1
|
Vinnakota C, Hudson MR, Ikeda K, Ide S, Mishina M, Sundram S, Jones NC, Hill RA. Effects of NMDA receptor antagonists on working memory and gamma oscillations, and the mediating role of the GluN2D subunit. Neuropsychopharmacology 2025:10.1038/s41386-025-02129-9. [PMID: 40374854 DOI: 10.1038/s41386-025-02129-9] [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: 01/02/2025] [Revised: 05/04/2025] [Accepted: 05/06/2025] [Indexed: 05/18/2025]
Abstract
Working memory relies on synchronised network oscillations involving complex interplay between pyramidal cells and GABAergic interneurons. NMDA receptor (NMDAR) antagonists influence both network oscillations and working memory, but the relationship between these two consequences has not been elucidated. This study aimed to determine the effect of NMDAR antagonists on network oscillations during a working memory task in mice, and the contribution of the GluN2D receptor subunit. After training wildtype (WT) and GluN2D-knockout (KO) mice on the Trial-Unique-Non-match to Location (TUNL) touchscreen task of working memory, recording electrodes were implanted into the prefrontal cortex (PFC) and hippocampus. Mice were challenged with either (S)-ketamine (30 mg/kg), (R)-ketamine (30 mg/kg), phencyclidine (PCP, 1 mg/kg), MK-801 (0.3 mg/kg) or saline prior to TUNL testing while simultaneous local field potential recordings were acquired. PCP disrupted working memory accuracy in WT (p = 0.001) but not GluN2D-KO mice (p = 0.79). MK-801 (p < 0.0001), (S)-ketamine (p < 0.0001) and (R)-ketamine (p = 0.007) disrupted working memory accuracy in both genotypes. PCP increased baseline hippocampal gamma (30-80 Hz) power in WT (p = 0.0015) but not GluN2D-KO mice (p = 0.92). All drugs increased baseline gamma power in the PFC in both genotypes (p < 0.05). Low gamma was induced during the maintenance phase of the TUNL task and increased when mice correctly completed the task (p = 0.024). This response-dependent increase in low gamma was disrupted by all drugs. In summary, PCP action involves the GluN2D subunit of the NMDA receptor in the hippocampus to alter baseline gamma power and working memory. Task-induced low gamma activity during maintenance aligns with task performance, and is disrupted by all NMDAR antagonists.
Collapse
Affiliation(s)
- Chitra Vinnakota
- Department of Psychiatry, Monash University, Clayton, VIC, 3168, Australia
| | - Matthew R Hudson
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan
- Department of Neuropsychopharmacology, National Center of Neurology and Psychiatry, Tokyo, 187-8553, Japan
| | - Soichiro Ide
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan
| | - Masayoshi Mishina
- Brain Science Laboratory, The Research Organization of Science and Technology, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, Japan
| | - Suresh Sundram
- Department of Psychiatry, Monash University, Clayton, VIC, 3168, Australia
- Mental Health Program, Monash Health, Clayton, VIC, 3168, Australia
| | - Nigel C Jones
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia.
- Department of Neurology, The Alfred Hospital, Commercial Road, Melbourne, VIC, 3004, Australia.
| | - Rachel Anne Hill
- Department of Psychiatry, Monash University, Clayton, VIC, 3168, Australia.
| |
Collapse
|
2
|
Feng YF, Zeng ZK, Ni Y, Hu Y, Yang KX, Cai F, Zhou QM, Chen M, Zhu XN, Chen S, Hu J. Parvalbumin neurons mediate neurological phenotypes of anti-NMDAR encephalitis. Brain 2025; 148:1652-1664. [PMID: 40071389 PMCID: PMC12073974 DOI: 10.1093/brain/awae374] [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: 06/28/2024] [Revised: 09/17/2024] [Accepted: 10/08/2024] [Indexed: 05/15/2025] Open
Abstract
Patients with anti-N-methyl-D-aspartate receptor (anti-NMDAR) encephalitis, often present with severe psychiatric symptoms, yet the neuropathological mechanisms underlying their cognitive deficits remain insufficiently understood. In this study, we constructed an animal model using anti-NMDAR IgG purified from the serum of patients with anti-NMDAR encephalitis, and we used IgG obtained from healthy individuals as a control. Daily administration of anti-NMDAR IgG into the medial prefrontal cortex (mPFC) of mice for 7 days resulted in cognitive impairments resembling clinical symptoms, which spontaneously resolved 30 days after discontinuing the injections. Immunohistochemical staining and electrophysiological testing of parvalbumin neurons in the mPFC treated with anti-NMDAR IgG revealed significant cellular morphological damage, reduced excitability, synaptic dysfunction and a loss of NMDAR antagonist-induced gamma oscillations. Application of optogenetic and pharmacogenetic techniques to activate parvalbumin neurons in the mPFC successfully reversed the cognitive impairments observed in the anti-NMDAR-IgG-treated mice. Single-cell sequencing of anti-NMDAR-IgG-treated parvalbumin neurons identified differentially expressed genes and pathways related to synapses and neuronal development, offering potential targets for therapeutic intervention. Additionally, we showed that these alterations in parvalbumin neurons were not confined to the mPFC, as similar changes were detected in the hippocampus after anti-NMDAR IgG injections. In summary, our findings elucidate distinct alterations in parvalbumin neurons during the pathogenesis of anti-NMDAR encephalitis, providing preclinical rationale for exploring approaches to modulate parvalbumin neuronal function to treat anti-NMDAR encephalitis.
Collapse
Affiliation(s)
- Yi-Fan Feng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zi-Ke Zeng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - You Ni
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yue Hu
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ke-Xin Yang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Fang Cai
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qin-Ming Zhou
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ming Chen
- MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Xiao-Na Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Sheng Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
3
|
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
|
4
|
Liu S, Wang M, Han W, Chen A, Liu X, Liu K, Li X, Chen Y, Zhang L, Liu Q, Guo X, Wang X, Kang N, Han Y, Li Y, Su X, Lv L, Liu B, Li W, Yang Y. Prediction of antipsychotic drug efficacy for schizophrenia treatment based on neural features of the resting-state functional connectome. Transl Psychiatry 2025; 15:137. [PMID: 40210875 PMCID: PMC11985992 DOI: 10.1038/s41398-025-03355-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/12/2025] Open
Abstract
Neuroimaging studies have identified a large number of biomarkers associated with schizophrenia (SZ), but there is still a lack of biomarkers that can predict the efficacy of antipsychotic medication in SZ patients. The aim of this study was to identify neuroimaging biomarkers of antipsychotic drug response among features of the resting-state connectome. Resting-state functional magnetic resonance scans were acquired from a discovery cohort of 105 patients with SZ at baseline and after 8 weeks of antipsychotic medication treatment. Baseline clinical status and post-treatment outcome were assessed using the Positive and Negative Symptom Scale (PANSS), and clinical improvement was rated by the total score reduction. Based on acquired imaging data, a resting-state functional connectivity matrix was constructed for each patient, and a connectome-based predictive model was subsequently established and trained to predict individual PANSS total score reduction. Model performance was assessed by calculating Pearson correlation coefficients between predicted and true score reduction with leave-one-out cross-validation. Finally, the generalizability of the model was tested using an independent validation cohort of 52 SZ patients. The model incorporating resting-state connectome characteristics predicted individual treatment outcomes in both the discovery cohort (prediction vs. truth r = 0.59, mean squared error (MSE) = 0.021) and validation cohort (r = 0.41, MSE = 0.036). The model identified four positive features and eight negative features, which were respectively correlated positively and negatively with PANSS total score reduction. Among these positive features, the specific connections within the parietal lobe played a crucial role in the model's predictive performance. As for the negative features, they included the frontoparietal control network and the cerebello-thalamo-cortical connections. This study discovered and validated a set of functional features based on resting-state connectome, where higher connectivity of positive features and lower connectivity of negative features at baseline were associated with a higher reduction rate of PANSS total score in patients and a better therapeutic effect. These functional features can be used to predict the PANSS total score reduction rate of SZ patients through a model. Clinical doctors can potentially infer the individual treatment response of antipsychotic medication treatment for patients based on the predicted results.
Collapse
Affiliation(s)
- Song Liu
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Meng Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
| | - Weiyi Han
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Anran Chen
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Xuzhen Liu
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Kang Liu
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Xue Li
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Yi Chen
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Luwen Zhang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Qing Liu
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Xiaoge Guo
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Xiujuan Wang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Ning Kang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Yong Han
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Yuanbo Li
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Xi Su
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Luxian Lv
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Bing Liu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
| | - Wenqiang Li
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China.
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China.
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China.
- Brain Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, 450000, China.
| | - Yongfeng Yang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China.
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China.
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China.
- Brain Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, 450000, China.
- Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, Xinxiang, 453002, China.
| |
Collapse
|
5
|
Herrlinger SA, Wang J, Rao BY, Chang J, Gogos JA, Losonczy A, Vitkup D. Rare mutations implicate CGE interneurons as a vulnerable axis of cognitive deficits across psychiatric disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.645799. [PMID: 40236134 PMCID: PMC11996443 DOI: 10.1101/2025.03.28.645799] [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
Neuropsychiatric disorders such as autism spectrum disorder (ASD) and schizophrenia (SCZ) share genetic risk factors, including rare high penetrance single nucleotide variants and copy number variants (CNVs), and exhibit both overlapping and distinct clinical phenotypes. Cognitive deficits and intellectual disability-critical predictors of long-term outcomes-are common to both conditions. To investigate shared and disorder-specific neurobiological impact of highly penetrant rare mutations in ASD and SCZ, we analyzed human single-nucleus whole-brain sequencing data to identify strongly affected brain cell types. Our analysis revealed Caudal Ganglionic Eminence (CGE)-derived GABAergic interneurons as a key nexus for cognitive deficits across these disorders. Notably, genes within 22q11.2 deletions, known to confer a high risk of SCZ, ASD, and cognitive impairment, showed a strong expression bias toward vasoactive intestinal peptide-expressing cells (VIP+) among CGE subtypes. To explore VIP+ GABAergic interneuron perturbations in the 22q11.2 deletion syndrome in vivo , we examined their activity in the Df(16)A +/- mouse model during a spatial navigation task and observed reduced activity along with altered responses to random rewards. At the population level, VIP+ interneurons exhibited impaired spatial encoding and diminished subtype-specific activity suggesting deficient disinhibition in CA1 microcircuits in the hippocampus, a region essential for learning and memory. Overall, these results demonstrate the crucial role of CGE-derived interneurons in mediating cognitive processes that are disrupted across a range of psychiatric and neurodevelopmental disorders.
Collapse
|
6
|
Ge D, Han C, Liu C, Meng Z. Neural Oscillations in the Somatosensory and Motor Cortex Distinguish Dexmedetomidine-Induced Anesthesia and Sleep in Rats. CNS Neurosci Ther 2025; 31:e70262. [PMID: 39963924 PMCID: PMC11833454 DOI: 10.1111/cns.70262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 01/06/2025] [Accepted: 01/27/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Anesthesia is featured by behavioral and physiological characteristics such as decreased sensory and motor function, loss of consciousness, etc. Some anesthetics such as dexmedetomidine (DEX), induce electroencephalogram signatures close to non-rapid eye movement sleep. Studies have shown that sleep is primarily driven by the activation of subcortical sleep-promoting neural pathways. AIMS However, the neuronal level electrophysiology features of anesthesia and how they differ from sleep is still not fully understood. MATERIALS AND METHODS In the present study, we recorded neuronal activity simultaneously from somatosensory cortex (S1) and motor cortex (M1) during awake, sleep, and DEX-induced anesthesia in rats. RESULTS The results show that DEX increased local field potential (LFP) power across a relatively wide band (1-25 Hz) in both S1 and M1. The coherence between S1 LFP and M1 LFP increased significantly in the delta and alpha bands. Power spectrum analysis during DEX-induced anesthesia revealed relatively high power in the delta and alpha bands, but low power in the theta and beta bands. Overall, the firing rate of individual neurons decreased after DEX. Correlation analysis of firing rate and LFP power indicate that more neurons were correlated, either positively or negatively, with LFPs during DEX-induced anesthesia compared to sleep. DISCUSSION Although these results showed enhancement of cortical LFP power in both DEX-induced anesthesia and sleep, different patterns of spike-field correlation suggest that the two states may be regulated by different cortical mechanisms. CONCLUSION Distinguishing anesthesia from sleep with neural oscillations could lead to more personalized, safer, and more effective approaches to managing consciousness in medical settings, with the potential for broad applications in neuroscience and clinical practice.
Collapse
Affiliation(s)
- Dengyun Ge
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Chuanliang Han
- School of Biomedical Sciences and Gerald Choa Neuroscience InstituteThe Chinese University of Hong KongHong KongSARChina
| | - Chang Liu
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- CAS Key Laboratory of Brain Connectome and ManipulationChinese Academy of SciencesShenzhenChina
| | - Zhiqiang Meng
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- CAS Key Laboratory of Brain Connectome and ManipulationChinese Academy of SciencesShenzhenChina
| |
Collapse
|
7
|
Hu Y, Feng Y, Luo H, Zhu XN, Chen S, Yang K, Deng Z, Luo M, Du W, Wang Q, Wang S, Wei K, Hu J, Wang Y. Dissociation-related behaviors in mice emerge from the inhibition of retrosplenial cortex parvalbumin interneurons. Cell Rep 2025; 44:115086. [PMID: 39708317 DOI: 10.1016/j.celrep.2024.115086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/11/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024] Open
Abstract
Dissociation, characterized by altered consciousness and perception, underlies multiple mental disorders, but the specific neuronal subtypes involved remain elusive. In mice, we find that dissociation-inducing doses of ketamine significantly inhibit retrosplenial cortex (RSC) parvalbumin interneurons (PV-INs), enhancing delta oscillations (1-3 Hz) and delta-gamma phase-amplitude coupling (δ-γ PAC) and inducing dissociation-like behaviors. Optogenetic inhibition of RSC PV-INs triggers delta oscillations, δ-γ PAC, and some dissociation-like behaviors without ketamine. Furthermore, activation of RSC PV-INs or knockdown of the N-methyl-D-aspartate receptor subunit NR1 and the hyperpolarization-activated cyclic nucleotide-gated channel 1 (HCN1) in RSC PV-INs attenuates ketamine-induced delta oscillations, δ-γ PAC, and certain dissociation-like behaviors. These findings reveal that PV-INs regulate delta oscillations and δ-γ PAC and identify NR1 and HCN1 as ketamine targets in PV-INs that may cooperatively affect dissociation, possibly providing potential therapeutic targets for dissociative symptoms.
Collapse
Affiliation(s)
- Yue Hu
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yifan Feng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Huoqing Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiao-Na Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Siyu Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Kexin Yang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ziqing Deng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Mengqiang Luo
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wenjie Du
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qi Wang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shubai Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Kai Wei
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| | - Yingwei Wang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
8
|
Gee A, Dazzan P, Grace AA, Modinos G. Corticolimbic circuitry as a druggable target in schizophrenia spectrum disorders: a narrative review. Transl Psychiatry 2025; 15:21. [PMID: 39856031 PMCID: PMC11760974 DOI: 10.1038/s41398-024-03221-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 12/06/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
Schizophrenia spectrum disorders (SSD) involve disturbances in the integration of perception, emotion and cognition. The corticolimbic system is an interacting set of cortical and subcortical brain regions critically involved in this process. Understanding how neural circuitry and molecular mechanisms within this corticolimbic system may contribute to the development of not only positive symptoms but also negative and cognitive deficits in SSD has been a recent focus of intense research, as the latter are not adequately treated by current antipsychotic medications and are more strongly associated with poorer functioning and long-term outcomes. This review synthesises recent developments examining corticolimbic dysfunction in the pathophysiology of SSD, with a focus on neuroimaging advances and related novel methodologies that enable the integration of data across different scales. We then integrate how these findings may inform the identification of novel therapeutic and preventive targets for SSD symptomatology. A range of pharmacological interventions have shown initial promise in correcting corticolimbic dysfunction and improving negative, cognitive and treatment-resistant symptoms. We discuss current challenges and opportunities for improving the still limited translation of these research findings into clinical practice. We argue how our knowledge of the role of corticolimbic dysfunction can be improved by combining multiple research modalities to examine hypotheses across different spatial and temporal scales, combining neuroimaging with experimental interventions and utilising large-scale consortia to advance biomarker identification. Translation of these findings into clinical practice will be aided by consideration of optimal intervention timings, biomarker-led patient stratification, and the development of more selective medications.
Collapse
Affiliation(s)
- Abigail Gee
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Paola Dazzan
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gemma Modinos
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
9
|
Khosroshahi PA, Ghanbari M. MicroRNA dysregulation in glutamate and dopamine pathways of schizophrenia: From molecular pathways to diagnostic and therapeutic approaches. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111081. [PMID: 39002925 DOI: 10.1016/j.pnpbp.2024.111081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 06/28/2024] [Accepted: 07/07/2024] [Indexed: 07/15/2024]
Abstract
Schizophrenia is a complex psychiatric disorder, and genetic and environmental factors have been implicated in its development. Dysregulated glutamatergic and dopaminergic transmission pathways are involved in schizophrenia development. Besides genetic mutations, epigenetic dysregulation has a considerable role in dysregulating molecular pathways involved in schizophrenia. MicroRNAs (miRNAs) are small, non-coding RNAs that target specific mRNAs and inhibit their translation into proteins. As epigenetic factors, miRNAs regulate many genes involved in glutamate and dopamine signaling pathways; thereby, their dysregulation can contribute to the development of schizophrenia. Secretion of specific miRNAs from damaged cells into body fluids can make them one of the ideal non-invasive biomarkers in the early diagnosis of schizophrenia. Also, understanding the molecular mechanisms of miRNAs in schizophrenia pathogenesis can pave the way for developing novel treatments for patients with schizophrenia. In this study, we reviewed the glutamatergic and dopaminergic pathophysiology and highlighted the role of miRNA dysregulation in schizophrenia development. Besides, we shed light on the significance of circulating miRNAs for schizophrenia diagnosis and the recent findings on the miRNA-based treatment for schizophrenia.
Collapse
Affiliation(s)
| | - Mohammad Ghanbari
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
10
|
Brakatselos C, Polissidis A, Ntoulas G, Asprogerakas MZ, Tsarna O, Vamvaka-Iakovou A, Nakas G, Delis A, Tzimas P, Skaltsounis L, Silva J, Delis F, Oliveira JF, Sotiropoulos I, Antoniou K. Multi-level therapeutic actions of cannabidiol in ketamine-induced schizophrenia psychopathology in male rats. Neuropsychopharmacology 2024; 50:388-400. [PMID: 39242923 PMCID: PMC11631973 DOI: 10.1038/s41386-024-01977-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/19/2024] [Accepted: 08/19/2024] [Indexed: 09/09/2024]
Abstract
Repeated administration of ketamine (KET) has been used to model schizophrenia-like symptomatology in rodents, but the psychotomimetic neurobiological and neuroanatomical underpinnings remain elusive. In parallel, the unmet need for a better treatment of schizophrenia requires the development of novel therapeutic strategies. Cannabidiol (CBD), a major non-addictive phytocannabinoid has been linked to antipsychotic effects with unclear mechanistic basis. Therefore, this study aims to clarify the neurobiological substrate of repeated KET administration model and to evaluate CBD's antipsychotic potential and neurobiological basis. CBD-treated male rats with and without prior repeated KET administration underwent behavioral analyses, followed by multilevel analysis of different brain areas including dopaminergic and glutamatergic activity, synaptic signaling, as well as electrophysiological recordings for the assessment of corticohippocampal and corticostriatal network activity. Repeated KET model is characterized by schizophrenia-like symptomatology and alterations in glutamatergic and dopaminergic activity mainly in the PFC and the dorsomedial striatum (DMS), through a bi-directional pattern. These observations are accompanied by glutamatergic/GABAergic deviations paralleled to impaired function of parvalbumin- and cholecystokinin-positive interneurons, indicative of excitation/inhibition (E/I) imbalance. Moreover, CBD counteracted the schizophrenia-like behavioral phenotype as well as reverted prefrontal abnormalities and ventral hippocampal E/I deficits, while partially modulated dorsostriatal dysregulations. This study adds novel insights to our understanding of the KET-induced schizophrenia-related brain pathology, as well as the CBD antipsychotic action through a region-specific set of modulations in the corticohippocampal and costicostrtiatal circuitry of KET-induced profile contributing to the development of novel therapeutic strategies focused on the ECS and E/I imbalance restoration.
Collapse
Affiliation(s)
- Charalampos Brakatselos
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110, Ioannina, Greece
| | - Alexia Polissidis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527, Athens, Greece
- Department of Science and Mathematics, ACG-Research Center, Deree - American College of Greece, 15342, Athens, Greece
| | - George Ntoulas
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110, Ioannina, Greece
| | - Michail-Zois Asprogerakas
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110, Ioannina, Greece
| | - Olga Tsarna
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110, Ioannina, Greece
| | - Anastasia Vamvaka-Iakovou
- Institute of Biosciences & Applications, NCSR Demokritos, Athens, Greece
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Gerasimos Nakas
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110, Ioannina, Greece
| | - Anastasios Delis
- Center of Basic Research, Biological Imaging Unit, Biomedical Research Foundation Academy of Athens, 11527, Athens, Greece
| | - Petros Tzimas
- Department of Pharmacognosy and Natural Product Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Leandros Skaltsounis
- Department of Pharmacognosy and Natural Product Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Joana Silva
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Foteini Delis
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110, Ioannina, Greece
| | - Joao Filipe Oliveira
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- IPCA-EST-2Ai, Polytechnic Institute of Cávado and Ave, Applied Artificial Intelligence Laboratory, Campus of IPCA, Barcelos, Portugal
| | - Ioannis Sotiropoulos
- Institute of Biosciences & Applications, NCSR Demokritos, Athens, Greece
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Katerina Antoniou
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110, Ioannina, Greece.
| |
Collapse
|
11
|
Mediane DH, Basu S, Cahill EN, Anastasiades PG. Medial prefrontal cortex circuitry and social behaviour in autism. Neuropharmacology 2024; 260:110101. [PMID: 39128583 DOI: 10.1016/j.neuropharm.2024.110101] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
Autism spectrum disorder (ASD) has proven to be highly enigmatic due to the diversity of its underlying genetic causes and the huge variability in symptom presentation. Uncovering common phenotypes across people with ASD and pre-clinical models allows us to better understand the influence on brain function of the many different genetic and cellular processes thought to contribute to ASD aetiology. One such feature of ASD is the convergent evidence implicating abnormal functioning of the medial prefrontal cortex (mPFC) across studies. The mPFC is a key part of the 'social brain' and may contribute to many of the changes in social behaviour observed in people with ASD. Here we review recent evidence for mPFC involvement in both ASD and social behaviours. We also highlight how pre-clinical mouse models can be used to uncover important cellular and circuit-level mechanisms that may underly atypical social behaviours in ASD. This article is part of the Special Issue on "PFC circuit function in psychiatric disease and relevant models".
Collapse
Affiliation(s)
- Diego H Mediane
- Department of Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Shinjini Basu
- Department of Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Emma N Cahill
- Department of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, United Kingdom
| | - Paul G Anastasiades
- Department of Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom.
| |
Collapse
|
12
|
Umbricht D, Kas MJH, Dawson GR. The role of biomarkers in clinical development of drugs for neuropsychiatric disorders - A pragmatic guide. Eur Neuropsychopharmacol 2024; 88:66-77. [PMID: 39236552 DOI: 10.1016/j.euroneuro.2024.08.511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 09/07/2024]
Abstract
The failure rate of drugs being developed for neuropsychiatric indications remains high. Optimizing drug discovery and development requires not only a better neurobiological understanding of disease aetiology and development, but also the means by which we can measure relevant biological and clinical processes related to disease progression, drug target engagement, and sensitivity to treatment. Here we address the role and key considerations for the selection of biomarkers in clinical drug development for neuropsychiatric disorders. We do not provide an exhaustive list of biomarkers; rather we lay out a pragmatic, well-defined biomarker selection strategy that addresses the main goals for each of the phases in the drug development cycle. We discuss the key questions and issues that concern biomarker selection and implementation in each phase of development. For the better development of biomarkers, we emphasize the need to focus on discrete biological dysfunction and/or symptom domains rather than diagnoses. We also advocate the use of biomarker-based patient stratification in phase 2 and 3 to increase sensitivity and power and reduce costs. Our aim is to enhance precision and chances of success for these complex and heterogeneous brain disorders with a high unmet medical need.
Collapse
Affiliation(s)
- Daniel Umbricht
- xperimed GmbH, Basel, Switzerland; University of Zurich, Switzerland.
| | - Martien J H Kas
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, the Netherlands
| | | |
Collapse
|
13
|
Pellegrino G, Isabella SL, Ferrazzi G, Gschwandtner L, Tik M, Arcara G, Marinazzo D, Schuler AL. Reliable measurement of auditory-driven gamma synchrony with a single EEG electrode: A simultaneous EEG-MEG study. Neuroimage 2024; 300:120862. [PMID: 39305968 DOI: 10.1016/j.neuroimage.2024.120862] [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: 01/27/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024] Open
Abstract
OBJECTIVE Auditory-driven gamma synchrony (GS) is linked to the function of a specific cortical circuit based on a parvalbumin+ and pyramidal neuron loop. This circuit is impaired in neuropsychiatric conditions (i.e. schizophrenia, Alzheimer's disease, stroke etc.) and its relevance in clinical practice is increasingly being recognized. Auditory stimulation at a typical gamma frequency of 40 Hz can be applied as a 'stress test' of excitation/inhibition (E/I) of the entire cerebral cortex, to drive GS and record it with magnetoencephalography (MEG) or high-density electroencephalography (EEG). However, these two techniques are costly and not widely available. Therefore, we assessed whether a single EEG electrode is sufficient to provide an accurate estimate of the auditory-driven GS level of the entire cortical surface while expecting the highest correspondence in the auditory and somatosensory cortices. METHODS We measured simultaneous EEG-MEG in 29 healthy subjects, utilizing 3 EEG electrodes (C4, F4, O2) and a full MEG setup. Recordings were performed during binaural exposure to auditory gamma stimulation and during silence. We compared GS measurement of each of the three EEG electrodes separately against full MEG mapping. Time-resolved phase locking value (PLVt) was computed between EEG signals and cortex reconstructed MEG signals. RESULTS During auditory stimulation, but not at rest, EEG captures a significant amount of GS, especially from both auditory cortices and motor-premotor regions. This was especially true for frontal (C4) and central electrodes (F4). DISCUSSION AND CONCLUSIONS While hd-EEG and MEG are necessary for accurate spatial mapping of GS at rest and during auditory stimulation, a single EEG channel is sufficient to detect the global level of GS. These results have great translational potential for mapping GS in standard clinical settings.
Collapse
Affiliation(s)
- Giovanni Pellegrino
- Clinical Neurological Sciences Department, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Silvia L Isabella
- Campus Bio-Medico University of Rome, Rome, Italy; IRCCS San Camillo Hospital, Via Alberoni 80, 30126, Venice, Italy
| | | | - Laura Gschwandtner
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Austria
| | - Martin Tik
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Austria.
| | - Giorgio Arcara
- IRCCS San Camillo Hospital, Via Alberoni 80, 30126, Venice, Italy; Department of General Psychology, University of Padua, Padua, Italy
| | - Daniele Marinazzo
- Department of Data Analysis, Faculty of Psychology and Educational Sciences, Ghent University, Belgium
| | - Anna-Lisa Schuler
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| |
Collapse
|
14
|
Zhang T, Liu C, Zhong N, Wang Y, Huang Y, Zhang X. Advances in the Treatment of Cognitive Impairment in Schizophrenia: Targeting NMDA Receptor Pathways. Int J Mol Sci 2024; 25:10668. [PMID: 39408997 PMCID: PMC11477438 DOI: 10.3390/ijms251910668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Cognitive impairment is a core feature of schizophrenia, playing a pivotal role in the pathogenesis and prognosis of this disorder. Cognitive impairment in schizophrenia encompasses a wide range of domains, including processing speed, episodic memory, working memory, and executive function. These deficits persist throughout the course of the illness and significantly impact functional outcomes and quality of life. Therefore, it is imperative to identify the biological basis of cognitive deficits in schizophrenia and develop effective treatments. The role of N-methyl-D-aspartate (NMDA) receptors in synaptic transmission and plasticity has long been recognized, making them potential targets for schizophrenia treatment. This review will focus on emerging pharmacology targeting NMDA receptors, offering strategies for the prevention and treatment of cognitive deficits in schizophrenia.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaoqin Zhang
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo 315211, China; (T.Z.); (C.L.); (N.Z.); (Y.W.); (Y.H.)
| |
Collapse
|
15
|
Carrillo GL, Su J, Cawley ML, Wei D, Gill SK, Blader IJ, Fox MA. Complement-dependent loss of inhibitory synapses on pyramidal neurons following Toxoplasma gondii infection. J Neurochem 2024; 168:3365-3385. [PMID: 36683435 PMCID: PMC10363253 DOI: 10.1111/jnc.15770] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 01/06/2023] [Accepted: 01/15/2023] [Indexed: 01/24/2023]
Abstract
The apicomplexan parasite Toxoplasma gondii has developed mechanisms to establish a central nervous system infection in virtually all warm-blooded animals. Acute T. gondii infection can cause neuroinflammation, encephalitis, and seizures. Meanwhile, studies in humans, nonhuman primates, and rodents have linked chronic T. gondii infection with altered behavior and increased risk for neuropsychiatric disorders, including schizophrenia. These observations and associations raise questions about how this parasitic infection may alter neural circuits. We previously demonstrated that T. gondii infection triggers the loss of inhibitory perisomatic synapses, a type of synapse whose dysfunction or loss has been linked to neurological and neuropsychiatric disorders. We showed that phagocytic cells (including microglia and infiltrating monocytes) contribute to the loss of these inhibitory synapses. Here, we show that these phagocytic cells specifically ensheath excitatory pyramidal neurons, leading to the preferential loss of perisomatic synapses on these neurons and not those on cortical interneurons. Moreover, we show that infection induces an increased expression of the complement C3 gene, including by populations of these excitatory neurons. Infecting C3-deficient mice with T. gondii revealed that C3 is required for the loss of perisomatic inhibitory synapses. Interestingly, loss of C1q did not prevent the loss of perisomatic synapses following infection. Together, these findings provide evidence that T. gondii induces changes in excitatory pyramidal neurons that trigger the selective removal of inhibitory perisomatic synapses and provide a role for a nonclassical complement pathway in the remodeling of inhibitory circuits in the infected brain.
Collapse
Affiliation(s)
- Gabriela L. Carrillo
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, 24016, USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia, 24061, USA
| | - Jianmin Su
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, 24016, USA
- School of Neuroscience, College of Science, Virginia Tech, Blacksburg, Virginia, 24061, USA
| | - Mikel L. Cawley
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, 24016, USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia, 24061, USA
| | - Derek Wei
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, 24016, USA
- School of Neuroscience, College of Science, Virginia Tech, Blacksburg, Virginia, 24061, USA
| | - Simran K. Gill
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, 24016, USA
- Department of Psychology, Roanoke College, Salem, Virginia, 24153, USA
- NeuroSURF Program, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, 24016, USA
| | - Ira J. Blader
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, New York, 14203, USA
| | - Michael A. Fox
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, 24016, USA
- School of Neuroscience, College of Science, Virginia Tech, Blacksburg, Virginia, 24061, USA
- Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg, Virginia, 24061, USA
- Department of Pediatrics, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, 24016, USA
| |
Collapse
|
16
|
Faulkner IE, Pajak RZ, Harte MK, Glazier JD, Hager R. Voltage-gated potassium channels as a potential therapeutic target for the treatment of neurological and psychiatric disorders. Front Cell Neurosci 2024; 18:1449151. [PMID: 39411003 PMCID: PMC11473391 DOI: 10.3389/fncel.2024.1449151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
Voltage-gated potassium channels are a widely distributed subgroup of potassium channels responsible for the efflux of potassium in the repolarisation of the cell membrane, and hence contribute to the latency and propagation of action potentials. As they are causal to synaptic transmission, alterations to the structure of these channels can lead to a variety of neurological and psychiatric diseases. The Kv3 subfamily of voltage-gated potassium channels are found on many neurons in the brain, including inhibitory interneurons where they contribute to fast-frequency firing. Changes to the firing ability of these interneurons can lead to an imbalance of inhibitory and excitatory neurotransmission. To date, we have little understanding of the mechanism by which excitatory and inhibitory inputs become imbalanced. This imbalance is associated with cognitive deficits seen across neurological and neuropsychiatric disorders, which are currently difficult to treat. In this review, we collate evidence supporting the hypothesis that voltage-gated potassium channels, specifically the Kv3 subfamily, are central to many neurological and psychiatric disorders, and may thus be considered as an effective drug target. The collective evidence provided by the studies reviewed here demonstrates that Kv3 channels may be amenable to novel treatments that modulate the activity of these channels, with the prospect of improved patient outcome.
Collapse
Affiliation(s)
- Isabel E. Faulkner
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Rachael Z. Pajak
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Michael K. Harte
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Jocelyn D. Glazier
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Reinmar Hager
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
17
|
Uliana DL, Lisboa JRF, Gomes FV, Grace AA. The excitatory-inhibitory balance as a target for the development of novel drugs to treat schizophrenia. Biochem Pharmacol 2024; 228:116298. [PMID: 38782077 PMCID: PMC11410545 DOI: 10.1016/j.bcp.2024.116298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
The intricate balance between excitation and inhibition (E/I) in the brain plays a crucial role in normative information processing. Dysfunctions in the E/I balance have been implicated in various psychiatric disorders, including schizophrenia (SCZ). In particular, abnormalities in GABAergic signaling, specifically in parvalbumin (PV)-containing interneurons, have been consistently observed in SCZ pathophysiology. PV interneuron function is vital for maintaining an ideal E/I balance, and alterations in PV interneuron-mediated inhibition contribute to circuit deficits observed in SCZ, including hippocampus hyperactivity and midbrain dopamine system overdrive. While current antipsychotic medications primarily target D2 dopamine receptors and are effective primarily in treating positive symptoms, novel therapeutic strategies aiming to restore the E/I balance could potentially mitigate not only positive symptoms but also negative symptoms and cognitive deficits. This could involve, for instance, increasing the inhibitory drive onto excitatory neurons or decreasing the putative enhanced pyramidal neuron activity due to functional loss of PV interneurons. Compounds targeting the glycine site at glutamate NMDA receptors and muscarinic acetylcholine receptors on PV interneurons that can increase PV interneuron drive, as well as drugs that increase the postsynaptic action of GABA, such as positive allosteric modulators of α5-GABA-A receptors, and decrease glutamatergic output, such as mGluR2/3 agonists, represent promising approaches. Preventive strategies aiming at E/I balance also represent a path to reduce the risk of transitioning to SCZ in high-risk individuals. Therefore, compounds with novel mechanisms targeting E/I balance provide optimism for more effective and tailored interventions in the management of SCZ.
Collapse
Affiliation(s)
- Daniela L Uliana
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joao Roberto F Lisboa
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Felipe V Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
18
|
Sullivan PF, Yao S, Hjerling-Leffler J. Schizophrenia genomics: genetic complexity and functional insights. Nat Rev Neurosci 2024; 25:611-624. [PMID: 39030273 DOI: 10.1038/s41583-024-00837-7] [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: 06/04/2024] [Indexed: 07/21/2024]
Abstract
Determining the causes of schizophrenia has been a notoriously intractable problem, resistant to a multitude of investigative approaches over centuries. In recent decades, genomic studies have delivered hundreds of robust findings that implicate nearly 300 common genetic variants (via genome-wide association studies) and more than 20 rare variants (via whole-exome sequencing and copy number variant studies) as risk factors for schizophrenia. In parallel, functional genomic and neurobiological studies have provided exceptionally detailed information about the cellular composition of the brain and its interconnections in neurotypical individuals and, increasingly, in those with schizophrenia. Taken together, these results suggest unexpected complexity in the mechanisms that drive schizophrenia, pointing to the involvement of ensembles of genes (polygenicity) rather than single-gene causation. In this Review, we describe what we now know about the genetics of schizophrenia and consider the neurobiological implications of this information.
Collapse
Affiliation(s)
- Patrick F Sullivan
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA.
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| | - Shuyang Yao
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jens Hjerling-Leffler
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
19
|
Dwyer GE, Johnsen E, Hugdahl K. NMDAR dysfunction and the regulation of dopaminergic transmission in schizophrenia. Schizophr Res 2024; 271:19-27. [PMID: 39002526 DOI: 10.1016/j.schres.2024.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/27/2024] [Accepted: 07/07/2024] [Indexed: 07/15/2024]
Abstract
A substantial body of evidence implicates dysfunction in N-methyl-d-aspartate receptors (NMDARs) in the pathophysiology of schizophrenia. This article illustrates how NMDAR dysfunction may give rise to many of the neurobiological phenomena frequently associated with schizophrenia with a particular focus on how NMDAR dysfunction affects the thalamic reticular nucleus (nRT) and pedunculopontine tegmental nucleus (PPTg). Furthermore, this article presents a model for schizophrenia illustrating how dysfunction in the nRT may interrupt prefrontal regulation of midbrain dopaminergic neurons, and how dysfunction in the PPTg may drive increased, irregular burst firing.
Collapse
Affiliation(s)
- Gerard Eric Dwyer
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway; NORMENT Centre of Excellence, Haukeland University Hospital, Bergen, Norway.
| | - Erik Johnsen
- NORMENT Centre of Excellence, Haukeland University Hospital, Bergen, Norway; Division of Psychiatry, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Kenneth Hugdahl
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway; Division of Psychiatry, Haukeland University Hospital, Bergen, Norway; Department of Radiology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
20
|
Lee DA, Jang T, Kang J, Park S, Park KM. Functional Connectivity Alterations in Patients with Post-stroke Epilepsy Based on Source-level EEG and Graph Theory. Brain Topogr 2024; 37:921-930. [PMID: 38625521 DOI: 10.1007/s10548-024-01048-0] [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: 01/03/2023] [Accepted: 03/28/2024] [Indexed: 04/17/2024]
Abstract
We investigated the differences in functional connectivity based on the source-level electroencephalography (EEG) analysis between stroke patients with and without post-stroke epilepsy (PSE). Thirty stroke patients with PSE and 35 stroke patients without PSE were enrolled. EEG was conducted during a resting state period. We used a Brainstorm program for source estimation and the connectivity matrix. Data were processed according to EEG frequency bands. We used a BRAPH program to apply a graph theoretical analysis. In the beta band, radius and diameter were increased in patients with PSE than in those without PSE (2.699 vs. 2.579, adjusted p = 0.03; 2.261 vs. 2.171, adjusted p = 0.03). In the low gamma band, radius was increased in patients with PSE than in those without PSE (2.808 vs. 2.617, adjusted p = 0.03). In the high gamma band, the radius, diameter, average eccentricity, and characteristic path length were increased (1.828 vs. 1.559, adjusted p < 0.01; 2.653 vs. 2.306, adjusted p = 0.01; 2.212 vs. 1.913, adjusted p < 0.01; 1.425 vs. 1.286, adjusted p = 0.01), whereas average strength, mean clustering coefficient, and transitivity were decreased in patients with PSE than in those without PSE (49.955 vs. 55.055, adjusted p < 0.01; 0.727 vs. 0.810, adjusted p < 0.01; 1.091 vs. 1.215, adjusted p < 0.01). However, in the delta, theta, and alpha bands, none of the functional connectivity measures were different between groups. We demonstrated significant alterations of functional connectivity in patients with PSE, who have decreased segregation and integration in brain network, compared to those without PSE.
Collapse
Affiliation(s)
- Dong Ah Lee
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Haeundae-ro 875, Haeundae-gu, Busan, 48108, Korea
| | - Taeik Jang
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Haeundae-ro 875, Haeundae-gu, Busan, 48108, Korea
| | - Jaeho Kang
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Haeundae-ro 875, Haeundae-gu, Busan, 48108, Korea
| | - Seongho Park
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Haeundae-ro 875, Haeundae-gu, Busan, 48108, Korea
| | - Kang Min Park
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Haeundae-ro 875, Haeundae-gu, Busan, 48108, Korea.
| |
Collapse
|
21
|
Ponserre M, Ionescu TM, Franz AA, Deiana S, Schuelert N, Lamla T, Williams RH, Wotjak CT, Hobson S, Dine J, Omrani A. Long-term adaptation of prefrontal circuits in a mouse model of NMDAR hypofunction. Neuropharmacology 2024; 254:109970. [PMID: 38685343 DOI: 10.1016/j.neuropharm.2024.109970] [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: 02/15/2024] [Revised: 04/12/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
Pharmacological approaches to induce N-methyl-d-aspartate receptor (NMDAR) hypofunction have been intensively used to understand the aetiology and pathophysiology of schizophrenia. Yet, the precise cellular and molecular mechanisms that relate to brain network dysfunction remain largely unknown. Here, we used a set of complementary approaches to assess the functional network abnormalities present in male mice that underwent a 7-day subchronic phencyclidine (PCP 10 mg/kg, subcutaneously, once daily) treatment. Our data revealed that pharmacological intervention with PCP affected cognitive performance and auditory evoked gamma oscillations in the prefrontal cortex (PFC) mimicking endophenotypes of some schizophrenia patients. We further assessed PFC cellular function and identified altered neuronal intrinsic membrane properties, reduced parvalbumin (PV) immunostaining and diminished inhibition onto L5 PFC pyramidal cells. A decrease in the strength of optogenetically-evoked glutamatergic current at the ventral hippocampus to PFC synapse was also demonstrated, along with a weaker shunt of excitatory transmission by local PFC interneurons. On a macrocircuit level, functional ultrasound measurements indicated compromised functional connectivity within several brain regions particularly involving PFC and frontostriatal circuits. Herein, we reproduced a panel of schizophrenia endophenotypes induced by subchronic PCP application in mice. We further recapitulated electrophysiological signatures associated with schizophrenia and provided an anatomical reference to critical elements in the brain circuitry. Together, our findings contribute to a better understanding of the physiological underpinnings of deficits induced by subchronic NMDAR antagonist regimes and provide a test system for characterization of pharmacological compounds.
Collapse
Affiliation(s)
- Marion Ponserre
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Tudor M Ionescu
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Alessa A Franz
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Serena Deiana
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Niklas Schuelert
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Thorsten Lamla
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | | | - Carsten T Wotjak
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Scott Hobson
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Julien Dine
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Azar Omrani
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany.
| |
Collapse
|
22
|
Dugan C, Zikopoulos B, Yazdanbakhsh A. A neural modeling approach to study mechanisms underlying the heterogeneity of visual spatial frequency sensitivity in schizophrenia. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:63. [PMID: 39013944 PMCID: PMC11252134 DOI: 10.1038/s41537-024-00480-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 06/14/2024] [Indexed: 07/18/2024]
Abstract
Patients with schizophrenia exhibit abnormalities in spatial frequency sensitivity, and it is believed that these abnormalities indicate more widespread dysfunction and dysregulation of bottom-up processing. The early visual system, including the first-order Lateral Geniculate Nucleus of the thalamus (LGN) and the primary visual cortex (V1), are key contributors to spatial frequency sensitivity. Medicated and unmedicated patients with schizophrenia exhibit contrasting changes in spatial frequency sensitivity, thus making it a useful probe for examining potential effects of the disorder and antipsychotic medications in neural processing. We constructed a parameterized, rate-based neural model of on-center/off-surround neurons in the early visual system to investigate the impacts of changes to the excitatory and inhibitory receptive field subfields. By incorporating changes in both the excitatory and inhibitory subfields that are associated with pathophysiological findings in schizophrenia, the model successfully replicated perceptual data from behavioral/functional studies involving medicated and unmedicated patients. Among several plausible mechanisms, our results highlight the dampening of excitation and/or increase in the spread and strength of the inhibitory subfield in medicated patients and the contrasting decreased spread and strength of inhibition in unmedicated patients. Given that the model was successful at replicating results from perceptual data under a variety of conditions, these elements of the receptive field may be useful markers for the imbalances seen in patients with schizophrenia.
Collapse
Affiliation(s)
- Caroline Dugan
- Program in Neuroscience, Boston University, Boston, MA, USA
| | - Basilis Zikopoulos
- Human Systems Neuroscience Laboratory, Department of Health Sciences, Boston University, Boston, MA, USA.
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA.
- Center for Systems Neuroscience, Boston University, Boston, MA, USA.
- Graduate Program for Neuroscience, Boston University, Boston, MA, USA.
| | - Arash Yazdanbakhsh
- Center for Systems Neuroscience, Boston University, Boston, MA, USA.
- Graduate Program for Neuroscience, Boston University, Boston, MA, USA.
- Computational Neuroscience and Vision Laboratory, Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA.
| |
Collapse
|
23
|
Oliver D, Chesney E, Cullen AE, Davies C, Englund A, Gifford G, Kerins S, Lalousis PA, Logeswaran Y, Merritt K, Zahid U, Crossley NA, McCutcheon RA, McGuire P, Fusar-Poli P. Exploring causal mechanisms of psychosis risk. Neurosci Biobehav Rev 2024; 162:105699. [PMID: 38710421 PMCID: PMC11250118 DOI: 10.1016/j.neubiorev.2024.105699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/17/2024] [Accepted: 04/28/2024] [Indexed: 05/08/2024]
Abstract
Robust epidemiological evidence of risk and protective factors for psychosis is essential to inform preventive interventions. Previous evidence syntheses have classified these risk and protective factors according to their strength of association with psychosis. In this critical review we appraise the distinct and overlapping mechanisms of 25 key environmental risk factors for psychosis, and link these to mechanistic pathways that may contribute to neurochemical alterations hypothesised to underlie psychotic symptoms. We then discuss the implications of our findings for future research, specifically considering interactions between factors, exploring universal and subgroup-specific factors, improving understanding of temporality and risk dynamics, standardising operationalisation and measurement of risk and protective factors, and developing preventive interventions targeting risk and protective factors.
Collapse
Affiliation(s)
- Dominic Oliver
- Department of Psychiatry, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Oxford, UK; OPEN Early Detection Service, Oxford Health NHS Foundation Trust, Oxford, UK; Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
| | - Edward Chesney
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Addictions Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 4 Windsor Walk, London SE5 8AF, UK
| | - Alexis E Cullen
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Clinical Neuroscience, Karolinska Institutet, Sweden
| | - Cathy Davies
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Amir Englund
- Addictions Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 4 Windsor Walk, London SE5 8AF, UK
| | - George Gifford
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Sarah Kerins
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Paris Alexandros Lalousis
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Yanakan Logeswaran
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Biostatistics & Health Informatics, King's College London, London, UK
| | - Kate Merritt
- Division of Psychiatry, Institute of Mental Health, UCL, London, UK
| | - Uzma Zahid
- Department of Psychology, King's College London, London, UK
| | - Nicolas A Crossley
- Department of Psychiatry, University of Oxford, Oxford, UK; Department of Psychiatry, School of Medicine, Pontificia Universidad Católica de Chile, Chile
| | - Robert A McCutcheon
- Department of Psychiatry, University of Oxford, Oxford, UK; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Oxford Health NHS Foundation Trust, Oxford, UK
| | - Philip McGuire
- Department of Psychiatry, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Oxford, UK; OPEN Early Detection Service, Oxford Health NHS Foundation Trust, Oxford, UK
| | - Paolo Fusar-Poli
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; OASIS Service, South London and Maudsley NHS Foundation Trust, London SE11 5DL, UK
| |
Collapse
|
24
|
Mana L, Schwartz-Pallejà M, Vila-Vidal M, Deco G. Overview on cognitive impairment in psychotic disorders: From impaired microcircuits to dysconnectivity. Schizophr Res 2024; 269:132-143. [PMID: 38788432 DOI: 10.1016/j.schres.2024.05.008] [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: 01/15/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Schizophrenia's cognitive deficits, often overshadowed by positive symptoms, significantly contribute to the disorder's morbidity. Increasing attention highlights these deficits as reflections of neural circuit dysfunction across various cortical regions. Numerous connectivity alterations linked to cognitive symptoms in psychotic disorders have been reported, both at the macroscopic and microscopic level, emphasizing the potential role of plasticity and microcircuits impairment during development and later stages. However, the heterogeneous clinical presentation of cognitive impairment and diverse connectivity findings pose challenges in summarizing them into a cohesive picture. This review aims to synthesize major cognitive alterations, recent insights into network structural and functional connectivity changes and proposed mechanisms and microcircuit alterations underpinning these symptoms, particularly focusing on neurodevelopmental impairment, E/I balance, and sleep disturbances. Finally, we will also comment on some of the most recent and promising therapeutic approaches that aim to target these mechanisms to address cognitive symptoms. Through this comprehensive exploration, we strive to provide an updated and nuanced overview of the multiscale connectivity impairment underlying cognitive impairment in psychotic disorders.
Collapse
Affiliation(s)
- L Mana
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain.
| | - M Schwartz-Pallejà
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Department of Experimental and Health Science, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Eurecat, Technology Center of Catalonia, Multimedia Technologies, Barcelona, Spain.
| | - M Vila-Vidal
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Computational Biology and Complex Systems Group, Department of Physics, Universitat Politècnica de Catalunya, Barcelona, Spain.
| | - G Deco
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Institució Catalana de la Recerca i Estudis Avançats (ICREA), Passeig Lluís Companys 23, Barcelona 08010, Spain.
| |
Collapse
|
25
|
Black T, Jenkins BW, Laprairie RB, Howland JG. Therapeutic potential of gamma entrainment using sensory stimulation for cognitive symptoms associated with schizophrenia. Neurosci Biobehav Rev 2024; 161:105681. [PMID: 38641090 DOI: 10.1016/j.neubiorev.2024.105681] [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: 01/25/2024] [Revised: 03/27/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
Schizophrenia is a complex neuropsychiatric disorder with significant morbidity. Treatment options that address the spectrum of symptoms are limited, highlighting the need for innovative therapeutic approaches. Gamma Entrainment Using Sensory Stimulation (GENUS) is an emerging treatment for neuropsychiatric disorders that uses sensory stimulation to entrain impaired oscillatory network activity and restore brain function. Aberrant oscillatory activity often underlies the symptoms experienced by patients with schizophrenia. We propose that GENUS has therapeutic potential for schizophrenia. This paper reviews the current status of schizophrenia treatment and explores the use of sensory stimulation as an adjunctive treatment, specifically through gamma entrainment. Impaired gamma frequency entrainment is observed in patients, particularly in response to auditory and visual stimuli. Thus, sensory stimulation, such as music listening, may have therapeutic potential for individuals with schizophrenia. GENUS holds novel therapeutic potential to improve the lives of individuals with schizophrenia, but further research is required to determine the efficacy of GENUS, optimize its delivery and therapeutic window, and develop strategies for its implementation in specific patient populations.
Collapse
Affiliation(s)
- Tallan Black
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada.
| | - Bryan W Jenkins
- Division of Behavioral Biology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Robert B Laprairie
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada; Department of Pharmacology, College of Medicine, Dalhousie University, Halifax, NS, Canada
| | - John G Howland
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
26
|
Faris P, Pischedda D, Palesi F, D’Angelo E. New clues for the role of cerebellum in schizophrenia and the associated cognitive impairment. Front Cell Neurosci 2024; 18:1386583. [PMID: 38799988 PMCID: PMC11116653 DOI: 10.3389/fncel.2024.1386583] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Schizophrenia (SZ) is a complex neuropsychiatric disorder associated with severe cognitive dysfunction. Although research has mainly focused on forebrain abnormalities, emerging results support the involvement of the cerebellum in SZ physiopathology, particularly in Cognitive Impairment Associated with SZ (CIAS). Besides its role in motor learning and control, the cerebellum is implicated in cognition and emotion. Recent research suggests that structural and functional changes in the cerebellum are linked to deficits in various cognitive domains including attention, working memory, and decision-making. Moreover, cerebellar dysfunction is related to altered cerebellar circuit activities and connectivity with brain regions associated with cognitive processing. This review delves into the role of the cerebellum in CIAS. We initially consider the major forebrain alterations in CIAS, addressing impairments in neurotransmitter systems, synaptic plasticity, and connectivity. We then focus on recent findings showing that several mechanisms are also altered in the cerebellum and that cerebellar communication with the forebrain is impaired. This evidence implicates the cerebellum as a key component of circuits underpinning CIAS physiopathology. Further studies addressing cerebellar involvement in SZ and CIAS are warranted and might open new perspectives toward understanding the physiopathology and effective treatment of these disorders.
Collapse
Affiliation(s)
- Pawan Faris
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Doris Pischedda
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Fulvia Palesi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Egidio D’Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Digital Neuroscience Center, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
27
|
Dharmasri PA, Levy AD, Blanpied TA. Differential nanoscale organization of excitatory synapses onto excitatory vs. inhibitory neurons. Proc Natl Acad Sci U S A 2024; 121:e2315379121. [PMID: 38625946 PMCID: PMC11047112 DOI: 10.1073/pnas.2315379121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 03/14/2024] [Indexed: 04/18/2024] Open
Abstract
A key feature of excitatory synapses is the existence of subsynaptic protein nanoclusters (NCs) whose precise alignment across the cleft in a transsynaptic nanocolumn influences the strength of synaptic transmission. However, whether nanocolumn properties vary between excitatory synapses functioning in different cellular contexts is unknown. We used a combination of confocal and DNA-PAINT super-resolution microscopy to directly compare the organization of shared scaffold proteins at two important excitatory synapses-those forming onto excitatory principal neurons (Ex→Ex synapses) and those forming onto parvalbumin-expressing interneurons (Ex→PV synapses). As in Ex→Ex synapses, we find that in Ex→PV synapses, presynaptic Munc13-1 and postsynaptic PSD-95 both form NCs that demonstrate alignment, underscoring synaptic nanostructure and the transsynaptic nanocolumn as conserved organizational principles of excitatory synapses. Despite the general conservation of these features, we observed specific differences in the characteristics of pre- and postsynaptic Ex→PV nanostructure. Ex→PV synapses contained larger PSDs with fewer PSD-95 NCs when accounting for size than Ex→Ex synapses. Furthermore, the PSD-95 NCs were larger and denser. The identity of the postsynaptic cell was also represented in Munc13-1 organization, as Ex→PV synapses hosted larger Munc13-1 puncta that contained less dense but larger and more numerous Munc13-1 NCs. Moreover, we measured the spatial variability of transsynaptic alignment in these synapse types, revealing protein alignment in Ex→PV synapses over a distinct range of distances compared to Ex→Ex synapses. We conclude that while general principles of nanostructure and alignment are shared, cell-specific elements of nanodomain organization likely contribute to functional diversity of excitatory synapses.
Collapse
Affiliation(s)
- Poorna A. Dharmasri
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD21201
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD21201
- University of Maryland-Medicine Institute of Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, MD21201
| | - Aaron D. Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD21201
- University of Maryland-Medicine Institute of Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, MD21201
| | - Thomas A. Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD21201
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD21201
- University of Maryland-Medicine Institute of Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, MD21201
| |
Collapse
|
28
|
Nakhnikian A, Oribe N, Hirano S, Fujishima Y, Hirano Y, Nestor PG, Francis GA, Levin M, Spencer KM. Spectral decomposition of resting state electroencephalogram reveals unique theta/alpha activity in schizophrenia. Eur J Neurosci 2024; 59:1946-1960. [PMID: 38217348 DOI: 10.1111/ejn.16244] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 11/18/2023] [Accepted: 12/16/2023] [Indexed: 01/15/2024]
Abstract
Resting state electroencephalographic (EEG) activity in schizophrenia (SZ) is frequently characterised by increased power at slow frequencies and/or a reduction of peak alpha frequency. Here we investigated the nature of these effects. As most studies to date have been limited by reliance on a priori frequency bands which impose an assumed structure on the data, we performed a data-driven analysis of resting EEG recorded in SZ patients and healthy controls (HC). The sample consisted of 39 chronic SZ and 36 matched HC. The EEG was recorded with a dense electrode array. Power spectral densities were decomposed via Varimax-rotated principal component analysis (PCA) over all participants and for each group separately. Spectral PCA was repeated at the cortical level on cortical current source density computed from standardised low resolution brain electromagnetic tomography. There was a trend for power in the theta/alpha range to be increased in SZ compared to HC, and peak alpha frequency was significantly reduced in SZ. PCA revealed that this frequency shift was because of the presence of a spectral component in the theta/alpha range (6-9 Hz) that was unique to SZ. The source distribution of the SZ > HC theta/alpha effect involved mainly prefrontal and parahippocampal areas. Abnormal low frequency resting EEG activity in SZ was accounted for by a unique theta/alpha oscillation. Other reports have described a similar phenomenon suggesting that the neural circuits oscillating in this range are relevant to SZ pathophysiology.
Collapse
Affiliation(s)
- Alexander Nakhnikian
- Neural Dynamics Laboratory, Research Service, VA Boston Healthcare System, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Naoya Oribe
- Neural Dynamics Laboratory, Research Service, VA Boston Healthcare System, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
- Japan Imaging Center of Psychiatry and Neurology, Fukuoka, Japan
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shogo Hirano
- Neural Dynamics Laboratory, Research Service, VA Boston Healthcare System, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuki Fujishima
- Neural Dynamics Laboratory, Research Service, VA Boston Healthcare System, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Yoji Hirano
- Neural Dynamics Laboratory, Research Service, VA Boston Healthcare System, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Psychiatry, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Paul G Nestor
- Department of Psychology, University of Massachusetts, Boston, Massachusetts, USA
| | - Grace A Francis
- Neural Dynamics Laboratory, Research Service, VA Boston Healthcare System, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Kevin M Spencer
- Neural Dynamics Laboratory, Research Service, VA Boston Healthcare System, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
29
|
Holter KM, Lekander AD, Pierce BE, Sands LP, Gould RW. Use of Quantitative Electroencephalography to Inform Age- and Sex-Related Differences in NMDA Receptor Function Following MK-801 Administration. Pharmaceuticals (Basel) 2024; 17:237. [PMID: 38399452 PMCID: PMC10892193 DOI: 10.3390/ph17020237] [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: 01/19/2024] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Sex- and age-related differences in symptom prevalence and severity have been widely reported in patients with schizophrenia, yet the underlying mechanisms contributing to these differences are not well understood. N-methyl-D-aspartate (NMDA) receptor hypofunction contributes to schizophrenia pathology, and preclinical models often use NMDA receptor antagonists, including MK-801, to model all symptom clusters. Quantitative electroencephalography (qEEG) represents a translational approach to measure neuronal activity, identify targetable biomarkers in neuropsychiatric disorders and evaluate possible treatments. Abnormalities in gamma power have been reported in patients with schizophrenia and correspond to psychosis and cognitive impairment. Further, as gamma power reflects cortical glutamate and GABA signaling, it is highly sensitive to changes in NMDA receptor function, and NMDA receptor antagonists aberrantly increase gamma power in rodents and humans. To evaluate the role of sex and age on NMDA receptor function, MK-801 (0.03-0.3 mg/kg, SC) was administered to 3- and 9-month-old male and female Sprague-Dawley rats that were implanted with wireless EEG transmitters to measure cortical brain function. MK-801-induced elevations in gamma power were observed in 3-month-old male and female and 9-month-old male rats. In contrast, 9-month-old female rats demonstrated blunted maximal elevations across a wide dose range. Importantly, MK-801-induced hyperlocomotor effects, a common behavioral screen used to examine antipsychotic-like activity, were similar across all groups. Overall, sex-by-age-related differences in gamma power support using qEEG as a translational tool to evaluate pathological progression and predict treatment response across a heterogeneous population.
Collapse
Affiliation(s)
| | | | | | | | - Robert W. Gould
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (K.M.H.)
| |
Collapse
|
30
|
Díez Á, Gomez-Pilar J, Poza J, Beño-Ruiz-de-la-Sierra R, Fernández-Linsenbarth I, Recio-Barbero M, Núñez P, Holgado-Madera P, Molina V. Functional network properties in schizophrenia and bipolar disorder assessed with high-density electroencephalography. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110902. [PMID: 38036032 DOI: 10.1016/j.pnpbp.2023.110902] [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: 07/31/2023] [Revised: 11/10/2023] [Accepted: 11/24/2023] [Indexed: 12/02/2023]
Abstract
BACKGROUND The study of the cortical functional network properties in schizophrenia (SZ) may benefit from the use of graph theory parameters applied to high-density electroencephalography (EEG). Connectivity Strength (CS) assesses global synchrony of the network, and Shannon Graph Complexity (SGC) summarizes the network distribution of link weights and allows distinguishing between primary and secondary pathways. Their joint use may help in understanding the underpinnings of the functional network hyperactivation and task-related hypomodulation previously described in psychoses. METHODS We used 64-sensor EEG recordings during a P300 oddball task in 128 SZ patients (96 chronic, CR, and 32 first episodes, FE), as well as 46 bipolar disorder (BD) patients, and 92 healthy controls (HC). Pre-stimulus and modulation (task-response minus pre-stimulus windows values) of CS and SGC were assessed in the theta band (4-8 Hz) and the broadband (4-70 Hz). RESULTS Compared to HC, SZ patients (CR and FE) showed significantly higher pre-stimulus CS values in the broadband, and both SZ and BD patients showed lower theta-band CS modulation. SGC modulation values, both theta-band and broadband, were also abnormally reduced in CR patients. Statistically significant relationships were found in the theta band between SGC modulation and both CS pre-stimulus and modulation values in patients. CS altered measures in patients were additionally related to their cognitive outcome and negative symptoms. A primary role of antipsychotics in these results was ruled out. CONCLUSIONS Our results linking SGC and CS alterations in psychotic patients supported a hyperactive and hypomodulatory network mainly involving connections in secondary pathways.
Collapse
Affiliation(s)
- Álvaro Díez
- Psychiatry Department, School of Medicine, University of Valladolid, Valladolid, Spain
| | - Javier Gomez-Pilar
- Biomedical Engineering Group, University of Valladolid, Valladolid, Spain.; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Valladolid, Spain
| | - Jesús Poza
- Biomedical Engineering Group, University of Valladolid, Valladolid, Spain.; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Valladolid, Spain
| | | | | | | | - Pablo Núñez
- Biomedical Engineering Group, University of Valladolid, Valladolid, Spain.; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Valladolid, Spain.; Coma Science Group, GIGA-Consciousness, University of Liège, Liège, Belgium
| | | | - Vicente Molina
- Psychiatry Department, School of Medicine, University of Valladolid, Valladolid, Spain.; Psychiatry Service, Clinical University Hospital of Valladolid, Valladolid, Spain..
| |
Collapse
|
31
|
Spencer KM. Gamma Oscillations as a Biomarker of Neural Circuit Function in Psychosis: Where Are We, and Where Do We Go from Here? ADVANCES IN NEUROBIOLOGY 2024; 40:321-349. [PMID: 39562450 DOI: 10.1007/978-3-031-69491-2_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
This chapter is a selective and critical review of the literature on gamma oscillations in schizophrenia and related studies in other relevant fields that pertain to the hypothesis that abnormal gamma oscillations underlie symptoms of psychosis in individuals with schizophrenia. These gamma abnormalities result from deficient recurrent inhibition, in which parvalbumin-expressing, fast-spiking inhibitory interneurons do not receive sufficient excitation from N-methyl-D-aspartate receptors, resulting in a loss of phasic control over pyramidal cell spiking and impairment of gamma generation. The evidence for this hypothesis is critically reviewed, focusing on studies in the areas of visual feature binding, auditory steady-state response, and spontaneous gamma activity. The current state of the field is discussed, and recommendations for future directions are presented.
Collapse
Affiliation(s)
- Kevin M Spencer
- Research Service, VA Boston Healthcare System, Boston, MA, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
32
|
Hamilton HK, Mathalon DH. Neurophysiological Models in Individuals at Clinical High Risk for Psychosis: Using Translational EEG Paradigms to Forecast Psychosis Risk and Resilience. ADVANCES IN NEUROBIOLOGY 2024; 40:385-410. [PMID: 39562452 DOI: 10.1007/978-3-031-69491-2_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Over the last several decades, there have been major research efforts to improve the identification of youth and young adults at clinical high-risk for psychosis (CHR-P). Among individuals identified as CHR-P based on clinical criteria, approximately 20% progress to full-blown psychosis over 2-3 years and 30% achieve remission. In more recent years, neurophysiological measures with established sensitivity to schizophrenia have gained traction in the study of CHR-P and its range of clinical outcomes, with the goal of identifying specific biomarkers that precede psychosis onset that 7 chapter, we review studies examining several translational electroencephalography (EEG) and event-related potential (ERP) measures, which have known sensitivity to schizophrenia and reflect abnormal sensory, perceptual, and cognitive processing of task stimuli, as predictors of future clinical outcomes in CHR-P individuals. We discuss the promise of these EEG/ERP biomarkers of psychosis risk, including their potential to provide (a) translational bridges between human studies and animal models focused on drug development for early psychosis, (b) target engagement measures for clinical trials, and (c) prognostic indicators that could enhance personalized treatment planning.
Collapse
Affiliation(s)
- Holly K Hamilton
- Department of Psychiatry & Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Psychiatry & Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
| | - Daniel H Mathalon
- Department of Psychiatry & Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA.
- San Francisco Veterans Affairs Health Care System, San Francisco, CA, USA.
| |
Collapse
|
33
|
Tranter MM, Faget L, Hnasko TS, Powell SB, Dillon DG, Barnes SA. Postnatal Phencyclidine-Induced Deficits in Decision Making Are Ameliorated by Optogenetic Inhibition of Ventromedial Orbitofrontal Cortical Glutamate Neurons. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:264-274. [PMID: 38298783 PMCID: PMC10829674 DOI: 10.1016/j.bpsgos.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/12/2023] [Accepted: 08/01/2023] [Indexed: 02/02/2024] Open
Abstract
Background The orbitofrontal cortex (OFC) is essential for decision making, and functional disruptions within the OFC are evident in schizophrenia. Postnatal phencyclidine (PCP) administration in rats is a neurodevelopmental manipulation that induces schizophrenia-relevant cognitive impairments. We aimed to determine whether manipulating OFC glutamate cell activity could ameliorate postnatal PCP-induced deficits in decision making. Methods Male and female Wistar rats (n = 110) were administered saline or PCP on postnatal days 7, 9, and 11. In adulthood, we expressed YFP (yellow fluorescent protein) (control), ChR2 (channelrhodopsin-2) (activation), or eNpHR 3.0 (enhanced halorhodopsin) (inhibition) in glutamate neurons within the ventromedial OFC (vmOFC). Rats were tested on the probabilistic reversal learning task once daily for 20 days while we manipulated the activity of vmOFC glutamate cells. Behavioral performance was analyzed using a Q-learning computational model of reinforcement learning. Results Compared with saline-treated rats expressing YFP, PCP-treated rats expressing YFP completed fewer reversals, made fewer win-stay responses, and had lower learning rates. We induced similar performance impairments in saline-treated rats by activating vmOFC glutamate cells (ChR2). Strikingly, PCP-induced performance deficits were ameliorated when the activity of vmOFC glutamate cells was inhibited (halorhodopsin). Conclusions Postnatal PCP-induced deficits in decision making are associated with hyperactivity of vmOFC glutamate cells. Thus, normalizing vmOFC activity may represent a potential therapeutic target for decision-making deficits in patients with schizophrenia.
Collapse
Affiliation(s)
- Michael M. Tranter
- Department of Psychiatry, University of California San Diego, La Jolla, California
- Research Service, VA San Diego Healthcare System, La Jolla, California
| | - Lauren Faget
- Department of Neurosciences, University of California San Diego, La Jolla, California
| | - Thomas S. Hnasko
- Research Service, VA San Diego Healthcare System, La Jolla, California
- Department of Neurosciences, University of California San Diego, La Jolla, California
| | - Susan B. Powell
- Department of Psychiatry, University of California San Diego, La Jolla, California
- Research Service, VA San Diego Healthcare System, La Jolla, California
| | - Daniel G. Dillon
- Center for Depression, Anxiety and Stress Research, McLean Hospital, Belmont, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Samuel A. Barnes
- Department of Psychiatry, University of California San Diego, La Jolla, California
- Research Service, VA San Diego Healthcare System, La Jolla, California
| |
Collapse
|
34
|
Santos-Silva T, dos Santos Fabris D, de Oliveira CL, Guimarães FS, Gomes FV. Prefrontal and Hippocampal Parvalbumin Interneurons in Animal Models for Schizophrenia: A Systematic Review and Meta-analysis. Schizophr Bull 2024; 50:210-223. [PMID: 37584417 PMCID: PMC10754178 DOI: 10.1093/schbul/sbad123] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
BACKGROUND Consistent with postmortem findings in patients, most animal models for schizophrenia (SCZ) present abnormal levels of parvalbumin (PV), a marker of fast-spiking GABAergic interneurons, in the prefrontal cortex (PFC) and hippocampus (HIP). However, there are discrepancies in the literature. PV reductions lead to a functional loss of PV interneurons, which is proposed to underly SCZ symptoms. Given its complex etiology, different categories of animal models have been developed to study SCZ, which may distinctly impact PV levels in rodent brain areas. STUDY DESIGN We performed a quantitative meta-analysis on PV-positive cell number/density and expression levels in the PFC and HIP of animal models for SCZ based on pharmacological, neurodevelopmental, and genetic manipulations. RESULTS Our results confirmed that PV levels are significantly reduced in the PFC and HIP regardless of the animal model. By categorizing into subgroups, we found that all pharmacological models based on NMDA receptor antagonism decreased PV-positive cell number/density or PV expression levels in both brain areas examined. In neurodevelopmental models, abnormal PV levels were confirmed in both brain areas in maternal immune activation models and HIP of the methylazoxymethanol acetate model. In genetic models, negative effects were found in neuregulin 1 and ERBB4 mutant mice in both brain regions and the PFC of dysbindin mutant mice. Regarding sex differences, male rodents exhibited PV reductions in both brain regions only in pharmacological models, while few studies have been conducted in females. CONCLUSION Overall, our findings support deficits in prefrontal and hippocampal PV interneurons in animal models for SCZ.
Collapse
Affiliation(s)
- Thamyris Santos-Silva
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Débora dos Santos Fabris
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Cilene Lino de Oliveira
- Department of Physiological Sciences, Center of Biological Sciences, University of Santa Catarina, Florianópolis,Brazil
| | - Francisco S Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Felipe V Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
35
|
Zhang XQ, Xu L, Zhu XY, Tang ZH, Dong YB, Yu ZP, Shang Q, Wang ZC, Shen HW. D-serine reconstitutes synaptic and intrinsic inhibitory control of pyramidal neurons in a neurodevelopmental mouse model for schizophrenia. Nat Commun 2023; 14:8255. [PMID: 38086803 PMCID: PMC10716516 DOI: 10.1038/s41467-023-43930-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
The hypothesis of N-methyl-D-aspartate receptor (NMDAR) dysfunction for cognitive impairment in schizophrenia constitutes the theoretical basis for the translational application of NMDAR co-agonist D-serine or its analogs. However, the cellular mechanism underlying the therapeutic effect of D-serine remains unclear. In this study, we utilize a mouse neurodevelopmental model for schizophrenia that mimics prenatal pathogenesis and exhibits hypoexcitability of parvalbumin-positive (PV) neurons, as well as PV-preferential NMDAR dysfunction. We find that D-serine restores excitation/inhibition balance by reconstituting both synaptic and intrinsic inhibitory control of cingulate pyramidal neurons through facilitating PV excitability and activating small-conductance Ca2+-activated K+ (SK) channels in pyramidal neurons, respectively. Either amplifying inhibitory drive via directly strengthening PV neuron activity or inhibiting pyramidal excitability via activating SK channels is sufficient to improve cognitive function in this model. These findings unveil a dual mechanism for how D-serine improves cognitive function in this model.
Collapse
Affiliation(s)
- Xiao-Qin Zhang
- Department of Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Le Xu
- Department of Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Xin-Yi Zhu
- Department of Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Zi-Hang Tang
- Department of Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Yi-Bei Dong
- Department of Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Zhi-Peng Yu
- Department of Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Qing Shang
- Department of Neurology, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, Zhejiang, 315211, China
| | - Zheng-Chun Wang
- Department of Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Hao-Wei Shen
- Department of Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China.
| |
Collapse
|
36
|
Cattarinussi G, Grimaldi DA, Sambataro F. Spontaneous Brain Activity Alterations in First-Episode Psychosis: A Meta-analysis of Functional Magnetic Resonance Imaging Studies. Schizophr Bull 2023; 49:1494-1507. [PMID: 38029279 PMCID: PMC10686347 DOI: 10.1093/schbul/sbad044] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
BACKGROUND AND HYPOTHESIS Several studies have shown that spontaneous brain activity, including the total and fractional amplitude of low-frequency fluctuations (LFF) and regional homogeneity (ReHo), is altered in psychosis. Nonetheless, neuroimaging results show a high heterogeneity. For this reason, we gathered the extant literature on spontaneous brain activity in first-episode psychosis (FEP), where the effects of long-term treatment and chronic disease are minimal. STUDY DESIGN A systematic research was conducted on PubMed, Scopus, and Web of Science to identify studies exploring spontaneous brain activity and local connectivity in FEP estimated using functional magnetic resonance imaging. 20 LFF and 15 ReHo studies were included. Coordinate-Based Activation Likelihood Estimation Meta-Analyses stratified by brain measures, age (adolescent vs adult), and drug-naïve status were performed to identify spatially-convergent alterations in spontaneous brain activity in FEP. STUDY RESULTS We found a significant increase in LFF in FEP compared to healthy controls (HC) in the right striatum and in ReHo in the left striatum. When pooling together all studies on LFF and ReHo, spontaneous brain activity was increased in the bilateral striatum and superior and middle frontal gyri and decreased in the right precentral gyrus and the right inferior frontal gyrus compared to HC. These results were also replicated in the adult and drug-naïve samples. CONCLUSIONS Abnormalities in the frontostriatal circuit are present in early psychosis independently of treatment status. Our findings support the view that altered frontostriatal can represent a core neural alteration of the disorder and could be a target of treatment.
Collapse
Affiliation(s)
- Giulia Cattarinussi
- Department of Neuroscience (DNS), University of Padova, Padua, Italy
- Department of Neuroscience (DNS), Padova Neuroscience Center, University of Padova, Padua, Italy
| | | | - Fabio Sambataro
- Department of Neuroscience (DNS), University of Padova, Padua, Italy
- Department of Neuroscience (DNS), Padova Neuroscience Center, University of Padova, Padua, Italy
| |
Collapse
|
37
|
Susin E, Destexhe A. A Network Model of the Modulation of γ Oscillations by NMDA Receptors in Cerebral Cortex. eNeuro 2023; 10:ENEURO.0157-23.2023. [PMID: 37940562 PMCID: PMC10668239 DOI: 10.1523/eneuro.0157-23.2023] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 11/10/2023] Open
Abstract
Psychotic drugs such as ketamine induce symptoms close to schizophrenia and stimulate the production of γ oscillations, as also seen in patients, but the underlying mechanisms are still unclear. Here, we have used computational models of cortical networks generating γ oscillations, and have integrated the action of drugs such as ketamine to partially block NMDA receptors (NMDARs). The model can reproduce the paradoxical increase of γ oscillations by NMDA receptor antagonists, assuming that antagonists affect NMDA receptors with higher affinity on inhibitory interneurons. We next used the model to compare the responsiveness of the network to external stimuli, and found that when NMDA channels are blocked, an increase of γ power is observed altogether with an increase of network responsiveness. However, this responsiveness increase applies not only to γ states, but also to asynchronous states with no apparent γ. We conclude that NMDA antagonists induce an increased excitability state, which may or may not produce γ oscillations, but the response to external inputs is exacerbated, which may explain phenomena such as altered perception or hallucinations.
Collapse
Affiliation(s)
- Eduarda Susin
- Institute of Neuroscience (NeuroPSI), Paris-Saclay University, Centre National de la Recherche Scientifique (CNRS), Saclay, France 91400
| | - Alain Destexhe
- Institute of Neuroscience (NeuroPSI), Paris-Saclay University, Centre National de la Recherche Scientifique (CNRS), Saclay, France 91400
| |
Collapse
|
38
|
Cao H, Lencz T, Gallego JA, Rubio JM, John M, Barber AD, Birnbaum ML, Robinson DG, Malhotra AK. A Functional Connectome-Based Neural Signature for Individualized Prediction of Antipsychotic Response in First-Episode Psychosis. Am J Psychiatry 2023; 180:827-835. [PMID: 37644811 PMCID: PMC11104773 DOI: 10.1176/appi.ajp.20220719] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
OBJECTIVE Identification of robust biomarkers that predict individualized response to antipsychotic treatment at the early stage of psychotic disorders remains a challenge in precision psychiatry. The aim of this study was to investigate whether any functional connectome-based neural traits could serve as such a biomarker. METHODS In a discovery sample, 49 patients with first-episode psychosis received multi-paradigm fMRI scans at baseline and were clinically followed up for 12 weeks under antipsychotic monotherapies. Treatment response was evaluated at the individual level based on the psychosis score of the Brief Psychiatric Rating Scale. Cross-paradigm connectivity and connectome-based predictive modeling were employed to train a predictive model that uses baseline connectomic measures to predict individualized change rates of psychosis scores, with model performance evaluated as the Pearson correlations between the predicted change rates and the observed change rates, based on cross-validation. The model generalizability was further examined in an independent validation sample of 24 patients in a similar design. RESULTS The results revealed a paradigm-independent connectomic trait that significantly predicted individualized treatment outcome in both the discovery sample (predicted-versus-observed r=0.41) and the validation sample (predicted-versus-observed r=0.47, mean squared error=0.019). Features that positively predicted psychosis change rates primarily involved connections related to the cerebellar-cortical circuitry, and features that negatively predicted psychosis change rates were chiefly connections within the cortical cognitive systems. CONCLUSIONS This study discovers and validates a connectome-based functional signature as a promising early predictor for individualized response to antipsychotic treatment in first-episode psychosis, thus highlighting the potential clinical value of this biomarker in precision psychiatry.
Collapse
Affiliation(s)
- Hengyi Cao
- Institute of Behavioral Sciences, Feinstein Institutes for Medical Research, Manhasset, N.Y. (all authors); Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, N.Y. (all authors); Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, N.Y. (all authors)
| | - Todd Lencz
- Institute of Behavioral Sciences, Feinstein Institutes for Medical Research, Manhasset, N.Y. (all authors); Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, N.Y. (all authors); Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, N.Y. (all authors)
| | - Juan A Gallego
- Institute of Behavioral Sciences, Feinstein Institutes for Medical Research, Manhasset, N.Y. (all authors); Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, N.Y. (all authors); Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, N.Y. (all authors)
| | - Jose M Rubio
- Institute of Behavioral Sciences, Feinstein Institutes for Medical Research, Manhasset, N.Y. (all authors); Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, N.Y. (all authors); Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, N.Y. (all authors)
| | - Majnu John
- Institute of Behavioral Sciences, Feinstein Institutes for Medical Research, Manhasset, N.Y. (all authors); Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, N.Y. (all authors); Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, N.Y. (all authors)
| | - Anita D Barber
- Institute of Behavioral Sciences, Feinstein Institutes for Medical Research, Manhasset, N.Y. (all authors); Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, N.Y. (all authors); Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, N.Y. (all authors)
| | - Michael L Birnbaum
- Institute of Behavioral Sciences, Feinstein Institutes for Medical Research, Manhasset, N.Y. (all authors); Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, N.Y. (all authors); Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, N.Y. (all authors)
| | - Delbert G Robinson
- Institute of Behavioral Sciences, Feinstein Institutes for Medical Research, Manhasset, N.Y. (all authors); Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, N.Y. (all authors); Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, N.Y. (all authors)
| | - Anil K Malhotra
- Institute of Behavioral Sciences, Feinstein Institutes for Medical Research, Manhasset, N.Y. (all authors); Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, N.Y. (all authors); Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, N.Y. (all authors)
| |
Collapse
|
39
|
Flynn LT, Gao WJ. DNA methylation and the opposing NMDAR dysfunction in schizophrenia and major depression disorders: a converging model for the therapeutic effects of psychedelic compounds in the treatment of psychiatric illness. Mol Psychiatry 2023; 28:4553-4567. [PMID: 37679470 PMCID: PMC11034997 DOI: 10.1038/s41380-023-02235-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/09/2023]
Abstract
Psychedelic compounds are being increasingly explored as a potential therapeutic option for treating several psychiatric conditions, despite relatively little being known about their mechanism of action. One such possible mechanism, DNA methylation, is a process of epigenetic regulation that changes gene expression via chemical modification of nitrogenous bases. DNA methylation has been implicated in the pathophysiology of several psychiatric conditions, including schizophrenia (SZ) and major depressive disorder (MDD). In this review, we propose alterations to DNA methylation as a converging model for the therapeutic effects of psychedelic compounds, highlighting the N-methyl D-aspartate receptor (NMDAR), a crucial mediator of synaptic plasticity with known dysfunction in both diseases, as an example and anchoring point. We review the established evidence relating aberrant DNA methylation to NMDAR dysfunction in SZ and MDD and provide a model asserting that psychedelic substances may act through an epigenetic mechanism to provide therapeutic effects in the context of these disorders.
Collapse
Affiliation(s)
- L Taylor Flynn
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
- MD/PhD program, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Wen-Jun Gao
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
40
|
Dugan C, Zikopoulos B, Yazdanbakhsh A. A neural modeling approach to study mechanisms underlying the heterogeneity of visual spatial frequency sensitivity in schizophrenia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.18.563001. [PMID: 37904992 PMCID: PMC10614973 DOI: 10.1101/2023.10.18.563001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Patients with schizophrenia exhibit abnormalities in spatial frequency sensitivity, and it is believed that these abnormalities indicate more widespread dysfunction and dysregulation of bottom-up processing. The early visual system, including the first-order Lateral Geniculate Nucleus of the thalamus (LGN) and the primary visual cortex (V1), are key contributors to spatial frequency sensitivity. Medicated and unmedicated patients with schizophrenia exhibit contrasting changes in spatial frequency sensitivity, thus making it a useful probe for examining potential effects of the disorder and antipsychotic medications in neural processing. We constructed a parameterized, rate-based neural model of on-center/off-surround neurons in the early visual system to investigate the impacts of changes to the excitatory and inhibitory receptive field subfields. By incorporating changes in both the excitatory and inhibitory subfields that are associated with pathophysiological findings in schizophrenia, the model successfully replicated perceptual data from behavioral/functional studies involving medicated and unmedicated patients. Among several plausible mechanisms, our results highlight the dampening of excitation and/or increase in the spread and strength of the inhibitory subfield in medicated patients and the contrasting decreased spread and strength of inhibition in unmedicated patients. Given that the model was successful at replicating results from perceptual data under a variety of conditions, these elements of the receptive field may be useful markers for the imbalances seen in patients with schizophrenia.
Collapse
Affiliation(s)
- Caroline Dugan
- Program in Neuroscience, Boston University, Boston, MA, United States
| | - Basilis Zikopoulos
- Human Systems Neuroscience Laboratory, Department of Health Sciences, Boston University, Boston, MA, United States
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
- Center for Systems Neuroscience, Boston University, Boston, MA, United States
- Graduate Program for Neuroscience, Boston University, Boston, MA, United States
| | - Arash Yazdanbakhsh
- Center for Systems Neuroscience, Boston University, Boston, MA, United States
- Graduate Program for Neuroscience, Boston University, Boston, MA, United States
- Computational Neuroscience and Vision Laboratory, Department of Psychological and Brain Sciences, Boston University, Boston, MA, United States
| |
Collapse
|
41
|
Al-youzbaki MU, Schormans AL, Allman BL. Past and present experience shifts audiovisual temporal perception in rats. Front Behav Neurosci 2023; 17:1287587. [PMID: 37908200 PMCID: PMC10613659 DOI: 10.3389/fnbeh.2023.1287587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 09/25/2023] [Indexed: 11/02/2023] Open
Abstract
Our brains have a propensity to integrate closely-timed auditory and visual stimuli into a unified percept; a phenomenon that is highly malleable based on prior sensory experiences, and is known to be altered in clinical populations. While the neural correlates of audiovisual temporal perception have been investigated using neuroimaging and electroencephalography techniques in humans, animal research will be required to uncover the underlying cellular and molecular mechanisms. Prior to conducting such mechanistic studies, it is important to first confirm the translational potential of any prospective animal model. Thus, in the present study, we conducted a series of experiments to determine if rats show the hallmarks of audiovisual temporal perception observed in neurotypical humans, and whether the rat behavioral paradigms could reveal when they experienced perceptual disruptions akin to those observed in neurodevelopmental disorders. After training rats to perform a temporal order judgment (TOJ) or synchrony judgment (SJ) task, we found that the rats' perception was malleable based on their past and present sensory experiences. More specifically, passive exposure to asynchronous audiovisual stimulation in the minutes prior to behavioral testing caused the rats' perception to predictably shift in the direction of the leading stimulus; findings which represent the first time that this form of audiovisual perceptual malleability has been reported in non-human subjects. Furthermore, rats performing the TOJ task also showed evidence of rapid recalibration, in which their audiovisual temporal perception on the current trial was predictably influenced by the timing lag between the auditory and visual stimuli in the preceding trial. Finally, by manipulating either experimental testing parameters or altering the rats' neurochemistry with a systemic injection of MK-801, we showed that the TOJ and SJ tasks could identify when the rats had difficulty judging the timing of audiovisual stimuli. These findings confirm that the behavioral paradigms are indeed suitable for future testing of rats with perceptual disruptions in audiovisual processing. Overall, our collective results highlight that rats represent an excellent animal model to study the cellular and molecular mechanisms underlying the acuity and malleability of audiovisual temporal perception, as they showcase the perceptual hallmarks commonly observed in humans.
Collapse
|
42
|
Rosenbrock H, Desch M, Wunderlich G. Development of the novel GlyT1 inhibitor, iclepertin (BI 425809), for the treatment of cognitive impairment associated with schizophrenia. Eur Arch Psychiatry Clin Neurosci 2023; 273:1557-1566. [PMID: 36971864 PMCID: PMC10465677 DOI: 10.1007/s00406-023-01576-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
Schizophrenia is a psychiatric disorder characterised by symptoms in three domains: positive (e.g. delusions, hallucinations), negative (e.g. social withdrawal, lack of motivation) and cognitive (e.g. working memory and executive function impairment). Cognitive impairment associated with schizophrenia (CIAS) is a major burden for patients and negatively impacts many aspects of a patient's life. Antipsychotics are the standard-of-care treatment for schizophrenia but only address positive symptoms. So far there are no approved pharmacotherapies for the treatment of CIAS. Iclepertin (BI 425809) is a novel, potent and selective glycine transporter 1 (GlyT1) inhibitor, under development by Boehringer Ingelheim for the treatment of CIAS. Phase I studies have shown it to be safe and well tolerated in healthy volunteers, and central target engagement (inhibition of GlyT1) was achieved in a dose-dependent manner from 5 to 50 mg in healthy volunteers. A Phase II study has demonstrated that iclepertin is safe and well tolerated in patients with schizophrenia and improves cognition at doses of 10 mg and 25 mg. Phase III studies are ongoing to confirm these initial positive safety and efficacy findings with the 10 mg dose, and if successful, iclepertin could become the first approved pharmacotherapy used to treat CIAS.
Collapse
Affiliation(s)
- Holger Rosenbrock
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Michael Desch
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Glen Wunderlich
- Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Road, Ridgefield, CT 06877, USA.
| |
Collapse
|
43
|
Dharmasri PA, Levy AD, Blanpied TA. Differential nanoscale organization of excitatory synapses onto excitatory vs inhibitory neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.06.556279. [PMID: 37732271 PMCID: PMC10508768 DOI: 10.1101/2023.09.06.556279] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
A key feature of excitatory synapses is the existence of subsynaptic protein nanoclusters whose precise alignment across the cleft in a trans-synaptic nanocolumn influences the strength of synaptic transmission. However, whether nanocolumn properties vary between excitatory synapses functioning in different cellular contexts is unknown. We used a combination of confocal and DNA-PAINT super-resolution microscopy to directly compare the organization of shared scaffold proteins at two important excitatory synapses - those forming onto excitatory principal neurons (Ex→Ex synapses) and those forming onto parvalbumin-expressing interneurons (Ex→PV synapses). As in Ex→Ex synapses, we find that in Ex→PV synapses presynaptic Munc13-1 and postsynaptic PSD-95 both form nanoclusters that demonstrate alignment, underscoring synaptic nanostructure and the trans-synaptic nanocolumn as conserved organizational principles of excitatory synapses. Despite the general conservation of these features, we observed specific differences in the characteristics of pre- and postsynaptic Ex→PV nanostructure. Ex→PV synapses contained larger PSDs with fewer PSD-95 NCs when accounting for size than Ex→Ex synapses. Furthermore, the PSD-95 NCs were larger and denser. The identity of the postsynaptic cell also had a retrograde impact on Munc13-1 organization, as Ex→PV synapses hosted larger Munc13-1 puncta that contained less dense but larger and more numerous Munc13-1 NCs. Moreover, we measured the spatial variability of transsynaptic alignment in these synapse types, revealing protein alignment in Ex→PV synapses over a distinct range of distances compared to Ex→Ex synapses. We conclude that while general principles of nanostructure and alignment are shared, cell-specific elements of nanodomain organization likely contribute to functional diversity of excitatory synapses. Understanding the rules of synapse nanodomain assembly, which themselves are cell-type specific, will be essential for illuminating brain network dynamics.
Collapse
Affiliation(s)
- Poorna A Dharmasri
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201
- University of Maryland Medicine Institute of Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Aaron D Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- University of Maryland Medicine Institute of Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201
- University of Maryland Medicine Institute of Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
44
|
Anand R, Turolla A, Chinellato G, Roy A, Hartman RD. Phase 2 Results Indicate Evenamide, A Selective Modulator of Glutamate Release, Is Associated With Clinically Important Long-Term Efficacy When Added to an Antipsychotic in Patients With Treatment-Resistant Schizophrenia. Int J Neuropsychopharmacol 2023; 26:523-528. [PMID: 37349110 PMCID: PMC10464926 DOI: 10.1093/ijnp/pyad035] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/21/2023] [Indexed: 06/24/2023] Open
Abstract
Results from a pilot, 6-week, randomized, open-label, rater-blinded study, with 46-week extension, indicate very good tolerability with exceptional, clinically important, increasing efficacy of evenamide (7.5, 15, and 30 mg bid), a glutamate modulator, as add-on treatment to antipsychotics in 161 treatment-resistant, schizophrenia patients. Ninety-five percent of patients completed 6 weeks (1 discontinued for adverse event), and 89% continued in the extension. Results from the first 100 patients enrolled showed very low attrition over 1 year (77 completers); data pooled from all dose groups showed the Positive and Negative Syndrome Scale total score improved significantly (P < .001; paired t test; last observation carried forward [LOCF]) from baseline at 6 weeks (-9.4), 6 months (-12.7), and 1 year (-14.7); similarly, the proportion of responders (≥20% improvement) increased over time from 6 weeks (16.5%) to 6 months (39%) to 1 year (47.4%). Noteworthy improvement was also observed at each timepoint on the Clinical Global Impression - Severity scale and Clinical Global Impression of Change, indicating progressively increasing efficacy of evenamide up to 1 year.
Collapse
Affiliation(s)
| | | | | | - Arjun Roy
- CliniRx Research Pvt Ltd, New Delhi, India
| | | |
Collapse
|
45
|
Jiang LX, Huang GD, Tian YL, Cong RX, Meng X, Wang HL, Zhang C, Yu X. Diminished activation of excitatory neurons in the prelimbic cortex leads to impaired working memory capacity in mice. BMC Biol 2023; 21:171. [PMID: 37568146 PMCID: PMC10416384 DOI: 10.1186/s12915-023-01674-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Working memory capacity impairment is an early sign of Alzheimer's disease, but the underlying mechanisms remain unclear. Clarifying how working memory capacity is affected will help us better understand the pathological mechanism of Alzheimer's disease. We used the olfactory working memory capacity paradigm to evaluate memory capacity in 3-month-old 5XFAD (an animal model of Alzheimer's disease) mice. Immunofluorescence staining of the prefrontal cortex was performed to detect the number of FOS-positive neurons, calmodulin-dependent protein kinase II-positive neurons, and glutamate decarboxylase-positive neurons in the prelimbic cortex and infralimbic cortex. A chemogenetic method was then used to modulate the inhibition and activation of excitatory neurons in the prelimbic cortex of wild-type and 5XFAD mice and to measure the memory capacity of mice. RESULTS Working memory capacity was significantly diminished in 5XFAD mice compared to littermate wild-type mice. Neuronal activation of the prelimbic cortex, but not the infralimbic cortex, was attenuated in 5XFAD mice performing the olfactory working memory capacity task. Subsequently, the FOS-positive neurons were co-localized with both calmodulin-dependent protein kinase II-positive neurons and glutamate decarboxylase-positive neurons. The results showed that the activation of excitatory neurons in the prelimbic cortex was correlated with working memory capacity in mice. Our results further demonstrate that the chemogenetic inhibition of prelimbic cortex excitatory neurons resulted in reduced working memory capacity in wild-type mice, while the chemogenetic activation of prelimbic cortex excitatory neurons improved the working memory capacity of 5XFAD mice. CONCLUSION The diminished activation of prelimbic cortex excitatory neurons in 5XFAD mice during task performance is associated with reduced working memory capacity, and activation modulation of excitatory neurons by chemogenetic methods can improve memory capacity impairment in 5XFAD mice. These findings may provide a new direction for exploring Alzheimer's disease therapeutic approaches.
Collapse
Affiliation(s)
- Li-Xin Jiang
- Peking University Institute of Mental Health (Sixth Hospital), No.51 Huayuanbei Road, Haidian District, Beijing, 100191, China
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University), Beijing, 100191, China
- Beijing Municipal Key Laboratory for Translational Research On Diagnosis and Treatment of Dementia, Beijing, 100191, China
| | - Geng-Di Huang
- Department of Addiction Medicine, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Mental Health Center, Shenzhen Kangning Hospital, No.77 Zhenbi Road, Pingshan District, Shenzhen, 518118, China
- Affiliated Mental Health Center, Southern University of Science and Technology, No.1088 Xueyuan Avenue, Fuguang Community, Taoyuan Street, Nanshan District, Shenzhen, 518118, China
| | - Yong-Lu Tian
- School of Psychological and Cognitive Sciences, Peking University, No.5 Summer Palace Road, Haidian District, Beijing, 100871, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Ri-Xu Cong
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Xue Meng
- National Center of Gerontology, Beijing Hospital, No.1 Dahua Road, Dongdan, Dongcheng District, Beijing, 100005, China
| | - Hua-Li Wang
- Peking University Institute of Mental Health (Sixth Hospital), No.51 Huayuanbei Road, Haidian District, Beijing, 100191, China.
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University), Beijing, 100191, China.
- Beijing Municipal Key Laboratory for Translational Research On Diagnosis and Treatment of Dementia, Beijing, 100191, China.
| | - Chen Zhang
- Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, School of Basic Medical Sciences, Capital Medical University, No.10 Xitoutiao, You'anmenwai, Fengtai District, Beijing, 100069, China.
| | - Xin Yu
- Peking University Institute of Mental Health (Sixth Hospital), No.51 Huayuanbei Road, Haidian District, Beijing, 100191, China.
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University), Beijing, 100191, China.
- Beijing Municipal Key Laboratory for Translational Research On Diagnosis and Treatment of Dementia, Beijing, 100191, China.
| |
Collapse
|
46
|
Maness EB, Blumenthal SA, Burk JA. Dual orexin/hypocretin receptor antagonism attenuates NMDA receptor hypofunction-induced attentional impairments in a rat model of schizophrenia. Behav Brain Res 2023; 450:114497. [PMID: 37196827 PMCID: PMC10330488 DOI: 10.1016/j.bbr.2023.114497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/19/2023]
Abstract
Schizophrenia is a neuropsychiatric condition that is associated with impaired attentional processing and performance. Failure to support increasing attentional load may result, in part, from inhibitory failure in attention-relevant cortical regions, and available antipsychotics often fail to address this issue. Orexin/hypocretin receptors are found throughout the brain and are expressed on neurons relevant to both attention and schizophrenia, highlighting them as a potential target to treat schizophrenia-associated attentional dysfunction. In the present experiment, rats (N = 14) trained in a visual sustained attention task that required discrimination of trials which presented a visual signal from trials during which no signal was presented. Once trained, rats were then co-administered the psychotomimetic N-methyl-D-aspartate (NMDA) receptor antagonist dizocilpine (MK-801: 0 or 0.1 mg/kg, intraperitoneal injections) and the dual orexin receptor antagonist filorexant (MK-6096: 0, 0.1, or 1 mM, intracerebroventricular infusions) prior to task performance across six sessions. Dizocilpine impaired overall accuracy during signal trials, slowed reaction times for correctly-responded trials, and increased the number of omitted trials throughout the task. Dizocilpine-induced increases in signal trial deficits, correct response latencies, and errors of omission were reduced following infusions of the 0.1 mM, but not 1 mM, dose of filorexant. As such, orexin receptor blockade may improve attentional deficits in a state of NMDA receptor hypofunction.
Collapse
Affiliation(s)
- Eden B Maness
- Department of Psychological Sciences, College of William and Mary, Williamsburg, VA 23187, USA; VA Boston Healthcare System and Department of Psychiatry, Harvard Medical School, West Roxbury, MA 02132, USA.
| | - Sarah A Blumenthal
- Center for Translational Social Neuroscience, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Joshua A Burk
- Department of Psychological Sciences, College of William and Mary, Williamsburg, VA 23187, USA
| |
Collapse
|
47
|
Deriha K, Hashimoto E, Ukai W, Marchisella F, Nishimura E, Hashiguchi H, Tayama M, Ishii T, Riva MA, Kawanishi C. Reduced sociability in a prenatal immune activation model: Modulation by a chronic blonanserin treatment through the amygdala-hippocampal axis. J Psychiatr Res 2023; 164:209-220. [PMID: 37379611 DOI: 10.1016/j.jpsychires.2023.06.014] [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: 11/21/2022] [Revised: 06/01/2023] [Accepted: 06/15/2023] [Indexed: 06/30/2023]
Abstract
The environmental disturbances in a critical neurodevelopmental period exert organizational effects on brain intrinsic plasticity including excitatory and inhibitory (E/I) neurotransmission those can cause the onset of psychiatric illness. We previously reported that treatment of neural precursor cells with N-methyl-D-aspartate (NMDA) receptor antagonist MK-801 induced reduction of GABAergic interneuron differentiation, and these changes recovered by atypical antipsychotic blonanserin treatment in vitro. However, it remains unclear how this treatment affects neural circuit changes in hippocampus and amygdala, which might contribute to the prevention of onset process of schizophrenia. To elucidate the pathogenic/preventive mechanisms underlying prenatal environmental adversity-induced schizophrenia in more detail, we administered poly (I:C) followed by antipsychotics and examined alterations in social/cognitive behaviors, GABA/glutamate-related gene expressions with cell density and E/I ratio, and brain-derived neurotrophic factor (Bdnf) transcript levels, particularly in limbic areas. Treatment with antipsychotic blonanserin ameliorated impaired social/cognitive behaviors and increased parvalbumin (PV)-positive (+) cell density and its mRNA levels as well as Bdnf with long 3'UTR mRNA levels, particularly in the dorsal hippocampus, in rats exposed to maternal immune activation (MIA). Low dose of blonanserin and haloperidol altered GABA and glutamate-related mRNA levels, the E/I ratio, and Bdnf long 3'UTR mRNA levels in the ventral hippocampus and amygdala, but did not attenuate behavioral impairments. These results strongly implicate changes in PV expression, PV(+) GABAergic interneuron density, and Bdnf long 3'UTR expression levels, particularly in the dorsal hippocampus, in the pathophysiology and treatment responses of MIA-induced schizophrenia and highlight the therapeutic potential of blonanserin for developmental stress-related schizophrenia.
Collapse
Affiliation(s)
- Kenta Deriha
- Department of Neuropsychiatry, Graduate School of Medicine, Sapporo Medical University, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Eri Hashimoto
- Department of Neuropsychiatry, Graduate School of Medicine, Sapporo Medical University, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Wataru Ukai
- Department of Neuropsychiatry, Graduate School of Medicine, Sapporo Medical University, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan; Department of Institutional Research, Center for Medical Education, Sapporo Medical University, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Francesca Marchisella
- Department of Pharmacological and Biomolecular Sciences University of Milan Via Balzaretti 9, 20133, Milan, Italy.
| | - Emi Nishimura
- Department of Neuropsychiatry, Graduate School of Medicine, Sapporo Medical University, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Hanako Hashiguchi
- Department of Neuropsychiatry, Graduate School of Medicine, Sapporo Medical University, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Masaya Tayama
- Department of Neuropsychiatry, Graduate School of Medicine, Sapporo Medical University, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Takao Ishii
- Department of Neuropsychiatry, Graduate School of Medicine, Sapporo Medical University, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan; Department of Occupational Therapy, Graduate School of Health Sciences, Sapporo Medical University, S-1, W-17, Chuo-ku, Sapporo, 0608556, Japan
| | - Marco A Riva
- Department of Pharmacological and Biomolecular Sciences University of Milan Via Balzaretti 9, 20133, Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| | - Chiaki Kawanishi
- Department of Neuropsychiatry, Graduate School of Medicine, Sapporo Medical University, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| |
Collapse
|
48
|
McCutcheon RA, Keefe RSE, McGuire PK. Cognitive impairment in schizophrenia: aetiology, pathophysiology, and treatment. Mol Psychiatry 2023; 28:1902-1918. [PMID: 36690793 PMCID: PMC10575791 DOI: 10.1038/s41380-023-01949-9] [Citation(s) in RCA: 233] [Impact Index Per Article: 116.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/25/2023]
Abstract
Cognitive deficits are a core feature of schizophrenia, account for much of the impaired functioning associated with the disorder and are not responsive to existing treatments. In this review, we first describe the clinical presentation and natural history of these deficits. We then consider aetiological factors, highlighting how a range of similar genetic and environmental factors are associated with both cognitive function and schizophrenia. We then review the pathophysiological mechanisms thought to underlie cognitive symptoms, including the role of dopamine, cholinergic signalling and the balance between GABAergic interneurons and glutamatergic pyramidal cells. Finally, we review the clinical management of cognitive impairments and candidate novel treatments.
Collapse
Affiliation(s)
- Robert A McCutcheon
- Department of Psychiatry, University of Oxford, Oxford, UK.
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, London, UK.
- Oxford health NHS Foundation Trust, Oxford health NHS Foundation Trust, Oxford, UK.
| | - Richard S E Keefe
- Departments of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Philip K McGuire
- Department of Psychiatry, University of Oxford, Oxford, UK
- Oxford health NHS Foundation Trust, Oxford health NHS Foundation Trust, Oxford, UK
- NIHR Oxford Health Biomedical Research Centre, Oxford, UK
| |
Collapse
|
49
|
Herzog LE, Wang L, Yu E, Choi S, Farsi Z, Song BJ, Pan JQ, Sheng M. Mouse mutants in schizophrenia risk genes GRIN2A and AKAP11 show EEG abnormalities in common with schizophrenia patients. Transl Psychiatry 2023; 13:92. [PMID: 36914641 PMCID: PMC10011509 DOI: 10.1038/s41398-023-02393-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/16/2023] Open
Abstract
Schizophrenia is a heterogeneous psychiatric disorder with a strong genetic basis, whose etiology and pathophysiology remain poorly understood. Exome sequencing studies have uncovered rare, loss-of-function variants that greatly increase risk of schizophrenia [1], including loss-of-function mutations in GRIN2A (aka GluN2A or NR2A, encoding the NMDA receptor subunit 2A) and AKAP11 (A-Kinase Anchoring Protein 11). AKAP11 and GRIN2A mutations are also associated with bipolar disorder [2], and epilepsy and developmental delay/intellectual disability [1, 3, 4], respectively. Accessible in both humans and rodents, electroencephalogram (EEG) recordings offer a window into brain activity and display abnormal features in schizophrenia patients. Does loss of Grin2a or Akap11 in mice also result in EEG abnormalities? We monitored EEG in heterozygous and homozygous knockout Grin2a and Akap11 mutant mice compared with their wild-type littermates, at 3- and 6-months of age, across the sleep/wake cycle and during auditory stimulation protocols. Grin2a and Akap11 mutants exhibited increased resting gamma power, attenuated auditory steady-state responses (ASSR) at gamma frequencies, and reduced responses to unexpected auditory stimuli during mismatch negativity (MMN) tests. Sleep spindle density was reduced in a gene dose-dependent manner in Akap11 mutants, whereas Grin2a mutants showed increased sleep spindle density. The EEG phenotypes of Grin2a and Akap11 mutant mice show a variety of abnormal features that overlap considerably with human schizophrenia patients, reflecting systems-level changes caused by Grin2a and Akap11 deficiency. These neurophysiologic findings further substantiate Grin2a and Akap11 mutants as genetic models of schizophrenia and identify potential biomarkers for stratification of schizophrenia patients.
Collapse
Affiliation(s)
- Linnea E Herzog
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Lei Wang
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eunah Yu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Soonwook Choi
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zohreh Farsi
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bryan J Song
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jen Q Pan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Morgan Sheng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
50
|
Gamma oscillations provide insights into cortical circuit development. Pflugers Arch 2023; 475:561-568. [PMID: 36864347 PMCID: PMC10105678 DOI: 10.1007/s00424-023-02801-3] [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: 01/24/2023] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023]
Abstract
Rhythmic coordination in gamma oscillations provides temporal structure to neuronal activity. Gamma oscillations are commonly observed in the mammalian cerebral cortex, are altered early on in several neuropsychiatric disorders, and provide insights into the development of underlying cortical networks. However, a lack of knowledge on the developmental trajectory of gamma oscillations prevented the combination of findings from the immature and the adult brain. This review is intended to provide an overview on the development of cortical gamma oscillations, the maturation of the underlying network, and the implications for cortical function and dysfunction. The majority of information is drawn from work in rodents with particular emphasis on the prefrontal cortex, the developmental trajectory of gamma oscillations, and potential implications for neuropsychiatric disorders. Current evidence supports the idea that fast oscillations during development are indeed an immature form of adult gamma oscillations and can help us understand the pathology of neuropsychiatric disorders.
Collapse
|