1
|
Szabó D, Franke V, Bianco S, Batiuk MY, Paul EJ, Kukalev A, Pfisterer UG, Irastorza-Azcarate I, Chiariello AM, Demharter S, Zea-Redondo L, Lopez-Atalaya JP, Nicodemi M, Akalin A, Khodosevich K, Ungless MA, Winick-Ng W, Pombo A. A single dose of cocaine rewires the 3D genome structure of midbrain dopamine neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.593308. [PMID: 38766140 PMCID: PMC11100777 DOI: 10.1101/2024.05.10.593308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Midbrain dopamine neurons (DNs) respond to a first exposure to addictive drugs and play key roles in chronic drug usage1-3. As the synaptic and transcriptional changes that follow an acute cocaine exposure are mostly resolved within a few days4,5, the molecular changes that encode the long-term cellular memory of the exposure within DNs remain unknown. To investigate whether a single cocaine exposure induces long-term changes in the 3D genome structure of DNs, we applied Genome Architecture Mapping and single nucleus transcriptomic analyses in the mouse midbrain. We found extensive rewiring of 3D genome architecture at 24 hours past exposure which remains or worsens by 14 days, outlasting transcriptional responses. The cocaine-induced chromatin rewiring occurs at all genomic scales and affects genes with major roles in cocaine-induced synaptic changes. A single cocaine exposure triggers extensive long-lasting changes in chromatin condensation in post-synaptic and post-transcriptional regulatory genes, for example the unfolding of Rbfox1 which becomes most prominent 14 days post exposure. Finally, structurally remodeled genes are most expressed in a specific DN sub-type characterized by low expression of the dopamine auto-receptor Drd2, a key feature of highly cocaine-sensitive cells. These results reveal an important role for long-lasting 3D genome remodelling in the cellular memory of a single cocaine exposure, providing new hypotheses for understanding the inception of drug addiction and 3D genome plasticity.
Collapse
Affiliation(s)
- Dominik Szabó
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, 10115 Berlin, Germany
- Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Vedran Franke
- Bioinformatics & Omics Data Science platform, Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, 10115 Berlin, Germany
| | - Simona Bianco
- Dipartimento di Fisica, Università di Napoli Federico II, and INFN Napoli, Complesso Universitario di Monte Sant’Angelo, 80126 Naples, Italy
| | - Mykhailo Y. Batiuk
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Eleanor J. Paul
- MRC London Institute of Medical Sciences (LMS), London W12 0HS, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Alexander Kukalev
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, 10115 Berlin, Germany
| | - Ulrich G. Pfisterer
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Ibai Irastorza-Azcarate
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, 10115 Berlin, Germany
| | - Andrea M. Chiariello
- Dipartimento di Fisica, Università di Napoli Federico II, and INFN Napoli, Complesso Universitario di Monte Sant’Angelo, 80126 Naples, Italy
| | - Samuel Demharter
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Luna Zea-Redondo
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, 10115 Berlin, Germany
- Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Jose P. Lopez-Atalaya
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), 03550, Sant Joan d’Alacant, Spain
| | - Mario Nicodemi
- Dipartimento di Fisica, Università di Napoli Federico II, and INFN Napoli, Complesso Universitario di Monte Sant’Angelo, 80126 Naples, Italy
- Berlin Institute of Health, 10178 Berlin, Germany
| | - Altuna Akalin
- Bioinformatics & Omics Data Science platform, Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, 10115 Berlin, Germany
| | - Konstantin Khodosevich
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Mark A. Ungless
- MRC London Institute of Medical Sciences (LMS), London W12 0HS, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Warren Winick-Ng
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, 10115 Berlin, Germany
- Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Toronto, Canada
| | - Ana Pombo
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, 10115 Berlin, Germany
- Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| |
Collapse
|
2
|
Mocellin P, Barnstedt O, Luxem K, Kaneko H, Vieweg S, Henschke JU, Dalügge D, Fuhrmann F, Karpova A, Pakan JMP, Kreutz MR, Mikulovic S, Remy S. A septal-ventral tegmental area circuit drives exploratory behavior. Neuron 2024; 112:1020-1032.e7. [PMID: 38266645 DOI: 10.1016/j.neuron.2023.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/10/2023] [Accepted: 12/20/2023] [Indexed: 01/26/2024]
Abstract
To survive, animals need to balance their exploratory drive with their need for safety. Subcortical circuits play an important role in initiating and modulating movement based on external demands and the internal state of the animal; however, how motivation and onset of locomotion are regulated remain largely unresolved. Here, we show that a glutamatergic pathway from the medial septum and diagonal band of Broca (MSDB) to the ventral tegmental area (VTA) controls exploratory locomotor behavior in mice. Using a self-supervised machine learning approach, we found an overrepresentation of exploratory actions, such as sniffing, whisking, and rearing, when this projection is optogenetically activated. Mechanistically, this role relies on glutamatergic MSDB projections that monosynaptically target a subset of both glutamatergic and dopaminergic VTA neurons. Taken together, we identified a glutamatergic basal forebrain to midbrain circuit that initiates locomotor activity and contributes to the expression of exploration-associated behavior.
Collapse
Affiliation(s)
- Petra Mocellin
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; International Max Planck Research School for Brain and Behavior (IMPRS), Bonn 53175, Germany.
| | - Oliver Barnstedt
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Kevin Luxem
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Hiroshi Kaneko
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Silvia Vieweg
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Julia U Henschke
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Dennis Dalügge
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; International Max Planck Research School for Brain and Behavior (IMPRS), Bonn 53175, Germany
| | - Falko Fuhrmann
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Anna Karpova
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg 39106, Germany
| | - Janelle M P Pakan
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg 39106, Germany
| | - Michael R Kreutz
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg 39106, Germany; German Center for Mental Health (DZPG), Magdeburg 39106, Germany
| | - Sanja Mikulovic
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg 39106, Germany; German Center for Mental Health (DZPG), Magdeburg 39106, Germany
| | - Stefan Remy
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg 39106, Germany; German Center for Mental Health (DZPG), Magdeburg 39106, Germany.
| |
Collapse
|
3
|
Frase S, Steddin J, Paschen E, Lenz M, Conforti P, Haas CA, Vlachos A, Schachtrup C, Hosp JA. Dense dopaminergic innervation of the peri-infarct cortex despite dopaminergic cell loss after a pure motor-cortical stroke in rats. J Neurochem 2023; 167:427-440. [PMID: 37735852 DOI: 10.1111/jnc.15970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 08/28/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023]
Abstract
After ischemic stroke, the cortex directly adjacent to the ischemic core (i.e., the peri-infarct cortex, PIC) undergoes plastic changes that facilitate motor recovery. Dopaminergic signaling is thought to support this process. However, ischemic stroke also leads to the remote degeneration of dopaminergic midbrain neurons, possibly interfering with this beneficial effect. In this study, we assessed the reorganization of dopaminergic innervation of the PIC in a rat model of focal cortical stroke. Adult Sprague-Dawley rats either received a photothrombotic stroke (PTS) in the primary motor cortex (M1) or a sham operation. 30 days after PTS or sham procedure, the retrograde tracer Micro Ruby (MR) was injected into the PIC of stroke animals or into homotopic cortical areas of matched sham rats. Thus, dopaminergic midbrain neurons projecting into the PIC were identified based on MR signal and immunoreactivity against tyrosine hydroxylase (TH), a marker for dopaminergic neurons. The density of dopaminergic innervation within the PIC was assessed by quantification of dopaminergic boutons indicated by TH-immunoreactivity. Regarding postsynaptic processes, expression of dopamine receptors (D1- and D2) and a marker of the functional signal cascade (DARPP-32) were visualized histologically. Despite a 25% ipsilesional loss of dopaminergic midbrain neurons after PTS, the number and spatial distribution of dopaminergic neurons projecting to the PIC was not different compared to sham controls. Moreover, the density of dopaminergic innervation in the PIC was significantly higher than in homotopic cortical areas of the sham group. Within the PIC, D1-receptors were expressed in neurons, whereas D2-receptors were confined to astrocytes. The intensity of D1- and DARPP-32 expression appeared to be higher in the PIC compared to the contralesional homotopic cortex. Our data suggest a sprouting of dopaminergic fibers into the PIC and point to a role for dopaminergic signaling in reparative mechanisms post-stroke, potentially related to recovery.
Collapse
Affiliation(s)
- Sibylle Frase
- Department of Neurology and Neuroscience, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julius Steddin
- Department of Neurology and Neuroscience, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Enya Paschen
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Pasquale Conforti
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Carola A Haas
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Schachtrup
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jonas A Hosp
- Department of Neurology and Neuroscience, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
4
|
Ma S, Zhong H, Liu X, Wang L. Spatial Distribution of Neurons Expressing Single, Double, and Triple Molecular Characteristics of Glutamatergic, Dopaminergic, or GABAergic Neurons in the Mouse Ventral Tegmental Area. J Mol Neurosci 2023; 73:345-362. [PMID: 37243808 DOI: 10.1007/s12031-023-02121-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/16/2023] [Indexed: 05/29/2023]
Abstract
The ventral tegmental area (VTA) is a heterogeneous midbrain area that plays a significant role in diverse neural processes such as reward, aversion, and motivation. The VTA contains three main neuronal populations, namely, dopamine (DA), γ-aminobutyric acid (GABA), and glutamate neurons, but some neurons exhibit combinatorial molecular characteristics of dopaminergic, GABAergic, or glutamatergic neurons. However, little information is available regarding detailed distribution of neurons with single, double, and triple molecular characteristics of glutamatergic, dopaminergic, or GABAergic neurons in mice. We present a topographical distribution map of three main neuronal populations expressing a single molecular characteristic of dopaminergic, GABAergic, or glutamatergic neurons, and four neuronal populations co-expressing double or triple molecular characteristics in combinatorial manners, in the mouse VTA, following analysis of triple fluorescent in situ hybridization for the simultaneous detection of tyrosine hydroxylase (TH, marker for dopaminergic neurons), vesicular glutamate transporter 2 (VGLUT2, marker for glutamatergic neurons), and glutamic acid decarboxylase 2 (GAD2, marker for GABAergic neurons) mRNA. We found that the vast majority of neurons expressed a single type of mRNA, and these neurons were intermingled with neurons co-expressing double or triple combinations of VGLUT2, TH, or GAD2 in the VTA. These seven neuronal populations were differentially distributed in the VTA sub-nuclei across the rostro-caudal and latero-medial axes. This histochemical study will lead to a deeper understanding of the complexity of neuronal molecular characteristics in different VTA sub-nuclei, and potentially facilitate clarification of diverse functions of the VTA.
Collapse
Affiliation(s)
- Shaohua Ma
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Hao Zhong
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xuemei Liu
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Liping Wang
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
5
|
Frase S, Löffler F, Hosp JA. Enhancing Post-Stroke Rehabilitation and Preventing Exo-Focal Dopaminergic Degeneration in Rats-A Role for Substance P. Int J Mol Sci 2022; 23:ijms23073848. [PMID: 35409207 PMCID: PMC8999050 DOI: 10.3390/ijms23073848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 12/10/2022] Open
Abstract
Dopaminergic signaling is a prerequisite for motor learning. Delayed degeneration of dopaminergic neurons after stroke is linked to motor learning deficits impairing motor rehabilitation. This study investigates safety and efficacy of substance P (SP) treatment on post-stroke rehabilitation, as this neuropeptide combines neuroprotective and plasticity-promoting properties. Male Sprague Dawley rats received a photothrombotic stroke within the primary motor cortex (M1) after which a previously acquired skilled reaching task was rehabilitated. Rats were treated with intraperitoneal saline (control group, n = 7) or SP-injections (250 µg/kg) 30 min before (SP-pre; n = 7) or 16 h (SP-post; n = 6) after rehabilitation training. Dopaminergic neurodegeneration, microglial activation and substance P-immunoreactivity (IR) were analyzed immunohistochemically. Systemic SP significantly facilitated motor rehabilitation. This effect was more pronounced in SP-pre compared to SP-post animals. SP prevented dopaminergic cell loss after stroke, particularly in the SP-pre condition. Despite its proinflammatory propensity, SP administration did not increase stroke volumes, post-stroke deficits or activation of microglia in the midbrain. Finally, SP administration prevented ipsilesional hypertrophy of striatal SPergic innervation, particularly in the SP-post condition. Mechanistically, SP-pre likely involved plasticity-promoting effects in the early phase of rehabilitation, whereas preservation of dopaminergic signaling may have ameliorated rehabilitative success in both SP groups during later stages of training. Our results demonstrate the facilitating effect of SP treatment on motor rehabilitation after stroke, especially if administered prior to training. SP furthermore prevented delayed dopaminergic degeneration and preserved physiological endogenous SPergic innervation.
Collapse
|
6
|
The Role of NEDD4 E3 Ubiquitin–Protein Ligases in Parkinson’s Disease. Genes (Basel) 2022; 13:genes13030513. [PMID: 35328067 PMCID: PMC8950476 DOI: 10.3390/genes13030513] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/03/2022] [Indexed: 01/25/2023] Open
Abstract
Parkinson’s disease (PD) is a debilitating neurodegenerative disease that causes a great clinical burden. However, its exact molecular pathologies are not fully understood. Whilst there are a number of avenues for research into slowing, halting, or reversing PD, one central idea is to enhance the clearance of the proposed aetiological protein, oligomeric α-synuclein. Oligomeric α-synuclein is the main constituent protein in Lewy bodies and neurites and is considered neurotoxic. Multiple E3 ubiquitin-protein ligases, including the NEDD4 (neural precursor cell expressed developmentally downregulated protein 4) family, parkin, SIAH (mammalian homologues of Drosophila seven in absentia), CHIP (carboxy-terminus of Hsc70 interacting protein), and SCFFXBL5 SCF ubiquitin ligase assembled by the S-phase kinase-associated protein (SKP1), cullin-1 (Cul1), a zinc-binding RING finger protein, and the F-box domain/Leucine-rich repeat protein 5-containing protein FBXL5), have been shown to be able to ubiquitinate α-synuclein, influencing its subsequent degradation via the proteasome or lysosome. Here, we explore the link between NEDD4 ligases and PD, which is not only via α-synuclein but further strengthened by several additional substrates and interaction partners. Some members of the NEDD4 family of ligases are thought to crosstalk even with PD-related genes and proteins found to be mutated in familial forms of PD. Mutations in NEDD4 family genes have not been observed in PD patients, most likely because of their essential survival function during development. Following further in vivo studies, it has been thought that NEDD4 ligases may be viable therapeutic targets in PD. NEDD4 family members could clear toxic proteins, enhancing cell survival and slowing disease progression, or might diminish beneficial proteins, reducing cell survival and accelerating disease progression. Here, we review studies to date on the expression and function of NEDD4 ubiquitin ligases in the brain and their possible impact on PD pathology.
Collapse
|
7
|
Avegno EM, Gilpin NW. Reciprocal midbrain-extended amygdala circuit activity in preclinical models of alcohol use and misuse. Neuropharmacology 2022; 202:108856. [PMID: 34710467 PMCID: PMC8627447 DOI: 10.1016/j.neuropharm.2021.108856] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 01/03/2023]
Abstract
Alcohol dependence is characterized by a shift in motivation to consume alcohol from positive reinforcement (i.e., increased likelihood of future alcohol drinking based on its rewarding effects) to negative reinforcement (i.e., increased likelihood of future alcohol drinking based on alcohol-induced reductions in negative affective symptoms, including but not limited to those experienced during alcohol withdrawal). The neural adaptations that occur during this transition are not entirely understood. Mesolimbic reinforcement circuitry (i.e., ventral tegmental area [VTA] neurons) is activated during early stages of alcohol use, and may be involved in the recruitment of brain stress circuitry (i.e., extended amygdala) during the transition to alcohol dependence, after chronic periods of high-dose alcohol exposure. Here, we review the literature regarding the role of canonical brain reinforcement (VTA) and brain stress (extended amygdala) systems, and the connections between them, in acute, sub-chronic, and chronic alcohol response. Particular emphasis is placed on preclinical models of alcohol use.
Collapse
Affiliation(s)
- Elizabeth M Avegno
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA,Department of Alcohol & Drug Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA,Corresponding author: Correspondence should be addressed to Elizabeth Avegno, 1901 Perdido St, Room 7205, New Orleans, LA 70112,
| | - Nicholas W Gilpin
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA,Department of Alcohol & Drug Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA,Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA,Southeast Louisiana VA Healthcare System (SLVHCS), New Orleans, LA
| |
Collapse
|
8
|
Ndyabawe K, Cipriano M, Zhao W, Haidekker M, Yao K, Mao L, Kisaalita WS. Brain-on-a-Chip Device for Modeling Multiregional Networks. ACS Biomater Sci Eng 2020; 7:350-359. [PMID: 33320530 DOI: 10.1021/acsbiomaterials.0c00895] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Animal models are frequently used in drug discovery because they represent a mammalian in vivo model system, they are the closest approximation to the human brain, and experimentation in humans is not ethical. Working with postmortem human brain samples is challenging and developing human in vitro systems, which mimic the in vivo human brain, has been challenging. However, the use of animal models in drug discovery for human neurological diseases is currently under scrutiny because data from animal models has come with variations due to genetic differences. Evidence from the literature suggests that techniques to reconstruct multiple neurotransmission projections, which characterize neurological disease circuits in humans, in vitro, have not been demonstrated. This paper presents a multicompartment microdevice for patterning neurospheres and specification of neural stem cell fate toward networks of multiple neuronal phenotypes. We validated our design by specification of human neural stem cells to dopaminergic and GABAergic neurons in different compartments of the device, simultaneously. The neurospheres formed unrestricted robust neuronal circuits between arrays of neurospheres in all compartments of the device. Such a device design may provide a basis for formation of multineurotransmission circuits to model functional connectivity between specific human brain regions, in vitro, using human-derived neural stem cells. This work finds relevance in neurological disease modeling and drug screening using human cell-based assays and may provide the impetus for shifting from animal-based models.
Collapse
|
9
|
Protocol for Recording from Ventral Tegmental Area Dopamine Neurons in Mice while Measuring Force during Head-Fixation. STAR Protoc 2020; 1:100091. [PMID: 33111123 PMCID: PMC7580214 DOI: 10.1016/j.xpro.2020.100091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Many studies in systems neuroscience use head-fixation preparations for in vivo experimentation. While head-fixation confers several advantages, one major limitation is the lack of behavioral measures that quantify whole-body movements. Here, we detail a step-by-step protocol for using a novel head-fixation device that measures the forces exerted by head-fixed mice in multiple dimensions. We further detail how this system can be used in conjunction with in vivo electrophysiology and optogenetics to study dopamine neurons in the ventral tegmental area. For complete details on the use and execution of this protocol, please refer to Hughes et al. (2020a, 2020b) Protocol for using a novel head-fixation device to measure head and body forces in mice Protocol allows recording from optogenetically tagged dopamine neurons Device can identify start and end of movements for correlation with neuron activity
Collapse
|
10
|
Namba H, Nawa H. Post-pubertal Difference in Nigral Dopaminergic Cells Firing in the Schizophrenia Model Prepared by Perinatal Challenges of a Cytokine, EGF. Neuroscience 2020; 441:22-32. [PMID: 32531471 DOI: 10.1016/j.neuroscience.2020.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 12/21/2022]
Abstract
Schizophrenia in humans typically develops during and after adolescence; however, the biological underpinning for the specificity of this onset time window remains to be determined. In the present study, we investigated this knowledge gap using our own animal model for schizophrenia. Rodents and monkeys challenged with a cytokine, epidermal growth factor (EGF), as neonates are known to exhibit various behavioral and cognitive abnormalities at the post-pubertal stage. We used the EGF-challenged mice as an animal model for schizophrenia to evaluate the electrophysiological impact of this modeling on nigral dopamine neurons before and after puberty. In vivo single unit recording revealed that the burst firing of putative dopamine neurons in substantia nigra pars compacta was significantly higher in the post-pubertal stage of the EGF model than in that of control mice; in contrast, this difference was not observed in the pre-pubertal stage. The increase in burst firing was accompanied by a decline in Ca2+-activated K+ (ISK) currents, which influence the firing pattern of dopamine neurons. In vivo local application of the SK channel blocker apamin (80 μM) to the substantia nigra was less effective at increasing burst firing in the EGF model than in control mice, suggesting the pathologic role of the ISK decrease in this model. Thus, these results suggest that the aberrant post-pubertal hyperactivity of midbrain dopaminergic neurons is associated with the temporal specificity of the behavioral deficit of this model, and support the hypothesis that this dopaminergic aberration could be implicated in the adolescent onset of schizophrenia.
Collapse
Affiliation(s)
- Hisaaki Namba
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan.
| | - Hiroyuki Nawa
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan.
| |
Collapse
|
11
|
Hosp JA, Greiner KL, Martinez Arellano L, Roth F, Löffler F, Reis J, Fritsch B. Progressive secondary exo-focal dopaminergic neurodegeneration occurs in not directly connected midbrain nuclei after pure motor-cortical stroke. Exp Neurol 2020; 327:113211. [PMID: 31987834 DOI: 10.1016/j.expneurol.2020.113211] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 01/20/2020] [Accepted: 01/24/2020] [Indexed: 10/25/2022]
Abstract
Transsynaptic anterograde and retrograde degeneration of neurons and neural fibers are assumed to trigger local excitotoxicity and inflammatory processes. These processes in turn are thought to drive exo-focal neurodegeneration in remote areas connected to the infarcted tissue after ischemic stroke. In the case of middle cerebral artery occlusion (MCAO), in which striato-nigral connections are affected, the hypothesis of inflammation-induced remote neurodegeneration is based on the temporal dynamics of an early appearance of inflammatory markers in midbrain followed by dopaminergic neuronal loss. To test the hypothesis of a direct transsynaptic mediation of secondary exo-focal post-ischemic neurodegeneration, we used a photochemical induction of a stroke (PTS) in Sprague-Dawley rats restricted to motor cortex (MC), thereby sparing the striatal connections to dopaminergic midbrain nuclei. To dissect the temporal dynamics of post-ischemic neurodegeneration, we analyzed brain sections harvested at day 7 and 14 post stroke. Here, an unexpectedly pronounced and widespread loss of dopaminergic neurons occurred 14 days after stroke also affecting dopaminergic nuclei that are not directly coupled to MC. Since the pattern of neurodegeneration in case of a pure motor stroke is similar to a major stroke including the striatum, it is unlikely that direct synaptic coupling is a prerequisite for delayed secondary exo-focal post ischemic neurodegeneration. Furthermore, dopaminergic neurodegeneration was already detected by Fluoro-Jade C staining at day 7, coinciding with a solely slight inflammatory response. Thus, inflammation cannot be assumed to be the primary driver of exo-focal post-ischemic cell death. Moreover, nigral substance P (SP) expression indicated intact striato-nigral innervation after PTS, whereas opposing effects on SP expression after striatal infarcts argue against a critical role of SP in neurodegenerative or inflammatory processes during exo-focal neurodegeneration.
Collapse
Affiliation(s)
- J A Hosp
- Department of Neurology and Neuroscience, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - K L Greiner
- Department of Neurology and Neuroscience, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - L Martinez Arellano
- Department of Neurology and Neuroscience, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - F Roth
- Department of Neurology and Neuroscience, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - F Löffler
- Department of Neurology and Neuroscience, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - J Reis
- Department of Neurology and Neuroscience, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - B Fritsch
- Department of Neurology and Neuroscience, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
12
|
Zhang J, Lv S, Tang G, Bian G, Yang Y, Li R, Yang J, Liu J. Activation of calcium-impermeable GluR2-containing AMPA receptors in the lateral habenula produces antidepressant-like effects in a rodent model of Parkinson's disease. Exp Neurol 2019; 322:113058. [DOI: 10.1016/j.expneurol.2019.113058] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/05/2019] [Accepted: 09/05/2019] [Indexed: 12/11/2022]
|
13
|
Rassouli H, Khalaj M, Hassani SN, Nemati SH, Hosseini Salekdeh GH, Baharvand H. Gene Expression Patterns of Royan Human Embryonic Stem Cells Correlate with Their Propensity and Culture Systems. CELL JOURNAL 2019; 21:290-299. [PMID: 31210435 PMCID: PMC6582416 DOI: 10.22074/cellj.2019.6128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 11/20/2018] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Human embryonic stem cells (hESCs) have the potential to give rise to all types of cells in the human body when appropriately induced to differentiate. Stem cells can differentiate spontaneously into the three-germ layer derivatives by embryoid bodies (EBs) formation. However, the two-dimensional (2D) adherent culture of hESCs under defined conditions is commonly used for directed differentiation toward a specific type of mature cells. In this study, we aimed to determine the propensity of the Royan hESC lines based on comparison of expression levels of 46 lineage specific markers. MATERIALS AND METHODS In this experimental study, we have compared the expression of lineage-specific markers in hESC lines during EB versus adherent-based spontaneous differentiation. We used quantitative real-time polymerase chain reaction (qRT-PCR) to assess expressions of 46 lineage-specific markers in 4 hESC lines, Royan H1 (RH1), RH2, RH5, and RH6, during spontaneous differentiation in both EB and adherent cultures at 0, 10, and 30 days after initiation of differentiation. RESULTS Based on qRT-PCR data analysis, the liver and neuronal markers had higher expression levels in EBs, whereas skin-specific markers expressed at higher levels in the adherent culture. The results showed differential expression patterns of some lineage-specific markers in EBs compared with the adherent cultures. CONCLUSION According to these results, possibly the spontaneous differentiation technique could be a useful method for optimization of culture conditions to differentiate stem cells into specific cell types such ectoderm, neuron, endoderm and hepatocyte. This approach might prove beneficial for further work on maximizing the efficiency of directed differentiation and development of novel differentiation protocols.
Collapse
Affiliation(s)
- Hassan Rassouli
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mona Khalaj
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Seyedeh-Nafiseh Hassani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - S Hiva Nemati
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - G Hasem Hosseini Salekdeh
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Systems Biology, Agricultural Biotechnology Research Institute of Iran, Agricultural Research, Education, and Extension Organization, Karaj, Iran. Electronic Address:
- Department of Molecular Sciences, Macquarie University, Sydney, NSW, Australia
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. Electronic Address:
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
14
|
Tomagra G, Picollo F, Battiato A, Picconi B, De Marchis S, Pasquarelli A, Olivero P, Marcantoni A, Calabresi P, Carbone E, Carabelli V. Quantal Release of Dopamine and Action Potential Firing Detected in Midbrain Neurons by Multifunctional Diamond-Based Microarrays. Front Neurosci 2019; 13:288. [PMID: 31024230 PMCID: PMC6465646 DOI: 10.3389/fnins.2019.00288] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/11/2019] [Indexed: 12/20/2022] Open
Abstract
Micro-Graphitic Single Crystal Diamond Multi Electrode Arrays (μG-SCD-MEAs) have so far been used as amperometric sensors to detect catecholamines from chromaffin cells and adrenal gland slices. Besides having time resolution and sensitivity that are comparable with carbon fiber electrodes, that represent the gold standard for amperometry, μG-SCD-MEAs also have the advantages of simultaneous multisite detection, high biocompatibility and implementation of amperometric/potentiometric protocols, aimed at monitoring exocytotic events and neuronal excitability. In order to adapt diamond technology to record neuronal activity, the μG-SCD-MEAs in this work have been interfaced with cultured midbrain neurons to detect electrical activity as well as quantal release of dopamine (DA). μG-SCD-MEAs are based on graphitic sensing electrodes that are embedded into the diamond matrix and are fabricated using MeV ion beam lithography. Two geometries have been adopted, with 4 × 4 and 8 × 8 microelectrodes (20 μm × 3.5 μm exposed area, 200 μm spacing). In the amperometric configuration, the 4 × 4 μG-SCD-MEAs resolved quantal exocytosis from midbrain dopaminergic neurons. KCl-stimulated DA release occurred as amperometric spikes of 15 pA amplitude and 0.5 ms half-width, at a mean frequency of 0.4 Hz. When used as potentiometric multiarrays, the 8 × 8 μG-SCD-MEAs detected the spontaneous firing activity of midbrain neurons. Extracellularly recorded action potentials (APs) had mean amplitude of ∼-50 μV and occurred at a mean firing frequency of 0.7 Hz in 67% of neurons, while the remaining fired at 6.8 Hz. Comparable findings were observed using conventional MEAs (0.9 and 6.4 Hz, respectively). To test the reliability of potentiometric recordings with μG-SCD-MEAs, the D2-autoreceptor modulation of firing was investigated by applying levodopa (L-DOPA, 20 μM), and comparing μG-SCD-MEAs, conventional MEAs and current-clamp recordings. In all cases, L-DOPA reduced the spontaneous spiking activity in most neurons by 70%, while the D2-antagonist sulpiride reversed this effect. Cell firing inhibition was generally associated with increased APs amplitude. A minority of neurons was either insensitive to, or potentiated by L-DOPA, suggesting that AP recordings originate from different midbrain neuronal subpopulations and reveal different modulatory pathways. Our data demonstrate, for the first time, that μG-SCD-MEAs are multi-functional biosensors suitable to resolve real-time DA release and AP firing in in vitro neuronal networks.
Collapse
Affiliation(s)
- Giulia Tomagra
- Department of Drug and Science Technology and "NIS" Inter-departmental Centre, University of Torino, Turin, Italy
| | - Federico Picollo
- Department of Physics and "NIS" Inter-departmental Centre, University of Torino, Turin, Italy.,Istituto Nazionale di Fisica Nucleare - Sezione di Torino, Turin, Italy
| | - Alfio Battiato
- Istituto Nazionale di Fisica Nucleare - Sezione di Torino, Turin, Italy
| | - Barbara Picconi
- Experimental Neurophysiology Laboratory, IRCCS San Raffaele Pisana, University San Raffaele, Rome, Italy.,University San Raffaele, Rome, Italy
| | - Silvia De Marchis
- Department of Life Sciences and Systems Biology and "NICO" Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Turin, Italy
| | | | - Paolo Olivero
- Department of Physics and "NIS" Inter-departmental Centre, University of Torino, Turin, Italy.,Istituto Nazionale di Fisica Nucleare - Sezione di Torino, Turin, Italy
| | - Andrea Marcantoni
- Department of Drug and Science Technology and "NIS" Inter-departmental Centre, University of Torino, Turin, Italy
| | - Paolo Calabresi
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy
| | - Emilio Carbone
- Department of Drug and Science Technology and "NIS" Inter-departmental Centre, University of Torino, Turin, Italy
| | - Valentina Carabelli
- Department of Drug and Science Technology and "NIS" Inter-departmental Centre, University of Torino, Turin, Italy
| |
Collapse
|
15
|
Wang HL, Zhang S, Qi J, Wang H, Cachope R, Mejias-Aponte CA, Gomez JA, Mateo-Semidey GE, Beaudoin GMJ, Paladini CA, Cheer JF, Morales M. Dorsal Raphe Dual Serotonin-Glutamate Neurons Drive Reward by Establishing Excitatory Synapses on VTA Mesoaccumbens Dopamine Neurons. Cell Rep 2019; 26:1128-1142.e7. [PMID: 30699344 PMCID: PMC6489450 DOI: 10.1016/j.celrep.2019.01.014] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 10/12/2018] [Accepted: 01/03/2019] [Indexed: 11/26/2022] Open
Abstract
Dorsal raphe (DR) serotonin neurons provide a major input to the ventral tegmental area (VTA). Here, we show that DR serotonin transporter (SERT) neurons establish both asymmetric and symmetric synapses on VTA dopamine neurons, but most of these synapses are asymmetric. Moreover, the DR-SERT terminals making asymmetric synapses on VTA dopamine neurons coexpress vesicular glutamate transporter 3 (VGluT3; transporter for accumulation of glutamate for its synaptic release), suggesting the excitatory nature of these synapses. VTA photoactivation of DR-SERT fibers promotes conditioned place preference, elicits excitatory currents on mesoaccumbens dopamine neurons, increases their firing, and evokes dopamine release in nucleus accumbens. These effects are blocked by VTA inactivation of glutamate and serotonin receptors, supporting the idea of glutamate release in VTA from dual DR SERT-VGluT3 inputs. Our findings suggest a path-specific input from DR serotonergic neurons to VTA that promotes reward by the release of glutamate and activation of mesoaccumbens dopamine neurons.
Collapse
Affiliation(s)
- Hui-Ling Wang
- National Institute on Drug Abuse, Neuronal Networks Section, NIH, Baltimore, MD, USA
| | - Shiliang Zhang
- National Institute on Drug Abuse, Electron Microscopy Core, NIH, Baltimore, MD, USA
| | - Jia Qi
- National Institute on Drug Abuse, Neuronal Networks Section, NIH, Baltimore, MD, USA
| | - Huikun Wang
- National Institute on Drug Abuse, Neuronal Networks Section, NIH, Baltimore, MD, USA
| | - Roger Cachope
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Jorge A Gomez
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | | | - Gerard M J Beaudoin
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | - Carlos A Paladini
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | - Joseph F Cheer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marisela Morales
- National Institute on Drug Abuse, Neuronal Networks Section, NIH, Baltimore, MD, USA.
| |
Collapse
|
16
|
Breton JM, Charbit AR, Snyder BJ, Fong PTK, Dias EV, Himmels P, Lock H, Margolis EB. Relative contributions and mapping of ventral tegmental area dopamine and GABA neurons by projection target in the rat. J Comp Neurol 2018; 527:916-941. [PMID: 30393861 DOI: 10.1002/cne.24572] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/08/2018] [Accepted: 10/21/2018] [Indexed: 12/12/2022]
Abstract
The ventral tegmental area (VTA) is a heterogeneous midbrain structure that contains dopamine (DA), GABA, and glutamate neurons that project to many different brain regions. Here, we combined retrograde tracing with immunocytochemistry against tyrosine hydroxylase (TH) or glutamate decarboxylase (GAD) to systematically compare the proportion of dopaminergic and GABAergic VTA projections to 10 target nuclei: anterior cingulate, prelimbic, and infralimbic cortex; nucleus accumbens core, medial shell, and lateral shell; anterior and posterior basolateral amygdala; ventral pallidum; and periaqueductal gray. Overall, the non-dopaminergic component predominated VTA efferents, accounting for more than 50% of all projecting neurons to each region except the nucleus accumbens core. In addition, GABA neurons contributed no more than 20% to each projection, with the exception of the projection to the ventrolateral periaqueductal gray, where the GABAergic contribution approached 50%. Therefore, there is likely a significant glutamatergic component to many of the VTA's projections. We also found that VTA cell bodies retrogradely labeled from the various target brain regions had distinct distribution patterns within the VTA, including in the locations of DA and GABA neurons. Despite this patterned organization, VTA neurons comprising these different projections were intermingled and never limited to any one subregion. These anatomical results are consistent with the idea that VTA neurons participate in multiple distinct, parallel circuits that differentially contribute to motivation and reward. While attention has largely focused on VTA DA neurons, a better understanding of VTA subpopulations, especially the contribution of non-DA neurons to projections, will be critical for future work.
Collapse
Affiliation(s)
- Jocelyn M Breton
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California
| | - Annabelle R Charbit
- Department of Neurology and Wheeler Center for the Neurobiology of Addiction, University of California, San Francisco, California
| | - Benjamin J Snyder
- Department of Neurology and Wheeler Center for the Neurobiology of Addiction, University of California, San Francisco, California
| | - Peter T K Fong
- Department of Neurology and Wheeler Center for the Neurobiology of Addiction, University of California, San Francisco, California.,Ernest Gallo Clinic and Research Center, University of California, San Francisco, Emeryville, California
| | - Elayne V Dias
- Ernest Gallo Clinic and Research Center, University of California, San Francisco, Emeryville, California
| | - Patricia Himmels
- Ernest Gallo Clinic and Research Center, University of California, San Francisco, Emeryville, California
| | - Hagar Lock
- Ernest Gallo Clinic and Research Center, University of California, San Francisco, Emeryville, California
| | - Elyssa B Margolis
- Department of Neurology and Wheeler Center for the Neurobiology of Addiction, University of California, San Francisco, California.,Ernest Gallo Clinic and Research Center, University of California, San Francisco, Emeryville, California
| |
Collapse
|
17
|
Hasani S, Boroujeni ME, Aliaghaei A, Baghai K, Rostami A. Dopaminergic induction of human adipose-derived mesenchymal stem cells is accompanied by transcriptional activation of autophagy. Cell Biol Int 2018; 42:1688-1694. [DOI: 10.1002/cbin.11056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 09/16/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Sanaz Hasani
- Faculty of Medical Biotechnology; Department of Stem Cells and Regenerative Medicine; National Institute of Genetic Engineering and Biotechnology; Tehran Iran
| | - Mahdi Eskandarian Boroujeni
- Faculty of Medical Biotechnology; Department of Stem Cells and Regenerative Medicine; National Institute of Genetic Engineering and Biotechnology; Tehran Iran
| | - Abbas Aliaghaei
- Cell Biology and Anatomical Sciences; School of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Kaveh Baghai
- Basic and Molecular Epidemiology of Gastrointestinal Disorder Research center; Research institute for Gastroenterology and Liver Diseases; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Amin Rostami
- Gastroenterology and Liver Disease Research Center; Research institute for Gastroenterology and Liver Diseases; Shahid Beheshti University of Medical Sciences; Tehran Iran
| |
Collapse
|
18
|
Heterogeneous dopamine signals support distinct features of motivated actions: implications for learning and addiction. ACTA ACUST UNITED AC 2018; 25:416-424. [PMID: 30115763 PMCID: PMC6097772 DOI: 10.1101/lm.047019.117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 06/15/2018] [Indexed: 01/05/2023]
Abstract
Despite decades of research, investigations into effective neural and pharmacological therapies for many drugs of abuse, such as cocaine, have produced no FDA-approved approaches. This difficulty derives from the complexity of substance use disorders, which encompass a variety of behavioral, psychological, and neural circuit-based changes that occur as a result of repeated experience with the drug. Dopamine signaling has been demonstrated to play a key role in several aspects of drug abuse—from mediating its reinforcing properties and drug-seeking to triggering relapse—while also mediating a number of important aspects of normal (nondrug related) motivated behaviors and actions. Real-time recording methods such as in vivo voltammetry, electrophysiology, and calcium imaging demonstrate that the signaling properties of dopamine for motivationally relevant stimuli are highly dynamic and spatiotemporally circumscribed within afferent target regions. In this review, we identify the origins and functional consequences of heterogeneous dopamine release in the limbic system, and how these properties are persistently altered in the drug-experienced brain. We propose that these spatiotemporally parallel dopaminergic signals are simultaneously available to the animal, but that these circuits are impaired following prolonged drug experience by disrupting the location and content of dopamine signals in afferent target regions. These findings are discussed in the context of relapse and pathways to discovering new treatments for addiction disorders.
Collapse
|
19
|
Zhong P, Vickstrom CR, Liu X, Hu Y, Yu L, Yu HG, Liu QS. HCN2 channels in the ventral tegmental area regulate behavioral responses to chronic stress. eLife 2018; 7:32420. [PMID: 29256865 PMCID: PMC5749952 DOI: 10.7554/elife.32420] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 12/18/2017] [Indexed: 12/17/2022] Open
Abstract
Dopamine neurons in the ventral tegmental area (VTA) are powerful regulators of depression-related behavior. Dopamine neuron activity is altered in chronic stress-based models of depression, but the underlying mechanisms remain incompletely understood. Here, we show that mice subject to chronic mild unpredictable stress (CMS) exhibit anxiety- and depressive-like behavior, which was associated with decreased VTA dopamine neuron firing in vivo and ex vivo. Dopamine neuron firing is governed by voltage-gated ion channels, in particular hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. Following CMS, HCN-mediated currents were decreased in nucleus accumbens-projecting VTA dopamine neurons. Furthermore, shRNA-mediated HCN2 knockdown in the VTA was sufficient to recapitulate CMS-induced depressive- and anxiety-like behavior in stress-naïve mice, whereas VTA HCN2 overexpression largely prevented CMS-induced behavioral deficits. Together, these results reveal a critical role for HCN2 in regulating VTA dopamine neuronal activity and depressive-related behaviors.
Collapse
Affiliation(s)
- Peng Zhong
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| | - Casey R Vickstrom
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| | - Xiaojie Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| | - Ying Hu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| | - Laikang Yu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| | - Han-Gang Yu
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, United States
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| |
Collapse
|
20
|
Vandegrift BJ, You C, Satta R, Brodie MS, Lasek AW. Estradiol increases the sensitivity of ventral tegmental area dopamine neurons to dopamine and ethanol. PLoS One 2017; 12:e0187698. [PMID: 29107956 PMCID: PMC5673180 DOI: 10.1371/journal.pone.0187698] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/24/2017] [Indexed: 12/19/2022] Open
Abstract
Gender differences in psychiatric disorders such as addiction may be modulated by the steroid hormone estrogen. For instance, 17β-estradiol (E2), the predominant form of circulating estrogen in pre-menopausal females, increases ethanol consumption, suggesting that E2 may affect the rewarding properties of ethanol and thus the development of alcohol use disorder in females. The ventral tegmental area (VTA) is critically involved in the rewarding and reinforcing effects of ethanol. In order to determine the role of E2 in VTA physiology, gonadally intact female mice were sacrificed during diestrus II (high E2) or estrus (low E2) for electrophysiology recordings. We measured the excitation by ethanol and inhibition by dopamine (DA) of VTA DA neurons and found that both excitation by ethanol and inhibition by dopamine were greater in diestrus II compared with estrus. Treatment of VTA slices from mice in diestrus II with an estrogen receptor antagonist (ICI 182,780) reduced ethanol-stimulated neuronal firing, but had no effect on ethanol-stimulated firing of neurons in slices from mice in estrus. Surprisingly, ICI 182,780 did not affect the inhibition by DA, indicating different mechanisms of action of estrogen receptors in altering ethanol and DA responses. We also examined the responses of VTA DA neurons to ethanol and DA in ovariectomized mice treated with E2 and found that E2 treatment enhanced the responses to ethanol and DA in a manner similar to what we observed in mice in diestrus II. Our data indicate that E2 modulates VTA neuron physiology, which may contribute to both the enhanced reinforcing and rewarding effects of alcohol and the development of other psychiatric disorders in females that involve alterations in DA neurotransmission.
Collapse
Affiliation(s)
- Bertha J. Vandegrift
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Chang You
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Rosalba Satta
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Mark S. Brodie
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Amy W. Lasek
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
21
|
Mingote S, Chuhma N, Kalmbach A, Thomsen GM, Wang Y, Mihali A, Sferrazza C, Zucker-Scharff I, Siena AC, Welch MG, Lizardi-Ortiz J, Sulzer D, Moore H, Gaisler-Salomon I, Rayport S. Dopamine neuron dependent behaviors mediated by glutamate cotransmission. eLife 2017; 6. [PMID: 28703706 PMCID: PMC5599237 DOI: 10.7554/elife.27566] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/06/2017] [Indexed: 12/11/2022] Open
Abstract
Dopamine neurons in the ventral tegmental area use glutamate as a cotransmitter. To elucidate the behavioral role of the cotransmission, we targeted the glutamate-recycling enzyme glutaminase (gene Gls1). In mice with a dopamine transporter (Slc6a3)-driven conditional heterozygous (cHET) reduction of Gls1 in their dopamine neurons, dopamine neuron survival and transmission were unaffected, while glutamate cotransmission at phasic firing frequencies was reduced, enabling a selective focus on the cotransmission. The mice showed normal emotional and motor behaviors, and an unaffected response to acute amphetamine. Strikingly, amphetamine sensitization was reduced and latent inhibition potentiated. These behavioral effects, also seen in global GLS1 HETs with a schizophrenia resilience phenotype, were not seen in mice with an Emx1-driven forebrain reduction affecting most brain glutamatergic neurons. Thus, a reduction in dopamine neuron glutamate cotransmission appears to mediate significant components of the GLS1 HET schizophrenia resilience phenotype, and glutamate cotransmission appears to be important in attribution of motivational salience.
Collapse
Affiliation(s)
- Susana Mingote
- Department of Psychiatry, Columbia University, New York, United States.,Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, United States
| | - Nao Chuhma
- Department of Psychiatry, Columbia University, New York, United States.,Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, United States
| | - Abigail Kalmbach
- Department of Psychiatry, Columbia University, New York, United States.,Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, United States
| | | | - Yvonne Wang
- Department of Psychiatry, Columbia University, New York, United States
| | - Andra Mihali
- Department of Psychiatry, Columbia University, New York, United States
| | | | | | - Anna-Claire Siena
- Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, United States
| | - Martha G Welch
- Department of Psychiatry, Columbia University, New York, United States.,Department of Pediatrics, Columbia University, New York, United States.,Department of Developmental Neuroscience, NYS Psychiatric Institute, New York, United States
| | | | - David Sulzer
- Department of Psychiatry, Columbia University, New York, United States.,Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, United States.,Department of Neurology, Columbia University, New York, United States.,Department of Pharmacology, Columbia University, New York, United States
| | - Holly Moore
- Department of Psychiatry, Columbia University, New York, United States.,Department of Integrative Neuroscience, NYS Psychiatric Institute, New York, United States
| | - Inna Gaisler-Salomon
- Department of Psychiatry, Columbia University, New York, United States.,Department of Psychology, University of Haifa, Haifa, Israel
| | - Stephen Rayport
- Department of Psychiatry, Columbia University, New York, United States.,Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, United States
| |
Collapse
|
22
|
Carbone C, Costa A, Provensi G, Mannaioni G, Masi A. The Hyperpolarization-Activated Current Determines Synaptic Excitability, Calcium Activity and Specific Viability of Substantia Nigra Dopaminergic Neurons. Front Cell Neurosci 2017; 11:187. [PMID: 28701928 PMCID: PMC5487410 DOI: 10.3389/fncel.2017.00187] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/16/2017] [Indexed: 12/28/2022] Open
Abstract
Differential vulnerability between Substantia Nigra pars compacta (SNpc) and Ventral Tegmental Area (VTA) dopaminergic (DAergic) neurons is a hallmark of Parkinson’s disease (PD). Understanding the molecular bases of this key histopathological aspect would foster the development of much-needed disease-modifying therapies. Non-heterogeneous DAergic degeneration is present in both toxin-based and genetic animal models, suggesting that cellular specificity, rather than causing factors, constitutes the background for differential vulnerability. In this regard, we previously demonstrated that MPP+, a neurotoxin able to cause selective nigrostriatal degeneration in animal rodents and primates, inhibits the Hyperpolarization-activated current (Ih) in SNpc DAergic neurons and that pharmacological Ih antagonism causes potentiation of evoked Excitatory post-synaptic potentials (EPSPs). Of note, the magnitude of such potentiation is greater in the SNpc subfield, consistent with higher Ih density. In the present work, we show that Ih block-induced synaptic potentiation leads to the amplification of somatic calcium responses (SCRs) in vitro. This effect is specific for the SNpc subfield and largely mediated by L-Type calcium channels, as indicated by sensitivity to the CaV 1 blocker isradipine. Furthermore, Ih is downregulated by low intracellular ATP and determines the efficacy of GABAergic inhibition in SNpc DAergic neurons. Finally, we show that stereotaxic administration of Ih blockers causes SNpc-specific neurodegeneration and hemiparkinsonian motor phenotype in rats. During PD progression, Ih downregulation may result from mitochondrial dysfunction and, in concert with PD-related disinhibition of excitatory inputs, determine a SNpc-specific disease pathway.
Collapse
Affiliation(s)
- Carmen Carbone
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of FlorenceFlorence, Italy
| | - Alessia Costa
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of FlorenceFlorence, Italy
| | - Gustavo Provensi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of FlorenceFlorence, Italy
| | - Guido Mannaioni
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of FlorenceFlorence, Italy.,Toxicology Unit, Azienda Ospedaliero-Universitaria CareggiFlorence, Italy
| | - Alessio Masi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of FlorenceFlorence, Italy.,Toxicology Unit, Azienda Ospedaliero-Universitaria CareggiFlorence, Italy
| |
Collapse
|
23
|
Margolis EB, Fujita W, Devi LA, Fields HL. Two delta opioid receptor subtypes are functional in single ventral tegmental area neurons, and can interact with the mu opioid receptor. Neuropharmacology 2017. [PMID: 28645621 DOI: 10.1016/j.neuropharm.2017.06.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The mu and delta opioid receptors (MOR and DOR) are highly homologous members of the opioid family of GPCRs. There is evidence that MOR and DOR interact, however the extent to which these interactions occur in vivo and affect synaptic function is unknown. There are two stable DOR subtypes: DPDPE sensitive (DOR1) and deltorphin II sensitive (DOR2); both agonists are blocked by DOR selective antagonists. Robust motivational effects are produced by local actions of both MOR and DOR ligands in the ventral tegmental area (VTA). Here we demonstrate that a majority of both dopaminergic and non-dopaminergic VTA neurons express combinations of functional DOR1, DOR2, and/or MOR, and that within a single VTA neuron, DOR1, DOR2, and MOR agonists can differentially couple to downstream signaling pathways. As reported for the MOR agonist DAMGO, DPDPE and deltorphin II produced either a predominant K+ dependent hyperpolarization or a Cav2.1 mediated depolarization in different neurons. In some neurons DPDPE and deltorphin II produced opposite responses. Excitation, inhibition, or no effect by DAMGO did not predict the response to DPDPE or deltorphin II, arguing against a MOR-DOR interaction generating DOR subtypes. However, in a subset of VTA neurons the DOR antagonist TIPP-Ψ augmented DAMGO responses; we also observed DPDPE or deltorphin II responses augmented by the MOR selective antagonist CTAP. These findings directly support the existence of two independent, stable forms of the DOR, and show that MOR and DOR can interact in some neurons to alter downstream signaling.
Collapse
Affiliation(s)
- Elyssa B Margolis
- Department of Neurology, The Wheeler Center for the Neurobiology of Addiction, Alcoholism and Addiction Research Group, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Wakako Fujita
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lakshmi A Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Howard L Fields
- Department of Neurology, The Wheeler Center for the Neurobiology of Addiction, Alcoholism and Addiction Research Group, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
24
|
Chuhma N, Mingote S, Kalmbach A, Yetnikoff L, Rayport S. Heterogeneity in Dopamine Neuron Synaptic Actions Across the Striatum and Its Relevance for Schizophrenia. Biol Psychiatry 2017; 81:43-51. [PMID: 27692238 PMCID: PMC5121049 DOI: 10.1016/j.biopsych.2016.07.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 07/03/2016] [Accepted: 07/07/2016] [Indexed: 02/06/2023]
Abstract
Brain imaging has revealed alterations in dopamine uptake, release, and receptor levels in patients with schizophrenia that have been resolved on the scale of striatal subregions. However, the underlying synaptic mechanisms are on a finer scale. Dopamine neuron synaptic actions vary across the striatum, involving variations not only in dopamine release but also in dopamine neuron connectivity, cotransmission, modulation, and activity. Optogenetic studies have revealed that dopamine neurons release dopamine in a synaptic signal mode, and that the neurons also release glutamate and gamma-aminobutyric acid as cotransmitters, with striking regional variation. Fast glutamate and gamma-aminobutyric acid cotransmission convey discrete patterns of dopamine neuron activity to striatal neurons. Glutamate may function not only in a signaling role at a subset of dopamine neuron synapses, but also in mediating vesicular synergy, contributing to regional differences in loading of dopamine into synaptic vesicles. Regional differences in dopamine neuron signaling are likely to be differentially involved in the schizophrenia disease process and likely determine the subregional specificity of the action of psychostimulants that exacerbate the disorder, and antipsychotics that ameliorate the disorder. Elucidating dopamine neuron synaptic signaling offers the potential for achieving greater pharmacological specificity through intersectional pharmacological actions targeting subsets of dopamine neuron synapses.
Collapse
Affiliation(s)
- Nao Chuhma
- Department of Psychiatry, Columbia University, New York, New York; Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, New York
| | - Susana Mingote
- Department of Psychiatry, Columbia University, New York, New York; Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, New York
| | - Abigail Kalmbach
- Department of Psychiatry, Columbia University, New York, New York; Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, New York
| | - Leora Yetnikoff
- Department of Psychiatry, Columbia University, New York, New York; Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, New York
| | - Stephen Rayport
- Department of Psychiatry, Columbia University, New York, New York; Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, New York.
| |
Collapse
|
25
|
Monoaminergic control of brain states and sensory processing: Existing knowledge and recent insights obtained with optogenetics. Prog Neurobiol 2016; 151:237-253. [PMID: 27634227 DOI: 10.1016/j.pneurobio.2016.09.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/18/2016] [Accepted: 09/10/2016] [Indexed: 01/18/2023]
Abstract
Monoamines are key neuromodulators involved in a variety of physiological and pathological brain functions. Classical studies using physiological and pharmacological tools have revealed several essential aspects of monoaminergic involvement in regulating the sleep-wake cycle and influencing sensory responses but many features have remained elusive due to technical limitations. The application of optogenetic tools led to the ability of monitoring and controlling neuronal populations with unprecedented temporal precision and neurochemical specificity. Here, we focus on recent advances in revealing the roles of some monoamines in brain state control and sensory information processing. We summarize the central position of monoamines in integrating sensory processing across sleep-wake states with an emphasis on research conducted using optogenetic techniques. Finally, we discuss the limitations and perspectives of new integrated experimental approaches in understanding the modulatory mechanisms of monoaminergic systems in the mammalian brain.
Collapse
|
26
|
Diversity of Dopaminergic Neural Circuits in Response to Drug Exposure. Neuropsychopharmacology 2016; 41:2424-46. [PMID: 26934955 PMCID: PMC4987841 DOI: 10.1038/npp.2016.32] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 02/05/2016] [Accepted: 02/22/2016] [Indexed: 01/09/2023]
Abstract
Addictive substances are known to increase dopaminergic signaling in the mesocorticolimbic system. The origin of this dopamine (DA) signaling originates in the ventral tegmental area (VTA), which sends afferents to various targets, including the nucleus accumbens, the medial prefrontal cortex, and the basolateral amygdala. VTA DA neurons mediate stimuli saliency and goal-directed behaviors. These neurons undergo robust drug-induced intrinsic and extrinsic synaptic mechanisms following acute and chronic drug exposure, which are part of brain-wide adaptations that ultimately lead to the transition into a drug-dependent state. Interestingly, recent investigations of the differential subpopulations of VTA DA neurons have revealed projection-specific functional roles in mediating reward, aversion, and stress. It is now critical to view drug-induced neuroadaptations from a circuit-level perspective to gain insight into how differential dopaminergic adaptations and signaling to targets of the mesocorticolimbic system mediates drug reward. This review hopes to describe the projection-specific intrinsic characteristics of these subpopulations, the differential afferent inputs onto these VTA DA neuron subpopulations, and consolidate findings of drug-induced plasticity of VTA DA neurons and highlight the importance of future projection-based studies of this system.
Collapse
|
27
|
Dopaminergic Neurons Exhibit an Age-Dependent Decline in Electrophysiological Parameters in the MitoPark Mouse Model of Parkinson's Disease. J Neurosci 2016; 36:4026-37. [PMID: 27053209 DOI: 10.1523/jneurosci.1395-15.2016] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 02/26/2016] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Dopaminergic neurons of the substantia nigra (SN) play a vital role in everyday tasks, such as reward-related behavior and voluntary movement, and excessive loss of these neurons is a primary hallmark of Parkinson's disease (PD). Mitochondrial dysfunction has long been implicated in PD and many animal models induce parkinsonian features by disrupting mitochondrial function. MitoPark mice are a recently developed genetic model of PD that lacks the gene for mitochondrial transcription factor A specifically in dopaminergic neurons. This model mimics many distinct characteristics of PD including progressive and selective loss of SN dopamine neurons, motor deficits that are improved byl-DOPA, and development of inclusion bodies. Here, we used brain slice electrophysiology to construct a timeline of functional decline in SN dopaminergic neurons from MitoPark mice. Dopaminergic neurons from MitoPark mice exhibited decreased cell capacitance and increased input resistance that became more severe with age. Pacemaker firing regularity was disrupted in MitoPark mice and ion channel conductances associated with firing were decreased. Additionally, dopaminergic neurons from MitoPark mice showed a progressive decrease of endogenous dopamine levels, decreased dopamine release, and smaller D2 dopamine receptor-mediated outward currents. Interestingly, expression of ion channel subunits associated with impulse activity (Cav1.2, Cav1.3, HCN1, Nav1.2, and NavB3) was upregulated in older MitoPark mice. The results describe alterations in intrinsic and synaptic properties of dopaminergic neurons in MitoPark mice occurring at ages both before and concurrent with motor impairment. These findings may help inform future investigations into treatment targets for prodromal PD. SIGNIFICANCE STATEMENT Parkinson's disease (PD) is the second most diagnosed neurodegenerative disorder, and the classic motor symptoms of the disease are attributed to selective loss of dopaminergic neurons of the substantia nigra. The MitoPark mouse is a genetic model of PD that mimics many of the key characteristics of the disease and enables the study of progressive neurodegeneration in parkinsonism. Here we have identified functional deficits in the ion channel physiology of dopaminergic neurons from MitoPark mice that both precede and are concurrent with the time course of behavioral symptomatology. Because PD is a progressive disease with a long asymptomatic phase, identification of early functional adaptations could lay the groundwork to test therapeutic interventions that halt or reverse disease progression.
Collapse
|
28
|
Sulzer D, Cragg SJ, Rice ME. Striatal dopamine neurotransmission: regulation of release and uptake. ACTA ACUST UNITED AC 2016; 6:123-148. [PMID: 27141430 DOI: 10.1016/j.baga.2016.02.001] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Dopamine (DA) transmission is governed by processes that regulate release from axonal boutons in the forebrain and the somatodendritic compartment in midbrain, and by clearance by the DA transporter, diffusion, and extracellular metabolism. We review how axonal DA release is regulated by neuronal activity and by autoreceptors and heteroreceptors, and address how quantal release events are regulated in size and frequency. In brain regions densely innervated by DA axons, DA clearance is due predominantly to uptake by the DA transporter, whereas in cortex, midbrain, and other regions with relatively sparse DA inputs, the norepinephrine transporter and diffusion are involved. We discuss the role of DA uptake in restricting the sphere of influence of DA and in temporal accumulation of extracellular DA levels upon successive action potentials. The tonic discharge activity of DA neurons may be translated into a tonic extracellular DA level, whereas their bursting activity can generate discrete extracellular DA transients.
Collapse
Affiliation(s)
- David Sulzer
- Depts of Psychiatry, Neurology, & Pharmacology, NY State Psychiatric Institute, Columbia University, New York, NY, USA
| | - Stephanie J Cragg
- Dept Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Margaret E Rice
- Depts of Neurosurgery & Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
29
|
Tung LW, Lu GL, Lee YH, Yu L, Lee HJ, Leishman E, Bradshaw H, Hwang LL, Hung MS, Mackie K, Zimmer A, Chiou LC. Orexins contribute to restraint stress-induced cocaine relapse by endocannabinoid-mediated disinhibition of dopaminergic neurons. Nat Commun 2016; 7:12199. [PMID: 27448020 PMCID: PMC4961842 DOI: 10.1038/ncomms12199] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 06/10/2016] [Indexed: 12/31/2022] Open
Abstract
Orexins are associated with drug relapse in rodents. Here, we show that acute restraint stress in mice activates lateral hypothalamic (LH) orexin neurons, increases levels of orexin A and 2-arachidonoylglycerol (2-AG) in the ventral tegmental area (VTA), and reinstates extinguished cocaine-conditioned place preference (CPP). This stress-induced reinstatement of cocaine CPP depends on type 1 orexin receptors (OX1Rs), type 1 cannabinoid receptors (CB1Rs) and diacylglycerol lipase (DAGL) in the VTA. In dopaminergic neurons of VTA slices, orexin A presynaptically inhibits GABAergic transmission. This effect is prevented by internal GDP-β-S or inhibiting OX1Rs, CB1Rs, phospholipase C or DAGL, and potentiated by inhibiting 2-AG degradation. These results suggest that restraint stress activates LH orexin neurons, releasing orexins into the VTA to activate postsynaptic OX1Rs of dopaminergic neurons and generate 2-AG through a Gq-protein-phospholipase C-DAGL cascade. 2-AG retrogradely inhibits GABA release through presynaptic CB1Rs, leading to VTA dopaminergic disinhibition and reinstatement of cocaine CPP. Stress is a major cause of relapse to cocaine seeking behaviour. Tung et al. show that orexin mediates stress-induced reinstatement of cocaine seeking behaviour in mice by endocannabinoid-dependent disinhibition in the ventral tegmental area.
Collapse
Affiliation(s)
- Li-Wei Tung
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Guan-Ling Lu
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Yen-Hsien Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250 Wuxing Street, Taipei 11031, Taiwan
| | - Lung Yu
- Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City 70101, Taiwan
| | - Hsin-Jung Lee
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Emma Leishman
- Gill Center and the Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana 47405, USA
| | - Heather Bradshaw
- Gill Center and the Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana 47405, USA
| | - Ling-Ling Hwang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250 Wuxing Street, Taipei 11031, Taiwan.,Department of Physiology, College of Medicine, Taipei Medical University, No. 250 Wuxing Street, Taipei 11031, Taiwan
| | - Ming-Shiu Hung
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| | - Ken Mackie
- Gill Center and the Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana 47405, USA
| | - Andreas Zimmer
- Institute for Molecular Psychiatry, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Lih-Chu Chiou
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei 100, Taiwan.,Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 100, Taiwan.,Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 100, Taiwan.,Research Center for Chinese Medicine &Acupuncture, China Medical University, Taichung 40447, Taiwan
| |
Collapse
|
30
|
Duda J, Pötschke C, Liss B. Converging roles of ion channels, calcium, metabolic stress, and activity pattern of Substantia nigra dopaminergic neurons in health and Parkinson's disease. J Neurochem 2016; 139 Suppl 1:156-178. [PMID: 26865375 PMCID: PMC5095868 DOI: 10.1111/jnc.13572] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 02/03/2016] [Accepted: 02/05/2016] [Indexed: 12/18/2022]
Abstract
Dopamine‐releasing neurons within the Substantia nigra (SN DA) are particularly vulnerable to degeneration compared to other dopaminergic neurons. The age‐dependent, progressive loss of these neurons is a pathological hallmark of Parkinson's disease (PD), as the resulting loss of striatal dopamine causes its major movement‐related symptoms. SN DA neurons release dopamine from their axonal terminals within the dorsal striatum, and also from their cell bodies and dendrites within the midbrain in a calcium‐ and activity‐dependent manner. Their intrinsically generated and metabolically challenging activity is created and modulated by the orchestrated function of different ion channels and dopamine D2‐autoreceptors. Here, we review increasing evidence that the mechanisms that control activity patterns and calcium homeostasis of SN DA neurons are not only crucial for their dopamine release within a physiological range but also modulate their mitochondrial and lysosomal activity, their metabolic stress levels, and their vulnerability to degeneration in PD. Indeed, impaired calcium homeostasis, lysosomal and mitochondrial dysfunction, and metabolic stress in SN DA neurons represent central converging trigger factors for idiopathic and familial PD. We summarize double‐edged roles of ion channels, activity patterns, calcium homeostasis, and related feedback/feed‐forward signaling mechanisms in SN DA neurons for maintaining and modulating their physiological function, but also for contributing to their vulnerability in PD‐paradigms. We focus on the emerging roles of maintained neuronal activity and calcium homeostasis within a physiological bandwidth, and its modulation by PD‐triggers, as well as on bidirectional functions of voltage‐gated L‐type calcium channels and metabolically gated ATP‐sensitive potassium (K‐ATP) channels, and their probable interplay in health and PD.
We propose that SN DA neurons possess several feedback and feed‐forward mechanisms to protect and adapt their activity‐pattern and calcium‐homeostasis within a physiological bandwidth, and that PD‐trigger factors can narrow this bandwidth. We summarize roles of ion channels in this view, and findings documenting that both, reduced as well as elevated activity and associated calcium‐levels can trigger SN DA degeneration.
This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Johanna Duda
- Department of Applied Physiology, Ulm University, Ulm, Germany
| | | | - Birgit Liss
- Department of Applied Physiology, Ulm University, Ulm, Germany.
| |
Collapse
|
31
|
He ZG, Liu BW, Xiang HB. Cross interaction of melanocortinergic and dopaminergic systems in neural modulation. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2015; 7:152-157. [PMID: 26823964 PMCID: PMC4697671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 10/21/2015] [Indexed: 06/05/2023]
Abstract
Melanocortinergic and dopaminergic systems are widely distributed in the CNS and have been established as a crucial regulatory component in diverse physiological functions. The pharmacology of both melanocortinergic and dopaminergic systems including their individual receptors, signaling mechanisms, agonists and antagonists has been extensively studied. Several lines of evidence showed that there existed a cross interaction between the receptors of melanocortinergic and dopaminergic systems. The data available at present had expanded our understanding of melanocortinergic and dopaminergic system interaction in neural modulation, which will be main discussed in this paper.
Collapse
Affiliation(s)
- Zhi-Gang He
- Department of Anesthesiology and Pain Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030, Hubei, PR China
| | - Bao-Wen Liu
- Department of Anesthesiology and Pain Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030, Hubei, PR China
| | - Hong-Bing Xiang
- Department of Anesthesiology and Pain Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030, Hubei, PR China
| |
Collapse
|
32
|
Greenberg GD, Steinman MQ, Doig IE, Hao R, Trainor BC. Effects of social defeat on dopamine neurons in the ventral tegmental area in male and female California mice. Eur J Neurosci 2015; 42:3081-94. [PMID: 26469289 DOI: 10.1111/ejn.13099] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/06/2015] [Accepted: 10/10/2015] [Indexed: 12/11/2022]
Abstract
Dopamine neurons in the ventral tegmental area (VTA) have important functions related to rewards but are also activated in aversive contexts. Electrophysiology studies suggest that the degree to which VTA dopamine neurons respond to noxious stimuli is topographically organized across the dorsal-ventral extent. We used c-fos immunohistochemistry to examine the responses of VTA dopamine neurons in contexts of social defeat and social approach. Studying monogamous California mice (Peromyscus californicus) allowed us to observe the effects of social defeat on both males and females. Females exposed to three episodes of defeat, but not a single episode, had more tyrosine hydroxylase (TH)/c-fos-positive cells in the ventral (but not dorsal) VTA compared with controls. This observation suggests that repeated exposure to aversive contexts is necessary to trigger activation of VTA dopamine neurons. Defeat did not affect TH/c-fos colocalizations in males. We also examined the long-term effects of defeat on c-fos expression in a social interaction test. As previously reported, defeat reduced social interaction in females but not males. Surprisingly, there were no effects of defeat stress on TH/c-fos colocalizations in any subregion of the VTA. However, females had more TH/c-fos-positive cells than males across the entire VTA, and also had greater c-fos-positive cell counts in posterior subregions of the nucleus accumbens shell. Our results show that dopamine neurons in the VTA are more responsive to social contexts in females and that the ventral VTA in particular is sensitive to aversive contexts.
Collapse
Affiliation(s)
- Gian D Greenberg
- Neuroscience Graduate Group, University of California, Davis, CA, USA.,Department of Psychology, University of California, 1 Shields Avenue, Davis, CA, 95616, USA.,Center for Neuroscience, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - Michael Q Steinman
- Department of Psychology, University of California, 1 Shields Avenue, Davis, CA, 95616, USA.,Molecular, Cellular and Integrative Physiology Graduate Group, University of California, Davis, CA, USA
| | - Ian E Doig
- Department of Psychology, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - Rebecca Hao
- Department of Psychology, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - Brian C Trainor
- Neuroscience Graduate Group, University of California, Davis, CA, USA.,Department of Psychology, University of California, 1 Shields Avenue, Davis, CA, 95616, USA.,Center for Neuroscience, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| |
Collapse
|
33
|
Bose P, Rompré PP, Warren RA. Neurotensin enhances glutamatergic EPSCs in VTA neurons by acting on different neurotensin receptors. Peptides 2015; 73:43-50. [PMID: 26296323 DOI: 10.1016/j.peptides.2015.08.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Revised: 08/11/2015] [Accepted: 08/13/2015] [Indexed: 11/26/2022]
Abstract
Neurotensin (NT) is an endogenous neuropeptide that modulates dopamine and glutamate neurotransmission in several limbic regions innervated by neurons located in the ventral tegmental area (VTA). While several studies showed that NT exerted a direct modulation on VTA dopamine neurons less is known about its role in the modulation of glutamatergic neurotransmission in this region. The present study was aimed at characterising the effects of NT on glutamate-mediated responses in different populations of VTA neurons. Using whole cell patch clamp recording technique in horizontal rat brain slices, we measured the amplitude of glutamatergic excitatory post-synaptic currents (EPSCs) evoked by electrical stimulation of VTA afferents before and after application of different concentrations of NT1-13 or its C-terminal fragment, NT8-13. Neurons were classified as either Ih(+) or Ih(-) based on the presence or absence of a hyperpolarisation activated cationic current (Ih). We found that NT1-13 and NT8-13 produced comparable concentration dependent increase in the amplitude of EPSCs in both Ih(+) and Ih(-) neurons. In Ih(+) neurons, the enhancement effect of NT8-13 was blocked by both antagonists, while in Ih(-) neurons it was blocked by the NTS1/NTS2 antagonist, SR142948A, but not the preferred NTS1 antagonist, SR48692. In as much as Ih(-) neurons are non-dopaminergic neurons and Ih(+) neurons represent both dopamine and non-dopamine neurons, we can conclude that NT enhances glutamatergic mediated responses in dopamine, and in a subset of non-dopamine, neurons by acting respectively on NTS1 and an NT receptor other than NTS1.
Collapse
Affiliation(s)
- Poulomee Bose
- Department of Psychiatry, University of Montreal, Quebec, Canada
| | | | - Richard A Warren
- Department of Psychiatry, University of Montreal, Quebec, Canada.
| |
Collapse
|
34
|
Cre-driven optogenetics in the heterogeneous genetic panorama of the VTA. Trends Neurosci 2015; 38:375-86. [PMID: 25962754 DOI: 10.1016/j.tins.2015.04.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 04/08/2015] [Accepted: 04/10/2015] [Indexed: 11/24/2022]
Abstract
The selectivity of optogenetics commonly relies on genetic promoters to manipulate specific populations of neurons through the use of Cre-driver lines. All studies performed in the ventral tegmental area (VTA) so far have utilized promoters present in groups of cells that release dopamine (DA), GABA, or glutamate. However, neurons that co-release neurotransmitters and variabilities within groups of neurons that release the same neurotransmitter present challenges when evaluating the results. Further complexity is introduced by ectopic expression patterns often occurring in transgenic Cre-drivers. New perspectives could be unfolded by identifying and selecting different types of promoter for driving the Cre recombinase. Here, we discuss some promising candidates and highlight the advantages or disadvantages of different methods for creating novel transgenic lines.
Collapse
|
35
|
A concurrent excitation and inhibition of dopaminergic subpopulations in response to nicotine. Sci Rep 2015; 5:8184. [PMID: 25640814 PMCID: PMC4313096 DOI: 10.1038/srep08184] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 12/29/2014] [Indexed: 02/05/2023] Open
Abstract
Midbrain dopamine (DA) neurons are key players in motivation and reward processing. Increased DA release is thought to be central in the initiation of drug addiction. Whereas dopamine neurons are generally considered to be activated by drugs such as nicotine, we report here that nicotine not only induces excitation of ventral tegmental area (VTA) DA cells but also induces inhibition of a subset of VTA DA neurons that are anatomically segregated in the medial part of the VTA. These opposite responses do not correlate with the inhibition and excitation induced by noxious stimuli. We show that this inhibition requires D2 receptor (D2-R) activation, suggesting that a dopaminergic release is involved in the mechanism. Our findings suggest a principle of concurrent excitation and inhibition of VTA DA cells in response to nicotine. It promotes unexplored roles for DA release in addiction contrasting with the classical views of reinforcement and motivation, and give rise to a new interpretation of the mode of operation of the reward system.
Collapse
|
36
|
Hosp JA, Nolan HE, Luft AR. Topography and collateralization of dopaminergic projections to primary motor cortex in rats. Exp Brain Res 2015; 233:1365-75. [PMID: 25633321 DOI: 10.1007/s00221-015-4211-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 01/16/2015] [Indexed: 11/30/2022]
Abstract
Dopaminergic signaling within the primary motor cortex (M1) is necessary for successful motor skill learning. Dopaminergic neurons projecting to M1 are located in the ventral tegmental area (VTA, nucleus A10) of the midbrain. It is unknown which behavioral correlates are encoded by these neurons. The objective here is to investigate whether VTA-M1 fibers are collaterals of projections to prefrontal cortex (PFC) or nucleus accumbens (NAc) or if they form a distinct pathway. In rats, multiple-site retrograde fluorescent tracers were injected into M1, PFC and the core region of the NAc and VTA sections investigated for concomitant labeling of different tracers. Dopaminergic neurons projecting to M1, PFC and NAc were found in nucleus A10 and to a lesser degree in the medial nucleus A9. Neurons show high target specificity, minimal collateral branching to other than their target area and hardly cross the midline. Whereas PFC- and NAc-projecting neurons are indistinguishably intermingled within the ventral portion of dopaminergic nuclei in middle and caudal midbrain, M1-projecting neurons are only located within the dorsal part of the rostral midbrain. Within M1, the forelimb representation receives sevenfold more dopaminergic projections than the hindlimb representation. This strong rostro-caudal gradient as well as the topographical preference to dorsal structures suggest that projections to M1 emerged late in the development of the dopaminergic systems in and form a functionally distinct system.
Collapse
Affiliation(s)
- Jonas A Hosp
- Clinical Neurorehabilitation, Department of Neurology, University of Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland
| | | | | |
Collapse
|
37
|
Abstract
The ventral tegmental area (VTA) is required for the rewarding and motivational actions of opioids and activation of dopamine neurons has been implicated in these effects. The canonical model posits that opioid activation of VTA dopamine neurons is indirect, through inhibition of GABAergic inputs. However, VTA dopamine neurons also express postsynaptic μ-opioid peptide (MOP) receptors. We report here that in Sprague Dawley rat, the MOP receptor-selective agonist DAMGO (0.5-3 μM) depolarized or increased the firing rate of 87 of 451 VTA neurons (including 22 of 110 dopamine neurons). This DAMGO excitation occurs in the presence of GABAA receptor blockade and its EC50 value is two orders of magnitude lower than for presynaptic inhibition of GABA release on to VTA neurons. Consistent with a postsynaptic channel opening, excitations were accompanied by a decrease in input resistance. Excitations were blocked by CdCl2 (100 μM, n = 5) and ω-agatoxin-IVA (100 nM, n = 3), nonselective and Cav2.1 Ca(2+) channel blockers, respectively. DAMGO also produced a postsynaptic inhibition in 233 of 451 VTA neurons, including 45 of 110 dopamine neurons. The mean reversal potential of the inhibitory current was -78 ± 7 mV and inhibitions were blocked by the K(+) channel blocker BaCl2 (100 μM, n = 7). Blockade of either excitation or inhibition unmasked the opposite effect, suggesting that MOP receptors activate concurrent postsynaptic excitatory and inhibitory processes in most VTA neurons. These results provide a novel direct mechanism for MOP receptor control of VTA dopamine neurons.
Collapse
|
38
|
Dopamine midbrain neurons in health and Parkinson’s disease: Emerging roles of voltage-gated calcium channels and ATP-sensitive potassium channels. Neuroscience 2015; 284:798-814. [DOI: 10.1016/j.neuroscience.2014.10.037] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/20/2014] [Accepted: 10/22/2014] [Indexed: 12/14/2022]
|
39
|
Hohmann M, Rumpel R, Fischer M, Donert M, Ratzka A, Klein A, Wesemann M, Effenberg A, Fahlke C, Grothe C. Electrophysiological Characterization of eGFP-Labeled Intrastriatal Dopamine Grafts. Cell Transplant 2014; 24:1451-67. [PMID: 25199117 DOI: 10.3727/096368914x683034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Substitution of degenerated dopaminergic (DA) neurons by intrastriatally transplanted ventral mesencephalon (VM)-derived progenitor cells has been shown to improve motor functions in parkinsonian patients and animal models, whereas investigations of electrophysiological properties of the grafted DA neurons have been rarely performed. Here we show electrophysiological properties of grafted VM progenitor cells at different time intervals up to 12 weeks after transplantation measured in acute brain slices using eGFP-Flag transfection to identify the graft. We were able to classify typical DA neurons according to the biphasic progression (voltage "sag") to hyperpolarizing current injections. Two types of DA-like neurons were classified. Whereas type 1 neurons were characterized by delayed action potentials after hyperpolarization and irregular spontaneous firing, type 2 neurons displayed burst firing after hyperpolarization, spontaneous bursts, and regular firing. Comparison to identified DA neurons in vitro indicates a high integration of the intrastriatally grafted neurons, since in vitro cultures displayed regular firing spontaneously, whereas grafted identified DA neurons showed irregular firing. Additionally, type 1 and type 2 neurons exhibited a slight increase in the spontaneous firing frequency over time intervals after grafting, which might reflect a progressive integration of the grafted DA neurons. Our results provide evidence of the differentiation of grafted VM progenitor cells into mature integrated DA neurons, which are shown to replace the missing DA neurons functionally early after grafting.
Collapse
Affiliation(s)
- Meltem Hohmann
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Marinelli M, McCutcheon JE. Heterogeneity of dopamine neuron activity across traits and states. Neuroscience 2014; 282:176-97. [PMID: 25084048 DOI: 10.1016/j.neuroscience.2014.07.034] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 07/21/2014] [Accepted: 07/22/2014] [Indexed: 12/29/2022]
Abstract
Midbrain dopamine neurons fire irregularly, with interspersed clusters of high-frequency spikes, commonly called 'bursts'. In this review we examine such heterogeneity in activity, and provide insight into how it can participate in psychiatric conditions such as drug addiction. We first describe several techniques used to evaluate dopamine neuron activity, and comment on the different measures that each provides. We next describe the activity of dopamine neurons in 'basal' conditions. Specifically, we discuss how the use of anesthesia and reduced preparations may alter aspects of dopamine cell activity, and how there is heterogeneity across species and regions. We also describe how dopamine cell firing changes throughout the peri-adolescent period and how dopamine neuron activity differs across the population. In the final section, we discuss how dopamine neuron activity changes in response to life events. First, we focus attention on drugs of abuse. Drugs themselves change firing activity through a variety of mechanisms, with effects on firing while drug is present differing from those seen after drug discontinuation. We then review how stimuli that are rewarding, aversive, or salient can evoke changes in firing rate and discharge pattern of dopamine neurons, and provide behavioral relevance of dopamine signaling. Finally, we discuss how stress can modulate dopamine neuron firing and how this may contribute to the role that stressful experiences play in psychiatric disorders such as addiction and depression.
Collapse
Affiliation(s)
- M Marinelli
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, 107 W. Dean Keeton, C0875, BME 6.114A, Austin, TX 78756, USA.
| | - J E McCutcheon
- Department of Cell Physiology and Pharmacology, College of Medicine, Biological Sciences and Psychology, University of Leicester, Maurice Shock Medical Sciences Building, University Road, P.O. Box 138, Leicester LE1 9HN, UK.
| |
Collapse
|
41
|
Acute high fat diet consumption activates the mesolimbic circuit and requires orexin signaling in a mouse model. PLoS One 2014; 9:e87478. [PMID: 24466352 PMCID: PMC3900715 DOI: 10.1371/journal.pone.0087478] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 12/24/2013] [Indexed: 12/03/2022] Open
Abstract
Overconsumption of palatable energy-dense foods has negative health implications and it is associated with obesity and several eating disorders. Currently, little is known about the neuronal circuitries activated by the acute ingestion of a rewarding stimulus. Here, we used a combination of immunohistochemistry, pharmacology and neuronal tracing analyses to examine the role of the mesolimbic system in general, and the orexin neurons in particular, in a simple experimental test in which naïve mice are allowed to spontaneously eat a pellet of a high fat diet (HFD) for 2 h. We found that acute HFD activates c-Fos expression in several reward-related brain areas, including the ventral tegmental area (VTA), nucleus accumbens, central amygdala and lateral hypothalamic area. We also found that: i- HFD-mediated orosensory stimulation was required for the mesolimbic pathway activation, ii- acute HFD differentially activates dopamine neurons of the paranigral, parabrachial pigmented and interfascicular sub-regions of the VTA, and iii- orexin neurons of the lateral hypothalamic area are responsive to acute HFD. Moreover, orexin signaling blockade, with the orexin 1 receptor antagonist SB-334867, reduces acute HFD consumption and c-Fos induction in the VTA but not in the other mesolimbic nuclei under study. Finally, we found that most orexin neurons responsive to acute HFD innervate the VTA. Our results show that acute HFD consumption recruits the mesolimbic system and that the full manifestation of this eating behavior requires the activation of orexin signaling.
Collapse
|
42
|
Stamatakis AM, Jennings JH, Ung RL, Blair GA, Weinberg RJ, Neve RL, Boyce F, Mattis J, Ramakrishnan C, Deisseroth K, Stuber GD. A unique population of ventral tegmental area neurons inhibits the lateral habenula to promote reward. Neuron 2014; 80:1039-53. [PMID: 24267654 DOI: 10.1016/j.neuron.2013.08.023] [Citation(s) in RCA: 259] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2013] [Indexed: 11/28/2022]
Abstract
Lateral habenula (LHb) neurons convey aversive and negative reward conditions through potent indirect inhibition of ventral tegmental area (VTA) dopaminergic neurons. Although VTA dopaminergic neurons reciprocally project to the LHb, the electrophysiological properties and the behavioral consequences associated with selective manipulations of this circuit are unknown. Here, we identify an inhibitory input to the LHb arising from a unique population of VTA neurons expressing dopaminergic markers. Optogenetic activation of this circuit resulted in no detectable dopamine release in LHb brain slices. Instead, stimulation produced GABA-mediated inhibitory synaptic transmission, which suppressed the firing of postsynaptic LHb neurons in brain slices and increased the spontaneous firing rate of VTA dopaminergic neurons in vivo. Furthermore, in vivo activation of this pathway produced reward-related phenotypes that were dependent on intra-LHb GABAA receptor signaling. These results suggest that noncanonical inhibitory signaling by these hybrid dopaminergic-GABAergic neurons act to suppress LHb output under rewarding conditions.
Collapse
Affiliation(s)
- Alice M Stamatakis
- Neurobiology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
New insights into the specificity and plasticity of reward and aversion encoding in the mesolimbic system. J Neurosci 2013; 33:17569-76. [PMID: 24198347 DOI: 10.1523/jneurosci.3250-13.2013] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The mesocorticolimbic system, consisting, at its core, of the ventral tegmental area, the nucleus accumbens, and medial prefrontal cortex, has historically been investigated primarily for its role in positively motivated behaviors and reinforcement learning, and its dysfunction in addiction, schizophrenia, depression, and other mood disorders. Recently, researchers have undertaken a more comprehensive analysis of this system, including its role in not only reward but also punishment, as well as in both positive and negative reinforcement. This focus has been facilitated by new anatomical, physiological, and behavioral approaches to delineate functional circuits underlying behaviors and to determine how this system flexibly encodes and responds to positive and negative states and events, beyond simple associative learning. This review is a summary of topics covered in a mini-symposium at the 2013 Society for Neuroscience annual meeting.
Collapse
|
44
|
Brain neuronal activation induced by flibanserin treatment in female rats. Psychopharmacology (Berl) 2013; 230:639-52. [PMID: 23857113 DOI: 10.1007/s00213-013-3194-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 06/17/2013] [Indexed: 12/19/2022]
Abstract
RATIONALE Flibanserin, a 5-HT1A agonist and 5-HT2A antagonist, is developed for the treatment of hypoactive sexual desire disorder in women, and its efficacy has been evidenced in several clinical studies. Flibanserin prosexual effects have been also evidenced in preclinical animal models. However, the mechanism of action of flibanserin remains not fully understood. OBJECTIVE The aim of the present study was to examine brain neuronal activation in female rats treated with flibanserin, using single immunocytochemical labeling of Fos protein, a marker of neuronal activation, and co-localization of Fos and catecholaminergic marker. METHOD Six groups of female rats received either acute or chronic administrations of vehicle, flibanserin 15 mg/kg or flibanserin 45 mg/kg. The brains were collected and processed for immunocytochemical labeling. RESULTS Acute flibanserin increased levels of Fos immunoreactivity in the nucleus accumbens, arcuate hypothalamic nucleus, locus coeruleus, lateral paragigantocellular nucleus, and nucleus of the solitary tract. Chronic 22-day treatment with flibanserin increased Fos expression in the medial preoptic area and arcuate nucleus of the hypothalamus, ventral tegmental area, locus coeruleus, and lateral paragigantocellular nucleus. Both acute and chronic flibanserin increased the density of activated catecholaminergic neurons in the ventral tegmental area but not in the locus coeruleus. CONCLUSION Altogether, our results showed that flibanserin, at the dose known to enhance female sexual motivation, preferentially activated the brain regions belonging to the mesolimbic dopaminergic pathway and hypothalamic structures involved in the integration of sexual cues related to sexual motivation.
Collapse
|
45
|
Grover ND, Limaye RP, Gokhale DV, Patil TR. Zonisamide: a review of the clinical and experimental evidence for its use in Parkinson's disease. Indian J Pharmacol 2013; 45:547-55. [PMID: 24347760 PMCID: PMC3847242 DOI: 10.4103/0253-7613.121266] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 03/24/2013] [Accepted: 08/14/2013] [Indexed: 12/18/2022] Open
Abstract
The limitations of currently available therapies in addressing the non motor symptoms of Parkinson's disease (PD) have egged on the search for newer options. Zonisamide has been in use for epilepsy and it was serendipitously found to improve the symptoms of PD in a patient who had both epilepsy and PD. Thereafter, various trials were designed to assess the use of zonisamide in PD. The present article investigates the evidence for use of zonisamide in PD from the various clinical trials that were designed to address this issue. Furthermore, the article also summarizes the various mechanisms of its use in PD as described in various animal experiments. A search protocol was designed with predefined inclusion and exclusion criteria. The databases searched were Pubmed, Ovid medline, Cochrane and clinicaltrials.gov. The data thus generated, was fed into a predesigned format. Most of the clinical trials on zonisamide in PD have come from Japan. Most of these trials used the changes in the Unified Parkinson's Disease Rating Scale (UPDRS) score as the endpoints and the most conclusive evidence is for a dose of 25-50 mg, which caused a change in UPDRS part III (motor symptoms). These patients were on levodopa and other drugs used for PD during the trials. One of the clinical trials conducted in Spain investigates the use of zonisamide in impulse control disorders among 15 patients of PD. Among the many mechanisms postulated, a reduction in levodopa induced quinone formation, protection against mitochondrial impairment and an increase in astroglial cysteine transport, an inhibition of microglial activation, monoamine oxidase-B (MAO-B) inhibition, an increased dopamine release and blockade of calcium channels are the most cited. There is evidence for use of zonisamide in PD in addition to levodopa and other therapies for control of motor symptoms. For now, the evidence for its use in control of non motor symptoms in PD is not enough and needs to be investigated further.
Collapse
Affiliation(s)
- Neeta D. Grover
- Department of Pharmacology, Bharati Vidyapeeth Deemed University Medical College, Sangli, Maharashtra, India
| | - Ramachandra P. Limaye
- Department of Pharmacology, Bharati Vidyapeeth Deemed University Medical College, Sangli, Maharashtra, India
| | - Dilip V. Gokhale
- Department of Pharmacology, Bharati Vidyapeeth Deemed University Medical College, Sangli, Maharashtra, India
| | - Tatyasaheb R. Patil
- Department of Pharmacology, Bharati Vidyapeeth Deemed University Medical College, Sangli, Maharashtra, India
| |
Collapse
|
46
|
Santra S, Xu L, Shah M, Johnson M, Dutta A. D-512 and D-440 as novel multifunctional dopamine agonists: characterization of neuroprotection properties and evaluation of in vivo efficacy in a Parkinson's disease animal model. ACS Chem Neurosci 2013; 4:1382-92. [PMID: 23906010 DOI: 10.1021/cn400106n] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In this article, we have demonstrated the in vivo efficacy of D-512 and D-440 in a 6-OHDA-induced unilaterally lesioned rat model experiment, a Parkinson's disease animal model. D-512 is a novel highly potent D2/D3 agonist, and D-440 is a novel highly selective D3 agonist. We evaluated the neuroprotective properties of D-512 and D-440 in the dopaminergic MN9D cells. Cotreatment of these two drugs with 6-OHDA and MPP+ significantly attenuated and reversed 6-OHDA- and MPP+-induced toxicity in a dose-dependent manner in the dopaminergic MN9D cells. The inhibition of caspase 3/7 and lipid peroxidation activities along with the restoration of tyrosine hydroxylase levels by D-512 in 6-OHDA-treated cells may partially explain the mechanism of its neuroprotective property. Furthermore, studies were carried out to elucidate the time-dependent changes in the pERK1/2 and pAkt, two kinases implicated in cell survival and apoptosis, levels upon treatment with 6-OHDA in presence of D-512. The neuroprotective property exhibited by these drugs was independent of their dopamine-agonist activity, which is consistent with our multifunctional drug-development approach that is focused on the generation of disease-modifying symptomatic-treatment agents for Parkinson's disease.
Collapse
Affiliation(s)
- Soumava Santra
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202, United
States
| | - Liping Xu
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202, United
States
| | - Mrudang Shah
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202, United
States
| | - Mark Johnson
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202, United
States
| | - Aloke Dutta
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202, United
States
| |
Collapse
|
47
|
Day JJ, Childs D, Guzman-Karlsson MC, Kibe M, Moulden J, Song E, Tahir A, Sweatt JD. DNA methylation regulates associative reward learning. Nat Neurosci 2013; 16:1445-52. [PMID: 23974711 PMCID: PMC3785567 DOI: 10.1038/nn.3504] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 07/20/2013] [Indexed: 12/13/2022]
Abstract
Reward-related memories are essential for adaptive behavior and evolutionary fitness, but they are also a core component of maladaptive brain diseases such as addiction. Reward learning requires dopamine neurons located in the ventral tegmental area (VTA), which encode relationships between predictive cues and future rewards. Recent evidence suggests that epigenetic mechanisms, including DNA methylation, are essential regulators of neuronal plasticity and experience-driven behavioral change. However, the role of epigenetic mechanisms in reward learning is poorly understood. Here we show that the formation of reward-related associative memories in rats upregulates key plasticity genes in the VTA, which are correlated with memory strength and associated with gene-specific changes in DNA methylation. Moreover, DNA methylation in the VTA is required for the formation of stimulus-reward associations. These results provide the first evidence that that activity-dependent methylation and demethylation of DNA is an essential substrate for the behavioral and neuronal plasticity driven by reward-related experiences.
Collapse
Affiliation(s)
- Jeremy J. Day
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL
| | - Daniel Childs
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL
| | - Mikael C. Guzman-Karlsson
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL
| | - Mercy Kibe
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL
| | - Jerome Moulden
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL
| | - Esther Song
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL
| | - Absar Tahir
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL
| | - J. David Sweatt
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
48
|
Melis M, Scheggi S, Carta G, Madeddu C, Lecca S, Luchicchi A, Cadeddu F, Frau R, Fattore L, Fadda P, Ennas MG, Castelli MP, Fratta W, Schilstrom B, Banni S, De Montis MG, Pistis M. PPARα regulates cholinergic-driven activity of midbrain dopamine neurons via a novel mechanism involving α7 nicotinic acetylcholine receptors. J Neurosci 2013; 33:6203-11. [PMID: 23554501 PMCID: PMC6618938 DOI: 10.1523/jneurosci.4647-12.2013] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 02/15/2013] [Accepted: 02/21/2013] [Indexed: 11/21/2022] Open
Abstract
Ventral tegmental area dopamine neurons control reward-driven learning, and their dysregulation can lead to psychiatric disorders. Tonic and phasic activity of these dopaminergic neurons depends on cholinergic tone and activation of nicotinic acetylcholine receptors (nAChRs), particularly those containing the β2 subunit (β2*-nAChRs). Nuclear peroxisome proliferator-activated receptors type-α (PPARα) tonically regulate β2*-nAChRs and thereby control dopamine neuron firing activity. However, it is unknown how and when PPARα endogenous ligands are synthesized by dopamine cells. Using ex vivo and in vivo electrophysiological techniques combined with biochemical and behavioral analysis, we show that activation of α7-nAChRs increases in the rat VTA both the tyrosine phosphorylation of the β2 subunit of nAChRs and the levels of two PPARα endogenous ligands in a Ca(2+)-dependent manner. Accordingly, in vivo production of endogenous PPARα ligands, triggered by α7-nAChR activation, blocks in rats nicotine-induced increased firing activity of dopamine neurons and displays antidepressant-like properties. These data demonstrate that endogenous PPARα ligands are effectors of α7-nAChRs and that their neuromodulatory properties depend on phosphorylation of β2*-nAChRs on VTA dopamine cells. This reveals an autoinhibitory mechanism aimed at reducing dopamine cell overexcitation engaged during hypercholinergic drive. Our results unveil important physiological functions of nAChR/PPARα signaling in dopamine neurons and how behavioral output can change after modifications of this signaling pathway. Overall, the present study suggests PPARα as new therapeutic targets for disorders associated with unbalanced dopamine-acetylcholine systems.
Collapse
Affiliation(s)
- Miriam Melis
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, 09042 Monserrato, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Inyushin MU, Arencibia-Albite F, de la Cruz A, Vázquez-Torres R, Colon K, Sanabria P, Jiménez-Rivera CA. New method to visualize neurons with DAT in slices of rat VTA using fluorescent substrate for DAT, ASP+. JOURNAL OF NEUROSCIENCE AND NEUROENGINEERING 2013; 2:98-103. [PMID: 24052926 PMCID: PMC3773718 DOI: 10.1166/jnsne.2013.1040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The ventral tegmental area (VTA), and in particular dopamine (DA) neurons in this region of midbrain, has been shown to play an important role in motivation (goal-directed behavior), reward, and drug addiction. Most evidence that implicates VTA DA neurons in these functions are based on widely accepted but indirect electrophysiological characterization, including the hyperpolarization activated non-specific cation current (Ih), spike frequency, and inhibition by D2 receptor agonists. In this study, we used a known neuronal dopamine transporter (DAT) fluorescent substrate [4-(4- (dimethylamino) styryl)-N-methylpyridinium iodide] (ASP+) to visualize DAT-containing cell bodies of DA neurons in VTA region in rat brain slices. Uptake of 100 nM of ASP+ in brain slices of rat VTA region marked 38% of visible neurons, while other neurons from this region and 100% neurons from hippocampus slices were not fluorescent. Using patch-clamp techniques, we have found that pronounced Ih current was present in all fluorescent neurons from VTA area, also spike frequency was similar to the widely accepted values for DA neurons. Furthermore, additional study has shown that there are 84% coincidence of ASP+ fluorescence in neuronal cell bodies and Falck-Hillarp labeling of DA cells. Electrophysiological recordings during ASP+ application have confirmed that low concentrations (100 nM) of ASP+ have no visible effect on neuronal activity during 1-2 hours after staining. Thus, uptake of fluorescent monoamine analog ASP+ by DAT can be an additional criterion for identification of DAT-containing neurons in slices.
Collapse
Affiliation(s)
- Mikhail U. Inyushin
- Department of Physiology, Universidad Central del Caribe, Medical School P.O.Box 60-327, Bayamón, Puerto Rico
| | - Francisco Arencibia-Albite
- Department of Physiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico, 00936-5067
| | - Angel de la Cruz
- Department of Physiology, Universidad Central del Caribe, Medical School P.O.Box 60-327, Bayamón, Puerto Rico
| | - Rafael Vázquez-Torres
- Department of Physiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico, 00936-5067
| | - Katiria Colon
- Department of Physiology, Universidad Central del Caribe, Medical School P.O.Box 60-327, Bayamón, Puerto Rico
| | - Priscila Sanabria
- Department of Physiology, Universidad Central del Caribe, Medical School P.O.Box 60-327, Bayamón, Puerto Rico
| | - Carlos A. Jiménez-Rivera
- Department of Physiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico, 00936-5067
| |
Collapse
|
50
|
Saddoris MP, Sugam JA, Cacciapaglia F, Carelli RM. Rapid dopamine dynamics in the accumbens core and shell: learning and action. Front Biosci (Elite Ed) 2013; 5:273-288. [PMID: 23276989 PMCID: PMC3897221 DOI: 10.2741/e615] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The catecholamine dopamine (DA) has been implicated in a host of neural processes as diverse as schizophrenia, parkinsonism and reward encoding. Importantly, these distinct features of DA function are due in large part to separate neural circuits involving connections arising from different DA-releasing nuclei and projections to separate afferent targets. Emerging data has suggested that this same principle of separate neural circuits may be applicable within structural subregions, such as the core and shell of the nucleus accumbens (NAc). Further, DA may act selectively on smaller ensembles of cells (or, microcircuits) via differential DA receptor density and distinct inputs and outputs of the microcircuits, thus enabling new learning about Pavlovian cues, instrumental responses, subjective reward processing and decision-making. In this review, by taking advantage of studies using subsecond voltammetric techniques in behaving animals to study how rapid changes in DA levels affect behavior, we examine the spatial and temporal features of DA release and how it relates to both normal learning and similarities to pathological learning in the form of addiction.
Collapse
Affiliation(s)
- Michael P Saddoris
- Department of Psychology, University of North Carolina, Chapel Hill, NC 27599-3270, USA.
| | | | | | | |
Collapse
|