Genéticamente , Antígenos CD8/genética , Calcio/metabolismo , Colforsina/farmacología , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas ELAV/genética , Proteínas ELAV/metabolismo , Larva , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Electrónica de Transmisión de Rastreo , Fotoblanqueo , Terminales Presinápticos/ultraestructura , Sinapsinas/genética , Vesículas Sinápticas/genética , Vesículas Sinápticas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
J Neurosci ; 34(7): 2652-9, 2014 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-24523554

RESUMEN

A large number of studies suggest that calcium triggers and accelerates vesicle endocytosis at many synapses and non-neuronal secretory cells. However, many studies show that prolonging the duration of the stimulation train, which induces more calcium influx, slows down endocytosis; and several studies suggest that instead of triggering endocytosis, calcium actually inhibits endocytosis. Here we addressed this apparent conflict at a large nerve terminal, the calyx of Held in rat brainstem, in which recent studies suggest that transient calcium increase up to tens of micromolar concentration at the micro/nano domain triggers endocytosis. By dialyzing 0-1 µM calcium into the calyx via a whole-cell pipette, we found that slow endocytosis was inhibited by calcium dialysis in a concentration-dependent manner. Thus, prolonged, small, and global calcium increase inhibits endocytosis, whereas transient and large calcium increase at the micro/nano domain triggers endocytosis and facilitates endocytosis. This yin and yang effect of calcium may reconcile apparent conflicts regarding whether calcium accelerates or inhibits endocytosis. Whether endocytosis is fast or slow depends on the net outcome between the yin and yang effect of calcium.


Asunto(s)
Tronco Encefálico/metabolismo , Calcio/metabolismo , Endocitosis/fisiología , Vesículas Sinápticas/metabolismo , Animales , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Ratas
16.
Mol Psychiatry ; 19(4): 433-43, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24535456

RESUMEN

Stress and glucocorticoids alter glutamatergic transmission, and the outcome of stress may range from plasticity enhancing effects to noxious, maladaptive changes. We have previously demonstrated that acute stress rapidly increases glutamate release in prefrontal and frontal cortex via glucocorticoid receptor and accumulation of presynaptic SNARE complex. Here we compared the ex vivo effects of acute stress on glutamate release with those of in vitro application of corticosterone, to analyze whether acute effect of stress on glutamatergic transmission is mediated by local synaptic action of corticosterone. We found that acute stress increases both the readily releasable pool (RRP) of vesicles and depolarization-evoked glutamate release, while application in vitro of corticosterone rapidly increases the RRP, an effect dependent on synaptic receptors for the hormone, but does not induce glutamate release for up to 20 min. These findings indicate that corticosterone mediates the enhancement of glutamate release induced by acute stress, and the rapid non-genomic action of the hormone is necessary but not sufficient for this effect.


Asunto(s)
Corticosterona/metabolismo , Lóbulo Frontal/patología , Neuronas/patología , Terminales Presinápticos/metabolismo , Estrés Psicológico/patología , Análisis de Varianza , Animales , Ácido Aspártico/metabolismo , Corticosterona/farmacología , Relación Dosis-Respuesta a Droga , Electrochoque/efectos adversos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Técnicas In Vitro , Masculino , Neuronas/metabolismo , Terminales Presinápticos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Estrés Psicológico/etiología , Sinapsinas/metabolismo , Vesículas Sinápticas/efectos de los fármacos , Vesículas Sinápticas/metabolismo , Sinaptosomas/metabolismo , Tritio/farmacocinética
17.
Neuropsychopharmacology ; 39(4): 841-54, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24108067

RESUMEN

The aim of the present work was to shed light on the role played by the isoprenoid/cholesterol biosynthetic pathway in the modulation of emotional reactivity and memory consolidation in rodents through the inhibition of the key and rate-limiting enzyme 3-hydroxy 3-methylglutaryl Coenzyme A reductase (HMGR) both in vivo and in vitro with simvastatin. Three-month-old male Wistar rats treated for 21 days with simvastatin or vehicle were tested in the social interaction, elevated plus-maze, and inhibitory avoidance tasks; after behavioral testing, the amygdala, hippocampus, prefrontal cortex, dorsal, and ventral striatum were dissected out for biochemical assays. In order to delve deeper into the molecular mechanisms underlying the observed effects, primary rat hippocampal neurons were used. Our results show that HMGR inhibition by simvastatin induces anxiogenic-like effects in the social interaction but not in the elevated plus-maze test, and improves memory consolidation in the inhibitory avoidance task. These effects are accompanied by imbalances in the activity of specific prenylated proteins, Rab3 and RhoA, involved in neurotransmitter release, and synaptic plasticity, respectively. Taken together, the present findings indicate that the isoprenoid/cholesterol biosynthetic pathway is critically involved in the physiological modulation of both emotional and cognitive processes in rodents.


Asunto(s)
Anticolesterolemiantes/farmacología , Colesterol/metabolismo , Cognición/efectos de los fármacos , Emociones/efectos de los fármacos , Simvastatina/farmacología , Terpenos/metabolismo , Animales , Reacción de Prevención/efectos de los fármacos , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/ultraestructura , Proteína de Unión a CREB/metabolismo , Células Cultivadas , Homólogo 4 de la Proteína Discs Large , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Ratas Wistar , Vesículas Sinápticas/efectos de los fármacos , Vesículas Sinápticas/metabolismo , Triglicéridos/sangre
18.
Neuron ; 78(5): 839-54, 2013 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-23684785

RESUMEN

Highly topographic organization of neural circuits exists for the regulation of various brain functions in corticobasal ganglia circuits. Although neural circuit-specific refinement during synapse development is essential for the execution of particular neural functions, the molecular and cellular mechanisms for synapse refinement are largely unknown. Here, we show that protocadherin 17 (PCDH17), one of the nonclustered δ2-protocadherin family members, is enriched along corticobasal ganglia synapses in a zone-specific manner during synaptogenesis and regulates presynaptic assembly in these synapses. PCDH17 deficiency in mice causes facilitated presynaptic vesicle accumulation and enhanced synaptic transmission efficacy in corticobasal ganglia circuits. Furthermore, PCDH17(-/-) mice exhibit antidepressant-like phenotypes that are known to be regulated by corticobasal ganglia circuits. Our findings demonstrate a critical role for PCDH17 in the synaptic development of specific corticobasal ganglia circuits and suggest the involvement of PCDH17 in such circuits in depressive behaviors.


Asunto(s)
Ganglios Basales/citología , Cadherinas/fisiología , Corteza Cerebral/citología , Neuronas/fisiología , Terminales Presinápticos/fisiología , Sinapsis/genética , Estimulación Acústica , Animales , Animales Recién Nacidos , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Transformada , Condicionamiento Psicológico/fisiología , Cricetinae , Cricetulus , Homólogo 4 de la Proteína Discs Large , Conducta Exploratoria , Miedo/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Guanilato-Quinasas/metabolismo , Suspensión Trasera/fisiología , Humanos , Técnicas In Vitro , Macaca mulatta , Masculino , Aprendizaje por Laberinto/fisiología , Potenciales de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Microscopía Electrónica , Red Nerviosa/fisiología , Neuronas/metabolismo , Neuronas/ultraestructura , Técnicas de Placa-Clamp , Protocadherinas , Natación/fisiología , Sinapsis/metabolismo , Sinapsis/ultraestructura , Transmisión Sináptica/genética , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestructura , Proteínas de Transporte Vesicular de Glutamato/metabolismo
19.
Front Neural Circuits ; 7: 202, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24391549

RESUMEN

During development, axons form branches in response to extracellular molecules. Little is known about the underlying molecular mechanisms. Here, we investigate how neurotrophin-induced axon branching is related to synaptic vesicle cycling for thalamocortical axons. The exogenous application of brain-derived neurotrophic factor (BDNF) markedly increased axon branching in thalamocortical co-cultures, while removal of endogenous BDNF reduced branching. Over-expression of a C-terminal fragment of AP180 that inhibits clathrin-mediated endocytosis affected the laminar distribution and the number of branch points. A dominant-negative synaptotagmin mutant that selectively targets synaptic vesicle cycling, strongly suppressed axon branching. Moreover, axons expressing the mutant synaptotagmin were resistant to the branch-promoting effect of BDNF. These results suggest that synaptic vesicle cycling might regulate BDNF induced branching during the development of the axonal arbor.


Asunto(s)
Axones/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Corteza Cerebral/metabolismo , Vesículas Sinápticas/metabolismo , Tálamo/metabolismo , Animales , Axones/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/farmacología , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Vesículas Sinápticas/efectos de los fármacos , Tálamo/efectos de los fármacos
20.
J Neurosci ; 32(47): 16586-96, 2012 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-23175814

RESUMEN

Synaptic communication requires the controlled release of synaptic vesicles from presynaptic axon terminals. Release efficacy is regulated by the many proteins that comprise the presynaptic release apparatus, including Ca(2+) channels and proteins that influence Ca(2+) channel accumulation at release sites. Here we identify Drosophila RIM (Rab3 interacting molecule) and demonstrate that it localizes to active zones at the larval neuromuscular junction. In Drosophila RIM mutants, there is a large decrease in evoked synaptic transmission because of a significant reduction in both the clustering of Ca(2+) channels and the size of the readily releasable pool of synaptic vesicles at active zones. Hence, RIM plays an evolutionarily conserved role in regulating synaptic calcium channel localization and readily releasable pool size. Because RIM has traditionally been studied as an effector of Rab3 function, we investigate whether RIM is involved in the newly identified function of Rab3 in the distribution of presynaptic release machinery components across release sites. Bruchpilot (Brp), an essential component of the active zone cytomatrix T bar, is unaffected by RIM disruption, indicating that Brp localization and distribution across active zones does not require wild-type RIM. In addition, larvae containing mutations in both RIM and rab3 have reduced Ca(2+) channel levels and a Brp distribution that is very similar to that of the rab3 single mutant, indicating that RIM functions to regulate Ca(2+) channel accumulation but is not a Rab3 effector for release machinery distribution across release sites.


Asunto(s)
Canales de Calcio/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Unión Neuromuscular/metabolismo , Proteínas de Unión al GTP rab3/genética , Proteínas de Unión al GTP rab3/metabolismo , Animales , Clonación Molecular , Análisis Mutacional de ADN , ADN Complementario/biosíntesis , ADN Complementario/genética , Proteínas de Drosophila/fisiología , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Larva , Microscopía Confocal , Microscopía Electrónica , Técnicas de Placa-Clamp , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestructura , Proteínas de Unión al GTP rab3/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA