201
|
Abstract
UNLABELLED Recent evidence has resurrected the idea that the amino acid aspartate, a selective NMDA receptor agonist, is a neurotransmitter. Using a mouse that lacks the glutamate-selective vesicular transporter VGLUT1, we find that glutamate alone fully accounts for the activation of NMDA receptors at excitatory synapses in the hippocampus. This excludes a role for aspartate and, by extension, a recently proposed role for the sialic acid transporter sialin in excitatory transmission. SIGNIFICANCE STATEMENT It has been proposed that the amino acid aspartate serves as a neurotransmitter. Although aspartate is a selective agonist for NMDA receptors, we find that glutamate alone fully accounts for neurotransmission at excitatory synapses in the hippocampus, excluding a role for aspartate.
Collapse
|
202
|
Zimmermann J, Herman MA, Rosenmund C. Co-release of glutamate and GABA from single vesicles in GABAergic neurons exogenously expressing VGLUT3. Front Synaptic Neurosci 2015; 7:16. [PMID: 26441632 PMCID: PMC4585203 DOI: 10.3389/fnsyn.2015.00016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/04/2015] [Indexed: 12/17/2022] Open
Abstract
The identity of the vesicle neurotransmitter transporter expressed by a neuron largely corresponds with the primary neurotransmitter that cell releases. However, the vesicular glutamate transporter subtype 3 (VGLUT3) is mainly expressed in non-glutamatergic neurons, including cholinergic, serotonergic, or GABAergic neurons. Though a functional role for glutamate release from these non-glutamatergic neurons has been demonstrated, the interplay between VGLUT3 and the neuron’s characteristic neurotransmitter transporter, particularly in the case of GABAergic neurons, at the synaptic and vesicular level is less clear. In this study, we explore how exogenous expression of VGLUT3 in striatal GABAergic neurons affects the packaging and release of glutamate and GABA in synaptic vesicles (SVs). We found that VGLUT3 expression in isolated, autaptic GABAergic neurons leads to action potential evoked release of glutamate. Under these conditions, glutamate and GABA could be packaged together in single vesicles release either spontaneously or asynchronously. However, the presence of glutamate in GABAergic vesicles did not affect uptake of GABA itself, suggesting a lack of synergy in vesicle filling for these transmitters. Finally, we found postsynaptic detection of glutamate released from GABAergic terminals difficult when bona fide glutamatergic synapses were present, suggesting that co-released glutamate cannot induce postsynaptic glutamate receptor clustering.
Collapse
Affiliation(s)
- Johannes Zimmermann
- Neurowissenschaftliches Forschungszentrum (NWFZ), NeuroCure Exzellenzcluster, CCO Charité Universitätsmedizin Berlin, Germany
| | - Melissa A Herman
- Neurowissenschaftliches Forschungszentrum (NWFZ), NeuroCure Exzellenzcluster, CCO Charité Universitätsmedizin Berlin, Germany
| | - Christian Rosenmund
- Neurowissenschaftliches Forschungszentrum (NWFZ), NeuroCure Exzellenzcluster, CCO Charité Universitätsmedizin Berlin, Germany
| |
Collapse
|
203
|
Cheung CC, Krause WC, Edwards RH, Yang CF, Shah NM, Hnasko TS, Ingraham HA. Sex-dependent changes in metabolism and behavior, as well as reduced anxiety after eliminating ventromedial hypothalamus excitatory output. Mol Metab 2015; 4:857-66. [PMID: 26629409 PMCID: PMC4632173 DOI: 10.1016/j.molmet.2015.09.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 08/26/2015] [Accepted: 09/02/2015] [Indexed: 12/17/2022] Open
Abstract
Objectives The ventromedial hypothalamic nucleus (VMH) regulates energy homeostasis as well as social and emotional behaviors. Nearly all VMH neurons, including those in the sexually dimorphic ventrolateral VMH (VMHvl) subregion, release the excitatory neurotransmitter glutamate and use the vesicular glutamate transporter 2 (Vglut2). Here, we asked how glutamatergic signaling contributes to the collective metabolic and behavioral responses attributed to the VMH and VMHvl. Methods Using Sf1-Cre and a Vglut2 floxed allele, Vglut2 was knocked-out in SF-1 VMH neurons (Vglut2Sf1-Cre). Metabolic and neurobehavioral assays were carried out initially on Vglut2fl/fl and Vglut2Sf1-Cre mice in a mixed, and then in the C57BL/6 genetic background, which is prone to hyperglycemia and diet induced obesity (DIO). Results Several phenotypes observed in Vglut2Sf1-Cre mice were largely unexpected based on prior studies that have perturbed VMH development or VMH glutamate signaling. In our hands, Vglut2Sf1-Cre mice failed to exhibit the anticipated increase in body weight after high fat diet (HFD) or the impaired glucose homeostasis after fasting. Instead, there was a significant sex-dependent attenuation of DIO in Vglut2Sf1-Cre females. Vglut2Sf1-Cre males also display a sex-specific loss of conditioned-fear responses and aggression accompanied by more novelty-associated locomotion. Finally, unlike the higher anxiety noted in Sf1Nestin-Cre mice that lack a fully formed VMH, both male and female Vglut2Sf1-Cre mice were less anxious. Conclusions Loss of VMH glutamatergic signaling sharply decreased DIO in females, attenuated aggression and learned fear in males, and was anxiolytic in males and females. Collectively, our findings demonstrate that while glutamatergic output from the VMH appears largely dispensable for counter regulatory responses to hypoglycemia, it drives sex-dependent differences in metabolism and social behaviors and is essential for adaptive responses to anxiety-provoking stimuli in both sexes. Excitatory VMH output controls sex-dependent metabolic and behavioral phenotypes. Vglut2Sf1-Cre mice are not prone to diet-induced obesity or glucose misregulation. Loss of VMH glutamatergic signaling leads to negative energy state in females. Aggression and learned fear are lower in males lacking VMH excitatory output. VMH glutamatergic signaling drives normal anxiety responses in both sexes.
Collapse
Affiliation(s)
- Clement C Cheung
- Department of Cellular and Molecular Pharmacology, Mission Bay Campus, University of California, San Francisco 94143, United States
| | - William C Krause
- Department of Cellular and Molecular Pharmacology, Mission Bay Campus, University of California, San Francisco 94143, United States
| | - Robert H Edwards
- Department of Physiology and Neurology, Mission Bay Campus, University of California, San Francisco 94143, United States
| | - Cindy F Yang
- Department of Anatomy, Mission Bay Campus, University of California, San Francisco 94143, United States
| | - Nirao M Shah
- Department of Anatomy, Mission Bay Campus, University of California, San Francisco 94143, United States
| | - Thomas S Hnasko
- Department of Physiology and Neurology, Mission Bay Campus, University of California, San Francisco 94143, United States
| | - Holly A Ingraham
- Department of Cellular and Molecular Pharmacology, Mission Bay Campus, University of California, San Francisco 94143, United States
| |
Collapse
|
204
|
Rooney KE, Wallace LJ. Computational modeling of extracellular dopamine kinetics suggests low probability of neurotransmitter release. Synapse 2015; 69:515-25. [PMID: 26248886 DOI: 10.1002/syn.21845] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 06/25/2015] [Accepted: 07/11/2015] [Indexed: 02/03/2023]
Abstract
Dopamine in the striatum signals the saliency of current environmental input and is involved in learned formation of appropriate responses. The regular baseline-firing rate of dopaminergic neurons suggests that baseline dopamine is essential for proper brain function. The first goal of the study was to estimate the likelihood of full exocytotic dopamine release associated with each firing event under baseline conditions. A computer model of extracellular space associated with a single varicosity was developed using the program MCell to estimate kinetics of extracellular dopamine. Because the literature provides multiple kinetic values for dopamine uptake depending on the system tested, simulations were run using different kinetic parameters. With all sets of kinetic parameters evaluated, at most, 25% of a single vesicle per varicosity would need to be released per firing event to maintain a 5-10 nM extracellular dopamine concentration, the level reported by multiple microdialysis experiments. The second goal was to estimate the fraction of total amount of stored dopamine released during a highly stimulated condition. This was done using the same model system to simulate published measurements of extracellular dopamine following electrical stimulation of striatal slices in vitro. The results suggest the amount of dopamine release induced by a single electrical stimulation may be as large as the contents of two vesicles per varicosity. We conclude that dopamine release probability at any particular varicosity is low. This suggests that factors capable of increasing release probability could have a powerful effect on sculpting dopamine signals.
Collapse
Affiliation(s)
- Katherine E Rooney
- Division of Pharmacology, College of Pharmacy, the Ohio State University, 500 W. 12th Avenue Columbus, Ohio, 43210
| | - Lane J Wallace
- Division of Pharmacology, College of Pharmacy, the Ohio State University, 500 W. 12th Avenue Columbus, Ohio, 43210.,500 West 12th Avenue Columbus, Ohio, 43210
| |
Collapse
|
205
|
Girasole AE, Nelson AB. Probing striatal microcircuitry to understand the functional role of cholinergic interneurons. Mov Disord 2015; 30:1306-18. [PMID: 26227561 DOI: 10.1002/mds.26340] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 06/12/2015] [Accepted: 06/21/2015] [Indexed: 12/23/2022] Open
Affiliation(s)
- Allison E Girasole
- Department of Neurology, University of California, San Francisco, USA.,Neuroscience Graduate Program, University of California, San Francisco, USA
| | - Alexandra B Nelson
- Department of Neurology, University of California, San Francisco, USA.,Neuroscience Graduate Program, University of California, San Francisco, USA
| |
Collapse
|
206
|
Holloway BB, Viar KE, Stornetta RL, Guyenet PG. The retrotrapezoid nucleus stimulates breathing by releasing glutamate in adult conscious mice. Eur J Neurosci 2015; 42:2271-82. [PMID: 26096172 DOI: 10.1111/ejn.12996] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/10/2015] [Accepted: 06/17/2015] [Indexed: 11/30/2022]
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-β-hydroxylase (DβH)-positive neurons no longer expressed Vglut2 (DβH(C) (re/0);;Vglut2(fl/fl)). In a third group of mice, i.e. DβH(+/+);;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.
Collapse
Affiliation(s)
- Benjamin B Holloway
- Department of Pharmacology, University of Virginia Health System, P.O. Box 800735, 1340 Jefferson Park Avenue, Charlottesville, VA, 22908-0735, USA
| | - Kenneth E Viar
- Department of Pharmacology, University of Virginia Health System, P.O. Box 800735, 1340 Jefferson Park Avenue, Charlottesville, VA, 22908-0735, USA
| | - Ruth L Stornetta
- Department of Pharmacology, University of Virginia Health System, P.O. Box 800735, 1340 Jefferson Park Avenue, Charlottesville, VA, 22908-0735, USA
| | - Patrice G Guyenet
- Department of Pharmacology, University of Virginia Health System, P.O. Box 800735, 1340 Jefferson Park Avenue, Charlottesville, VA, 22908-0735, USA
| |
Collapse
|
207
|
Fyk-Kolodziej BE, Shimano T, Gafoor D, Mirza N, Griffith RD, Gong TW, Holt AG. Dopamine in the auditory brainstem and midbrain: co-localization with amino acid neurotransmitters and gene expression following cochlear trauma. Front Neuroanat 2015; 9:88. [PMID: 26257610 PMCID: PMC4510424 DOI: 10.3389/fnana.2015.00088] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 06/19/2015] [Indexed: 11/13/2022] Open
Abstract
Dopamine (DA) modulates the effects of amino acid neurotransmitters (AANs), including GABA and glutamate, in motor, visual, olfactory, and reward systems (Hnasko et al., 2010; Stuber et al., 2010; Hnasko and Edwards, 2012). The results suggest that DA may play a similar modulatory role in the auditory pathways. Previous studies have shown that deafness results in decreased GABA release, changes in excitatory neurotransmitter levels, and increased spontaneous neuronal activity within brainstem regions related to auditory function. Modulation of the expression and localization of tyrosine hydroxylase (TH; the rate limiting enzyme in the production of DA) in the IC following cochlear trauma has been previously reported (Tong et al., 2005). In the current study the possibility of co-localization of TH with AANs was examined. Changes in the gene expression of TH were compared with changes in the gene expression of markers for AANs in the cochlear nucleus (CN) and inferior colliculus (IC) to determine whether those deafness related changes occur concurrently. The results indicate that bilateral cochlear ablation significantly reduced TH gene expression in the CN after 2 months while in the IC the reduction in TH was observed at both 3 days and 2 months following ablation. Furthermore, in the CN, glycine transporter 2 (GLYT2) and the GABA transporter (GABAtp) were also significantly reduced only after 2 months. However, in the IC, DA receptor 1 (DRDA1), vesicular glutamate transporters 2 and 3 (VGLUT2, VGLUT3), GABAtp and GAD67 were reduced in expression both at the 3 days and 2 months time points. A close relationship between the distribution of TH and several of the AANs was determined in both the CN and the IC. In addition, GLYT2 and VGLUT3 each co-localized with TH within IC somata and dendrites. Therefore, the results of the current study suggest that DA is spatially well positioned to influence the effects of AANs on auditory neurons.
Collapse
Affiliation(s)
- Bozena E Fyk-Kolodziej
- Molecular Anatomy of Auditory-related Central Systems, Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit MI, USA
| | - Takashi Shimano
- Department of Otolaryngology, Kansai Medical University Osaka, Japan
| | - Dana Gafoor
- Molecular Anatomy of Auditory-related Central Systems, Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit MI, USA
| | - Najab Mirza
- Molecular Anatomy of Auditory-related Central Systems, Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit MI, USA
| | - Ronald D Griffith
- Molecular Anatomy of Auditory-related Central Systems, Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit MI, USA
| | - Tzy-Wen Gong
- Kresge Hearing Research Institute, University of Michigan School of Medicine, Ann Arbor MI, USA
| | - Avril Genene Holt
- Molecular Anatomy of Auditory-related Central Systems, Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit MI, USA
| |
Collapse
|
208
|
Stensrud MJ, Sogn CJ, Gundersen V. Immunogold characteristics of VGLUT3-positive GABAergic nerve terminals suggest corelease of glutamate. J Comp Neurol 2015; 523:2698-713. [PMID: 26010578 DOI: 10.1002/cne.23811] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 05/18/2015] [Accepted: 05/18/2015] [Indexed: 12/20/2022]
Abstract
There is compelling evidence that glutamate can act as a cotransmitter in the mammalian brain. Interestingly, the third vesicular glutamate transporter (VGLUT3) is primarily found in neurons that were anticipated to be nonglutamatergic. Whereas the function of VGLUT3 in acetylcholinergic and serotoninergic neurons has been elucidated, the role of VGLUT3 in neurons releasing gamma-aminobutyric acid (GABA) is not settled. We have previously shown that VGLUT3 is found together with the vesicular GABA transporter (VIAAT) on synaptic vesicle membranes in the hippocampus. Now we provide novel electron microscopic data from the rat hippocampus suggesting that glutamate is enriched in inhibitory nerve terminals containing VGLUT3 compared to those lacking VGLUT3. The opposite was found for GABA; VGLUT3-positive inhibitory terminals contained lower density of GABA labeling compared to VGLUT3-negative inhibitory terminals. In addition, semiquantitative confocal immunofluorescence showed that N-methyl-D-aspartate (NMDA)-receptor labeling was present more frequently in VGLUT3-positive/VIAAT-positive synapses versus in VGLUT3-negative/VIAAT-positive synapses. Electron microscopic immunogold data further suggest that NMDA receptors are enriched in VGLUT3 containing inhibitory terminals. Our data reveal new chemical characteristics of a subset of GABAergic interneurons in the hippocampus. The analyses suggest that glutamate is coreleased with GABA from hippocampal basket cell-synapses to act on NMDA receptors.
Collapse
Affiliation(s)
- Mats Julius Stensrud
- Department of Anatomy and Healthy Brain Ageing Centre Regional Research Network, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Carl Johan Sogn
- Department of Anatomy and Healthy Brain Ageing Centre Regional Research Network, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Vidar Gundersen
- Department of Anatomy and Healthy Brain Ageing Centre Regional Research Network, Institute of Basic Medical Sciences, University of Oslo, Norway.,Department of Neurology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
209
|
Morgan RG, Gibbs JT, Melief EJ, Postupna NO, Sherfield EE, Wilson A, Keene CD, Montine TJ, Palmiter RD, Darvas M. Relative contributions of severe dopaminergic neuron ablation and dopamine depletion to cognitive impairment. Exp Neurol 2015; 271:205-14. [PMID: 26079646 DOI: 10.1016/j.expneurol.2015.06.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/22/2015] [Accepted: 06/12/2015] [Indexed: 10/23/2022]
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons and produces a movement disorder and cognitive impairment that becomes more extensive with the duration of the disease. To what extent cognitive impairment in advanced PD can be attributed to severe loss of dopamine (DA) signaling is not well understood. Furthermore, it is unclear if the loss of DA neurons contributes to the cognitive impairment caused by the reduction in DA signaling. We generated genetic mouse models with equally severe chronic loss of DA achieved by either extensive ablation of DA neurons or inactivation of DA synthesis from preserved neurons and compared their motor and cognitive performance. Motor behaviors were equally blunted in both models, but we observed that DA neuron ablation caused more severe cognitive deficits than DA depletion. Both models had marked deficits in cue-discrimination learning. Yet, deficits in cue-discrimination learning were more severe in mice with DA neuron ablation and only mice with DA neuron ablation had drastically impaired performance in spatial learning, spatial memory and object memory tests. These results indicate that while a severe reduction in DA signaling results in motor and cognitive impairments, the loss of DA neurons promotes more extensive cognitive deficits and suggest that a loss of additional factors that depend on DA neurons may participate in the progressive cognitive decline found in patients with PD.
Collapse
Affiliation(s)
- R Garrett Morgan
- Department of Pathology, University of Washington School of Medicine, University of Washington, Box 357470, Seattle, WA 98195, USA
| | - Jeffrey T Gibbs
- Department of Pathology, University of Washington School of Medicine, University of Washington, Box 357470, Seattle, WA 98195, USA
| | - Erica J Melief
- Department of Pathology, University of Washington School of Medicine, University of Washington, Box 357470, Seattle, WA 98195, USA
| | - Nadia O Postupna
- Department of Pathology, University of Washington School of Medicine, University of Washington, Box 357470, Seattle, WA 98195, USA
| | - Emily E Sherfield
- Department of Pathology, University of Washington School of Medicine, University of Washington, Box 357470, Seattle, WA 98195, USA
| | - Angela Wilson
- Department of Pathology, University of Washington School of Medicine, University of Washington, Box 357470, Seattle, WA 98195, USA
| | - C Dirk Keene
- Department of Pathology, University of Washington School of Medicine, University of Washington, Box 357470, Seattle, WA 98195, USA
| | - Thomas J Montine
- Department of Pathology, University of Washington School of Medicine, University of Washington, Box 357470, Seattle, WA 98195, USA
| | - Richard D Palmiter
- Department of Biochemistry, University of Washington School of Medicine, University of Washington, Box 357350, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington School of Medicine, University of Washington, Box 357350, Seattle, WA 98195, USA
| | - Martin Darvas
- Department of Pathology, University of Washington School of Medicine, University of Washington, Box 357470, Seattle, WA 98195, USA.
| |
Collapse
|
210
|
Gu H, Lazarenko RM, Koktysh D, Iacovitti L, Zhang Q. A Stem Cell-Derived Platform for Studying Single Synaptic Vesicles in Dopaminergic Synapses. Stem Cells Transl Med 2015; 4:887-93. [PMID: 26025981 DOI: 10.5966/sctm.2015-0005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 04/13/2015] [Indexed: 11/16/2022] Open
Abstract
The exocytotic release of dopamine is one of the most characteristic but also one of the least appreciated processes in dopaminergic neurotransmission. Fluorescence imaging has yielded rich information about the properties of synaptic vesicles and the release of neurotransmitters in excitatory and inhibitory neurons. In contrast, imaging-based studies for in-depth understanding of synaptic vesicle behavior in dopamine neurons are lagging largely because of a lack of suitable preparations. Midbrain culture has been one of the most valuable preparations for the subcellular investigation of dopaminergic transmission; however, the paucity and fragility of cultured dopaminergic neurons limits their use for live cell imaging. Recent developments in stem cell technology have led to the successful production of dopamine neurons from embryonic or induced pluripotent stem cells. Although the dopaminergic identity of these stem cell-derived neurons has been characterized in different ways, vesicle-mediated dopamine release from their axonal terminals has been barely assessed. We report a more efficient procedure to reliably generate dopamine neurons from embryonic stem cells, and it yields more dopamine neurons with more dopaminergic axon projections than midbrain culture does. Using a collection of functional measurements, we show that stem cell-derived dopamine neurons are indistinguishable from those in midbrain culture. Taking advantage of this new preparation, we simultaneously tracked the turnover of hundreds of synaptic vesicles individually using pH-sensitive quantum dots. By doing so, we revealed distinct fusion kinetics of the dopamine-secreting vesicles, which is consistent within both preparations.
Collapse
Affiliation(s)
- Haigang Gu
- Department of Pharmacology and Vanderbilt Institute of Nanoscale Science and Engineering, Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Neuroscience, Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Roman M Lazarenko
- Department of Pharmacology and Vanderbilt Institute of Nanoscale Science and Engineering, Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Neuroscience, Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Dmitry Koktysh
- Department of Pharmacology and Vanderbilt Institute of Nanoscale Science and Engineering, Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Neuroscience, Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Lorraine Iacovitti
- Department of Pharmacology and Vanderbilt Institute of Nanoscale Science and Engineering, Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Neuroscience, Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Qi Zhang
- Department of Pharmacology and Vanderbilt Institute of Nanoscale Science and Engineering, Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Neuroscience, Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
211
|
Werlen E, Jones MW. Modulating the map: dopaminergic tuning of hippocampal spatial coding and interactions. PROGRESS IN BRAIN RESEARCH 2015; 219:187-216. [PMID: 26072240 DOI: 10.1016/bs.pbr.2015.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Salient events activate the midbrain dopaminergic system and have important impacts on various aspects of mnemonic function, including the stability of hippocampus-dependent memories. Dopamine is also central to modulation of neocortical memory processing, particularly during prefrontal cortex-dependent working memory. Here, we review the current state of the circuitry and physiology underlying dopamine's actions, suggesting that--alongside local effects within hippocampus and prefrontal cortex--dopamine released from the midbrain ventral tegmental area is well positioned to dynamically tune interactions between limbic-cortical circuits through modulation of rhythmic network activity.
Collapse
Affiliation(s)
- Emilie Werlen
- School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol, UK.
| | - Matthew W Jones
- School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol, UK
| |
Collapse
|
212
|
Cre-driven optogenetics in the heterogeneous genetic panorama of the VTA. Trends Neurosci 2015; 38:375-86. [PMID: 25962754 DOI: 10.1016/j.tins.2015.04.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 04/08/2015] [Accepted: 04/10/2015] [Indexed: 11/24/2022]
Abstract
The selectivity of optogenetics commonly relies on genetic promoters to manipulate specific populations of neurons through the use of Cre-driver lines. All studies performed in the ventral tegmental area (VTA) so far have utilized promoters present in groups of cells that release dopamine (DA), GABA, or glutamate. However, neurons that co-release neurotransmitters and variabilities within groups of neurons that release the same neurotransmitter present challenges when evaluating the results. Further complexity is introduced by ectopic expression patterns often occurring in transgenic Cre-drivers. New perspectives could be unfolded by identifying and selecting different types of promoter for driving the Cre recombinase. Here, we discuss some promising candidates and highlight the advantages or disadvantages of different methods for creating novel transgenic lines.
Collapse
|
213
|
Antidepressant Effects of Ketamine Are Not Related to 18F-FDG Metabolism or Tyrosine Hydroxylase Immunoreactivity in the Ventral Tegmental Area of Wistar Rats. Neurochem Res 2015; 40:1153-64. [DOI: 10.1007/s11064-015-1576-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 03/16/2015] [Accepted: 04/06/2015] [Indexed: 12/28/2022]
|
214
|
Oswald MJ, Schulz JM, Kelsch W, Oorschot DE, Reynolds JNJ. Potentiation of NMDA receptor-mediated transmission in striatal cholinergic interneurons. Front Cell Neurosci 2015; 9:116. [PMID: 25914618 PMCID: PMC4391264 DOI: 10.3389/fncel.2015.00116] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 03/13/2015] [Indexed: 12/30/2022] Open
Abstract
Pauses in the tonic firing of striatal cholinergic interneurons (CINs) emerge during reward-related learning in response to conditioning of a neutral cue. We have previously reported that augmenting the postsynaptic response to cortical afferents in CINs is coupled to the emergence of a cell-intrinsic afterhyperpolarization (AHP) underlying pauses in tonic activity. Here we investigated in a bihemispheric rat-brain slice preparation the mechanisms of synaptic plasticity of excitatory afferents to CINs and the association with changes in the AHP. We found that high frequency stimulation (HFS) of commissural corticostriatal afferents from the contralateral hemisphere induced a robust long-term depression (LTD) of postsynaptic potentials (PSP) in CINs. Depression of the PSP of smaller magnitude and duration was observed in response to HFS of the ipsilateral white matter or cerebral cortex. In Mg2+-free solution HFS induced NMDA receptor-dependent potentiation of the PSP, evident in both the maximal slope and amplitude of the PSP. The increase in maximal slope corroborates previous findings, and was blocked by antagonism of either D1-like dopamine receptors with SCH23390 or D2-like dopamine receptors with sulpiride during HFS in Mg2+-free solution. Potentiation of the slower PSP amplitude component was due to augmentation of the NMDA receptor-mediated potential as this was completely reversed on subsequent application of the NMDA receptor antagonist AP5. HFS similarly potentiated NMDA receptor currents isolated by blockade of AMPA/kainate receptors with CNQX. The plasticity-induced increase in the slow PSP component was directly associated with an increase in the subsequent AHP. Thus plasticity of cortical afferent synapses is ideally suited to influence the cue-induced firing dynamics of CINs, particularly through potentiation of NMDA receptor-mediated synaptic transmission.
Collapse
Affiliation(s)
- Manfred J Oswald
- Department of Anatomy and the Brain Health Research Centre, University of Otago Dunedin, New Zealand
| | - Jan M Schulz
- Department of Anatomy and the Brain Health Research Centre, University of Otago Dunedin, New Zealand
| | - Wolfgang Kelsch
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University Mannheim, Germany
| | - Dorothy E Oorschot
- Department of Anatomy and the Brain Health Research Centre, University of Otago Dunedin, New Zealand
| | - John N J Reynolds
- Department of Anatomy and the Brain Health Research Centre, University of Otago Dunedin, New Zealand
| |
Collapse
|
215
|
Taylor SR, Badurek S, Dileone RJ, Nashmi R, Minichiello L, Picciotto MR. GABAergic and glutamatergic efferents of the mouse ventral tegmental area. J Comp Neurol 2015; 522:3308-34. [PMID: 24715505 DOI: 10.1002/cne.23603] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 04/02/2014] [Accepted: 04/03/2014] [Indexed: 12/11/2022]
Abstract
The role of dopaminergic (DA) projections from the ventral tegmental area (VTA) in appetitive and rewarding behavior has been widely studied, but the VTA also has documented DA-independent functions. Several drugs of abuse, act on VTA GABAergic neurons, and most studies have focused on local inhibitory connections. Relatively little is known about VTA GABA projection neurons and their connections to brain sites outside the VTA. This study employed viral-vector-mediated cell-type-specific anterograde tracing, classical retrograde tracing, and immunohistochemistry to characterize VTA GABA efferents throughout the brain. We found that VTA GABA neurons project widely to forebrain and brainstem targets, including the ventral pallidum, lateral and magnocellular preoptic nuclei, lateral hypothalamus, and lateral habenula. Minor projections also go to central amygdala, mediodorsal thalamus, dorsal raphe, and deep mesencephalic nuclei, and sparse projections go to prefrontal cortical regions and to nucleus accumbens shell and core. These projections differ from the major VTA DA target regions. Retrograde tracing studies confirmed results from the anterograde experiments and differences in projections from VTA subnuclei. Retrogradely labeled GABA neurons were not numerous, and most non-tyrosine hydroxylase/retrogradely labeled cells lacked GABAergic markers. Many non-TH/retrogradely labeled cells projecting to several areas expressed VGluT2. VTA GABA and glutamate neurons project throughout the brain, most prominently to regions with reciprocal connections to the VTA. These data indicate that VTA GABA and glutamate neurons may have more DA-independent functions than previously recognized.
Collapse
Affiliation(s)
- Seth R Taylor
- Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut, 06519
| | | | | | | | | | | |
Collapse
|
216
|
Stuber GD, Stamatakis AM, Kantak PA. Considerations when using cre-driver rodent lines for studying ventral tegmental area circuitry. Neuron 2015; 85:439-45. [PMID: 25611514 DOI: 10.1016/j.neuron.2014.12.034] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2014] [Indexed: 01/20/2023]
Abstract
The use of Cre-driver rodent lines for targeting ventral tegmental area (VTA) cell types has generated important and novel insights into how precise neurocircuits regulate physiology and behavior. While this approach generally results in enhanced cellular specificity, an important issue has recently emerged related to the selectivity and penetrance of viral targeting of VTA neurons using several Cre-driver transgenic mouse lines. Here, we highlight several considerations when utilizing these tools to study the function of genetically defined neurocircuits. While VTA dopaminergic neurons have previously been targeted and defined by the expression of single genes important for aspects of dopamine neurotransmission, many VTA and neighboring cells display dynamic gene expression phenotypes that are partially consistent with both classically described dopaminergic and non-dopaminergic neurons. Thus, in addition to varying degrees of selectivity and penetrance, distinct Cre lines likely permit targeting of partially overlapping, but not identical VTA cell populations. This Matters Arising Response paper addresses the Lammel et al. (2015) Matters Arising paper, published concurrently in Neuron.
Collapse
Affiliation(s)
- Garret D Stuber
- Departments of Psychiatry and Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Alice M Stamatakis
- Curriculum in Neurobiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Pranish A Kantak
- Departments of Psychiatry and Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
217
|
Homeostatic regulation of excitatory synapses on striatal medium spiny neurons expressing the D2 dopamine receptor. Brain Struct Funct 2015; 221:2093-107. [PMID: 25782435 DOI: 10.1007/s00429-015-1029-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 03/06/2015] [Indexed: 10/23/2022]
Abstract
Striatal medium spiny neurons (MSNs) are contacted by glutamatergic axon terminals originating from cortex, thalamus and other regions. The striatum is also innervated by dopaminergic (DAergic) terminals, some of which release glutamate as a co-transmitter. Despite evidence for functional DA release at birth in the striatum, the role of DA in the establishment of striatal circuitry is unclear. In light of recent work suggesting activity-dependent homeostatic regulation of glutamatergic terminals on MSNs expressing the D2 DA receptor (D2-MSNs), we used primary co-cultures to test the hypothesis that stimulation of DA and glutamate receptors regulates the homeostasis of glutamatergic synapses on MSNs. Co-culture of D2-MSNs with mesencephalic DA neurons or with cortical neurons produced an increase in spines and functional glutamate synapses expressing VGLUT2 or VGLUT1, respectively. The density of VGLUT2-positive terminals was reduced by the conditional knockout of this gene from DA neurons. In the presence of both mesencephalic and cortical neurons, the density of synapses reached the same total, compatible with the possibility of a homeostatic mechanism capping excitatory synaptic density. Blockade of D2 receptors increased the density of cortical and mesencephalic glutamatergic terminals, without changing MSN spine density or mEPSC frequency. Combined blockade of AMPA and NMDA glutamate receptors increased the density of cortical terminals and decreased that of mesencephalic VGLUT2-positive terminals, with no net change in total excitatory terminal density or in mEPSC frequency. These results suggest that DA and glutamate signaling regulate excitatory inputs to striatal D2-MSNs at both the pre- and postsynaptic level, under the influence of a homeostatic mechanism controlling functional output of the circuit.
Collapse
|
218
|
Zhang S, Qi J, Li X, Wang HL, Britt JP, Hoffman AF, Bonci A, Lupica CR, Morales M. Dopaminergic and glutamatergic microdomains in a subset of rodent mesoaccumbens axons. Nat Neurosci 2015; 18:386-92. [PMID: 25664911 PMCID: PMC4340758 DOI: 10.1038/nn.3945] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/13/2015] [Indexed: 12/14/2022]
Abstract
Mesoaccumbens fibers are thought to co-release dopamine and glutamate. However, the mechanism is unclear, and co-release by mesoaccumbens fibers has not been documented. Using electron microcopy, we found that some mesoaccumbens fibers have vesicular transporters for dopamine (VMAT2) in axon segments that are continuous with axon terminals that lack VMAT2, but contain vesicular glutamate transporters type 2 (VGluT2). In vivo overexpression of VMAT2 did not change the segregation of the two vesicular types, suggesting the existence of highly regulated mechanisms for maintaining this segregation. The mesoaccumbens axon terminals containing VGluT2 vesicles make asymmetric synapses, commonly associated with excitatory signaling. Using optogenetics, we found that dopamine and glutamate were released from the same mesoaccumbens fibers. These findings reveal a complex type of signaling by mesoaccumbens fibers in which dopamine and glutamate can be released from the same axons, but are not normally released at the same site or from the same synaptic vesicles.
Collapse
Affiliation(s)
- Shiliang Zhang
- National Institute on Drug Abuse, Neuronal Networks Section, US National Institutes of Health, Baltimore, Maryland, USA
| | - Jia Qi
- National Institute on Drug Abuse, Neuronal Networks Section, US National Institutes of Health, Baltimore, Maryland, USA
| | - Xueping Li
- National Institute on Drug Abuse, Neuronal Networks Section, US National Institutes of Health, Baltimore, Maryland, USA
| | - Hui-Ling Wang
- National Institute on Drug Abuse, Neuronal Networks Section, US National Institutes of Health, Baltimore, Maryland, USA
| | - Jonathan P. Britt
- National Institute on Drug Abuse, Synaptic Plasticity Section, US National Institutes of Health, Baltimore, Maryland, USA
| | - Alexander F. Hoffman
- National Institute on Drug Abuse, Electrophysiology Research Section, US National Institutes of Health, Baltimore, Maryland, USA
| | - Antonello Bonci
- National Institute on Drug Abuse, Synaptic Plasticity Section, US National Institutes of Health, Baltimore, Maryland, USA
| | - Carl R. Lupica
- National Institute on Drug Abuse, Electrophysiology Research Section, US National Institutes of Health, Baltimore, Maryland, USA
| | - Marisela Morales
- National Institute on Drug Abuse, Neuronal Networks Section, US National Institutes of Health, Baltimore, Maryland, USA
| |
Collapse
|
219
|
Jasinska AJ, Chen BT, Bonci A, Stein EA. Dorsal medial prefrontal cortex (MPFC) circuitry in rodent models of cocaine use: implications for drug addiction therapies. Addict Biol 2015; 20:215-26. [PMID: 24620898 DOI: 10.1111/adb.12132] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although the importance of the medial prefrontal cortex (MPFC) in cocaine addiction is well established, its precise contribution to cocaine seeking, taking and relapse remains incompletely understood. In particular, across two different models of cocaine self-administration, pharmacological or optogenetic activation of the dorsal MPFC has been reported to sometimes promote and sometimes inhibit cocaine seeking. We highlight important methodological differences between the two experimental paradigms and propose a framework to potentially reconcile the apparent discrepancy. We also draw parallels between these pre-clinical models of cocaine self-administration and human neuro-imaging studies in cocaine users, and argue that both lines of evidence point to dynamic interactions between cue-reactivity processes and control processes within the dorsal MPFC circuitry. From a translational perspective, these findings underscore the importance of interventions and therapeutics targeting not just a brain region, but a specific computational process within that brain region, and may have implications for the design and implementation of more effective treatments for human cocaine addiction.
Collapse
Affiliation(s)
- Agnes J. Jasinska
- Intramural Research Program; National Institute on Drug Abuse; Baltimore MD USA
| | - Billy T. Chen
- Intramural Research Program; National Institute on Drug Abuse; Baltimore MD USA
| | - Antonello Bonci
- Solomon Snyder Department of Neuroscience; The Johns Hopkins University School of Medicine; Baltimore MD USA
- Department of Psychiatry; The Johns Hopkins University School of Medicine; Baltimore MD USA
- Intramural Research Program; National Institute on Drug Abuse; Baltimore MD USA
| | - Elliot A. Stein
- Intramural Research Program; National Institute on Drug Abuse; Baltimore MD USA
| |
Collapse
|
220
|
Yamaguchi T, Qi J, Wang HL, Zhang S, Morales M. Glutamatergic and dopaminergic neurons in the mouse ventral tegmental area. Eur J Neurosci 2015; 41:760-72. [PMID: 25572002 PMCID: PMC4363208 DOI: 10.1111/ejn.12818] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 12/02/2014] [Indexed: 12/31/2022]
Abstract
The ventral tegmental area (VTA) comprises dopamine (DA), γ-aminobutyric acid (GABA) and glutamate (Glu) neurons. Some rat VTA Glu neurons, expressing vesicular glutamate transporter 2 (VGluT2), co-express tyrosine hydroxylase (TH). While transgenic mice are now being used in attempts to determine the role of VGluT2/TH neurons in reward and neuronal signaling, such neurons have not been characterized in mouse tissue. By cellular detection of VGluT2 mRNA and TH immunoreactivity (TH-IR), we determined the cellular expression of VGluT2 mRNA within VTA TH-IR neurons in the mouse. We found that some mouse VGluT2 neurons coexpressed TH-IR, but their frequency was lower than in the rat. To determine whether low expression of TH mRNA or TH-IR accounts for this low frequency, we evaluated VTA cellular coexpression of TH transcripts and TH protein. Within the medial aspects of the VTA, some neurons expressed TH mRNA but lacked TH-IR; among them a subset coexpressed VGluT2 mRNA. To determine if lack of VTA TH-IR was due to TH trafficking, we tagged VTA TH neurons by Cre-inducible expression of mCherry in TH::Cre mice. By dual immunofluorescence, we detected axons containing mCherry, but lacking TH-IR, in the lateral habenula, indicating that low frequency of VGluT2 mRNA (+)/TH-IR (+) neurons in the mouse is due to lack of synthesis of TH protein, rather than TH protein trafficking. In conclusion, VGluT2 neurons are present in the rat and mouse VTA, but they differ in the populations of VGluT2/TH and TH neurons. Under normal conditions, the translation of TH protein is suppressed in the mouse mesohabenular TH neurons.
Collapse
Affiliation(s)
- Tsuyoshi Yamaguchi
- TY. Current address, Department of Histology and Neurobiology, Dokkyo Medical University, 880 Kitakobayashi, Mibu-machi, Shimotsuga-gun, Tochigi, 321-0293 JAPAN,
| | | | - Hui-Ling Wang
- National Institutes of Health. National Institute on Drug Abuse, Intramural Research Program. Neuronal Networks Section. 251 Bayview Boulevard, Baltimore, MD 21224
| | - Shiliang Zhang
- National Institutes of Health. National Institute on Drug Abuse, Intramural Research Program. Neuronal Networks Section. 251 Bayview Boulevard, Baltimore, MD 21224
| | - Marisela Morales
- National Institutes of Health. National Institute on Drug Abuse, Intramural Research Program. Neuronal Networks Section. 251 Bayview Boulevard, Baltimore, MD 21224
| |
Collapse
|
221
|
Mrejeru A, Martí-Prats L, Avegno EM, Harrison NL, Sulzer D. A subset of ventral tegmental area dopamine neurons responds to acute ethanol. Neuroscience 2015; 290:649-58. [PMID: 25660505 DOI: 10.1016/j.neuroscience.2014.12.081] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 12/16/2014] [Accepted: 12/18/2014] [Indexed: 10/24/2022]
Abstract
The mechanisms by which alcohol drinking promotes addiction in humans and self-administration in rodents remain obscure, but it is well known that alcohol can enhance dopamine (DA) neurotransmission from neurons of the ventral tegmental area (VTA) and increase DA levels within the nucleus accumbens and prefrontal cortex. We recorded from identified DA neuronal cell bodies within ventral midbrain slices prepared from a transgenic mouse line (TH-GFP) using long-term stable extracellular recordings in a variety of locations and carefully mapped the responses to applied ethanol (EtOH). We identified a subset of DA neurons in the medial VTA located within the rostral linear and interfascicular nuclei that fired spontaneously and exhibited a concentration-dependent increase of firing frequency in response to EtOH, with some neurons responsive to as little as 20mM EtOH. Many of these medial VTA DA neurons were also insensitive to the D2 receptor agonist quinpirole. In contrast, DA neurons in the lateral VTA (located within the parabrachial pigmented and paranigral nuclei) were either unresponsive or responded only to 100mM EtOH. Typically, these lateral VTA DA cells had very slow firing rates, and all exhibited inhibition by quinpirole via D2 "autoreceptors". VTA non-DA cells did not show any significant response to low levels of EtOH. These findings are consistent with evidence for heterogeneity among midbrain DA neurons and provide an anatomical and pharmacological distinction between DA neuron sub-populations that will facilitate future mechanistic studies on the actions of EtOH in the VTA.
Collapse
Affiliation(s)
- A Mrejeru
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | - L Martí-Prats
- Departament de Farmàcia i Tecnologia Farmacèutica, Universitat de València, Burjassot, Spain
| | - E M Avegno
- Departments of Anesthesiology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA
| | - N L Harrison
- Departments of Anesthesiology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA.
| | - D Sulzer
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA; Department of Psychiatry and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
222
|
Filippi A, Mueller T, Driever W. vglut2 and gad expression reveal distinct patterns of dual GABAergic versus glutamatergic cotransmitter phenotypes of dopaminergic and noradrenergic neurons in the zebrafish brain. J Comp Neurol 2015; 522:2019-37. [PMID: 24374659 PMCID: PMC4288968 DOI: 10.1002/cne.23524] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 11/27/2013] [Accepted: 11/27/2013] [Indexed: 01/22/2023]
Abstract
Throughout the vertebrate lineage, dopaminergic neurons form important neuromodulatory systems that influence motor behavior, mood, cognition, and physiology. Studies in mammals have established that dopaminergic neurons often use γ-aminobutyric acid (GABA) or glutamatergic cotransmission during development and physiological function. Here, we analyze vglut2, gad1b and gad2 expression in combination with tyrosine hydroxylase immunoreactivity in 4-day-old larval and 30-day-old juvenile zebrafish brains to determine which dopaminergic and noradrenergic groups may use GABA or glutamate as a second transmitter. Our results show that most dopaminergic neurons also express GABAergic markers, including the dopaminergic groups of the olfactory bulb (homologous to mammalian A16) and the subpallium, the hypothalamic groups (A12, A14), the prethalamic zona incerta group (A13), the preoptic groups (A15), and the pretectal group. Thus, the majority of catecholaminergic neurons are gad1b/2-positive and coexpress GABA. A very few gad1/2-negative dopaminergic groups, however, express vglut2 instead and use glutamate as a second transmitter. These glutamatergic dual transmitter phenotypes are the Orthopedia transcription factor–dependent, A11-type dopaminergic neurons of the posterior tuberculum. All together, our results demonstrate that all catecholaminergic groups in zebrafish are either GABAergic or glutamatergic. Thus, cotransmission of dopamine and noradrenaline with either GABA or glutamate appears to be a regular feature of zebrafish catecholaminergic systems. We compare our results with those that have been described for mammalian systems, discuss the phenomenon of transmitter dualism in the context of developmental specification of GABAergic and glutamatergic regions in the brain, and put this phenomenon in an evolutionary perspective. J. Comp. Neurol. 522:2019–2037, 2014.
Collapse
Affiliation(s)
- Alida Filippi
- Developmental Biology, Institute of Biology I, Faculty of Biology, Albert-Ludwigs-University Freiburg, 79104, Freiburg, Germany
| | | | | |
Collapse
|
223
|
Abstract
Disruptions in mitochondrial dynamics may contribute to the selective degeneration of dopamine (DA) neurons in Parkinson's disease (PD). However, little is known about the normal functions of mitochondrial dynamics in these neurons, especially in axons where degeneration begins, and this makes it difficult to understand the disease process. To study one aspect of mitochondrial dynamics-mitochondrial fission-in mouse DA neurons, we deleted the central fission protein dynamin-related protein 1 (Drp1). Drp1 loss rapidly eliminates the DA terminals in the caudate-putamen and causes cell bodies in the midbrain to degenerate and lose α-synuclein. Without Drp1, mitochondrial mass dramatically decreases, especially in axons, where the mitochondrial movement becomes uncoordinated. However, in the ventral tegmental area (VTA), a subset of midbrain DA neurons characterized by small hyperpolarization-activated cation currents (Ih) is spared, despite near complete loss of their axonal mitochondria. Drp1 is thus critical for targeting mitochondria to the nerve terminal, and a disruption in mitochondrial fission can contribute to the preferential death of nigrostriatal DA neurons.
Collapse
|
224
|
Nordenankar K, Bergfors A, Wallén-Mackenzie Å. Targeted deletion of Vglut2 expression in the embryonal telencephalon promotes an anxiolytic phenotype of the adult mouse. Ups J Med Sci 2015; 120:144-56. [PMID: 25857802 PMCID: PMC4526870 DOI: 10.3109/03009734.2015.1032454] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Anxiety is a natural emotion experienced by all individuals. However, when anxiety becomes excessive, it contributes to the substantial group of anxiety disorders that affect one in three people and thus are among the most common psychiatric disorders. Anxiolysis, the reduction of anxiety, is mediated via several large groups of therapeutical compounds, but the relief is often only temporary, and increased knowledge of the neurobiology underlying anxiety is needed in order to improve future therapies. AIM We previously demonstrated that mice lacking forebrain expression of the Vesicular glutamate transporter 2 (Vglut2) from adolescence showed a strong anxiolytic behaviour as adults. In the current study, we wished to analyse if removal of Vglut2 expression already from mid-gestation of the mouse embryo would give rise to similar anxiolysis in the adult mouse. METHODS We produced transgenic mice lacking Vglut2 from mid-gestation and analysed their affective behaviour, including anxiety, when they had reached adulthood. RESULTS The transgenic mice lacking Vglut2 expression from mid-gestation showed certain signs of anxiolytic behaviour, but this phenotype was not as prominent as when Vglut2 was removed during adolescence. CONCLUSION Our results suggest that both embryonal and adolescent forebrain expression of Vglut2 normally contributes to balancing the level of anxiety. As the neurobiological basis for anxiety is similar across species, our results in mice may help improve the current understanding of the neurocircuitry of anxiety, and hence anxiolysis, also in humans.
Collapse
Affiliation(s)
- Karin Nordenankar
- Department of Neuroscience, Unit of Functional Neurobiology and Unit of Developmental Genetics, Uppsala University, Box 593, S-75214 Uppsala, Sweden
| | - Assar Bergfors
- Department of Neuroscience, Unit of Functional Neurobiology and Unit of Developmental Genetics, Uppsala University, Box 593, S-75214 Uppsala, Sweden
| | - Åsa Wallén-Mackenzie
- Department of Neuroscience, Unit of Functional Neurobiology and Unit of Developmental Genetics, Uppsala University, Box 593, S-75214 Uppsala, Sweden
| |
Collapse
|
225
|
de Velasco EMF, McCall N, Wickman K. GIRK Channel Plasticity and Implications for Drug Addiction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 123:201-38. [DOI: 10.1016/bs.irn.2015.05.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
226
|
Abstract
Enzyme-linked signal amplification is a key technique used to enhance the immunohistochemical detection of protein, mRNA, and other molecular species. Tyramide signal amplification (TSA) is based on a catalytic reporter deposit in close vicinity to the epitope of interest. The advantages of this technique are its simplicity, enhanced sensitivity, high specificity, and compatibility with modern multi-label fluorescent microscopy. Here, we describe the use of a TSA kit to increase the signal of enhanced green fluorescent protein (eGFP) expressed under the control of Slc17a6 regulatory elements in the brain of a transgenic mouse. The labeling procedure consists of 6 basic steps: (1) tissue preparation, (2) blocking of nonspecific epitopes, (3) binding with primary antibody, (4) binding with horseradish peroxidase-conjugated secondary antibody, (5) reacting with fluorescent tyramide substrate, and (6) imaging of the signal. The procedures described herein detail these steps and provide additional guidance and background to assist novice users.
Collapse
|
227
|
Pistillo F, Clementi F, Zoli M, Gotti C. Nicotinic, glutamatergic and dopaminergic synaptic transmission and plasticity in the mesocorticolimbic system: focus on nicotine effects. Prog Neurobiol 2014; 124:1-27. [PMID: 25447802 DOI: 10.1016/j.pneurobio.2014.10.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 10/08/2014] [Accepted: 10/24/2014] [Indexed: 01/11/2023]
Abstract
Cigarette smoking is currently the leading cause of preventable deaths and disability throughout the world, being responsible for about five million premature deaths/year. Unfortunately, fewer than 10% of tobacco users who try to stop smoking actually manage to do so. The main addictive agent delivered by cigarette smoke is nicotine, which induces psychostimulation and reward, and reduces stress and anxiety. The use of new technologies (including optogenetics) and the development of mouse models characterised by cell-specific deletions of receptor subtype genes or the expression of gain-of-function nAChR subunits has greatly increased our understanding of the molecular mechanisms and neural substrates of nicotine addiction first revealed by classic electrophysiological, neurochemical and behavioural approaches. It is now becoming clear that various aspects of nicotine dependence are mediated by close interactions of the glutamatergic, dopaminergic and γ-aminobutyric acidergic systems in the mesocorticolimbic system. This review is divided into two parts. The first provides an updated overview of the circuitry of the ventral tegmental area, ventral striatum and prefrontal cortex, the neurotransmitter receptor subtypes expressed in these areas, and their physiological role in the mesocorticolimbic system. The second will focus on the molecular, functional and behavioural mechanisms involved in the acute and chronic effects of nicotine on the mesocorticolimbic system.
Collapse
Affiliation(s)
- Francesco Pistillo
- CNR, Neuroscience Institute-Milano, Biometra University of Milan, Milan, Italy
| | - Francesco Clementi
- CNR, Neuroscience Institute-Milano, Biometra University of Milan, Milan, Italy
| | - Michele Zoli
- Department of Biomedical, Metabolic and Neural Sciences, Section of Physiology and Neurosciences, University of Modena and Reggio Emilia, Modena, Italy.
| | - Cecilia Gotti
- CNR, Neuroscience Institute-Milano, Biometra University of Milan, Milan, Italy.
| |
Collapse
|
228
|
Comasco E, Hallman J, Wallén-Mackenzie A. Haplotype-tag single nucleotide polymorphism analysis of the vesicular glutamate transporter (VGLUT) genes in severely alcoholic women. Psychiatry Res 2014; 219:403-5. [PMID: 24953423 DOI: 10.1016/j.psychres.2014.05.052] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 05/18/2014] [Accepted: 05/28/2014] [Indexed: 11/26/2022]
Affiliation(s)
- Erika Comasco
- Department of Neuroscience, Uppsala University, BMC, Box 593, SE-75124 Uppsala, Sweden
| | - Jarmila Hallman
- Department of Neuroscience, Uppsala University, BMC, Box 593, SE-75124 Uppsala, Sweden
| | - Asa Wallén-Mackenzie
- Department of Neuroscience, Uppsala University, BMC, Box 593, SE-75124 Uppsala, Sweden.
| |
Collapse
|
229
|
Phasic dopaminergic activity exerts fast control of cholinergic interneuron firing via sequential NMDA, D2, and D1 receptor activation. J Neurosci 2014; 34:11549-59. [PMID: 25164653 DOI: 10.1523/jneurosci.1175-14.2014] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Phasic increases in dopamine (DA) are involved in the detection and selection of relevant sensory stimuli. The DAergic and cholinergic system dynamically interact to gate and potentiate sensory inputs to striatum. Striatal cholinergic interneurons (CINs) respond to relevant sensory stimuli with an initial burst, a firing pause, or a late burst, or a combination of these three components. CIN responses coincide with phasic firing of DAergic neurons in vivo. In particular, the late burst of CINs codes for the anticipated reward. To examine whether DAergic midbrain afferents can evoke the different CIN responses, we recorded from adult olfactory tubercle slices in the mouse ventral striatum. Olfactory inputs to striatal projection neurons were gated by the cholinergic tone. Phasic optogenetic activation of DAergic terminals evoked combinations of initial bursts, pauses, and late bursts in subsets of CINs by distinct receptor pathways. Glutamate release from midbrain afferents evoked an NMDAR-dependent initial burst followed by an afterhyperpolarization-induced pause. Phasic release of DA itself evoked acute changes in CIN firing. In particular, in CINs without an initial burst, phasic DA release evoked a pause through D2-type DA receptor activation. Independently, phasic DA activated a slow depolarizing conductance and the late burst through a D1-type DA receptor pathway. In summary, DAergic neurons elicit transient subsecond firing responses in CINs by sequential activation of NMDA, D2-type, and D1-type receptors. This fast control of striatal cholinergic tone by phasic DA provides a novel dynamic link of two transmitter systems central to the detection and selection of relevant stimuli.
Collapse
|
230
|
Quantitative Multi-modal Brain Autoradiography of Glutamatergic, Dopaminergic, Cannabinoid, and Nicotinic Receptors in Mutant Disrupted-In-Schizophrenia-1 (DISC1) Mice. Mol Imaging Biol 2014; 17:355-63. [DOI: 10.1007/s11307-014-0786-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
231
|
Iwashita M. Phasic activation of ventral tegmental neurons increases response and pattern similarity in prefrontal cortex neurons. eLife 2014; 3. [PMID: 25269147 PMCID: PMC4206826 DOI: 10.7554/elife.02726] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 09/28/2014] [Indexed: 12/15/2022] Open
Abstract
Dopamine is critical for higher neural processes and modifying the activity of the prefrontal cortex (PFC). However, the mechanism of dopamine contribution to the modification of neural representation is unclear. Using in vivo two-photon population Ca2+ imaging in awake mice, this study investigated how neural representation of visual input to PFC neurons is regulated by dopamine. Phasic stimulation of dopaminergic neurons in the ventral tegmental area (VTA) evoked prolonged Ca2+ transients, lasting ∼30 s in layer 2/3 neurons of the PFC, which are regulated by a dopamine D1 receptor-dependent pathway. Furthermore, only a conditioning protocol with visual sensory input applied 0.5 s before the VTA dopaminergic input could evoke enhanced Ca2+ transients and increased pattern similarity (or establish a neural representation) of PFC neurons to the same sensory input. By increasing both the level of neuronal response and pattern similarity, dopaminergic input may establish robust and reliable cortical representation. DOI:http://dx.doi.org/10.7554/eLife.02726.001 Around 120 years ago, Ivan Pavlov unintentionally sparked a new field of psychology research. He did so by noting that his dogs had learned to associate the sound of the bell that he rang before feeding them with the food itself, such that they would salivate upon hearing the bell even when there was no food present. This form of learning—now known as associative learning—has since been demonstrated in species from honeybees to humans. For the brain to associate two events, such as the sound of a bell and the delivery of food, it must encode the first event and keep that information available or ‘on-line’ until the occurrence of the second event, at which point the two can be linked together. This process takes place in part of the brain called the prefrontal cortex, but the mechanism by which it occurs is largely unclear. Now, Iwashita has obtained new insights into the molecular basis of associative learning by studying how the activity of the prefrontal cortex is affected by the activity of a second region of the brain. This second region, called the ventral tegmental area, is part of the brain's reward circuit: it becomes active whenever an animal experiences a desirable event, such as receiving food, and supplies a neurotransmitter called dopamine to its target areas, which include the prefrontal cortex. Electrodes were used to mimic the changes in brain activity that occur when a mouse learns to associate a visual stimulus with a reward: this involved repeatedly activating the visual cortex in a conscious mouse, followed by activation of the ventral tegmental area. Short-lived increases in calcium levels were seen in the prefrontal cortex, raising the possibility that these ‘calcium transients’ are the signal that enables the brain to link two events. Moreover, blocking proteins called dopamine D1 receptors in the prefrontal cortex reduced the calcium transients, which is consistent with existing evidence that dopamine from the ventral tegmental area is required for associative learning. Intriguingly, the calcium transients lasted for roughly 30 s, which is also the maximum length of time by which a stimulus and a reward can be separated and still be associated. Given that the calcium transients could not be detected in anesthetized mice, a full understanding of the mechanisms underlying associative learning may require studies of the conscious brain. DOI:http://dx.doi.org/10.7554/eLife.02726.002
Collapse
Affiliation(s)
- Motoko Iwashita
- 1National Institute of Mental Health, National Institutes of Health, Bethesda, United States
| |
Collapse
|
232
|
Abstract
It is now clear that many neuronal populations release more than one classical neurotransmitter, yet in most cases the functional role of corelease is unknown. Striatal cholinergic interneurons release both glutamate and acetylcholine, and vesicular loading of glutamate has been shown to enhance acetylcholine content. Using a combination of optogenetics and whole-cell recordings in mice, we now provide physiological evidence that optogenetic stimulation of cholinergic interneurons triggers monosynaptic glutamate- and acetylcholine-mediated currents in striatal fast-spiking interneurons (FSIs), both of which depend on the expression of the vesicular glutamate transporter 3 (VGLUT3). In contrast to corticostriatal glutamatergic inputs onto FSIs, which are mediated primarily by AMPA-type glutamate receptors, glutamate release by cholinergic interneurons activates both AMPA- and NMDA-type glutamate receptors, suggesting a unique role for these inputs in the modulation of FSI activity. Importantly, we find that the loss of VGLUT3 not only markedly attenuates glutamatergic and cholinergic inputs on FSIs, but also significantly decreases disynaptic GABAergic input onto medium spiny neurons (MSNs), the major output neurons of the striatum. Our data demonstrate that VGLUT3 is required for normal cholinergic signaling onto FSIs, as well as for acetylcholine-dependent disynaptic inhibition of MSNs. Thus, by supporting fast glutamatergic transmission as well as by modulating the strength of cholinergic signaling, VGLUT3 has the capacity to exert widespread influence on the striatal network.
Collapse
|
233
|
Vezoli J, Dzahini K, Costes N, Wilson CRE, Fifel K, Cooper HM, Kennedy H, Procyk E. Increased DAT binding in the early stage of the dopaminergic lesion: a longitudinal [11C]PE2I binding study in the MPTP-monkey. Neuroimage 2014; 102 Pt 2:249-61. [PMID: 25108180 DOI: 10.1016/j.neuroimage.2014.07.059] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Revised: 07/24/2014] [Accepted: 07/30/2014] [Indexed: 12/13/2022] Open
Abstract
The delayed appearance of motor symptoms in PD poses a crucial challenge for early detection of the disease. We measured the binding potential of the selective dopamine active transporter (DAT) radiotracer [(11)C]PE2I in MPTP-treated macaque monkeys, thus establishing a detailed profile of the nigrostriatal DA status following MPTP intoxication and its relation to induced motor and non-motor symptoms. Clinical score and cognitive performance were followed throughout the study. We measured longitudinally in vivo the non-displaceable binding potential to DAT in premotor, motor-recovered (i.e. both non-symptomatic) and symptomatic MPTP-treated monkeys. Results show an unexpected and pronounced dissociation between clinical scores and [(11)C]PE2I-BP(ND) during the premotor phase i.e. DAT binding in the striatum of premotor animals was increased around 20%. Importantly, this broad increase of DAT binding in the caudate, ventral striatum and anterior putamen was accompanied by i) deteriorated cognitive performance, showing a likely causal role of the observed hyperdopaminergic state (Cools, 2011; Cools and D'Esposito, 2011) and ii) an asymmetric decrease of DAT binding at a focal point of the posterior putamen, suggesting that increased DAT is one of the earliest, intrinsic compensatory mechanisms. Following spontaneous recovery from motor deficits, DAT binding was greatly reduced as recently shown in-vivo with other radiotracers (Blesa et al., 2010, 2012). Finally, high clinical scores were correlated to considerably low levels of DAT only after the induction of a stable parkinsonian state. We additionally show that the only striatal region which was significantly correlated to the degree of motor impairments is the ventral striatum. Further research on this period should allow better understanding of DA compensation at premature stages of PD and potentially identify new diagnosis and therapeutic index.
Collapse
Affiliation(s)
- Julien Vezoli
- INSERM U846, Stem Cell and Brain Research Institute, Bron, France; Université de Lyon, Université Lyon1, Lyon, France.
| | - Kwamivi Dzahini
- INSERM U846, Stem Cell and Brain Research Institute, Bron, France; Université de Lyon, Université Lyon1, Lyon, France; Primastem (LifeStemCells), Bron, France
| | | | - Charles R E Wilson
- INSERM U846, Stem Cell and Brain Research Institute, Bron, France; Université de Lyon, Université Lyon1, Lyon, France
| | - Karim Fifel
- INSERM U846, Stem Cell and Brain Research Institute, Bron, France; Université de Lyon, Université Lyon1, Lyon, France
| | - Howard M Cooper
- INSERM U846, Stem Cell and Brain Research Institute, Bron, France; Université de Lyon, Université Lyon1, Lyon, France
| | - Henry Kennedy
- INSERM U846, Stem Cell and Brain Research Institute, Bron, France; Université de Lyon, Université Lyon1, Lyon, France
| | - Emmanuel Procyk
- INSERM U846, Stem Cell and Brain Research Institute, Bron, France; Université de Lyon, Université Lyon1, Lyon, France
| |
Collapse
|
234
|
Marinelli M, McCutcheon JE. Heterogeneity of dopamine neuron activity across traits and states. Neuroscience 2014; 282:176-97. [PMID: 25084048 DOI: 10.1016/j.neuroscience.2014.07.034] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 07/21/2014] [Accepted: 07/22/2014] [Indexed: 12/29/2022]
Abstract
Midbrain dopamine neurons fire irregularly, with interspersed clusters of high-frequency spikes, commonly called 'bursts'. In this review we examine such heterogeneity in activity, and provide insight into how it can participate in psychiatric conditions such as drug addiction. We first describe several techniques used to evaluate dopamine neuron activity, and comment on the different measures that each provides. We next describe the activity of dopamine neurons in 'basal' conditions. Specifically, we discuss how the use of anesthesia and reduced preparations may alter aspects of dopamine cell activity, and how there is heterogeneity across species and regions. We also describe how dopamine cell firing changes throughout the peri-adolescent period and how dopamine neuron activity differs across the population. In the final section, we discuss how dopamine neuron activity changes in response to life events. First, we focus attention on drugs of abuse. Drugs themselves change firing activity through a variety of mechanisms, with effects on firing while drug is present differing from those seen after drug discontinuation. We then review how stimuli that are rewarding, aversive, or salient can evoke changes in firing rate and discharge pattern of dopamine neurons, and provide behavioral relevance of dopamine signaling. Finally, we discuss how stress can modulate dopamine neuron firing and how this may contribute to the role that stressful experiences play in psychiatric disorders such as addiction and depression.
Collapse
Affiliation(s)
- M Marinelli
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, 107 W. Dean Keeton, C0875, BME 6.114A, Austin, TX 78756, USA.
| | - J E McCutcheon
- Department of Cell Physiology and Pharmacology, College of Medicine, Biological Sciences and Psychology, University of Leicester, Maurice Shock Medical Sciences Building, University Road, P.O. Box 138, Leicester LE1 9HN, UK.
| |
Collapse
|
235
|
Inhibition of vesicular glutamate transporters contributes to attenuate methamphetamine-induced conditioned place preference in rats. Behav Brain Res 2014; 267:1-5. [DOI: 10.1016/j.bbr.2014.02.047] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/19/2014] [Accepted: 02/22/2014] [Indexed: 11/23/2022]
|
236
|
Morales M, Root DH. Glutamate neurons within the midbrain dopamine regions. Neuroscience 2014; 282:60-8. [PMID: 24875175 DOI: 10.1016/j.neuroscience.2014.05.032] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 05/15/2014] [Accepted: 05/18/2014] [Indexed: 12/26/2022]
Abstract
Midbrain dopamine systems play important roles in Parkinson's disease, schizophrenia, addiction, and depression. The participation of midbrain dopamine systems in diverse clinical contexts suggests these systems are highly complex. Midbrain dopamine regions contain at least three neuronal phenotypes: dopaminergic, GABAergic, and glutamatergic. Here, we review the locations, subtypes, and functions of glutamatergic neurons within midbrain dopamine regions. Vesicular glutamate transporter 2 (VGluT2) mRNA-expressing neurons are observed within each midbrain dopamine system. Within rat retrorubral field (RRF), large populations of VGluT2 neurons are observed throughout its anteroposterior extent. Within rat substantia nigra pars compacta (SNC), VGluT2 neurons are observed centrally and caudally, and are most dense within the laterodorsal subdivision. RRF and SNC rat VGluT2 neurons lack tyrosine hydroxylase (TH), making them an entirely distinct population of neurons from dopaminergic neurons. The rat ventral tegmental area (VTA) contains the most heterogeneous populations of VGluT2 neurons. VGluT2 neurons are found in each VTA subnucleus but are most dense within the anterior midline subnuclei. Some subpopulations of rat VGluT2 neurons co-express TH or glutamic acid decarboxylase (GAD), but most of the VGluT2 neurons lack TH or GAD. Different subsets of rat VGluT2-TH neurons exist based on the presence or absence of vesicular monoamine transporter 2, dopamine transporter, or D2 dopamine receptor. Thus, the capacity by which VGluT2-TH neurons may release dopamine will differ based on their capacity to accumulate vesicular dopamine, uptake extracellular dopamine, or be autoregulated by dopamine. Rat VTA VGluT2 neurons exhibit intrinsic VTA projections and extrinsic projections to the accumbens and to the prefrontal cortex. Mouse VTA VGluT2 neurons project to accumbens shell, prefrontal cortex, ventral pallidum, amygdala, and lateral habenula. Given their molecular diversity and participation in circuits involved in addiction, we hypothesize that individual VGluT2 subpopulations of neurons play unique roles in addiction and other disorders.
Collapse
Affiliation(s)
- M Morales
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Boulevard, Suite 200, Baltimore, MD 21224, United States.
| | - D H Root
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Boulevard, Suite 200, Baltimore, MD 21224, United States
| |
Collapse
|
237
|
Cachope R, Cheer JF. Local control of striatal dopamine release. Front Behav Neurosci 2014; 8:188. [PMID: 24904339 PMCID: PMC4033078 DOI: 10.3389/fnbeh.2014.00188] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 05/07/2014] [Indexed: 11/17/2022] Open
Abstract
The mesolimbic and nigrostriatal dopamine (DA) systems play a key role in the physiology of reward seeking, motivation and motor control. Importantly, they are also involved in the pathophysiology of Parkinson’s and Huntington’s disease, schizophrenia and addiction. Control of DA release in the striatum is tightly linked to firing of DA neurons in the ventral tegmental area (VTA) and the substantia nigra (SN). However, local influences in the striatum affect release by exerting their action directly on axon terminals. For example, endogenous glutamatergic and cholinergic activity is sufficient to trigger striatal DA release independently of cell body firing. Recent developments involving genetic manipulation, pharmacological selectivity or selective stimulation have allowed for better characterization of these phenomena. Such termino-terminal forms of control of DA release transform considerably our understanding of the mesolimbic and nigrostriatal systems, and have strong implications as potential mechanisms to modify impaired control of DA release in the diseased brain. Here, we review these and related mechanisms and their implications in the physiology of ascending DA systems.
Collapse
Affiliation(s)
- Roger Cachope
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine Baltimore, MD, USA ; CHDI Foundation Los Angeles, CA, USA
| | - Joseph F Cheer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine Baltimore, MD, USA ; Department of Psychiatry, University of Maryland School of Medicine Baltimore, MD, USA
| |
Collapse
|
238
|
Vaaga CE, Borisovska M, Westbrook GL. Dual-transmitter neurons: functional implications of co-release and co-transmission. Curr Opin Neurobiol 2014; 29:25-32. [PMID: 24816154 DOI: 10.1016/j.conb.2014.04.010] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 04/16/2014] [Accepted: 04/17/2014] [Indexed: 11/18/2022]
Abstract
Co-transmission, the ability of a neuron to release multiple transmitters, has long been recognized in selected circuits. However, the release of multiple primary neurotransmitters from a single neuron is only beginning to be appreciated. Here we consider recent examples of co-transmission as well as co-release-the packaging of multiple neurotransmitters into a single vesicle. The properties associated with each mode of release greatly enhance the possible action of such neurons within circuits. The functional importance of dual- (or multi-) transmitter neurons extends beyond actions on postsynaptic receptors, due in part to differential spatial and temporal profiles of each neurotransmitter. Recent evidence also suggests that the dual-transmitter phenotype can be dynamically regulated during development and following injury or disease.
Collapse
Affiliation(s)
- Christopher E Vaaga
- Neuroscience Graduate Program, Oregon Health and Science University, Portland, OR, USA; Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Maria Borisovska
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Gary L Westbrook
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
239
|
Nordenankar K, Smith-Anttila CJA, Schweizer N, Viereckel T, Birgner C, Mejia-Toiber J, Morales M, Leao RN, Wallén-Mackenzie Å. Increased hippocampal excitability and impaired spatial memory function in mice lacking VGLUT2 selectively in neurons defined by tyrosine hydroxylase promoter activity. Brain Struct Funct 2014; 220:2171-90. [PMID: 24802380 PMCID: PMC4481332 DOI: 10.1007/s00429-014-0778-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 04/11/2014] [Indexed: 02/01/2023]
Abstract
Three populations of neurons expressing the vesicular glutamate transporter 2 (Vglut2) were recently described in the A10 area of the mouse midbrain, of which two populations were shown to express the gene encoding, the rate-limiting enzyme for catecholamine synthesis, tyrosine hydroxylase (TH).One of these populations ("TH-Vglut2 Class1") also expressed the dopamine transporter (DAT) gene while one did not ("TH-Vglut2 Class2"), and the remaining population did not express TH at all ("Vglut2-only"). TH is known to be expressed by a promoter which shows two phases of activation, a transient one early during embryonal development, and a later one which gives rise to stable endogenous expression of the TH gene. The transient phase is, however, not specific to catecholaminergic neurons, a feature taken to advantage here as it enabled Vglut2 gene targeting within all three A10 populations expressing this gene, thus creating a new conditional knockout. These knockout mice showed impairment in spatial memory function. Electrophysiological analyses revealed a profound alteration of oscillatory activity in the CA3 region of the hippocampus. In addition to identifying a novel role for Vglut2 in hippocampus function, this study points to the need for improved genetic tools for targeting of the diversity of subpopulations of the A10 area.
Collapse
Affiliation(s)
- Karin Nordenankar
- Unit of Functional Neurobiology and Unit of Developmental Genetics, Biomedical Center, Department of Neuroscience, Uppsala University, Box 593, S-751 24, Uppsala, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
240
|
Tritsch NX, Oh WJ, Gu C, Sabatini BL. Midbrain dopamine neurons sustain inhibitory transmission using plasma membrane uptake of GABA, not synthesis. eLife 2014; 3:e01936. [PMID: 24843012 PMCID: PMC4001323 DOI: 10.7554/elife.01936] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Synaptic transmission between midbrain dopamine neurons and target neurons in the striatum is essential for the selection and reinforcement of movements. Recent evidence indicates that nigrostriatal dopamine neurons inhibit striatal projection neurons by releasing a neurotransmitter that activates GABAA receptors. Here, we demonstrate that this phenomenon extends to mesolimbic afferents, and confirm that the released neurotransmitter is GABA. However, the GABA synthetic enzymes GAD65 and GAD67 are not detected in midbrain dopamine neurons. Instead, these cells express the membrane GABA transporters mGAT1 (Slc6a1) and mGAT4 (Slc6a11) and inhibition of these transporters prevents GABA co-release. These findings therefore indicate that GABA co-release is a general feature of midbrain dopaminergic neurons that relies on GABA uptake from the extracellular milieu as opposed to de novo synthesis. This atypical mechanism may confer dopaminergic neurons the flexibility to differentially control GABAergic transmission in a target-dependent manner across their extensive axonal arbors. DOI:http://dx.doi.org/10.7554/eLife.01936.001 The electrical signals that are fired along neurons cannot be transmitted across the small gaps, called synapses that are found between neurons. Instead, the neuron sending the signal releases chemicals called neurotransmitters into the synapse. These neurotransmitters bind to receptor proteins on the surface of the second neuron and control how it fires. A neurotransmitter called dopamine plays a key role in the circuits of the brain that control how we learn certain tasks involving movement. In particular, two populations of neurons from the midbrain that release dopamine target the striatum, an area of the brain that is responsible for motor control. These neurons also release other neurotransmitters, but the identity of these other chemicals is not known, and the details of the interaction between the neurons and the striatum are poorly understood. Previous research showed that some of the midbrain neurons activate receptors that normally respond to a neurotransmitter called gamma-aminobutyric acid (GABA). However, several different chemicals can trigger this receptor. Using a range of techniques, Tritsch et al. now confirm that dopamine neurons release GABA alongside dopamine, and that this applies to both sets of the dopamine-producing neurons that feed into the striatum. Some neurons can manufacture GABA from amino acids found in their internal fluid. However, Tritsch et al. could not detect the enzymes needed for this reaction in dopamine-producing neurons. Instead, these neurons contain proteins that can transport GABA across the cell membrane, which suggests that the neurons collect GABA from the extracellular fluid that surrounds them. DOI:http://dx.doi.org/10.7554/eLife.01936.002
Collapse
Affiliation(s)
- Nicolas X Tritsch
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| | - Won-Jong Oh
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Chenghua Gu
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Bernardo L Sabatini
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| |
Collapse
|
241
|
Chuhma N, Mingote S, Moore H, Rayport S. Dopamine neurons control striatal cholinergic neurons via regionally heterogeneous dopamine and glutamate signaling. Neuron 2014; 81:901-12. [PMID: 24559678 DOI: 10.1016/j.neuron.2013.12.027] [Citation(s) in RCA: 169] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2013] [Indexed: 12/12/2022]
Abstract
Midbrain dopamine neurons fire in bursts conveying salient information. Bursts are associated with pauses in tonic firing of striatal cholinergic interneurons. Although the reciprocal balance of dopamine and acetylcholine in the striatum is well known, how dopamine neurons control cholinergic neurons has not been elucidated. Here, we show that dopamine neurons make direct fast dopaminergic and glutamatergic connections with cholinergic interneurons, with regional heterogeneity. Dopamine neurons drive a burst-pause firing sequence in cholinergic interneurons in the medial shell of the nucleus accumbens, mixed actions in the accumbens core, and a pause in the dorsal striatum. This heterogeneity is due mainly to regional variation in dopamine-neuron glutamate cotransmission. A single dose of amphetamine attenuates dopamine neuron connections to cholinergic interneurons with dose-dependent regional specificity. Overall, the present data indicate that dopamine neurons control striatal circuit function via discrete, plastic connections with cholinergic interneurons.
Collapse
Affiliation(s)
- Nao Chuhma
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, NY 10032, USA.
| | - Susana Mingote
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, NY 10032, USA
| | - Holly Moore
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Department of Integrative Neuroscience, NYS Psychiatric Institute, New York, NY 10032, USA
| | - Stephen Rayport
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, NY 10032, USA.
| |
Collapse
|
242
|
Glutamate and dopamine transmission from midbrain dopamine neurons share similar release properties but are differentially affected by cocaine. J Neurosci 2014; 34:3183-92. [PMID: 24573277 DOI: 10.1523/jneurosci.4958-13.2014] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Synaptic transmission between ventral tegmental area and nucleus accumbens (NAc) is critically involved in reward-motivated behaviors and thought to be altered in addiction. In addition to dopamine (DA), glutamate is packaged and released by a subset of mesolimbic DA neurons, eliciting EPSCs onto medium spiny neurons in NAc. Little is known about the properties and modulation of glutamate release from DA midbrain terminals and the effect of cocaine. Using an optogenetic approach to selectively activate midbrain DA fibers, we compared the properties and modulation of DA transients and EPSCs measured using fast-scan cyclic voltammetry and whole-cell recordings in mouse brain slices. DA transients and EPSCs were inhibited by DA receptor D2R agonist and showed a marked paired-pulse depression that required 2 min for full recovery. Cocaine depressed EPSCs amplitude by 50% but enhanced the overall DA transmission from midbrain DA neurons. AMPA and NMDA receptor-mediated EPSCs were equally inhibited by cocaine, suggesting a presynaptic mechanism of action. Pharmacological blockage and genetic deletion of D2R in DA neurons prevented the cocaine-induced inhibition of EPSCs and caused a larger increase in DA transient peak, confirming the involvement of presynaptic D2R. These findings demonstrate that acute cocaine inhibits DA and glutamate release from midbrain DA neurons via presynaptic D2R but has differential overall effects on their transmissions in the NAc. We postulate that cocaine, by blocking DA reuptake, prolongs DA transients and facilitates the feedback inhibition of DA and glutamate release from these terminals.
Collapse
|
243
|
Yetnikoff L, Lavezzi HN, Reichard RA, Zahm DS. An update on the connections of the ventral mesencephalic dopaminergic complex. Neuroscience 2014; 282:23-48. [PMID: 24735820 DOI: 10.1016/j.neuroscience.2014.04.010] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 04/03/2014] [Accepted: 04/04/2014] [Indexed: 12/21/2022]
Abstract
This review covers the intrinsic organization and afferent and efferent connections of the midbrain dopaminergic complex, comprising the substantia nigra, ventral tegmental area and retrorubral field, which house, respectively, the A9, A10 and A8 groups of nigrostriatal, mesolimbic and mesocortical dopaminergic neurons. In addition, A10dc (dorsal, caudal) and A10rv (rostroventral) extensions into, respectively, the ventrolateral periaqueductal gray and supramammillary nucleus are discussed. Associated intrinsic and extrinsic connections of the midbrain dopaminergic complex that utilize gamma-aminobutyric acid (GABA), glutamate and neuropeptides and various co-expressed combinations of these compounds are considered in conjunction with the dopamine-containing systems. A framework is provided for understanding the organization of massive afferent systems descending and ascending to the midbrain dopaminergic complex from the telencephalon and brainstem, respectively. Within the context of this framework, the basal ganglia direct and indirect output pathways are treated in some detail. Findings from rodent brain are briefly compared with those from primates, including humans. Recent literature is emphasized, including traditional experimental neuroanatomical and modern gene transfer and optogenetic studies. An attempt was made to provide sufficient background and cite a representative sampling of earlier primary papers and reviews so that people new to the field may find this to be a relatively comprehensive treatment of the subject.
Collapse
Affiliation(s)
- L Yetnikoff
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, 1402 S. Grand Boulevard, Saint Louis, MO 63104, United States.
| | - H N Lavezzi
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, 1402 S. Grand Boulevard, Saint Louis, MO 63104, United States
| | - R A Reichard
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, 1402 S. Grand Boulevard, Saint Louis, MO 63104, United States
| | - D S Zahm
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, 1402 S. Grand Boulevard, Saint Louis, MO 63104, United States.
| |
Collapse
|
244
|
Gagnon D, Parent M. Distribution of VGLUT3 in highly collateralized axons from the rat dorsal raphe nucleus as revealed by single-neuron reconstructions. PLoS One 2014; 9:e87709. [PMID: 24504335 PMCID: PMC3913638 DOI: 10.1371/journal.pone.0087709] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 01/03/2014] [Indexed: 12/11/2022] Open
Abstract
This study aimed at providing the first detailed morphological description, at the single-cell level, of the rat dorsal raphe nucleus neurons, including the distribution of the VGLUT3 protein within their axons. Electrophysiological guidance procedures were used to label dorsal raphe nucleus neurons with biotinylated dextran amine. The somatodendritic and axonal arborization domains of labeled neurons were reconstructed entirely from serial sagittal sections using a computerized image analysis system. Under anaesthesia, dorsal raphe nucleus neurons display highly regular (1.72±0.50 Hz) spontaneous firing patterns. They have a medium size cell body (9.8±1.7 µm) with 2–4 primary dendrites mainly oriented anteroposteriorly. The ascending axons of dorsal raphe nucleus are all highly collateralized and widely distributed (total axonal length up to 18.7 cm), so that they can contact, in various combinations, forebrain structures as diverse as the striatum, the prefrontal cortex and the amygdala. Their morphological features and VGLUT3 content vary significantly according to their target sites. For example, high-resolution confocal analysis of the distribution of VGLUT3 within individually labeled-axons reveals that serotonin axon varicosities displaying VGLUT3 are larger (0.74±0.03 µm) than those devoid of this protein (0.55±0.03 µm). Furthermore, the percentage of axon varicosities that contain VGLUT3 is higher in the striatum (93%) than in the motor cortex (75%), suggesting that a complex trafficking mechanism of the VGLUT3 protein is at play within highly collateralized axons of the dorsal raphe nucleus neurons. Our results provide the first direct evidence that the dorsal raphe nucleus ascending projections are composed of widely distributed neuronal systems, whose capacity to co-release serotonin and glutamate varies from one forebrain locus to the other.
Collapse
Affiliation(s)
- Dave Gagnon
- Centre de recherche de l’Institut universitaire en santé mentale de Québec, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC, Canada
| | - Martin Parent
- Centre de recherche de l’Institut universitaire en santé mentale de Québec, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC, Canada
- * E-mail:
| |
Collapse
|
245
|
Trudeau LE, Hnasko TS, Wallén-Mackenzie A, Morales M, Rayport S, Sulzer D. The multilingual nature of dopamine neurons. PROGRESS IN BRAIN RESEARCH 2014; 211:141-64. [PMID: 24968779 DOI: 10.1016/b978-0-444-63425-2.00006-4] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The ability of dopamine (DA) neurons to release other transmitters in addition to DA itself has been increasingly recognized, hence the concept of their multilingual nature. A subset of DA neurons, mainly found in the ventral tegmental area, express VGLUT2, allowing them to package and release glutamate onto striatal spiny projection neurons and cholinergic interneurons. Some dopaminergic axon terminals release GABA. Glutamate release by DA neurons has a developmental role, facilitating axonal growth and survival, and may determine in part the critical contribution of the ventral striatum to psychostimulant-induced behavior. Vesicular glutamate coentry may have synergistic effects on vesicular DA filling. The multilingual transmission of DA neurons across multiple striatal domains and the increasing insight into the role of glutamate cotransmission in the ventral striatum highlight the importance of analyzing DA neuron transmission at the synaptic level.
Collapse
Affiliation(s)
- Louis-Eric Trudeau
- Department of Pharmacology, Neuroscience Research Group, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada; Department of Neurosciences, Neuroscience Research Group, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada.
| | - Thomas S Hnasko
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Asa Wallén-Mackenzie
- Unit of Functional Neurobiology, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Marisela Morales
- National Institute on Drug Abuse, Intramural Research Program, Neuronal Networks Section, Baltimore, MD, USA
| | - Steven Rayport
- Department of Psychiatry, Columbia University, New York, NY, USA; Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, NY, USA
| | - David Sulzer
- Department of Psychiatry, Columbia University, New York, NY, USA; Department of Neurology, Columbia University, New York, NY, USA; Department of Pharmacology, Columbia University, New York, NY, USA; Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, NY, USA
| |
Collapse
|
246
|
Delwig A, Majumdar S, Ahern K, LaVail MM, Edwards R, Hnasko TS, Copenhagen DR. Glutamatergic neurotransmission from melanopsin retinal ganglion cells is required for neonatal photoaversion but not adult pupillary light reflex. PLoS One 2013; 8:e83974. [PMID: 24391855 PMCID: PMC3877098 DOI: 10.1371/journal.pone.0083974] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 11/11/2013] [Indexed: 12/29/2022] Open
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.
Collapse
Affiliation(s)
- Anton Delwig
- Department of Ophthalmology, University of California San Francisco, San Francisco, California, United States of America
| | - Sriparna Majumdar
- Department of Ophthalmology, University of California San Francisco, San Francisco, California, United States of America
| | - Kelly Ahern
- Department of Anatomy, University of California San Francisco, San Francisco, California United States of America
| | - Matthew M. LaVail
- Department of Ophthalmology, University of California San Francisco, San Francisco, California, United States of America
- Department of Anatomy, University of California San Francisco, San Francisco, California United States of America
| | - Robert Edwards
- Department of Physiology, University of California San Francisco, San Francisco, California, United States of America
- Department of Neurology, University of California, San Francisco San Francisco, California, United States of America
| | - Thomas S. Hnasko
- Department of Neurology, University of California, San Francisco San Francisco, California, United States of America
- Department of Neurosciences, University of California San Diego, San Diego, California, United States of America
| | - David R. Copenhagen
- Department of Ophthalmology, University of California San Francisco, San Francisco, California, United States of America
- Department of Physiology, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
247
|
Glycine transporter-1 inhibition promotes striatal axon sprouting via NMDA receptors in dopamine neurons. J Neurosci 2013; 33:16778-89. [PMID: 24133278 DOI: 10.1523/jneurosci.3041-12.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
NMDA receptor activity is involved in shaping synaptic connections throughout development and adulthood. We recently reported that brief activation of NMDA receptors on cultured ventral midbrain dopamine neurons enhanced their axon growth rate and induced axonal branching. To test whether this mechanism was relevant to axon regrowth in adult animals, we examined the reinnervation of dorsal striatum following nigral dopamine neuron loss induced by unilateral intrastriatal injections of the toxin 6-hydroxydopamine. We used a pharmacological approach to enhance NMDA receptor-dependent signaling by treatment with an inhibitor of glycine transporter-1 that elevates levels of extracellular glycine, a coagonist required for NMDA receptor activation. All mice displayed sprouting of dopaminergic axons from spared fibers in the ventral striatum to the denervated dorsal striatum at 7 weeks post-lesion, but the reinnervation in mice treated for 4 weeks with glycine uptake inhibitor was approximately twice as dense as in untreated mice. The treated mice also displayed higher levels of striatal dopamine and a complete recovery from lateralization in a test of sensorimotor behavior. We confirmed that the actions of glycine uptake inhibition on reinnervation and behavioral recovery required NMDA receptors in dopamine neurons using targeted deletion of the NR1 NMDA receptor subunit in dopamine neurons. Glycine transport inhibitors promote functionally relevant sprouting of surviving dopamine axons and could provide clinical treatment for disorders such as Parkinson's disease.
Collapse
|
248
|
Münster-Wandowski A, Gómez-Lira G, Gutiérrez R. Mixed neurotransmission in the hippocampal mossy fibers. Front Cell Neurosci 2013; 7:210. [PMID: 24319410 PMCID: PMC3837298 DOI: 10.3389/fncel.2013.00210] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/23/2013] [Indexed: 01/14/2023] Open
Abstract
The hippocampal mossy fibers (MFs), the axons of the granule cells (GCs) of the dentate gyrus, innervate mossy cells and interneurons in the hilus on their way to CA3 where they innervate interneurons and pyramidal cells. Synapses on each target cell have distinct anatomical and functional characteristics. In recent years, the paradigmatic view of the MF synapses being only glutamatergic and, thus, excitatory has been questioned. Several laboratories have provided data supporting the hypothesis that the MFs can transiently release GABA during development and, in the adult, after periods of enhanced excitability. This transient glutamate-GABA co-transmission coincides with the transient up-regulation of the machinery for the synthesis and release of GABA in the glutamatergic GCs. Although some investigators have deemed this evidence controversial, new data has appeared with direct evidence of co-release of glutamate and GABA from single, identified MF boutons. However, this must still be confirmed by other groups and with other methodologies. A second, intriguing observation is that MF activation produced fast spikelets followed by excitatory postsynaptic potentials in a number of pyramidal cells, which, unlike the spikelets, underwent frequency potentiation and were strongly depressed by activation of metabotropic glutamate receptors. The spikelets persisted during blockade of chemical transmission and were suppressed by the gap junction blocker carbenoxolone. These data are consistent with the hypothesis of mixed electrical-chemical synapses between MFs and some pyramidal cells. Dye coupling between these types of principal cells and ultrastructural studies showing the co-existence of AMPA receptors and connexin 36 in this synapse corroborate their presence. A deeper consideration of mixed neurotransmission taking place in this synapse may expand our search and understanding of communication channels between different regions of the mammalian CNS.
Collapse
|
249
|
Abbott SBG, Holloway BB, Viar KE, Guyenet PG. Vesicular glutamate transporter 2 is required for the respiratory and parasympathetic activation produced by optogenetic stimulation of catecholaminergic neurons in the rostral ventrolateral medulla of mice in vivo. Eur J Neurosci 2013; 39:98-106. [PMID: 24236954 DOI: 10.1111/ejn.12421] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 09/17/2013] [Accepted: 10/11/2013] [Indexed: 11/27/2022]
Abstract
Catecholaminergic neurons of the rostral ventrolateral medulla (RVLM-CA neurons; C1 neurons) contribute to the sympathetic, parasympathetic and neuroendocrine responses elicited by physical stressors such as hypotension, hypoxia, hypoglycemia, and infection. Most RVLM-CA neurons express vesicular glutamate transporter (VGLUT)2, and may use glutamate as a ionotropic transmitter, but the importance of this mode of transmission in vivo is uncertain. To address this question, we genetically deleted VGLUT2 from dopamine-β-hydroxylase-expressing neurons in mice [DβH(Cre/0) ;VGLUT2(flox/flox) mice (cKO mice)]. We compared the in vivo effects of selectively stimulating RVLM-CA neurons in cKO vs. control mice (DβH(Cre/0) ), using channelrhodopsin-2 (ChR2-mCherry) optogenetics. ChR2-mCherry was expressed by similar numbers of rostral ventrolateral medulla (RVLM) neurons in each strain (~400 neurons), with identical selectivity for catecholaminergic neurons (90-99% colocalisation with tyrosine hydroxylase). RVLM-CA neurons had similar morphology and axonal projections in DβH(Cre/0) and cKO mice. Under urethane anesthesia, photostimulation produced a similar pattern of activation of presumptive ChR2-positive RVLM-CA neurons in DβH(Cre/0) and cKO mice. Photostimulation in conscious mice produced frequency-dependent respiratory activation in DβH(Cre/0) mice but no effect in cKO mice. Similarly, photostimulation under urethane anesthesia strongly activated efferent vagal nerve activity in DβH(Cre/0) mice only. Vagal responses were unaffected by α1 -adrenoreceptor blockade. In conclusion, two responses evoked by RVLM-CA neuron stimulation in vivo require the expression of VGLUT2 by these neurons, suggesting that the acute autonomic responses driven by RVLM-CA neurons are mediated by glutamate.
Collapse
Affiliation(s)
- Stephen B G Abbott
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | | | | | | |
Collapse
|
250
|
Laminar and columnar development of barrel cortex relies on thalamocortical neurotransmission. Neuron 2013; 79:970-86. [PMID: 24012009 DOI: 10.1016/j.neuron.2013.06.043] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2013] [Indexed: 12/21/2022]
Abstract
A dynamic interplay between intrinsic regional molecular cues and extrinsic factors from the thalamus shape multiple features of early cortical development. It remains uncertain and controversial, however, whether the initial formation of cortical columns depends on neuronal activity, and there is little evidence that cortical lamination or neuronal differentiation is influenced by extrinsic activity. We examined the role of thalamic-derived factors in cortical development by selectively eliminating glutamatergic synaptic transmission from thalamocortical neurons in mice and found that eliminating thalamocortical neurotransmission prevented the formation of "barrel" columns in somatosensory cortex. Interestingly, based on cytoarchitectonic criteria and genetic markers, blocking thalamocortical neurotransmission also perturbed the development of superficial cortical lamina and the morphologic development of neurons. These experiments demonstrate that barrels and aspects of the layer-dependent pattern of cortical cytoarchitecture, gene expression, and neuronal differentiation depend on thalamocortical neurotransmission, extending the apparent influence of extrinsic, presumably activity-dependent factors, on cortical development.
Collapse
|