1
|
Oliveira A, Azevedo M, Seixas R, Silva S, Martinho R, Serrão P, Silva E, Moreira-Rodrigues M. Hippocampus muscarinic M4 receptor mRNA expression may influence central cholinergic activity, causing fear memory strengthening by peripheral adrenaline. Neuropharmacology 2025; 271:110382. [PMID: 39988278 DOI: 10.1016/j.neuropharm.2025.110382] [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: 12/03/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Adrenaline (Ad) strengthens contextual fear memory by increasing blood glucose, possibly enhancing hippocampus acetylcholine synthesis. Nevertheless, it is unclear if peripheral Ad influences the cholinergic system, contributing to contextual fear memory strengthening. We aimed to evaluate whether peripheral Ad alters muscarinic receptor expression and if the cholinergic system is involved in peripheral Ad contextual fear memory strengthening effect. Wild-type (WT) and Ad-deficient male mice (129 × 1/SvJ) underwent a fear conditioning procedure followed by intraperitoneal pre-training and pre-context administration of Ad (0.1 mg/kg), atropine (10 mg/kg), methylatropine (0.5 mg/kg), Ad (0.1 mg/kg) plus atropine (10 mg/kg) or vehicle (NaCl, 0.9%). Shock responsiveness and freezing behaviour were accessed. Hippocampal M1, M2, and M4 mRNA expression were evaluated. Ad-deficient mice presented decreased hippocampal muscarinic M4 subtype receptor mRNA expression compared to WT mice. In Ad-administered Ad-deficient mice, hippocampal muscarinic M4 subtype receptor mRNA expression increased compared with vehicle-administered Ad-deficient mice. On the context day, atropine-administered WT mice presented decreased freezing behaviour compared to vehicle or methylatropine-administered WT mice. Moreover, Ad plus atropine-administered Ad-deficient mice led to decreased freezing behaviour compared to Ad-administered Ad-deficient mice. In conclusion, Ad-deficient mice's contextual fear memory impairment was associated with hippocampal muscarinic M4 subtype receptor down expression, which was reversed by Ad. This may be related to contextual fear memory consolidation or retrieval induced by peripheral Ad. Furthermore, the effect of Ad contextual fear memory might be due to increased hippocampus muscarinic subtype M4 expression, which may contribute to increased cholinergic activity in the central nervous system.
Collapse
Affiliation(s)
- Ana Oliveira
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Márcia Azevedo
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Rafaela Seixas
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Soraia Silva
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Raquel Martinho
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Paula Serrão
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Biomedicine, Faculty of Medicine, University of Porto (FMUP), Porto, Portugal
| | - Elisabete Silva
- Ageing and Stress Group, i3S- Institute for Research and Innovation in Health, University of Porto, Porto, Portugal; Faculty of Veterinary Medicine, Lusófona University, Lisbon, Portugal
| | - Mónica Moreira-Rodrigues
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal.
| |
Collapse
|
2
|
Moran PM, Granger KT. IUPHAR review: Moving beyond dopamine-based therapeutic strategies for schizophrenia. Pharmacol Res 2025; 216:107727. [PMID: 40320224 DOI: 10.1016/j.phrs.2025.107727] [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: 03/19/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 05/12/2025]
Abstract
In the following we comprehensively review approaches to treating schizophrenia that do not primarily involve dopamine antagonism or partial agonism. Following 70 years of broadly similar dopamine D2 receptor antagonist/partial agonist drugs, Cobenfy™ was approved as a novel antipsychotic in September 2024. Cobenfy™ is a combination formulation of xanomeline, a muscarinic cholinergic M1/M4 receptor agonist and trospium, a peripherally restricted muscarinic antagonist included to offset peripheral side effects of xanomeline. This approval has reinvigorated optimism in the field and raised important questions for the future direction of antipsychotic drug development. We review therapeutic strategies beyond dopamine that have been and are currently being investigated to address whether there are a sufficient number of novel approaches to maintain the momentum of this breakthrough and question why it has taken so long. The current pipeline of late-stage compounds is low and potentially constrained by historical setbacks and challenges in clinical trial design for schizophrenia. This success rate has future potential to improve given the range of biomarkers in development designed to enable greater precision in future clinical trials. Cobenfy™ approval demonstrates that with combination formulations designed to improve side effect profiles and optimised clinical trial design it is possible to generate tolerable and efficacious treatment options for patients beyond a solely dopaminergic framework. We conclude that advances in understanding the neurobiology of schizophrenia, while not complete, has generated a diverse and well justified pool of potentially novel and repurpose-ready approaches, with mechanisms beyond simple dopamine D2 antagonism/partial agonism.
Collapse
Affiliation(s)
- Paula M Moran
- School of Psychology, University of Nottingham, University Park, NG72RD, UK.
| | - Kiri T Granger
- Monument Therapeutics Ltd., Alderley Park, Congleton Road, Cheshire, Macclesfield SK10 4TG, UK.
| |
Collapse
|
3
|
Liu B, Christopoulos A, Thal DM, Capuano B, Valant C, Scammells PJ. The Prosperity and Adversity of M 4 Muscarinic Acetylcholine Receptor Activators in the Treatment of Neuropsychiatric Disorders. J Med Chem 2025; 68:7932-7954. [PMID: 40237346 DOI: 10.1021/acs.jmedchem.5c00678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Since the serendipitous discovery of chlorpromazine in the 1950s, almost all current anti-schizophrenia drugs utilize the same mode of action by blocking the dopamine receptors in the brain. Unfortunately, these only treat part of the symptoms and are ineffective in almost 30% of patients. The recent FDA approval of Cobenfy, a coformulation of xanomeline, a M1/M4 muscarinic acetylcholine receptor (mAChR) agonist, and a peripherally restricted pan-mAChR blocker, has propelled the M4R as a validated and novel antipsychotic target. With >25 years of history in developing xanomeline, significant challenges remain in developing M4R activators, either at the ACh orthosteric binding site or allosterically via secondary less-conserved binding sites. Herein, we summarize recent successes and failures of M4R agonists and positive allosteric modulators, along with the progress in structure-activity relationship studies on both orthosteric and allosteric scaffolds to offer pathways for future therapeutics to this novel biological target for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Boqun Liu
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
- Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - David M Thal
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Ben Capuano
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Celine Valant
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Peter J Scammells
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| |
Collapse
|
4
|
Meyer JM, Kramer K, Vuocolo S, Kaul I, Miller AC. From theory to therapy: unlocking the potential of muscarinic receptor activation in schizophrenia with the dual M1/M4 muscarinic receptor agonist xanomeline and trospium chloride and insights from clinical trials. Int J Neuropsychopharmacol 2025; 28:pyaf015. [PMID: 40056428 PMCID: PMC11997306 DOI: 10.1093/ijnp/pyaf015] [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: 12/11/2024] [Accepted: 03/03/2025] [Indexed: 03/10/2025] Open
Abstract
Since the 1950s, understanding of antipsychotic activity in schizophrenia has been largely grounded in the dopamine (DA) hypothesis. Most antipsychotics approved for schizophrenia interact with D2 DA receptors as an important part of their mechanism of action. While antipsychotics blocking D2 DA receptors can be effective for positive symptoms of schizophrenia, none are approved by regulatory authorities for predominant negative or cognitive symptoms. Moreover, many of these agents induce a range of problematic side effects related to D2 DA receptor blockade (eg, drug-induced parkinsonism, akathisia, tardive dyskinesia, hyperprolactinemia and related sexual side effects, sedation). This has prompted the search for novel mechanisms with improved efficacy and tolerability based on evidence supporting involvement of other neurotransmitter systems in schizophrenia pathophysiology, including acetylcholine, gamma-aminobutyric acid, and glutamate. Among these options, targeting muscarinic receptors emerged as a promising treatment strategy. In September 2024, the U.S. Food and Drug Administration approved xanomeline and trospium chloride for treatment of adults with schizophrenia based on results from three 5-week, randomized, double-blind, placebo-controlled trials and two 52-week open-label trials. In the placebo-controlled trials, xanomeline/trospium reduced symptoms of schizophrenia, was generally well tolerated, and was not associated with clinically meaningful motor symptoms, hyperprolactinemia, sexual side effects, or weight gain compared with placebo. The long-term safety of xanomeline/trospium was also confirmed in two 52-week, open-label trials. This paper reviews the preclinical and clinical rationale for muscarinic receptor activation as a treatment for schizophrenia and the efficacy, safety, and tolerability profile of xanomeline/trospium.
Collapse
Affiliation(s)
- Jonathan M Meyer
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - Ken Kramer
- Bristol Myers Squibb, Princeton, NJ, United States
| | | | - Inder Kaul
- Bristol Myers Squibb, Princeton, NJ, United States
| | | |
Collapse
|
5
|
Conley AC, Key AP, Blackford JU, Russell JK, Albert KM, Gong X, Bubser M, Rook JM, Conn PJ, Lindsley CW, Jones CK, Newhouse PA. Safety, Tolerability, Pharmacokinetic and Pharmacodynamic Effects of the Muscarinic M 1 Positive Allosteric Modulator VU0467319 for Alzheimer's disease: A Single Ascending-Dose Study in Healthy Participants. RESEARCH SQUARE 2025:rs.3.rs-6271510. [PMID: 40235479 PMCID: PMC11998771 DOI: 10.21203/rs.3.rs-6271510/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The development of cholinergic neurotransmitter based cognitive enhancers for Alzheimer's disease and other neuropsychiatric disorders have focused recently on allosteric modulation of specific muscarinic acetylcholine receptor (mAChR) subtypes to reduce dose-limiting side-effects that have been the hallmark of earlier orthosteric mAChR agonists. VU0467319 (VU319) is an investigational positive allosteric modulator of the M 1 mAChR. A Phase 1 first-in-human study was conducted assessing safety and brain activity utilizing cognitive tasks and event-related potentials (ERPs) in single-ascending dose and food effect studies. VU319 was given orally to 52 healthy volunteers aged 18-55 years. The single ascending dose study tested 40 participants in five dose escalating cohorts (60, 120, 240, 400, 600 mg; 6 VU319/2 placebo per dose). The food effect study involved 12 participants, 10 VU319 (120 mg)/2 placebo. Exploratory cognitive and electrophysiological tasks were examined pre-dosing and at 5 hours post-dose. Tolerability was good with no observed dose limiting side effects throughout the full dose range tested. Drug exposure increased with dose in a less than dose-proportional manner with a half-life ranging from 36 to 43 hours. Absorption was increased with food. Exploratory cognitive/ERP testing showed evidence for drug-induced CNS activity on higher doses of VU319 compared to placebo. Single dose VU319 across five ascending cohorts appeared to have a favorable safety profile and a PK profile consistent with once daily dosing. Target engagement results suggest stimulation of the cholinergic system functioning in healthy adults following a single dose of VU319. These results provide a strong foundation for further studies of positive allosteric modulators of muscarinic M 1 receptors for potential cognitive or behavioral benefits.
Collapse
|
6
|
Mandai T, Simen AA, Laurenza A, Kimura H. M1 receptor positive allosteric modulators discovery approaches. Trends Pharmacol Sci 2025; 46:298-302. [PMID: 40133192 DOI: 10.1016/j.tips.2025.03.001] [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: 02/03/2025] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/27/2025]
Abstract
The development of M1 muscarinic acetylcholine-receptor-positive allosteric modulators has been hindered by their limited cognitive efficacy and cholinergic side effects. We discuss two unique approaches - low intrinsic agonism and low binding cooperativity - that were developed to address these issues and their therapeutic implications.
Collapse
Affiliation(s)
- Takao Mandai
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Arthur A Simen
- Neuroscience Therapeutic Area Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Antonio Laurenza
- Neuroscience Therapeutic Area Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Haruhide Kimura
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan.
| |
Collapse
|
7
|
Mancini V, Latreche C, Fanshawe JB, Varvari I, Zauchenberger CZ, McGinn N, Catalan A, Pillinger T, McGuire PK, McCutcheon RA. Anticholinergic Burden and Cognitive Function in Psychosis: A Systematic Review and Meta-Analysis. Am J Psychiatry 2025; 182:349-359. [PMID: 40007252 DOI: 10.1176/appi.ajp.20240260] [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: 02/27/2025]
Abstract
OBJECTIVE The authors synthesized evidence from studies quantifying the relationship between anticholinergic medication and cognitive function in psychosis, and additionally explored studies that investigated whether reducing anticholinergic medications affects cognitive function in individuals with psychosis. METHODS A database search was conducted in MEDLINE, Embase, and PsycINFO, from database inception to October 2023, for studies reporting objective cognitive assessment and quantification of anticholinergic burden using clinical scales, serological anticholinergic activity, or tapering of anticholinergic medications. Analyses were carried out in R using the metafor package. Random-effects meta-analysis models were employed, along with assessment of heterogeneity, study quality, and meta-regressions (age, sex, and antipsychotic dosage in chlorpromazine equivalents). RESULTS Of 1,337 citations retrieved, 40 met inclusion criteria, comprising 25 anticholinergic burden studies (4,620 patients), six serological anticholinergic activity studies (382 patients), and nine tapering studies (186 patients). A negative correlation was identified between anticholinergic burden and global cognition (r=-0.37, 95% CI=-0.48, -0.25), verbal learning (r=-0.28, 95% CI=-0.36, -0.21), visual learning (r=-0.17, 95% CI=-0.28, -0.06), working memory (r=-0.22, 95% CI=-0.29, -0.14), processing speed (r=-0.24, 95% CI=-0.35, -0.13), attention (r=-0.19, 95% CI=-0.29, -0.08), executive functions (r=-0.17, 95% CI=-0.27, -0.06), and social cognition (r=-0.12, 95% CI=-0.19, -0.05), and between serological anticholinergic activity and verbal learning (r=-0.26, 95% CI=-0.38, -0.14), working memory (r=-0.19, 95% CI=-0.35, -0.03), and executive functions (r=-0.16, 95% CI=-0.27, -0.04). Finally, tapering off anticholinergic medication improved the scores in verbal learning (d=0.77, 95% CI=0.44, 1.1), working memory (d=0.94, 95% CI=0.63, 1.26), and executive functions (d=0.44, 95% CI=0.26, 0.62). CONCLUSIONS Anticholinergic burden is associated with the cognitive impairments observed in psychosis. From a clinical perspective, tapering off anticholinergic medication in patients with psychosis may improve cognition. However, randomized clinical trials are needed for an unbiased quantification of benefit.
Collapse
Affiliation(s)
- Valentina Mancini
- Department of Psychiatry (Mancini, Fanshawe, Varvari, Zauchenberger, Catalan, McGuire, McCutcheon), MRC Brain Network Dynamics Unit (Mancini), and Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging (Mancini), University of Oxford, Oxford, UK; TUNEUP, Oxford Health NHS Foundation Trust, Oxford, UK (Mancini, Varvari, Zauchenberger, McCutcheon); Department of Psychiatry, University of Geneva, Geneva (Latreche, McGinn); Oxford Health NHS Foundation Trust, Oxford, UK (Fanshawe, McGuire); Department of Psychosis Studies, Institute of Psychiatry, Psychology, and Neuroscience, King's College London (Catalan, Pillinger, McGuire, McCutcheon); Basurto University Hospital, OSI Bilbao-Basurto, Biobizkaia Health Research Institute, University of the Basque Country UPV/EHU, Centro de Investigación en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Bilbao, Spain (Catalan); South London and Maudsley NHS Foundation Trust, London (Pillinger)
| | - Caren Latreche
- Department of Psychiatry (Mancini, Fanshawe, Varvari, Zauchenberger, Catalan, McGuire, McCutcheon), MRC Brain Network Dynamics Unit (Mancini), and Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging (Mancini), University of Oxford, Oxford, UK; TUNEUP, Oxford Health NHS Foundation Trust, Oxford, UK (Mancini, Varvari, Zauchenberger, McCutcheon); Department of Psychiatry, University of Geneva, Geneva (Latreche, McGinn); Oxford Health NHS Foundation Trust, Oxford, UK (Fanshawe, McGuire); Department of Psychosis Studies, Institute of Psychiatry, Psychology, and Neuroscience, King's College London (Catalan, Pillinger, McGuire, McCutcheon); Basurto University Hospital, OSI Bilbao-Basurto, Biobizkaia Health Research Institute, University of the Basque Country UPV/EHU, Centro de Investigación en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Bilbao, Spain (Catalan); South London and Maudsley NHS Foundation Trust, London (Pillinger)
| | - Jack B Fanshawe
- Department of Psychiatry (Mancini, Fanshawe, Varvari, Zauchenberger, Catalan, McGuire, McCutcheon), MRC Brain Network Dynamics Unit (Mancini), and Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging (Mancini), University of Oxford, Oxford, UK; TUNEUP, Oxford Health NHS Foundation Trust, Oxford, UK (Mancini, Varvari, Zauchenberger, McCutcheon); Department of Psychiatry, University of Geneva, Geneva (Latreche, McGinn); Oxford Health NHS Foundation Trust, Oxford, UK (Fanshawe, McGuire); Department of Psychosis Studies, Institute of Psychiatry, Psychology, and Neuroscience, King's College London (Catalan, Pillinger, McGuire, McCutcheon); Basurto University Hospital, OSI Bilbao-Basurto, Biobizkaia Health Research Institute, University of the Basque Country UPV/EHU, Centro de Investigación en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Bilbao, Spain (Catalan); South London and Maudsley NHS Foundation Trust, London (Pillinger)
| | - Ioana Varvari
- Department of Psychiatry (Mancini, Fanshawe, Varvari, Zauchenberger, Catalan, McGuire, McCutcheon), MRC Brain Network Dynamics Unit (Mancini), and Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging (Mancini), University of Oxford, Oxford, UK; TUNEUP, Oxford Health NHS Foundation Trust, Oxford, UK (Mancini, Varvari, Zauchenberger, McCutcheon); Department of Psychiatry, University of Geneva, Geneva (Latreche, McGinn); Oxford Health NHS Foundation Trust, Oxford, UK (Fanshawe, McGuire); Department of Psychosis Studies, Institute of Psychiatry, Psychology, and Neuroscience, King's College London (Catalan, Pillinger, McGuire, McCutcheon); Basurto University Hospital, OSI Bilbao-Basurto, Biobizkaia Health Research Institute, University of the Basque Country UPV/EHU, Centro de Investigación en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Bilbao, Spain (Catalan); South London and Maudsley NHS Foundation Trust, London (Pillinger)
| | - Chambrez-Zita Zauchenberger
- Department of Psychiatry (Mancini, Fanshawe, Varvari, Zauchenberger, Catalan, McGuire, McCutcheon), MRC Brain Network Dynamics Unit (Mancini), and Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging (Mancini), University of Oxford, Oxford, UK; TUNEUP, Oxford Health NHS Foundation Trust, Oxford, UK (Mancini, Varvari, Zauchenberger, McCutcheon); Department of Psychiatry, University of Geneva, Geneva (Latreche, McGinn); Oxford Health NHS Foundation Trust, Oxford, UK (Fanshawe, McGuire); Department of Psychosis Studies, Institute of Psychiatry, Psychology, and Neuroscience, King's College London (Catalan, Pillinger, McGuire, McCutcheon); Basurto University Hospital, OSI Bilbao-Basurto, Biobizkaia Health Research Institute, University of the Basque Country UPV/EHU, Centro de Investigación en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Bilbao, Spain (Catalan); South London and Maudsley NHS Foundation Trust, London (Pillinger)
| | - Nova McGinn
- Department of Psychiatry (Mancini, Fanshawe, Varvari, Zauchenberger, Catalan, McGuire, McCutcheon), MRC Brain Network Dynamics Unit (Mancini), and Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging (Mancini), University of Oxford, Oxford, UK; TUNEUP, Oxford Health NHS Foundation Trust, Oxford, UK (Mancini, Varvari, Zauchenberger, McCutcheon); Department of Psychiatry, University of Geneva, Geneva (Latreche, McGinn); Oxford Health NHS Foundation Trust, Oxford, UK (Fanshawe, McGuire); Department of Psychosis Studies, Institute of Psychiatry, Psychology, and Neuroscience, King's College London (Catalan, Pillinger, McGuire, McCutcheon); Basurto University Hospital, OSI Bilbao-Basurto, Biobizkaia Health Research Institute, University of the Basque Country UPV/EHU, Centro de Investigación en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Bilbao, Spain (Catalan); South London and Maudsley NHS Foundation Trust, London (Pillinger)
| | - Ana Catalan
- Department of Psychiatry (Mancini, Fanshawe, Varvari, Zauchenberger, Catalan, McGuire, McCutcheon), MRC Brain Network Dynamics Unit (Mancini), and Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging (Mancini), University of Oxford, Oxford, UK; TUNEUP, Oxford Health NHS Foundation Trust, Oxford, UK (Mancini, Varvari, Zauchenberger, McCutcheon); Department of Psychiatry, University of Geneva, Geneva (Latreche, McGinn); Oxford Health NHS Foundation Trust, Oxford, UK (Fanshawe, McGuire); Department of Psychosis Studies, Institute of Psychiatry, Psychology, and Neuroscience, King's College London (Catalan, Pillinger, McGuire, McCutcheon); Basurto University Hospital, OSI Bilbao-Basurto, Biobizkaia Health Research Institute, University of the Basque Country UPV/EHU, Centro de Investigación en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Bilbao, Spain (Catalan); South London and Maudsley NHS Foundation Trust, London (Pillinger)
| | - Toby Pillinger
- Department of Psychiatry (Mancini, Fanshawe, Varvari, Zauchenberger, Catalan, McGuire, McCutcheon), MRC Brain Network Dynamics Unit (Mancini), and Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging (Mancini), University of Oxford, Oxford, UK; TUNEUP, Oxford Health NHS Foundation Trust, Oxford, UK (Mancini, Varvari, Zauchenberger, McCutcheon); Department of Psychiatry, University of Geneva, Geneva (Latreche, McGinn); Oxford Health NHS Foundation Trust, Oxford, UK (Fanshawe, McGuire); Department of Psychosis Studies, Institute of Psychiatry, Psychology, and Neuroscience, King's College London (Catalan, Pillinger, McGuire, McCutcheon); Basurto University Hospital, OSI Bilbao-Basurto, Biobizkaia Health Research Institute, University of the Basque Country UPV/EHU, Centro de Investigación en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Bilbao, Spain (Catalan); South London and Maudsley NHS Foundation Trust, London (Pillinger)
| | - Philip K McGuire
- Department of Psychiatry (Mancini, Fanshawe, Varvari, Zauchenberger, Catalan, McGuire, McCutcheon), MRC Brain Network Dynamics Unit (Mancini), and Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging (Mancini), University of Oxford, Oxford, UK; TUNEUP, Oxford Health NHS Foundation Trust, Oxford, UK (Mancini, Varvari, Zauchenberger, McCutcheon); Department of Psychiatry, University of Geneva, Geneva (Latreche, McGinn); Oxford Health NHS Foundation Trust, Oxford, UK (Fanshawe, McGuire); Department of Psychosis Studies, Institute of Psychiatry, Psychology, and Neuroscience, King's College London (Catalan, Pillinger, McGuire, McCutcheon); Basurto University Hospital, OSI Bilbao-Basurto, Biobizkaia Health Research Institute, University of the Basque Country UPV/EHU, Centro de Investigación en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Bilbao, Spain (Catalan); South London and Maudsley NHS Foundation Trust, London (Pillinger)
| | - Robert A McCutcheon
- Department of Psychiatry (Mancini, Fanshawe, Varvari, Zauchenberger, Catalan, McGuire, McCutcheon), MRC Brain Network Dynamics Unit (Mancini), and Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging (Mancini), University of Oxford, Oxford, UK; TUNEUP, Oxford Health NHS Foundation Trust, Oxford, UK (Mancini, Varvari, Zauchenberger, McCutcheon); Department of Psychiatry, University of Geneva, Geneva (Latreche, McGinn); Oxford Health NHS Foundation Trust, Oxford, UK (Fanshawe, McGuire); Department of Psychosis Studies, Institute of Psychiatry, Psychology, and Neuroscience, King's College London (Catalan, Pillinger, McGuire, McCutcheon); Basurto University Hospital, OSI Bilbao-Basurto, Biobizkaia Health Research Institute, University of the Basque Country UPV/EHU, Centro de Investigación en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Bilbao, Spain (Catalan); South London and Maudsley NHS Foundation Trust, London (Pillinger)
| |
Collapse
|
8
|
Horvath G, Ducza E, Adlan LG, Büki A, Kekesi G. Distinct Effects of Olanzapine Depot Treatment on Behavior and Muscarinic M1 Receptor Expression in the Triple-Hit Wisket Rat Model of Schizophrenia. GENES, BRAIN, AND BEHAVIOR 2025; 24:e70015. [PMID: 39844699 PMCID: PMC11754962 DOI: 10.1111/gbb.70015] [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: 10/15/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/24/2025]
Abstract
This study aimed to characterize the triple-hit schizophrenia-like model rats (Wisket) by the assessment of (1) behavioral parameters in different test conditions (reward-based Ambitus test and HomeManner system) for a prolonged period, (2) cerebral muscarinic M1 receptor (M1R) expression, and (3) the effects of olanzapine treatment on these parameters. Wistar (control) and Wisket rats were injected for three consecutive weeks with olanzapine depot (100 mg/kg) and spent 4 weeks in large cages with environmental enrichment (HomeManner). The vehicle-treated Wisket rats spent longer time awake with decreased grooming activity compared to controls, without changes in their active social behavior (sniffing, playing, fighting) obtained in HomeManner. Olanzapine treatment decreased most of these parameters, only the passive social interaction (huddling during sleeping) enhanced mostly in the Wisket rats on the injection day, which recovered within 4 days. In the Ambitus test, vehicle-treated Wisket rats showed lower locomotor and exploratory activities and impaired cognition compared to control rats, deteriorating by olanzapine in both groups. In Wisket brain samples, the M1R mRNA expression was significantly lower in the cerebral cortex and elevated in the hippocampus, with no difference in the prefrontal cortex versus control. Olanzapine normalized the hippocampal M1R expression, but enhanced it in the prefrontal cortex. The triple-hit Wisket model rats had impaired behavioral characteristics in both acute reward-based test and undisturbed circumstances investigated for prolonged periods, and altered cerebral M1R expression. Chronic olanzapine treatment resulted deterioration of some parameters in control group, and could restore only few negative signs in model rats.
Collapse
Affiliation(s)
- Gyongyi Horvath
- Department of PhysiologyAlbert Szent‐Györgyi Medical School, University of SzegedSzegedHungary
| | - Eszter Ducza
- Department of Pharmacodynamics and Biopharmacy, Faculty of PharmacyUniversity of SzegedSzegedHungary
| | | | - Alexandra Büki
- Department of PhysiologyAlbert Szent‐Györgyi Medical School, University of SzegedSzegedHungary
| | - Gabriella Kekesi
- Department of PhysiologyAlbert Szent‐Györgyi Medical School, University of SzegedSzegedHungary
| |
Collapse
|
9
|
Incontro S, Musella ML, Sammari M, Di Scala C, Fantini J, Debanne D. Lipids shape brain function through ion channel and receptor modulations: physiological mechanisms and clinical perspectives. Physiol Rev 2025; 105:137-207. [PMID: 38990068 DOI: 10.1152/physrev.00004.2024] [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: 01/16/2024] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Lipids represent the most abundant molecular type in the brain, with a fat content of ∼60% of the dry brain weight in humans. Despite this fact, little attention has been paid to circumscribe the dynamic role of lipids in brain function and disease. Membrane lipids such as cholesterol, phosphoinositide, sphingolipids, arachidonic acid, and endocannabinoids finely regulate both synaptic receptors and ion channels that ensure critical neural functions. After a brief introduction on brain lipids and their respective properties, we review here their role in regulating synaptic function and ion channel activity, action potential propagation, neuronal development, and functional plasticity and their contribution in the development of neurological and neuropsychiatric diseases. We also provide possible directions for future research on lipid function in brain plasticity and diseases.
Collapse
Affiliation(s)
| | | | - Malika Sammari
- UNIS, INSERM, Aix-Marseille Université, Marseille, France
| | | | | | | |
Collapse
|
10
|
Marques SI, Sá SI, Carmo H, Carvalho F, Silva JP. Pharmaceutical-mediated neuroimmune modulation in psychiatric/psychological adverse events. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111114. [PMID: 39111563 DOI: 10.1016/j.pnpbp.2024.111114] [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: 05/10/2024] [Revised: 07/21/2024] [Accepted: 08/03/2024] [Indexed: 08/13/2024]
Abstract
The therapeutic use of many pharmaceuticals, including small molecules and biological therapies, has been associated with the onset of psychiatric and psychological adverse events (PPAEs), posing substantial concerns to patients' health and safety. These events, which encompass mood (e.g., depression, schizophrenia, suicidal ideation) and cognitive changes (e.g., learning and memory impairment, dementia) often remain undetected until advanced stages of clinical trials or pharmacovigilance, mostly because the mechanisms underlying the onset of PPAEs remain poorly understood. In recent years, the role of neuroimmune modulation (comprising an intricate interplay between various cell types and signaling pathways) in PPAEs has garnered substantial interest. Indeed, understanding these complex interactions would substantially contribute to increase the ability to predict the potential onset of PPAEs during preclinical stages of a new drug's R&D. This review provides a comprehensive summary of the most recent advances in neuroimmune modulation-related mechanisms contributing to the onset of PPAEs and their association with specific pharmaceuticals. Reported data strongly support an association between neuroimmune modulation and the onset of PPAEs. Pharmaceuticals may target specific molecular pathways and pathway elements (e.g., cholinergic and serotonergic systems), which in turn may directly or indirectly impact the inflammatory status and the homeostasis of the brain, regulating inflammation and neuronal function. Also, modulation of the peripheral immune system by pharmaceuticals that do not permeate the blood-brain barrier (e.g., monoclonal antibodies) may alter the neuroimmunomodulatory status of the brain, leading to PPAEs. In summary, this review underscores the diverse pathways through which drugs can influence brain inflammation, shedding light on potential targeted interventions.
Collapse
Affiliation(s)
- Sandra I Marques
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Susana I Sá
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.
| | - Helena Carmo
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Félix Carvalho
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - João P Silva
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
11
|
Dean B. IUPHAR Review on muscarinic M1 and M4 receptors as drug treatment targets relevant to the molecular pathology of schizophrenia. Pharmacol Res 2024; 210:107510. [PMID: 39566671 DOI: 10.1016/j.phrs.2024.107510] [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: 10/28/2024] [Revised: 11/13/2024] [Accepted: 11/16/2024] [Indexed: 11/22/2024]
Abstract
Cobenfy, a co-formulation of xanomeline and trospium, is the first drug not acting on the dopaminergic system of the CNS approved for the treatment of schizophrenia by the FDA. Xanomeline is a muscarinic M1 and M4 receptor (CHRM1 and CHRM4) agonist whilst trospium is a peripherally active CHRM antagonist that reduces the unwanted peripheral side-effects of xanomeline. Relevant to this exciting development, this review details the human CNS cholinergic systems and how those systems are affected by the molecular pathology of schizophrenia in a way suggesting activating the CHRM1 and 4 would be beneficial in treating the disorder. The CNS distribution of CHRMs is presented along with findings using CHRM knockout mice and mice treated with drugs that activate the CHRM1 and / or M4, these data explain why these CHRMs could be involved in the genesis of the symptoms of schizophrenia. Next, the process leading to the formulation of Cobenfy and the preclinical data on xanomeline are reviewed showing why Cobenfy was expected to be useful in treating schizophrenia. The pipeline of drugs targeting CHRM1 and /or M4 receptors to treat schizophrenia are discussed. Finally, the molecular pathology of two sub-groups within schizophrenia, separated based on the presence or absence of a deficit of cortical CHRM1, are reviewed to show how such approaches could identify new drug targets. In conclusion, the history of the development of Cobenfy highlights how a growing understanding the pathophysiology of schizophrenia will suggest new treatment targets for the disorder and that pharmacologists can synthesise drugs to target these sites.
Collapse
Affiliation(s)
- Brian Dean
- The Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia.
| |
Collapse
|
12
|
Moyano P, Guzmán G, Flores A, García J, Guerra-Menéndez L, Sanjuan J, Plaza JC, Abascal L, Mateo O, Del Pino J. Thyroid Hormone Neuroprotection Against Perfluorooctane Sulfonic Acid Cholinergic and Glutamatergic Disruption and Neurodegeneration Induction. Biomedicines 2024; 12:2441. [PMID: 39595009 PMCID: PMC11591898 DOI: 10.3390/biomedicines12112441] [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: 09/22/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Perfluorooctane sulfonic acid (PFOS), a widely used industrial chemical, was reported to induce memory and learning process dysfunction. Some studies tried to reveal the mechanisms that mediate these effects, but how they are produced is still unknown. Basal forebrain cholinergic neurons (BFCN) maintain cognitive function and their selective neurodegeneration induces cognitive decline, as observed in Alzheimer's disease. PFOS was reported to disrupt cholinergic and glutamatergic transmissions and thyroid hormone action, which regulate cognitive processes and maintain BFCN viability. Objective/Methods: To evaluate PFOS neurodegenerative effects on BFCN and the mechanisms that mediate them, SN56 cells (a neuroblastoma cholinergic cell line from the basal forebrain) were treated with PFOS (0.1 µM to 40 µM) with or without thyroxine (T3; 15 nM), MK-801 (20 µM) or acetylcholine (ACh; 10 µM). Results: In the present study, we found that PFOS treatment (1 or 14 days) decreased thyroid receptor α (TRα) activity by decreasing its protein levels and increased T3 metabolism through increased deiodinase 3 (D3) levels. Further, we observed that PFOS treatment disrupted cholinergic transmission by decreasing ACh content through decreased choline acetyltransferase (ChAT) activity and protein levels and through decreasing muscarinic receptor 1 (M1R) binding and protein levels. PFOS also disrupted glutamatergic transmission by decreasing glutamate content through increased glutaminase activity and protein levels and through decreasing N-methyl-D-aspartate receptor subunit 1 (NMDAR1); effects mediated through M1R disruption. All these effects were mediated through decreased T3 activity and T3 supplementation partially restored to the normal state. Conclusions: These findings may assist in understanding how PFOS induces neurodegeneration, and the mechanisms involved, especially in BFCN, to explain the process that could lead to cognitive dysfunction and provide new therapeutic tools to treat and prevent its neurotoxic effects.
Collapse
Affiliation(s)
- Paula Moyano
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Gabriela Guzmán
- Departamento de Ciencias Meìdicas Baìsicas, Facultad de Medicina, Universidad San Pablo-CEU, Urbanizacioìn Montepriìncipe, 28660 Boadilla del Monte, Spain
| | - Andrea Flores
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Jimena García
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Lucia Guerra-Menéndez
- Departamento de Ciencias Meìdicas Baìsicas, Facultad de Medicina, Universidad San Pablo-CEU, Urbanizacioìn Montepriìncipe, 28660 Boadilla del Monte, Spain
| | - Javier Sanjuan
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - José Carlos Plaza
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Luisa Abascal
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Olga Mateo
- Department of Surgery, Medicine School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Javier Del Pino
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
13
|
Yohn SE, Harvey PD, Brannan SK, Horan WP. The potential of muscarinic M 1 and M 4 receptor activators for the treatment of cognitive impairment associated with schizophrenia. Front Psychiatry 2024; 15:1421554. [PMID: 39483736 PMCID: PMC11525114 DOI: 10.3389/fpsyt.2024.1421554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/03/2024] [Indexed: 11/03/2024] Open
Abstract
Cognitive impairment is a core symptom of schizophrenia and a major determinant of poor long-term functional outcomes. Despite considerable efforts, we do not yet have any approved pharmacological treatments for cognitive impairment associated with schizophrenia (CIAS). A combination of advances in pre-clinical research and recent clinical trial findings have led to a resurgence of interest in the cognition-enhancing potential of novel muscarinic acetylcholine receptor (mAChR) agonists in schizophrenia. This article provides an overview of the scientific rationale for targeting M1 and M4 mAChRs. We describe the evolution of neuroscience research on these receptors since early drug discovery efforts focused on the mAChR agonist xanomeline. This work has revealed that M1 and M4 mAChRs are highly expressed in brain regions that are implicated in cognition. The functional significance of M1 and M4 mAChRs has been extensively characterized in animal models via use of selective receptor subtype compounds through neuronal and non-neuronal mechanisms. Recent clinical trials of a dual M1/M4 mAChR agonist show promising, replicable evidence of potential pro-cognitive effects in schizophrenia, with several other mAChR agonists in clinical development.
Collapse
Affiliation(s)
| | - Phillip D. Harvey
- Division of Psychology, University of Miami, Miami, FL, United States
| | | | - William P. Horan
- Bristol Myers Squibb, Princeton, NJ, United States
- Department of Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
14
|
Tobin AB. A golden age of muscarinic acetylcholine receptor modulation in neurological diseases. Nat Rev Drug Discov 2024; 23:743-758. [PMID: 39143241 DOI: 10.1038/s41573-024-01007-1] [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] [Accepted: 06/28/2024] [Indexed: 08/16/2024]
Abstract
Over the past 40 years, the muscarinic acetylcholine receptor family, particularly the M1-receptor and M4-receptor subtypes, have emerged as validated targets for the symptomatic treatment of neurological diseases such as schizophrenia and Alzheimer disease. However, despite considerable effort and investment, no drugs have yet gained clinical approval. This is largely attributable to cholinergic adverse effects that have halted the majority of programmes and resulted in a waning of interest in these G-protein-coupled receptor targets. Recently, this trend has been reversed. Driven by advances in structure-based drug design and an appreciation of the optimal pharmacological properties necessary to deliver clinical efficacy while minimizing adverse effects, a new generation of M1-receptor and M4-receptor orthosteric agonists and positive allosteric modulators are now entering the clinic. These agents offer the prospect of novel therapeutic solutions for 'hard to treat' neurological diseases, heralding a new era of muscarinic drug discovery.
Collapse
Affiliation(s)
- Andrew B Tobin
- Centre for Translational Pharmacology, School of Molecular Biosciences, The Advanced Research Centre, University of Glasgow, Glasgow, UK.
| |
Collapse
|
15
|
Poon MM, Lorrain KI, Stebbins KJ, Edu GC, Broadhead AR, Lorenzana AJ, Roppe JR, Baccei JM, Baccei CS, Chen AC, Green AJ, Lorrain DS, Chan JR. Targeting the muscarinic M1 receptor with a selective, brain-penetrant antagonist to promote remyelination in multiple sclerosis. Proc Natl Acad Sci U S A 2024; 121:e2407974121. [PMID: 39083422 PMCID: PMC11317586 DOI: 10.1073/pnas.2407974121] [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: 04/22/2024] [Accepted: 06/25/2024] [Indexed: 08/02/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic and debilitating neurological disease that results in inflammatory demyelination. While endogenous remyelination helps to recover function, this restorative process tends to become less efficient over time. Currently, intense efforts aimed at the mechanisms that promote remyelination are being considered promising therapeutic approaches. The M1 muscarinic acetylcholine receptor (M1R) was previously identified as a negative regulator of oligodendrocyte differentiation and myelination. Here, we validate M1R as a target for remyelination by characterizing expression in human and rodent oligodendroglial cells (including those in human MS tissue) using a highly selective M1R probe. As a breakthrough to conventional methodology, we conjugated a fluorophore to a highly M1R selective peptide (MT7) which targets the M1R in the subnanomolar range. This allows for exceptional detection of M1R protein expression in the human CNS. More importantly, we introduce PIPE-307, a brain-penetrant, small-molecule antagonist with favorable drug-like properties that selectively targets M1R. We evaluate PIPE-307 in a series of in vitro and in vivo studies to characterize potency and selectivity for M1R over M2-5R and confirm the sufficiency of blocking this receptor to promote differentiation and remyelination. Further, PIPE-307 displays significant efficacy in the mouse experimental autoimmune encephalomyelitis model of MS as evaluated by quantifying disability, histology, electron microscopy, and visual evoked potentials. Together, these findings support targeting M1R for remyelination and support further development of PIPE-307 for clinical studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Ari J. Green
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA94158
| | | | - Jonah R. Chan
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA94158
| |
Collapse
|
16
|
Watamura N, Kakiya N, Fujioka R, Kamano N, Takahashi M, Nilsson P, Saito T, Iwata N, Fujisawa S, Saido TC. The dopaminergic system promotes neprilysin-mediated degradation of amyloid-β in the brain. Sci Signal 2024; 17:eadk1822. [PMID: 39106321 DOI: 10.1126/scisignal.adk1822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 04/21/2024] [Accepted: 07/01/2024] [Indexed: 08/09/2024]
Abstract
Deposition of amyloid-β (Aβ) in the brain can impair neuronal function and contribute to cognitive decline in Alzheimer's disease (AD). Here, we found that dopamine and the dopamine precursor levodopa (also called l-DOPA) induced Aβ degradation in the brain. Chemogenetic approaches in mice revealed that the activation of dopamine release from ventral tegmental area (VTA) neurons increased the abundance and activity of the Aβ-degrading enzyme neprilysin and reduced the amount of Aβ deposits in the prefrontal cortex in a neprilysin-dependent manner. Aged mice had less dopamine and neprilysin in the anterior cortex, a decrease that was accentuated in AD model mice. Treating AD model mice with levodopa reduced Aβ deposition and improved cognitive function. These observations demonstrate that dopamine promotes brain region-specific, neprilysin-dependent degradation of Aβ, suggesting that dopamine-associated strategies have the potential to treat this aspect of AD pathology.
Collapse
Affiliation(s)
- Naoto Watamura
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Naomasa Kakiya
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Ryo Fujioka
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Naoko Kamano
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Mika Takahashi
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division for Neurogeriatrics, Karolinska Institutet, 171 64, Solna, Sweden
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi 464-8601, Japan
| | - Nobuhisa Iwata
- Department of Genome-based Drug Discovery & Leading Medical Research Core Unit, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8521, Japan
| | - Shigeyoshi Fujisawa
- Laboratory for Systems Neurophysiology, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
17
|
Lee J, Jeong Y, Park S, Kim S, Oh H, Jin JA, Sohn JW, Kim D, Shin HS, Do Heo W. Phospholipase C beta 1 in the dentate gyrus gates fear memory formation through regulation of neuronal excitability. SCIENCE ADVANCES 2024; 10:eadj4433. [PMID: 38959322 PMCID: PMC11221510 DOI: 10.1126/sciadv.adj4433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 05/28/2024] [Indexed: 07/05/2024]
Abstract
Memory processes rely on a molecular signaling system that balances the interplay between positive and negative modulators. Recent research has focused on identifying memory-regulating genes and their mechanisms. Phospholipase C beta 1 (PLCβ1), highly expressed in the hippocampus, reportedly serves as a convergence point for signal transduction through G protein-coupled receptors. However, the detailed role of PLCβ1 in memory function has not been elucidated. Here, we demonstrate that PLCβ1 in the dentate gyrus functions as a memory suppressor. We reveal that mice lacking PLCβ1 in the dentate gyrus exhibit a heightened fear response and impaired memory extinction, and this excessive fear response is repressed by upregulation of PLCβ1 through its overexpression or activation using a newly developed optogenetic system. Last, our results demonstrate that PLCβ1 overexpression partially inhibits exaggerated fear response caused by traumatic experience. Together, PLCβ1 is crucial in regulating contextual fear memory formation and potentially enhancing the resilience to trauma-related conditions.
Collapse
Affiliation(s)
- Jinsu Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Yeonji Jeong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Seahyung Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Sungsoo Kim
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Hyunsik Oh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Ju-Ae Jin
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jong-Woo Sohn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Daesoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Department of Brain and Cognitive Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Hee-Sup Shin
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34141, Korea
| | - Won Do Heo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Department of Brain and Cognitive Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- KAIST Institute for the BioCentury (KIB), Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
Widman CJ, Ventresca S, Dietrich J, Elmslie G, Smith H, Kaup G, Wesley A, Doenecke M, Williams FE, Schiefer IT, Ellis J, Messer WS. Allosteric modulators of M 1 muscarinic receptors enhance acetylcholine efficacy and decrease locomotor activity and turning behaviors in zebrafish. Sci Rep 2024; 14:14901. [PMID: 38942828 PMCID: PMC11213934 DOI: 10.1038/s41598-024-65445-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: 01/26/2024] [Accepted: 06/20/2024] [Indexed: 06/30/2024] Open
Abstract
Allosteric modulation of muscarinic acetylcholine receptors (mAChR) has been identified as a potential strategy for regulating cholinergic signaling in the treatment of various neurological disorders. Most positive allosteric modulators (PAMs) of mAChR enhance agonist affinity and potency, while very few PAMs (e.g., amiodarone) selectively enhance G protein coupling efficacy. The key structural features of amiodarone responsible for enhancement of mAChR efficacy were examined in CHO cells expressing M1 receptors. Subsequent incorporation of these structural features into previously identified allosteric modulators of potency (i.e., n-benzyl isatins) generated ligands that demonstrated similar or better enhancement of mAChR efficacy, lower in vivo toxicity, and higher allosteric binding affinity relative to amiodarone. Notable ligands include 8a, c which respectively demonstrated the strongest binding affinity and the most robust enhancement of mAChR efficacy as calculated from an allosteric operational model. Amiodarone derivatives and hybrid ligands were additionally screened in wildtype zebrafish (Danio rerio) to provide preliminary in vivo toxicity data as well as to observe effects on locomotor and turning behaviors relative to other mAChR PAMs. Several compounds, including 8a, c, reduced locomotor activity and increased measures of turning behaviors in zebrafish, suggesting that allosteric modulation of muscarinic receptor efficacy might be useful in the treatment of repetitive behaviors associated with autism spectrum disorder (ASD) and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Corey J Widman
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Mail Stop #1015, 3000 Arlington Ave., Toledo, OH, 43614, USA
| | - Sestina Ventresca
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43614, USA
| | - Jillian Dietrich
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43614, USA
| | - Gwendolynne Elmslie
- Departments of Psychiatry and Pharmacology, College of Medicine, Penn State University, Hershey, PA, 17033, USA
| | - Hazel Smith
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Mail Stop #1015, 3000 Arlington Ave., Toledo, OH, 43614, USA
| | - Gina Kaup
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Mail Stop #1015, 3000 Arlington Ave., Toledo, OH, 43614, USA
| | - Aaron Wesley
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43614, USA
| | - Madeline Doenecke
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43614, USA
| | - Frederick E Williams
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43614, USA
| | - Isaac T Schiefer
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Mail Stop #1015, 3000 Arlington Ave., Toledo, OH, 43614, USA
- Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43614, USA
| | - John Ellis
- Departments of Psychiatry and Pharmacology, College of Medicine, Penn State University, Hershey, PA, 17033, USA
| | - William S Messer
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Mail Stop #1015, 3000 Arlington Ave., Toledo, OH, 43614, USA.
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43614, USA.
| |
Collapse
|
21
|
Tamura H, Miyazaki A, Kawamura T, Gotoh H, Yamamoto N, Narita M. Chronic ingestion of soy peptide supplementation reduces aggressive behavior and abnormal fear memory caused by juvenile social isolation. Sci Rep 2024; 14:11557. [PMID: 38773352 PMCID: PMC11109177 DOI: 10.1038/s41598-024-62534-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/17/2024] [Indexed: 05/23/2024] Open
Abstract
Juvenile loneliness is a risk factor for psychopathology in later life. Deprivation of early social experience due to peer rejection has a detrimental impact on emotional and cognitive brain function in adulthood. Accumulating evidence indicates that soy peptides have many positive effects on higher brain function in rodents and humans. However, the effects of soy peptide use on juvenile social isolation are unknown. Here, we demonstrated that soy peptides reduced the deterioration of behavioral and cellular functions resulting from juvenile socially-isolated rearing. We found that prolonged social isolation post-weaning in male C57BL/6J mice resulted in higher aggression and impulsivity and fear memory deficits at 7 weeks of age, and that these behavioral abnormalities, except impulsivity, were mitigated by ingestion of soy peptides. Furthermore, we found that daily intake of soy peptides caused upregulation of postsynaptic density 95 in the medial prefrontal cortex and phosphorylation of the cyclic adenosine monophosphate response element binding protein in the hippocampus of socially isolated mice, increased phosphorylation of the adenosine monophosphate-activated protein kinase in the hippocampus, and altered the microbiota composition. These results suggest that soy peptides have protective effects against juvenile social isolation-induced behavioral deficits via synaptic maturation and cellular functionalization.
Collapse
Affiliation(s)
- Hideki Tamura
- Laboratory of Biofunctional Science, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa, Tokyo, 142-8501, Japan.
- Institute for Advanced Life Sciences, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan.
| | - Akiko Miyazaki
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan
| | - Takashi Kawamura
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan
| | - Hikaru Gotoh
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan
| | - Naoki Yamamoto
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, 819-0395, Japan
| | - Minoru Narita
- Institute for Advanced Life Sciences, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan
- Department of Pharmacy, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
22
|
Reza ASMA, Raihan R, Azam S, Shahanewz M, Nasrin MS, Siddique MAB, Uddin MN, Dey AK, Sadik MG, Alam AK. Experimental and pharmacoinformatic approaches unveil the neuropharmacological and analgesic potential of chloroform fraction of Roktoshirinchi (Achyranthes ferruginea Roxb.). JOURNAL OF ETHNOPHARMACOLOGY 2024; 324:117769. [PMID: 38219886 DOI: 10.1016/j.jep.2024.117769] [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: 10/29/2023] [Revised: 01/07/2024] [Accepted: 01/11/2024] [Indexed: 01/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Achyranthes ferruginea (A. ferruginea) Roxb. is a common plant used in traditional medicine in Asia and Africa. It has a variety of local names, including "Gulmanci" in Nigeria, "Dangar" in Pakistan, "Thola" in Ethiopia, and "Roktoshirinchi" in Bangladesh. It is edible and has several ethnomedical uses for a wide range of illnesses, including hysteria, dropsy, constipation, piles, boils, asthma, and shigellosis. However, the neuropharmacological and analgesic potential of A. ferruginea remains uninvestigated. AIM OF THE STUDY To assess the neuropharmacological and analgesic potential of A. ferruginea through a multifaceted approach encompassing both experimental and computational models. MATERIALS AND METHODS Methanol was used to extract the leaves of A. ferruginea. It was then fractionated with low to high polar solvents (n-hexane, chloroform, ethyl acetate, and water) to get different fractions, including chloroform fraction (CLF). The study selected CLF at different doses and conducted advanced chemical element and proximate analyses, as well as phytochemical profiling using GC-MS. Toxicological studies were done at 300 μg per rat per day for 14 days. Cholinesterase inhibitory potential was checked using an in-vitro colorimetric assay. Acetic acid-induced writhing (AAWT) and formalin-induced licking tests (FILT) were used to assess anti-nociceptive effects. The forced swim test (FST), tail suspension test (TST), elevated plus maze (EPM), hole board test (HBT), and light and dark box test (LDB) were among the behavioral tests used to assess depression and anxiolytic activity. Network pharmacology-based analysis was performed on selected compounds using the search tool for interacting chemicals-5 (STITCH 5), Swiss target prediction tool, and search tool for the retrieval of interacting genes and proteins (STRING) database to link their role with genes involved in neurological disorders through gene ontology and reactome analysis. RESULTS Qualitative chemical element analysis revealed the presence of 15 elements, including Na, K, Ca, Mg, P, and Zn. The moisture content, ash value, and organic matter were found to be 11.12, 11.03, and 88.97%, respectively. GC-MS data revealed that the CLF possesses 25 phytoconstituents. Toxicological studies suggested the CLF has no effects on normal growth, hematological and biochemical parameters, or cellular organs after 14 days at 300 μg per rat. The CLF markedly reduced the activity of both acetylcholinesterase and butyrylcholinesterase (IC50: 56.22 and 13.22 μg/mL, respectively). Promising dose-dependent analgesic activity (p < 0.05) was observed in chemically-induced pain models. The TST and FST showed a dose-dependent substantial reduction in immobility time due to the CLF. Treatment with CLF notably increased the number of open arm entries and time spent in the EPM test at doses of 200 and 400 mg/kg b.w. The CLF showed significant anxiolytic activity at 200 mg/kg b.w. in the HBT test, whereas a similar activity was observed at 400 mg/kg b.w. in the EPM test. A notable increase in the amount of time spent in the light compartment was observed in the LDB test by mice treated with CLF, suggesting an anxiolytic effect. A network pharmacology study demonstrated the relationship between the phytochemicals and a number of targets, such as PPARA, PPARG, CHRM1, and HTR2, which are connected to the shown bioactivities. CONCLUSIONS This study demonstrated the safety of A. ferruginea and its efficacy in attenuating cholinesterase inhibitory activity, central and peripheral pain, anxiety, and depression, warranting further exploration of its therapeutic potential.
Collapse
Affiliation(s)
- A S M Ali Reza
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, 4318, Bangladesh.
| | - Riaj Raihan
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, 4318, Bangladesh.
| | - Saidul Azam
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, 4318, Bangladesh.
| | - Mohammed Shahanewz
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, 4318, Bangladesh.
| | - Mst Samima Nasrin
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, 4318, Bangladesh.
| | - Md Abu Bakar Siddique
- Institute of National Analytical Research and Service (INARS), Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhanmondi, Dhaka, 1205, Bangladesh.
| | - Md Nazim Uddin
- Institute of Food Science and Technology (IFST), Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhanmondi, Dhaka, 1205, Bangladesh.
| | - Anik Kumar Dey
- Department of Pharmacy, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| | - Md Golam Sadik
- Department of Pharmacy, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| | - Ahm Khurshid Alam
- Department of Pharmacy, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| |
Collapse
|
23
|
Widman CJ, Ventresca S, Dietrich J, Elmslie G, Smith H, Kaup G, Wesley A, Doenecke M, Williams FE, Schiefer IT, Ellis J, Messer WS. Hybrid Allosteric Modulators of M1 Muscarinic Receptors Enhance Acetylcholine Efficacy and Decrease Locomotor Activity and Turning Behaviors in Zebrafish. RESEARCH SQUARE 2024:rs.3.rs-3901189. [PMID: 38410427 PMCID: PMC10896388 DOI: 10.21203/rs.3.rs-3901189/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Allosteric modulation of muscarinic acetylcholine receptors (mAChR) has been identified as a potential strategy for regulating cholinergic signaling in the treatment of various neurological disorders. Most positive allosteric modulators (PAMs) of mAChR enhance agonist affinity and potency, while very few PAMs selectively enhance G-protein coupling efficacy (e.g., amiodarone). The key structural features of amiodarone responsible for enhancement of mAChR efficacy were examined in CHO cells expressing M1 receptors. Subsequent incorporation of these structural features into previously identified allosteric modulators of potency (i.e., n-benzyl isatins) generated hybrid ligands that demonstrated similar or better enhancement of mAChR efficacy, lower in vivo toxicity, and higher allosteric binding affinity relative to amiodarone. Notable hybrid ligands include 8a and 8b which respectively demonstrated the strongest binding affinity and the most robust enhancement of mAChR efficacy as calculated from an allosteric operational model. Amiodarone derivatives and hybrid ligands were additionally screened in wildtype zebrafish (Danio rerio) to provide preliminary in vivo toxicity data as well as to observe effects on locomotor and turning behaviors relative to other mAChR PAMs. Several compounds, including 8a and 8c, reduced locomotor activity and increased measures of turning behaviors in zebrafish, suggesting that allosteric modulation of muscarinic receptor efficacy might be useful in the treatment of repetitive behaviors associated with autism spectrum disorder (ASD) and other neuropsychiatric disorders.
Collapse
|
24
|
Palma JA. Muscarinic control of cardiovascular function in humans: a review of current clinical evidence. Clin Auton Res 2024; 34:31-44. [PMID: 38305989 PMCID: PMC10994193 DOI: 10.1007/s10286-024-01016-5] [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: 10/10/2023] [Accepted: 01/11/2024] [Indexed: 02/03/2024]
Abstract
PURPOSE To review the available evidence on the impact of muscarinic receptor modulation on cardiovascular control in humans. METHODS In this narrative Review we summarize data on cardiovascular endpoints from clinical trials of novel subtype-selective or quasi-selective muscarinic modulators, mostly PAMs, performed in the last decade. We also review the cardiovascular phenotype in recently described human genetic and autoimmune disorders affecting muscarinic receptors. RESULTS Recent advancements in the development of compounds that selectively target muscarinic acetylcholine receptors are expanding our knowledge about the physiological function of each muscarinic receptor subtype (M1, M2, M3, M4, M5). Among these novel compounds, positive allosteric modulators (PAMs) have emerged as the preferred therapeutic to regulate muscarinic receptor subtype function. Many muscarinic allosteric and orthosteric modulators (including but not limited to xanomeline-trospium and emraclidine) are now in clinical development and approaching regulatory approval for multiple indications, including the treatment of cognitive and psychiatric symptoms in patients with schizophrenia as well as Alzheimer's disease and other dementias. The results of these clinical trials provide an opportunity to understand the influence of muscarinic modulation on cardiovascular autonomic control in humans. While the results and the impact of each of these therapies on heart rate and blood pressure control have been variable, in part because the clinical trials were not specifically designed to measure cardiovascular endpoints, the emerging data is valuable to elucidate the relative cardiovascular contributions of each muscarinic receptor subtype. CONCLUSION Understanding the muscarinic control of cardiovascular function is of paramount importance and may contribute to the development of novel therapeutic strategies for treating cardiovascular disease.
Collapse
Affiliation(s)
- Jose-Alberto Palma
- Department of Neurology, NYU Dysautonomia Center, New York University School of Medicine, 530 First Av, Suite 9Q, New York, 10016, USA.
| |
Collapse
|
25
|
Nimgampalle M, Chakravarthy H, Sharma S, Shree S, Bhat AR, Pradeepkiran JA, Devanathan V. Neurotransmitter systems in the etiology of major neurological disorders: Emerging insights and therapeutic implications. Ageing Res Rev 2023; 89:101994. [PMID: 37385351 DOI: 10.1016/j.arr.2023.101994] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/21/2023] [Accepted: 06/25/2023] [Indexed: 07/01/2023]
Abstract
Neurotransmitters serve as chemical messengers playing a crucial role in information processing throughout the nervous system, and are essential for healthy physiological and behavioural functions in the body. Neurotransmitter systems are classified as cholinergic, glutamatergic, GABAergic, dopaminergic, serotonergic, histaminergic, or aminergic systems, depending on the type of neurotransmitter secreted by the neuron, allowing effector organs to carry out specific functions by sending nerve impulses. Dysregulation of a neurotransmitter system is typically linked to a specific neurological disorder. However, more recent research points to a distinct pathogenic role for each neurotransmitter system in more than one neurological disorder of the central nervous system. In this context, the review provides recently updated information on each neurotransmitter system, including the pathways involved in their biochemical synthesis and regulation, their physiological functions, pathogenic roles in diseases, current diagnostics, new therapeutic targets, and the currently used drugs for associated neurological disorders. Finally, a brief overview of the recent developments in neurotransmitter-based therapeutics for selected neurological disorders is offered, followed by future perspectives in that area of research.
Collapse
Affiliation(s)
- Mallikarjuna Nimgampalle
- Department of Biology, Indian Institute of Science Education and Research Tirupati (IISER T), Transit campus, Karakambadi Road, Mangalam, Tirupati 517507, Andhra Pradesh, India
| | - Harshini Chakravarthy
- Department of Biology, Indian Institute of Science Education and Research Tirupati (IISER T), Transit campus, Karakambadi Road, Mangalam, Tirupati 517507, Andhra Pradesh, India.
| | - Sapana Sharma
- Department of Biology, Indian Institute of Science Education and Research Tirupati (IISER T), Transit campus, Karakambadi Road, Mangalam, Tirupati 517507, Andhra Pradesh, India
| | - Shruti Shree
- Department of Biology, Indian Institute of Science Education and Research Tirupati (IISER T), Transit campus, Karakambadi Road, Mangalam, Tirupati 517507, Andhra Pradesh, India
| | - Anoop Ramachandra Bhat
- Department of Biology, Indian Institute of Science Education and Research Tirupati (IISER T), Transit campus, Karakambadi Road, Mangalam, Tirupati 517507, Andhra Pradesh, India
| | | | - Vasudharani Devanathan
- Department of Biology, Indian Institute of Science Education and Research Tirupati (IISER T), Transit campus, Karakambadi Road, Mangalam, Tirupati 517507, Andhra Pradesh, India.
| |
Collapse
|
26
|
Zheng F, Wess J, Alzheimer C. Long-Term-But Not Short-Term-Plasticity at the Mossy Fiber-CA3 Pyramidal Cell Synapse in Hippocampus Is Altered in M1/M3 Muscarinic Acetylcholine Receptor Double Knockout Mice. Cells 2023; 12:1890. [PMID: 37508553 PMCID: PMC10378318 DOI: 10.3390/cells12141890] [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: 06/13/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Muscarinic acetylcholine receptors are well-known for their crucial involvement in hippocampus-dependent learning and memory, but the exact roles of the various receptor subtypes (M1-M5) are still not fully understood. Here, we studied how M1 and M3 receptors affect plasticity at the mossy fiber (MF)-CA3 pyramidal cell synapse. In hippocampal slices from M1/M3 receptor double knockout (M1/M3-dKO) mice, the signature short-term plasticity of the MF-CA3 synapse was not significantly affected. However, the rather unique NMDA receptor-independent and presynaptic form of long-term potentiation (LTP) of this synapse was much larger in M1/M3-deficient slices compared to wild-type slices in both field potential and whole-cell recordings. Consistent with its presynaptic origin, induction of MF-LTP strongly enhanced the excitatory drive onto single CA3 pyramidal cells, with the effect being more pronounced in M1/M3-dKO cells. In an earlier study, we found that the deletion of M2 receptors in mice disinhibits MF-LTP in a similar fashion, suggesting that endogenous acetylcholine employs both M1/M3 and M2 receptors to constrain MF-LTP. Importantly, such synergism was not observed for MF long-term depression (LTD). Low-frequency stimulation, which reliably induced LTD of MF synapses in control slices, failed to do so in M1/M3-dKO slices and gave rise to LTP instead. In striking contrast, loss of M2 receptors augmented LTD when compared to control slices. Taken together, our data demonstrate convergence of M1/M3 and M2 receptors on MF-LTP, but functional divergence on MF-LTD, with the net effect resulting in a well-balanced bidirectional plasticity of the MF-CA3 pyramidal cell synapse.
Collapse
Affiliation(s)
- Fang Zheng
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Biological Chemistry, NIDDK, NIH, Bethesda, MD 20892, USA
| | - Christian Alzheimer
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
27
|
Sabbir MG, Swanson M, Speth RC, Albensi BC. Hippocampal versus cortical deletion of cholinergic receptor muscarinic 1 in mice differentially affects post-translational modifications and supramolecular assembly of respiratory chain-associated proteins, mitochondrial ultrastructure, and respiration: implications in Alzheimer's disease. Front Cell Dev Biol 2023; 11:1179252. [PMID: 37293125 PMCID: PMC10246746 DOI: 10.3389/fcell.2023.1179252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/02/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction: In a previous retrospective study using postmortem human brain tissues, we demonstrated that loss of Cholinergic Receptor Muscarinic 1 (CHRM1) in the temporal cortex of a subset of Alzheimer's patients was associated with poor survival, whereas similar loss in the hippocampus showed no such association. Mitochondrial dysfunction underlies Alzheimer's pathogenesis. Therefore, to investigate the mechanistic basis of our findings, we evaluated cortical mitochondrial phenotypes in Chrm1 knockout (Chrm1-/-) mice. Cortical Chrm1 loss resulted in reduced respiration, reduced supramolecular assembly of respiratory protein complexes, and caused mitochondrial ultrastructural abnormalities. These mouse-based findings mechanistically linked cortical CHRM1 loss with poor survival of Alzheimer's patients. However, evaluation of the effect of Chrm1 loss on mouse hippocampal mitochondrial characteristics is necessary to fully understand our retrospective human tissue-based observations. This is the objective of this study. Methods: Enriched hippocampal and cortical mitochondrial fractions (EHMFs/ECMFs, respectively) derived from wild-type and Chrm1-/- mice were used to measure respiration by quantifying real-time oxygen consumption, supramolecular assembly of oxidative phosphorylation (OXPHOS)-associated proteins by blue native polyacrylamide gel electrophoresis, post-translational modifications (PTMs) by isoelectric focusing (IEF), and mitochondrial ultrastructure by electron microscopy. Results: In contrast to our previous observations in Chrm1-/- ECMFs, EHMFs of Chrm1-/- mice significantly increased respiration with a concomitant increase in the supramolecular assembly of OXPHOS-associated proteins, specifically Atp5a and Uqcrc2, with no mitochondrial ultrastructural alterations. IEF of ECMFs and EHMFs from Chrm1-/- mice showed a decrease and an increase, respectively in a negatively charged (pH∼3) fraction of Atp5a relative to the wild-type mice, with a corresponding decrease or increase in the supramolecular assembly of Atp5a and respiration indicating a tissue-specific signaling effect. Discussion: Our findings indicate that loss of Chrm1 in the cortex causes structural, and physiological alterations to mitochondria that compromise neuronal function, whereas Chrm1 loss in the hippocampus may benefit neuronal function by enhancing mitochondrial function. This brain region-specific differential effect of Chrm1 deletion on mitochondrial function supports our human brain region-based findings and Chrm1-/- mouse behavioral phenotypes. Furthermore, our study indicates that Chrm1-mediated brain region-specific differential PTMs of Atp5a may alter complex-V supramolecular assembly which in turn regulates mitochondrial structure-function.
Collapse
Affiliation(s)
- Mohammad Golam Sabbir
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
- Alzo Biosciences Inc., San Diego, CA, United States
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
- Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Mamiko Swanson
- Alzo Biosciences Inc., San Diego, CA, United States
- Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Robert C. Speth
- Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
- Department of Pharmacology and Physiology, School of Medicine, Georgetown University, Washington, DC, United States
| | - Benedict C. Albensi
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
- Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
28
|
Jones SE, Harvey PD. Cross-diagnostic determinants of cognitive functioning: the muscarinic cholinergic receptor as a model system. Transl Psychiatry 2023; 13:100. [PMID: 36973270 PMCID: PMC10042838 DOI: 10.1038/s41398-023-02400-x] [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: 01/11/2022] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
Cognitive impairment is a predictor of disability across different neuropsychiatric conditions, and cognitive abilities are also strongly related to educational attainment and indices of life success in the general population. Previous attempts at drug development for cognitive enhancement have commonly attempted to remedy defects in transmitters systems putatively associated with the conditions of interest such as the glutamate system in schizophrenia. Recent studies of the genomics of cognitive performance have suggested influences that are common in the general population and in different neuropsychiatric conditions. Thus, it seems possible that transmitter systems that are implicated for cognition across neuropsychiatric conditions and the general population would be a viable treatment target. We review the scientific data on cognition and the muscarinic cholinergic receptor system (M1 and M4) across different diagnoses, in aging, and in the general population. We suggest that there is evidence suggesting potential beneficial impacts of stimulation of critical muscarinic receptors for the enhancement of cognition in a broad manner, as well as the treatment of psychotic symptoms. Recent developments make stimulation of the M1 receptor more tolerable, and we identify the potential benefits of M1 and M4 receptor stimulation as a trans-diagnostic treatment model.
Collapse
Affiliation(s)
- Sara E Jones
- Department of Psychiatry, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - Philip D Harvey
- Department of Psychiatry, University of Miami Miller School of Medicine, Miami, FL, USA.
- Research Service, Bruce W. Carter VA Medical Center, Miami, FL, USA.
| |
Collapse
|
29
|
Gimenez-Gomez P, Le T, Martin GE. Modulation of neuronal excitability by binge alcohol drinking. Front Mol Neurosci 2023; 16:1098211. [PMID: 36866357 PMCID: PMC9971943 DOI: 10.3389/fnmol.2023.1098211] [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: 11/14/2022] [Accepted: 01/19/2023] [Indexed: 02/16/2023] Open
Abstract
Drug use poses a serious threat to health systems throughout the world. The number of consumers rises every year being alcohol the drug of abuse most consumed causing 3 million deaths (5.3% of all deaths) worldwide and 132.6 million disability-adjusted life years. In this review, we present an up-to-date summary about what is known regarding the global impact of binge alcohol drinking on brains and how it affects the development of cognitive functions, as well as the various preclinical models used to probe its effects on the neurobiology of the brain. This will be followed by a detailed report on the state of our current knowledge of the molecular and cellular mechanisms underlying the effects of binge drinking on neuronal excitability and synaptic plasticity, with an emphasis on brain regions of the meso-cortico limbic neurocircuitry.
Collapse
Affiliation(s)
- Pablo Gimenez-Gomez
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
- The Brudnick Neuropsychiatric Research Institute, Worcester, MA, United States
| | - Timmy Le
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
- The Brudnick Neuropsychiatric Research Institute, Worcester, MA, United States
- Graduate Program in Neuroscience, Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA, United States
| | - Gilles E. Martin
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
- The Brudnick Neuropsychiatric Research Institute, Worcester, MA, United States
| |
Collapse
|
30
|
Juza R, Musilek K, Mezeiova E, Soukup O, Korabecny J. Recent advances in dopamine D 2 receptor ligands in the treatment of neuropsychiatric disorders. Med Res Rev 2023; 43:55-211. [PMID: 36111795 DOI: 10.1002/med.21923] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 02/04/2023]
Abstract
Dopamine is a biologically active amine synthesized in the central and peripheral nervous system. This biogenic monoamine acts by activating five types of dopamine receptors (D1-5 Rs), which belong to the G protein-coupled receptor family. Antagonists and partial agonists of D2 Rs are used to treat schizophrenia, Parkinson's disease, depression, and anxiety. The typical pharmacophore with high D2 R affinity comprises four main areas, namely aromatic moiety, cyclic amine, central linker and aromatic/heteroaromatic lipophilic fragment. From the literature reviewed herein, we can conclude that 4-(2,3-dichlorophenyl), 4-(2-methoxyphenyl)-, 4-(benzo[b]thiophen-4-yl)-1-substituted piperazine, and 4-(6-fluorobenzo[d]isoxazol-3-yl)piperidine moieties are critical for high D2 R affinity. Four to six atoms chains are optimal for D2 R affinity with 4-butoxyl as the most pronounced one. The bicyclic aromatic/heteroaromatic systems are most frequently occurring as lipophilic appendages to retain high D2 R affinity. In this review, we provide a thorough overview of the therapeutic potential of D2 R modulators in the treatment of the aforementioned disorders. In addition, this review summarizes current knowledge about these diseases, with a focus on the dopaminergic pathway underlying these pathologies. Major attention is paid to the structure, function, and pharmacology of novel D2 R ligands, which have been developed in the last decade (2010-2021), and belong to the 1,4-disubstituted aromatic cyclic amine group. Due to the abundance of data, allosteric D2 R ligands and D2 R modulators from patents are not discussed in this review.
Collapse
Affiliation(s)
- Radomir Juza
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Kamil Musilek
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Eva Mezeiova
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Ondrej Soukup
- Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jan Korabecny
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
31
|
Dean B, Bakker G, Ueda HR, Tobin AB, Brown A, Kanaan RAA. A growing understanding of the role of muscarinic receptors in the molecular pathology and treatment of schizophrenia. Front Cell Neurosci 2023; 17:1124333. [PMID: 36909280 PMCID: PMC9992992 DOI: 10.3389/fncel.2023.1124333] [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: 12/15/2022] [Accepted: 02/06/2023] [Indexed: 02/24/2023] Open
Abstract
Pre-clinical models, postmortem and neuroimaging studies all support a role for muscarinic receptors in the molecular pathology of schizophrenia. From these data it was proposed that activation of the muscarinic M1 and/or M4 receptor would reduce the severity of the symptoms of schizophrenia. This hypothesis is now supported by results from two clinical trials which indicate that activating central muscarinic M1 and M4 receptors can reduce the severity of positive, negative and cognitive symptoms of the disorder. This review will provide an update on a growing body of evidence that argues the muscarinic M1 and M4 receptors have critical roles in CNS functions that are dysregulated by the pathophysiology of schizophrenia. This realization has been made possible, in part, by the growing ability to visualize and quantify muscarinic M1 and M4 receptors in the human CNS using molecular neuroimaging. We will discuss how these advances have provided evidence to support the notion that there is a sub-group of patients within the syndrome of schizophrenia that have a unique molecular pathology driven by a marked loss of muscarinic M1 receptors. This review is timely, as drugs targeting muscarinic receptors approach clinical use for the treatment of schizophrenia and here we outline the background biology that supported development of such drugs to treat the disorder.
Collapse
Affiliation(s)
- Brian Dean
- Synaptic Biology and Cognition Laboratory, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | | | - Hiroki R Ueda
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan
| | - Andrew B Tobin
- Advanced Research Centre (ARC), School of Molecular Bioscience, University of Glasgow, Glasgow, United Kingdom
| | | | - Richard A A Kanaan
- Department of Psychiatry, Austin Health, The University of Melbourne, Heidelberg, VIC, Australia
| |
Collapse
|
32
|
Yao R, Yamada K, Kito T, Aizu N, Iwata D, Izawa S, Nishii K, Sawada H, Chihara T. Novel shaking exercises for hippocampal and medial prefrontal cortex functioning maintain spatial working memory. Exp Gerontol 2023; 171:112024. [PMID: 36372283 DOI: 10.1016/j.exger.2022.112024] [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: 09/18/2022] [Revised: 10/27/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
INTRODUCTION The decline in spatial working memory is one of the earliest signs of normal brain aging. OBJECTIVE We developed a novel physical exercise method, termed the "shaking exercise," to slow down this process. METHODS The experimental protocol included administering the shaking exercise for 8-32 weeks in male senescence-accelerated mouse prone 10 (SAMP-10). They were subjected to the T-maze test, followed by immunohistochemical analysis, to assess the influence of the shaking exercise on the M1 muscarinic acetylcholine receptor (CHRM1) and α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) of the dorsal hippocampus and medial prefrontal cortex (dHC-mPFC). RESULTS The T-maze test demonstrated that the shaking group had less hesitation in the face of selecting direction at week 24. In the immunohistochemical analysis, more CHRM1s were in the CA3 subregion and more AMPARs were in the subiculum. CHRM1s and AMPARs were maintained in the CA1 region and the mPFC. The CHRM1s seem to have a positive effect on the AMPAR in the dentate gyrus (DG) region and the CA3 region. In the CA1 region, CHRM1s were negatively correlated with AMPARs. In addition, high-density neurons were expressed in the shaking group in the upstream DG, the middle part and the distal part of CA3, the distal part of CA1, and the mPFC. CONCLUSIONS Our results raise the possibility that maintenance of the spatial working memory effect observed with the shaking exercise is driven in part by the uneven affection of CHRM1s and AMPARs in the dHC-mPFC circuit system and significantly maintains the neuronal expression in the dHC-mPFC.
Collapse
Affiliation(s)
- Runhong Yao
- Department of Physical Therapy, School of Health Sciences, Japan University of Health Sciences, Satte, Saitama 340-0145, Japan; Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan.
| | - Kouji Yamada
- Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan.
| | - Takumi Kito
- Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan; Department of Physical Therapy, Faculty of Health Sciences, Kinjo University, Hakusan, Ishikawa 924-8511, Japan
| | - Naoki Aizu
- Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan.
| | - Daiki Iwata
- Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan.
| | - Sho Izawa
- Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan.
| | - Kazuhiro Nishii
- Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan.
| | - Hirohide Sawada
- Department of Medical Technology, School of Nursing and Medical Care, Yokkaichi Nursing and Medical Care University, Yokkaichi, Mie 512-8045, Japan.
| | - Takeshi Chihara
- Department of Medical Technology, School of Nursing and Medical Care, Yokkaichi Nursing and Medical Care University, Yokkaichi, Mie 512-8045, Japan.
| |
Collapse
|
33
|
Sabbir MG, Speth RC, Albensi BC. Loss of Cholinergic Receptor Muscarinic 1 (CHRM1) Protein in the Hippocampus and Temporal Cortex of a Subset of Individuals with Alzheimer’s Disease, Parkinson’s Disease, or Frontotemporal Dementia: Implications for Patient Survival. J Alzheimers Dis 2022; 90:727-747. [DOI: 10.3233/jad-220766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Dysfunction of cholinergic neurotransmission is a hallmark of Alzheimer’s disease (AD); forming the basis for using acetylcholine (ACh) esterase (AChE) inhibitors to mitigate symptoms of ACh deficiency in AD. The Cholinergic Receptor Muscarinic 1 (CHRM1) is highly expressed in brain regions impaired by AD. Previous analyses of postmortem AD brains revealed unaltered CHRM1 mRNA expression compared to normal brains. However, the CHRM1 protein level in AD and other forms of dementia has not been extensively studied. Reduced expression of CHRM1 in AD patients may explain the limited clinical efficacy of AChE inhibitors. Objective: To quantify CHRM1 protein in the postmortem hippocampus and temporal cortex of AD, Parkinson’s disease (PD), and frontotemporal dementia (FTD) patients. Methods: Western blotting was performed on postmortem hippocampus (N = 19/73/7/9: unaffected/AD/FTD/PD) and temporal cortex (N = 9/74/27: unaffected/AD/PD) using a validated anti-CHRM1 antibody. Results: Quantification based on immunoblotting using a validated anti-CHRM1 antibody revealed a significant loss of CHRM1 protein level (<50%) in the hippocampi (78% AD, 66% PD, and 85% FTD) and temporal cortices (56% AD and 42% PD) of dementia patients. Loss of CHRM1 in the temporal cortex was significantly associated with early death (<65–75 years) for both AD and PD patients. Conclusion: Severe reduction of CHRM1 in a subset of AD and PD patients can explain the reported low efficacy of AChE inhibitors as a mitigating treatment for dementia patients. Based on this study, it can be suggested that future research should prioritize therapeutic restoration of CHRM1 protein levels in cholinergic neurons.
Collapse
Affiliation(s)
- Mohammad Golam Sabbir
- Alzo Biosciences Inc., San Diego, CA, USA
- St. Boniface Hospital Albrechtsen Research Centre, Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, Manitoba, Canada
- Nova Southeastern University, College of Pharmacy, Davie, FL, USA
| | - Robert C. Speth
- Nova Southeastern University, College of Pharmacy, Davie, FL, USA
- Department of Pharmacology and Physiology, School of Medicine, Georgetown University, Washington, DC, USA
| | - Benedict C. Albensi
- Nova Southeastern University, College of Pharmacy, Davie, FL, USA
- St. Boniface Hospital Albrechtsen Research Centre, Division of Neurodegenerative Disorders, Winnipeg, Manitoba, Canada
- University of Manitoba, College of Medicine, Winnipeg, Manitoba, Canada
| |
Collapse
|
34
|
Chandramouleeshwaran S, Ghazala Z, Nobrega JN, Raymond R, Gambino S, Pollock BG, Rajji TK. Cell-based serum anticholinergic activity assay and working memory in cognitively healthy older adults before and after scopolamine: An exploratory study. J Psychopharmacol 2022; 36:1070-1076. [PMID: 36112867 DOI: 10.1177/02698811221122019] [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] [Indexed: 11/17/2022]
Abstract
BACKGROUND A new cell-based serum anticholinergic activity (cSAA) assay that measures anticholinergic activity specifically at muscarinic M1 receptors and eliminates many of the drawbacks of the existing assay was developed by our team. AIMS We aimed to study the relationship between changes in working memory and executive function with changes in cSAA using the new assay in cognitively healthy older adults. METHODS Cognitively healthy participants aged 50 years and above, received a single dose of 0.4 mg of intravenous scopolamine. Cognition and cSAA levels were measured before and 30 min after receiving scopolamine. Cognition was measured using the Cambridge Neuropsychological Test Automated Battery. RESULTS Ten participants were recruited, and nine (mean age = 69.8, SD = 9.5, range 59-86 years) completed the study. Following scopolamine, participants experienced an increase in cSAA (cSAA pre = 0.90 ± 0.97 vs cSAA post = 12.0 ± 3.70 pmol/L; t-test (df = (8) = -9.5, p < 0.001). In addition, there was an association between change in cSAA and changes in working memory (Spearman's ρ = 0.68, p = 0.042) and executive function (Spearman's ρ = 0.72, p = 0.027). CONCLUSIONS In our sample of cognitively healthy older adults, the new cSAA assay was able to quantify the scopolamine induced increase in anticholinergic load which correlated significantly with the observed decline in working memory and executive function.
Collapse
Affiliation(s)
- Susmita Chandramouleeshwaran
- Centre for Addiction and Mental Health, Toronto, ON, Canada.,University of Toronto, Toronto, ON, Canada.,University of Ottawa, Ottawa, ON, Canada
| | - Zaid Ghazala
- Centre for Addiction and Mental Health, Toronto, ON, Canada.,University of Toronto, Toronto, ON, Canada
| | - José N Nobrega
- Centre for Addiction and Mental Health, Toronto, ON, Canada.,University of Toronto, Toronto, ON, Canada
| | - Roger Raymond
- Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Sara Gambino
- Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Bruce G Pollock
- Centre for Addiction and Mental Health, Toronto, ON, Canada.,University of Toronto, Toronto, ON, Canada
| | - Tarek K Rajji
- Centre for Addiction and Mental Health, Toronto, ON, Canada.,University of Toronto, Toronto, ON, Canada
| |
Collapse
|
35
|
Paul SM, Yohn SE, Popiolek M, Miller AC, Felder CC. Muscarinic Acetylcholine Receptor Agonists as Novel Treatments for Schizophrenia. Am J Psychiatry 2022; 179:611-627. [PMID: 35758639 DOI: 10.1176/appi.ajp.21101083] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Schizophrenia remains a challenging disease to treat effectively with current antipsychotic medications due to their limited efficacy across the entire spectrum of core symptoms as well as their often burdensome side-effect profiles and poor tolerability. An unmet need remains for novel, mechanistically unique, and better tolerated therapeutic agents for treating schizophrenia, especially those that treat not only positive symptoms but also the negative and cognitive symptoms of the disease. Almost 25 years ago, the muscarinic acetylcholine receptor (mAChR) agonist xanomeline was reported to reduce psychotic symptoms and improve cognition in patients with Alzheimer's disease. The antipsychotic and procognitive properties of xanomeline were subsequently confirmed in a small study of acutely psychotic patients with chronic schizophrenia. These unexpected clinical findings have prompted considerable efforts across academia and industry to target mAChRs as a new approach to potentially treat schizophrenia and other psychotic disorders. The authors discuss recent advances in mAChR biology and pharmacology and the current understanding of the relative roles of the various mAChR subtypes, their downstream cellular effectors, and key neural circuits mediating the reduction in the core symptoms of schizophrenia in patients treated with xanomeline. They also provide an update on the status of novel mAChR agonists currently in development for potential treatment of schizophrenia and other neuropsychiatric disorders.
Collapse
|
36
|
Bae YS, Yoon SH, Kim YS, Oh SP, Song WS, Cha JH, Kim MH. Suppression of exaggerated NMDAR activity by memantine treatment ameliorates neurological and behavioral deficits in aminopeptidase P1-deficient mice. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1109-1124. [PMID: 35922532 PMCID: PMC9440093 DOI: 10.1038/s12276-022-00818-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 11/25/2022]
Abstract
Inborn errors of metabolism (IEMs) are common causes of neurodevelopmental disorders, including microcephaly, hyperactivity, and intellectual disability. However, the synaptic mechanisms of and pharmacological interventions for the neurological complications of most IEMs are unclear. Here, we report that metabolic dysfunction perturbs neuronal NMDA receptor (NMDAR) homeostasis and that the restoration of NMDAR signaling ameliorates neurodevelopmental and cognitive deficits in IEM model mice that lack aminopeptidase P1. Aminopeptidase P1-deficient (Xpnpep1–/–) mice, with a disruption of the proline-specific metalloprotease gene Xpnpep1, exhibit hippocampal neurodegeneration, behavioral hyperactivity, and impaired hippocampus-dependent learning. In this study, we found that GluN1 and GluN2A expression, NMDAR activity, and the NMDAR-dependent long-term potentiation (LTP) of excitatory synaptic transmission were markedly enhanced in the hippocampi of Xpnpep1–/– mice. The exaggerated NMDAR activity and NMDAR-dependent LTP were reversed by the NMDAR antagonist memantine. A single administration of memantine reversed hyperactivity in adult Xpnpep1–/– mice without improving learning and memory. Furthermore, chronic administration of memantine ameliorated hippocampal neurodegeneration, hyperactivity, and impaired learning and memory in Xpnpep1–/– mice. In addition, abnormally enhanced NMDAR-dependent LTP and NMDAR downstream signaling in the hippocampi of Xpnpep1–/– mice were reversed by chronic memantine treatment. These results suggest that the metabolic dysfunction caused by aminopeptidase P1 deficiency leads to synaptic dysfunction with excessive NMDAR activity, and the restoration of synaptic function may be a potential therapeutic strategy for the treatment of neurological complications related to IEMs. Addressing neurological symptoms may offer new treatments for inborn errors of metabolism (IEMs) affecting neurodevelopment. In such IEMs, mutation of an enzyme disrupts a metabolic pathway, causing buildup or lack of key molecules, with symptoms including hyperactivity, developmental delay, and intellectual disability. Because the detailed pathological mechanisms of most IEMs are unknown, there are no treatments for resulting neurological issues. Myoung-Hwan Kim at Seoul National University and co-workers investigated whether they could treat the neurological symptoms of the IEM, aminopeptidase P1 (APP1) deficiency. They found that APP1 deficiency in mice caused an increase in the neural receptor NMDAR. Suppressing NMDAR reduced both neurological and behavioral symptoms. These findings suggest potential treatments for APP1 deficiency, and indicate that neurodevelopmental disorders in IEMs may be treated by repairing the neural circuitry instead of the root metabolic cause.
Collapse
Affiliation(s)
- Young-Soo Bae
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Sang Ho Yoon
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.,Neuroscience Research Institute, Seoul National University Medical Research Center, Seoul, 03080, Korea
| | - Young Sook Kim
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Sung Pyo Oh
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Woo Seok Song
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.,Neuroscience Research Institute, Seoul National University Medical Research Center, Seoul, 03080, Korea
| | - Jin Hee Cha
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Myoung-Hwan Kim
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea. .,Neuroscience Research Institute, Seoul National University Medical Research Center, Seoul, 03080, Korea. .,Seoul National University Bundang Hospital, Seongnam, Gyeonggi, 13620, Korea.
| |
Collapse
|
37
|
Kirstein M, Cambrils A, Segarra A, Melero A, Varea E. Cholinergic Senescence in the Ts65Dn Mouse Model for Down Syndrome. Neurochem Res 2022; 47:3076-3092. [PMID: 35767135 PMCID: PMC9470680 DOI: 10.1007/s11064-022-03659-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/08/2022] [Accepted: 06/14/2022] [Indexed: 11/29/2022]
Abstract
Down syndrome (DS) induces a variable phenotype including intellectual disabilities and early development of Alzheimer’s disease (AD). Moreover, individuals with DS display accelerated aging that affects diverse organs, among them the brain. The Ts65Dn mouse is the most widely used model to study DS. Progressive loss of cholinergic neurons is one of the hallmarks of AD present in DS and in the Ts65Dn model. In this study, we quantify the number of cholinergic neurons in control and Ts65Dn mice, observing a general reduction in their number with age but in particular, a greater loss in old Ts65Dn mice. Increased expression of the m1 muscarinic receptor in the hippocampus counteracts this loss. Cholinergic neurons in the Ts65Dn mice display overexpression of the early expression gene c-fos and an increase in the expression of β-galactosidase, a marker of senescence. A possible mechanism for senescence induction could be phosphorylation of the transcription factor FOXO1 and its retention in the cytoplasm, which we are able to confirm in the Ts65Dn model. In our study, using Ts65Dn mice, we observe increased cholinergic activity, which induces a process of early senescence that culminates in the loss of these neurons.
Collapse
Affiliation(s)
- Martina Kirstein
- Cell Biology Department, Universitat de València, Dr. Moliner, 50, Burjassot, 46100, València, Spain
| | - Alba Cambrils
- Cell Biology Department, Universitat de València, Dr. Moliner, 50, Burjassot, 46100, València, Spain
| | - Ana Segarra
- Cell Biology Department, Universitat de València, Dr. Moliner, 50, Burjassot, 46100, València, Spain
| | - Ana Melero
- Cell Biology Department, Universitat de València, Dr. Moliner, 50, Burjassot, 46100, València, Spain
| | - Emilio Varea
- Cell Biology Department, Universitat de València, Dr. Moliner, 50, Burjassot, 46100, València, Spain.
| |
Collapse
|
38
|
VGLUT3 Ablation Differentially Modulates Glutamate Receptor Densities in Mouse Brain. eNeuro 2022; 9:ENEURO.0041-22.2022. [PMID: 35443989 PMCID: PMC9087739 DOI: 10.1523/eneuro.0041-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/04/2022] [Accepted: 04/10/2022] [Indexed: 11/21/2022] Open
Abstract
Type 3 vesicular glutamate transporter (VGLUT3) represents a unique modulator of glutamate release from both nonglutamatergic and glutamatergic varicosities within the brain. Despite its limited abundance, VGLUT3 is vital for the regulation of glutamate signaling and, therefore, modulates the activity of various brain microcircuits. However, little is known about how glutamate receptors are regulated by VGLUT3 across different brain regions. Here, we used VGLUT3 constitutive knock-out (VGLUT3-/-) mice and explored how VGLUT3 deletion influences total and cell surface expression of different ionotropic and metabotropic glutamate receptors. VGLUT3 deletion upregulated the overall expression of metabotropic glutamate receptors mGluR5 and mGluR2/3 in the cerebral cortex. In contrast, no change in the total expression of ionotropic NMDAR glutamate receptors were observed in the cerebral cortex of VGLUT3-/- mice. We noted significant reduction in cell surface levels of mGluR5, NMDAR2A, NMDAR2B, as well as reductions in dopaminergic D1 receptors and muscarinic M1 acetylcholine receptors in the hippocampus of VGLUT3-/- mice. Furthermore, mGluR2/3 total expression and mGluR5 cell surface levels were elevated in the striatum of VGLUT3-/- mice. Last, AMPAR subunit GluA1 was significantly upregulated throughout cortical, hippocampal, and striatal brain regions of VGLUT3-/- mice. Together, these findings complement and further support the evidence that VGLUT3 dynamically regulates glutamate receptor densities in several brain regions. These results suggest that VGLUT3 may play an intricate role in shaping glutamatergic signaling and plasticity in several brain areas.
Collapse
|
39
|
Asogwa NC, Toji N, He Z, Shao C, Shibata Y, Tatsumoto S, Ishikawa H, Go Y, Wada K. Nicotinic acetylcholine receptors in a songbird brain. J Comp Neurol 2022; 530:1966-1991. [PMID: 35344610 DOI: 10.1002/cne.25314] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 11/07/2022]
Abstract
Nicotinic acetylcholine receptors (nAChRs) are ligand-gated ion channels that mediate fast synaptic transmission and cell signaling, which contribute to learning, memory, and the execution of motor skills. Birdsong is a complex learned motor skill in songbirds. Although the existence of 15 nAChR subunits has been predicted in the avian genome, their expression patterns and potential contributions to song learning and production have not been comprehensively investigated. Here, we cloned all the 15 nAChR subunits (ChrnA1-10, B2-4, D, and G) from the zebra finch brain and investigated the mRNA expression patterns in the neural pathways responsible for the learning and production of birdsong during a critical period of song learning. Although there were no detectable hybridization signals for ChrnA1, A6, A9, and A10, the other 11 nAChR subunits were uniquely expressed in one or more major subdivisions in the song nuclei of the songbird brain. Of these 11 subunits, ChrnA3-5, A7, and B2 were differentially regulated in the song nuclei compared with the surrounding anatomically related regions. ChrnA5 was upregulated during the critical period of song learning in the lateral magnocellular nucleus of the anterior nidopallium. Furthermore, single-cell RNA sequencing revealed ChrnA7 and B2 to be the major subunits expressed in neurons of the vocal motor nuclei HVC and robust nucleus of the arcopallium, indicating the potential existence of ChrnA7-homomeric and ChrnB2-heteromeric nAChRs in limited cell populations. These results suggest that relatively limited types of nAChR subunits provide functional contributions to song learning and production in songbirds.
Collapse
Affiliation(s)
| | - Noriyuki Toji
- Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Ziwei He
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Chengru Shao
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Yukino Shibata
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Shoji Tatsumoto
- Cognitive Genomics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Hiroe Ishikawa
- Cognitive Genomics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Yasuhiro Go
- Cognitive Genomics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
- School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Japan
- Department of Physiological Sciences, National Institute for Physiological Sciences, Okazaki, Japan
| | - Kazuhiro Wada
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
- Faculty of Science, Hokkaido University, Sapporo, Japan
| |
Collapse
|
40
|
TrkA-cholinergic signaling modulates fear encoding and extinction learning in PTSD-like behavior. Transl Psychiatry 2022; 12:111. [PMID: 35301275 PMCID: PMC8931170 DOI: 10.1038/s41398-022-01869-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 02/19/2022] [Accepted: 02/24/2022] [Indexed: 11/25/2022] Open
Abstract
Recent studies have suggested that the use of cognitive enhancers as adjuncts to exposure-based therapy in individuals suffering from post-traumatic stress disorder (PTSD) may be beneficial. Brain cholinergic signaling through basal forebrain projections to the hippocampus is an established pathway mediating fear response and cognitive flexibility. Here we employed a genetic strategy to enhance cholinergic activity through increased signaling of the NGF receptor TrkA. This strategy leads to increased levels of the marker of cholinergic activation, acetylcholine synthesizing enzyme choline acetyltransferase, in forebrain cholinergic regions and their projection areas such as the hippocampus. Mice with increased cholinergic activity do not display any neurobehavioral abnormalities except a selective attenuation of fear response and lower fear expression in extinction trials. Reduction in fear response is rescued by the GABA antagonist picrotoxin in mutant mice, and, in wild-type mice, is mimicked by the GABA agonist midazolam suggesting that GABA can modulate cholinergic functions on fear circuitries. Importantly, mutant mice also show a reduction in fear processing under stress conditions in a single prolonged stress (SPS) model of PTSD-like behavior, and augmentation of cholinergic signaling by the drug donepezil in wild-type mice promotes extinction learning in a similar SPS model of PTSD-like behavior. Donepezil is already in clinical use for the treatment of dementia suggesting a new translational application of this drug for improving exposure-based psychotherapy in PTSD patients.
Collapse
|
41
|
Drug Design Targeting the Muscarinic Receptors and the Implications in Central Nervous System Disorders. Biomedicines 2022; 10:biomedicines10020398. [PMID: 35203607 PMCID: PMC8962391 DOI: 10.3390/biomedicines10020398] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 11/16/2022] Open
Abstract
There is substantial evidence that cholinergic system function impairment plays a significant role in many central nervous system (CNS) disorders. During the past three decades, muscarinic receptors (mAChRs) have been implicated in various pathologies and have been prominent targets of drug-design efforts. However, due to the high sequence homology of the orthosteric binding site, many drug candidates resulted in limited clinical success. Although several advances in treating peripheral pathologies have been achieved, targeting CNS pathologies remains challenging for researchers. Nevertheless, significant progress has been made in recent years to develop functionally selective orthosteric and allosteric ligands targeting the mAChRs with limited side effect profiles. This review highlights past efforts and focuses on recent advances in drug design targeting these receptors for Alzheimer’s disease (AD), schizophrenia (SZ), and depression.
Collapse
|
42
|
Sola E, Moyano P, Flores A, García J, García JM, Anadon MJ, Frejo MT, Pelayo A, de la Cabeza Fernandez M, Del Pino J. Cadmium-induced neurotoxic effects on rat basal forebrain cholinergic system through thyroid hormones disruption. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 90:103791. [PMID: 34968718 DOI: 10.1016/j.etap.2021.103791] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 06/14/2023]
Abstract
Cadmium (Cd) single and repeated exposure produces cognitive dysfunctions. Basal forebrain cholinergic neurons (BFCN) regulate cognitive functions. BFCN loss or cholinergic neurotransmission dysfunction leads to cognitive disabilities. Thyroid hormones (THs) maintain BFCN viability and functions, and Cd disrupts their levels. However, Cd-induced BFCN damages and THs disruption involvement was not studied. To research this we treated male Wistar rats intraperitoneally with Cd once (1 mg/kg) or repetitively for 28 days (0.1 mg/kg) with/without triiodothyronine (T3, 40 µg/kg/day). Cd increased thyroid-stimulating-hormone (TSH) and decreased T3 and tetraiodothyronine (T4). Cd altered cholinergic transmission and induced a more pronounced neurodegeneration on BFCN, mediated partially by THs reduction. Additionally, Cd antagonized muscarinic 1 receptor (M1R), overexpressed acetylcholinesterase S variant (AChE-S), downregulated AChE-R, M2R, M3R and M4R, and reduced AChE and choline acetyltransferase activities through THs disruption. These results may assist to discover cadmium mechanisms that induce cognitive disabilities, revealing a new possible therapeutic tool.
Collapse
Affiliation(s)
- Emma Sola
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School, Complutense University of Madrid, 28041 Madrid, Spain
| | - Paula Moyano
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Andrea Flores
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Jimena García
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - José Manuel García
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - María José Anadon
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School, Complutense University of Madrid, 28041 Madrid, Spain
| | - María Teresa Frejo
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Adela Pelayo
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School, Complutense University of Madrid, 28041 Madrid, Spain
| | - Maria de la Cabeza Fernandez
- Department of Chemistry in Pharmaceutical Sciences, Pharnacy School, Complutense University of Madrid, 28041 Madrid, Spain
| | - Javier Del Pino
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain.
| |
Collapse
|
43
|
Tian T, Lai X, Xiang K, Han X, Yin S, Cabrera RM, Steele JW, Lei Y, Cao X, Finnell RH, Wang L, Ren A. Hypermethylation of PI3K-AKT signalling pathway genes is associated with human neural tube defects. Epigenetics 2022; 17:133-146. [PMID: 33491544 PMCID: PMC8865305 DOI: 10.1080/15592294.2021.1878725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/25/2020] [Accepted: 01/07/2021] [Indexed: 10/22/2022] Open
Abstract
Neural tube defects (NTDs) are a group of common and severe congenital malformations. The PI3K-AKT signalling pathway plays a crucial role in the neural tube development. There is limited evidence concerning any possible association between aberrant methylation in PI3K-AKT signalling pathway genes and NTDs. Therefore, we aimed to investigate potential associations between aberrant methylation of PI3K-AKT pathway genes and NTDs. Methylation studies of PI3K-AKT pathway genes utilizing microarray genome-methylation data derived from neural tissues of ten NTD cases and eight non-malformed controls were performed. Targeted DNA methylation analysis was subsequently performed in an independent cohort of 73 NTD cases and 32 controls to validate the methylation levels of identified genes. siRNAs were used to pull-down the target genes in human embryonic stem cells (hESCs) to examine the effects of the aberrant expression of target genes on neural cells. As a result, 321 differentially hypermethylated CpG sites in the promoter regions of 30 PI3K-AKT pathway genes were identified in the microarray data. In target methylation analysis, CHRM1, FGF19, and ITGA7 were confirmed to be significantly hypermethylated in NTD cases and were associated with increased risk for NTDs. The down-regulation of FGF19, CHRM1, and ITGA7 impaired the formation of rosette-like cell aggregates. The down-regulation of those three genes affected the expression of PAX6, SOX2 and MAP2, implying their influence on the differentiation of neural cells. This study for the first time reported that hypermethylation of PI3K-AKT pathway genes such as CHRM1, FGF19, and ITGA7 is associated with human NTDs.
Collapse
Affiliation(s)
- Tian Tian
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Xinyuan Lai
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Kuanhui Xiang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiao Han
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Shengju Yin
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Robert M. Cabrera
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - John W. Steele
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Yunping Lei
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Xuanye Cao
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Richard H. Finnell
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Departments of Molecular and Human Genetics and Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Linlin Wang
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Aiguo Ren
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| |
Collapse
|
44
|
Walker LC, Campbell EJ, Huckstep KL, Chen NA, Langmead CJ, Lawrence AJ. M 1 muscarinic receptor activation decreases alcohol consumption via a reduction in consummatory behavior. Pharmacol Res Perspect 2021; 10:e00907. [PMID: 34962108 PMCID: PMC8929368 DOI: 10.1002/prp2.907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 11/21/2021] [Indexed: 11/07/2022] Open
Abstract
Muscarinic acetylcholine receptors (mAChRs) have been shown to mediate alcohol consumption and seeking. Both M4 and M5 mAChRs have been highlighted as potential novel treatment targets for alcohol use disorders (AUD). Similarly, M1 mAChRs are expressed throughout reward circuitry, and their signaling has been implicated in cocaine consumption. However, whether the same effects are seen for alcohol consumption, or whether natural reward intake is inadvertently impacted is still unknown. To determine the role of M1 mAChRs in alcohol consumption, we tested operant self-administration of alcohol under both fixed ratio (FR3) and progressive ratio (PR3-4) schedules. Enhancing M1 mAChR signaling (via the M1 PAM-Agonist PF-06767832, 1 mg/kg, i.p.) reduced operant alcohol consumption on a fixed schedule but had no effect on motivation to acquire alcohol. To determine whether these actions were specific to alcohol, we examined the effects of M1 enhancement on natural reward (sucrose) self-administration. Systemic administration of PF-06767832 (1 mg/kg, i.p.) also reduced operant sucrose self-administration, suggesting the actions of the M1 receptor may be non-selective across drug and natural rewards. Finally, to understand whether this reduction extended to natural consummatory behaviors, we assessed home cage standard chow and water consumption. M1 enhancement via systemic PF-06767832 administration reduced food and water consumption. Together our results suggest the M1 PAM-agonist, PF-06767832, non-specifically reduces consummatory behaviors that are not associated with motivational strength for the reward. These data highlight the need to further characterize M1 agonists, PAMs, and PAM-agonists, which may have varying degrees of utility in the treatment of neuropsychiatric disorders including AUD.
Collapse
Affiliation(s)
- Leigh C. Walker
- Florey Institute of Neuroscience and Mental HealthParkvilleAustralia
- Florey Department of Neuroscience and Mental HealthUniversity of MelbourneParkvilleAustralia
| | - Erin J. Campbell
- Florey Institute of Neuroscience and Mental HealthParkvilleAustralia
- Florey Department of Neuroscience and Mental HealthUniversity of MelbourneParkvilleAustralia
| | - Kate L. Huckstep
- Florey Institute of Neuroscience and Mental HealthParkvilleAustralia
- Florey Department of Neuroscience and Mental HealthUniversity of MelbourneParkvilleAustralia
| | - Nicola A. Chen
- Florey Institute of Neuroscience and Mental HealthParkvilleAustralia
- Florey Department of Neuroscience and Mental HealthUniversity of MelbourneParkvilleAustralia
| | - Christopher J. Langmead
- Drug Discovery BiologyMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVictoriaAustralia
| | - Andrew J. Lawrence
- Florey Institute of Neuroscience and Mental HealthParkvilleAustralia
- Florey Department of Neuroscience and Mental HealthUniversity of MelbourneParkvilleAustralia
| |
Collapse
|
45
|
Mori C, Aoki N, Fujita T, Yamaguchi S, Matsushima T, Homma KJ. Gene expression profiles of the muscarinic acetylcholine receptors in brain regions relating to filial imprinting of newly-hatched domestic chicks. Behav Brain Res 2021; 420:113708. [PMID: 34902480 DOI: 10.1016/j.bbr.2021.113708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/24/2021] [Accepted: 12/05/2021] [Indexed: 12/01/2022]
Abstract
Muscarinic acetylcholine receptors (mAChRs) in the central nervous system play an important role in regulating complex functions such as learning, memory, and selective attention. Five subtypes of the mAChRs (M1-M5) have been identified in mammals, and are classified into two subfamilies: excitatory (M1, M3, and M5) and inhibitory (M2 and M4) subfamilies. Filial imprinting of domestic chicks is a useful model in the laboratory to investigate the mechanisms of memory formation in early learning. We recently found that mAChRs in the intermediate medial mesopallium (IMM) are involved in the memory formation of imprinting. However, expression profiles of each mAChR subtype in the brain regions including the IMM remain unexplored. Here we show the unique gene expression of each mAChR subtype in the pallial regions involved in imprinting. In terms of the excitatory mAChRs, M5 was expressed in the IMM region and other parts of the pallium, whereas M3 was less expressed in the IMM but highly expressed in the hyperpallium and nidopallium. Regarding the inhibitory mAChRs, M2 was sparsely distributed but clearly in some cells throughout the pallial regions. M4 was highly expressed in the IMM region and other parts of the pallium. These expression profiles can be used as a basis for understanding cholinergic modulation in the memory formation of imprinting and other learning processes in birds, and compared to those of mammals.
Collapse
Affiliation(s)
- Chihiro Mori
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Naoya Aoki
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Toshiyuki Fujita
- Department of Biological Sciences, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Shinji Yamaguchi
- Department of Biological Sciences, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Toshiya Matsushima
- Department of Biology, Faculty of Science, Hokkaido University, Hokkaido 060-0810, Japan
| | - Koichi J Homma
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| |
Collapse
|
46
|
Schledwitz A, Sundel MH, Alizadeh M, Hu S, Xie G, Raufman JP. Differential Actions of Muscarinic Receptor Subtypes in Gastric, Pancreatic, and Colon Cancer. Int J Mol Sci 2021; 22:ijms222313153. [PMID: 34884958 PMCID: PMC8658119 DOI: 10.3390/ijms222313153] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/17/2022] Open
Abstract
Cancers arising from gastrointestinal epithelial cells are common, aggressive, and difficult to treat. Progress in this area resulted from recognizing that the biological behavior of these cancers is highly dependent on bioactive molecules released by neurocrine, paracrine, and autocrine mechanisms within the tumor microenvironment. For many decades after its discovery as a neurotransmitter, acetylcholine was thought to be synthesized and released uniquely from neurons and considered the sole physiological ligand for muscarinic receptor subtypes, which were believed to have similar or redundant actions. In the intervening years, we learned this former dogma is not tenable. (1) Acetylcholine is not produced and released only by neurons. The cellular machinery required to synthesize and release acetylcholine is present in immune, cancer, and other cells, as well as in lower organisms (e.g., bacteria) that inhabit the gut. (2) Acetylcholine is not the sole physiological activator of muscarinic receptors. For example, selected bile acids can modulate muscarinic receptor function. (3) Muscarinic receptor subtypes anticipated to have overlapping functions based on similar G protein coupling and downstream signaling may have unexpectedly diverse actions. Here, we review the relevant research findings supporting these conclusions and discuss how the complexity of muscarinic receptor biology impacts health and disease, focusing on their role in the initiation and progression of gastric, pancreatic, and colon cancers.
Collapse
Affiliation(s)
- Alyssa Schledwitz
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.S.); (M.A.); (S.H.); (G.X.)
| | - Margaret H. Sundel
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Madeline Alizadeh
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.S.); (M.A.); (S.H.); (G.X.)
- The Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Shien Hu
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.S.); (M.A.); (S.H.); (G.X.)
- VA Maryland Healthcare System, Baltimore, MD 21201, USA
| | - Guofeng Xie
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.S.); (M.A.); (S.H.); (G.X.)
- VA Maryland Healthcare System, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jean-Pierre Raufman
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.S.); (M.A.); (S.H.); (G.X.)
- VA Maryland Healthcare System, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Correspondence: ; Tel.: +1-410-328-8728
| |
Collapse
|
47
|
Chandramouleeshwaran S, Ahsan N, Raymond R, Nobrega JN, Wang W, Fischer CE, Flint AJ, Herrmann N, Kumar S, Lanctôt K, Mah L, Mulsant BH, Pollock BG, Rajji TK. Relationships Between a New Cultured Cell-Based Serum Anticholinergic Activity Assay and Anticholinergic Burden Scales or Cognitive Performance in Older Adults. Am J Geriatr Psychiatry 2021; 29:1239-1252. [PMID: 33846084 DOI: 10.1016/j.jagp.2021.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/10/2021] [Accepted: 03/12/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVES Anticholinergic burden has been associated with deleterious effects on cognition particularly in those with an underlying brain disorder. We developed a new assay based on cultured cells to measure serum anticholinergic activity (cSAA). We report on its relationships with established anticholinergic burden rating scales and cognitive assessments in older patients with mild cognitive impairment (MCI) or major depressive disorder (MDD) in remission or both. DESIGN The study was cross sectional in nature. SETTING This was a five-centre study conducted in Toronto, Canada. PARTICIPANTS Serum samples were collected and cSAA levels were measured in 311 participants aged 60 years or older (154 with MCI, 57 with MDD, and 100 with MCI + MDD). MEASUREMENTS The cSAA assay uses radio-ligand binding to cultured cells stably expressing the muscarinic M1 receptors, with an added procedure to remove potential confounds associated with serum proteins. Lists of medications were used to calculate Anticholinergic Burden and Anticholinergic Drug Scale total scores. Participants also completed a comprehensive cognitive battery. RESULTS Higher cSAA levels were associated with higher anticholinergic burden and anticholinergic drug scale scores, and also with lower performance on executive function tests, after adjusting for age, gender, education, and diagnosis. CONCLUSIONS These results support the use of the cSAA assay as a laboratory measure of anticholinergic burden.
Collapse
Affiliation(s)
- Susmita Chandramouleeshwaran
- Centre for Addiction and Mental Health (SC, NA, RR, JNN, WW, SK, BHM, BGP, TKR), Toronto, Canada; Department of Psychiatry (JNB, WW, CEF, AJF, NH, SK, KL, LM, BHM, BGP, TKR), University of Toronto, Toronto, Canada
| | - Naba Ahsan
- Centre for Addiction and Mental Health (SC, NA, RR, JNN, WW, SK, BHM, BGP, TKR), Toronto, Canada
| | - Roger Raymond
- Centre for Addiction and Mental Health (SC, NA, RR, JNN, WW, SK, BHM, BGP, TKR), Toronto, Canada
| | - Jose N Nobrega
- Centre for Addiction and Mental Health (SC, NA, RR, JNN, WW, SK, BHM, BGP, TKR), Toronto, Canada; Department of Psychiatry (JNB, WW, CEF, AJF, NH, SK, KL, LM, BHM, BGP, TKR), University of Toronto, Toronto, Canada
| | - Wei Wang
- Centre for Addiction and Mental Health (SC, NA, RR, JNN, WW, SK, BHM, BGP, TKR), Toronto, Canada; Department of Psychiatry (JNB, WW, CEF, AJF, NH, SK, KL, LM, BHM, BGP, TKR), University of Toronto, Toronto, Canada
| | - Corinne E Fischer
- Department of Psychiatry (JNB, WW, CEF, AJF, NH, SK, KL, LM, BHM, BGP, TKR), University of Toronto, Toronto, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital (CEF), Toronto, Canada; Toronto Dementia Research Alliance (CEF, SK, KL, BHM, BGP, TKR), University of Toronto, Canada
| | - Alastair J Flint
- Department of Psychiatry (JNB, WW, CEF, AJF, NH, SK, KL, LM, BHM, BGP, TKR), University of Toronto, Toronto, Canada; Centre for Mental Health (AJF), University Health Network, Toronto, Canada
| | - Nathan Herrmann
- Department of Psychiatry (JNB, WW, CEF, AJF, NH, SK, KL, LM, BHM, BGP, TKR), University of Toronto, Toronto, Canada; Sunnybrook Health Sciences Centre, (NH, KL) Toronto, Canada
| | - Sanjeev Kumar
- Centre for Addiction and Mental Health (SC, NA, RR, JNN, WW, SK, BHM, BGP, TKR), Toronto, Canada; Department of Psychiatry (JNB, WW, CEF, AJF, NH, SK, KL, LM, BHM, BGP, TKR), University of Toronto, Toronto, Canada; Toronto Dementia Research Alliance (CEF, SK, KL, BHM, BGP, TKR), University of Toronto, Canada
| | - Krista Lanctôt
- Department of Psychiatry (JNB, WW, CEF, AJF, NH, SK, KL, LM, BHM, BGP, TKR), University of Toronto, Toronto, Canada; Toronto Dementia Research Alliance (CEF, SK, KL, BHM, BGP, TKR), University of Toronto, Canada; Sunnybrook Health Sciences Centre, (NH, KL) Toronto, Canada
| | - Linda Mah
- Department of Psychiatry (JNB, WW, CEF, AJF, NH, SK, KL, LM, BHM, BGP, TKR), University of Toronto, Toronto, Canada; Rotman Research Institute, Baycrest Health Sciences Centre, (LM) Toronto, Canada
| | - Benoit H Mulsant
- Centre for Addiction and Mental Health (SC, NA, RR, JNN, WW, SK, BHM, BGP, TKR), Toronto, Canada; Department of Psychiatry (JNB, WW, CEF, AJF, NH, SK, KL, LM, BHM, BGP, TKR), University of Toronto, Toronto, Canada; Toronto Dementia Research Alliance (CEF, SK, KL, BHM, BGP, TKR), University of Toronto, Canada
| | - Bruce G Pollock
- Centre for Addiction and Mental Health (SC, NA, RR, JNN, WW, SK, BHM, BGP, TKR), Toronto, Canada; Department of Psychiatry (JNB, WW, CEF, AJF, NH, SK, KL, LM, BHM, BGP, TKR), University of Toronto, Toronto, Canada; Toronto Dementia Research Alliance (CEF, SK, KL, BHM, BGP, TKR), University of Toronto, Canada
| | - Tarek K Rajji
- Centre for Addiction and Mental Health (SC, NA, RR, JNN, WW, SK, BHM, BGP, TKR), Toronto, Canada; Department of Psychiatry (JNB, WW, CEF, AJF, NH, SK, KL, LM, BHM, BGP, TKR), University of Toronto, Toronto, Canada; Toronto Dementia Research Alliance (CEF, SK, KL, BHM, BGP, TKR), University of Toronto, Canada.
| |
Collapse
|
48
|
Abd-Elrahman KS, Sarasija S, Colson TLL, Ferguson SSG. A M1 muscarinic acetylcholine receptor positive allosteric modulator improves pathology and cognitive deficits in female APPswe/PSEN1ΔE9 mice. Br J Pharmacol 2021; 179:1769-1783. [PMID: 34820835 DOI: 10.1111/bph.15750] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 11/09/2021] [Accepted: 11/13/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive cognitive decline and women account for 60% of diagnosed cases. Beta-amyloid (Aβ) oligomers is considered the principal neurotoxic species in AD brains. The M1 muscarinic acetylcholine receptor (M1 mAChR) plays a key role in memory and learning. M1 mAChR agonists show pro-cognitive activity but cause many adverse off-target effects. A new orally bioavailable M1 mAChR positive allosteric modulator (PAM), VU0486846, is devoid of direct agonist activity or adverse effects but was not tested for disease-modifying efficacy in female AD mice. EXPERIMENTAL APPROACH Nine-month-old female APPswe/PSEN1ΔE9 (APPswe) and wildtype mice were treated with VU0486846 in drinking water (10mg/kg/day) for 4 or 8 weeks. Cognitive function of mice was assessed after treatment and brains were harvested for biochemical and immunohistochemical assessment. KEY RESULTS VU0486846 improved cognitive function of APPswe mice when tested in novel object recognition and Morris water maze. This was paralleled by a significant reduction in Aβ oligomers and plaques and neuronal loss in hippocampus. VU0486846 reduced Aβ oligomer production in APPswe mice by increasing M1 mAChR expression and shifting the processing of amyloid precursor protein from amyloidogenic cleavage to non-amyloidogenic cleavage. Specifically, VU0486846 reduced the expression of β-secretase 1 (BACE1), whereas it enhanced the expression of the α-secretase ADAM10 in APPswe hippocampus. CONCLUSION AND IMPLICATIONS Using M1 mAChR PAMs can be a viable disease-modifying approach that should be exploited clinically to slow AD in women.
Collapse
Affiliation(s)
- Khaled S Abd-Elrahman
- University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario, Canada.,Departments of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Shaarika Sarasija
- University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario, Canada.,Departments of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Tash-Lynn L Colson
- University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario, Canada.,Departments of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Stephen S G Ferguson
- University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario, Canada.,Departments of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
49
|
Reale M, Carrarini C, Russo M, Dono F, Ferri L, Pietro MD, Costantini E, Porreca A, Nicola MD, Onofrj M, Bonanni L. Muscarinic Receptors Expression in the Peripheral Blood Cells Differentiate Dementia with Lewy Bodies from Alzheimer's Disease. J Alzheimers Dis 2021; 85:323-330. [PMID: 34806612 DOI: 10.3233/jad-215285] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Central nervous system disruption of cholinergic (ACh) signaling, which plays a major role in cognitive processes, is well documented in dementia with Lewy bodies (DLB) and Alzheimer's disease (AD). The expression of muscarinic ACh receptors type 1 and 4 (CHRM1 and CHRM4) has been reported to be altered in the brain of DLB patients. OBJECTIVE We aim to assess the peripheral gene expression of CHRM1 and 4 in DLB as a possible marker as compared to AD and healthy control (HC) subjects. METHODS Peripheral blood mononuclear cells were collected from 21 DLB, 13 AD, and 8 HC matched subjects. RT-PCR was performed to estimate gene expression of CHRM1 and CHRM4. RESULTS Peripheral CHRM1 expression was higher and CHRM4 was lower in DLB and AD compared to HC, whereas both CHRM1 and CHRM4 levels were higher in AD compared to DLB patients. Receiver operating characteristics curves, with logistic regression analysis, showed that combining peripheral CHRM1 and CHRM4 levels, DLB and AD subjects were classified with an accuracy of 76.0%. CONCLUSION Alterations of peripheral CHRM1 and CHRM4 was found in both AD and DLB patients as compared to HC. CHRM1 and CHRM4 gene expression resulted to be lower in DLB patients compared to AD. In the future, peripheral CHRM expression could be studied as a possible marker of neurodegenerative conditions associated with cholinergic deficit and a possible marker of response to acetylcholinesterase inhibitors.
Collapse
Affiliation(s)
- Marcella Reale
- Department of Innovative Technologies in Medicine and Dentistry, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Claudia Carrarini
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Mirella Russo
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Fedele Dono
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Laura Ferri
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Martina Di Pietro
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Erica Costantini
- Department of Medicine and Aging Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Annamaria Porreca
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Marta Di Nicola
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Marco Onofrj
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Laura Bonanni
- Department of Medicine and Aging Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
50
|
Humphries R, Mellor JR, O'Donnell C. Acetylcholine Boosts Dendritic NMDA Spikes in a CA3 Pyramidal Neuron Model. Neuroscience 2021; 489:69-83. [PMID: 34780920 DOI: 10.1016/j.neuroscience.2021.11.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 10/12/2021] [Accepted: 11/05/2021] [Indexed: 11/25/2022]
Abstract
Acetylcholine has been proposed to facilitate the formation of memory ensembles within the hippocampal CA3 network, by enhancing plasticity at CA3-CA3 recurrent synapses. Regenerative NMDA receptor (NMDAR) activation in CA3 neuron dendrites (NMDA spikes) increase synaptic Ca2+ influx and can trigger this synaptic plasticity. Acetylcholine inhibits potassium channels which enhances dendritic excitability and therefore could facilitate NMDA spike generation. Here, we investigate NMDAR-mediated nonlinear synaptic integration in stratum radiatum (SR) and stratum lacunosum moleculare (SLM) dendrites in a reconstructed CA3 neuron computational model and study the effect of cholinergic inhibition of potassium conductances on this nonlinearity. We found that distal SLM dendrites, with a higher input resistance, had a lower threshold for NMDA spike generation compared to SR dendrites. Simulating acetylcholine by blocking potassium channels (M-type, A-type, Ca2+-activated, and inwardly-rectifying) increased dendritic excitability and reduced the number of synapses required to generate NMDA spikes, particularly in the SR dendrites. The magnitude of this effect was heterogeneous across different dendritic branches within the same neuron. These results predict that acetylcholine facilitates dendritic integration and NMDA spike generation in selected CA3 dendrites which could strengthen connections between specific CA3 neurons to form memory ensembles.
Collapse
Affiliation(s)
- Rachel Humphries
- Center for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol BS8 1TD, UK; Computational Neuroscience Unit, School of Computer Science, Electrical and Electronic Engineering, and Engineering Mathematics, University of Bristol, Bristol BS8 1UB, UK
| | - Jack R Mellor
- Center for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - Cian O'Donnell
- Computational Neuroscience Unit, School of Computer Science, Electrical and Electronic Engineering, and Engineering Mathematics, University of Bristol, Bristol BS8 1UB, UK; School of Computing, Engineering and Intelligent Systems, Ulster University, Magee Campus, Northland Road, Derry/Londonderry BT48 7JL, UK.
| |
Collapse
|