51
|
Anastasiades PG, Boada C, Carter AG. Cell-Type-Specific D1 Dopamine Receptor Modulation of Projection Neurons and Interneurons in the Prefrontal Cortex. Cereb Cortex 2020; 29:3224-3242. [PMID: 30566584 DOI: 10.1093/cercor/bhy299] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/01/2018] [Accepted: 11/07/2018] [Indexed: 11/14/2022] Open
Abstract
Dopamine modulation in the prefrontal cortex (PFC) mediates diverse effects on neuronal physiology and function, but the expression of dopamine receptors at subpopulations of projection neurons and interneurons remains unresolved. Here, we examine D1 receptor expression and modulation at specific cell types and layers in the mouse prelimbic PFC. We first show that D1 receptors are enriched in pyramidal cells in both layers 5 and 6, and that these cells project to intratelencephalic targets including contralateral cortex, striatum, and claustrum rather than to extratelencephalic structures. We then find that D1 receptors are also present in interneurons and enriched in superficial layer VIP-positive (VIP+) interneurons that coexpresses calretinin but absent from parvalbumin-positive (PV+) and somatostatin-positive (SOM+) interneurons. Finally, we determine that D1 receptors strongly and selectively enhance action potential firing in only a subset of these corticocortical neurons and VIP+ interneurons. Our findings define several novel subpopulations of D1+ neurons, highlighting how modulation via D1 receptors can influence both excitatory and disinhibitory microcircuits in the PFC.
Collapse
Affiliation(s)
- Paul G Anastasiades
- Center for Neural Science, New York University, 4 Washington Place, New York, NY, USA
| | - Christina Boada
- Center for Neural Science, New York University, 4 Washington Place, New York, NY, USA
| | - Adam G Carter
- Center for Neural Science, New York University, 4 Washington Place, New York, NY, USA
| |
Collapse
|
52
|
Kumar SP, Babu PP. Aberrant Dopamine Receptor Signaling Plays Critical Role in the Impairment of Striatal Neurons in Experimental Cerebral Malaria. Mol Neurobiol 2020; 57:5069-5083. [PMID: 32833186 DOI: 10.1007/s12035-020-02076-0] [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: 03/02/2020] [Accepted: 08/14/2020] [Indexed: 01/19/2023]
Abstract
One-fourth survivors of cerebral malaria (CM) retain long-term cognitive and behavioral deficits. Structural abnormalities in striatum are reported in 80% of children with CM. Dopamine receptors (D1 and D2) are widely expressed in striatal medium spiny neurons (MSNs) that regulate critical physiological functions related to behavior and cognition. Dysregulation of dopamine receptors alters the expression of downstream proteins such as dopamine- and cAMP-regulated phosphoprotein (DARPP), Ca2+/calmodulin-dependent protein kinase II alpha (CaMKIIα), and p25/cyclin-dependent kinase 5 (cdk5). However, the role of dopamine receptor signaling dysfunction on the outcome of striatal neuron degeneration is unknown underlying the pathophysiology of CM. Using experimental CM (ECM), the present study attempted to understand the role of aberrant dopamine receptor signaling and its possible relation in causing MSNs morphological impairment. The effect of antimalarial drug artemether (ARM) rescue therapy was also assessed after ECM on the outcome of dopamine receptors downstream signaling. ECM was induced in C57BL/6 mice (male and female) infecting with Plasmodium berghei ANKA (PbA) parasite that reiterates the clinical setting of CM. We demonstrated that ECM caused a significant increase in the expression of D1, D2 receptors, phosphorylated DARPP, p25, cdk5, CaMKIIα, and D1-D2 heteromers. A substantial increase in neuronal damage observed in the dorsolateral striatum region of ECM brains (particularly in MSNs) as revealed by increased Fluoro-Jade C staining, reduced dendritic spine density, and impaired dendritic arborization with varicosities. While the ARM rescue therapy significantly altered the effects of ECM induced dopamine receptor signaling dysfunction and neurodegeneration. Overall, our data suggest that dysregulation of dopamine receptor signaling plays an important role in the degeneration of MSNs, and the ARM rescue therapy might provide better insights to develop effective therapeutic strategies for CM.
Collapse
Affiliation(s)
- Simhadri Praveen Kumar
- Neuroscience Laboratory (F-23/71), Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500046, India
| | - Phanithi Prakash Babu
- Neuroscience Laboratory (F-23/71), Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500046, India.
| |
Collapse
|
53
|
Vigil JM, Montera MA, Pentkowski NS, Diviant JP, Orozco J, Ortiz AL, Rael LJ, Westlund KN. The Therapeutic Effectiveness of Full Spectrum Hemp Oil Using a Chronic Neuropathic Pain Model. Life (Basel) 2020; 10:E69. [PMID: 32443500 PMCID: PMC7281216 DOI: 10.3390/life10050069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/09/2020] [Accepted: 05/14/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Few models exist that can control for placebo and expectancy effects commonly observed in clinical trials measuring 'Cannabis' pharmacodynamics. We used the Foramen Rotundum Inflammatory Constriction Trigeminal Infraorbital Nerve injury (FRICT-ION) model to measure the effect of "full-spectrum" whole plant extracted hemp oil on chronic neuropathic pain sensitivity in mice. METHODS Male BALBc mice were submitted to the FRICT-ION chronic neuropathic pain model with oral insertion through an incision in the buccal/cheek crease of 3 mm of chromic gut suture (4-0). The suture, wedged along the V2 trigeminal nerve branch, creates a continuous irritation that develops into secondary mechanical hypersensitivity on the snout. Von Frey filament stimuli on the mouse whisker pad was used to assess the mechanical pain threshold from 0-6 h following dosing among animals (n = 6) exposed to 5 μL of whole plant extracted hemp oil combined with a peanut butter vehicle (0.138 mg/kg), the vehicle alone (n = 3) 7 weeks post-surgery, or a naïve control condition (n = 3). RESULTS Mechanical allodynia was alleviated within 1 h (d = 2.50, p < 0.001) with a peak reversal effect at 4 h (d = 7.21, p < 0.001) and remained significant throughout the 6 h observation window. There was no threshold change on contralateral whisker pad after hemp oil administration, demonstrating the localization of anesthetic response to affected areas. CONCLUSION Future research should focus on how whole plant extracted hemp oil affects multi-sensory and cognitive-attentional systems that process pain.
Collapse
Affiliation(s)
- Jacob M. Vigil
- Department of Psychology, University of New Mexico, Albuquerque, NM 87131, USA; (J.M.V.); (N.S.P.); (J.P.D.); (J.O.)
| | - Marena A. Montera
- Department of Anesthesiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA;
| | - Nathan S. Pentkowski
- Department of Psychology, University of New Mexico, Albuquerque, NM 87131, USA; (J.M.V.); (N.S.P.); (J.P.D.); (J.O.)
| | - Jegason P. Diviant
- Department of Psychology, University of New Mexico, Albuquerque, NM 87131, USA; (J.M.V.); (N.S.P.); (J.P.D.); (J.O.)
| | - Joaquin Orozco
- Department of Psychology, University of New Mexico, Albuquerque, NM 87131, USA; (J.M.V.); (N.S.P.); (J.P.D.); (J.O.)
| | - Anthony L. Ortiz
- Organic-Energetic Solutions, Albuquerque, NM 87108, USA; (A.L.O.); (L.J.R.)
| | - Lawrence J. Rael
- Organic-Energetic Solutions, Albuquerque, NM 87108, USA; (A.L.O.); (L.J.R.)
| | - Karin N. Westlund
- Department of Anesthesiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA;
| |
Collapse
|
54
|
Kummer KK, Mitrić M, Kalpachidou T, Kress M. The Medial Prefrontal Cortex as a Central Hub for Mental Comorbidities Associated with Chronic Pain. Int J Mol Sci 2020; 21:E3440. [PMID: 32414089 PMCID: PMC7279227 DOI: 10.3390/ijms21103440] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic pain patients frequently develop and suffer from mental comorbidities such as depressive mood, impaired cognition, and other significant constraints of daily life, which can only insufficiently be overcome by medication. The emotional and cognitive components of pain are processed by the medial prefrontal cortex, which comprises the anterior cingulate cortex, the prelimbic, and the infralimbic cortex. All three subregions are significantly affected by chronic pain: magnetic resonance imaging has revealed gray matter loss in all these areas in chronic pain conditions. While the anterior cingulate cortex appears hyperactive, prelimbic, and infralimbic regions show reduced activity. The medial prefrontal cortex receives ascending, nociceptive input, but also exerts important top-down control of pain sensation: its projections are the main cortical input of the periaqueductal gray, which is part of the descending inhibitory pain control system at the spinal level. A multitude of neurotransmitter systems contributes to the fine-tuning of the local circuitry, of which cholinergic and GABAergic signaling are particularly emerging as relevant components of affective pain processing within the prefrontal cortex. Accordingly, factors such as distraction, positive mood, and anticipation of pain relief such as placebo can ameliorate pain by affecting mPFC function, making this cortical area a promising target region for medical as well as psychosocial interventions for pain therapy.
Collapse
Affiliation(s)
| | | | | | - Michaela Kress
- Institute of Physiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.K.K.); (M.M.); (T.K.)
| |
Collapse
|
55
|
Li YC, Panikker P, Xing B, Yang SS, Alexandropoulos C, McEachern EP, Akumuo R, Zhao E, Gulchina Y, Pletnikov MV, Urs NM, Caron MG, Elefant F, Gao WJ. Deletion of Glycogen Synthase Kinase-3β in D 2 Receptor-Positive Neurons Ameliorates Cognitive Impairment via NMDA Receptor-Dependent Synaptic Plasticity. Biol Psychiatry 2020; 87:745-755. [PMID: 31892408 PMCID: PMC7103512 DOI: 10.1016/j.biopsych.2019.10.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cortical dopaminergic systems are critically involved in prefrontal cortex (PFC) functions, especially in working memory and neurodevelopmental disorders such as schizophrenia. GSK-3β (glycogen synthase kinase-3β) is highly associated with cAMP (cyclic adenosine monophosphate)-independent dopamine D2 receptor (D2R)-mediated signaling to affect dopamine-dependent behaviors. However, the mechanisms underlying the GSK-3β modulation of cognitive function via D2Rs remains unclear. METHODS This study explored how conditional cell-type-specific ablation of GSK-3β in D2R+ neurons (D2R-GSK-3β-/-) in the brain affects synaptic function in the medial PFC (mPFC). Both male and female (postnatal days 60-90) mice, including 140 D2R, 24 D1R, and 38 DISC1 mice, were used. RESULTS This study found that NMDA receptor (NMDAR) function was significantly increased in layer V pyramidal neurons in mPFC of D2R-GSK-3β-/- mice, along with increased dopamine modulation of NMDAR-mediated current. Consistently, NR2A and NR2B protein levels were elevated in mPFC of D2R-GSK-3β-/- mice. This change was accompanied by a significant increase in enrichment of activator histone mark H3K27ac at the promoters of both Grin2a and Grin2b genes. In addition, altered short- and long-term synaptic plasticity, along with an increased spine density in layer V pyramidal neurons, were detected in D2R-GSK-3β-/- mice. Indeed, D2R-GSK-3β-/- mice also exhibited a resistance of working memory impairment induced by injection of NMDAR antagonist MK-801. Notably, either inhibiting GSK-3β or disrupting the D2R-DISC1 complex was able to reverse the mutant DISC1-induced decrease of NMDAR-mediated currents in the mPFC. CONCLUSIONS This study demonstrates that GSK-3β modulates cognition via D2R-DISC1 interaction and epigenetic regulation of NMDAR expression and function.
Collapse
Affiliation(s)
- Yan-Chun Li
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania.
| | - Priyalakshmi Panikker
- Department of Biology, Drexel University College of Medicine, Philadelphia, PA 19104, USA
| | - Bo Xing
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Sha-Sha Yang
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Cassandra Alexandropoulos
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Erin P McEachern
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Rita Akumuo
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Elise Zhao
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Yelena Gulchina
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Mikhail V. Pletnikov
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Nikhil M. Urs
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | - Marc G. Caron
- Departments of Cell Biology, Neurobiology, and Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Felice Elefant
- Department of Biology, Drexel University College of Medicine, Philadelphia, PA 19104, USA
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
56
|
The prefrontal cortex and the caudate nucleus respond conjointly to methylphenidate (Ritalin). Concomitant behavioral and neuronal recording study. Brain Res Bull 2020; 157:77-89. [PMID: 31987926 DOI: 10.1016/j.brainresbull.2019.10.009] [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: 07/29/2019] [Revised: 09/15/2019] [Accepted: 10/23/2019] [Indexed: 01/07/2023]
Abstract
Methylphenidate (MPD) is commonly used to treat attention-deficit hyperactivity disorder (ADHD). Recently, it is being abused for cognitive enhancement and recreation leading to concerns regarding its addictive potential. The prefrontal cortex (PFC) and caudate nucleus (CN) are two of the brain structures involved in the motive/reward circuit most affected by MPD and are also thought to be responsible for ADHD phenomena. This study is unique in that it investigated acute and chronic, dose-response MPD exposure on animals' behavior activity concomitantly with PFC and CN neuronal circuitry in freely behaving adult animals without the interference of anesthesia. Further, it compared acute and chronic MPD action on over 1,000 subcortical and cortical neurons simultaneously, allowing for a more accurate interpretation of drug action on corticostriatal neuronal circuitry. For this experiment, four groups of animals were used: saline (control), 0.6, 2.5, and 10.0 mg/kg MPD following acute and repetitive exposure. The data shows that the same MPD dose elicits behavioral sensitization in some animals and tolerance in others and that the PFC and CN neuronal activity correlates with the animals' behavioral responses to MPD. The expression of sensitization and tolerance are experimental biomarkers indicating that a drug has addictive potential. In general, a greater percentage of CN units responded to both acute and chronic MPD exposure as compared to PFC units. Dose response differences between the PFC and the CN units were observed. The dichotomy that some PFC and CN units responded to the same MPD dose by excitation and other units by attenuation in neuronal firing rate is discussed. In conclusion, to understand the mechanism of action of the drug, it is essential to study, simultaneously, on more than one brain site, the electrophysiological and behavioral effects of acute and chronic drug exposure, as sensitization and tolerance are experimental biomarkers indicating that a drug has addictive potential. The behavioral and neuronal data obtained from this study indicates that chronic MPD exposure results in behavioral and biochemical changes consistent with a substance abuse disorder.
Collapse
|
57
|
Mueller A, Krock RM, Shepard S, Moore T. Dopamine Receptor Expression Among Local and Visual Cortex-Projecting Frontal Eye Field Neurons. Cereb Cortex 2020; 30:148-164. [PMID: 31038690 PMCID: PMC7029694 DOI: 10.1093/cercor/bhz078] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/14/2019] [Accepted: 03/14/2019] [Indexed: 12/21/2022] Open
Abstract
Dopaminergic modulation of prefrontal cortex plays an important role in numerous cognitive processes, including attention. The frontal eye field (FEF) is modulated by dopamine and has an established role in visual attention, yet the underlying circuitry upon which dopamine acts is not known. We compared the expression of D1 and D2 dopamine receptors (D1Rs and D2Rs) across different classes of FEF neurons, including those projecting to dorsal or ventral extrastriate cortex. First, we found that both D1Rs and D2Rs are more prevalent on pyramidal neurons than on several classes of interneurons and are particularly prevalent on putatively long-range projecting pyramidals. Second, higher proportions of pyramidal neurons express D1Rs than D2Rs. Third, overall a higher proportion of inhibitory neurons expresses D2Rs than D1Rs. Fourth, among inhibitory interneurons, a significantly higher proportion of parvalbumin+ neurons expresses D2Rs than D1Rs, and a significantly higher proportion of calbindin+ neurons expresses D1Rs than D2Rs. Finally, compared with D2Rs, virtually all of the neurons with identified projections to both dorsal and ventral extrastriate visual cortex expressed D1Rs. Our results demonstrate that dopamine tends to act directly on the output of the FEF and that dopaminergic modulation of top-down projections to visual cortex is achieved predominately via D1Rs.
Collapse
Affiliation(s)
- Adrienne Mueller
- Howard Hughes Medical Institute and Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rebecca M Krock
- Howard Hughes Medical Institute and Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Steven Shepard
- Howard Hughes Medical Institute and Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tirin Moore
- Howard Hughes Medical Institute and Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
58
|
Jager A, Amiri H, Bielczyk N, van Heukelum S, Heerschap A, Aschrafi A, Poelmans G, Buitelaar JK, Kozicz T, Glennon JC. Cortical control of aggression: GABA signalling in the anterior cingulate cortex. Eur Neuropsychopharmacol 2020; 30:5-16. [PMID: 29274996 DOI: 10.1016/j.euroneuro.2017.12.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 11/14/2017] [Accepted: 12/02/2017] [Indexed: 11/28/2022]
Abstract
Reduced top-down control by cortical areas is assumed to underlie pathological forms of aggression. While the precise underlying molecular mechanisms are still elusive, it seems that balancing the excitatory and inhibitory tones of cortical brain areas has a role in aggression control. The molecular mechanisms underpinning aggression control were examined in the BALB/cJ mouse model. First, these mice were extensively phenotyped for aggression and anxiety in comparison to BALB/cByJ controls. Microarray data was then used to construct a molecular landscape, based on the mRNAs that were differentially expressed in the brains of BALB/cJ mice. Subsequently, we provided corroborating evidence for the key findings from the landscape through 1H-magnetic resonance imaging and quantitative polymerase chain reactions, specifically in the anterior cingulate cortex (ACC). The molecular landscape predicted that altered GABA signalling may underlie the observed increased aggression and anxiety in BALB/cJ mice. This was supported by a 40% reduction of 1H-MRS GABA levels and a 20-fold increase of the GABA-degrading enzyme Abat in the ventral ACC. As a possible compensation, Kcc2, a potassium-chloride channel involved in GABA-A receptor signalling, was found increased. Moreover, we observed aggressive behaviour that could be linked to altered expression of neuroligin-2, a membrane-bound cell adhesion protein that mediates synaptogenesis of mainly inhibitory synapses. In conclusion, Abat and Kcc2 seem to be involved in modulating aggressive and anxious behaviours observed in BALB/cJ mice through affecting GABA signalling in the ACC.
Collapse
Affiliation(s)
- Amanda Jager
- Department of Cognitive Neuroscience, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands.
| | - Houshang Amiri
- Department of Cognitive Neuroscience, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands; Neuroscience Research Centre, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran; Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Natalia Bielczyk
- Department of Cognitive Neuroscience, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Sabrina van Heukelum
- Department of Cognitive Neuroscience, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Arend Heerschap
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Armaz Aschrafi
- Laboratory of Molecular Biology, National Institute of Mental Health, Bethesda, United States
| | - Geert Poelmans
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University, Nijmegen, The Netherlands
| | - Jan K Buitelaar
- Department of Cognitive Neuroscience, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Tamas Kozicz
- Department of Anatomy, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Jeffrey C Glennon
- Department of Cognitive Neuroscience, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| |
Collapse
|
59
|
Frau R, Traccis F, Bortolato M. Neurobehavioural complications of sleep deprivation: Shedding light on the emerging role of neuroactive steroids. J Neuroendocrinol 2020; 32:e12792. [PMID: 31505075 PMCID: PMC6982588 DOI: 10.1111/jne.12792] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/06/2019] [Accepted: 09/05/2019] [Indexed: 01/05/2023]
Abstract
Sleep deprivation (SD) is associated with a broad spectrum of cognitive and behavioural complications, including emotional lability and enhanced stress reactivity, as well as deficits in executive functions, decision making and impulse control. These impairments, which have profound negative consequences on the health and productivity of many individuals, reflect alterations of the prefrontal cortex (PFC) and its connectivity with subcortical regions. However, the molecular underpinnings of these alterations remain elusive. Our group and others have begun examining how the neurobehavioural outcomes of SD may be influenced by neuroactive steroids, a family of molecules deeply implicated in sleep regulation and the stress response. These studies have revealed that, similar to other stressors, acute SD leads to increased synthesis of the neurosteroid allopregnanolone in the PFC. Whereas this up-regulation is likely aimed at counterbalancing the detrimental impact of oxidative stress induced by SD, the increase in prefrontal allopregnanolone levels contributes to deficits in sensorimotor gating and impulse control, signalling a functional impairment of PFC. This scenario suggests that the synthesis of neuroactive steroids during acute SD may be enacted as a neuroprotective response in the PFC; however, such compensation may in turn set off neurobehavioural complications by interfering with the corticolimbic connections responsible for executive functions and emotional regulation.
Collapse
Affiliation(s)
- Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato (CA), Italy
- National Institute of Neuroscience (INN), University of Cagliari, Monserrato (CA), Italy
| | - Francesco Traccis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato (CA), Italy
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City (UT), USA
| |
Collapse
|
60
|
Dubovyk V, Manahan-Vaughan D. Gradient of Expression of Dopamine D2 Receptors Along the Dorso-Ventral Axis of the Hippocampus. Front Synaptic Neurosci 2019; 11:28. [PMID: 31680927 PMCID: PMC6803426 DOI: 10.3389/fnsyn.2019.00028] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 09/24/2019] [Indexed: 01/11/2023] Open
Abstract
Dopamine D2-like receptors (D2R) play an important role in the regulation of hippocampal neuronal excitability and contribute to the regulation of synaptic plasticity, the encoding of hippocampus-dependent memories and the regulation of affective state. In line with this, D2R are targeted in the treatment of psychosis and affective disorders. It has been proposed that the dorso-ventral axis of the hippocampus can be functionally delineated into the dorsal pole that predominantly processes spatial information and the ventral pole that mainly addresses hippocampal processing of emotional and affective state. Although dopaminergic control of hippocampal information processing has been the focus of a multitude of studies, very little is known about the precise distribution of D2R both within anatomically defined sublayers of the hippocampus and along its dorsoventral axis, that could in turn yield insights as to the functional significance of this receptor in supporting hippocampal processing of spatial and affective information. Here, we used an immunohistochemical approach to precisely scrutinize the protein expression of D2R both within the cellular and dendritic layers of the hippocampal subfields, and along the dorso-ventral hippocampal axis. In general, we detected significantly higher levels of protein expression of D2R in the ventral, compared to the dorsal poles with regard to the CA1, CA2, CA3 and dentate gyrus (DG) regions. Effects were very consistent: the molecular layer, granule cell layer and polymorphic layer of the DG exhibited higher D2R levels in the ventral compared to dorsal hippocampus. D2R levels were also significantly higher in the ventral Stratum oriens, Stratum radiatum, and Stratum lacunosum-moleculare layers of the CA1 and CA3 regions. The apical dendrites of the ventral CA2 region also exhibited higher D2R expression compared to the dorsal pole. Taken together, our study suggests that the higher D2R expression levels of the ventral hippocampus may contribute to reported gradients in the degree of expression of synaptic plasticity along the dorso-ventral hippocampal axis, and may support behavioral information processing by the ventral hippocampus.
Collapse
Affiliation(s)
- Valentyna Dubovyk
- Medical Faculty, Department of Neurophysiology, Ruhr University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
| | | |
Collapse
|
61
|
Serotonin 5-HT 1A, 5-HT 2A and dopamine D 2 receptors strongly influence prefronto-hippocampal neural networks in alert mice: Contribution to the actions of risperidone. Neuropharmacology 2019; 158:107743. [PMID: 31430459 DOI: 10.1016/j.neuropharm.2019.107743] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/22/2019] [Accepted: 08/13/2019] [Indexed: 12/26/2022]
Abstract
Atypical antipsychotic drugs (APDs) used to treat positive and negative symptoms in schizophrenia block serotonin receptors 5-HT2AR and dopamine receptors D2R and stimulate 5-HT1AR directly or indirectly. However, the exact cellular mechanisms mediating their therapeutic actions remain unresolved. We recorded neural activity in the prefrontal cortex (PFC) and hippocampus (HPC) of freely-moving mice before and after acute administration of 5-HT1AR, 5-HT2AR and D2R selective agonists and antagonists and atypical APD risperidone. We then investigated the contribution of the three receptors to the actions of risperidone on brain activity via statistical modeling and pharmacological reversal (risperidone + 5-HT1AR antagonist WAY-100635, risperidone + 5-HT2A/2CR agonist DOI, risperidone + D2R agonist quinpirole). Risperidone, 5-HT1AR agonism with 8-OH-DPAT, 5-HT2AR antagonism with M100907, and D2R antagonism with haloperidol reduced locomotor activity of mice that correlated with a suppression of neural spiking, power of theta and gamma oscillations in PFC and HPC, and reduction of PFC-HPC theta phase synchronization. By contrast, activation of 5-HT2AR with DOI enhanced high-gamma oscillations in PFC and PFC-HPC high gamma functional connectivity, likely related to its hallucinogenic effects. Together, power changes, regression modeling and pharmacological reversals suggest an important role of 5-HT1AR agonism and 5-HT2AR antagonism in risperidone-induced alterations of delta, beta and gamma oscillations, while D2R antagonism may contribute to risperidone-mediated changes in delta oscillations. This study provides novel insight into the neural mechanisms for widely prescribed psychiatric medication targeting the serotonin and dopamine systems in two regions involved in the pathophysiology of schizophrenia.
Collapse
|
62
|
Su P, Lai TKY, Lee FHF, Abela AR, Fletcher PJ, Liu F. Disruption of SynGAP–dopamine D1 receptor complexes alters actin and microtubule dynamics and impairs GABAergic interneuron migration. Sci Signal 2019; 12:12/593/eaau9122. [DOI: 10.1126/scisignal.aau9122] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Disruption of γ-aminobutyric acid (GABA)–ergic interneuron migration is implicated in various neurodevelopmental disorders, including autism spectrum disorder and schizophrenia. The dopamine D1 receptor (D1R) promotes GABAergic interneuron migration, which is disrupted in various neurological disorders, some of which are also associated with mutations in the gene encoding synaptic Ras–guanosine triphosphatase–activating protein (SynGAP). Here, we explored the mechanisms underlying these associations and their possible connection. In prenatal mouse brain tissue, we found a previously unknown interaction between the D1R and SynGAP. This D1R-SynGAP interaction facilitated D1R localization to the plasma membrane and promoted D1R-mediated downstream signaling pathways, including phosphorylation of protein kinase A and p38 mitogen-activated protein kinase. These effects were blocked by a peptide (TAT-D1Rpep) that disrupted the D1R-SynGAP interaction. Furthermore, disrupting this complex in mice during embryonic development resulted in pronounced and selective deficits in the tangential migration of GABAergic interneurons, possibly due to altered actin and microtubule dynamics. Our results provide insights into the molecular mechanisms regulating interneuron development and suggest that disruption of the D1R-SynGAP interaction may underlie SYNGAP1 mutation–related neurodevelopmental disorders.
Collapse
|
63
|
Elevated dopamine signaling from ventral tegmental area to prefrontal cortical parvalbumin neurons drives conditioned inhibition. Proc Natl Acad Sci U S A 2019; 116:13077-13086. [PMID: 31182594 DOI: 10.1073/pnas.1901902116] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Conditioned inhibition is an important process to suppress learned responses for optimal adaptation, but its underlying biological mechanism is poorly understood. Here we used safety learning (SL)/fear discrimination after fear conditioning as a conditioned inhibition model because it demonstrates the essential properties of summation and retardation. Activity of the dorsomedial prefrontal cortex (dmPFC) parvalbumin (PV) neurons bidirectionally regulates spiking levels of dmPFC excitatory neurons and fear states. Responses to safety cues are increased in dopaminergic (DA) neurons in the ventral tegmental area (VTA) and in PV neurons in dmPFC after SL. Plasticity in the VTA is implicated, since SL requires activation of N-methyl-d-aspartate receptors. Furthermore, in a posttraumatic stress disorder model, impaired SL is associated with impaired potentiation of VTA DA neuron activity. Our results demonstrate a DA-dependent learning process that targets prefrontal inhibitory neurons for suppression of learned responses, and have implications for the pathogenesis and treatment of various psychiatric diseases.
Collapse
|
64
|
Miguel PM, Deniz BF, Confortim HD, Bronauth LP, de Oliveira BC, Alves MB, Silveira PP, Pereira LO. Methylphenidate administration reverts attentional inflexibility in adolescent rats submitted to a model of neonatal hypoxia-ischemia: Predictive validity for ADHD study. Exp Neurol 2019; 315:88-99. [DOI: 10.1016/j.expneurol.2019.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/04/2019] [Accepted: 02/08/2019] [Indexed: 12/29/2022]
|
65
|
The allosteric dopamine D1 receptor potentiator, DETQ, ameliorates subchronic phencyclidine-induced object recognition memory deficits and enhances cortical acetylcholine efflux in male humanized D1 receptor knock-in mice. Behav Brain Res 2019; 361:139-150. [DOI: 10.1016/j.bbr.2018.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/30/2018] [Accepted: 12/01/2018] [Indexed: 12/13/2022]
|
66
|
Chronic unpredictable stress promotes cell-specific plasticity in prefrontal cortex D1 and D2 pyramidal neurons. Neurobiol Stress 2019; 10:100152. [PMID: 30937357 PMCID: PMC6430618 DOI: 10.1016/j.ynstr.2019.100152] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/04/2019] [Accepted: 03/04/2019] [Indexed: 11/24/2022] Open
Abstract
Exposure to unpredictable environmental stress is widely recognized as a major determinant for risk and severity in neuropsychiatric disorders such as major depressive disorder, anxiety, schizophrenia, and PTSD. The ability of ostensibly unrelated disorders to give rise to seemingly similar psychiatric phenotypes highlights a need to identify circuit-level concepts that could unify diverse factors under a common pathophysiology. Although difficult to disentangle a causative effect of stress from other factors on medial prefrontal cortex (PFC) dysfunction, a wealth of data from humans and rodents demonstrates that the PFC is a key target of stress. The present study sought to identify a model of chronic unpredictable stress (CUS) which induces affective behaviors in C57BL6J mice and once established, measure stress-related alterations in intrinsic excitability and synaptic regulation of mPFC layer 5/6 pyramidal neurons. Adult male mice received 2 weeks of 'less intense' stress or 2 or 4 weeks of 'more intense' CUS followed by sucrose preference for assessment of anhedonia, elevated plus maze for assessment of anxiety and forced swim test for assessment of depressive-like behaviors. Our findings indicate that more intense CUS exposure results in increased anhedonia, anxiety, and depressive behaviors, while the less intense stress results in no measured behavioral phenotypes. Once a behavioral model was established, mice were euthanized approximately 21 days post-stress for whole-cell patch clamp recordings from layer 5/6 pyramidal neurons in the prelimbic (PrL) and infralimbic (IL) cortices. No significant differences were initially observed in intrinsic cell excitability in either region. However, post-hoc analysis and subsequent confirmation using transgenic mice expressing tdtomato or eGFP under control of dopamine D1-or D2-type receptor showed that D1-expressing pyramidal neurons (D1-PYR) in the PrL exhibit reduced thresholds to fire an action potential (increased excitability) but impaired firing capacity at more depolarized potentials, whereas D2-expressing pyramidal neurons (D2-PYR) showed an overall reduction in excitability and spike firing frequency. Examination of synaptic transmission showed that D1-and D2-PYR exhibit differences in basal excitatory and inhibitory signaling under naïve conditions. In CUS mice, D1-PYR showed increased frequency of both miniature excitatory and inhibitory postsynaptic currents, whereas D2-PYR only showed a reduction in excitatory currents. These findings demonstrate that D1-and D2-PYR subpopulations differentially undergo stress-induced intrinsic and synaptic plasticity that may have functional implications for stress-related pathology, and that these adaptations may reflect unique differences in basal properties regulating output of these cells.
Collapse
|
67
|
Hare BD, Shinohara R, Liu RJ, Pothula S, DiLeone RJ, Duman RS. Optogenetic stimulation of medial prefrontal cortex Drd1 neurons produces rapid and long-lasting antidepressant effects. Nat Commun 2019; 10:223. [PMID: 30644390 PMCID: PMC6333924 DOI: 10.1038/s41467-018-08168-9] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 12/11/2018] [Indexed: 12/13/2022] Open
Abstract
Impaired function in the medial prefrontal cortex (mPFC) contributes to depression, and the therapeutic response produced by novel rapid-acting antidepressants such as ketamine are mediated by mPFC activity. The mPFC contains multiple types of pyramidal cells, but it is unclear whether a particular subtype mediates the rapid antidepressant actions of ketamine. Here we tested two major subtypes, Drd1 and Drd2 dopamine receptor expressing pyramidal neurons and found that activating Drd1 expressing pyramidal cells in the mPFC produces rapid and long-lasting antidepressant and anxiolytic responses. In contrast, photostimulation of Drd2 expressing pyramidal cells was ineffective across anxiety-like and depression-like measures. Disruption of Drd1 activity also blocked the rapid antidepressant effects of ketamine. Finally, we demonstrate that stimulation of mPFC Drd1 terminals in the BLA recapitulates the antidepressant effects of somatic stimulation. These findings aid in understanding the cellular target neurons in the mPFC and the downstream circuitry involved in rapid antidepressant responses.
Collapse
Affiliation(s)
- Brendan D Hare
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Ryota Shinohara
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Rong Jian Liu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Santosh Pothula
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Ralph J DiLeone
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Ronald S Duman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA.
| |
Collapse
|
68
|
Genetic labeling reveals temporal and spatial expression pattern of D2 dopamine receptor in rat forebrain. Brain Struct Funct 2019; 224:1035-1049. [PMID: 30604007 PMCID: PMC6499762 DOI: 10.1007/s00429-018-01824-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 12/20/2018] [Indexed: 01/11/2023]
Abstract
The D2 dopamine receptor (Drd2) is implicated in several brain disorders such as schizophrenia, Parkinson’s disease, and drug addiction. Drd2 is also the primary target of both antipsychotics and Parkinson’s disease medications. Although the expression pattern of Drd2 is relatively well known in mouse brain, the temporal and spatial distribution of Drd2 is lesser clear in rat brain due to the lack of Drd2 reporter rat lines. Here, we used CRISPR/Cas9 techniques to generate two knockin rat lines: Drd2::Cre and Rosa26::loxp-stop-loxp-tdTomato. By crossing these two lines, we produced Drd2 reporter rats expressing the fluorescence protein tdTomato under the control of the endogenous Drd2 promoter. Using fluorescence imaging and unbiased stereology, we revealed the cellular expression pattern of Drd2 in adult and postnatal rat forebrain. Strikingly, the Drd2 expression pattern differs between Drd2 reporter rats and Drd2 reporter mice generated by BAC transgene in prefrontal cortex and hippocampus. These results provide fundamental information needed for the study of Drd2 function in rat forebrain. The Drd2::Cre rats generated here may represent a useful tool to study the function of neuronal populations expressing Drd2.
Collapse
|
69
|
Quintana C, Beaulieu JM. A fresh look at cortical dopamine D2 receptor expressing neurons. Pharmacol Res 2018; 139:440-445. [PMID: 30528973 DOI: 10.1016/j.phrs.2018.12.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/28/2018] [Accepted: 12/03/2018] [Indexed: 01/02/2023]
Abstract
The dopamine D2 receptor (DRD2) remains the principal target of antipsychotic drugs used for the management of schizophrenia and other psychotic disorders. This receptor is highly expressed within the basal ganglia, more specifically the striatal caudate nucleus and the nucleus accumbens. The general functions, signaling and behavioral contributions of striatal DRD2 are well understood. However, the study of cortical DRD2 expression and functions has for the most part been restricted to a subset of pyramidal neurons and interneurons (e.g. parvalbumine positive) of the pre frontal cortex where DRD2 regulated local circuits are believed to contribute to the regulation of emotional and cognitive functions. The further investigations of cortical DRD2 functions have been hindered by relatively low receptor expression and the sensitivity of detection methods. Here we report recent findings by our group using high sensitivity approaches to map cortical DRD2 expression. Results from these investigations revealed different scales of heterogeneity within DRD2 expressing neurons. These variations affected the types of neurons expressing DRD2 as well as the co-expression of DRD2 with other receptors across several cortical regions. Furthermore several cortical regions showing higher clusters of DRD2 expressing neurons are involved in the regulation of emotional, cognitive and sensory functions that can be involved in the expression of psychotic symptoms. These findings underscore the need for a reexamination of cortical DRD2 mediated synaptic plasticity in the context of schizophrenia and other psychotic disorders.
Collapse
Affiliation(s)
- Clémentine Quintana
- Department of Pharmacology & Toxicology, University of Toronto, Medical Sciences Building, Toronto, Ontario, M5S 1A8, Canada
| | - Jean-Martin Beaulieu
- Department of Pharmacology & Toxicology, University of Toronto, Medical Sciences Building, Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
70
|
Weele CMV, Siciliano CA, Tye KM. Dopamine tunes prefrontal outputs to orchestrate aversive processing. Brain Res 2018; 1713:16-31. [PMID: 30513287 DOI: 10.1016/j.brainres.2018.11.044] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/25/2018] [Accepted: 11/30/2018] [Indexed: 01/06/2023]
Abstract
Decades of research suggest that the mesocortical dopamine system exerts powerful control over mPFC physiology and function. Indeed, dopamine signaling in the medial prefrontal cortex (mPFC) is implicated in a vast array of processes, including working memory, stimulus discrimination, stress responses, and emotional and behavioral control. Consequently, even slight perturbations within this delicate system result in profound disruptions of mPFC-mediated processes. Many neuropsychiatric disorders are associated with dysregulation of mesocortical dopamine, including schizophrenia, depression, attention deficit hyperactivity disorder, post-traumatic stress disorder, among others. Here, we review the anatomy and functions of the mesocortical dopamine system. In contrast to the canonical role of striatal dopamine in reward-related functions, recent work has revealed that mesocortical dopamine fine-tunes distinct efferent projection populations in a manner that biases subsequent behavior towards responding to stimuli associated with potentially aversive outcomes. We propose a framework wherein dopamine can serve as a signal for switching mPFC states by orchestrating how information is routed to the rest of the brain.
Collapse
Affiliation(s)
- Caitlin M Vander Weele
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Cody A Siciliano
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kay M Tye
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
71
|
Downregulation of Dopamine D1-like Receptor Pathways of GABAergic Interneurons in the Anterior Cingulate Cortex of Spontaneously Hypertensive Rats. Neuroscience 2018; 394:267-285. [DOI: 10.1016/j.neuroscience.2018.10.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 09/28/2018] [Accepted: 10/22/2018] [Indexed: 01/09/2023]
|
72
|
Huang S, Borgland SL, Zamponi GW. Dopaminergic modulation of pain signals in the medial prefrontal cortex: Challenges and perspectives. Neurosci Lett 2018; 702:71-76. [PMID: 30503912 DOI: 10.1016/j.neulet.2018.11.043] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Chronic pain is a massive socieoeconomic burden and is often refractory to treatment. To devise novel therapeutic interventions, it is important to understand in detail the processing of pain signals in the brain. Recent studies have revealed shared features between the brain's reward and pain systems. Dopamine (DA) is a key neuromodulator in the mesocorticolimbic system that has been implicated not only in motivated behaviours, reinforcement learning and reward processing, but also in the pain axis. The medial prefrontal cortex (mPFC) is an important region for mediating executive functions including attention, judgement, and learning. Studies have revealed that the mPFC undergoes plasticity during the development of chronic pain. The mPFC receives dopaminergic input from the ventral tegmental area (VTA), and stimulation of these inputs has been shown to modulate the plasticity of the mPFC and anxiety and aversive behaviour. Here, we review the role of the mPFC and its dopaminergic modulation in chronic pain.
Collapse
Affiliation(s)
- Shuo Huang
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada.
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada.
| |
Collapse
|
73
|
Hearing M. Prefrontal-accumbens opioid plasticity: Implications for relapse and dependence. Pharmacol Res 2018; 139:158-165. [PMID: 30465850 DOI: 10.1016/j.phrs.2018.11.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/05/2018] [Accepted: 11/07/2018] [Indexed: 01/12/2023]
Abstract
In addiction, an individual's ability to inhibit drug seeking and drug taking is thought to reflect a pathological strengthening of drug-seeking behaviors or impairments in the capacity to control maladaptive behavior. These processes are not mutually exclusive and reflect drug-induced modifications within prefrontal cortical and nucleus accumbens circuits, however unlike psychostimulants such as cocaine, far less is known about the temporal, anatomical, and cellular dynamics of these changes. We discuss what is known regarding opioid-induced adaptations in intrinsic membrane physiology and pre-/postsynaptic neurotransmission in principle pyramidal and medium spiny neurons in the medial prefrontal cortex and nucleus accumbens from electrophysiological studies and explore how circuit specific adaptations may contribute to unique facets of opioid addiction.
Collapse
Affiliation(s)
- Matthew Hearing
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, 53233, USA.
| |
Collapse
|
74
|
Kang S, Cox CL, Gulley JM. High frequency stimulation-induced plasticity in the prelimbic cortex of rats emerges during adolescent development and is associated with an increase in dopamine receptor function. Neuropharmacology 2018; 141:158-166. [PMID: 30165079 DOI: 10.1016/j.neuropharm.2018.08.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 08/13/2018] [Accepted: 08/26/2018] [Indexed: 01/10/2023]
Abstract
Recent studies in rats suggest that high frequency stimulation (HFS) in the ventral hippocampus induces long-term depression (LTD) in the deep layer of the medial prefrontal cortex (mPFC), but only after the prefrontal GABA system has sufficiently developed during early-to mid-adolescence. It is not clear whether this LTD is specific to the hippocampus-mPFC circuit or is instead an intrinsitc regulatory mechanism for the developed mPFC neuro-network. The potential mechanisms underlying this HFS-induced LTD are also largely unknown. In the current study, naïve male Sprague Dawley rats were sacrificed during peri-adolescence or young adulthood for in vitro extracellular recording to determine if HFS delivered in the prelimbic cortex (PLC) would induce LTD in an age-dependent manner and if dopamine receptors are involved in the expression of this LTD. We found four trains of stimulation at 50 Hz induced an LTD in the PFC of adult, but not peri-adolescent, rats. This LTD required intact GABAA receptor functioning and could also be blocked by dopamine D1 or D2 receptor antagonists. Bath application of selective D1 or D2 receptor agonists produced a significant facilitation or suppression in the field potential, respectively, and these effects were only observed in the adult PLC. Furthermore, neither D1 nor D2 stimualtion prior to HFS was able to facilitate LTD in the peri-adolescent PLC. Together, these results suggest dopamine receptor functionality in the PLC increases during adolescent development and it plays an important role in this late-maturating form of plasticity.
Collapse
Affiliation(s)
- Shuo Kang
- Neuroscience Program, University of Illinois, Urbana-Champaign, USA
| | - Charles L Cox
- Neuroscience Program, University of Illinois, Urbana-Champaign, USA; Department of Molecular and Integrative Physiology, University of Illinois, Urbana-Champaign, USA; Department of Pharmacology, University of Illinois, Urbana-Champaign, USA; Beckman Institute for Advanced Science, University of Illinois, Urbana-Champaign, USA.
| | - Joshua M Gulley
- Neuroscience Program, University of Illinois, Urbana-Champaign, USA; Department of Psychology, University of Illinois, Urbana-Champaign, USA; Institute for Genomic Biology, University of Illinois, Urbana-Champaign, USA.
| |
Collapse
|
75
|
Selleck RA, Giacomini J, Buchholtz BD, Lake C, Sadeghian K, Baldo BA. Modulation of appetitive motivation by prefrontal cortical mu-opioid receptors is dependent upon local dopamine D1 receptor signaling. Neuropharmacology 2018; 140:302-309. [PMID: 30086291 DOI: 10.1016/j.neuropharm.2018.07.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/25/2018] [Accepted: 07/26/2018] [Indexed: 10/28/2022]
Abstract
Opioid neurotransmission has been implicated in psychiatric disorders featuring impaired control over appetitive motivation, such as addiction and binge-eating disorder. We have previously shown that infusions of the μ-opioid receptor (μOR) agonist DAMGO into the ventromedial prefrontal cortex (vmPFC) induced hyperphagia, increased motor activity, and augmented sucrose-reinforced responding in the task progressive ratio (PR) task, which assesses the motivational value of an incentive. These effects were not reproduced by intra-PFC infusion of a variety of dopamine (DA) agonists and antagonists, suggesting that manipulation of intra-PFC DA systems alone is not sufficient to reproduce μOR-like effects. Nevertheless, this does not rule out interactions between PFC DA and μ-opioid systems. Here we used intra-vmPFC drug cocktails containing DAMGO and SCH 23390 (a DA D1 receptor antagonist) to determine whether increases in appetitive motivation and motor activity elicited by intra-vmPFC μOR stimulation require intact signaling through vmPFC D1 receptors. Blockade of D1 receptors with SCH 23390 attenuated the enhancement of PR breakpoint, and increases in exploratory-like behavior and feeding initiation elicited by intra-vmPFC μOR stimulation. These results establish that intra-vmPFC D1 signaling is required for the expression of behavioral effects evoked by μOR stimulation within the PFC, and further suggest that D1 tone plays an enabling or permissive role in the expression of μOR -elicited effects. Simultaneous targeting of both μ-opioid and D1 systems may represent a more efficacious treatment strategy (compared to μOR blockade alone) for psychiatric disorders characterized by dysregulated appetitive motivation.
Collapse
Affiliation(s)
- Ryan A Selleck
- Dept. Cellular & Molecular Pharmacology, Rosalind Franklin, Univ.of Medicine & Science, USA
| | - Juliana Giacomini
- Physiology Graduate Training Program, Univ. Wisconsin-Madison, School of Medicine &Public Health, USA
| | | | - Curtis Lake
- College of Agricultural & Life Sciences, Univ. Wisconsin-Madison, USA
| | - Ken Sadeghian
- Dept. Psychiatry, Univ. Wisconsin-Madison, School of Medicine & Public Health, USA
| | - Brian A Baldo
- Dept. Psychiatry, Univ. Wisconsin-Madison, School of Medicine & Public Health, USA.
| |
Collapse
|
76
|
Leyrer‐Jackson JM, Thomas MP. Layer-specific effects of dopaminergic D1 receptor activation on excitatory synaptic trains in layer V mouse prefrontal cortical pyramidal cells. Physiol Rep 2018; 6:e13806. [PMID: 30073790 PMCID: PMC6072891 DOI: 10.14814/phy2.13806] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/03/2018] [Accepted: 04/30/2018] [Indexed: 11/24/2022] Open
Abstract
In humans, executive functions (e.g., working memory [WM]) are mediated in part by prefrontal cortical areas (PFC), where ventromedial areas may be homologous to ventromedial areas (mPFC) in rodents. Many executive functions are critically dependent on optimal dopamine levels within the PFC; however, our understanding of the role of dopamine in modulating PFC-mediated tasks is incomplete. Stable patterns of neuronal activity have been associated with WM processes, and recurrent excitatory synaptic activity has been proposed to play a role in this sustained activity. This excitatory activity may be regulated in a frequency-dependent manner. Thus, we examined the effects of dopamine D1-like receptor (D1R) activation on short-term excitatory postsynaptic potential (EPSP) dynamics in two subtypes of mouse layer V mPFC pyramidal neurons by varying evoked train frequency from 10 to 50 Hz. We isolated non-NMDA receptor (non-NMDAR) and NMDA receptor (NMDAR)-mediated components of EPSP trains, which were evoked by stimulating fibers located either within layer V or layer I of the mPFC. Interestingly, no differences in the effects of D1R activation were observed between subcortically projecting (PT or pyramidal tract) and contralaterally projecting (IT or intratelencephalic) layer V pyramidal cells. However, we found that D1R activation had layer-specific effects on NMDAR- and non-NMDAR-mediated EPSP trains: while D1R activation increased the amplitude of both components with layer V stimulation, with layer I stimulation D1R activation had no effect on non-NMDAR-mediated EPSP trains but decreased the amplitude of NMDAR-mediated EPSP trains. Our results suggest that dopamine, acting at D1-like receptors, increases the influence of local inputs from other layer V pyramidal cells, but may restrict the influence of layer I (tuft) inputs. Our demonstration of differential D1R regulation of excitatory synaptic dynamics in distinct compartments of mPFC layer V neurons may provide another important aspect linking cellular mechanisms of dopaminergic modulation to PFC network functioning, and ultimately to executive functions such as working memory.
Collapse
Affiliation(s)
- Jonna M. Leyrer‐Jackson
- University of NorthernColorado School of Biological SciencesUniversity of Northern ColoradoGreeleyColorado
| | - Mark P. Thomas
- University of NorthernColorado School of Biological SciencesUniversity of Northern ColoradoGreeleyColorado
| |
Collapse
|
77
|
Dopamine D1 receptor subtype mediates acute stress-induced dendritic growth in excitatory neurons of the medial prefrontal cortex and contributes to suppression of stress susceptibility in mice. Mol Psychiatry 2018; 23:1717-1730. [PMID: 28924188 DOI: 10.1038/mp.2017.177] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/18/2017] [Accepted: 07/28/2017] [Indexed: 12/12/2022]
Abstract
Dopamine in prefrontal cortices is implicated in cognitive and emotional functions, and the dysfunction of prefrontal dopamine has been associated with cognitive and emotional deficits in mental illnesses. These findings have led to clinical trials of dopamine-targeting drugs and brain imaging of dopamine receptors in patients with mental illnesses. Rodent studies have suggested that dopaminergic pathway projecting to the medial prefrontal cortex (mPFC) suppresses stress susceptibility. Although various types of mPFC neurons express several dopamine receptor subtypes, previous studies neither isolated a role of dopamine receptor subtype nor identified the site of its action in mPFC. Using social defeat stress (SDS) in mice, here we identified a role of dopamine D1 receptor subtype in mPFC excitatory neurons in suppressing stress susceptibility. Repeated social defeat stress (R-SDS) reduces the expression of D1 receptor subtype in mPFC of mice susceptible to R-SDS. Knockdown of D1 receptor subtype in whole neuronal populations or excitatory neurons in mPFC facilitates the induction of social avoidance by SDS. Single social defeat stress (S-SDS) induces D1 receptor-mediated extracellular signal-regulated kinase phosphorylation and c-Fos expression in mPFC neurons. Whereas R-SDS reduces dendritic lengths of mPFC layer II/III pyramidal neurons, S-SDS increases arborization and spines of apical dendrites of these neurons in a D1 receptor-dependent manner. Collectively, our findings show that D1 receptor subtype and related signaling in mPFC excitatory neurons mediate acute stress-induced dendritic growth of these neurons and contribute to suppression of stress susceptibility. Therefore, we propose that D1 receptor-mediated dendritic growth in mPFC excitatory neurons suppresses stress susceptibility.
Collapse
|
78
|
Jacob SN, Nienborg H. Monoaminergic Neuromodulation of Sensory Processing. Front Neural Circuits 2018; 12:51. [PMID: 30042662 PMCID: PMC6048220 DOI: 10.3389/fncir.2018.00051] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 06/11/2018] [Indexed: 12/17/2022] Open
Abstract
All neuronal circuits are subject to neuromodulation. Modulatory effects on neuronal processing and resulting behavioral changes are most commonly reported for higher order cognitive brain functions. Comparatively little is known about how neuromodulators shape processing in sensory brain areas that provide the signals for downstream regions to operate on. In this article, we review the current knowledge about how the monoamine neuromodulators serotonin, dopamine and noradrenaline influence the representation of sensory stimuli in the mammalian sensory system. We review the functional organization of the monoaminergic brainstem neuromodulatory systems in relation to their role for sensory processing and summarize recent neurophysiological evidence showing that monoamines have diverse effects on early sensory processing, including changes in gain and in the precision of neuronal responses to sensory inputs. We also highlight the substantial evidence for complementarity between these neuromodulatory systems with different patterns of innervation across brain areas and cortical layers as well as distinct neuromodulatory actions. Studying the effects of neuromodulators at various target sites is a crucial step in the development of a mechanistic understanding of neuronal information processing in the healthy brain and in the generation and maintenance of mental diseases.
Collapse
Affiliation(s)
- Simon N Jacob
- Department of Neurosurgery, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Hendrikje Nienborg
- Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| |
Collapse
|
79
|
Murphy MJM, Deutch AY. Organization of afferents to the orbitofrontal cortex in the rat. J Comp Neurol 2018; 526:1498-1526. [PMID: 29524205 PMCID: PMC5899655 DOI: 10.1002/cne.24424] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/29/2018] [Accepted: 02/09/2018] [Indexed: 01/05/2023]
Abstract
The prefrontal cortex (PFC) is usually defined as the frontal cortical area receiving a mediodorsal thalamic (MD) innervation. Certain areas in the medial wall of the rat frontal area receive a MD innervation. A second frontal area that is the target of MD projections is located dorsal to the rhinal sulcus and often referred to as the orbitofrontal cortex (OFC). Both the medial PFC and OFC are comprised of a large number of cytoarchitectonic regions. We assessed the afferent innervation of the different areas of the OFC, with a focus on projections arising from the mediodorsal thalamic nucleus, the basolateral nucleus of the amygdala, and the midbrain dopamine neurons. Although there are specific inputs to various OFC areas, a simplified organizational scheme could be defined, with the medial areas of the OFC receiving thalamic inputs, the lateral areas of the OFC being the recipient of amygdala afferents, and a central zone that was the target of midbrain dopamine neurons. Anterograde tracer data were consistent with this organization of afferents, and revealed that the OFC inputs from these three subcortical sites were largely spatially segregated. This spatial segregation suggests that the central portion of the OFC (pregenual agranular insular cortex) is the only OFC region that is a prefrontal cortical area, analogous to the prelimbic cortex in the medial prefrontal cortex. These findings highlight the heterogeneity of the OFC, and suggest possible functional attributes of the three different OFC areas.
Collapse
Affiliation(s)
| | - Ariel Y. Deutch
- Neuroscience Program, Vanderbilt University, Nashville, TN
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN
- Department of Pharmacology, Vanderbilt University, Nashville, TN
| |
Collapse
|
80
|
Cheng Y, Wang X, Wei X, Xie X, Melo S, Miranda RC, Wang J. Prenatal Exposure to Alcohol Induces Functional and Structural Plasticity in Dopamine D1 Receptor-Expressing Neurons of the Dorsomedial Striatum. Alcohol Clin Exp Res 2018; 42:10.1111/acer.13806. [PMID: 29870053 PMCID: PMC6281858 DOI: 10.1111/acer.13806] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/29/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) is a leading cause of hyperactivity in children. Excitation of dopamine D1 receptor-expressing medium spiny neurons (D1-MSNs) of the dorsomedial striatum (DMS), a brain region that controls voluntary behavior, is known to induce hyperactivity in mice. We therefore hypothesized that PAE-linked hyperactivity was due to persistently altered glutamatergic activity in DMS D1-MSNs. METHODS Female Ai14 tdTomato reporter mice were given access to alcohol in an intermittent access, 2-bottle choice paradigm before pregnancy, and following mating with male D1-Cre mice, through the pregnancy period, and until postnatal day (P) 10. Locomotor activity was tested in juvenile (P21) and adult (P133) offspring, and alcohol-conditioned place preference (CPP) was measured in adult offspring. Glutamatergic activity in DMS D1-MSNs of adult PAE and control mice was measured by slice electrophysiology, followed by measurements of dendritic morphology. RESULTS Our voluntary maternal alcohol consumption model resulted in increased locomotor activity in juvenile PAE mice, and this hyperactivity was maintained into adulthood. Furthermore, PAE resulted in a higher alcohol-induced CPP in adult offspring. Glutamatergic activity onto DMS D1-MSNs was also enhanced by PAE. Finally, PAE increased dendritic complexity in DMS D1-MSNs in adult offspring. CONCLUSIONS Our model of PAE does result in persistent hyperactivity in offspring. In adult PAE offspring, hyperactivity is accompanied by potentiated glutamatergic strength and afferent connectivity in DMS D1-MSNs, an outcome that is also consistent with the observed increase in alcohol preference in PAE offspring. Consequently, a PAE-sensitive circuit, centered within the D1-MSN, may be linked to behavioral outcomes of PAE.
Collapse
Affiliation(s)
- Yifeng Cheng
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas 77807
| | - Xuehua Wang
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas 77807
| | - Xiaoyan Wei
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas 77807
| | - Xueyi Xie
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas 77807
| | - Sebastian Melo
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas 77807
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas 77807
| | - Jun Wang
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas 77807
| |
Collapse
|
81
|
What does the Fos say? Using Fos-based approaches to understand the contribution of stress to substance use disorders. Neurobiol Stress 2018; 9:271-285. [PMID: 30450391 PMCID: PMC6234265 DOI: 10.1016/j.ynstr.2018.05.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/08/2018] [Accepted: 05/25/2018] [Indexed: 02/06/2023] Open
Abstract
Despite extensive research efforts, drug addiction persists as a largely unmet medical need. Perhaps the biggest challenge for treating addiction is the high rate of recidivism. While many factors can promote relapse in abstinent drug users, the contribution of stress is particularly problematic, as stress is uncontrollable and pervasive in the lives of those struggling with addiction. Thus, understanding the neurocircuitry that underlies the influence of stress on drug seeking is critical for guiding treatment. Preclinical research aimed at defining this neurocircuitry has, in part, relied upon the use of experimental approaches that allow visualization of cellular and circuit activity that corresponds to stressor-induced drug seeking in rodent relapse models. Much of what we have learned about the mechanisms that mediate stressor-induced relapse has been informed by studies that have used the expression of the immediate early gene, cfos, or its protein product, Fos, as post-mortem activity markers. In this review we provide an overview of the rodent models used to study stressor-induced relapse and briefly summarize what is known about the underlying neurocircuitry before describing the use of cfos/Fos-based approaches. In addition to reviewing findings obtained using this approach, its advantages and limitations are considered. Moreover, new techniques that leverage the expression profile of cfos to tag and manipulate cells based on their activity patterns are discussed. The intent of the review is to guide the interpretation of old and design of new studies that utilize cfos/Fos-based strategies to study the neurocircuitry that contributes to stress-related drug use.
Collapse
|
82
|
Choi SJ, Mukai J, Kvajo M, Xu B, Diamantopoulou A, Pitychoutis PM, Gou B, Gogos JA, Zhang H. A Schizophrenia-Related Deletion Leads to KCNQ2-Dependent Abnormal Dopaminergic Modulation of Prefrontal Cortical Interneuron Activity. Cereb Cortex 2018; 28:2175-2191. [PMID: 28525574 PMCID: PMC6018968 DOI: 10.1093/cercor/bhx123] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 03/25/2017] [Indexed: 02/06/2023] Open
Abstract
Altered prefrontal cortex function is implicated in schizophrenia (SCZ) pathophysiology and could arise from imbalance between excitation and inhibition (E/I) in local circuits. It remains unclear whether and how such imbalances relate to genetic etiologies. We used a mouse model of the SCZ-predisposing 22q11.2 deletion (Df(16)A+/- mice) to evaluate how this genetic lesion affects the excitability of layer V prefrontal pyramidal neurons and its modulation by dopamine (DA). Df(16)A+/- mice have normal balance between E/I at baseline but are unable to maintain it upon dopaminergic challenge. Specifically, in wild-type mice, D1 receptor (D1R) activation enhances excitability of layer V prefrontal pyramidal neurons and D2 receptor (D2R) activation reduces it. Whereas the excitatory effect upon D1R activation is enhanced in Df(16)A+/- mice, the inhibitory effect upon D2R activation is reduced. The latter is partly due to the inability of mutant mice to activate GABAergic parvalbumin (PV)+ interneurons through D2Rs. We further demonstrate that reduced KCNQ2 channel function in PV+ interneurons in Df(16)A+/- mice renders them less capable of inhibiting pyramidal neurons upon D2 modulation. Thus, DA modulation of PV+ interneurons and control of E/I are altered in Df(16)A+/- mice with a higher excitation and lower inhibition during dopaminergic modulation.
Collapse
Affiliation(s)
- Se Joon Choi
- Department of Neurology, Columbia University, New York, NY10032, USA
| | - Jun Mukai
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Mirna Kvajo
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Bin Xu
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Anastasia Diamantopoulou
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Pothitos M Pitychoutis
- Department of Biology, Center for Tissue Regeneration and Engineering (TREND), University of Dayton, 300 College Park, Dayton, OH 45469, USA
| | - Bin Gou
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Joseph A Gogos
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
- Department of Neuroscience, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Hui Zhang
- Department of Neurology, Columbia University, New York, NY10032, USA
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
83
|
Paroxetine and Low-dose Risperidone Induce Serotonin 5-HT1A and Dopamine D2 Receptor Heteromerization in the Mouse Prefrontal Cortex. Neuroscience 2018; 377:184-196. [DOI: 10.1016/j.neuroscience.2018.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 02/12/2018] [Accepted: 03/04/2018] [Indexed: 01/10/2023]
|
84
|
Radnikow G, Feldmeyer D. Layer- and Cell Type-Specific Modulation of Excitatory Neuronal Activity in the Neocortex. Front Neuroanat 2018; 12:1. [PMID: 29440997 PMCID: PMC5797542 DOI: 10.3389/fnana.2018.00001] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 01/04/2018] [Indexed: 01/08/2023] Open
Abstract
From an anatomical point of view the neocortex is subdivided into up to six layers depending on the cortical area. This subdivision has been described already by Meynert and Brodmann in the late 19/early 20. century and is mainly based on cytoarchitectonic features such as the size and location of the pyramidal cell bodies. Hence, cortical lamination is originally an anatomical concept based on the distribution of excitatory neuron. However, it has become apparent in recent years that apart from the layer-specific differences in morphological features, many functional properties of neurons are also dependent on cortical layer or cell type. Such functional differences include changes in neuronal excitability and synaptic activity by neuromodulatory transmitters. Many of these neuromodulators are released from axonal afferents from subcortical brain regions while others are released intrinsically. In this review we aim to describe layer- and cell-type specific differences in the effects of neuromodulator receptors in excitatory neurons in layers 2–6 of different cortical areas. We will focus on the neuromodulator systems using adenosine, acetylcholine, dopamine, and orexin/hypocretin as examples because these neuromodulator systems show important differences in receptor type and distribution, mode of release and functional mechanisms and effects. We try to summarize how layer- and cell type-specific neuromodulation may affect synaptic signaling in cortical microcircuits.
Collapse
Affiliation(s)
- Gabriele Radnikow
- Research Centre Jülich, Institute of Neuroscience and Medicine, INM-10, Jülich, Germany
| | - Dirk Feldmeyer
- Research Centre Jülich, Institute of Neuroscience and Medicine, INM-10, Jülich, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, Medical School, RWTH Aachen University, Aachen, Germany.,Jülich-Aachen Research Alliance - Translational Brain Medicine, Jülich, Germany
| |
Collapse
|
85
|
Zbukvic IC, Hyun Kim J. Divergent prefrontal dopaminergic mechanisms mediate drug- and fear-associated cue extinction during adolescence versus adulthood. Eur Neuropsychopharmacol 2018; 28:1-12. [PMID: 29174948 DOI: 10.1016/j.euroneuro.2017.11.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/08/2017] [Accepted: 11/03/2017] [Indexed: 01/21/2023]
Abstract
Cue-associated learning is vital to guiding behaviour for survival. Adolescence represents a key developmental stage for perturbations in cue-related learning, including a characteristic deficit in cue extinction learning. The present review summarizes evidence from animal and human literature that cue extinction is critically mediated by prefrontal dopamine, a system that undergoes dramatic reorganization during adolescence. We propose that extinction learning and memory is governed by a developmentally dynamic balance of dopamine receptors in the prefrontal cortex, which changes across adolescence into adulthood. This is contrary to the previous idea that extinction deficits during adolescence reflect inefficiency in the same neural circuitry as adults. This leads to proposal of the novel theory that cue extinction involves divergent prefrontal dopaminergic mechanisms depending on the age of extinction.
Collapse
Affiliation(s)
- Isabel C Zbukvic
- Black Dog Institute, Randwick 2031, NSW, Australia; Behavioral Neuroscience Division, The Florey Institute of Neuroscience & Mental Health, Parkville 3051, VIC, Australia.
| | - Jee Hyun Kim
- Behavioral Neuroscience Division, The Florey Institute of Neuroscience & Mental Health, Parkville 3051, VIC, Australia; The Florey Department of Neuroscience & Mental Health, University of Melbourne, Parkville 3010, VIC, Australia
| |
Collapse
|
86
|
van den Munkhof HE, Arnt J, Celada P, Artigas F. The antipsychotic drug brexpiprazole reverses phencyclidine-induced disruptions of thalamocortical networks. Eur Neuropsychopharmacol 2017; 27:1248-1257. [PMID: 29128144 DOI: 10.1016/j.euroneuro.2017.10.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 10/22/2017] [Indexed: 12/22/2022]
Abstract
Brexpiprazole (BREX), a recently approved antipsychotic drug in the US and Canada, improves cognitive dysfunction in animal models, by still largely unknown mechanisms. BREX is a partial agonist at 5-HT1A and D2 receptors and antagonist at α1B- and α2C-adrenergic and 5-HT2A receptors all with a similar potency. The NMDA receptor antagonist phencyclidine (PCP), used as pharmacological model of schizophrenia, activates thalamocortical networks and decreases low frequency oscillations (LFO; <4 Hz). These effects are reversed by antipsychotics. Here we assessed the ability of BREX to reverse PCP-induced hyperactivity of thalamocortical circuits, and the involvement of 5-HT1A receptors in its therapeutic action. BREX reversed PCP-induced neuronal activation at a lower dose in centromedial/mediodorsal thalamic nuclei (CM/MD; 0.5mg/kg) than in pyramidal medial prefrontal cortex neurons (mPFC, 2mg/kg), perhaps due to antagonism at α1B-adrenoceptors, abundantly expressed in the thalamus. Conversely, a cumulative 0.5 mg/kg dose reversed a PCP-induced LFO decrease in mPFC but not in CM/MD. BREX reduced LFO in both areas, yet with a different dose-response, and moderately excited mPFC neurons. The latter effect was reversed by the 5-HT1A receptor antagonist WAY-100635. Thus, BREX partly antagonizes PCP-induced thalamocortical hyperactivity, differentially in mPFC versus CM/MD. This regional selectivity may be related to the differential expression of α1B-, α2C-adrenergic and 5-HT2A receptors in both regions and/or different neuronal types. Furthermore, the pro-cognitive properties of BREX may be related to the 5-HT1A receptor-mediated increase in mPFC pyramidal neuron activity. Overall, the present data provide new insight on the brain elements involved in BREX's therapeutic actions.
Collapse
Affiliation(s)
- Hanna E van den Munkhof
- Department of Neurochemistry and Neuropharmacology, Institut d'Investigacions Biomèdiques de Barcelona, CSIC-IDIBAPS, Barcelona, Spain; CIBERSAM (Centro de Investigación Biomédica en Red de Salud Mental), Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jørn Arnt
- Lundbeck: Synaptic Transmission, Neuroscience Drug Discovery, H. Lundbeck A/S, Ottiliavej 9, DK-2500 Valby, Denmark; Sunred Pharma Consulting ApS, Svend Gonges Vej 11A, DK-2680 Solrod Strand, Denmark
| | - Pau Celada
- Department of Neurochemistry and Neuropharmacology, Institut d'Investigacions Biomèdiques de Barcelona, CSIC-IDIBAPS, Barcelona, Spain; CIBERSAM (Centro de Investigación Biomédica en Red de Salud Mental), Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Francesc Artigas
- Department of Neurochemistry and Neuropharmacology, Institut d'Investigacions Biomèdiques de Barcelona, CSIC-IDIBAPS, Barcelona, Spain; CIBERSAM (Centro de Investigación Biomédica en Red de Salud Mental), Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
87
|
Madsen HB, Guerin AA, Kim JH. Investigating the role of dopamine receptor- and parvalbumin-expressing cells in extinction of conditioned fear. Neurobiol Learn Mem 2017; 145:7-17. [DOI: 10.1016/j.nlm.2017.08.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 08/09/2017] [Accepted: 08/21/2017] [Indexed: 12/16/2022]
|
88
|
Bezu M, Maliković J, Kristofova M, Engidawork E, Höger H, Lubec G, Korz V. Spatial Working Memory in Male Rats: Pre-Experience and Task Dependent Roles of Dopamine D1- and D2-Like Receptors. Front Behav Neurosci 2017; 11:196. [PMID: 29081740 PMCID: PMC5645514 DOI: 10.3389/fnbeh.2017.00196] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 10/03/2017] [Indexed: 01/01/2023] Open
Abstract
The dopaminergic system is known to be involved in working memory processed by several brain regions like prefrontal cortex (PFC), hippocampus, striatum. In an earlier study we could show that Levodopa but not Modafinil enhanced working memory in a T-maze only during the early phase of training (day 3), whereas the later phase remained unaffected. Rats treated with a higher dose performed better than low dose treated rats. Here we could more specifically segregate the contributions of dopamine type 1- and 2- like receptors (D1R; D2R) to the training state dependent modulation of spatial working memory by intracerebroventricular (ICV) application of a D1R-like (SKF81297) and D2R-like agonist (Sumanirole) and antagonist (SCH23390, Remoxipride) at a low and high dose through 3 days of training. The D1R-like-agonist at both doses enhanced working memory at day 1 but only in the low dose treated rats enhancement persists over training compared to control rats. Rats treated with a high dose of a D1R-like-antagonist show persistent enhancement of working memory over training, whereas in low dose treated rats no statistical difference at any time point could be determined compared to controls. The D2R-like-agonist at both doses does not show an effect at any time point when compared to control animals, whereas the D2R-like antagonist at a low dose enhanced working memory at day 2. For the most effective D1R-like agonist, we repeated the experiments in a water maze working memory task, to test for task dependent differences in working memory modulations. Treated rats at both doses did not differ as compared to controls, but the temporal behavioral performance of all groups was different compared to T-maze trained rats. The results are in line with the view that spatial working memory is optimized within a limited range of dopaminergic transmission, however suggest that these ranges vary during spatial training.
Collapse
Affiliation(s)
- Mekite Bezu
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Jovana Maliković
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Martina Kristofova
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Ephrem Engidawork
- School of Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Harald Höger
- Core Unit of Biomedical Research, Division of Laboratory Animal Science and Genetics, Department of Biomedicine, Medical University of Vienna, Vienna, Austria
| | - Gert Lubec
- Paracelsus Medical University, Salzburg, Austria
| | - Volker Korz
- Brain Research Center, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
89
|
Santana N, Artigas F. Laminar and Cellular Distribution of Monoamine Receptors in Rat Medial Prefrontal Cortex. Front Neuroanat 2017; 11:87. [PMID: 29033796 PMCID: PMC5625028 DOI: 10.3389/fnana.2017.00087] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 09/15/2017] [Indexed: 01/03/2023] Open
Abstract
The prefrontal cortex (PFC) is deeply involved in higher brain functions, many of which are altered in psychiatric conditions. The PFC exerts a top-down control of most cortical and subcortical areas through descending pathways and is densely innervated by axons emerging from the brainstem monoamine cell groups, namely, the dorsal and median raphe nuclei (DR and MnR, respectively), the ventral tegmental area and the locus coeruleus (LC). In turn, the activity of these cell groups is tightly controlled by afferent pathways arising from layer V PFC pyramidal neurons. The reciprocal connectivity between PFC and monoamine cell groups is of interest to study the pathophysiology and treatment of severe psychiatric disorders, such as major depression and schizophrenia, inasmuch as antidepressant and antipsychotic drugs target monoamine receptors/transporters expressed in these areas. Here we review previous reports examining the presence of monoamine receptors in pyramidal and GABAergic neurons of the PFC using double in situ hybridization. Additionally, we present new data on the quantitative layer distribution (layers I, II-III, V, and VI) of monoamine receptor-expressing cells in the cingulate (Cg), prelimbic (PrL) and infralimbic (IL) subfields of the medial PFC (mPFC). The receptors examined include serotonin 5-HT1A, 5-HT2A, 5-HT2C, and 5-HT3, dopamine D1 and D2 receptors, and α1A-, α1B-, and α1D-adrenoceptors. With the exception of 5-HT3 receptors, selectively expressed by layers I-III GABA interneurons, the rest of monoamine receptors are widely expressed by pyramidal and GABAergic neurons in intermediate and deep layers of mPFC (5-HT2C receptors are also expressed in layer I). This complex distribution suggests that monoamines may modulate the communications between PFC and cortical/subcortical areas through the activation of receptors expressed by neurons in intermediate (e.g., 5-HT1A, 5-HT2A, α1D-adrenoceptors, dopamine D1 receptors) and deep layers (e.g., 5-HT1A, 5-HT2A, α1A-adrenoceptors, dopamine D2 receptors), respectively. Overall, these data provide a detailed framework to better understand the role of monoamines in the processing of cognitive and emotional signals by the PFC. Likewise, they may be helpful to characterize brain circuits relevant for the therapeutic action of antidepressant and antipsychotic drugs and to improve their therapeutic action, overcoming the limitations of current drugs.
Collapse
Affiliation(s)
- Noemí Santana
- Systems Neuropharmacology, Department of Neurochemistry and Neuropharmacology, Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental, Madrid, Spain
| | - Francesc Artigas
- Systems Neuropharmacology, Department of Neurochemistry and Neuropharmacology, Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental, Madrid, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| |
Collapse
|
90
|
Özkan M, Johnson NW, Sehirli US, Woodhall GL, Stanford IM. Dopamine acting at D1-like, D2-like and α1-adrenergic receptors differentially modulates theta and gamma oscillatory activity in primary motor cortex. PLoS One 2017; 12:e0181633. [PMID: 28732063 PMCID: PMC5521821 DOI: 10.1371/journal.pone.0181633] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/04/2017] [Indexed: 11/30/2022] Open
Abstract
The loss of dopamine (DA) in Parkinson’s is accompanied by the emergence of exaggerated theta and beta frequency neuronal oscillatory activity in the primary motor cortex (M1) and basal ganglia. DA replacement therapy or deep brain stimulation reduces the power of these oscillations and this is coincident with an improvement in motor performance implying a causal relationship. Here we provide in vitro evidence for the differential modulation of theta and gamma activity in M1 by DA acting at receptors exhibiting conventional and non-conventional DA pharmacology. Recording local field potentials in deep layer V of rat M1, co-application of carbachol (CCh, 5 μM) and kainic acid (KA, 150 nM) elicited simultaneous oscillations at a frequency of 6.49 ± 0.18 Hz (theta, n = 84) and 34.97 ± 0.39 Hz (gamma, n = 84). Bath application of DA resulted in a decrease in gamma power with no change in theta power. However, application of either the D1-like receptor agonist SKF38393 or the D2-like agonist quinpirole increased the power of both theta and gamma suggesting that the DA-mediated inhibition of oscillatory power is by action at other sites other than classical DA receptors. Application of amphetamine, which promotes endogenous amine neurotransmitter release, or the adrenergic α1-selective agonist phenylephrine mimicked the action of DA and reduced gamma power, a result unaffected by prior co-application of D1 and D2 receptor antagonists SCH23390 and sulpiride. Finally, application of the α1-adrenergic receptor antagonist prazosin blocked the action of DA on gamma power suggestive of interaction between α1 and DA receptors. These results show that DA mediates complex actions acting at dopamine D1-like and D2-like receptors, α1 adrenergic receptors and possibly DA/α1 heteromultimeric receptors to differentially modulate theta and gamma activity in M1.
Collapse
Affiliation(s)
- Mazhar Özkan
- Aston Brain Centre, Aston University, School of Life and Health Sciences, Birmingham, United Kingdom
- Department of Anatomy, School of Medicine, Marmara University, Istanbul, Turkey
| | - Nicholas W. Johnson
- Aston Brain Centre, Aston University, School of Life and Health Sciences, Birmingham, United Kingdom
| | - Umit S. Sehirli
- Department of Anatomy, School of Medicine, Marmara University, Istanbul, Turkey
| | - Gavin L. Woodhall
- Aston Brain Centre, Aston University, School of Life and Health Sciences, Birmingham, United Kingdom
| | - Ian M. Stanford
- Aston Brain Centre, Aston University, School of Life and Health Sciences, Birmingham, United Kingdom
- * E-mail:
| |
Collapse
|
91
|
Jenni NL, Larkin JD, Floresco SB. Prefrontal Dopamine D 1 and D 2 Receptors Regulate Dissociable Aspects of Decision Making via Distinct Ventral Striatal and Amygdalar Circuits. J Neurosci 2017; 37:6200-6213. [PMID: 28546312 PMCID: PMC6705698 DOI: 10.1523/jneurosci.0030-17.2017] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 05/15/2017] [Accepted: 05/18/2017] [Indexed: 01/04/2023] Open
Abstract
Mesocortical dopamine (DA) regulates a variety of cognitive functions via actions on D1 and/or D2 receptors. For example, risk/reward decision making is modulated differentially by these two receptors within the prefrontal cortex (PFC), with D2 receptors enabling flexible decision making and D1 receptors promoting persistence in choice biases. However, it is unclear how DA mediates opposing patterns of behavior by acting on different receptors within the same terminal region. We explored the possibility that DA may act on separate networks of PFC neurons that are modulated by D1 or D2 receptors and in turn interface with divergent downstream structures such as the basolateral amygdala (BLA) or nucleus accumbens (NAc). Decision making was assessed using a probabilistic discounting task in which well trained male rats chose between small/certain or large/risky rewards, with the odds of obtaining the larger reward changing systematically within a session. Selective disruption of D1 or D2 modulation of separate PFC output pathways was achieved using unilateral intra-PFC infusions of DA antagonists combined with contralateral inactivation of the BLA or NAc. Disrupting D2 (but not D1) modulation of PFC→BLA circuitry impaired adjustments in decision biases in response to changes in reward probabilities. In contrast, disrupting D1 modulation of PFC→NAc networks reduced risky choice, attenuating reward sensitivity and increasing sensitivity to reward omissions. These findings reveal that mesocortical DA can facilitate dissociable components of reward seeking and action selection by acting on different functional networks of PFC neurons that can be distinguished by the subcortical projection targets with which they interface.SIGNIFICANCE STATEMENT Prefrontal cortical dopamine regulates a variety of executive functions governed by the frontal lobes via actions on D1 and D2 receptors. These receptors can in some instances mediate different patterns of behavior, but the mechanisms underlying these dissociable actions are unclear. Using a selective disconnection approach, we reveal that D1 and D2 receptors can facilitate diverse aspects of decision making by acting on separate networks of prefrontal neurons that interface with distinct striatal or amygdalar targets. These findings reveal an additional level of complexity in how mesocortical DA regulates different forms of cognition via actions on different receptors, highlighting how it may act upon distinct cortical microcircuits to drive different patterns of behavior.
Collapse
Affiliation(s)
- Nicole L Jenni
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Joshua D Larkin
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Stan B Floresco
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| |
Collapse
|
92
|
Lupinsky D, Moquin L, Gratton A. Interhemispheric regulation of the rat medial prefrontal cortical glutamate stress response: role of local GABA- and dopamine-sensitive mechanisms. Psychopharmacology (Berl) 2017; 234:353-363. [PMID: 27822602 DOI: 10.1007/s00213-016-4468-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 10/21/2016] [Indexed: 11/26/2022]
Abstract
RATIONALE We previously reported that stressors increase medial prefrontal cortex (PFC) glutamate (GLU) levels as a result of activating callosal neurons located in the opposite hemisphere and that this PFC GLU stress response is regulated by GLU-, dopamine- (DA-), and GABA-sensitive mechanisms (Lupinsky et al. 2010). OBJECTIVES Here, we examine the possibility that PFC DA regulates the stress responsivity of callosal neurons indirectly by acting at D1 and D2 receptors located on GABA interneurons. METHODS Microdialysis combined with drug perfusion (reverse dialysis) or microinjections was used in adult male Long-Evans rats to characterize D1, D2, and GABAB receptor-mediated regulation of the PFC GABA response to tail-pinch (TP) stress. RESULTS We report that TP stress reliably elicited comparable increases in extracellular GABA in the left and right PFCs. SCH23390 (D1 antagonist; 100 μM perfusate concentration) perfused by reverse microdialysis attenuated the local GABA stress responses equally in the left and right PFCs. Intra-PFC raclopride perfusion (D2 antagonist; 100 μM) had the opposite effect, not only potentiating the local GABA stress response but also causing a transient elevation in basal (pre-stress) GABA. Moreover, unilateral PFC raclopride microinjection (6 nmol) attenuated the GLU response to TP stress in the contralateral PFC. Finally, intra-PFC baclofen perfusion (GABAB agonist; 100 μM) inhibited the local GLU and GABA stress responses. CONCLUSIONS Taken together, these findings implicate PFC GABA interneurons in processing stressful stimuli, showing that local D1, D2, and GABAB receptor-mediated changes in PFC GABA transmission play a crucial role in the interhemispheric regulation of GLU stress responsivity.
Collapse
Affiliation(s)
- Derek Lupinsky
- Department of Psychiatry, McGill University, Montréal, Québec, H4H 1R3, Canada.
- Douglas Institute Research Center, 6875 LaSalle Blvd, Montréal, Québec, H4H 1R3, Canada.
| | - Luc Moquin
- Douglas Institute Research Center, 6875 LaSalle Blvd, Montréal, Québec, H4H 1R3, Canada
| | - Alain Gratton
- Department of Psychiatry, McGill University, Montréal, Québec, H4H 1R3, Canada
- Douglas Institute Research Center, 6875 LaSalle Blvd, Montréal, Québec, H4H 1R3, Canada
| |
Collapse
|
93
|
Meunier CNJ, Chameau P, Fossier PM. Modulation of Synaptic Plasticity in the Cortex Needs to Understand All the Players. Front Synaptic Neurosci 2017; 9:2. [PMID: 28203201 PMCID: PMC5285384 DOI: 10.3389/fnsyn.2017.00002] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/13/2017] [Indexed: 12/19/2022] Open
Abstract
The prefrontal cortex (PFC) is involved in cognitive tasks such as working memory, decision making, risk assessment and regulation of attention. These functions performed by the PFC are supposed to rely on rhythmic electrical activity generated by neuronal network oscillations determined by a precise balance between excitation and inhibition balance (E/I balance) resulting from the coordinated activities of recurrent excitation and feedback and feedforward inhibition. Functional alterations in PFC functions have been associated with cognitive deficits in several pathologies such as major depression, anxiety and schizophrenia. These pathological situations are correlated with alterations of different neurotransmitter systems (i.e., serotonin (5-HT), dopamine (DA), acetylcholine…) that result in alterations of the E/I balance. The aim of this review article is to cover the basic aspects of the regulation of the E/I balance as well as to highlight the importance of the complementarity role of several neurotransmitters in the modulation of the plasticity of excitatory and inhibitory synapses. We illustrate our purpose by recent findings that demonstrate that 5-HT and DA cooperate to regulate the plasticity of excitatory and inhibitory synapses targeting layer 5 pyramidal neurons (L5PyNs) of the PFC and to fine tune the E/I balance. Using a method based on the decomposition of the synaptic conductance into its excitatory and inhibitory components, we show that concomitant activation of D1-like receptors (D1Rs) and 5-HT1ARs, through a modulation of NMDA receptors, favors long term potentiation (LTP) of both excitation and inhibition and consequently does not modify the E/I balance. We also demonstrate that activation of D2-receptors requires functional 5-HT1ARs to shift the E-I balance towards more inhibition and to favor long term depression (LTD) of excitatory synapses through the activation of glycogen synthase kinase 3β (GSK3β). This cooperation between different neurotransmitters is particularly relevant in view of pathological situations in which alterations of one neurotransmitter system will also have consequences on the regulation of synaptic efficacy by other neurotransmitters. This opens up new perspectives in the development of therapeutic strategies for the pharmacological treatment of neuronal disorders.
Collapse
Affiliation(s)
- Claire N J Meunier
- Institut de Neurosciences Paris-Saclay (NeuroPSI), UMR 91197 CNRS-Université Paris-Saclay Paris, France
| | - Pascal Chameau
- Swammerdam Institute for Life Sciences, Center for NeuroScience, University of Amsterdam Amsterdam, Netherlands
| | - Philippe M Fossier
- Institut de Neurosciences Paris-Saclay (NeuroPSI), UMR 91197 CNRS-Université Paris-Saclay Paris, France
| |
Collapse
|
94
|
Abstract
Hallucinogens evoke sensory, perceptual, affective, and cognitive effects that may be useful to understand the neurobiological basis of mood and psychotic disorders. The present chapter reviews preclinical research carried out in recent years in order to better understand the action of psychotomimetic agents such as the noncompetitive NMDA receptor (NMDA-R) antagonists and serotonergic hallucinogens. Our studies have focused on the mechanisms through which these agents alter cortical activity. Noncompetitive NMDA-R antagonists, such as phencyclidine (PCP) and MK-801 (dizocilpine), as well as the serotonergic hallucinogens DOI and 5-MeO-DMT, produce similar effects on cellular and population activity in prefrontal cortex (PFC); these effects include alterations of pyramidal neuron discharge (with an overall increase in firing), as well as a marked attenuation of the low frequency oscillations (0.2-4 Hz) to which neuronal discharge is coupled in anesthetized rodents. PCP increases c-fos expression in excitatory neurons from various cortical and subcortical areas, particularly the thalamus. This effect of PCP involves the preferential blockade of NMDA-R on GABAergic neurons of the reticular nucleus of the thalamus, which provides feedforward inhibition to the rest of thalamic nuclei. It is still unknown whether serotonergic hallucinogens also affect thalamocortical networks. However, when examined, similar alterations in other cortical areas, such as the primary visual cortex (V1), have been observed, suggesting that these agents affect cortical activity in sensory and associative areas. Interestingly, the disruption of PFC activity induced by PCP, DOI and 5-MeO-DMT is reversed by classical and atypical antipsychotic drugs. This effect suggests a possible link between the mechanisms underlying the disruption of perception by multiple classes of hallucinogenic agents and the therapeutic efficacy of antipsychotic agents.
Collapse
|
95
|
Distinct cortical and striatal actions of a β-arrestin-biased dopamine D2 receptor ligand reveal unique antipsychotic-like properties. Proc Natl Acad Sci U S A 2016; 113:E8178-E8186. [PMID: 27911814 DOI: 10.1073/pnas.1614347113] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The current dopamine (DA) hypothesis of schizophrenia postulates striatal hyperdopaminergia and cortical hypodopaminergia. Although partial agonists at DA D2 receptors (D2Rs), like aripiprazole, were developed to simultaneously target both phenomena, they do not effectively improve cortical dysfunction. In this study, we investigate the potential for newly developed β-arrestin2 (βarr2)-biased D2R partial agonists to simultaneously target hyper- and hypodopaminergia. Using neuron-specific βarr2-KO mice, we show that the antipsychotic-like effects of a βarr2-biased D2R ligand are driven through both striatal antagonism and cortical agonism of D2R-βarr2 signaling. Furthermore, βarr2-biased D2R agonism enhances firing of cortical fast-spiking interneurons. This enhanced cortical agonism of the biased ligand can be attributed to a lack of G-protein signaling and elevated expression of βarr2 and G protein-coupled receptor (GPCR) kinase 2 in the cortex versus the striatum. Therefore, we propose that βarr2-biased D2R ligands that exert region-selective actions could provide a path to develop more effective antipsychotic therapies.
Collapse
|
96
|
Lack of GSK3β activation and modulation of synaptic plasticity by dopamine in 5-HT1A-receptor KO mice. Neuropharmacology 2016; 113:124-136. [PMID: 27678414 DOI: 10.1016/j.neuropharm.2016.09.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 09/13/2016] [Accepted: 09/23/2016] [Indexed: 12/31/2022]
Abstract
Psychiatric disorders are associated with excitation-inhibition (E-I) balance impairment in the prefrontal cortex. However, how the E-I balance is regulated is poorly known. The E-I balance of neuronal networks is linked to the action of numerous neuromodulators such as dopamine and 5-HT. We investigated the role of D2-receptors in tuning the E-I balance in a mouse model of anxiety, the 5-HT1A-receptor KO mice. We focused on synaptic plasticity of excitation and inhibition on layer 5 pyramidal neurons. We show that D2-receptor activation decreases the excitation and favors HFS-induced LTD of excitatory synapses via the activation of GSK3β. This effect is absent in 5-HT1A-receptor KO mice. Our data show that the fine control of excitatory transmission by GSK3β requires recruitment of D2-receptors and depends on the presence of 5-HT1A-receptors. In psychiatric disorders in which the number of 5-HT1A-receptors decreased, therapies should reconsider how serotonin and dopamine receptors interact and control neuronal network activity.
Collapse
|
97
|
Santana N, Artigas F. Expression of Serotonin2CReceptors in Pyramidal and GABAergic Neurons of Rat Prefrontal Cortex: A Comparison with Striatum. Cereb Cortex 2016; 27:3125-3139. [DOI: 10.1093/cercor/bhw148] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
98
|
Zbukvic IC, Ganella DE, Perry CJ, Madsen HB, Bye CR, Lawrence AJ, Kim JH. Role of Dopamine 2 Receptor in Impaired Drug-Cue Extinction in Adolescent Rats. Cereb Cortex 2016; 26:2895-904. [PMID: 26946126 PMCID: PMC4869820 DOI: 10.1093/cercor/bhw051] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Adolescent drug users display resistance to treatment such as cue exposure therapy (CET), as well as increased liability to relapse. The basis of CET is extinction learning, which involves dopamine signaling in the medial prefrontal cortex (mPFC). This system undergoes dramatic alterations during adolescence. Therefore, we investigated extinction of a cocaine-associated cue in adolescent and adult rats. While cocaine self-administration and lever-alone extinction were not different between the two ages, we observed that cue extinction reduced cue-induced reinstatement in adult but not adolescent rats. Infusion of the selective dopamine 2 receptor (D2R)-like agonist quinpirole into the infralimbic cortex (IL) of the mPFC prior to cue extinction significantly reduced cue-induced reinstatement in adolescents. This effect was replicated by acute systemic treatment with the atypical antipsychotic aripiprazole (Abilify), a partial D2R-like agonist. These data suggest that adolescents may be more susceptible to relapse due to a deficit in cue extinction learning, and highlight the significance of D2R signaling in the IL for cue extinction during adolescence. These findings inspire new tactics for improving adolescent CET, with aripiprazole representing an exciting potential pharmacological adjunct for behavioral therapy.
Collapse
Affiliation(s)
- Isabel C. Zbukvic
- Behavioral Neuroscience Division, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3051, Australia
- The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Despina E. Ganella
- Behavioral Neuroscience Division, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3051, Australia
- The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Christina J. Perry
- Behavioral Neuroscience Division, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3051, Australia
- The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Heather B. Madsen
- Behavioral Neuroscience Division, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3051, Australia
- The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Christopher R. Bye
- Behavioral Neuroscience Division, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3051, Australia
- The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Andrew J. Lawrence
- Behavioral Neuroscience Division, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3051, Australia
- The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Jee Hyun Kim
- Behavioral Neuroscience Division, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3051, Australia
- The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
99
|
Caballero A, Granberg R, Tseng KY. Mechanisms contributing to prefrontal cortex maturation during adolescence. Neurosci Biobehav Rev 2016; 70:4-12. [PMID: 27235076 DOI: 10.1016/j.neubiorev.2016.05.013] [Citation(s) in RCA: 201] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/17/2016] [Accepted: 05/18/2016] [Indexed: 12/17/2022]
Abstract
Adolescence is defined as a transitional period between childhood and adulthood characterized by changes in social interaction and acquisition of mature cognitive abilities. These changes have been associated with the maturation of brain regions involved in the control of motivation, emotion, and cognition. Among these regions, the protracted development of the human prefrontal cortex during adolescence has been proposed to underlie the maturation of cognitive functions and the regulation of affective responses. Studies in animal models allow us to test the causal contribution of specific neural processes in the development of the prefrontal cortex and the acquisition of adult behavior. This review summarizes the cellular and synaptic mechanisms occurring in the rodent prefrontal cortex during adolescence as a model for understanding the changes underlying human prefrontal development.
Collapse
Affiliation(s)
- Adriana Caballero
- Department of Cellular & Molecular Pharmacology, The Chicago Medical School at Rosalind Franklin University of Medicine & Science, North Chicago, IL 60064, USA
| | - Rachel Granberg
- Department of Cellular & Molecular Pharmacology, The Chicago Medical School at Rosalind Franklin University of Medicine & Science, North Chicago, IL 60064, USA
| | - Kuei Y Tseng
- Department of Cellular & Molecular Pharmacology, The Chicago Medical School at Rosalind Franklin University of Medicine & Science, North Chicago, IL 60064, USA.
| |
Collapse
|
100
|
Effects of Chronic Dopamine D2R Agonist Treatment and Polysialic Acid Depletion on Dendritic Spine Density and Excitatory Neurotransmission in the mPFC of Adult Rats. Neural Plast 2016; 2016:1615363. [PMID: 27110404 PMCID: PMC4821975 DOI: 10.1155/2016/1615363] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 02/25/2016] [Indexed: 12/23/2022] Open
Abstract
Dopamine D2 receptors (D2R) in the medial prefrontal cortex (mPFC) are key players in the etiology and therapeutics of schizophrenia. The overactivation of these receptors contributes to mPFC dysfunction. Chronic treatment with D2R agonists modifies the expression of molecules implicated in neuronal structural plasticity, synaptic function, and inhibitory neurotransmission, which are also altered in schizophrenia. These changes are dependent on the expression of the polysialylated form of the neural cell adhesion molecule (PSA-NCAM), a plasticity-related molecule, but nothing is known about the effects of D2R and PSA-NCAM on excitatory neurotransmission and the structure of mPFC pyramidal neurons, two additional features affected in schizophrenia. To evaluate these parameters, we have chronically treated adult rats with PPHT (a D2R agonist) after enzymatic removal of PSA with Endo-N. Both treatments decreased spine density in apical dendrites of pyramidal neurons without affecting their inhibitory innervation. Endo-N also reduced the expression of vesicular glutamate transporter-1. These results indicate that D2R and PSA-NCAM are important players in the regulation of the structural plasticity of mPFC excitatory neurons. This is relevant to our understanding of the neurobiological basis of schizophrenia, in which structural alterations of pyramidal neurons and altered expression of D2R and PSA-NCAM have been found.
Collapse
|