1
|
Chen C, Masotti M, Shepard N, Promes V, Tombesi G, Arango D, Manzoni C, Greggio E, Hilfiker S, Kozorovitskiy Y, Parisiadou L. LRRK2 mediates haloperidol-induced changes in indirect pathway striatal projection neurons. Mol Psychiatry 2025:10.1038/s41380-025-03030-z. [PMID: 40269187 DOI: 10.1038/s41380-025-03030-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025]
Abstract
Haloperidol is used to manage psychotic symptoms in several neurological disorders through mechanisms that involve antagonism of dopamine D2 receptors that are highly expressed in the striatum. Significant side effects of haloperidol, known as extrapyramidal symptoms, lead to motor deficits similar to those seen in Parkinson's disease and present a major challenge in clinical settings. The underlying molecular mechanisms responsible for these side effects remain poorly understood. Parkinson's disease-associated leucine-rich repeat kinase 2 (LRRK2) has an essential role in striatal physiology and a known link to dopamine D2 receptor signaling. Here, we systematically explore convergent signaling of haloperidol and LRRK2 through pharmacological or genetic inhibition of LRRK2 kinase, as well as knock-in mouse models expressing pathogenic mutant LRRK2 with increased kinase activity. Behavioral assays show that LRRK2 kinase inhibition ameliorates haloperidol-induced motor changes in mice. A combination of electrophysiological and anatomical approaches reveals that LRRK2 kinase inhibition interferes with haloperidol-induced changes, specifically in striatal neurons of the indirect pathway. Proteomic studies and targeted intracellular pathway analyses demonstrate that haloperidol induces a similar pattern of intracellular signaling as increased LRRK2 kinase activity. Our study suggests that LRRK2 kinase plays a key role in striatal dopamine D2 receptor signaling underlying the undesirable motor side effects of haloperidol. This work opens up new therapeutic avenues for dopamine-related disorders, such as psychosis, also furthering our understanding of Parkinson's disease pathophysiology.
Collapse
Affiliation(s)
- Chuyu Chen
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Meghan Masotti
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Nathaniel Shepard
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Vanessa Promes
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Giulia Tombesi
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Daniel Arango
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | | | - Elisa Greggio
- Department of Biology, University of Padova, Padova, Italy
| | - Sabine Hilfiker
- Department of Anesthesiology, Rutgers, New Jersey Medical School, Newark, NJ, USA
| | | | - Loukia Parisiadou
- Department of Pharmacology, Northwestern University, Chicago, IL, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
2
|
Ko SY, Kim DG, Lee H, Jung SJ, Son H. Extinction of contextual fear memory is facilitated in TRPM2 knockout mice. Mol Brain 2025; 18:16. [PMID: 40016847 PMCID: PMC11869647 DOI: 10.1186/s13041-025-01181-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 01/31/2025] [Indexed: 03/01/2025] Open
Abstract
Transient receptor potential melastatin type 2 (TRPM2) is a nonselective cation channel involved in synaptic plasticity. We investigated its role in contextual fear conditioning and extinction of conditioned fear using Trpm2-deficient (Trpm2-/-) mice. Trpm2-/- mice exhibited reduced acquisition of contextual fear memory during conditioning but had an intact freezing response to conditioning context 24 h after conditioning. They also showed a reduced freezing response to extinction training, indicating facilitated extinction. Consistent with this, infusion of flufenamic acid (FFA), a TRPM2 antagonist, into the dentate gyrus (DG) of the hippocampus in fear-conditioned mice facilitated extinction of contextual fear. The enhanced extinction in Trpm2-/- and FFA-treated mice was associated with down-regulation of immediate-early genes (IEGs) including Npas4, c-Fos, Arc and Egr1 in the hippocampus after extinction training. Our results indicate that TRPM2 plays a positive role in retention of contextual fear memory by modulating neuronal activity in the hippocampus, and suggest that TRPM2 activity could potentially be targeted to strengthen extinction-based exposure therapies for post-traumatic stress disorder (PTSD).
Collapse
Affiliation(s)
- Seung Yeon Ko
- Hanyang Biomedical Research Institute, Hanyang University, Seongdong-gu, Seoul, 04763, Korea
| | - Do Gyeong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seongdong-gu, Seoul, 04763, Korea
| | - Huiju Lee
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seongdong-gu, Seoul, 04763, Korea
| | - Sung Jun Jung
- Department of Physiology, College of Medicine, Hanyang University, Seongdong-gu, Seoul, 04763, Korea.
- College of Medicine, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea.
| | - Hyeon Son
- Hanyang Biomedical Research Institute, Hanyang University, Seongdong-gu, Seoul, 04763, Korea.
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seongdong-gu, Seoul, 04763, Korea.
- College of Medicine, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea.
| |
Collapse
|
3
|
Chen C, Masotti M, Shepard N, Promes V, Tombesi G, Arango D, Manzoni C, Greggio E, Hilfiker S, Kozorovitskiy Y, Parisiadou L. LRRK2 mediates haloperidol-induced changes in indirect pathway striatal projection neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597594. [PMID: 38895420 PMCID: PMC11185612 DOI: 10.1101/2024.06.06.597594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Haloperidol is used to manage psychotic symptoms in several neurological disorders through mechanisms that involve antagonism of dopamine D2 receptors that are highly expressed in the striatum. Significant side effects of haloperidol, known as extrapyramidal symptoms, lead to motor deficits similar to those seen in Parkinson's disease and present a major challenge in clinical settings. The underlying molecular mechanisms responsible for these side effects remain poorly understood. Parkinson's disease-associated LRRK2 kinase has an important role in striatal physiology and a known link to dopamine D2 receptor signaling. Here, we systematically explore convergent signaling of haloperidol and LRRK2 through pharmacological or genetic inhibition of LRRK2 kinase, as well as knock-in mouse models expressing pathogenic mutant LRRK2 with increased kinase activity. Behavioral assays show that LRRK2 kinase inhibition ameliorates haloperidol-induced motor changes in mice. A combination of electrophysiological and anatomical approaches reveals that LRRK2 kinase inhibition interferes with haloperidol-induced changes, specifically in striatal neurons of the indirect pathway. Proteomic studies and targeted intracellular pathway analyses demonstrate that haloperidol induces a similar pattern of intracellular signaling as increased LRRK2 kinase activity. Our study suggests that LRRK2 kinase plays a key role in striatal dopamine D2 receptor signaling underlying the undesirable motor side effects of haloperidol. This work opens up new therapeutic avenues for dopamine-related disorders, such as psychosis, also furthering our understanding of Parkinson's disease pathophysiology.
Collapse
Affiliation(s)
- Chuyu Chen
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Meghan Masotti
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Nathaniel Shepard
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Vanessa Promes
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Giulia Tombesi
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Daniel Arango
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | | | | | - Sabine Hilfiker
- Department of Anesthesiology, Rutgers, New Jersey Medical School, NJ, USA
| | | | - Loukia Parisiadou
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
4
|
Ali S, Wang P, Murphy RE, Allen JA, Zhou J. Orphan GPR52 as an emerging neurotherapeutic target. Drug Discov Today 2024; 29:103922. [PMID: 38387741 DOI: 10.1016/j.drudis.2024.103922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/13/2024] [Accepted: 02/18/2024] [Indexed: 02/24/2024]
Abstract
GPR52 is a highly conserved, brain-enriched, Gs/olf-coupled orphan G protein-coupled receptor (GPCR) that controls various cyclic AMP (cAMP)-dependent physiological and pathological processes. Stimulation of GPR52 activity might be beneficial for the treatment of schizophrenia, psychiatric disorders and other human neurological diseases, whereas inhibition of its activity might provide a potential therapeutic approach for Huntington's disease. Excitingly, HTL0048149 (HTL'149), an orally available GPR52 agonist, has been advanced into phase I human clinical trials for the treatment of schizophrenia. In this concise review, we summarize the current understanding of GPR52 receptor distribution as well as its structure and functions, highlighting the recent advances in drug discovery efforts towards small-molecule GPR52 ligands. The opportunities and challenges presented by targeting GPR52 for novel therapeutics are also briefly discussed.
Collapse
Affiliation(s)
- Saghir Ali
- Chemical Biology Program, Department of Pharmacology and Toxicology, and Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Pingyuan Wang
- Chemical Biology Program, Department of Pharmacology and Toxicology, and Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ryan E Murphy
- Chemical Biology Program, Department of Pharmacology and Toxicology, and Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - John A Allen
- Chemical Biology Program, Department of Pharmacology and Toxicology, and Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, and Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
5
|
Rahman MM, Islam MR, Mim SA, Sultana N, Chellappan DK, Dua K, Kamal MA, Sharma R, Emran TB. Insights into the Promising Prospect of G Protein and GPCR-Mediated Signaling in Neuropathophysiology and Its Therapeutic Regulation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8425640. [PMID: 36187336 PMCID: PMC9519337 DOI: 10.1155/2022/8425640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/23/2022] [Indexed: 11/18/2022]
Abstract
G protein-coupled receptors (GPCRs) are intricately involved in the conversion of extracellular feedback to intracellular responses. These specialized receptors possess a crucial role in neurological and psychiatric disorders. Most nonsensory GPCRs are active in almost 90% of complex brain functions. At the time of receptor phosphorylation, a GPCR pathway is essentially activated through a G protein signaling mechanism via a G protein-coupled receptor kinase (GRK). Dopamine, an important neurotransmitter, is primarily involved in the pathophysiology of several CNS disorders; for instance, bipolar disorder, schizophrenia, Parkinson's disease, and ADHD. Since dopamine, acetylcholine, and glutamate are potent neuropharmacological targets, dopamine itself has potential therapeutic effects in several CNS disorders. GPCRs essentially regulate brain functions by modulating downstream signaling pathways. GPR6, GPR52, and GPR8 are termed orphan GPCRs because they colocalize with dopamine D1 and D2 receptors in neurons of the basal ganglia, either alone or with both receptors. Among the orphan GPCRs, the GPR52 is recognized for being an effective psychiatric receptor. Various antipsychotics like aripiprazole and quetiapine mainly target GPCRs to exert their actions. One of the most important parts of signal transduction is the regulation of G protein signaling (RGS). These substances inhibit the activation of the G protein that initiates GPCR signaling. Developing a combination of RGS inhibitors with GPCR agonists may prove to have promising therapeutic potential. Indeed, several recent studies have suggested that GPCRs represent potentially valuable therapeutic targets for various psychiatric disorders. Molecular biology and genetically modified animal model studies recommend that these enriched GPCRs may also act as potential therapeutic psychoreceptors. Neurotransmitter and neuropeptide GPCR malfunction in the frontal cortex and limbic-related regions, including the hippocampus, hypothalamus, and brainstem, is likely responsible for the complex clinical picture that includes cognitive, perceptual, emotional, and motor symptoms. G protein and GPCR-mediated signaling play a critical role in developing new treatment options for mental health issues, and this study is aimed at offering a thorough picture of that involvement. For patients who are resistant to current therapies, the development of new drugs that target GPCR signaling cascades remains an interesting possibility. These discoveries might serve as a fresh foundation for the creation of creative methods for pharmacologically useful modulation of GPCR function.
Collapse
Affiliation(s)
- Md. Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Sadia Afsana Mim
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Nasrin Sultana
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Mohammad Amjad Kamal
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
- King Fahd Medical Research Center, King Abdulaziz University, Saudi Arabia
- Enzymoics, Novel Global Community Educational Foundation, Australia
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005 Uttar Pradesh, India
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| |
Collapse
|
6
|
Present and future antipsychotic drugs: a systematic review of the putative mechanisms of action for efficacy and a critical appraisal under a translational perspective. Pharmacol Res 2022; 176:106078. [PMID: 35026403 DOI: 10.1016/j.phrs.2022.106078] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/23/2021] [Accepted: 01/07/2022] [Indexed: 01/10/2023]
Abstract
Antipsychotics represent the mainstay of schizophrenia pharmacological therapy, and their role has been expanded in the last years to mood disorders treatment. Although introduced in 1952, many years of research were required before an accurate picture of how antipsychotics work began to emerge. Despite the well-recognized characterization of antipsychotics in typical and atypical based on their liability to induce motor adverse events, their main action at dopamine D2R to elicit the "anti-psychotic" effect, as well as the multimodal action at other classes of receptors, their effects on intracellular mechanisms starting with receptor occupancy is still not completely understood. Significant lines of evidence converge on the impact of these compounds on multiple molecular signaling pathways implicated in the regulation of early genes and growth factors, dendritic spine shape, brain inflammation, and immune response, tuning overall the function and architecture of the synapse. Here we present, based on PRISMA approach, a comprehensive and systematic review of the above mechanisms under a translational perspective to disentangle those intracellular actions and signaling that may underline clinically relevant effects and represent potential targets for further innovative strategies in antipsychotic therapy.
Collapse
|
7
|
Kunst RF, Langlais AL, Barlow D, Houseknecht KL, Motyl KJ. Housing Temperature Influences Atypical Antipsychotic Drug-Induced Bone Loss in Female C57BL/6J Mice. JBMR Plus 2021; 5:e10541. [PMID: 34693191 PMCID: PMC8520062 DOI: 10.1002/jbm4.10541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/01/2021] [Accepted: 08/13/2021] [Indexed: 12/15/2022] Open
Abstract
Atypical antipsychotic (AA) drugs, such as risperidone, are associated with endocrine and metabolic side effects, including impaired bone mineral density (BMD) acquisition and increased fracture risk. We have previously shown that risperidone causes bone loss through the sympathetic nervous system and that bone loss is associated with elevated markers of thermogenesis in brown and white adipose tissue. Because rodents are normally housed in sub‐thermoneutral conditions, we wanted to test whether increasing housing temperature would protect against bone loss from risperidone. Four weeks of risperidone treatment in female C57BL/6J mice at thermoneutral (28°C) housing attenuated risperidone‐induced trabecular bone loss and led to a low‐turnover bone phenotype, with indices of both bone formation and resorption suppressed in mice with risperidone treatment at thermoneutrality, whereas indices of bone resorption were elevated by risperidone at room temperature. Protection against trabecular bone loss was not absolute, however, and additional evidence of cortical bone loss emerged in risperidone‐treated mice at thermoneutrality. Taken together, these findings suggest thermal challenge may be in part responsible for bone loss with risperidone treatment and that housing temperature should be considered when assessing bone outcomes of treatments that impact thermogenic pathways. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Roni F Kunst
- Center for Molecular Medicine Maine Medical Center Research Institute Scarborough ME USA
| | - Audrie L Langlais
- Center for Molecular Medicine Maine Medical Center Research Institute Scarborough ME USA.,Graduate School of Biomedical Sciences and Engineering, University of Maine Orono ME USA
| | - Deborah Barlow
- College of Osteopathic Medicine, University of New England Biddeford ME USA
| | | | - Katherine J Motyl
- Center for Molecular Medicine Maine Medical Center Research Institute Scarborough ME USA.,Graduate School of Biomedical Sciences and Engineering, University of Maine Orono ME USA.,Tufts University School of Medicine, Tufts University Boston MA USA
| |
Collapse
|
8
|
Marchisella F, Paladini MS, Guidi A, Begni V, Brivio P, Spero V, Calabrese F, Molteni R, Riva MA. Chronic treatment with the antipsychotic drug blonanserin modulates the responsiveness to acute stress with anatomical selectivity. Psychopharmacology (Berl) 2020; 237:1783-1793. [PMID: 32296859 DOI: 10.1007/s00213-020-05498-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 02/27/2020] [Indexed: 12/29/2022]
Abstract
RATIONALE Patients diagnosed with schizophrenia typically receive life-long treatments with antipsychotic drugs (APDs). However, the impact of chronic APDs treatment on neuroplastic mechanisms in the brain remains largely elusive. OBJECTIVE Here, we focused on blonanserin, a second-generation antipsychotic (SGA) that acts as an antagonist at dopamine D2, D3, and serotonin 5-HT2A receptors, and represents an important tool for the treatment of schizophrenia. METHODS We used rats to investigate the ability of chronic treatment blonanserin to modulate the activity of brain structures relevant for schizophrenia, under baseline conditions or in response to an acute forced swim session (FSS). We measured the expression of different immediate early genes (IEGs), including c-Fos, Arc/Arg 3.1, Zif268 and Npas4. RESULTS Blonanserin per se produced limited changes in the expression of these genes under basal conditions, while, as expected, FSS produced a significant elevation of IEGs transcription in different brain regions. The response of blonanserin-treated rats to FSS show anatomical and gene-selective differences. Indeed, the upregulation of IEGs was greatly reduced in the striatum, a brain structure enriched in dopamine receptors, whereas the upregulation of some genes (Zif268, Npas4) was largely preserved in other regions, such as the prefrontal cortex and the ventral hippocampus. CONCLUSIONS Taken together, our findings show that chronic exposure to blonanserin modulates selective IEGs with a specific anatomical profile. Moreover, the differential activation of specific brain regions under challenging conditions may contribute to specific clinical features of the drug.
Collapse
Affiliation(s)
- Francesca Marchisella
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133, Milan, Italy
| | - Maria Serena Paladini
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli 32, 20129, Milan, Italy
| | - Alice Guidi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli 32, 20129, Milan, Italy
| | - Veronica Begni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133, Milan, Italy
| | - Paola Brivio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133, Milan, Italy
| | - Vittoria Spero
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli 32, 20129, Milan, Italy
| | - Francesca Calabrese
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133, Milan, Italy
| | - Raffaella Molteni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli 32, 20129, Milan, Italy.
| | - Marco Andrea Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133, Milan, Italy
| |
Collapse
|
9
|
Omics in schizophrenia: current progress and future directions of antipsychotic treatments. JOURNAL OF BIO-X RESEARCH 2019. [DOI: 10.1097/jbr.0000000000000049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
10
|
Davies W. An Analysis of Cellular Communication Network Factor Proteins as Candidate Mediators of Postpartum Psychosis Risk. Front Psychiatry 2019; 10:876. [PMID: 31849729 PMCID: PMC6901936 DOI: 10.3389/fpsyt.2019.00876] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/06/2019] [Indexed: 12/21/2022] Open
Abstract
Postpartum (or puerperal) psychosis (PP) is a severe psychiatric condition associated with hallucinations, delusions, cognitive disorganization, and mood problems, which affects approximately 1-2 out of every 1,000 mothers shortly after childbirth. While the risk factors for, and co-morbidities of, PP are relatively well-defined, currently, the pathophysiology underlying the disorder is very poorly-specified. Here, I argue, on the basis of multiple lines of new evidence, that altered expression of the Cellular Communication Network (CCN) factor proteins (and of the heterodimerizing CCN2 and CCN3 proteins in particular), may be associated with, and possibly causal for, increased PP risk. Future preclinical and clinical studies should aim to test this hypothesis as empirical support for it would provide much-needed clues regarding the biological substrates of PP, and could point to predictive biomarkers for the condition.
Collapse
Affiliation(s)
- William Davies
- MRC Centre for Neuropsychiatric Genetics and Genomics and Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
- School of Psychology, Cardiff University, Cardiff, United Kingdom
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
11
|
Li X, Marshall PR, Leighton LJ, Zajaczkowski EL, Wang Z, Madugalle SU, Yin J, Bredy TW, Wei W. The DNA Repair-Associated Protein Gadd45γ Regulates the Temporal Coding of Immediate Early Gene Expression within the Prelimbic Prefrontal Cortex and Is Required for the Consolidation of Associative Fear Memory. J Neurosci 2019; 39:970-983. [PMID: 30545945 PMCID: PMC6363930 DOI: 10.1523/jneurosci.2024-18.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/11/2018] [Accepted: 12/04/2018] [Indexed: 02/04/2023] Open
Abstract
We have identified a member of the growth arrest and DNA damage (Gadd45) protein family, Gadd45γ, which is known to be critically involved in DNA repair, as a key player in the regulation of immediate early gene (IEG) expression underlying the consolidation of associative fear memory in adult male C57BL/6 mice. Gadd45γ temporally influences learning-induced IEG expression in the prelimbic prefrontal cortex (PLPFC) through its interaction with DNA double-strand break (DSB)-mediated changes in DNA methylation. Our findings suggest a two-hit model of experience-dependent IEG activity and learning that comprises (1) a first wave of IEG expression governed by DSBs and followed by a rapid increase in DNA methylation, and (2) a second wave of IEG expression associated with the recruitment of Gadd45γ and active DNA demethylation at the same site, which is necessary for memory consolidation.SIGNIFICANCE STATEMENT How does the pattern of immediate early gene transcription in the brain relate to the storage and accession of information, and what controls these patterns? This paper explores how Gadd45γ, a gene that is known to be involved with DNA modification and repair, regulates the temporal coding of IEGs underlying associative learning and memory. We reveal that, during fear learning, Gadd45γ serves to act as a coordinator of IEG expression and subsequent memory consolidation by directing temporally specific changes in active DNA demethylation at the promoter of plasticity-related IEGs.
Collapse
Affiliation(s)
- Xiang Li
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Paul R Marshall
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Laura J Leighton
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Esmi L Zajaczkowski
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Ziqi Wang
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Sachithrani U Madugalle
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Jiayu Yin
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Timothy W Bredy
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Wei Wei
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| |
Collapse
|
12
|
Zygmunt M, Piechota M, Rodriguez Parkitna J, Korostyński M. Decoding the transcriptional programs activated by psychotropic drugs in the brain. GENES BRAIN AND BEHAVIOR 2018; 18:e12511. [PMID: 30084543 DOI: 10.1111/gbb.12511] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 07/25/2018] [Accepted: 08/03/2018] [Indexed: 01/01/2023]
Abstract
Analysis of drug-induced gene expression in the brain has long held the promise of revealing the molecular mechanisms of drug actions as well as predicting their long-term clinical efficacy. However, despite some successes, this promise has yet to be fulfilled. Here, we present an overview of the current state of understanding of drug-induced gene expression in the brain and consider the obstacles to achieving a robust prediction of the properties of psychoactive compounds based on gene expression profiles. We begin with a comprehensive overview of the mechanisms controlling drug-inducible transcription and the complexity resulting from expression of noncoding RNAs and alternative gene isoforms. Particular interest is placed on studies that examine the associations within drug classes with regard to the effects on gene transcription, alterations in cell signaling and neuropharmacological drug properties. While the ability of gene expression signatures to distinguish specific clinical classes of psychotropic and addictive drugs remains unclear, some reports show that under specific constraints, drug properties can be predicted based on gene expression. Such signatures offer a simple and effective way to classify psychotropic drugs and screen novel psychoactive compounds. Finally, we note that the amount of data regarding molecular programs activated in the brain by drug treatment has grown exponentially in recent years and that future advances may therefore come in large part from integrating the currently available high-throughput data sets.
Collapse
Affiliation(s)
- Magdalena Zygmunt
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Krakow, Poland
| | - Marcin Piechota
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Krakow, Poland
| | - Jan Rodriguez Parkitna
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Krakow, Poland
| | - Michał Korostyński
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Krakow, Poland
| |
Collapse
|
13
|
Maruyama M, Hotta N, Nio Y, Hamagami K, Nagi T, Funata M, Sakamoto J, Nakakariya M, Amano N, Nishida M, Okawa T, Arikawa Y, Sasaki S, Kasai S, Nagisa Y, Habata Y, Mori M. Bombesin receptor subtype-3-expressing neurons regulate energy homeostasis through a novel neuronal pathway in the hypothalamus. Brain Behav 2018; 8:e00881. [PMID: 29568682 PMCID: PMC5853643 DOI: 10.1002/brb3.881] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 10/16/2017] [Accepted: 10/19/2017] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVES Bombesin receptor subtype-3 (BRS-3) has been suggested to play a potential role in energy homeostasis. However, the physiological mechanism of BRS-3 on energy homeostasis remains unknown. Thus, we investigated the BRS-3-mediated neuronal pathway involved in food intake and energy expenditure. MATERIALS AND METHODS Expression of BRS-3 in the rat brain was histologically examined. The BRS-3 neurons activated by refeeding-induced satiety or a BRS-3 agonist were identified by c-Fos immunostaining. We also analyzed expression changes in feeding-relating peptides in the brain of fasted rats administered with the BRS-3 agonist. RESULTS In the paraventricular hypothalamic nucleus (PVH), dorsomedial hypothalamic nucleus (DMH), and medial preoptic area (MPA), strong c-Fos induction was observed in the BRS-3 neurons especially in PVH after refeeding. However, the BRS-3 neurons in the PVH did not express feeding-regulating peptides, while the BRS-3 agonist administration induced c-Fos expression in the DMH and MPA, which were not refeeding-sensitive, as well as in the PVH. The BRS-3 agonist administration changed the Pomc and Cart mRNA level in several brain regions of fasted rats. CONCLUSION These results suggest that BRS-3 neurons in the PVH are a novel functional subdivision in the PVH that regulates feeding behavior. As the MPA and DMH are reportedly involved in thermoregulation and energy metabolism, the BRS-3 neurons in the MPA/DMH might mediate the energy expenditure control. POMC and CART may contribute to BRS-3 neuron-mediated energy homeostasis regulation. In summary, BRS-3-expressing neurons could regulate energy homeostasis through a novel neuronal pathway.
Collapse
Affiliation(s)
- Minoru Maruyama
- Cardiovascular and Metabolic Drug Discovery Unit Takeda Pharmaceutical Company Limited Kanagawa Japan
| | - Natsu Hotta
- Cardiovascular and Metabolic Drug Discovery Unit Takeda Pharmaceutical Company Limited Kanagawa Japan
| | - Yasunori Nio
- Extra Value Generation & General Medicine Drug Discovery Unit Takeda Pharmaceutical Company Limited Kanagawa Japan
| | - Kenichi Hamagami
- Cardiovascular and Metabolic Drug Discovery Unit Takeda Pharmaceutical Company Limited Kanagawa Japan
| | - Toshimi Nagi
- Central Nervous System Drug Discovery Unit Takeda Pharmaceutical Company Limited Kanagawa Japan
| | - Masaaki Funata
- Biomolecular Research Laboratories Takeda Pharmaceutical Company Limited Kanagawa Japan
| | - Junichi Sakamoto
- Biomolecular Research Laboratories Takeda Pharmaceutical Company Limited Kanagawa Japan
| | - Masanori Nakakariya
- Drug Metabolism and Pharmacokinetics Research LaboratoriesTakeda Pharmaceutical Company Limited Kanagawa Japan
| | - Nobuyuki Amano
- Drug Metabolism and Pharmacokinetics Research LaboratoriesTakeda Pharmaceutical Company Limited Kanagawa Japan
| | - Mayumi Nishida
- Integrated Technology Research Laboratories Takeda Pharmaceutical Company Limited Kanagawa Japan
| | - Tomohiro Okawa
- Central Nervous System Drug Discovery Unit Takeda Pharmaceutical Company Limited Kanagawa Japan
| | - Yasuyoshi Arikawa
- Central Nervous System Drug Discovery Unit Takeda Pharmaceutical Company Limited Kanagawa Japan
| | - Shinobu Sasaki
- Medicinal Chemistry Research Laboratories Pharmaceutical Research Division Takeda Pharmaceutical Company Limited Kanagawa Japan
| | - Shizuo Kasai
- Cardiovascular and Metabolic Drug Discovery Unit Takeda Pharmaceutical Company Limited Kanagawa Japan
| | - Yasutaka Nagisa
- Cardiovascular and Metabolic Drug Discovery Unit Takeda Pharmaceutical Company Limited Kanagawa Japan.,Present address: CVM Marketing Japan Pharma Business UnitTakeda Pharmaceutical Co. Ltd.12-10, Nihonbashi 2-Chome, Chuo-ku Tokyo 103-8686 Japan
| | - Yugo Habata
- Cardiovascular and Metabolic Drug Discovery Unit Takeda Pharmaceutical Company Limited Kanagawa Japan.,Present address: Foods & Nutrients Yamanashi Gakuin Junior College Sakaori 2-4-5, Kofu Yamanashi 400-8575 Japan
| | - Masaaki Mori
- Cardiovascular and Metabolic Drug Discovery Unit Takeda Pharmaceutical Company Limited Kanagawa Japan
| |
Collapse
|
14
|
de Bartolomeis A, Buonaguro EF, Latte G, Rossi R, Marmo F, Iasevoli F, Tomasetti C. Immediate-Early Genes Modulation by Antipsychotics: Translational Implications for a Putative Gateway to Drug-Induced Long-Term Brain Changes. Front Behav Neurosci 2017; 11:240. [PMID: 29321734 PMCID: PMC5732183 DOI: 10.3389/fnbeh.2017.00240] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 11/22/2017] [Indexed: 12/12/2022] Open
Abstract
An increasing amount of research aims at recognizing the molecular mechanisms involved in long-lasting brain architectural changes induced by antipsychotic treatments. Although both structural and functional modifications have been identified following acute antipsychotic administration in humans, currently there is scarce knowledge on the enduring consequences of these acute changes. New insights in immediate-early genes (IEGs) modulation following acute or chronic antipsychotic administration may help to fill the gap between primary molecular response and putative long-term changes. Moreover, a critical appraisal of the spatial and temporal patterns of IEGs expression may shed light on the functional "signature" of antipsychotics, such as the propensity to induce motor side effects, the potential neurobiological mechanisms underlying the differences between antipsychotics beyond D2 dopamine receptor affinity, as well as the relevant effects of brain region-specificity in their mechanisms of action. The interest for brain IEGs modulation after antipsychotic treatments has been revitalized by breakthrough findings such as the role of early genes in schizophrenia pathophysiology, the involvement of IEGs in epigenetic mechanisms relevant for cognition, and in neuronal mapping by means of IEGs expression profiling. Here we critically review the evidence on the differential modulation of IEGs by antipsychotics, highlighting the association between IEGs expression and neuroplasticity changes in brain regions impacted by antipsychotics, trying to elucidate the molecular mechanisms underpinning the effects of this class of drugs on psychotic, cognitive and behavioral symptoms.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University School of Medicine "Federico II", Naples, Italy
| | - Elisabetta F Buonaguro
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University School of Medicine "Federico II", Naples, Italy
| | - Gianmarco Latte
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University School of Medicine "Federico II", Naples, Italy
| | - Rodolfo Rossi
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University School of Medicine "Federico II", Naples, Italy
| | - Federica Marmo
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University School of Medicine "Federico II", Naples, Italy
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University School of Medicine "Federico II", Naples, Italy
| | - Carmine Tomasetti
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University School of Medicine "Federico II", Naples, Italy
| |
Collapse
|
15
|
Nishiyama K, Suzuki H, Harasawa T, Suzuki N, Kurimoto E, Kawai T, Maruyama M, Komatsu H, Sakuma K, Shimizu Y, Shimojo M. FTBMT, a Novel and Selective GPR52 Agonist, Demonstrates Antipsychotic-Like and Procognitive Effects in Rodents, Revealing a Potential Therapeutic Agent for Schizophrenia. J Pharmacol Exp Ther 2017; 363:253-264. [PMID: 28851764 DOI: 10.1124/jpet.117.242925] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/23/2017] [Indexed: 01/23/2023] Open
Abstract
GPR52 is a Gs-coupled G protein-coupled receptor that is predominantly expressed in the striatum and nucleus accumbens (NAc) and was recently proposed as a potential therapeutic target for schizophrenia. In the current study, we investigated the in vitro and in vivo pharmacologic activities of a novel GPR52 agonist, 4-(3-(3-fluoro-5-(trifluoromethyl)benzyl)-5-methyl-1H-1,2,4-triazol-1-yl)-2-methylbenzamide (FTBMT). FTBMT functioned as a selective GPR52 agonist in vitro and in vivo, as demonstrated by the activation of Camp signaling in striatal neurons. FTBMT inhibited MK-801-induced hyperactivity, an animal model for acute psychosis, without causing catalepsy in mice. The c-fos expression also revealed that FTBMT preferentially induced neuronal activation in the shell of the Nac compared with the striatum, thereby supporting its antipsychotic-like activity with less catalepsy. Furthermore, FTBMT improved recognition memory in a novel object-recognition test and attenuated MK-801-induced working memory deficits in a radial arm maze test in rats. These recognitive effects were supported by the results of FTBMT-induced c-fos expression in the brain regions related to cognition, including the medial prefrontal cortex, entorhinal cortex, and hippocampus. Taken together, these findings suggest that FTBMT shows antipsychotic and recognitive properties without causing catalepsy in rodents. Given its unique pharmacologic profile, which differs from that of current antipsychotics, FTBMT may provide a new therapeutic option for the treatment of positive and cognitive symptoms of schizophrenia.
Collapse
MESH Headings
- Animals
- Antipsychotic Agents/chemistry
- Antipsychotic Agents/pharmacology
- Antipsychotic Agents/therapeutic use
- Benzamides/chemistry
- Benzamides/pharmacology
- Benzamides/therapeutic use
- CHO Cells
- Cricetinae
- Cricetulus
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Female
- Locomotion/drug effects
- Locomotion/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred ICR
- Mice, Knockout
- Nootropic Agents/chemistry
- Nootropic Agents/pharmacology
- Nootropic Agents/therapeutic use
- Nucleus Accumbens/drug effects
- Nucleus Accumbens/physiology
- Organ Culture Techniques
- Rats
- Rats, Long-Evans
- Rats, Sprague-Dawley
- Rats, Wistar
- Receptors, G-Protein-Coupled/agonists
- Receptors, G-Protein-Coupled/physiology
- Schizophrenia/drug therapy
- Treatment Outcome
- Triazoles/chemistry
- Triazoles/pharmacology
- Triazoles/therapeutic use
Collapse
Affiliation(s)
- Keiji Nishiyama
- CNS Drug Discovery Unit, Research (K.N., H.S., T.H., N.S., E.K., T.K., M.M., H.K., Y.S., M.S.) and Regenerative Medicine Unit (K.S.), Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hirobumi Suzuki
- CNS Drug Discovery Unit, Research (K.N., H.S., T.H., N.S., E.K., T.K., M.M., H.K., Y.S., M.S.) and Regenerative Medicine Unit (K.S.), Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Toshiya Harasawa
- CNS Drug Discovery Unit, Research (K.N., H.S., T.H., N.S., E.K., T.K., M.M., H.K., Y.S., M.S.) and Regenerative Medicine Unit (K.S.), Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Noriko Suzuki
- CNS Drug Discovery Unit, Research (K.N., H.S., T.H., N.S., E.K., T.K., M.M., H.K., Y.S., M.S.) and Regenerative Medicine Unit (K.S.), Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Emi Kurimoto
- CNS Drug Discovery Unit, Research (K.N., H.S., T.H., N.S., E.K., T.K., M.M., H.K., Y.S., M.S.) and Regenerative Medicine Unit (K.S.), Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Takayuki Kawai
- CNS Drug Discovery Unit, Research (K.N., H.S., T.H., N.S., E.K., T.K., M.M., H.K., Y.S., M.S.) and Regenerative Medicine Unit (K.S.), Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Minoru Maruyama
- CNS Drug Discovery Unit, Research (K.N., H.S., T.H., N.S., E.K., T.K., M.M., H.K., Y.S., M.S.) and Regenerative Medicine Unit (K.S.), Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hidetoshi Komatsu
- CNS Drug Discovery Unit, Research (K.N., H.S., T.H., N.S., E.K., T.K., M.M., H.K., Y.S., M.S.) and Regenerative Medicine Unit (K.S.), Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Kensuke Sakuma
- CNS Drug Discovery Unit, Research (K.N., H.S., T.H., N.S., E.K., T.K., M.M., H.K., Y.S., M.S.) and Regenerative Medicine Unit (K.S.), Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yuji Shimizu
- CNS Drug Discovery Unit, Research (K.N., H.S., T.H., N.S., E.K., T.K., M.M., H.K., Y.S., M.S.) and Regenerative Medicine Unit (K.S.), Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Masato Shimojo
- CNS Drug Discovery Unit, Research (K.N., H.S., T.H., N.S., E.K., T.K., M.M., H.K., Y.S., M.S.) and Regenerative Medicine Unit (K.S.), Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
16
|
Xavier RM, Vorderstrasse A. Genetic Basis of Positive and Negative Symptom Domains in Schizophrenia. Biol Res Nurs 2017; 19:559-575. [PMID: 28691507 DOI: 10.1177/1099800417715907] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Schizophrenia is a highly heritable disorder, the genetic etiology of which has been well established. Yet despite significant advances in genetics research, the pathophysiological mechanisms of this disorder largely remain unknown. This gap has been attributed to the complexity of the polygenic disorder, which has a heterogeneous clinical profile. Examining the genetic basis of schizophrenia subphenotypes, such as those based on particular symptoms, is thus a useful strategy for decoding the underlying mechanisms. This review of literature examines the recent advances (from 2011) in genetic exploration of positive and negative symptoms in schizophrenia. We searched electronic databases PubMed, Web of Science, and Cumulative Index to Nursing and Allied Health Literature using key words schizophrenia, symptoms, positive symptoms, negative symptoms, cognition, genetics, genes, genetic predisposition, and genotype in various combinations. We identified 115 articles, which are included in the review. Evidence from these studies, most of which are genetic association studies, identifies shared and unique gene associations for the symptom domains. Genes associated with neurotransmitter systems and neuronal development/maintenance primarily constitute the shared associations. Needed are studies that examine the genetic basis of specific symptoms within the broader domains in addition to functional mechanisms. Such investigations are critical to developing precision treatment and care for individuals afflicted with schizophrenia.
Collapse
Affiliation(s)
| | - Allison Vorderstrasse
- 2 Duke Center for Applied Genomics and Precision Medicine, Duke University School of Nursing, Durham, NC, USA
| |
Collapse
|
17
|
Davies W. Understanding the pathophysiology of postpartum psychosis: Challenges and new approaches. World J Psychiatry 2017; 7:77-88. [PMID: 28713685 PMCID: PMC5491479 DOI: 10.5498/wjp.v7.i2.77] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/25/2017] [Accepted: 04/20/2017] [Indexed: 02/05/2023] Open
Abstract
Postpartum psychosis is a severe psychiatric condition which affects 1-2 of every 1000 mothers shortly after childbirth. Whilst there is convincing evidence that the condition is precipitated by a complex combination of biological and environmental factors, as yet the pathophysiological mechanisms remain extremely poorly defined. Here, I critically review approaches that have been, or are being, employed to identify and characterise such mechanisms; I also review a recent animal model approach, and describe a novel biological risk model that it suggests. Clarification of biological risk mechanisms underlying disorder risk should permit the identification of relevant predictive biomarkers which will ensure that “at risk” subjects receive prompt clinical intervention if required.
Collapse
|
18
|
Waters S, Svensson P, Kullingsjö J, Pontén H, Andreasson T, Sunesson Y, Ljung E, Sonesson C, Waters N. In Vivo Systems Response Profiling and Multivariate Classification of CNS Active Compounds: A Structured Tool for CNS Drug Discovery. ACS Chem Neurosci 2017; 8:785-797. [PMID: 27997108 DOI: 10.1021/acschemneuro.6b00371] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This paper describes the application of in vivo systems response profiling in CNS drug discovery by a process referred to as the Integrative Screening Process. The biological response profile, treated as an array, is used as major outcome for selection of candidate drugs. Dose-response data, including ex vivo brain monoaminergic biomarkers and behavioral descriptors, are systematically collected and analyzed by principal component analysis (PCA) and partial least-squares (PLS) regression, yielding multivariate characterization across compounds. The approach is exemplified by assessing a new class of CNS active compounds, the dopidines, compared to other monoamine modulating compounds including antipsychotics, antidepressants, and procognitive agents. Dopidines display a distinct phenotypic profile which has prompted extensive further preclinical and clinical investigations. In summary, in vivo profiles of CNS compounds are mapped, based on dose response studies in the rat. Applying a systematic and standardized work-flow, a database of in vivo systems response profiles is compiled, enabling comparisons and classification. This creates a framework for translational mapping, a crucial component in CNS drug discovery.
Collapse
Affiliation(s)
- Susanna Waters
- Department
of Pharmacology, Gothenburg University, SE-405 30 Gothenburg, Sweden
- Integrative Research Laboratories Sweden AB, Gothenburg SE-413 46, Sweden
| | - Peder Svensson
- Integrative Research Laboratories Sweden AB, Gothenburg SE-413 46, Sweden
| | - Johan Kullingsjö
- Integrative Research Laboratories Sweden AB, Gothenburg SE-413 46, Sweden
| | - Henrik Pontén
- Department
of Pharmacology, Gothenburg University, SE-405 30 Gothenburg, Sweden
| | | | | | - Elisabeth Ljung
- Integrative Research Laboratories Sweden AB, Gothenburg SE-413 46, Sweden
| | - Clas Sonesson
- Integrative Research Laboratories Sweden AB, Gothenburg SE-413 46, Sweden
| | - Nicholas Waters
- Integrative Research Laboratories Sweden AB, Gothenburg SE-413 46, Sweden
| |
Collapse
|
19
|
Humby T, Cross ES, Messer L, Guerrero S, Davies W. A pharmacological mouse model suggests a novel risk pathway for postpartum psychosis. Psychoneuroendocrinology 2016; 74:363-370. [PMID: 27728876 PMCID: PMC5094271 DOI: 10.1016/j.psyneuen.2016.09.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/08/2016] [Accepted: 09/21/2016] [Indexed: 01/04/2023]
Abstract
Postpartum psychosis (PP) is a severe psychiatric disorder affecting a small proportion of new mothers shortly after childbirth. The molecular pathophysiology underlying the disorder is currently poorly understood, and there are no amenable animal models for the condition; maternal deficiency for the enzyme steroid sulfatase has been proposed as a potential risk mechanism. Here we show that inhibition of steroid sulfatase with 667-COUMATE (10mg/kg p.o.) in new mouse mothers results in behavioural abnormalities that can be partially alleviated by the administration of the clinically-efficacious antipsychotic ziprasidone (0.3-1.0mg/kg i.p.). The pattern of behavioural abnormalities in 667-COUMATE-treated mice implicated a genetic substrate at 21-23cM on chromosome 15; of the 17 genes within this chromosomal interval, only one (Nov/Ccn3) was significantly differentially expressed in the brains of vehicle and 667-COUMATE-treated mice. Two additional members of the Ccn family (Ccn2/Ctgf and Ccn4/Wisp1) were also significantly differentially expressed between the two groups, as were three further genes co-expressed with Nov/Ccn3 in brain (Arhgdig) or previously implicated in disorder risk by clinical studies (Adcy8 and Ccl2). The expression of Nov/Ccn3, but not of the other differentially-expressed genes, could be normalised by ziprasidone administration (1.0mg/kg). NOV/CCN3 lies directly under a linkage peak for PP risk at 8q24, and the associated protein possesses numerous characteristics that make it an excellent candidate mediator of PP risk. Our data suggest the 667-COUMATE-treated mouse as a model for PP with some degree of face, construct, and predictive validity, and implicate a novel, and biologically-plausible, molecular risk pathway for PP.
Collapse
Affiliation(s)
- Trevor Humby
- School of Psychology, Cardiff University, Tower Building, 70, Park Place, Cardiff, CF10 3AT, UK; Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK; Medical Research Council Centre for Neuropsychiatric Genetics and Genomics and Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.
| | - Ellen S. Cross
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Lauren Messer
- School of Psychology, Cardiff University, Tower Building, 70, Park Place, Cardiff, CF10 3AT, UK.
| | - Silvia Guerrero
- University of Barcelona, Gran Via de les Corts Catalanes, 585 08007 Barcelona, Spain.
| | - William Davies
- School of Psychology, Cardiff University, Tower Building, 70, Park Place, Cardiff, CF10 3AT, UK; Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK; Medical Research Council Centre for Neuropsychiatric Genetics and Genomics and Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.
| |
Collapse
|
20
|
Chen Y, Brooks MJ, Gieser L, Swaroop A, Palczewski K. Transcriptome profiling of NIH3T3 cell lines expressing opsin and the P23H opsin mutant identifies candidate drugs for the treatment of retinitis pigmentosa. Pharmacol Res 2016; 115:1-13. [PMID: 27838510 DOI: 10.1016/j.phrs.2016.10.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 10/18/2016] [Accepted: 10/26/2016] [Indexed: 01/01/2023]
Abstract
Mammalian cells are commonly employed in screening assays to identify active compounds that could potentially affect the progression of different human diseases including retinitis pigmentosa (RP), a class of inherited diseases causing retinal degeneration with compromised vision. Using transcriptome analysis, we compared NIH3T3 cells expressing wildtype (WT) rod opsin with a retinal disease-causing single P23H mutation. Surprisingly, heterologous expression of WT opsin in NIH3T3 cells caused more than a 2-fold change in 783 out of 16,888 protein coding transcripts. The perturbed genes encoded extracellular matrix proteins, growth factors, cytoskeleton proteins, glycoproteins and metalloproteases involved in cell adhesion, morphology and migration. A different set of 347 transcripts was either up- or down-regulated when the P23H mutant opsin was expressed suggesting an altered molecular perturbation compared to WT opsin. Transcriptome perturbations elicited by drug candidates aimed at mitigating the effects of the mutant protein revealed that different drugs targeted distinct molecular pathways that resulted in a similar phenotype selected by a cell-based high-throughput screen. Thus, transcriptome profiling can provide essential information about the therapeutic potential of a candidate drug to restore normal gene expression in pathological conditions.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Matthew J Brooks
- Neurobiology-Neurodegeneration & Repair Laboratory (N-NRL), National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda, MD 20892, United States
| | - Linn Gieser
- Neurobiology-Neurodegeneration & Repair Laboratory (N-NRL), National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda, MD 20892, United States
| | - Anand Swaroop
- Neurobiology-Neurodegeneration & Repair Laboratory (N-NRL), National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda, MD 20892, United States
| | - Krzysztof Palczewski
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States.
| |
Collapse
|
21
|
A novel anesthesia regime enables neurofunctional studies and imaging genetics across mouse strains. Sci Rep 2016; 6:24523. [PMID: 27080031 PMCID: PMC4832200 DOI: 10.1038/srep24523] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 03/30/2016] [Indexed: 12/18/2022] Open
Abstract
Functional magnetic resonance imaging (fMRI) has revolutionized neuroscience by opening a unique window that allows neurocircuitry function and pathological alterations to be probed non-invasively across brain disorders. Here we report a novel sustainable anesthesia procedure for small animal neuroimaging that overcomes shortcomings of anesthetics commonly used in rodent fMRI. The significantly improved preservation of cerebrovascular dynamics enhances sensitivity to neural activity changes for which it serves as a proxy in fMRI readouts. Excellent cross-species/strain applicability provides coherence among preclinical findings and is expected to improve translation to clinical fMRI investigations. The novel anesthesia procedure based on the GABAergic anesthetic etomidate was extensively validated in fMRI studies conducted in a range of genetically engineered rodent models of autism and strains commonly used for transgenic manipulations. Etomidate proved effective, yielded long-term stable physiology with basal cerebral blood flow of ~0.5 ml/g/min and full recovery. Cerebrovascular responsiveness of up to 180% was maintained as demonstrated with perfusion- and BOLD-based fMRI upon hypercapnic, pharmacological and sensory stimulation. Hence, etomidate lends itself as an anesthetic-of-choice for translational neuroimaging studies across rodent models of brain disorders.
Collapse
|
22
|
Malik AR, Liszewska E, Jaworski J. Matricellular proteins of the Cyr61/CTGF/NOV (CCN) family and the nervous system. Front Cell Neurosci 2015; 9:237. [PMID: 26157362 PMCID: PMC4478388 DOI: 10.3389/fncel.2015.00237] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Accepted: 06/12/2015] [Indexed: 12/22/2022] Open
Abstract
Matricellular proteins are secreted proteins that exist at the border of cells and the extracellular matrix (ECM). However, instead of playing a role in structural integrity of the ECM, these proteins, that act as modulators of various surface receptors, have a regulatory function and instruct a multitude of cellular responses. Among matricellular proteins are members of the Cyr61/CTGF/NOV (CCN) protein family. These proteins exert their activity by binding directly to integrins and heparan sulfate proteoglycans and activating multiple intracellular signaling pathways. CCN proteins also influence the activity of growth factors and cytokines and integrate their activity with integrin signaling. At the cellular level, CCN proteins regulate gene expression and cell survival, proliferation, differentiation, senescence, adhesion, and migration. To date, CCN proteins have been extensively studied in the context of osteo- and chondrogenesis, angiogenesis, and carcinogenesis, but the expression of these proteins is also observed in a variety of tissues. The role of CCN proteins in the nervous system has not been systematically studied or described. Thus, the major aim of this review is to introduce the CCN protein family to the neuroscience community. We first discuss the structure, interactions, and cellular functions of CCN proteins and then provide a detailed review of the available data on the neuronal expression and contribution of CCN proteins to nervous system development, function, and pathology.
Collapse
Affiliation(s)
- Anna R Malik
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology Warsaw, Poland
| | - Ewa Liszewska
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology Warsaw, Poland
| | - Jacek Jaworski
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology Warsaw, Poland
| |
Collapse
|
23
|
Novel Therapeutic GPCRs for Psychiatric Disorders. Int J Mol Sci 2015; 16:14109-21. [PMID: 26101869 PMCID: PMC4490542 DOI: 10.3390/ijms160614109] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 05/25/2015] [Accepted: 06/09/2015] [Indexed: 02/04/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are the most common targets of the neuropharmacological drugs in the central nervous system (CNS). GPCRs are activated by manifold neurotransmitters, and their activation in turn evokes slow synaptic transmission. They are deeply involved in multiple neurological and psychiatric disorders such as Parkinson’s disease and schizophrenia. In the brain, the striatum is strongly innervated by the ventral tegmental area (VTA) and plays a central role in manifestation of psychiatric disorders. Recently, anatomical and comprehensive transcriptome analysis of the non-odorant GPCR superfamily revealed that the orphan GPCRs GPR88, GPR6, and GPR52, as well as dopamine D1 and D2 receptors and the adenosine A2a receptor, are the most highly enriched in the rodent striatum. Genetically engineered animal models and molecular biological studies have suggested that these striatally enriched GPCRs have a potential to be therapeutic psychiatric receptors. This review summarizes the current understanding of the therapeutic GPCR candidates for psychiatric disorders.
Collapse
|