1
|
Makino Y, Hodgson NW, Doenier E, Serbin AV, Osada K, Artoni P, Dickey M, Sullivan B, Potter-Dickey A, Komanchuk J, Sekhon B, Letourneau N, Ryan ND, Trauth J, Cameron JL, Hensch TK. Sleep-sensitive dopamine receptor expression in male mice underlies attention deficits after a critical period of early adversity. Sci Transl Med 2024; 16:eadh9763. [PMID: 39383245 DOI: 10.1126/scitranslmed.adh9763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/03/2024] [Accepted: 09/13/2024] [Indexed: 10/11/2024]
Abstract
Early life stress (ELS) yields cognitive impairments of unknown molecular and physiological origin. We found that fragmented maternal care of mice during a neonatal critical period from postnatal days P2-9 elevated dopamine receptor D2R and suppressed D4R expression, specifically within the anterior cingulate cortex (ACC) in only the male offspring. This was associated with poor performance on a two-choice visual attention task, which was acutely rescued in adulthood by local or systemic pharmacological rebalancing of D2R/D4R activity. Furthermore, ELS male mice demonstrated heightened hypothalamic orexin and persistently disrupted sleep. Given that acute sleep deprivation in normally reared male mice mimicked the ACC dopamine receptor subtype modulation and disrupted attention of ELS mice, sleep loss likely underlies cognitive deficits in ELS mice. Likewise, sleep impairment mediated the attention deficits associated with early adversity in human children, as demonstrated by path analysis on data collected with multiple questionnaires for a large child cohort. A deeper understanding of the sex-specific cognitive consequences of ELS thus has the potential to reveal therapeutic strategies for overcoming them.
Collapse
Affiliation(s)
- Yuichi Makino
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
- International Research Center for Neurointelligence, UTIAS, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Nathaniel W Hodgson
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Emma Doenier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Anna Victoria Serbin
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Koya Osada
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Pietro Artoni
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Matthew Dickey
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Breanna Sullivan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Jelena Komanchuk
- School of Nursing, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Bikram Sekhon
- School of Nursing, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Nicole Letourneau
- School of Nursing, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Neal D Ryan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jeanette Trauth
- Department of Behavioral and Community Health Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Judy L Cameron
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Takao K Hensch
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
- International Research Center for Neurointelligence, UTIAS, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| |
Collapse
|
2
|
Homberg JR, Brivio P, Greven CU, Calabrese F. Individuals being high in their sensitivity to the environment: Are sensitive period changes in play? Neurosci Biobehav Rev 2024; 159:105605. [PMID: 38417743 DOI: 10.1016/j.neubiorev.2024.105605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/13/2024] [Accepted: 02/25/2024] [Indexed: 03/01/2024]
Abstract
All individuals on planet earth are sensitive to the environment, but some more than others. These individual differences in sensitivity to environments are seen across many animal species including humans, and can influence personalities as well as vulnerability and resilience to mental disorders. Yet, little is known about the underlying brain mechanisms. Key genes that contribute to individual differences in environmental sensitivity are the serotonin transporter, dopamine D4 receptor and brain-derived neurotrophic factor genes. By synthesizing neurodevelopmental findings of these genetic factors, and discussing them through the lens of mechanisms related to sensitive periods, which are phases of heightened neuronal plasticity during which a certain network is being finetuned by experiences, we propose that these genetic factors delay but extend postnatal sensitive periods. This may explain why sensitive individuals show behavioral features that are characteristic of a young brain state at the level of sensory information processing, such as reduced filtering or blockade of irrelevant information, resulting in a sensory processing system that 'keeps all options open'.
Collapse
Affiliation(s)
- Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Paola Brivio
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Corina U Greven
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands; Karakter Child and Adolescent Psychiatry University Center, Nijmegen, the Netherlands; King's College London, Institute of Psychiatry, Psychology and Neuroscience, Social, Genetic and Developmental Psychiatry Center, London, United Kingdom
| | - Francesca Calabrese
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
3
|
Hirato Y, Seiriki K, Kojima L, Yamada S, Rokujo H, Takemoto T, Nakazawa T, Kasai A, Hashimoto H. Clozapine Induces Neuronal Activation in the Medial Prefrontal Cortex in a Projection Target-Biased Manner. Biol Pharm Bull 2024; 47:478-485. [PMID: 38382927 DOI: 10.1248/bpb.b23-00898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
The medial prefrontal cortex (mPFC) is associated with various behavioral controls via diverse projections to cortical and subcortical areas of the brain. Dysfunctions and modulations of this circuitry are related to the pathophysiology of schizophrenia and its pharmacotherapy, respectively. Clozapine is an atypical antipsychotic drug used for treatment-resistant schizophrenia and is known to modulate neuronal activity in the mPFC. However, it remains unclear which prefrontal cortical projections are activated by clozapine among the various projection targets. To identify the anatomical characteristics of neurons activated by clozapine at the mesoscale level, we investigated the brain-wide projection patterns of neurons with clozapine-induced c-Fos expression in the mPFC. Using a whole-brain imaging and virus-mediated genetic tagging of activated neurons, we found that clozapine-responsive neurons in the mPFC had a wide range of projections to the mesolimbic, amygdala and thalamic areas, especially the mediodorsal thalamus. These results may provide key insights into the neuronal basis of the therapeutic action of clozapine.
Collapse
Affiliation(s)
- Yumi Hirato
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Kaoru Seiriki
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Leo Kojima
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Shohei Yamada
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Hiroki Rokujo
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Tomoya Takemoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Takanobu Nakazawa
- Department of Bioscience, Graduate School of Life Sciences, Tokyo University of Agriculture
| | - Atsushi Kasai
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University
- Systems Brain Science Project, Drug Innovation Center, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University, and University of Fukui
- Institute for Datability Science, Osaka University
- Department of Molecular Pharmaceutical Sciences, Graduate School of Medicine, Osaka University
| |
Collapse
|
4
|
Zhong P, Cao Q, Yan Z. Distinct and Convergent Alterations of Entorhinal Cortical Circuits in Two Mouse Models for Alzheimer's Disease and Related Disorders. J Alzheimers Dis 2024; 98:1121-1131. [PMID: 38489190 PMCID: PMC11432142 DOI: 10.3233/jad-231413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Background The impairment of neural circuits controlling cognitive processes has been implicated in the pathophysiology of Alzheimer's disease and related disorders (ADRD). However, it is largely unclear what circuits are specifically changed in ADRD, particularly at the early stage. Objective Our goal of this study is to reveal the functional changes in the circuit of entorhinal cortex (EC), an interface between neocortex and hippocampus, in AD. Methods Electrophysiological, optogenetic and chemogenetic approaches were used to examine and manipulate entorhinal cortical circuits in amyloid-β familial AD model (5×FAD) and tauopathy model (P301S Tau). Results We found that, compared to wild-type mice, electrical stimulation of EC induced markedly smaller responses in subiculum (hippocampal output) of 5×FAD mice (6-month-old), suggesting that synaptic communication in the EC to subiculum circuit is specifically blocked in this AD model. In addition, optogenetic stimulation of glutamatergic terminals from prefrontal cortex (PFC) induced smaller responses in EC of 5×FAD and P301S Tau mice (6-month-old), suggesting that synaptic communication in the PFC to EC pathway is compromised in both ADRD models. Chemogenetic activation of PFC to EC pathway did not affect the bursting activity of EC neurons in 5×FAD mice, but partially restored the diminished EC neuronal activity in P301S Tau mice. Conclusions These data suggest that 5×FAD mice has a specific impairment of short-range hippocampal gateway (EC to subiculum), which may be caused by amyloid-β deposits; while two ADRD models have a common impairment of long-range cortical to hippocampal circuit (PFC to EC), which may be caused by microtubule/tau-based transport deficits. These circuit deficits provide a pathophysiological basis for unique and common impairments of various cognitive processes in ADRD conditions.
Collapse
Affiliation(s)
- Ping Zhong
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Qing Cao
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| |
Collapse
|
5
|
Ott T, Stein AM, Nieder A. Dopamine receptor activation regulates reward expectancy signals during cognitive control in primate prefrontal neurons. Nat Commun 2023; 14:7537. [PMID: 37985776 PMCID: PMC10661983 DOI: 10.1038/s41467-023-43271-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 11/06/2023] [Indexed: 11/22/2023] Open
Abstract
Dopamine neurons respond to reward-predicting cues but also modulate information processing in the prefrontal cortex essential for cognitive control. Whether dopamine controls reward expectation signals in prefrontal cortex that motivate cognitive control is unknown. We trained two male macaques on a working memory task while varying the reward size earned for successful task completion. We recorded neurons in lateral prefrontal cortex while simultaneously stimulating dopamine D1 receptor (D1R) or D2 receptor (D2R) families using micro-iontophoresis. We show that many neurons predict reward size throughout the trial. D1R stimulation showed mixed effects following reward cues but decreased reward expectancy coding during the memory delay. By contrast, D2R stimulation increased reward expectancy coding in multiple task periods, including cueing and memory periods. Stimulation of either dopamine receptors increased the neurons' selective responses to reward size upon reward delivery. The differential modulation of reward expectancy by dopamine receptors suggests that dopamine regulates reward expectancy necessary for successful cognitive control.
Collapse
Affiliation(s)
- Torben Ott
- Animal Physiology, Institute of Neurobiology, Auf der Morgenstelle 28, University of Tübingen, 72076, Tübingen, Germany.
- Bernstein Center for Computational Neuroscience and Institute of Biology, Humboldt-University of Berlin, 10099, Berlin, Germany.
| | - Anna Marlina Stein
- Animal Physiology, Institute of Neurobiology, Auf der Morgenstelle 28, University of Tübingen, 72076, Tübingen, Germany
| | - Andreas Nieder
- Animal Physiology, Institute of Neurobiology, Auf der Morgenstelle 28, University of Tübingen, 72076, Tübingen, Germany.
| |
Collapse
|
6
|
Wang W, Tan T, Cao Q, Zhang F, Rein B, Duan WM, Yan Z. Histone Deacetylase Inhibition Restores Behavioral and Synaptic Function in a Mouse Model of 16p11.2 Deletion. Int J Neuropsychopharmacol 2022; 25:877-889. [PMID: 35907244 PMCID: PMC9593221 DOI: 10.1093/ijnp/pyac048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/21/2022] [Accepted: 07/27/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Microdeletion of the human 16p11.2 gene locus confers risk for autism spectrum disorders and intellectual disability. How 16p11.2 deletion is linked to these neurodevelopmental disorders and whether there are treatment avenues for the manifested phenotypes remain to be elucidated. Emerging evidence suggests that epigenetic aberrations are strongly implicated in autism. METHODS We performed behavioral and electrophysiological experiments to examine the therapeutic effects of epigenetic drugs in transgenic mice carrying 16p11.2 deletion (16p11del/+). RESULTS We found that 16p11del/+ mice exhibited a significantly reduced level of histone acetylation in the prefrontal cortex (PFC). A short (3-day) treatment with class I histone deacetylase (HDAC) inhibitor MS-275 or Romidepsin led to the prolonged (3-4 weeks) rescue of social and cognitive deficits in 16p11del/+ mice. Concomitantly, MS-275 treatment reversed the hypoactivity of PFC pyramidal neurons and the hyperactivity of PFC fast-spiking interneurons. Moreover, the diminished N-methyl-D-aspartate (NMDA) receptor-mediated synaptic currents and the elevated GABAA receptor-mediated synaptic currents in PFC pyramidal neurons of 16p11del/+ mice were restored to control levels by MS-275 treatment. CONCLUSIONS Our results suggest that HDAC inhibition provides a highly effective therapeutic strategy for behavioral deficits and excitation/inhibition imbalance in 16p11del/+ mice, likely via normalization of synaptic function in the PFC.
Collapse
Affiliation(s)
- Wei Wang
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Tao Tan
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Qing Cao
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Freddy Zhang
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Benjamin Rein
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Wei-Ming Duan
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
7
|
Thörn CW, Kafetzopoulos V, Kocsis B. Differential Effect of Dopamine D4 Receptor Activation on Low-Frequency Oscillations in the Prefrontal Cortex and Hippocampus May Bias the Bidirectional Prefrontal–Hippocampal Coupling. Int J Mol Sci 2022; 23:ijms231911705. [PMID: 36233007 PMCID: PMC9569525 DOI: 10.3390/ijms231911705] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/18/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
Dopamine D4 receptor (D4R) mechanisms are implicated in psychiatric diseases characterized by cognitive deficits, including schizophrenia, ADHD, and autism. The cellular mechanisms are poorly understood, but impaired neuronal synchronization in cortical networks was proposed to contribute to these deficits. In animal experiments, D4R activation was shown to generate aberrant increased gamma oscillations and to reduce performance on cognitive tasks requiring functional prefrontal cortex (PFC) and hippocampus (HPC) networks. While fast oscillations in the gamma range are important for local synchronization within neuronal ensembles, long-range synchronization between distant structures is achieved by slow rhythms in the delta, theta, alpha ranges. The characteristics of slow oscillations vary between structures during cognitive tasks. HPC activity is dominated by theta rhythm, whereas PFC generates unique oscillations in the 2–4 Hz range. In order to investigate the role of D4R on slow rhythms, cortical activity was recorded in rats under urethane anesthesia in which slow oscillations can be elicited in a controlled manner without behavioral confounds, by electrical stimulation of the brainstem reticular formation. The local field potential segments during stimulations were extracted and subjected to fast Fourier transform to obtain power density spectra. The selective D4R agonist A-412997 (5 and 10 mg/kg) and antagonists L-745870 (5 and 10 mg/kg) were injected systemically and the peak power in the two frequency ranges were compared before and after the injection. We found that D4R compounds significantly changed the activity of both HPC and PFC, but the direction of the effect was opposite in the two structures. D4R agonist enhanced PFC slow rhythm (delta, 2–4 Hz) and suppressed HPC theta, whereas the antagonist had an opposite effect. Analogous changes of the two slow rhythms were also found in the thalamic nucleus reuniens, which has connections to both forebrain structures. Slow oscillations play a key role in interregional cortical coupling; delta and theta oscillations were shown in particular, to entrain neuronal firing and to modulate gamma activity in interconnected forebrain structures with a relative HPC theta dominance over PFC. Thus, the results of this study indicate that D4R activation may introduce an abnormal bias in the bidirectional PFC–HPC coupling which can be reversed by D4R antagonists.
Collapse
Affiliation(s)
| | - Vasilios Kafetzopoulos
- Department Psychiatry at BIDMC, Harvard Medical School, Boston, MA 02215, USA
- Department of Psychiatry, Medical School, University of Ioannina, 45110 Ioannina, Greece
| | - Bernat Kocsis
- Department Psychiatry at BIDMC, Harvard Medical School, Boston, MA 02215, USA
- Correspondence: ; Tel.: +617-331-1782
| |
Collapse
|
8
|
Zhong P, Cao Q, Yan Z. Selective impairment of circuits between prefrontal cortex glutamatergic neurons and basal forebrain cholinergic neurons in a tauopathy mouse model. Cereb Cortex 2022; 32:5569-5579. [PMID: 35235649 PMCID: PMC9753040 DOI: 10.1093/cercor/bhac036] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 01/25/2023] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder linked to cognitive decline. To understand how specific neuronal circuits are impaired in AD, we have used optogenetic and electrophysiological approaches to reveal the functional changes between prefrontal cortex (PFC) and basal forebrain (BF), 2 key regions controlling cognitive processes, in a tauopathy mouse model. We found that the glutamatergic synaptic responses in BF cholinergic neurons from P301S Tau mice (6-8 months old) were markedly diminished. The attenuated long-range PFC to BF pathway in the AD model significantly increased the failure rate of action potential firing of BF cholinergic neurons triggered by optogenetic stimulations of glutamatergic terminals from PFC. In contrast, the projection from PFC to other regions, such as amygdala and striatum, was largely unaltered. On the other hand, optogenetic stimulation of cholinergic terminals from BF induced a persistent reduction of the excitability of PFC pyramidal neurons from Tau mice, instead of the transient reduction exhibited in wild-type mice. Taken together, these data have revealed a selective aberration of the pathway between PFC pyramidal neurons and BF cholinergic neurons in a tauopathy mouse model. This circuit deficit may underlie the loss of attention and executive function in AD.
Collapse
Affiliation(s)
- Ping Zhong
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, United States
| | - Qing Cao
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, United States
| | - Zhen Yan
- Corresponding author: State University of New York at Buffalo, 955 Main St., Room 3102, Buffalo, NY 14203, United States.
| |
Collapse
|
9
|
Ferré S, Belcher AM, Bonaventura J, Quiroz C, Sánchez-Soto M, Casadó-Anguera V, Cai NS, Moreno E, Boateng CA, Keck TM, Florán B, Earley CJ, Ciruela F, Casadó V, Rubinstein M, Volkow ND. Functional and pharmacological role of the dopamine D 4 receptor and its polymorphic variants. Front Endocrinol (Lausanne) 2022; 13:1014678. [PMID: 36267569 PMCID: PMC9578002 DOI: 10.3389/fendo.2022.1014678] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
The functional and pharmacological significance of the dopamine D4 receptor (D4R) has remained the least well understood of all the dopamine receptor subtypes. Even more enigmatic has been the role of the very prevalent human DRD4 gene polymorphisms in the region that encodes the third intracellular loop of the receptor. The most common polymorphisms encode a D4R with 4 or 7 repeats of a proline-rich sequence of 16 amino acids (D4.4R and D4.7R). DRD4 polymorphisms have been associated with individual differences linked to impulse control-related neuropsychiatric disorders, with the most consistent associations established between the gene encoding D4.7R and attention-deficit hyperactivity disorder (ADHD) and substance use disorders. The function of D4R and its polymorphic variants is being revealed by addressing the role of receptor heteromerization and the relatively avidity of norepinephrine for D4R. We review the evidence conveying a significant and differential role of D4.4R and D4.7R in the dopaminergic and noradrenergic modulation of the frontal cortico-striatal pyramidal neuron, with implications for the moderation of constructs of impulsivity as personality traits. This differential role depends on their ability to confer different properties to adrenergic α2A receptor (α2AR)-D4R heteromers and dopamine D2 receptor (D2R)-D4R heteromers, preferentially localized in the perisomatic region of the frontal cortical pyramidal neuron and its striatal terminals, respectively. We also review the evidence to support the D4R as a therapeutic target for ADHD and other impulse-control disorders, as well as for restless legs syndrome.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, United States
- *Correspondence: Sergi Ferré,
| | - Annabelle M. Belcher
- Division of Addiction Research and Treatment, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jordi Bonaventura
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, United States
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain
| | - César Quiroz
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, United States
| | - Marta Sánchez-Soto
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, United States
| | - Verònica Casadó-Anguera
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - Ning-Sheng Cai
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, United States
| | - Estefanía Moreno
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - Comfort A. Boateng
- Department of Basic Pharmaceutical Sciences, Fred Wilson School of Pharmacy, High Point, NC, United States
| | - Thomas M. Keck
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ, United States
| | - Benjamín Florán
- Departament of Physiology, Biophysics and Neurosciences, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Christopher J. Earley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain
| | - Vicent Casadó
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - Marcelo Rubinstein
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas and, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nora D. Volkow
- National Institute on Drug Abuse, National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
10
|
Yan Z, Rein B. Mechanisms of synaptic transmission dysregulation in the prefrontal cortex: pathophysiological implications. Mol Psychiatry 2022; 27:445-465. [PMID: 33875802 PMCID: PMC8523584 DOI: 10.1038/s41380-021-01092-3] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 03/13/2021] [Accepted: 03/29/2021] [Indexed: 02/02/2023]
Abstract
The prefrontal cortex (PFC) serves as the chief executive officer of the brain, controlling the highest level cognitive and emotional processes. Its local circuits among glutamatergic principal neurons and GABAergic interneurons, as well as its long-range connections with other brain regions, have been functionally linked to specific behaviors, ranging from working memory to reward seeking. The efficacy of synaptic signaling in the PFC network is profundedly influenced by monoaminergic inputs via the activation of dopamine, adrenergic, or serotonin receptors. Stress hormones and neuropeptides also exert complex effects on the synaptic structure and function of PFC neurons. Dysregulation of PFC synaptic transmission is strongly linked to social deficits, affective disturbance, and memory loss in brain disorders, including autism, schizophrenia, depression, and Alzheimer's disease. Critical neural circuits, biological pathways, and molecular players that go awry in these mental illnesses have been revealed by integrated electrophysiological, optogenetic, biochemical, and transcriptomic studies of PFC. Novel epigenetic mechanism-based strategies are proposed as potential avenues of therapeutic intervention for PFC-involved diseases. This review provides an overview of PFC network organization and synaptic modulation, as well as the mechanisms linking PFC dysfunction to the pathophysiology of neurodevelopmental, neuropsychiatric, and neurodegenerative diseases. Insights from the preclinical studies offer the potential for discovering new medical treatments for human patients with these brain disorders.
Collapse
Affiliation(s)
- Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, USA.
| | | |
Collapse
|
11
|
Joffe ME, Winder DG, Conn PJ. Contrasting sex-dependent adaptations to synaptic physiology and membrane properties of prefrontal cortex interneuron subtypes in a mouse model of binge drinking. Neuropharmacology 2020; 178:108126. [PMID: 32781000 DOI: 10.1016/j.neuropharm.2020.108126] [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] [Received: 01/05/2020] [Revised: 04/18/2020] [Accepted: 04/29/2020] [Indexed: 12/24/2022]
Abstract
Alcohol use disorder (AUD) affects all sexes, however women who develop AUD may be particularly susceptible to cravings and other components of the disease. While many brain regions are involved in AUD etiology, proper prefrontal cortex (PFC) function is particularly important for top-down craving management and the moderation of drinking behaviors. Essential regulation of PFC output is provided by local inhibitory interneurons, yet how drinking affects interneuron physiology remains poorly understood, particularly in female individuals. To address this gap, we generated fluorescent reporter transgenic mice to label the two major classes of interneuron in deep layer prelimbic PFC, based on expression of parvalbumin (PV-IN) or somatostatin (SST-IN). We then interrogated PV-IN and SST-IN membrane and synaptic physiology in a rodent model of binge drinking. Beginning in late adolescence, mice received 3-4 weeks of intermittent access (IA) ethanol. We prepared acute brain slices one day after the last drinking session. PV-INs but not SST-INs from IA ethanol mice displayed increased excitability relative to controls, regardless of sex. On the contrary, synaptic adaptations to PV-INs differed based on sex. While drinking decreased excitatory synaptic strength onto PV-INs from female mice, PV-INs from IA ethanol male mice exhibited potentiated excitatory transmission relative to controls. In contrast, decreased synaptic strength onto SST-INs was observed following IA ethanol in all groups of mice. Together, these findings illustrate novel sex differences in drinking-related PFC pathophysiology. Discovering means to restore PV-IN and SST-IN dysfunction following extended drinking provides opportunities for developing new treatments for all AUD patients.
Collapse
Affiliation(s)
- Max E Joffe
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA; Vanderbilt Center for Addiction Research, Nashville, TN, 37232, USA.
| | - Danny G Winder
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Vanderbilt Center for Addiction Research, Nashville, TN, 37232, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA; Vanderbilt Center for Addiction Research, Nashville, TN, 37232, USA
| |
Collapse
|
12
|
Lei G, Liu F, Liu P, Jiao T, Yang L, Chu Z, Deng LS, Li Y, Dang YH. Does genetic mouse model of constitutive Hint1 deficiency exhibit schizophrenia-like behaviors? Schizophr Res 2020; 222:304-318. [PMID: 32439293 DOI: 10.1016/j.schres.2020.05.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/03/2020] [Accepted: 05/06/2020] [Indexed: 01/13/2023]
Abstract
The histidine triad nucleotide binding protein 1 (HINT1) is closely related to many neuropsychiatric disorders. Clinical studies supported that mutations in the Hint1 gene correlated potentially with schizophrenia. In addition, Hint1 gene knockout (KO) mice exhibited hyperactivity induced by amphetamine and apomorphine. However, it is still unclear whether this animal model exhibits schizophrenia-like behaviors and, if so, their underlying mechanisms remain to be elucidated. Thus, our study sought to evaluate schizophrenia-like behaviors in Hint1-KO mice, and explore the associated changes in neuronal structural plasticity and schizophrenia-related molecules. A series of behavioral tests were used to compare Hint1-KO and their wild-type (WT) littermates, alongside a number of morphological and molecular biological methods. Relative to WT mice, Hint1-KO mice exhibited reduced social interaction behaviors, aggressive behavior, sensorimotor gating deficits, apathetic and self-neglect behaviors, and increased MK-801-induced hyperactivity. Hint1-KO mice also showed partly increased dendritic complexity in the hippocampus (Hip) relative to WT mice. Total glutamate was decreased in the medial prefrontal cortex, nucleus accumbens (NAc), and Hip of KO mice. Expression of NR1, NR2A, and D4R was decreased whereas that of D1R was increased in the NAc of KO relative to WT mice. The expression level of NR2B was increased whereas that of D1R was decreased in the Hip of KO mice. Hint1-KO mice exhibited schizophrenia-like behaviors. Partly increased dendritic complexity and dysfunction in both the dopaminergic and glutamatergic systems may be involved in the abnormalities in Hint1-KO mice.
Collapse
Affiliation(s)
- Gang Lei
- College of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Fei Liu
- College of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China; Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Peng Liu
- College of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Tong Jiao
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Liu Yang
- College of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Zheng Chu
- College of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Li-Sha Deng
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Yan Li
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Yong-Hui Dang
- College of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China; Key Laboratory of the Health Ministry for Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China; Key Laboratory of Shaanxi Province for Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China; State Key Laboratory for Manufacturing Systems Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China.
| |
Collapse
|
13
|
Zhong P, Qin L, Yan Z. Dopamine Differentially Regulates Response Dynamics of Prefrontal Cortical Principal Neurons and Interneurons to Optogenetic Stimulation of Inputs from Ventral Tegmental Area. Cereb Cortex 2020; 30:4402-4409. [PMID: 32236403 PMCID: PMC7325712 DOI: 10.1093/cercor/bhaa027] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/06/2019] [Accepted: 01/11/2020] [Indexed: 01/15/2023] Open
Abstract
Prefrontal cortex (PFC) is highly influenced by the inputs from ventral tegmental area (VTA); however, how the projection from VTA impacts PFC neurons and how the synaptically released dopamine affects PFC activity are largely unclear. Using optogenetics and electrophysiological approaches, we examined the impact of VTA stimulation on PFC principal neurons and parvalbumin-positive (PV+) interneurons and the modulatory role of dopamine. We found that the brief activation of the VTA-PFC circuit immediately induced action potential firing, which was mediated by glutamatergic transmission. However, strong stimulation of VTA gradually induced a marked and prolonged enhancement of the excitability of PFC PV+ interneurons and a modest and short-lived enhancement of the excitability of PFC principal neurons. Blocking dopamine receptors (DARs) shortened the VTA excitation of PFC PV+ interneurons and prolonged the VTA excitation of PFC principal neurons. Blocking GABAA receptors induced a similar effect as DAR antagonists in PFC principal neurons, suggesting that the dopaminergic effect is through influencing the inhibitory transmission system. These results have revealed a role of dopamine in regulating the temporal dynamics of excitation/inhibition balance in VTA-PFC circuit, which provides insights into the functional consequence of activating dopamine system in the mesocortical system.
Collapse
Affiliation(s)
- Ping Zhong
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo 14203, NY, USA
| | - Luye Qin
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo 14203, NY, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo 14203, NY, USA
| |
Collapse
|
14
|
Sadeghian A, Salari Z, Azizi H, Raoufy MR, Shojaei A, Kosarmadar N, Zare M, Rezaei M, Barkley V, Javan M, Fathollahi Y, Mirnajafi-Zadeh J. The role of dopamine D 2-like receptors in a "depotentiation-like effect" of deep brain stimulation in kindled rats. Brain Res 2020; 1738:146820. [PMID: 32251663 DOI: 10.1016/j.brainres.2020.146820] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 01/12/2023]
Abstract
The mechanisms involved in the anti-seizure effects of low-frequency stimulation (LFS) have not been completely determined. However, Gi-protein-coupled receptors, including D2-like receptors, may have a role in mediating these effects. In the present study, the role of D2-like receptors in LFS' anti-seizure action was investigated. Rats were kindled with semi-rapid (6 stimulations per day), electrical stimulation of the hippocampal CA1 area. In LFS-treated groups, subjects received four trials of LFS at 5 min, 6 h, 24 h, and 30 h following the last kindling stimulation. Each LFS set occurred at 5 min intervals, and consisted of 4 trains. Each train contained 200, 0/1 ms long, monophasic square wave pulses at 1 Hz. Haloperidol (D2-like receptors antagonist, 2 µm) and/or bromocriptine (D2-like receptors agonist 2 µg/µlit) were microinjected into the lateral ventricle immediately after the last kindling, before applying LFS. Obtained results showed that applying LFS in fully-kindled subjects led to a depotentiation-like decrease in kindling-induced potentiation and reduced the amplitude and rise slope of excitatory and inhibitory post-synaptic currents in whole-cell recordings from CA1 pyramidal neurons. In addition, LFS restored the kindling-induced, spatial learning and memory impairments in the Barnes maze test. A D2-like receptor antagonist inhibited these effects of LFS, while a D2-like receptor agonist mimicked these effects. In conclusion, a depotentiation-like mechanism may be involved in restoring LFS' effects on learning and memory, and synaptic plasticity. These effects depend on D2-like receptors activity.
Collapse
Affiliation(s)
- Azam Sadeghian
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Salari
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Azizi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Reza Raoufy
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Shojaei
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Nastaran Kosarmadar
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Meysam Zare
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahmoud Rezaei
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Victoria Barkley
- Krembil Research Institute, University Health Network, Toronto, Canada
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Yaghoub Fathollahi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
15
|
Methylation-related metabolic effects of D4 dopamine receptor expression and activation. Transl Psychiatry 2019; 9:295. [PMID: 31719518 PMCID: PMC6851363 DOI: 10.1038/s41398-019-0630-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 10/13/2019] [Accepted: 10/20/2019] [Indexed: 11/16/2022] Open
Abstract
D4 dopamine receptor (D4R) activation uniquely promotes methylation of plasma membrane phospholipids, utilizing folate-derived methyl groups provided by methionine synthase (MS). We evaluated the impact of D4R expression on folate-dependent phospholipid methylation (PLM) and MS activity, as well as cellular redox and methylation status, in transfected CHO cells expressing human D4R variants containing 2, 4, or 7 exon III repeats (D4.2R, D4.4R, D4.7R). Dopamine had no effect in non-transfected CHO cells, but increased PLM to a similar extent for both D4.2R- and D4.4R-expressing cells, while the maximal increase was for D4.7R was significantly lower. D4R expression in CHO cells decreased basal MS activity for all receptor subtypes and conferred dopamine-sensitive MS activity, which was greater with a higher number of repeats. Consistent with decreased MS activity, D4R expression decreased basal levels of methylation cycle intermediates methionine, S-adenosylmethionine (SAM), and S-adenosylhomocysteine (SAH), as well as cysteine and glutathione (GSH). Conversely, dopamine stimulation increased GSH, SAM, and the SAM/SAH ratio, which was associated with a more than 2-fold increase in global DNA methylation. Our findings illustrate a profound influence of D4R expression and activation on MS activity, coupled with the ability of dopamine to modulate cellular redox and methylation status. These previously unrecognized signaling activities of the D4R provide a unique link between neurotransmission and metabolism.
Collapse
|
16
|
Neuromodulators and Long-Term Synaptic Plasticity in Learning and Memory: A Steered-Glutamatergic Perspective. Brain Sci 2019; 9:brainsci9110300. [PMID: 31683595 PMCID: PMC6896105 DOI: 10.3390/brainsci9110300] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/24/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022] Open
Abstract
The molecular pathways underlying the induction and maintenance of long-term synaptic plasticity have been extensively investigated revealing various mechanisms by which neurons control their synaptic strength. The dynamic nature of neuronal connections combined with plasticity-mediated long-lasting structural and functional alterations provide valuable insights into neuronal encoding processes as molecular substrates of not only learning and memory but potentially other sensory, motor and behavioural functions that reflect previous experience. However, one key element receiving little attention in the study of synaptic plasticity is the role of neuromodulators, which are known to orchestrate neuronal activity on brain-wide, network and synaptic scales. We aim to review current evidence on the mechanisms by which certain modulators, namely dopamine, acetylcholine, noradrenaline and serotonin, control synaptic plasticity induction through corresponding metabotropic receptors in a pathway-specific manner. Lastly, we propose that neuromodulators control plasticity outcomes through steering glutamatergic transmission, thereby gating its induction and maintenance.
Collapse
|
17
|
Trajectories of brain remodeling in temporal lobe epilepsy. J Neurol 2019; 266:3150-3159. [PMID: 31549200 DOI: 10.1007/s00415-019-09546-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 09/12/2019] [Accepted: 09/14/2019] [Indexed: 12/24/2022]
Abstract
Temporal lobe epilepsy has been usually associated with progressive brain atrophy due to neuronal cell loss. However, recent animal models demonstrated a dual effect of epileptic seizures with initial enhancement of hippocampal neurogenesis followed by abnormal astrocyte proliferation and neurogenesis depletion in the chronic stage. Our aim was to test for the hypothesized bidirectional pattern of epilepsy-associated brain remodeling in the context of the presence and absence of mesial temporal lobe sclerosis. We acquired MRIs from a large cohort of mesial temporal lobe epilepsy patients with or without hippocampus sclerosis on radiological examination. The statistical analysis tested explicitly for common and differential brain patterns between the two patients' cohorts and healthy controls within the computational anatomy framework of voxel-based morphometry. The main effect of disease was associated with continuous hippocampus volume loss ipsilateral to the seizure onset zone in both temporal lobe epilepsy cohorts. The post hoc simple effects tests demonstrated bilateral hippocampus volume increase in the early epilepsy stages in patients without hippocampus sclerosis. Early age of onset and longer disease duration correlated with volume decrease in the ipsilateral hippocampus. Our findings of seizure-induced hippocampal remodeling are associated with specific patterns of mesial temporal lobe atrophy that are modulated by individual clinical phenotype features. Directionality of hippocampus volume changes strongly depends on the chronicity of disease. Specific anatomy differences represent a snapshot within a progressive continuum of seizure-induced structural remodeling.
Collapse
|
18
|
HPO-Shuffle: an associated gene prioritization strategy and its application in drug repurposing for the treatment of canine epilepsy. Biosci Rep 2019; 39:BSR20191247. [PMID: 31427480 PMCID: PMC6732366 DOI: 10.1042/bsr20191247] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/03/2019] [Accepted: 08/12/2019] [Indexed: 12/16/2022] Open
Abstract
Epilepsy is a common neurological disorder that affects mammalian species including human beings and dogs. In order to discover novel drugs for the treatment of canine epilepsy, multiomics data were analyzed to identify epilepsy associated genes. In this research, the original ranking of associated genes was obtained by two high-throughput bioinformatics experiments: Genome Wide Association Study (GWAS) and microarray analysis. The association ranking of genes was enhanced by a re-ranking system, HPO-Shuffle, which integrated information from GWAS, microarray analysis and Human Phenotype Ontology database for further downstream analysis. After applying HPO-Shuffle, the association ranking of epilepsy genes were improved. Afterward, a weighted gene coexpression network analysis (WGCNA) led to a set of gene modules, which hinted a clear relevance between the high ranked genes and the target disease. Finally, WGCNA and connectivity map (CMap) analysis were performed on the integrated dataset to discover a potential drug list, in which a well-known anticonvulsant phensuximide was included.
Collapse
|
19
|
Tan T, Wang W, Williams J, Ma K, Cao Q, Yan Z. Stress Exposure in Dopamine D4 Receptor Knockout Mice Induces Schizophrenia-Like Behaviors via Disruption of GABAergic Transmission. Schizophr Bull 2019; 45:1012-1023. [PMID: 30476265 PMCID: PMC6737476 DOI: 10.1093/schbul/sby163] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
A combination of genetic and environmental risk factors has been considered as the pathogenic cause for mental disorders including schizophrenia. Here, we sought to find out whether the abnormality of the dopamine system, coupled with the exposure to modest stress, is sufficient to trigger the manifestation of schizophrenia-like behaviors. We found that exposing dopamine D4 receptor knockout (D4KO) mice with 1-week restraint stress (2 h/d) induced significant deficits in sensorimotor gating, cognitive processes, social engagement, as well as the elevated exploratory behaviors, which are reminiscent to schizophrenia phenotypes. Electrophysiological studies found that GABAergic transmission was significantly reduced in prefrontal cortical neurons from stressed D4KO mice. Additionally, administration of diazepam, a GABA enhancer, restored GABAergic synaptic responses and ameliorated some behavioral abnormalities in stressed D4KO mice. These results have revealed that the combination of 2 key genetic and environmental susceptibility factors, dopamine dysfunction and stress, is a crucial trigger for schizophrenia-like phenotypes, and GABA system in the prefrontal cortex is a downstream convergent target that mediates some behavioral outcomes.
Collapse
Affiliation(s)
- Tao Tan
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY,Sichuan Provincial Hospital for Women and Children, Chengdu, China
| | - Wei Wang
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY
| | - Jamal Williams
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY
| | - Kaijie Ma
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY
| | - Qing Cao
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY
| | - Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY,To whom correspondence should be addressed; tel: 716-829-3058, fax: 716-829-2344, e-mail:
| |
Collapse
|
20
|
Meda S, Freund N, Norman KJ, Thompson BS, Sonntag KC, Andersen SL. The use of laser capture microdissection to identify specific pathways and mechanisms involved in impulsive choice in rats. Heliyon 2019; 5:e02254. [PMID: 31485508 PMCID: PMC6716106 DOI: 10.1016/j.heliyon.2019.e02254] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/29/2019] [Accepted: 08/05/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Microinjections, lesions, viral-mediated gene transfer, or designer receptors exclusively activated by designer drugs (DREADDs) can identify brain signaling pathways and their pharmacology in research animals. Genetically modified animals are used for more precise assessment of neural circuits. However, only a few of the gene-based pathway modifications are available for use in outbred rat strains. NEW METHOD Behaviorally characterized Sprague-Dawley rats undergo tract tracing through microinjection of fluorospheres, followed by laser capture microdissection (LCM) and qPCR for detecting mRNA of pathway-associated gene products. Correlations between mRNA expression and behavior identify specific involvement of pharmacologically relevant molecules within cells of interest. Here, we examined this methodology in an impulsive choice paradigm and targeted projections from the orbital and medial prefrontal cortex. RESULTS In this proof of concept study, we demonstrate relationships between measures of impulsive choice with distinct neurotransmitter receptor expression in cell populations from four different signaling pathways. COMPARISONS WITH EXISTING METHODS Combining behavior, tract tracing, LCM, and gene expression profiling provides more cellular selectivity than localized lesions and DREADDs, and greater pharmacological specificity than microinjections and viral-mediated gene transfer due to targeting identified neurons. Furthermore, the assessment of inter-individual pathways provides insight into the complex nature of underlying mechanisms involved in typical and atypical behavior. CONCLUSIONS The novel combination of behavior, tract tracing, LCM, and single gene or potential whole genome transcriptome analysis allows for a more targeted understanding of the interconnection of neural circuitry with behavior, and holds promise to identify more specific drug targets that are relevant to behavioral phenotypes.
Collapse
Affiliation(s)
- Shirisha Meda
- Laboratory of Developmental Neuropharmacology, McLean Hospital, Harvard Medical School, USA
| | - Nadja Freund
- Laboratory of Developmental Neuropharmacology, McLean Hospital, Harvard Medical School, USA
- Experimental and Molecular Psychiatry, Ruhr-University, Germany
| | - Kevin J. Norman
- Laboratory of Developmental Neuropharmacology, McLean Hospital, Harvard Medical School, USA
| | - Britta S. Thompson
- Laboratory of Developmental Neuropharmacology, McLean Hospital, Harvard Medical School, USA
| | - Kai-C. Sonntag
- Laboratory for Translational Research on Neurodegeneration, Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, USA
- Basic Neuroscience Division, McLean Hospital, Harvard Medical School, USA
- Department of Psychiatry, McLean Hospital, Harvard Medical School, USA
| | - Susan L. Andersen
- Laboratory of Developmental Neuropharmacology, McLean Hospital, Harvard Medical School, USA
- Basic Neuroscience Division, McLean Hospital, Harvard Medical School, USA
- Department of Psychiatry, McLean Hospital, Harvard Medical School, USA
| |
Collapse
|
21
|
Qin L, Ma K, Yan Z. Chemogenetic Activation of Prefrontal Cortex in Shank3-Deficient Mice Ameliorates Social Deficits, NMDAR Hypofunction, and Sgk2 Downregulation. iScience 2019; 17:24-35. [PMID: 31247448 PMCID: PMC6599088 DOI: 10.1016/j.isci.2019.06.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/21/2019] [Accepted: 06/06/2019] [Indexed: 12/15/2022] Open
Abstract
Haploinsufficiency of the SHANK3 gene is causally linked to autism spectrum disorders (ASDs) in human genetic studies. Here we found that chemogenetic activation of pyramidal neurons in the prefrontal cortex (PFC) of Shank3-deficient mice with the hM3D (Gq) DREADD restored social preference behaviors and elevated glutamatergic synaptic function in PFC. Moreover, the expression of Sgk2 (serum- and glucocorticoid-inducible kinase 2), a member of the Sgk family, which plays a key role in regulating the membrane trafficking of glutamate receptors, was diminished by Shank3 deficiency and rescued by Gq DREADD activation of PFC. Blocking Sgk function in Shank3-deficient mice prevented Gq DREADD from rescuing social and synaptic deficits, whereas blocking Sgk function in wild-type mice led to the attenuation of PFC glutamatergic signaling and the induction of autism-like social deficits. These results have provided a potential circuit intervention and molecular target for autism treatment.
Collapse
Affiliation(s)
- Luye Qin
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA
| | - Kaijie Ma
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA.
| |
Collapse
|
22
|
Ott T, Nieder A. Dopamine and Cognitive Control in Prefrontal Cortex. Trends Cogn Sci 2019; 23:213-234. [PMID: 30711326 DOI: 10.1016/j.tics.2018.12.006] [Citation(s) in RCA: 301] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/20/2018] [Accepted: 12/28/2018] [Indexed: 12/16/2022]
Abstract
Cognitive control, the ability to orchestrate behavior in accord with our goals, depends on the prefrontal cortex. These cognitive functions are heavily influenced by the neuromodulator dopamine. We review here recent insights exploring the influence of dopamine on neuronal response properties in prefrontal cortex (PFC) during ongoing behaviors in primates. This review suggests three major computational roles of dopamine in cognitive control: (i) gating sensory input, (ii) maintaining and manipulating working memory contents, and (iii) relaying motor commands. For each of these roles, we propose a neuronal microcircuit based on known mechanisms of action of dopamine in PFC, which are corroborated by computational network models. This conceptual approach accounts for the various roles of dopamine in prefrontal executive functioning.
Collapse
Affiliation(s)
- Torben Ott
- Animal Physiology Unit, Institute of Neurobiology, University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany; Present address: Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Andreas Nieder
- Animal Physiology Unit, Institute of Neurobiology, University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany.
| |
Collapse
|
23
|
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: 1.9] [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
|
24
|
Intellectual Investment, Dopaminergic Gene Variation, and Life Events: A Critical Examination. PERSONALITY NEUROSCIENCE 2018; 1:e3. [PMID: 32435725 PMCID: PMC7219688 DOI: 10.1017/pen.2018.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 01/17/2018] [Indexed: 11/05/2022]
Abstract
Need for Cognition (NFC) and Openness to Ideas are intellectual investment traits that are characterized by a tendency to seek out, engage in and enjoy effortful cognitive activity. Little, however, is known about the extent to which they are influenced by genetic and environmental factors. With the present contribution, we aim at furthering our knowledge on the mechanisms underlying intellectual investment traits by following-up on a recent investigation of the role of dopaminergic gene variation in intellectual investment. Employing a standard approach that relied on null-hypothesis significance testing, we found that, first, two dopaminergic genetic variants interacted in modulating individual differences in NFC, but not in Openness to Ideas; that, second, negative life events played a role in the modulation of Openness to Ideas, but not of NFC; and that, third, negative life events as assessed using another measure were only marginally related to Openness to Ideas while positive life events were associated with both Openness to Ideas and NFC, with the latter effect being also dependent on DRD4 exon III genotype. However, employing a Bayesian approach, the assumption of a genetic effect on investment traits was overall not supported, while the assumption of a role of positive life events in the modulation of investment traits could be confirmed, with a tentative increment in the prediction of NFC by adding an interaction of positive life events and DRD4 variation to the main effect of positive life events. Our findings underscore the importance to use different approaches in the field of personality neuroscience. To gain deeper insight into the basis of personality traits does not only require to consider genetic as well as environmental influences and their interplay, but also requires more differentiated statistical analyses that can at least in part tackle the often inconsistent findings in this field.
Collapse
|
25
|
Ott T, Nieder A. Dopamine D2 Receptors Enhance Population Dynamics in Primate Prefrontal Working Memory Circuits. Cereb Cortex 2018; 27:4423-4435. [PMID: 27591146 DOI: 10.1093/cercor/bhw244] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 07/13/2016] [Indexed: 01/05/2023] Open
Abstract
Working memory is associated with persistent activity in the prefrontal cortex (PFC). The neuromodulator dopamine, which is released by midbrain neurons projecting into the frontal lobe, influences PFC neurons and networks via the dopamine D1 (D1R) and the D2 receptor (D2R) families. Although behavioral, clinical and computational evidence suggest an involvement of D2Rs in working memory, a neuronal explanation is missing. We report an enhancement of persistent working memory responses of PFC neurons after iontophoretically stimulating D2Rs in monkeys memorizing the number of items in a display. D2R activation improved working memory representation at the population level and increased population dynamics during the transition from visual to mnemonic representations. Computational modeling suggests that D2Rs act by modulating interneuron-to-pyramidal signaling. By increasing the population's response dynamics, D2Rs might put PFC networks in a more flexible state and enhance the neurons' working memory coding, thereby controlling dynamic cognitive control.
Collapse
Affiliation(s)
- Torben Ott
- Animal Physiology, Institute of Neurobiology, Auf der Morgenstelle 28, University of Tübingen, 72076Tübingen, Germany
| | - Andreas Nieder
- Animal Physiology, Institute of Neurobiology, Auf der Morgenstelle 28, University of Tübingen, 72076Tübingen, Germany
| |
Collapse
|
26
|
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: 19] [Impact Index Per Article: 2.7] [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
|
27
|
Wei J, Zhong P, Qin L, Tan T, Yan Z. Chemicogenetic Restoration of the Prefrontal Cortex to Amygdala Pathway Ameliorates Stress-Induced Deficits. Cereb Cortex 2018; 28:1980-1990. [PMID: 28498919 PMCID: PMC6018994 DOI: 10.1093/cercor/bhx104] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/27/2017] [Indexed: 01/11/2023] Open
Abstract
Corticosteroid stress hormones exert a profound impact on cognitive and emotional processes. Understanding the neuronal circuits that are altered by chronic stress is important for counteracting the detrimental effects of stress in a brain region- and cell type-specific manner. Using the chemogenetic tool, Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), which enables the remote, noninvasive and long-lasting modulation of cellular activity and signal transduction in discrete neuronal populations in vivo, we sought to identify the specific pathways that play an essential role in stress responses. We found that prolonged severe stress induced the diminished glutamatergic projection from pyramidal neurons in prefrontal cortex (PFC) to GABAergic interneurons in basolateral amygdala (BLA), leading to the loss of feedforward inhibition and ensuing hyperexcitability of BLA principal neurons, which caused a variety of behavioral abnormalities. Activating PFC pyramidal neurons with hM3D(Gq) DREADD restored the functional connection between PFC and BLA in stressed animals, resulting in the rescue of recognition memory, normalization of locomotor activity and reduction of aggressive behaviors. Inhibiting BLA principal neurons directly with hM4D(Gi) DREADD also blocked BLA hyperactivity and aggressive behaviors in stressed animals. These results have offered an effective avenue to counteract the stress-induced disruption of circuitry homeostasis.
Collapse
Affiliation(s)
- Jing Wei
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
- Medical Research, VA Western New York Healthcare System, Buffalo, NY 14215, USA
| | - Ping Zhong
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
- Medical Research, VA Western New York Healthcare System, Buffalo, NY 14215, USA
| | - Luye Qin
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Tao Tan
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
- Medical Research, VA Western New York Healthcare System, Buffalo, NY 14215, USA
| |
Collapse
|
28
|
Chemogenetic Activation of Prefrontal Cortex Rescues Synaptic and Behavioral Deficits in a Mouse Model of 16p11.2 Deletion Syndrome. J Neurosci 2018; 38:5939-5948. [PMID: 29853627 DOI: 10.1523/jneurosci.0149-18.2018] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/30/2018] [Accepted: 05/21/2018] [Indexed: 01/27/2023] Open
Abstract
Microdeletion of the human 16p11.2 gene locus has been linked to autism spectrum disorder (ASD) and intellectual disability and confers risk for a number of other neurodevelopmental deficits. Transgenic mice carrying 16p11.2 deletion (16p11+/-) display phenotypes reminiscent of those in human patients with 16p11.2 deletion syndrome, but the molecular mechanisms and treatment strategies for these phenotypes remain unknown. In this study, we have found that both male and female 16p11+/- mice exhibit deficient NMDA receptor (NMDAR) function in the medial prefrontal cortex (mPFC), a brain region critical for high-level "executive" functions. Elevating the activity of mPFC pyramidal neurons with a CaMKII-driven Gq-DREADD (Gq-coupled designer receptors exclusively activated by designer drugs) led to the significant increase of NR2B subunit phosphorylation and the restoration of NMDAR function, as well as the amelioration of cognitive and social impairments in 16p11+/- mice. These results suggest that NMDAR hypofunction in PFC may contribute to the pathophysiology of 16p11.2 deletion syndrome and that restoring PFC activity is sufficient to rescue the behavioral deficits.SIGNIFICANCE STATEMENT The 16p11.2 deletion syndrome is strongly associated with autism spectrum disorder and intellectual disability. Using a mouse model carrying the 16p11.2 deletion, 16p11+/-, we identified NMDA receptor hypofunction in the prefrontal cortex (PFC). Elevating the activity of PFC pyramidal neurons with a chemogenetic tool, Gq-DREADD, led to the restoration of NMDA receptor function and the amelioration of cognitive and social impairments in 16p11+/- mice. These results have revealed a novel route for potential therapeutic intervention of 16p11.2 deletion syndrome.
Collapse
|
29
|
Jing Li J, Szkudlarek H, Renard J, Hudson R, Rushlow W, Laviolette SR. Fear Memory Recall Potentiates Opiate Reward Sensitivity through Dissociable Dopamine D1 versus D4 Receptor-Dependent Memory Mechanisms in the Prefrontal Cortex. J Neurosci 2018; 38:4543-4555. [PMID: 29686048 PMCID: PMC6705931 DOI: 10.1523/jneurosci.3113-17.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 03/09/2018] [Accepted: 04/04/2018] [Indexed: 12/18/2022] Open
Abstract
Disturbances in prefrontal cortical (PFC) dopamine (DA) transmission are well established features of psychiatric disorders involving pathological memory processing, such as post-traumatic stress disorder and opioid addiction. Transmission through PFC DA D4 receptors (D4Rs) has been shown to potentiate the emotional salience of normally nonsalient emotional memories, whereas transmission through PFC DA D1 receptors (D1Rs) has been demonstrated to selectively block recall of reward- or aversion-related associative memories. In the present study, using a combination of fear conditioning and opiate reward conditioning in male rats, we examined the role of PFC D4/D1R signaling during the processing of fear-related memory acquisition and recall and subsequent sensitivity to opiate reward memory formation. We report that PFC D4R activation potentiates the salience of normally subthreshold fear conditioning memory cues and simultaneously potentiates the rewarding effects of systemic or intra-ventral tegmental area (VTA) morphine conditioning cues. In contrast, blocking the recall of salient fear memories with intra-PFC D1R activation, blocks the ability of fear memory recall to potentiate systemic or intra-VTA morphine place preference. These effects were dependent upon dissociable PFC phosphorylation states involving calcium-calmodulin-kinase II or extracellular signal-related kinase 1-2, following intra-PFC D4 or D1R activation, respectively. Together, these findings reveal new insights into how aberrant PFC DAergic transmission and associated downstream molecular signaling pathways may modulate fear-related emotional memory processing and concomitantly increase opioid addiction vulnerability.SIGNIFICANCE STATEMENT Post-traumatic stress disorder is highly comorbid with addiction. In this study, we use a translational model of fear memory conditioning to examine how transmission through dopamine D1 or D4 receptors, in the prefrontal cortex (PFC), may differentially control acquisition or recall of fear memories and how these mechanisms might regulate sensitivity to the rewarding effects of opioids. We demonstrate that PFC D4 activation not only controls the salience of fear memory acquisition, but potentiates the rewarding effects of opioids. In contrast, PFC D1 receptor activation blocks recall of fear memories and prevents potentiation of opioid reward effects. Together, these findings demonstrate novel PFC mechanisms that may account for how emotional memory disturbances might increase the addictive liability of opioid-class drugs.
Collapse
Affiliation(s)
| | | | | | - Roger Hudson
- Departments of Anatomy and Cell Biology, and
- Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Walter Rushlow
- Departments of Anatomy and Cell Biology, and
- Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Steven R Laviolette
- Departments of Anatomy and Cell Biology, and
- Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| |
Collapse
|
30
|
Navakkode S, Chew KCM, Tay SJN, Lin Q, Behnisch T, Soong TW. Bidirectional modulation of hippocampal synaptic plasticity by Dopaminergic D4-receptors in the CA1 area of hippocampus. Sci Rep 2017; 7:15571. [PMID: 29138490 PMCID: PMC5686203 DOI: 10.1038/s41598-017-15917-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/03/2017] [Indexed: 11/09/2022] Open
Abstract
Long-term potentiation (LTP) is the persistent increase in the strength of the synapses. However, the neural networks would become saturated if there is only synaptic strenghthening. Synaptic weakening could be facilitated by active processes like long-term depression (LTD). Molecular mechanisms that facilitate the weakening of synapses and thereby stabilize the synapses are also important in learning and memory. Here we show that blockade of dopaminergic D4 receptors (D4R) promoted the formation of late-LTP and transformed early-LTP into late-LTP. This effect was dependent on protein synthesis, activation of NMDA-receptors and CaMKII. We also show that GABAA-receptor mediated mechanisms are involved in the enhancement of late-LTP. We could show that short-term plasticity and baseline synaptic transmission were unaffected by D4R inhibition. On the other hand, antagonizing D4R prevented both early and late forms of LTD, showing that activation of D4Rs triggered a dual function. Synaptic tagging experiments on LTD showed that D4Rs act as plasticity related proteins rather than the setting of synaptic tags. D4R activation by PD 168077 induced a slow-onset depression that was protein synthesis, NMDAR and CaMKII dependent. The D4 receptors, thus exert a bidirectional modulation of CA1 pyramidal neurons by restricting synaptic strengthening and facilitating synaptic weakening.
Collapse
Affiliation(s)
- Sheeja Navakkode
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.,Neurobiology/Aging Program, Centre for Life Sciences, National University of Singapore, Singapore, 117456, Singapore
| | - Katherine C M Chew
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.,Neurobiology/Aging Program, Centre for Life Sciences, National University of Singapore, Singapore, 117456, Singapore
| | - Sabrina Jia Ning Tay
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.,Neurobiology/Aging Program, Centre for Life Sciences, National University of Singapore, Singapore, 117456, Singapore
| | - Qingshu Lin
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.,Neurobiology/Aging Program, Centre for Life Sciences, National University of Singapore, Singapore, 117456, Singapore
| | - Thomas Behnisch
- The Institutes of Brain Science, The State Key Laboratory of Medical Neurobiology, and the Collaborative Innovation Center for Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Tuck Wah Soong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore. .,Neurobiology/Aging Program, Centre for Life Sciences, National University of Singapore, Singapore, 117456, Singapore. .,National Neuroscience Institute, Singapore, 308433, Singapore.
| |
Collapse
|
31
|
Wang YL, Wang JG, Guo FL, Gao XH, Zhao DD, Zhang L, Wang JZ, Lu CB. Selective dopamine receptor 4 activation mediates the hippocampal neuronal calcium response via IP 3 and ryanodine receptors. Brain Res 2017; 1670:1-5. [DOI: 10.1016/j.brainres.2017.05.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 01/21/2023]
|
32
|
Andino-Pavlovsky V, Souza AC, Scheffer-Teixeira R, Tort ABL, Etchenique R, Ribeiro S. Dopamine Modulates Delta-Gamma Phase-Amplitude Coupling in the Prefrontal Cortex of Behaving Rats. Front Neural Circuits 2017; 11:29. [PMID: 28536507 PMCID: PMC5422429 DOI: 10.3389/fncir.2017.00029] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 04/10/2017] [Indexed: 11/13/2022] Open
Abstract
Dopamine release and phase-amplitude cross-frequency coupling (CFC) have independently been implicated in prefrontal cortex (PFC) functioning. To causally investigate whether dopamine release affects phase-amplitude comodulation between different frequencies in local field potentials (LFP) recorded from the medial PFC (mPFC) of behaving rats, we used RuBiDopa, a light-sensitive caged compound that releases the neurotransmitter dopamine when irradiated with visible light. LFP power did not change in any frequency band after the application of light-uncaged dopamine, but significantly strengthened phase-amplitude comodulation between delta and gamma oscillations. Saline did not exert significant changes, while injections of dopamine and RuBiDopa produced a slow increase in comodulation for several minutes after the injection. The results show that dopamine release in the medial PFC shifts phase-amplitude comodulation from theta-gamma to delta-gamma. Although being preliminary results due to the limitation of the low number of animals present in this study, our findings suggest that dopamine-mediated modification of the frequencies involved in comodulation could be a mechanism by which this neurotransmitter regulates functioning in mPFC.
Collapse
Affiliation(s)
- Victoria Andino-Pavlovsky
- Departamento de Química Inorganica, Analítica y Química Física, Instituto de Química Física de los Materiales, Medio Ambiente y Energía (INQUIMAE), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos AiresBuenos Aires, Argentina
| | - Annie C Souza
- Instituto do Cérebro, Federal University of Rio Grande do NorteNatal, Brazil
| | | | - Adriano B L Tort
- Instituto do Cérebro, Federal University of Rio Grande do NorteNatal, Brazil
| | - Roberto Etchenique
- Departamento de Química Inorganica, Analítica y Química Física, Instituto de Química Física de los Materiales, Medio Ambiente y Energía (INQUIMAE), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos AiresBuenos Aires, Argentina
| | - Sidarta Ribeiro
- Instituto do Cérebro, Federal University of Rio Grande do NorteNatal, Brazil
| |
Collapse
|
33
|
Miyauchi M, Neugebauer NM, Meltzer HY. Dopamine D 4 receptor stimulation contributes to novel object recognition: Relevance to cognitive impairment in schizophrenia. J Psychopharmacol 2017; 31:442-452. [PMID: 28347261 DOI: 10.1177/0269881117693746] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Several atypical antipsychotic drugs (APDs) have high affinity for the dopamine (DA) D4 receptor, but the relevance to the efficacy for the treatment of cognitive impairment associated with schizophrenia (CIAS) is poorly understood. The aim of this study was to investigate the effects of D4 receptor stimulation or blockade on novel object recognition (NOR) in normal rats and on the sub-chronic phencyclidine (PCP)-induced novel object recognition deficit. The effect of the D4 agonist, PD168077, and the D4 antagonist, L-745,870, were studied alone, and in combination with clozapine and lurasidone. In normal rats, L-745,870 impaired novel object recognition, whereas PD168077 had no effect. PD168077 acutely reversed the sub-chronic phencyclidine-induced novel object recognition deficit. Co-administration of a sub-effective dose (SED) of PD168077 with a sub-effective dose of lurasidone also reversed this deficit, but a sub-effective dose of PD168077 with a sub-effective dose of clozapine, a more potent D4 antagonist than lurasidone, did not reverse the sub-chronic phencyclidine-induced novel object recognition deficit. At a dose that did not induce a novel object recognition deficit, L-745,870 blocked the ability of clozapine, but not lurasidone, to reverse the novel object recognition deficit. D4 receptor agonism has a beneficial effect on novel object recognition in sub-chronic PCP-treated rats and augments the cognitive enhancing efficacy of an atypical antipsychotic drug that lacks affinity for the D4 receptor, lurasidone.
Collapse
Affiliation(s)
- Masanori Miyauchi
- 1 Department of Psychiatry and Behavioral Sciences, Northwestern Feinberg School of Medicine, Chicago, USA.,2 Sumitomo Dainippon Pharma Co. Ltd, Suita, Osaka, Japan
| | - Nichole M Neugebauer
- 1 Department of Psychiatry and Behavioral Sciences, Northwestern Feinberg School of Medicine, Chicago, USA
| | - Herbert Y Meltzer
- 1 Department of Psychiatry and Behavioral Sciences, Northwestern Feinberg School of Medicine, Chicago, USA
| |
Collapse
|
34
|
Lukkes JL, Freund N, Thompson BS, Meda S, Andersen SL. Preventative treatment in an animal model of ADHD: Behavioral and biochemical effects of methylphenidate and its interactions with ovarian hormones in female rats. Eur Neuropsychopharmacol 2016; 26:1496-1506. [PMID: 27397110 PMCID: PMC5204118 DOI: 10.1016/j.euroneuro.2016.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/06/2016] [Accepted: 06/10/2016] [Indexed: 12/26/2022]
Abstract
Clinical and preclinical studies on attention deficit hyperactivity disorder (ADHD) show that juvenile males that are exposed to methylphenidate (MPH) show reduced risk for substance use later in life. In contrast, little is known about whether females have the same enduring treatment response to stimulants and how gonadal hormones influence their behavior later in life. Females received either a sham or 6-hydroxydopamine (6-OHDA) microinjection in the prefrontal cortex (PFC) at postnatal day (P)10. Subjects were then treated with Vehicle or MPH (2mg/kg, p.o.) between P20-35 and tested during late adolescence/young adulthood (P60); half of these subjects underwent ovariectomy at P55 to determine hormonal influences. Females with 6-OHDA were depleted of PFC dopamine by 61% and demonstrated increased impulsive choice (delayed discounting) and preferences for cocaine-associated environments relative to control females. Both MPH and ovariectomy reduced impulsive choice and cocaine preferences in 6-OHDA females, but had no enduring effect in Sham females. Ovariectomy itself did not significantly affect impulsivity. Juvenile MPH interacted strongly with 6-OHDA to increase D4, D5, Alpha-1A, Alpha-2A, and 5-HT-1A mRNA receptor expression in the PFC. MPH alone effected D1 mRNA, while 6-OHDA increased BDNF; all markers were decreased by ovariectomy. Together, these data suggest that 6-OHDA changes in dopamine are not only relevant for ADHD-like behaviors, but their long-term modulation by treatment and the influence of cyclical differences in menstrual cycle.
Collapse
Affiliation(s)
- Jodi L Lukkes
- Laboratory for Developmental Neuropharmacology, McLean Hospital, USA; Harvard Medical School, USA
| | - Nadja Freund
- Laboratory for Developmental Neuropharmacology, McLean Hospital, USA; Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Britta S Thompson
- Laboratory for Developmental Neuropharmacology, McLean Hospital, USA
| | - Shirisha Meda
- Laboratory for Developmental Neuropharmacology, McLean Hospital, USA
| | - Susan L Andersen
- Laboratory for Developmental Neuropharmacology, McLean Hospital, USA; Harvard Medical School, USA.
| |
Collapse
|
35
|
Zhong P, Liu W, Yan Z. Aberrant regulation of synchronous network activity by the attention-deficit/hyperactivity disorder-associated human dopamine D4 receptor variant D4.7 in the prefrontal cortex. J Physiol 2015; 594:135-47. [PMID: 26541360 DOI: 10.1113/jp271317] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/29/2015] [Indexed: 01/28/2023] Open
Abstract
KEY POINTS The hD4.7 variant has been linked to attention-deficit/hyperactivity disorder (ADHD); however, the underlying mechanism is unknown. We found that activation of hD4.7 induced over-suppression of glutamatergic excitatory network bursts and under-suppression of GABAergic inhibitory network bursts in the prefrontal cortex (PFC) circuitry. Methylphenidate, a psychostimulant drug used to treat ADHD, normalized the effects of hD4.7 on synchronous network bursts in PFC pyramidal neurons. The findings of the present study suggest that the aberrant regulation of PFC synchronous network activity by hD4.7 may underlie its involvement in ADHD. A unique feature of the human D4 receptor (hD4 R) gene is the existence of a large number of polymorphisms in exon 3 coding for the third intracellular loop, which consists of a variable number of tandem repeats. The hD4 R variants with long repeats have been linked to attention-deficit/hyperactivity disorder (ADHD); however, the underlying mechanism is unknown. Emerging evidence suggests that selective attention is controlled by the rhythmic synchronization in the prefrontal cortex (PFC) and its connected networks. In the present study, we examined the role of hD4 R variants in regulating PFC synchronous network activity. D4 R knockout mice with viral infection of hD4.4 or hD4.7 in the medial PFC were used. Whole-cell patch-clamp recordings were performed to examine the effects of activating hD4.x on the spontaneous large scale correlated activity in PFC pyramidal neurons. We found that, compared to the normal four-repeat variant hD4.4, the ADHD-linked variant hD4.7 induces more suppression of glutamatergic excitatory network bursts and less suppression of GABAergic inhibitory network bursts in the PFC circuitry. Methylphenidate, a psychostimulant drug used to treat ADHD, normalized the effects of hD4.7 on synchronous network bursts in PFC pyramidal neurons. These results reveal the differential effects of hD4 R variants on the integrated excitability of PFC circuits. It is suggested that the aberrant regulation of PFC network activity by hD4.7 may underlie its involvement in ADHD. The methylphenidate-induced normalization of synaptic circuitry regulation may contribute to its effectiveness in ADHD treatment.
Collapse
Affiliation(s)
- Ping Zhong
- Department of Physiology & Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY, USA.,VA Western New York Healthcare System, Buffalo, NY, USA
| | - Wenhua Liu
- Department of Physiology & Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY, USA.,School of Lifescience, Zhaoqing University, Zhaoqing, China
| | - Zhen Yan
- Department of Physiology & Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY, USA.,VA Western New York Healthcare System, Buffalo, NY, USA
| |
Collapse
|
36
|
Nakazawa S, Nakamichi K, Imai H, Ichihara J. Effect of dopamine D4 receptor agonists on sleep architecture in rats. Prog Neuropsychopharmacol Biol Psychiatry 2015; 63:6-13. [PMID: 25985889 DOI: 10.1016/j.pnpbp.2015.05.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 04/28/2015] [Accepted: 05/11/2015] [Indexed: 01/11/2023]
Abstract
Dopamine plays a key role in the regulation of sleep-wake states, as revealed by the observation that dopamine-releasing agents such as methylphenidate have wake-promoting effects. However, the precise mechanisms for the wake-promoting effect produced by the enhancement of dopamine transmission are not fully understood. Although dopamine D1, D2, and D3 receptors are known to have differential effects on sleep architecture, the role of D4 receptors (D4Rs), and particularly the influence of D4R activation on the sleep-wake state, has not been studied so far. In this study, we investigated for the first time the effects of two structurally different D4R agonists, Ro 10-5824 and A-412997, on the sleep-wake states in rats. We found that both D4R agonists generally increased waking duration, and conversely, reduced non-rapid eye movement (NREM) sleep duration in rats. The onset of NREM sleep was also generally delayed. However, only the A-412997 agonist (but not the Ro 10-5824) influenced rapid eye movement sleep onset and duration. Furthermore, these effects were accompanied with an enhancement of EEG spectral power in the theta and the gamma bands. Our results suggest the involvement of dopamine D4R in the regulation of sleep-wake states. The activation of the D4R could enhance the arousal states as revealed by the behavioral and electrophysiological patterns in this study. Dopamine D4R may contribute to the arousal effects of dopamine-releasing agents such as methylphenidate.
Collapse
Affiliation(s)
- Shunsuke Nakazawa
- Drug Development Research Laboratories, Sumitomo Dainippon Pharma, Co., Ltd., Osaka, Japan.
| | - Keiko Nakamichi
- Drug Development Research Laboratories, Sumitomo Dainippon Pharma, Co., Ltd., Osaka, Japan
| | - Hideaki Imai
- Drug Development Research Laboratories, Sumitomo Dainippon Pharma, Co., Ltd., Osaka, Japan
| | - Junji Ichihara
- Drug Development Research Laboratories, Sumitomo Dainippon Pharma, Co., Ltd., Osaka, Japan
| |
Collapse
|
37
|
Berridge CW, Spencer RC. Differential cognitive actions of norepinephrine a2 and a1 receptor signaling in the prefrontal cortex. Brain Res 2015; 1641:189-96. [PMID: 26592951 DOI: 10.1016/j.brainres.2015.11.024] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 10/06/2015] [Accepted: 11/12/2015] [Indexed: 10/22/2022]
Abstract
The prefrontal cortex (PFC) supports cognitive and behavioral processes that guide goal directed behavior. Moreover, dysregulated prefrontal cognitive dysfunction is associated with multiple psychiatric disorders. Norepinephrine (NE) signaling in the PFC is a critical modulator of prefrontal cognition and is targeted by a variety of drugs used to treat PFC-dependent cognitive dysfunction. Noradrenergic modulation of PFC-dependent cognition is complex, with concentration and receptor-specific actions that are likely dependent on neuronal activity state. Recent studies indicate that within the PFC, noradrenergic α1 and α2 receptors exert unique modulatory actions across distinct cognitive processes that allow for context-dependent modulation of cognition. Specifically, high affinity post-synaptic α2 receptors, engaged at moderate rates of NE release associated with moderate arousal levels, promote working memory. In contrast, lower affinity α1 receptors, engaged at higher rates of release associated with high arousal conditions (e.g. stress), impair working memory performance while promoting flexible attention. While these and other observations were initially interpreted to indicate high rates of NE release promotes the transition from focused to flexible/scanning attention, recent findings indicate that α1 receptors promote both focused and flexible attention. Collectively, these observations indicate that while α2 and α1 receptors in the PFC differentially modulate distinct cognitive processes, this cannot be simply ascribed to differential roles of these receptors in 'focused' vs. 'flexible' cognitive processes. Translationally, this information indicates that: (1) not all tests of prefrontal cognitive function may be appropriate for preclinical programs aimed at specific PFC-dependent disorders and (2) the treatment of specific PFC cognitive deficits may require the differential targeting of noradrenergic receptor subtypes. This article is part of a Special Issue entitled SI: Noradrenergic System.
Collapse
Affiliation(s)
- Craig W Berridge
- Department of Psychology, University of Wisconsin-Madison, Madison, WI 53706, United States.
| | - Robert C Spencer
- Department of Psychology, University of Wisconsin-Madison, Madison, WI 53706, United States
| |
Collapse
|