1
|
Gruber J, Hanssen R, Qubad M, Bouzouina A, Schack V, Sochor H, Schiweck C, Aichholzer M, Matura S, Slattery DA, Zopf Y, Borgland SL, Reif A, Thanarajah SE. Impact of insulin and insulin resistance on brain dopamine signalling and reward processing- an underexplored mechanism in the pathophysiology of depression? Neurosci Biobehav Rev 2023; 149:105179. [PMID: 37059404 DOI: 10.1016/j.neubiorev.2023.105179] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 04/16/2023]
Abstract
Type 2 diabetes and major depressive disorder (MDD) are the leading causes of disability worldwide and have a high comorbidity rate with fatal outcomes. Despite the long-established association between these conditions, the underlying molecular mechanisms remain unknown. Since the discovery of insulin receptors in the brain and the brain's reward system, evidence has accumulated indicating that insulin modulates dopaminergic (DA) signalling and reward behaviour. Here, we review the evidence from rodent and human studies, that insulin resistance directly alters central DA pathways, which may result in motivational deficits and depressive symptoms. Specifically, we first elaborate on the differential effects of insulin on DA signalling in the ventral tegmental area (VTA) - the primary DA source region in the midbrain - and the striatum as well as its effects on behaviour. We then focus on the alterations induced by insulin deficiency and resistance. Finally, we review the impact of insulin resistance in DA pathways in promoting depressive symptoms and anhedonia on a molecular and epidemiological level and discuss its relevance for stratified treatment strategies.
Collapse
Affiliation(s)
- Judith Gruber
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Ruth Hanssen
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Policlinic for Endocrinology, Diabetology and Prevention Medicine, Germany
| | - Mishal Qubad
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Aicha Bouzouina
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Vivi Schack
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Hannah Sochor
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Carmen Schiweck
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Mareike Aichholzer
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Silke Matura
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Yurdaguel Zopf
- Hector-Center for Nutrition, Exercise and Sports, Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, Canada
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Sharmili Edwin Thanarajah
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany.
| |
Collapse
|
2
|
Effect of Renal Ischemia Reperfusion on Brain Neuroinflammation. Biomedicines 2022; 10:biomedicines10112993. [PMID: 36428560 PMCID: PMC9687457 DOI: 10.3390/biomedicines10112993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Acute kidney injury (AKI) is an inflammatory sequence. It can lead to distant organ injury, including damage to the central nervous system (CNS), mediated by increased circulating cytokines and other inflammatory mediators. It can also lead to increased blood-brain barrier (BBB) permeability. However, the effect of AKI on the inflammatory response of the brain has not yet been investigated. Therefore, we observed the effect of AKI on BBB permeability, microglia and astrocyte activation, and neuronal toxicity in the brain. The striatum and ventral midbrain, known to control overall movement, secrete the neurotransmitter dopamine. The activation of microglia and astrocytes present in this area causes neuro-degenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD). The activation of astrocytes and microglia in the hippocampus and cerebral cortex, which are responsible for important functions, including memory, learning, concentration, and language, can trigger nerve cell apoptosis. The activation of astrocytes and microglia at this site is also involved in the inflammatory response associated with the accumulation of beta-amyloid. In the situation of kidney ischemia reperfusion (IR)-induced AKI, activation of microglia and astrocytes were observed in the striatum, ventral midbrain, hippocampus, and cortex. However, neuronal cell death was not observed until 48 h.
Collapse
|
3
|
Gunasekera B, Diederen K, Bhattacharyya S. Cannabinoids, reward processing, and psychosis. Psychopharmacology (Berl) 2022; 239:1157-1177. [PMID: 33644820 PMCID: PMC9110536 DOI: 10.1007/s00213-021-05801-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 02/10/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Evidence suggests that an overlap exists between the neurobiology of psychotic disorders and the effects of cannabinoids on neurocognitive and neurochemical substrates involved in reward processing. AIMS We investigate whether the psychotomimetic effects of delta-9-tetrahydrocannabinol (THC) and the antipsychotic potential of cannabidiol (CBD) are underpinned by their effects on the reward system and dopamine. METHODS This narrative review focuses on the overlap between altered dopamine signalling and reward processing induced by cannabinoids, pre-clinically and in humans. A systematic search was conducted of acute cannabinoid drug-challenge studies using neuroimaging in healthy subjects and those with psychosis RESULTS: There is evidence of increased striatal presynaptic dopamine synthesis and release in psychosis, as well as abnormal engagement of the striatum during reward processing. Although, acute THC challenges have elicited a modest effect on striatal dopamine, cannabis users generally indicate impaired presynaptic dopaminergic function. Functional MRI studies have identified that a single dose of THC may modulate regions involved in reward and salience processing such as the striatum, midbrain, insular, and anterior cingulate, with some effects correlating with the severity of THC-induced psychotic symptoms. CBD may modulate brain regions involved in reward/salience processing in an opposite direction to that of THC. CONCLUSIONS There is evidence to suggest modulation of reward processing and its neural substrates by THC and CBD. Whether such effects underlie the psychotomimetic/antipsychotic effects of these cannabinoids remains unclear. Future research should address these unanswered questions to understand the relationship between endocannabinoid dysfunction, reward processing abnormalities, and psychosis.
Collapse
Affiliation(s)
- Brandon Gunasekera
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 16 De Crespigny Park, Box P067, London, SE5 8AF, UK
| | - Kelly Diederen
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 16 De Crespigny Park, Box P067, London, SE5 8AF, UK
| | - Sagnik Bhattacharyya
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 16 De Crespigny Park, Box P067, London, SE5 8AF, UK.
| |
Collapse
|
4
|
Pavia-Collado R, Rodríguez-Aller R, Alarcón-Arís D, Miquel-Rio L, Ruiz-Bronchal E, Paz V, Campa L, Galofré M, Sgambato V, Bortolozzi A. Up and Down γ-Synuclein Transcription in Dopamine Neurons Translates into Changes in Dopamine Neurotransmission and Behavioral Performance in Mice. Int J Mol Sci 2022; 23:ijms23031807. [PMID: 35163729 PMCID: PMC8836558 DOI: 10.3390/ijms23031807] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 12/15/2022] Open
Abstract
The synuclein family consists of α-, β-, and γ-Synuclein (α-Syn, β-Syn, and γ-Syn) expressed in the neurons and concentrated in synaptic terminals. While α-Syn is at the center of interest due to its implication in the pathogenesis of Parkinson's disease (PD) and other synucleinopathies, limited information exists on the other members. The current study aimed at investigating the biological role of γ-Syn controlling the midbrain dopamine (DA) function. We generated two different mouse models with: (i) γ-Syn overexpression induced by an adeno-associated viral vector and (ii) γ-Syn knockdown induced by a ligand-conjugated antisense oligonucleotide, in order to modify the endogenous γ-Syn transcription levels in midbrain DA neurons. The progressive overexpression of γ-Syn decreased DA neurotransmission in the nigrostriatal and mesocortical pathways. In parallel, mice evoked motor deficits in the rotarod and impaired cognitive performance as assessed by novel object recognition, passive avoidance, and Morris water maze tests. Conversely, acute γ-Syn knockdown selectively in DA neurons facilitated forebrain DA neurotransmission. Importantly, modifications in γ-Syn expression did not induce the loss of DA neurons or changes in α-Syn expression. Collectively, our data strongly suggest that DA release/re-uptake processes in the nigrostriatal and mesocortical pathways are partially dependent on substantia nigra pars compacta /ventral tegmental area (SNc/VTA) γ-Syn transcription levels, and are linked to modulation of DA transporter function, similar to α-Syn.
Collapse
Affiliation(s)
- Rubén Pavia-Collado
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (R.P.-C.); (D.A.-A.); (L.M.-R.); (E.R.-B.); (V.P.); (L.C.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain
- miCure Therapeutics Ltd., Tel Aviv 6423902, Israel
| | - Raquel Rodríguez-Aller
- CHU de Quebec Research Center, Axe Neurosciences, Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1V 4G2, Canada;
- CERVO Brain Research Centre, Quebec City, QC G1J 2G3, Canada
| | - Diana Alarcón-Arís
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (R.P.-C.); (D.A.-A.); (L.M.-R.); (E.R.-B.); (V.P.); (L.C.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain
| | - Lluís Miquel-Rio
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (R.P.-C.); (D.A.-A.); (L.M.-R.); (E.R.-B.); (V.P.); (L.C.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain
| | - Esther Ruiz-Bronchal
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (R.P.-C.); (D.A.-A.); (L.M.-R.); (E.R.-B.); (V.P.); (L.C.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain
| | - Verónica Paz
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (R.P.-C.); (D.A.-A.); (L.M.-R.); (E.R.-B.); (V.P.); (L.C.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain
| | - Leticia Campa
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (R.P.-C.); (D.A.-A.); (L.M.-R.); (E.R.-B.); (V.P.); (L.C.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain
| | - Mireia Galofré
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedicine, Faculty of Medicine and Health Science, University of Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28029 Madrid, Spain
| | - Véronique Sgambato
- CNRS, Institut des Sciences Cognitives Marc Jeannerod, UMR 5229, 69675 Bron, France;
| | - Analia Bortolozzi
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (R.P.-C.); (D.A.-A.); (L.M.-R.); (E.R.-B.); (V.P.); (L.C.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-93-363-8313
| |
Collapse
|
5
|
Gui Y, Grzyb K, Thomas MH, Ohnmacht J, Garcia P, Buttini M, Skupin A, Sauter T, Sinkkonen L. Single-nuclei chromatin profiling of ventral midbrain reveals cell identity transcription factors and cell-type-specific gene regulatory variation. Epigenetics Chromatin 2021; 14:43. [PMID: 34503558 PMCID: PMC8427957 DOI: 10.1186/s13072-021-00418-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/24/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Cell types in ventral midbrain are involved in diseases with variable genetic susceptibility, such as Parkinson's disease and schizophrenia. Many genetic variants affect regulatory regions and alter gene expression in a cell-type-specific manner depending on the chromatin structure and accessibility. RESULTS We report 20,658 single-nuclei chromatin accessibility profiles of ventral midbrain from two genetically and phenotypically distinct mouse strains. We distinguish ten cell types based on chromatin profiles and analysis of accessible regions controlling cell identity genes highlights cell-type-specific key transcription factors. Regulatory variation segregating the mouse strains manifests more on transcriptome than chromatin level. However, cell-type-level data reveals changes not captured at tissue level. To discover the scope and cell-type specificity of cis-acting variation in midbrain gene expression, we identify putative regulatory variants and show them to be enriched at differentially expressed loci. Finally, we find TCF7L2 to mediate trans-acting variation selectively in midbrain neurons. CONCLUSIONS Our data set provides an extensive resource to study gene regulation in mesencephalon and provides insights into control of cell identity in the midbrain and identifies cell-type-specific regulatory variation possibly underlying phenotypic and behavioural differences between mouse strains.
Collapse
Affiliation(s)
- Yujuan Gui
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Belvaux, Luxembourg
| | - Kamil Grzyb
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Mélanie H Thomas
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Jochen Ohnmacht
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Belvaux, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Pierre Garcia
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Manuel Buttini
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Thomas Sauter
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Belvaux, Luxembourg
| | - Lasse Sinkkonen
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Belvaux, Luxembourg.
| |
Collapse
|
6
|
Expression of a novel brain specific isoform of C3G is regulated during development. Sci Rep 2020; 10:18838. [PMID: 33139841 PMCID: PMC7606606 DOI: 10.1038/s41598-020-75813-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 10/01/2020] [Indexed: 12/13/2022] Open
Abstract
Mice lacking C3G (RapGEF1), a ubiquitously expressed protein essential for neuronal differentiation, show multiple defects in brain development. Function of C3G in neurogenesis is poorly defined. Here, we identify brain specific expression of a novel C3G isoform in mice and humans. This isoform has an insert in the Crk-binding region, generating a polypeptide of 175 kDa, unlike the previously known 140 kDa form expressed in all other tissues. In the adult mouse brain, C3G expression is seen in neurons, but was not detectable in GFAP-positive cells. C3G levels were high in the CA3 region of hippocampus and in mitral cells of olfactory bulb. Neural progenitor cells positive for Doublecortin and Nestin, show expression of C3G. During development, C3G is expressed in precursor cells prior to their differentiation into mature neurons or astrocytes. The 175 kDa as well as 140 kDa forms are seen in embryonic mouse brain, while only the 175 kDa variant is seen in post-natal brain. Human cerebral organoids generated from induced pluripotent stem cells predominantly expressed the 140 kDa polypeptides, and the 175 kDa isoform appeared upon maturation. This study describes developmental regulation and neuronal expression of a brain specific isoform of C3G, a molecule essential for normal development of the mammalian brain.
Collapse
|
7
|
Single-cell transcriptomics reveals multiple neuronal cell types in human midbrain-specific organoids. Cell Tissue Res 2020; 382:463-476. [PMID: 32737576 PMCID: PMC7683480 DOI: 10.1007/s00441-020-03249-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023]
Abstract
Human stem cell-derived organoids have great potential for modelling physiological and pathological processes. They recapitulate in vitro the organization and function of a respective organ or part of an organ. Human midbrain organoids (hMOs) have been described to contain midbrain-specific dopaminergic neurons that release the neurotransmitter dopamine. However, the human midbrain contains also additional neuronal cell types, which are functionally interacting with each other. Here, we analysed hMOs at high-resolution by means of single-cell RNA sequencing (scRNA-seq), imaging and electrophysiology to unravel cell heterogeneity. Our findings demonstrate that hMOs show essential neuronal functional properties as spontaneous electrophysiological activity of different neuronal subtypes, including dopaminergic, GABAergic, glutamatergic and serotonergic neurons. Recapitulating these in vivo features makes hMOs an excellent tool for in vitro disease phenotyping and drug discovery.
Collapse
|
8
|
Smits LM, Schwamborn JC. Midbrain Organoids: A New Tool to Investigate Parkinson's Disease. Front Cell Dev Biol 2020; 8:359. [PMID: 32509785 PMCID: PMC7248385 DOI: 10.3389/fcell.2020.00359] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 04/22/2020] [Indexed: 12/12/2022] Open
Abstract
The study of human 3D cell culture models not only bridges the gap between traditional 2D in vitro experiments and in vivo animal models, it also addresses processes that cannot be recapitulated by either of these traditional models. Therefore, it offers an opportunity to better understand complex biology including brain development. The brain organoid technology provides a physiologically relevant context, which holds great potential for its application in modeling neurological diseases. Here, we compare different methods to obtain highly specialized structures that resemble specific features of the human midbrain. Regionally patterned neural stem cells (NSCs) were utilized to derive such human midbrain-specific organoids (hMO). The resulting neural tissue exhibited abundant neurons with midbrain dopaminergic neuron identity, as well as astroglia and oligodendrocyte differentiation. Within the midbrain organoids, neurite myelination, and the formation of synaptic connections were observed. Regular neuronal fire patterning and neural network synchronicity were determined by multielectrode array recordings. In addition to electrophysiologically functional neurons producing and secreting dopamine, responsive neuronal subtypes, such as GABAergic and glutamatergic neurons were also detected. In order to model disorders like Parkinson's disease (PD) in vitro, midbrain organoids carrying a disease specific mutation were derived and compared to healthy control organoids to investigate relevant neurodegenerative pathophysiology. In this way midbrain-specific organoids constitute a powerful tool for human-specific in vitro modeling of neurological disorders with a great potential to be utilized in advanced therapy development.
Collapse
|
9
|
Bambico FR, Li Z, Oliveira C, McNeill S, Diwan M, Raymond R, Nobrega JN. Rostrocaudal subregions of the ventral tegmental area are differentially impacted by chronic stress. Psychopharmacology (Berl) 2019; 236:1917-1929. [PMID: 30796492 DOI: 10.1007/s00213-019-5177-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 01/21/2019] [Indexed: 01/17/2023]
Abstract
RATIONALE The ventral tegmental area (VTA) is implicated in the pathophysiology of depression and addictive disorders and is subject to the detrimental effects of stress. Chronic stress may differentially alter the activity pattern of its different subregions along the rostrocaudal and dorsoventral axes, which may relate to the variable behavioral sensitivity to stress mediated by these subregions. OBJECTIVES Here, chronic stress-exposed rats were tested for depressive-like reactivity. In situ hybridization for zif268 as a marker of neuronal activation was combined with in vivo single-unit recording of dopaminergic neurons to assess modifications in the activity of the rostral VTA (rVTA) and caudal VTA (cVTA). Changes in the expression of stress-responsive glucocorticoid receptors (GR) and brain-derived neurotrophic factor (BDNF) were also assessed. RESULTS Stress-induced anhedonia-like, hyper-anxious, and passive-like responding were associated with reductions in dopaminergic burst activity in the cVTA and an increase in local GABAergic activity, particularly in GABAA receptor sensitivity. On the other hand, stress increased single-spiking activity, burst activity, and zif268 mRNA levels in the rVTA, which were associated with increased glutamatergic tonus and enhanced GR and AMPA receptor (AMPAR) expression. rVTA and cVTA activity differentially correlated with sucrose preference and passivity measures. CONCLUSIONS These data demonstrate that the rVTA and cVTA respond differently to stress and suggest that while cVTA activity may be related to passivity-like states, the activity of both subregions appears to be related to anhedonia and the processing of incentive value. These region-dependent abnormalities indicate the multi-modular composition of the VTA, which could provide multiple substrates for different symptom features.
Collapse
Affiliation(s)
- Francis Rodriguez Bambico
- Behavioural Neurobiology Laboratory, Campbell Family Mental Health Research Institute and Research Imaging Center, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R82, Canada. .,Department of Psychology, Memorial University of Newfoundland, St John's, NL, A1B 3X9, Canada.
| | - Zhuoliang Li
- Behavioural Neurobiology Laboratory, Campbell Family Mental Health Research Institute and Research Imaging Center, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R82, Canada
| | - Caio Oliveira
- Behavioural Neurobiology Laboratory, Campbell Family Mental Health Research Institute and Research Imaging Center, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R82, Canada
| | - Sean McNeill
- Behavioural Neurobiology Laboratory, Campbell Family Mental Health Research Institute and Research Imaging Center, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R82, Canada
| | - Mustansir Diwan
- Behavioural Neurobiology Laboratory, Campbell Family Mental Health Research Institute and Research Imaging Center, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R82, Canada
| | - Roger Raymond
- Behavioural Neurobiology Laboratory, Campbell Family Mental Health Research Institute and Research Imaging Center, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R82, Canada
| | - José N Nobrega
- Behavioural Neurobiology Laboratory, Campbell Family Mental Health Research Institute and Research Imaging Center, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R82, Canada
| |
Collapse
|
10
|
Scherma M, Masia P, Satta V, Fratta W, Fadda P, Tanda G. Brain activity of anandamide: a rewarding bliss? Acta Pharmacol Sin 2019; 40:309-323. [PMID: 30050084 DOI: 10.1038/s41401-018-0075-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 05/20/2018] [Indexed: 12/11/2022] Open
Abstract
Anandamide is a lipid mediator that acts as an endogenous ligand of CB1 receptors. These receptors are also the primary molecular target responsible for the pharmacological effects of Δ9-tetrahydrocannabinol, the psychoactive ingredient in Cannabis sativa. Several studies demonstrate that anandamide exerts an overall modulatory effect on the brain reward circuitry. Several reports suggest its involvement in the addiction-producing actions of other abused drugs, and it can also act as a behavioral reinforcer in animal models of drug abuse. Importantly, all these effects of anandamide appear to be potentiated by pharmacological inhibition of its metabolic degradation. Enhanced brain levels of anandamide after treatment with inhibitors of fatty acid amide hydrolase, the main enzyme responsible for its degradation, seem to affect the rewarding and reinforcing actions of many drugs of abuse. In this review, we will provide an overview from a preclinical perspective of the current state of knowledge regarding the behavioral pharmacology of anandamide, with a particular emphasis on its motivational/reinforcing properties. We will also discuss how modulation of anandamide levels through inhibition of enzymatic metabolic pathways could provide a basis for developing new pharmaco-therapeutic tools for the treatment of substance use disorders.
Collapse
|
11
|
You C, Savarese A, Vandegrift BJ, He D, Pandey SC, Lasek AW, Brodie MS. Ethanol acts on KCNK13 potassium channels in the ventral tegmental area to increase firing rate and modulate binge-like drinking. Neuropharmacology 2019; 144:29-36. [PMID: 30332606 PMCID: PMC6286249 DOI: 10.1016/j.neuropharm.2018.10.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/27/2018] [Accepted: 10/10/2018] [Indexed: 12/13/2022]
Abstract
Alcohol excitation of the ventral tegmental area (VTA) is important in neurobiological processes related to the development of alcoholism. The ionotropic receptors on VTA neurons that mediate ethanol-induced excitation have not been identified. Quinidine blocks ethanol excitation of VTA neurons, and blockade of two-pore potassium channels is among the actions of quinidine. Therefore two-pore potassium channels in the VTA may be potential targets for the action of ethanol. Here, we explored whether ethanol activation of VTA neurons is mediated by the two-pore potassium channel KCNK13. Extracellular recordings of the response of VTA neurons to ethanol were performed in combination with knockdown of Kcnk13 using a short hairpin RNA (shRNA) in C57BL/6 J mice. Real-time PCR and immunohistochemistry were used to examine expression of this channel in the VTA. Finally, the role of KCNK13 in binge-like drinking was examined in the drinking in the dark test after knockdown of the channel. Kcnk13 expression in the VTA was increased by acute ethanol exposure. Ethanol-induced excitation of VTA neurons was selectively reduced by shRNA targeting Kcnk13. Importantly, knockdown of Kcnk13 in the VTA resulted in increased alcohol drinking. These results are consistent with the idea that ethanol stimulates VTA neurons at least in part by inhibiting KCNK13, a specific two-pore potassium channel, and that KCNK13 can control both VTA neuronal activity and binge drinking. KCNK13 is a novel alcohol-sensitive molecular target and may be amenable to the development of pharmacotherapies for alcoholism treatment.
Collapse
Affiliation(s)
- Chang You
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, 60612, USA; Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Antonia Savarese
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Bertha J Vandegrift
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, 60612, USA; Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Donghong He
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA; Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
| | - Amy W Lasek
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Mark S Brodie
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, 60612, USA; Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
12
|
N-oleoyldopamine modulates activity of midbrain dopaminergic neurons through multiple mechanisms. Neuropharmacology 2017; 119:111-122. [PMID: 28400256 DOI: 10.1016/j.neuropharm.2017.04.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/07/2017] [Accepted: 04/07/2017] [Indexed: 11/23/2022]
Abstract
N-oleoyl-dopamine (OLDA) is an amide of dopamine and oleic acid, synthesized in catecholaminergic neurons. The present study investigates OLDA targets in midbrain dopaminergic (DA) neurons. Substantia Nigra compacta (SNc) DA neurons recorded in brain slices were excited by OLDA in wild type mice. In transient receptor potential vanilloid 1 (TRPV1) knockout (KO) mice, however, SNc DA neurons displayed sustained inhibition of firing. In the presence of the dopamine type 2 receptor (D2R) antagonist sulpiride or the dopamine transporter blocker nomifensine no such inhibition was observed. Under sulpiride OLDA slightly excited SNc DA neurons, an action abolished upon combined application of the cannabinoid1 and 2 receptor antagonists AM251 and AM630. In ventral tegmental area (VTA) DA neurons from TRPV1 KO mice a transient inhibition of firing by OLDA was observed. Thus OLDA modulates the firing of nigrostriatal DA neurons through interactions with TRPV1, cannabinoid receptors and dopamine uptake. These findings suggest further development of OLDA-like tandem molecules for the treatment of movement disorders including Parkinson's disease.
Collapse
|
13
|
Vena AA, Mangieri R, Gonzales RA. Regional Analysis of the Pharmacological Effects of Acute Ethanol on Extracellular Striatal Dopamine Activity. Alcohol Clin Exp Res 2016; 40:2528-2536. [PMID: 27785807 PMCID: PMC5133149 DOI: 10.1111/acer.13246] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 09/20/2016] [Indexed: 01/20/2023]
Abstract
BACKGROUND The objective of this study was to characterize the acute pharmacological effects of ethanol (EtOH) on extracellular dopamine in the dorsomedial and dorsolateral striata. This is the first study to quantify and directly compare the effects of acute EtOH on dopamine in these subregions. Therefore, we also tested the nucleus accumbens as a positive control. We hypothesized that while EtOH may increase extracellular dopamine in the dorsomedial striatum and dorsolateral striatum, the magnitude of this increase and the temporal profiles of extracellular dopamine concentrations would differ among the dorsomedial striatum, dorsolateral striatum, and nucleus accumbens. METHODS We performed in vivo microdialysis in adult, male Long Evans rats as they received a single (experiment 1) or repeated (experiment 2) doses of EtOH. RESULTS The results of our positive control study validate earlier work by our laboratory demonstrating that acute intravenous EtOH immediately and robustly increases extracellular dopamine in the nucleus accumbens (Howard et al., ). In contrast, a single 1-g/kg dose of intravenous EtOH did not significantly affect extracellular dopamine in the dorsomedial striatum or the dorsolateral striatum. However, following a cumulative EtOH dosing protocol, we observed a ramping up of tonic dopamine activity in both the dorsomedial striatum and dorsolateral striatum over the course of the experiment, but this effect was more robust in the dorsomedial striatum. CONCLUSIONS These results suggest that distinct mechanisms underlie the stimulating effects of acute EtOH on extracellular dopamine in striatal subregions. Additionally, our findings suggest a role for the dorsomedial striatum and minimal-to-no role for the dorsolateral striatum in mediating the intoxicating effects of acute moderate to high doses of EtOH.
Collapse
Affiliation(s)
- Ashley A Vena
- Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas at Austin, Austin, Texas
| | - Regina Mangieri
- Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas at Austin, Austin, Texas
| | - Rueben A Gonzales
- Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas at Austin, Austin, Texas
| |
Collapse
|
14
|
Vogt Weisenhorn DM, Giesert F, Wurst W. Diversity matters - heterogeneity of dopaminergic neurons in the ventral mesencephalon and its relation to Parkinson's Disease. J Neurochem 2016; 139 Suppl 1:8-26. [PMID: 27206718 PMCID: PMC5096020 DOI: 10.1111/jnc.13670] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/25/2016] [Accepted: 05/17/2016] [Indexed: 12/25/2022]
Abstract
Dopaminergic neurons in the ventral mesencephalon (the ventral mesencephalic dopaminergic complex) are known for their role in a multitude of behaviors, including cognition, reward, addiction and voluntary movement. Dysfunctions of these neurons are the underlying cause of various neuropsychiatric disorders, such as depression, addiction and schizophrenia. In addition, Parkinson's disease (PD), which is the second most common degenerative disease in developed countries, is characterized by the degeneration of dopaminergic neurons, leading to the core motor symptoms of the disease. However, only a subset of dopaminergic neurons in the ventral mesencephalon is highly vulnerable to the disease process. Indeed, research over several decades revealed that the neurons in the ventral mesencephalic dopaminergic complex do not form a homogeneous group with respect to anatomy, physiology, function, molecular identity or vulnerability/dysfunction in different diseases. Here, we review how the concept of dopaminergic neuron diversity, assisted by the advent and application of new technologies, evolved and was refined over time and how it shaped our understanding of PD pathogenesis. Understanding this diversity of neurons in the ventral mesencephalic dopaminergic complex at all levels is imperative for the development of new and more selective drugs for both PD and various other neuropsychiatric diseases. Several decades of research revealed that the neurons in the ventral mesencephalic dopaminergic complex do not form a homogeneous group in respect to anatomy, physiology, function, molecular identity or vulnerability/dysfunction in diseases like Parkinson's disease (PD). Here, we review how this concept evolved and was refined over time and how it shaped our understanding of the pathogenesis of PD. Source of the midbrain image: www.wikimd.org/wiki/index.php/The_Midbrain_or_Mesencephalon; downloaded 28.01.2016. See also Figures and of the paper. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Daniela Maria Vogt Weisenhorn
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany
- Technische Universität München-Weihenstephan, Lehrstuhl für Entwicklungsgenetik, c/o Helmholtz Zentrum München, Neuherberg, Germany
| | - Florian Giesert
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany
- Technische Universität München-Weihenstephan, Lehrstuhl für Entwicklungsgenetik, c/o Helmholtz Zentrum München, Neuherberg, Germany
| | - Wolfgang Wurst
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany.
- Technische Universität München-Weihenstephan, Lehrstuhl für Entwicklungsgenetik, c/o Helmholtz Zentrum München, Neuherberg, Germany.
- Deutsches Zentrum für Neurodegenerative Erkrankungen e. V. (DZNE), Standort München, München, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, München, Germany.
| |
Collapse
|
15
|
Jo J, Xiao Y, Sun AX, Cukuroglu E, Tran HD, Göke J, Tan ZY, Saw TY, Tan CP, Lokman H, Lee Y, Kim D, Ko HS, Kim SO, Park JH, Cho NJ, Hyde TM, Kleinman JE, Shin JH, Weinberger DR, Tan EK, Je HS, Ng HH. Midbrain-like Organoids from Human Pluripotent Stem Cells Contain Functional Dopaminergic and Neuromelanin-Producing Neurons. Cell Stem Cell 2016; 19:248-257. [PMID: 27476966 DOI: 10.1016/j.stem.2016.07.005] [Citation(s) in RCA: 542] [Impact Index Per Article: 67.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 04/22/2016] [Accepted: 07/08/2016] [Indexed: 12/13/2022]
Abstract
Recent advances in 3D culture systems have led to the generation of brain organoids that resemble different human brain regions; however, a 3D organoid model of the midbrain containing functional midbrain dopaminergic (mDA) neurons has not been reported. We developed a method to differentiate human pluripotent stem cells into a large multicellular organoid-like structure that contains distinct layers of neuronal cells expressing characteristic markers of human midbrain. Importantly, we detected electrically active and functionally mature mDA neurons and dopamine production in our 3D midbrain-like organoids (MLOs). In contrast to human mDA neurons generated using 2D methods or MLOs generated from mouse embryonic stem cells, our human MLOs produced neuromelanin-like granules that were structurally similar to those isolated from human substantia nigra tissues. Thus our MLOs bearing features of the human midbrain may provide a tractable in vitro system to study the human midbrain and its related diseases.
Collapse
Affiliation(s)
- Junghyun Jo
- Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672, Singapore
| | - Yixin Xiao
- Signature Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Alfred Xuyang Sun
- Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672, Singapore; National Neuroscience Institute, 20 College Road, Singapore 169856, Singapore
| | - Engin Cukuroglu
- Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672, Singapore
| | - Hoang-Dai Tran
- Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672, Singapore; Department of Biochemistry, National University of Singapore, 8 Medical Drive, Singapore 117597, Singapore
| | - Jonathan Göke
- Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672, Singapore
| | - Zi Ying Tan
- Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672, Singapore; Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore
| | - Tzuen Yih Saw
- Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672, Singapore
| | - Cheng-Peow Tan
- Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672, Singapore
| | - Hidayat Lokman
- Signature Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Younghwan Lee
- Signature Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Donghoon Kim
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Han Seok Ko
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Seong-Oh Kim
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Jae Hyeon Park
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Nam-Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 136-701, Korea
| | - Thomas M Hyde
- The Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA; Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joel E Kleinman
- The Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA; Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joo Heon Shin
- The Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Daniel R Weinberger
- The Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA; Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Eng King Tan
- National Neuroscience Institute, 20 College Road, Singapore 169856, Singapore
| | - Hyunsoo Shawn Je
- Signature Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
| | - Huck-Hui Ng
- Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672, Singapore; Department of Biochemistry, National University of Singapore, 8 Medical Drive, Singapore 117597, Singapore; Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore; School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 639798, Singapore.
| |
Collapse
|
16
|
Kanlikilicer P, Zhang D, Dragomir A, Akay YM, Akay M. Gene expression profiling of midbrain dopamine neurons upon gestational nicotine exposure. Med Biol Eng Comput 2016; 55:467-482. [PMID: 27255453 DOI: 10.1007/s11517-016-1531-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 05/18/2016] [Indexed: 12/11/2022]
Abstract
Maternal smoking during pregnancy is associated with low birth weight, increased risk of stillbirth, conduct disorder, attention-deficit/hyperactivity disorder and neurocognitive deficits. Ventral tegmental area dopamine (DA) neurons in the mesocorticolimbic pathway were suggested to play a critical role in these pathological mechanisms induced by nicotine. Nicotine-mediated changes in genetic expression during pregnancy are of great interest for current researchers. We used patch clamp methods to identify and harvest DA and non-DA neurons separately and assayed them using oligonucleotide arrays to elucidate the alterations in gene expressions in these cells upon gestational nicotine exposure. Microarray analysis identified a set of 135 genes as significantly differentially expressed between DA and non-DA neurons. Some of the genes were found to be related to neurological disease pathways, such as Alzheimer's disease, Parkinson's disease and Huntington's disease. Significantly up-/down-regulated genes found in DA neurons were mostly related to G-protein-coupled protein receptor signaling and developmental processes. These alterations in gene expressions may explain, partially at least, the possible pathological mechanisms for the diseases induced by maternal smoking.
Collapse
Affiliation(s)
- Pınar Kanlikilicer
- Department of Biomedical Engineering, University of Houston, SERC Building, 3605 Cullen Blvd, Houston, TX, 77204, USA
| | - Die Zhang
- Department of Biomedical Engineering, University of Houston, SERC Building, 3605 Cullen Blvd, Houston, TX, 77204, USA
| | - Andrei Dragomir
- Department of Biomedical Engineering, University of Houston, SERC Building, 3605 Cullen Blvd, Houston, TX, 77204, USA
| | - Yasemin M Akay
- Department of Biomedical Engineering, University of Houston, SERC Building, 3605 Cullen Blvd, Houston, TX, 77204, USA
| | - Metin Akay
- Department of Biomedical Engineering, University of Houston, SERC Building, 3605 Cullen Blvd, Houston, TX, 77204, USA.
| |
Collapse
|
17
|
Guilarte TR, Gonzales KK. Manganese-Induced Parkinsonism Is Not Idiopathic Parkinson's Disease: Environmental and Genetic Evidence. Toxicol Sci 2016. [PMID: 26220508 DOI: 10.1093/toxsci/kfv099] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Movement abnormalities caused by chronic manganese (Mn) intoxication clinically resemble but are not identical to those in idiopathic Parkinson's disease. In fact, the most successful parkinsonian drug treatment, the dopamine precursor levodopa, is ineffective in alleviating Mn-induced motor symptoms, implying that parkinsonism in Mn-exposed individuals may not be linked to midbrain dopaminergic neuron cell loss. Over the last decade, supporting evidence from human and nonhuman primates has emerged that Mn-induced parkinsonism partially results from damage to basal ganglia nuclei of the striatal "direct pathway" (ie, the caudate/putamen, internal globus pallidus, and substantia nigra pars reticulata) and a marked inhibition of striatal dopamine release in the absence of nigrostriatal dopamine terminal degeneration. Recent neuroimaging studies have revealed similar findings in a particular group of young drug users intravenously injecting the Mn-containing psychostimulant ephedron and in individuals with inherited mutations of the Mn transporter gene SLC30A10. This review will provide a detailed discussion about the aforementioned studies, followed by a comparison with their rodent analogs and idiopathic parkinsonism. Together, these findings in combination with a limited knowledge about the underlying neuropathology of Mn-induced parkinsonism strongly support the need for a more complete understanding of the neurotoxic effects of Mn on basal ganglia function to uncover the appropriate cellular and molecular therapeutic targets for this disorder.
Collapse
Affiliation(s)
- Tomás R Guilarte
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032
| | - Kalynda K Gonzales
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032
| |
Collapse
|
18
|
Abstract
Variations in the fat mass and obesity-associated (FTO) gene are linked to obesity. However, the underlying neurobiological mechanisms by which these genetic variants influence obesity, behavior, and brain are unknown. Given that Fto regulates D2/3R signaling in mice, we tested in humans whether variants in FTO would interact with a variant in the ANKK1 gene, which alters D2R signaling and is also associated with obesity. In a behavioral and fMRI study, we demonstrate that gene variants of FTO affect dopamine (D2)-dependent midbrain brain responses to reward learning and behavioral responses associated with learning from negative outcome in humans. Furthermore, dynamic causal modeling confirmed that FTO variants modulate the connectivity in a basic reward circuit of meso-striato-prefrontal regions, suggesting a mechanism by which genetic predisposition alters reward processing not only in obesity, but also in other disorders with altered D2R-dependent impulse control, such as addiction. Significance statement: Variations in the fat mass and obesity-associated (FTO) gene are associated with obesity. Here we demonstrate that variants of FTO affect dopamine-dependent midbrain brain responses and learning from negative outcomes in humans during a reward learning task. Furthermore, FTO variants modulate the connectivity in a basic reward circuit of meso-striato-prefrontal regions, suggesting a mechanism by which genetic vulnerability in reward processing can increase predisposition to obesity.
Collapse
|
19
|
Merrill CB, Friend LN, Newton ST, Hopkins ZH, Edwards JG. Ventral tegmental area dopamine and GABA neurons: Physiological properties and expression of mRNA for endocannabinoid biosynthetic elements. Sci Rep 2015; 5:16176. [PMID: 26553597 PMCID: PMC4639757 DOI: 10.1038/srep16176] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/08/2015] [Indexed: 11/12/2022] Open
Abstract
The ventral tegmental area (VTA) is involved in adaptive reward and motivation processing and is composed of dopamine (DA) and GABA neurons. Defining the elements regulating activity and synaptic plasticity of these cells is critical to understanding mechanisms of reward and addiction. While endocannabinoids (eCBs) that potentially contribute to addiction are known to be involved in synaptic plasticity mechanisms in the VTA, where they are produced is poorly understood. In this study, DA and GABAergic cells were identified using electrophysiology, cellular markers, and a transgenic mouse model that specifically labels GABA cells. Using single-cell RT-qPCR and immunohistochemistry, we investigated mRNA and proteins involved in eCB signaling such as diacylglycerol lipase α, N-acyl-phosphatidylethanolamine-specific phospholipase D, and 12-lipoxygenase, as well as type I metabotropic glutamate receptors (mGluRs). Our results demonstrate the first molecular evidence of colocalization of eCB biosynthetic enzyme and type I mGluR mRNA in VTA neurons. Further, these data reveal higher expression of mGluR1 in DA neurons, suggesting potential differences in eCB synthesis between DA and GABA neurons. These data collectively suggest that VTA GABAergic and DAergic cells have the potential to produce various eCBs implicated in altering neuronal activity or plasticity in adaptive motivational reward or addiction.
Collapse
Affiliation(s)
- Collin B Merrill
- Brigham Young University Department of Physiology and Developmental Biology Provo, UT 84602 USA
| | - Lindsey N Friend
- Brigham Young University Neuroscience Center Provo, UT 84602 USA
| | - Scott T Newton
- Brigham Young University Neuroscience Center Provo, UT 84602 USA
| | | | - Jeffrey G Edwards
- Brigham Young University Department of Physiology and Developmental Biology Provo, UT 84602 USA.,Brigham Young University Neuroscience Center Provo, UT 84602 USA
| |
Collapse
|
20
|
Lammel S, Steinberg EE, Földy C, Wall NR, Beier K, Luo L, Malenka RC. Diversity of transgenic mouse models for selective targeting of midbrain dopamine neurons. Neuron 2015; 85:429-38. [PMID: 25611513 DOI: 10.1016/j.neuron.2014.12.036] [Citation(s) in RCA: 238] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2014] [Indexed: 10/24/2022]
Abstract
Ventral tegmental area (VTA) dopamine (DA) neurons have been implicated in reward, aversion, salience, cognition, and several neuropsychiatric disorders. Optogenetic approaches involving transgenic Cre-driver mouse lines provide powerful tools for dissecting DA-specific functions. However, the emerging complexity of VTA circuits requires Cre-driver mouse lines that restrict transgene expression to a precisely defined cell population. Because of recent work reporting that VTA DA neurons projecting to the lateral habenula release GABA, but not DA, we performed an extensive anatomical, molecular, and functional characterization of prominent DA transgenic mouse driver lines. We find that transgenes under control of the tyrosine hydroxylase, but not the dopamine transporter, promoter exhibit dramatic non-DA cell-specific expression patterns within and around VTA nuclei. Our results demonstrate how Cre expression in unintentionally targeted cells in transgenic mouse lines can confound the interpretation of supposedly cell-type-specific experiments. This Matters Arising paper is in response to Stamatakis et al. (2013), published in Neuron. See also the Matters Arising Response paper by Stuber et al. (2015), published concurrently with this Matters Arising in Neuron.
Collapse
Affiliation(s)
- Stephan Lammel
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford CA 94305-5453, USA.
| | - Elizabeth E Steinberg
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford CA 94305-5453, USA
| | - Csaba Földy
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford CA 94305-5453, USA; Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford CA 94305-5453, USA
| | - Nicholas R Wall
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford CA 94305-5453, USA
| | - Kevin Beier
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford CA 94305-5453, USA; Howard Hughes Medical Institute and Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Liqun Luo
- Howard Hughes Medical Institute and Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Robert C Malenka
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford CA 94305-5453, USA.
| |
Collapse
|
21
|
Liechti R, Ducray AD, Jensen P, Di Santo S, Seiler S, Jensen CH, Meyer M, Widmer HR. Characterization of fetal antigen 1/delta-like 1 homologue expressing cells in the rat nigrostriatal system: effects of a unilateral 6-hydroxydopamine lesion. PLoS One 2015; 10:e0116088. [PMID: 25723595 PMCID: PMC4344227 DOI: 10.1371/journal.pone.0116088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/04/2014] [Indexed: 11/18/2022] Open
Abstract
Fetal antigen 1/delta-like 1 homologue (FA1/dlk1) belongs to the epidermal growth factor superfamily and is considered to be a non-canonical ligand for the Notch receptor. Interactions between Notch and its ligands are crucial for the development of various tissues. Moreover, FA1/dlk1 has been suggested as a potential supplementary marker of dopaminergic neurons. The present study aimed at investigating the distribution of FA1/dlk1-immunoreactive (-ir) cells in the early postnatal and adult midbrain as well as in the nigrostriatal system of 6-hydroxydopamine (6-OHDA)-lesioned hemiparkinsonian adult rats. FA1/dlk1-ir cells were predominantly distributed in the substantia nigra (SN) pars compacta (SNc) and in the ventral tegmental area. Interestingly, the expression of FA1/dlk1 significantly increased in tyrosine hydroxylase (TH)-ir cells during early postnatal development. Co-localization and tracing studies demonstrated that FA1/dlk1-ir cells in the SNc were nigrostriatal dopaminergic neurons, and unilateral 6-OHDA lesions resulted in loss of both FA1/dlk1-ir and TH-ir cells in the SNc. Surprisingly, increased numbers of FA1/dlk1-ir cells (by 70%) were detected in dopamine-depleted striata as compared to unlesioned controls. The higher number of FA1/dlk1-ir cells was likely not due to neurogenesis as colocalization studies for proliferation markers were negative. This suggests that FA1/dlk1 was up-regulated in intrinsic cells in response to the 6-OHDA-mediated loss of FA1/dlk1-expressing SNc dopaminergic neurons and/or due to the stab wound. Our findings hint to a significant role of FA1/dlk1 in the SNc during early postnatal development. The differential expression of FA1/dlk1 in the SNc and the striatum of dopamine-depleted rats could indicate a potential involvement of FA1/dlk1 in the cellular response to the degenerative processes.
Collapse
Affiliation(s)
- Rémy Liechti
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster University of Bern, Inselspital, CH-3010 Berne, Switzerland
| | - Angélique D. Ducray
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster University of Bern, Inselspital, CH-3010 Berne, Switzerland
| | - Pia Jensen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Winsløwparken 21, DK-5000 Odense C, Denmark
| | - Stefano Di Santo
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster University of Bern, Inselspital, CH-3010 Berne, Switzerland
| | - Stefanie Seiler
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster University of Bern, Inselspital, CH-3010 Berne, Switzerland
| | - Charlotte H. Jensen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Sdr. Boulevard 29, DK-5000, Odense C, Denmark
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Winsløwparken 21, DK-5000 Odense C, Denmark
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Winsløwparken 21, DK-5000 Odense C, Denmark
| | - Hans Rudolf Widmer
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster University of Bern, Inselspital, CH-3010 Berne, Switzerland
- * E-mail:
| |
Collapse
|
22
|
Simão D, Pinto C, Piersanti S, Weston A, Peddie CJ, Bastos AE, Licursi V, Schwarz SC, Collinson LM, Salinas S, Serra M, Teixeira AP, Saggio I, Lima PA, Kremer EJ, Schiavo G, Brito C, Alves PM. Modeling Human Neural Functionality In Vitro: Three-Dimensional Culture for Dopaminergic Differentiation. Tissue Eng Part A 2015; 21:654-68. [DOI: 10.1089/ten.tea.2014.0079] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Daniel Simão
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Catarina Pinto
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Stefania Piersanti
- Dipartimento di Biologia e Biotecnologie “Charles Darwin,” Università di Roma La Sapienza, Rome, Italy
| | - Anne Weston
- Lincoln's Inn Fields Laboratories, Cancer Research UK London Research Institute, London, United Kingdom
| | - Christopher J. Peddie
- Lincoln's Inn Fields Laboratories, Cancer Research UK London Research Institute, London, United Kingdom
| | - André E.P. Bastos
- NOVA Medical School, Faculdade de Ciências Médicas da Universidade Nova de Lisboa, Lisboa, Portugal
- Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade de Lisboa, Lisboa, Portugal
| | - Valerio Licursi
- Dipartimento di Biologia e Biotecnologie “Charles Darwin,” Università di Roma La Sapienza, Rome, Italy
| | | | - Lucy M. Collinson
- Lincoln's Inn Fields Laboratories, Cancer Research UK London Research Institute, London, United Kingdom
| | - Sara Salinas
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, France
- Université Montpellier I and II, Montpellier, France
| | - Margarida Serra
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ana P. Teixeira
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Isabella Saggio
- Dipartimento di Biologia e Biotecnologie “Charles Darwin,” Università di Roma La Sapienza, Rome, Italy
- Istituto Pasteur Fondazione Cenci Bolognetti, Università di Roma La Sapienza, Rome, Italy
- Istituto di Biologia e Patologia Molecolari del CNR, Università di Roma La Sapienza, Rome, Italy
| | - Pedro A. Lima
- NOVA Medical School, Faculdade de Ciências Médicas da Universidade Nova de Lisboa, Lisboa, Portugal
| | - Eric J. Kremer
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, France
- Université Montpellier I and II, Montpellier, France
| | - Giampietro Schiavo
- Lincoln's Inn Fields Laboratories, Cancer Research UK London Research Institute, London, United Kingdom
- Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, University College London, London, United Kingdom
| | - Catarina Brito
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Paula M. Alves
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
23
|
Vashchinkina E, Panhelainen A, Aitta-Aho T, Korpi ER. GABAA receptor drugs and neuronal plasticity in reward and aversion: focus on the ventral tegmental area. Front Pharmacol 2014; 5:256. [PMID: 25505414 PMCID: PMC4243505 DOI: 10.3389/fphar.2014.00256] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 11/03/2014] [Indexed: 12/13/2022] Open
Abstract
GABAA receptors are the main fast inhibitory neurotransmitter receptors in the mammalian brain, and targets for many clinically important drugs widely used in the treatment of anxiety disorders, insomnia and in anesthesia. Nonetheless, there are significant risks associated with the long-term use of these drugs particularly related to development of tolerance and addiction. Addictive mechanisms of GABAA receptor drugs are poorly known, but recent findings suggest that those drugs may induce aberrant neuroadaptations in the brain reward circuitry. Recently, benzodiazepines, acting on synaptic GABAA receptors, and modulators of extrasynaptic GABAA receptors (THIP and neurosteroids) have been found to induce plasticity in the ventral tegmental area (VTA) dopamine neurons and their main target projections. Furthermore, depending whether synaptic or extrasynaptic GABAA receptor populations are activated, the behavioral outcome of repeated administration seems to correlate with rewarding or aversive behavioral responses, respectively. The VTA dopamine neurons project to forebrain centers such as the nucleus accumbens and medial prefrontal cortex, and receive afferent projections from these brain regions and especially from the extended amygdala and lateral habenula, forming the major part of the reward and aversion circuitry. Both synaptic and extrasynaptic GABAA drugs inhibit the VTA GABAergic interneurons, thus activating the VTA DA neurons by disinhibition and this way inducing glutamatergic synaptic plasticity. However, the GABAA drugs failed to alter synaptic spine numbers as studied from Golgi-Cox-stained VTA dendrites. Since the GABAergic drugs are known to depress the brain metabolism and gene expression, their likely way of inducing neuroplasticity in mature neurons is by disinhibiting the principal neurons, which remains to be rigorously tested for a number of clinically important anxiolytics, sedatives and anesthetics in different parts of the circuitry.
Collapse
Affiliation(s)
- Elena Vashchinkina
- Department of Pharmacology, Institute of Biomedicine, University of Helsinki Helsinki, Finland
| | - Anne Panhelainen
- Institute of Biotechnology, University of Helsinki Helsinki, Finland
| | - Teemu Aitta-Aho
- Department of Pharmacology, Institute of Biomedicine, University of Helsinki Helsinki, Finland ; Department of Pharmacology, University of Cambridge Cambridge, UK
| | - Esa R Korpi
- Department of Pharmacology, Institute of Biomedicine, University of Helsinki Helsinki, Finland ; Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Neurobiology and Ageing Programme, Life Sciences Institute, National University of Singapore, and SINAPSE, Singapore Institute for Neurotechnology Singapore, Singapore
| |
Collapse
|
24
|
Yetnikoff L, Lavezzi HN, Reichard RA, Zahm DS. An update on the connections of the ventral mesencephalic dopaminergic complex. Neuroscience 2014; 282:23-48. [PMID: 24735820 DOI: 10.1016/j.neuroscience.2014.04.010] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 04/03/2014] [Accepted: 04/04/2014] [Indexed: 12/21/2022]
Abstract
This review covers the intrinsic organization and afferent and efferent connections of the midbrain dopaminergic complex, comprising the substantia nigra, ventral tegmental area and retrorubral field, which house, respectively, the A9, A10 and A8 groups of nigrostriatal, mesolimbic and mesocortical dopaminergic neurons. In addition, A10dc (dorsal, caudal) and A10rv (rostroventral) extensions into, respectively, the ventrolateral periaqueductal gray and supramammillary nucleus are discussed. Associated intrinsic and extrinsic connections of the midbrain dopaminergic complex that utilize gamma-aminobutyric acid (GABA), glutamate and neuropeptides and various co-expressed combinations of these compounds are considered in conjunction with the dopamine-containing systems. A framework is provided for understanding the organization of massive afferent systems descending and ascending to the midbrain dopaminergic complex from the telencephalon and brainstem, respectively. Within the context of this framework, the basal ganglia direct and indirect output pathways are treated in some detail. Findings from rodent brain are briefly compared with those from primates, including humans. Recent literature is emphasized, including traditional experimental neuroanatomical and modern gene transfer and optogenetic studies. An attempt was made to provide sufficient background and cite a representative sampling of earlier primary papers and reviews so that people new to the field may find this to be a relatively comprehensive treatment of the subject.
Collapse
Affiliation(s)
- L Yetnikoff
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, 1402 S. Grand Boulevard, Saint Louis, MO 63104, United States.
| | - H N Lavezzi
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, 1402 S. Grand Boulevard, Saint Louis, MO 63104, United States
| | - R A Reichard
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, 1402 S. Grand Boulevard, Saint Louis, MO 63104, United States
| | - D S Zahm
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, 1402 S. Grand Boulevard, Saint Louis, MO 63104, United States.
| |
Collapse
|
25
|
Tønnesen J, Kokaia M. Electrophysiological investigations of synaptic connectivity between host and graft neurons. PROGRESS IN BRAIN RESEARCH 2013. [PMID: 23195416 DOI: 10.1016/b978-0-444-59575-1.00005-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
The functional synaptic integration of grafted stem cell-derived neurons is one of the key aspects of neural cell replacement therapies for neurological disorders such as Parkinson's disease. However, little is currently known about the synaptic connectivity between graft and host cells after transplantation, not only in the settings of clinical trials but also in experimental studies. This knowledge gap is primarily due to the lack of experimental electrophysiological approaches allowing interrogation of synaptic connectivity between prospectively identified host and graft neurons and hampers our understanding of the mechanisms underlying functional integration of stem cell-derived neurons in the host brain, as well as the optimization of protocols for deriving stem cells for neural cell replacement therapy. Recent optogenetic tools allow for direct investigation of connectivity between host and graft neural populations and have already been applied to show bidirectional integration of dopaminergic neurons in a host tissue. These new tools have potential to advance our understanding of functional integration in the near future. Here, we provide an overview of the current literature addressing functional integration of stem cell-derived neurons in the settings of Parkinson's disease models and discuss some experimental paradigms to approach this issue.
Collapse
Affiliation(s)
- Jan Tønnesen
- Synaptic Plasticity and Superresolution Microscopy Group, Interdisciplinary Institute for Neurosciences, Université de Bordeaux Segalen, Bordeaux, France
| | | |
Collapse
|
26
|
Li C, McCall NM, Lopez AJ, Kash TL. Alcohol effects on synaptic transmission in periaqueductal gray dopamine neurons. Alcohol 2013; 47:279-87. [PMID: 23597415 DOI: 10.1016/j.alcohol.2013.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 02/22/2013] [Accepted: 02/22/2013] [Indexed: 12/15/2022]
Abstract
The role of dopamine (DA) signaling in regulating the rewarding properties of drugs, including alcohol, has been widely studied. The majority of these studies, however, have focused on the DA neurons located in the ventral tegmental area (VTA), and their projections to the nucleus accumbens. DA neurons within the ventral periaqueductal gray (vPAG) have been shown to regulate reward but little is known about the functional properties of these neurons, or how they are modified by drugs of abuse. This lack of knowledge is likely due to the highly heterogeneous cell composition of the vPAG, with both γ-aminobutyric acid (GABA) and glutamate neurons present in addition to DA neurons. In this study, we performed whole-cell recordings in a TH-eGFP transgenic mouse line to evaluate the properties of vPAG-DA neurons. Following this initial characterization, we examined how both acute and chronic alcohol exposure modify synaptic transmission onto vPAG-DA neurons. We found minimal effects of acute alcohol exposure on GABA transmission, but a robust enhancement of glutamatergic synaptic transmission in vPAG-DA. Consistent with this effect on excitatory transmission, we also found that alcohol caused an increase in firing rate. These data were in contrast to the effects of chronic intermittent alcohol exposure, which had no significant impact on either inhibitory or excitatory synaptic transmission on the vPAG-DA neurons. These data add to a growing body of literature that points to alcohol having both region-dependent and cell-type dependent effects on function.
Collapse
Affiliation(s)
- Chia Li
- Curriculum in Neurobiology, University of North Carolina-Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | | | | | | |
Collapse
|
27
|
Lammel S, Lim BK, Malenka RC. Reward and aversion in a heterogeneous midbrain dopamine system. Neuropharmacology 2013; 76 Pt B:351-9. [PMID: 23578393 DOI: 10.1016/j.neuropharm.2013.03.019] [Citation(s) in RCA: 522] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 03/11/2013] [Accepted: 03/26/2013] [Indexed: 01/14/2023]
Abstract
The ventral tegmental area (VTA) is a heterogeneous brain structure that serves a central role in motivation and reward processing. Abnormalities in the function of VTA dopamine (DA) neurons and the targets they influence are implicated in several prominent neuropsychiatric disorders including addiction and depression. Recent studies suggest that the midbrain DA system is composed of anatomically and functionally heterogeneous DA subpopulations with different axonal projections. These findings may explain a number of previously confusing observations that suggested a role for DA in processing both rewarding as well as aversive events. Here we will focus on recent advances in understanding the neural circuits mediating reward and aversion in the VTA and how stress as well as drugs of abuse, in particular cocaine, alter circuit function within a heterogeneous midbrain DA system. This article is part of a Special Issue entitled 'NIDA 40th Anniversary Issue'.
Collapse
Affiliation(s)
- Stephan Lammel
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | | | | |
Collapse
|
28
|
Luk KC, Rymar VV, van den Munckhof P, Nicolau S, Steriade C, Bifsha P, Drouin J, Sadikot AF. The transcription factor Pitx3 is expressed selectively in midbrain dopaminergic neurons susceptible to neurodegenerative stress. J Neurochem 2013; 125:932-43. [DOI: 10.1111/jnc.12160] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Revised: 12/17/2012] [Accepted: 01/09/2013] [Indexed: 12/12/2022]
Affiliation(s)
- Kelvin C. Luk
- Department of Neurology and Neurosurgery; Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| | - Vladimir V. Rymar
- Department of Neurology and Neurosurgery; Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| | - Pepijn van den Munckhof
- Unité genetique moleculaire; Institut de recherches cliniques de Montreal; Montreal Quebec Canada
| | - Stefan Nicolau
- Department of Neurology and Neurosurgery; Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| | - Claude Steriade
- Department of Neurology and Neurosurgery; Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| | - Panojot Bifsha
- Unité genetique moleculaire; Institut de recherches cliniques de Montreal; Montreal Quebec Canada
| | - Jacques Drouin
- Unité genetique moleculaire; Institut de recherches cliniques de Montreal; Montreal Quebec Canada
| | - Abbas F. Sadikot
- Department of Neurology and Neurosurgery; Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| |
Collapse
|
29
|
Gambardella C, Pignatelli A, Belluzzi O. The h-current in the substantia Nigra pars compacta neurons: a re-examination. PLoS One 2012; 7:e52329. [PMID: 23284989 PMCID: PMC3528748 DOI: 10.1371/journal.pone.0052329] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 11/12/2012] [Indexed: 01/15/2023] Open
Abstract
The properties of the hyperpolarization-activated cation current (Ih) were investigated in rat substantia nigra - pars compacta (SNc) principal neurons using patch-clamp recordings in thin slices. A reliable identification of single dopaminergic neurons was made possible by the use of a transgenic line of mice expressing eGFP under the tyrosine hydroxylase promoter. The effects of temperature and different protocols on the Ih kinetics showed that, at 37°C and minimizing the disturbance of the intracellular milieu with perforated patch, this current actually activates at potentials more positive than what is generally indicated, with a half-activation potential of −77.05 mV and with a significant level of opening already at rest, thereby substantially contributing to the control of membrane potential, and ultimately playing a relevant function in the regulation of the cell excitability. The implications of the known influence of intracellular cAMP levels on Ih amplitude and kinetics were examined. The direct application of neurotransmitters (DA, 5-HT and noradrenaline) physiologically released onto SNc neurons and known to act on metabotropic receptors coupled to the cAMP pathway modify the Ih amplitude. Here, we show that direct activation of dopaminergic and of 5-HT receptors results in Ih inhibition of SNc DA cells, whereas noradrenaline has the opposite effect. Together, these data suggest that the modulation of Ih by endogenously released neurotransmitters acting on metabotropic receptors –mainly but not exclusively linked to the cAMP pathway- could contribute significantly to the control of SNc neuron excitability.
Collapse
Affiliation(s)
- Cristina Gambardella
- Dipartimento di Scienze della Vita e Biotecnologie, University of Ferrara and Istituto Nazionale di Neuroscienze, Ferrara, Italy
| | - Angela Pignatelli
- Dipartimento di Scienze della Vita e Biotecnologie, University of Ferrara and Istituto Nazionale di Neuroscienze, Ferrara, Italy
| | - Ottorino Belluzzi
- Dipartimento di Scienze della Vita e Biotecnologie, University of Ferrara and Istituto Nazionale di Neuroscienze, Ferrara, Italy
- * E-mail:
| |
Collapse
|
30
|
Vasudevan A, Won C, Li S, Erdélyi F, Szabó G, Kim KS. Dopaminergic neurons modulate GABA neuron migration in the embryonic midbrain. Development 2012; 139:3136-41. [PMID: 22872083 DOI: 10.1242/dev.078394] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neuronal migration, a key event during brain development, remains largely unexplored in the mesencephalon, where dopaminergic (DA) and GABA neurons constitute two major neuronal populations. Here we study the migrational trajectories of DA and GABA neurons and show that they occupy ventral mesencephalic territory in a temporally and spatially specific manner. Our results from the Pitx3-deficient aphakia mouse suggest that pre-existing DA neurons modulate GABA neuronal migration to their final destination, providing novel insights and fresh perspectives concerning neuronal migration and connectivity in the mesencephalon in normal as well as diseased brains.
Collapse
Affiliation(s)
- Anju Vasudevan
- Angiogenesis and Brain Development Laboratory, Division of Basic Neuroscience, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Aggarwal M, Hyland BI, Wickens JR. Neural control of dopamine neurotransmission: implications for reinforcement learning. Eur J Neurosci 2012; 35:1115-23. [DOI: 10.1111/j.1460-9568.2012.08055.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
32
|
Functional alterations to the nigrostriatal system in mice lacking all three members of the synuclein family. J Neurosci 2011; 31:7264-74. [PMID: 21593311 DOI: 10.1523/jneurosci.6194-10.2011] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The synucleins (α, β, and γ) are highly homologous proteins thought to play a role in regulating neurotransmission and are found abundantly in presynaptic terminals. To overcome functional overlap between synuclein proteins and to understand their role in presynaptic signaling from mesostriatal dopaminergic neurons, we produced mice lacking all three members of the synuclein family. The effect on the mesostriatal system was assessed in adult (4- to 14-month-old) animals using a combination of behavioral, biochemical, histological, and electrochemical techniques. Adult triple-synuclein-null (TKO) mice displayed no overt phenotype and no change in the number of midbrain dopaminergic neurons. TKO mice were hyperactive in novel environments and exhibited elevated evoked release of dopamine in the striatum detected with fast-scan cyclic voltammetry. Elevated dopamine release was specific to the dorsal not ventral striatum and was accompanied by a decrease of dopamine tissue content. We confirmed a normal synaptic ultrastructure and a normal abundance of SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) protein complexes in the dorsal striatum. Treatment of TKO animals with drugs affecting dopamine metabolism revealed normal rate of synthesis, enhanced turnover, and reduced presynaptic striatal dopamine stores. Our data uniquely reveal the importance of the synuclein proteins in regulating neurotransmitter release from specific populations of midbrain dopamine neurons through mechanisms that differ from those reported in other neurons. The finding that the complete loss of synucleins leads to changes in dopamine handling by presynaptic terminals specifically in those regions preferentially vulnerable in Parkinson's disease may ultimately inform on the selectivity of the disease process.
Collapse
|
33
|
Chieng B, Azriel Y, Mohammadi S, Christie MJ. Distinct cellular properties of identified dopaminergic and GABAergic neurons in the mouse ventral tegmental area. J Physiol 2011; 589:3775-87. [PMID: 21646409 DOI: 10.1113/jphysiol.2011.210807] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The midbrain ventral tegmental area (VTA) contains neurons largely with either a dopaminergic (DAergic) or GABAergic phenotype. Physiological and pharmacological properties of DAergic neurons have been determined using tyrosine hydroxylase (TH) immunohistochemistry but many properties overlap with non-DAergic neurons presumed to be GABAergic. This study examined properties of GABAergic neurons, non-GABAergic neurons and TH-immunopositive neurons in VTA of GAD67-GFP knock-in mice. Ninety-eight per cent of VTA neurons were either GAD-GFP or TH positive,with the latter being five times more abundant. During cell-attached patch-clamp recordings, GAD-GFP neurons fired brief action potentials that could be completely distinguished from those of non-GFP neurons. Pharmacologically, the μ-opioid agonist DAMGO inhibited firing of action potentials in 92% of GAD-GFP neurons but had no effect in non-GFP neurons. By contrast, dopamine invariably inhibited action potentials in non-GFP neurons but only did so in 8% of GAD-GFP neurons. During whole-cell recordings, the narrower width of action potential in GAD-GFP neurons was also evident but there was considerable overlap with non-GFP neurons. GAD-GFP neurons invariably failed to exhibit the potassium-mediated slow depolarizing potential during injection of positive current that was present in all non-GFP neurons. Under voltage-clamp the cationic current, I(h), was found in both types of neurons with considerable overlap in both amplitude and kinetics. These distinct cellular properties may thus be used to confidently discriminate GABAergic and DAergic neurons in VTA during in vitro electrophysiological recordings.
Collapse
Affiliation(s)
- Billy Chieng
- Brain and Mind Research Institute, University of Sydney, Level 6, Building F, 94 Mallett Street, M02F, Camperdown, NSW 2050, Australia.
| | | | | | | |
Collapse
|
34
|
She H, Yang Q, Shepherd K, Smith Y, Miller G, Testa C, Mao Z. Direct regulation of complex I by mitochondrial MEF2D is disrupted in a mouse model of Parkinson disease and in human patients. J Clin Invest 2011; 121:930-40. [PMID: 21393861 DOI: 10.1172/jci43871] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Accepted: 12/15/2010] [Indexed: 12/21/2022] Open
Abstract
The transcription factors in the myocyte enhancer factor 2 (MEF2) family play important roles in cell survival by regulating nuclear gene expression. Here, we report that MEF2D is present in rodent neuronal mitochondria, where it can regulate the expression of a gene encoded within mitochondrial DNA (mtDNA). Immunocytochemical, immunoelectron microscopic, and biochemical analyses of rodent neuronal cells showed that a portion of MEF2D was targeted to mitochondria via an N-terminal motif and the chaperone protein mitochondrial heat shock protein 70 (mtHsp70). MEF2D bound to a MEF2 consensus site in the region of the mtDNA that contained the gene NADH dehydrogenase 6 (ND6), which encodes an essential component of the complex I enzyme of the oxidative phosphorylation system; MEF2D binding induced ND6 transcription. Blocking MEF2D function specifically in mitochondria decreased complex I activity, increased cellular H(2)O(2) level, reduced ATP production, and sensitized neurons to stress-induced death. Toxins known to affect complex I preferentially disrupted MEF2D function in a mouse model of Parkinson disease (PD). In addition, mitochondrial MEF2D and ND6 levels were decreased in postmortem brain samples of patients with PD compared with age-matched controls. Thus, direct regulation of complex I by mitochondrial MEF2D underlies its neuroprotective effects, and dysregulation of this pathway may contribute to PD.
Collapse
Affiliation(s)
- Hua She
- Department of Pharmacology, Rollins School of Public Health, Atlanta, Georgia, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Aumann TD, Egan K, Lim J, Boon WC, Bye CR, Chua HK, Baban N, Parish CL, Bobrovskaya L, Dickson P, Horne MK. Neuronal activity regulates expression of tyrosine hydroxylase in adult mouse substantia nigra pars compacta neurons. J Neurochem 2011; 116:646-58. [PMID: 21166807 DOI: 10.1111/j.1471-4159.2010.07151.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Striatal delivery of dopamine (DA) by midbrain substantia nigra pars compacta (SNc) neurons is vital for motor control and its depletion causes the motor symptoms of Parkinson's disease. While membrane potential changes or neuronal activity regulates tyrosine hydroxylase (TH, the rate limiting enzyme in catecholamine synthesis) expression in other catecholaminergic cells, it is not known whether the same occurs in adult SNc neurons. We administered drugs known to alter neuronal activity to mouse SNc DAergic neurons in various experimental preparations and measured changes in their TH expression. In cultured midbrain neurons, blockade of action potentials with 1 μM tetrodotoxin decreased TH expression beginning around 20 h later (as measured in real time by green fluorescent protein (GFP) expression driven off TH promoter activity). By contrast, partial blockade of small-conductance, Ca(2+) -activated potassium channels with 300 nM apamin increased TH mRNA and protein between 12 and 24 h later in slices of adult midbrain. Two-week infusions of 300 nM apamin directly to the adult mouse midbrain in vivo also increased TH expression in SNc neurons, measured immunohistochemically. Paradoxically, the number of TH immunoreactive (TH+) SNc neurons decreased in these animals. Similar in vivo infusions of drugs affecting other ion-channels and receptors (L-type voltage-activated Ca(2+) channels, GABA(A) receptors, high K(+) , DA receptors) also increased or decreased cellular TH immunoreactivity but decreased or increased, respectively, the number of TH+ cells in SNc. We conclude that in adult SNc neurons: (i) TH expression is activity-dependent and begins to change ∼20 h following sustained changes in neuronal activity; (ii) ion-channels and receptors mediating cell-autonomous activity or synaptic input are equally potent in altering TH expression; and (iii) activity-dependent changes in TH expression are balanced by opposing changes in the number of TH+ SNc cells.
Collapse
Affiliation(s)
- Tim D Aumann
- Florey Neuroscience Institutes, The University of Melbourne, Parkville, Victoria, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Stahl SM, Sommer B, Allers KA. Multifunctional Pharmacology of Flibanserin: Possible Mechanism of Therapeutic Action in Hypoactive Sexual Desire Disorder. J Sex Med 2011; 8:15-27. [DOI: 10.1111/j.1743-6109.2010.02032.x] [Citation(s) in RCA: 248] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
37
|
Zhang TA, Placzek AN, Dani JA. In vitro identification and electrophysiological characterization of dopamine neurons in the ventral tegmental area. Neuropharmacology 2010; 59:431-6. [PMID: 20600174 DOI: 10.1016/j.neuropharm.2010.06.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 06/08/2010] [Accepted: 06/10/2010] [Indexed: 01/21/2023]
Abstract
Dopamine (DA) neurons in the ventral tegmental area (VTA) have been implicated in brain mechanisms related to motivation, reward, and drug addiction. Successful identification of these neurons in vitro has historically depended upon the expression of a hyperpolarization-activated current (I(h)) and immunohistochemical demonstration of the presence of tyrosine hydroxylase (TH), the rate-limiting enzyme for DA synthesis. Recent findings suggest that electrophysiological criteria may be insufficient for distinguishing DA neurons from non-DA neurons in the VTA. In this study, we sought to determine factors that could potentially account for the apparent discrepancies in the literature regarding DA neuron identification in the rodent brain slice preparation. We found that confirmed DA neurons from the lateral VTA generally displayed a larger amplitude I(h) relative to DA neurons located in the medial VTA. Measurement of a large amplitude I(h) (>100 pA) consistently indicated a dopaminergic phenotype, but non-dopamine neurons also can have I(h) current. The data also showed that immunohistochemical TH labeling of DA neurons can render false negative results after relatively long duration (>15 min) whole-cell patch clamp recordings. We conclude that whole-cell patch clamp recording in combination with immunohistochemical detection of TH expression can guarantee positive but not negative DA identification in the VTA.
Collapse
Affiliation(s)
- Tao A Zhang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030-3498, USA
| | | | | |
Collapse
|
38
|
Pacemaking in dopaminergic ventral tegmental area neurons: depolarizing drive from background and voltage-dependent sodium conductances. J Neurosci 2010; 30:7401-13. [PMID: 20505107 DOI: 10.1523/jneurosci.0143-10.2010] [Citation(s) in RCA: 187] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Dopaminergic neurons in the ventral tegmental area (VTA) fire spontaneously in a pacemaker-like manner. We analyzed the ionic currents that drive pacemaking in dopaminergic VTA neurons, studied in mouse brain slices. Pacemaking was not inhibited by blocking hyperpolarization-activated cation current (I(h)) or blocking all calcium current by Mg(2+) replacement of Ca(2+). Tetrodotoxin (TTX) stopped spontaneous activity and usually resulted in stable resting potentials near -60 mV to -55 mV, 10-15 mV below the action potential threshold. When external sodium was replaced by N-methyl-D-glucamine (NMDG) with TTX present, cells hyperpolarized by an average of -11 mV, suggesting a significant resting sodium conductance not sensitive to TTX. Voltage-clamp experiments using slow (10 mV/s) ramps showed a steady-state, steeply voltage-dependent current blocked by TTX that activates near -60 mV, as well as a sodium "background" current with little voltage sensitivity, revealed by NMDG replacement for sodium with TTX present. We quantified these two components of sodium current during the pacemaking trajectory using action potential clamp. The initial phase of depolarization, up to approximately -55 mV, is driven mainly by non-voltage-dependent sodium background current. Above -55 mV, TTX-sensitive voltage-dependent "persistent" Na current helps drive the final phase of depolarization to the spike threshold. Voltage-dependent calcium current is small at all subthreshold voltages. The pacemaking mechanism in VTA neurons differs from that in substantia nigra pars compacta (SNc) neurons, where subthreshold calcium current plays a dominant role. In addition, we found that non-voltage-dependent background sodium current is much smaller in SNc neurons than VTA neurons.
Collapse
|
39
|
Melis M, Pistis M. Endocannabinoid signaling in midbrain dopamine neurons: more than physiology? Curr Neuropharmacol 2010; 5:268-77. [PMID: 19305743 PMCID: PMC2644494 DOI: 10.2174/157015907782793612] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2006] [Revised: 03/30/2007] [Accepted: 04/03/2007] [Indexed: 11/22/2022] Open
Abstract
Different classes of neurons in the CNS utilize endogenous cannabinoids as retrograde messengers to shape afferent activity in a short- and long-lasting fashion. Transient suppression of excitation and inhibition as well as long-term depression or potentiation in many brain regions require endocannabinoids to be released by the postsynaptic neurons and activate presynaptic CB1 receptors. Memory consolidation and/or extinction and habit forming have been suggested as the potential behavioral consequences of endocannabinoid-mediated synaptic modulation. HOWEVER, ENDOCANNABINOIDS HAVE A DUAL ROLE: beyond a physiological modulation of synaptic functions, they have been demonstrated to participate in the mechanisms of neuronal protection under circumstances involving excessive excitatory drive, glutamate excitotoxicity, hypoxia-ischemia, which are key features of several neurodegenerative disorders. In this framework, the recent discovery that the endocannabinoid 2-arachidonoyl-glycerol is released by midbrain dopaminergic neurons, under both physiological synaptic activity to modulate afferent inputs and pathological conditions such as ischemia, is particularly interesting for the possible implication of these molecules in brain functions and dysfunctions. Since dopamine dysfunctions underlie diverse neuropsychiatric disorders including schizophrenia, psychoses, and drug addiction, the importance of better understanding the correlation between an unbalanced endocannabinoid signal and the dopamine system is even greater. Additionally, we will review the evidence of the involvement of the endocannabinoid system in the pathogenesis of Parkinson's disease, where neuroprotective actions of cannabinoid-acting compounds may prove beneficial.The modulation of the endocannabinoid system by pharmacological agents is a valuable target in protection of dopamine neurons against functional abnormalities as well as against their neurodegeneration.
Collapse
Affiliation(s)
- M Melis
- B.B. Brodie Department of Neuroscience and Center of Excellence for the Neurobiology of Addiction, University of Cagliari, Monserrato, 09042, Italy
| | | |
Collapse
|
40
|
Allers KA, Dremencov E, Ceci A, Flik G, Ferger B, Cremers TIFH, Ittrich C, Sommer B. Acute and repeated flibanserin administration in female rats modulates monoamines differentially across brain areas: a microdialysis study. J Sex Med 2010; 7:1757-67. [PMID: 20163532 DOI: 10.1111/j.1743-6109.2010.01763.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Hypoactive sexual desire disorder (HSDD) is defined as persistent lack of sexual fantasies or desire marked by distress. With a prevalence of 10% it is the most common form of female sexual dysfunction. Recently, the serotonin-1A (5-HT(1A)) receptor agonist and the serotonin-2A (5-HT(2A)) receptor antagonist flibanserin were shown to be safe and efficacious in premenopausal women suffering from HSDD in phase III clinical trials. AIM The current study aims to assess the effect of flibanserin on neurotransmitters serotonin (5-HT), norepinephrine (NE), dopamine (DA), glutamate, and gamma-aminobutyric acid (GABA) in brain areas associated with sexual behavior. METHODS Flibanserin was administered to female Wistar rats (280-350 g). Microdialysis probes were stereotactically inserted into the mPFC, NAC, or MPOA, under isoflurane anesthesia. The extracellular levels of neurotransmitters were assessed in freely moving animals, 24 hours after the surgery. MAIN OUTCOME MEASURES Dialysate levels of DA, NE, and serotonin from medial prefrontal cortex (mPFC), nucleus accumbens (NAC), and hypothalamic medial preoptic area (MPOA) from female rats. RESULTS Acute flibanserin administration decreased 5-HT and increased NE levels in all tested areas. DA was increased in mPFC and MPOA, but not in the NAC. Basal levels of NE in mPFC and NAC and of DA in mPFC were increased upon repeated flibanserin administration, when compared to vehicle-treated animals. The basal levels of 5-HT were not altered by repeated flibanserin administration, but basal DA and NE levels were increased in the mPFC. Glutamate and GABA levels remained unchanged following either repeated or acute flibanserin treatment. CONCLUSIONS Systemic administration of flibanserin to female rats differentially affects the monoamine systems of the brain. This may be the mechanistic underpinning of flibanserin's therapeutic efficacy in HSDD, as sexual behavior is controlled by an intricate interplay between stimulatory (catecholaminergic) and inhibitory (serotonergic) systems.
Collapse
Affiliation(s)
- Kelly A Allers
- Boehringer Ingelheim Pharma GmbH, Biberach an der Riss, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Zellner MR, Ranaldi R. How conditioned stimuli acquire the ability to activate VTA dopamine cells: a proposed neurobiological component of reward-related learning. Neurosci Biobehav Rev 2009; 34:769-80. [PMID: 19914285 DOI: 10.1016/j.neubiorev.2009.11.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Revised: 11/09/2009] [Accepted: 11/10/2009] [Indexed: 02/04/2023]
Abstract
The ability to learn about conditioned stimuli (CS) associated with rewards is a crucial adaptive mechanism. Activity in the mesocorticolimbic dopamine (DA) system, as well as in the ventral tegmental area (VTA), is correlated with responding to and learning about CSs. The mechanism by which VTA neurons become activated by signals associated with conditioned stimuli is not fully understood. Our model suggests that NMDA receptor stimulation in the VTA allows originally weak glutamate signals carrying information about environmental stimuli, coincident with strong excitation correlated with primary rewards, to be strengthened and thereby acquire the ability to activate VTA neurons in themselves, producing approach. Furthermore, once synaptic strengthening occurs, the model suggests that NMDA receptor stimulation in VTA is not necessary for the expression of reward-related learning. In this review we survey evidence that VTA cells respond to cues associated with primary rewards, that this responding is acquired, and that the VTA possesses the attributes to function as a site of integration of signals of primary and conditioned stimuli.
Collapse
Affiliation(s)
- Margaret R Zellner
- Laboratory of Neurobiology & Behavior, The Rockefeller University, New York, NY, United States
| | | |
Collapse
|
42
|
Chuhma N, Choi WY, Mingote S, Rayport S. Dopamine neuron glutamate cotransmission: frequency-dependent modulation in the mesoventromedial projection. Neuroscience 2009; 164:1068-83. [PMID: 19729052 DOI: 10.1016/j.neuroscience.2009.08.057] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 07/22/2009] [Accepted: 08/26/2009] [Indexed: 10/20/2022]
Abstract
Mesoventromedial dopamine neurons projecting from the medial ventral tegmental area to the ventromedial shell of the nucleus accumbens play a role in attributing incentive salience to environmental stimuli that predict important events, and appear to be particularly sensitive to the effects of psychostimulant drugs. Despite the observation that these dopamine neurons make up almost the entire complement of neurons in the projection, stimulating their cell bodies evokes a fast glutamatergic response in accumbens neurons. This is apparently due to dopamine neuron glutamate cotransmission, suggested by the extensive coexpression of vesicular glutamate transporter 2 (VGLUT2) in the neurons. To examine the interplay between the dopamine and glutamate signals, we used acute quasi-horizontal brain slices made from DAT-YFP mice in which the intact mesoventromedial projection can be visualized. Under current clamp, when dopamine neurons were stimulated repeatedly, dopamine neuron glutamate transmission showed dopamine-mediated facilitation, solely at higher, burst-firing frequencies. Facilitation was diminished under voltage clamp and flipped to inhibition by intracellular Cs(+) or GDPbetaS, indicating that it was mediated postsynaptically. Postsynaptic facilitation was D1 mediated, required activation of NMDA receptors and closure of voltage gated K(+)-channels. When postsynaptic facilitation was blocked, D2-mediated presynaptic inhibition became apparent. These counterbalanced pre- and postsynaptic actions determine the frequency dependence of dopamine modulation; at lower firing frequencies dopamine modulation is not apparent, while at burst firing frequency postsynaptic facilitation dominates and dopamine becomes facilitatory. Dopamine neuron glutamate cotransmission may play an important role in encoding the incentive salience value of conditioned stimuli that activate goal-directed behaviors, and may be an important subtract for enduring drug-seeking behaviors.
Collapse
Affiliation(s)
- N Chuhma
- Department of Psychiatry, Columbia University; Department of Molecular Therapeutics, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, USA.
| | | | | | | |
Collapse
|
43
|
NR2A/B-containing NMDA receptors mediate cocaine-induced synaptic plasticity in the VTA and cocaine psychomotor sensitization. Neurosci Lett 2009; 461:159-62. [PMID: 19524640 DOI: 10.1016/j.neulet.2009.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Accepted: 06/02/2009] [Indexed: 01/21/2023]
Abstract
Cocaine-induced modifications of glutamatergic synaptic transmission in the mesolimbic system play a key role in adaptations that promote addictive behaviors. In particular, the activation of ionotropic glutamate N-methyl-D-aspartate receptor (NMDAR) in the ventral tegmental area (VTA) is critical for both cocaine-induced synaptic plasticity induced by a single cocaine injection and for the initiation of cocaine psychomotor sensitization. In this study, we set to determine whether the NR2 subunits of the NMDAR play a specific role in triggering cocaine-induced alterations in synaptic plasticity and the development of psychomotor sensitization. We found that inhibition of NR2A-containing NMDARs by NVP-AAM077, or NR2B-containing receptors by ifenprodil, blocked cocaine-induced increase in the AMPAR/NMDAR currents ratio, a measure of long-term potentiation (LTP) in vivo, in VTA neurons 24h following a single cocaine injection. Furthermore, inhibition of the NR2A subunit during the development of psychomotor sensitization attenuated the enhanced locomotor activity following repeated cocaine injections. Together, these results suggest that NR2-containing NMDA receptors play an important role in the machinery that triggers synaptic and behavioral adaptations to drugs of abuse such as cocaine.
Collapse
|
44
|
Zweifel LS, Argilli E, Bonci A, Palmiter RD. Role of NMDA receptors in dopamine neurons for plasticity and addictive behaviors. Neuron 2008; 59:486-96. [PMID: 18701073 DOI: 10.1016/j.neuron.2008.05.028] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2007] [Revised: 05/19/2008] [Accepted: 05/20/2008] [Indexed: 11/18/2022]
Abstract
A single exposure to drugs of abuse produces an NMDA receptor (NMDAR)-dependent long-term potentiation (LTP) of AMPA receptor (AMPAR) currents in DA neurons; however, the importance of LTP for various aspects of drug addiction is unclear. To test the role of NMDAR-dependent plasticity in addictive behavior, we genetically inactivated functional NMDAR signaling exclusively in DA neurons (KO mice). Inactivation of NMDARs results in increased AMPAR-mediated transmission that is indistinguishable from the increases associated with a single cocaine exposure, yet locomotor responses to multiple drugs of abuse were unaltered in the KO mice. The initial phase of locomotor sensitization to cocaine is intact; however, the delayed sensitization that occurs with prolonged cocaine withdrawal did not occur. Conditioned behavioral responses for cocaine-testing environment were also absent in the KO mice. These findings provide evidence for a role of NMDAR signaling in DA neurons for specific behavioral modifications associated with drug seeking behaviors.
Collapse
Affiliation(s)
- Larry S Zweifel
- Howard Hughes Medical Institute and Department of Biochemistry, University of Washington, Seattle, WA 98195, USA.
| | | | | | | |
Collapse
|
45
|
Liss B, Roeper J. Individual dopamine midbrain neurons: Functional diversity and flexibility in health and disease. ACTA ACUST UNITED AC 2008; 58:314-21. [DOI: 10.1016/j.brainresrev.2007.10.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2007] [Revised: 10/03/2007] [Accepted: 10/15/2007] [Indexed: 11/30/2022]
|
46
|
Dal Bo G, Bérubé-Carrière N, Mendez JA, Leo D, Riad M, Descarries L, Lévesque D, Trudeau LE. Enhanced glutamatergic phenotype of mesencephalic dopamine neurons after neonatal 6-hydroxydopamine lesion. Neuroscience 2008; 156:59-70. [PMID: 18706980 DOI: 10.1016/j.neuroscience.2008.07.032] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Revised: 07/14/2008] [Accepted: 07/15/2008] [Indexed: 10/21/2022]
Abstract
There is increasing evidence that a subset of midbrain dopamine (DA) neurons uses glutamate as a co-transmitter and expresses vesicular glutamate transporter (VGLUT) 2, one of the three vesicular glutamate transporters. In the present study, double in situ hybridization was used to examine tyrosine hydroxylase (TH) and VGLUT2 mRNA expression during the embryonic development of these neurons, and postnatally, in normal rats and rats injected with 6-hydroxydopamine (6-OHDA) at P4 to destroy partially DA neurons. At embryonic days 15 and 16, there was a regional overlap in the labeling of TH and VGLUT2 mRNA in the ventral mesencephalon, which was no longer found at late embryonic stages (E18-E21) and postnatally. In normal pups from P5 to P15, only 1-2% of neurons containing TH mRNA in the ventral tegmental area (VTA) and substantia nigra, pars compacta, also displayed VGLUT2 mRNA. In contrast, after the cerebroventricular administration of 6-OHDA at P4, 26% of surviving DA neurons in the VTA of P15 rats expressed VGLUT2. To search for a colocalization of TH and VGLUT2 protein in axon terminals of these neurons, the nucleus accumbens of normal and 6-OHDA-lesioned P15 rats was examined by electron microscopy after dual immunocytochemical labeling. In normal rats, VGLUT2 protein was found in 28% of TH positive axon terminals in the core of nucleus accumbens. In 6-OHDA-lesioned rats, the total number of TH positive terminals was considerably reduced, and yet the proportion also displaying VGLUT2 immunoreactivity was modestly but significantly increased (37%). These results lead to the suggestion that the glutamatergic phenotype of a VTA DA neurons is highly plastic, repressed toward the end of normal embryonic development, and derepressed postnatally following injury. They also support the hypothesis of co-release of glutamate and DA by mesencephalic neurons in vivo, at least in the developing brain.
Collapse
Affiliation(s)
- G Dal Bo
- Department of Pharmacology, Faculty of Medicine, Université de Montréal, C.P. 6128, Succursale Centre-Ville, Montréal, QC, Canada H3C 3J7
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Nair-Roberts RG, Chatelain-Badie SD, Benson E, White-Cooper H, Bolam JP, Ungless MA. Stereological estimates of dopaminergic, GABAergic and glutamatergic neurons in the ventral tegmental area, substantia nigra and retrorubral field in the rat. Neuroscience 2008; 152:1024-31. [PMID: 18355970 PMCID: PMC2575227 DOI: 10.1016/j.neuroscience.2008.01.046] [Citation(s) in RCA: 421] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2007] [Revised: 12/14/2007] [Accepted: 01/11/2008] [Indexed: 02/06/2023]
Abstract
Midbrain dopamine neurons in the ventral tegmental area, substantia nigra and retrorubral field play key roles in reward processing, learning and memory, and movement. Within these midbrain regions and admixed with the dopamine neurons, are also substantial populations of GABAergic neurons that regulate dopamine neuron activity and have projection targets similar to those of dopamine neurons. Additionally, there is a small group of putative glutamatergic neurons within the ventral tegmental area whose function remains unclear. Although dopamine neurons have been intensively studied and quantified, there is little quantitative information regarding the GABAergic and glutamatergic neurons. We therefore used unbiased stereological methods to estimate the number of dopaminergic, GABAergic and glutamatergic cells in these regions in the rat. Neurons were identified using a combination of immunohistochemistry (tyrosine hydroxylase) and in situ hybridization (glutamic acid decarboxylase mRNA and vesicular glutamate transporter 2 mRNA). In substantia nigra pars compacta 29% of cells were glutamic acid decarboxylase mRNA-positive, 58% in the retrorubral field and 35% in the ventral tegmental area. There were further differences in the relative sizes of the GABAergic populations in subnuclei of the ventral tegmental area. Thus, glutamic acid decarboxylase mRNA-positive neurons represented 12% of cells in the interfascicular nucleus, 30% in the parabrachial nucleus, and 45% in the parainterfascicular nucleus. Vesicular glutamate transporter 2 mRNA-positive neurons were present in the ventral tegmental area, but not substantia nigra or retrorubral field. They were mainly confined to the rostro-medial region of the ventral tegmental area, and represented approximately 2-3% of the total neurons counted ( approximately 1600 cells). These results demonstrate that GABAergic and glutamatergic neurons represent large proportions of the neurons in what are traditionally considered as dopamine nuclei and that there are considerable heterogeneities in the proportions of cell types in the different dopaminergic midbrain regions.
Collapse
Affiliation(s)
- R G Nair-Roberts
- Department of Zoology, University of Oxford, South Parks Rd, Oxford, OX1 3PS, UK; Cardiff School of Biosciences, Cardiff University, Cardiff, CF10 3US, UK
| | | | | | | | | | | |
Collapse
|
48
|
Olson VG, Nestler EJ. Topographical organization of GABAergic neurons within the ventral tegmental area of the rat. Synapse 2007; 61:87-95. [PMID: 17117419 DOI: 10.1002/syn.20345] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The ventral tegmental area (VTA), the origin of dopaminergic cell bodies that comprise the mesocorticolimibic DA system, is widely implicated in drug and natural reward, cognition, and several psychiatric disorders. In addition to dopaminergic neurons, this region is populated by GABAergic neurons, which both regulate the firing of their dopaminergic counterparts and send projections throughout the brain. Although the dopaminergic neurons of the VTA have been extensively characterized neuroanatomically, much less is known about the GABAergic neurons in this region. Recent data suggest that the rostro-caudal topographic organization of these GABAergic neurons may correspond to their ability to regulate drug reward. In the present study, we used immunohistochemical techniques to examine the frequency and topography of GABAergic neurons throughout the rostro-caudal axis of the VTA and the extent to which they coexpress other proteins, including tyrosine hydroxylase (a marker of DA neurons), cholecystokinin, parvalbumin, calretinin, and calbindin d 28k.
Collapse
Affiliation(s)
- Valerie G Olson
- Department of Psychiatry and Center for Basic Neuroscience, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | |
Collapse
|
49
|
Yanagihara S, Hessler NA. Modulation of singing-related activity in the songbird ventral tegmental area by social context. Eur J Neurosci 2006; 24:3619-27. [PMID: 17229110 DOI: 10.1111/j.1460-9568.2006.05228.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Successful reproduction depends critically on social interactions. To understand the neural mechanisms underlying such interactions, the study of courtship singing of songbirds has many advantages. Male zebra finches produce a similar song during courtship of a female and while alone. However, singing-related neural activity in the anterior forebrain pathway (AFP), a basal ganglia-forebrain circuit, is markedly dependent on the social context in which singing occurs. Thus, the AFP should receive a signal of social context from outside the song system. Here, we have begun to investigate the neural source of such a signal by recording from neurons in the ventral tegmental area (VTA), which provides dopaminergic input to Area X, a striatal nucleus of the AFP. The level of activity of most VTA neurons we recorded (32/35) was clearly modulated during singing, especially when males sang to a female bird. Modulation of the level of activity could occur in the presence of a female without singing, but typically was further increased when males sang to the female. In addition, activity of some neurons was patterned in relation to song elements, and appeared related to motor output. These results suggest that VTA activity could carry signals related to motivational aspects of singing, as well as more primary sensory and motor signals.
Collapse
Affiliation(s)
- Shin Yanagihara
- Laboratory for Vocal Behavior Mechanisms, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| | | |
Collapse
|
50
|
Schulz DJ, Baines RA, Hempel CM, Li L, Liss B, Misonou H. Cellular excitability and the regulation of functional neuronal identity: from gene expression to neuromodulation. J Neurosci 2006; 26:10362-7. [PMID: 17035518 PMCID: PMC6674680 DOI: 10.1523/jneurosci.3194-06.2006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- David J Schulz
- Biological Sciences, University of Missouri, Columbia, Missouri 65211, USA.
| | | | | | | | | | | |
Collapse
|