1
|
Düdükcü Ö, Raj DDA, van de Haar LL, Grossouw LM, Linders LE, Garritsen O, Adolfs Y, van Kronenburg NCH, Broekhoven MH, Kapteijns THW, Meye FJ, Pasterkamp RJ. Molecular diversity and migration of GABAergic neurons in the developing ventral midbrain. iScience 2024; 27:111239. [PMID: 39569362 PMCID: PMC11576407 DOI: 10.1016/j.isci.2024.111239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/30/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
Dopaminergic neurons in the ventral midbrain (mDA) are surrounded by GABAergic neurons. The full extent of GABAergic neuron subtypes occupying this region and the mechanisms that underlie their development and function are largely unknown. Therefore, we performed single-cell RNA sequencing (scRNA-seq) of fluorescence-activated cell sorting (FACS)-isolated GABAergic neurons in the developing mouse ventral midbrain. Several distinct GABAergic neuron subtypes were identified based on transcriptomic profiles and spatially assigned to the ventral midbrain using in situ hybridization and immunohistochemistry for specific markers. A subset of GABAergic clusters that co-expressed mDA markers was studied in more detail and showed distinctive molecular, functional, and wiring properties. Finally, migration of different GABAergic neuron subtypes required netrin-1 from different cellular sources acting via distinct receptor mechanisms. Overall, our work provides insight into the heterogeneity and spatial organization of GABAergic neurons in the developing ventral midbrain and begins to dissect the mechanisms that underlie their development.
Collapse
Affiliation(s)
- Özge Düdükcü
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Divya D A Raj
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Lieke L van de Haar
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Laurens M Grossouw
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Louisa E Linders
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Oxana Garritsen
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Nicky C H van Kronenburg
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Mark H Broekhoven
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Troy H W Kapteijns
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Frank J Meye
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584 CG Utrecht, the Netherlands
| |
Collapse
|
2
|
Barbano MF, Qi J, Chen E, Mohammad U, Espinoza O, Candido M, Wang H, Liu B, Hahn S, Vautier F, Morales M. VTA glutamatergic projections to the nucleus accumbens suppress psychostimulant-seeking behavior. Neuropsychopharmacology 2024; 49:1905-1915. [PMID: 38926603 PMCID: PMC11473768 DOI: 10.1038/s41386-024-01905-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024]
Abstract
Converging evidence indicates that both dopamine and glutamate neurotransmission within the nucleus accumbens (NAc) play a role in psychostimulant self-administration and relapse in rodent models. Increased NAc dopamine release from ventral tegmental area (VTA) inputs is critical to psychostimulant self-administration and NAc glutamate release from prelimbic prefrontal cortex (PFC) inputs synapsing on medium spiny neurons (MSNs) is critical to reinstatement of psychostimulant-seeking after extinction. The regulation of the activity of MSNs by VTA dopamine inputs has been extensively studied, and recent findings have demonstrated that VTA glutamate neurons target the NAc medial shell. Here, we determined whether the mesoaccumbal glutamatergic pathway plays a role in psychostimulant conditioned place preference and self-administration in mice. We used optogenetics to induce NAc release of glutamate from VTA inputs during the acquisition, expression, and reinstatement phases of cocaine- or methamphetamine-induced conditioned place preference (CPP), and during priming-induced reinstatement of cocaine-seeking behavior. We found that NAc medial shell release of glutamate resulting from the activation of VTA glutamatergic fibers did not affect the acquisition of cocaine-induced CPP, but it blocked the expression, stress- and priming-induced reinstatement of cocaine- and methamphetamine CPP, as well as it blocked the priming-induced reinstatement of cocaine-seeking behavior after extinction. These findings indicate that in contrast to the well-recognized mesoaccumbal dopamine system that is critical to psychostimulant reward and relapse, there is a parallel mesoaccumbal glutamatergic system that suppresses reward and psychostimulant-seeking behavior.
Collapse
Affiliation(s)
- M Flavia Barbano
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Jia Qi
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Emma Chen
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Uzma Mohammad
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Orlando Espinoza
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Marcos Candido
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Huiling Wang
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Bing Liu
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Suyun Hahn
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - François Vautier
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Marisela Morales
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
3
|
Beauséjour PA, Veilleux JC, Condamine S, Zielinski BS, Dubuc R. Olfactory Projections to Locomotor Control Centers in the Sea Lamprey. Int J Mol Sci 2024; 25:9370. [PMID: 39273317 PMCID: PMC11395479 DOI: 10.3390/ijms25179370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Although olfaction is well known to guide animal behavior, the neural circuits underlying the motor responses elicited by olfactory inputs are poorly understood. In the sea lamprey, anatomical evidence shows that olfactory inputs project to the posterior tuberculum (PT), a structure containing dopaminergic (DA) neurons homologous to the mammalian ventral tegmental area and the substantia nigra pars compacta. Olfactory inputs travel directly from the medial olfactory bulb (medOB) or indirectly through the main olfactory bulb and the lateral pallium (LPal). Here, we characterized the transmission of olfactory inputs to the PT in the sea lamprey, Petromyzon marinus. Abundant projections from the medOB were observed close to DA neurons of the PT. Moreover, electrophysiological experiments revealed that PT neurons are activated by both the medOB and LPal, and calcium imaging indicated that the olfactory signal is then relayed to the mesencephalic locomotor region to initiate locomotion. In semi-intact preparations, stimulation of the medOB and LPal induced locomotion that was tightly associated with neural activity in the PT. Moreover, PT neurons were active throughout spontaneously occurring locomotor bouts. Altogether, our observations suggest that the medOB and LPal convey olfactory inputs to DA neurons of the PT, which in turn activate the brainstem motor command system to elicit locomotion.
Collapse
Affiliation(s)
| | - Jean-Christophe Veilleux
- Research Group in Adapted Physical Activity, Department of Exercise Sciences, Faculty of Sciences, University of Quebec in Montreal, Montreal, QC H2X 1Y4, Canada
| | - Steven Condamine
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Barbara S Zielinski
- Department of Integrative Biology, Faculty of Science, University of Windsor, Windsor, ON N9B 3P4, Canada
| | - Réjean Dubuc
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
- Research Group in Adapted Physical Activity, Department of Exercise Sciences, Faculty of Sciences, University of Quebec in Montreal, Montreal, QC H2X 1Y4, Canada
| |
Collapse
|
4
|
McGovern DJ, Phillips A, Ly A, Prévost ED, Ward L, Siletti K, Kim YS, Fenno LE, Ramakrishnan C, Deisseroth K, Ford CP, Root DH. Salience signaling and stimulus scaling of ventral tegmental area glutamate neuron subtypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598688. [PMID: 38915564 PMCID: PMC11195246 DOI: 10.1101/2024.06.12.598688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Ventral tegmental area (VTA) glutamatergic neurons participate in reward, aversion, drug-seeking, and stress. Subsets of VTA VGluT2+ neurons are capable of co-transmitting glutamate and GABA (VGluT2+VGaT+ neurons), transmitting glutamate without GABA (VGluT2+VGaT- neurons), or co-transmitting glutamate and dopamine (VGluT2+TH+ neurons), but whether these molecularly distinct subpopulations show behavior-related differences is not wholly understood. We identified that neuronal activity of each VGluT2+ subpopulation is sensitive to reward value but signaled this in different ways. The phasic maximum activity of VGluT2+VGaT+ neurons increased with sucrose concentration, whereas VGluT2+VGaT- neurons increased maximum and sustained activity with sucrose concentration, and VGluT2+TH+ neurons increased sustained but not maximum activity with sucrose concentration. Additionally, VGluT2+ subpopulations signaled consummatory preferences in different ways. VGluT2+VGaT- neurons and VGluT2+TH+ neurons showed a signaling preference for a behaviorally-preferred fat reward over sucrose, but in temporally-distinct ways. In contrast, VGluT2+VGaT+ neurons uniquely signaled a less behaviorally-preferred sucrose reward compared with fat. Further experiments suggested that VGluT2+VGaT+ consummatory reward-related activity was related to sweetness, partially modulated by hunger state, and not dependent on caloric content or behavioral preference. All VGluT2+ subtypes increased neuronal activity following aversive stimuli but VGluT2+VGaT+ neurons uniquely scaled their magnitude and sustained activity with footshock intensity. Optogenetic activation of VGluT2+VGaT+ neurons during low intensity footshock enhanced fear-related behavior without inducing place preference or aversion. We interpret these data such that VTA glutamatergic subpopulations signal different elements of rewarding and aversive experiences and highlight the unique role of VTA VGluT2+VGaT+ neurons in enhancing the salience of behavioral experiences.
Collapse
Affiliation(s)
- Dillon J. McGovern
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301
| | - Alysabeth Phillips
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301
| | - Annie Ly
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301
| | - Emily D. Prévost
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301
| | - Lucy Ward
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301
| | - Kayla Siletti
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301
| | - Yoon Seok Kim
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Lief E. Fenno
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
- Current address: Department of Neuroscience, Dell Medical School, The University of Texas at Austin 78712
| | - Charu Ramakrishnan
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Christopher P. Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045
| | - David H. Root
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301
| |
Collapse
|
5
|
McGovern DJ, Ly A, Ecton KL, Huynh DT, Prévost ED, Gonzalez SC, McNulty CJ, Rau AR, Hentges ST, Daigle TL, Tasic B, Baratta MV, Root DH. Ventral tegmental area glutamate neurons mediate nonassociative consequences of stress. Mol Psychiatry 2024; 29:1671-1682. [PMID: 36437312 PMCID: PMC10375863 DOI: 10.1038/s41380-022-01858-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/29/2022]
Abstract
Exposure to trauma is a risk factor for the development of a number of mood disorders, and may enhance vulnerability to future adverse life events. Recent data demonstrate that ventral tegmental area (VTA) neurons expressing the vesicular glutamate transporter 2 (VGluT2) signal and causally contribute to behaviors that involve aversive or threatening stimuli. However, it is unknown whether VTA VGluT2 neurons regulate transsituational outcomes of stress and whether these neurons are sensitive to stressor controllability. This work adapted an operant mouse paradigm to examine the impact of stressor controllability on VTA VGluT2 neuron function as well as the role of VTA VGluT2 neurons in mediating transsituational stressor outcomes. Uncontrollable (inescapable) stress, but not physically identical controllable (escapable) stress, produced social avoidance and exaggerated fear in male mice. Uncontrollable stress in females led to exploratory avoidance of a novel brightly lit environment. Both controllable and uncontrollable stressors increased VTA VGluT2 neuronal activity, and chemogenetic silencing of VTA VGluT2 neurons prevented the behavioral sequelae of uncontrollable stress in male and female mice. Further, we show that stress activates multiple genetically-distinct subtypes of VTA VGluT2 neurons, especially those that are VGluT2+VGaT+, as well as lateral habenula neurons receiving synaptic input from VTA VGluT2 neurons. Our results provide causal evidence that mice can be used for identifying stressor controllability circuitry and that VTA VGluT2 neurons contribute to transsituational stressor outcomes, such as social avoidance, exaggerated fear, or anxiety-like behavior that are observed within trauma-related disorders.
Collapse
Affiliation(s)
- Dillon J McGovern
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US
| | - Annie Ly
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US
| | - Koy L Ecton
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US
| | - David T Huynh
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US
| | - Emily D Prévost
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US
| | - Shamira C Gonzalez
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US
| | - Connor J McNulty
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US
| | - Andrew R Rau
- Department of Biomedical Sciences, Colorado State University, 1617 Campus Delivery, Fort Collins, 80523, CO, US
- Center for Structural and Functional Neuroscience, Division of Biological Sciences, University of Montana, Missoula, 59812, MT, US
| | - Shane T Hentges
- Department of Biomedical Sciences, Colorado State University, 1617 Campus Delivery, Fort Collins, 80523, CO, US
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, 99164, WA, US
| | - Tanya L Daigle
- Allen Institute for Brain Science, 615 Westlake. Avenue North, Seattle, 98109, WA, US
| | - Bosiljka Tasic
- Allen Institute for Brain Science, 615 Westlake. Avenue North, Seattle, 98109, WA, US
| | - Michael V Baratta
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US.
| | - David H Root
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US.
| |
Collapse
|
6
|
Gao R, Schneider AM, Mulloy SM, Lee AM. Expression pattern of nicotinic acetylcholine receptor subunit transcripts in neurons and astrocytes in the ventral tegmental area and locus coeruleus. Eur J Neurosci 2024; 59:2225-2239. [PMID: 37539749 PMCID: PMC10838369 DOI: 10.1111/ejn.16109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 06/06/2023] [Accepted: 07/20/2023] [Indexed: 08/05/2023]
Abstract
Acetylcholine is the endogenous agonist for the neuronal nicotinic acetylcholine receptor (nAChR) system, which is involved in attention, memory, affective behaviours and substance use disorders. Brain nAChRs are highly diverse with 11 different subunits that can form multiple receptor subtypes, each with distinct receptor and pharmacological properties. Different neuronal cell types can also express different nAChR subtypes, resulting in highly complex cholinergic signalling. Identifying which nAChR subunit transcripts are expressed in cell types can provide an indication of which nAChR combinations are possible and which receptor subtypes may be most pharmacologically relevant to target. In addition to differences in expression across cell types, nAChRs also undergo changes in expression levels from adolescence to adulthood. In this study, we used fluorescent in situ hybridization to identify and quantify the expression of α4, α5, α6, β2 and β3 nAChR subunit transcripts in dopaminergic, GABAergic, glutamatergic and noradrenergic neurons and astrocytes in the ventral tegmental area (VTA) and locus coeruleus (LC) in adult and adolescent, male and female C57BL/6J mice. There were distinct differences in the pattern of nAChR subunit transcript expression between the two brain regions. LC noradrenergic neurons had high prevalence of α6, β2 and β3 expression, with very low expression of α4, suggesting the α6(non-α4)β2β3 receptor as a main subtype in these neurons. VTA astrocytes from adult mice showed greater prevalence of α5, α6, β2 and β3 transcript compared with adolescent mice. These data highlight the complex nAChR expression patterns across brain region and cell type.
Collapse
Affiliation(s)
- Runbo Gao
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Amelia M. Schneider
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sarah M. Mulloy
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | - Anna M. Lee
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
7
|
Seiler JL, Zhuang X, Nelson AB, Lerner TN. Dopamine across timescales and cell types: Relevance for phenotypes in Parkinson's disease progression. Exp Neurol 2024; 374:114693. [PMID: 38242300 DOI: 10.1016/j.expneurol.2024.114693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/21/2024]
Abstract
Dopamine neurons in the substantia nigra pars compacta (SNc) synthesize and release dopamine, a critical neurotransmitter for movement and learning. SNc dopamine neurons degenerate in Parkinson's Disease (PD), causing a host of motor and non-motor symptoms. Here, we review recent conceptual advances in our basic understanding of the dopamine system - including our rapidly advancing knowledge of dopamine neuron heterogeneity - with special attention to their importance for understanding PD. In PD patients, dopamine neuron degeneration progresses from lateral SNc to medial SNc, suggesting clinically relevant heterogeneity in dopamine neurons. With technical advances in dopamine system interrogation, we can understand the relevance of this heterogeneity for PD progression and harness it to develop new treatments.
Collapse
Affiliation(s)
- Jillian L Seiler
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Xiaowen Zhuang
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Alexandra B Nelson
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA; Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Talia N Lerner
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern University Interdepartmental Neuroscience Program (NUIN), Evanston, IL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
8
|
Fudge JL, Kelly EA, Love TM. Amygdalo-nigral inputs target dopaminergic and GABAergic neurons in the primate: a view from dendrites and soma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.16.575910. [PMID: 38293165 PMCID: PMC10827221 DOI: 10.1101/2024.01.16.575910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The central nucleus (CeN) of the amygdala is an important afferent to the DA system that mediates motivated learning. We previously found that CeN terminals in nonhuman primates primarily overlap the elongated lateral VTA (parabrachial pigmented nucleus, PBP, A10), and retrorubral field(A8) subregion. Here, we examined CeN afferent contacts on cell somata and proximal dendrites of DA and GABA neurons, and distal dendrites of each, using confocal and electron microscopy (EM) methods, respectively. At the soma/proximal dendrites, the proportion of TH+ and GAD1+ cells receiving at least one CeN afferent contact was surprisingly similar (TH = 0.55: GAD1=0.55 in PBP; TH = 0.56; GAD1 =0.51 in A8), with the vast majority of contacted TH+ and GAD1+ soma/proximal dendrites received 1-2 contacts. Similar numbers of tracer-labeled terminals also contacted TH-positive and GAD1-positive small dendrites and/or spines (39% of all contacted dendrites were either TH- or GAD1-labeled). Overall, axon terminals had more symmetric (putative inhibitory) axonal contacts with no difference in the relative distribution in the PBP versus A8, or onto TH+ versus GAD1+ dendrites/spines in either region. The striking uniformity in the amygdalonigral projection across the PBP-A8 terminal field suggests that neither neurotransmitter phenotype nor midbrain location dictates likelihood of a terminal contact. We discuss how this afferent uniformity can play out in recently discovered differences in DA:GABA cell densities between the PBP and A8, and affect specific outputs. Significance statement The amygdala's central nucleus (CeN) channels salient cues to influence both appetitive and aversive responses via DA outputs. In higher species, the broad CeN terminal field overlaps the parabrachial pigmented nucleus ('lateral A10') and the retrorubral field (A8). We quantified terminal contacts in each region on DA and GABAergic soma/proximal dendrites and small distal dendrites. There was striking uniformity in contacts on DA and GABAergic cells, regardless of soma and dendritic compartment, in both regions. Most contacts were symmetric (putative inhibitory) with little change in the ratio of inhibitory to excitatory contacts by region.We conclude that post-synaptic shifts in DA-GABA ratios are key to understanding how these relatively uniform inputs can produce diverse effects on outputs.
Collapse
|
9
|
Barbano MF, Zhang S, Chen E, Espinoza O, Mohammad U, Alvarez-Bagnarol Y, Liu B, Hahn S, Morales M. Lateral hypothalamic glutamatergic inputs to VTA glutamatergic neurons mediate prioritization of innate defensive behavior over feeding. Nat Commun 2024; 15:403. [PMID: 38195566 PMCID: PMC10776608 DOI: 10.1038/s41467-023-44633-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 12/20/2023] [Indexed: 01/11/2024] Open
Abstract
The lateral hypothalamus (LH) is involved in feeding behavior and defense responses by interacting with different brain structures, including the Ventral Tegmental Area (VTA). Emerging evidence indicates that LH-glutamatergic neurons infrequently synapse on VTA-dopamine neurons but preferentially establish multiple synapses on VTA-glutamatergic neurons. Here, we demonstrated that LH-glutamatergic inputs to VTA promoted active avoidance, long-term aversion, and escape attempts. By testing feeding in the presence of a predator, we observed that ongoing feeding was decreased, and that this predator-induced decrease in feeding was abolished by photoinhibition of the LH-glutamatergic inputs to VTA. By VTA specific neuronal ablation, we established that predator-induced decreases in feeding were mediated by VTA-glutamatergic neurons but not by dopamine or GABA neurons. Thus, we provided evidence for an unanticipated neuronal circuitry between LH-glutamatergic inputs to VTA-glutamatergic neurons that plays a role in prioritizing escape, and in the switch from feeding to escape in mice.
Collapse
Affiliation(s)
- M Flavia Barbano
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Shiliang Zhang
- Confocal and Electron Microscopy Core, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Emma Chen
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
- Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Orlando Espinoza
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Uzma Mohammad
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Yocasta Alvarez-Bagnarol
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
- Department of Anatomy and Neurobiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico, USA
| | - Bing Liu
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Suyun Hahn
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Marisela Morales
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
10
|
Kelly EA, Love TM, Fudge JL. Corticotropin-releasing factor-dopamine interactions in male and female macaque: Beyond the classic VTA. Synapse 2024; 78:e22284. [PMID: 37996987 PMCID: PMC10842953 DOI: 10.1002/syn.22284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 11/25/2023]
Abstract
Dopamine (DA) is involved in stress and stress-related illnesses, including many psychiatric disorders. Corticotropin-releasing factor (CRF) plays a role in stress responses and targets the ventral midbrain DA system, which is composed of DA and non-DA cells, and divided into specific subregions. Although CRF inputs to the midline A10 nuclei ("classic VTA") are known, in monkeys, CRF-containing terminals are also highly enriched in the expanded A10 parabrachial pigmented nucleus (PBP) and in the A8 retrorubral field subregions. We characterized CRF-labeled synaptic terminals on DA (tyrosine hydroxylase, TH+) and non-DA (TH-) cell types in the PBP and A8 regions using immunoreactive electron microscopy (EM) in male and female macaques. CRF labeling was present mostly in axon terminals, which mainly contacted TH-negative dendrites in both subregions. Most CRF-positive terminals had symmetric profiles. In both PBP and A8, CRF symmetric (putative inhibitory) synapses onto TH-negative dendrites were significantly greater than asymmetric (putative excitatory) profiles. This overall pattern was similar in males and females, despite shifts in the size of these effects between regions depending on sex. Because stress and gonadal hormone shifts can influence CRF expression, we also did hormonal assays over a 6-month time period and found little variability in basal cortisol across similarly housed animals at the same age. Together our findings suggest that at baseline, CRF-positive synaptic terminals in the primate PBP and A8 are poised to regulate DA indirectly through synaptic contacts onto non-DA neurons.
Collapse
Affiliation(s)
- E A Kelly
- Departments of Neuroscience, Del Monte Institute of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - T M Love
- Department of Biostatistics, Del Monte Institute of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - J L Fudge
- Departments of Neuroscience, Del Monte Institute of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Department of Psychiatry, Del Monte Institute of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
11
|
Barcomb K, Ford CP. Alterations in neurotransmitter co-release in Parkinson's disease. Exp Neurol 2023; 370:114562. [PMID: 37802381 PMCID: PMC10842357 DOI: 10.1016/j.expneurol.2023.114562] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/18/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
Parkinson's disease is a neurological disorder characterized by degeneration of midbrain dopamine neurons, which results in numerous adaptations in basal ganglia circuits. Research over the past twenty-five years has identified that midbrain dopamine neurons of the substantia nigra pars compacta (SNc) and ventral tegmental area (VTA) co-release multiple other transmitters including glutamate and GABA, in addition to their canonical transmitter, dopamine. This review summarizes previous work characterizing neurotransmitter co-release from dopamine neurons, work examining potential changes in co-release dynamics that result in animal models of Parkinson's disease, and future opportunities for determining how dysfunction in co-release may contribute to circuit dysfunction in Parkinson's disease.
Collapse
Affiliation(s)
- Kelsey Barcomb
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Christopher P Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
12
|
Strong CE, Zhang J, Carrasco M, Kundu S, Boutin M, Vishwasrao HD, Liu J, Medina A, Chen YC, Wilson K, Lee EM, Ferrer M. Functional brain region-specific neural spheroids for modeling neurological diseases and therapeutics screening. Commun Biol 2023; 6:1211. [PMID: 38017066 PMCID: PMC10684574 DOI: 10.1038/s42003-023-05582-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 11/14/2023] [Indexed: 11/30/2023] Open
Abstract
3D spheroids have emerged as powerful drug discovery tools given their high-throughput screening (HTS) compatibility. Here, we describe a method for generating functional neural spheroids by cell-aggregation of differentiated human induced pluripotent stem cell (hiPSC)-derived neurons and astrocytes at cell type compositions mimicking specific regions of the human brain. Recordings of intracellular calcium oscillations were used as functional assays, and the utility of this spheroids system was shown through disease modeling, drug testing, and formation of assembloids to model neurocircuitry. As a proof of concept, we generated spheroids incorporating neurons with Alzheimer's disease-associated alleles, as well as opioid use disorder modeling spheroids induced by chronic treatment of a mu-opioid receptor agonist. We reversed baseline functional deficits in each pilot disease model with clinically approved treatments and showed that assembloid activity can be chemogenetically manipulated. Here, we lay the groundwork for brain region-specific neural spheroids as a robust functional assay platform for HTS studies.
Collapse
Affiliation(s)
- Caroline E Strong
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Jiajing Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Martin Carrasco
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Srikanya Kundu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Molly Boutin
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Harshad D Vishwasrao
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jiamin Liu
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Angelica Medina
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Yu-Chi Chen
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Kelli Wilson
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Emily M Lee
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA.
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA.
| |
Collapse
|
13
|
Zell V, Teuns G, Needham AS, Mukherjee S, Roscoe N, Le M, Fourgeaud L, Woodruff G, Bhattacharya A, Marella M, Bonaventure P, Drevets WC, Balana B. Characterization of Selective M 5 Acetylcholine Muscarinic Receptor Modulators on Dopamine Signaling in the Striatum. J Pharmacol Exp Ther 2023; 387:226-234. [PMID: 37679045 DOI: 10.1124/jpet.123.001737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/04/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
The type-5 muscarinic acetylcholine receptor (mAChR, M5) is almost exclusively expressed in dopamine (DA) neurons of the ventral tegmental area and substantia nigra pars compacta; therefore, they are ideally located to modulate DA signaling and underlying behaviors. However, the role of M5 in shaping DA release is still poorly characterized. In this study, we first quantitatively mapped the expression of M5 in different neurons of the mouse midbrain, then used voltammetry in mouse striatum to evaluate the effect of M5-selective modulators on DA release. The M5 negative allosteric modulator ML375 significantly decreased electrically evoked DA release and blocked the effect of Oxotremorine-M (Oxo-M; nonselective mAChR agonist) on DA release in the presence of an acetylcholine nicotinic receptor blocker. Conversely, the M5 positive allosteric modulator VU 0365114 significantly increased electrically evoked DA release and the Oxo-M effect on DA release. We then assessed M5's impact on mesolimbic circuit function in vivo. Although psychostimulant-induced locomotor activity models in knockout mice have previously been used to characterize the role of M5 in DA transmission, the results of these studies conflict, leading us to select a different in vivo model, namely a cocaine self-administration paradigm. In contrast to a previous study that also used this model, in the current study, administration of ML375 did not decrease cocaine self-administration in rats (using fixed and progressive ratio). These conflicting results illustrate the complexity of M5 modulation and the need to further characterize its involvement in the regulation of dopamine signaling, central to multiple neuropsychiatric diseases. SIGNIFICANCE STATEMENT: This work describes the type-5 muscarinic receptor (M5) pattern of expression within the midbrain as well as its physiological modulation by selective compounds at the axon terminal level in the striatum, where M5 directly shapes dopamine transmission. It offers the first direct readout of mesolimbic dopamine release modulation by M5, highlighting its role in regulating neurocircuits implicated in the pathophysiology of neuropsychiatric disorders such as substance use disorders, major depressive disorder, and schizophrenia.
Collapse
Affiliation(s)
- Vivien Zell
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Greetje Teuns
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Alexandra Stormy Needham
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Sruti Mukherjee
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Nathaniel Roscoe
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Michelle Le
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Lawrence Fourgeaud
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Grace Woodruff
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Anindya Bhattacharya
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Mathieu Marella
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Pascal Bonaventure
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Wayne C Drevets
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Bartosz Balana
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| |
Collapse
|
14
|
Barker DJ, Zhang S, Wang H, Estrin DJ, Miranda-Barrientos J, Liu B, Kulkarni RJ, de Deus JL, Morales M. Lateral preoptic area glutamate neurons relay nociceptive information to the ventral tegmental area. Cell Rep 2023; 42:113029. [PMID: 37632750 PMCID: PMC10584074 DOI: 10.1016/j.celrep.2023.113029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/28/2023] [Accepted: 08/09/2023] [Indexed: 08/28/2023] Open
Abstract
The ventral tegmental area (VTA) has been proposed to play a role in pain, but the brain structures modulating VTA activity in response to nociceptive stimuli remain unclear. Here, we demonstrate that the lateral preoptic area (LPO) glutamate neurons relay nociceptive information to the VTA. These LPO glutamatergic neurons synapsing on VTA neurons respond to nociceptive stimulation and conditioned stimuli predicting nociceptive stimulation and also mediate aversion. In contrast, LPO GABA neurons synapsing in the VTA mediate reward. By ultrastructural quantitative synaptic analysis, ex vivo electrophysiology, and functional neuroanatomy we identify a complex circuitry between LPO glutamatergic and GABAergic neurons and VTA dopaminergic, GABAergic, and glutamatergic neurons. We conclude that LPO glutamatergic neurons play a causal role in the processing of nociceptive stimuli and in relaying information about nociceptive stimuli. The pathway from LPO glutamatergic neurons to the VTA represents an unpredicted interface between peripheral nociceptive information and the limbic system.
Collapse
Affiliation(s)
- David J Barker
- Integrative Neuroscience Branch, Neuronal Networks Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
| | - Shiliang Zhang
- Confocal and Electron Microscopy Core, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
| | - Huiling Wang
- Integrative Neuroscience Branch, Neuronal Networks Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
| | - David J Estrin
- Integrative Neuroscience Branch, Neuronal Networks Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
| | - Jorge Miranda-Barrientos
- Integrative Neuroscience Branch, Neuronal Networks Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
| | - Bing Liu
- Integrative Neuroscience Branch, Neuronal Networks Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
| | - Rucha J Kulkarni
- Integrative Neuroscience Branch, Neuronal Networks Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
| | - Junia Lara de Deus
- Integrative Neuroscience Branch, Neuronal Networks Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
| | - Marisela Morales
- Integrative Neuroscience Branch, Neuronal Networks Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA.
| |
Collapse
|
15
|
Devoto F, Ferrulli A, Banfi G, Luzi L, Zapparoli L, Paulesu E. How images of food become cravingly salient in obesity. Obesity (Silver Spring) 2023; 31:2294-2303. [PMID: 37605635 DOI: 10.1002/oby.23834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 08/23/2023]
Abstract
OBJECTIVE This case-control study was aimed at testing two main hypotheses: (i) obesity is characterized by neurofunctional alterations within the mesocorticolimbic reward system, a brain network originating from the midbrain ventral tegmental area (VTA); and (ii) these alterations are associated with a bias for food-related stimuli and craving. METHODS Normal-weight individuals and individuals with obesity underwent a resting-state functional magnetic resonance imaging scan and the assessment of impulsivity, food craving, appetite, and implicit bias for food and non-food stimuli. The VTA was used as a seed to map, for each participant, the strength of its functional connections with the rest of the brain. The between-group difference in functional connectivity was then computed, and brain-behavior correlations were performed. RESULTS Individuals with obesity showed hyper-connectivity of the VTA with part of the ventral occipitotemporal cortex, recently found to be specialized for food images, and hypo-connectivity with the left inferior frontal gyrus, devoted to cognitive control. VTA-ventral occipitotemporal cortex connectivity was positively associated with food craving and food-related bias; the reverse correlation was observed for VTA-inferior frontal gyrus connectivity. CONCLUSIONS These findings reveal that, in obesity, food-related visual stimuli become cravingly salient through an imbalanced connectivity of the reward system with sensory-specific regions and the frontal cortex involved in cognitive control.
Collapse
Affiliation(s)
| | - Anna Ferrulli
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Giuseppe Banfi
- IRCCS Orthopedic Institute Galeazzi, Milan, Italy
- University Vita e Salute San Raffaele, Milan, Italy
| | - Livio Luzi
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Laura Zapparoli
- Department of Psychology, University of Milano-Bicocca, Milan, Italy
- IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| | - Eraldo Paulesu
- Department of Psychology, University of Milano-Bicocca, Milan, Italy
- IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| |
Collapse
|
16
|
Brown NK, Roche JK, Farmer CB, Roberts RC. Evidence for upregulation of excitatory synaptic transmission in the substantia nigra in Schizophrenia: a postmortem ultrastructural study. J Neural Transm (Vienna) 2023; 130:561-573. [PMID: 36735096 DOI: 10.1007/s00702-023-02593-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/14/2023] [Indexed: 02/04/2023]
Abstract
The dopamine hypothesis of schizophrenia suggests that psychotic symptoms originate from dysregulation of dopaminergic activity, which may be controlled by upstream innervation. We hypothesized that we would find anatomical evidence for the hyperexcitability seen in the SN. We examined and quantified synaptic morphology, which correlates with function, in the postmortem substantia nigra (SN) from 15 schizophrenia and 12 normal subjects. Synapses were counted using stereological techniques and classified based on the morphology of the post-synaptic density (PSD) and the presence or absence of a presynaptic density. The density and proportion of excitatory synapses was higher in the schizophrenia group than in controls, while the proportion (but not density) of inhibitory synapses was lower. We also detected in the schizophrenia group an increase in density of synapses with a PSD of intermediate thickness, which may represent excitatory synapses. The density of synapses with presynaptic densities was similar in both groups. The density of synapses with mixed morphologies was higher in the schizophrenia group than in controls. The human SN contains atypical synaptic morphology. We found an excess amount and proportion of excitatory synapses in the SN in schizophrenia that could result in hyperactivity and drive the psychotic symptoms of schizophrenia. The sources of afferent excitatory inputs to the SN arise from the subthalamic nucleus, the pedunculopontine nucleus, and the ventral tegmental area (VTA), areas that could be the source of excess excitation. Synapses with mixed morphologies may represent inputs from the VTA, which release multiple transmitters.
Collapse
Affiliation(s)
- Nicole K Brown
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Sparks Center 835C, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
| | - Joy K Roche
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Sparks Center 835C, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
| | - Charlene B Farmer
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Sparks Center 835C, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
| | - Rosalinda C Roberts
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Sparks Center 835C, 1720 2nd Avenue South, Birmingham, AL, 35294, USA.
| |
Collapse
|
17
|
Neuronal Dot1l Activity Acts as a Mitochondrial Gene-Repressor Associated with Human Brain Aging via H3K79 Hypermethylation. Int J Mol Sci 2023; 24:ijms24021387. [PMID: 36674903 PMCID: PMC9862808 DOI: 10.3390/ijms24021387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/02/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
Methylation of histone 3 at lysine 79 (H3K79) and its catalyst, a disrupter of telomeric silencing (DOT1l), have been coupled to multiple forms of stress, such as bioenergetic and ER challenges. However, studies on H3K79 methylation and Dot1l in the (aging) brain and neurons are limited. This, together with the increasing evidence of a dynamic neuroepigenome, made us wonder if H3K79 methylation and its activator Dot1l could play important roles in brain aging and associated disorders. In aged humans, we found strong and consistent global hypermethylation of H3K79 in neurons. Specific in dopaminergic neurons, we found a strong increase in H3K79 methylation in lipofucsin positive neurons, which are linked to pathology. In animals, where we conditionally removed Dot1l, we found a rapid loss of H3K79 methylation. As a consequence, we found some decrease in specific dopaminergic genes, and surprisingly, a clear up-regulation of almost all genes belonging to the family of the respiratory chain. These data, in relation to the observed increase in global H3K79 methylation, suggest that there is an inverse relationship between H3K79 methylation and the capacity of energy metabolism in neuronal systems.
Collapse
|
18
|
Adamson A, Buck SA, Freyberg Z, De Miranda BR. Sex Differences in Dopaminergic Vulnerability to Environmental Toxicants - Implications for Parkinson's Disease. Curr Environ Health Rep 2022; 9:563-573. [PMID: 36201109 PMCID: PMC10201647 DOI: 10.1007/s40572-022-00380-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW Sex dimorphism in Parkinson's disease (PD) is an ostensible feature of the neurological disorder, particularly as men are 1.5-2 times more likely to develop PD than women. Clinical features of the disease, such as presentation at onset, most prevalent symptoms, and response to treatment, are also affected by sex. Despite these well-known sex differences in PD risk and phenotype, the mechanisms that impart sex dimorphisms in PD remain poorly understood. RECENT FINDINGS As PD incidence is influenced by environmental factors, an intriguing pattern has recently emerged in research studies suggesting a male-specific vulnerability to dopaminergic neurodegeneration caused by neurotoxicant exposure, with relative protection in females. These new experimental data have uncovered potential mechanisms that provide clues to the source of sex differences in dopaminergic neurodegeneration and other PD pathology such as alpha-synuclein toxicity. In this review, we discuss the emerging evidence of increased male sensitivity to neurodegeneration from environmental exposures. We examine mechanisms underlying dopaminergic neurodegeneration and PD-related pathologies with evidence supporting the roles of estrogen, SRY expression, the vesicular glutamate transporter VGLUT2, and the microbiome as prospective catalysts for male vulnerability. We also highlight the importance of including sex as a biological variable, particularly when evaluating dopaminergic neurotoxicity in the context of PD.
Collapse
Affiliation(s)
- Ashley Adamson
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1719 6th Ave South, CIRC 560, Birmingham, AL, 35294, USA
| | - Silas A Buck
- Center for Neuroscience, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zachary Freyberg
- Center for Neuroscience, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Briana R De Miranda
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1719 6th Ave South, CIRC 560, Birmingham, AL, 35294, USA.
| |
Collapse
|
19
|
Gaertner Z, Azcorra M, Dombeck DA, Awatramani R. Molecular heterogeneity in the substantia nigra: A roadmap for understanding PD motor pathophysiology. Neurobiol Dis 2022; 175:105925. [DOI: 10.1016/j.nbd.2022.105925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022] Open
|
20
|
Roberts RC, McCollum LA, Schoonover KE, Mabry SJ, Roche JK, Lahti AC. Ultrastructural evidence for glutamatergic dysregulation in schizophrenia. Schizophr Res 2022; 249:4-15. [PMID: 32014360 PMCID: PMC7392793 DOI: 10.1016/j.schres.2020.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/16/2020] [Accepted: 01/18/2020] [Indexed: 12/14/2022]
Abstract
The aim of this paper is to summarize ultrastructural evidence for glutamatergic dysregulation in several linked regions in postmortem schizophrenia brain. Following a brief summary of glutamate circuitry and how synapses are identified at the electron microscopic (EM) level, we will review EM pathology in the cortex and basal ganglia. We will include the effects of antipsychotic drugs and the relation of treatment response. We will discuss how these findings support or confirm other postmortem findings as well as imaging results. Briefly, synaptic and mitochondrial density in anterior cingulate cortex was decreased in schizophrenia, versus normal controls (NCs), in a selective layer specific pattern. In dorsal striatum, increases in excitatory synaptic density were detected in caudate matrix, a compartment associated with cognitive and motor function, and in the putamen patches, a region associated with limbic function and in the core of the nucleus accumbens. Patients who were treatment resistant or untreated had significantly elevated numbers of excitatory synapses in limbic striatal areas in comparison to NCs and responders. Protein levels of vGLUT2, found in subcortical glutamatergic neurons, were increased in the nucleus accumbens in schizophrenia. At the EM level, schizophrenia subjects had an increase in density of excitatory synapses in several areas of the basal ganglia. In the substantia nigra, the protein levels of vGLUT2 were elevated in untreated patients compared to NCs. The density of inhibitory synapses was decreased in schizophrenia versus NCs. In schizophrenia, glutamatergic synapses are differentially affected depending on the brain region, treatment status, and treatment response.
Collapse
Affiliation(s)
- Rosalinda C Roberts
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama, Birmingham, AL 35294, United States of America.
| | - Lesley A McCollum
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama, Birmingham, AL 35294, United States of America
| | - Kirsten E Schoonover
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama, Birmingham, AL 35294, United States of America
| | - Samuel J Mabry
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama, Birmingham, AL 35294, United States of America
| | - Joy K Roche
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama, Birmingham, AL 35294, United States of America
| | - Adrienne C Lahti
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama, Birmingham, AL 35294, United States of America
| |
Collapse
|
21
|
Buck SA, Quincy Erickson-Oberg M, Logan RW, Freyberg Z. Relevance of interactions between dopamine and glutamate neurotransmission in schizophrenia. Mol Psychiatry 2022; 27:3583-3591. [PMID: 35681081 PMCID: PMC9712151 DOI: 10.1038/s41380-022-01649-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/16/2022] [Accepted: 05/25/2022] [Indexed: 02/08/2023]
Abstract
Dopamine (DA) and glutamate neurotransmission are strongly implicated in schizophrenia pathophysiology. While most studies focus on contributions of neurons that release only DA or glutamate, neither DA nor glutamate models alone recapitulate the full spectrum of schizophrenia pathophysiology. Similarly, therapeutic strategies limited to either system cannot effectively treat all three major symptom domains of schizophrenia: positive, negative, and cognitive symptoms. Increasing evidence suggests extensive interactions between the DA and glutamate systems and more effective treatments may therefore require the targeting of both DA and glutamate signaling. This offers the possibility that disrupting DA-glutamate circuitry between these two systems, particularly in the striatum and forebrain, culminate in schizophrenia pathophysiology. Yet, the mechanisms behind these interactions and their contributions to schizophrenia remain unclear. In addition to circuit- or system-level interactions between neurons that solely release either DA or glutamate, here we posit that functional alterations involving a subpopulation of neurons that co-release both DA and glutamate provide a novel point of integration between DA and glutamate systems, offering a key missing link in our understanding of schizophrenia pathophysiology. Better understanding of mechanisms underlying DA/glutamate co-release from these neurons may therefore shed new light on schizophrenia pathophysiology and lead to more effective therapeutics.
Collapse
Affiliation(s)
- Silas A Buck
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - M Quincy Erickson-Oberg
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Ryan W Logan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, 02118, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
22
|
Kelly EA, Contreras J, Duan A, Vassell R, Fudge JL. Unbiased Stereological Estimates of Dopaminergic and GABAergic Neurons in the A10, A9, and A8 Subregions in the Young Male Macaque. Neuroscience 2022; 496:152-164. [PMID: 35738547 PMCID: PMC9329254 DOI: 10.1016/j.neuroscience.2022.06.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/25/2022] [Accepted: 06/10/2022] [Indexed: 11/28/2022]
Abstract
The ventral midbrain is the primary source of dopamine- (DA) expressing neurons in most species. GABA-ergic and glutamatergic cell populations are intermixed among DA-expressing cells and purported to regulate both local and long-range dopamine neuron activity. Most work has been conducted in rodent models, however due to evolutionary expansion of the ventral midbrain in primates, the increased size and complexity of DA subpopulations warrants further investigation. Here, we quantified the number of DA neurons, and their GABA-ergic complement in classic DA cell groups A10 (midline ventral tegmental area nuclei [VTA] and parabrachial pigmented nucleus [PBP]), A9 (substantia nigra, pars compacta [SNc]) and A8 (retrorubral field [RRF]) in the macaque. Because the PBP is a disproportionately expanded feature of the A10 group, and has unique connectional features in monkeys, we analyzed A10 data by dividing it into 'classic' midline nuclei and the PBP. Unbiased stereology revealed total putative DA neuron counts to be 210,238 ± 17,127 (A10 = 110,319 ± 9649, A9 = 87,399 ± 7751 and A8 = 12,520 ± 827). Putative GABAergic neurons were fewer overall, and evenly dispersed across the DA subpopulations (GAD67 = 71,215 ± 5663; A10 = 16,836 ± 2743; A9 = 24,855 ± 3144 and A8 = 12,633 ± 3557). Calculating the GAD67/TH ratio for each subregion revealed differential balances of these two cell types across the DA subregions. The A8 subregion had the highest complement of GAD67-positive neurons compared to TH-positive neurons (1:1), suggesting a potentially high capacity for GABAergic inhibition of DA output in this region.
Collapse
Affiliation(s)
- Emily A Kelly
- Department of Neuroscience, University of Rochester Medical Center, United States
| | - Jancy Contreras
- Department of Neuroscience, The City University of New York, United States
| | - Annie Duan
- Department of Neuroscience, University of Rochester Medical Center, United States
| | - Rochelle Vassell
- Department of Neuroscience, University of Rochester Medical Center, United States
| | - Julie L Fudge
- Department of Neuroscience, University of Rochester Medical Center, United States; Department of Psychiatry, University of Rochester Medical Center, United States.
| |
Collapse
|
23
|
Cai J, Tong Q. Anatomy and Function of Ventral Tegmental Area Glutamate Neurons. Front Neural Circuits 2022; 16:867053. [PMID: 35669454 PMCID: PMC9164627 DOI: 10.3389/fncir.2022.867053] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/30/2022] [Indexed: 11/29/2022] Open
Abstract
The ventral tegmental area (VTA) is well known for regulating reward consumption, learning, memory, and addiction behaviors through mediating dopamine (DA) release in downstream regions. Other than DA neurons, the VTA is known to be heterogeneous and contains other types of neurons, including glutamate neurons. In contrast to the well-studied and established functions of DA neurons, the role of VTA glutamate neurons is understudied, presumably due to their relatively small quantity and a lack of effective means to study them. Yet, emerging studies have begun to reveal the importance of glutamate release from VTA neurons in regulating diverse behavioral repertoire through a complex intra-VTA and long-range neuronal network. In this review, we summarize the features of VTA glutamate neurons from three perspectives, namely, cellular properties, neural connections, and behavioral functions. Delineation of VTA glutamatergic pathways and their interactions with VTA DA neurons in regulating behaviors may reveal previously unappreciated functions of the VTA in other physiological processes.
Collapse
Affiliation(s)
- Jing Cai
- Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, UTHealth McGovern Medical School, Houston, TX, United States
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, UTHealth McGovern Medical School, Houston, TX, United States
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| |
Collapse
|
24
|
Steinkellner T, Conrad WS, Kovacs I, Rissman RA, Lee EB, Trojanowski JQ, Freyberg Z, Roy S, Luk KC, Lee VM, Hnasko TS. Dopamine neurons exhibit emergent glutamatergic identity in Parkinson's disease. Brain 2022; 145:879-886. [PMID: 35258081 PMCID: PMC9050538 DOI: 10.1093/brain/awab373] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 01/07/2023] Open
Abstract
Loss of midbrain dopamine neurons causes the cardinal symptoms of Parkinson's disease. However, not all dopamine neurons are equally vulnerable and a better understanding of the cell-type specific properties relating to selective dopamine neuron degeneration is needed. Most midbrain dopamine neurons express the vesicular glutamate transporter VGLUT2 during development and a subset continue to express low levels of VGLUT2 in adulthood, enabling the co-release of glutamate. Moreover, VGLUT2 expression in dopamine neurons can be neuroprotective since its genetic disruption was shown to sensitize dopamine neurons to neurotoxins. Here, we show that in response to toxic insult, and in two distinct models of alpha-synuclein stress, VGLUT2 dopamine neurons were resilient to degeneration. Dopamine neurons expressing VGLUT2 were enriched whether or not insult induced dopamine neuron loss, suggesting that while VGLUT2 dopamine neurons are more resilient, VGLUT2 expression can also be transcriptionally upregulated by injury. Finally, we observed that VGLUT2 expression was enhanced in surviving dopamine neurons from post-mortem Parkinson's disease individuals. These data indicate that emergence of a glutamatergic identity in dopamine neurons may be part of a neuroprotective response in Parkinson's disease.
Collapse
Affiliation(s)
- Thomas Steinkellner
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - William S Conrad
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Imre Kovacs
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zachary Freyberg
- Departments of Psychiatry and Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Subhojit Roy
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Kelvin C Luk
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Virginia M Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas S Hnasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
25
|
Glycation modulates glutamatergic signaling and exacerbates Parkinson's disease-like phenotypes. NPJ Parkinsons Dis 2022; 8:51. [PMID: 35468899 PMCID: PMC9038780 DOI: 10.1038/s41531-022-00314-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/31/2022] [Indexed: 01/17/2023] Open
Abstract
Alpha-synuclein (aSyn) is a central player in the pathogenesis of synucleinopathies due to its accumulation in typical protein aggregates in the brain. However, it is still unclear how it contributes to neurodegeneration. Type-2 diabetes mellitus is a risk factor for Parkinson's disease (PD). Interestingly, a common molecular alteration among these disorders is the age-associated increase in protein glycation. We hypothesized that glycation-induced neuronal dysfunction is a contributing factor in synucleinopathies. Here, we dissected the impact of methylglyoxal (MGO, a glycating agent) in mice overexpressing aSyn in the brain. We found that MGO-glycation potentiates motor, cognitive, olfactory, and colonic dysfunction in aSyn transgenic (Thy1-aSyn) mice that received a single dose of MGO via intracerebroventricular injection. aSyn accumulates in the midbrain, striatum, and prefrontal cortex, and protein glycation is increased in the cerebellum and midbrain. SWATH mass spectrometry analysis, used to quantify changes in the brain proteome, revealed that MGO mainly increase glutamatergic-associated proteins in the midbrain (NMDA, AMPA, glutaminase, VGLUT and EAAT1), but not in the prefrontal cortex, where it mainly affects the electron transport chain. The glycated proteins in the midbrain of MGO-injected Thy1-aSyn mice strongly correlate with PD and dopaminergic pathways. Overall, we demonstrated that MGO-induced glycation accelerates PD-like sensorimotor and cognitive alterations and suggest that the increase of glutamatergic signaling may underly these events. Our study sheds new light into the enhanced vulnerability of the midbrain in PD-related synaptic dysfunction and suggests that glycation suppressors and anti-glutamatergic drugs may hold promise as disease-modifying therapies for synucleinopathies.
Collapse
|
26
|
Phillips RA, Tuscher JJ, Black SL, Andraka E, Fitzgerald ND, Ianov L, Day JJ. An atlas of transcriptionally defined cell populations in the rat ventral tegmental area. Cell Rep 2022; 39:110616. [PMID: 35385745 PMCID: PMC10888206 DOI: 10.1016/j.celrep.2022.110616] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/28/2021] [Accepted: 03/11/2022] [Indexed: 01/06/2023] Open
Abstract
The ventral tegmental area (VTA) is a complex brain region that is essential for reward function and frequently implicated in neuropsychiatric disease. While decades of research on VTA function have focused on dopamine neurons, recent evidence has identified critical roles for GABAergic and glutamatergic neurons in reward processes. Additionally, although subsets of VTA neurons express genes involved in the synthesis and transport of multiple neurotransmitters, characterization of these combinatorial populations has largely relied on low-throughput methods. To comprehensively define the molecular architecture of the VTA, we performed single-nucleus RNA sequencing on 21,600 cells from the rat VTA. Analysis of neuronal subclusters identifies selective markers for dopamine and combinatorial neurons, reveals expression profiles for receptors targeted by drugs of abuse, and demonstrates population-specific enrichment of gene sets linked to brain disorders. These results highlight the heterogeneity of the VTA and provide a resource for further exploration of VTA gene expression.
Collapse
Affiliation(s)
- Robert A Phillips
- Department of Neurobiology & Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer J Tuscher
- Department of Neurobiology & Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Samantha L Black
- Department of Neurobiology & Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Emma Andraka
- Department of Neurobiology & Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - N Dalton Fitzgerald
- Department of Neurobiology & Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lara Ianov
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeremy J Day
- Department of Neurobiology & Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
27
|
Dvořáček J, Bednářová A, Krishnan N, Kodrík D. Dopaminergic muhsroom body neurons in Drosophila: flexibility of neuron identity in a model organism? Neurosci Biobehav Rev 2022; 135:104570. [DOI: 10.1016/j.neubiorev.2022.104570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/03/2022] [Accepted: 02/03/2022] [Indexed: 11/28/2022]
|
28
|
Buck SA, Erickson-Oberg MQ, Bhatte SH, McKellar CD, Ramanathan VP, Rubin SA, Freyberg Z. Roles of VGLUT2 and Dopamine/Glutamate Co-Transmission in Selective Vulnerability to Dopamine Neurodegeneration. ACS Chem Neurosci 2022; 13:187-193. [PMID: 34994539 PMCID: PMC9242677 DOI: 10.1021/acschemneuro.1c00741] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Growing evidence has established that a subset of dopamine (DA) neurons co-release glutamate and express vesicular glutamate transporter 2 (VGLUT2). VGLUT2 expression in DA neurons plays a key role in selective vulnerability to DA neurodegeneration in Parkinson's disease (PD). In this review, we summarize recent findings on impacts of VGLUT2 expression and glutamate co-release from DA neurons on selective DA neuron vulnerability. We present evidence that DA neuron VGLUT2 expression may be neuroprotective, boosting DA neuron resilience in the context of ongoing neurodegenerative processes in PD. We highlight genetic and pesticide models of PD that have provided mechanistic insights into selective DA neuron vulnerability. Finally, we discuss potential neuroprotective mechanisms, focusing on roles of VGLUT2 and glutamate in promoting mitochondrial health and diminishing oxidative stress and excitotoxicity. Elucidating these mechanisms may ultimately lead to more effective treatments to boost DA neuron resilience that can slow or even prevent DA neurodegeneration.
Collapse
Affiliation(s)
- Silas A. Buck
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - M. Quincy Erickson-Oberg
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sai H. Bhatte
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chase D. McKellar
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Sophie A. Rubin
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
29
|
Li J, Curley WH, Guerin B, Dougherty DD, Dalca AV, Fischl B, Horn A, Edlow BL. Mapping the subcortical connectivity of the human default mode network. Neuroimage 2021; 245:118758. [PMID: 34838949 PMCID: PMC8945548 DOI: 10.1016/j.neuroimage.2021.118758] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/29/2021] [Accepted: 11/23/2021] [Indexed: 01/17/2023] Open
Abstract
The default mode network (DMN) mediates self-awareness and introspection, core components of human consciousness. Therapies to restore consciousness in patients with severe brain injuries have historically targeted subcortical sites in the brainstem, thalamus, hypothalamus, basal forebrain, and basal ganglia, with the goal of reactivating cortical DMN nodes. However, the subcortical connectivity of the DMN has not been fully mapped, and optimal subcortical targets for therapeutic neuromodulation of consciousness have not been identified. In this work, we created a comprehensive map of DMN subcortical connectivity by combining high-resolution functional and structural datasets with advanced signal processing methods. We analyzed 7 Tesla resting-state functional MRI (rs-fMRI) data from 168 healthy volunteers acquired in the Human Connectome Project. The rs-fMRI blood-oxygen-level-dependent (BOLD) data were temporally synchronized across subjects using the BrainSync algorithm. Cortical and subcortical DMN nodes were jointly analyzed and identified at the group level by applying a novel Nadam-Accelerated SCAlable and Robust (NASCAR) tensor decomposition method to the synchronized dataset. The subcortical connectivity map was then overlaid on a 7 Tesla 100 µm ex vivo MRI dataset for neuroanatomic analysis using automated segmentation of nuclei within the brainstem, thalamus, hypothalamus, basal forebrain, and basal ganglia. We further compared the NASCAR subcortical connectivity map with its counterpart generated from canonical seed-based correlation analyses. The NASCAR method revealed that BOLD signal in the central lateral nucleus of the thalamus and ventral tegmental area of the midbrain is strongly correlated with that of the DMN. In an exploratory analysis, additional subcortical sites in the median and dorsal raphe, lateral hypothalamus, and caudate nuclei were correlated with the cortical DMN. We also found that the putamen and globus pallidus are negatively correlated (i.e., anti-correlated) with the DMN, providing rs-fMRI evidence for the mesocircuit hypothesis of human consciousness, whereby a striatopallidal feedback system modulates anterior forebrain function via disinhibition of the central thalamus. Seed-based analyses yielded similar subcortical DMN connectivity, but the NASCAR result showed stronger contrast and better spatial alignment with dopamine immunostaining data. The DMN subcortical connectivity map identified here advances understanding of the subcortical regions that contribute to human consciousness and can be used to inform the selection of therapeutic targets in clinical trials for patients with disorders of consciousness.
Collapse
Affiliation(s)
- Jian Li
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - William H Curley
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Bastien Guerin
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Darin D Dougherty
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA; Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Adrian V Dalca
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bruce Fischl
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andreas Horn
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Center for Brain Circuit Therapeutics, Department of Neurology, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Movement Disorders & Neuromodulation Section, Department of Neurology, Charité - Universitätsmedizin, Berlin, Germany
| | - Brian L Edlow
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
30
|
Pereira Luppi M, Azcorra M, Caronia-Brown G, Poulin JF, Gaertner Z, Gatica S, Moreno-Ramos OA, Nouri N, Dubois M, Ma YC, Ramakrishnan C, Fenno L, Kim YS, Deisseroth K, Cicchetti F, Dombeck DA, Awatramani R. Sox6 expression distinguishes dorsally and ventrally biased dopamine neurons in the substantia nigra with distinctive properties and embryonic origins. Cell Rep 2021; 37:109975. [PMID: 34758317 PMCID: PMC8607753 DOI: 10.1016/j.celrep.2021.109975] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 09/15/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Dopamine (DA) neurons in the ventral tier of the substantia nigra pars compacta (SNc) degenerate prominently in Parkinson's disease, while those in the dorsal tier are relatively spared. Defining the molecular, functional, and developmental characteristics of each SNc tier is crucial to understand their distinct susceptibility. We demonstrate that Sox6 expression distinguishes ventrally and dorsally biased DA neuron populations in the SNc. The Sox6+ population in the ventral SNc includes an Aldh1a1+ subset and is enriched in gene pathways that underpin vulnerability. Sox6+ neurons project to the dorsal striatum and show activity correlated with acceleration. Sox6- neurons project to the medial, ventral, and caudal striatum and respond to rewards. Moreover, we show that this adult division is encoded early in development. Overall, our work demonstrates a dual origin of the SNc that results in DA neuron cohorts with distinct molecular profiles, projections, and functions.
Collapse
Affiliation(s)
- Milagros Pereira Luppi
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Maite Azcorra
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Giuliana Caronia-Brown
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jean-Francois Poulin
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Zachary Gaertner
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Serafin Gatica
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Navid Nouri
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Marilyn Dubois
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Yongchao C Ma
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Charu Ramakrishnan
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Lief Fenno
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Yoon Seok Kim
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Francesca Cicchetti
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Daniel A Dombeck
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
| | - Rajeshwar Awatramani
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
31
|
Islam KUS, Meli N, Blaess S. The Development of the Mesoprefrontal Dopaminergic System in Health and Disease. Front Neural Circuits 2021; 15:746582. [PMID: 34712123 PMCID: PMC8546303 DOI: 10.3389/fncir.2021.746582] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 09/10/2021] [Indexed: 12/18/2022] Open
Abstract
Midbrain dopaminergic neurons located in the substantia nigra and the ventral tegmental area are the main source of dopamine in the brain. They send out projections to a variety of forebrain structures, including dorsal striatum, nucleus accumbens, and prefrontal cortex (PFC), establishing the nigrostriatal, mesolimbic, and mesoprefrontal pathways, respectively. The dopaminergic input to the PFC is essential for the performance of higher cognitive functions such as working memory, attention, planning, and decision making. The gradual maturation of these cognitive skills during postnatal development correlates with the maturation of PFC local circuits, which undergo a lengthy functional remodeling process during the neonatal and adolescence stage. During this period, the mesoprefrontal dopaminergic innervation also matures: the fibers are rather sparse at prenatal stages and slowly increase in density during postnatal development to finally reach a stable pattern in early adulthood. Despite the prominent role of dopamine in the regulation of PFC function, relatively little is known about how the dopaminergic innervation is established in the PFC, whether and how it influences the maturation of local circuits and how exactly it facilitates cognitive functions in the PFC. In this review, we provide an overview of the development of the mesoprefrontal dopaminergic system in rodents and primates and discuss the role of altered dopaminergic signaling in neuropsychiatric and neurodevelopmental disorders.
Collapse
Affiliation(s)
- K Ushna S Islam
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Norisa Meli
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany.,Institute of Neuropathology, Section for Translational Epilepsy Research, Medical Faculty, University of Bonn, Bonn, Germany
| | - Sandra Blaess
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
32
|
Pérez-Fernández J, Barandela M, Jiménez-López C. The Dopaminergic Control of Movement-Evolutionary Considerations. Int J Mol Sci 2021; 22:11284. [PMID: 34681941 PMCID: PMC8541398 DOI: 10.3390/ijms222011284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 12/11/2022] Open
Abstract
Dopamine is likely the most studied modulatory neurotransmitter, in great part due to characteristic motor deficits in Parkinson's disease that arise after the degeneration of the dopaminergic neurons in the substantia nigra pars compacta (SNc). The SNc, together with the ventral tegmental area (VTA), play a key role modulating motor responses through the basal ganglia. In contrast to the large amount of existing literature addressing the mammalian dopaminergic system, comparatively little is known in other vertebrate groups. However, in the last several years, numerous studies have been carried out in basal vertebrates, allowing a better understanding of the evolution of the dopaminergic system, especially the SNc/VTA. We provide an overview of existing research in basal vertebrates, mainly focusing on lampreys, belonging to the oldest group of extant vertebrates. The lamprey dopaminergic system and its role in modulating motor responses have been characterized in significant detail, both anatomically and functionally, providing the basis for understanding the evolution of the SNc/VTA in vertebrates. When considered alongside results from other early vertebrates, data in lampreys show that the key role of the SNc/VTA dopaminergic neurons modulating motor responses through the basal ganglia was already well developed early in vertebrate evolution.
Collapse
Affiliation(s)
- Juan Pérez-Fernández
- Center for Biomedical Research (CINBIO), Neurocircuits Group, Department of Functional Biology and Health Sciences, Campus Universitario Lagoas, Marcosende, Universidade de Vigo, 36310 Vigo, Spain; (M.B.); (C.J.-L.)
| | | | | |
Collapse
|
33
|
Kushwaha A, Singh G, Sharma M. Designing of cerium phosphate nanorods decorated reduced graphene oxide nanostructures as modified electrode: An effective mode of dopamine sensing. Microchem J 2021. [DOI: 10.1016/j.microc.2021.106224] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
34
|
Neurochemical Signaling of Reward and Aversion to Ventral Tegmental Area Glutamate Neurons. J Neurosci 2021; 41:5471-5486. [PMID: 34001626 DOI: 10.1523/jneurosci.1419-20.2021] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 12/21/2022] Open
Abstract
Ventral tegmental area (VTA) glutamate neurons signal and participate in reward and aversion-based behaviors. However, the neurochemical mechanisms that underlie how these neurons contribute to motivated behaviors is unknown. We used a combination of optical sensors to identify how distinct neurochemical inputs to VTA glutamate neurons participate in motivated behavior within female and male transgenic mice. Activity of glutamate inputs to VTA glutamate neurons increased for both reward-predicting and aversion-predicting cues and aversive outcomes, but subpopulations of glutamate inputs were increased or decreased by reward. For both reward and aversion-based cues and outcomes, activity of GABA inputs to VTA glutamate neurons mostly decreased. GCaMP recordings showed overall population increases in VTA glutamate neuron intracellular calcium during reward and aversion-based cues and outcomes. Electrophysiological recordings of VTA VGluT2 neurons showed that glutamate receptor activation increases firing while loss of excitation via glutamate receptor blockade decreases firing. GABA-A receptor activation decreased VTA glutamate neuron firing but GABA-A receptor blockade did not significantly change VTA glutamate neuron firing. Electrophysiological recordings in coordination with our sensor data suggest that glutamate inputs strongly regulate VTA glutamate neuron participation in diverse motivated behaviors.SIGNIFICANCE STATEMENT Glutamate and GABA are the primary excitatory and inhibitory neurotransmitters of the nervous system. However, identifying how these neurotransmitters regulate motivated behavior has remained challenging because of a lack of tools (1) capable of measuring neurotransmission at the temporal scale of motivated behaviors and (2) capable of capturing chemical signaling onto genetically-distinct neuronal populations. We have overcome these obstacles by implementing genetically-encoded fluorescent indicators to monitor both glutamate and GABA input dynamics exclusively to ventral tegmental area (VTA) glutamate neurons during reward and aversion-based behaviors. We identify that glutamate and GABA inputs to VTA glutamate neurons differentially and dynamically signal reward and aversion-based cues and outcomes. This research provides foundational evidence that links distinct neurotransmitters to motivated behaviors regulated by VTA glutamate neurons.
Collapse
|
35
|
Root DH, Barker DJ, Estrin DJ, Miranda-Barrientos JA, Liu B, Zhang S, Wang HL, Vautier F, Ramakrishnan C, Kim YS, Fenno L, Deisseroth K, Morales M. Distinct Signaling by Ventral Tegmental Area Glutamate, GABA, and Combinatorial Glutamate-GABA Neurons in Motivated Behavior. Cell Rep 2021; 32:108094. [PMID: 32877676 PMCID: PMC7556367 DOI: 10.1016/j.celrep.2020.108094] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/02/2020] [Accepted: 08/10/2020] [Indexed: 12/22/2022] Open
Abstract
Ventral tegmental area (VTA) neurons play roles in reward and aversion. We recently discovered that the VTA has neurons that co-transmit glutamate and GABA (glutamate-GABA co-transmitting neurons), transmit glutamate without GABA (glutamate-transmitting neurons), or transmit GABA without glutamate (GABA-transmitting neurons). However, the functions of these VTA cell types in motivated behavior are unclear. To identify the functions of these VTA cell types, we combine recombinase mouse lines with INTRSECT2.0 vectors to selectively target these neurons. We find that VTA cell types have unique signaling patterns for reward, aversion, and learned cues. Whereas VTA glutamate-transmitting neurons signal cues predicting reward, VTA GABA-transmitting neurons signal cues predicting the absence of reward, and glutamate-GABA co-transmitting neurons signal rewarding and aversive outcomes without signaling learned cues related to those outcomes. Thus, we demonstrate that genetically defined subclasses of VTA glutamate and GABA neurons signal different aspects of motivated behavior. Root et al. examine ventral tegmental area (VTA) neurons that release glutamate without GABA, GABA without glutamate, or both glutamate and GABA. Cell types have differential projection densities and unique neuronal activity profiles related to cues predicting rewarding, nonreward, or aversive outcomes and in the receipt of predicted rewards.
Collapse
Affiliation(s)
- David H Root
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Boulevard Suite 200, Baltimore, MD 21224, USA
| | - David J Barker
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Boulevard Suite 200, Baltimore, MD 21224, USA
| | - David J Estrin
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Boulevard Suite 200, Baltimore, MD 21224, USA
| | - Jorge A Miranda-Barrientos
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Boulevard Suite 200, Baltimore, MD 21224, USA
| | - Bing Liu
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Boulevard Suite 200, Baltimore, MD 21224, USA
| | - Shiliang Zhang
- Confocal and Electron Microscopy Core, National Institute on Drug Abuse, 251 Bayview Boulevard Suite 200, Baltimore, MD 21224, USA
| | - Hui-Ling Wang
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Boulevard Suite 200, Baltimore, MD 21224, USA
| | - Francois Vautier
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Boulevard Suite 200, Baltimore, MD 21224, USA
| | - Charu Ramakrishnan
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Yoon Seok Kim
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Lief Fenno
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Marisela Morales
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Boulevard Suite 200, Baltimore, MD 21224, USA.
| |
Collapse
|
36
|
Eskenazi D, Malave L, Mingote S, Yetnikoff L, Ztaou S, Velicu V, Rayport S, Chuhma N. Dopamine Neurons That Cotransmit Glutamate, From Synapses to Circuits to Behavior. Front Neural Circuits 2021; 15:665386. [PMID: 34093138 PMCID: PMC8170480 DOI: 10.3389/fncir.2021.665386] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/16/2021] [Indexed: 11/21/2022] Open
Abstract
Discovered just over 20 years ago, dopamine neurons have the ability to cotransmit both dopamine and glutamate. Yet, the functional roles of dopamine neuron glutamate cotransmission and their implications for therapeutic use are just emerging. This review article encompasses the current body of evidence investigating the functions of dopamine neurons of the ventral midbrain that cotransmit glutamate. Since its discovery in dopamine neuron cultures, further work in vivo confirmed dopamine neuron glutamate cotransmission across species. From there, growing interest has led to research related to neural functioning including roles in synaptic signaling, development, and behavior. Functional connectome mapping reveals robust connections in multiple forebrain regions to various cell types, most notably to cholinergic interneurons in both the medial shell of the nucleus accumbens and the lateral dorsal striatum. Glutamate markers in dopamine neurons reach peak levels during embryonic development and increase in response to various toxins, suggesting dopamine neuron glutamate cotransmission may serve neuroprotective roles. Findings from behavioral analyses reveal prominent roles for dopamine neuron glutamate cotransmission in responses to psychostimulants, in positive valence and cognitive systems and for subtle roles in negative valence systems. Insight into dopamine neuron glutamate cotransmission informs the pathophysiology of neuropsychiatric disorders such as addiction, schizophrenia and Parkinson Disease, with therapeutic implications.
Collapse
Affiliation(s)
- Daniel Eskenazi
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Lauren Malave
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Susana Mingote
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY, United States
| | - Leora Yetnikoff
- Department of Psychology, College of Staten Island, City University of New York, Staten Island, NY, United States
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, New York, NY, United States
| | - Samira Ztaou
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Vlad Velicu
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Stephen Rayport
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Nao Chuhma
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| |
Collapse
|
37
|
Buck SA, Steinkellner T, Aslanoglou D, Villeneuve M, Bhatte SH, Childers VC, Rubin SA, De Miranda BR, O'Leary EI, Neureiter EG, Fogle KJ, Palladino MJ, Logan RW, Glausier JR, Fish KN, Lewis DA, Greenamyre JT, McCabe BD, Cheetham CEJ, Hnasko TS, Freyberg Z. Vesicular glutamate transporter modulates sex differences in dopamine neuron vulnerability to age-related neurodegeneration. Aging Cell 2021; 20:e13365. [PMID: 33909313 PMCID: PMC8135008 DOI: 10.1111/acel.13365] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/22/2021] [Accepted: 03/31/2021] [Indexed: 12/30/2022] Open
Abstract
Age is the greatest risk factor for Parkinson's disease (PD) which causes progressive loss of dopamine (DA) neurons, with males at greater risk than females. Intriguingly, some DA neurons are more resilient to degeneration than others. Increasing evidence suggests that vesicular glutamate transporter (VGLUT) expression in DA neurons plays a role in this selective vulnerability. We investigated the role of DA neuron VGLUT in sex- and age-related differences in DA neuron vulnerability using the genetically tractable Drosophila model. We found sex differences in age-related DA neurodegeneration and its associated locomotor behavior, where males exhibit significantly greater decreases in both DA neuron number and locomotion during aging compared with females. We discovered that dynamic changes in DA neuron VGLUT expression mediate these age- and sex-related differences, as a potential compensatory mechanism for diminished DA neurotransmission during aging. Importantly, female Drosophila possess higher levels of VGLUT expression in DA neurons compared with males, and this finding is conserved across flies, rodents, and humans. Moreover, we showed that diminishing VGLUT expression in DA neurons eliminates females' greater resilience to DA neuron loss across aging. This offers a new mechanism for sex differences in selective DA neuron vulnerability to age-related DA neurodegeneration. Finally, in mice, we showed that the ability of DA neurons to achieve optimal control over VGLUT expression is essential for DA neuron survival. These findings lay the groundwork for the manipulation of DA neuron VGLUT expression as a novel therapeutic strategy to boost DA neuron resilience to age- and PD-related neurodegeneration.
Collapse
Affiliation(s)
- Silas A. Buck
- Center for NeuroscienceUniversity of PittsburghPittsburghPAUSA
- Department of PsychiatryUniversity of PittsburghPittsburghPAUSA
| | - Thomas Steinkellner
- Department of NeurosciencesUniversity of California, San DiegoLa JollaCAUSA
- Institute of PharmacologyCenter for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | | | | | - Sai H. Bhatte
- Department of PsychiatryUniversity of PittsburghPittsburghPAUSA
| | | | - Sophie A. Rubin
- Department of PsychiatryUniversity of PittsburghPittsburghPAUSA
| | - Briana R. De Miranda
- Department of NeurologyUniversity of PittsburghPittsburghPAUSA
- Present address:
Department of NeurologyCenter for Neurodegeneration and Experimental TherapeuticsUniversity of Alabama at BirminghamBirminghamALUSA
| | - Emma I. O'Leary
- Center for NeuroscienceUniversity of PittsburghPittsburghPAUSA
| | - Elizabeth G. Neureiter
- Center for NeuroscienceUniversity of PittsburghPittsburghPAUSA
- Department of PsychiatryUniversity of PittsburghPittsburghPAUSA
| | - Keri J. Fogle
- Department of Pharmacology & Chemical BiologyUniversity of PittsburghPittsburghPAUSA
- Pittsburgh Institute for Neurodegenerative DiseasesUniversity of PittsburghPittsburghPAUSA
| | - Michael J. Palladino
- Department of Pharmacology & Chemical BiologyUniversity of PittsburghPittsburghPAUSA
- Pittsburgh Institute for Neurodegenerative DiseasesUniversity of PittsburghPittsburghPAUSA
| | - Ryan W. Logan
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMAUSA
- Center for Systems Neurogenetics of AddictionThe Jackson LaboratoryBar HarborMEUSA
| | | | - Kenneth N. Fish
- Department of PsychiatryUniversity of PittsburghPittsburghPAUSA
| | - David A. Lewis
- Department of PsychiatryUniversity of PittsburghPittsburghPAUSA
| | - J. Timothy Greenamyre
- Department of NeurologyUniversity of PittsburghPittsburghPAUSA
- Pittsburgh Institute for Neurodegenerative DiseasesUniversity of PittsburghPittsburghPAUSA
- Geriatric Research, Education and Clinical CenterVA Pittsburgh Healthcare SystemPittsburghPAUSA
| | - Brian D. McCabe
- Brain Mind InstituteSwiss Federal Institute of Technology (EPFL)LausanneSwitzerland
| | | | - Thomas S. Hnasko
- Department of NeurosciencesUniversity of California, San DiegoLa JollaCAUSA
- Research ServiceVA San Diego Healthcare SystemSan DiegoCAUSA
| | - Zachary Freyberg
- Department of PsychiatryUniversity of PittsburghPittsburghPAUSA
- Department of Cell BiologyUniversity of PittsburghPittsburghPAUSA
| |
Collapse
|
38
|
Björklund A, Parmar M. Dopamine Cell Therapy: From Cell Replacement to Circuitry Repair. JOURNAL OF PARKINSONS DISEASE 2021; 11:S159-S165. [PMID: 33814467 PMCID: PMC8543294 DOI: 10.3233/jpd-212609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cell therapy for Parkinson's disease (PD) is aimed to replace the degenerated midbrain dopamine (mDA) neurons and restore DA neurotransmission in the denervated forebrain targets. A limitation of the intrastriatal grafting approach, which is currently used in clinical trials, is that the mDA neurons are implanted into the target area, in most cases the putamen, and not in the ventral midbrain where they normally reside. This ectopic location of the cells may limit their functionality due to the lack of appropriate afferent regulation from the host. Homotopic transplantation, into the substantia nigra, is now being pursued in rodent PD models as a way to achieve more complete circuitry repair. Intranigral grafts of mDA neurons, derived from human embryonic stem cells, have the capacity to re-establish the nigrostriatal and mesolimbic pathways in their entirety and restore dense functional innervations in striatal, limbic and cortical areas. Tracing of host afferent inputs using the rabies tracing technique shows that the afferent connectivity of grafts implanted in the nigra matches closely that of the intrinsic mDA system, suggesting a degree of circuitry reconstruction that exceeds what has been achieved before. This approach holds great promise, but to match the larger size of the human brain, and the 10 times greater distance between substantia nigra and its forebrain targets, it may be necessary to find ways to improve the growth capacity of the grafted mDA neurons, pointing to a combined approach where growth promoting factors are used to enhance the performance of mDA neuron grafts.
Collapse
Affiliation(s)
- Anders Björklund
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Malin Parmar
- Department of Experimental Medical Science, Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| |
Collapse
|
39
|
Buck SA, Torregrossa MM, Logan RW, Freyberg Z. Roles of dopamine and glutamate co-release in the nucleus accumbens in mediating the actions of drugs of abuse. FEBS J 2021; 288:1462-1474. [PMID: 32702182 PMCID: PMC7854787 DOI: 10.1111/febs.15496] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/30/2020] [Accepted: 07/21/2020] [Indexed: 12/20/2022]
Abstract
Projections of ventral tegmental area dopamine (DA) neurons to the medial shell of the nucleus accumbens have been increasingly implicated as integral to the behavioral and physiological changes involved in the development of substance use disorders (SUDs). Recently, many of these nucleus accumbens-projecting DA neurons were found to also release the neurotransmitter glutamate. This glutamate co-release from DA neurons is critical in mediating the effect of drugs of abuse on addiction-related behaviors. Potential mechanisms underlying the role(s) of dopamine/glutamate co-release in contributing to SUDs are unclear. Nevertheless, an important clue may relate to glutamate's ability to potentiate loading of DA into synaptic vesicles within terminals in the nucleus accumbens in response to drug-induced elevations in neuronal activity, enabling a more robust release of DA after stimulation. Here, we summarize how drugs of abuse, particularly cocaine, opioids, and alcohol, alter DA release in the nucleus accumbens medial shell, examine the potential role of DA/glutamate co-release in mediating these effects, and discuss future directions for further investigating these mechanisms.
Collapse
Affiliation(s)
- Silas A. Buck
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mary M. Torregrossa
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan W. Logan
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Zachary Freyberg
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
40
|
Fischer KD, Knackstedt LA, Rosenberg PA. Glutamate homeostasis and dopamine signaling: Implications for psychostimulant addiction behavior. Neurochem Int 2021; 144:104896. [PMID: 33159978 PMCID: PMC8489281 DOI: 10.1016/j.neuint.2020.104896] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 02/06/2023]
Abstract
Cocaine, amphetamine, and methamphetamine abuse disorders are serious worldwide health problems. To date, there are no FDA-approved medications for the treatment of these disorders. Elucidation of the biochemical underpinnings contributing to psychostimulant addiction is critical for the development of effective therapies. Excitatory signaling and glutamate homeostasis are well known pathophysiological substrates underlying addiction-related behaviors spanning multiple types of psychostimulants. To alleviate relapse behavior to psychostimulants, considerable interest has focused on GLT-1, the major glutamate transporter in the brain. While many brain regions are implicated in addiction behavior, this review focuses on two regions well known for their role in mediating the effects of cocaine and amphetamines, namely the nucleus accumbens (NAc) and the ventral tegmental area (VTA). In addition, because many investigators have utilized Cre-driver lines to selectively control gene expression in defined cell populations relevant for psychostimulant addiction, we discuss potential off-target effects of Cre-recombinase that should be considered in the design and interpretation of such experiments.
Collapse
Affiliation(s)
- Kathryn D Fischer
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Lori A Knackstedt
- Psychology Department, University of Florida, Gainesville, FL, 32611, USA
| | - Paul A Rosenberg
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA; Program in Neuroscience, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
41
|
Muckom RJ, Sampayo RG, Johnson HJ, Schaffer DV. Advanced Materials to Enhance Central Nervous System Tissue Modeling and Cell Therapy. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2002931. [PMID: 33510596 PMCID: PMC7840150 DOI: 10.1002/adfm.202002931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Indexed: 05/04/2023]
Abstract
The progressively deeper understanding of mechanisms underlying stem cell fate decisions has enabled parallel advances in basic biology-such as the generation of organoid models that can further one's basic understanding of human development and disease-and in clinical translation-including stem cell based therapies to treat human disease. Both of these applications rely on tight control of the stem cell microenvironment to properly modulate cell fate, and materials that can be engineered to interface with cells in a controlled and tunable manner have therefore emerged as valuable tools for guiding stem cell growth and differentiation. With a focus on the central nervous system (CNS), a broad range of material solutions that have been engineered to overcome various hurdles in constructing advanced organoid models and developing effective stem cell therapeutics is reviewed. Finally, regulatory aspects of combined material-cell approaches for CNS therapies are considered.
Collapse
Affiliation(s)
- Riya J Muckom
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94704, USA
| | - Rocío G Sampayo
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94704, USA
| | - Hunter J Johnson
- Department of Bioengineering, UC Berkeley, Berkeley, CA 94704, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94704, USA
| |
Collapse
|
42
|
Gordon-Fennell A, Gordon-Fennell L, Desaivre S, Marinelli M. The Lateral Preoptic Area and Its Projection to the VTA Regulate VTA Activity and Drive Complex Reward Behaviors. Front Syst Neurosci 2020; 14:581830. [PMID: 33224029 PMCID: PMC7669548 DOI: 10.3389/fnsys.2020.581830] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/27/2020] [Indexed: 11/22/2022] Open
Abstract
The ventral tegmental area (VTA) underlies motivation and reinforcement of natural rewards. The lateral preoptic area (LPO) is an anterior hypothalamic brain region that sends direct projections to the VTA and to other brain structures known to regulate VTA activity. Here, we investigated the functional connection between the LPO and subpopulations of VTA neurons and explored the reinforcing and valence qualities of the LPO in rats. We found that the LPO and the LPO→VTA pathway inhibit the activity of VTA GABA neurons and have mixed effects on VTA dopamine neurons. Furthermore, we found that the LPO supports operant responding but drives avoidance, and we explored the apparent discrepancy between these two results. Finally, using fiber photometry, we show that the LPO signals aversive events but not rewarding events. Together, our findings demonstrate that the LPO modulates the activity of the VTA and drives motivated behavior and represents an overlooked modulator of reinforcement.
Collapse
Affiliation(s)
- Adam Gordon-Fennell
- Department of Neuroscience, College of Natural Sciences, University of Texas at Austin, Austin, TX, United States
| | - Lydia Gordon-Fennell
- Department of Neuroscience, College of Natural Sciences, University of Texas at Austin, Austin, TX, United States
| | - Stève Desaivre
- Department of Neuroscience, College of Natural Sciences, University of Texas at Austin, Austin, TX, United States
| | - Michela Marinelli
- Department of Neuroscience, College of Natural Sciences, University of Texas at Austin, Austin, TX, United States.,Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States.,Department of Psychiatry, Dell Medical School, University of Texas at Austin, Austin, TX, United States.,Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
43
|
Hernández RB, Carrascal M, Abian J, Michalke B, Farina M, Gonzalez YR, Iyirhiaro GO, Moteshareie H, Burnside D, Golshani A, Suñol C. Manganese-induced neurotoxicity in cerebellar granule neurons due to perturbation of cell network pathways with potential implications for neurodegenerative disorders. Metallomics 2020; 12:1656-1678. [PMID: 33206086 DOI: 10.1039/d0mt00085j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Manganese (Mn) is essential for living organisms, playing an important role in nervous system function. Nevertheless, chronic and/or acute exposure to this metal, especially during early life stages, can lead to neurotoxicity and dementia by unclear mechanisms. Thus, based on previous works of our group with yeast and zebrafish, we hypothesized that the mechanisms mediating manganese-induced neurotoxicity can be associated with the alteration of protein metabolism. These mechanisms may also depend on the chemical speciation of manganese. Therefore, the current study aimed at investigating the mechanisms mediating the toxic effects of manganese in primary cultures of cerebellar granule neurons (CGNs). By exposing cultured CGNs to different chemical species of manganese ([[2-[(dithiocarboxy)amino]ethyl]carbamodithioato]](2-)-kS,kS']manganese, named maneb (MB), and [[1,2-ethanediylbis[carbamodithioato]](2-)]manganese mixture with [[1,2-ethanediylbis[carbamodithioato]](2-)]zinc, named mancozeb (MZ), and manganese chloride (MnCl2)), and using the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, we observed that both MB and MZ induced similar cytotoxicity (LC50∼ 7-9 μM), which was higher than that of MnCl2 (LC50∼ 27 μM). Subsequently, we applied systems biology approaches, including metallomics, proteomics, gene expression and bioinformatics, and revealed that independent of chemical speciation, for non-cytotoxic concentrations (0.3-3 μM), Mn-induced neurotoxicity in CGNs is associated with metal dyshomeostasis and impaired protein metabolism. In this way, we verified that MB induced more post-translational alterations than MnCl2, which can be a plausible explanation for cytotoxic differences between both chemical species. The metabolism of proteins is one of the most energy consuming cellular processes and its impairment appears to be a key event of some cellular stress processes reported separately in other studies such as cell cycle arrest, energy impairment, cell signaling, excitotoxicity, immune response, potential protein accumulation and apoptosis. Interestingly, we verified that Mn-induced neurotoxicity shares pathways associated with the development of Alzheimer's disease, Amyotrophic Lateral Sclerosis, Huntington's disease, and Parkinson's disease. This has been observed in baker's yeast and zebrafish suggesting that the mode of action of Mn may be evolutionarily conserved.
Collapse
Affiliation(s)
- Raúl Bonne Hernández
- Laboratory of Bioinorganic and Environmental Toxicology - LABITA, Department of Exact and Earth Sciences, Federal University of São Paulo, Rua Prof. Artur Riedel, 275, CEP 09972-270, Diadema, SP, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mabry SJ, McCollum LA, Farmer CB, Bloom ES, Roberts RC. Evidence for altered excitatory and inhibitory tone in the post-mortem substantia nigra in schizophrenia. World J Biol Psychiatry 2020; 21:339-356. [PMID: 31062628 PMCID: PMC6891153 DOI: 10.1080/15622975.2019.1615638] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 03/21/2019] [Accepted: 05/02/2019] [Indexed: 12/14/2022]
Abstract
Objectives: The substantia nigra (SN) receives glutamatergic and GABAergic inputs that regulate dopaminergic neuronal activity. Imaging studies have shown hyperactivity of the SN in schizophrenia (SZ) patients. We examined neurochemically defined inputs to the SN, synaptic density, and neuromelanin content that might contribute to or reflect this hyperexcitability.Methods: Glutamatergic axon terminals were identified by the immunohistochemical localisation of vGLUT1 and vGLUT2; GABA inputs were identified by the immunohistochemical localisation of GAD67. Neuromelanin granules are visible in unstained sections and thus were assessed in unstained sections. Optical densitometry was measured to assess the density of vGLUT1, vGLUT2 or GAD67 immunolabelled axon terminals and neuromelanin granules. Electron microscopy was used to quantify synaptic and mitochondrial density.Results: Compared to controls, SZ subjects had nonsignificant trends toward a decrease in vGLUT1, and an increase in both vGLUT2 and GAD67. vGLUT1 was negatively correlated with GAD67 in normal controls (NCs) and positively correlated in SZ subjects. A correlation of coefficient analysis showed a significant difference between the negative correlation in NCs and the positive correlation in SZ subjects. Frequency histograms showed the distribution of neuromelanin density was different in SZ subjects compared to NCs. Synaptic density data showed a decrease in inhibitory synapses in SZ subjects. Mitochondrial density was normal in SZ subjects.Conclusions: Synaptic density alterations and the lack of a positive correlation between GAD67 and vGLUT1 could contribute to hyperactivity in the SN.
Collapse
Affiliation(s)
- Samuel J. Mabry
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, 1720 7 Ave. South, Birmingham AL, 35294
| | - Lesley A. McCollum
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, 1720 7 Ave. South, Birmingham AL, 35294
| | - Charlene B. Farmer
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, 1720 7 Ave. South, Birmingham AL, 35294
| | - Emma S. Bloom
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, 1720 7 Ave. South, Birmingham AL, 35294
| | - Rosalinda C. Roberts
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, 1720 7 Ave. South, Birmingham AL, 35294
| |
Collapse
|
45
|
Alijanpour S, Zarrindast MR. Potentiation of morphine-induced antinociception by harmaline: involvement of μ-opioid and ventral tegmental area NMDA receptors. Psychopharmacology (Berl) 2020; 237:557-570. [PMID: 31740992 DOI: 10.1007/s00213-019-05389-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 11/04/2019] [Indexed: 12/23/2022]
Abstract
RATIONAL Morphine is one of the most well-known and potent analgesic agents; however, it can also induce various side effects. Thus, finding drugs and mechanisms which can potentiate the analgesic effects of low doses of morphine will be a good strategy for pain management. OBJECTIVE The involvement of μ-opioid receptors and ventral tegmental area (VTA) glutamatergic system in harmaline and morphine combination on the nociceptive response were investigated. Also, we examined reward efficacy and tolerance expression following the drugs. METHODS Animals were bilaterally cannulated in the VTA by stereotaxic instrument. A tail-flick (TF) apparatus and conditioned place preference (CPP) paradigm were used to measure nociceptive response and rewarding effects in male NMRI mice respectively. RESULTS Morphine (2 mg/kg, i.p.) had no effect in TF test. Also, harmaline (1.25 and 5 mg/kg, i.p.) could not change pain threshold. Combination of a non-effective dose of harmaline (5 mg/kg) and morphine (2 mg/kg) produced antinociception and also prevented morphine tolerance but had no effect on the acquisition of CPP. Systemic administration of naloxone (0.5 and 1 mg/kg) and intra-VTA microinjection of NMDA (0.06 and 0.1 μg/mouse) before harmaline (5 mg/kg) plus morphine (2 mg/kg) prevented antinociception induced by the drugs. D-AP5 (0.5 and 1 μg/mouse, intra-VTA) potentiated the effect of low-dose harmaline (1.25 mg/kg) and morphine (2 mg/kg) and induced antinociception. Microinjection of the same doses of NMDA or D-AP5 into the VTA alone had no effect on pain threshold. CONCLUSION The findings showed that harmaline potentiated the analgesic effect of morphine and reduced morphine tolerance. Glutamatergic and μ-opioidergic system interactions in the VTA seem to have a modulatory role in harmaline plus morphine-induced analgesia.
Collapse
Affiliation(s)
- Sakineh Alijanpour
- Department of Biology, Faculty of Science, Gonbad Kavous University, P. O. Box 163, Gonbad Kavous, Iran.
| | - Mohammad-Reza Zarrindast
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran.,Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Bimpisidis Z, Wallén-Mackenzie Å. Neurocircuitry of Reward and Addiction: Potential Impact of Dopamine-Glutamate Co-release as Future Target in Substance Use Disorder. J Clin Med 2019; 8:E1887. [PMID: 31698743 PMCID: PMC6912639 DOI: 10.3390/jcm8111887] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 10/29/2019] [Accepted: 11/01/2019] [Indexed: 12/21/2022] Open
Abstract
Dopamine-glutamate co-release is a unique property of midbrain neurons primarily located in the ventral tegmental area (VTA). Dopamine neurons of the VTA are important for behavioral regulation in response to rewarding substances, including natural rewards and addictive drugs. The impact of glutamate co-release on behaviors regulated by VTA dopamine neurons has been challenging to probe due to lack of selective methodology. However, several studies implementing conditional knockout and optogenetics technologies in transgenic mice have during the past decade pointed towards a role for glutamate co-release in multiple physiological and behavioral processes of importance to substance use and abuse. In this review, we discuss these studies to highlight findings that may be critical when considering mechanisms of importance for prevention and treatment of substance abuse.
Collapse
|
47
|
Gangarossa G, Castell L, Castro L, Tarot P, Veyrunes F, Vincent P, Bertaso F, Valjent E. Contrasting patterns of ERK activation in the tail of the striatum in response to aversive and rewarding signals. J Neurochem 2019; 151:204-226. [PMID: 31245856 DOI: 10.1111/jnc.14804] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/13/2019] [Accepted: 06/19/2019] [Indexed: 01/08/2023]
Abstract
The caudal part of the striatum, also named the tail of the striatum (TS), defines a fourth striatal domain. Determining whether rewarding, aversive and salient stimuli regulate the activity of striatal spiny projections neurons (SPNs) of the TS is therefore of paramount importance to understand its functions, which remain largely elusive. Taking advantage of genetically encoded biosensors (A-kinase activity reporter 3) to record protein kinase A signals and by analyzing the distribution of dopamine D1R- and D2R-SPNs in the TS, we characterized three subterritories: a D2R/A2aR-lacking, a D1R/D2R-intermingled and a D1R/D2R-SPNs-enriched area (corresponding to the amygdalostriatal transition). In addition, we provide evidence that the distribution of D1R- and D2R-SPNs in the TS is evolutionarily conserved (mouse, rat, gerbil). The in vivo analysis of extracellular signal-regulated kinase (ERK) phosphorylation in these TS subterritories in response to distinct appetitive, aversive and pharmacological stimuli revealed that SPNs of the TS are not recruited by stimuli triggering innate aversive responses, fasting, satiety, or palatable signals whereas a reduction in ERK phosphorylation occurred following learned avoidance. In contrast, D1R-SPNs of the intermingled and D2R/A2aR-lacking areas were strongly activated by both D1R agonists and psychostimulant drugs (d-amphetamine, cocaine, 3,4-methyl enedioxy methamphetamine, or methylphenidate), but not by hallucinogens. Finally, a similar pattern of ERK activation was observed by blocking selectively dopamine reuptake. Together, our results reveal that the caudal TS might participate in the processing of specific reward signals and discrete aversive stimuli. Cover Image for this issue: doi: 10.1111/jnc.14526. Open Science: This manuscript was awarded with the Open Materials Badge For more information see: https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Giuseppe Gangarossa
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France.,Université de Paris, BFA, UMR 8251, CNRS, Paris, France
| | - Laia Castell
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Liliana Castro
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, Paris, France
| | - Pauline Tarot
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Frederic Veyrunes
- Institut des Sciences de l'Evolution de Montpellier, ISEM, Université de Montpellier, CNRS, EPHE, IRD, Montpellier, France
| | - Pierre Vincent
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, Paris, France
| | - Federica Bertaso
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Emmanuel Valjent
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| |
Collapse
|
48
|
Cassidy CM, Zucca FA, Girgis RR, Baker SC, Weinstein JJ, Sharp ME, Bellei C, Valmadre A, Vanegas N, Kegeles LS, Brucato G, Kang UJ, Sulzer D, Zecca L, Abi-Dargham A, Horga G. Neuromelanin-sensitive MRI as a noninvasive proxy measure of dopamine function in the human brain. Proc Natl Acad Sci U S A 2019; 116:5108-5117. [PMID: 30796187 PMCID: PMC6421437 DOI: 10.1073/pnas.1807983116] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Neuromelanin-sensitive MRI (NM-MRI) purports to detect the content of neuromelanin (NM), a product of dopamine metabolism that accumulates with age in dopamine neurons of the substantia nigra (SN). Interindividual variability in dopamine function may result in varying levels of NM accumulation in the SN; however, the ability of NM-MRI to measure dopamine function in nonneurodegenerative conditions has not been established. Here, we validated that NM-MRI signal intensity in postmortem midbrain specimens correlated with regional NM concentration even in the absence of neurodegeneration, a prerequisite for its use as a proxy for dopamine function. We then validated a voxelwise NM-MRI approach with sufficient anatomical sensitivity to resolve SN subregions. Using this approach and a multimodal dataset of molecular PET and fMRI data, we further showed the NM-MRI signal was related to both dopamine release in the dorsal striatum and resting blood flow within the SN. These results suggest that NM-MRI signal in the SN is a proxy for function of dopamine neurons in the nigrostriatal pathway. As a proof of concept for its clinical utility, we show that the NM-MRI signal correlated to severity of psychosis in schizophrenia and individuals at risk for schizophrenia, consistent with the well-established dysfunction of the nigrostriatal pathway in psychosis. Our results indicate that noninvasive NM-MRI is a promising tool that could have diverse research and clinical applications to investigate in vivo the role of dopamine in neuropsychiatric illness.
Collapse
Affiliation(s)
- Clifford M Cassidy
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032;
- University of Ottawa Institute of Mental Health Research, affiliated with The Royal, Ottawa, ON K1Z 8N3, Canada
| | - Fabio A Zucca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, 20090 Milan, Italy
| | - Ragy R Girgis
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032
| | - Seth C Baker
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032
| | - Jodi J Weinstein
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032
- Department of Psychiatry, Stony Brook University, Stony Brook, NY 11794
| | - Madeleine E Sharp
- Department of Neurology, Columbia University Medical Center, New York, NY 10032
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Chiara Bellei
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, 20090 Milan, Italy
| | - Alice Valmadre
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, 20090 Milan, Italy
| | - Nora Vanegas
- Department of Neurology, Columbia University Medical Center, New York, NY 10032
| | - Lawrence S Kegeles
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032
| | - Gary Brucato
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032
| | - Un Jung Kang
- Department of Neurology, Columbia University Medical Center, New York, NY 10032
| | - David Sulzer
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Medical Center, New York, NY 10032
| | - Luigi Zecca
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, 20090 Milan, Italy
| | - Anissa Abi-Dargham
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032
- Department of Psychiatry, Stony Brook University, Stony Brook, NY 11794
| | - Guillermo Horga
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032;
| |
Collapse
|
49
|
von Twickel A, Kowatschew D, Saltürk M, Schauer M, Robertson B, Korsching S, Walkowiak W, Grillner S, Pérez-Fernández J. Individual Dopaminergic Neurons of Lamprey SNc/VTA Project to Both the Striatum and Optic Tectum but Restrict Co-release of Glutamate to Striatum Only. Curr Biol 2019; 29:677-685.e6. [DOI: 10.1016/j.cub.2019.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/23/2018] [Accepted: 01/02/2019] [Indexed: 10/27/2022]
|
50
|
Functional neuroanatomical review of the ventral tegmental area. Neuroimage 2019; 191:258-268. [PMID: 30710678 DOI: 10.1016/j.neuroimage.2019.01.062] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 01/22/2019] [Accepted: 01/23/2019] [Indexed: 12/19/2022] Open
Abstract
The ventral tegmental area (VTA) and substantia nigra pars compacta (SNc) are assumed to play a key role in dopamine-related functions such as reward-related behaviour, motivation, addiction and motor functioning. Although dopamine-producing midbrain structures are bordering, they show significant differences in structure and function that argue for a distinction when studying the functions of the dopaminergic midbrain, especially by means of neuroimaging. First, unlike the SNc, the VTA is not a nucleus, which makes it difficult to delineate the structure due to lack of clear anatomical borders. Second, there is no consensus in the literature about the anatomical nomenclature to describe the VTA. Third, these factors in combination with limitations in magnetic resonance imaging (MRI) complicate VTA visualization. We suggest that developing an MRI-compatible probabilistic atlas of the VTA will help to overcome these issues. Such an atlas can be used to identify the individual VTA and serve as region-of-interest for functional MRI.
Collapse
|