1
|
Chen C, Masotti M, Shepard N, Promes V, Tombesi G, Arango D, Manzoni C, Greggio E, Hilfiker S, Kozorovitskiy Y, Parisiadou L. LRRK2 mediates haloperidol-induced changes in indirect pathway striatal projection neurons. Mol Psychiatry 2025:10.1038/s41380-025-03030-z. [PMID: 40269187 DOI: 10.1038/s41380-025-03030-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025]
Abstract
Haloperidol is used to manage psychotic symptoms in several neurological disorders through mechanisms that involve antagonism of dopamine D2 receptors that are highly expressed in the striatum. Significant side effects of haloperidol, known as extrapyramidal symptoms, lead to motor deficits similar to those seen in Parkinson's disease and present a major challenge in clinical settings. The underlying molecular mechanisms responsible for these side effects remain poorly understood. Parkinson's disease-associated leucine-rich repeat kinase 2 (LRRK2) has an essential role in striatal physiology and a known link to dopamine D2 receptor signaling. Here, we systematically explore convergent signaling of haloperidol and LRRK2 through pharmacological or genetic inhibition of LRRK2 kinase, as well as knock-in mouse models expressing pathogenic mutant LRRK2 with increased kinase activity. Behavioral assays show that LRRK2 kinase inhibition ameliorates haloperidol-induced motor changes in mice. A combination of electrophysiological and anatomical approaches reveals that LRRK2 kinase inhibition interferes with haloperidol-induced changes, specifically in striatal neurons of the indirect pathway. Proteomic studies and targeted intracellular pathway analyses demonstrate that haloperidol induces a similar pattern of intracellular signaling as increased LRRK2 kinase activity. Our study suggests that LRRK2 kinase plays a key role in striatal dopamine D2 receptor signaling underlying the undesirable motor side effects of haloperidol. This work opens up new therapeutic avenues for dopamine-related disorders, such as psychosis, also furthering our understanding of Parkinson's disease pathophysiology.
Collapse
Affiliation(s)
- Chuyu Chen
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Meghan Masotti
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Nathaniel Shepard
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Vanessa Promes
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Giulia Tombesi
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Daniel Arango
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | | | - Elisa Greggio
- Department of Biology, University of Padova, Padova, Italy
| | - Sabine Hilfiker
- Department of Anesthesiology, Rutgers, New Jersey Medical School, Newark, NJ, USA
| | | | - Loukia Parisiadou
- Department of Pharmacology, Northwestern University, Chicago, IL, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
2
|
Kaplan CA, Poels EMP, van den Heuvel MI, Bijma HH, Bergink V, Rommel AS, Robakis T. Systematic Review: Antipsychotic Medication in Pregnancy and Neurodevelopmental Outcomes in Children. J Am Acad Child Adolesc Psychiatry 2025:S0890-8567(25)00211-4. [PMID: 40274065 DOI: 10.1016/j.jaac.2025.04.008] [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: 09/24/2024] [Revised: 01/10/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025]
Abstract
OBJECTIVE Antipsychotic medications are widely prescribed, including during pregnancy, and pregnant individuals worry about the potential sequelae for the child. While antipsychotics do not seem to be teratogenic, the long-term neurodevelopmental impact of prenatal exposure remains unclear. A systematic review was conducted to determine if intrauterine antipsychotic exposure increases the risk of adverse neurodevelopmental outcomes. METHOD A systematic search was performed in MEDLINE, Cochrane, Embase, and PsycINFO for studies published before September 7th, 2024. We included original studies assessing cognitive, motor, behavioral, social, and psychiatric outcomes in children prenatally exposed to antipsychotics, excluding case reports, reviews, pre-clinical studies, and studies without a control group. Quality and risk of bias were assessed using the Newcastle-Ottawa Scale (NOS). RESULTS Of 1349 studies identified, 56 underwent full-text screening, and 16 were included in the review. The number of exposed participants ranged from 11 to >15,000. In the eight studies assessing motor development, early motor delays were observed but did not persist into later childhood. Neurodevelopmental disorders were assessed in seven studies. Crude estimates showed greater risk in exposed children, but after adjusting for confounders, most studies found no significant risk. The mean NOS score was 7.1. CONCLUSION Transient motor delays may be associated with antipsychotic use during pregnancy, although future studies adjusting for confounding factors should clarify this risk. After adjustment for confounders, the risk of neurodevelopmental disorders in school-age children does not seem to be increased. Studies with longer follow-up time are required to further investigate the risk of neurodevelopmental disorders. STUDY REGISTRATION INFORMATION Neurodevelopmental Consequences of Antenatal Exposure to Antipsychotic Medication: A Systematic Review and Meta-Analysis; https://www.crd.york.ac.uk/PROSPERO/view/CRD42024499352.
Collapse
|
3
|
Ilango MS, Desagani D, Jagadeesan S, Snezhko A, Vatine G, Ben-Yoav H. Membrane Permeability Monitoring to Antipsychotic Olanzapine Using Platinum Black-Modified Electrodes. SENSORS (BASEL, SWITZERLAND) 2025; 25:2266. [PMID: 40218778 PMCID: PMC11991649 DOI: 10.3390/s25072266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025]
Abstract
The blood-brain barrier (BBB) is key to the regular functioning of the central nervous system. The dysfunction of the BBB has been described in various neurological disorders, including schizophrenia. Schizophrenia (SCZ) is a chronic psychiatric disorder described by hallucinations, delusions, and negative symptoms. The Olanzapine (OLZ) drug is an electroactive species, and its levels can be monitored using electrochemical sensors. The detection of OLZ was demonstrated previously by using electrochemical sensors, and this technique can be used to monitor the levels of OLZ in real time. The challenge is to identify the permeability of OLZ through the BBB, so a replica model was designed with the BBB based on a Transwell membrane seeded with endothelial cells. A microfabricated electrode consisting of a 3 mm Au disk was modified with platinum black; this enables higher selectivity of electrochemical signals from OLZ. The dose-response of OLZ was characterized in phosphate buffer saline solution (10 mM, pH 7.4) by adding 20-200 nM (in steps 20) of OLZ stock solution. The observed chronoamperometric electrochemical signals showed an increasing current at 0.45 V vs. Ag/AgCl with an increasing OLZ concentration. The controls for the experiments were performed in phosphate-buffered saline solution (10 mM, pH 7.4). The detection limit was calculated as 9.96 ± 7.35 × 10-6 nM from the calibration curve. The membrane permeability of the OLZ drug tested with five SCZ patients was monitored by studying the TEER measurements and permeability rate constant data.
Collapse
Affiliation(s)
- Murugaiya Sridar Ilango
- Nanobioelectronics Laboratory, Department of Biomedical Engineering, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (M.S.I.); (D.D.)
| | - Dayananda Desagani
- Nanobioelectronics Laboratory, Department of Biomedical Engineering, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (M.S.I.); (D.D.)
| | - Srikanth Jagadeesan
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
| | - Alexander Snezhko
- Nanobioelectronics Laboratory, Department of Biomedical Engineering, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (M.S.I.); (D.D.)
| | - Gad Vatine
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
| | - Hadar Ben-Yoav
- Nanobioelectronics Laboratory, Department of Biomedical Engineering, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (M.S.I.); (D.D.)
| |
Collapse
|
4
|
Lakshminarasimhan K, Buck J, Kellendonk C, Horga G. A corticostriatal learning mechanism linking excess striatal dopamine and auditory hallucinations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643990. [PMID: 40166304 PMCID: PMC11956939 DOI: 10.1101/2025.03.18.643990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Auditory hallucinations are linked to elevated striatal dopamine, but their underlying computational mechanisms have been obscured by regional heterogeneity in striatal dopamine signaling. To address this, we developed a normative circuit model in which corticostriatal plasticity in the ventral striatum is modulated by reward prediction errors to drive reinforcement learning while that in the sensory-dorsal striatum is modulated by sensory prediction errors derived from internal belief to drive self-supervised learning. We then validate the key predictions of this model using dopamine recordings across striatal regions in mice, as well as human behavior in a hybrid learning task. Finally, we find that changes in learning resulting from optogenetic stimulation of the sensory striatum in mice and individual variability in hallucination proneness in humans are best explained by selectively enhancing dopamine levels in the model sensory striatum. These findings identify plasticity mechanisms underlying biased learning of sensory expectations as a biologically plausible link between excess dopamine and hallucinations.
Collapse
Affiliation(s)
- Kaushik Lakshminarasimhan
- Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY, USA
| | - Justin Buck
- Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - Christoph Kellendonk
- Department of Psychiatry, Columbia University, New York, NY, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, USA
| | - Guillermo Horga
- Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
| |
Collapse
|
5
|
Javitt DC. Xanomeline-Trospium Treatment of Cognitive Impairments of Schizophrenia: Hope for Some, or Hope for All? Am J Psychiatry 2025; 182:237-239. [PMID: 40022530 DOI: 10.1176/appi.ajp.20241187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/03/2025]
Affiliation(s)
- Daniel C Javitt
- Department of Psychiatry, Columbia University Medical Center, New York, and Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY
| |
Collapse
|
6
|
Fujimoto A, Elorette C, Fujimoto SH, Fleysher L, Rudebeck PH, Russ BE. Pharmacological Modulation of Dopamine Receptors Reveals Distinct Brain-Wide Networks Associated with Learning and Motivation in Nonhuman Primates. J Neurosci 2025; 45:e1301242024. [PMID: 39730205 PMCID: PMC11800751 DOI: 10.1523/jneurosci.1301-24.2024] [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: 07/08/2024] [Revised: 11/07/2024] [Accepted: 11/25/2024] [Indexed: 12/29/2024] Open
Abstract
The neurotransmitter dopamine (DA) has a multifaceted role in healthy and disordered brains through its action on multiple subtypes of dopaminergic receptors. How the modulation of these receptors influences learning and motivation by altering intrinsic brain-wide networks remains unclear. Here, we performed parallel behavioral and resting-state functional MRI experiments after administration of two different DA receptor antagonists in male and female macaque monkeys. Systemic administration of SCH-23390 (D1 antagonist) slowed probabilistic learning when subjects had to learn new stimulus-reward associations and diminished functional connectivity (FC) in corticocortical and frontostriatal connections. In contrast, haloperidol (D2 antagonist) improved learning and broadly enhanced FC in cortical connections. Further comparisons between the effect of SCH-23390/haloperidol on behavioral and resting-state FC revealed specific cortical and subcortical networks associated with the cognitive and motivational effects of DA manipulation, respectively. Thus, we reveal distinct brain-wide networks that are associated with the dopaminergic control of learning and motivation via DA receptors.
Collapse
Affiliation(s)
- Atsushi Fujimoto
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Lipschultz Center for Cognitive Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Catherine Elorette
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Lipschultz Center for Cognitive Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Satoka H Fujimoto
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Lipschultz Center for Cognitive Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Lazar Fleysher
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Peter H Rudebeck
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Lipschultz Center for Cognitive Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Brian E Russ
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute, Orangeburg, New York 10962
- Department of Psychiatry, New York University at Langone, New York, New York 10016
| |
Collapse
|
7
|
De Risi M, Cusimano L, Bujanda Cundin X, Pizzo M, Gigante Y, Monaco M, Di Eugenio C, De Leonibus E. D1 dopamine receptor antagonists as a new therapeutic strategy to treat autistic-like behaviours in lysosomal storage disorders. Mol Psychiatry 2025:10.1038/s41380-025-02904-6. [PMID: 39865184 DOI: 10.1038/s41380-025-02904-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 12/11/2024] [Accepted: 01/20/2025] [Indexed: 01/28/2025]
Abstract
Lysosomal storage disorders characterized by defective heparan sulfate (HS) degradation, such as Mucopolysaccharidosis type IIIA-D (MPS-IIIA-D), result in neurodegeneration and dementia in children. However, dementia is preceded by severe autistic-like behaviours (ALBs), presenting as hyperactivity, stereotypies, social interaction deficits, and sleep disturbances. The absence of experimental studies on ALBs' mechanisms in MPS-III has led clinicians to adopt symptomatic treatments, such as antipsychotics, which are used for non-genetic neuropsychiatric disorders. However, they have limited efficacy in MPS-III and lead to higher extrapyramidal effects, leaving ALBs in MPS-IIIA as an unmet medical need with a significant burden on patients and their families. Using mouse and cellular models of MPS-IIIA, we have previously shown that ALBs result from increased proliferation of mesencephalic dopamine neurons during embryogenesis. In adulthood, MPS-IIIA mice exhibit an imbalance of dopaminergic receptor subtypes, resulting in striatal overstimulation of the D1 dopamine receptor (D1R)-direct pathway, contrasting with a downregulation of the D2 dopamine receptor (D2R)-indirect pathway. In this study, we aimed to provide an evidence-based pharmacological approach for managing ALBs in MPS-IIIA. We hypothesized that rebalancing dopaminergic receptor signalling with a D1R antagonist, rather than a D2 antagonist, would lead to safe and effective treatment. Neither risperidone nor methylphenidate improves ALBs in the MPS-IIIA mouse model, with the former showing increased cataleptic (extrapyramidal-like) side effects compared to littermate wild-type animals. Methylphenidate, however, showed some beneficial effects on neuroinflammation and later manifesting dementia-like behaviours. In contrast, ecopipam, a D1 antagonist already used in the clinic for other neuropsychiatric disorders, rescues ALBs, cognition, D1 hyperactivity, and does not worsen neurodegenerative signs. These results align with recent evidence highlighting the clinical relevance of D1 antagonists for neuropsychiatric disorders and pave the way for their use in managing psychotic symptoms in neurodegenerative disorders such as dementia with Lewy bodies.
Collapse
Affiliation(s)
- Maria De Risi
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy.
- Institute of Biochemistry and Cell Biology, CNR, Via Ramarini 32, Monterotondo Scalo, 00015, Rome, Italy.
| | - Lorenzo Cusimano
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
| | - Xabier Bujanda Cundin
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
| | - Mariateresa Pizzo
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
| | - Ylenia Gigante
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
| | - Mariagrazia Monaco
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
| | - Chiara Di Eugenio
- Institute of Biochemistry and Cell Biology, CNR, Via Ramarini 32, Monterotondo Scalo, 00015, Rome, Italy
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy.
- Institute of Biochemistry and Cell Biology, CNR, Via Ramarini 32, Monterotondo Scalo, 00015, Rome, Italy.
| |
Collapse
|
8
|
Goto S. Functional pathology of neuroleptic-induced dystonia based on the striatal striosome-matrix dopamine system in humans. J Neurol Neurosurg Psychiatry 2025; 96:177-183. [PMID: 39631787 DOI: 10.1136/jnnp-2024-334545] [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: 06/27/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
Neuroleptic-induced dystonia is a source of great concern in clinical practice because of its iatrogenic nature which can potentially lead to life-threatening conditions. Since all neuroleptics (antipsychotics) share the ability to block the dopamine D2-type receptors (D2Rs) that are highly enriched in the striatum, this drug-induced dystonia is thought to be caused by decreased striatal D2R activity. However, how associations of striatal D2R inactivation with dystonia are formed remains elusive.A growing body of evidence suggests that imbalanced activities between D1R-expressing medium spiny neurons and D2R-expressing medium spiny neurons (D1-MSNs and D2-MSNs) in the striatal striosome-matrix system underlie the pathophysiology of various basal ganglia disorders including dystonia. Given the specificity of the striatal dopamine D1 system in 'humans', this article highlights the striatal striosome hypothesis in causing 'repetitive' and 'stereotyped' motor symptoms which are key clinical features of dystonia. It is suggested that exposure to neuroleptics may reduce striosomal D1-MSN activity and thereby cause dystonia symptoms. This may occur through an increase in the striatal cholinergic activity and the collateral inhibitory action of D2-MSNs onto neighbouring D1-MSNs within the striosome subfields. The article proposes a functional pathology of the striosome-matrix dopamine system for neuroleptic-induced acute dystonia or neuroleptic-withdrawal dystonia. A rationale for the effectiveness of dopaminergic or cholinergic pharmacotherapy is also provided for treating dystonias. This narrative review covers various aspects of the relevant field and provides a detailed discussion of the mechanisms of neuroleptic-induced dystonia.
Collapse
Affiliation(s)
- Satoshi Goto
- Research Organization of Science and Technology, Ritsumeikan University, Kyoto, Japan
| |
Collapse
|
9
|
Bruce RA, Weber M, Bova A, Volkman R, Jacobs C, Sivakumar K, Stutt H, Kim Y, Curtu R, Narayanan K. Complementary cognitive roles for D2-MSNs and D1-MSNs during interval timing. eLife 2025; 13:RP96287. [PMID: 39812105 PMCID: PMC11735027 DOI: 10.7554/elife.96287] [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: 01/16/2025] Open
Abstract
The role of striatal pathways in cognitive processing is unclear. We studied dorsomedial striatal cognitive processing during interval timing, an elementary cognitive task that requires mice to estimate intervals of several seconds and involves working memory for temporal rules as well as attention to the passage of time. We harnessed optogenetic tagging to record from striatal D2-dopamine receptor-expressing medium spiny neurons (D2-MSNs) in the indirect pathway and from D1-dopamine receptor-expressing MSNs (D1-MSNs) in the direct pathway. We found that D2-MSNs and D1-MSNs exhibited distinct dynamics over temporal intervals as quantified by principal component analyses and trial-by-trial generalized linear models. MSN recordings helped construct and constrain a four-parameter drift-diffusion computational model in which MSN ensemble activity represented the accumulation of temporal evidence. This model predicted that disrupting either D2-MSNs or D1-MSNs would increase interval timing response times and alter MSN firing. In line with this prediction, we found that optogenetic inhibition or pharmacological disruption of either D2-MSNs or D1-MSNs increased interval timing response times. Pharmacologically disrupting D2-MSNs or D1-MSNs also changed MSN dynamics and degraded trial-by-trial temporal decoding. Together, our findings demonstrate that D2-MSNs and D1-MSNs had opposing dynamics yet played complementary cognitive roles, implying that striatal direct and indirect pathways work together to shape temporal control of action. These data provide novel insight into basal ganglia cognitive operations beyond movement and have implications for human striatal diseases and therapies targeting striatal pathways.
Collapse
Affiliation(s)
- Robert A Bruce
- Department of Neurology, University of IowaIowa CityUnited States
| | - Matthew Weber
- Department of Neurology, University of IowaIowa CityUnited States
| | - Alexandra Bova
- Department of Neurology, University of IowaIowa CityUnited States
| | - Rachael Volkman
- Department of Neurology, University of IowaIowa CityUnited States
| | - Casey Jacobs
- Department of Neurology, University of IowaIowa CityUnited States
| | - Kartik Sivakumar
- Department of Neurology, University of IowaIowa CityUnited States
| | - Hannah Stutt
- Department of Neurology, University of IowaIowa CityUnited States
| | - Youngcho Kim
- Department of Neurology, University of IowaIowa CityUnited States
| | - Rodica Curtu
- Department of Mathematics, University of IowaIowa CityUnited States
- The Iowa Neuroscience InstituteIowa CityUnited States
| | - Kumar Narayanan
- Department of Neurology, University of IowaIowa CityUnited States
- The Iowa Neuroscience InstituteIowa CityUnited States
| |
Collapse
|
10
|
Varin C, de Kerchove d'Exaerde A. Neuronal encoding of behaviors and instrumental learning in the dorsal striatum. Trends Neurosci 2025; 48:77-91. [PMID: 39632222 DOI: 10.1016/j.tins.2024.11.003] [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: 07/16/2024] [Revised: 10/08/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024]
Abstract
The dorsal striatum is instrumental in regulating motor control and goal-directed behaviors. The classical description of the two output pathways of the dorsal striatum highlights their antagonistic control over actions. However, recent experimental evidence implicates both pathways and their coordinated activities during actions. In this review, we examine the different models proposed for striatal encoding of actions during self-paced behaviors and how they can account for evidence harvested during goal-directed behaviors. We also discuss how the activation of striatal ensembles can be reshaped and reorganized to support the formation of instrumental learning and behavioral flexibility. Future work integrating these considerations may resolve controversies regarding the control of actions by striatal networks.
Collapse
Affiliation(s)
- Christophe Varin
- Université Libre de Bruxelles (ULB), ULB Neuroscience Institute, Neurophysiology Laboratory, Brussels, Belgium.
| | - Alban de Kerchove d'Exaerde
- Université Libre de Bruxelles (ULB), ULB Neuroscience Institute, Neurophysiology Laboratory, Brussels, Belgium.
| |
Collapse
|
11
|
Fujimoto A, Elorette C, Fujimoto SH, Fleysher L, Rudebeck PH, Russ BE. Pharmacological modulation of dopamine receptors reveals distinct brain-wide networks associated with learning and motivation in non-human primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.27.573487. [PMID: 38234858 PMCID: PMC10793459 DOI: 10.1101/2023.12.27.573487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
The neurotransmitter dopamine (DA) has a multifaceted role in healthy and disordered brains through its action on multiple subtypes of dopaminergic receptors. How modulation of these receptors influences learning and motivation by altering intrinsic brain-wide networks remains unclear. Here we performed parallel behavioral and resting-state functional MRI experiments after administration of two different DA receptor antagonists in macaque monkeys. Systemic administration of SCH-23390 (D1 antagonist) slowed probabilistic learning when subjects had to learn new stimulus-reward associations and diminished functional connectivity (FC) in cortico-cortical and fronto-striatal connections. By contrast, haloperidol (D2 antagonist) improved learning and broadly enhanced FC in cortical connections. Further comparisons between the effect of SCH-23390/haloperidol on behavioral and resting-state FC revealed specific cortical and subcortical networks associated with the cognitive and motivational effects of DA manipulation, respectively. Thus, we reveal distinct brain-wide networks that are associated with the dopaminergic control of learning and motivation via DA receptors.
Collapse
Affiliation(s)
- Atsushi Fujimoto
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029
- Lipschultz Center for Cognitive Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Catherine Elorette
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029
- Lipschultz Center for Cognitive Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Satoka H. Fujimoto
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029
- Lipschultz Center for Cognitive Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Lazar Fleysher
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029
| | - Peter H. Rudebeck
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029
- Lipschultz Center for Cognitive Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Brian E. Russ
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY 10962
- Department of Psychiatry, New York University at Langone, One, 8, Park Ave, New York, NY 10016
| |
Collapse
|
12
|
Degel C, Zitelli K, Zapata J, Nassi J, Botta P. Harnessing Miniscope Imaging in Freely Moving Animals to Unveil Migraine Pathophysiology and Validate Novel Therapeutic Strategies. Synapse 2024; 78:e70001. [PMID: 39567365 PMCID: PMC11578939 DOI: 10.1002/syn.70001] [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: 07/17/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/22/2024]
Abstract
Migraine is a debilitating neurological disorder that affects millions worldwide. Elucidating its underlying mechanisms is crucial for developing effective therapeutic interventions. In this editorial, we discuss the potential applications of one-photon miniscopes, which enable minimally invasive, high spatiotemporal resolution fluorescence imaging in freely moving animals. By providing real-time visualization of vascular dynamics and neuronal activity, these cutting-edge techniques can offer unique insights into migraine pathophysiology. We explore the significance of these applications in preclinical research with a case study demonstrating their potential to drive the development of novel therapeutic strategies for effective migraine management.
Collapse
Affiliation(s)
- Caroline Degel
- H. Lundbeck A/S, Neuroscience Research, Circuit BiologyValbyDenmark
| | - Kevin Zitelli
- Inscopix ‐ A Bruker CompanyMountain ViewCaliforniaUSA
| | | | | | - Paolo Botta
- H. Lundbeck A/S, Neuroscience Research, Circuit BiologyValbyDenmark
| |
Collapse
|
13
|
Kielhold ML, Jacobs DS, Torrado-Pacheco A, Lebowitz JJ, Langdon AJ, Williams JT, Zweifel LS, Moghaddam B. Reductions of Grin2a in adolescent dopamine neurons confers aberrant salience and related psychosis phenotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620713. [PMID: 39554173 PMCID: PMC11565768 DOI: 10.1101/2024.10.28.620713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Psychosis is a hallmark of schizophrenia. It typically emerges in late adolescence and is associated with dopamine abnormalities and aberrant salience. Most genes associated with schizophrenia risk involve ubiquitous targets that may not explain delayed emergence of dopaminergic disruptions. This includes GRIN2A, the gene encoding the GluN2A subunit of the NMDA receptor. Both common and rare variants of GRIN2A are considered genetic risk factors for schizophrenia diagnosis. We find that Grin2a knockout in dopamine neurons during adolescence is sufficient to produce a behavioral phenotype that mirrors aspects of psychosis. These include disruptions in effort optimization, salience attribution, and ability to utilize feedback to guide behavior. We also find a selective effect of this manipulation on dopamine release during prediction error signaling. These data provide mechanistic insight into how variants of GRIN2A may lead to the latent presentation of aberrant salience and abnormalities in dopamine dynamics. This etiologically relevant model may aid future discovery of course altering treatments for schizophrenia.
Collapse
|
14
|
Paul SM, Yohn SE, Brannan SK, Neugebauer NM, Breier A. Muscarinic Receptor Activators as Novel Treatments for Schizophrenia. Biol Psychiatry 2024; 96:627-637. [PMID: 38537670 DOI: 10.1016/j.biopsych.2024.03.014] [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: 11/17/2023] [Revised: 03/08/2024] [Accepted: 03/17/2024] [Indexed: 05/26/2024]
Abstract
Achieving optimal treatment outcomes for individuals living with schizophrenia remains challenging, despite 70 years of drug development efforts. Many chemically distinct antipsychotics have been developed over the past 7 decades with improved safety and tolerability but with only slight variation in efficacy. All antipsychotics currently approved for the treatment of schizophrenia act as antagonists or partial agonists at the dopamine D2 receptor. With only a few possible exceptions, antipsychotic drugs have similar and modest efficacy for treating positive symptoms and are relatively ineffective in addressing the negative and cognitive symptoms of the disease. The development of novel treatments focused on targeting muscarinic acetylcholine receptors (mAChRs) has been of interest for more than 25 years following reports that treatment with a dual M1/M4-preferring mAChR agonist resulted in antipsychotic-like effects and procognitive properties in individuals living with Alzheimer's disease and schizophrenia; more recent clinical trials have confirmed these findings. In addition, advances in our understanding of the receptor binding and activation properties of xanomeline at specific mAChRs have the potential to inform future drug design targeting mAChRs.
Collapse
Affiliation(s)
- Steven M Paul
- Karuna Therapeutics, Boston, Massachusetts; Department of Psychiatry and Neurology, Washington University of St. Louis, St. Louis, Missouri.
| | | | | | | | - Alan Breier
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
15
|
Bruce R, Weber MA, Bova A, Volkman R, Jacobs C, Sivakumar K, Kim Y, Curtu R, Narayanan N. Complementary cognitive roles for D2-MSNs and D1-MSNs during interval timing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.25.550569. [PMID: 37546735 PMCID: PMC10402049 DOI: 10.1101/2023.07.25.550569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
The role of striatal pathways in cognitive processing is unclear. We studied dorsomedial striatal cognitive processing during interval timing, an elementary cognitive task that requires mice to estimate intervals of several seconds and involves working memory for temporal rules as well as attention to the passage of time. We harnessed optogenetic tagging to record from striatal D2-dopamine receptor-expressing medium spiny neurons (D2-MSNs) in the indirect pathway and from D1-dopamine receptor-expressing MSNs (D1-MSNs) in the direct pathway. We found that D2-MSNs and D1-MSNs exhibited distinct dynamics over temporal intervals as quantified by principal component analyses and trial-by-trial generalized linear models. MSN recordings helped construct and constrain a four-parameter drift-diffusion computational model. This model predicted that disrupting either D2-MSNs or D1-MSNs would increase interval timing response times and alter MSN firing. In line with this prediction, we found that optogenetic inhibition or pharmacological disruption of either D2-MSNs or D1-MSNs increased interval timing response times. Pharmacologically disrupting D2-MSNs or D1-MSNs also changed MSN dynamics and degraded trial-by-trial temporal decoding. Together, our findings demonstrate that D2-MSNs and D1-MSNs make complementary contributions to interval timing despite opposing dynamics, implying that striatal direct and indirect pathways work together to shape temporal control of action. These data provide novel insight into basal ganglia cognitive operations beyond movement and have implications for human striatal diseases and therapies targeting striatal pathways.
Collapse
|
16
|
Cameron D, Vinh NN, Prapaiwongs P, Perry EA, Walters JTR, Li M, O’Donovan MC, Bray NJ. Genetic Implication of Prenatal GABAergic and Cholinergic Neuron Development in Susceptibility to Schizophrenia. Schizophr Bull 2024; 50:1171-1184. [PMID: 38869145 PMCID: PMC11349020 DOI: 10.1093/schbul/sbae083] [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] [Indexed: 06/14/2024]
Abstract
BACKGROUND The ganglionic eminences (GE) are fetal-specific structures that give rise to gamma-aminobutyric acid (GABA)- and acetylcholine-releasing neurons of the forebrain. Given the evidence for GABAergic, cholinergic, and neurodevelopmental disturbances in schizophrenia, we tested the potential involvement of GE neuron development in mediating genetic risk for the condition. STUDY DESIGN We combined data from a recent large-scale genome-wide association study of schizophrenia with single-cell RNA sequencing data from the human GE to test the enrichment of schizophrenia risk variation in genes with high expression specificity for developing GE cell populations. We additionally performed the single nuclei Assay for Transposase-Accessible Chromatin with Sequencing (snATAC-Seq) to map potential regulatory genomic regions operating in individual cell populations of the human GE, using these to test for enrichment of schizophrenia common genetic variant liability and to functionally annotate non-coding variants-associated with the disorder. STUDY RESULTS Schizophrenia common variant liability was enriched in genes with high expression specificity for developing neuron populations that are predicted to form dopamine D1 and D2 receptor-expressing GABAergic medium spiny neurons of the striatum, cortical somatostatin-positive GABAergic interneurons, calretinin-positive GABAergic neurons, and cholinergic neurons. Consistent with these findings, schizophrenia genetic risk was concentrated in predicted regulatory genomic sequence mapped in developing neuronal populations of the GE. CONCLUSIONS Our study implicates prenatal development of specific populations of GABAergic and cholinergic neurons in later susceptibility to schizophrenia, and provides a map of predicted regulatory genomic elements operating in cells of the GE.
Collapse
Affiliation(s)
- Darren Cameron
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, UK
| | - Ngoc-Nga Vinh
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, UK
| | - Parinda Prapaiwongs
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK
| | - Elizabeth A Perry
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, UK
| | - James T R Walters
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, UK
| | - Meng Li
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, UK
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK
| | - Michael C O’Donovan
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, UK
| | - Nicholas J Bray
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, UK
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK
| |
Collapse
|
17
|
Díaz-Holguín A, Saarinen M, Vo DD, Sturchio A, Branzell N, Cabeza de Vaca I, Hu H, Mitjavila-Domènech N, Lindqvist A, Baranczewski P, Millan MJ, Yang Y, Carlsson J, Svenningsson P. AlphaFold accelerated discovery of psychotropic agonists targeting the trace amine-associated receptor 1. SCIENCE ADVANCES 2024; 10:eadn1524. [PMID: 39110804 PMCID: PMC11305387 DOI: 10.1126/sciadv.adn1524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 06/28/2024] [Indexed: 08/10/2024]
Abstract
Artificial intelligence is revolutionizing protein structure prediction, providing unprecedented opportunities for drug design. To assess the potential impact on ligand discovery, we compared virtual screens using protein structures generated by the AlphaFold machine learning method and traditional homology modeling. More than 16 million compounds were docked to models of the trace amine-associated receptor 1 (TAAR1), a G protein-coupled receptor of unknown structure and target for treating neuropsychiatric disorders. Sets of 30 and 32 highly ranked compounds from the AlphaFold and homology model screens, respectively, were experimentally evaluated. Of these, 25 were TAAR1 agonists with potencies ranging from 12 to 0.03 μM. The AlphaFold screen yielded a more than twofold higher hit rate (60%) than the homology model and discovered the most potent agonists. A TAAR1 agonist with a promising selectivity profile and drug-like properties showed physiological and antipsychotic-like effects in wild-type but not in TAAR1 knockout mice. These results demonstrate that AlphaFold structures can accelerate drug discovery.
Collapse
Affiliation(s)
- Alejandro Díaz-Holguín
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Box 596, SE-751 24 Uppsala, Sweden
| | - Marcus Saarinen
- Neuro Svenningsson, Department of Clinical Neuroscience, Karolinska Institute, SE-171 76 Stockholm, Sweden
| | - Duc Duy Vo
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Box 596, SE-751 24 Uppsala, Sweden
| | - Andrea Sturchio
- Neuro Svenningsson, Department of Clinical Neuroscience, Karolinska Institute, SE-171 76 Stockholm, Sweden
- Department of Neurology, James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, University of Cincinnati, Cincinnati, OH, USA
| | - Niclas Branzell
- Neuro Svenningsson, Department of Clinical Neuroscience, Karolinska Institute, SE-171 76 Stockholm, Sweden
| | - Israel Cabeza de Vaca
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Box 596, SE-751 24 Uppsala, Sweden
| | - Huabin Hu
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Box 596, SE-751 24 Uppsala, Sweden
| | - Núria Mitjavila-Domènech
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Box 596, SE-751 24 Uppsala, Sweden
| | - Annika Lindqvist
- Department of Pharmacy, SciLifeLab Drug Discovery and Development Platform, Uppsala University, Box 580, SE-751 23 Uppsala, Sweden
| | - Pawel Baranczewski
- Department of Pharmacy, SciLifeLab Drug Discovery and Development Platform, Uppsala University, Box 580, SE-751 23 Uppsala, Sweden
| | - Mark J. Millan
- Neuroinflammation Therapeutic Area, Institut de Recherches Servier, Centre de Recherches de Croissy, Paris, France and Institute of Neuroscience and Psychology, College of Medicine, Vet and Life Sciences, Glasgow University, Scotland, Glasgow, UK
| | - Yunting Yang
- Neuro Svenningsson, Department of Clinical Neuroscience, Karolinska Institute, SE-171 76 Stockholm, Sweden
| | - Jens Carlsson
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Box 596, SE-751 24 Uppsala, Sweden
| | - Per Svenningsson
- Neuro Svenningsson, Department of Clinical Neuroscience, Karolinska Institute, SE-171 76 Stockholm, Sweden
| |
Collapse
|
18
|
Bari BA, Gershman SJ. Resource-rational psychopathology. Behav Neurosci 2024; 138:221-234. [PMID: 38753400 PMCID: PMC11423359 DOI: 10.1037/bne0000600] [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: 09/10/2024]
Abstract
Psychopathology is vast and diverse. Across distinct disease states, individuals exhibit symptoms that appear counter to the standard view of rationality (expected utility maximization). We argue that some aspects of psychopathology can be described as resource-rational, reflecting a rational trade-off between reward and cognitive resources. We review work on two theories of this kind: rational inattention, where a capacity limit applies to perceptual channels, and policy compression, where the capacity limit applies to action channels. We show how these theories can parsimoniously explain many forms of psychopathology, including affective, primary psychotic, and neurodevelopmental disorders, as well as many effects of psychoactive medications on these disorders. While there are important disorder-specific differences and the theories are by no means universal, we argue that resource rationality offers a useful new perspective on psychopathology. By emphasizing the role of cognitive resource constraints, this approach offers a more inclusive picture of rationality. Some aspects of psychopathology may reflect rational trade-offs rather than the breakdown of rationality. (PsycInfo Database Record (c) 2024 APA, all rights reserved).
Collapse
Affiliation(s)
- Bilal A. Bari
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114
| | - Samuel J. Gershman
- Department of Psychology and Center for Brain Science, Harvard University, Cambridge, MA 02138
- Center for Brains, Minds, and Machines, MIT, Cambridge, MA 02139
| |
Collapse
|
19
|
Keller GB, Sterzer P. Predictive Processing: A Circuit Approach to Psychosis. Annu Rev Neurosci 2024; 47:85-101. [PMID: 38424472 DOI: 10.1146/annurev-neuro-100223-121214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Predictive processing is a computational framework that aims to explain how the brain processes sensory information by making predictions about the environment and minimizing prediction errors. It can also be used to explain some of the key symptoms of psychotic disorders such as schizophrenia. In recent years, substantial advances have been made in our understanding of the neuronal circuitry that underlies predictive processing in cortex. In this review, we summarize these findings and how they might relate to psychosis and to observed cell type-specific effects of antipsychotic drugs. We argue that quantifying the effects of antipsychotic drugs on specific neuronal circuit elements is a promising approach to understanding not only the mechanism of action of antipsychotic drugs but also psychosis. Finally, we outline some of the key experiments that should be done. The aims of this review are to provide an overview of the current circuit-based approaches to psychosis and to encourage further research in this direction.
Collapse
Affiliation(s)
- Georg B Keller
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland;
- Faculty of Natural Science, University of Basel, Basel, Switzerland
| | - Philipp Sterzer
- Department of Psychiatry, University of Basel, Basel, Switzerland
| |
Collapse
|
20
|
Correll CU, Tusconi M, Carta MG, Dursun SM. What Remains to Be Discovered in Schizophrenia Therapeutics: Contributions by Advancing the Molecular Mechanisms of Drugs for Psychosis and Schizophrenia. Biomolecules 2024; 14:906. [PMID: 39199294 PMCID: PMC11353083 DOI: 10.3390/biom14080906] [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: 06/17/2024] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 09/01/2024] Open
Abstract
Schizophrenia is a frequently debilitating and complex mental disorder affecting approximately 1% of the global population, characterized by symptoms such as hallucinations, delusions, disorganized thoughts and behaviors, cognitive dysfunction, and negative symptoms. Traditional treatment has centered on postsynaptic dopamine antagonists, commonly known as antipsychotic drugs, which aim to alleviate symptoms and improve functioning and the quality of life. Despite the availability of these medications, significant challenges remain in schizophrenia therapeutics, including incomplete symptom relief, treatment resistance, and medication side effects. This opinion article explores advancements in schizophrenia treatment, emphasizing molecular mechanisms, novel drug targets, and innovative delivery methods. One promising approach is novel strategies that target neural networks and circuits rather than single neurotransmitters, acknowledging the complexity of brain region interconnections involved in schizophrenia. Another promising approach is the development of biased agonists, which selectively activate specific signaling pathways downstream of receptors, offering potential for more precise pharmacological interventions with fewer side effects. The concept of molecular polypharmacy, where a single drug targets multiple molecular pathways, is exemplified by KarXT, a novel drug combining xanomeline and trospium to address both psychosis and cognitive dysfunction. This approach represents a comprehensive strategy for schizophrenia treatment, potentially improving outcomes for patients. In conclusion, advancing the molecular understanding of schizophrenia and exploring innovative therapeutic strategies hold promise for addressing the unmet needs in schizophrenia treatment, aiming for more effective and tailored interventions. Future research should focus on these novel approaches to achieve better clinical outcomes and improve the functional level and quality of life for individuals with schizophrenia.
Collapse
Affiliation(s)
- Christoph U. Correll
- Department of Psychiatry, Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY 10128, USA;
- Department of Psychiatry and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
- Department of Child and Adolescent Psychiatry, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | | | - Mauro Giovanni Carta
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy;
| | - Serdar M. Dursun
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB T6G 2G5, Canada;
| |
Collapse
|
21
|
Marshall RD, Menniti FS, Tepper MA. A Novel PDE10A Inhibitor for Tourette Syndrome and Other Movement Disorders. Cells 2024; 13:1230. [PMID: 39056811 PMCID: PMC11274801 DOI: 10.3390/cells13141230] [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: 06/06/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Tourette syndrome is a neurodevelopmental movement disorder involving basal ganglia dysfunction. PDE10A inhibitors modulate signaling in the striatal basal ganglia nuclei and are thus of interest as potential therapeutics in treating Tourette syndrome and other movement disorders. METHODS The preclinical pharmacology and toxicology, human safety and tolerability, and human PET striatal enzyme occupancy data for the PDE10A inhibitor EM-221 are presented. RESULTS EM-221 inhibited PDE10A with an in vitro IC50 of 9 pM and was >100,000 selective vs. other PDEs and other CNS receptors and enzymes. In rats, at doses of 0.05-0.50 mg/kg, EM-221 reduced hyperlocomotion and the disruption of prepulse inhibition induced by MK-801, attenuated conditioned avoidance, and facilitated novel object recognition, consistent with PDE10A's inhibition. EM-221 displayed no genotoxicity and was well tolerated up to 300 mg/kg in rats and 100 mg/kg in dogs. In single- and multiple-day ascending dose studies in healthy human volunteers, EM-221 was well tolerated up to 10 mg, with a maximum tolerated dose of 15 mg. PET imaging indicated that a PDE10A enzyme occupancy of up to 92.8% was achieved with a ~24 h half-life. CONCLUSIONS The preclinical and clinical data presented here support the study of EM-221 in phase 2 trials of Tourette syndrome and other movement disorders.
Collapse
Affiliation(s)
| | - Frank S. Menniti
- MindImmune Therapeutics, Inc., Kingston, RI 02881, USA;
- The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
| | - Mark A. Tepper
- EuMentis Therapeutics Inc., 275 Grove Street, 2-400, Newton, MA 02466, USA;
| |
Collapse
|
22
|
McCutcheon RA, Weber LAE, Nour MM, Cragg SJ, McGuire PM. Psychosis as a disorder of muscarinic signalling: psychopathology and pharmacology. Lancet Psychiatry 2024; 11:554-565. [PMID: 38795721 DOI: 10.1016/s2215-0366(24)00100-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 05/28/2024]
Abstract
Dopaminergic receptor antagonism is a crucial component of all licensed treatments for psychosis, and dopamine dysfunction has been central to pathophysiological models of psychotic symptoms. Some clinical trials, however, indicate that drugs that act through muscarinic receptor agonism can also be effective in treating psychosis, potentially implicating muscarinic abnormalities in the pathophysiology of psychosis. Here, we discuss understanding of the central muscarinic system, and we examine preclinical, behavioural, post-mortem, and neuroimaging evidence for its involvement in psychosis. We then consider how altered muscarinic signalling could contribute to the genesis and maintenance of psychotic symptoms, and we review the clinical evidence for muscarinic agents as treatments. Finally, we discuss future research that could clarify the relationship between the muscarinic system and psychotic symptoms.
Collapse
Affiliation(s)
- Robert A McCutcheon
- Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health, Oxford Health NHS Foundation Trust, Oxford, UK; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
| | - Lilian A E Weber
- Department of Psychiatry, University of Oxford, Oxford, UK; Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| | - Matthew M Nour
- Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health, Oxford Health NHS Foundation Trust, Oxford, UK; Max Planck UCL Centre for Computational Psychiatry and Ageing Research, University College London, London, UK
| | - Stephanie J Cragg
- Department of Physiology, Anatomy and Genetics, Centre for Cellular and Molecular Neurobiology, University of Oxford, UK; Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, MD, USA
| | - Philip M McGuire
- Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health, Oxford Health NHS Foundation Trust, Oxford, UK
| |
Collapse
|
23
|
Oliver D, Chesney E, Cullen AE, Davies C, Englund A, Gifford G, Kerins S, Lalousis PA, Logeswaran Y, Merritt K, Zahid U, Crossley NA, McCutcheon RA, McGuire P, Fusar-Poli P. Exploring causal mechanisms of psychosis risk. Neurosci Biobehav Rev 2024; 162:105699. [PMID: 38710421 PMCID: PMC11250118 DOI: 10.1016/j.neubiorev.2024.105699] [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: 11/01/2023] [Revised: 02/17/2024] [Accepted: 04/28/2024] [Indexed: 05/08/2024]
Abstract
Robust epidemiological evidence of risk and protective factors for psychosis is essential to inform preventive interventions. Previous evidence syntheses have classified these risk and protective factors according to their strength of association with psychosis. In this critical review we appraise the distinct and overlapping mechanisms of 25 key environmental risk factors for psychosis, and link these to mechanistic pathways that may contribute to neurochemical alterations hypothesised to underlie psychotic symptoms. We then discuss the implications of our findings for future research, specifically considering interactions between factors, exploring universal and subgroup-specific factors, improving understanding of temporality and risk dynamics, standardising operationalisation and measurement of risk and protective factors, and developing preventive interventions targeting risk and protective factors.
Collapse
Affiliation(s)
- Dominic Oliver
- Department of Psychiatry, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Oxford, UK; OPEN Early Detection Service, Oxford Health NHS Foundation Trust, Oxford, UK; Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
| | - Edward Chesney
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Addictions Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 4 Windsor Walk, London SE5 8AF, UK
| | - Alexis E Cullen
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Clinical Neuroscience, Karolinska Institutet, Sweden
| | - Cathy Davies
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Amir Englund
- Addictions Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 4 Windsor Walk, London SE5 8AF, UK
| | - George Gifford
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Sarah Kerins
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Paris Alexandros Lalousis
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Yanakan Logeswaran
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Biostatistics & Health Informatics, King's College London, London, UK
| | - Kate Merritt
- Division of Psychiatry, Institute of Mental Health, UCL, London, UK
| | - Uzma Zahid
- Department of Psychology, King's College London, London, UK
| | - Nicolas A Crossley
- Department of Psychiatry, University of Oxford, Oxford, UK; Department of Psychiatry, School of Medicine, Pontificia Universidad Católica de Chile, Chile
| | - Robert A McCutcheon
- Department of Psychiatry, University of Oxford, Oxford, UK; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Oxford Health NHS Foundation Trust, Oxford, UK
| | - Philip McGuire
- Department of Psychiatry, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Oxford, UK; OPEN Early Detection Service, Oxford Health NHS Foundation Trust, Oxford, UK
| | - Paolo Fusar-Poli
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; OASIS Service, South London and Maudsley NHS Foundation Trust, London SE11 5DL, UK
| |
Collapse
|
24
|
Chen C, Masotti M, Shepard N, Promes V, Tombesi G, Arango D, Manzoni C, Greggio E, Hilfiker S, Kozorovitskiy Y, Parisiadou L. LRRK2 mediates haloperidol-induced changes in indirect pathway striatal projection neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597594. [PMID: 38895420 PMCID: PMC11185612 DOI: 10.1101/2024.06.06.597594] [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
Haloperidol is used to manage psychotic symptoms in several neurological disorders through mechanisms that involve antagonism of dopamine D2 receptors that are highly expressed in the striatum. Significant side effects of haloperidol, known as extrapyramidal symptoms, lead to motor deficits similar to those seen in Parkinson's disease and present a major challenge in clinical settings. The underlying molecular mechanisms responsible for these side effects remain poorly understood. Parkinson's disease-associated LRRK2 kinase has an important role in striatal physiology and a known link to dopamine D2 receptor signaling. Here, we systematically explore convergent signaling of haloperidol and LRRK2 through pharmacological or genetic inhibition of LRRK2 kinase, as well as knock-in mouse models expressing pathogenic mutant LRRK2 with increased kinase activity. Behavioral assays show that LRRK2 kinase inhibition ameliorates haloperidol-induced motor changes in mice. A combination of electrophysiological and anatomical approaches reveals that LRRK2 kinase inhibition interferes with haloperidol-induced changes, specifically in striatal neurons of the indirect pathway. Proteomic studies and targeted intracellular pathway analyses demonstrate that haloperidol induces a similar pattern of intracellular signaling as increased LRRK2 kinase activity. Our study suggests that LRRK2 kinase plays a key role in striatal dopamine D2 receptor signaling underlying the undesirable motor side effects of haloperidol. This work opens up new therapeutic avenues for dopamine-related disorders, such as psychosis, also furthering our understanding of Parkinson's disease pathophysiology.
Collapse
Affiliation(s)
- Chuyu Chen
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Meghan Masotti
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Nathaniel Shepard
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Vanessa Promes
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Giulia Tombesi
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Daniel Arango
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | | | | | - Sabine Hilfiker
- Department of Anesthesiology, Rutgers, New Jersey Medical School, NJ, USA
| | | | - Loukia Parisiadou
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
25
|
Yang SM, Ghoshal A, Hubbard JM, Gackière F, Teyssié R, Neale SA, Hopkins SC, Koblan KS, Bristow LJ, Dedic N. TAAR1 agonist ulotaront modulates striatal and hippocampal glutamate function in a state-dependent manner. Neuropsychopharmacology 2024; 49:1091-1103. [PMID: 38110609 PMCID: PMC11109157 DOI: 10.1038/s41386-023-01779-x] [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: 09/12/2023] [Revised: 11/08/2023] [Accepted: 11/21/2023] [Indexed: 12/20/2023]
Abstract
Aberrant dopaminergic and glutamatergic function, particularly within the striatum and hippocampus, has repeatedly been associated with the pathophysiology of schizophrenia. Supported by preclinical and recent clinical data, trace amine-associated receptor 1 (TAAR1) agonism has emerged as a potential new treatment approach for schizophrenia. While current evidence implicates TAAR1-mediated regulation of dopaminergic tone as the primary circuit mechanism, little is known about the effects of TAAR1 agonists on the glutamatergic system and excitation-inhibition balance. Here we assessed the impact of ulotaront (SEP-363856), a TAAR1 agonist in Phase III clinical development for schizophrenia, on glutamate function in the mouse striatum and hippocampus. Ulotaront reduced spontaneous glutamatergic synaptic transmission and neuronal firing in striatal and hippocampal brain slices, respectively. Interestingly, ulotaront potentiated electrically-evoked excitatory synaptic transmission in both brain regions, suggesting the ability to modulate glutamatergic signaling in a state-dependent manner. Similar striatal effects were also observed with the TAAR1 agonist, RO5166017. Furthermore, we show that ulotaront regulates excitation-inhibition balance in the striatum by specifically modulating glutamatergic, but not GABAergic, spontaneous synaptic events. These findings expand the mechanistic circuit hypothesis of ulotaront and TAAR1 agonists, which may be uniquely positioned to normalize both the excessive dopaminergic tone and regulate abnormal glutamatergic function associated with schizophrenia.
Collapse
Affiliation(s)
- Sung M Yang
- Sumitomo Pharma America, Inc., Marlborough, MA, USA
| | - Ayan Ghoshal
- Sumitomo Pharma America, Inc., Marlborough, MA, USA
| | | | | | | | | | | | | | | | - Nina Dedic
- Sumitomo Pharma America, Inc., Marlborough, MA, USA.
| |
Collapse
|
26
|
Ricci V, De Berardis D, Maina G. Third-Generation Antipsychotics and Lurasidone in the Treatment of Substance-Induced Psychoses: A Narrative Review. Healthcare (Basel) 2024; 12:339. [PMID: 38338224 PMCID: PMC10855531 DOI: 10.3390/healthcare12030339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/09/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
This narrative review explores the efficacy and tolerability of third-generation antipsychotics (TGAs)-aripiprazole, cariprazine, brexpiprazole, and lurasidone-for the management of substance-induced psychosis (SIP). SIP is a psychiatric condition triggered by substance misuse or withdrawal, characterized by unique features distinct from those of primary psychotic disorders. These distinctive features include a heightened prevalence of positive symptoms, such as hallucinations and delusions, in addition to a spectrum of mood and cognitive disturbances. This review comprehensively investigates various substances, such as cannabinoids, cocaine, amphetamines, and LSD, which exhibit a greater propensity for inducing psychosis. TGAs exhibit substantial promise in addressing both psychotic symptoms and issues related to substance misuse. This review elucidates the distinctive pharmacological properties of each TGA, their intricate interactions with neurotransmitters, and their potential utility in the treatment of SIP. We advocate for further research to delineate the long-term effects of TGAs in this context and underscore the necessity for adopting an integrated approach that combines pharmacological and psychological interventions. Our findings underscore the intricate and multifaceted nature of treating SIP, highlighting the potential role of TGAs within therapeutic strategies.
Collapse
Affiliation(s)
- Valerio Ricci
- San Luigi Gonzaga Hospital, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy;
| | - Domenico De Berardis
- NHS, Department of Mental Health, Psychiatric Service for Diagnosis and Treatment, Hospital “G. Mazzini”, ASL 4, 64100 Teramo, Italy;
| | - Giuseppe Maina
- San Luigi Gonzaga Hospital, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy;
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, 10124 Torino, Italy
| |
Collapse
|
27
|
Howes OD, Bukala BR, Beck K. Schizophrenia: from neurochemistry to circuits, symptoms and treatments. Nat Rev Neurol 2024; 20:22-35. [PMID: 38110704 DOI: 10.1038/s41582-023-00904-0] [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: 11/08/2023] [Indexed: 12/20/2023]
Abstract
Schizophrenia is a leading cause of global disability. Current pharmacotherapy for the disease predominantly uses one mechanism - dopamine D2 receptor blockade - but often shows limited efficacy and poor tolerability. These limitations highlight the need to better understand the aetiology of the disease to aid the development of alternative therapeutic approaches. Here, we review the latest meta-analyses and other findings on the neurobiology of prodromal, first-episode and chronic schizophrenia, and the link to psychotic symptoms, focusing on imaging evidence from people with the disorder. This evidence demonstrates regionally specific neurotransmitter alterations, including higher glutamate and dopamine measures in the basal ganglia, and lower glutamate, dopamine and γ-aminobutyric acid (GABA) levels in cortical regions, particularly the frontal cortex, relative to healthy individuals. We consider how dysfunction in cortico-thalamo-striatal-midbrain circuits might alter brain information processing to underlie psychotic symptoms. Finally, we discuss the implications of these findings for developing new, mechanistically based treatments and precision medicine for psychotic symptoms, as well as negative and cognitive symptoms.
Collapse
Affiliation(s)
- Oliver D Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- Faculty of Medicine, Institute of Clinical Sciences, Imperial College London, London, UK.
| | - Bernard R Bukala
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Katherine Beck
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| |
Collapse
|
28
|
Cameron D, Vinh NN, Prapaiwongs P, Perry EA, Walters JTR, Li M, O’Donovan MC, Bray NJ. Genetic implication of prenatal GABAergic and cholinergic neuron development in susceptibility to schizophrenia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.14.23299948. [PMID: 38168283 PMCID: PMC10760267 DOI: 10.1101/2023.12.14.23299948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Background The ganglionic eminences are fetal-specific structures that give rise to gamma-aminobutyric acid (GABA)- and acetylcholine- releasing neurons of the forebrain. Given evidence for GABAergic and cholinergic disturbances in schizophrenia, as well as an early neurodevelopmental component to the disorder, we tested the potential involvement of developing cells of the ganglionic eminences in mediating genetic risk for the condition. Study Design We combined data from a recent large-scale genome-wide association study of schizophrenia with single cell RNA sequencing data from the human ganglionic eminences to test enrichment of schizophrenia risk variation in genes with high expression specificity for particular developing cell populations within these structures. We additionally performed the single nuclei Assay for Transposase-Accessible Chromatin with Sequencing (snATAC-Seq) to map potential regulatory genomic regions operating in individual cell populations of the human ganglionic eminences, using these to additionally test for enrichment of schizophrenia common genetic variant liability and to functionally annotate non-coding variants associated with the disorder. Study Results Schizophrenia common variant liability was enriched in genes with high expression specificity for developing neuron populations that are predicted to form dopamine D1 and D2 receptor expressing GABAergic medium spiny neurons of the striatum, cortical somatostatin-positive GABAergic interneurons, calretinin-positive GABAergic neurons and cholinergic neurons. Consistent with these findings, schizophrenia genetic risk was also concentrated in predicted regulatory genomic sequence mapped in developing neuronal populations of the ganglionic eminences. Conclusions Our study provides evidence for a role of prenatal GABAergic and cholinergic neuron development in later susceptibility to schizophrenia.
Collapse
Affiliation(s)
- Darren Cameron
- Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Ngoc-Nga Vinh
- Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Parinda Prapaiwongs
- Neuroscience & Mental Health Innovation Institute, Cardiff University, Cardiff, United Kingdom
| | - Elizabeth A. Perry
- Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - James T. R. Walters
- Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Meng Li
- Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
- Neuroscience & Mental Health Innovation Institute, Cardiff University, Cardiff, United Kingdom
| | - Michael C. O’Donovan
- Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Nicholas J. Bray
- Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
- Neuroscience & Mental Health Innovation Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
29
|
Conn KA, Alexander S, Burne THJ, Kesby JP. Antagonism of D2 receptors via raclopride ameliorates amphetamine-induced associative learning deficits in male mice. Behav Brain Res 2023; 454:114649. [PMID: 37643667 DOI: 10.1016/j.bbr.2023.114649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/09/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023]
Abstract
Dopamine levels in the dorsomedial striatum (DMS) are highly dynamic and are thought to underly the encoding of action-outcome associations. Although it is known that amphetamine disrupts the learning that is required for goal-directed action, the role of D1 and D2 receptors in this process has not been established. In this study, we examined the role of D1 and D2 receptor antagonists on learning in response to amphetamine. We used the outcome-specific devaluation task to examine goal-directed action in male C57BL6/J mice treated systemically with either a D1 antagonist (SCH-23990; 0.01 mg/kg) or a D2 antagonist (raclopride; 0.5 mg/kg) and then administered amphetamine (1 mg/kg). The mice were injected repeatedly throughout the instrumental training phase of the task to assess the impact on the learning of action-outcomes, and the subsequent choice test assessing performance of goal-directed action was conducted drug free. Effects of chronic drug administration on locomotor behaviour was assessed before and after the choice test. Treatment during learning with either amphetamine, or the D1 or D2 antagonists, impaired the subsequent performance of goal-directed action. The amphetamine-induced impairment in goal-directed action was reversed in mice treated with raclopride, but not when treated with SCH-23990. By contrast, amphetamine-induced hyperactivity was reversed in mice treated with SCH-23990, but not in mice treated with raclopride. Taken together, these data support the role of a balance of dopamine receptor signalling after amphetamine treatment. While overall D1 receptor availability is necessary to promote learning, in a state of elevated dopamine, modifying D2 receptor function can ameliorate learning deficits.
Collapse
Affiliation(s)
- Kyna-Anne Conn
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Suzy Alexander
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Thomas H J Burne
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia; Queensland Centre for Mental Health Research, Wacol, QLD 4076, Australia
| | - James P Kesby
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia; Queensland Centre for Mental Health Research, Wacol, QLD 4076, Australia.
| |
Collapse
|