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/metabolismoRESUMEN
Alzheimer's disease (AD) is the most common form of dementia and the most prevalent neurodegenerative disease. Genome-wide association studies have linked PICALM to AD risk. PICALM has been implicated in Aß42 production and turnover, but whether it plays a direct role in modulating Aß42 toxicity remains unclear. We found that increased expression of the Drosophila PICALM orthologue lap could rescue Aß42 toxicity in an adult-onset model of AD, without affecting Aß42 level. Imbalances in the glutamatergic system, leading to excessive, toxic stimulation, have been associated with AD. We found that Aß42 caused the accumulation of presynaptic vesicular glutamate transporter (VGlut) and increased spontaneous glutamate release. Increased lap expression reversed these phenotypes back to control levels, suggesting that lap may modulate glutamatergic transmission. We also found that lap modulated the localization of amphiphysin (Amph), the homologue of another AD risk factor BIN1, and that Amph itself modulated postsynaptic glutamate receptor (GluRII) localization. We propose a model where PICALM modulates glutamatergic transmission, together with BIN1, to ameliorate synaptic dysfunction and disease progression.
Asunto(s)
Enfermedad de Alzheimer/genética , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteínas de Drosophila/genética , Receptores Ionotrópicos de Glutamato/genética , Factores de Transcripción/genética , Proteínas de Transporte Vesicular de Glutamato/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Conducta Animal , Drosophila melanogaster/genética , Fármacos actuantes sobre Aminoácidos Excitadores , Humanos , Proteínas de Ensamble de Clatrina Monoméricas/genética , Proteínas del Tejido Nervioso/genética , Fragmentos de Péptidos/genética , Transmisión Sináptica/genéticaRESUMEN
Glutamate is the most abundant excitatory amino acid in the central nervous system. Neurons using glutamate as a neurotransmitter can be characterised by vesicular glutamate transporters (VGLUTs). Among the three subtypes, VGLUT3 is unique, co-localising with other "classical" neurotransmitters, such as the inhibitory GABA. Glutamate, manipulated by VGLUT3, can modulate the packaging as well as the release of other neurotransmitters and serve as a retrograde signal through its release from the somata and dendrites. Its contribution to sensory processes (including seeing, hearing, and mechanosensation) is well characterised. However, its involvement in learning and memory can only be assumed based on its prominent hippocampal presence. Although VGLUT3-expressing neurons are detectable in the hippocampus, most of the hippocampal VGLUT3 positivity can be found on nerve terminals, presumably coming from the median raphe. This hippocampal glutamatergic network plays a pivotal role in several important processes (e.g., learning and memory, emotions, epilepsy, cardiovascular regulation). Indirect information from anatomical studies and KO mice strains suggests the contribution of local VGLUT3-positive hippocampal neurons as well as afferentations in these events. However, further studies making use of more specific tools (e.g., Cre-mice, opto- and chemogenetics) are needed to confirm these assumptions.
Asunto(s)
Ácido Glutámico/metabolismo , Hipocampo/fisiología , Células Piramidales/metabolismo , Proteínas de Transporte Vesicular de Glutamato/genética , Proteínas de Transporte Vesicular de Glutamato/metabolismo , Animales , Biomarcadores , Fenómenos Electrofisiológicos , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones Noqueados , Neurotransmisores/metabolismo , Transducción de Señal , Transmisión SinápticaRESUMEN
BACKGROUND: The carotid body (CB) plays a critical role in cyclic intermittent hypoxia (CIH)-induced chemosensitivity; however, the underlying mechanism remains uncertain. We have demonstrated the presence of multiple inotropic glutamate receptors (iGluRs) in CB, and that CIH exposure alters the level of some iGluRs in CB. This result implicates glutamatergic signaling in the CB response to hypoxia. The glutamatergic neurotransmission is not only dependent on glutamate and glutamate receptors, but is also dependent on glutamate transporters, including vesicular glutamate transporters (VGluTs) and excitatory amino acid transporters (EAATs). Here, we have further assessed the expression and distribution of VGluTs and EAATs in human and rat CB and the effect of CIH exposure on glutamate transporters expression. METHODS: The mRNA of VGluTs and EAATs in the human CB were detected by RT-PCR. The protein expression of VGluTs and EAATs in the human and rat CB were detected by Western blot. The distribution of VGluT3, EAAT2 and EAAT3 were observed by immunohistochemistry staining and immunofluorescence staining. Male Sprague-Dawley (SD) rats were exposed to CIH (FIO2 10-21%, 3 min/3 min for 8 h per day) for 2 weeks. The unpaired Student's t-test was performed. RESULTS: Here, we report on the presence of mRNAs for VGluT1-3 and EAAT1-3 in human CB, which is consistent with our previous results in rat CB. The proteins of VGluT1 and 3, EAAT2 and 3, but not VGluT2 and EAAT1, were detected with diverse levels in human and rat CB. Immunostaining showed that VGluT3, the major type of VGluTs in CB, was co-localized with tyrosine hydroxylase (TH) in type I cells. EAAT2 and EAAT3 were distributed not only in type I cells, but also in glial fibrillary acidic protein (GFAP) positive type II cells. Moreover, we found that exposure of SD rats to CIH enhanced the protein level of EAAT3 as well as TH, but attenuated the levels of VGluT3 and EAAT2 in CB. CONCLUSIONS: Our study suggests that glutamate transporters are expressed in the CB, and that glutamate transporters may contribute to glutamatergic signaling-dependent carotid chemoreflex to CIH.
Asunto(s)
Cuerpo Carotídeo/metabolismo , Células Quimiorreceptoras/metabolismo , Proteínas de Transporte de Glutamato en la Membrana Plasmática/biosíntesis , Proteínas de Transporte Vesicular de Glutamato/biosíntesis , Sistema de Transporte de Aminoácidos X-AG/análisis , Sistema de Transporte de Aminoácidos X-AG/biosíntesis , Sistema de Transporte de Aminoácidos X-AG/genética , Animales , Cuerpo Carotídeo/química , Células Quimiorreceptoras/química , Expresión Génica , Proteínas de Transporte de Glutamato en la Membrana Plasmática/análisis , Proteínas de Transporte de Glutamato en la Membrana Plasmática/genética , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Proteínas de Transporte Vesicular de Glutamato/análisis , Proteínas de Transporte Vesicular de Glutamato/genéticaRESUMEN
Many neurons receive synchronous input from heterogeneous presynaptic neurons with distinct properties. An instructive example is the crustacean stomatogastric pyloric circuit pacemaker group, consisting of the anterior burster (AB) and pyloric dilator (PD) neurons, which are active synchronously and exert a combined synaptic action on most pyloric follower neurons. Previous studies in lobster have indicated that AB is glutamatergic, whereas PD is cholinergic. However, although the stomatogastric system of the crab Cancer borealis has become a preferred system for exploration of cellular and synaptic basis of circuit dynamics, the pacemaker synaptic output has not been carefully analyzed in this species. We examined the synaptic properties of these neurons using a combination of single-cell mRNA analysis, electrophysiology, and pharmacology. The crab PD neuron expresses high levels of choline acetyltransferase and the vesicular acetylcholine transporter mRNAs, hallmarks of cholinergic neurons. In contrast, the AB neuron expresses neither cholinergic marker but expresses high levels of vesicular glutamate transporter mRNA, consistent with a glutamatergic phenotype. Notably, in the combined synapses to follower neurons, 70-75% of the total current was blocked by putative glutamatergic blockers, but short-term synaptic plasticity remained unchanged, and although the total pacemaker current in two follower neuron types was different, this difference did not contribute to the phasing of the follower neurons. These findings provide a guide for similar explorations of heterogeneous synaptic connections in other systems and a baseline in this system for the exploration of the differential influence of neuromodulators.NEW & NOTEWORTHY The pacemaker-driven pyloric circuit of the Jonah crab stomatogastric nervous system is a well-studied model system for exploring circuit dynamics and neuromodulation, yet the understanding of the synaptic properties of the two pacemaker neuron types is based on older analyses in other species. We use single-cell PCR and electrophysiology to explore the neurotransmitters used by the pacemaker neurons and their distinct contribution to the combined synaptic potentials.
Asunto(s)
Relojes Biológicos , Ganglios de Invertebrados/fisiología , Neuronas/clasificación , Píloro/inervación , Transmisión Sináptica , Acetilcolina/metabolismo , Animales , Braquiuros , Colina O-Acetiltransferasa/genética , Colina O-Acetiltransferasa/metabolismo , Ganglios de Invertebrados/citología , Ácido Glutámico/metabolismo , Neuronas/metabolismo , Neuronas/fisiología , Píloro/fisiología , Proteínas de Transporte Vesicular de Acetilcolina/genética , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo , Proteínas de Transporte Vesicular de Glutamato/genética , Proteínas de Transporte Vesicular de Glutamato/metabolismoRESUMEN
The atypical vesicular glutamate transporter type 3 (VGLUT3) is expressed by subpopulations of neurons using acetylcholine, GABA, or serotonin as neurotransmitters. In addition, VGLUT3 is expressed in the inner hair cells of the auditory system. A mutation (p.A211V) in the gene that encodes VGLUT3 is responsible for progressive deafness in two unrelated families. In this study, we investigated the consequences of the p.A211V mutation in cell cultures and in the CNS of a mutant mouse. The mutation substantially decreased VGLUT3 expression (-70%). We measured VGLUT3-p.A211V activity by vesicular uptake in BON cells, electrophysiological recording of isolated neurons, and its ability to stimulate serotonergic accumulation in cortical synaptic vesicles. Despite a marked loss of expression, the activity of the mutated isoform was only minimally altered. Furthermore, mutant mice displayed none of the behavioral alterations that have previously been reported in VGLUT3 knock-out mice. Finally, we used stimulated emission depletion microscopy to analyze how the mutation altered VGLUT3 distribution within the terminals of mice expressing the mutated isoform. The mutation appeared to reduce the expression of the VGLUT3 transporter by simultaneously decreasing the number of VGLUT3-positive synaptic vesicles and the amount of VGLUT3 per synapses. These observations suggested that VGLUT3 global activity is not linearly correlated with VGLUT3 expression. Furthermore, our data unraveled a nonuniform distribution of VGLUT3 in synaptic vesicles. Identifying the mechanisms responsible for this complex vesicular sorting will be critical to understand VGLUT's involvement in normal and pathological conditions.SIGNIFICANCE STATEMENT VGLUT3 is an atypical member of the vesicular glutamate transporter family. A point mutation of VGLUT3 (VGLUT3-p.A211V) responsible for a progressive loss of hearing has been identified in humans. We observed that this mutation dramatically reduces VGLUT3 expression in terminals (â¼70%) without altering its function. Furthermore, using stimulated emission depletion microscopy, we found that reducing the expression levels of VGLUT3 diminished the number of VGLUT3-positive vesicles at synapses. These unexpected findings challenge the vision of a uniform distribution of synaptic vesicles at synapses. Therefore, the overall activity of VGLUT3 is not proportional to the level of VGLUT3 expression. These data will be key in interpreting the role of VGLUTs in human pathologies.
Asunto(s)
Encéfalo/metabolismo , Mutación Puntual/fisiología , Vesículas Sinápticas/genética , Vesículas Sinápticas/metabolismo , Proteínas de Transporte Vesicular de Glutamato/genética , Proteínas de Transporte Vesicular de Glutamato/metabolismo , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Células Cultivadas , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Distribución AleatoriaRESUMEN
Animals show various behaviors in response to environmental chemicals. These behaviors are often plastic depending on previous experiences. Caenorhabditis elegans, which has highly developed chemosensory system with a limited number of sensory neurons, is an ideal model for analyzing the role of each neuron in innate and learned behaviors. Here, we report a new type of memory-dependent behavioral plasticity in Na+ chemotaxis generated by the left member of bilateral gustatory neuron pair ASE (ASEL neuron). When worms were cultivated in the presence of Na+, they showed positive chemotaxis toward Na+, but when cultivated under Na+-free conditions, they showed no preference regarding Na+ concentration. Both channelrhodopsin-2 (ChR2) activation with blue light and up-steps of Na+ concentration activated ASEL only after cultivation with Na+, as judged by increase in intracellular Ca2+ Under cultivation conditions with Na+, photoactivation of ASEL caused activation of its downstream interneurons AIY and AIA, which stimulate forward locomotion, and inhibition of its downstream interneuron AIB, which inhibits the turning/reversal behavior, and overall drove worms toward higher Na+ concentrations. We also found that the Gq signaling pathway and the neurotransmitter glutamate are both involved in the behavioral response generated by ASEL.SIGNIFICANCE STATEMENT Animals have acquired various types of behavioral plasticity during their long evolutionary history. Caenorhabditis elegans prefers odors associated with food, but plastically changes its behavioral response according to previous experience. Here, we report a new type of behavioral response generated by a single gustatory sensory neuron, the ASE-left (ASEL) neuron. ASEL did not respond to photostimulation or upsteps of Na+ concentration when worms were cultivated in Na+-free conditions; however, when worms were cultivated with Na+, ASEL responded and inhibited AIB to avoid turning and stimulated AIY and AIA to promote forward locomotion, which collectively drove worms toward higher Na+ concentrations. Glutamate and the Gq signaling pathway are essential for driving worms toward higher Na+ concentrations.
Asunto(s)
Quimiotaxis/efectos de los fármacos , Tracto Gastrointestinal/citología , Memoria/fisiología , Red Nerviosa/fisiología , Células Receptoras Sensoriales/fisiología , Cloruro de Sodio/farmacología , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Calcio/metabolismo , Quimiotaxis/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Locomoción/efectos de los fármacos , Locomoción/genética , Memoria/efectos de los fármacos , Microscopía Confocal , Mutación/genética , Red Nerviosa/efectos de los fármacos , Optogenética , Rodopsina/genética , Rodopsina/metabolismo , Células Receptoras Sensoriales/citología , Células Receptoras Sensoriales/efectos de los fármacos , Proteínas de Transporte Vesicular de Glutamato/genética , Proteínas de Transporte Vesicular de Glutamato/metabolismoRESUMEN
The synaptic cleft is the space through which neurotransmitters convey neural information between two synaptic terminals. This space is presumably filled with extracellular matrix molecules involved in synaptic function or differentiation. However, little is known about the identities of the matrix components, and it remains unclear how these molecules organize the matrix in synaptic clefts. In this study, we identified Hasp, a Drosophila secretory protein containing CCP and WAP domains. Molecular genetic analysis revealed that Hasp diffuses extracellularly and is predominantly captured at synaptic clefts of cholinergic synapses. Furthermore, Hasp regulates levels of DLG and the nAChR subunits Dα6 and Dα7 at postsynaptic terminals. Hasp is required for trapping of another matrix protein, Hig, which is also secreted and diffused in the brain, at synaptic clefts of cholinergic synapses; however, Hig is dispensable for localization of Hasp at synaptic clefts. In addition, in the brains of triple mutants for the nAChR subunits Dα5, Dα6, and Dα7, the level of Hig, but not Hasp, was markedly reduced in synaptic regions, indicating that these nAChR subunits are required to anchor Hig to synaptic clefts. High-resolution microscopy revealed that Hasp and Hig exhibit segregated distribution within individual synaptic clefts, reflecting their differing roles in synaptogenesis. These data provide insight into how Hasp and Hig construct the synaptic cleft matrix and regulate the differentiation of cholinergic synapses, and also illuminate a previously unidentified architecture within synaptic clefts. SIGNIFICANCE STATEMENT: The synapse has been extensively studied because it is essential for neurotransmission. By contrast, the space between the synaptic terminals, the synaptic cleft, is still an undeveloped research area despite its ubiquity in synapses. In fruit fly brains, we obtained evidence that the matrix protein Hasp and the previously identified Hig, both of which are secreted extracellularly, localize predominantly to synaptic clefts of cholinergic synapses, and modulate the levels of nAChR subunits on postsynaptic membranes. However, Hasp and Hig play differential roles in matrix formation and exhibit segregated distribution within synaptic clefts. These results reveal the molecular mechanisms of synaptic matrix construction and illuminate a molecular architecture within synaptic clefts previously unrevealed in any animal species.
Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/genética , Neuronas/ultraestructura , Proteínas Serina-Treonina Quinasas/metabolismo , Sinapsis/metabolismo , Sinapsis/ultraestructura , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Encéfalo/citología , Drosophila , Proteínas de Drosophila/genética , Regulación de la Expresión Génica/genética , Masculino , Proteínas del Tejido Nervioso/genética , Neurogénesis/fisiología , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Proteínas de Transporte Vesicular de Glutamato/genética , Proteínas de Transporte Vesicular de Glutamato/metabolismoRESUMEN
Alcohol use disorder is the outcome of both genetic and environmental influences and their interaction via epigenetic mechanisms. The neurotransmitter glutamate is an important regulator of reward circuits and implicated in adaptive changes induced by ethanol intake. The present study aimed at investigating corticolimbic and corticostriatal genetic signatures focusing on the glutamatergic phenotype in relation to early-life stress (ELS) and consequent adult ethanol consumption. A rodent maternal separation model was employed to mimic ELS, and a free-choice paradigm was used to assess ethanol intake in adulthood. Gene expression levels of the Vesicular Glutamate Transporters (Vglut) 1, 2 and 3, as well as two key regulators of DNA methylation, DNA (cytosine-5)-methyltransferase 1 (Dnmt1) and methyl-CpG-binding protein 2 (Mecp2), were analyzed. Brain regions of interest were the ventral tegmental area (VTA), nucleus accumbens (Acb), medial prefrontal cortex (mPFC) and dorsal striatum (dStr), all involved in mediating aspects of ethanol reward. Region-specific Vglut, Dnmt1 and Mecp2 expression patterns were observed. ELS was associated with down-regulated expression of Vglut2 in the VTA and mPFC. Rats exposed to ELS were more sensitive to ethanol-induced changes in Vglut expression in the VTA, Acb, and dStr and in Dnmt1 and Mecp2 expression in the striatal regions. These findings suggest long-term glutamatergic and DNA methylation neuroadaptations as a consequence of ELS, and show an association between voluntary drinking in non-preferring, non-dependent, rodents and different Vglut, Dnmt1 and Mecp2 expression depending on early-life history.
Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Cuerpo Estriado/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Etanol/farmacología , Expresión Génica/efectos de los fármacos , Sistema Límbico/efectos de los fármacos , Privación Materna , Terminales Presinápticos/efectos de los fármacos , Estrés Psicológico/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Conducta de Elección , Cuerpo Estriado/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasa 1/genética , Femenino , Ácido Glutámico/metabolismo , Sistema Límbico/metabolismo , Masculino , Proteína 2 de Unión a Metil-CpG/efectos de los fármacos , Proteína 2 de Unión a Metil-CpG/genética , Neostriado/efectos de los fármacos , Neostriado/metabolismo , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Terminales Presinápticos/metabolismo , Ratas , Ratas Wistar , Recompensa , Estrés Psicológico/metabolismo , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/efectos de los fármacos , Proteína 1 de Transporte Vesicular de Glutamato/genética , Proteína 2 de Transporte Vesicular de Glutamato/efectos de los fármacos , Proteína 2 de Transporte Vesicular de Glutamato/genética , Proteínas de Transporte Vesicular de Glutamato/efectos de los fármacos , Proteínas de Transporte Vesicular de Glutamato/genéticaRESUMEN
BACKGROUND: One of the causes of sensorineural hearing loss (SNHL) is degeneration of the inner hair cells in the organ of Corti in the cochlea. The SLC17A8 (solute carrier family 17, member 8) gene encodes vesicular glutamate transporter 3 (VGLUT3), and among its isoforms (VGLUT1-3), only VGLUT3 is expressed selectively in the inner hair cells (IHCs). VGLUT3 transports the neurotransmitter glutamate into the synaptic vesicles of the IHCs. Mutation of the SLC17A8 gene is reported to be associated with DFNA25 (deafness, autosomal dominant 25), an autosomal dominant non-syndromic hearing loss (ADNSHL) in humans. METHODS: In this study, we performed a genetic analysis of 87 unrelated Korean patients with ADNSHL to determine whether the SLC17A8 gene affects hearing ability in the Korean population. RESULTS: We found a novel heterozygous frameshift mutation, 2 non-synonymous variations, and a synonymous variation. The novel frameshift mutation, p.M206Nfs*4, in which methionine is changed to asparagine at amino acid position 206, resulted in a termination codon at amino acid position 209. This alteration is predicted to encode a truncated protein lacking transmembrane domains 5 to 12. This mutation is located in a highly conserved region in VGLUT3 across multiple amino acid alignments in different vertebrate species, but it was not detected in 100 unrelated controls who had normal hearing ability. The results from our study suggest that the p.M206Nfs*4 mutation in the SLC17A8 gene is likely a pathogenic mutation that causes ADNSHL. CONCLUSION: Our findings can facilitate the prediction of the primary cause of ADNSHL in Korean patients.
Asunto(s)
Pueblo Asiatico/genética , Pérdida Auditiva Sensorineural/genética , Proteínas de Transporte Vesicular de Glutamato/genética , Secuencia de Aminoácidos , Estudios de Casos y Controles , Femenino , Mutación del Sistema de Lectura , Pruebas Genéticas , Genómica , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación , Linaje , Polimorfismo de Longitud del Fragmento de Restricción , República de Corea , Alineación de Secuencia , Análisis de Secuencia de ADNRESUMEN
Tonically active cholinergic interneurons (TANs) from the nucleus accumbens (NAc) are centrally involved in reward behavior. TANs express a vesicular glutamate transporter referred to as VGLUT3 and thus use both acetylcholine and glutamate as neurotransmitters. The respective roles of each transmitter in the regulation of reward and addiction are still unknown. In this study, we showed that disruption of the gene that encodes VGLUT3 (Slc17a8) markedly increased cocaine self-administration in mice. Concomitantly, the amount of dopamine (DA) release was strongly augmented in the NAc of VGLUT3(-/-) mice because of a lack of signaling by metabotropic glutamate receptors. Furthermore, dendritic spines and glutamatergic synaptic transmission on medium spiny neurons were increased in the NAc of VGLUT3(-/-) mice. Increased DA and glutamate signaling in the NAc are hallmarks of addiction. Our study shows that TANs use glutamate to reduce DA release and decrease reinforcing properties of cocaine in mice. Interestingly, we also observed an increased frequency of rare variations in SLC17A8 in a cohort of severe drug abusers compared with controls. Our findings identify VGLUT3 as an unexpected regulator of drug abuse.
Asunto(s)
Trastornos Relacionados con Cocaína/genética , Trastornos Relacionados con Cocaína/patología , Dopamina/metabolismo , Predisposición Genética a la Enfermedad/genética , Ácido Glutámico/metabolismo , Núcleo Accumbens/metabolismo , Transducción de Señal/fisiología , Proteínas de Transporte Vesicular de Glutamato/genética , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Adulto , Animales , Cocaína/farmacología , Condicionamiento Operante/efectos de los fármacos , Inhibidores de Captación de Dopamina/farmacología , Humanos , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Núcleo Accumbens/citología , Núcleo Accumbens/efectos de los fármacos , Trastornos Relacionados con Opioides/genética , Trastornos Relacionados con Opioides/patología , Autoadministración , Potenciales Sinápticos/efectos de los fármacos , Potenciales Sinápticos/genética , Proteínas de Transporte Vesicular de Glutamato/deficienciaRESUMEN
Adenocarcinoma (AC) and squamous cell carcinoma (SqCC) are two major histological subtypes of lung cancer. Genome-wide association studies (GWAS) have made considerable advances in the understanding of lung cancer susceptibility. Obvious heterogeneity has been observed between different histological subtypes of lung cancer, but genetic determinants in specific to lung SqCC have not been systematically investigated. Here, we performed the GWAS analysis specifically for lung SqCC in 833 SqCC cases and 3,094 controls followed by a two-stage replication in additional 2,223 lung SqCC cases and 6,409 controls from Chinese populations. We found that rs12296850 in SLC17A8-NR1H4 gene region at12q23.1 was significantly associated with risk of lung SqCC at genome-wide significance level [additive model: odds ratio (OR)â=â0.78, 95% confidence interval (CI)â=â0.72-0.84, Pâ=â1.19×10(-10)]. Subjects carrying AG or GG genotype had a 26% (ORâ=â0.74, 95% CIâ=â0.67-0.81) or 32% (ORâ=â0.68, 95% CIâ=â0.56-0.83) decreased risk of lung SqCC, respectively, as compared with AA genotype. However, we did not observe significant association between rs12296850 and risk of lung AC in a total of 4,368 cases with lung AC and 9,486 controls (ORâ=â0.96, 95% CIâ=â0.90-1.02, Pâ=â0.173). These results indicate that genetic variations on chromosome 12q23.1 may specifically contribute to lung SqCC susceptibility in Chinese population.
Asunto(s)
Carcinoma de Células Escamosas , Estudio de Asociación del Genoma Completo , Neoplasias Pulmonares , Receptores Citoplasmáticos y Nucleares/genética , Proteínas de Transporte Vesicular de Glutamato/genética , Pueblo Asiatico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Cromosomas Humanos Par 12/genética , Predisposición Genética a la Enfermedad , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Polimorfismo de Nucleótido Simple , Factores de RiesgoRESUMEN
Using riboprobe in situ hybridization, we studied the localization of the transcripts for the neuropeptide galanin and its receptors (GalR1-R3), tryptophan hydroxylase 2, tyrosine hydroxylase, and nitric oxide synthase as well as the three vesicular glutamate transporters (VGLUT 1-3) in the locus coeruleus (LC) and the dorsal raphe nucleus (DRN) regions of postmortem human brains. Quantitative real-time PCR (qPCR) was used also. Galanin and GalR3 mRNA were found in many noradrenergic LC neurons, and GalR3 overlapped with serotonin neurons in the DRN. The qPCR analysis at the LC level ranked the transcripts in the following order in the LC: galanin >> GalR3 >> GalR1 > GalR2; in the DRN the ranking was galanin >> GalR3 >> GalR1 = GalR2. In forebrain regions the ranking was GalR1 > galanin > GalR2. VGLUT1 and -2 were strongly expressed in the pontine nuclei but could not be detected in LC or serotonin neurons. VGLUT2 transcripts were found in very small, nonpigmented cells in the LC and in the lateral and dorsal aspects of the periaqueductal central gray. Nitric oxide synthase was not detected in serotonin neurons. These findings show distinct differences between the human brain and rodents, especially rat, in the distribution of the galanin system and some other transmitter systems. For example, GalR3 seems to be the important galanin receptor in both the human LC and DRN versus GalR1 and -2 in the rodent brain. Such knowledge may be important when considering therapeutic principles and drug development.
Asunto(s)
Encéfalo/metabolismo , Galanina/genética , Galanina/metabolismo , Neurotransmisores/metabolismo , Animales , Humanos , Hibridación in Situ , Locus Coeruleus/metabolismo , Neurotransmisores/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Núcleos del Rafe/metabolismo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Galanina Tipo 1/genética , Receptor de Galanina Tipo 1/metabolismo , Receptor de Galanina Tipo 2/genética , Receptor de Galanina Tipo 2/metabolismo , Receptor de Galanina Tipo 3/genética , Receptor de Galanina Tipo 3/metabolismo , Especificidad de la Especie , Distribución Tisular , Proteína 1 de Transporte Vesicular de Glutamato/genética , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/genética , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Proteínas de Transporte Vesicular de Glutamato/genética , Proteínas de Transporte Vesicular de Glutamato/metabolismoRESUMEN
The concept of a tripartite synapse including a presynaptic terminal, a postsynaptic spine, and an astrocytic process that responds to neuronal activity by fast gliotransmitter release, confers to the electrically silent astrocytes an active role in information processing. However, the mechanisms of gliotransmitter release are still highly controversial. The reported expression of all three vesicular glutamate transporters (VGLUT1-3) by astrocytes suggests that astrocytes, like neurons, may release glutamate by exocytosis. However, the demonstration of astrocytic VGLUT expression is largely based on immunostaining, and the possibility of nonspecific labeling needs to be systematically addressed. We therefore examined the expression of VGLUT1-3 in astrocytes, both in culture and in situ. We used Western blots and single-vesicle imaging by total internal reflection fluorescence microscopy in live cultured astrocytes, and confocal microscopy, at the cellular level in cortical, hippocampal, and cerebellar brain slices, combined with quantitative image analysis. Control experiments were systematically performed in cultured astrocytes using wild-type, VGLUT1-3 knock-out, VGLUT1(Venus) knock-in, and VGLUT2-EGFP transgenic mice. In fixed brain slices, we quantified the degree of overlap between VGLUT1-3 and neuronal or astrocytic markers, both in an object-based manner using fluorescence line profiles, and in a pixel-based manner using dual-color scatter plots followed by the calculation of Pearson's correlation coefficient over all pixels with intensities significantly different from background. Our data provide no evidence in favor of the expression of any of the three VGLUTs by gray matter protoplasmic astrocytes of the primary somatosensory cortex, the thalamic ventrobasal nucleus, the hippocampus, and the cerebellum.
Asunto(s)
Astrocitos/metabolismo , Proteínas de Transporte Vesicular de Glutamato/metabolismo , Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Corteza Cerebral/citología , Homólogo 4 de la Proteína Discs Large , Transportador 2 de Aminoácidos Excitadores/metabolismo , Femenino , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Guanilato-Quinasas/metabolismo , Hipocampo/citología , Procesamiento de Imagen Asistido por Computador , Técnicas In Vitro , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Microscopía Confocal , Proteínas del Tejido Nervioso/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Proteínas de Transporte Vesicular de Glutamato/clasificación , Proteínas de Transporte Vesicular de Glutamato/genéticaRESUMEN
Increasing evidence indicates that the Eph receptors and their ephrin ligands are involved in the regulation of interactions between neurons and astrocytes. Moreover, astrocytic ephrin-A3 reverse signaling mediated by EphA4 receptors is necessary for controlling the abundance of glial glutamate transporters. However, the role of ephrin-A3 reverse signaling in astrocytic function and neuronal death under ischemic conditions remains unclear. In the present study, we found that the EphA4 receptor and its ephrin-A3 ligand, which were distributed in neurons and astrocytes, respectively, in the hippocampus showed a coincident up-regulation of protein expression in the early stage of ischemia. Application of clustered EphA4 decreased the expressions of astrocytic glutamate transporters together with astrocytic glutamate uptake capacity through activating ephrin-A3 reverse signaling. In consequence, neuronal loss was aggravated in the CA1 region of the hippocampus accompanied by impaired hippocampus-dependent spatial memory when clustered EphA4 treatment was administered prior to transient global ischemia. These findings indicate that EphA4-mediated ephrin-A3 reverse signaling is a crucial mechanism for astrocytes to control glial glutamate transporters and prevent glutamate excitotoxicity under pathological conditions. Astrocytic ephrin-A3 reverse signaling mediated by EphA4 receptor is necessary for controlling the abundance of glial glutamate transporters under physiological conditions. However, the role of ephrin-A3 reverse signaling in astrocytic function and neuronal death under ischemic conditions remains unclear. We found EphA4-mediated ephrin-A3 reverse signaling to be a crucial mechanism for astrocytes to control glial glutamate transporters and protect hippocampal neurons from glutamate excitotoxicity under ischemic conditions, this cascade representing a potential therapeutic target for stroke.
Asunto(s)
Astrocitos/metabolismo , Efrina-A3/farmacología , Hipocampo/patología , Ataque Isquémico Transitorio/metabolismo , Ataque Isquémico Transitorio/patología , Neuronas/patología , Proteínas de Transporte Vesicular de Glutamato/metabolismo , Animales , Inyecciones Intraventriculares , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Receptores de la Familia Eph/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas de Transporte Vesicular de Glutamato/genéticaRESUMEN
Disrupted-in-schizophrenia 1 (DISC1) is a genetic risk factor that has been implicated in major mental disorders. DISC1 binds to and stabilizes serine racemase to regulate production of D-serine by astrocytes, contributing to glutamate (GLU) neurotransmission. However, the possible involvement of astrocytic DISC1 in synthesis, metabolism, reuptake, or secretion of GLU remains unexplored. Therefore, we studied the effects of dominant-negative mutant DISC1 on various aspects of GLU metabolism by using primary astrocyte cultures and hippocampal tissue from transgenic mice with astrocyte-restricted expression of mutant DISC1. Although mutant DISC1 had no significant effects on astrocyte proliferation, GLU reuptake, glutaminase, or glutamate carboxypeptidase II activity, expression of mutant DISC1 was associated with increased levels of alanine-serine-cysteine transporter 2, vesicular glutamate transporters 1 and 3 in primary astrocytes and in the hippocampus, and elevated expression of the NR1 subunit and diminished expression of the NR2A subunit of N-methyl-D-aspartate (NMDA) receptors in the hippocampus, at postnatal day 21. Our findings indicate that decreased D-serine production by astrocytic mutant DISC1 might lead to compensatory changes in levels of the amino acid transporters and NMDA receptors in the context of tripartite synapse.
Asunto(s)
Astrocitos/metabolismo , Ácido Glutámico/metabolismo , Mutación/genética , Proteínas del Tejido Nervioso/genética , Animales , Animales Recién Nacidos , Bromodesoxiuridina/metabolismo , Proliferación Celular/genética , Células Cultivadas , Proteína Ácida Fibrilar de la Glía/genética , Glutamato Carboxipeptidasa II/metabolismo , Glutaminasa/metabolismo , Hipocampo/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Prosencéfalo/citología , Sinaptosomas/metabolismo , Proteínas de Transporte Vesicular de Glutamato/genética , Proteínas de Transporte Vesicular de Glutamato/metabolismoRESUMEN
Recurrent inhibition, wherein excitatory principal neurons stimulate inhibitory interneurons that feedback on the same principal cells, occurs ubiquitously in the brain. However, the regulation and function of recurrent inhibition are poorly understood in terms of the contributing interneuron subtypes as well as their effect on neural and cognitive outputs. In the Drosophila olfactory system, odorants activate olfactory sensory neurons (OSNs), which stimulate projection neurons (PNs) in the antennal lobe. Both OSNs and PNs activate local inhibitory neurons (LNs) that provide either feedforward or recurrent/feedback inhibition in the lobe. During olfactory habituation, prior exposure to an odorant selectively decreases the animal's subsequent response to the odorant. We show here that habituation occurs in response to feedback from PNs. Output from PNs is necessary for olfactory habituation and, in the absence of odorant, direct PN activation is sufficient to induce the odorant-selective behavioral attenuation characteristic of olfactory habituation. PN-induced habituation occludes further odor-induced habituation and similarly requires GABA(A)Rs and NMDARs in PNs, as well as VGLUT and cAMP signaling in the multiglomerular inhibitory local interneurons (LN1) type of LN. Thus, PN output is monitored by an LN subtype whose resultant plasticity underlies behavioral habituation. We propose that recurrent inhibitory motifs common in neural circuits may similarly underlie habituation to other complex stimuli.
Asunto(s)
Antenas de Artrópodos/fisiología , Conexinas/fisiología , Proteínas de Drosophila/fisiología , Retroalimentación Sensorial/fisiología , Proteínas del Tejido Nervioso/fisiología , Inhibición Neural/fisiología , Vías Olfatorias/fisiología , Animales , Animales Modificados Genéticamente , Conexinas/genética , AMP Cíclico/fisiología , Drosophila , Proteínas de Drosophila/genética , Dinaminas/genética , Dinaminas/fisiología , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/fisiología , Habituación Psicofisiológica/fisiología , Canales Iónicos , Proteínas del Tejido Nervioso/genética , Neuronas/fisiología , Receptores de GABA-A/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Receptores Odorantes/fisiología , Células Receptoras Sensoriales/fisiología , Olfato/fisiología , Canal Catiónico TRPA1 , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/fisiología , Proteínas de Transporte Vesicular de Glutamato/genética , Proteínas de Transporte Vesicular de Glutamato/fisiologíaRESUMEN
Dopaminergic neurons of the substantia nigra compacta (SNC), ventral tegmental area (VTA) and retrorubral field (RRF) play a role in reward, motivation, learning, memory, and movement. These neurons are intermingled with GABAergic neurons. Recent evidence shows that the VTA contains glutamatergic neurons expressing vesicular glutamate transporter type 2 (VGluT2); some of them co-express tyrosine hydroxylase (TH). Here, we used a combination of radioactive in situ hybridisation and immunohistochemistry to explore whether any of the vesicular glutamate transporters [vesicular glutamate transporter type 1 (VGluT1), VGluT2, or vesicular glutamate transporter type 3 (VGluT3)] were encoded by neurons in the SNC or RRF. We found expression of VGluT2 mRNA, but not of VGluT1 or VGluT3, in the SNC and RRF. These VGluT2 neurons rarely showed TH immunoreactivity. Within the SNC, the VGluT2 neurons were infrequently found at the rostral level, but were often seen at the medial and caudal levels intercalated in the mediolateral portion of the dorsal tier, at a ratio of one VGluT2 neuron per 4.4 TH neurons. At this level, VGluT2 neurons were also found in the adjacent substantia nigra reticulata and substantia nigra pars lateralis. Within the RRF, the VGluT2 neurons showed an increasing rostrocaudal gradient of distribution. The RRF proportion of VGluT2 neurons in relation to TH neurons was constant throughout the rostrocaudal levels, showing an average ratio of one VGluT2 neuron per 1.7 TH neurons. In summary, we provide evidence indicating that the SNC and RRF, which are traditionally considered to be dopaminergic areas, have neurons with the ability to participate in glutamate signaling.
Asunto(s)
Ácido Glutámico/metabolismo , Neuronas/metabolismo , Sustancia Negra/metabolismo , Proteínas de Transporte Vesicular de Glutamato/metabolismo , Animales , Neuronas GABAérgicas/metabolismo , Neuronas/clasificación , Especificidad de Órganos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Núcleo Rojo/citología , Núcleo Rojo/metabolismo , Sustancia Negra/citología , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo , Área Tegmental Ventral/citología , Área Tegmental Ventral/metabolismo , Proteínas de Transporte Vesicular de Glutamato/genéticaRESUMEN
PURPOSE: VGLUTs, which are essential for loading glutamate into synaptic vesicles, are present in various neuronal systems. However, to our knowledge the expression of VGLUTs in neurons innervating the bladder has not yet been analyzed. We studied VGLUT1, VGLUT2 and VGLUT3 in mouse bladder neurons. MATERIALS AND METHODS: We analyzed the expression of VGLUT1, VGLUT2 and calcitonin gene-related peptide by immunohistochemistry in the retrograde labeled primary afferent and autonomic neurons of BALB/c mice after injecting fast blue in the bladder wall. To study VGLUT3 we traced the bladder of transgenic mice, in which VGLUT3 is identified by enhanced green fluorescent protein detection. RESULTS: Most bladder dorsal root ganglion neurons expressed VGLUT2. A smaller percentage of neurons also expressed VGLUT1 or VGLUT3. Co-expression with calcitonin gene-related peptide was only observed for VGLUT2. Occasional VGLUT2 immunoreactive neurons were seen in the major pelvic ganglia. Abundant VGLUT2 immunoreactive nerves were detected in the bladder dome and trigone, and the urethra. VGLUT1 immunoreactive nerves were discretely present. CONCLUSIONS: We present what are to our knowledge novel data on VGLUT expression in sensory and autonomic neurons innervating the mouse bladder. The frequent association of VGLUT2 and calcitonin gene-related peptide in sensory neurons suggests interactions between glutamatergic and peptidergic neurotransmissions, potentially influencing commonly perceived sensations in the bladder, such as discomfort and pain.
Asunto(s)
Péptido Relacionado con Gen de Calcitonina/metabolismo , Ganglios Espinales/metabolismo , Vejiga Urinaria/inervación , Proteínas de Transporte Vesicular de Glutamato/metabolismo , Animales , Sistema Nervioso Autónomo/fisiología , Péptido Relacionado con Gen de Calcitonina/genética , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Animales , Neuronas Aferentes/metabolismo , Neuronas Aferentes/fisiología , Sensibilidad y Especificidad , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología , Vejiga Urinaria/metabolismo , Proteínas de Transporte Vesicular de Glutamato/genéticaRESUMEN
Visceral pain is among the most prevalent and bothersome forms of chronic pain, but their transmission in the spinal cord is still poorly understood. Here, we conducted focal colorectal distention (fCRD) to drive both visceromotor responses (VMRs) and aversion. We first found that spinal CCK neurons were necessary for noxious fCRD to drive both VMRs and aversion under naive conditions. We next showed that spinal VGLUT3 neurons mediate visceral allodynia, whose ablation caused loss of aversion evoked by low-intensity fCRD in mice with gastrointestinal (GI) inflammation or spinal circuit disinhibition. Importantly, these neurons were dispensable for driving sensitized VMRs under both inflammatory and central disinhibition conditions. Anatomically, a subset of VGLUT3 neurons projected to parabrachial nuclei, whose photoactivation sufficiently generated aversion in mice with GI inflammation, without influencing VMRs. Our studies suggest the presence of different spinal substrates that transmit nociceptive versus affective dimensions of visceral sensory information.