1
|
Requejo-Mendoza N, Arias-Montaño JA, Gutierrez R. Nucleus accumbens D2-expressing neurons: Balancing reward and licking disruption through rhythmic optogenetic stimulation. PLoS One 2025; 20:e0317605. [PMID: 39919051 PMCID: PMC11805367 DOI: 10.1371/journal.pone.0317605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/31/2024] [Indexed: 02/09/2025] Open
Abstract
Nucleus accumbens (NAc) dopamine D1 receptor-expressing neurons are known to be critical for processing reward and regulating food intake. However, the role of D2-expressing neurons in this nucleus remains less understood. This study employed optogenetic manipulations to investigate the role of NAc D2-expressing neurons in reward processing and sucrose consumption. Optogenetic activation of these neurons decreased sucrose preference (at 20 Hz), disrupted licking patterns (particularly at 8 and 20 Hz), and increased self-stimulation. Conversely, synchronizing stimulation with the animal licking rhythm mitigated licking disruption and even increased sucrose intake, suggesting a rewarding effect. Furthermore, 20 Hz stimulation (but not 8 Hz) induced place preference in a real-time place preference (RTPP) test. In contrast, inhibiting D2 neurons produced a negative hedonic state, although not reaching complete aversion, influencing food choices in specific contexts. For instance, while the RTPP test per se was not sensitive enough to observe place aversion when mice could choose between consuming a high-fat diet (HFD) pellet in a context associated with or without inhibition of D2 neurons, they preferred to consume HFD on the non-inhibited side. This suggests that the palatability of HFD can unmask (but also overshadow) the negative hedonic state associated with D2 neuron inhibition. A negative reinforcement paradigm further confirmed the active avoidance behavior induced by D2 neuron inhibition. In conclusion, NAc D2 neuron inhibition induces a negative hedonic state, while activation has a dual effect-it is rewarding yet disrupts licking behavior-highlighting its complex role in reward and consummatory behavior. Importantly, self-paced stimulation, where the animal controls the timing of the stimulation through its licking behavior, offers a more efficient and natural approach for stimulating NAc activity.
Collapse
Affiliation(s)
- Nikte Requejo-Mendoza
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados (Cinvestav), Ciudad de México, México
- Laboratory Neurobiology of Appetite; Departamento Farmacología, Centro de Investigación y de Estudios Avanzados (Cinvestav), Ciudad de México, México
| | - José-Antonio Arias-Montaño
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados (Cinvestav), Ciudad de México, México
| | - Ranier Gutierrez
- Laboratory Neurobiology of Appetite; Departamento Farmacología, Centro de Investigación y de Estudios Avanzados (Cinvestav), Ciudad de México, México
- Laboratory Neurobiology of Appetite; Centro de Investigación sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados (CIE, Cinvestav Sede sur), Ciudad de México, México
| |
Collapse
|
2
|
Tavakoli NS, Malone SG, Anderson TL, Neeley RE, Asadipooya A, Bardo MT, Ortinski PI. Astrocyte Ca 2+ in the dorsal striatum suppresses neuronal activity to oppose cue-induced reinstatement of cocaine seeking. Front Cell Neurosci 2024; 18:1347491. [PMID: 39280793 PMCID: PMC11393831 DOI: 10.3389/fncel.2024.1347491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 08/12/2024] [Indexed: 09/18/2024] Open
Abstract
Recent literature supports a prominent role for astrocytes in regulation of drug-seeking behaviors. The dorsal striatum, specifically, is known to play a role in reward processing with neuronal activity that can be influenced by astrocyte Ca2+. However, the manner in which Ca2+ in dorsal striatum astrocytes impacts neuronal signaling after exposure to self-administered cocaine remains unclear. We addressed this question following over-expression of the Ca2+ extrusion pump, hPMCA2w/b, in dorsal striatum astrocytes and the Ca2+ indicator, GCaMP6f, in dorsal striatum neurons of rats that were trained to self-administer cocaine. Following extinction of cocaine-seeking behavior, the rats over-expressing hMPCA2w/b showed a significant increase in cue-induced reinstatement of cocaine seeking. Suppression of astrocyte Ca2+ increased the amplitude of neuronal Ca2+ transients in brain slices, but only after cocaine self-administration. This was accompanied by decreased duration of neuronal Ca2+ events in the cocaine group and no changes in Ca2+ event frequency. Acute administration of cocaine to brain slices decreased amplitude of neuronal Ca2+ in both the control and cocaine self-administration groups regardless of hPMCA2w/b expression. These results indicated that astrocyte Ca2+ control over neuronal Ca2+ transients was enhanced by cocaine self-administration experience, although sensitivity to acutely applied cocaine remained comparable across all groups. To explore this further, we found that neither the hMPCA2w/b expression nor the cocaine self-administration experience altered regulation of neuronal Ca2+ events by NPS-2143, a Ca2+ sensing receptor (CaSR) antagonist, suggesting that plasticity of neuronal signaling after hPMCA2w/b over-expression was unlikely to result from elevated extracellular Ca2+. We conclude that astrocyte Ca2+ in the dorsal striatum impacts neurons via cell-intrinsic mechanisms (e.g., gliotransmission, metabolic coupling, etc.) and impacts long-term neuronal plasticity after cocaine self-administration differently from neuronal response to acute cocaine. Overall, astrocyte Ca2+ influences neuronal output in the dorsal striatum to promote resistance to cue-induced reinstatement of cocaine seeking.
Collapse
Affiliation(s)
- Navid S Tavakoli
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Samantha G Malone
- Department of Psychology, University of Kentucky, Lexington, KY, United States
| | - Tanner L Anderson
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Ryson E Neeley
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Artin Asadipooya
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Michael T Bardo
- Department of Psychology, University of Kentucky, Lexington, KY, United States
| | - Pavel I Ortinski
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
3
|
Aceto G, Nardella L, Nanni S, Pecci V, Bertozzi A, Nutarelli S, Viscomi MT, Colussi C, D'Ascenzo M, Grassi C. Glycine-induced activation of GPR158 increases the intrinsic excitability of medium spiny neurons in the nucleus accumbens. Cell Mol Life Sci 2024; 81:268. [PMID: 38884814 PMCID: PMC11335193 DOI: 10.1007/s00018-024-05260-w] [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: 02/02/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 06/18/2024]
Abstract
It has been recently established that GPR158, a class C orphan G protein-coupled receptor, serves as a metabotropic glycine receptor. GPR158 is highly expressed in the nucleus accumbens (NAc), a major input structure of the basal ganglia that integrates information from cortical and subcortical structures to mediate goal-directed behaviors. However, whether glycine modulates neuronal activity in the NAc through GPR158 activation has not been investigated yet. Using whole-cell patch-clamp recordings, we found that glycine-dependent activation of GPR158 increased the firing rate of NAc medium spiny neurons (MSNs) while it failed to significantly affect the excitability of cholinergic interneurons (CIN). In MSNs GPR158 activation reduced the latency to fire, increased the action potential half-width, and reduced action potential afterhyperpolarization, effects that are all consistent with negative modulation of potassium M-currents, that in the central nervous system are mainly carried out by Kv7/KCNQ-channels. Indeed, we found that the GPR158-induced increase in MSN excitability was associated with decreased M-current amplitude, and selective pharmacological inhibition of the M-current mimicked and occluded the effects of GPR158 activation. In addition, when the protein kinase A (PKA) or extracellular signal-regulated kinase (ERK) signaling was pharmacologically blocked, modulation of MSN excitability by GPR158 activation was suppressed. Moreover, GPR158 activation increased the phosphorylation of ERK and Kv7.2 serine residues. Collectively, our findings suggest that GPR158/PKA/ERK signaling controls MSN excitability via Kv7.2 modulation. Glycine-dependent activation of GPR158 may significantly affect MSN firing in vivo, thus potentially mediating specific aspects of goal-induced behaviors.
Collapse
Affiliation(s)
- Giuseppe Aceto
- Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, 00168, Italy
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
| | - Luca Nardella
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
| | - Simona Nanni
- Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, 00168, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
| | - Valeria Pecci
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
| | - Alessia Bertozzi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
- Istituto di Analisi dei Sistemi ed Informatica "Antonio Ruberti", National Research Council, Rome, Italy
| | - Sofia Nutarelli
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
| | - Maria Teresa Viscomi
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
| | - Claudia Colussi
- Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, 00168, Italy
- Istituto di Analisi dei Sistemi ed Informatica "Antonio Ruberti", National Research Council, Rome, Italy
| | - Marcello D'Ascenzo
- Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, 00168, Italy.
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, 00168, Italy.
| | - Claudio Grassi
- Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, 00168, Italy
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
| |
Collapse
|
4
|
Bezerra TO, Roque AC, Salum C. A Computational Model for the Simulation of Prepulse Inhibition and Its Modulation by Cortical and Subcortical Units. Brain Sci 2024; 14:502. [PMID: 38790479 PMCID: PMC11118907 DOI: 10.3390/brainsci14050502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/10/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024] Open
Abstract
The sensorimotor gating is a nervous system function that modulates the acoustic startle response (ASR). Prepulse inhibition (PPI) phenomenon is an operational measure of sensorimotor gating, defined as the reduction of ASR when a high intensity sound (pulse) is preceded in milliseconds by a weaker stimulus (prepulse). Brainstem nuclei are associated with the mediation of ASR and PPI, whereas cortical and subcortical regions are associated with their modulation. However, it is still unclear how the modulatory units can influence PPI. In the present work, we developed a computational model of a neural circuit involved in the mediation (brainstem units) and modulation (cortical and subcortical units) of ASR and PPI. The activities of all units were modeled by the leaky-integrator formalism for neural population. The model reproduces basic features of PPI observed in experiments, such as the effects of changes in interstimulus interval, prepulse intensity, and habituation of ASR. The simulation of GABAergic and dopaminergic drugs impaired PPI by their effects over subcortical units activity. The results show that subcortical units constitute a central hub for PPI modulation. The presented computational model offers a valuable tool to investigate the neurobiology associated with disorder-related impairments in PPI.
Collapse
Affiliation(s)
- Thiago Ohno Bezerra
- Center of Mathematics, Computation and Cognition, Universidade Federal do ABC, São Bernardo do Campo 09606-045, Brazil
| | - Antonio C. Roque
- Department of Physics, School of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-901, Brazil
| | - Cristiane Salum
- Center of Mathematics, Computation and Cognition, Universidade Federal do ABC, São Bernardo do Campo 09606-045, Brazil
- Interdisciplinary Applied Neuroscience Unit, Universidade Federal do ABC, São Bernardo do Campo 09606-045, Brazil
| |
Collapse
|
5
|
Valyear MD, Brown A, Deyab G, Villaruel FR, Lahlou S, Caporicci-Dinucci N, Chaudhri N. Augmenting glutamatergic, but not dopaminergic, activity in the nucleus accumbens shell disrupts responding to a discrete alcohol cue in an alcohol context. Eur J Neurosci 2024; 59:1500-1518. [PMID: 38185906 DOI: 10.1111/ejn.16231] [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: 09/08/2023] [Revised: 11/27/2023] [Accepted: 12/02/2023] [Indexed: 01/09/2024]
Abstract
Discrete alcohol cues and contexts are relapse triggers for people with alcohol use disorder exerting particularly powerful control over behaviour when they co-occur. Here, we investigated the neural substrates subserving the capacity for alcohol-associated contexts to elevate responding to an alcohol-predictive conditioned stimulus (CS). Specifically, rats were trained in a distinct 'alcohol context' to respond by entering a fluid port during a discrete auditory CS that predicted the delivery of alcohol and were familiarized with a 'neutral context' wherein alcohol was never available. When conditioned CS responding was tested by presenting the CS without alcohol, we found that augmenting glutamatergic activity in the nucleus accumbens (NAc) shell by microinfusing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) reduced responding to an alcohol CS in an alcohol, but not neutral, context. Further, AMPA microinfusion robustly affected behaviour, attenuating the number, duration and latency of CS responses selectively in the alcohol context. Although dopaminergic inputs to the NAc shell were previously shown to be necessary for CS responding in an alcohol context, here, chemogenetic excitation of ventral tegmental area (VTA) dopamine neurons and their inputs to the NAc shell did not affect CS responding. Critically, chemogenetic excitation of VTA dopamine neurons affected feeding behaviour and elevated c-fos immunoreactivity in the VTA and NAc shell, validating the chemogenetic approach. These findings enrich our understanding of the substrates underlying Pavlovian responding for alcohol and reveal that the capacity for contexts to modulate responding to discrete alcohol cues is delicately underpinned by the NAc shell.
Collapse
Affiliation(s)
- Milan D Valyear
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
- Department of Psychology, McGill University, Montreal, Quebec, Canada
| | - Alexa Brown
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Ghislaine Deyab
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Franz R Villaruel
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Soraya Lahlou
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Nina Caporicci-Dinucci
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Nadia Chaudhri
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| |
Collapse
|
6
|
Renu K, Myakala H, Chakraborty R, Bhattacharya S, Abuwani A, Lokhandwala M, Vellingiri B, Gopalakrishnan AV. Molecular mechanisms of alcohol's effects on the human body: A review and update. J Biochem Mol Toxicol 2023; 37:e23502. [PMID: 37578200 DOI: 10.1002/jbt.23502] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 07/18/2023] [Accepted: 07/31/2023] [Indexed: 08/15/2023]
Abstract
Alcohol consumption has been linked to numerous negative health outcomes although it has some beneficial effects on moderate dosages, the most severe of which being alcohol-induced hepatitis. The number of people dying from this liver illness has been shown to climb steadily over time, and its prevalence has been increasing. Researchers have found that alcohol consumption primarily affects the brain, leading to a wide range of neurological and psychological diseases. High-alcohol-consumption addicts not only experienced seizures, but also ataxia, aggression, social anxiety, and variceal hemorrhage that ultimately resulted in death, ascites, and schizophrenia. Drugs treating this liver condition are limited and can cause serious side effects like depression. Serine-threonine kinases, cAMP protein kinases, protein kinase C, ERK, RACK 1, Homer 2, and more have all been observed to have their signaling pathways disrupted by alcohol, and alcohol has also been linked to epigenetic changes. In addition, alcohol consumption induces dysbiosis by changing the composition of the microbiome found in the gastrointestinal tract. Although more studies are needed, those that have been done suggest that probiotics aid in keeping the various microbiota concentrations stable. It has been argued that reducing one's alcohol intake may seem less harmful because excessive drinking is a lifestyle disorder.
Collapse
Affiliation(s)
- Kaviyarasi Renu
- Department of Biochemistry, Centre of Molecular Medicine and Diagnostics (COMManD), Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Haritha Myakala
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Rituraj Chakraborty
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Sharmishtha Bhattacharya
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Asmita Abuwani
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Mariyam Lokhandwala
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Balachandar Vellingiri
- Department of Zoology, Stem Cell and Regenerative Medicine/Translational Research, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda, Punjab, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| |
Collapse
|
7
|
Zhang Y, Wang H, Sun Y, Huang Z, Tao Y, Wang Y, Jiang X, Tao J. Trace amine-associated receptor 1 regulation of Kv1.4 channels in trigeminal ganglion neurons contributes to nociceptive behaviors. J Headache Pain 2023; 24:49. [PMID: 37158881 PMCID: PMC10165857 DOI: 10.1186/s10194-023-01582-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/21/2023] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND Trace amines, such as tyramine, are endogenous amino acid metabolites that have been hypothesized to promote headache. However, the underlying cellular and molecular mechanisms remain unknown. METHODS Using patch-clamp recording, immunostaining, molecular biological approaches and behaviour tests, we elucidated a critically functional role of tyramine in regulating membrane excitability and pain sensitivity by manipulating Kv1.4 channels in trigeminal ganglion (TG) neurons. RESULTS Application of tyramine to TG neurons decreased the A-type K+ current (IA) in a manner dependent on trace amine-associated receptor 1 (TAAR1). Either siRNA knockdown of Gαo or chemical inhibition of βγ subunit (Gβγ) signaling abrogated the response to tyramine. Antagonism of protein kinase C (PKC) prevented the tyramine-induced IA response, while inhibition of conventional PKC isoforms or protein kinase A elicited no such effect. Tyramine increased the membrane abundance of PKCθ in TG neurons, and either pharmacological or genetic inhibition of PKCθ blocked the TAAR1-mediated IA decrease. Furthermore, PKCθ-dependent IA suppression was mediated by Kv1.4 channels. Knockdown of Kv1.4 abrogated the TAAR1-induced IA decrease, neuronal hyperexcitability, and pain hypersensitivity. In a mouse model of migraine induced by electrical stimulation of the dura mater surrounding the superior sagittal sinus, blockade of TAAR1 signaling attenuated mechanical allodynia; this effect was occluded by lentiviral overexpression of Kv1.4 in TG neurons. CONCLUSION These results suggest that tyramine induces Kv1.4-mediated IA suppression through stimulation of TAAR1 coupled to the Gβγ-dependent PKCθ signaling cascade, thereby enhancing TG neuronal excitability and mechanical pain sensitivity. Insight into TAAR1 signaling in sensory neurons provides attractive targets for the treatment of headache disorders such as migraine.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Geriatrics & Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, 1055 San-Xiang Road, Suzhou, 215004, P.R. China.
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, P.R. China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123, P.R. China.
| | - Hua Wang
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P.R. China
| | - Yufang Sun
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, P.R. China
| | - Zitong Huang
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, P.R. China
| | - Yu Tao
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, P.R. China
| | - Yiru Wang
- Department of Geriatrics & Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, 1055 San-Xiang Road, Suzhou, 215004, P.R. China
| | - Xinghong Jiang
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, P.R. China
| | - Jin Tao
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, P.R. China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123, P.R. China.
| |
Collapse
|
8
|
Chapp AD, Collins AR, Driscoll KM, Behnke JE, Shan Z, Zhang L, Chen QH. Ethanol Metabolite, Acetate, Increases Excitability of the Central Nucleus of Amygdala Neurons through Activation of NMDA Receptors. ACS Chem Neurosci 2023; 14:1278-1290. [PMID: 36957993 PMCID: PMC11163875 DOI: 10.1021/acschemneuro.2c00784] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023] Open
Abstract
The central nucleus of the amygdala (CeA) is a key brain region involved in emotional and stressor responses due to its many projections to autonomic regulatory centers. It is also a primary site of action from ethanol consumption. However, the influence of active metabolites of ethanol such as acetate on the CeA neural circuitry has yet to be elucidated. Here, we investigated the effect of acetate on CeA neurons with the axon projecting to the rostral ventrolateral medulla (CeA-RVLM), as well as quantified cytosolic calcium responses in primary neuronal cultures. Whole-cell patch-clamp recordings in brain slices containing autonomic CeA-RVLM neurons revealed a dose-dependent increase in neuronal excitability in response to acetate. N-Methyl-d-aspartate receptor (NMDAR) antagonists suppressed the acetate-induced increase in CeA-RVLM neuronal excitability and memantine suppressed the direct activation of NMDAR-dependent inward currents by acetate in brain slices. We observed that acetate increased cytosolic Ca2+ in a time-dependent manner in primary neuronal cell cultures. The acetate enhancement of calcium signaling was abolished by memantine. Computational modeling of acetic acid at NMDAR/NR1 glutamatergic and glycinergic sites suggests potential active site interactions. These findings suggest that within the CeA, acetate is excitatory at least partially through activation of NMDAR, which may underlie the impact of ethanol consumption on autonomic circuitry.
Collapse
Affiliation(s)
- Andrew D Chapp
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, SDC, 1400 Townsend Drive, Houghton, Michigan 49931, United States
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931, United States
- Department of Neuroscience, University of Minnesota, Twin Cities, 321 Church Street SE, Minneapolis, Minnesota 55455, United States
| | - Andréa R Collins
- Department of Psychiatry, University of California, San Francisco, Fresno, California 93701, United States
| | - Kyle M Driscoll
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Jessica E Behnke
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, SDC, 1400 Townsend Drive, Houghton, Michigan 49931, United States
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Zhiying Shan
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, SDC, 1400 Townsend Drive, Houghton, Michigan 49931, United States
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Li Zhang
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland 20852, United States
| | - Qing-Hui Chen
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, SDC, 1400 Townsend Drive, Houghton, Michigan 49931, United States
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931, United States
| |
Collapse
|
9
|
Anderson EM, Tsvetkov E, Galante A, DeVries D, McCue LM, Wood D, Barry S, Berto S, Lavin A, Taniguchi M, Cowan CW. Epigenetic function during heroin self-administration controls future relapse-associated behavior in a cell type-specific manner. Proc Natl Acad Sci U S A 2023; 120:e2210953120. [PMID: 36745812 PMCID: PMC9963300 DOI: 10.1073/pnas.2210953120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 01/06/2023] [Indexed: 02/08/2023] Open
Abstract
Opioid use produces enduring associations between drug reinforcement/euphoria and discreet or diffuse cues in the drug-taking environment. These powerful associations can trigger relapse in individuals recovering from opioid use disorder (OUD). Here, we sought to determine whether the epigenetic enzyme, histone deacetylase 5 (HDAC5), regulates relapse-associated behavior in an animal model of OUD. We examined the effects of nucleus accumbens (NAc) HDAC5 on both heroin- and sucrose-seeking behaviors using operant self-administration paradigms. We utilized cre-dependent viral-mediated approaches to investigate the cell-type-specific effects of HDAC5 on heroin-seeking behavior, gene expression, and medium spiny neuron (MSN) cell and synaptic physiology. We found that NAc HDAC5 functions during the acquisition phase of heroin self-administration to limit future relapse-associated behavior. Moreover, overexpressing HDAC5 in the NAc suppressed context-associated and reinstated heroin-seeking behaviors, but it did not alter sucrose seeking. We also found that HDAC5 functions within dopamine D1 receptor-expressing MSNs to suppress cue-induced heroin seeking, and within dopamine D2 receptor-expressing MSNs to suppress drug-primed heroin seeking. Assessing cell-type-specific transcriptomics, we found that HDAC5 reduced expression of multiple ion transport genes in both D1- and D2-MSNs. Consistent with this observation, HDAC5 also produced firing rate depression in both MSN classes. These findings revealed roles for HDAC5 during active heroin use in both D1- and D2-MSNs to limit distinct triggers of drug-seeking behavior. Together, our results suggest that HDAC5 might limit relapse vulnerability through regulation of ion channel gene expression and suppression of MSN firing rates during active heroin use.
Collapse
Affiliation(s)
- Ethan M. Anderson
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Evgeny Tsvetkov
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Allison Galante
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Derek DeVries
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Lauren M. McCue
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Daniel Wood
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Sarah Barry
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Stefano Berto
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Antonieta Lavin
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Makoto Taniguchi
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Christopher W. Cowan
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| |
Collapse
|
10
|
Li Y, Wang JZ, Deng YM, Wang K, Yang L, Long C. Amyloid-β Protein Precursor Regulates Electrophysiological Properties in the Hippocampus via Altered Kv1.4 Expression and Function in Mice. J Alzheimers Dis 2023; 92:1241-1256. [PMID: 36872774 DOI: 10.3233/jad-220606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
BACKGROUND Amyloid-β protein precursor (AβPP) is enriched in neurons. However, the mechanism underlying AβPP regulation of neuronal activity is poorly understood. Potassium channels are critically involved in neuronal excitability. In hippocampus, A-type potassium channels are highly expressed and involved in determining neuronal spiking. OBJECTIVE We explored hippocampal local field potential (LFP) and spiking in the presence and absence of AβPP, and the potential involvement of an A-type potassium channel. METHODS We used in vivo extracellular recording and whole-cell patch-clamp recording to determine neuronal activity, current density of A-type potassium currents, and western blot to detect changes in related protein levels. RESULTS Abnormal LFP was observed in AβPP-/- mice, including reduced beta and gamma power, and increased epsilon and ripple power. The firing rate of glutamatergic neurons reduced significantly, in line with an increased action potential rheobase. Given that A-type potassium channels regulate neuronal firing, we measured the protein levels and function of two major A-type potassium channels and found that the post-transcriptional level of Kv1.4, but not Kv4.2, was significantly increased in the AβPP-/- mice. This resulted in a marked increase in the peak time of A-type transient outward potassium currents in both glutamatergic and gamma-aminobutyric acid-ergic (GABAergic) neurons. Furthermore, a mechanistic experiment using human embryonic kidney 293 (HEK293) cells revealed that the AβPP deficiency-induced increase in Kv1.4 may not involve protein-protein interaction between AβPP and Kv1.4. CONCLUSION This study suggests that AβPP modulates neuronal firing and oscillatory activity in the hippocampus, and Kv1.4 may be involved in mediating the modulation.
Collapse
Affiliation(s)
- Yi Li
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, China
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Jin-Zhao Wang
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Yue-Ming Deng
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Kun Wang
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Li Yang
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Cheng Long
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, China
- School of Life Sciences, South China Normal University, Guangzhou, China
| |
Collapse
|
11
|
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
|
12
|
Sanchez EO, Bangasser DA. The effects of early life stress on impulsivity. Neurosci Biobehav Rev 2022; 137:104638. [PMID: 35341796 DOI: 10.1016/j.neubiorev.2022.104638] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 01/19/2022] [Accepted: 03/21/2022] [Indexed: 01/08/2023]
Abstract
Elevated impulsivity is a symptom shared by various psychiatric disorders such as substance use disorder, bipolar disorder, and attention-deficit/hyperactivity disorder. However, impulsivity is not a unitary construct and impulsive behaviors fall into two subcategories: impulsive action and impulsive choice. Impulsive choice refers to the tendency to prefer immediate, small rewards over delayed, large rewards, whereas impulsive action involves difficulty inhibiting rash, premature, or mistimed behaviors. These behaviors are mediated by the mesocorticolimbic dopamine (DA) system, which consists of projections from the ventral tegmental area to the nucleus accumbens and prefrontal cortex. Early life stress (ELS) alters both impulsive choice and impulsive action in rodents. ELS also changes DA receptor expression, transmission, and activity within the mesocorticolimbic system. This review integrates the dopamine, impulsivity, and ELS literature to provide evidence that ELS alters impulsivity via inducing changes in the mesocorticolimbic DA system. Understanding how ELS affects brain circuits associated with impulsivity can help advance treatments aimed towards reducing impulsivity symptoms in a variety of psychiatric disorders.
Collapse
Affiliation(s)
- Evelyn Ordoñes Sanchez
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA.
| | - Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA.
| |
Collapse
|
13
|
Trigo S, Silva PA, Cardoso GC, Soares MC. A test of context and sex-dependent dopaminergic effects on the behavior of a gregarious bird, the common waxbill Estrilda astrild. J Exp Biol 2022; 225:274524. [PMID: 35202471 DOI: 10.1242/jeb.243861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/14/2022] [Indexed: 11/20/2022]
Abstract
The Dopaminergic (DAergic) system has well known influences on behavioral and cognitive functions. Previous work with common waxbills (Estrilda astrild) reported context-specific DAergic effects that could have been due to social environment. Manipulating the dopamine D2-like receptor family (D2R) pathways had opposed effects on behavior depending on whether waxbills were tested alone or in a small cage with a mirror as social stimulus. Since waxbills are highly gregarious, it was hypothesized that being alone or perceiving to have a companion might explain this context-dependence. To test context-dependent DAergic effects, we compared behavioral effects of D2R manipulation in waxbills in the same familiar environment, but either alone or with a familiar, same-sex companion. We found that D2R agonism decreased movement and feeding, similarly to previous results when testing waxbills alone. However, contrary to the hypothesis of dependence on social context, we found that the behavioral effects of the D2R agonist were unchanged when waxbills were tested with a companion. The context-dependence reported earlier might thus be due to other factors, such as the stress of being in a novel environment (small cage) or with an unfamiliar social stimulus (mirror image). In tests with a companion, we also found a sex-specific social effect of D2R manipulation: D2R blocking tended to decrease aggression in males but to increase in females. Together with past work, our results suggest that DAergic effects on behavior involve different types of context- or sex-dependence.
Collapse
Affiliation(s)
- Sandra Trigo
- CIBIO/InBIO-Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, Campus Agrário de Vairão, 4485-661 Vairão, Portugal
| | - Paulo A Silva
- CIBIO/InBIO-Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, Campus Agrário de Vairão, 4485-661 Vairão, Portugal
| | - Gonçalo C Cardoso
- CIBIO/InBIO-Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, Campus Agrário de Vairão, 4485-661 Vairão, Portugal
| | - Marta C Soares
- CIBIO/InBIO-Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, Campus Agrário de Vairão, 4485-661 Vairão, Portugal
| |
Collapse
|
14
|
Allam SL, Rumbell TH, Hoang-Trong T, Parikh J, Kozloski JR. Neuronal population models reveal specific linear conductance controllers sufficient to rescue preclinical disease phenotypes. iScience 2021; 24:103279. [PMID: 34778727 PMCID: PMC8577087 DOI: 10.1016/j.isci.2021.103279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/05/2021] [Accepted: 10/13/2021] [Indexed: 12/23/2022] Open
Abstract
Preclinical drug candidates are screened for their ability to ameliorate in vitro neuronal electrophysiology, and go/no-go decisions progress drugs to clinical trials based on population means across cells and animals. However, these measures do not mitigate clinical endpoint risk. Population-based modeling captures variability across multiple electrophysiological measures from healthy, disease, and drug phenotypes. We pursued optimizing therapeutic targets by identifying coherent sets of ion channel target modulations for recovering heterogeneous wild-type (WT) population excitability profiles from a heterogeneous Huntington's disease (HD) population. Our approach combines mechanistic simulations with population modeling of striatal neurons using evolutionary optimization algorithms to design 'virtual drugs'. We introduce efficacy metrics to score populations and rank virtual drug candidates. We found virtual drugs using heuristic approaches that performed better than single target modulators and standard classification methods. We compare a real drug to virtual candidates and demonstrate a novel in silico triaging method.
Collapse
Affiliation(s)
- Sushmita L. Allam
- IBM T.J. Watson Research Center, 13-158B, P.O. Box 218, 1101 Kitchawan Road, Yorktown Heights, NY 10598, USA
| | - Timothy H. Rumbell
- IBM T.J. Watson Research Center, 13-158B, P.O. Box 218, 1101 Kitchawan Road, Yorktown Heights, NY 10598, USA
| | - Tuan Hoang-Trong
- IBM T.J. Watson Research Center, 13-158B, P.O. Box 218, 1101 Kitchawan Road, Yorktown Heights, NY 10598, USA
| | - Jaimit Parikh
- IBM T.J. Watson Research Center, 13-158B, P.O. Box 218, 1101 Kitchawan Road, Yorktown Heights, NY 10598, USA
| | - James R. Kozloski
- IBM T.J. Watson Research Center, 13-158B, P.O. Box 218, 1101 Kitchawan Road, Yorktown Heights, NY 10598, USA
| |
Collapse
|
15
|
Yang Y. Functional Selectivity of Dopamine D 1 Receptor Signaling: Retrospect and Prospect. Int J Mol Sci 2021; 22:ijms222111914. [PMID: 34769344 PMCID: PMC8584964 DOI: 10.3390/ijms222111914] [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: 08/17/2021] [Revised: 10/18/2021] [Accepted: 11/01/2021] [Indexed: 11/16/2022] Open
Abstract
Research progress on dopamine D1 receptors indicates that signaling no longer is limited to G protein-dependent cyclic adenosine monophosphate phosphorylation but also includes G protein-independent β-arrestin-related mitogen-activated protein kinase activation, regulation of ion channels, phospholipase C activation, and possibly more. This review summarizes recent studies revealing the complexity of D1 signaling and its clinical implications, and suggests functional selectivity as a promising strategy for drug discovery to magnify the merit of D1 signaling. Functional selectivity/biased receptor signaling has become a major research front because of its potential to improve therapeutics through precise targeting. Retrospective pharmacological review indicated that many D1 ligands have some degree of mild functional selectivity, and novel compounds with extreme bias at D1 signaling were reported recently. Behavioral and neurophysiological studies inspired new methods to investigate functional selectivity and gave insight into the biased signaling of several drugs. Results from recent clinical trials also supported D1 functional selectivity signaling as a promising strategy for discovery and development of better therapeutics.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pharmacology, Penn State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
16
|
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
|
17
|
Ferré S, Guitart X, Quiroz C, Rea W, García-Malo C, Garcia-Borreguero D, Allen RP, Earley CJ. Akathisia and Restless Legs Syndrome: Solving the Dopaminergic Paradox. Sleep Med Clin 2021; 16:249-267. [PMID: 33985651 DOI: 10.1016/j.jsmc.2021.02.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Akathisia is an urgent need to move that is associated with treatment with dopamine receptor blocking agents (DRBAs) and with restless legs syndrome (RLS). The pathogenetic mechanism of akathisia has not been resolved. This article proposes that it involves an increased presynaptic dopaminergic transmission in the ventral striatum and concomitant strong activation of postsynaptic dopamine D1 receptors, which form complexes (heteromers) with dopamine D3 and adenosine A1 receptors. It also proposes that in DRBA-induced akathisia, increased dopamine release depends on inactivation of autoreceptors, whereas in RLS it depends on a brain iron deficiency-induced down-regulation of striatal presynaptic A1 receptors.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Building, 333 Cassell Drive, Baltimore, MD 21224, USA.
| | - Xavier Guitart
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Building, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - César Quiroz
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Building, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - William Rea
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Building, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Celia García-Malo
- Sleep Research Institute, Paseo de la Habana 151, Madrid 28036, Spain
| | | | - Richard P Allen
- Department of Neurology, Johns Hopkins University, Johns Hopkins Bayview Medical Center, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA
| | - Christopher J Earley
- Department of Neurology, Johns Hopkins University, Johns Hopkins Bayview Medical Center, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA
| |
Collapse
|
18
|
Eck SR, Bangasser DA. The effects of early life stress on motivated behaviors: A role for gonadal hormones. Neurosci Biobehav Rev 2020; 119:86-100. [PMID: 33022296 PMCID: PMC7744121 DOI: 10.1016/j.neubiorev.2020.09.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/22/2020] [Accepted: 09/09/2020] [Indexed: 12/14/2022]
Abstract
Motivated behaviors are controlled by the mesocorticolimbic dopamine (DA) system, consisting of projections from the ventral tegmental area (VTA) to the nucleus accumbens (NAc) and prefrontal cortex (PFC), with input from structures including the medial preoptic area (mPOA). Sex differences are present in this circuit, and gonadal hormones (e.g., estradiol and testosterone) are important for regulating DA transmission. Early life stress (ELS) also regulates the mesocorticolimbic DA system. ELS modifies motivated behaviors and the underlying DA circuitry, increasing risk for disorders such as substance use disorder, major depression, and schizophrenia. ELS has been shown to change gonadal hormone signaling in both sexes. Thus, one way that ELS could impact mesocorticolimbic DA is by altering the efficacy of gonadal hormones. This review provides evidence for this idea by integrating the gonadal hormone, motivation, and ELS literature to argue that ELS alters gonadal hormone signaling to impact motivated behavior. We also discuss the importance of these effects in the context of understanding risk and treatments for psychiatric disorders in men and women.
Collapse
Affiliation(s)
- Samantha R Eck
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA.
| | - Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| |
Collapse
|
19
|
Strauss M, O'Donovan B, Ma Y, Xiao Z, Lin S, Bardo MT, Ortinski PI, McLaughlin JP, Zhu J. [ 3H]Dopamine Uptake through the Dopamine and Norepinephrine Transporters is Decreased in the Prefrontal Cortex of Transgenic Mice Expressing HIV-1 Transactivator of Transcription Protein. J Pharmacol Exp Ther 2020; 374:241-251. [PMID: 32461322 PMCID: PMC7366287 DOI: 10.1124/jpet.120.266023] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/21/2020] [Indexed: 01/16/2023] Open
Abstract
Dysregulation of dopamine neurotransmission has been linked to the development of human immunodeficiency virus (HIV)-associated neurocognitive disorder (HAND). To investigate the mechanisms underlying this phenomenon, this study used an inducible HIV-1 transactivator of transcription (Tat) transgenic (iTat-tg) mouse model, which demonstrates brain-specific Tat expression induced by administration of doxycycline. We found that induction of Tat expression in the iTat-tg mice for either 7 or 14 days resulted in a decrease (∼30%) in the V max of [3H]dopamine uptake via both the dopamine transporter (DAT) and norepinephrine transporter (NET) in the prefrontal cortex (PFC), which was comparable to the magnitude (∼35%) of the decrease in B max for [3H]WIN 35,428 and [3H]nisoxetine binding to DAT and NET, respectively. The decreased V max was not accompanied by a reduction of total or plasma membrane expression of DAT and NET. Consistent with the decreased V max for DAT and NET in the PFC, the current study also found an increase in the tissue content of DA and dihydroxyphenylacetic acid in the PFC of iTat-tg mice after 7 days' administration of doxycycline. Electrophysiological recordings in layer V pyramidal neurons of the prelimbic cortex from iTat-tg mice found a significant reduction in action potential firing, which was not sensitive to selective inhibitors for DAT and NET, respectively. These findings provide a molecular basis for using the iTat-tg mouse model in the studies of NeuroHIV. Determining the mechanistic basis underlying the interaction between Tat and DAT/NET may reveal novel therapeutic possibilities for preventing the increase in comorbid conditions as well as HAND. SIGNIFICANCE STATEMENT: Human immunodeficiency virus (HIV)-1 infection disrupts dopaminergic neurotransmission, leading to HIV-associated neurocognitive disorders (HANDs). Based on our in vitro and in vivo studies, dopamine uptake via both dopamine and norepinephrine transporters is decreased in the prefrontal cortex of HIV-1 Tat transgenic mice, which is consistent with the increased dopamine and dihydroxyphenylacetic acid contents in this brain region. Thus, these plasma membrane transporters are an important potential target for therapeutic intervention for patients with HAND.
Collapse
Affiliation(s)
- Matthew Strauss
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| | - Bernadette O'Donovan
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| | - Yizhi Ma
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| | - Ziyu Xiao
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| | - Steven Lin
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| | - Michael T Bardo
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| | - Pavel I Ortinski
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| | - Jay P McLaughlin
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| |
Collapse
|
20
|
Valyear MD, Glovaci I, Zaari A, Lahlou S, Trujillo-Pisanty I, Andrew Chapman C, Chaudhri N. Dissociable mesolimbic dopamine circuits control responding triggered by alcohol-predictive discrete cues and contexts. Nat Commun 2020; 11:3764. [PMID: 32724058 PMCID: PMC7534644 DOI: 10.1038/s41467-020-17543-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/06/2020] [Indexed: 01/01/2023] Open
Abstract
Context can influence reactions to environmental cues and this elemental process has implications for substance use disorder. Using an animal model, we show that an alcohol-associated context elevates entry into a fluid port triggered by a conditioned stimulus (CS) that predicted alcohol (CS-triggered alcohol-seeking). This effect persists across multiple sessions and, after it diminishes in extinction, the alcohol context retains the capacity to augment reinstatement. Systemically administered eticlopride and chemogenetic inhibition of ventral tegmental area (VTA) dopamine neurons reduce CS-triggered alcohol-seeking. Chemogenetically silencing VTA dopamine terminals in the nucleus accumbens (NAc) core reduces CS-triggered alcohol-seeking, irrespective of context, whereas silencing VTA dopamine terminals in the NAc shell selectively reduces the elevation of CS-triggered alcohol-seeking in an alcohol context. This dissociation reveals new roles for divergent mesolimbic dopamine circuits in the control of responding to a discrete cue for alcohol and in the amplification of this behaviour in an alcohol context.
Collapse
Affiliation(s)
- Milan D Valyear
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Iulia Glovaci
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Audrey Zaari
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Soraya Lahlou
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Ivan Trujillo-Pisanty
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - C Andrew Chapman
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Nadia Chaudhri
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada.
| |
Collapse
|
21
|
Asfestani MA, Brechtmann V, Santiago J, Peter A, Born J, Feld GB. Consolidation of Reward Memory during Sleep Does Not Require Dopaminergic Activation. J Cogn Neurosci 2020; 32:1688-1703. [PMID: 32459129 DOI: 10.1162/jocn_a_01585] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Sleep enhances memories, especially if they are related to future rewards. Although dopamine has been shown to be a key determinant during reward learning, the role of dopaminergic neurotransmission for amplifying reward-related memories during sleep remains unclear. In this study, we scrutinize the idea that dopamine is needed for the preferential consolidation of rewarded information. We impaired dopaminergic neurotransmission, thereby aiming to wipe out preferential sleep-dependent consolidation of high- over low-rewarded memories during sleep. Following a double-blind, balanced, crossover design, 17 young healthy men received the dopamine d2-like receptor blocker sulpiride (800 mg) or placebo, after learning a motivated learning task. The task required participants to memorize 80 highly and 80 lowly rewarded pictures. Half of them were presented for a short (750 msec) and a long (1500 msec) duration, respectively, which permitted dissociation of the effects of reward on sleep-associated consolidation from those of mere encoding depth. Retrieval was tested after a retention interval of approximately 22 hr that included 8 hr of nocturnal sleep. As expected, at retrieval, highly rewarded memories were remembered better than lowly rewarded memories, under placebo. However, there was no evidence for an effect of reducing dopaminergic neurotransmission with sulpiride during sleep on this differential retention of rewarded information. This result indicates that dopaminergic activation likely is not required for the preferential consolidation of reward-associated memory. Rather, it appears that dopaminergic activation only tags such memories at encoding for intensified reprocessing during sleep.
Collapse
Affiliation(s)
| | | | - João Santiago
- University of Tübingen.,German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Andreas Peter
- German Center for Diabetes Research (DZD), Tübingen, Germany.,University Hospital of Tübingen
| | - Jan Born
- University of Tübingen.,German Center for Diabetes Research (DZD), Tübingen, Germany
| | | |
Collapse
|
22
|
Yano H, Adhikari P, Naing S, Hoffman AF, Baumann MH, Lupica CR, Shi L. Positive Allosteric Modulation of the 5-HT 1A Receptor by Indole-Based Synthetic Cannabinoids Abused by Humans. ACS Chem Neurosci 2020; 11:1400-1405. [PMID: 32324370 DOI: 10.1021/acschemneuro.0c00034] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The nonmedical (i.e., recreational) misuse of synthetic cannabinoids (SCs) is a worldwide public health problem. When compared to cannabis, the misuse of SCs is associated with a higher incidence of serious adverse effects, suggesting the possible involvement of noncannabinoid sites of action. Here, we find that, unlike the phytocannabinoid Δ9-tetrahydrocannabinol, the indole-moiety containing SCs, AM2201 and JWH-018, act as positive allosteric modulators (PAMs) at the 5-HT1A receptor (5-HT1AR). This suggests that some biological effects of SCs might involve allosteric interactions with 5-HT1ARs. To test this hypothesis, we examined effects of AM2201 on 5-HT1AR agonist-activated G protein-coupled inwardly rectifying potassium channel currents in neurons in vitro and on the hypothermic response to 5-HT1AR stimulation in mice lacking the cannabinoid receptor 1. We found that both 5-HT1AR effects were potentiated by AM2201, suggesting that PAM activity at 5-HT1AR may represent a novel noncannabinoid receptor mechanism underlying the complex profile of effects for certain SCs.
Collapse
|
23
|
Chronic mild stress alters synaptic plasticity in the nucleus accumbens through GSK3β-dependent modulation of Kv4.2 channels. Proc Natl Acad Sci U S A 2020; 117:8143-8153. [PMID: 32209671 DOI: 10.1073/pnas.1917423117] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Although major depressive disorder (MDD) is highly prevalent, its pathophysiology is poorly understood. Recent evidence suggests that glycogen-synthase kinase 3β (GSK3β) plays a key role in memory formation, yet its role in mood regulation remains controversial. Here, we investigated whether GSK3β activity in the nucleus accumbens (NAc) is associated with depression-like behaviors and synaptic plasticity. We performed whole-cell patch-clamp recordings of medium spiny neurons (MSNs) in the NAc and determined the role of GSK3β in spike timing-dependent long-term potentiation (tLTP) in the chronic unpredictable mild stress (CUMS) mouse model of depression. To assess the specific role of GSK3β in tLTP, we used in vivo genetic silencing by an adeno-associated viral vector (AAV2) short hairpin RNA against GSK3β. In addition, we examined the role of the voltage-gated potassium Kv4.2 subunit, a molecular determinant of A-type K+ currents, as a potential downstream target of GSK3β. We found increased levels of active GSK3β and augmented tLTP in CUMS mice, a phenotype that was prevented by selective GSK3β knockdown. Furthermore, knockdown of GSK3β in the NAc ameliorated depressive-like behavior in CUMS mice. Electrophysiological, immunohistochemical, biochemical, and pharmacological experiments revealed that inhibition of the Kv4.2 channel through direct phosphorylation at Ser-616 mediated the GSK3β-dependent tLTP changes in CUMS mice. Our results identify GSK3β regulation of Kv4.2 channels as a molecular mechanism of MSN maladaptive plasticity underlying depression-like behaviors and suggest that the GSK3β-Kv4.2 axis may be an attractive therapeutic target for MDD.
Collapse
|
24
|
Delint-Ramirez I, Garcia-Oscos F, Segev A, Kourrich S. Cocaine engages a non-canonical, dopamine-independent, mechanism that controls neuronal excitability in the nucleus accumbens. Mol Psychiatry 2020; 25:680-691. [PMID: 29880884 PMCID: PMC7042730 DOI: 10.1038/s41380-018-0092-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 04/03/2018] [Accepted: 04/13/2018] [Indexed: 11/25/2022]
Abstract
Drug-induced enhanced dopamine (DA) signaling in the brain is a canonical mechanism that initiates addiction processes. However, indirect evidence suggests that cocaine also triggers non-canonical, DA-independent, mechanisms that contribute to behavioral responses to cocaine, including psychomotor sensitization and cocaine self-administration. Identifying these mechanisms and determining how they are initiated is fundamental to further our understanding of addiction processes. Using physiologically relevant in vitro tractable models, we found that cocaine-induced hypoactivity of nucleus accumbens shell (NAcSh) medium spiny neurons (MSNs), one hallmark of cocaine addiction, is independent of DA signaling. Combining brain slice studies and site-directed mutagenesis in HEK293T cells, we found that cocaine binding to intracellular sigma-1 receptor (σ1) initiates this mechanism. Subsequently, σ1 binds to Kv1.2 potassium channels, followed by accumulation of Kv1.2 in the plasma membrane, thereby depressing NAcSh MSNs firing. This mechanism is specific to D1 receptor-expressing MSNs. Our study uncovers a mechanism for cocaine that bypasses DA signaling and leads to addiction-relevant neuroadaptations, thereby providing combinatorial strategies for treating stimulant abuse.
Collapse
Affiliation(s)
- Ilse Delint-Ramirez
- 0000 0000 9482 7121grid.267313.2Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Francisco Garcia-Oscos
- 0000 0000 9482 7121grid.267313.2Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Amir Segev
- 0000 0000 9482 7121grid.267313.2Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Saïd Kourrich
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
25
|
McDevitt DS, Jonik B, Graziane NM. Morphine Differentially Alters the Synaptic and Intrinsic Properties of D1R- and D2R-Expressing Medium Spiny Neurons in the Nucleus Accumbens. Front Synaptic Neurosci 2019; 11:35. [PMID: 31920618 PMCID: PMC6932971 DOI: 10.3389/fnsyn.2019.00035] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 12/06/2019] [Indexed: 12/31/2022] Open
Abstract
Exposure to opioids reshapes future reward and motivated behaviors partially by altering the functional output of medium spiny neurons (MSNs) in the nucleus accumbens shell. Here, we investigated how morphine, a highly addictive opioid, alters synaptic transmission and intrinsic excitability on dopamine D1-receptor (D1R) expressing and dopamine D2-receptor (D2R) expressing MSNs, the two main output neurons in the nucleus accumbens shell. Using whole-cell electrophysiology recordings, we show, that 24 h abstinence following repeated non-contingent administration of morphine (10 mg/kg, i.p.) in mice reduces the miniature excitatory postsynaptic current (mEPSC) frequency and miniature inhibitory postsynaptic current (mIPSC) frequency on D2R-MSNs, with concomitant increases in D2R-MSN intrinsic membrane excitability. We did not observe any changes in synaptic or intrinsic changes on D1R-MSNs. Last, in an attempt to determine the integrated effect of the synaptic and intrinsic alterations on the overall functional output of D2R-MSNs, we measured the input-output efficacy by measuring synaptically-driven action potential firing. We found that both D1R-MSN and D2R-MSN output was unchanged following morphine treatment.
Collapse
Affiliation(s)
- Dillon S McDevitt
- Departments of Anesthesiology and Perioperative Medicine, and Pharmacology, Penn State College of Medicine, Hershey, PA, United States.,Neuroscience Graduate Program, Penn State College of Medicine, Hershey, PA, United States
| | - Benjamin Jonik
- Medical Student Research Program, Penn State College of Medicine, Hershey, PA, United States
| | - Nicholas M Graziane
- Departments of Anesthesiology and Perioperative Medicine, and Pharmacology, Penn State College of Medicine, Hershey, PA, United States
| |
Collapse
|
26
|
Soriani O, Kourrich S. The Sigma-1 Receptor: When Adaptive Regulation of Cell Electrical Activity Contributes to Stimulant Addiction and Cancer. Front Neurosci 2019; 13:1186. [PMID: 31780884 PMCID: PMC6861184 DOI: 10.3389/fnins.2019.01186] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/21/2019] [Indexed: 12/17/2022] Open
Abstract
The sigma-1 receptor (σ1R) is an endoplasmic reticulum (ER)-resident chaperone protein that acts like an inter-organelle signaling modulator. Among its several functions such as ER lipid metabolisms/transports and indirect regulation of genes transcription, one of its most intriguing feature is the ability to regulate the function and trafficking of a variety of functional proteins. To date, and directly relevant to the present review, σ1R has been found to regulate both voltage-gated ion channels (VGICs) belonging to distinct superfamilies (i.e., sodium, Na+; potassium, K+; and calcium, Ca2+ channels) and non-voltage-gated ion channels. This regulatory function endows σ1R with a powerful capability to fine tune cells’ electrical activity and calcium homeostasis—a regulatory power that appears to favor cell survival in pathological contexts such as stroke or neurodegenerative diseases. In this review, we present the current state of knowledge on σ1R’s role in the regulation of cellular electrical activity, and how this seemingly adaptive function can shift cell homeostasis and contribute to the development of very distinct chronic pathologies such as psychostimulant abuse and tumor cell growth in cancers.
Collapse
Affiliation(s)
| | - Saïd Kourrich
- Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, QC, Canada.,Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois, Université du Québec à Montréal, Montréal, QC, Canada.,Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
27
|
O'Donovan B, Adeluyi A, Anderson EL, Cole RD, Turner JR, Ortinski PI. Altered gating of K v1.4 in the nucleus accumbens suppresses motivation for reward. eLife 2019; 8:e47870. [PMID: 31487241 PMCID: PMC6728144 DOI: 10.7554/elife.47870] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 08/22/2019] [Indexed: 12/13/2022] Open
Abstract
Deficient motivation contributes to numerous psychiatric disorders, including withdrawal from drug use, depression, schizophrenia, and others. Nucleus accumbens (NAc) has been implicated in motivated behavior, but it remains unclear whether motivational drive is linked to discrete neurobiological mechanisms within the NAc. To examine this, we profiled cohorts of Sprague-Dawley rats in a test of motivation to consume sucrose. We found that substantial variability in willingness to exert effort for reward was not associated with operant responding under low-effort conditions or stress levels. Instead, effort-based motivation was mirrored by a divergent NAc shell transcriptome with differential regulation at potassium and dopamine signaling genes. Functionally, motivation was inversely related to excitability of NAc principal neurons. Furthermore, neuronal and behavioral outputs associated with low motivation were linked to faster inactivation of a voltage-gated potassium channel, Kv1.4. These results raise the prospect of targeting Kv1.4 gating in psychiatric conditions associated with motivational dysfunction.
Collapse
Affiliation(s)
| | - Adewale Adeluyi
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of PharmacyUniversity of South CarolinaColumbiaUnited States
| | - Erin L Anderson
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of PharmacyUniversity of South CarolinaColumbiaUnited States
| | - Robert D Cole
- Department of NeuroscienceUniversity of KentuckyLexingtonUnited States
| | - Jill R Turner
- College of PharmacyUniversity of KentuckyLexingtonUnited States
| | - Pavel I Ortinski
- Department of NeuroscienceUniversity of KentuckyLexingtonUnited States
| |
Collapse
|
28
|
Porzionato A, Stocco E, Guidolin D, Agnati L, Macchi V, De Caro R. Receptor-Receptor Interactions of G Protein-Coupled Receptors in the Carotid Body: A Working Hypothesis. Front Physiol 2018; 9:697. [PMID: 29930516 PMCID: PMC6000251 DOI: 10.3389/fphys.2018.00697] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 05/18/2018] [Indexed: 12/15/2022] Open
Abstract
In the carotid body (CB), a wide series of neurotransmitters and neuromodulators have been identified. They are mainly produced and released by type I cells and act on many different ionotropic and metabotropic receptors located in afferent nerve fibers, type I and II cells. Most metabotropic receptors are G protein-coupled receptors (GPCRs). In other transfected or native cells, GPCRs have been demonstrated to establish physical receptor–receptor interactions (RRIs) with formation of homo/hetero-complexes (dimers or receptor mosaics) in a dynamic monomer/oligomer equilibrium. RRIs modulate ligand binding, signaling, and internalization of GPCR protomers and they are considered of relevance for physiology, pharmacology, and pathology of the nervous system. We hypothesize that RRI may also occur in the different structural elements of the CB (type I cells, type II cells, and afferent fibers), with potential implications in chemoreception, neuromodulation, and tissue plasticity. This ‘working hypothesis’ is supported by literature data reporting the contemporary expression, in type I cells, type II cells, or afferent terminals, of GPCRs which are able to physically interact with each other to form homo/hetero-complexes. Functional data about cross-talks in the CB between different neurotransmitters/neuromodulators also support the hypothesis. On the basis of the above findings, the most significant homo/hetero-complexes which could be postulated in the CB include receptors for dopamine, adenosine, ATP, opioids, histamine, serotonin, endothelin, galanin, GABA, cannabinoids, angiotensin, neurotensin, and melatonin. From a methodological point of view, future studies should demonstrate the colocalization in close proximity (less than 10 nm) of the above receptors, through biophysical (i.e., bioluminescence/fluorescence resonance energy transfer, protein-fragment complementation assay, total internal reflection fluorescence microscopy, fluorescence correlation spectroscopy and photoactivated localization microscopy, X-ray crystallography) or biochemical (co-immunoprecipitation, in situ proximity ligation assay) methods. Moreover, functional approaches will be able to show if ligand binding to one receptor produces changes in the biochemical characteristics (ligand recognition, decoding, and trafficking processes) of the other(s). Plasticity aspects would be also of interest, as development and environmental stimuli (chronic continuous or intermittent hypoxia) produce changes in the expression of certain receptors which could potentially invest the dynamic monomer/oligomer equilibrium of homo/hetero-complexes and the correlated functional implications.
Collapse
Affiliation(s)
| | - Elena Stocco
- Department of Neuroscience, University of Padua, Padua, Italy
| | - Diego Guidolin
- Department of Neuroscience, University of Padua, Padua, Italy
| | - Luigi Agnati
- Department of Diagnostic, Clinical Medicine and Public Health, University of Modena and Reggio Emilia, Modena, Italy
| | - Veronica Macchi
- Department of Neuroscience, University of Padua, Padua, Italy
| | | |
Collapse
|
29
|
Herrera-López G, Galván EJ. Modulation of hippocampal excitability via the hydroxycarboxylic acid receptor 1. Hippocampus 2018; 28:557-567. [PMID: 29704292 DOI: 10.1002/hipo.22958] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 04/19/2018] [Accepted: 04/21/2018] [Indexed: 01/15/2023]
Abstract
In addition to its prominent role as an energetic substrate in the brain, lactate is emerging as a signaling molecule capable of controlling neuronal excitability. The finding that the lactate-activated receptor (hydroxycarboxylic acid receptor 1; HCA1) is widely expressed in the brain opened up the possibility that lactate exerts modulation of neuronal activity via a transmembranal receptor-linked mechanism. Here, we show that lactate causes biphasic modulation of the intrinsic excitability of CA1 pyramidal cells. In the low millimolar range, lactate or the HCA1 agonist 3,5-DHBA reduced the input resistance and membrane time constant. In addition, activation of HCA1 significantly blocked the fast inactivating sodium current and increased the delay from inactivation to a conducting state of the sodium channel. As the observed actions occurred in the presence of 4-CIN, a blocker of the neuronal monocarboxylate transporter, the possibility that lactate acted via neuronal metabolism is unlikely. Consistently, modulation of the intrinsic excitability was abolished when CA1 pyramidal cells were dialyzed with pertussis toxin, indicating the dependency of a Gαi/o -protein-coupled receptor. The activation of HCA1 appears to serve as a restraining mechanism during enhanced network activity and may function as a negative feedback for the astrocytic production of lactate.
Collapse
Affiliation(s)
- Gabriel Herrera-López
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Calzada de los Tenorios No. 235, México City 14330, México
| | - Emilio J Galván
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Calzada de los Tenorios No. 235, México City 14330, México
| |
Collapse
|
30
|
Yano H, Cai NS, Xu M, Verma RK, Rea W, Hoffman AF, Shi L, Javitch JA, Bonci A, Ferré S. Gs- versus Golf-dependent functional selectivity mediated by the dopamine D 1 receptor. Nat Commun 2018; 9:486. [PMID: 29402888 PMCID: PMC5799184 DOI: 10.1038/s41467-017-02606-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 12/09/2017] [Indexed: 12/13/2022] Open
Abstract
The two highly homologous subtypes of stimulatory G proteins Gαs (Gs) and Gαolf (Golf) display contrasting expression patterns in the brain. Golf is predominant in the striatum, while Gs is predominant in the cortex. Yet, little is known about their functional distinctions. The dopamine D1 receptor (D1R) couples to Gs/olf and is highly expressed in cortical and striatal areas, making it an important therapeutic target for neuropsychiatric disorders. Using novel drug screening methods that allow analysis of specific G-protein subtype coupling, we found that, relative to dopamine, dihydrexidine and N-propyl-apomorphine behave as full D1R agonists when coupled to Gs, but as partial D1R agonists when coupled to Golf. The Gs/Golf-dependent biased agonism by dihydrexidine was consistently observed at the levels of cellular signaling, neuronal function, and behavior. Our findings of Gs/Golf-dependent functional selectivity in D1R ligands open a new avenue for the treatment of cortex-specific or striatum-specific neuropsychiatric dysfunction.
Collapse
Affiliation(s)
- Hideaki Yano
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA.
| | - Ning-Sheng Cai
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Min Xu
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Ravi Kumar Verma
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - William Rea
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Alexander F Hoffman
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Lei Shi
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Jonathan A Javitch
- Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Antonello Bonci
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Sergi Ferré
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| |
Collapse
|
31
|
Wen RT, Liang JH, Zhang HT. Targeting Phosphodiesterases in Pharmacotherapy for Substance Dependence. ADVANCES IN NEUROBIOLOGY 2018; 17:413-444. [PMID: 28956341 DOI: 10.1007/978-3-319-58811-7_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Substance dependence is a chronic relapsing brain disorder associated with adaptational changes in synaptic plasticity and neuronal functions. The high levels of substance consumption and relapse rate suggest more reliable medications are in need to better address the underlying causes of this disease. It has been well established that the intracellular second messengers cyclic AMP (cAMP) and cyclic GMP (cGMP) and their signaling systems play an important role in the molecular mechanisms of substance taking behaviors. On this basis, the phosphodiesterase (PDE) superfamily, which crucially controls cyclic nucleotide levels by catalyzing their hydrolysis, has been proposed as a novel class of therapeutic targets for substance use disorders. This chapter reviews the expression patterns of PDEs in the brain with regard to neural structures underlying the dependent process and highlights available evidence for a modulatory role of PDEs in substance dependence.
Collapse
Affiliation(s)
- Rui-Ting Wen
- Department of Pharmacy, Peking University People's Hospital, Beijing, 100044, China
| | - Jian-Hui Liang
- Department of Molecular and Cellular Pharmacology, Peking University School of Pharmaceutical Sciences, Beijing, 100191, China.
| | - Han-Ting Zhang
- Department of Behavioral Medicine and Psychiatry, West Virginia University Health Sciences Center, 1 Medical Center Drive, Morgantown, WV, 26506, USA. .,Department of Physiology and Pharmacology, West Virginia University Health Sciences Center, 1 Medical Center Drive, Morgantown, WV, 26506, USA. .,Institute of Pharmacology, Taishan Medical University, Taian, 271016, China.
| |
Collapse
|
32
|
Wang ZY, Lian H, Zhou L, Zhang YM, Cai QQ, Zheng LF, Zhu JX. Altered Expression of D1 and D2 Dopamine Receptors in Vagal Neurons Innervating the Gastric Muscularis Externa in a Parkinson's Disease Rat Model. JOURNAL OF PARKINSONS DISEASE 2017; 6:317-23. [PMID: 27164043 DOI: 10.3233/jpd-160817] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Parkinson's disease (PD) is characterized by dopaminergic neuron degeneration in the substantia nigra (SN) accompanied by pathology of the dorsal motor nucleus of the vagus (DMV). Gastroparesis is a common non-motor system symptom of PD in patients and in animal models. However, the underlying mechanism of this symptom is not clear. We previously reported on the expression of enhanced tyrosine hydroxylase (TH) and decreased choline acetyltransferase (ChAT) in the DMV of a PD animal model and colocalization of TH and ChAT with the dopamine receptors D1 and D2. We hypothesize that these receptors might contribute to the delayed gastric emptying observed in PD. OBJECTIVE To investigate the distribution of D1 and D2 in gastric-projecting DMV neurons and alteration of their distribution in a PD rat model. METHODS Retrograde tracing, double-labeling immunofluorescence techniques and western blotting were used. RESULTS After injection of the retrograde tracer fluoro-gold (FG) into the gastric wall, FG-labeled gastric-projecting motoneurons were observed in the caudal and rostral parts of the DMV, and neurons with D1-, D2- and ChAT- immunoreactivity (IR) were widely colocalized in the DMV. Many TH-IR fibers were observed around the D1- and D2-IR neurons. Moreover, decreased D1 and enhanced D2 expression in the DMV was observed in 6-hydroxydopamine (6-OHDA) rats that were treated with a bilateral microinjection of 6-OHDA in the SN. CONCLUSIONS The results indicate that dopamine receptors might affect the activity of gastric-projecting neurons in the DMV, their altered expression may contribute to the gastroparesis observed in PD.
Collapse
Affiliation(s)
- Zhi-Yong Wang
- Key Lab for Medical Tissue Regeneration of Henan Province, Department of Anatomy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Hui Lian
- Key Lab for Medical Tissue Regeneration of Henan Province, Department of Anatomy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Li Zhou
- Key Lab for Medical Tissue Regeneration of Henan Province, Department of Anatomy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yi-Min Zhang
- Key Lab for Medical Tissue Regeneration of Henan Province, Department of Anatomy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Qing-Qing Cai
- Key Lab for Medical Tissue Regeneration of Henan Province, Department of Anatomy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Li-Fei Zheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jin-Xia Zhu
- Key Lab for Medical Tissue Regeneration of Henan Province, Department of Anatomy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
33
|
Han J, Gao L, Dong J, Wang Y, Zhang M, Zheng J. Dopamine attenuates ethanol-induced neuroapoptosis in the developing rat retina via the cAMP/PKA pathway. Mol Med Rep 2017; 16:1982-1990. [PMID: 28656313 PMCID: PMC5561998 DOI: 10.3892/mmr.2017.6823] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 04/24/2017] [Indexed: 01/11/2023] Open
Abstract
Apoptosis has been identified as the primary cause of fetal alcohol spectrum disorder (FASD), and the development of methods to prevent and treat FASD have been based on the mechanisms of alcohol-induced apoptosis. The present study aimed to explore the effects of dopamine on alcohol-induced neuronal apoptosis using whole-mount cultures of rat retinas (postnatal day 7). Retinas were initially incubated with ethanol (100, 200 or 500 mM), and in subsequent analyses retinas were co-incubated with ethanol (200 mM) and dopamine (10 µM). In addition, several antagonists and inhibitors were used, including a D1 dopamine receptor (D1R) antagonist (SCH23390; 10 µM), a D2R antagonist (raclopride; 40 µM), an adenosine A2A receptor (AA2AR) antagonist (SCH58261; 100 nM), an adenylyl cyclase (AC) inhibitor (SQ22536; 100 µM) and a PKA inhibitor (H-89; 1 µM). The results demonstrated that exposure increased neuroapoptosis in the retinal ganglion cell layer (GCL) in a dose-dependent manner. Dopamine treatment significantly attenuated ethanol-induced neuronal apoptosis. D1R, D2R and AA2AR antagonists partially inhibited the protective effects of dopamine against ethanol-induced apoptosis; similar results were observed with AC and PKA inhibitor treatments. In summary, the present study demonstrated that dopamine treatment may be able to attenuate alcohol-induced neuroapoptosis in the developing rat retina by activating D1R, D2R and AA2AR, and by upregulating cyclic AMP/protein kinase A signaling.
Collapse
Affiliation(s)
- Junde Han
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, P.R. China
| | - Lingqi Gao
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, P.R. China
| | - Jing Dong
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, P.R. China
| | - Yingtian Wang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, P.R. China
| | - Mazhong Zhang
- Department of Anesthesiology and Pediatric Clinical Pharmacology Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Jijian Zheng
- Department of Anesthesiology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| |
Collapse
|
34
|
Sebastianutto I, Cenci MA, Fieblinger T. Alterations of striatal indirect pathway neurons precede motor deficits in two mouse models of Huntington's disease. Neurobiol Dis 2017; 105:117-131. [PMID: 28578004 DOI: 10.1016/j.nbd.2017.05.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 04/26/2017] [Accepted: 05/29/2017] [Indexed: 11/15/2022] Open
Abstract
Striatal neurons forming the indirect pathway (iSPNs) are particularly vulnerable in Huntington's disease (HD). In this study we set out to investigate morphological and physiological alterations of iSPNs in two mouse models of HD with relatively slow disease progression (long CAG repeat R6/2 and zQ175-KI). Both were crossed with a transgenic mouse line expressing eGFP in iSPNs. Using the open-field and rotarod tests, we first defined two time points in relation to the occurrence of motor deficits in each model. Then, we investigated electrophysiological and morphological properties of iSPNs at both ages. Both HD models exhibited increased iSPN excitability already before the onset of motor deficits, associated with a reduced number of primary dendrites and decreased function of Kir- and voltage-gated potassium channels. Alterations that specifically occurred at symptomatic ages included increased calcium release by back-propagating action potentials in proximal dendrites, due to enhanced engagement of intracellular calcium stores. Moreover, motorically impaired mice of both HD models showed a reduction in iSPN spine density and progressive formation of huntingtin (Htt) aggregates in the striatum. Our study therefore reports iSPN-specific alterations relative to the development of a motor phenotype in two different mouse models of HD. While some alterations occur early and are partly non-progressive, others potentially provide a pathophysiological marker of an overt disease state.
Collapse
Affiliation(s)
- Irene Sebastianutto
- Basal Ganglia Pathophysiology Unit, Dept. of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| | - Maria Angela Cenci
- Basal Ganglia Pathophysiology Unit, Dept. of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| | - Tim Fieblinger
- Basal Ganglia Pathophysiology Unit, Dept. of Experimental Medical Science, Lund University, 22184 Lund, Sweden.
| |
Collapse
|
35
|
Nicola SM. Reassessing wanting and liking in the study of mesolimbic influence on food intake. Am J Physiol Regul Integr Comp Physiol 2016; 311:R811-R840. [PMID: 27534877 PMCID: PMC5130579 DOI: 10.1152/ajpregu.00234.2016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/09/2016] [Indexed: 01/12/2023]
Abstract
Humans and animals such as rats and mice tend to overconsume calorie-dense foods, a phenomenon that likely contributes to obesity. One often-advanced explanation for why we preferentially consume sweet and fatty foods is that they are more "rewarding" than low-calorie foods. "Reward" has been subdivided into three interdependent psychological processes: hedonia (liking a food), reinforcement (formation of associations among stimuli, actions, and/or the food), and motivation (wanting the food). Research into these processes has focused on the mesolimbic system, which comprises both dopamine neurons in the ventral tegmental area and neurons in their major projection target, the nucleus accumbens. The mesolimbic system and closely connected structures are commonly referred to as the brain's "reward circuit." Implicit in this title is the assumption that "rewarding" experiences are generally the result of activity in this circuit. In this review, I argue that food intake and the preference for calorie-dense foods can be explained without reference to subjective emotions. Furthermore, the contribution of mesolimbic dopamine to food intake and preference may not be a general one of promoting or coordinating behaviors that result in the most reward or caloric intake but may instead be limited to the facilitation of a specific form of neural computation that results in conditioned approach behavior. Studies on the neural mechanisms of caloric intake regulation must address how sensory information about calorie intake affects not just the mesolimbic system but also many other forms of computation that govern other types of food-seeking and food-oriented behaviors.
Collapse
Affiliation(s)
- Saleem M Nicola
- Departments of Neuroscience and Psychiatry, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
36
|
Nagai T, Yoshimoto J, Kannon T, Kuroda K, Kaibuchi K. Phosphorylation Signals in Striatal Medium Spiny Neurons. Trends Pharmacol Sci 2016; 37:858-871. [DOI: 10.1016/j.tips.2016.07.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/18/2016] [Accepted: 07/21/2016] [Indexed: 12/21/2022]
|
37
|
Lei K, Wegner SA, Yu JH, Mototake A, Hu B, Hopf FW. Nucleus Accumbens Shell and mPFC but Not Insula Orexin-1 Receptors Promote Excessive Alcohol Drinking. Front Neurosci 2016; 10:400. [PMID: 27625592 PMCID: PMC5004043 DOI: 10.3389/fnins.2016.00400] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/15/2016] [Indexed: 12/22/2022] Open
Abstract
Addiction to alcohol remains a major social and economic problem, in part because of the high motivation for alcohol that humans exhibit and the hazardous binge intake this promotes. Orexin-1-type receptors (OX1Rs) promote reward intake under conditions of strong drives for reward, including excessive alcohol intake. While systemic modulation of OX1Rs can alter alcohol drinking, the brain regions that mediate this OX1R enhancement of excessive drinking remain unknown. Given the importance of the nucleus accumbens (NAc) and anterior insular cortex (aINS) in driving many addictive behaviors, including OX1Rs within these regions, we examined the importance of OX1Rs in these regions on excessive alcohol drinking in C57BL/6 mice during limited-access alcohol drinking in the dark cycle. Inhibition of OX1Rs with the widely used SB-334867 within the medial NAc Shell (mNAsh) significantly reduced drinking of alcohol, with no effect on saccharin intake, and no effect on alcohol consumption when infused above the mNAsh. In contrast, intra-mNAsh infusion of the orexin-2 receptor TCS-OX2-29 had no impact on alcohol drinking. In addition, OX1R inhibition within the aINS had no effect on excessive drinking, which was surprising given the importance of aINS-NAc circuits in promoting alcohol consumption and the role for aINS OX1Rs in driving nicotine intake. However, OX1R inhibition within the mPFC did reduce alcohol drinking, indicating cortical OXR involvement in promoting intake. Also, in support of the critical role for mNAsh OX1Rs, SB within the mNAsh also significantly reduced operant alcohol self-administration in rats. Finally, orexin ex vivo enhanced firing in mNAsh neurons from alcohol-drinking mice, with no effect on evoked EPSCs or input resistance; a similar orexin increase in firing without a change in input resistance was observed in alcohol-naïve mice. Taken together, our results suggest that OX1Rs within the mNAsh and mPFC, but not the aINS, play a central role in driving excessive alcohol drinking.
Collapse
Affiliation(s)
- Kelly Lei
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco San Francisco, CA, USA
| | - Scott A Wegner
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco San Francisco, CA, USA
| | - Ji Hwan Yu
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco San Francisco, CA, USA
| | - Arisa Mototake
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco San Francisco, CA, USA
| | - Bing Hu
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco San Francisco, CA, USA
| | - Frederic W Hopf
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco San Francisco, CA, USA
| |
Collapse
|
38
|
du Hoffmann J, Nicola SM. Activation of Dopamine Receptors in the Nucleus Accumbens Promotes Sucrose-Reinforced Cued Approach Behavior. Front Behav Neurosci 2016; 10:144. [PMID: 27471453 PMCID: PMC4943936 DOI: 10.3389/fnbeh.2016.00144] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 06/23/2016] [Indexed: 01/19/2023] Open
Abstract
Dopamine receptor activation in the nucleus accumbens (NAc) promotes vigorous environmentally-cued food-seeking in hungry rats. Rats fed ad libitum, however, respond to fewer food-predictive cues, particularly when the value of food reward is low. Here, we investigated whether this difference could be due to differences in the degree of dopamine receptor activation in the NAc. First, we observed that although rats given ad libitum access to chow in their home cages approached a food receptacle in response to reward-predictive cues, the number of such approaches declined as animals accumulated food rewards. Intriguingly, cued approach to food occurred in clusters, with several cued responses followed by successive non-responses. This pattern suggested that behavior was dictated by transitions between two states, responsive and non-responsive. Injection of D1 or D2 dopamine receptor agonists into the NAc dose-dependently increased cue responding by promoting transitions to the responsive state and by preventing transitions to the non-responsive state. In contrast, antagonists of either D1 or D2 receptors promoted long bouts of non-responding by inducing transitions to the non-responsive state and by preventing transitions to the responsive state. Moreover, locomotor behavior during the inter-trial interval was correlated with the responsive state, and was also increased by dopamine receptor agonists. These results suggest that activation of NAc dopamine receptors plays an important role in regulating the probability of approach to food under conditions of normative satiety.
Collapse
Affiliation(s)
- Johann du Hoffmann
- Department of Neuroscience and Psychiatry, Albert Einstein College of Medicine Bronx, NY, USA
| | - Saleem M Nicola
- Department of Neuroscience and Psychiatry, Albert Einstein College of Medicine Bronx, NY, USA
| |
Collapse
|
39
|
Nagai T, Nakamuta S, Kuroda K, Nakauchi S, Nishioka T, Takano T, Zhang X, Tsuboi D, Funahashi Y, Nakano T, Yoshimoto J, Kobayashi K, Uchigashima M, Watanabe M, Miura M, Nishi A, Kobayashi K, Yamada K, Amano M, Kaibuchi K. Phosphoproteomics of the Dopamine Pathway Enables Discovery of Rap1 Activation as a Reward Signal In Vivo. Neuron 2016; 89:550-65. [DOI: 10.1016/j.neuron.2015.12.019] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 10/17/2015] [Accepted: 12/10/2015] [Indexed: 12/21/2022]
|
40
|
Thompson JL, Yang J, Lau B, Liu S, Baimel C, Kerr LE, Liu F, Borgland SL. Age-Dependent D1-D2 Receptor Coactivation in the Lateral Orbitofrontal Cortex Potentiates NMDA Receptors and Facilitates Cognitive Flexibility. Cereb Cortex 2015; 26:4524-4539. [PMID: 26405054 DOI: 10.1093/cercor/bhv222] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The orbitofrontal cortex (OFC) integrates information about the environment to guide decision-making. Glutamatergic synaptic transmission mediated through N-methyl-d-aspartate receptors is required for optimal functioning of the OFC. Additionally, abnormal dopamine signaling in this region has been implicated in impulsive behavior and poor cognitive flexibility. Yet, despite the high prevalence of psychostimulants prescribed for attention deficit/hyperactivity disorder, there is little information on how dopamine modulates synaptic transmission in the juvenile or the adult OFC. Using whole-cell patch-clamp recordings in OFC pyramidal neurons, we demonstrated that while dopamine or selective D2-like receptor (D2R) agonists suppress excitatory synaptic transmission of juvenile or adult lateral OFC neurons; in juvenile lateral OFC neurons, higher concentrations of dopamine can target dopamine receptors that couple to a phospholipase C (PLC) signaling pathway to enhance excitatory synaptic transmission. Interfering with the formation of a putative D1R-D2R interaction blocked the potentiation of excitatory synaptic transmission. Furthermore, targeting the putative D1R-D2R complex with a biased agonist, SKF83959, not only enhanced excitatory synaptic transmission in a PLC-dependent manner, but also improved the performance of juvenile rats on a reversal-learning task. Our results demonstrate that dopamine signaling in the lateral OFC differs between juveniles and adults, through potential crosstalk between dopamine receptor subtypes.
Collapse
Affiliation(s)
- Jennifer L Thompson
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada V6T 1Z3.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Jinhui Yang
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Benjamin Lau
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Shuai Liu
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Corey Baimel
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada V6T 1Z3.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Lauren E Kerr
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Fang Liu
- Department of Neuroscience, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | | |
Collapse
|
41
|
Dias EV, Sartori CR, Marião PR, Vieira AS, Camargo LC, Athie MCP, Pagliusi MO, Tambeli CH, Parada CA. Nucleus accumbens dopaminergic neurotransmission switches its modulatory action in chronification of inflammatory hyperalgesia. Eur J Neurosci 2015; 42:2380-9. [DOI: 10.1111/ejn.13015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 07/03/2015] [Accepted: 07/06/2015] [Indexed: 12/30/2022]
Affiliation(s)
- Elayne Vieira Dias
- Department of Structural and Functional Biology; State University of Campinas; Rua Monteiro Lobato 255, Cidade Universitaria Zeferino Vaz Box 6109 13083-865 Campinas SP Brazil
| | - César Renato Sartori
- Department of Structural and Functional Biology; State University of Campinas; Rua Monteiro Lobato 255, Cidade Universitaria Zeferino Vaz Box 6109 13083-865 Campinas SP Brazil
| | - Paula Ramos Marião
- Department of Structural and Functional Biology; State University of Campinas; Rua Monteiro Lobato 255, Cidade Universitaria Zeferino Vaz Box 6109 13083-865 Campinas SP Brazil
| | - André Schwambach Vieira
- Department of Structural and Functional Biology; State University of Campinas; Rua Monteiro Lobato 255, Cidade Universitaria Zeferino Vaz Box 6109 13083-865 Campinas SP Brazil
| | - Lilian Calili Camargo
- Department of Structural and Functional Biology; State University of Campinas; Rua Monteiro Lobato 255, Cidade Universitaria Zeferino Vaz Box 6109 13083-865 Campinas SP Brazil
| | - Maria Carolina Pedro Athie
- Department of Structural and Functional Biology; State University of Campinas; Rua Monteiro Lobato 255, Cidade Universitaria Zeferino Vaz Box 6109 13083-865 Campinas SP Brazil
| | - Marco Oreste Pagliusi
- Department of Structural and Functional Biology; State University of Campinas; Rua Monteiro Lobato 255, Cidade Universitaria Zeferino Vaz Box 6109 13083-865 Campinas SP Brazil
| | - Claudia Herrera Tambeli
- Department of Structural and Functional Biology; State University of Campinas; Rua Monteiro Lobato 255, Cidade Universitaria Zeferino Vaz Box 6109 13083-865 Campinas SP Brazil
| | - Carlos Amilcar Parada
- Department of Structural and Functional Biology; State University of Campinas; Rua Monteiro Lobato 255, Cidade Universitaria Zeferino Vaz Box 6109 13083-865 Campinas SP Brazil
| |
Collapse
|
42
|
Ortinski PI, Briand LA, Pierce RC, Schmidt HD. Cocaine-seeking is associated with PKC-dependent reduction of excitatory signaling in accumbens shell D2 dopamine receptor-expressing neurons. Neuropharmacology 2015; 92:80-9. [PMID: 25596492 PMCID: PMC4346508 DOI: 10.1016/j.neuropharm.2015.01.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 11/25/2014] [Accepted: 01/07/2015] [Indexed: 01/11/2023]
Abstract
Stimulation of D1-like dopamine receptors (D1DRs) or D2-like dopamine receptors (D2DRs) in the nucleus accumbens (NAc) shell reinstates cocaine seeking in rats, an animal model of relapse. D2DRs and D1DRs activate protein kinase C (PKC) and recent studies indicate that activation of PKC in the NAc plays an important role in the reinstatement of drug seeking induced by a systemic cocaine priming injection. In the present study, pharmacological inhibition of PKC in the NAc shell attenuated cocaine seeking induced by intra-accumbens shell microinjection of a D2DR agonist, but not a D1DR agonist. D1DRs and D2DRs are primarily expressed on different accumbens medium spiny (MSN) neurons. Neuronal signaling and activity were assessed in these two populations of NAc neurons with transgenic mice expressing fluorescent labels under the control of D1DR and D2DR promoters. Following the extinction of cocaine self-administration, bath application of a PKC inhibitor produced similar effects on single evoked excitatory and inhibitory post-synaptic currents in D1DR- and D2DR-positive MSNs in the NAc shell. However, inhibition of PKC preferentially improved the ability of excitatory, but not inhibitory, synapses to sustain responding to brief train of stimuli specifically in D2DR-positive MSNs. This effect did not appear to involve modulation of presynaptic release mechanisms. Taken together, these findings indicate that the reinstatement of cocaine seeking is at least partially due to D2DR-dependent increases in PKC signaling in the NAc shell, which reduce excitatory synaptic efficacy in D2DR-expressing MSNs.
Collapse
Affiliation(s)
- Pavel I Ortinski
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Lisa A Briand
- Department of Psychology, Temple University, Philadelphia, PA 19122, USA
| | - R Christopher Pierce
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Heath D Schmidt
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
43
|
Kv4 channel blockade reduces motor and neuropsychiatric symptoms in rodent models of Parkinson’s disease. Behav Pharmacol 2015; 26:91-100. [DOI: 10.1097/fbp.0000000000000107] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
44
|
Ji X, Martin GE. BK channels mediate dopamine inhibition of firing in a subpopulation of core nucleus accumbens medium spiny neurons. Brain Res 2014; 1588:1-16. [PMID: 25219484 DOI: 10.1016/j.brainres.2014.09.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 08/25/2014] [Accepted: 09/04/2014] [Indexed: 10/24/2022]
Abstract
Dopamine, a key neurotransmitter mediating the rewarding properties of drugs of abuse, is widely believed to exert some of its effects by modulating neuronal activity of nucleus accumbens (NAcc) medium spiny neurons (MSNs). Although its effects on synaptic transmission have been well documented, its regulation of intrinsic neuronal excitability is less understood. In this study, we examined the cellular mechanisms of acute dopamine effects on core accumbens MSNs evoked firing. We found that 0.5 µM A-77636 and 10 µM quinpirole, dopamine D1 (DR1s) and D2 receptor (D2Rs) agonists, respectively, markedly inhibited MSN evoked action potentials. This effect, observed only in about 25% of all neurons, was associated with spike-timing-dependent (STDP) long-term potentiation (tLTP), but not long-term depression (tLTD). Dopamine inhibits evoked firing by compromising subthreshold depolarization, not by altering action potentials themselves. Recordings in voltage-clamp mode revealed that all MSNs expressed fast (IA), slowly inactivating delayed rectifier (Idr), and large conductance voltage- and calcium-activated potassium (BKs) channels. Although A-77636 and quinpirole enhanced IA, its selective blockade by 0.5 µM phrixotoxin-1 had no effect on evoked firing. In contrast, exposing tissue to low TEA concentrations and to 10 µM paxilline, a selective BK channel blocker, prevented D1R agonist from inhibiting MSN firing. This result indicates that dopamine inhibits MSN firing through BK channels in a subpopulation of core accumbens MSNs exclusively associated with spike-timing-dependent long-term potentiation.
Collapse
Affiliation(s)
- Xincai Ji
- University of Massachusetts Medical School, The Brudnick Neuropsychiatric Research Institute, Department of Psychiatry, 303 Belmont Street, Worcester, MA 01604
| | - Gilles E Martin
- University of Massachusetts Medical School, The Brudnick Neuropsychiatric Research Institute, Department of Psychiatry, 303 Belmont Street, Worcester, MA 01604.
| |
Collapse
|
45
|
Krawczyk M, deBacker J, Mason X, Jones AA, Dumont EC. Dopamine decreases NMDA currents in the oval bed nucleus of the stria terminalis of cocaine self-administering rats. Prog Neuropsychopharmacol Biol Psychiatry 2014; 51:83-8. [PMID: 24472317 PMCID: PMC4011798 DOI: 10.1016/j.pnpbp.2014.01.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 01/17/2014] [Accepted: 01/19/2014] [Indexed: 11/17/2022]
Abstract
Dopamine (DA) and N-methyl-D-aspartate receptors (NMDARs) contribute in the neural processes underlying drug-driven behaviors. DA is a potent modulator of NMDAR, but few studies have investigated the functional interaction between DA and NMDAR in the context of substance abuse. We combined the rat model of cocaine self-administration with brain slice electrophysiology to study DA modulation of NMDA currents in the oval bed nucleus of the stria terminalis (ovBNST), a dense DA terminal field involved in maintenance of cocaine self-administration amongst other drug related behaviors. Long-Evans rats self-administered intravenous cocaine (0.75 mg/kg/injection) on a progressive ratio (PR) schedule of reinforcement for 15 days and whole-cell patch-clamp recordings were done on the 16th day. DA reduced NMDA currents in brain-slices from cocaine self-administering rats, but not in those of drug-naïve and sucrose self-administering, or when cocaine exposure was passive (yoked), revealing a mechanism unique to voluntary cocaine intake. DA reduced NMDA currents by activating G-protein-coupled D1- and D2-like receptors that converged on phospholipase C and protein phosphatases. Accordingly, our study reveals a mechanism that may contribute to dysfunctional synaptic plasticity associated with drug-driven behaviors during acute withdrawal.
Collapse
Affiliation(s)
- Michal Krawczyk
- Department of Biomedical and Molecular Sciences, Center for Neuroscience Studies, Queen's University, 99 University Avenue, Kingston, Ontario K7L 3N6, Canada
| | - Julian deBacker
- Department of Biomedical and Molecular Sciences, Center for Neuroscience Studies, Queen's University, 99 University Avenue, Kingston, Ontario K7L 3N6, Canada
| | - Xenos Mason
- Department of Biomedical and Molecular Sciences, Center for Neuroscience Studies, Queen's University, 99 University Avenue, Kingston, Ontario K7L 3N6, Canada
| | - Andrea A Jones
- Department of Biomedical and Molecular Sciences, Center for Neuroscience Studies, Queen's University, 99 University Avenue, Kingston, Ontario K7L 3N6, Canada
| | - Eric C Dumont
- Department of Biomedical and Molecular Sciences, Center for Neuroscience Studies, Queen's University, 99 University Avenue, Kingston, Ontario K7L 3N6, Canada.
| |
Collapse
|
46
|
Etter G, Krezel W. Dopamine D2 receptor controls hilar mossy cells excitability. Hippocampus 2014; 24:725-32. [PMID: 24753432 DOI: 10.1002/hipo.22280] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2014] [Indexed: 12/12/2022]
Abstract
Hippocampal control of memory formation is regulated by dopaminergic signaling. Whereas the role of dopamine D1 receptors is well documented in such regulations, functions of dopamine D2 receptors (DRD2) are not fully understood. Using fluorescence in situ hybridization we demonstrate that Drd2 expression in the hippocampus of wild-type mice is limited to glutamatergic hilar mossy cells. Using whole cell electrophysiological recordings in hippocampal slice preparations, we provide evidence that unlike in basal ganglia, activation of DRD2 by the selective agonist, quinpirole, induces a long-lasting increase in excitability of hilar mossy cells, which can be blocked by the DRD2 antagonist raclopride. Such activity is mediated by the Akt/GSK pathway, as application of specific inhibitors such as A1070722 or SB216763 prevented quinpirole activity. Long-term effects of acute DRD2 activation in vitro suggest that volume transmission of dopamine may modulate mossy cell activities in vivo. This is supported by the presence of dense tyrosine hydroxylase positive varicosities in the hilus, which are rarely seen in the vicinity of mossy cell dendrites. From these data we discuss how dopamine could control mossy cell activity and thus dentate gyrus functions.
Collapse
Affiliation(s)
- Guillaume Etter
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, UMR 7104 CNRS, U 964 INSERM, Université de Strasbourg, B.P. 10142, 67404 Illkirch, Cedex, France
| | | |
Collapse
|
47
|
Nguyen CL, Tran AH, Matsumoto J, Hori E, Uwano T, Ono T, Nishijo H. Hippocampal place cell responses to distal and proximal cue manipulations in dopamine D2 receptor-knockout mice. Brain Res 2014; 1567:13-27. [PMID: 24747614 DOI: 10.1016/j.brainres.2014.04.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 04/11/2014] [Accepted: 04/13/2014] [Indexed: 10/25/2022]
Abstract
The human hippocampus is critical for learning and memory. In rodents, hippocampal pyramidal neurons fire in a location-specific manner and form relational representations of environmental cues. The important roles of dopaminergic D1 receptors in learning and in hippocampal neural synaptic plasticity in novel environments have been previously shown. However, the roles of D2 receptors in hippocampal neural plasticity in response to novel and familiar spatial stimuli remain unclear. In order to clarify this issue, we recorded from hippocampal neurons in dopamine D2 receptor-knockout (D2R-KO) mice and their wild-type (WT) littermates during manipulations of distinct spatial cues in familiar and novel environments. Here, we report that D2R-KO mice showed substantial deficits in place-cell properties (number of place cells, intra-field firing rates, spatial tuning, and spatial coherence). Furthermore, although place cells in D2R-KO mice responded to manipulations of distal and proximal cues in both familiar and novel environments in a manner that was similar to place cells in WT mice, place fields were less stable in the D . The axes represent the differences between the peak and the valley of each waveform of EL2 and EL3.2R-KO mice in the familiar environment, but not in the novel environment. The present results suggested that D2 receptors in the hippocampus are important for place response stability. The place-cell properties of D2R-KO mice were similar to aged animals, suggesting that the alterations of place-cell properties in aged animals might be ascribed partly to alterations in the D2R in the HF of aged animals.
Collapse
Affiliation(s)
- Chien Le Nguyen
- System Emotional Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Anh Hai Tran
- System Emotional Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Jumpei Matsumoto
- System Emotional Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Etsuro Hori
- System Emotional Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Teruko Uwano
- Integrative Neuroscience, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Taketoshi Ono
- Integrative Neuroscience, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Hisao Nishijo
- System Emotional Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan.
| |
Collapse
|
48
|
Steinberg EE, Boivin JR, Saunders BT, Witten IB, Deisseroth K, Janak PH. Positive reinforcement mediated by midbrain dopamine neurons requires D1 and D2 receptor activation in the nucleus accumbens. PLoS One 2014; 9:e94771. [PMID: 24733061 PMCID: PMC3986242 DOI: 10.1371/journal.pone.0094771] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 03/20/2014] [Indexed: 12/17/2022] Open
Abstract
The neural basis of positive reinforcement is often studied in the laboratory using intracranial self-stimulation (ICSS), a simple behavioral model in which subjects perform an action in order to obtain exogenous stimulation of a specific brain area. Recently we showed that activation of ventral tegmental area (VTA) dopamine neurons supports ICSS behavior, consistent with proposed roles of this neural population in reinforcement learning. However, VTA dopamine neurons make connections with diverse brain regions, and the specific efferent target(s) that mediate the ability of dopamine neuron activation to support ICSS have not been definitively demonstrated. Here, we examine in transgenic rats whether dopamine neuron-specific ICSS relies on the connection between the VTA and the nucleus accumbens (NAc), a brain region also implicated in positive reinforcement. We find that optogenetic activation of dopaminergic terminals innervating the NAc is sufficient to drive ICSS, and that ICSS driven by optical activation of dopamine neuron somata in the VTA is significantly attenuated by intra-NAc injections of D1 or D2 receptor antagonists. These data demonstrate that the NAc is a critical efferent target sustaining dopamine neuron-specific ICSS, identify receptor subtypes through which dopamine acts to promote this behavior, and ultimately help to refine our understanding of the neural circuitry mediating positive reinforcement.
Collapse
Affiliation(s)
- Elizabeth E. Steinberg
- Ernest Gallo Clinic and Research Center, Department of Neurology, University of California at San Francisco, San Francisco, California, United States of America
- Graduate Program in Neuroscience, University of California at San Francisco, San Francisco, California, United States of America
- * E-mail:
| | - Josiah R. Boivin
- Ernest Gallo Clinic and Research Center, Department of Neurology, University of California at San Francisco, San Francisco, California, United States of America
- Graduate Program in Neuroscience, University of California at San Francisco, San Francisco, California, United States of America
| | - Benjamin T. Saunders
- Ernest Gallo Clinic and Research Center, Department of Neurology, University of California at San Francisco, San Francisco, California, United States of America
| | - Ilana B. Witten
- Princeton Neuroscience Institute and Department of Psychology, Princeton University, Princeton, New Jersey, United States of America
| | - Karl Deisseroth
- Department of Bioengineering, Department of Psychiatry and Behavioral Sciences, Howard Hughes Medical Institute, and CNC Program, Stanford University, Stanford, California, United States of America
| | - Patricia H. Janak
- Ernest Gallo Clinic and Research Center, Department of Neurology, University of California at San Francisco, San Francisco, California, United States of America
- Wheeler Center for the Neurobiology of Addiction, University of California at San Francisco, San Francisco, California, United States of America
| |
Collapse
|
49
|
James AS, Chen JY, Cepeda C, Mittal N, Jentsch JD, Levine MS, Evans CJ, Walwyn W. Opioid self-administration results in cell-type specific adaptations of striatal medium spiny neurons. Behav Brain Res 2013; 256:279-83. [PMID: 23968589 DOI: 10.1016/j.bbr.2013.08.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 08/02/2013] [Accepted: 08/05/2013] [Indexed: 11/28/2022]
Abstract
Medium-sized spiny neurons (MSNs), the predominant neuronal population of the striatum, are an integral component of the many cortical and limbic pathways associated with reward-related behaviors. A differential role of the D1 receptor-enriched (D1) MSNs of the striatonigral direct pathway, as compared with the D2 receptor-enriched (D2) MSNs of the striatopallidal indirect pathway, in mediating the addictive behaviors associated with cocaine is beginning to emerge. However, whether opioids, well-known analgesics with euphoric properties, similarly induce dissociable signaling adaptations in these neurons remains unclear. Transgenic mice expressing green fluorescent protein (GFP)-labeled D1 or D2 neurons were implanted with intravenous jugular catheters. Mice learned to self-administer 0.1mg/kg/infusion of the opioid remifentanil during 2h sessions over 13 contiguous days. Thereafter, the electrophysiological properties of D1- and D2-MSNs in the shell region of the nucleus accumbens (NAc) were assessed. We found that prior opioid exposure did not alter the basic membrane properties nor the kinetics or amplitude of miniature excitatory postsynaptic currents (mEPSCs). However, when challenged with the mu opioid receptor (μOR) agonist DAMGO, the characteristic inhibitory profile of this receptor was altered. DAMGO inhibited the frequency of mEPSCs in D1-MSNs from control mice receiving saline and in D2-MSNs from mice exposed to remifentanil or saline, but this inhibitory profile was reduced in D1-MSNs from mice receiving remifentanil. Remifentanil exposure also altered the probability of glutamate release onto D1-, but not D2-MSNs. Together these results suggest a D1-pathway specific effect associated with the acquisition of opioid-seeking behaviors.
Collapse
Affiliation(s)
- Alex S James
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Hopf FW, Seif T, Chung S, Civelli O. MCH and apomorphine in combination enhance action potential firing of nucleus accumbens shell neurons in vitro. PeerJ 2013; 1:e61. [PMID: 23646281 PMCID: PMC3642701 DOI: 10.7717/peerj.61] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 03/12/2013] [Indexed: 11/20/2022] Open
Abstract
The MCH and dopamine receptor systems have been shown to modulate a number of behaviors related to reward processing, addiction, and neuropsychiatric conditions such as schizophrenia and depression. In addition, MCH and dopamine receptors can interact in a positive manner, for example in the expression of cocaine self-administration. A recent report (Chung et al., 2011a) showed that the DA1/DA2 dopamine receptor activator apomorphine suppresses pre-pulse inhibition, a preclinical model for some aspects of schizophrenia. Importantly, MCH can enhance the effects of lower doses of apomorphine, suggesting that co-modulation of dopamine and MCH receptors might alleviate some symptoms of schizophrenia with a lower dose of dopamine receptor modulator and thus fewer potential side effects. Here, we investigated whether MCH and apomorphine could enhance action potential firing in vitro in the nucleus accumbens shell (NAshell), a region which has previously been shown to mediate some behavioral effects of MCH. Using whole-cell patch-clamp electrophysiology, we found that MCH, which has no effect on firing on its own, was able to increase NAshell firing when combined with a subthreshold dose of apomorphine. Further, this MCH/apomorphine increase in firing was prevented by an antagonist of either a DA1 or a DA2 receptor, suggesting that apomorphine acts through both receptor types to enhance NAshell firing. The MCH/apomorphine-mediated firing increase was also prevented by an MCH receptor antagonist or a PKA inhibitor. Taken together, our results suggest that MCH can interact with lower doses of apomorphine to enhance NAshell firing, and thus that MCH and apomorphine might interact in vivo within the NAshell to suppress pre-pulse inhibition.
Collapse
Affiliation(s)
- F Woodward Hopf
- Ernest Gallo Clinic and Research Center, Department of Neurology, University of California , San Francisco, Emeryville, CA , USA
| | | | | | | |
Collapse
|