Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-32116571

ABSTRACT

The piriform cortex (PC) is a key brain area involved in both processing and coding of olfactory information. It is implicated in various brain disorders, such as epilepsy, Alzheimer's disease, and autism. The PC consists of the anterior (APC) and posterior (PPC) parts, which are different anatomically and functionally. However, the direct input networks to specific neuronal populations within the APC and PPC remain poorly understood. Here, we mapped the whole-brain direct inputs to the two major neuronal populations, the excitatory glutamatergic principal neurons and inhibitory γ-aminobutyric acid (GABA)-ergic interneurons within the APC and PPC using the rabies virus (RV)-mediated retrograde trans-synaptic tracing system. We found that for both types of neurons, APC and PPC share some similarities in input networks, with dominant inputs originating from the olfactory region (OLF), followed by the cortical subplate (CTXsp), isocortex, cerebral nuclei (CNU), hippocampal formation (HPF) and interbrain (IB), whereas the midbrain (MB) and hindbrain (HB) were rarely labeled. However, APC and PPC also show distinct features in their input distribution patterns. For both types of neurons, the input proportion from the OLF to the APC was higher than that to the PPC; while the PPC received higher proportions of inputs from the HPF and CNU than the APC did. Overall, our results revealed the direct input networks of both excitatory and inhibitory neuronal populations of different PC subareas, providing a structural basis to analyze the diverse PC functions.


Subject(s)
GABAergic Neurons/physiology , Glutamic Acid/physiology , Piriform Cortex/cytology , Piriform Cortex/physiology , Animals , Cell Count/methods , Female , GABAergic Neurons/chemistry , Glutamate Decarboxylase/analysis , Glutamate Decarboxylase/physiology , Glutamic Acid/analysis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Piriform Cortex/chemistry , Vesicular Glutamate Transport Protein 2/analysis , Vesicular Glutamate Transport Protein 2/physiology
2.
FASEB J ; 33(1): 400-417, 2019 01.
Article in English | MEDLINE | ID: mdl-30011230

ABSTRACT

Dopamine (DA) is a key regulator of circuits controlling movement and motivation. A subset of midbrain DA neurons has been shown to express the vesicular glutamate transporter (VGLUT)2, underlying their capacity for glutamate release. Glutamate release is found mainly by DA neurons of the ventral tegmental area (VTA) and can be detected at terminals contacting ventral, but not dorsal, striatal neurons, suggesting the possibility that target-derived signals regulate the neurotransmitter phenotype of DA neurons. Whether glutamate can be released from the same terminals that release DA or from a special subset of axon terminals is unclear. Here, we provide in vitro and in vivo data supporting the hypothesis that DA and glutamate-releasing terminals in mice are mostly segregated and that striatal neurons regulate the cophenotype of midbrain DA neurons and the segregation of release sites. Our work unveils a fundamental feature of dual neurotransmission and plasticity of the DA system.-Fortin, G. M., Ducrot, C., Giguère, N., Kouwenhoven, W. M., Bourque, M.-J., Pacelli, C., Varaschin, R. K., Brill, M., Singh, S., Wiseman, P. W., Trudeau, L.-E. Segregation of dopamine and glutamate release sites in dopamine neuron axons: regulation by striatal target cells.


Subject(s)
Corpus Striatum/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Glutamic Acid/metabolism , Synaptic Transmission , Ventral Tegmental Area/metabolism , Vesicular Glutamate Transport Protein 2/physiology , Animals , Corpus Striatum/cytology , Dopaminergic Neurons/cytology , Male , Mice , Mice, Knockout , Ventral Tegmental Area/cytology
3.
Neurobiol Dis ; 121: 230-239, 2019 01.
Article in English | MEDLINE | ID: mdl-30308244

ABSTRACT

Brain microvascular endothelial cells (BMEC) are highly complex regulatory cells that communicate with other cells in the neurovascular unit. Cerebral ischemic injury is known to produce detectable synaptic dysfunction. This study aims to investigate whether endothelial cells in the brain regulate postnatal synaptic development and to elucidate their role in functional recovery after ischemia. Here, we found that in vivo engraftment of endothelial cells increased synaptic puncta and excitatory postsynaptic currents in layers 2/3 of the motor cortex. This pro-synaptogenic effect was blocked by the depletion of VEGF in the grafted BMEC. The in vitro results showed that BMEC conditioned medium enhanced spine and synapse formation but conditioned medium without VEGF had no such effects. Moreover, under pathological conditions, transplanted endothelial cells were capable of enhancing angiogenesis and synaptogenesis and improved motor function in the ischemic injury model. Collectively, our findings suggest that endothelial cells promote excitatory synaptogenesis via the paracrine factor VEGF during postnatal development and exert repair functions in hypoxia-ischemic neonatal mice. This study highlights the importance of the endothelium-neuron interaction not only in regulating neuronal development but also in maintaining healthy brain function.


Subject(s)
Brain Ischemia/physiopathology , Endothelial Cells/physiology , Excitatory Postsynaptic Potentials , Motor Cortex/blood supply , Motor Cortex/growth & development , Motor Disorders/physiopathology , Synapses/physiology , Animals , Animals, Newborn , Brain Ischemia/complications , Cells, Cultured , Culture Media, Conditioned , Female , Male , Mice, Inbred C57BL , Microvessels/physiology , Motor Disorders/etiology , Neovascularization, Physiologic , Thalamus/growth & development , Vascular Endothelial Growth Factor A/physiology , Vesicular Glutamate Transport Protein 2/physiology
4.
Hear Res ; 376: 1-10, 2019 05.
Article in English | MEDLINE | ID: mdl-30314930

ABSTRACT

The inferior colliculus occupies a central position in ascending and descending auditory pathways. A substantial proportion of its neurons are GABAergic, and these neurons contribute to intracollicular circuits as well as to extrinsic projections to numerous targets. A variety of types of evidence - morphology, physiology, molecular markers - indicate that the GABAergic cells can be divided into at least four subtypes that serve different functions. However, there has yet to emerge a unified scheme for distinguishing these subtypes. The present review discusses these criteria and, where possible, relates the different properties. In contrast to GABAergic cells in cerebral cortex, where subtypes are much more thoroughly characterized, those in the inferior colliculus contribute substantially to numerous long range extrinsic projections. At present, the best characterized subtype is a GABAergic cell with a large soma, dense perisomatic synaptic inputs and a large axon that provides rapid auditory input to the thalamus. This large GABAergic subtype projects to additional targets, and other subtypes also project to the thalamus. The eventual characterization of these subtypes can be expected to reveal multiple functions of these inhibitory cells and the many circuits to which they contribute.


Subject(s)
GABAergic Neurons/classification , GABAergic Neurons/physiology , Inferior Colliculi/cytology , Inferior Colliculi/physiology , Animals , Auditory Pathways/cytology , Auditory Pathways/physiology , Calcium-Binding Proteins/physiology , Cell Surface Extensions/physiology , Cell Surface Extensions/ultrastructure , GABAergic Neurons/cytology , Models, Neurological , Receptors, Neurotransmitter/physiology , Vesicular Glutamate Transport Protein 2/physiology
5.
eNeuro ; 5(3)2018.
Article in English | MEDLINE | ID: mdl-29756029

ABSTRACT

Intrinsically photosensitive retinal ganglion cells (ipRGCs) innervate the hypothalamic suprachiasmatic nucleus (SCN), a circadian oscillator that functions as a biological clock. ipRGCs use vesicular glutamate transporter 2 (vGlut2) to package glutamate into synaptic vesicles and light-evoked resetting of the SCN circadian clock is widely attributed to ipRGC glutamatergic neurotransmission. Pituitary adenylate cyclase-activating polypeptide (PACAP) is also packaged into vesicles in ipRGCs and PACAP may be coreleased with glutamate in the SCN. vGlut2 has been conditionally deleted in ipRGCs in mice [conditional knock-outs (cKOs)] and their aberrant photoentrainment and residual attenuated light responses have been ascribed to ipRGC PACAP release. However, there is no direct evidence that all ipRGC glutamatergic neurotransmission is eliminated in vGlut2 cKOs. Here, we examined two lines of ipRGC vGlut2 cKO mice for SCN-mediated behavioral responses under several lighting conditions and for ipRGC glutamatergic neurotransmission in the SCN. Circadian behavioral responses varied from a very limited response to light to near normal photoentrainment. After collecting behavioral data, hypothalamic slices were prepared and evoked EPSCs (eEPSCs) were recorded from SCN neurons by stimulating the optic chiasm. In cKOs, glutamatergic eEPSCs were recorded and all eEPSC parameters examined (stimulus threshold, amplitude, rise time or time-to-peak and stimulus strength to evoke a maximal response) were similar to controls. We conclude that a variable number but functionally significant percentage of ipRGCs in two vGlut2 cKO mouse lines continue to release glutamate. Thus, the residual SCN-mediated light responses in these cKO mouse lines cannot be attributed solely to ipRGC PACAP release.


Subject(s)
Behavior, Animal , Circadian Rhythm , Excitatory Postsynaptic Potentials , Glutamic Acid/metabolism , Optic Chiasm/physiology , Retinal Ganglion Cells/physiology , Suprachiasmatic Nucleus/physiology , Vesicular Glutamate Transport Protein 2/physiology , Animals , Female , Male , Mice, Knockout , Motor Activity , Photic Stimulation
6.
Nat Commun ; 8(1): 1405, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29123082

ABSTRACT

Basic and clinical observations suggest that the caudal hypothalamus comprises a key node of the ascending arousal system, but the cell types underlying this are not fully understood. Here we report that glutamate-releasing neurons of the supramammillary region (SuMvglut2) produce sustained behavioral and EEG arousal when chemogenetically activated. This effect is nearly abolished following selective genetic disruption of glutamate release from SuMvglut2 neurons. Inhibition of SuMvglut2 neurons decreases and fragments wake, also suppressing theta and gamma frequency EEG activity. SuMvglut2 neurons include a subpopulation containing both glutamate and GABA (SuMvgat/vglut2) and another also expressing nitric oxide synthase (SuMNos1/Vglut2). Activation of SuMvgat/vglut2 neurons produces minimal wake and optogenetic stimulation of SuMvgat/vglut2 terminals elicits monosynaptic release of both glutamate and GABA onto dentate granule cells. Activation of SuMNos1/Vglut2 neurons potently drives wakefulness, whereas inhibition reduces REM sleep theta activity. These results identify SuMvglut2 neurons as a key node of the wake-sleep regulatory system.


Subject(s)
Arousal/physiology , Glutamic Acid/physiology , Hypothalamus, Posterior/physiology , Neurons/physiology , Animals , Hypothalamus, Posterior/cytology , Male , Mice , Mice, Knockout , Mice, Transgenic , Nitric Oxide Synthase Type I/physiology , Sleep, REM/physiology , Theta Rhythm/physiology , Vesicular Glutamate Transport Protein 2/deficiency , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Protein 2/physiology , Vesicular Inhibitory Amino Acid Transport Proteins/physiology , Wakefulness/physiology
7.
Neuron ; 95(5): 1074-1088.e7, 2017 Aug 30.
Article in English | MEDLINE | ID: mdl-28823729

ABSTRACT

The ability of presynaptic dopamine terminals to tune neurotransmitter release to meet the demands of neuronal activity is critical to neurotransmission. Although vesicle content has been assumed to be static, in vitro data increasingly suggest that cell activity modulates vesicle content. Here, we use a coordinated genetic, pharmacological, and imaging approach in Drosophila to study the presynaptic machinery responsible for these vesicular processes in vivo. We show that cell depolarization increases synaptic vesicle dopamine content prior to release via vesicular hyperacidification. This depolarization-induced hyperacidification is mediated by the vesicular glutamate transporter (VGLUT). Remarkably, both depolarization-induced dopamine vesicle hyperacidification and its dependence on VGLUT2 are seen in ventral midbrain dopamine neurons in the mouse. Together, these data suggest that in response to depolarization, dopamine vesicles utilize a cascade of vesicular transporters to dynamically increase the vesicular pH gradient, thereby increasing dopamine vesicle content.


Subject(s)
Dopamine/metabolism , Neurons/metabolism , Synaptic Vesicles/metabolism , Vesicular Glutamate Transport Protein 2/physiology , Animals , Animals, Genetically Modified , Dextroamphetamine/pharmacology , Drosophila , Drosophila Proteins/metabolism , Hydrogen-Ion Concentration , Locomotion/drug effects , Mesencephalon/metabolism , Mice , Neurons/physiology , Presynaptic Terminals/metabolism , Vesicular Glutamate Transport Protein 2/genetics
8.
J Neurosci ; 35(43): 14533-43, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-26511244

ABSTRACT

Circulating estradiol exerts a profound influence on the activity of the gonadotropin-releasing hormone (GnRH) neuronal network controlling fertility. Using genetic strategies enabling neuron-specific deletion of estrogen receptor α (Esr1), we examine here whether estradiol-modulated GABA and glutamate transmission are critical for the functioning of the GnRH neuron network in the female mouse. Using Vgat- and Vglut2-ires-Cre knock-in mice and ESR1 immunohistochemistry, we demonstrate that subpopulations of GABA and glutamate neurons throughout the limbic forebrain express ESR1, with ESR1-GABAergic neurons being more widespread and numerous than ESR1-glutamatergic neurons. We crossed Vgat- and Vglut2-ires-Cre mice with an Esr1(lox/lox) line to generate animals with GABA-neuron-specific or glutamate-neuron-specific deletion of Esr1. Vgat-ires-Cre;Esr1(lox/lox) mice were infertile, with abnormal estrous cycles, and exhibited a complete failure of the estrogen positive feedback mechanism responsible for the preovulatory GnRH surge. However, puberty onset and estrogen negative feedback were normal. Vglut2-ires-Cre;Esr1(lox/lox) mice were also infertile but displayed a wider range of deficits, including advanced puberty onset, abnormal negative feedback, and abolished positive feedback. Whereas <25% of preoptic kisspeptin neurons expressed Cre in Vgat- and Vglut2-ires-Cre lines, ∼70% of arcuate kisspeptin neurons were targeted in Vglut2-ires-Cre;Esr1(lox/lox) mice, possibly contributing to their advanced puberty phenotype. These observations show that, unexpectedly, ESR1-GABA neurons are only essential for the positive feedback mechanism. In contrast, we reveal the key importance of ESR1 in glutamatergic neurons for multiple estrogen feedback loops within the GnRH neuronal network required for fertility in the female mouse.


Subject(s)
Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/physiology , Estrogens/physiology , Feedback, Physiological/physiology , Fertility/genetics , Fertility/physiology , Glutamates/physiology , Neurons/metabolism , Sexual Maturation/genetics , Sexual Maturation/physiology , gamma-Aminobutyric Acid/physiology , Animals , Estrogen Receptor alpha/biosynthesis , Estrous Cycle/genetics , Estrous Cycle/physiology , Female , Follicular Phase/genetics , Follicular Phase/physiology , Gene Knock-In Techniques , Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/metabolism , Kisspeptins/physiology , Limbic System/metabolism , Mice , Prosencephalon/metabolism , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Protein 2/physiology , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
9.
Eur J Neurosci ; 42(6): 2271-82, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26096172

ABSTRACT

The retrotrapezoid nucleus (RTN) is a bilateral cluster of neurons located at the ventral surface of the brainstem below the facial nucleus. The RTN is activated by hypercapnia and stabilises arterial Pco2 by adjusting lung ventilation in a feedback manner. RTN neurons contain vesicular glutamate transporter-2 (Vglut2) transcripts (Slc17a6), and their synaptic boutons are Vglut2-immunoreactive. Here, we used optogenetics to test whether the RTN increases ventilation in conscious adult mice by releasing glutamate. Neurons located below the facial motor nucleus were transduced unilaterally to express channelrhodopsin-2 (ChR2)-enhanced yellow fluorescent protein, with lentiviral vectors that employ the Phox2b-activated artificial promoter PRSx8. The targeted population consisted of two types of Phox2b-expressing neuron: non-catecholaminergic neurons (putative RTN chemoreceptors) and catecholaminergic (C1) neurons. Opto-activation of a mix of ChR2-expressing RTN and C1 neurons produced a powerful stimulus frequency-dependent (5-15 Hz) stimulation of breathing in control conscious mice. Respiratory stimulation was comparable in mice in which dopamine-ß-hydroxylase (DßH)-positive neurons no longer expressed Vglut2 (DßH(C) (re/0);;Vglut2(fl/fl)). In a third group of mice, i.e. DßH(+/+);;Vglut2(fl/fl) mice, we injected a mixture of PRSx8-Cre lentiviral vector and Cre-dependent ChR2 adeno-associated virus 2 unilaterally into the RTN; this procedure deleted Vglut2 from ChR2-expressing neurons regardless of whether or not they were catecholaminergic. The ventilatory response elicited by photostimulation of ChR2-positive neurons was almost completely absent in these mice. Resting ventilatory parameters were identical in the three groups of mice, and their brains contained similar numbers of ChR2-positive catecholaminergic and non-catecholaminergic neurons. From these results, we conclude that RTN neurons increase breathing in conscious adult mice by releasing glutamate.


Subject(s)
Glutamic Acid/physiology , Medulla Oblongata/physiology , Neurons/physiology , Respiration , Vesicular Glutamate Transport Protein 2/physiology , Animals , Catecholamines/physiology , Chemoreceptor Cells/metabolism , Chemoreceptor Cells/physiology , Female , Male , Medulla Oblongata/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Optogenetics , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Protein 2/metabolism
10.
J Neurosci ; 34(42): 13906-10, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25319687

ABSTRACT

The ventral tegmental area (VTA) plays roles in both reward and aversion. The participation of VTA in diverse behaviors likely reflects its heterogeneous neuronal phenotypes and circuits. Recent findings indicate that VTA GABAergic neurons that coexpress tyrosine hydroxylase (TH) projecting to lateral habenula (LHb) play a role in reward. In addition to these mesohabenular TH-GABAergic neurons, the VTA has many neurons expressing vesicular glutamate transporter 2 (VGluT2) that also project to LHb. To determine the behavioral role of mesohabenular VGluT2 neurons, we targeted channelrhodopsin2 to VTA VGluT2 neurons of VGluT2::Cre mice. These mice were tested in an apparatus where moving into one chamber stimulated VTA VGluT2 projections within the LHb, and exiting the chamber inactivated the stimulation. We found that mice spent significantly less time in the chamber where VGluT2 mesohabenular fiber stimulation occurred. Mice that received injections of mixed AMPA and NMDA glutamate receptor antagonists in LHb were unresponsive to VGluT2-mesohabenular fiber stimulation, demonstrating the participation of LHb glutamate receptors in mesohabenular stimulation-elicited aversion. In the absence of light stimulation, mice showed a conditioned place aversion to the chamber that was previously associated with VGluT2-mesohabenular fiber stimulation. We conclude that there is a glutamatergic signal from VTA VGluT2-mesohabenular neurons that plays a role in aversion by activating LHb glutamatergic receptors.


Subject(s)
Avoidance Learning/physiology , Conditioning, Psychological/physiology , Glutamic Acid/physiology , Habenula/physiology , Ventral Tegmental Area/physiology , Vesicular Glutamate Transport Protein 2/physiology , Animals , Avoidance Learning/drug effects , Conditioning, Psychological/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Habenula/drug effects , Male , Mice , Mice, Transgenic , Neural Pathways/drug effects , Neural Pathways/physiology , Ventral Tegmental Area/drug effects , Vesicular Glutamate Transport Protein 2/antagonists & inhibitors
11.
Mol Pharmacol ; 85(2): 322-34, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24275230

ABSTRACT

The transient receptor potential cation channel subfamily V member 1 (TRPV1) is known as a thermosensor and integrator of inflammation-induced hyperalgesia. TRPV1 is expressed in a subpopulation of primary afferent neurons that express several different neurotransmitters. The role of the TRPV1 channel in the development of hyperalgesia is established, but the role of the neurotransmitter glutamate, used partially by the same neuronal population and thus probably mediating the response, is still under investigation. We have used a Trpv1-Cre mouse line in which we either ablated Trpv1-Cre expressing neurons or induced vesicular glutamate transporter 2 (Vglut2) deficiency in Trpv1-Cre expressing neurons and investigated specific states of hyperalgesia after persistent inflammation. Furthermore, by pharmacologic inhibition of substance P (SP) or calcitonin gene-related peptide (CGRP) signaling in Vglut2-deficient mice, we also evaluated the contribution of SP or CGRP to inflammation-induced hyperalgesia, with or without the presence of vesicular glutamate transporter 2 (VGLUT2)-mediated glutamatergic transmission in Trpv1-Cre neurons. This examination, together with c-Fos analyses, showed that VGLUT2-mediated glutamatergic transmission in Trpv1-Cre afferents together with SP or CGRP is essential for the development of the heat hyperalgesia associated with persistent inflammation. Additionally, SP-, CGRP-, and VGLUT2-mediated transmission together were found to play a role in the development of mechanical hyperalgesia after persistent inflammation.


Subject(s)
Calcitonin Gene-Related Peptide/physiology , Glutamic Acid/physiology , Hyperalgesia/etiology , Inflammation/complications , Substance P/physiology , Androstanes/pharmacology , Animals , Benzimidazoles/pharmacology , Female , Hot Temperature , Male , Mice , Nerve Growth Factor/physiology , Neurons, Afferent/physiology , Piperazines/pharmacology , Quinazolines/pharmacology , TRPV Cation Channels/physiology , Vesicular Glutamate Transport Protein 2/physiology
12.
Neuron ; 80(4): 920-33, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24267650

ABSTRACT

Locomotion is controlled by spinal networks that generate rhythm and coordinate left-right and flexor-extensor patterning. Defined populations of spinal interneurons have been linked to patterning circuits; however, neurons comprising the rhythm-generating kernel have remained elusive. Here, we identify an ipsilaterally projecting excitatory interneuron population, marked by the expression of Shox2 that overlaps partially with V2a interneurons. Optogenetic silencing or blocking synaptic output of Shox2 interneurons (INs) in transgenic mice perturbed rhythm without an effect on pattern generation, whereas ablation of the Shox2 IN subset coinciding with the V2a population was without effect. Most Shox2 INs are rhythmically active during locomotion and analysis of synaptic connectivity showed that Shox2 INs contact other Shox2 INs, commissural neurons, and motor neurons, with preference for flexor motor neurons. Our findings focus attention on a subset of Shox2 INs that appear to participate in the rhythm-generating kernel for spinal locomotion.


Subject(s)
Homeodomain Proteins/physiology , Interneurons/physiology , Locomotion/physiology , Animals , Axons/physiology , Dependovirus/genetics , Electrophysiological Phenomena , Excitatory Amino Acid Agonists/pharmacology , Gene Silencing , Glutamic Acid/physiology , Immunohistochemistry , In Situ Hybridization , Locomotion/drug effects , Male , Mice , Motor Neurons/physiology , N-Methylaspartate/pharmacology , Neural Pathways/physiology , Optogenetics , Serotonin/pharmacology , Spinal Cord/cytology , Spinal Cord/physiology , Vesicular Glutamate Transport Protein 2/physiology
13.
J Neurosci ; 33(26): 10647-60, 2013 Jun 26.
Article in English | MEDLINE | ID: mdl-23804088

ABSTRACT

The vesicular glutamate transporters (VGLUTs) package glutamate into synaptic vesicles, and the two principal isoforms VGLUT1 and VGLUT2 have been suggested to influence the properties of release. To understand how a VGLUT isoform might influence transmitter release, we have studied their trafficking and previously identified a dileucine-like endocytic motif in the C terminus of VGLUT1. Disruption of this motif impairs the activity-dependent recycling of VGLUT1, but does not eliminate its endocytosis. We now report the identification of two additional dileucine-like motifs in the N terminus of VGLUT1 that are not well conserved in the other isoforms. In the absence of all three motifs, rat VGLUT1 shows limited accumulation at synaptic sites and no longer responds to stimulation. In addition, shRNA-mediated knockdown of clathrin adaptor proteins AP-1 and AP-2 shows that the C-terminal motif acts largely via AP-2, whereas the N-terminal motifs use AP-1. Without the C-terminal motif, knockdown of AP-1 reduces the proportion of VGLUT1 that responds to stimulation. VGLUT1 thus contains multiple sorting signals that engage distinct trafficking mechanisms. In contrast to VGLUT1, the trafficking of VGLUT2 depends almost entirely on the conserved C-terminal dileucine-like motif: without this motif, a substantial fraction of VGLUT2 redistributes to the plasma membrane and the transporter's synaptic localization is disrupted. Consistent with these differences in trafficking signals, wild-type VGLUT1 and VGLUT2 differ in their response to stimulation.


Subject(s)
Leucine/genetics , Leucine/physiology , Vesicular Glutamate Transport Protein 1/physiology , Adaptor Protein Complex 2/metabolism , Amino Acid Motifs/physiology , Amino Acid Sequence , Animals , Cells, Cultured , Clathrin/metabolism , Endocytosis/physiology , Hippocampus/cytology , Hippocampus/metabolism , Hydrogen-Ion Concentration , Immunohistochemistry , Molecular Sequence Data , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/physiology , Polymerase Chain Reaction , RNA Interference , Rats , Vesicular Glutamate Transport Protein 1/genetics , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Protein 2/physiology
14.
J Neurosci ; 33(2): 734-47, 2013 Jan 09.
Article in English | MEDLINE | ID: mdl-23303951

ABSTRACT

Identification of the neural pathways involved in retraining the spinal central pattern generators (CPGs) by afferent input in the absence of descending supraspinal control is feasible in isolated rodent spinal cords where the locomotor CPGs are potently activated by sacrocaudal afferent (SCA) input. Here we study the involvement of sacral neurons projecting rostrally through the ventral funiculi (VF) in activation of the CPGs by sensory stimulation. Fluorescent labeling and immunostaining showed that VF neurons are innervated by primary afferents immunoreactive for vesicular glutamate transporters 1 and 2 and by intraspinal neurons. Calcium imaging revealed that 55% of the VF neurons were activated by SCA stimulation. The activity of VF neurons and the sacral and lumbar CPGs was abolished when non-NMDA receptors in the sacral segments were blocked by the antagonist CNQX. When sacral NMDA receptors were blocked by APV, the sacral CPGs were suppressed, VF neurons with nonrhythmic activity were recruited and a moderate-drive locomotor rhythm developed during SCA stimulation. In contrast, when the sacral CPGs were activated by SCA stimulation, rhythmic and nonrhythmic VF neurons were recruited and the locomotor rhythm was most powerful. The activity of 73 and 27% of the rhythmic VF neurons was in-phase with the ipsilateral and contralateral motor output, respectively. Collectively, our studies indicate that sacral VF neurons serve as a major link between SCA and the hindlimb CPGs and that the ability of SCA to induce stepping can be enhanced by the sacral CPGs. The nature of the ascending drive to lumbar CPGs, the identity of subpopulations of VF neurons, and their potential role in activating the locomotor rhythm are discussed.


Subject(s)
Afferent Pathways/physiology , Interneurons/physiology , Locomotion/physiology , Spinal Cord/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Afferent Pathways/cytology , Afferent Pathways/drug effects , Animals , Calcium/physiology , Data Interpretation, Statistical , Electric Stimulation , Electrodes , Excitatory Amino Acid Antagonists/pharmacology , Female , Fluorescent Antibody Technique , Functional Laterality/drug effects , Functional Laterality/physiology , Glutamic Acid/physiology , Hindlimb/innervation , Hindlimb/physiology , Immunohistochemistry , Interneurons/drug effects , Locomotion/drug effects , Male , Microscopy, Fluorescence , Rats , Receptors, N-Methyl-D-Aspartate/drug effects , Sacrococcygeal Region/physiology , Spinal Cord/cytology , Spinal Cord/drug effects , Vesicular Glutamate Transport Protein 1/physiology , Vesicular Glutamate Transport Protein 2/physiology
15.
PLoS One ; 8(12): e83974, 2013.
Article in English | MEDLINE | ID: mdl-24391855

ABSTRACT

Melanopsin-expressing retinal ganglion cells (mRGCs) in the eye play an important role in many light-activated non-image-forming functions including neonatal photoaversion and the adult pupillary light reflex (PLR). MRGCs rely on glutamate and possibly PACAP (pituitary adenylate cyclase-activating polypeptide) to relay visual signals to the brain. However, the role of these neurotransmitters for individual non-image-forming responses remains poorly understood. To clarify the role of glutamatergic signaling from mRGCs in neonatal aversion to light and in adult PLR, we conditionally deleted vesicular glutamate transporter (VGLUT2) selectively from mRGCs in mice. We found that deletion of VGLUT2 in mRGCs abolished negative phototaxis and light-induced distress vocalizations in neonatal mice, underscoring a necessary role for glutamatergic signaling. In adult mice, loss of VGLUT2 in mRGCs resulted in a slow and an incomplete PLR. We conclude that glutamatergic neurotransmission from mRGCs is required for neonatal photoaversion but is complemented by another non-glutamatergic signaling mechanism for the pupillary light reflex in adult mice. We speculate that this complementary signaling might be due to PACAP neurotransmission from mRGCs.


Subject(s)
Light , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Reflex, Pupillary/physiology , Retinal Ganglion Cells/metabolism , Rod Opsins/physiology , Synaptic Transmission/physiology , Vesicular Glutamate Transport Protein 2/physiology , Animals , Animals, Newborn , Behavior, Animal , Female , Immunoenzyme Techniques , Integrases/metabolism , Light Signal Transduction , Male , Mice , Mice, Knockout , Neurotransmitter Agents/metabolism , Photic Stimulation , Reflex, Pupillary/radiation effects , Retinal Ganglion Cells/radiation effects , Vision Disorders , Vision, Ocular/physiology , Vision, Ocular/radiation effects
16.
J Comp Neurol ; 521(6): 1354-77, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23047588

ABSTRACT

We examined thalamic input to striatum in rats using immunolabeling for the vesicular glutamate transporter (VGLUT2). Double immunofluorescence viewed with confocal laser scanning microscopy (CLSM) revealed that VGLUT2+ terminals are distinct from VGLUT1+ terminals. CLSM of Phaseolus vulgaris-leucoagglutinin (PHAL)-labeled cortical or thalamic terminals revealed that VGLUT2 is rare in corticostriatal terminals but nearly always present in thalamostriatal terminals. Electron microscopy revealed that VGLUT2+ terminals made up 39.4% of excitatory terminals in striatum (with VGLUT1+ corticostriatal terminals constituting the rest), and 66.8% of VGLUT2+ terminals synapsed on spines and the remainder on dendrites. VGLUT2+ axospinous terminals had a mean diameter of 0.624 µm, while VGLUT2+ axodendritic terminals a mean diameter of 0.698 µm. In tissue in which we simultaneously immunolabeled thalamostriatal terminals for VGLUT2 and striatal neurons for D1 (with about half of spines immunolabeled for D1), 54.6% of VGLUT2+ terminals targeted D1+ spines (i.e., direct pathway striatal neurons), and 37.3% of D1+ spines received VGLUT2+ synaptic contacts. By contrast, 45.4% of VGLUT2+ terminals targeted D1-negative spines (i.e., indirect pathway striatal neurons), and only 25.8% of D1-negative spines received VGLUT2+ synaptic contacts. Similarly, among VGLUT2+ axodendritic synaptic terminals, 59.1% contacted D1+ dendrites, and 40.9% contacted D1-negative dendrites. VGLUT2+ terminals on D1+ spines and dendrites tended to be slightly smaller than those on D1-negative spines and dendrites. Thus, thalamostriatal terminals contact both direct and indirect pathway striatal neurons, with a slight preference for direct. These results are consistent with physiological studies indicating slightly different effects of thalamic input on the two types of striatal projection neurons.


Subject(s)
Corpus Striatum/ultrastructure , Neurons/ultrastructure , Thalamus/ultrastructure , Vesicular Glutamate Transport Protein 2/ultrastructure , Animals , Corpus Striatum/chemistry , Corpus Striatum/cytology , Excitatory Postsynaptic Potentials/physiology , Male , Microscopy, Confocal/methods , Neural Pathways/chemistry , Neural Pathways/cytology , Neural Pathways/ultrastructure , Neurons/chemistry , Rats , Rats, Sprague-Dawley , Thalamus/chemistry , Thalamus/cytology , Vesicular Glutamate Transport Protein 2/physiology
17.
Adv Exp Med Biol ; 758: 115-22, 2012.
Article in English | MEDLINE | ID: mdl-23080151

ABSTRACT

The retrotrapezoid nucleus (RTN) is located in the rostral medulla oblongata close to the ventral surface and consists of a bilateral cluster of glutamatergic neurons that are non-aminergic and express homeodomain transcription factor Phox2b throughout life. These neurons respond vigorously to increases in local pCO(2) via cell-autonomous and paracrine (glial) mechanisms and receive additional chemosensory information from the carotid bodies. RTN neurons exclusively innervate the regions of the brainstem that contain the respiratory pattern generator (RPG). Lesion or inhibition of RTN neurons largely attenuates the respiratory chemoreflex of adult rats whereas their activation increases respiratory rate, inspiratory amplitude and active expiration. Phox2b mutations that cause congenital central hypoventilation syndrome in humans prevent the development of RTN neurons in mice. Selective deletion of the RTN Phox2b-VGLUT2 neurons by genetic means in mice eliminates the respiratory chemoreflex in neonates.In short, RTN Phox2b-VGLUT2 neurons are a major nodal point of the CNS network that regulates pCO(2) via breathing and these cells are probable central chemoreceptors.


Subject(s)
Chemoreceptor Cells/physiology , Medulla Oblongata/physiology , Respiration , Animals , Carbon Dioxide/metabolism , Homeodomain Proteins/physiology , Humans , Reflex , Transcription Factors/physiology , Vesicular Glutamate Transport Protein 2/physiology
18.
Neuroscience ; 226: 253-69, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-22989920

ABSTRACT

Spinocerebellar tract neurons are inhibited by various sources of input via pathways activated by descending tracts as well as peripheral afferents. Inhibition may be used to modulate transmission of excitatory information forwarded to the cerebellum. However it may also provide information on the degree of inhibition of motoneurons and on the operation of inhibitory premotor neurons. Our aim was to extend previous comparisons of morphological substrates of excitation of spinocerebellar neurons to inhibitory input. Contacts formed by inhibitory axon terminals were characterised as either GABAergic, glycinergic or both GABAergic/glycinergic by using antibodies against vesicular GABA transporter, glutamic acid decarboxylase and gephyrin. Quantitative analysis revealed the presence of much higher proportions of inhibitory contacts when compared with excitatory contacts on spinal border (SB) neurons. However similar proportions of inhibitory and excitatory contacts were associated with ventral spinocerebellar tract (VSCT) and dorsal spinocerebellar tract neurons located in Clarke's column (ccDSCT) and the dorsal horn (dhDSCT). In all of the cells, the majority of inhibitory terminals were glycinergic. The density of contacts was higher on somata and proximal versus distal dendrites of SB and VSCT neurons but more evenly distributed in ccDSCT and dhDSCT neurons. Variations in the density and distribution of inhibitory contacts found in this study may reflect differences in information on inhibitory processes forwarded by subtypes of spinocerebellar tract neurons to the cerebellum.


Subject(s)
Neurons/physiology , Spinal Cord/physiology , Spinocerebellar Tracts/physiology , Animals , Cats , Electric Stimulation , Glutamic Acid/physiology , Glycine/physiology , Image Processing, Computer-Assisted , Immunohistochemistry , Microscopy, Confocal , Nerve Endings/physiology , Peripheral Nerves/physiology , Vesicular Glutamate Transport Protein 1/physiology , Vesicular Glutamate Transport Protein 2/physiology , Vesicular Inhibitory Amino Acid Transport Proteins/physiology , gamma-Aminobutyric Acid/physiology
19.
Nat Neurosci ; 15(9): 1192-4, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22885848

ABSTRACT

We found that, during the formation of the mouse barrel cortex, NG2 cells received glutamatergic synapses from thalamocortical fibers and preferentially accumulated along septa separating the barrels. Sensory deprivation reduced thalamocortical inputs on NG2 cells and increased their proliferation, leading to a more uniform distribution in the deprived barrels. Thus, early sensory experience regulates thalamocortical innervation on NG2 cells, as well as their proliferation and distribution during development.


Subject(s)
Neural Stem Cells/physiology , Somatosensory Cortex/physiology , Animals , Cell Count , Cell Proliferation , DNA-Binding Proteins , Darkness , Excitatory Postsynaptic Potentials/physiology , Glutamates/physiology , Immunohistochemistry , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Fluorescence , Nerve Fibers/physiology , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Oligodendroglia/physiology , Patch-Clamp Techniques , Thalamus/physiology , Vesicular Glutamate Transport Protein 2/physiology , Vibrissae/innervation , Vibrissae/physiology
20.
J Physiol ; 590(7): 1737-55, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22371473

ABSTRACT

The cerebellum receives information from the hindlimbs through several populations of spinocerebellar tract neurons. Although the role of these neurons has been established in electrophysiological experiments, the relative contribution of afferent fibres and central neurons to their excitatory input has only been estimated approximately so far. Taking advantage of differences in the immunohistochemistry of glutamatergic terminals of peripheral afferents and of central neurons (with vesicular glutamate transporters VGLUT1 or VGLUT2, respectively), we compared sources of excitatory input to four populations of spinocerebellar neurons in the thoraco-lumbar spinal cord: dorsal spinocerebellar tract neurons located in Clarke's column (ccDSCT) and in the dorsal horn (dhDSCT) and ventral spinocerebellar tract (VSCT) neurons including spinal border (SB) neurons. This was done on 22 electrophysiologically identified intracellularly labelled neurons in cats and on 80 neurons labelled by retrograde transport of cholera toxin b subunit injected into the cerebellum of rats. In both species distribution of antibodies against VGLUT1 and VGLUT2 on SB neurons (which have dominating inhibitory input from limb muscles), revealed very few VGLUT1 contacts and remarkably high numbers of VGLUT2 contacts. In VSCT neurons with excitatory afferent input, the number of VGLUT1 contacts was relatively high although VGLUT2 contacts likewise dominated, while the proportions of VGLUT1 and VGLUT2 immunoreactive terminals were the reverse on the two populations of DSCT neurons. These findings provide morphological evidence that SB neurons principally receive excitatory inputs from central neurons and provide the cerebellum with information regarding central neuronal activity.


Subject(s)
Movement/physiology , Neurons/physiology , Spinal Cord/physiology , Spinocerebellar Tracts/physiology , Animals , Cats , Electric Stimulation , Hindlimb/innervation , Hindlimb/physiology , Male , Rats , Rats, Sprague-Dawley , Spinal Cord/cytology , Spinocerebellar Tracts/cytology , Vesicular Glutamate Transport Protein 1/physiology , Vesicular Glutamate Transport Protein 2/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...