1
|
Khosroshahi PA, Ghanbari M. MicroRNA dysregulation in glutamate and dopamine pathways of schizophrenia: From molecular pathways to diagnostic and therapeutic approaches. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111081. [PMID: 39002925 DOI: 10.1016/j.pnpbp.2024.111081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 06/28/2024] [Accepted: 07/07/2024] [Indexed: 07/15/2024]
Abstract
Schizophrenia is a complex psychiatric disorder, and genetic and environmental factors have been implicated in its development. Dysregulated glutamatergic and dopaminergic transmission pathways are involved in schizophrenia development. Besides genetic mutations, epigenetic dysregulation has a considerable role in dysregulating molecular pathways involved in schizophrenia. MicroRNAs (miRNAs) are small, non-coding RNAs that target specific mRNAs and inhibit their translation into proteins. As epigenetic factors, miRNAs regulate many genes involved in glutamate and dopamine signaling pathways; thereby, their dysregulation can contribute to the development of schizophrenia. Secretion of specific miRNAs from damaged cells into body fluids can make them one of the ideal non-invasive biomarkers in the early diagnosis of schizophrenia. Also, understanding the molecular mechanisms of miRNAs in schizophrenia pathogenesis can pave the way for developing novel treatments for patients with schizophrenia. In this study, we reviewed the glutamatergic and dopaminergic pathophysiology and highlighted the role of miRNA dysregulation in schizophrenia development. Besides, we shed light on the significance of circulating miRNAs for schizophrenia diagnosis and the recent findings on the miRNA-based treatment for schizophrenia.
Collapse
Affiliation(s)
| | - Mohammad Ghanbari
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
2
|
Littlepage-Saunders M, Hochstein MJ, Chang DS, Johnson KA. G protein-coupled receptor modulation of striatal dopamine transmission: Implications for psychoactive drug effects. Br J Pharmacol 2024; 181:4399-4413. [PMID: 37258878 PMCID: PMC10687321 DOI: 10.1111/bph.16151] [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: 01/31/2023] [Revised: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023] Open
Abstract
Dopamine transmission in the striatum is a critical mediator of the rewarding and reinforcing effects of commonly misused psychoactive drugs. G protein-coupled receptors (GPCRs) that bind a variety of neuromodulators including dopamine, endocannabinoids, acetylcholine and endogenous opioid peptides regulate dopamine release by acting on several components of dopaminergic circuitry. Striatal dopamine release can be driven by both somatic action potential firing and local mechanisms that depend on acetylcholine released from striatal cholinergic interneurons. GPCRs that primarily regulate somatic firing of dopamine neurons via direct effects or modulation of synaptic inputs are likely to affect distinct aspects of behaviour and psychoactive drug actions compared with those GPCRs that primarily regulate local acetylcholine-dependent dopamine release in striatal regions. This review will highlight mechanisms by which GPCRs modulate dopaminergic transmission and the relevance of these findings to psychoactive drug effects on physiology and behaviour.
Collapse
Affiliation(s)
- Mydirah Littlepage-Saunders
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Neuroscience Graduate Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Michael J Hochstein
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Doris S Chang
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Kari A Johnson
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Neuroscience Graduate Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Spicer MM, Weber MA, Luo Z, Yang J, Narayanan NS, Fisher RA. Regulator of G protein signaling 6 (RGS6) in dopamine neurons promotes EtOH seeking, behavioral reward, and susceptibility to relapse. Psychopharmacology (Berl) 2024; 241:2255-2269. [PMID: 38856764 PMCID: PMC11518640 DOI: 10.1007/s00213-024-06631-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 05/31/2024] [Indexed: 06/11/2024]
Abstract
Mesolimbic dopamine (DA) transmission is believed to play a critical role in mediating reward responses to drugs of abuse, including alcohol (EtOH). The neurobiological mechanisms underlying EtOH-seeking behavior and dependence are not fully understood, and abstinence remains the only effective way to prevent alcohol use disorders (AUDs). Here, we developed novel RGS6fl/fl; DAT-iCreER mice to determine the role of RGS6 in DA neurons on EtOH consumption, reward, and relapse behaviors. We found that RGS6 is expressed in DA neurons in both human and mouse ventral tegmental area (VTA), and that RGS6 loss in mice upregulates DA transporter (DAT) expression in VTA DA neuron synaptic terminals. Remarkably, loss of RGS6 in DA neurons significantly reduced EtOH consumption, preference, and reward in a manner indistinguishable from that seen in RGS6-/- mice. Strikingly, RGS6 loss from DA neurons before or after EtOH behavioral reward is established significantly reduced (~ 50%) re-instatement of reward following extinguishment, demonstrating distinct roles of RGS6 in promoting reward and relapse susceptibility to EtOH. These studies identify DA neurons as the locus of RGS6 action in promoting EtOH consumption, preference, reward, and relapse. RGS6 is unique among R7 RGS proteins in promoting rather than suppressing behavioral responses to drugs of abuse and to modulate EtOH behavioral reward. This is a result of RGS6's pre-synaptic actions that we hypothesize promote VTA DA transmission by suppressing GPCR-Gαi/o-DAT signaling in VTA DA neurons. These studies identify RGS6 as a potential therapeutic target for behavioral reward and relapse to EtOH.
Collapse
Affiliation(s)
- Mackenzie M Spicer
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 51 Newton Rd. BSB 2-512, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA
- Department of Neurology, University of Iowa Carver College of Medicine, 200 Hawkins Dr., Iowa City, IA, 52242, USA
| | - Matthew A Weber
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA
- Department of Neurology, University of Iowa Carver College of Medicine, 200 Hawkins Dr., Iowa City, IA, 52242, USA
| | - Zili Luo
- Department of Pediatrics, University of Iowa Carver College of Medicine, 200 Hawkins Dr., Iowa City, IA, 52242, USA
| | - Jianqi Yang
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 51 Newton Rd. BSB 2-512, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA
| | - Nandakumar S Narayanan
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA
- Department of Neurology, University of Iowa Carver College of Medicine, 200 Hawkins Dr., Iowa City, IA, 52242, USA
| | - Rory A Fisher
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 51 Newton Rd. BSB 2-512, Iowa City, IA, 52242, USA.
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA.
| |
Collapse
|
4
|
Haas S, Bravo F, Ionescu TM, Gonzalez-Menendez I, Quintanilla-Martinez L, Dunkel G, Kuebler L, Hahn A, Lanzenberger R, Weigelin B, Reischl G, Pichler BJ, Herfert K. Functional PET/MRI reveals active inhibition of neuronal activity during optogenetic activation of the nigrostriatal pathway. SCIENCE ADVANCES 2024; 10:eadn2776. [PMID: 39454014 PMCID: PMC11506239 DOI: 10.1126/sciadv.adn2776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 09/23/2024] [Indexed: 10/27/2024]
Abstract
The dopaminergic system is a central component of the brain's neurobiological framework, governing motor control and reward responses and playing an essential role in various brain disorders. Within this complex network, the nigrostriatal pathway represents a critical circuit for dopamine neurotransmission from the substantia nigra to the striatum. However, stand-alone functional magnetic resonance imaging is unable to study the intricate interplay between brain activation and its molecular underpinnings. In our study, the use of a functional [fluorine-18]2-fluor-2-deoxy-d-glucose positron emission tomography approach, simultaneously with blood oxygen level-dependent functional magnetic resonance imaging, provided an important insight that demonstrates an active suppression of the nigrostriatal activity during optogenetic stimulation. This result increases our understanding of the molecular mechanisms of brain function and provides an important perspective on how dopamine influences hemodynamic responses in the brain.
Collapse
Affiliation(s)
- Sabrina Haas
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Fernando Bravo
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Tudor M. Ionescu
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Irene Gonzalez-Menendez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, Eberhard Karls University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, Eberhard Karls University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Gina Dunkel
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Laura Kuebler
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Andreas Hahn
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Bettina Weigelin
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Gerald Reischl
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Bernd J. Pichler
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Kristina Herfert
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| |
Collapse
|
5
|
Lee NC, Hsu PC, Liu YH, Wang HC, Chen TI, Chien YH, Hwu WL. Nigrostriatal tract defects in mice with aromatic l-amino acid decarboxylase deficiency. Neurobiol Dis 2024; 202:106707. [PMID: 39433135 DOI: 10.1016/j.nbd.2024.106707] [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: 07/21/2024] [Revised: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 10/23/2024] Open
Abstract
The development of the nigrostriatal dopaminergic (DA) pathway in the brain involves many transcriptional and chemotactic molecules, and a deficiency of these molecules can cause nigrostriatal tract defects. However, the role of the end product, dopamine, in nigrostriatal pathway development has not been described. In the present study, we analyzed a mouse model of congenital dopamine and serotonin deficiency, namely, the aromatic l-amino acid decarboxylase (AADC) deficiency (DdcKI) mouse model. We found via tyrosine hydroxylase (TH) immunofluorescence staining that the number of DA fibers in the stratum of 14-day-old DdcKI mice decreased. In TH-stained cleared whole brains of DdcKI mice, the numbers of DA neurons in the substantia nigra (SN) and the number of DA nerve bundles leaving the SN were both normal. However, we found that the nigrostriatal bundles in DdcKI mice were dispersed, taking aberrant routes to the striatum and spreading over a wide area. The total volume occupied by the nigrostriatal tract was increased, and the fraction of TH staining in the tract was decreased in DdcKI mice. Single-nucleus RNA sequencing analysis for mice 0, 7, and 14 days of age, revealed delayed axonogenesis and synapse formation in the striatum of DdcKI mice. The CellChat program inferred less cell-cell communication between striatal D1/D2 neurons but increased cell-cell communication involving neural precursors in DdcKI mice. Therefore, a congenital deficiency in dopamine affects nigrostriatal axon extension and striatal innervation. These nigrostriatal tract defects may limit the treatment efficacy for patients with TH or AADC deficiency.
Collapse
Affiliation(s)
- Ni-Chung Lee
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Pei-Chun Hsu
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Han Liu
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hao-Chun Wang
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Tsu-I Chen
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yin-Hsiu Chien
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wuh-Liang Hwu
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; Center for Precision Medicine, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
6
|
Michorowska S, Kucharski D, Chojnacka J, Nałęcz-Jawecki G, Marek D, Giebułtowicz J. Metabolomic study on ostracods exposed to environmentally relevant concentrations of five pharmaceuticals selected via a novel approach. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 946:174036. [PMID: 38889824 DOI: 10.1016/j.scitotenv.2024.174036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
Pharmaceuticals (PhACs) are increasingly detected in aquatic ecosystems, yet their effects on biota remain largely unknown. The environmentally relevant concentrations of many PhACs may not result in individual-level responses, like mortality or growth inhibition, traditional toxicity endpoints. However, this doesn't imply the absence of negative effects on biota. Metabolomics offers a more sensitive approach, detecting responses at molecular and cellular levels and providing mechanistic understanding of adverse effects. We evaluated bioaccumulation and metabolic alterations in a benthic ostracod, Heterocypris incongruens, exposed to a mixture of five PhACs (carbamazepine, tiapride, tolperisone, propranolol and amlodipine) at environmentally relevant concentrations for 7 days using liquid chromatography coupled with mass spectrometry. The selection of PhACs was based, among other factors, on risk quotient values determined using toxicological data available in the literature and concentrations of PhACs quantified in our previous research in the sediments of the Odra River estuary. This represents a novel approach to PhACs selection for metabolomic studies that considers strictly quantitative data. Amlodipine and tolperisone exhibited the highest bioaccumulation. Significant impacts were observed in Alanine, aspartate and glutamate metabolism, Starch and sucrose metabolism, Arginine biosynthesis, Histidine metabolism, Tryptophan metabolism, Glycerophospholipid metabolism, and Glutathione metabolism pathways. Most of the below-individual-level responses were likely nonspecific and related to dysregulation in energy metabolism and oxidative stress response. Additionally, some pharmaceutical-specific responses were also observed. Therefore, untargeted metabolomics can be used to detect metabolic changes resulting from environmentally relevant concentrations of PhACs in aquatic ecosystems and to understand their underlying mechanism.
Collapse
Affiliation(s)
- Sylwia Michorowska
- Department of Drug Chemistry, Pharmaceutical and Biomedical Analysis, Medical University of Warsaw, Banacha 1 Str., 02-097 Warszawa, Poland
| | - Dawid Kucharski
- Department of Drug Chemistry, Pharmaceutical and Biomedical Analysis, Medical University of Warsaw, Banacha 1 Str., 02-097 Warszawa, Poland
| | - Justyna Chojnacka
- Department of Toxicology and Food Science, Medical University of Warsaw, Banacha 1 Str., 02-097 Warszawa, Poland
| | - Grzegorz Nałęcz-Jawecki
- Department of Toxicology and Food Science, Medical University of Warsaw, Banacha 1 Str., 02-097 Warszawa, Poland
| | - Dominik Marek
- Department of Drug Chemistry, Pharmaceutical and Biomedical Analysis, Medical University of Warsaw, Banacha 1 Str., 02-097 Warszawa, Poland
| | - Joanna Giebułtowicz
- Department of Drug Chemistry, Pharmaceutical and Biomedical Analysis, Medical University of Warsaw, Banacha 1 Str., 02-097 Warszawa, Poland.
| |
Collapse
|
7
|
Gowripriya T, Yashwanth R, James PB, Suresh R, Balamurugan K. Dopaminergic neuronal regulation determines innate immunity of Caenorhabditis elegans during Klebsiella aerogenes infection. Microbes Infect 2024:105430. [PMID: 39369984 DOI: 10.1016/j.micinf.2024.105430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 09/06/2024] [Accepted: 10/02/2024] [Indexed: 10/08/2024]
Abstract
The innate immune signals are the front line of host defense against bacterial pathogens. Pathogen-induced harmful effects, such as reduced neuronal signals to the intestine, affect the host's food sensing and dwelling behavior. Here, we report that dopamine and kpc-1 signals control the intestinal innate immune responses through the p38/PMK-1 MAPK signaling pathway in C. elegans. K. aerogenes infection in C. elegans affects the food-dwelling behavior, which depends on dopamine regulation. The absence of the dopamine receptor (dop-1) and transporter (dat-1) increases attraction to the pathogen instead of avoidance. The K. aerogenes infection affects age-1 regulation through the furin-like proprotein convertase (kpc-1); the absence of kpc-1 affects environment-dependent dauer formation. In contrast, the dop-1 mutation antagonistically regulates intestinal immune regulation, while the kpc-1 mutation partially regulates the p38/PMK-1 MAPK pathway. Our findings indicate that dopamine and kpc-1signaling from the nervous system control intestinal immunity in an antagonistic and agonistic manner, respectively.
Collapse
Affiliation(s)
- Thirumugam Gowripriya
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, 630 003, India
| | - Radhakrishnan Yashwanth
- ITC Life Sciences and Technology Centre, Peenya Industrial Area, Bangalore, 560 058, Karnataka, India
| | - Prabhanand Bhaskar James
- ITC Life Sciences and Technology Centre, Peenya Industrial Area, Bangalore, 560 058, Karnataka, India
| | - Ramamurthi Suresh
- ITC Life Sciences and Technology Centre, Peenya Industrial Area, Bangalore, 560 058, Karnataka, India
| | | |
Collapse
|
8
|
D'Aquila PS. Licking microstructure in response to novel rewards, reward devaluation and dopamine antagonists: Possible role of D1 and D2 medium spiny neurons in the nucleus accumbens. Neurosci Biobehav Rev 2024; 165:105861. [PMID: 39159734 DOI: 10.1016/j.neubiorev.2024.105861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/29/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
Evidence on the effect of dopamine D1 and D2-like antagonists and of manipulations of reward value on licking microstructure is reanalysed considering recent findings on the role of nucleus accumbens (NAc) medium spiny neurons (MSNs) in the control of sugar intake. The results of this analysis suggest that D1 MSN activation, which is involved in the emission of licking bursts, might play a crucial role in response to novel rewards. D2 MSN activation, which results in reduction of burst size and suppression of licking, might mediate the response to reward devaluation. Elucidating the neural mechanisms underlying the licking response might lead to a better definition of its microstructural measures in behaviourally and psychologically meaningful functional terms. This could further support its use as a behavioural substrate in the study of the neural mechanisms of ingestive behaviour and motivation, as well as in animal models of pathological conditions such as eating disorders and obesity.
Collapse
Affiliation(s)
- Paolo S D'Aquila
- Dipartimento di Scienze Biomediche, Università di Sassari, Viale S. Pietro 43/b, Sassari 07100, Italy.
| |
Collapse
|
9
|
Bassareo V, Maccioni R, Talani G, Zuffa S, El Abiead Y, Lorrai I, Kawamura T, Pantis S, Puliga R, Vargiu R, Lecca D, Enrico P, Peana A, Dazzi L, Dorrestein PC, Sanna PP, Sanna E, Acquas E. Receptor and metabolic insights on the ability of caffeine to prevent alcohol-induced stimulation of mesolimbic dopamine transmission. Transl Psychiatry 2024; 14:391. [PMID: 39341817 PMCID: PMC11438888 DOI: 10.1038/s41398-024-03112-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024] Open
Abstract
The consumption of alcohol and caffeine affects the lives of billions of individuals worldwide. Although recent evidence indicates that caffeine impairs the reinforcing properties of alcohol, a characterization of its effects on alcohol-stimulated mesolimbic dopamine (DA) function was lacking. Acting as the pro-drug of salsolinol, alcohol excites DA neurons in the posterior ventral tegmental area (pVTA) and increases DA release in the nucleus accumbens shell (AcbSh). Here we show that caffeine, via antagonistic activity on A2A adenosine receptors (A2AR), prevents alcohol-dependent activation of mesolimbic DA function as assessed, in-vivo, by brain microdialysis of AcbSh DA and, in-vitro, by electrophysiological recordings of pVTA DA neuronal firing. Accordingly, while the A1R antagonist DPCPX fails to prevent the effects of alcohol on DA function, both caffeine and the A2AR antagonist SCH 58261 prevent alcohol-dependent pVTA generation of salsolinol and increase in AcbSh DA in-vivo, as well as alcohol-dependent excitation of pVTA DA neurons in-vitro. However, caffeine also prevents direct salsolinol- and morphine-stimulated DA function, suggesting that it can exert these inhibitory effects also independently from affecting alcohol-induced salsolinol formation or bioavailability. Finally, untargeted metabolomics of the pVTA showcases that caffeine antagonizes alcohol-mediated effects on molecules (e.g. phosphatidylcholines, fatty amides, carnitines) involved in lipid signaling and energy metabolism, which could represent an additional salsolinol-independent mechanism of caffeine in impairing alcohol-mediated stimulation of mesolimbic DA transmission. In conclusion, the outcomes of this study strengthen the potential of caffeine, as well as of A2AR antagonists, for future development of preventive/therapeutic strategies for alcohol use disorder.
Collapse
Affiliation(s)
- Valentina Bassareo
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Riccardo Maccioni
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.
| | - Giuseppe Talani
- Institute of Neuroscience - National Research Council (C.N.R.) of Italy, Cagliari, Italy
| | - Simone Zuffa
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Yasin El Abiead
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Irene Lorrai
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Tomoya Kawamura
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Sofia Pantis
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- Department of Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Roberta Puliga
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Romina Vargiu
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Daniele Lecca
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Paolo Enrico
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Alessandra Peana
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Laura Dazzi
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Pietro Paolo Sanna
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Enrico Sanna
- Institute of Neuroscience - National Research Council (C.N.R.) of Italy, Cagliari, Italy
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Elio Acquas
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| |
Collapse
|
10
|
Saggu S, Bai A, Aida M, Rehman H, Pless A, Ware D, Deak F, Jiao K, Wang Q. Monoamine alterations in Alzheimer's disease and their implications in comorbid neuropsychiatric symptoms. GeroScience 2024:10.1007/s11357-024-01359-x. [PMID: 39331291 DOI: 10.1007/s11357-024-01359-x] [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: 05/22/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder characterized by relentless cognitive decline and the emergence of profoundly disruptive neuropsychiatric symptoms. As the disease progresses, it unveils a formidable array of neuropsychiatric manifestations, including debilitating depression, anxiety, agitation, and distressing episodes of psychosis. The intricate web of the monoaminergic system, governed by serotonin, dopamine, and norepinephrine, significantly influences our mood, cognition, and behavior. Emerging evidence suggests that dysregulation and degeneration of this system occur early in AD, leading to notable alterations in these critical neurotransmitters' levels, metabolism, and receptor function. However, how the degeneration of monoaminergic neurons and subsequent compensatory changes contribute to the presentation of neuropsychiatric symptoms observed in Alzheimer's disease remains elusive. This review synthesizes current findings on monoamine alterations in AD and explores how these changes contribute to the neuropsychiatric symptomatology of the disease. By elucidating the biological underpinnings of AD-related psychiatric symptoms, we aim to underscore the complexity and inform innovative approaches for treating neuropsychiatric symptoms in AD.
Collapse
Affiliation(s)
- Shalini Saggu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| | - Ava Bai
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Mae Aida
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Hasibur Rehman
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Andrew Pless
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Destany Ware
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Ferenc Deak
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Kai Jiao
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Qin Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
11
|
Bardgett ME, Griffith MS, Robinson KR, Stevens RM, Gannon MA, Knuth MD, Hawk GS, Pauly JR. Early-life risperidone alters locomotor responses to apomorphine and quinpirole in adulthood. Behav Brain Res 2024; 473:115171. [PMID: 39094954 PMCID: PMC11345744 DOI: 10.1016/j.bbr.2024.115171] [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: 04/12/2024] [Revised: 07/12/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
An escalating trend of antipsychotic drug use in children with ADHD, disruptive behavior disorder, or mood disorders has raised concerns about the impact of these drugs on brain development. Since antipsychotics chiefly target dopamine receptors, it is important to assay the function of these receptors after early-life antipsychotic administration. Using rats as a model, we examined the effects of early-life risperidone, the most prescribed antipsychotic drug in children, on locomotor responses to the dopamine D1/D2 receptor agonist, apomorphine, and the D2/D3 receptor agonist, quinpirole. Female and male Long-Evans rats received daily subcutaneous injections of risperidone (1.0 and 3.0 mg/kg) or vehicle from postnatal day 14-42. Locomotor responses to one of three doses (0.03, 0.1, and 0.3 mg/kg) of apomorphine or quinpirole were tested once a week for four weeks beginning on postnatal day 76 and 147 for each respective drug. The locomotor activity elicited by the two lower doses of apomorphine was significantly greater in adult rats, especially females, administered risperidone early in life. Adult rats administered risperidone early in life also showed more locomotor activity after the low dose of quinpirole. Overall, female rats were more sensitive to the locomotor effects of each agonist. In a separate group of rats administered risperidone early in life, autoradiography of forebrain D2 receptors at postnatal day 62 revealed a modest increase in D2 receptor density in the medial caudate. These results provide evidence that early-life risperidone administration can produce long-lasting changes in dopamine receptor function and density.
Collapse
Affiliation(s)
- Mark E Bardgett
- Department of Psychological Science, Northern Kentucky University, Highland Heights, KY 41076, United States.
| | - Molly S Griffith
- Department of Psychological Science, Northern Kentucky University, Highland Heights, KY 41076, United States
| | - Kathleen R Robinson
- Department of Psychological Science, Northern Kentucky University, Highland Heights, KY 41076, United States
| | - Rachel M Stevens
- Department of Psychological Science, Northern Kentucky University, Highland Heights, KY 41076, United States
| | - Matthew A Gannon
- Department of Psychological Science, Northern Kentucky University, Highland Heights, KY 41076, United States
| | - Meghan D Knuth
- Department of Psychological Science, Northern Kentucky University, Highland Heights, KY 41076, United States
| | - Gregory S Hawk
- Department of Statistics, College of Arts and Sciences, University of Kentucky, Lexington, KY 40504, United States; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40504, United States
| | - James R Pauly
- Department of Statistics, College of Arts and Sciences, University of Kentucky, Lexington, KY 40504, United States; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40504, United States
| |
Collapse
|
12
|
Zack M, Behzadi A, Biback C, Chugani B, DiGiacomo D, Fang T, Houle S, Kalia A, Lobo D, Payer D, Poulos CX, Rusjan PM, Smart K, Tatone D, Warsh J, Wilson AA, Kennedy JL. Dopamine mediates a directionally opposite correlation between empathy and the reinforcing effects of amphetamine and gambling in people with gambling disorder vs. healthy controls. Pharmacol Biochem Behav 2024; 245:173865. [PMID: 39236810 DOI: 10.1016/j.pbb.2024.173865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 06/05/2024] [Accepted: 08/25/2024] [Indexed: 09/07/2024]
Abstract
Understanding the relationship between empathy, subjective effects of addictive reinforcers and dopamine function in people with gambling disorder (PGD) vs. healthy controls (HCs) may inform GD treatment. The current investigation addressed this issue via retrospective analysis of data from three studies using amphetamine and a slot machine (SLOTS) as reinforcers in PGD and HCs. The Empathy scale of Eysenck's Impulsiveness Questionnaire assessed trait Empathy. The Gamblers Beliefs Questionnaire assessed cognitive distortions. The Eysenck Lie scale assessed socially desirable responding. PET scans quantified dopamine receptor expression and amphetamine-induced dopamine release in Study 1. Pre-treatment with the D2-receptor (D2R)-preferring antagonist, haloperidol or D1R-D2R antagonist, fluphenazine before SLOTS tested the role of D2 autoreceptors and post-synaptic D2R in Study 2. Pre-treatment with the multi-system indirect dopamine agonist, modafinil before SLOTS assessed the reliability of correlations in PGD. Striatal D2R expression predicted greater Empathy and lower amphetamine 'Liking' in HCs, and predicted greater symptom severity in PGD. Empathy predicted lower 'Exciting' effects of SLOTS under placebo in HCs; no correlation emerged under either antagonist. Relative to placebo, haloperidol decreased, whereas fluphenazine increased, the positive correlation between Empathy and Exciting effects of SLOTS in PGD. Modafinil markedly reduced the positive correlation between Empathy and Exciting effects of SLOTS seen under placebo in PGD. Empathy predicted greater cognitive distortions in PGD in all studies. Lie scale variance influenced several primary effects. Prior research linking the insula with Empathy, reactivity to interoceptive signals for risky rewards (uncertainty), and cognitive distortions, provides a parsimonious account for these results.
Collapse
Affiliation(s)
- Martin Zack
- Molecular Brain Sciences Dept., Centre for Addiction & Mental Health, Toronto, ON M5T 1R8, Canada; Dept. of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Arian Behzadi
- Vivian M. Rakoff PET Centre, Centre for Addiction & Mental Health, Toronto, ON M5T 1R8, Canada
| | - Candice Biback
- Dept. of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Bindiya Chugani
- Dept. of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Dan DiGiacomo
- Addiction Psychiatry Service, Centre for Addiction & Mental Health, Toronto, ON M6J 1H4, Canada
| | - Tim Fang
- Dept. of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sylvain Houle
- Vivian M. Rakoff PET Centre, Centre for Addiction & Mental Health, Toronto, ON M5T 1R8, Canada
| | - Aditi Kalia
- Dept. of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Daniela Lobo
- Addiction Psychiatry Service, Centre for Addiction & Mental Health, Toronto, ON M6J 1H4, Canada
| | - Doris Payer
- Vivian M. Rakoff PET Centre, Centre for Addiction & Mental Health, Toronto, ON M5T 1R8, Canada
| | - Constantine X Poulos
- Molecular Brain Sciences Dept., Centre for Addiction & Mental Health, Toronto, ON M5T 1R8, Canada
| | - Pablo M Rusjan
- Vivian M. Rakoff PET Centre, Centre for Addiction & Mental Health, Toronto, ON M5T 1R8, Canada
| | - Kelly Smart
- Dept. of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada; Vivian M. Rakoff PET Centre, Centre for Addiction & Mental Health, Toronto, ON M5T 1R8, Canada
| | - Daniel Tatone
- Dept. of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jerry Warsh
- Vivian M. Rakoff PET Centre, Centre for Addiction & Mental Health, Toronto, ON M5T 1R8, Canada; Dept. of Psychiatry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Alan A Wilson
- Vivian M. Rakoff PET Centre, Centre for Addiction & Mental Health, Toronto, ON M5T 1R8, Canada
| | - James L Kennedy
- Molecular Brain Sciences Dept., Centre for Addiction & Mental Health, Toronto, ON M5T 1R8, Canada; Dept. of Psychiatry, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
13
|
Saenz J, Khezerlou E, Aggarwal M, Shaikh A, Ganti N, Herborg F, Pan PY. Parkinson's disease gene, Synaptojanin1, dysregulates the surface maintenance of the dopamine transporter. NPJ Parkinsons Dis 2024; 10:148. [PMID: 39117637 PMCID: PMC11310474 DOI: 10.1038/s41531-024-00769-0] [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: 03/06/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
Missense mutations of PARK20/SYNJ1 (synaptojanin1/Synj1) were found in complex forms of familial Parkinsonism. However, the Synj1-regulated molecular and cellular changes associated with dopaminergic dysfunction remain unknown. We now report a fast depletion of evoked dopamine and impaired maintenance of the axonal dopamine transporter (DAT) in the Synj1 haploinsufficient (Synj1+/-) neurons. While Synj1 has been traditionally known to facilitate the endocytosis of synaptic vesicles, we provide in vitro and in vivo evidence demonstrating that Synj1 haploinsufficiency results in an increase of total DAT but a reduction of the surface DAT. Synj1+/- neurons exhibit maladaptive DAT trafficking, which could contribute to the altered DA release. We showed that the loss of surface DAT is associated with the impaired 5'-phosphatase activity and the hyperactive PI(4,5)P2-PKCβ pathway downstream of Synj1 deficiency. Thus, our findings provided important mechanistic insight for Synj1-regulated DAT trafficking integral to dysfunctional DA signaling, which might be relevant to early Parkinsonism.
Collapse
Affiliation(s)
- Jacqueline Saenz
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ, 08854, USA
- Rutgers Graduate School of Biomedical Sciences, Molecular Biosciences Graduate Program, 675 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Elnaz Khezerlou
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Meha Aggarwal
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Amina Shaikh
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Naga Ganti
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Freja Herborg
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen, Denmark
| | - Ping-Yue Pan
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ, 08854, USA.
| |
Collapse
|
14
|
Apuschkin M, Burm HB, Schmidt JH, Skov LJ, Andersen RC, Bowin CF, Støier JF, Jensen KL, Posselt LP, Dmytriyeva O, Sørensen AT, Egerod KL, Holst B, Rickhag M, Schwartz TW, Gether U. An atlas of GPCRs in dopamine neurons: Identification of the free fatty acid receptor 4 as a regulator of food and water intake. Cell Rep 2024; 43:114509. [PMID: 39003735 DOI: 10.1016/j.celrep.2024.114509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 04/03/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Midbrain dopaminergic neurons (DANs) are subject to extensive metabotropic regulation, but the repertoire of G protein-coupled receptors (GPCRs) present in these neurons has not been mapped. Here, we isolate DANs from Dat-eGFP mice to generate a GPCR atlas by unbiased qPCR array expression analysis of 377 GPCRs. Combined with data mining of scRNA-seq databases, we identify multiple receptors in DAN subpopulations with 38 of these receptors representing the majority of transcripts. We identify 41 receptors expressed in midbrain DANs but not in non-DAN midbrain cells, including the free fatty acid receptor 4 (FFAR4). Functional expression of FFAR4 is validated by ex vivo Ca2+ imaging, and in vivo experiments support that FFAR4 negatively regulates food and water intake and bodyweight. In addition to providing a critical framework for understanding metabotropic DAN regulation, our data suggest fatty acid sensing by FFAR4 as a mechanism linking high-energy intake to the dopamine-reward pathway.
Collapse
Affiliation(s)
- Mia Apuschkin
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Hayley B Burm
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jan H Schmidt
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Louise J Skov
- Novo Nordic Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Rita C Andersen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Carl-Fredrik Bowin
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jonatan F Støier
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kathrine L Jensen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Leonie P Posselt
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Oksana Dmytriyeva
- Novo Nordic Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Andreas T Sørensen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kristoffer L Egerod
- Novo Nordic Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Birgitte Holst
- Department of Biomedical Sciences, Laboratory for Molecular Pharmacology and Novo Nordic Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Mattias Rickhag
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; Danish Research Centre for Magnetic Resonance (DRCMR), Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Thue W Schwartz
- Novo Nordic Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ulrik Gether
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
15
|
Hamilton AR, Vishwanath A, Weintraub NC, Cowen SL, Heien ML. Dopamine Release Dynamics in the Nucleus Accumbens Are Modulated by the Timing of Electrical Stimulation Pulses When Applied to the Medial Forebrain Bundle and Medial Prefrontal Cortex. ACS Chem Neurosci 2024; 15:2643-2653. [PMID: 38958080 DOI: 10.1021/acschemneuro.4c00115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024] Open
Abstract
Electrical brain stimulation has been used in vivo and in vitro to investigate neural circuitry. Historically, stimulation parameters such as amplitude, frequency, and pulse width were varied to investigate their effects on neurotransmitter release and behavior. These experiments have traditionally employed fixed-frequency stimulation patterns, but it has previously been found that neurons are more precisely tuned to variable input. Introducing variability into the interpulse interval of stimulation pulses will inform on how dopaminergic release can be modulated by variability in pulse timing. Here, dopaminergic release in rats is monitored in the nucleus accumbens (NAc), a key dopaminergic center which plays a role in learning and motivation, by fast-scan cyclic voltammetry. Dopaminergic release in the NAc could also be modulated by stimulation region due to differences in connectivity. We targeted two regions for stimulation─the medial forebrain bundle (MFB) and the medial prefrontal cortex (mPFC)─due to their involvement in reward processing and projections to the NAc. Our goal is to investigate how variable interpulse interval stimulation patterns delivered to these regions affect the time course of dopamine release in the NAc. We found that stimulating the MFB with these variable stimulation patterns saw a highly responsive, frequency-driven dopaminergic response. In contrast, variable stimulation patterns applied to the mPFC were not as sensitive to the variable frequency changes. This work will help inform on how stimulation patterns can be tuned specifically to the stimulation region to improve the efficiency of electrical stimulation and control dopamine release.
Collapse
Affiliation(s)
- Andrea R Hamilton
- Department of Chemistry & Biochemistry, University of Arizona, 1306 East University Boulevard, Tucson, Arizona 85721, United States
| | - Abhilasha Vishwanath
- Department of Psychology, University of Arizona, 1306 East University Boulevard, Tucson, Arizona 85721, United States
| | - Nathan C Weintraub
- Department of Chemistry & Biochemistry, University of Arizona, 1306 East University Boulevard, Tucson, Arizona 85721, United States
| | - Stephen L Cowen
- Department of Psychology, University of Arizona, 1306 East University Boulevard, Tucson, Arizona 85721, United States
- Evelyn F. McKnight Brain Institute, University of Arizona, 1306 East University Boulevard, Tucson, Arizona 85721, United States
| | - M Leandro Heien
- Department of Chemistry & Biochemistry, University of Arizona, 1306 East University Boulevard, Tucson, Arizona 85721, United States
| |
Collapse
|
16
|
Szabadi E. Three paradoxes related to the mode of action of pramipexole: The path from D2/D3 dopamine receptor stimulation to modification of dopamine-modulated functions. J Psychopharmacol 2024; 38:581-596. [PMID: 39041250 DOI: 10.1177/02698811241261022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Pramipexole, a D2/D3 dopamine receptor agonist, is used to treat the motor symptoms of Parkinson's disease, caused by degeneration of the dopaminergic nigrostriatal pathway. There are three paradoxes associated with its mode of action. Firstly, stimulation of D2/D3 receptors leads to neuronal inhibition, although pramipexole does not inhibit but promotes some dopamine-modulated functions, such as locomotion and reinforcement. Secondly, another dopamine-modulated function, arousal, is not promoted but inhibited by pramipexole, leading to sedation. Thirdly, pramipexole-evoked sedation is associated with an increase in pupil diameter, although sedation is expected to cause pupil constriction. To resolve these paradoxes, the path from stimulation of D2/D3 receptors to the modification of dopamine-modulated functions has been tracked. The functions considered are modulated by midbrain dopaminergic nuclei: locomotion - substantia nigra pars compacta (SNc), reinforcement/motivation - ventral tegmental area (VTA), sympathetic activity (as reflected in pupil function) - VTA; arousal - ventral periaqueductal grey (vPAG), with contributions from VTA and SNc. The application of genetics-based molecular techniques (optogenetics and chemogenetics) has enabled tracing the chains of neurones from the dopaminergic nuclei to their final targets executing the functions. The functional neuronal circuits linked to the D2/D3 receptors in the dorsal and ventral striata, stimulated by inputs from SNc and VTA, respectively, may explain how neuronal inhibition induced by pramipexole is translated into the promotion of locomotion, reinforcement/motivation and sympathetic activity. As the vPAG may increase arousal mainly by stimulating cortical D1 dopamine receptors, pramipexole would stimulate only presynaptic D2/D3 receptors on vPAG neurones, curtailing their activity and leading to sedation.
Collapse
Affiliation(s)
- Elemer Szabadi
- Developmental Psychiatry, University of Nottingham, Nottingham, UK
| |
Collapse
|
17
|
Azargoonjahromi A. Current Findings and Potential Mechanisms of KarXT (Xanomeline-Trospium) in Schizophrenia Treatment. Clin Drug Investig 2024; 44:471-493. [PMID: 38904739 DOI: 10.1007/s40261-024-01377-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 06/22/2024]
Abstract
Standard schizophrenia treatment involves antipsychotic medications that target D2 dopamine receptors. However, these drugs have limitations in addressing all symptoms and can lead to adverse effects such as motor impairments, metabolic effects, sedation, sexual dysfunction, cognitive impairment, and tardive dyskinesia. Recently, KarXT has emerged as a novel drug for schizophrenia. KarXT combines xanomeline, a muscarinic receptor M1 and M4 agonist, with trospium, a nonselective antimuscarinic agent. Of note, xanomeline can readily cross blood-brain barrier (BBB) and, thus, enter into the brain, thereby stimulating muscarinic receptors (M1 and M4). By doing so, xanomeline has been shown to target negative symptoms and potentially improve positive symptoms. Trospium, on the other hand, is not able to cross BBB, thereby not affecting M1 and M4 receptors; instead, it acts as an antimuscarinic agent and, hence, diminishes peripheral activity of muscarinic receptors to minimize side effects probably stemming from xanomeline in other organs. Accordingly, ongoing clinical trials investigating KarXT's efficacy in schizophrenia have demonstrated positive outcomes, including significant improvements in the Positive and Negative Syndrome Scale (PANSS) total score and cognitive function compared with placebo. These findings emphasize the potential of KarXT as a promising treatment for schizophrenia, providing symptom relief while minimizing side effects associated with xanomeline monotherapy. Despite such promising evidence, further research is needed to confirm the efficacy, safety, and tolerability of KarXT in managing schizophrenia. This review article explores the current findings and potential mechanisms of KarXT in the treatment of schizophrenia.
Collapse
Affiliation(s)
- Ali Azargoonjahromi
- Shiraz University of Medical Sciences, Janbazan Blv, 14th Alley, Jahrom, Shiraz, 7417773539, Fars, Iran.
| |
Collapse
|
18
|
Oriol L, Chao M, Kollman GJ, Dowlat DS, Singhal SM, Steinkellner T, Hnasko TS. Ventral tegmental area interneurons revisited: GABA and glutamate projection neurons make local synapses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.597996. [PMID: 38895464 PMCID: PMC11185768 DOI: 10.1101/2024.06.07.597996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The ventral tegmental area (VTA) contains projection neurons that release the neurotransmitters dopamine, GABA, and/or glutamate from distal synapses. VTA also contains GABA neurons that synapse locally on to VTA dopamine neurons, synapses widely credited to a population of so-called VTA interneurons. Interneurons in cortex, striatum, and elsewhere have well-defined morphological features, physiological properties, and molecular markers, but such features have not been clearly described in VTA. Indeed, there is scant evidence that local and distal synapses originate from separate populations of VTA GABA neurons. In this study we tested whether several markers expressed in non-dopamine VTA neurons are selective markers of interneurons, defined as neurons that synapse locally but not distally. Challenging previous assumptions, we found that VTA neurons genetically defined by expression of parvalbumin, somatostatin, neurotensin, or mu-opioid receptor project to known VTA targets including nucleus accumbens, ventral pallidum, lateral habenula, and prefrontal cortex. Moreover, we provide evidence that VTA GABA and glutamate projection neurons make functional inhibitory or excitatory synapses locally within VTA. These findings suggest that local collaterals of VTA projection neurons could mediate functions prior attributed to VTA interneurons. This study underscores the need for a refined understanding of VTA connectivity to explain how heterogeneous VTA circuits mediate diverse functions related to reward, motivation, or addiction.
Collapse
Affiliation(s)
- Lucie Oriol
- Department of Neurosciences, University of California, San Diego, La Jolla, United States
| | - Melody Chao
- Department of Neurosciences, University of California, San Diego, La Jolla, United States
| | - Grace J Kollman
- Department of Neurosciences, University of California, San Diego, La Jolla, United States
| | - Dina S Dowlat
- Department of Neurosciences, University of California, San Diego, La Jolla, United States
| | - Sarthak M Singhal
- Department of Neurosciences, University of California, San Diego, La Jolla, United States
| | - Thomas Steinkellner
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Thomas S Hnasko
- Department of Neurosciences, University of California, San Diego, La Jolla, United States
- Research Service VA San Diego Healthcare System, San Diego, United States
| |
Collapse
|
19
|
Piantadosi SC, Lee MK, Wu M, Huynh H, Avila R, Pizzano C, Zamorano CA, Wu Y, Xavier R, Stanslaski M, Kang J, Thai S, Kim Y, Zhang J, Huang Y, Kozorovitskiy Y, Good CH, Banks AR, Rogers JA, Bruchas MR. An integrated microfluidic and fluorescence platform for probing in vivo neuropharmacology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594203. [PMID: 38798493 PMCID: PMC11118345 DOI: 10.1101/2024.05.14.594203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Neurotechnologies and genetic tools for dissecting neural circuit functions have advanced rapidly over the past decade, although the development of complementary pharmacological method-ologies has comparatively lagged. Understanding the precise pharmacological mechanisms of neuroactive compounds is critical for advancing basic neurobiology and neuropharmacology, as well as for developing more effective treatments for neurological and neuropsychiatric disorders. However, integrating modern tools for assessing neural activity in large-scale neural networks with spatially localized drug delivery remains a major challenge. Here, we present a dual microfluidic-photometry platform that enables simultaneous intracranial drug delivery with neural dynamics monitoring in the rodent brain. The integrated platform combines a wireless, battery-free, miniaturized fluidic microsystem with optical probes, allowing for spatially and temporally specific drug delivery while recording activity-dependent fluorescence using genetically encoded calcium indicators (GECIs), neurotransmitter sensors GRAB NE and GRAB DA , and neuropeptide sensors. We demonstrate the performance this platform for investigating neuropharmacological mechanisms in vivo and characterize its efficacy in probing precise mechanistic actions of neuroactive compounds across several rapidly evolving neuroscience domains.
Collapse
|
20
|
Costa KM, Zhang Z, Zhuo Y, Li G, Li Y, Schoenbaum G. Dopamine and acetylcholine correlations in the nucleus accumbens depend on behavioral task states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592439. [PMID: 38746204 PMCID: PMC11092761 DOI: 10.1101/2024.05.03.592439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Dopamine in the nucleus accumbens ramps up as animals approach desired goals. These ramps have received intense scrutiny because they seem to violate long-held hypotheses on dopamine function. Furthermore, it has been proposed that they are driven by local acetylcholine release, i.e., that they are mechanistically separate from dopamine signals related to reward prediction errors. Here, we tested this hypothesis by simultaneously recording accumbal dopamine and acetylcholine signals in rats executing a task involving motivated approach. Contrary to recent reports, we found that dopamine ramps were not coincidental with changes in acetylcholine. Instead, we found that acetylcholine could be positively, negatively, or uncorrelated with dopamine depending on whether the task phase was determined by a salient cue, reward prediction error, or active approach, respectively. Our results suggest that accumbal dopamine and acetylcholine are largely independent but may combine to engage different postsynaptic mechanisms depending on the behavioral task states.
Collapse
|
21
|
Zhang AQ, Ralph MR, Stinchcombe AR. A mathematical model for the role of dopamine-D2 self-regulation in the production of ultradian rhythms. PLoS Comput Biol 2024; 20:e1012082. [PMID: 38701077 PMCID: PMC11095719 DOI: 10.1371/journal.pcbi.1012082] [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: 11/09/2023] [Revised: 05/15/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024] Open
Abstract
Many self-motivated and goal-directed behaviours display highly flexible, approximately 4 hour ultradian (shorter than a day) oscillations. Despite lacking direct correspondence to physical cycles in the environment, these ultradian rhythms may be involved in optimizing functional interactions with the environment and reflect intrinsic neural dynamics. Current evidence supports a role of mesostriatal dopamine (DA) in the expression and propagation of ultradian rhythmicity, however, the biochemical processes underpinning these oscillations remain to be identified. Here, we use a mathematical model to investigate D2 autoreceptor-dependent DA self-regulation as the source of ultradian behavioural rhythms. DA concentration at the midbrain-striatal synapses is governed through a dual-negative feedback-loop structure, which naturally gives rise to rhythmicity. This model shows the propensity of striatal DA to produce an ultradian oscillation characterized by a flexible period that is highly sensitive to parameter variations. Circadian (approximately 24 hour) regulation consolidates the ultradian oscillations and alters their response to the phase-dependent, rapid-resetting effect of a transient excitatory stimulus. Within a circadian framework, the ultradian rhythm orchestrates behavioural activity and enhances responsiveness to an external stimulus. This suggests a role for the circadian-ultradian timekeeping hierarchy in governing organized behaviour and shaping daily experience through coordinating the motivation to engage in recurring, albeit not highly predictable events, such as social interactions.
Collapse
Affiliation(s)
- An Qi Zhang
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Martin R. Ralph
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
22
|
Harada M, Capdevila LS, Wilhelm M, Burdakov D, Patriarchi T. Stimulation of VTA dopamine inputs to LH upregulates orexin neuronal activity in a DRD2-dependent manner. eLife 2024; 12:RP90158. [PMID: 38567902 PMCID: PMC10990487 DOI: 10.7554/elife.90158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024] Open
Abstract
Dopamine and orexins (hypocretins) play important roles in regulating reward-seeking behaviors. It is known that hypothalamic orexinergic neurons project to dopamine neurons in the ventral tegmental area (VTA), where they can stimulate dopaminergic neuronal activity. Although there are reciprocal connections between dopaminergic and orexinergic systems, whether and how dopamine regulates the activity of orexin neurons is currently not known. Here we implemented an opto-Pavlovian task in which mice learn to associate a sensory cue with optogenetic dopamine neuron stimulation to investigate the relationship between dopamine release and orexin neuron activity in the lateral hypothalamus (LH). We found that dopamine release can be evoked in LH upon optogenetic stimulation of VTA dopamine neurons and is also naturally evoked by cue presentation after opto-Pavlovian learning. Furthermore, orexin neuron activity could also be upregulated by local stimulation of dopaminergic terminals in the LH in a way that is partially dependent on dopamine D2 receptors (DRD2). Our results reveal previously unknown orexinergic coding of reward expectation and unveil an orexin-regulatory axis mediated by local dopamine inputs in the LH.
Collapse
Affiliation(s)
- Masaya Harada
- Institute of Pharmacology and Toxicology, University of ZürichZürichSwitzerland
| | | | - Maria Wilhelm
- Institute of Pharmacology and Toxicology, University of ZürichZürichSwitzerland
| | - Denis Burdakov
- Neuroscience Center Zürich, University and ETH ZürichZürichSwitzerland
- Department of Health Sciences and Technology, ETH ZürichZürichSwitzerland
| | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of ZürichZürichSwitzerland
- Neuroscience Center Zürich, University and ETH ZürichZürichSwitzerland
| |
Collapse
|
23
|
Li YT, Huang YL, Chen JJJ, Hyland BI, Wickens JR. Phasic dopamine signals are reduced in the spontaneously hypertensive rat and increased by methylphenidate. Eur J Neurosci 2024; 59:1567-1584. [PMID: 38314648 DOI: 10.1111/ejn.16269] [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: 09/02/2023] [Revised: 12/27/2023] [Accepted: 01/17/2024] [Indexed: 02/06/2024]
Abstract
The spontaneously hypertensive rat (SHR) is a selectively bred animal strain that is frequently used to model attention-deficit hyperactivity disorder (ADHD) because of certain genetically determined behavioural characteristics. To test the hypothesis that the characteristically altered response to positive reinforcement in SHRs may be due to altered phasic dopamine response to reward, we measured phasic dopamine signals in the SHRs and Sprague Dawley (SD) rats using in vivo fast-scan cyclic voltammetry. The effects of the dopamine reuptake inhibitor, methylphenidate, on these signals were also studied. Phasic dopamine signals during the pairing of a sensory cue with electrical stimulation of midbrain dopamine neurons were significantly smaller in the SHRs than in the SD rats. Over repeated pairings, the dopamine response to the sensory cue increased, whereas the response to the electrical stimulation of dopamine neurons decreased, similarly in both strains. However, the final amplitude of the response to the sensory cue after pairing was significantly smaller in SHRs than in the SD rats. Methylphenidate increased responses to sensory cues to a significantly greater extent in the SHRs than in the SD rats, due largely to differences in the low dose effect. At a higher dose, methylphenidate increased responses to sensory cues and electrical stimulation similarly in SHRs and SD rats. The smaller dopamine responses may explain the reduced salience of reward-predicting cues previously reported in the SHR, whereas the action of methylphenidate on the cue response suggests a potential mechanism for the therapeutic effects of low-dose methylphenidate in ADHD.
Collapse
Affiliation(s)
- Yu-Ting Li
- Neurobiology Research Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
- Taiwan Instrument Research Institute, National Applied Research Laboratories, Hsinchu, Taiwan
| | - Yi-Ling Huang
- Neurobiology Research Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
| | - Jia-Jin Jason Chen
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Brian Ian Hyland
- Department of Physiology, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Jeffery R Wickens
- Neurobiology Research Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
| |
Collapse
|
24
|
Rocha GS, Freire MAM, Paiva KM, Oliveira RF, Morais PLAG, Santos JR, Cavalcanti JRLP. The neurobiological effects of senescence on dopaminergic system: A comprehensive review. J Chem Neuroanat 2024; 137:102415. [PMID: 38521203 DOI: 10.1016/j.jchemneu.2024.102415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/26/2024] [Accepted: 03/15/2024] [Indexed: 03/25/2024]
Abstract
Over time, the body undergoes a natural, multifactorial, and ongoing process named senescence, which induces changes at the molecular, cellular, and micro-anatomical levels in many body systems. The brain, being a highly complex organ, is particularly affected by this process, potentially impairing its numerous functions. The brain relies on chemical messengers known as neurotransmitters to function properly, with dopamine being one of the most crucial. This catecholamine is responsible for a broad range of critical roles in the central nervous system, including movement, learning, cognition, motivation, emotion, reward, hormonal release, memory consolidation, visual performance, sexual drive, modulation of circadian rhythms, and brain development. In the present review, we thoroughly examine the impact of senescence on the dopaminergic system, with a primary focus on the classic delimitations of the dopaminergic nuclei from A8 to A17. We provide in-depth information about their anatomy and function, particularly addressing how senescence affects each of these nuclei.
Collapse
Affiliation(s)
- Gabriel S Rocha
- Behavioral and Evolutionary Neurobiology Laboratory, Federal University of Sergipe (UFS), Itabaiana, Brazil
| | - Marco Aurelio M Freire
- Behavioral and Evolutionary Neurobiology Laboratory, Federal University of Sergipe (UFS), Itabaiana, Brazil
| | - Karina M Paiva
- Laboratory of Experimental Neurology, State University of Rio Grande do Norte (UERN), Mossoró, Brazil
| | - Rodrigo F Oliveira
- Laboratory of Experimental Neurology, State University of Rio Grande do Norte (UERN), Mossoró, Brazil
| | - Paulo Leonardo A G Morais
- Laboratory of Experimental Neurology, State University of Rio Grande do Norte (UERN), Mossoró, Brazil
| | - José Ronaldo Santos
- Behavioral and Evolutionary Neurobiology Laboratory, Federal University of Sergipe (UFS), Itabaiana, Brazil
| | | |
Collapse
|
25
|
Foldi CJ. Taking better advantage of the activity-based anorexia model. Trends Mol Med 2024; 30:330-338. [PMID: 38103992 DOI: 10.1016/j.molmed.2023.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/02/2023] [Accepted: 11/17/2023] [Indexed: 12/19/2023]
Abstract
The lack of specific treatments for anorexia nervosa (AN) is partly driven by an inadequate understanding of the neurobiological drivers of the condition. The activity-based anorexia (ABA) model recapitulates key characteristics of AN in rats and mice, and can be used to understand factors that predispose, maintain, and rescue anorectic behaviour. With the rapidly evolving suite of technologies to manipulate and record neural activity during the development of ABA, we are better placed than ever before to take advantage of this unique biobehavioural model in order to develop and refine novel treatments for AN. This will require a collective effort to bridge research disciplines in order to capitalise on knowledge gains from genetics, neurobiology, metabolism, and cognition.
Collapse
Affiliation(s)
- Claire J Foldi
- Monash University, Department of Physiology, 26 Innovation Walk, Clayton, VIC 3800, Australia; Monash Biomedicine Discovery Institute, 23 Innovation Walk, Clayton, VIC 3800, Australia.
| |
Collapse
|
26
|
Farahbakhsh ZZ, Holleran KM, Sens JP, Fordahl SC, Mauterer MI, López AJ, Cuzon Carlson VC, Kiraly DD, Grant KA, Jones SR, Siciliano CA. Synchrony between midbrain gene transcription and dopamine terminal regulation is modulated by chronic alcohol drinking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.584711. [PMID: 38559169 PMCID: PMC10979957 DOI: 10.1101/2024.03.15.584711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Alcohol use disorder is marked by disrupted behavioral and emotional states which persist into abstinence. The enduring synaptic alterations that remain despite the absence of alcohol are of interest for interventions to prevent relapse. Here, 28 male rhesus macaques underwent over 20 months of alcohol drinking interspersed with three 30-day forced abstinence periods. After the last abstinence period, we paired direct sub-second dopamine monitoring via ex vivo voltammetry in nucleus accumbens slices with RNA-sequencing of the ventral tegmental area. We found persistent augmentation of dopamine transporter function, kappa opioid receptor sensitivity, and dynorphin release - all inhibitory regulators which act to decrease extracellular dopamine. Surprisingly, though transcript expression was not altered, the relationship between gene expression and functional readouts of these encoded proteins was highly dynamic and altered by drinking history. These results outline the long-lasting synaptic impact of alcohol use and suggest that assessment of transcript-function relationships is critical for the rational design of precision therapeutics.
Collapse
|
27
|
Sang Y, Niu C, Xu J, Zhu T, You S, Wang J, Zhang L, Du X, Zhang H. PI4KIIIβ-Mediated Phosphoinositides Metabolism Regulates Function of the VTA Dopaminergic Neurons and Depression-Like Behavior. J Neurosci 2024; 44:e0555232024. [PMID: 38267258 PMCID: PMC10941068 DOI: 10.1523/jneurosci.0555-23.2024] [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: 03/27/2023] [Revised: 12/18/2023] [Accepted: 01/11/2024] [Indexed: 01/26/2024] Open
Abstract
Phosphoinositides, including phosphatidylinositol-4,5-bisphosphate (PIP2), play a crucial role in controlling key cellular functions such as membrane and vesicle trafficking, ion channel, and transporter activity. Phosphatidylinositol 4-kinases (PI4K) are essential enzymes in regulating the turnover of phosphoinositides. However, the functional role of PI4Ks and mediated phosphoinositide metabolism in the central nervous system has not been fully revealed. In this study, we demonstrated that PI4KIIIβ, one of the four members of PI4Ks, is an important regulator of VTA dopaminergic neuronal activity and related depression-like behavior of mice by controlling phosphoinositide turnover. Our findings provide new insights into possible mechanisms and potential drug targets for neuropsychiatric diseases, including depression. Both sexes were studied in basic behavior tests, but only male mice could be used in the social defeat depression model.
Collapse
Affiliation(s)
- Yuqi Sang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric Diseases, Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Chenxu Niu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric Diseases, Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Jiaxi Xu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University Health Science Center, Xi'an, Shanxi 710061, China
| | - Tiantian Zhu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric Diseases, Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Shuangzhu You
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric Diseases, Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Jing Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric Diseases, Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Ludi Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric Diseases, Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Xiaona Du
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric Diseases, Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric Diseases, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Department of Psychiatry, The First Hospital of Hebei Medical University, Mental Health Institute of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
28
|
Saenz J, Khezerlou E, Aggarwal M, Shaikh A, Ganti N, Herborg F, Pan PY. Parkinson's disease gene, Synaptojanin1, dysregulates the surface maintenance of the dopamine transporter. RESEARCH SQUARE 2024:rs.3.rs-4021466. [PMID: 38559229 PMCID: PMC10980101 DOI: 10.21203/rs.3.rs-4021466/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Missense mutations of PARK20/SYNJ1 (synaptojanin1/Synj1) have been linked to complex forms of familial parkinsonism, however, the molecular and cellular changes associated with dopaminergic dysfunction remains unknown. We now report fast depletion of evoked dopamine (DA) and altered maintenance of the axonal dopamine transporter (DAT) in the Synj1+/- neurons. While Synj1 has been traditionally known to facilitate the endocytosis of synaptic vesicles, we demonstrated that axons of cultured Synj1+/- neurons exhibit an increase of total DAT but a reduction of the surface DAT, which could be exacerbated by neuronal activity. We revealed that the loss of surface DAT is specifically associated with the impaired 5'-phosphatase activity of Synj1 and the hyperactive downstream PI(4,5)P2-PKCβ pathway. Thus, our findings provided important mechanistic insight for Synj1-regulated DAT trafficking integral to dysfunctional DA signaling in early parkinsonism.
Collapse
Affiliation(s)
- Jacqueline Saenz
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA
- Rutgers Graduate School of Biomedical Sciences, Molecular Biosciences Graduate Program, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Elnaz Khezerlou
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Meha Aggarwal
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Amina Shaikh
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Naga Ganti
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Freja Herborg
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark
| | - Ping-Yue Pan
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| |
Collapse
|
29
|
Tsuboi D, Nagai T, Yoshimoto J, Kaibuchi K. Neuromodulator regulation and emotions: insights from the crosstalk of cell signaling. Front Mol Neurosci 2024; 17:1376762. [PMID: 38516040 PMCID: PMC10954900 DOI: 10.3389/fnmol.2024.1376762] [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: 01/26/2024] [Accepted: 02/26/2024] [Indexed: 03/23/2024] Open
Abstract
The unraveling of the regulatory mechanisms that govern neuronal excitability is a major challenge for neuroscientists worldwide. Neurotransmitters play a critical role in maintaining the balance between excitatory and inhibitory activity in the brain. The balance controls cognitive functions and emotional responses. Glutamate and γ-aminobutyric acid (GABA) are the primary excitatory and inhibitory neurotransmitters of the brain, respectively. Disruptions in the balance between excitatory and inhibitory transmission are implicated in several psychiatric disorders, including anxiety disorders, depression, and schizophrenia. Neuromodulators such as dopamine and acetylcholine control cognition and emotion by regulating the excitatory/inhibitory balance initiated by glutamate and GABA. Dopamine is closely associated with reward-related behaviors, while acetylcholine plays a role in aversive and attentional behaviors. Although the physiological roles of neuromodulators have been extensively studied neuroanatomically and electrophysiologically, few researchers have explored the interplay between neuronal excitability and cell signaling and the resulting impact on emotion regulation. This review provides an in-depth understanding of "cell signaling crosstalk" in the context of neuronal excitability and emotion regulation. It also anticipates that the next generation of neurochemical analyses, facilitated by integrated phosphorylation studies, will shed more light on this topic.
Collapse
Affiliation(s)
- Daisuke Tsuboi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi, Japan
| | - Taku Nagai
- Division of Behavioral Neuropharmacology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi, Japan
| | - Junichiro Yoshimoto
- Department of Biomedical Data Science, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Kozo Kaibuchi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi, Japan
| |
Collapse
|
30
|
Brimblecombe KR, Connor-Robson N, Bataille CJR, Roberts BM, Gracie C, O'Connor B, Te Water Naude R, Karthik G, Russell AJ, Wade-Martins R, Cragg SJ. Inhibition of striatal dopamine release by the L-type calcium channel inhibitor isradipine co-varies with risk factors for Parkinson's. Eur J Neurosci 2024; 59:1242-1259. [PMID: 37941514 PMCID: PMC11426196 DOI: 10.1111/ejn.16180] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/25/2023] [Accepted: 10/14/2023] [Indexed: 11/10/2023]
Abstract
Ca2+ entry into nigrostriatal dopamine (DA) neurons and axons via L-type voltage-gated Ca2+ channels (LTCCs) contributes, respectively, to pacemaker activity and DA release and has long been thought to contribute to vulnerability to degeneration in Parkinson's disease. LTCC function is greater in DA axons and neurons from substantia nigra pars compacta than from ventral tegmental area, but this is not explained by channel expression level. We tested the hypothesis that LTCC control of DA release is governed rather by local mechanisms, focussing on candidate biological factors known to operate differently between types of DA neurons and/or be associated with their differing vulnerability to parkinsonism, including biological sex, α-synuclein, DA transporters (DATs) and calbindin-D28k (Calb1). We detected evoked DA release ex vivo in mouse striatal slices using fast-scan cyclic voltammetry and assessed LTCC support of DA release by detecting the inhibition of DA release by the LTCC inhibitors isradipine or CP8. Using genetic knockouts or pharmacological manipulations, we identified that striatal LTCC support of DA release depended on multiple intersecting factors, in a regionally and sexually divergent manner. LTCC function was promoted by factors associated with Parkinsonian risk, including male sex, α-synuclein, DAT and a dorsolateral co-ordinate, but limited by factors associated with protection, that is, female sex, glucocerebrosidase activity, Calb1 and ventromedial co-ordinate. Together, these data show that LTCC function in DA axons and isradipine effect are locally governed and suggest they vary in a manner that in turn might impact on, or reflect, the cellular stress that leads to parkinsonian degeneration.
Collapse
Affiliation(s)
- Katherine R Brimblecombe
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Natalie Connor-Robson
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
| | - Carole J R Bataille
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Oxford, UK
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Bradley M Roberts
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
| | - Caitlin Gracie
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Bethan O'Connor
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | | | - Gayathri Karthik
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Angela J Russell
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Oxford, UK
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Richard Wade-Martins
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Stephanie J Cragg
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| |
Collapse
|
31
|
Wallace CW, Holleran KM, Slinkard CY, Centanni SW, Jones SR. Kappa Opioid Receptors Negatively Regulate Real Time Spontaneous Dopamine Signals by Reducing Release and Increasing Uptake. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.05.578840. [PMID: 38370660 PMCID: PMC10871279 DOI: 10.1101/2024.02.05.578840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
The role of the dynorphin/kappa opioid receptor (KOR) system in dopamine (DA) regulation has been extensively investigated. KOR activation reduces extracellular DA concentrations and increases DA transporter (DAT) activity and trafficking to the membrane. To explore KOR influences on real-time DA fluctuations, we used the photosensor dLight1.2 with fiber photometry in the nucleus accumbens (NAc) core of freely moving male and female C57BL/6 mice. First, we established that the rise and fall of spontaneous DA signals were due to DA release and reuptake, respectively. Then mice were systemically administered the KOR agonist U50,488H (U50), with or without pretreatment with the KOR antagonist aticaprant (ATIC). U50 reduced both the amplitude and width of spontaneous signals in males, but only reduced width in females. Further, the slope of the correlation between amplitude and width was increased in both sexes, suggesting that DA uptake rates were increased. U50 also reduced the frequency of signals in both males and females. All effects of KOR activation were stronger in males. Overall, KORs exerted significant inhibitory control over spontaneous DA signaling, acting through at least three mechanisms - inhibiting DA release, promoting DAT-mediated uptake, and reducing the frequency of signals.
Collapse
Affiliation(s)
- Conner W Wallace
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Katherine M Holleran
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Clare Y Slinkard
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Samuel W Centanni
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Sara R Jones
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC
| |
Collapse
|
32
|
Everett T, Ten Eyck TW, Wu CH, Shelowitz AL, Stansbury SM, Firek A, Setlow B, McIntyre JC. Cilia loss on distinct neuron populations differentially alters cocaine-induced locomotion and reward. J Psychopharmacol 2024; 38:200-212. [PMID: 38151883 PMCID: PMC11078551 DOI: 10.1177/02698811231219058] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
BACKGROUND Neuronal primary cilia are being recognized for their role in mediating signaling associated with a variety of neurobehaviors, including responses to drugs of abuse. They function as signaling hubs, enriched with a diverse array of G-protein coupled receptors (GPCRs), including several associated with motivation and drug-related behaviors. However, our understanding of how cilia regulate neuronal function and behavior is still limited. AIMS The objective of the current study was to investigate the contributions of primary cilia on specific neuronal populations to behavioral responses to cocaine. METHODS To test the consequences of cilia loss on cocaine-induced locomotion and reward-related behavior, we selectively ablated cilia from dopaminergic or GAD2-GABAergic neurons in mice. RESULTS Cilia ablation on either population of neurons failed to significantly alter acute locomotor responses to cocaine at a range of doses. With repeated administration, mice lacking cilia on GAD2-GABAergic neurons showed no difference in locomotor sensitization to cocaine compared to wild-type (WT) littermates, whereas mice lacking cilia on dopaminergic neurons exhibited reduced locomotor sensitization to cocaine at 10 and 30 mg/kg. Mice lacking cilia on GAD2-GABAergic neurons showed no difference in cocaine conditioned place preference (CPP), whereas mice lacking cilia on dopaminergic neurons exhibited reduced CPP compared to WT littermates. CONCLUSIONS Combined with previous findings using amphetamine, our results show that behavioral effects of cilia ablation are cell- and drug type-specific, and that neuronal cilia contribute to modulation of both the locomotor-inducing and rewarding properties of cocaine.
Collapse
Affiliation(s)
- Thomas Everett
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Tyler W. Ten Eyck
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Chang-Hung Wu
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | | | - Sofia M. Stansbury
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Alexandra Firek
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Barry Setlow
- Department of Psychiatry, University of Florida, Gainesville, FL 32610
- Center for Addiction Research and Education, University of Florida, Gainesville, FL 32610
| | - Jeremy C. McIntyre
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
- Center for Addiction Research and Education, University of Florida, Gainesville, FL 32610
| |
Collapse
|
33
|
Ike KGO, Lamers SJC, Kaim S, de Boer SF, Buwalda B, Billeter JC, Kas MJH. The human neuropsychiatric risk gene Drd2 is necessary for social functioning across evolutionary distant species. Mol Psychiatry 2024; 29:518-528. [PMID: 38114631 PMCID: PMC11116113 DOI: 10.1038/s41380-023-02345-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 12/21/2023]
Abstract
The Drd2 gene, encoding the dopamine D2 receptor (D2R), was recently indicated as a potential target in the etiology of lowered sociability (i.e., social withdrawal), a symptom of several neuropsychiatric disorders such as Schizophrenia and Major Depression. Many animal species show social withdrawal in response to stimuli, including the vinegar fly Drosophila melanogaster and mice, which also share most human disease-related genes. Here we will test for causality between Drd2 and sociability and for its evolutionary conserved function in these two distant species, as well as assess its mechanism as a potential therapeutic target. During behavioral observations in groups of freely interacting D. melanogaster, Drd2 homologue mutant showed decreased social interactions and locomotor activity. After confirming Drd2's social effects in flies, conditional transgenic mice lacking Drd2 in dopaminergic cells (autoreceptor KO) or in serotonergic cells (heteroreceptor KO) were studied in semi-natural environments, where they could freely interact. Autoreceptor KOs showed increased sociability, but reduced activity, while no overall effect of Drd2 deletion was observed in heteroreceptor KOs. To determine acute effects of D2R signaling on sociability, we also showed that a direct intervention with the D2R agonist Sumanirole decreased sociability in wild type mice, while the antagonist showed no effects. Using a computational ethological approach, this study demonstrates that Drd2 regulates sociability across evolutionary distant species, and that activation of the mammalian D2R autoreceptor, in particular, is necessary for social functioning.
Collapse
Affiliation(s)
- Kevin G O Ike
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Sanne J C Lamers
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Soumya Kaim
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Sietse F de Boer
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Bauke Buwalda
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Jean-Christophe Billeter
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Martien J H Kas
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
34
|
Mayer FP, Stewart A, Blakely RD. Leaky lessons learned: Efflux prone dopamine transporter variant reveals sex and circuit specific contributions of D2 receptor signalling to neuropsychiatric disease. Basic Clin Pharmacol Toxicol 2024; 134:206-218. [PMID: 37987120 DOI: 10.1111/bcpt.13964] [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: 07/07/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
Aberrant dopamine (DA) signalling has been implicated in various neuropsychiatric disorders, including attention-deficit/hyperactivity disorder (ADHD), autism spectrum disorder (ASD), schizophrenia, bipolar disorder (BPD) and addiction. The availability of extracellular DA is sculpted by the exocytotic release of vesicular DA and subsequent transporter-mediated clearance, rendering the presynaptic DA transporter (DAT) a crucial regulator of DA neurotransmission. D2-type DA autoreceptors (D2ARs) regulate multiple aspects of DA homeostasis, including (i) DA synthesis, (ii) vesicular release, (iii) DA neuron firing and (iv) the surface expression of DAT and DAT-mediated DA clearance. The DAT Val559 variant, identified in boys with ADHD or ASD, as well as in a girl with BPD, supports anomalous DA efflux (ADE), which we have shown drives tonic activation of D2ARs. Through ex vivo and in vivo studies of the DAT Val559 variant using transgenic knock-in mice, we have uncovered a circuit and sex-specific capacity of D2ARs to regulate DAT, which consequently disrupts DA signalling and behaviour differently in males and females. Our studies reveal the ability of the construct-valid DAT Val559 model to elucidate endogenous mechanisms that support DA signalling, findings that may be of translational and/or therapeutic importance.
Collapse
Affiliation(s)
- Felix P Mayer
- Department of Biomedical Science, Florida Atlantic University, Jupiter, Florida, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida, USA
| | - Adele Stewart
- Department of Biomedical Science, Florida Atlantic University, Jupiter, Florida, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida, USA
| | - Randy D Blakely
- Department of Biomedical Science, Florida Atlantic University, Jupiter, Florida, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida, USA
| |
Collapse
|
35
|
Urueña-Méndez G, Arrondeau C, Bellés L, Ginovart N. Decoupling Dopamine Synthesis from Impulsive Action, Risk-Related Decision-Making, and Propensity to Cocaine Intake: A Longitudinal [ 18F]-FDOPA PET Study in Roman High- and Low-Avoidance Rats. eNeuro 2024; 11:ENEURO.0492-23.2023. [PMID: 38253584 PMCID: PMC10867553 DOI: 10.1523/eneuro.0492-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/20/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Impulsive action and risk-related decision-making (RDM) are two facets of impulsivity linked to a hyperdopaminergic release in the striatum and an increased propensity to cocaine intake. We previously showed that with repeated cocaine exposure, this initial hyperdopaminergic release is blunted in impulsive animals, potentially signaling drug-induced tolerance. Whether such dopaminergic dynamics involve changes in dopamine (DA) synthesis as a function of impulsivity is currently unknown. Here, we investigated the predictive value of DA synthesis for impulsive action, RDM, and the propensity to take cocaine in a rat model of vulnerability to cocaine abuse. Additionally, we assessed the effects of cocaine intake on these variables. Rats were tested sequentially in the rat Gambling Task (rGT) and were scanned with positron emission tomography and [18F]-FDOPA to respectively assess both impulsivity facets and striatal DA synthesis before and after cocaine self-administration (SA). Our results revealed that baseline striatal levels of DA synthesis did not significantly predict impulsive action, RDM, or a greater propensity to cocaine SA in impulsive animals. Besides, we showed that impulsive action, but not RDM, predicted higher rates of cocaine taking. However, chronic cocaine exposure had no impact on DA synthesis, nor affected impulsive action and RDM. These findings indicate that the hyper-responsive DA system associated with impulsivity and a propensity for cocaine consumption, along with the reduction in this hyper-responsive DA state in impulsive animals with a history of cocaine use, might not be mediated by dynamic changes in DA synthesis.
Collapse
Affiliation(s)
- Ginna Urueña-Méndez
- Departments of Psychiatry, Faculty of Medicine, University of Geneva, Geneva CH1206, Switzerland
- Basic Neuroscience, Faculty of Medicine, University of Geneva, Geneva CH1206, Switzerland
| | - Chloé Arrondeau
- Departments of Psychiatry, Faculty of Medicine, University of Geneva, Geneva CH1206, Switzerland
- Basic Neuroscience, Faculty of Medicine, University of Geneva, Geneva CH1206, Switzerland
| | - Lidia Bellés
- Departments of Psychiatry, Faculty of Medicine, University of Geneva, Geneva CH1206, Switzerland
- Basic Neuroscience, Faculty of Medicine, University of Geneva, Geneva CH1206, Switzerland
| | - Nathalie Ginovart
- Departments of Psychiatry, Faculty of Medicine, University of Geneva, Geneva CH1206, Switzerland
- Basic Neuroscience, Faculty of Medicine, University of Geneva, Geneva CH1206, Switzerland
| |
Collapse
|
36
|
Xue Z, Zhang Y, Zhao R, Liu X, Grützmann K, Klink B, Zhang X, Wang S, Zhao W, Sun Y, Han M, Wang X, Hu Y, Liu X, Yang N, Qiu C, Li W, Huang B, Li X, Bjerkvig R, Wang J, Zhou W. The dopamine receptor D1 inhibitor, SKF83566, suppresses GBM stemness and invasion through the DRD1-c-Myc-UHRF1 interactions. J Exp Clin Cancer Res 2024; 43:25. [PMID: 38246990 PMCID: PMC10801958 DOI: 10.1186/s13046-024-02947-7] [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] [Accepted: 01/06/2024] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Extensive local invasion of glioblastoma (GBM) cells within the central nervous system (CNS) is one factor that severely limits current treatments. The aim of this study was to uncover genes involved in the invasion process, which could also serve as therapeutic targets. For the isolation of invasive GBM cells from non-invasive cells, we used a three-dimensional organotypic co-culture system where glioma stem cell (GSC) spheres were confronted with brain organoids (BOs). Using ultra-low input RNA sequencing (ui-RNA Seq), an invasive gene signature was obtained that was exploited in a therapeutic context. METHODS GFP-labeled tumor cells were sorted from invasive and non-invasive regions within co-cultures. Ui-RNA sequencing analysis was performed to find a gene cluster up-regulated in the invasive compartment. This gene cluster was further analyzed using the Connectivity MAP (CMap) database. This led to the identification of SKF83566, an antagonist of the D1 dopamine receptor (DRD1), as a candidate therapeutic molecule. Knockdown and overexpression experiments were performed to find molecular pathways responsible for the therapeutic effects of SKF83566. Finally, the effects of SKF83566 were validated in orthotopic xenograft models in vivo. RESULTS Ui-RNA seq analysis of three GSC cell models (P3, BG5 and BG7) yielded a set of 27 differentially expressed genes between invasive and non-invasive cells. Using CMap analysis, SKF83566 was identified as a selective inhibitor targeting both DRD1 and DRD5. In vitro studies demonstrated that SKF83566 inhibited tumor cell proliferation, GSC sphere formation, and invasion. RNA sequencing analysis of SKF83566-treated P3, BG5, BG7, and control cell populations yielded a total of 32 differentially expressed genes, that were predicted to be regulated by c-Myc. Of these, the UHRF1 gene emerged as the most downregulated gene following treatment, and ChIP experiments revealed that c-Myc binds to its promoter region. Finally, SKF83566, or stable DRD1 knockdown, inhibited the growth of orthotopic GSC (BG5) derived xenografts in nude mice. CONCLUSIONS DRD1 contributes to GBM invasion and progression by regulating c-Myc entry into the nucleus that affects the transcription of the UHRF1 gene. SKF83566 inhibits the transmembrane protein DRD1, and as such represents a candidate small therapeutic molecule for GBMs.
Collapse
Affiliation(s)
- Zhiyi Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Yan Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Ruiqi Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Xiaofei Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Konrad Grützmann
- Core Unit for Molecular Tumour Diagnostics (CMTD), National Center for Tumour Diseases (NCT) Dresden, Dresden, Germany
- Institute for Medical Informatics and Biometry, Medical Faculty, TU Dresden, Dresden, Germany
| | - Barbara Klink
- Department of Genetics, Laboratoire National de Santé, Dudelange, Luxembourg
| | - Xun Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Shuai Wang
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Wenbo Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Yanfei Sun
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Mingzhi Han
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Xu Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yaotian Hu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Xuemeng Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Ning Yang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Chen Qiu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenjie Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Rolf Bjerkvig
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, Bergen, 5009, Norway
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, Bergen, 5009, Norway.
| | - Wenjing Zhou
- Department of Blood Transfusion, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
37
|
Kolacheva A, Pavlova E, Bannikova A, Bogdanov V, Ugrumov M. Initial Molecular Mechanisms of the Pathogenesis of Parkinson's Disease in a Mouse Neurotoxic Model of the Earliest Preclinical Stage of This Disease. Int J Mol Sci 2024; 25:1354. [PMID: 38279354 PMCID: PMC10816442 DOI: 10.3390/ijms25021354] [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: 12/20/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Studying the initial molecular mechanisms of the pathogenesis of Parkinson's disease (PD), primarily in the nigrostriatal dopaminergic system, is one of the priorities in neurology. Of particular interest is elucidating these mechanisms in the preclinical stage of PD, which lasts decades before diagnosis and is therefore not available for study in patients. Therefore, our main goal was to study the initial molecular mechanisms of the pathogenesis of PD in the striatum, the key center for dopamine regulation in motor function, in a mouse model of the earliest preclinical stage of PD, from 1 to 24 h after the administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). It was shown that the content of tyrosine hydroxylase (TH), the first enzyme in dopamine synthesis, does not change within 6 h after the administration of MPTP, but decreases after 24 h. In turn, TH activity increases after 1 h, decreases after 3 h, remains at the control level after 6 h, and decreases 24 h after the administration of MPTP. The concentration of dopamine in the striatum gradually decreases after MPTP administration, despite a decrease in its degradation. The identified initial molecular mechanisms of PD pathogenesis are considered as potential targets for the development of preventive neuroprotective treatment.
Collapse
Affiliation(s)
| | | | | | | | - Michael Ugrumov
- Laboratory of Neural and Neuroendocrine Regulations, Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, 119334 Moscow, Russia; (A.K.); (E.P.); (A.B.); (V.B.)
| |
Collapse
|
38
|
Patterson JR, Kochmanski J, Stoll AC, Kubik M, Kemp CJ, Duffy MF, Thompson K, Howe JW, Cole-Strauss A, Kuhn NC, Miller KM, Nelson S, Onyekpe CU, Beck JS, Counts SE, Bernstein AI, Steece-Collier K, Luk KC, Sortwell CE. Transcriptomic profiling of early synucleinopathy in rats induced with preformed fibrils. NPJ Parkinsons Dis 2024; 10:7. [PMID: 38172128 PMCID: PMC10764951 DOI: 10.1038/s41531-023-00620-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Examination of early phases of synucleinopathy when inclusions are present, but long before neurodegeneration occurs, is critical to both understanding disease progression and the development of disease modifying therapies. The rat alpha-synuclein (α-syn) preformed fibril (PFF) model induces synchronized synucleinopathy that recapitulates the pathological features of Parkinson's disease (PD) and can be used to study synucleinopathy progression. In this model, phosphorylated α-syn (pSyn) inclusion-containing neurons and reactive microglia (major histocompatibility complex-II immunoreactive) peak in the substantia nigra pars compacta (SNpc) months before appreciable neurodegeneration. However, it remains unclear which specific genes are driving these phenotypic changes. To identify transcriptional changes associated with early synucleinopathy, we used laser capture microdissection of the SNpc paired with RNA sequencing (RNASeq). Precision collection of the SNpc allowed for the assessment of differential transcript expression in the nigral dopamine neurons and proximal glia. Transcripts upregulated in early synucleinopathy were mainly associated with an immune response, whereas transcripts downregulated were associated with neurotransmission and the dopamine pathway. A subset of 29 transcripts associated with neurotransmission/vesicular release and the dopamine pathway were verified in a separate cohort of males and females to confirm reproducibility. Within this subset, fluorescent in situ hybridization (FISH) was used to localize decreases in the Syt1 and Slc6a3 transcripts to pSyn inclusion-containing neurons. Identification of transcriptional changes in early synucleinopathy provides insight into the molecular mechanisms driving neurodegeneration.
Collapse
Affiliation(s)
- Joseph R Patterson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA.
- Neuroscience Program, Michigan State University, East Lansing, MI, USA.
| | - Joseph Kochmanski
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Anna C Stoll
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Michael Kubik
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Christopher J Kemp
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Megan F Duffy
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Kajene Thompson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Jacob W Howe
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Allyson Cole-Strauss
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Nathan C Kuhn
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Kathryn M Miller
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Seth Nelson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Christopher U Onyekpe
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - John S Beck
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Scott E Counts
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Alison I Bernstein
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
- Environmental and Occupational Health Science Institute, Rutgers University, Piscataway, NJ, USA
| | - Kathy Steece-Collier
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Kelvin C Luk
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Caryl E Sortwell
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
39
|
Rasmi Y, Shokati A, Hatamkhani S, Farnamian Y, Naderi R, Jalali L. Assessment of the relationship between the dopaminergic pathway and severe acute respiratory syndrome coronavirus 2 infection, with related neuropathological features, and potential therapeutic approaches in COVID-19 infection. Rev Med Virol 2024; 34:e2506. [PMID: 38282395 DOI: 10.1002/rmv.2506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 07/06/2023] [Accepted: 12/17/2023] [Indexed: 01/30/2024]
Abstract
Dopamine is a known catecholamine neurotransmitter involved in several physiological processes, including motor control, motivation, reward, cognition, and immune function. Dopamine receptors are widely distributed throughout the nervous system and in immune cells. Several viruses, including human immunodeficiency virus and Japanese encephalitis virus, can use dopaminergic receptors to replicate in the nervous system and are involved in viral neuropathogenesis. In addition, studies suggest that dopaminergic receptors may play a role in the progression and pathogenesis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. When SARS-CoV-2 binds to angiotensin-converting enzyme 2 receptors on the surface of neuronal cells, the spike protein of the virus can bind to dopaminergic receptors on neighbouring cells to accelerate its life cycle and exacerbate neurological symptoms. In addition, recent research has shown that dopamine is an important regulator of the immune-neuroendocrine system. Most immune cells express dopamine receptors and other dopamine-related proteins, indicating the importance of dopaminergic immune regulation. The increase in dopamine concentration during SARS-CoV2 infection may reduce immunity (innate and adaptive) that promotes viral spread, which could lead to neuronal damage. In addition, dopaminergic signalling in the nervous system may be affected by SARS-CoV-2 infection. COVID -19 can cause various neurological symptoms as it interacts with the immune system. One possible treatment strategy for COVID -19 patients could be the use of dopamine antagonists. To fully understand how to protect the neurological system and immune cells from the virus, we need to study the pathophysiology of the dopamine system in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Yousef Rasmi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ameneh Shokati
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Shima Hatamkhani
- Experimental and Applied Pharmaceutical Sciences Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Pharmacy, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Yeganeh Farnamian
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Roya Naderi
- Nephrology and Kidney Transplant Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Physiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ladan Jalali
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
40
|
Wojtas A, Gołembiowska K. Molecular and Medical Aspects of Psychedelics. Int J Mol Sci 2023; 25:241. [PMID: 38203411 PMCID: PMC10778977 DOI: 10.3390/ijms25010241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/17/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Psychedelics belong to the oldest psychoactive drugs. They arouse recent interest due to their therapeutic applications in the treatment of major depressive disorder, substance use disorder, end-of-life anxiety,= and anxiety symptoms, and obsessive-compulsive disorder. In this review, the current state of preclinical research on the mechanism of action, neurotoxicity, and behavioral impact of psychedelics is summarized. The effect of selective 5-HT2A receptor agonists, 25I- and 25B-NBOMe, after acute and repeated administration is characterized and compared with the effects of a less selective drug, psilocybin. The data show a significant effect of NBOMes on glutamatergic, dopaminergic, serotonergic, and cholinergic neurotransmission in the frontal cortex, striatum, and nucleus accumbens. The increases in extracellular levels of neurotransmitters were not dose-dependent, which most likely resulted from the stimulation of the 5-HT2A receptor and subsequent activation of the 5-HT2C receptors. This effect was also observed in the wet dog shake test and locomotor activity. Chronic administration of NBOMes elicited rapid development of tolerance, genotoxicity, and activation of microglia. Acute treatment with psilocybin affected monoaminergic and aminoacidic neurotransmitters in the frontal cortex, nucleus accumbens, and hippocampus but not in the amygdala. Psilocybin exhibited anxiolytic properties resulting from intensification of GABAergic neurotransmission. The data indicate that NBOMes as selective 5-HT2A agonists exert a significant effect on neurotransmission and behavior of rats while also inducing oxidative DNA damage. In contrast to NBOMes, the effects induced by psilocybin suggest a broader therapeutic index of this drug.
Collapse
Affiliation(s)
| | - Krystyna Gołembiowska
- Unit II, Department of Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland;
| |
Collapse
|
41
|
Qi-Lytle X, Sayers S, Wagner EJ. Current Review of the Function and Regulation of Tuberoinfundibular Dopamine Neurons. Int J Mol Sci 2023; 25:110. [PMID: 38203281 PMCID: PMC10778701 DOI: 10.3390/ijms25010110] [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: 10/30/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
Tuberoinfundibular dopamine (TIDA) neurons have cell bodies located in the arcuate nucleus of the mediobasal hypothalamus. They project to the external zone of the median eminence, and the dopamine (DA) released there is carried by the hypophysial portal vasculature to the anterior pituitary. The DA then activates D2 receptors to inhibit prolactin (PRL) secretion from lactotrophs. The TIDA neuronal population is the principal regulatory factor controlling PRL secretion. The neuroendocrine role subserved by TIDA neurons sets them apart from other dopaminergic populations like the nigrostriatal and mesolimbic DA neurons. TIDA neurons exhibit intrinsic oscillatory fluctuations in their membrane potential that give rise to phasic firing and bursting activity. TIDA neuronal activity is sexually differentiated and modulated by gonadal hormones and PRL, as well as an array of small molecule and peptide neurotransmitters. This review covers these characteristics.
Collapse
Affiliation(s)
- Xiaojun Qi-Lytle
- Department of Medical Education, Geisinger Commonwealth School of Medicine, 525 Pine St., Scranton, PA 18509, USA;
| | - Sarah Sayers
- Department of Basic Medical Science, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second St., Pomona, CA 91766, USA;
| | - Edward J. Wagner
- Department of Basic Medical Science, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second St., Pomona, CA 91766, USA;
| |
Collapse
|
42
|
Zald DH. The influence of dopamine autoreceptors on temperament and addiction risk. Neurosci Biobehav Rev 2023; 155:105456. [PMID: 37926241 PMCID: PMC11330662 DOI: 10.1016/j.neubiorev.2023.105456] [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: 07/31/2023] [Revised: 10/22/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
As a major regulator of dopamine (DA), DA autoreceptors (DAARs) exert substantial influence over DA-mediated behaviors. This paper reviews the physiological and behavioral impact of DAARs. Individual differences in DAAR functioning influences temperamental traits such as novelty responsivity and impulsivity, both of which are associated with vulnerability to addictive behavior in animal models and a broad array of externalizing behaviors in humans. DAARs additionally impact the response to psychostimulants and other drugs of abuse. Human PET studies of D2-like receptors in the midbrain provide evidence for parallels to the animal literature. These data lead to the proposal that weak DAAR regulation is a risk factor for addiction and externalizing problems. The review highlights the potential to build translational models of the functional role of DAARs in behavior. It also draws attention to key limitations in the current literature that would need to be addressed to further advance a weak DAAR regulation model of addiction and externalizing risk.
Collapse
Affiliation(s)
- David H Zald
- Center for Advanced Human Brain Imaging and Department of Psychiatry, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
43
|
Kearney PJ, Zhang Y, Tan Y, Kahuno E, Conklin TL, Fagan RR, Pavchinskiy RG, Shafer SA, Yue Z, Melikian HE. Rit2 silencing in dopamine neurons drives a Parkinsonian phenotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.26.538430. [PMID: 37162843 PMCID: PMC10168302 DOI: 10.1101/2023.04.26.538430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disease and arises from dopamine (DA) neuron death selectively in the substantia nigra pars compacta (SNc). Rit2 is a reported PD risk allele, and recent single cell transcriptomic studies identified a major RIT2 cluster in PD DA neurons, potentially linking Rit2 expression loss to a PD patient cohort. However, it is still unknown whether Rit2 loss itself is causative for PD or PD-like symptoms. Here we report that conditional Rit2 silencing in mouse DA neurons drove motor dysfunction that occurred earlier in males than females and was rescued at early stages by either inhibiting the DA transporter (DAT) or with L-DOPA treatment. Motor dysfunction was accompanied by decreased DA release, striatal DA content, phenotypic DAergic markers, DA neurons, and DAergic terminals, with increased pSer129-alpha synuclein and pSer935-LRRK2 expression. These results provide the first evidence that Rit2 loss is causal for SNc cell death and a PD-like phenotype, and reveal key sex-specific differences in the response to Rit2 loss.
Collapse
|
44
|
Burkert N, Roy S, Häusler M, Wuttke D, Müller S, Wiemer J, Hollmann H, Oldrati M, Ramirez-Franco J, Benkert J, Fauler M, Duda J, Goaillard JM, Pötschke C, Münchmeyer M, Parlato R, Liss B. Deep learning-based image analysis identifies a DAT-negative subpopulation of dopaminergic neurons in the lateral Substantia nigra. Commun Biol 2023; 6:1146. [PMID: 37950046 PMCID: PMC10638391 DOI: 10.1038/s42003-023-05441-6] [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: 12/08/2022] [Accepted: 10/10/2023] [Indexed: 11/12/2023] Open
Abstract
Here we present a deep learning-based image analysis platform (DLAP), tailored to autonomously quantify cell numbers, and fluorescence signals within cellular compartments, derived from RNAscope or immunohistochemistry. We utilised DLAP to analyse subtypes of tyrosine hydroxylase (TH)-positive dopaminergic midbrain neurons in mouse and human brain-sections. These neurons modulate complex behaviour, and are differentially affected in Parkinson's and other diseases. DLAP allows the analysis of large cell numbers, and facilitates the identification of small cellular subpopulations. Using DLAP, we identified a small subpopulation of TH-positive neurons (~5%), mainly located in the very lateral Substantia nigra (SN), that was immunofluorescence-negative for the plasmalemmal dopamine transporter (DAT), with ~40% smaller cell bodies. These neurons were negative for aldehyde dehydrogenase 1A1, with a lower co-expression rate for dopamine-D2-autoreceptors, but a ~7-fold higher likelihood of calbindin-d28k co-expression (~70%). These results have important implications, as DAT is crucial for dopamine signalling, and is commonly used as a marker for dopaminergic SN neurons.
Collapse
Affiliation(s)
- Nicole Burkert
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Shoumik Roy
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany.
| | - Max Häusler
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | | | - Sonja Müller
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Johanna Wiemer
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Helene Hollmann
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Marvin Oldrati
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Jorge Ramirez-Franco
- UMR_S 1072, Aix Marseille Université, INSERM, Faculté de Médecine Secteur Nord, Marseille, France
- INT, Aix Marseille Université, CNRS, Campus Santé Timone, Marseille, France
| | - Julia Benkert
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Michael Fauler
- Institute of General Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Johanna Duda
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Jean-Marc Goaillard
- UMR_S 1072, Aix Marseille Université, INSERM, Faculté de Médecine Secteur Nord, Marseille, France
- INT, Aix Marseille Université, CNRS, Campus Santé Timone, Marseille, France
| | - Christina Pötschke
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Moritz Münchmeyer
- Wolution GmbH & Co. KG, 82152, Munich, Germany
- Department of Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Rosanna Parlato
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
- Division of Neurodegenerative Disorders, Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, 68167, Mannheim, Germany
| | - Birgit Liss
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany.
- Linacre College & New College, Oxford University, OX1 2JD, Oxford, UK.
| |
Collapse
|
45
|
Fernandes L, Kleene R, Congiu L, Freitag S, Kneussel M, Loers G, Schachner M. CHL1 depletion affects dopamine receptor D2-dependent modulation of mouse behavior. Front Behav Neurosci 2023; 17:1288509. [PMID: 38025382 PMCID: PMC10665519 DOI: 10.3389/fnbeh.2023.1288509] [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: 09/04/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction The dopaminergic system plays a key role in the appropriate functioning of the central nervous system, where it is essential for emotional balance, arousal, reward, and motor control. The cell adhesion molecule close homolog of L1 (CHL1) contributes to dopaminergic system development, and CHL1 and the dopamine receptor D2 (D2R) are associated with mental disorders like schizophrenia, addiction, autism spectrum disorder and depression. Methods Here, we investigated how the interplay between CHL1 and D2R affects the behavior of young adult male and female wild-type (CHL+/+) and CHL1-deficient (CHL1-/-) mice, when D2R agonist quinpirole and antagonist sulpiride are applied. Results Low doses of quinpirole (0.02 mg/kg body weight) induced hypolocomotion of CHL1+/+ and CHL1-/- males and females, but led to a delayed response in CHL1-/- mice. Sulpiride (1 mg/kg body weight) affected locomotion of CHL1-/- females and social interaction of CHL1+/+ females as well as social interactions of CHL1-/- and CHL1+/+ males. Quinpirole increased novelty-seeking behavior of CHL1-/- males compared to CHL1+/+ males. Vehicle-treated CHL1-/- males and females showed enhanced working memory and reduced stress-related behavior. Discussion We propose that CHL1 regulates D2R-dependent functions in vivo. Deficiency of CHL1 leads to abnormal locomotor activity and emotionality, and to sex-dependent behavioral differences.
Collapse
Affiliation(s)
- Luciana Fernandes
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Ludovica Congiu
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Sandra Freitag
- Institut für Molekulare Neurogenetik, Zentrum für Molekulare Neurobiologie Hamburg, ZMNH, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Kneussel
- Institut für Molekulare Neurogenetik, Zentrum für Molekulare Neurobiologie Hamburg, ZMNH, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Melitta Schachner
- Department of Cell Biology and Neuroscience, Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, United States
| |
Collapse
|
46
|
Manza P, Tomasi D, Shokri-Kojori E, Zhang R, Kroll D, Feldman D, McPherson K, Biesecker C, Dennis E, Johnson A, Yuan K, Wang WT, Yonga MV, Wang GJ, Volkow ND. Neural circuit selective for fast but not slow dopamine increases in drug reward. Nat Commun 2023; 14:6408. [PMID: 37938560 PMCID: PMC10632365 DOI: 10.1038/s41467-023-41972-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 09/20/2023] [Indexed: 11/09/2023] Open
Abstract
The faster a drug enters the brain, the greater its addictive potential, yet the brain circuits underlying the rate dependency to drug reward remain unresolved. With simultaneous PET-fMRI we linked dynamics of dopamine signaling, brain activity/connectivity, and self-reported 'high' in 20 adults receiving methylphenidate orally (results in slow delivery) and intravenously (results in fast delivery) (trial NCT03326245). We estimated speed of striatal dopamine increases to oral and IV methylphenidate and then tested where brain activity was associated with slow and fast dopamine dynamics (primary endpoint). We then tested whether these brain circuits were temporally associated with individual 'high' ratings to methylphenidate (secondary endpoint). A corticostriatal circuit comprising the dorsal anterior cingulate cortex and insula and their connections with dorsal caudate was activated by fast (but not slow) dopamine increases and paralleled 'high' ratings. These data provide evidence in humans for a link between dACC/insula activation and fast but not slow dopamine increases and document a critical role of the salience network in drug reward.
Collapse
Affiliation(s)
- Peter Manza
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| | - Dardo Tomasi
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Ehsan Shokri-Kojori
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Rui Zhang
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Danielle Kroll
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Dana Feldman
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Katherine McPherson
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Catherine Biesecker
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Evan Dennis
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Allison Johnson
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Kai Yuan
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710071, PR China
| | - Wen-Tung Wang
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Michele-Vera Yonga
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Gene-Jack Wang
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
47
|
Mann LG, Servant M, Hay KR, Song AK, Trujillo P, Yan B, Kang H, Zald D, Donahue MJ, Logan GD, Claassen DO. The Role of a Dopamine-Dependent Limbic-Motor Network in Sensory Motor Processing in Parkinson Disease. J Cogn Neurosci 2023; 35:1806-1822. [PMID: 37677065 PMCID: PMC10594953 DOI: 10.1162/jocn_a_02048] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Limbic and motor integration is enabled by a mesial temporal to motor cortex network. Parkinson disease (PD) is characterized by a loss of dorsal striatal dopamine but relative preservation of mesolimbic dopamine early in disease, along with changes to motor action control. Here, we studied 47 patients with PD using the Simon conflict task and [18F]fallypride PET imaging. Additionally, a cohort of 16 patients participated in a single-blinded dextroamphetamine (dAMPH) study. Task performance was evaluated using the diffusion model for conflict tasks, which allows for an assessment of interpretable action control processes. First, a voxel-wise examination disclosed a negative relationship, such that longer non-decision time is associated with reduced D2-like binding potential (BPND) in the bilateral putamen, left globus pallidus, and right insula. Second, an ROI analysis revealed a positive relationship, such that shorter non-decision time is associated with reduced D2-like BPND in the amygdala and ventromedial OFC. The difference in non-decision time between off-dAMPH and on-dAMPH trials was positively associated with D2-like BPND in the globus pallidus. These findings support the idea that dysfunction of the traditional striatal-motor loop underlies action control deficits but also suggest that a compensatory parallel limbic-motor loop regulates motor output.
Collapse
Affiliation(s)
- Leah G. Mann
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Mathieu Servant
- Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive, Université de Franche-Comté, 25000 Besançon, France
| | - Kaitlyn R. Hay
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Alexander K. Song
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Paula Trujillo
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Bailu Yan
- Deparment of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Hakmook Kang
- Deparment of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - David Zald
- Department of Psychiatry, Rutgers University, Piscataway, NJ 08854, USA
| | - Manus J. Donahue
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Gordon D. Logan
- Department of Psychology, Vanderbilt University, Nashville, TN 37240, USA
| | - Daniel O. Claassen
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
48
|
Kim HY, Lee J, Kim HJ, Lee BE, Jeong J, Cho EJ, Jang HJ, Shin KJ, Kim MJ, Chae YC, Lee SE, Myung K, Baik JH, Suh PG, Kim JI. PLCγ1 in dopamine neurons critically regulates striatal dopamine release via VMAT2 and synapsin III. Exp Mol Med 2023; 55:2357-2375. [PMID: 37907739 PMCID: PMC10689754 DOI: 10.1038/s12276-023-01104-y] [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: 01/13/2023] [Revised: 08/05/2023] [Accepted: 08/06/2023] [Indexed: 11/02/2023] Open
Abstract
Dopamine neurons are essential for voluntary movement, reward learning, and motivation, and their dysfunction is closely linked to various psychological and neurodegenerative diseases. Hence, understanding the detailed signaling mechanisms that functionally modulate dopamine neurons is crucial for the development of better therapeutic strategies against dopamine-related disorders. Phospholipase Cγ1 (PLCγ1) is a key enzyme in intracellular signaling that regulates diverse neuronal functions in the brain. It was proposed that PLCγ1 is implicated in the development of dopaminergic neurons, while the physiological function of PLCγ1 remains to be determined. In this study, we investigated the physiological role of PLCγ1, one of the key effector enzymes in intracellular signaling, in regulating dopaminergic function in vivo. We found that cell type-specific deletion of PLCγ1 does not adversely affect the development and cellular morphology of midbrain dopamine neurons but does facilitate dopamine release from dopaminergic axon terminals in the striatum. The enhancement of dopamine release was accompanied by increased colocalization of vesicular monoamine transporter 2 (VMAT2) at dopaminergic axon terminals. Notably, dopamine neuron-specific knockout of PLCγ1 also led to heightened expression and colocalization of synapsin III, which controls the trafficking of synaptic vesicles. Furthermore, the knockdown of VMAT2 and synapsin III in dopamine neurons resulted in a significant attenuation of dopamine release, while this attenuation was less severe in PLCγ1 cKO mice. Our findings suggest that PLCγ1 in dopamine neurons could critically modulate dopamine release at axon terminals by directly or indirectly interacting with synaptic machinery, including VMAT2 and synapsin III.
Collapse
Affiliation(s)
- Hye Yun Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Jieun Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Hyun-Jin Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Byeong Eun Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Jaewook Jeong
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Eun Jeong Cho
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Hyun-Jun Jang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, 58245, Republic of Korea
| | - Kyeong Jin Shin
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Min Ji Kim
- Department of Life Sciences, Korea University, Seoul, 02841, Korea
| | - Young Chan Chae
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Seung Eun Lee
- Research Animal Resource Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Ja-Hyun Baik
- Department of Life Sciences, Korea University, Seoul, 02841, Korea
| | - Pann-Ghill Suh
- Korea Brain Research Institute (KBRI), Daegu, 41062, Republic of Korea
| | - Jae-Ick Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| |
Collapse
|
49
|
Spicer MM, Weber MA, Luo Z, Yang J, Narayanan NS, Fisher RA. Regulator of G protein signaling 6 (RGS6) in ventral tegmental area (VTA) dopamine neurons promotes EtOH seeking, behavioral reward and susceptibility to relapse. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563844. [PMID: 37961154 PMCID: PMC10634791 DOI: 10.1101/2023.10.24.563844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Mesolimbic dopamine (DA) transmission is believed to play a critical role in mediating reward responses to drugs of abuse, including alcohol (EtOH). EtOH is the most abused substance worldwide with chronic consumption often leading to the development of dependence and abuse. Unfortunately, the neurobiological mechanisms underlying EtOH-seeking behavior and dependence are not fully understood, and abstinence remains the only effective way to prevent alcohol use disorders (AUDs). Here, we developed novel RGS6 fl/fl ; DAT-iCreER mice to determine the role of RGS6 in VTA DA neurons on EtOH consumption and reward behaviors. We found that RGS6 is expressed in DA neurons in both human and mouse VTA, and that RGS6 loss in mice upregulates DA transporter (DAT) expression in VTA DA neuron synaptic terminals. Remarkably, loss of RGS6 in VTA DA neurons significantly reduced EtOH consumption, preference, and reward in a manner indistinguishable from that seen in RGS6 -/- mice. Strikingly, RGS6 loss from VTA DA neurons before or after EtOH behavioral reward is established significantly reduced (∼50%) re-instatement of reward following extinguishment, demonstrating distinct roles of RGS6 in promoting reward and relapse susceptibility to EtOH. These studies illuminate a critical role of RGS6 in the mesolimbic circuit in promoting EtOH seeking, reward, and reinstatement. We propose that RGS6 functions to promote DA transmission through its function as a negative modulator of GPCR-Gα i/o -DAT signaling in VTA DA neurons. These studies identify RGS6 as a potential therapeutic target for behavioral reward and relapse to EtOH.
Collapse
|
50
|
Vahdat Z, Gambrell O, Singh A. Characterizing the role of autaptic feedback in enhancing precision of neuronal firing times. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.06.561207. [PMID: 37873216 PMCID: PMC10592613 DOI: 10.1101/2023.10.06.561207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
In a chemical synapse, information flow occurs via the release of neurotransmitters from a presynaptic neuron that triggers an Action potential (AP) in the postsynaptic neuron. At its core, this occurs via the postsynaptic membrane potential integrating neurotransmitter-induced synaptic currents, and AP generation occurs when potential reaches a critical threshold. This manuscript investigates feedback implementation via an autapse, where the axon from the postsynaptic neuron forms an inhibitory synapse onto itself. Using a stochastic model of neuronal synaptic transmission, we formulate AP generation as a first-passage time problem and derive expressions for both the mean and noise of AP-firing times. Our analytical results supported by stochastic simulations identify parameter regimes where autaptic feedback transmission enhances the precision of AP firing times consistent with experimental data. These noise attenuating regimes are intuitively based on two orthogonal mechanisms - either expanding the time window to integrate noisy upstream signals; or by linearizing the mean voltage increase over time. Interestingly, we find regimes for noise amplification that specifically occur when the inhibitory synapse has a low probability of release for synaptic vesicles. In summary, this work explores feedback modulation of the stochastic dynamics of autaptic neurotransmission and reveals its function of creating more regular AP firing patterns.
Collapse
Affiliation(s)
- Zahra Vahdat
- Department of Electrical and Computer Engineering, University of Delaware, Newark, DE USA 19716
| | - Oliver Gambrell
- Department of Electrical and Computer Engineering, University of Delaware, Newark, DE USA 19716
| | - Abhyudai Singh
- Department of Electrical and Computer Engineering, Biomedical Engineering, Mathematical Sciences, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE USA 19716
| |
Collapse
|