1
|
María-Ríos CE, Murphy GG, Morrow JD. Individual Variation in Intrinsic Neuronal Properties of Nucleus Accumbens Core and Shell Medium Spiny Neurons in Animals Prone to Sign- or Goal-Track. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.644332. [PMID: 40236090 PMCID: PMC11996421 DOI: 10.1101/2025.03.24.644332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The "sign-tracking" and "goal-tracking" model of individual variation in associative learning permits the identification of rats with different cue-reactivity and predisposition to addiction-like behaviors. Certainly, compared to "goal-trackers" (GTs), "sign-trackers" (STs) show more susceptibility traits such as increased cue-induced 'relapse' of drugs of abuse. Different cue- and reward-evoked patterns of activity in the nucleus accumbens (NAc) have been a hallmark of the ST/GT phenotype. However, it is unknown whether differences in the intrinsic neuronal properties of NAc medium spiny neurons (MSNs) in the core and shell subregions are also a physiological correlate of these phenotypes. We performed whole-cell slice electrophysiology in outbred male rats and found that STs exhibited the lowest excitability in the NAc core, with lower number of action potentials and firing frequency as well as a blunted voltage/current relationship curve in response to hyperpolarized potentials in both the NAc core and shell. Although firing properties of shell MSNs did not differ between STs and GTs, intermediate responders that engage in both behaviors showed greater excitability compared to both STs and GTs. These findings suggest that intrinsic excitability in the NAc may contribute to individual differences in the attribution of incentive salience. Significance Statement During associative learning, cues acquire predictive value, but in some instances, they also acquire incentive salience, meaning they take on some of the motivational properties of the reward. The propensity to attribute cues with incentive salience varies between individuals, and excessive attribution can lead to maladaptive behaviors. The "sign-and goal-tracking" model allows us to isolate these two properties and disambiguate the neurobiological processes that govern them. To our knowledge this is the first study characterizing passive and active membrane properties of MSNs in the NAc core and shell of STs and GTs, as well as IRs. These findings are meant to better inform investigations of the distinct role of the NAc in reward learning, particularly in the attribution of incentive salience and addiction predisposition.
Collapse
|
2
|
Donders Z, Skorupska IJ, Willems E, Mussen F, Broeckhoven JV, Carlier A, Schepers M, Vanmierlo T. Beyond PDE4 inhibition: A comprehensive review on downstream cAMP signaling in the central nervous system. Biomed Pharmacother 2024; 177:117009. [PMID: 38908196 DOI: 10.1016/j.biopha.2024.117009] [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: 03/28/2024] [Revised: 05/27/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is a key second messenger that regulates signal transduction pathways pivotal for numerous biological functions. Intracellular cAMP levels are spatiotemporally regulated by their hydrolyzing enzymes called phosphodiesterases (PDEs). It has been shown that increased cAMP levels in the central nervous system (CNS) promote neuroplasticity, neurotransmission, neuronal survival, and myelination while suppressing neuroinflammation. Thus, elevating cAMP levels through PDE inhibition provides a therapeutic approach for multiple CNS disorders, including multiple sclerosis, stroke, spinal cord injury, amyotrophic lateral sclerosis, traumatic brain injury, and Alzheimer's disease. In particular, inhibition of the cAMP-specific PDE4 subfamily is widely studied because of its high expression in the CNS. So far, the clinical translation of full PDE4 inhibitors has been hampered because of dose-limiting side effects. Hence, focusing on signaling cascades downstream activated upon PDE4 inhibition presents a promising strategy, offering novel and pharmacologically safe targets for treating CNS disorders. Yet, the underlying downstream signaling pathways activated upon PDE(4) inhibition remain partially elusive. This review provides a comprehensive overview of the existing knowledge regarding downstream mediators of cAMP signaling induced by PDE4 inhibition or cAMP stimulators. Furthermore, we highlight existing gaps and future perspectives that may incentivize additional downstream research concerning PDE(4) inhibition, thereby providing novel therapeutic approaches for CNS disorders.
Collapse
Affiliation(s)
- Zoë Donders
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium
| | - Iga Joanna Skorupska
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht 6629ER, the Netherlands
| | - Emily Willems
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium
| | - Femke Mussen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium
| | - Jana Van Broeckhoven
- Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; University MS Centre (UMSC) Hasselt - Pelt, Belgium
| | - Aurélie Carlier
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht 6629ER, the Netherlands
| | - Melissa Schepers
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; University MS Centre (UMSC) Hasselt - Pelt, Belgium
| | - Tim Vanmierlo
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; University MS Centre (UMSC) Hasselt - Pelt, Belgium.
| |
Collapse
|
3
|
Labouesse MA, Wilhelm M, Kagiampaki Z, Yee AG, Denis R, Harada M, Gresch A, Marinescu AM, Otomo K, Curreli S, Serratosa Capdevila L, Zhou X, Cola RB, Ravotto L, Glück C, Cherepanov S, Weber B, Zhou X, Katner J, Svensson KA, Fellin T, Trudeau LE, Ford CP, Sych Y, Patriarchi T. A chemogenetic approach for dopamine imaging with tunable sensitivity. Nat Commun 2024; 15:5551. [PMID: 38956067 PMCID: PMC11219860 DOI: 10.1038/s41467-024-49442-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 06/05/2024] [Indexed: 07/04/2024] Open
Abstract
Genetically-encoded dopamine (DA) sensors enable high-resolution imaging of DA release, but their ability to detect a wide range of extracellular DA levels, especially tonic versus phasic DA release, is limited by their intrinsic affinity. Here we show that a human-selective dopamine receptor positive allosteric modulator (PAM) can be used to boost sensor affinity on-demand. The PAM enhances DA detection sensitivity across experimental preparations (in vitro, ex vivo and in vivo) via one-photon or two-photon imaging. In vivo photometry-based detection of optogenetically-evoked DA release revealed that DETQ administration produces a stable 31 minutes window of potentiation without effects on animal behavior. The use of the PAM revealed region-specific and metabolic state-dependent differences in tonic DA levels and enhanced single-trial detection of behavior-evoked phasic DA release in cortex and striatum. Our chemogenetic strategy can potently and flexibly tune DA imaging sensitivity and reveal multi-modal (tonic/phasic) DA signaling across preparations and imaging approaches.
Collapse
Affiliation(s)
- Marie A Labouesse
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Maria Wilhelm
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
- Institute for Neuroscience, ETH Zurich, Zurich, Switzerland
| | | | - Andrew G Yee
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Raphaelle Denis
- Department of Pharmacology & Physiology, Faculty of Medicine, SNC and CIRCA Research groups, Université de Montréal, Montréal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, SNC and CIRCA Research groups, Université de Montréal, Montréal, QC, Canada
| | - Masaya Harada
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Andrea Gresch
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | | | - Kanako Otomo
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Sebastiano Curreli
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | | | - Xuehan Zhou
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Reto B Cola
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Luca Ravotto
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Chaim Glück
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Stanislav Cherepanov
- Institute of Cellular and Integrative Neuroscience, University of Strasbourg, Strasbourg, France
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland
| | - Xin Zhou
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | - Tommaso Fellin
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | - Louis-Eric Trudeau
- Department of Pharmacology & Physiology, Faculty of Medicine, SNC and CIRCA Research groups, Université de Montréal, Montréal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, SNC and CIRCA Research groups, Université de Montréal, Montréal, QC, Canada
| | - Christopher P Ford
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Yaroslav Sych
- Institute of Cellular and Integrative Neuroscience, University of Strasbourg, Strasbourg, France
| | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland.
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
4
|
Szabadi E. Three paradoxes related to the mode of action of pramipexole: The path from D2/D3 dopamine receptor stimulation to modification of dopamine-modulated functions. J Psychopharmacol 2024; 38:581-596. [PMID: 39041250 DOI: 10.1177/02698811241261022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Pramipexole, a D2/D3 dopamine receptor agonist, is used to treat the motor symptoms of Parkinson's disease, caused by degeneration of the dopaminergic nigrostriatal pathway. There are three paradoxes associated with its mode of action. Firstly, stimulation of D2/D3 receptors leads to neuronal inhibition, although pramipexole does not inhibit but promotes some dopamine-modulated functions, such as locomotion and reinforcement. Secondly, another dopamine-modulated function, arousal, is not promoted but inhibited by pramipexole, leading to sedation. Thirdly, pramipexole-evoked sedation is associated with an increase in pupil diameter, although sedation is expected to cause pupil constriction. To resolve these paradoxes, the path from stimulation of D2/D3 receptors to the modification of dopamine-modulated functions has been tracked. The functions considered are modulated by midbrain dopaminergic nuclei: locomotion - substantia nigra pars compacta (SNc), reinforcement/motivation - ventral tegmental area (VTA), sympathetic activity (as reflected in pupil function) - VTA; arousal - ventral periaqueductal grey (vPAG), with contributions from VTA and SNc. The application of genetics-based molecular techniques (optogenetics and chemogenetics) has enabled tracing the chains of neurones from the dopaminergic nuclei to their final targets executing the functions. The functional neuronal circuits linked to the D2/D3 receptors in the dorsal and ventral striata, stimulated by inputs from SNc and VTA, respectively, may explain how neuronal inhibition induced by pramipexole is translated into the promotion of locomotion, reinforcement/motivation and sympathetic activity. As the vPAG may increase arousal mainly by stimulating cortical D1 dopamine receptors, pramipexole would stimulate only presynaptic D2/D3 receptors on vPAG neurones, curtailing their activity and leading to sedation.
Collapse
Affiliation(s)
- Elemer Szabadi
- Developmental Psychiatry, University of Nottingham, Nottingham, UK
| |
Collapse
|
5
|
Wang Q, Wang Y, Liao FF, Zhou FM. Dopaminergic inhibition of the inwardly rectifying potassium current in direct pathway medium spiny neurons in normal and parkinsonian striatum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.29.590632. [PMID: 38746264 PMCID: PMC11092482 DOI: 10.1101/2024.04.29.590632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Despite the profound behavioral effects of the striatal dopamine (DA) activity and the inwardly rectifying potassium channel ( Kir ) being a key determinant of striatal medium spiny neuron (MSN) activity that also profoundly affects behavior, previously reported DA regulations of Kir are conflicting and incompatible with MSN function in behavior. Here we show that in normal mice with an intact striatal DA system, the predominant effect of DA activation of D1Rs in D1-MSNs is to cause a modest depolarization and increase in input resistance by inhibiting Kir, thus moderately increasing the spike outputs from behavior-promoting D1-MSNs. In parkinsonian (DA-depleted) striatum, DA increases D1-MSN intrinsic excitability more strongly than in normal striatum, consequently strongly increasing D1-MSN spike firing that is behavior-promoting; this DA excitation of D1-MSNs is stronger when the DA depletion is more severe. The DA inhibition of Kir is occluded by the Kir blocker barium chloride (BaCl 2 ). In behaving parkinsonian mice, BaCl 2 microinjection into the dorsal striatum stimulates movement but occludes the motor stimulation of D1R agonism. Taken together, our results resolve the long-standing question about what D1R agonism does to D1-MSN excitability in normal and parkinsonian striatum and strongly indicate that D1R inhibition of Kir is a key ion channel mechanism that mediates D1R agonistic behavioral stimulation in normal and parkinsonian animals.
Collapse
|
6
|
Fang LZ, Creed MC. Updating the striatal-pallidal wiring diagram. Nat Neurosci 2024; 27:15-27. [PMID: 38057614 PMCID: PMC11892008 DOI: 10.1038/s41593-023-01518-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 11/06/2023] [Indexed: 12/08/2023]
Abstract
The striatal and pallidal complexes are basal ganglia structures that orchestrate learning and execution of flexible behavior. Models of how the basal ganglia subserve these functions have evolved considerably, and the advent of optogenetic and molecular tools has shed light on the heterogeneity of subcircuits within these pathways. However, a synthesis of how molecularly diverse neurons integrate into existing models of basal ganglia function is lacking. Here, we provide an overview of the neurochemical and molecular diversity of striatal and pallidal neurons and synthesize recent circuit connectivity studies in rodents that takes this diversity into account. We also highlight anatomical organizational principles that distinguish the dorsal and ventral basal ganglia pathways in rodents. Future work integrating the molecular and anatomical properties of striatal and pallidal subpopulations may resolve controversies regarding basal ganglia network function.
Collapse
Affiliation(s)
- Lisa Z Fang
- Washington University Pain Center, Department of Anesthesiology, St. Louis, MO, USA
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, Newfoundland and Labrador, Canada
| | - Meaghan C Creed
- Washington University Pain Center, Department of Anesthesiology, St. Louis, MO, USA.
- Departments of Psychiatry, Neuroscience and Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
7
|
Li C, Saliba NB, Martin H, Losurdo NA, Kolahdouzan K, Siddiqui R, Medeiros D, Li W. Purkinje cell dopaminergic inputs to astrocytes regulate cerebellar-dependent behavior. Nat Commun 2023; 14:1613. [PMID: 36959176 PMCID: PMC10036610 DOI: 10.1038/s41467-023-37319-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/13/2023] [Indexed: 03/25/2023] Open
Abstract
Dopamine has a significant role in motor and cognitive function. The dopaminergic pathways originating from the midbrain have received the most attention; however, the relevance of the cerebellar dopaminergic system is largely undiscovered. Here, we show that the major cerebellar astrocyte type Bergmann glial cells express D1 receptors. Dopamine can be synthesized in Purkinje cells by cytochrome P450 and released in an activity-dependent fashion. We demonstrate that activation of D1 receptors induces membrane depolarization and Ca2+ release from the internal store. These astrocytic activities in turn modify Purkinje cell output by altering its excitatory and inhibitory synaptic input. Lastly, we show that conditional knockout of D1 receptors in Bergmann glial cells results in decreased locomotor activity and impaired social activity. These results contribute to the understanding of the molecular, cellular, and circuit mechanisms underlying dopamine function in the cerebellum, revealing a critical role for the cerebellar dopaminergic system in motor and social behavior.
Collapse
Affiliation(s)
- Chang Li
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Natalie B Saliba
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hannah Martin
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Nicole A Losurdo
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Neuroscience Program, The University of Utah, Salt Lake City, UT, USA
| | - Kian Kolahdouzan
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Riyan Siddiqui
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Destynie Medeiros
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Wei Li
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
8
|
Tsuboi D, Otsuka T, Shimomura T, Faruk MO, Yamahashi Y, Amano M, Funahashi Y, Kuroda K, Nishioka T, Kobayashi K, Sano H, Nagai T, Yamada K, Tzingounis AV, Nambu A, Kubo Y, Kawaguchi Y, Kaibuchi K. Dopamine drives neuronal excitability via KCNQ channel phosphorylation for reward behavior. Cell Rep 2022; 40:111309. [PMID: 36070693 DOI: 10.1016/j.celrep.2022.111309] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/22/2022] [Accepted: 08/12/2022] [Indexed: 11/25/2022] Open
Abstract
Dysfunctional dopamine signaling is implicated in various neuropsychological disorders. Previously, we reported that dopamine increases D1 receptor (D1R)-expressing medium spiny neuron (MSN) excitability and firing rates in the nucleus accumbens (NAc) via the PKA/Rap1/ERK pathway to promote reward behavior. Here, the results show that the D1R agonist, SKF81297, inhibits KCNQ-mediated currents and increases D1R-MSN firing rates in murine NAc slices, which is abolished by ERK inhibition. In vitro ERK phosphorylates KCNQ2 at Ser414 and Ser476; in vivo, KCNQ2 is phosphorylated downstream of dopamine signaling in NAc slices. Conditional deletion of Kcnq2 in D1R-MSNs reduces the inhibitory effect of SKF81297 on KCNQ channel activity, while enhancing neuronal excitability and cocaine-induced reward behavior. These effects are restored by wild-type, but not phospho-deficient KCNQ2. Hence, D1R-ERK signaling controls MSN excitability via KCNQ2 phosphorylation to regulate reward behavior, making KCNQ2 a potential therapeutical target for psychiatric diseases with a dysfunctional reward circuit.
Collapse
Affiliation(s)
- Daisuke Tsuboi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, 1-98 Dengakugakubo, Kusukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Takeshi Otsuka
- Division of Cerebral Circuitry, National Institute for Physiological Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi, 444-8787, Japan
| | - Takushi Shimomura
- Division of Biophysics and Neurobiology, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan
| | - Md Omar Faruk
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, 65 Tsuruma-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Yukie Yamahashi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, 1-98 Dengakugakubo, Kusukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Mutsuki Amano
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, 65 Tsuruma-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Yasuhiro Funahashi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, 1-98 Dengakugakubo, Kusukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Keisuke Kuroda
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, 65 Tsuruma-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Tomoki Nishioka
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, 1-98 Dengakugakubo, Kusukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan
| | - Hiromi Sano
- Division of System Neurophysiology, National Institute for Physiological Sciences and Department of Physiological Sciences, Sokendai, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan; Division of Behavioral Neuropharmacology, International Center for Brain Science, Fujita Health University, 1-98 Dengakugakubo, Kusukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Taku Nagai
- Division of Behavioral Neuropharmacology, International Center for Brain Science, Fujita Health University, 1-98 Dengakugakubo, Kusukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Graduate School of Medicine, Nagoya University, 65 Tsuruma-cho, Showa-ku, Nagoya, Aichi 466-8560, Japan
| | | | - Atsushi Nambu
- Division of System Neurophysiology, National Institute for Physiological Sciences and Department of Physiological Sciences, Sokendai, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan
| | - Yoshihiro Kubo
- Division of Biophysics and Neurobiology, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan
| | - Yasuo Kawaguchi
- Division of Cerebral Circuitry, National Institute for Physiological Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi, 444-8787, Japan; Brain Science Institute, Tamagawa University, Machida, Tokyo 194-8610, Japan
| | - Kozo Kaibuchi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, 1-98 Dengakugakubo, Kusukake-cho, Toyoake, Aichi 470-1192, Japan; Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, 65 Tsuruma-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan.
| |
Collapse
|
9
|
The effects of ninjin'yoeito on the electrophysiological properties of dopamine neurons in the ventral tegmental area/substantia nigra pars compacta and medium spiny neurons in the nucleus accumbens. Aging (Albany NY) 2022; 14:4634-4652. [PMID: 35660668 PMCID: PMC9217710 DOI: 10.18632/aging.204109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 05/23/2022] [Indexed: 11/25/2022]
Abstract
The ventral tegmental area (VTA), substantia nigra pars compacta (SNpc) and nucleus accumbens (NAc) are involved in the regulation of appetite and motivational behaviors. A traditional Japanese (Kampo) medicine, ninjin'yoeito (NYT), has been reported to improve decreased motivation and anorexia in patients with Alzheimer's disease and apathy-like model mice. Thus, NYT may affect the activities of neurons in the VTA, SNpc and NAc. However, little is known about the underlying mechanisms of NYT. Here, we investigated the effects of NYT on the electrophysiological properties of dopaminergic neurons in the VTA and SNpc, as well as on those of medium spiny neurons (MSNs) in the NAc (core and shell subregions), by applying the patch-clamp technique in the brain slices. NYT reduced the resting membrane potential of VTA and SNpc dopaminergic neurons. In contrast, NYT increased the firing frequency of NAc MSNs accompanied by shortened first spike latency and interspike interval. Furthermore, NYT attenuated the inward rectification and sustained outward currents. In conclusion, NYT may directly influence the excitability of dopaminergic neurons in the VTA and SNpc, as well as MSNs in the NAc (core and shell). NYT may modulate dopamine signals in appetite and motivational behaviors.
Collapse
|
10
|
Dairaghi L, Constantin S, Oh A, Shostak D, Wray S. The Dopamine D4 Receptor Regulates Gonadotropin-Releasing Hormone Neuron Excitability in Male Mice. eNeuro 2022; 9:ENEURO.0461-21.2022. [PMID: 35165199 PMCID: PMC8896547 DOI: 10.1523/eneuro.0461-21.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/26/2022] [Accepted: 02/02/2022] [Indexed: 12/30/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH)-secreting neurons control fertility. The release of GnRH peptide regulates the synthesis and release of both luteinizing hormone (LH) and Follicle stimulation hormone (FSH) from the anterior pituitary. While it is known that dopamine regulates GnRH neurons, the specific dopamine receptor subtype(s) involved remain unclear. Previous studies in adult rodents have reported juxtaposition of fibers containing tyrosine hydroxylase (TH), a marker of catecholaminergic cells, onto GnRH neurons and that exogenous dopamine inhibits GnRH neurons postsynaptically through dopamine D1-like and/or D2-like receptors. Our microarray data from GnRH neurons revealed a high level of Drd4 transcripts [i.e., dopamine D4 receptor (D4R)]. Single-cell RT-PCR and immunocytochemistry confirmed GnRH cells express the Drd4 transcript and protein, respectively. Calcium imaging identified changes in GnRH neuronal activity during application of subtype-specific dopamine receptor agonists and antagonists when GABAergic and glutamatergic transmission was blocked. Dopamine, dopamine with D1/5R-specific or D2/3R-specific antagonists or D4R-specific agonists decreased the frequency of calcium oscillations. In contrast, D1/5R-specific agonists increased the frequency of calcium oscillations. The D4R-mediated inhibition was dependent on Gαi/o protein coupling, while the D1/5R-mediated excitation required Gαs protein coupling. Together, these results indicate that D4R plays an important role in the dopaminergic inhibition of GnRH neurons.
Collapse
Affiliation(s)
| | | | - Andrew Oh
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892
| | - David Shostak
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892
| | | |
Collapse
|
11
|
Zhang Y, Gui H, Duan Z, Yu T, Zhang J, Liang X, Liu C. Dopamine D1 Receptor in the Nucleus Accumbens Modulates the Emergence from Propofol Anesthesia in Rat. Neurochem Res 2021; 46:1435-1446. [PMID: 33683630 DOI: 10.1007/s11064-021-03284-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 01/26/2021] [Accepted: 02/23/2021] [Indexed: 01/04/2023]
Abstract
It has been reported that systemic activation of D1 receptors promotes emergence from isoflurane-induced unconsciousness, suggesting that the central dopaminergic system is involved in the process of recovering from general anesthesia. The nucleus accumbens (NAc) contains abundant GABAergic medium spiny neurons (MSNs) expressing the D1 receptor (D1R), which plays a key role in sleep-wake behavior. However, the role of NAc D1 receptors in the process of emergence from general anesthesia has not been identified. Here, using real-time in vivo fiber photometry, we found that neuronal activity in the NAc was markedly disinhibited during recovery from propofol anesthesia. Subsequently, microinjection of a D1R selective agonist (chloro-APB hydrobromide) into the NAc notably reduced the time to emerge from propofol anesthesia with a decrease in δ-band power and an increase in β-band power evident in the cortical electroencephalogram. These effects were prevented by pretreatment with a D1R antagonist (SCH-23390). Whole-cell patch clamp recordings were performed to further explore the cellular mechanism underlying the modulation of D1 receptors on MSNs under propofol anesthesia. Our data primarily demonstrated that propofol increased the frequency and prolonged the decay time of spontaneous inhibitory postsynaptic currents (sIPSCs) and miniature IPSCs (mIPSCs) of MSNs expressing D1 receptors. A D1R agonist attenuated the effect of propofol on the frequency of sIPSCs and mIPSCs, and the effects of the agonist were eliminated by preapplication of SCH-23390. Collectively, these results indicate that modulation of the D1 receptor on the activity of NAc MSNs is vital for emergence from propofol-induced unconsciousness.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Huan Gui
- Guizhou Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zikun Duan
- Guizhou Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Tian Yu
- Guizhou Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jie Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaoli Liang
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chengxi Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China.
- Guizhou Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, The Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
12
|
Liu J, Jin Y, Wang B, Wang Y, Zuo S, Zhang J. Dopamine D1 receptor alleviates doxorubicin-induced cardiac injury by inhibiting NLRP3 inflammasome. Biochem Biophys Res Commun 2021; 561:7-13. [PMID: 33992835 DOI: 10.1016/j.bbrc.2021.04.098] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023]
Abstract
Doxorubicin (DOX) is a broad-spectrum antineoplastic drug; however, its serious cardiotoxic side effects in inflammatory responses limit its use in clinical applications. Dopamine D1 receptor (DRD1), a G protein-coupled receptor, is crucial for the development and function of the nervous system; additionally, it also play a role in immune regulation. However, the specific role of DRD1 in DOX-induced cardiac inflammation has not yet been clarified. Here, we discovered that DRD1 expression was induced by DOX treatment in H9C2 cardiomyocytes. DRD1 activation by A-68930, a DRD1-specific agonist, decreased DOX-induced nucleotide-binding domain-like receptor protein 3 (NLRP3) expression, caspase-1 activation, and IL-1β maturation in H9C2 cells. Expression of the cytokines IL-1β and IL-18 in the supernatants was also inhibited by A-68930 treatment. DRD1 knockdown, using siRNA, abolished the effects of A-68930 on the DOX-induced NLRP3 inflammasome. Furthermore, we found that DRD1 signaling downregulated the NLRP3 inflammasome in H9C2 cells through cyclic adenosine monophosphate (cAMP). Moreover, application of A-68930 to activate DRD1 reduced cardiac injury and fibrosis in a DOX-treated mouse model by suppressing the NLRP3 inflammasome in the heart. These findings indicate that DRD1 signaling may protect against DOX-induced cardiac injury by inhibiting the NLRP3 inflammasome-mediated inflammation.
Collapse
Affiliation(s)
- Jiao Liu
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
| | - Yuxuan Jin
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bei Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yiran Wang
- Division of Biological and Environmental Science and Engineering, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Saudi Arabia
| | - Shengkai Zuo
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jinying Zhang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China.
| |
Collapse
|
13
|
Zhang Y, Gui H, Hu L, Li C, Zhang J, Liang X. Dopamine D1 receptor in the NAc shell is involved in delayed emergence from isoflurane anesthesia in aged mice. Brain Behav 2021; 11:e01913. [PMID: 33094567 PMCID: PMC7821614 DOI: 10.1002/brb3.1913] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/24/2020] [Accepted: 10/07/2020] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Delayed emergence after general anesthesia tends to occur in the elderly population, but the mechanism remains unclear. Apart from age-related pharmacokinetic changes, the aging-induced structural and functional alterations in the arousal-promoting neural substrates should be considered. The nucleus accumbens (NAc) is a crucial arousal-related nucleus, in which activating medium spiny neurons (MSNs) expressing dopamine D1 receptor (D1R) could facilitate the arousal from natural sleep. Meanwhile, the dopaminergic systems decline with aging in multiple brain regions. However, whether the age-related decline in D1R in the NAc shell attenuates its arousal-promoting capacity from general anesthesia remains to be elucidated. METHODS We first verified the delayed emergence from isoflurane anesthesia and examined the corresponding changes of electroencephalogram (EEG) power in aged mice. In turn, the arousal-modulating capacity of D1R was characterized in the young and aged cohorts by microinjection of D1R agonist/antagonist into the NAc shell. Furthermore, to address the possible mechanism responsible for the attenuated arousal-modulating capacity of the aged NAc, the expression of D1R in the NAc shell was measured and compared between young and aged mice. RESULTS Our data indicated that compared with young mice, the emergence time in aged mice was notably longer, while EEG power in δ band (1-4Hz) was significantly higher and power in β band (12-25Hz) was lower. Activating or inhibiting D1R in the NAc shell by microinjection D1R agonist/antagonist promoted or delayed the emergence process in young mice. Nevertheless, this modulation capacity of D1R in the NAc shell declined in aged mice, respectively. Meanwhile, downregulation of D1R expression in the NAc shell was detected in the aged brain. CONCLUSION Together, these results suggest that aging attenuates the arousal-modulating capacity of D1R in the NAc shell probably through downregulation of D1R expression therein, which may provide a potential explanation and a therapeutic target for increased sensitivity to anesthetics in the elderly patients.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Zunyi Medical University, Zunyi, China.,Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, China.,School of Anesthesiology, Zunyi Medical University, Zunyi, China
| | - Huan Gui
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, China
| | - Lang Hu
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, China
| | - Chengxi Li
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, China
| | - Jie Zhang
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, China
| | - Xiaoli Liang
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, China.,School of Anesthesiology, Zunyi Medical University, Zunyi, China
| |
Collapse
|
14
|
Ferrario CR. Why did I eat that? Contributions of individual differences in incentive motivation and nucleus accumbens plasticity to obesity. Physiol Behav 2020; 227:113114. [DOI: 10.1016/j.physbeh.2020.113114] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 07/16/2020] [Accepted: 07/21/2020] [Indexed: 02/02/2023]
|
15
|
Lahiri AK, Bevan MD. Dopaminergic Transmission Rapidly and Persistently Enhances Excitability of D1 Receptor-Expressing Striatal Projection Neurons. Neuron 2020; 106:277-290.e6. [PMID: 32075716 PMCID: PMC7182485 DOI: 10.1016/j.neuron.2020.01.028] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 12/26/2019] [Accepted: 01/22/2020] [Indexed: 12/13/2022]
Abstract
Substantia nigra dopamine neurons have been implicated in the initiation and invigoration of movement, presumably through their modulation of striatal projection neuron (SPN) activity. However, the impact of native dopaminergic transmission on SPN excitability has not been directly demonstrated. Using perforated patch-clamp recording, we found that optogenetic stimulation of nigrostriatal dopamine axons rapidly and persistently elevated the excitability of D1 receptor-expressing SPNs (D1-SPNs). The evoked firing of D1-SPNs increased within hundreds of milliseconds of stimulation and remained elevated for ≥ 10 min. Consistent with the negative modulation of depolarization- and Ca2+-activated K+ currents, dopaminergic transmission accelerated subthreshold depolarization in response to current injection, reduced the latency to fire, and transiently diminished action potential afterhyperpolarization. Persistent modulation was protein kinase A dependent and associated with a reduction in action potential threshold. Together, these data demonstrate that dopaminergic transmission potently increases D1-SPN excitability with a time course that could support subsecond and sustained behavioral control.
Collapse
Affiliation(s)
- Asha K Lahiri
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Mark D Bevan
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
16
|
The projections from the anterior cingulate cortex to the nucleus accumbens and ventral tegmental area contribute to neuropathic pain-evoked aversion in rats. Neurobiol Dis 2020; 140:104862. [PMID: 32251841 DOI: 10.1016/j.nbd.2020.104862] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 03/26/2020] [Accepted: 03/31/2020] [Indexed: 02/08/2023] Open
Abstract
Although the anterior cingulate cortex (ACC) plays a vital role in neuropathic pain-related aversion, the underlying mechanisms haven't been fully studied. The mesolimbic dopamine system encodes reward and aversion, and participates in the exacerbation of chronic pain. Therefore, we investigated whether the ACC modulates aversion to neuropathic pain via control of the mesolimbic dopamine system, in a rat model of chronic constriction injury (CCI) to the sciatic nerve. Using anterograde and retrograde tracings, we confirmed that a subgroup of ACC neurons projected to the nucleus accumbens (NAc) and ventral tegmental area (VTA), which are two crucial nodes of the mesolimbic dopamine system. Combining electrophysiology in juvenile rats 7 days post-CCI, we found that the NAc/VTA-projecting neurons were hyperexcitable after CCI. Chemogenetic inhibition of these projections induced conditioned place preference in young adult rats 10-14 days post-CCI, without modulating the evoked pain threshold, whereas activation of these projections in sham rats mimicked aversive behavior. Furthermore, the function of the ACC projections was probably mediated by NAc D2-type medium spiny neurons and VTA GABAergic neurons. Taken together, our findings suggest that projections from the ACC to the NAc and VTA mediate neuropathic pain-related aversive behavior.
Collapse
|
17
|
Keren A, Gilhar A, Ullmann Y, Zlotkin-Frušić M, Soroka Y, Domb AJ, Levite M. Instantaneous depolarization of T cells via dopamine receptors, and inhibition of activated T cells of Psoriasis patients and inflamed human skin, by D1-like receptor agonist: Fenoldopam. Immunology 2020; 158:171-193. [PMID: 31424569 DOI: 10.1111/imm.13109] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 12/11/2022] Open
Abstract
Activated T cells are pathological in various autoimmune and inflammatory diseases including Psoriasis, and also in graft rejection and graft-versus-host-disease. In these pathological conditions, selective silencing of activated T cells through physiological receptors they express remains a clinical challenge. In our previous studies we found that activation of dopamine receptors (DRs) in resting human T cells activates these cells, and induces by itself many beneficial T cell functions. In this study, we found that normal human T cells express all types of DRs, and that expression of D1R, D4R and D5R increases profoundly after T cell receptor (TCR) activation. Interestingly, DR agonists shift the membrane potential (Vm ) of both resting and activated human T cells, and induces instantaneous T cell depolarization within 15 seconds only. Thus, activation of DRs in T cells depolarize these immune cells, alike activation of DRs in neural cells. The skin of Psoriasis patients contains 20-fold more D1R+ T cells than healthy human skin. In line with that, 25-fold more D1R+ T cells are present in Psoriasis humanized mouse model. Highly selective D1-like receptor agonists, primarily Fenoldopam (Corlopam) - a D1-like receptor agonist and a drug used in hypertension, induced the following suppressive effects on activated T cells of Psoriasis patients: reduced chemotactic migration towards the chemokine SDF-1/CXCL12; reduced dramatically the secretion of eight cytokines: tumor necrosis factor-α, interferon-γ, interleukin-1β (IL-1β), IL-2, IL-4, IL-6, IL-8 and IL-10; and reduced three T cell activation proteins/markers: CD69, CD28 and IL-2. Next, we invented a novel topical/dermal Fenoldopam formulation, allowing it to be spread on, and providing prolonged and regulated release in, diseased skin. Our novel topical/dermal Fenoldopam: reduced secretion of the eight cytokines by activated human T cells; reduced IL-1β and IL-6 secretion by human lipopolysaccharide-inflamed skin; eliminated preferentially >90% of live and large/proliferating human T cells. Together, our findings show for the first time that both resting and activated T cells are depolarized instantaneously via DRs, and that targeting D1-like receptors in activated T cells and inflamed human skin by Fenoldopam, in Psoriasis, and potentially in other T cell-mediated diseases, could be therapeutic. Validation in vivo is required.
Collapse
Affiliation(s)
- Aviad Keren
- Skin Research Laboratory, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Amos Gilhar
- Skin Research Laboratory, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yehuda Ullmann
- Skin Research Laboratory, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | | | - Yoram Soroka
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Abraham J Domb
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Mia Levite
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.,Institute of Gene Therapy, Hadassah University Hospital, Jerusalem, Israel
| |
Collapse
|
18
|
Alonso-Caraballo Y, Ferrario CR. Effects of the estrous cycle and ovarian hormones on cue-triggered motivation and intrinsic excitability of medium spiny neurons in the Nucleus Accumbens core of female rats. Horm Behav 2019; 116:104583. [PMID: 31454509 PMCID: PMC7256930 DOI: 10.1016/j.yhbeh.2019.104583] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/01/2019] [Accepted: 08/20/2019] [Indexed: 11/08/2022]
Abstract
Naturally occurring alterations in estradiol influence food intake in females. However, how motivational responses to food cues are affected by the estrous cycle or ovarian hormones is unknown. In addition, while individual susceptibility to obesity is accompanied by enhanced incentive motivational responses to food cues and increased NAc intrinsic excitability in males, studies in females are absent. Therefore, we examined basal differences in intrinsic NAc excitability of obesity-prone vs. obesity-resistant females and determined how conditioned approach (a measure of cue-triggered motivation), food intake, and motivation for food vary with the cycle in naturally cycling female obesity-prone, obesity-resistant, and outbred Sprague-Dawley rats. Finally, we used ovariectomy followed by hormone treatment to determine the role of ovarian hormones in cue-triggered motivation in selectively-bred and outbred female rats. We found that intrinsic excitability of NAc MSNs and conditioned approach are enhanced in female obesity-prone vs. obesity-resistant rats. These effects were driven by greater MSN excitability and conditioned approach behavior during metestrus/diestrus vs. proestrus/estrus in obesity-prone but not obesity-resistant rats, despite similar regulation of food intake and food motivation by the cycle in these groups. Furthermore, estradiol and progesterone treatment reduced conditioned approach behavior in obesity-prone and outbred Sprague-Dawley females. To our knowledge, these data are the first to demonstrate cycle- and hormone-dependent effects on the motivational response to a food cue, and the only studies to date to determine how individual susceptibility to obesity influences NAc excitability, cue-triggered food-seeking, and differences in the regulation of these neurobehavioral responses by the estrous cycle.
Collapse
Affiliation(s)
| | - Carrie R Ferrario
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, United States of America; Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States of America.
| |
Collapse
|
19
|
Oginsky MF, Ferrario CR. Eating "junk food" has opposite effects on intrinsic excitability of nucleus accumbens core neurons in obesity-susceptible versus -resistant rats. J Neurophysiol 2019; 122:1264-1273. [PMID: 31365322 DOI: 10.1152/jn.00361.2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The nucleus accumbens (NAc) plays critical roles in motivated behaviors, including food seeking and feeding. Differences in NAc function contribute to overeating that drives obesity, but the underlying mechanisms are poorly understood. In addition, there is a fair degree of variation in individual susceptibility versus resistance to obesity that is due in part to differences in NAc function. For example, using selectively bred obesity-prone and obesity-resistant rats, we have found that excitability of medium spiny neurons (MSNs) within the NAc core is enhanced in obesity-prone versus -resistant populations, before any diet manipulation. However, it is unknown whether consumption of sugary, fatty "junk food" alters MSN excitability. Here whole cell patch-clamp recordings were conducted to examine MSN intrinsic excitability in adult male obesity-prone and obesity-resistant rats with and without exposure to a sugary, fatty junk food diet. We replicated our initial finding that basal excitability is enhanced in obesity-prone versus obesity-resistant rats and determined that this is due to a lower fast transient potassium current (IA) in prone versus resistant groups. In addition, the junk food diet had opposite effects on excitability in obesity-prone versus obesity-resistant rats. Specifically, junk food enhanced excitability in MSNs of obesity-resistant rats; this was mediated by a reduction in IA. In contrast, junk food reduced excitability in MSNs from obesity-prone rats; this was mediated by an increase in inward-rectifying potassium current. Thus individual differences in obesity susceptibility influence both basal excitability and how MSN excitability adapts to junk food consumption.NEW & NOTEWORTHY Medium spiny neurons (MSNs) in the nucleus accumbens of obesity-prone rats are hyperexcitable compared with MSNs from obesity-resistant rats. We found that 10 days of "junk food" exposure reduces MSN excitability in obesity-prone rats by increasing inward-rectifying potassium current and increases MSN excitability in obesity-resistant rats by decreasing fast transient potassium current. These data show that there are basal and junk food diet-induced differences in MSN excitability in obesity-prone and obesity-resistant individuals; this may contribute to previously observed differences in incentive motivation.
Collapse
Affiliation(s)
- Max F Oginsky
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | - Carrie R Ferrario
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
20
|
Lindroos R, Dorst MC, Du K, Filipović M, Keller D, Ketzef M, Kozlov AK, Kumar A, Lindahl M, Nair AG, Pérez-Fernández J, Grillner S, Silberberg G, Hellgren Kotaleski J. Basal Ganglia Neuromodulation Over Multiple Temporal and Structural Scales-Simulations of Direct Pathway MSNs Investigate the Fast Onset of Dopaminergic Effects and Predict the Role of Kv4.2. Front Neural Circuits 2018; 12:3. [PMID: 29467627 PMCID: PMC5808142 DOI: 10.3389/fncir.2018.00003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/09/2018] [Indexed: 12/16/2022] Open
Abstract
The basal ganglia are involved in the motivational and habitual control of motor and cognitive behaviors. Striatum, the largest basal ganglia input stage, integrates cortical and thalamic inputs in functionally segregated cortico-basal ganglia-thalamic loops, and in addition the basal ganglia output nuclei control targets in the brainstem. Striatal function depends on the balance between the direct pathway medium spiny neurons (D1-MSNs) that express D1 dopamine receptors and the indirect pathway MSNs that express D2 dopamine receptors. The striatal microstructure is also divided into striosomes and matrix compartments, based on the differential expression of several proteins. Dopaminergic afferents from the midbrain and local cholinergic interneurons play crucial roles for basal ganglia function, and striatal signaling via the striosomes in turn regulates the midbrain dopaminergic system directly and via the lateral habenula. Consequently, abnormal functions of the basal ganglia neuromodulatory system underlie many neurological and psychiatric disorders. Neuromodulation acts on multiple structural levels, ranging from the subcellular level to behavior, both in health and disease. For example, neuromodulation affects membrane excitability and controls synaptic plasticity and thus learning in the basal ganglia. However, it is not clear on what time scales these different effects are implemented. Phosphorylation of ion channels and the resulting membrane effects are typically studied over minutes while it has been shown that neuromodulation can affect behavior within a few hundred milliseconds. So how do these seemingly contradictory effects fit together? Here we first briefly review neuromodulation of the basal ganglia, with a focus on dopamine. We furthermore use biophysically detailed multi-compartmental models to integrate experimental data regarding dopaminergic effects on individual membrane conductances with the aim to explain the resulting cellular level dopaminergic effects. In particular we predict dopaminergic effects on Kv4.2 in D1-MSNs. Finally, we also explore dynamical aspects of the onset of neuromodulation effects in multi-scale computational models combining biochemical signaling cascades and multi-compartmental neuron models.
Collapse
Affiliation(s)
- Robert Lindroos
- Department of Neuroscience, Nobel Institute for Neurophysiology, Stockholm, Sweden
| | - Matthijs C. Dorst
- Department of Neuroscience, Nobel Institute for Neurophysiology, Stockholm, Sweden
| | - Kai Du
- Department of Neuroscience, Nobel Institute for Neurophysiology, Stockholm, Sweden
| | - Marko Filipović
- Bernstein Center Freiburg, University of Freiburg, Freiburg im Breisgau, Germany
| | - Daniel Keller
- Blue Brain Project, Ecole Polytechnique Fédérale de Lausanne, Geneva, Switzerland
| | - Maya Ketzef
- Department of Neuroscience, Nobel Institute for Neurophysiology, Stockholm, Sweden
| | - Alexander K. Kozlov
- Science for Life Laboratory, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Solna, Sweden
| | - Arvind Kumar
- Bernstein Center Freiburg, University of Freiburg, Freiburg im Breisgau, Germany
- Department Computational Science and Technology, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Mikael Lindahl
- Science for Life Laboratory, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Solna, Sweden
| | - Anu G. Nair
- Science for Life Laboratory, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Solna, Sweden
| | - Juan Pérez-Fernández
- Department of Neuroscience, Nobel Institute for Neurophysiology, Stockholm, Sweden
| | - Sten Grillner
- Department of Neuroscience, Nobel Institute for Neurophysiology, Stockholm, Sweden
| | - Gilad Silberberg
- Department of Neuroscience, Nobel Institute for Neurophysiology, Stockholm, Sweden
| | - Jeanette Hellgren Kotaleski
- Department of Neuroscience, Nobel Institute for Neurophysiology, Stockholm, Sweden
- Science for Life Laboratory, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Solna, Sweden
| |
Collapse
|
21
|
Cui P, Li XY, Zhao Y, Li Q, Gao F, Li LZ, Yin N, Sun XH, Wang Z. Activation of dopamine D1 receptors enhances the temporal summation and excitability of rat retinal ganglion cells. Neuroscience 2017; 355:71-83. [DOI: 10.1016/j.neuroscience.2017.04.046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/24/2017] [Accepted: 04/30/2017] [Indexed: 01/11/2023]
|
22
|
Contestabile A, Magara S, Cancedda L. The GABAergic Hypothesis for Cognitive Disabilities in Down Syndrome. Front Cell Neurosci 2017; 11:54. [PMID: 28326014 PMCID: PMC5339239 DOI: 10.3389/fncel.2017.00054] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/14/2017] [Indexed: 12/04/2022] Open
Abstract
Down syndrome (DS) is a genetic disorder caused by the presence of a third copy of chromosome 21. DS affects multiple organs, but it invariably results in altered brain development and diverse degrees of intellectual disability. A large body of evidence has shown that synaptic deficits and memory impairment are largely determined by altered GABAergic signaling in trisomic mouse models of DS. These alterations arise during brain development while extending into adulthood, and include genesis of GABAergic neurons, variation of the inhibitory drive and modifications in the control of neural-network excitability. Accordingly, different pharmacological interventions targeting GABAergic signaling have proven promising preclinical approaches to rescue cognitive impairment in DS mouse models. In this review, we will discuss recent data regarding the complex scenario of GABAergic dysfunctions in the trisomic brain of DS mice and patients, and we will evaluate the state of current clinical research targeting GABAergic signaling in individuals with DS.
Collapse
Affiliation(s)
- Andrea Contestabile
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia (IIT) Genova, Italy
| | - Salvatore Magara
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia (IIT) Genova, Italy
| | - Laura Cancedda
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia (IIT)Genova, Italy; Dulbecco Telethon InstituteGenova, Italy
| |
Collapse
|
23
|
Suzuki K, Harada A, Suzuki H, Miyamoto M, Kimura H. TAK-063, a PDE10A Inhibitor with Balanced Activation of Direct and Indirect Pathways, Provides Potent Antipsychotic-Like Effects in Multiple Paradigms. Neuropsychopharmacology 2016; 41:2252-62. [PMID: 26849714 PMCID: PMC4946053 DOI: 10.1038/npp.2016.20] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 01/13/2016] [Accepted: 02/01/2016] [Indexed: 01/29/2023]
Abstract
Phosphodiesterase 10A (PDE10A) inhibitors are expected to be novel drugs for schizophrenia through activation of both direct and indirect pathway medium spiny neurons. However, excess activation of the direct pathway by a dopamine D1 receptor agonist SKF82958 canceled antipsychotic-like effects of a dopamine D2 receptor antagonist haloperidol in methamphetamine (METH)-induced hyperactivity in rats. Thus, balanced activation of these pathways may be critical for PDE10A inhibitors. Current antipsychotics and the novel PDE10A inhibitor TAK-063, but not the selective PDE10A inhibitor MP-10, produced dose-dependent antipsychotic-like effects in METH-induced hyperactivity and prepulse inhibition in rodents. TAK-063 and MP-10 activated the indirect pathway to a similar extent; however, MP-10 caused greater activation of the direct pathway than did TAK-063. Interestingly, the off-rate of TAK-063 from PDE10A in rat brain sections was faster than that of MP-10, and a slower off-rate PDE10A inhibitor with TAK-063-like chemical structure showed an MP-10-like pharmacological profile. In general, faster off-rate enzyme inhibitors are more sensitive than slower off-rate inhibitors to binding inhibition by enzyme substrates. As expected, TAK-063 was more sensitive than MP-10 to binding inhibition by cyclic nucleotides. Moreover, an immunohistochemistry study suggested that cyclic adenosine monophosphate levels in the direct pathway were higher than those in the indirect pathway. These data can explain why TAK-063 showed partial activation of the direct pathway compared with MP-10. The findings presented here suggest that TAK-063's antipsychotic-like efficacy may be attributable to its unique pharmacological properties, resulting in balanced activation of the direct and indirect striatal pathways.
Collapse
Affiliation(s)
- Kazunori Suzuki
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Akina Harada
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hirobumi Suzuki
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Maki Miyamoto
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan,Drug Metabolism and Pharmacokinetics Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Haruhide Kimura
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan,Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan, Tel: +81 466321859, Fax: +81 466294468, E-mail:
| |
Collapse
|
24
|
Domingos L, Desrus A, Même S, Même W. L-Phosphinothricin modulation of inwardly rectifying K+ channels increased excitability in striatal medium-sized spiny neurons. Arch Toxicol 2016; 90:1719-27. [DOI: 10.1007/s00204-016-1721-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 04/20/2016] [Indexed: 10/21/2022]
|
25
|
Zhao B, Zhu J, Dai D, Xing J, He J, Fu Z, Zhang L, Li Z, Wang W. Differential dopaminergic regulation of inwardly rectifying potassium channel mediated subthreshold dynamics in striatal medium spiny neurons. Neuropharmacology 2016; 107:396-410. [PMID: 27018450 DOI: 10.1016/j.neuropharm.2016.03.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 03/09/2016] [Accepted: 03/22/2016] [Indexed: 11/29/2022]
Abstract
The dorsal striatum plays a key role in motor control and cognitive processes. Proper functioning of the striatum relies on the fine dynamic balance between the direct pathway projection medium spiny neurons (MSNs) that express D1 dopamine receptor (D1 MSNs) and indirect pathway projection MSNs that express D2 dopamine receptor (D2 MSNs). The inwardly rectifying K(+) channels (Kir), which express on both D1 and D2 MSNs, participate in the subthreshold dynamics including the membrane resonance and dendritic integration. However, it remains unclear whether dopamine differentially regulates Kir mediated subthreshold dynamics in two subtypes MSNs. Using transgenic mice that express either tdTomato in D1 MSNs or eGFP in D2 MSNs, we explored the Kir mediated subthreshold dynamics in D1 or D2 MSNs with whole cell patch clamp recording in acute brain slices. We found that D1 receptor agonist increased the Kir current while D2 receptor activation decreased the Kir conductance. The dopamine regulation of the Kir enhanced the resonant frequency and reduced the resonant impedance of D1 MSNs. The converse is ture for D2 MSNs. It also caused an opposing effect on dendritic integration between D1 and D2 MSNs, which can promote stability of the two pathways. The D1 receptor activation modulated Kir through cAMP-PKA signaling, whereas the D2 receptor modulated Kir through PLC-PKC signaling. Our findings demonstrated the differential dopaminergic regulation role of Kir, which mediates distinct subthreshold dynamics, and thus, contributes to the role of dopamine in fine tuning the balance of the striatal direct and indirect pathway activities.
Collapse
Affiliation(s)
- Bo Zhao
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, PR China; Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, PR China; Department of Neurology, Anning Branch of Lanzhou General Hospital of Lanzhou Military Region, Lanzhou 730070, PR China
| | - Junling Zhu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, PR China
| | - Dongqing Dai
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, PR China; Cadet Brigade, Fourth Military Medical University, Xi'an 710032, PR China
| | - Junling Xing
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, PR China
| | - Jiahou He
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, PR China; Cadet Brigade, Fourth Military Medical University, Xi'an 710032, PR China
| | - Zhanyan Fu
- Model System and Neurobiology Department, Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Lei Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Zhuyi Li
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, PR China.
| | - Wenting Wang
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, PR China.
| |
Collapse
|
26
|
Li Q, Wu N, Cui P, Gao F, Qian WJ, Miao Y, Sun XH, Wang Z. Suppression of outward K(+) currents by activating dopamine D1 receptors in rat retinal ganglion cells through PKA and CaMKII signaling pathways. Brain Res 2016; 1635:95-104. [PMID: 26826585 DOI: 10.1016/j.brainres.2016.01.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/17/2016] [Accepted: 01/21/2016] [Indexed: 01/11/2023]
Abstract
Dopamine plays an important role in regulating neuronal functions in the central nervous system by activating the specific G-protein coupled receptors. Both D1 and D2 dopamine receptors are extensively distributed in the retinal neurons. In the present study, we investigated the effects of D1 receptor signaling on outward K(+) currents in acutely isolated rat retinal ganglion cells (RGCs) by patch-clamp techniques. Extracellular application of SKF81297 (10 μM), a specific D1 receptor agonist, significantly and reversibly suppressed outward K(+) currents of the cells, which was reversed by SCH23390 (10 μM), a selective D1 receptor antagonist. We further showed that SKF81297 mainly suppressed the glybenclamide (Gb)- and 4-aminopyridine (4-AP)-sensitive K(+) current components, but did not show effect on the tetraethylammonium (TEA)-sensitive one. Both protein kinase A (PKA) and calcium/calmodulin-dependent protein kinase II (CaMKII) signaling pathways were likely involved in the SKF81297-induced suppression of the K(+) currents since either Rp-cAMP (10 μM), a cAMP/PKA signaling inhibitor, or KN-93 (10 μM), a specific CaMKII inhibitor, eliminated the SKF81297 effect. In contrast, neither protein kinase C (PKC) nor mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling pathway seemed likely to be involved because both the PKC inhibitor bisindolylmaleimide IV (Bis IV) (10 μM) and the MAPK/ERK1/2 inhibitor U0126 (10 μM) did not block the SKF81297-induced suppression of the K(+) currents. These results suggest that activation of D1 receptors suppresses the Gb- and 4-AP-sensitive K(+) current components in rat RGCs through the intracellular PKA and CaMKII signaling pathways, thus modulating the RGC excitability.
Collapse
Affiliation(s)
- Qian Li
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Institute of Neurobiology, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Na Wu
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Department of Ophthalmology at Eye & ENT Hospital, Fudan University, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Peng Cui
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Institute of Neurobiology, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Feng Gao
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Department of Ophthalmology at Eye & ENT Hospital, Fudan University, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Wen-Jing Qian
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Institute of Neurobiology, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Yanying Miao
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Institute of Neurobiology, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Xing-Huai Sun
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Department of Ophthalmology at Eye & ENT Hospital, Fudan University, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Zhongfeng Wang
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Department of Ophthalmology at Eye & ENT Hospital, Fudan University, Shanghai 200031, China; Institute of Neurobiology, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
27
|
In vitro cardiomyocyte differentiation of umbilical cord blood cells: crucial role for c-kit+ cells. Cytotherapy 2015; 17:1627-37. [DOI: 10.1016/j.jcyt.2015.07.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 07/16/2015] [Accepted: 07/20/2015] [Indexed: 11/22/2022]
|
28
|
Hamao K, Kawaura K, Soeda F, Hamasaki R, Shirasaki T, Takahama K. Tipepidine increases dopamine level in the nucleus accumbens without methamphetamine-like behavioral sensitization. Behav Brain Res 2015; 284:118-24. [DOI: 10.1016/j.bbr.2015.02.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 01/10/2015] [Accepted: 02/05/2015] [Indexed: 11/27/2022]
|
29
|
Gray CL, Norvelle A, Larkin T, Huhman KL. Dopamine in the nucleus accumbens modulates the memory of social defeat in Syrian hamsters (Mesocricetus auratus). Behav Brain Res 2015; 286:22-8. [PMID: 25721736 DOI: 10.1016/j.bbr.2015.02.030] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 02/10/2015] [Accepted: 02/16/2015] [Indexed: 11/17/2022]
Abstract
Conditioned defeat (CD) is a behavioral response that occurs in Syrian hamsters after they experience social defeat. Subsequently, defeated hamsters no longer produce territorial aggression but instead exhibit heightened levels of avoidance and submission, even when confronted with a smaller, non-aggressive intruder. Dopamine in the nucleus accumbens is hypothesized to act as a signal of salience for both rewarding and aversive stimuli to promote memory formation and appropriate behavioral responses to significant events. The purpose of the present study was to test the hypothesis that dopamine in the nucleus accumbens modulates the acquisition and expression of behavioral responses to social defeat. In Experiment 1, bilateral infusion of the non-specific D1/D2 receptor antagonist cis(z)flupenthixol (3.75 μg/150 nl saline) into the nucleus accumbens 5 min prior to defeat training significantly reduced submissive and defensive behavior expressed 24h later in response to a non-aggressive intruder. In Experiment 2, infusion of 3.75 μg cis-(Z)-flupenthixol 5 min before conditioned defeat testing with a non-aggressive intruder significantly increased aggressive behavior in drug-infused subjects. In Experiment 3, we found that the effect of cis-(Z)-flupenthixol on aggression was specific to defeated animals as infusion of drug into the nucleus accumbens of non-defeated animals did not significantly alter their behavior in response to a non-aggressive intruder. These data demonstrate that dopamine in the nucleus accumbens modulates both acquisition and expression of social stress-induced behavioral changes and suggest that the nucleus accumbens plays an important role in the suppression of aggression that is observed after social defeat.
Collapse
Affiliation(s)
- C L Gray
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - A Norvelle
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302, USA
| | - T Larkin
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302, USA
| | - K L Huhman
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302, USA.
| |
Collapse
|
30
|
Sun N, Laviolette SR. Dopamine receptor blockade modulates the rewarding and aversive properties of nicotine via dissociable neuronal activity patterns in the nucleus accumbens. Neuropsychopharmacology 2014; 39:2799-815. [PMID: 24896614 PMCID: PMC4200490 DOI: 10.1038/npp.2014.130] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 05/29/2014] [Accepted: 05/30/2014] [Indexed: 11/09/2022]
Abstract
The mesolimbic pathway comprising the ventral tegmental area (VTA) and projection terminals in the nucleus accumbens (NAc) has been identified as a critical neural system involved in processing both the rewarding and aversive behavioral effects of nicotine. Transmission through dopamine (DA) receptors functionally modulates these effects directly within the NAc. Nevertheless, the neuronal mechanisms within the NAc responsible for these bivalent behavioral effects are presently not known. Using an unbiased conditioned place preference procedure combined with in vivo neuronal recordings, we examined the effects of nicotine reward and aversion conditioning on intra-NAc neuronal sub-population activity patterns. We report that intra-VTA doses of nicotine that differentially produce rewarding or aversive behavioral effects produce opposite effects on sub-populations of fast-spiking interneurons (FSIs) or medium spiny neurons (MSNs) within the shell region of the NAc (NAshell). Thus, while the rewarding effects of intra-VTA nicotine were associated with inhibition of FSI and activation of MSNs, the aversive effects of nicotine produced the opposite pattern of NAshell neuronal population activity. Blockade of DA transmission with a broad-spectrum DA receptor antagonist, α-flupenthixol, strongly inhibited the spontaneous activity of NAshell FSIs, and reversed the conditioning properties of intra-VTA nicotine, switching nicotine-conditioned responses from aversive to rewarding. Remarkably, DA receptor blockade switched intra-NAshell neuronal population activity from an aversion to a reward pattern, concomitant with the observed switch in behavioral conditioning effects.
Collapse
Affiliation(s)
- Ninglei Sun
- Department of Anatomy and Cell Biology, The Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Steven R Laviolette
- Department of Anatomy and Cell Biology, The Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada,Department of Psychiatry, The Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada,Department of Psychology, The Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada,Department of Anatomy and Cell Biology, The Schulich School of Medicine and Dentistry, University of Western Ontario, 468 Medical Science Building, London, ON, Canada N6A 5C1, Tel: +1 519 661 2111 ext. 80302, Fax: +1 519 661 3936, E-mail:
| | | |
Collapse
|
31
|
Glycyrrhizae radix methanol extract attenuates methamphetamine-induced locomotor sensitization and conditioned place preference. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:152063. [PMID: 25386216 PMCID: PMC4214053 DOI: 10.1155/2014/152063] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Revised: 09/09/2014] [Accepted: 09/18/2014] [Indexed: 11/29/2022]
Abstract
Glycyrrhizae Radix modulates the neurochemical and locomotor alterations induced by acute psychostimulants in rodents via GABAb receptors. This study investigated the influence of methanol extract from Glycyrrhizae Radix (MEGR) on repeated methamphetamine- (METH-) induced locomotor sensitization and conditioned place preference (CPP). A cohort of rats was treated with METH (1 mg/kg/day) for 6 consecutive days, subjected to 6 days of withdrawal, and then challenged with the same dose of METH to induce locomotor sensitization; during the withdrawal period, the rats were administered MEGR (60 or 180 mg/kg/day). A separate cohort of rats was treated with either METH or saline every other day for 6 days in METH-paired or saline-paired chambers, respectively, to induce CPP. These rats were also administered MEGR (180 mg/kg) prior to every METH or CPP expression test. Pretreatment with MEGR (60 and 180 mg/kg/day) attenuated the expression of METH-induced locomotor sensitization dose-dependently, and 180 mg/kg MEGR significantly inhibited the development and expression of METH-induced CPP. Furthermore, administration of a selective GABAb receptor antagonist (SCH50911) prior to MEGR treatment effectively blocked the inhibitory effects of MEGR on locomotor sensitization, but not CPP. These results suggest that Glycyrrhizae Radix blocked repeated METH-induced behavioral changes via GABAb receptors.
Collapse
|
32
|
Gunaydin LA, Grosenick L, Finkelstein JC, Kauvar IV, Fenno LE, Adhikari A, Lammel S, Mirzabekov JJ, Airan RD, Zalocusky KA, Tye KM, Anikeeva P, Malenka RC, Deisseroth K. Natural neural projection dynamics underlying social behavior. Cell 2014; 157:1535-51. [PMID: 24949967 DOI: 10.1016/j.cell.2014.05.017] [Citation(s) in RCA: 973] [Impact Index Per Article: 88.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 04/30/2014] [Accepted: 05/12/2014] [Indexed: 01/11/2023]
Abstract
Social interaction is a complex behavior essential for many species and is impaired in major neuropsychiatric disorders. Pharmacological studies have implicated certain neurotransmitter systems in social behavior, but circuit-level understanding of endogenous neural activity during social interaction is lacking. We therefore developed and applied a new methodology, termed fiber photometry, to optically record natural neural activity in genetically and connectivity-defined projections to elucidate the real-time role of specified pathways in mammalian behavior. Fiber photometry revealed that activity dynamics of a ventral tegmental area (VTA)-to-nucleus accumbens (NAc) projection could encode and predict key features of social, but not novel object, interaction. Consistent with this observation, optogenetic control of cells specifically contributing to this projection was sufficient to modulate social behavior, which was mediated by type 1 dopamine receptor signaling downstream in the NAc. Direct observation of deep projection-specific activity in this way captures a fundamental and previously inaccessible dimension of mammalian circuit dynamics.
Collapse
Affiliation(s)
- Lisa A Gunaydin
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Logan Grosenick
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Neuroscience Program, Stanford University, Stanford, CA 94305, USA
| | - Joel C Finkelstein
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Isaac V Kauvar
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Lief E Fenno
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Neuroscience Program, Stanford University, Stanford, CA 94305, USA
| | - Avishek Adhikari
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Stephan Lammel
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Julie J Mirzabekov
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Raag D Airan
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Kelly A Zalocusky
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Neuroscience Program, Stanford University, Stanford, CA 94305, USA
| | - Kay M Tye
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Polina Anikeeva
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Robert C Malenka
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
33
|
Borin M, Fogli Iseppe A, Pignatelli A, Belluzzi O. Inward rectifier potassium (Kir) current in dopaminergic periglomerular neurons of the mouse olfactory bulb. Front Cell Neurosci 2014; 8:223. [PMID: 25152712 PMCID: PMC4126183 DOI: 10.3389/fncel.2014.00223] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 07/21/2014] [Indexed: 11/23/2022] Open
Abstract
Dopaminergic (DA) periglomerular (PG) neurons are critically placed at the entry of the bulbar circuitry, directly in contact with both the terminals of olfactory sensory neurons and the apical dendrites of projection neurons; they are autorhythmic and are the target of numerous terminals releasing a variety of neurotransmitters. Despite the centrality of their position, suggesting a critical role in the sensory processing, their properties -and consequently their function- remain elusive. The current mediated by inward rectifier potassium (Kir) channels in DA-PG cells was recorded by adopting the perforated-patch configuration in thin slices; IKir could be distinguished from the hyperpolarization-activated current (I h ) by showing full activation in <10 ms, no inactivation, suppression by Ba(2+) in a typical voltage-dependent manner (IC50 208 μM) and reversal potential nearly coincident with EK. Ba(2+) (2 mM) induces a large depolarization of DA-PG cells, paralleled by an increase of the input resistance, leading to a block of the spontaneous activity, but the Kir current is not an essential component of the pacemaker machinery. The Kir current is negatively modulated by intracellular cAMP, as shown by a decrease of its amplitude induced by forskolin or 8Br-cAMP. We have also tested the neuromodulatory effects of the activation of several metabotropic receptors known to be present on these cells, showing that the current can be modulated by a multiplicity of pathways, whose activation in some case increases the amplitude of the current, as can be observed with agonists of D2, muscarinic, and GABAA receptors, whereas in other cases has the opposite effect, as it can be observed with agonists of α1 noradrenergic, 5-HT and histamine receptors. These characteristics of the Kir currents provide the basis for an unexpected plasticity of DA-PG cell function, making them potentially capable to reconfigure the bulbar network to allow a better flexibility.
Collapse
Affiliation(s)
| | | | | | - Ottorino Belluzzi
- Department of Life Sciences and Biotechnology, University of FerraraFerrara, Italy
| |
Collapse
|
34
|
D'Ascenzo M, Podda MV, Grassi C. The role of D-serine as co-agonist of NMDA receptors in the nucleus accumbens: relevance to cocaine addiction. Front Synaptic Neurosci 2014; 6:16. [PMID: 25076900 PMCID: PMC4100571 DOI: 10.3389/fnsyn.2014.00016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 06/29/2014] [Indexed: 12/20/2022] Open
Abstract
Cocaine addiction is characterized by compulsive drug use despite adverse consequences and high rate of relapse during periods of abstinence. Increasing consensus suggests that addiction to drugs of abuse usurps learning and memory mechanisms normally related to natural rewards, ultimately producing long-lasting neuroadaptations in the mesocorticolimbic system. This system, formed in part by the ventral tegmental area and nucleus accumbens (NAc), has a central role in the development and expression of addictive behaviors. In addition to a broad spectrum of changes that affect morphology and function of NAc excitatory circuits in cocaine–treated animals, impaired N-methyl-D-aspartate receptor (NMDAR)-dependent synaptic plasticity is a typical feature. D-serine, a D-amino acid that has been found at high levels in mammalian brain, binds with high affinity the co-agonist site of NMDAR and mediates, along with glutamate, several important processes including synaptic plasticity. Here we review recent literature focusing on cocaine-induced impairment in synaptic plasticity mechanisms in the NAc and on the fundamental role of D-serine as co-agonist of NMDAR in functional and dysfunctional synaptic plasticity within this nucleus. The emerging picture is that reduced D-serine levels play a crucial role in synaptic plasticity relevant to cocaine addiction. This finding opens new perspectives for therapeutic approaches to treat this addictive state.
Collapse
Affiliation(s)
- Marcello D'Ascenzo
- Institute of Human Physiology, Medical School, Universitá Cattolica "S. Cuore" Rome, Italy
| | - Maria Vittoria Podda
- Institute of Human Physiology, Medical School, Universitá Cattolica "S. Cuore" Rome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Medical School, Universitá Cattolica "S. Cuore" Rome, Italy
| |
Collapse
|
35
|
Piccart E, De Backer JF, Gall D, Lambot L, Raes A, Vanhoof G, Schiffmann S, D’Hooge R. Genetic deletion of PDE10A selectively impairs incentive salience attribution and decreases medium spiny neuron excitability. Behav Brain Res 2014; 268:48-54. [DOI: 10.1016/j.bbr.2014.03.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 03/02/2014] [Accepted: 03/12/2014] [Indexed: 01/04/2023]
|
36
|
Zhang L, Bose P, Warren RA. Dopamine preferentially inhibits NMDA receptor-mediated EPSCs by acting on presynaptic D1 receptors in nucleus accumbens during postnatal development. PLoS One 2014; 9:e86970. [PMID: 24784836 PMCID: PMC4006738 DOI: 10.1371/journal.pone.0086970] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 12/19/2013] [Indexed: 11/23/2022] Open
Abstract
Nucleus accumbens (nAcb), a major site of action of drugs of abuse and dopamine (DA) signalling in MSNs (medium spiny neurons), is critically involved in mediating behavioural responses of drug addiction. Most studies have evaluated the effects of DA on MSN firing properties but thus far, the effects of DA on a cellular circuit involving glutamatergic afferents to the nAcb have remained rather elusive. In this study we attempted to characterize the effects of dopamine (DA) on evoked glutamatergic excitatory postsynaptic currents (EPSCs) in nAcb medium spiny (MS) neurons in 1 to 21 day-old rat pups. The EPSCs evoked by local nAcb stimuli displayed both AMPA/KA and NMDA receptor-mediated components. The addition of DA to the superfusing medium produced a marked decrease of both components of the EPSCs that did not change during the postnatal period studied. Pharmacologically isolated AMPA/KA receptor-mediated response was inhibited on average by 40% whereas the isolated NMDA receptor-mediated EPSC was decreased by 90%. The effect of DA on evoked EPSCs were mimicked by the D1-like receptor agonist SKF 38393 and antagonized by the D1-like receptor antagonist SCH 23390 whereas D2-like receptor agonist or antagonist respectively failed to mimic or to block the action of DA. DA did not change the membrane input conductance of MS neurons or the characteristics of EPSCs produced by the local administration of glutamate in the presence of tetrodotoxin. In contrast, DA altered the paired-pulse ratio of evoked EPSCs. The present results show that the activation D1-like dopaminergic receptors modulate glutamatergic neurotransmission by preferentially inhibiting NMDA receptor-mediated EPSC through presynaptic mechanisms.
Collapse
Affiliation(s)
- Liming Zhang
- Centre de recherche Fernand-Seguin, University of Montreal, Montreal, Canada
- Department of Physiology, University of Montreal, Montreal, Canada
| | - Poulomee Bose
- Department of Psychiatry, University of Montreal, Montreal, Canada
| | - Richard A. Warren
- Centre de recherche Fernand-Seguin, University of Montreal, Montreal, Canada
- Department of Psychiatry, University of Montreal, Montreal, Canada
- * E-mail:
| |
Collapse
|
37
|
Maze I, Chaudhury D, Dietz DM, Von Schimmelmann M, Kennedy PJ, Lobo MK, Daws SE, Miller ML, Bagot RC, Sun H, Turecki G, Neve RL, Hurd YL, Shen L, Han MH, Schaefer A, Nestler EJ. G9a influences neuronal subtype specification in striatum. Nat Neurosci 2014; 17:533-9. [PMID: 24584053 PMCID: PMC3972624 DOI: 10.1038/nn.3670] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Accepted: 02/04/2014] [Indexed: 12/13/2022]
Abstract
Cocaine-mediated repression of the histone methyltransferase (HMT) G9a has recently been implicated in transcriptional, morphological and behavioral responses to chronic cocaine administration. Here, using a ribosomal affinity purification approach, we found that G9a repression by cocaine occurred in both Drd1-expressing (striatonigral) and Drd2-expressing (striatopallidal) medium spiny neurons. Conditional knockout and overexpression of G9a within these distinct cell types, however, revealed divergent behavioral phenotypes in response to repeated cocaine treatment. Our studies further indicated that such developmental deletion of G9a selectively in Drd2 neurons resulted in the unsilencing of transcriptional programs normally specific to striatonigral neurons and in the acquisition of Drd1-associated projection and electrophysiological properties. This partial striatopallidal to striatonigral 'switching' phenotype in mice indicates a new role for G9a in contributing to neuronal subtype identity and suggests a critical function for cell type-specific histone methylation patterns in the regulation of behavioral responses to environmental stimuli.
Collapse
Affiliation(s)
- Ian Maze
- Laboratory of Chromatin Biology and Epigenetics, The Rockefeller
University, New York, New York 10065, USA
| | - Dipesh Chaudhury
- Department of Pharmacology and Systems Therapeutics, Friedman Brain
Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029,
USA
| | - David M. Dietz
- Department of Pharmacology and Toxicology, University at Buffalo,
Buffalo, New York, 14214, USA
| | - Melanie Von Schimmelmann
- Fishberg Department of Neuroscience and Friedman Brain Institute,
Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Pamela J. Kennedy
- Department of Psychology, University of California, Los Angeles, Los
Angeles, California 90095, USA
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland
School of Medicine, Baltimore, Maryland 21201, USA
| | - Stephanie E. Daws
- Fishberg Department of Neuroscience and Friedman Brain Institute,
Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai,
New York, New York 10029, USA
| | - Michael L. Miller
- Fishberg Department of Neuroscience and Friedman Brain Institute,
Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Rosemary C. Bagot
- Fishberg Department of Neuroscience and Friedman Brain Institute,
Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - HaoSheng Sun
- Fishberg Department of Neuroscience and Friedman Brain Institute,
Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Gustavo Turecki
- Depressive Disorders Program, Douglas Mental Health University and
McGill University, Montréal, Québec H4H 1R3, Canada
| | - Rachael L. Neve
- Department of Brain and Cognitive Science, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, USA
| | - Yasmin L. Hurd
- Department of Pharmacology and Systems Therapeutics, Friedman Brain
Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029,
USA
- Fishberg Department of Neuroscience and Friedman Brain Institute,
Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai,
New York, New York 10029, USA
| | - Li Shen
- Fishberg Department of Neuroscience and Friedman Brain Institute,
Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Ming-Hu Han
- Department of Pharmacology and Systems Therapeutics, Friedman Brain
Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029,
USA
- Fishberg Department of Neuroscience and Friedman Brain Institute,
Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Anne Schaefer
- Fishberg Department of Neuroscience and Friedman Brain Institute,
Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Eric J. Nestler
- Department of Pharmacology and Systems Therapeutics, Friedman Brain
Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029,
USA
- Fishberg Department of Neuroscience and Friedman Brain Institute,
Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
38
|
Zaika OL, Mamenko M, Palygin O, Boukelmoune N, Staruschenko A, Pochynyuk O. Direct inhibition of basolateral Kir4.1/5.1 and Kir4.1 channels in the cortical collecting duct by dopamine. Am J Physiol Renal Physiol 2013; 305:F1277-F1287. [PMID: 23986512 PMCID: PMC3840222 DOI: 10.1152/ajprenal.00363.2013] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 08/16/2013] [Indexed: 12/11/2022] Open
Abstract
It is recognized that dopamine promotes natriuresis by inhibiting multiple transporting systems in the proximal tubule. In contrast, less is known about the molecular targets of dopamine actions on water-electrolyte transport in the cortical collecting duct (CCD). Epithelial cells in the CCD are exposed to dopamine, which is synthesized locally or secreted from sympathetic nerve endings. Basolateral K(+) channels in the distal renal tubule are critical for K(+) recycling and controlling basolateral membrane potential to establish the driving force for Na(+) reabsorption. Here, we demonstrate that Kir4.1 and Kir5.1 are highly expressed in the mouse kidney cortex and are localized to the basolateral membrane of the CCD. Using patch-clamp electrophysiology in freshly isolated CCDs, we detected highly abundant 40-pS and scarce 20-pS single channel conductances, most likely representing Kir4.1/5.1 and Kir4.1 channels, respectively. Dopamine reversibly decreased the open probability of both channels, with a relatively greater action on the Kir4.1/5.1 heterodimer. This effect was mediated by D2-like but not D1-like dopamine receptors. PKC blockade abolished the inhibition of basolateral K(+) channels by dopamine. Importantly, dopamine significantly decreased the amplitude of Kir4.1/5.1 and Kir4.1 unitary currents. Consistently, dopamine induced an acute depolarization of basolateral membrane potential, as directly monitored using current-clamp mode in isolated CCDs. Therefore, we demonstrate that dopamine inhibits basolateral Kir4.1/5.1 and Kir4.1 channels in CCD cells via stimulation of D2-like receptors and subsequently PKC. This leads to depolarization of the basolateral membrane and a decreased driving force for Na(+) reabsorption in the distal renal tubule.
Collapse
Affiliation(s)
- Oleg L Zaika
- Dept. of Integrative Biology and Pharmacology, Univ. of Texas Health Science Center, 6431 Fannin St., Houston, TX 77030.
| | | | | | | | | | | |
Collapse
|
39
|
Podda MV, Grassi C. New perspectives in cyclic nucleotide-mediated functions in the CNS: the emerging role of cyclic nucleotide-gated (CNG) channels. Pflugers Arch 2013; 466:1241-57. [PMID: 24142069 DOI: 10.1007/s00424-013-1373-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Revised: 09/27/2013] [Accepted: 09/28/2013] [Indexed: 01/07/2023]
Abstract
Cyclic nucleotides play fundamental roles in the central nervous system (CNS) under both physiological and pathological conditions. The impact of cAMP and cGMP signaling on neuronal and glial cell functions has been thoroughly characterized. Most of their effects have been related to cyclic nucleotide-dependent protein kinase activity. However, cyclic nucleotide-gated (CNG) channels, first described as key mediators of sensory transduction in retinal and olfactory receptors, have been receiving increasing attention as possible targets of cyclic nucleotides in the CNS. In the last 15 years, consistent evidence has emerged for their expression in neurons and astrocytes of the rodent brain. Far less is known, however, about the functional role of CNG channels in these cells, although several of their features, such as Ca(2+) permeability and prolonged activation in the presence of cyclic nucleotides, make them ideal candidates for mediators of physiological functions in the CNS. Here, we review literature suggesting the involvement of CNG channels in a number of CNS cellular functions (e.g., regulation of membrane potential, neuronal excitability, and neurotransmitter release) as well as in more complex phenomena, like brain plasticity, adult neurogenesis, and pain sensitivity. The emerging picture is that functional and dysfunctional cyclic nucleotide signaling in the CNS has to be reconsidered including CNG channels among possible targets. However, concerted efforts and multidisciplinary approaches are still needed to get more in-depth knowledge in this field.
Collapse
Affiliation(s)
- Maria Vittoria Podda
- Institute of Human Physiology, Medical School, Università Cattolica, Largo Francesco Vito 1, 00168, Rome, Italy
| | | |
Collapse
|
40
|
Hamasaki R, Shirasaki T, Soeda F, Takahama K. Tipepidine activates VTA dopamine neuron via inhibiting dopamine D₂ receptor-mediated inward rectifying K⁺ current. Neuroscience 2013; 252:24-34. [PMID: 23896570 DOI: 10.1016/j.neuroscience.2013.07.044] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 06/28/2013] [Accepted: 07/18/2013] [Indexed: 11/19/2022]
Abstract
We previously reported that the novel antidepressant-like effect of tipepidine may be produced at least partly through the activation of mesolimbic dopamine (DA) neurons via inhibiting G protein-coupled inwardly rectifying potassium (GIRK) channels. In this study, we investigated the action of tipepidine on DA D2 receptor-mediated GIRK currents (IDA(GIRK)) and membrane excitability in DA neurons using the voltage clamp and current clamp modes of the patch-clamp techniques, respectively. DA neurons were acutely dissociated from the ventral tegmental area (VTA) in rats and identified by the presence of the hyperpolarization-activated currents. Tipepidine reversibly inhibited IDA(GIRK) with IC50 7.0 μM and also abolished IDA(GIRK) irreversibly activated in the presence of intracellular GTPγS. Then tipepidine depolarized membrane potential and generated action potentials in the neurons current-clamped. Furthermore, the drug at 40 mg/kg, i.p. increased the number of cells immunopositive both for c-Fos and tyrosine hydroxylase (TH) in the VTA. These results suggest that tipepidine may activate DA neurons in VTA through the inhibition of GIRK channel-activated currents.
Collapse
Affiliation(s)
- R Hamasaki
- Department of Environmental and Molecular Health Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto 862-0973, Japan
| | | | | | | |
Collapse
|
41
|
Li J, Li J, Liu X, Qin S, Guan Y, Liu Y, Cheng Y, Chen X, Li W, Wang S, Xiong M, Kuzhikandathil EV, Ye JH, Zhang C. MicroRNA expression profile and functional analysis reveal that miR-382 is a critical novel gene of alcohol addiction. EMBO Mol Med 2013; 5:1402-14. [PMID: 23873704 PMCID: PMC3799494 DOI: 10.1002/emmm.201201900] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 05/02/2013] [Accepted: 06/17/2013] [Indexed: 12/31/2022] Open
Abstract
Alcohol addiction is a major social and health concern. Here, we determined the expression profile of microRNAs (miRNAs) in the nucleus accumbens (NAc) of rats treated with alcohol. The results suggest that multiple miRNAs were aberrantly expressed in rat NAc after alcohol injection. Among them, miR-382 was down-regulated in alcohol-treated rats. In both cultured neuronal cells in vitro and in the NAc in vivo, we identified that the dopamine receptor D1 (Drd1) is a direct target gene of miR-382. Via this target gene, miR-382 strongly modulated the expression of DeltaFosB. Moreover, overexpression of miR-382 significantly attenuated alcohol-induced up-regulation of DRD1 and DeltaFosB, decreased voluntary intake of and preference for alcohol and inhibited the DRD1-induced action potential responses. The results indicated that miRNAs are involved in and may represent novel therapeutic targets for alcoholism.
Collapse
Affiliation(s)
- Jingyuan Li
- Department of Pharmacology, Rush University Medical Center, Rush University, Chicago, IL, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Cilz NI, Kurada L, Hu B, Lei S. Dopaminergic modulation of GABAergic transmission in the entorhinal cortex: concerted roles of α1 adrenoreceptors, inward rectifier K⁺, and T-type Ca²⁺ channels. Cereb Cortex 2013; 24:3195-208. [PMID: 23843440 DOI: 10.1093/cercor/bht177] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Whereas the entorhinal cortex (EC) receives profuse dopaminergic innervations from the midbrain, the effects of dopamine (DA) on γ-Aminobutyric acid (GABA)ergic interneurons in this brain region have not been determined. We probed the actions of DA on GABAA receptor-mediated synaptic transmission in the EC. Application of DA increased the frequency, not the amplitude, of spontaneous IPSCs (sIPSCs) and miniature IPSCs (mIPSCs) recorded from entorhinal principal neurons, but slightly reduced the amplitude of the evoked IPSCs. The effects of DA were unexpectedly found to be mediated by α1 adrenoreceptors, but not by DA receptors. DA endogenously released by the application of amphetamine also increased the frequency of sIPSCs. Ca(2+) influx via T-type Ca(2+) channels was required for DA-induced facilitation of sIPSCs and mIPSCs. DA depolarized and enhanced the firing frequency of action potentials of interneurons. DA-induced depolarization was independent of extracellular Na(+) and Ca(2+) and did not require the functions of hyperpolarization-activated (Ih) channels and T-type Ca(2+) channels. DA-generated currents showed a reversal potential close to the K(+) reversal potential and inward rectification, suggesting that DA inhibits the inward rectifier K(+) channels (Kirs). Our results demonstrate that DA facilitates GABA release by activating α1 adrenoreceptors to inhibit Kirs, which further depolarize interneurons resulting in secondary Ca(2+) influx via T-type Ca(+) channels.
Collapse
Affiliation(s)
- Nicholas I Cilz
- Department of Pharmacology, Physiology and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Lalitha Kurada
- Department of Pharmacology, Physiology and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Binqi Hu
- Department of Pharmacology, Physiology and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Saobo Lei
- Department of Pharmacology, Physiology and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| |
Collapse
|
43
|
Curcio L, Podda MV, Leone L, Piacentini R, Mastrodonato A, Cappelletti P, Sacchi S, Pollegioni L, Grassi C, D'Ascenzo M. Reduced D-serine levels in the nucleus accumbens of cocaine-treated rats hinder the induction of NMDA receptor-dependent synaptic plasticity. ACTA ACUST UNITED AC 2013; 136:1216-30. [PMID: 23518710 DOI: 10.1093/brain/awt036] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cocaine seeking behaviour and relapse have been linked to impaired potentiation and depression at excitatory synapses in the nucleus accumbens, but the mechanism underlying this process is poorly understood. We show that, in the rat nucleus accumbens core, D-serine is the endogenous coagonist of N-methyl-D-aspartate receptors, and its presence is essential for N-methyl-D-aspartate receptor-dependent potentiation and depression of synaptic transmission. Nucleus accumbens core slices obtained from cocaine-treated rats after 1 day of abstinence presented significantly reduced D-serine concentrations, increased expression of the D-serine degrading enzyme, D-amino acid oxidase, and downregulated expression of serine racemase, the enzyme responsible for D-serine synthesis. The D-serine deficit was associated with impairment of potentiation and depression of glutamatergic synaptic transmission, which was restored by slice perfusion with exogenous D-serine. Furthermore, in vivo administration of D-serine directly into the nucleus accumbens core blocked behavioural sensitization to cocaine. These results provide evidence for a critical role of D-serine signalling in synaptic plasticity relevant to cocaine addiction.
Collapse
Affiliation(s)
- Livia Curcio
- Institute of Human Physiology, Medical School, Università Cattolica, 00168 Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Dvorzhak A, Semtner M, Faber DS, Grantyn R. Tonic mGluR5/CB1-dependent suppression of inhibition as a pathophysiological hallmark in the striatum of mice carrying a mutant form of huntingtin. J Physiol 2012; 591:1145-66. [PMID: 23230231 DOI: 10.1113/jphysiol.2012.241018] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Changes in the activity of striatal output neurons (SONs) have been implicated in the pathogenesis of Huntington's disease (HD). In this inherited polyglutamine disorder, accumulation of intracellular toxins causes a variety of deficits, including synaptic dysfunction, but it is still unclear to what extent striatal GABA release is afflicted as well. Two murine HD models were used, a recently created knock-in mouse (Z_Q175_KI) and an established model of HD (R6/2). In sagittal slices with relatively well-preserved glutamatergic connections throughout the basal ganglia, we have characterized the following: (i) the excitability of SONs; (ii) their spontaneous action potential-dependent GABAergic synaptic activity; (iii) the capacity of exogenous GABA to inhibit spontaneous action potential generation; and (iv) the properties of GABAergic unitary evoked responses (eIPSCs) in response to intrastriatal minimal stimulation at low and high frequency. The HD SONs exhibited enhanced intrisic excitability and higher levels of GABAergic spontaneous activity without presenting evidence for homeostatic upregulation of endogenous or exogenous GABA actions. Unitary eIPSC amplitudes were reduced, with a clear deficit in the probability of release, as indicated by a higher paired-pulse ratio, failure rate and coefficient of variation. In conditions of high-frequency activation, GABAergic connections of HD SONs were prone to asynchronous release and delayed IPSC generation at the expense of synchronized release. Both in wild-type and in HD SONs, GABA was inhibitory. Our results support the conclusion that the enhanced spontaneous synaptic activity in the HD striatum reflects disinhibition. Pharmacological tests identified the HD-related tonic suppression of synaptic inhibition as a glutamate- and endocannabinoid-dependent process.
Collapse
Affiliation(s)
- Anton Dvorzhak
- Cluster of Excellence Neurocure and Department of Experimental Neurology, University Medicine Charitè, Berlin, Germany
| | | | | | | |
Collapse
|
45
|
Austgen JR, Dantzler HA, Barger BK, Kline DD. 5-hydroxytryptamine 2C receptors tonically augment synaptic currents in the nucleus tractus solitarii. J Neurophysiol 2012; 108:2292-305. [PMID: 22855775 PMCID: PMC3545023 DOI: 10.1152/jn.00049.2012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 07/29/2012] [Indexed: 12/20/2022] Open
Abstract
The nucleus tractus solitarii (nTS) is the primary termination and integration point for visceral afferents in the brain stem. Afferent glutamate release and its efficacy on postsynaptic activity within this nucleus are modulated by additional neuromodulators and transmitters, including serotonin (5-HT) acting through its receptors. The 5-HT(2) receptors in the medulla modulate the cardiorespiratory system and autonomic reflexes, but the distribution of the 5-HT(2C) receptor and the role of these receptors during synaptic transmission in the nTS remain largely unknown. In the present study, we examined the distribution of 5-HT(2C) receptors in the nTS and their role in modulating excitatory postsynaptic currents (EPSCs) in monosynaptic nTS neurons in the horizontal brain stem slice. Real-time RT-PCR and immunohistochemistry identified 5-HT(2C) receptor message and protein in the nTS and suggested postsynaptic localization. In nTS neurons innervated by general visceral afferents, 5-HT(2C) receptor activation increased solitary tract (TS)-EPSC amplitude and input resistance and depolarized membrane potential. Conversely, 5-HT(2C) receptor blockade reduced TS-EPSC and miniature EPSC amplitude, as well as input resistance, and hyperpolarized membrane potential. Synaptic parameters in nTS neurons that receive sensory input from carotid body chemoafferents were also attenuated by 5-HT(2C) receptor blockade. Taken together, these data suggest that 5-HT(2C) receptors in the nTS are located postsynaptically and augment excitatory neurotransmission.
Collapse
Affiliation(s)
- James R Austgen
- Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | | | | | | |
Collapse
|
46
|
Involvement of nucleus accumbens dopamine D1 receptors in ethanol drinking, ethanol-induced conditioned place preference, and ethanol-induced psychomotor sensitization in mice. Psychopharmacology (Berl) 2012; 222:141-53. [PMID: 22222864 DOI: 10.1007/s00213-011-2630-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 12/19/2011] [Indexed: 12/24/2022]
Abstract
RATIONALE Dopamine D1 receptor (D1R) signaling has been associated to ethanol consumption and reward in laboratory animals. OBJECTIVES Here, we hypothesize that this receptor, which is located within the nucleus accumbens (NAc) neurons, modulates alcohol reward mechanisms. METHODS To test this hypothesis, we measured alcohol consumption and ethanol-induced psychomotor sensitization and conditioned place preference (CPP) in mice that received bilateral microinjections of small interference RNA (siRNA)-expressing lentiviral vectors (LV-siD1R) producing D1R knock-down. The other group received control (LV-Mock) viral vectors into the NAc. RESULTS There were no differences in the total fluid consumed and also no differences in the amount of ethanol consumed between groups prior to surgery. However, after surgery, the LV-siD1R group consumed less ethanol than the control group. This difference was not associated to taste neophobia. In addition, results have shown that down-regulation of endogenous D1R using viral-mediated siRNA in the NAc significantly decreased ethanol-induced behavioral sensitization as well as acquisition, but not expression, of ethanol-induced place preference. CONCLUSIONS We conclude that decreased D1R expression into the NAc led to reduced ethanol rewarding properties, thereby leading to lower voluntary ethanol consumption. Together, these findings demonstrate that the D1 receptor pathway within the NAc controls ethanol reward and intake.
Collapse
|
47
|
Sharopov S, Moser J, Chen R, Kolbaev SN, Bernedo VE, Werhahn KJ, Luhmann HJ, Kilb W. Dopaminergic modulation of low-Mg2+-induced epileptiform activity in the intact hippocampus of the newborn mouse in vitro. J Neurosci Res 2012; 90:2020-33. [DOI: 10.1002/jnr.23084] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 04/11/2012] [Accepted: 04/13/2012] [Indexed: 11/12/2022]
|
48
|
Effects of protein kinase A inhibitor and activator on rewarding effects of SKF-82958 microinjected into nucleus accumbens shell of ad libitum fed and food-restricted rats. Psychopharmacology (Berl) 2012; 221:589-99. [PMID: 22143580 PMCID: PMC3310955 DOI: 10.1007/s00213-011-2602-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Accepted: 11/27/2011] [Indexed: 10/14/2022]
Abstract
RATIONALE Previous studies indicate that the rewarding effect of D-1 dopamine receptor stimulation in nucleus accumbens (NAc) shell is greater in food-restricted (FR) than in ad libitum fed (AL) rats. The D-1 receptor is positively coupled to adenylyl cyclase and activates protein kinase A (PKA). OBJECTIVES The purpose of this study was to determine whether PKA is involved in the rewarding effect of D-1 receptor stimulation and, if so, whether it is involved in the enhanced response of FR rats. MATERIALS AND METHODS Rats were stereotaxically implanted with microinjection cannulae in NAc shell and a stimulating electrode in lateral hypothalamus. The rewarding effects of SKF-82958 (1.5 or 3.0 μg, bilaterally) in the presence and absence of PKA inhibitor, Rp-cAMPS (8.9 μg), and PKA activator, Sp-cAMPS (8.9 μg), were assessed using the curve-shift method of intracranial self-stimulation (ICSS). Basal NAc levels of DARPP-32 phosphorylated on Thr34 and Thr75 were measured. RESULTS Rp-cAMPS increased the rewarding effect of SKF-82958 in AL but not FR rats, doubling the ICSS threshold-lowering effect of the 3.0-μg dose. Sp-cAMPS decreased the rewarding effect of SKF-82958 in FR but not AL rats. Levels of phospho-DARPP-32 (Thr75), which inhibits PKA, were higher in FR than AL rats. CONCLUSIONS Results indicate that inhibition of PKA enhances the unconditioned rewarding effect of D-1 receptor stimulation and that decreased PKA may be involved in the effect of FR on drug reward. Evidence for involvement of D-2 receptor-expressing neurons in the enhancing effect of PKA inhibition is discussed.
Collapse
|
49
|
Podda MV, Leone L, Piacentini R, Cocco S, Mezzogori D, D'Ascenzo M, Grassi C. Expression of olfactory-type cyclic nucleotide-gated channels in rat cortical astrocytes. Glia 2012; 60:1391-405. [PMID: 22653779 DOI: 10.1002/glia.22360] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 05/03/2012] [Indexed: 12/31/2022]
Abstract
Cyclic nucleotide-gated (CNG) channels are nonselective cation channels activated by cyclic AMP (cAMP) or cyclic GMP (cGMP). They were originally identified in retinal and olfactory receptors, but evidence has also emerged for their expression in several mammalian brain areas. Because cGMP and cAMP control important aspects of glial cell physiology, we wondered whether CNG channels are expressed in astrocytes, the most functionally relevant glial cells in the CNS. Immunoblot and immunofluorescence experiments demonstrated expression of the CNG channel olfactory-type A subunit, CNGA2, in cultured rat cortical astrocytes. In patch-clamp experiments, currents elicited in these cells by voltage ramps from -100 to +100 mV in the presence of the cGMP analogue, dB-cGMP, were significantly reduced by the CNG channel blockers, L-cis-diltiazem (LCD) and Cd(2+) . The reversal potentials of the LCD- and Cd(2+) -sensitive currents were more positive than that of K(+) , as expected for a mixed cation current. Noninactivating, voltage-independent currents were also elicited by extracellular application of the membrane permeant cGMP analogue, 8-Br-cGMP. These effects were blocked by LCD and were mimicked by natriuretic peptide receptor activation and inhibition of phosphodiesterase activity. Voltage-independent, LCD-sensitive currents were also elicited by 8-Br-cGMP in astrocytes of hippocampal and neocortical brain slices. Immunohistochemistry confirmed a broad distribution of CNG channels in astrocytes of the rat forebrain, midbrain, and hindbrain. These findings suggest that CNG channels are downstream targets of cyclic nucleotides in astrocytes, and they may be involved in the glial-mediated regulation of CNS functions under physiological and pathological conditions.
Collapse
Affiliation(s)
- Maria Vittoria Podda
- Institute of Human Physiology, Medical School, Università Cattolica, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
50
|
Ranieri F, Podda MV, Riccardi E, Frisullo G, Dileone M, Profice P, Pilato F, Di Lazzaro V, Grassi C. Modulation of LTP at rat hippocampal CA3-CA1 synapses by direct current stimulation. J Neurophysiol 2012; 107:1868-80. [PMID: 22236710 DOI: 10.1152/jn.00319.2011] [Citation(s) in RCA: 160] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Transcranial direct current stimulation (tDCS) can produce a lasting polarity-specific modulation of cortical excitability in the brain, and it is increasingly used in experimental and clinical settings. Recent studies suggest that the after-effects of tDCS are related to molecular mechanisms of activity-dependent synaptic plasticity. Here we investigated the effect of DCS on the induction of one of the most studied N-methyl-d-aspartate receptor-dependent forms of long-term potentiation (LTP) of synaptic activity at CA3-CA1 synapses in the hippocampus. We show that DCS applied to rat brain slices determines a modulation of LTP that is increased by anodal and reduced by cathodal DCS. Immediate early genes, such as c-fos and zif268 (egr1/NGFI-A/krox24), are rapidly induced following neuronal activation, and a specific role of zif268 in the induction and maintenance of LTP has been demonstrated. We found that both anodal and cathodal DCS produce a marked subregion-specific increase in the expression of zif268 protein in the cornus ammonis (CA) region, whereas the same protocols of stimulation produce a less pronounced increase in c-fos protein expression in the CA and in dentate gyrus regions of the hippocampus. Brain-derived neurotrophic factor expression was also investigated, and it was found to be reduced in cathodal-stimulated slices. The present data demonstrate that it is possible to modulate LTP by using DCS and provide the rationale for the use of DCS in neurological diseases to promote the adaptive and suppress the maladaptive forms of brain plasticity.
Collapse
Affiliation(s)
- F Ranieri
- Istituto di Neurologia, Università Cattolica, L.go A. Gemelli 8, 00168 Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|