1
|
Govil P, Kantrowitz JT. Negative Symptoms in Schizophrenia: An Update on Research Assessment and the Current and Upcoming Treatment Landscape. CNS Drugs 2025:10.1007/s40263-024-01151-7. [PMID: 39799532 DOI: 10.1007/s40263-024-01151-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/01/2024] [Indexed: 01/15/2025]
Abstract
The negative symptoms of schizophrenia include diminished emotional expression, avolition, alogia, anhedonia, and asociality, and due to their low responsiveness to available treatments, are a primary driver of functional disability in schizophrenia. This narrative review has the aim of providing a comprehensive overview of the current research developments in the treatment of negative symptoms in schizophrenia, and begins by introducing the concepts of primary, secondary, prominent, predominant, and broadly defined negative symptoms. We then compare and contrast commonly used research assessment scales for negative symptoms and review the evidence for the specific utility of widely available off-label and investigational treatments that have been studied for negative symptoms. Mechanism of action/putative treatments included are antipsychotics (D2R antagonists), N-methyl-D-aspartate receptor (NMDAR) and other glutamatergic modulators, serotonin receptor (5-HTR) modulators, anti-inflammatory agents, antidepressants, pro-dopaminergic modulators (non-D2R antagonists), acetylcholine modulators, oxytocin, and phosphodiesterase (PDE) inhibitors. With the caveat that no compounds are definitively proven as gold-standard treatments for broadly defined negative symptoms, the evidence base supports several potentially beneficial off-label and investigational medications for treating negative symptoms in schizophrenia, such as monotherapy with cariprazine, olanzapine, clozapine, and amisulpride, or adjunctive use of memantine, setrons such as ondansetron, minocycline, and antidepressants. These medications are widely available worldwide, generally tolerable and could be considered for an off-label, time-limited trial for a predesignated period of time, after which a decision to switch or stay can be made based on clinical response. Among investigational medications, NMDAR agonists, muscarinic agonists, and LB-102 remain under study. Suggestions for future research include reducing placebo effects by designing studies with a smaller number of high-quality study sites, potentially increasing the use of more precise rating scales for negative symptoms, and focused studies in people with predominant negative symptoms.
Collapse
Affiliation(s)
- Preetika Govil
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA
| | - Joshua T Kantrowitz
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA.
- College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA.
- Nathan Kline Institute, Orangeburg, NY, 10962, USA.
| |
Collapse
|
2
|
Ansari U, Wen J, Syed B, Nadora D, Sedighi R, Nadora D, Chen V, Lui F. Analyzing the potential of neuronal pentraxin 2 as a biomarker in neurological disorders: A literature review. AIMS Neurosci 2024; 11:505-519. [PMID: 39801792 PMCID: PMC11712228 DOI: 10.3934/neuroscience.2024031] [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: 08/07/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
Neuronal pentraxin 2 (NP2) plays a significant role in synaptic plasticity, neuronal survival, and excitatory synapse regulation. Emerging research suggests that NP2 is implicated in the pathogenesis of various neurological disorders, including neurodegenerative diseases, neuropsychiatric disorders, and neuropathies. This literature review extensively analyzes NP2's role in these conditions, thereby highlighting its contributions to synaptic dysfunction, neuroinflammation, and neurotoxic protein aggregation. In Alzheimer's and Parkinson's diseases, NP2 is linked to amyloid-beta aggregation and dopaminergic neuron degeneration, respectively. Additionally, altered NP2 expression is observed in schizophrenia and bipolar disorder, thus suggesting its involvement in synaptic dysfunction and neurotransmitter imbalance. In neuropathic pain and epilepsy, NP2 modulates the synaptic plasticity and inflammatory responses, with altered levels correlating with disease severity. Furthermore, NP2's involvement in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) emphasizes its broad impact on neuronal health. Understanding NP2's multifaceted roles may reveal novel therapeutic targets and improve the clinical outcomes for these neurological disorders. Though the precise role of NP2 remains uncertain, its clinical potential and initial findings justify further investigations into neuronal pentraxins and other related neuroproteins.
Collapse
Affiliation(s)
- Ubaid Ansari
- California Northstate University College of Medicine, USA
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Sugiyama S, Inui K, Ohi K, Shioiri T. The influence of novelty detection on the 40-Hz auditory steady-state response in schizophrenia: A novel hypothesis from meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111096. [PMID: 39029650 DOI: 10.1016/j.pnpbp.2024.111096] [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: 04/07/2024] [Revised: 07/14/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
The 40-Hz auditory steady-state response (ASSR) is influenced not only by parameters such as attention, stimulus type, and analysis level but also by stimulus duration and inter-stimulus interval (ISI). In this meta-analysis, we examined these parameters in 33 studies that investigated 40-Hz ASSRs in patients with schizophrenia. The average Hedges' g random effect sizes were - 0.47 and - 0.43 for spectral power and phase-locking, respectively. We also found differences in ASSR measures based on stimulus duration and ISI. In particular, ISI was shown to significantly influence differences in the 40-Hz ASSR between healthy controls and patients with schizophrenia. We proposed a novel hypothesis focusing on the role of novelty detection, dependent on stimulus duration and ISI, as a critical factor in determining these differences. Specifically, longer stimulus durations and shorter ISIs under random presentation, or shorter stimulus durations and longer ISIs under repetitive presentation, decrease the 40-Hz ASSR in healthy controls. Patients with schizophrenia show minimal changes in response to stimulus duration and ISI, thus reducing the difference between controls and patients. This hypothesis can consistently explain most of the studies that have failed to show a reduction in 40-Hz ASSR in patients with schizophrenia. Increased novelty-related activity, reflected as an increase in auditory evoked potential components at stimulus onset, such as the N1, could suppress the 40-Hz ASSR, potentially reducing the peak measures of spectral power and phase-locking. To establish the 40-Hz ASSR as a truly valuable biomarker for schizophrenia, further systematic research using paradigms with various stimulus durations and ISIs is needed.
Collapse
Affiliation(s)
- Shunsuke Sugiyama
- Department of Psychiatry, Gifu University Graduate School of Medicine, Gifu, Japan.
| | - Koji Inui
- Department of Functioning and Disability, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai, Japan; Section of Brain Function Information, National Institute for Physiological Sciences, Okazaki, Japan
| | - Kazutaka Ohi
- Department of Psychiatry, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Toshiki Shioiri
- Department of Psychiatry, Gifu University Graduate School of Medicine, Gifu, Japan
| |
Collapse
|
4
|
Mouffok I, Lahogue C, Cailly T, Freret T, Bouet V, Boulouard M. A New Three-Hit Mouse Model of Neurodevelopmental Disorder with Cognitive Impairments and Persistent Sociability Deficits. Brain Sci 2024; 14:1281. [PMID: 39766480 PMCID: PMC11674404 DOI: 10.3390/brainsci14121281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Cognitive deficits and negative symptoms associated with schizophrenia are poorly managed by current antipsychotics. In order to develop effective treatments, refining animal models of neurodevelopmental disorders is essential. METHODS To address their multifactorial etiology, we developed a new three-hit mouse model based on the hypoglutamatergic hypothesis of the pathology combined with early stress, offering strong construct validity. Thus, a genetic susceptibility (serine racemase deletion) was associated with an early environmental stress (24 h maternal separation at 9 days of age) and a further pharmacological treatment with phencyclidine (PCP, a glutamate receptor antagonist treatment, 10 mg/kg/day, from 8 to 10 weeks of age). The face validity of this model was assessed in female mice 1 and 6 weeks after the end of PCP treatment by a set of behavioral experiments investigating positive- and negative-like symptoms and cognitive deficits. RESULTS Our results showed that the three-hit mice displayed persistent hyperlocomotion (positive-like symptoms) and social behavior impairment deficits (negative-like symptoms) but non-persistent spatial working memory deficits (cognitive symptoms). CONCLUSIONS Our work confirms the usefulness of a three-hit combination to model, particularly for negative-like symptoms associated with schizophrenia and other psychiatric disorders. The model therefore gathers powerful construct and face validities and supports an involvement of glutamate dysfunction in behavioral symptoms.
Collapse
Affiliation(s)
- Imane Mouffok
- Department of Health, Normandie Université, UNICAEN (Université de Caen Normandie), INSERM (Institut National de la Santé et de la Recherche Médicale), UMR (Unité Mixte de Recherche) 1075 COMETE, Campus 5, CYCERON, FHU (Fédération Hospitalo-Universitaire) A2M2P, CHU (Centre Hospitalo-Universitaire) Caen, 14000 Caen, France; (I.M.); (C.L.); (T.F.); (M.B.)
| | - Caroline Lahogue
- Department of Health, Normandie Université, UNICAEN (Université de Caen Normandie), INSERM (Institut National de la Santé et de la Recherche Médicale), UMR (Unité Mixte de Recherche) 1075 COMETE, Campus 5, CYCERON, FHU (Fédération Hospitalo-Universitaire) A2M2P, CHU (Centre Hospitalo-Universitaire) Caen, 14000 Caen, France; (I.M.); (C.L.); (T.F.); (M.B.)
| | - Thomas Cailly
- CERMN UR (Unité de Recherche) 4258, Campus 5, Université de Caen Normandie, 14000 Caen, France;
- CYCERON UAR (Unité d’Appui à la Recherche) 3408-US50, IMOGERE, Campus 1, Université de Caen Normandie, 14000 Caen, France
- Department of Nuclear Medicine, CHU Côte de Nacre, 14000 Caen, France
| | - Thomas Freret
- Department of Health, Normandie Université, UNICAEN (Université de Caen Normandie), INSERM (Institut National de la Santé et de la Recherche Médicale), UMR (Unité Mixte de Recherche) 1075 COMETE, Campus 5, CYCERON, FHU (Fédération Hospitalo-Universitaire) A2M2P, CHU (Centre Hospitalo-Universitaire) Caen, 14000 Caen, France; (I.M.); (C.L.); (T.F.); (M.B.)
| | - Valentine Bouet
- Department of Health, Normandie Université, UNICAEN (Université de Caen Normandie), INSERM (Institut National de la Santé et de la Recherche Médicale), UMR (Unité Mixte de Recherche) 1075 COMETE, Campus 5, CYCERON, FHU (Fédération Hospitalo-Universitaire) A2M2P, CHU (Centre Hospitalo-Universitaire) Caen, 14000 Caen, France; (I.M.); (C.L.); (T.F.); (M.B.)
| | - Michel Boulouard
- Department of Health, Normandie Université, UNICAEN (Université de Caen Normandie), INSERM (Institut National de la Santé et de la Recherche Médicale), UMR (Unité Mixte de Recherche) 1075 COMETE, Campus 5, CYCERON, FHU (Fédération Hospitalo-Universitaire) A2M2P, CHU (Centre Hospitalo-Universitaire) Caen, 14000 Caen, France; (I.M.); (C.L.); (T.F.); (M.B.)
| |
Collapse
|
5
|
Omori NE, Malys MK, Woo G, Mansor L. Exogenous ketone bodies and the ketogenic diet as a treatment option for neurodevelopmental disorders. Front Nutr 2024; 11:1485280. [PMID: 39749357 PMCID: PMC11693454 DOI: 10.3389/fnut.2024.1485280] [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: 08/30/2024] [Accepted: 11/13/2024] [Indexed: 01/04/2025] Open
Abstract
Background Despite being the most prevalent neurodevelopmental disorders, there are comparatively few treatment options available to patients presenting with autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD). The ketogenic diet has historically shown therapeutic utility in treating refractory epilepsy, an adjacent neuropsychiatric condition, in children, adolescents and adults. The following review explores preclinical and clinical literature focusing on the therapeutic potential of the ketogenic diet and exogenous ketone body supplementation in treating common neurodevelopmental disorders. Method A narrative review of extant literature was conducted across the domains of perinatal nutrition, ASD, and ADHD. Preclinical and clinical studies focusing on the effect of either the ketogenic diet or exogenous ketone supplementation as a treatment option were included for review. Results 14 preclinical and 10 clinical studies were included for discussion. Data supporting the use of a ketogenic intervention for neurodevelopmental disorders is mixed. High heterogeneity in study design was noted for preclinical models, ketogenic intervention, and outcomes measured. Conclusion Studies evaluating ketogenic interventions for neurodevelopmental disorders remain in their infancy in terms of both the depth and scope of available literature. The safety and tolerability of ketogenic diets and supplements means there would be value in exploring their effectiveness further in clinical studies.
Collapse
Affiliation(s)
- Naomi Elyse Omori
- Health Via Modern Nutrition Inc. (H.V.M.N.), San Francisco, CA, United States
| | - Mantas Kazimieras Malys
- Department of Psychological Medicine, King’s College London, Institute of Psychiatry, Psychology & Neuroscience, London, United Kingdom
| | - Geoffrey Woo
- Health Via Modern Nutrition Inc. (H.V.M.N.), San Francisco, CA, United States
| | - Latt Mansor
- Health Via Modern Nutrition Inc. (H.V.M.N.), San Francisco, CA, United States
| |
Collapse
|
6
|
Camporesi S, Xin L, Golay P, Eap CB, Cleusix M, Cuenod M, Fournier M, Hashimoto K, Jenni R, Ramain J, Restellini R, Solida A, Conus P, Do KQ, Khadimallah I. Neurocognition and NMDAR co-agonists pathways in individuals with treatment resistant first-episode psychosis: a 3-year follow-up longitudinal study. Mol Psychiatry 2024; 29:3669-3679. [PMID: 38849515 PMCID: PMC11541217 DOI: 10.1038/s41380-024-02631-4] [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: 01/09/2024] [Revised: 05/15/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024]
Abstract
This study aims to determine whether 1) individuals with treatment-resistant schizophrenia display early cognitive impairment compared to treatment-responders and healthy controls and 2) N-methyl-D-aspartate-receptor hypofunction is an underlying mechanism of cognitive deficits in treatment-resistance. In this case‒control 3-year-follow-up longitudinal study, n = 697 patients with first-episode psychosis, aged 18 to 35, were screened for Treatment Response and Resistance in Psychosis criteria through an algorithm that assigns patients to responder, limited-response or treatment-resistant category (respectively resistant to 0, 1 or 2 antipsychotics). Assessments at baseline: MATRICS Consensus Cognitive Battery; N-methyl-D-aspartate-receptor co-agonists biomarkers in brain by MRS (prefrontal glutamate levels) and plasma (D-serine and glutamate pathways key markers). Patients were compared to age- and sex-matched healthy controls (n = 114). Results: patient mean age 23, 27% female. Treatment-resistant (n = 51) showed lower scores than responders (n = 183) in processing speed, attention/vigilance, working memory, verbal learning and visual learning. Limited responders (n = 59) displayed an intermediary phenotype. Treatment-resistant and limited responders were merged in one group for the subsequent D-serine and glutamate pathway analyses. This group showed D-serine pathway dysregulation, with lower levels of the enzymes serine racemase and serine-hydroxymethyltransferase 1, and higher levels of the glutamate-cysteine transporter 3 than in responders. Better cognition was associated with higher D-serine and lower glutamate-cysteine transporter 3 levels only in responders; this association was disrupted in the treatment resistant group. Treatment resistant patients and limited responders displayed early cognitive and persistent functioning impairment. The dysregulation of NMDAR co-agonist pathways provides underlying molecular mechanisms for cognitive deficits in treatment-resistant first-episode psychosis. If replicated, our findings would open ways to mechanistic biomarkers guiding response-based patient stratification and targeting cognitive improvement in clinical trials.
Collapse
Affiliation(s)
- Sara Camporesi
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Department of psychiatry and Emergency Department, Geneva University Hospital, Geneva, Switzerland
| | - Lijing Xin
- Center for Biomedical Imaging, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Philippe Golay
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Chin Bin Eap
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, University of Lausanne, Lausanne, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Martine Cleusix
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Michel Cuenod
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Margot Fournier
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Raoul Jenni
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Julie Ramain
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Training and Research Institute in Mental Health (IFRSM), Neuchâtel Centre of Psychiatry, Neuchâtel, Switzerland
| | - Romeo Restellini
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Emergency medicine department, Geneva University Hospital, Geneva, Switzerland
| | - Alessandra Solida
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Psychiatry Department for Adults 2, Neuchâtel Centre of Psychiatry, Prefargier, Switzerland
| | - Philippe Conus
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Ines Khadimallah
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland.
- Translational Research Center, University Hospital of Psychiatry, University of Bern, Bern, Switzerland.
| |
Collapse
|
7
|
Preller KH, Scholpp J, Wunder A, Rosenbrock H. Neuroimaging Biomarkers for Drug Discovery and Development in Schizophrenia. Biol Psychiatry 2024; 96:666-673. [PMID: 38272287 DOI: 10.1016/j.biopsych.2024.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/19/2023] [Accepted: 01/14/2024] [Indexed: 01/27/2024]
Abstract
Schizophrenia is a chronic mental illness that affects up to 1% of the population. While efficacious therapies are available for positive symptoms, effective treatment of cognitive and negative symptoms remains an unmet need after decades of research. New developments in the field of neuroimaging are accelerating our knowledge gain regarding the underlying pathophysiology of symptoms in schizophrenia and psychosis spectrum disorders, inspiring new targets for drug development. However, no validated and qualified biomarkers are currently available to support the development of new therapeutics. This review summarizes the current use of neuroimaging technology in clinical drug development for psychotic disorders. As exemplified by drug development programs that target NMDA receptor hypofunction, neuroimaging results play a critical role in target discovery and establishing target engagement and dose selection. Furthermore, pharmacological neuroimaging may provide response biomarkers that allow for early decision making in proof-of-concept studies that leverage pharmacological challenge models in healthy volunteers. That said, while response and predictive biomarkers are starting to be evaluated in patient populations, they continue to play a limited role. Novel approaches to neuroimaging data acquisition and analysis may aid the establishment of biomarkers that are predictive at the individual level in the future. Nevertheless, various gaps in knowledge need to be addressed and biomarkers need to be validated to establish them as "fit for purpose" in drug development.
Collapse
Affiliation(s)
- Katrin H Preller
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany; Boehringer Ingelheim (Schweiz) GmbH, Basel, Switzerland.
| | - Joachim Scholpp
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Andreas Wunder
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Holger Rosenbrock
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| |
Collapse
|
8
|
Sharkey RJ, Bacon C, Peterson Z, Rootes-Murdy K, Salvador R, Pomarol-Clotet E, Karuk A, Homan P, Ji E, Omlor W, Homan S, Georgiadis F, Kaiser S, Kirschner M, Ehrlich S, Dannlowski U, Grotegerd D, Goltermann J, Meinert S, Kircher T, Stein F, Brosch K, Krug A, Nenadic I, Sim K, Spalletta G, Banaj N, Sponheim SR, Demro C, Ramsay IS, King M, Quidé Y, Green MJ, Nguyen D, Preda A, Calhoun V, Turner J, van Erp T, Nickl-Jockschat T. Differences in the neural correlates of schizophrenia with positive and negative formal thought disorder in patients with schizophrenia in the ENIGMA dataset. Mol Psychiatry 2024; 29:3086-3096. [PMID: 38671214 PMCID: PMC11449795 DOI: 10.1038/s41380-024-02563-z] [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: 07/17/2023] [Revised: 04/04/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024]
Abstract
Formal thought disorder (FTD) is a clinical key factor in schizophrenia, but the neurobiological underpinnings remain unclear. In particular, the relationship between FTD symptom dimensions and patterns of regional brain volume loss in schizophrenia remains to be established in large cohorts. Even less is known about the cellular basis of FTD. Our study addresses these major obstacles by enrolling a large multi-site cohort acquired by the ENIGMA Schizophrenia Working Group (752 schizophrenia patients and 1256 controls), to unravel the neuroanatomy of FTD in schizophrenia and using virtual histology tools on implicated brain regions to investigate the cellular basis. Based on the findings of previous clinical and neuroimaging studies, we decided to separately explore positive, negative and total formal thought disorder. We used virtual histology tools to relate brain structural changes associated with FTD to cellular distributions in cortical regions. We identified distinct neural networks positive and negative FTD. Both networks encompassed fronto-occipito-amygdalar brain regions, but positive and negative FTD demonstrated a dissociation: negative FTD showed a relative sparing of orbitofrontal cortical thickness, while positive FTD also affected lateral temporal cortices. Virtual histology identified distinct transcriptomic fingerprints associated for both symptom dimensions. Negative FTD was linked to neuronal and astrocyte fingerprints, while positive FTD also showed associations with microglial cell types. These results provide an important step towards linking FTD to brain structural changes and their cellular underpinnings, providing an avenue for a better mechanistic understanding of this syndrome.
Collapse
Affiliation(s)
- Rachel J Sharkey
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Chelsea Bacon
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Zeru Peterson
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | | | - Raymond Salvador
- FIDMAG Germanes Hospitalàries Research Foundation, CIBERSAM ISCIII, Barcelona, Spain
| | - Edith Pomarol-Clotet
- FIDMAG Germanes Hospitalàries Research Foundation, CIBERSAM ISCIII, Barcelona, Spain
| | - Andriana Karuk
- FIDMAG Germanes Hospitalàries Research Foundation, CIBERSAM ISCIII, Barcelona, Spain
| | - Philipp Homan
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich (PUK), Zurich, 8008, Switzerland
| | - Ellen Ji
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich (PUK), Zurich, 8008, Switzerland
| | - Wolfgang Omlor
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich (PUK), Zurich, 8008, Switzerland
| | - Stephanie Homan
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich (PUK), Zurich, 8008, Switzerland
| | - Foivos Georgiadis
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich (PUK), Zurich, 8008, Switzerland
| | - Stefan Kaiser
- Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | - Matthias Kirschner
- Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | - Stefan Ehrlich
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Janik Goltermann
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Katharina Brosch
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Axel Krug
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Igor Nenadic
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Kang Sim
- West Region, Institute of Mental Health, Singapore, Singapore
| | | | - Nerisa Banaj
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Scott R Sponheim
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Caroline Demro
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Ian S Ramsay
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | | | - Yann Quidé
- School of Psychiatry, University of New South Wales (UNSW) Sydney, Sydney, NSW, Australia
| | - Melissa Jane Green
- School of Psychiatry, University of New South Wales (UNSW) Sydney, Sydney, NSW, Australia
| | - Dana Nguyen
- Department of Pediatric Neurology, University of California Irvine, Irvine, CA, USA
| | - Adrian Preda
- Department of Pediatric Neurology, University of California Irvine, Irvine, CA, USA
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, USA
| | - Vince Calhoun
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GE, USA
| | - Jessica Turner
- Department of Psychiatry and Behavioral Medicine, Ohio State University, Columbus, OH, USA
| | - Theo van Erp
- Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA, USA
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, USA
| | - Thomas Nickl-Jockschat
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA.
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Department of Psychiatry and Psychotherapy, Otto-von-Guericke University, Magdeburg, Germany.
- German Center for Mental Health (DZPG), partner site Halle-Jena-Magdeburg, Magdeburg, Germany.
- Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Halle-Jena-Magdeburg, Magdeburg, Germany.
| |
Collapse
|
9
|
Aydıner Yılmaz M, Kaya H, Aşut G, Çiftçi H, Bayram Ş, Fırat Oğuz E, Turhan T, Göka E. Plasma G72 protein in schizophrenia: A comparative analysis of drug-naive schizophrenia patients, patients in acute exacerbation and healthy controls. J Psychiatr Res 2024; 178:8-14. [PMID: 39096761 DOI: 10.1016/j.jpsychires.2024.07.053] [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: 07/01/2024] [Revised: 07/20/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
OBJECTIVES Schizophrenia is a complex psychiatric disorder with an unclear etiopathogenesis. This study investigates the plasma G72 protein levels in drug-naive schizophrenia patients (DNS), those in acute psychotic episodes (AES), and healthy controls (HC). It also examines the correlation between the plasma G72 protein levels and Positive and Negative Syndrome Scale (PANSS) scores. METHODS The study included 138 schizophrenia patients (84 DNS, 54 AES) and 83 HCs. Plasma G72 protein levels were measured by ELISA. Statistical analyses, including log-transformation and correlation analysis, were conducted. RESULTS Schizophrenia patients had significantly lower plasma G72 levels than HCs (4.39 ± 5.38 vs. 8.06 ± 10.27 ng/mL, p < 0.001), while DNS and AES groups did not differ significantly. Log-transformed data confirmed these differences. Negative correlation was found between plasma G72 levels and age (r = -0.258, p = 0.02), PANSS-G (r = -0.249, p = 0.004), and total PANSS scores (r = -0.226, p = 0.008). ROC analysis showed poor discrimination between schizophrenia patients and controls (AUC: 0.587, p = 0.031). CONCLUSIONS This study's novel findings reveal that plasma G72 protein levels are significantly lower in schizophrenia patients and inversely correlated with age and symptom severity. However, the poor diagnostic accuracy observed in the ROC analysis suggests that G72 may not be a reliable biomarker for schizophrenia at this stage. These results underscore the need for further research to explore the potential clinical implications of these findings.
Collapse
Affiliation(s)
- Meltem Aydıner Yılmaz
- Department of Psychiatry, Samsun Mental Health and Diseases Hospital, Samsun, Turkey
| | - Hasan Kaya
- Department of Psychiatry, University of Health Sciences Ankara City Hospital, Ankara, Turkey
| | - Gonca Aşut
- Department of Psychiatry, Baskent University Faculty of Medicine, Ankara, Turkey.
| | - Hatice Çiftçi
- Department of Psychiatry, University of Health Sciences Ankara City Hospital, Ankara, Turkey
| | - Şenol Bayram
- Department of Psychiatry, Kocaeli City Hospital, Kocaeli, Turkey
| | - Esra Fırat Oğuz
- Department of Clinical Biochemistery, University of Health Sciences Ankara City Hospital, Ankara, Turkey
| | - Turan Turhan
- Department of Clinical Biochemistery, University of Health Sciences Ankara City Hospital, Ankara, Turkey
| | - Erol Göka
- Department of Psychiatry, University of Health Sciences Ankara City Hospital, Ankara, Turkey
| |
Collapse
|
10
|
Bianciardi B, Mastek H, Franka M, Uhlhaas PJ. Effects of N-Methyl-d-Aspartate Receptor Antagonists on Gamma-Band Activity During Auditory Stimulation Compared With Electro/Magneto-encephalographic Data in Schizophrenia and Early-Stage Psychosis: A Systematic Review and Perspective. Schizophr Bull 2024; 50:1104-1116. [PMID: 38934800 PMCID: PMC11349021 DOI: 10.1093/schbul/sbae090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
BACKGROUND AND HYPOTHESIS N-Methyl-d-aspartate receptor (NMDA-R) hypofunctioning has been hypothesized to be involved in circuit dysfunctions in schizophrenia (ScZ). Yet, it remains to be determined whether the physiological changes observed following NMDA-R antagonist administration are consistent with auditory gamma-band activity in ScZ which is dependent on NMDA-R activity. STUDY DESIGN This systematic review investigated the effects of NMDA-R antagonists on auditory gamma-band activity in preclinical (n = 15) and human (n = 3) studies and compared these data to electro/magneto-encephalographic measurements in ScZ patients (n = 37) and 9 studies in early-stage psychosis. The following gamma-band parameters were examined: (1) evoked spectral power, (2) intertrial phase coherence (ITPC), (3) induced spectral power, and (4) baseline power. STUDY RESULTS Animal and human pharmacological data reported a reduction, especially for evoked gamma-band power and ITPC, as well as an increase and biphasic effects of gamma-band activity following NMDA-R antagonist administration. In addition, NMDA-R antagonists increased baseline gamma-band activity in preclinical studies. Reductions in ITPC and evoked gamma-band power were broadly compatible with findings observed in ScZ and early-stage psychosis patients where the majority of studies observed decreased gamma-band spectral power and ITPC. In regard to baseline gamma-band power, there were inconsistent findings. Finally, a publication bias was observed in studies investigating auditory gamma-band activity in ScZ patients. CONCLUSIONS Our systematic review indicates that NMDA-R antagonists may partially recreate reductions in gamma-band spectral power and ITPC during auditory stimulation in ScZ. These findings are discussed in the context of current theories involving alteration in E/I balance and the role of NMDA hypofunction in the pathophysiology of ScZ.
Collapse
Affiliation(s)
- Bianca Bianciardi
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, UK
| | - Helena Mastek
- Department of Child and Adolescent Psychiatry, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Michelle Franka
- Department of Child and Adolescent Psychiatry, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Peter J Uhlhaas
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, UK
- Department of Child and Adolescent Psychiatry, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
11
|
Brickwedde M, Anders P, Kühn AA, Lofredi R, Holtkamp M, Kaindl AM, Grent-'t-Jong T, Krüger P, Sander T, Uhlhaas PJ. Applications of OPM-MEG for translational neuroscience: a perspective. Transl Psychiatry 2024; 14:341. [PMID: 39181883 PMCID: PMC11344782 DOI: 10.1038/s41398-024-03047-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 06/25/2024] [Accepted: 08/01/2024] [Indexed: 08/27/2024] Open
Abstract
Magnetoencephalography (MEG) allows the non-invasive measurement of brain activity at millisecond precision combined with localization of the underlying generators. So far, MEG-systems consisted of superconducting quantum interference devices (SQUIDS), which suffer from several limitations. Recent technological advances, however, have enabled the development of novel MEG-systems based on optically pumped magnetometers (OPMs), offering several advantages over conventional SQUID-MEG systems. Considering potential improvements in the measurement of neuronal signals as well as reduced operating costs, the application of OPM-MEG systems for clinical neuroscience and diagnostic settings is highly promising. Here we provide an overview of the current state-of-the art of OPM-MEG and its unique potential for translational neuroscience. First, we discuss the technological features of OPMs and benchmark OPM-MEG against SQUID-MEG and electroencephalography (EEG), followed by a summary of pioneering studies of OPMs in healthy populations. Key applications of OPM-MEG for the investigation of psychiatric and neurological conditions are then reviewed. Specifically, we suggest novel applications of OPM-MEG for the identification of biomarkers and circuit deficits in schizophrenia, dementias, movement disorders, epilepsy, and neurodevelopmental syndromes (autism spectrum disorder and attention deficit hyperactivity disorder). Finally, we give an outlook of OPM-MEG for translational neuroscience with a focus on remaining methodological and technical challenges.
Collapse
Affiliation(s)
- Marion Brickwedde
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Department of Child and Adolescent Psychiatry, 13353, Berlin, Germany.
- Physikalisch-Technische Bundesanstalt, Berlin, Germany.
| | - Paul Anders
- Physikalisch-Technische Bundesanstalt, Berlin, Germany
| | - Andrea A Kühn
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Sektion für Bewegungsstörungen und Neuromodulation, Klinik für Neurologie und Experimentelle Neurologie, 10117, Berlin, Germany
- Bernstein Center for Computational Neuroscience, Humboldt-Universität, Berlin, Germany
- NeuroCure, Exzellenzcluster, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZNE, German center for neurodegenerative diseases, Berlin, Germany
- Berlin School of Mind and Brain, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Roxanne Lofredi
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Sektion für Bewegungsstörungen und Neuromodulation, Klinik für Neurologie und Experimentelle Neurologie, 10117, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Martin Holtkamp
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Department of Neurology, Epilepsy-Center Berlin-Brandenburg, 10117, Berlin, Germany
| | - Angela M Kaindl
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Department of Pediatric Neurology, 13353, Berlin, Germany
- Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Center for Chronically Sick Children, 13353, Berlin, Germany
- Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Institute of Cell Biology and Neurobiology, 10117, Berlin, Germany
| | - Tineke Grent-'t-Jong
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Department of Child and Adolescent Psychiatry, 13353, Berlin, Germany
- Institute for Neuroscience and Psychology, Glasgow University, Scotland, United Kingdom
| | - Peter Krüger
- Physikalisch-Technische Bundesanstalt, Berlin, Germany
| | | | - Peter J Uhlhaas
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Department of Child and Adolescent Psychiatry, 13353, Berlin, Germany
- Institute for Neuroscience and Psychology, Glasgow University, Scotland, United Kingdom
| |
Collapse
|
12
|
Gao B, Li C, Qu Y, Cai M, Zhou Q, Zhang Y, Lu H, Tang Y, Li H, Shen H. Progress and trends of research on mineral elements for depression. Heliyon 2024; 10:e35469. [PMID: 39170573 PMCID: PMC11336727 DOI: 10.1016/j.heliyon.2024.e35469] [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: 12/14/2023] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 08/23/2024] Open
Abstract
Objective To explore the research progress and trends on mineral elements and depression. Methods After querying the MeSH database and referring to the search rules, the search terms were selected and optimized to obtain the target literature collection. We analyzed the general characteristics of the literature, conducted network clustering and co-occurrence analysis, and carried out a narrative review of crucial literature. Results Bipolar disorder was a dominant topic in the retrieved literature, which saw a significant increase in 2010 and 2019-2020. Most studies focused on mineral elements, including lithium, calcium, magnesium, zinc, and copper. The majority of journals and disciplines were in the fields of psychiatry, neuropsychology, neuropharmacology, nutrition, medical informatics, chemistry, and public health. The United States had the highest proportion in terms of paper sources, most-cited articles, high-frequency citations, frontier citations, and high centrality citation. Regarding the influence of academic institutions, the top five were King's College London, the Chinese Academy of Sciences, University of Barcelona, INSERM, and Heidelberg University. Frontier keywords included bipolar disorder, drinking water, (neuro)inflammation, gut microbiota, and systematic analysis. Research on lithium response, magnesium supplementation, and treatment-resistant unipolar depression increased significantly after 2013. Conclusion Global adverse events may have indirectly driven the progress in related research. Although the literature from the United States represents an absolute majority, its influence on academic institutions is relatively weaker. Multiple pieces of evidence support the efficacy of lithium in treating bipolar disorder (BD). A series of key discoveries have led to a paradigm shift in research, leading to increasingly detailed studies on the role of magnesium, calcium, zinc, and copper in the treatment of depression. Most studies on mineral elements remain diverse and inconclusive. The potential toxicity and side effects of some elements warrant careful attention.
Collapse
Affiliation(s)
- Biao Gao
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai, 200433, China
- Teaching and Research Support Center, Naval Medical University, Shanghai, 200433, China
| | - Chenqi Li
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai, 200433, China
- Department of Nutrition, The Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, China
| | - Yicui Qu
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai, 200433, China
| | - Mengyu Cai
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai, 200433, China
| | - Qicheng Zhou
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai, 200433, China
| | - Yinyin Zhang
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai, 200433, China
| | - Hongtao Lu
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai, 200433, China
| | - Yuxiao Tang
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai, 200433, China
| | - Hongxia Li
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai, 200433, China
| | - Hui Shen
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
13
|
Wynn JK, Green MF. An EEG-Based Neuroplastic Approach to Predictive Coding in People With Schizophrenia or Traumatic Brain Injury. Clin EEG Neurosci 2024; 55:445-454. [PMID: 38711326 DOI: 10.1177/15500594241252897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Despite different etiologies, people with schizophrenia (SCZ) or with traumatic brain injury (TBI) both show aberrant neuroplasticity. One neuroplastic mechanism that may be affected is prediction error coding. We used a roving mismatch negativity (rMMN) paradigm which uses different lengths of standard tone trains and is optimized to assess predictive coding. Twenty-five SCZ, 22 TBI (mild to moderate), and 25 healthy controls were assessed. We used a frequency-deviant rMMN in which the number of standards preceding the deviant was either 2, 6, or 36. We evaluated repetition positivity to the standard tone immediately preceding a deviant tone (repetition positivity [RP], to assess formation of the memory trace), deviant negativity to the deviant stimulus (deviant negativity [DN], which reflects signaling of a prediction error), and the difference wave between the 2 (the MMN). We found that SCZ showed reduced DN and MMN compared with healthy controls and with people with mild to moderate TBI. We did not detect impairments in any index (RP, DN, or MMN) in people with TBI compared to controls. Our findings suggest that prediction error coding assessed with rMMN is aberrant in SCZ but intact in TBI, though there is a suggestion that severity of head injury results in poorer prediction error coding.
Collapse
Affiliation(s)
- Jonathan K Wynn
- Center on Enhancement of Community Integration for Homeless Veterans, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Michael F Green
- Center on Enhancement of Community Integration for Homeless Veterans, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| |
Collapse
|
14
|
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
|
15
|
Qi X, Yu X, Wei L, Jiang H, Dong J, Li H, Wei Y, Zhao L, Deng W, Guo W, Hu X, Li T. Novel α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor (AMPAR) potentiator LT-102: A promising therapeutic agent for treating cognitive impairment associated with schizophrenia. CNS Neurosci Ther 2024; 30:e14713. [PMID: 38615362 PMCID: PMC11016348 DOI: 10.1111/cns.14713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 03/07/2024] [Accepted: 03/23/2024] [Indexed: 04/16/2024] Open
Abstract
AIMS We aimed to evaluate the potential of a novel selective α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor (AMPAR) potentiator, LT-102, in treating cognitive impairments associated with schizophrenia (CIAS) and elucidating its mechanism of action. METHODS The activity of LT-102 was examined by Ca2+ influx assays and patch-clamp in rat primary hippocampal neurons. The structure of the complex was determined by X-ray crystallography. The selectivity of LT-102 was evaluated by hERG tail current recording and kinase-inhibition assays. The electrophysiological characterization of LT-102 was characterized by patch-clamp recording in mouse hippocampal slices. The expression and phosphorylation levels of proteins were examined by Western blotting. Cognitive function was assessed using the Morris water maze and novel object recognition tests. RESULTS LT-102 is a novel and selective AMPAR potentiator with little agonistic effect, which binds to the allosteric site formed by the intradimer interface of AMPAR's GluA2 subunit. Treatment with LT-102 facilitated long-term potentiation in mouse hippocampal slices and reversed cognitive deficits in a phencyclidine-induced mouse model. Additionally, LT-102 treatment increased the protein level of brain-derived neurotrophic factor and the phosphorylation of GluA1 in primary neurons and hippocampal tissues. CONCLUSION We conclude that LT-102 ameliorates cognitive impairments in a phencyclidine-induced model of schizophrenia by enhancing synaptic function, which could make it a potential therapeutic candidate for CIAS.
Collapse
Affiliation(s)
- Xueyu Qi
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain‐Machine Integration, State Key Laboratory of Brain‐Machine IntelligenceZhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang UniversityHangzhouChina
| | - Xueli Yu
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain‐Machine Integration, State Key Laboratory of Brain‐Machine IntelligenceZhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang UniversityHangzhouChina
| | - Long Wei
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
| | - Han Jiang
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
| | - Jiangwen Dong
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
| | - Hongxing Li
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
| | - Yingying Wei
- The Psychiatric Laboratory, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Liansheng Zhao
- The Psychiatric Laboratory, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Wei Deng
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain‐Machine Integration, State Key Laboratory of Brain‐Machine IntelligenceZhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang UniversityHangzhouChina
| | - Wanjun Guo
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain‐Machine Integration, State Key Laboratory of Brain‐Machine IntelligenceZhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang UniversityHangzhouChina
| | - Xun Hu
- The Clinical Research Center and Department of Pathology, The Second Affiliated HospitalZhejiang University School of MedicineZhejiangHangzhouChina
| | - Tao Li
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain‐Machine Integration, State Key Laboratory of Brain‐Machine IntelligenceZhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang UniversityHangzhouChina
| |
Collapse
|
16
|
Lee J, Huh S, Park K, Kang N, Yu HS, Park HG, Kim YS, Kang UG, Won S, Kim SH. Behavioral and transcriptional effects of repeated electroconvulsive seizures in the neonatal MK-801-treated rat model of schizophrenia. Psychopharmacology (Berl) 2024; 241:817-832. [PMID: 38081977 DOI: 10.1007/s00213-023-06511-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 11/23/2023] [Indexed: 03/13/2024]
Abstract
RATIONALE Electroconvulsive therapy (ECT) is an effective treatment modality for schizophrenia. However, its antipsychotic-like mechanism remains unclear. OBJECTIVES To gain insight into the antipsychotic-like actions of ECT, this study investigated how repeated treatments of electroconvulsive seizure (ECS), an animal model for ECT, affect the behavioral and transcriptomic profile of a neurodevelopmental animal model of schizophrenia. METHODS Two injections of MK-801 or saline were administered to rats on postnatal day 7 (PN7), and either repeated ECS treatments (E10X) or sham shock was conducted daily from PN50 to PN59. Ultimately, the rats were divided into vehicle/sham (V/S), MK-801/sham (M/S), vehicle/ECS (V/E), and MK-801/ECS (M/E) groups. On PN59, prepulse inhibition and locomotor activity were tested. Prefrontal cortex transcriptomes were analyzed with mRNA sequencing and network and pathway analyses, and quantitative real-time polymerase chain reaction (qPCR) analyses were subsequently conducted. RESULTS Prepulse inhibition deficit was induced by MK-801 and normalized by E10X. In M/S vs. M/E model, Egr1, Mmp9, and S100a6 were identified as center genes, and interleukin-17 (IL-17), nuclear factor kappa B (NF-κB), and tumor necrosis factor (TNF) signaling pathways were identified as the three most relevant pathways. In the V/E vs. V/S model, mitophagy, NF-κB, and receptor for advanced glycation end products (RAGE) pathways were identified. qPCR analyses demonstrated that Igfbp6, Btf3, Cox6a2, and H2az1 were downregulated in M/S and upregulated in M/E. CONCLUSIONS E10X reverses the behavioral changes induced by MK-801 and produces transcriptional changes in inflammatory, insulin, and mitophagy pathways, which provide mechanistic insight into the antipsychotic-like mechanism of ECT.
Collapse
Affiliation(s)
- Jeonghoon Lee
- Department of Psychiatry, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seonghoo Huh
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyungtaek Park
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
| | - Nuree Kang
- Department of Psychiatry, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyun Sook Yu
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hong Geun Park
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yong Sik Kim
- Department of Psychiatry, Nowon Eulji Medical Center, Eulji University, Seoul, Republic of Korea
| | - Ung Gu Kang
- Department of Psychiatry, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Human Behavioral Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sungho Won
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
- Interdisciplinary Program of Bioinformatics, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
- RexSoft Inc., Seoul, Republic of Korea
| | - Se Hyun Kim
- Department of Psychiatry, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
17
|
Flynn LT, Bouras NN, Migovich VM, Clarin JD, Gao WJ. The "psychiatric" neuron: the psychic neuron of the cerebral cortex, revisited. Front Hum Neurosci 2024; 18:1356674. [PMID: 38562227 PMCID: PMC10982399 DOI: 10.3389/fnhum.2024.1356674] [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: 12/16/2023] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Nearly 25 years ago, Dr. Patricia Goldman-Rakic published her review paper, "The 'Psychic' Neuron of the Cerebral Cortex," outlining the circuit-level dynamics, neurotransmitter systems, and behavioral correlates of pyramidal neurons in the cerebral cortex, particularly as they relate to working memory. In the decades since the release of this paper, the existing literature and our understanding of the pyramidal neuron have increased tremendously, and research is still underway to better characterize the role of the pyramidal neuron in both healthy and psychiatric disease states. In this review, we revisit Dr. Goldman-Rakic's characterization of the pyramidal neuron, focusing on the pyramidal neurons of the prefrontal cortex (PFC) and their role in working memory. Specifically, we examine the role of PFC pyramidal neurons in the intersection of working memory and social function and describe how deficits in working memory may actually underlie the pathophysiology of social dysfunction in psychiatric disease states. We briefly describe the cortico-cortical and corticothalamic connections between the PFC and non-PFC brain regions, as well the microcircuit dynamics of the pyramidal neuron and interneurons, and the role of both these macro- and microcircuits in the maintenance of the excitatory/inhibitory balance of the cerebral cortex for working memory function. Finally, we discuss the consequences to working memory when pyramidal neurons and their circuits are dysfunctional, emphasizing the resulting social deficits in psychiatric disease states with known working memory dysfunction.
Collapse
Affiliation(s)
- L. Taylor Flynn
- Department of Neurobiology, Drexel University College of Medicine, Philadelphia, PA, United States
- Drexel University College of Medicine, Philadelphia, PA, United States
| | - Nadia N. Bouras
- Department of Neurobiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Volodar M. Migovich
- Department of Neurobiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Jacob D. Clarin
- Department of Neurobiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Wen-Jun Gao
- Department of Neurobiology, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
18
|
Laricchiuta D, Papi M, Decandia D, Panuccio A, Cutuli D, Peciccia M, Mazzeschi C, Petrosini L. The role of glial cells in mental illness: a systematic review on astroglia and microglia as potential players in schizophrenia and its cognitive and emotional aspects. Front Cell Neurosci 2024; 18:1358450. [PMID: 38419655 PMCID: PMC10899480 DOI: 10.3389/fncel.2024.1358450] [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: 12/19/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Schizophrenia is a complex and severe mental disorder that affects approximately 1% of the global population. It is characterized by a wide range of symptoms, including delusions, hallucinations, disorganized speech and behavior, and cognitive impairment. Recent research has suggested that the immune system dysregulation may play a significant role in the pathogenesis of schizophrenia, and glial cells, such as astroglia and microglia known to be involved in neuroinflammation and immune regulation, have emerged as potential players in this process. The aim of this systematic review is to summarize the glial hallmarks of schizophrenia, choosing as cellular candidate the astroglia and microglia, and focusing also on disease-associated psychological (cognitive and emotional) changes. We conducted a systematic review following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. We searched PubMed, Scopus, and Web of Science for articles that investigated the differences in astroglia and microglia in patients with schizophrenia, published in the last 5 years. The present systematic review indicates that changes in the density, morphology, and functioning of astroglia and microglia may be involved in the development of schizophrenia. The glial alterations may contribute to the pathogenesis of schizophrenia by dysregulating neurotransmission and immune responses, worsening cognitive capabilities. The complex interplay of astroglial and microglial activation, genetic/epigenetic variations, and cognitive assessments underscores the intricate relationship between biological mechanisms, symptomatology, and cognitive functioning in schizophrenia.
Collapse
Affiliation(s)
- Daniela Laricchiuta
- Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Martina Papi
- Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Davide Decandia
- Laboratory of Experimental and Behavioral Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Anna Panuccio
- Laboratory of Experimental and Behavioral Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Debora Cutuli
- Laboratory of Experimental and Behavioral Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Maurizio Peciccia
- Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Claudia Mazzeschi
- Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Laura Petrosini
- Laboratory of Experimental and Behavioral Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
19
|
O'Connell MN, Barczak A. Auditory Biomarkers of Neuropsychiatric Disorders in Nonhuman Primates. ADVANCES IN NEUROBIOLOGY 2024; 40:219-234. [PMID: 39562447 DOI: 10.1007/978-3-031-69491-2_9] [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
Animal models of neuropsychiatric disorders with appropriate biomarkers can greatly inform the neurobiological basis of disorder-related deficits of cognitive and/or sensory processes. Given the genetic, physiologic, and behavioral similarities between humans and nonhuman primates (NHPs), NHP studies are monumentally important for preclinical translational research. Capitalizing on the NHP's similarities with human systems provides one of the best opportunities to gain detailed insight into the mechanisms underlying disorder-related symptoms and to accumulate a foundation of information for the development of therapeutic interventions. Here, we discuss how results from NHP studies have provided insight into the generation and modulation of select auditory biomarkers of schizophrenia including auditory steady-state responses and mismatch negativity. Since neuro-oscillatory activity has been shown to be relatively preserved across species, we highlight how incorporating the analysis of local and network-level oscillations from multiple nodes across different pathways involved in auditory processing has been used to further the precision of translational comparisons across species.
Collapse
Affiliation(s)
- Monica N O'Connell
- Translational Neuroscience Division, Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute, Orangeburg, NY, USA.
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
| | - Annamaria Barczak
- Translational Neuroscience Division, Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute, Orangeburg, NY, USA
| |
Collapse
|
20
|
Javitt DC. Mismatch Negativity (MMN) as a Pharmacodynamic/Response Biomarker for NMDA Receptor and Excitatory/Inhibitory Imbalance-Targeted Treatments in Schizophrenia. ADVANCES IN NEUROBIOLOGY 2024; 40:411-451. [PMID: 39562453 DOI: 10.1007/978-3-031-69491-2_15] [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
Schizophrenia is a major mental disorder that affects approximately 0.5% of the population worldwide. Persistent negative symptoms and cognitive impairments associated with schizophrenia (CIAS) are key features of the disorder and primary predictors of long-term disability. At the neurochemical level, both CIAS and negative symptoms are potentially attributable to dysfunction or dysregulation of N-methyl-D-aspartate receptor (NMDAR)-mediated neurotransmission within cortical and subcortical brain regions. At present, there are no approved treatments for either CIAS or persistent negative symptoms. Development of novel treatments, moreover, is limited by the lack of biomarkers that can be used translationally across preclinical and early-stage clinical investigation. The present chapter describes the use of mismatch negativity (MMN) as a pharmacodynamic/response (PD/R) biomarker for early-stage clinical investigation of NMDAR targeted therapies for schizophrenia. MMN indexes dysfunction of early auditory processing (EAP) in schizophrenia. In humans, deficits in MMN generation contribute hierarchically to impaired cognition and functional outcome. Across humans, rodents, and primates, MMN has been linked to impaired NMDAR function and resultant disturbances in excitatory/inhibitory (E/I) balance involving interactions between glutamatergic (excitatory) pyramidal and GABAeric (inhibitory) local circuit neurons. In early-stage clinical trials, MMN has shown sensitivity to the acute effects of novel pharmacological treatments. These findings support use of MMN as a pharmacodynamic/response biomarker to support preclinical drug discovery and early-stage proof-of-mechanisms studies in schizophrenia and other related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Daniel C Javitt
- Division of Experimental Therapeutics, College of Physicians and Surgeons, Columbia University, New York, NY, USA.
- Schizophrenia Research Division, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA.
| |
Collapse
|
21
|
Bortolasci CC, Jaehne EJ, Hernández D, Spolding B, Connor T, Panizzutti B, Dean OM, Crowley TM, Yung AR, Gray L, Kim JH, van den Buuse M, Berk M, Walder K. Metergoline Shares Properties with Atypical Antipsychotic Drugs Identified by Gene Expression Signature Screen. Neurotox Res 2023; 41:502-513. [PMID: 37922109 PMCID: PMC10682262 DOI: 10.1007/s12640-023-00673-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 11/05/2023]
Abstract
Novel approaches are required to find new treatments for schizophrenia and other neuropsychiatric disorders. This study utilised a combination of in vitro transcriptomics and in silico analysis with the BROAD Institute's Connectivity Map to identify drugs that can be repurposed to treat psychiatric disorders. Human neuronal (NT2-N) cells were treated with a combination of atypical antipsychotic drugs commonly used to treat psychiatric disorders (such as schizophrenia, bipolar disorder, and major depressive disorder), and differential gene expression was analysed. Biological pathways with an increased gene expression included circadian rhythm and vascular endothelial growth factor signalling, while the adherens junction and cell cycle pathways were transcriptionally downregulated. The Connectivity Map (CMap) analysis screen highlighted drugs that affect global gene expression in a similar manner to these psychiatric disorder treatments, including several other antipsychotic drugs, confirming the utility of this approach. The CMap screen specifically identified metergoline, an ergot alkaloid currently used to treat seasonal affective disorder, as a drug of interest. In mice, metergoline dose-dependently reduced MK-801- or methamphetamine-induced locomotor hyperactivity confirming the potential of metergoline to treat positive symptoms of schizophrenia in an animal model. Metergoline had no effects on prepulse inhibition deficits induced by MK-801 or methamphetamine. Taken together, metergoline appears a promising drug for further studies to be repurposed as a treatment for schizophrenia and possibly other psychiatric disorders.
Collapse
Affiliation(s)
- Chiara C Bortolasci
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Emily J Jaehne
- School of Psychology and Public Health, La Trobe University, Bundoora, Australia
| | - Damián Hernández
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia.
| | - Briana Spolding
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Timothy Connor
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Bruna Panizzutti
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Olivia M Dean
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
- Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Tamsyn M Crowley
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Alison R Yung
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
- School of Health Sciences, University of Manchester, Manchester, UK
- Centre for Youth Mental Health, University of Melbourne, Parkville, Australia
| | - Laura Gray
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
- Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Jee Hyun Kim
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
- Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | | | - Michael Berk
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Ken Walder
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| |
Collapse
|
22
|
Kolar D, Krajcovic B, Kleteckova L, Kuncicka D, Vales K, Brozka H. Review: Genes Involved in Mitochondrial Physiology Within 22q11.2 Deleted Region and Their Relevance to Schizophrenia. Schizophr Bull 2023; 49:1637-1653. [PMID: 37379469 PMCID: PMC10686339 DOI: 10.1093/schbul/sbad066] [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] [Indexed: 06/30/2023]
Abstract
BACKGROUND AND HYPOTHESIS Schizophrenia is associated with altered energy metabolism, but the cause and potential impact of these metabolic changes remain unknown. 22q11.2 deletion syndrome (22q11.2DS) represents a genetic risk factor for schizophrenia, which is associated with the loss of several genes involved in mitochondrial physiology. Here we examine how the haploinsufficiency of these genes could contribute to the emergence of schizophrenia in 22q11.2DS. STUDY DESIGN We characterize changes in neuronal mitochondrial function caused by haploinsufficiency of mitochondria-associated genes within the 22q11.2 region (PRODH, MRPL40, TANGO2, ZDHHC8, SLC25A1, TXNRD2, UFD1, and DGCR8). For that purpose, we combine data from 22q11.2DS carriers and schizophrenia patients, in vivo (animal models) and in vitro (induced pluripotent stem cells, IPSCs) studies. We also review the current knowledge about seven non-coding microRNA molecules located in the 22q11.2 region that may be indirectly involved in energy metabolism by acting as regulatory factors. STUDY RESULTS We found that the haploinsufficiency of genes of interest is mainly associated with increased oxidative stress, altered energy metabolism, and calcium homeostasis in animal models. Studies on IPSCs from 22q11.2DS carriers corroborate findings of deficits in the brain energy metabolism, implying a causal role between impaired mitochondrial function and the development of schizophrenia in 22q11.2DS. CONCLUSIONS The haploinsufficiency of genes within the 22q11.2 region leads to multifaceted mitochondrial dysfunction with consequences to neuronal function, viability, and wiring. Overlap between in vitro and in vivo studies implies a causal role between impaired mitochondrial function and the development of schizophrenia in 22q11.2DS.
Collapse
Affiliation(s)
- David Kolar
- National Institute of Mental Health, Klecany, Czech Republic
| | - Branislav Krajcovic
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Daniela Kuncicka
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Karel Vales
- National Institute of Mental Health, Klecany, Czech Republic
| | - Hana Brozka
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
23
|
Nickl-Jockschat T, Sharkey R, Bacon C, Peterson Z, Rootes-Murdy K, Salvador R, Pomarol E, Karuk A, Homan P, Ji E, Omlor W, Homan S, Georgiadis F, Kaiser S, Kirschner M, Ehrlich S, Dannlowski U, Grotegerd D, Goltermann J, Meinert S, Kircher T, Stein F, Brosch K, Krug A, Nenadic I, Sim K, Piras F, Banaj N, Sponheim S, Demro C, Ramsay I, King M, Quidé Y, Green M, Nguyen D, Preda A, Calhoun V, Turner J, van Erp T, Spalletta G. Neural Correlates of Positive and Negative Formal Thought Disorder in Individuals with Schizophrenia: An ENIGMA Schizophrenia Working Group Study. RESEARCH SQUARE 2023:rs.3.rs-3179362. [PMID: 37841855 PMCID: PMC10571603 DOI: 10.21203/rs.3.rs-3179362/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Formal thought disorder (FTD) is a key clinical factor in schizophrenia, but the neurobiological underpinnings remain unclear. In particular, relationship between FTD symptom dimensions and patterns of regional brain volume deficiencies in schizophrenia remain to be established in large cohorts. Even less is known about the cellular basis of FTD. Our study addresses these major obstacles based on a large multi-site cohort through the ENIGMA Schizophrenia Working Group (752 individuals with schizophrenia and 1256 controls), to unravel the neuroanatomy of positive, negative and total FTD in schizophrenia and their cellular bases. We used virtual histology tools to relate brain structural changes associated with FTD to cellular distributions in cortical regions. We identified distinct neural networks for positive and negative FTD. Both networks encompassed fronto-occipito-amygdalar brain regions, but negative FTD showed a relative sparing of orbitofrontal cortical thickness, while positive FTD also affected lateral temporal cortices. Virtual histology identified distinct transcriptomic fingerprints associated for both symptom dimensions. Negative FTD was linked to neuronal and astrocyte fingerprints, while positive FTD was also linked to microglial cell types. These findings relate different dimensions of FTD to distinct brain structural changes and their cellular underpinnings, improve our mechanistic understanding of these key psychotic symptoms.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster
| | | | | | | | | | | | | | | | - Igor Nenadic
- Philipps University Marburg / Marburg University Hospital
| | | | | | | | | | | | | | | | | | | | | | | | - Vince Calhoun
- Georgia Institute of Technology, Emory University and Georgia State University
| | | | | | | |
Collapse
|
24
|
Gawande DY, S Narasimhan KK, Shelkar GP, Pavuluri R, Stessman HAF, Dravid SM. GluN2D Subunit in Parvalbumin Interneurons Regulates Prefrontal Cortex Feedforward Inhibitory Circuit and Molecular Networks Relevant to Schizophrenia. Biol Psychiatry 2023; 94:297-309. [PMID: 37004850 PMCID: PMC10524289 DOI: 10.1016/j.biopsych.2023.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/01/2023] [Accepted: 03/21/2023] [Indexed: 04/04/2023]
Abstract
BACKGROUND Parvalbumin interneuron (PVI) activity synchronizes the medial prefrontal cortex circuit for normal cognitive function, and its impairment may contribute to schizophrenia (SZ). NMDA receptors in PVIs participate in these activities and form the basis for the NMDA receptor hypofunction hypothesis of SZ. However, the role of the GluN2D subunit, which is enriched in PVIs, in regulating molecular networks relevant to SZ is unknown. METHODS Using electrophysiology and a mouse model with conditional deletion of GluN2D from PVIs (PV-GluN2D knockout [KO]), we examined the cell excitability and neurotransmission in the medial prefrontal cortex. Histochemical, RNA sequencing analysis and immunoblotting were conducted to understand molecular mechanisms. Behavioral analysis was conducted to test cognitive function. RESULTS PVIs in the medial prefrontal cortex were found to express putative GluN1/2B/2D receptors. In a PV-GluN2D KO model, PVIs were hypoexcitable, whereas pyramidal neurons were hyperexcitable. Excitatory neurotransmission was higher in both cell types in PV-GluN2D KO, whereas inhibitory neurotransmission showed contrasting changes, which could be explained by reduced somatostatin interneuron projections and increased PVI projections. Genes associated with GABA (gamma-aminobutyric acid) synthesis, vesicular release, and uptake as well as those involved in formation of inhibitory synapses, specifically GluD1-Cbln4 and Nlgn2, and regulation of dopamine terminals were downregulated in PV-GluN2D KO. SZ susceptibility genes including Disc1, Nrg1, and ErbB4 and their downstream targets were also downregulated. Behaviorally, PV-GluN2D KO mice showed hyperactivity and anxiety behavior and deficits in short-term memory and cognitive flexibility. CONCLUSIONS These findings demonstrate that GluN2D in PVIs serves as a point of convergence of pathways involved in the regulation of GABAergic synapses relevant to SZ.
Collapse
Affiliation(s)
- Dinesh Y Gawande
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska
| | | | - Gajanan P Shelkar
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska
| | - Ratnamala Pavuluri
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska
| | - Holly A F Stessman
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska
| | - Shashank M Dravid
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska.
| |
Collapse
|
25
|
Vinnakota C, Hudson MR, Jones NC, Sundram S, Hill RA. Potential Roles for the GluN2D NMDA Receptor Subunit in Schizophrenia. Int J Mol Sci 2023; 24:11835. [PMID: 37511595 PMCID: PMC10380280 DOI: 10.3390/ijms241411835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/19/2023] [Accepted: 07/22/2023] [Indexed: 07/30/2023] Open
Abstract
Glutamate N-methyl-D-aspartate receptor (NMDAR) hypofunction has been proposed to underlie schizophrenia symptoms. This theory arose from the observation that administration of NMDAR antagonists, which are compounds that inhibit NMDAR activity, reproduces behavioural and molecular schizophrenia-like phenotypes, including hallucinations, delusions and cognitive impairments in healthy humans and animal models. However, the role of specific NMDAR subunits in these schizophrenia-relevant phenotypes is largely unknown. Mounting evidence implicates the GluN2D subunit of NMDAR in some of these symptoms and pathology. Firstly, genetic and post-mortem studies show changes in the GluN2D subunit in people with schizophrenia. Secondly, the psychosis-inducing effects of NMDAR antagonists are blunted in GluN2D-knockout mice, suggesting that the GluN2D subunit mediates NMDAR-antagonist-induced psychotomimetic effects. Thirdly, in the mature brain, the GluN2D subunit is relatively enriched in parvalbumin (PV)-containing interneurons, a cell type hypothesized to underlie the cognitive symptoms of schizophrenia. Lastly, the GluN2D subunit is widely and abundantly expressed early in development, which could be of importance considering schizophrenia is a disorder that has its origins in early neurodevelopment. The limitations of currently available therapies warrant further research into novel therapeutic targets such as the GluN2D subunit, which may help us better understand underlying disease mechanisms and develop novel and more effective treatment options.
Collapse
Affiliation(s)
- Chitra Vinnakota
- Department of Psychiatry, School of Clinical Sciences, Faculty of Medical, Nursing and Health Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Matthew R Hudson
- Department of Neuroscience, Faculty of Medical, Nursing and Health Sciences, Monash University, Melbourne, VIC 3004, Australia
| | - Nigel C Jones
- Department of Neuroscience, Faculty of Medical, Nursing and Health Sciences, Monash University, Melbourne, VIC 3004, Australia
| | - Suresh Sundram
- Department of Psychiatry, School of Clinical Sciences, Faculty of Medical, Nursing and Health Sciences, Monash University, Clayton, VIC 3168, Australia
- Mental Health Program, Monash Health, Clayton, VIC 3168, Australia
| | - Rachel A Hill
- Department of Psychiatry, School of Clinical Sciences, Faculty of Medical, Nursing and Health Sciences, Monash University, Clayton, VIC 3168, Australia
| |
Collapse
|
26
|
Sehatpour P, Iosifescu DV, De Baun HM, Shope C, Mayer MR, Gangwisch J, Dias E, Sobeih T, Choo TH, Wall MM, Medalia A, Saperstein AM, Kegeles LS, Girgis RR, Carlson M, Kantrowitz JT. Dose-Dependent Augmentation of Neuroplasticity-Based Auditory Learning in Schizophrenia: A Double-Blind, Placebo-Controlled, Randomized, Target Engagement Clinical Trial of the NMDA Glutamate Receptor Agonist d-serine. Biol Psychiatry 2023; 94:164-173. [PMID: 36958998 PMCID: PMC10313776 DOI: 10.1016/j.biopsych.2023.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/30/2023]
Abstract
BACKGROUND Patients with schizophrenia show reduced NMDA glutamate receptor-dependent auditory plasticity, which is rate limiting for auditory cognitive remediation (AudRem). We evaluate the utility of behavioral and neurophysiological pharmacodynamic target engagement biomarkers, using a d-serine+AudRem combination. METHODS Forty-five participants with schizophrenia or schizoaffective disorder were randomized to 3 once-weekly AudRem visits + double-blind d-serine (80, 100, or 120 mg/kg) or placebo in 3 dose cohorts of 12 d-serine and 3 placebo-treated participants each. In AudRem, participants indicated which paired tone was higher in pitch. The primary outcome was plasticity improvement, operationalized as change in pitch threshold between AudRem tones [(test tone Hz - reference tone Hz)/reference tone Hz] between the initial plateau pitch threshold (mean of trials 20-30 of treatment visit 1) to pitch threshold at the end of visit(s). Target engagement was assessed by electroencephalography outcomes, including mismatch negativity (pitch primary). RESULTS There was a significant overall treatment effect for plasticity improvement (p = .014). Plasticity improvement was largest within the 80 and 100 mg/kg groups (p < .001, d > 0.67), while 120 mg/kg and placebo-treated participants showed nonsignificant within-group changes. Plasticity improvement was seen after a single treatment and was sustained on subsequent treatments. Target engagement was demonstrated by significantly larger mismatch negativity (p = .049, d = 1.0) for the 100 mg/kg dose versus placebo. CONCLUSIONS Our results demonstrate sufficient proof of principle for continued development of both the d-serine+AudRem combination and our target engagement methodology. The ultimate utility is dependent on the results of an ongoing larger, longer study of the combination for clinically relevant outcomes.
Collapse
Affiliation(s)
- Pejman Sehatpour
- Area Psychosis, New York State Psychiatric Institute, New York, New York; Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York; Nathan Kline Institute, Orangeburg, New York
| | - Dan V Iosifescu
- Nathan Kline Institute, Orangeburg, New York; Psychiatry, New York University Grossman School of Medicine, New York, New York
| | - Heloise M De Baun
- Area Psychosis, New York State Psychiatric Institute, New York, New York
| | | | - Megan R Mayer
- Area Psychosis, New York State Psychiatric Institute, New York, New York
| | - James Gangwisch
- Area Psychosis, New York State Psychiatric Institute, New York, New York; Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Elisa Dias
- Nathan Kline Institute, Orangeburg, New York; Psychiatry, New York University Grossman School of Medicine, New York, New York
| | | | - Tse-Hwei Choo
- Area Psychosis, New York State Psychiatric Institute, New York, New York; Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Melanie M Wall
- Area Psychosis, New York State Psychiatric Institute, New York, New York; Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Alice Medalia
- Area Psychosis, New York State Psychiatric Institute, New York, New York; Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Alice M Saperstein
- Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Lawrence S Kegeles
- Area Psychosis, New York State Psychiatric Institute, New York, New York; Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Ragy R Girgis
- Area Psychosis, New York State Psychiatric Institute, New York, New York; Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Marlene Carlson
- Area Psychosis, New York State Psychiatric Institute, New York, New York; Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Joshua T Kantrowitz
- Area Psychosis, New York State Psychiatric Institute, New York, New York; Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York; Nathan Kline Institute, Orangeburg, New York.
| |
Collapse
|
27
|
Wierońska JM, Cieślik P, Burnat G, Kalinowski L. Activation of Metabotropic Glutamate Receptor (mGlu 2) and Muscarinic Receptors (M 1, M 4, and M 5), Alone or in Combination, and Its Impact on the Acquisition and Retention of Learning in the Morris Water Maze, NMDA Expression and cGMP Synthesis. Biomolecules 2023; 13:1064. [PMID: 37509100 PMCID: PMC10377483 DOI: 10.3390/biom13071064] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/22/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
The Morris water maze (MWM) is regarded as one of the most popular tests for detecting spatial memory in rodents. Long-term potentiation and cGMP synthesis seem to be among the crucial factors involved in this type of learning. Muscarinic (M1, M4, and M5 receptors) and metabotropic glutamate (mGlu) receptors are important targets in the search for antipsychotic drugs with the potency to treat cognitive disabilities associated with the disorder. Here, we show that muscarinic receptor activators (VU0357017, VU0152100, and VU0238429) and an mGlu2 receptor activator, LY487379, dose-dependently prevented the development of cognitive disorders as a result of MK-801 administration in the MWM. The dose-ranges of the compounds were as follows: VU0357017, 0.25, 0.5, and 1 mg/kg; VU0152100, 0.05, 0.25, and 1 mg/kg; VU0238429, 1, 5, and 20 mg/kg; and LY487379, 0.5, 3, and 5 mg/kg. The co-administration of LY487379 with each of the individual muscarinic receptor ligands showed no synergistic effect, which contradicts the results obtained earlier in the novel object recognition (NOR) test. MWM learning resulted in increased cGMP synthesis, both in the cortex and hippocampi, when compared to that in intact animals, which was prevented by MK-801 administration. The investigated compounds at the highest doses reversed this MK-801-induced effect. Neither the procedure nor the treatment resulted in changes in GluN2B-NMDA expression.
Collapse
Affiliation(s)
- Joanna M Wierońska
- Maj Institute of Pharmacology Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Paulina Cieślik
- Maj Institute of Pharmacology Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Grzegorz Burnat
- Maj Institute of Pharmacology Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 7 Debinki Street, 80-211 Gdansk, Poland
- BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 11/12 Gabriela Narutowicza Street, 80-233 Gdansk, Poland
| |
Collapse
|
28
|
Dong B, Yue Y, Dong H, Wang Y. N-methyl-D-aspartate receptor hypofunction as a potential contributor to the progression and manifestation of many neurological disorders. Front Mol Neurosci 2023; 16:1174738. [PMID: 37396784 PMCID: PMC10308130 DOI: 10.3389/fnmol.2023.1174738] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/26/2023] [Indexed: 07/04/2023] Open
Abstract
N-methyl-D-aspartate receptors (NMDA) are glutamate-gated ion channels critical for synaptic transmission and plasticity. A slight variation of NMDAR expression and function can result in devastating consequences, and both hyperactivation and hypoactivation of NMDARs are detrimental to neural function. Compared to NMDAR hyperfunction, NMDAR hypofunction is widely implicated in many neurological disorders, such as intellectual disability, autism, schizophrenia, and age-related cognitive decline. Additionally, NMDAR hypofunction is associated with the progression and manifestation of these diseases. Here, we review the underlying mechanisms of NMDAR hypofunction in the progression of these neurological disorders and highlight that targeting NMDAR hypofunction is a promising therapeutic intervention in some neurological disorders.
Collapse
Affiliation(s)
- Bin Dong
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| | - Yang Yue
- School of Psychology, Northeast Normal University, Changchun, China
| | - Han Dong
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| | - Yuehui Wang
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
29
|
Sharkey RJ, Bacon C, Peterson Z, Rootes-Murdy K, Salvador R, Pomarol-Clotet E, Karuk A, Homan P, Ji E, Omlor W, Homan S, Georgiadis F, Kaiser S, Kirschner M, Ehrlich S, Dannlowski U, Grotegerd D, Goltermann J, Meinert S, Kircher T, Stein F, Brosch K, Krug A, Nenadić I, Sim K, Spalletta G, Piras F, Banaj N, Sponheim SR, Demro C, Ramsay IS, King M, Quidé Y, Green MJ, Nguyen D, Preda A, Calhoun VD, Turner JA, van Erp TGM, Nickl-Jockschat T. Neural Correlates of Positive and Negative Formal Thought Disorder in Individuals with Schizophrenia: An ENIGMA Schizophrenia Working Group Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.06.23291034. [PMID: 37333179 PMCID: PMC10274967 DOI: 10.1101/2023.06.06.23291034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Formal thought disorder (FTD) is a key clinical factor in schizophrenia, but the neurobiological underpinnings remain unclear. In particular, relationship between FTD symptom dimensions and patterns of regional brain volume deficiencies in schizophrenia remain to be established in large cohorts. Even less is known about the cellular basis of FTD. Our study addresses these major obstacles based on a large multi-site cohort through the ENIGMA Schizophrenia Working Group (752 individuals with schizophrenia and 1256 controls), to unravel the neuroanatomy of positive, negative and total FTD in schizophrenia and their cellular bases. We used virtual histology tools to relate brain structural changes associated with FTD to cellular distributions in cortical regions. We identified distinct neural networks for positive and negative FTD. Both networks encompassed fronto-occipito-amygdalar brain regions, but negative FTD showed a relative sparing of orbitofrontal cortical thickness, while positive FTD also affected lateral temporal cortices. Virtual histology identified distinct transcriptomic fingerprints associated for both symptom dimensions. Negative FTD was linked to neuronal and astrocyte fingerprints, while positive FTD was also linked to microglial cell types. These findings relate different dimensions of FTD to distinct brain structural changes and their cellular underpinnings, improve our mechanistic understanding of these key psychotic symptoms.
Collapse
|
30
|
Govani V, Shastry A, Iosifescu D, Govil P, Mayer M, Sobeih T, Choo T, Wall M, Sehatpour P, Kantrowitz J. Augmentation of learning in schizophrenia by D-serine is related to auditory and frontally-generated biomarkers: A randomized, double-blind, placebo-controlled study. RESEARCH SQUARE 2023:rs.3.rs-2943290. [PMID: 37293030 PMCID: PMC10246259 DOI: 10.21203/rs.3.rs-2943290/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Auditory cognition is impaired in schizophrenia, and typically engages a complex, distributed, hierarchical network, including both auditory and frontal input. We recently demonstrated proof of principle for the target engagement of an N-methyl-D-aspartate-type glutamate receptor (NMDAR) agonist + auditory targeted remediation (d-serine+AudRem) combination, showing significant improvement in auditory-learning induced plasticity and mismatch negativity. In this secondary analysis, we report on frontal EEG outcomes, assessing for both generalized effects and the mechanism of auditory plasticity. 21 schizophrenia or schizoaffective disorder participants were randomized to three 1x weekly AudRem + double-blind d-serine (100 mg/kg) visits. In AudRem, participants indicated which paired tone was higher in pitch. The focus of this secondary analysis was a frontally (premotor) mediated EEG outcome- event-related desynchronization in the b band (b-ERD), which was shown to be sensitive to AudRem in previous studies. d-Serine+AudRem led to significant improvement in b-ERD power across the retention and motor preparation intervals (F 1,18 =6.0, p=0.025) vs. AudRem alone. b-ERD was significantly related to baseline cognition, but not auditory-learning induced plasticity. The principal finding of this prespecified secondary analysis are that in addition to improving auditory based biomarkers, the d-serine+AudRem combination led to significant improvement in biomarkers thought to represent frontally mediated dysfunction, suggesting potential generalization of effects. Changes in auditory-learning induced plasticity were independent of these frontally mediated biomarkers. Ongoing work will assess whether d-serine+AudRem is sufficient to remediate cognition or whether targeting frontal NMDAR deficits with higher-level remediation may also be required. Trial Registration: NCT03711500.
Collapse
|
31
|
Dormann OD, Schuelert N, Rosenbrock H. Effects of the mGlu2/3 receptor agonist LY379268 on two models of disturbed auditory evoked brain oscillations in mice. Transl Psychiatry 2023; 13:150. [PMID: 37147311 PMCID: PMC10162958 DOI: 10.1038/s41398-023-02455-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 05/07/2023] Open
Abstract
Cognitive impairment is a core feature of schizophrenia and is poorly addressed by currently available medication. This is partly because the underlying circuits are insufficiently understood, and available animal models for brain dysfunction do not adequately mimic human pathology. To improve the translatability of animal studies and complement behavioral data, EEG measurements are being increasingly used in preclinical research. Brain oscillations are similar across species and can be impaired via several means. In this study, we used two approaches to impair early sensory processing and cortical oscillations in mice: a pharmacological model targeting NMDA receptor function in the whole brain via systemic MK-801 application and an optogenetic model targeting parvalbumin-positive (PV+) interneurons locally in the medial prefrontal cortex (mPFC). We evoked brain activity using auditory stimulation, a tool with high translatability from mouse to human. We then investigated the effect of LY379268, an agonist of mGlu2/3 receptors, a potential therapeutic target for schizophrenia, on single neuron and EEG responses. LY379268 was able to rescue MK-801-induced deficits for a variety of clinically relevant early sensory EEG biomarkers. Single neuron recordings revealed a strong effect of LY379268 on the signal-to-noise ratio during auditory stimulation and optogenetic inhibition of PV+ interneurons. Our results contribute to a better understanding of how group II metabotropic glutamate receptors modulate neuronal population and network activity under sensory stimulation while challenged pharmacologically or optogenetically.
Collapse
Affiliation(s)
- Oana-Daniela Dormann
- Central Nervous System Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach Riss, Germany.
| | - Niklas Schuelert
- Central Nervous System Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach Riss, Germany
| | - Holger Rosenbrock
- Central Nervous System Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach Riss, Germany
| |
Collapse
|
32
|
Jalewa J, Todd J, Michie PT, Hodgson DM, Harms L. The effect of schizophrenia risk factors on mismatch responses in a rat model. Psychophysiology 2023; 60:e14175. [PMID: 36087044 PMCID: PMC10909418 DOI: 10.1111/psyp.14175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 06/28/2022] [Accepted: 08/04/2022] [Indexed: 01/06/2023]
Abstract
Reduced mismatch negativity (MMN), a robust finding in schizophrenia, has prompted interest in MMN as a preclinical biomarker of schizophrenia. The rat brain can generate human-like mismatch responses (MMRs) which therefore enables the exploration of the neurobiology of reduced MMRs. Given epidemiological evidence that two developmental factors, maternal infection and adolescent cannabis use, increase the risk of schizophrenia, we determined the effect of these two developmental risk factors on rat MMR amplitude in different auditory contexts. MMRs were assessed in awake adult male and female Wistar rats that were offspring of pregnant dams treated with either a viral infection mimetic (poly I:C) inducing maternal immune activation (MIA) or saline control. In adolescence, subgroups of the prenatal treatment groups were exposed to either a synthetic cannabinoid (adolescent cannabinoid exposure: ACE) or vehicle. The context under which MMRs were obtained was manipulated by employing two different oddball paradigms, one that manipulated the physical difference between rare and common auditory stimuli, and another that manipulated the probability of the rare stimulus. The design of the multiple stimulus sequences across the two paradigms also allowed an investigation of context on MMRs to two identical stimulus sequences. Male offspring exposed to each of the risk factors for schizophrenia (MIA, ACE or both) showed a reduction in MMR, which was evident only in the probability paradigm, with no effects seen in the physical difference. Our findings highlight the importance of contextual factors induced by paradigm manipulations and sex for modeling schizophrenia-like MMN impairments in rats.
Collapse
Affiliation(s)
- Jaishree Jalewa
- School of Psychological Sciences, College of Engineering, Science and EnvironmentUniversity of NewcastleCallaghanNew South WalesAustralia
| | - Juanita Todd
- School of Psychological Sciences, College of Engineering, Science and EnvironmentUniversity of NewcastleCallaghanNew South WalesAustralia
- Hunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Patricia T. Michie
- School of Psychological Sciences, College of Engineering, Science and EnvironmentUniversity of NewcastleCallaghanNew South WalesAustralia
- Hunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Deborah M. Hodgson
- School of Psychological Sciences, College of Engineering, Science and EnvironmentUniversity of NewcastleCallaghanNew South WalesAustralia
- Hunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Lauren Harms
- Hunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
- School of Biomedical Science and Pharmacy, College of Health, Medicine and WellbeingUniversity of NewcastleCallaghanNew South WalesAustralia
| |
Collapse
|
33
|
Translational cognitive systems: focus on attention. Emerg Top Life Sci 2022; 6:529-539. [PMID: 36408755 DOI: 10.1042/etls20220009] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/22/2022]
Abstract
Cognitive dysfunction, particularly attentional impairment, is a core feature of many psychiatric disorders, yet is inadequately addressed by current treatments. Development of targeted therapeutics for the remediation of attentional deficits requires knowledge of underlying neurocircuit, cellular, and molecular mechanisms that cannot be directly assayed in the clinic. This level of detail can only be acquired by testing animals in cross-species translatable attentional paradigms, in combination with preclinical neuroscience techniques. The 5-choice continuous performance test (5C-CPT) and rodent continuous performance test (rCPT) represent the current state of the art of preclinical assessment of the most commonly studied subtype of attention: sustained attention, or vigilance. These tasks present animals with continuous streams of target stimuli to which they must respond (attention), in addition to non-target stimuli from which they must withhold responses (behavioral inhibition). The 5C-CPT and rCPT utilize the same measures as gold-standard clinical continuous performance tests and predict clinical efficacy of known pro-attentional drugs. They also engage common brain regions across species, although efforts to definitively establish neurophysiological construct validity are ongoing. The validity of these tasks as translational vigilance assessments enables their use in characterizing the neuropathology underlying attentional deficits of animal models of psychiatric disease, and in determining therapeutic potential of drugs ahead of clinical testing. Here, we briefly review the development and validation of such tests of attentional functioning, as well as the data they have generated pertaining to inattention, disinhibition, and impulsivity in psychiatric disorders.
Collapse
|
34
|
Antipsychotic prescription, assumption and conversion to psychosis: resolving missing clinical links to optimize prevention through precision. SCHIZOPHRENIA 2022; 8:48. [PMID: 35853891 PMCID: PMC9261109 DOI: 10.1038/s41537-022-00254-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/04/2022] [Indexed: 11/08/2022]
Abstract
AbstractThe current concept of clinical high-risk(CHR) of psychosis relies heavily on “below-threshold” (i.e. attenuated or limited and intermittent) psychotic positive phenomena as predictors of the risk for future progression to “above-threshold” positive symptoms (aka “transition” or “conversion”). Positive symptoms, even at attenuated levels are often treated with antipsychotics (AP) to achieve clinical stabilization and mitigate the psychopathological severity. The goal of this study is to contextually examine clinicians’ decision to prescribe AP, CHR individuals’ decision to take AP and psychosis conversion risk in relation to prodromal symptoms profiles. CHR individuals (n = 600) were recruited and followed up for 2 years between 2016 and 2021. CHR individuals were referred to the participating the naturalistic follow-up study, which research procedure was independent of the routine clinical treatment. Clinical factors from the Structured Interview for Prodromal Syndromes (SIPS) and global assessment of function (GAF) were profiled via exploratory factor analysis (EFA), then the extracted factor structure was used to investigate the relationship of prodromal psychopathology with clinicians’ decisions to AP-prescription, CHR individuals’ decisions to AP-taking and conversion to psychosis. A total of 427(71.2%) CHR individuals were prescribed AP at baseline, 532(88.7%) completed the 2-year follow-up, 377(377/532, 70.9%) were taken AP at least for 2 weeks during the follow-up. EFA identified six factors (Factor-1-Negative symptoms, Factor-2-Global functions, Factor-3-Disorganized communication & behavior, Factor-4-General symptoms, Factor-5-Odd thoughts, and Factor-6-Distorted cognition & perception). Positive symptoms (Factor-5 and 6) and global functions (Factor-2) factors were significant predictors for clinicians’ decisions to AP-prescription and CHR individuals’ decisions to assume AP, whereas negative symptoms (Factor-1) and global functions (Factor-2) factors predicted conversion. While decisions to AP-prescription, decisions to AP-taking were associated to the same factors (positive symptoms and global functions), only one of those was predictive of conversion, i.e. global functions. The other predictor of conversion, i.e. negative symptoms, did not seem to be contemplated both on the clinician and patients’ sides. Overall, the findings indicated that a realignment in the understanding of AP usage is warranted.
Collapse
|
35
|
Kuo CY, Lin CH, Lane HY. Targeting D-Amino Acid Oxidase (DAAO) for the Treatment of Schizophrenia: Rationale and Current Status of Research. CNS Drugs 2022; 36:1143-1153. [PMID: 36194364 DOI: 10.1007/s40263-022-00959-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/14/2022] [Indexed: 11/29/2022]
Abstract
In the brain, D-amino acid oxidase (DAAO) is a peroxisomal flavoenzyme. Through oxidative deamination by DAAO, D-serine, the main coagonist of synaptic N-methyl-D-aspartate receptors (NMDARs), is degraded into α-keto acids and ammonia; flavin adenine dinucleotide (FAD) is simultaneously reduced to dihydroflavine-adenine dinucleotide (FADH2), which is subsequently reoxidized to FAD, with hydrogen peroxide produced as a byproduct. NMDAR hypofunction is implicated in the pathogenesis of schizophrenia. In previous studies, compared with control subjects, patients with schizophrenia had lower D-serine levels in peripheral blood and cerebrospinal fluid but higher DAAO expression and activity in the brain. Inhibiting DAAO activity and slowing D-serine degradation by using DAAO inhibitors to enhance NMDAR function may be a new strategy for use in the treatment of schizophrenia. The aim of this leading article is to review the current research in DAAO inhibitors.
Collapse
Affiliation(s)
- Chien-Yi Kuo
- Department of Psychiatry, China Medical University Hospital, No. 2, Yude Rd., North Dist., Taichung City, 404327, Taiwan, ROC.,Graduate Institute of Biomedical Sciences, China Medical University, No. 91, Xueshi Rd., North Dis., Taichung City, 404333, Taiwan, ROC
| | - Chieh-Hsin Lin
- Graduate Institute of Biomedical Sciences, China Medical University, No. 91, Xueshi Rd., North Dis., Taichung City, 404333, Taiwan, ROC. .,Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, No. 123, Dapi Rd., Niaosong Dist., Kaohsiung City, 83301, Taiwan, ROC. .,School of Medicine, Chang Gung University, No. 259, Wenhua 1st Rd., Guishan Dist., Taoyuan City, 33302, Taiwan, ROC.
| | - Hsien-Yuan Lane
- Department of Psychiatry, China Medical University Hospital, No. 2, Yude Rd., North Dist., Taichung City, 404327, Taiwan, ROC. .,Graduate Institute of Biomedical Sciences, China Medical University, No. 91, Xueshi Rd., North Dis., Taichung City, 404333, Taiwan, ROC. .,Department of Psychology, College of Medical and Health Sciences, Asia University, No. 500, Lioufeng Rd., Wufeng Dist., Taichung City, 413305, Taiwan, ROC.
| |
Collapse
|
36
|
Omeiza NA, Bakre AG, Abdulrahim HA, Isibor H, Ezurike PU, Sowunmi AA, Ben-Azu B, Aderibigbe AO. Pretreatment with Carpolobia lutea ethanol extract prevents schizophrenia-like behavior in mice models of psychosis. JOURNAL OF ETHNOPHARMACOLOGY 2022; 295:115432. [PMID: 35659625 DOI: 10.1016/j.jep.2022.115432] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/28/2022] [Accepted: 05/31/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Carpolobia lutea decoction is widely used as a phytotherapeutic against central nervous system-related disorders including insomnia, migraine headache, and mental illness in West and Central Tropical Africa. AIM This study was designed to investigate the antipsychotic activity of Carpolobia lutea (EECL) in mice models of psychosis. METHODS Male Swiss mice (n = 5/group) were given EECL (100, 200, 400, and 800 mg/kg), haloperidol (1 mg/kg), clozapine (5 mg/kg) and vehicle (10 mL/kg) orally before amphetamine (5 mg/kg)-induced hyperlocomotion and stereotypy, apomorphine (2 mg/kg)-induced stereotypy, or ketamine (10, 30, and 100 mg/kg)-induced hyperlocomotion, enhancement of immobility and cognitive impairment. RESULTS EECL (200, 400, and 800 mg/kg) prevented amphetamine- and apomorphine-induced stereotypies, as well as reduced hyperlocomotion induced by amphetamine and ketamine, all of which are predictors of positive symptoms. Regardless of the dose administered, EECL prevented the index of negative symptoms induced by ketamine. Furthermore, higher doses of EECL (400 and 800 mg/kg) also prevented ketamine-induced cognitive impairment, a behavioral phenotype of cognitive symptoms. CONCLUSION Pretreatment with EECL demonstrated antipsychotic activity in mice, preventing amphetamine-, apomorphine-, and ketamine-induced schizophrenia-like symptoms, with 800 mg/kg being the most effective dose.
Collapse
Affiliation(s)
- Noah A Omeiza
- Department of Pharmacology and Therapeutics, Neuropharmacology Unit, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| | - Adewale G Bakre
- Department of Pharmacology and Therapeutics, Neuropharmacology Unit, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Halimat A Abdulrahim
- Department of Medical Biochemistry, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Happy Isibor
- Department of Pharmacology and Therapeutics, Neuropharmacology Unit, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Precious U Ezurike
- Department of Pharmacology and Therapeutics, Neuropharmacology Unit, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Abimbola A Sowunmi
- Department of Pharmacology and Therapeutics, Neuropharmacology Unit, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Benneth Ben-Azu
- Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Adegbuyi O Aderibigbe
- Department of Pharmacology and Therapeutics, Neuropharmacology Unit, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
37
|
Casas BS, Arancibia-Altamirano D, Acevedo-La Rosa F, Garrido-Jara D, Maksaev V, Pérez-Monje D, Palma V. It takes two to tango: Widening our understanding of the onset of schizophrenia from a neuro-angiogenic perspective. Front Cell Dev Biol 2022; 10:946706. [PMID: 36092733 PMCID: PMC9448889 DOI: 10.3389/fcell.2022.946706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Schizophrenia is a chronic debilitating mental disorder characterized by perturbations in thinking, perception, and behavior, along with brain connectivity deficiencies, neurotransmitter dysfunctions, and loss of gray brain matter. To date, schizophrenia has no cure and pharmacological treatments are only partially efficacious, with about 30% of patients describing little to no improvement after treatment. As in most neurological disorders, the main descriptions of schizophrenia physiopathology have been focused on neural network deficiencies. However, to sustain proper neural activity in the brain, another, no less important network is operating: the vast, complex and fascinating vascular network. Increasing research has characterized schizophrenia as a systemic disease where vascular involvement is important. Several neuro-angiogenic pathway disturbances have been related to schizophrenia. Alterations, ranging from genetic polymorphisms, mRNA, and protein alterations to microRNA and abnormal metabolite processing, have been evaluated in plasma, post-mortem brain, animal models, and patient-derived induced pluripotent stem cell (hiPSC) models. During embryonic brain development, the coordinated formation of blood vessels parallels neuro/gliogenesis and results in the structuration of the neurovascular niche, which brings together physical and molecular signals from both systems conforming to the Blood-Brain barrier. In this review, we offer an upfront perspective on distinctive angiogenic and neurogenic signaling pathways that might be involved in the biological causality of schizophrenia. We analyze the role of pivotal angiogenic-related pathways such as Vascular Endothelial Growth Factor and HIF signaling related to hypoxia and oxidative stress events; classic developmental pathways such as the NOTCH pathway, metabolic pathways such as the mTOR/AKT cascade; emerging neuroinflammation, and neurodegenerative processes such as UPR, and also discuss non-canonic angiogenic/axonal guidance factor signaling. Considering that all of the mentioned above pathways converge at the Blood-Brain barrier, reported neurovascular alterations could have deleterious repercussions on overall brain functioning in schizophrenia.
Collapse
|
38
|
Bauminger H, Gaisler-Salomon I. Beyond NMDA Receptors: Homeostasis at the Glutamate Tripartite Synapse and Its Contributions to Cognitive Dysfunction in Schizophrenia. Int J Mol Sci 2022; 23:8617. [PMID: 35955750 PMCID: PMC9368772 DOI: 10.3390/ijms23158617] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/30/2022] [Accepted: 08/01/2022] [Indexed: 02/01/2023] Open
Abstract
Cognitive deficits are core symptoms of schizophrenia but remain poorly addressed by dopamine-based antipsychotic medications. Glutamate abnormalities are implicated in schizophrenia-related cognitive deficits. While the role of the NMDA receptor has been extensively studied, less attention was given to other components that control glutamate homeostasis. Glutamate dynamics at the tripartite synapse include presynaptic and postsynaptic components and are tightly regulated by neuron-astrocyte crosstalk. Here, we delineate the role of glutamate homeostasis at the tripartite synapse in schizophrenia-related cognitive dysfunction. We focus on cognitive domains that can be readily measured in humans and rodents, i.e., working memory, recognition memory, cognitive flexibility, and response inhibition. We describe tasks used to measure cognitive function in these domains in humans and rodents, and the relevance of glutamate alterations in these domains. Next, we delve into glutamate tripartite synaptic components and summarize findings that implicate the relevance of these components to specific cognitive domains. These collective findings indicate that neuron-astrocyte crosstalk at the tripartite synapse is essential for cognition, and that pre- and postsynaptic components play a critical role in maintaining glutamate homeostasis and cognitive well-being. The contribution of these components to cognitive function should be considered in order to better understand the role played by glutamate signaling in cognition and develop efficient pharmacological treatment avenues for schizophrenia treatment-resistant symptoms.
Collapse
Affiliation(s)
- Hagar Bauminger
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel;
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| | - Inna Gaisler-Salomon
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel;
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| |
Collapse
|
39
|
Harrison PJ, Mould A, Tunbridge EM. New drug targets in psychiatry: Neurobiological considerations in the genomics era. Neurosci Biobehav Rev 2022; 139:104763. [PMID: 35787892 DOI: 10.1016/j.neubiorev.2022.104763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 05/15/2022] [Accepted: 06/14/2022] [Indexed: 01/11/2023]
Abstract
After a period of withdrawal, pharmaceutical companies have begun to reinvest in neuropsychiatric disorders, due to improvements in our understanding of these disorders, stimulated in part by genomic studies. However, translating this information into disease insights and ultimately into tractable therapeutic targets is a major challenge. Here we consider how different sources of information might be integrated to guide this process. We review how an understanding of neurobiology has been used to advance therapeutic candidates identified in the pre-genomic era, using catechol-O-methyltransferase (COMT) as an exemplar. We then contrast with ZNF804A, the first genome-wide significant schizophrenia gene, and draw on some of the lessons that these and other examples provide. We highlight that, at least in the short term, the translation of potential targets for which there is orthogonal neurobiological support is likely to be more straightforward and productive than that those relying solely on genomic information. Although we focus here on information from genomic studies of schizophrenia, the points are broadly applicable across major psychiatric disorders and their symptoms.
Collapse
Affiliation(s)
- Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health NHS Foundation Trust, Oxford, UK
| | - Arne Mould
- Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health NHS Foundation Trust, Oxford, UK
| | - Elizabeth M Tunbridge
- Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
40
|
de Bartolomeis A, Vellucci L, Austin MC, De Simone G, Barone A. Rational and Translational Implications of D-Amino Acids for Treatment-Resistant Schizophrenia: From Neurobiology to the Clinics. Biomolecules 2022; 12:biom12070909. [PMID: 35883465 PMCID: PMC9312470 DOI: 10.3390/biom12070909] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 12/13/2022] Open
Abstract
Schizophrenia has been conceptualized as a neurodevelopmental disorder with synaptic alterations and aberrant cortical–subcortical connections. Antipsychotics are the mainstay of schizophrenia treatment and nearly all share the common feature of dopamine D2 receptor occupancy, whereas glutamatergic abnormalities are not targeted by the presently available therapies. D-amino acids, acting as N-methyl-D-aspartate receptor (NMDAR) modulators, have emerged in the last few years as a potential augmentation strategy in those cases of schizophrenia that do not respond well to antipsychotics, a condition defined as treatment-resistant schizophrenia (TRS), affecting almost 30–40% of patients, and characterized by serious cognitive deficits and functional impairment. In the present systematic review, we address with a direct and reverse translational perspective the efficacy of D-amino acids, including D-serine, D-aspartate, and D-alanine, in poor responders. The impact of these molecules on the synaptic architecture is also considered in the light of dendritic spine changes reported in schizophrenia and antipsychotics’ effect on postsynaptic density proteins. Moreover, we describe compounds targeting D-amino acid oxidase and D-aspartate oxidase enzymes. Finally, other drugs acting at NMDAR and proxy of D-amino acids function, such as D-cycloserine, sarcosine, and glycine, are considered in the light of the clinical burden of TRS, together with other emerging molecules.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, 80131 Naples, Italy; (L.V.); (G.D.S.); (A.B.)
- Correspondence: ; Tel.: +39-081-7463673 or +39-081-7463884 or +39-3662745592; Fax: +39-081-7462644
| | - Licia Vellucci
- Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, 80131 Naples, Italy; (L.V.); (G.D.S.); (A.B.)
| | - Mark C. Austin
- Clinical Psychopharmacology Program, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA;
| | - Giuseppe De Simone
- Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, 80131 Naples, Italy; (L.V.); (G.D.S.); (A.B.)
| | - Annarita Barone
- Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, 80131 Naples, Italy; (L.V.); (G.D.S.); (A.B.)
| |
Collapse
|
41
|
Qi B, Boscenco S, Ramamurthy J, Trakadis YJ. Transcriptomics and machine learning to advance schizophrenia genetics: A case-control study using post-mortem brain data. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 214:106590. [PMID: 34954633 DOI: 10.1016/j.cmpb.2021.106590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 08/31/2021] [Accepted: 12/14/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND AND OBJECTIVE Alterations of the expression of a variety of genes have been reported in patients with schizophrenia (SCZ). Moreover, machine learning (ML) analysis of gene expression microarray data has shown promising preliminary results in the study of SCZ. Our objective was to evaluate the performance of ML in classifying SCZ cases and controls based on gene expression microarray data from the dorsolateral prefrontal cortex. METHODS We apply a state-of-the-art ML algorithm (XGBoost) to train and evaluate a classification model using 201 SCZ cases and 278 controls. We utilized 10-fold cross-validation for model selection, and a held-out testing set to evaluate the model. The performance metric utilizes to evaluate classification performance was the area under the receiver-operator characteristics curve (AUC). RESULTS We report an average AUC on 10-fold cross-validation of 0.76 and an AUC of 0.76 on testing data, not used during training. Analysis of the rolling balanced classification accuracy from high to low prediction confidence levels showed that the most certain subset of predictions ranged between 80-90%. The ML model utilized 182 gene expression probes. Further improvement to classification performance was observed when applying an automated ML strategy on the 182 features, which achieved an AUC of 0.79 on the same testing data. We found literature evidence linking all of the top ten ML ranked genes to SCZ. Furthermore, we leveraged information from the full set of microarray gene expressions available via univariate differential gene expression analysis. We then prioritized differentially expressed gene sets using the piano gene set analysis package. We augmented the ranking of the prioritized gene sets with genes from the complex multivariate ML model using hypergeometric tests to identify more robust gene sets. We identified two significant Gene Ontology molecular function gene sets: "oxidoreductase activity, acting on the CH-NH2 group of donors" and "integrin binding." Lastly, we present candidate treatments for SCZ based on findings from our study CONCLUSIONS: Overall, we observed above-chance performance from ML classification of SCZ cases and controls based on brain gene expression microarray data, and found that ML analysis of gene expressions could further our understanding of the pathophysiology of SCZ and help identify novel treatments.
Collapse
Affiliation(s)
- Bill Qi
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Sonia Boscenco
- Faculty of Science, McGill University, Montreal, QC, Canada
| | | | - Yannis J Trakadis
- Department of Human Genetics, McGill University, Montreal, QC, Canada; Department of Medical Genetics, McGill University Health Center, Montreal, QC, Canada.
| |
Collapse
|
42
|
Abstract
The cognitive dysfunction experienced by patients with schizophrenia represents a major unmet clinical need. We believe that enhancing synaptic function and plasticity by targeting kalirin may provide a novel means to remediate these symptoms. Karilin (a protein encoded by the KALRN gene) has multiple functional domains, including two Dbl homology (DH) guanine exchange factor (GEF) domains, which act to enhance the activity of the Rho family guanosine triphosphate (GTP)-ases. Here, we provide an overview of kalirin's roles in brain function and its therapeutic potential in schizophrenia. We outline how it mediates diverse effects via a suite of distinct isoforms that couple to members of the Rho GTPase family to regulate synapse formation and stabilisation, and how genomic and post-mortem data implicate it in schizophrenia. We then review the current state of knowledge about the influence of kalirin on brain function at a systems level, based largely on evidence from transgenic mouse models, which support its proposed role in regulating dendritic spine function and plasticity. We demonstrate that, whilst the GTPases are classically considered to be 'undruggable', targeting kalirin and other Rho GEFs provides a means to indirectly modulate their activity. Finally, we integrate across the information presented to assess the therapeutic potential of kalirin for schizophrenia and highlight the key outstanding questions required to advance it in this capacity; namely, the need for more information about the diversity and function of its isoforms, how these change across neurodevelopment, and how they affect brain function in vivo.
Collapse
|
43
|
Guerrin CGJ, Doorduin J, Sommer IE, de Vries EFJ. The dual hit hypothesis of schizophrenia: Evidence from animal models. Neurosci Biobehav Rev 2021; 131:1150-1168. [PMID: 34715148 DOI: 10.1016/j.neubiorev.2021.10.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/21/2021] [Accepted: 10/24/2021] [Indexed: 12/16/2022]
Abstract
Schizophrenia is a heterogeneous psychiatric disorder, which can severely impact social and professional functioning. Epidemiological and clinical studies show that schizophrenia has a multifactorial aetiology comprising genetic and environmental risk factors. Although several risk factors have been identified, it is still not clear how they result in schizophrenia. This knowledge gap, however, can be investigated in animal studies. In this review, we summarise animal studies regarding molecular and cellular mechanisms through which genetic and environmental factors may affect brain development, ultimately causing schizophrenia. Preclinical studies suggest that early environmental risk factors can affect the immune, GABAergic, glutamatergic, or dopaminergic system and thus increase the susceptibility to another risk factor later in life. A second insult, like social isolation, stress, or drug abuse, can further disrupt these systems and the interactions between them, leading to behavioural abnormalities. Surprisingly, first insults like maternal infection and early maternal separation can also have protective effects. Single gene mutations associated with schizophrenia did not have a major impact on the susceptibility to subsequent environmental hits.
Collapse
Affiliation(s)
- Cyprien G J Guerrin
- Department of Nuclear Medicine and Medical Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Medical Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Iris E Sommer
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Erik F J de Vries
- Department of Nuclear Medicine and Medical Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands.
| |
Collapse
|
44
|
Metzner C, Steuber V. The beta component of gamma-band auditory steady-state responses in patients with schizophrenia. Sci Rep 2021; 11:20387. [PMID: 34650135 PMCID: PMC8516862 DOI: 10.1038/s41598-021-99793-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 09/24/2021] [Indexed: 01/18/2023] Open
Abstract
The mechanisms underlying circuit dysfunctions in schizophrenia (SCZ) remain poorly understood. Auditory steady-state responses (ASSRs), especially in the gamma and beta band, have been suggested as a potential biomarker for SCZ. While the reduction of 40 Hz power for 40 Hz drive has been well established and replicated in SCZ patients, studies are inconclusive when it comes to an increase in 20 Hz power during 40 Hz drive. There might be several factors explaining the inconsistencies, including differences in the sensitivity of the recording modality (EEG vs MEG), differences in stimuli (click-trains vs amplitude-modulated tones) and large differences in the amplitude of the stimuli. Here, we used a computational model of ASSR deficits in SCZ and explored the effect of three SCZ-associated microcircuit alterations: reduced GABA activity, increased GABA decay times and NMDA receptor hypofunction. We investigated the effect of input strength on gamma (40 Hz) and beta (20 Hz) band power during gamma ASSR stimulation and saw that the pronounced increase in beta power during gamma stimulation seen experimentally could only be reproduced in the model when GABA decay times were increased and only for a specific range of input strengths. More specifically, when the input was in this specific range, the rhythmic drive at 40 Hz produced a strong 40 Hz rhythm in the control network; however, in the 'SCZ-like' network, the prolonged inhibition led to a so-called 'beat-skipping', where the network would only strongly respond to every other input. This mechanism was responsible for the emergence of the pronounced 20 Hz beta peak in the power spectrum. The other two microcircuit alterations were not able to produce a substantial 20 Hz component but they further narrowed the input strength range for which the network produced a beta component when combined with increased GABAergic decay times. Our finding that the beta component only existed for a specific range of input strengths might explain the seemingly inconsistent reporting in experimental studies and suggests that future ASSR studies should systematically explore different amplitudes of their stimuli. Furthermore, we provide a mechanistic link between a microcircuit alteration and an electrophysiological marker in schizophrenia and argue that more complex ASSR stimuli are needed to disentangle the nonlinear interactions of microcircuit alterations. The computational modelling approach put forward here is ideally suited to facilitate the development of such stimuli in a theory-based fashion.
Collapse
Affiliation(s)
- Christoph Metzner
- Neural Information Processing Group, Institute of Software Engineering and Theoretical Computer Science, Technische Universität Berlin, Berlin, Germany.
- School of Physics, Engineering and Computer Science, University of Hertfordshire, Hatfield, UK.
| | - Volker Steuber
- School of Physics, Engineering and Computer Science, University of Hertfordshire, Hatfield, UK
| |
Collapse
|
45
|
Li G, Dai J, Liu H, Lin Y, Liu Q, Zheng K, Li S, Chen S, Ye Y. Association study between genetic variants and the risk of schizophrenia in the Chinese population based on GWAS-implicated 6p21.3-23.1 human genome region: a case-control study. BMC Psychiatry 2021; 21:483. [PMID: 34607594 PMCID: PMC8489080 DOI: 10.1186/s12888-021-03496-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 09/20/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Schizophrenia is a polygenic disease; however, the specific risk genetic variants of schizophrenia are still largely unknown. Single nucleotide polymorphism (SNP) is important genetic factor for the susceptibility of schizophrenia. Investigating individual candidate gene contributing to disease risk remains important. METHODS In a case-control study, five SNPs located in 6p21.3-p23.1 including rs2021722 in human leukocyte antigen (HLA) locus and rs107822, rs383711, rs439205 and rs421446 within the upstream of microRNA-219a-1 were genotyped in 454 schizophrenia patients and 445 healthy controls to investigate the possible association between the loci and schizophrenia in a Han Chinese population. RESULTS Our results showed significant associations between the rs2021722 and schizophrenia in allele (A vs. G: adjusted OR = 1.661, 95%CI = 1.196-2.308), co-dominant (AG vs. GG: OR = 1.760, 95%CI = 1.234-2.510) and dominant genetic model (AG + AA vs. GG: OR = 1.756, 95%CI = 1.237-2.492), respectively. Haplotype analysis showed that TGGT and CAAC were protective factor for schizophrenia compared with TAAC haplotype (OR = 0.324, 95% CI = 0.157-0.672; OR = 0.423, 95% CI = 0.199-0.900). CONCLUSIONS These findings indicate that rs2021722 in HLA locus might be involved in pathogenesis of schizophrenia and that genotypes AG and allele A of the locus are risk factors for schizophrenia in the Han Chinese population, confirming the association between immune system and schizophrenia.
Collapse
Affiliation(s)
- Gangqin Li
- grid.13291.380000 0001 0807 1581Department of Forensic Psychiatry, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041 Sichuan China
| | - Jie Dai
- grid.415880.00000 0004 1755 2258Department of Pathology, Sichuan Cancer Hospital, Chengdu, Sichuan China
| | - Hao Liu
- grid.13291.380000 0001 0807 1581Department of Forensic Psychiatry, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041 Sichuan China
| | - Yushan Lin
- grid.13291.380000 0001 0807 1581West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan China
| | - Qiaoni Liu
- grid.13291.380000 0001 0807 1581West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan China
| | - Kaiyuan Zheng
- grid.13291.380000 0001 0807 1581West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan China
| | - Suyu Li
- grid.13291.380000 0001 0807 1581West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan China
| | - Siyu Chen
- Nanchong Psychosomatic Hospital, Nanchong, 637000, Sichuan, China.
| | - Yi Ye
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
46
|
N-methyl-D-aspartate receptor antibody and the choroid plexus in schizophrenia patients with tardive dyskinesia. J Psychiatr Res 2021; 142:290-298. [PMID: 34411812 DOI: 10.1016/j.jpsychires.2021.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/02/2021] [Accepted: 08/09/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND Immune disturbance has been postulated to be one of the mechanisms underlying the pathogenesis of tardive dyskinesia (TD). Recently, the role of autoimmune abnormality in TD has been increasingly recognized. Autoantibodies against neuronal N-methyl-D-aspartate receptor (NMDAR) may be cross-reactive in the brain in neuropsychiatric disorders, and the choroid plexus (CP) is a crucial immune barrier in the central nervous system (CNS). We supposed that NMDAR antibodies might underlie the pathophysiological process of TD through the mediation of CP. METHODS Serum NMDAR antibody levels were assessed by enzyme-linked immunosorbent assay, CP and ventricle volumes were assessed by magnetic resonance imaging in schizophrenia patients with TD (n = 61), without TD (NTD, n = 61), and in healthy controls (n = 74). Psychopathology and TD severity were assessed by the Positive and Negative Syndrome Scale and Abnormal Involuntary Movement Scale (AIMS). RESULTS NMDAR antibody levels were significantly higher, CP volumes were larger in the TD group than in the NTD group (p = 0.022; p = 0.019, respectively). In the TD group, higher NMDAR antibody level was correlated with larger CP volume (β = 0.406, p = 0.002). An elevated NMDAR antibody level and enlarged CP volume were correlated with orofacial AIMS score (β = 0.331, p = 0.011; β = 0.459, p = 3.34 × 10-4, respectively). In a mediation model, the effect of NMDAR antibody level on the orofacial AIMS score was mediated by the CP volume (indirect effect: β = 0.08, 95% confidence interval = 0.002-0.225; direct effect: β = 0.14, p = 0.154). CONCLUSIONS Our findings highlight a potential NMDAR antibody-associated mechanism in orofacial TD, which may be mediated by increased CP volume.
Collapse
|
47
|
Meltzer HY, Gadaleta E. Contrasting Typical and Atypical Antipsychotic Drugs. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2021; 19:3-13. [PMID: 34483761 DOI: 10.1176/appi.focus.20200051] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The beliefs that antipsychotic drugs (APDs) are 1) effective only to treat delusions and hallucinations (positive symptoms), 2) that typical and atypical APDs differ only in ability to cause extrapyramidal side effects, and 3) that their efficacy as antipsychotics is due solely to their dopamine D2 receptor blockade are outmoded concepts that prevent clinicians from achieving optimal clinical results when prescribing an APD. Atypical APDs are often more effective than typical APDs in treating negative symptoms, cognitive impairment, and mood symptoms as well as reducing the risk for suicide and decreasing aggression. This applies not only to those diagnosed with schizophrenia or schizoaffective disorder but also to bipolar disorder, major depression, and other psychiatric diagnoses. The greater advantage of an atypical APD is not evident in all patients for every atypical APD due, in part, to individual differences in genetic and epigenetic endowment and differences in the pharmacology of the atypical APDs, their mode of action being far more complex than that of the typical APDs. A common misconception is that among the atypical APDs, only clozapine is effective for reducing psychosis in treatment-resistant schizophrenia. Aripiprazole, lurasidone, olanzapine, and risperidone also can be more effective than typical APDs for treatment-resistant schizophrenia; clozapine is uniquely indicated for reducing the risk for suicide. The ability of the atypical APDs to improve cognition and negative symptoms in some patients together with lower propensity to cause tardive dyskinesia (an underappreciated advantage) leads to better overall outcomes. These advantages of the atypical APDs in efficacy and safety are due, in part, to initiation of synaptic plasticity via direct and indirect effects of the atypical APDs on a variety of proteins, especially G proteins, and release of neurotrophins (e.g., brain-derived neurotrophic factor). The typical APDs beneficial effects on psychosis are mainly the result of D2 receptor blockade, which can be associated with serious side effects and lack of tolerability.
Collapse
|
48
|
Meftah A, Hasegawa H, Kantrowitz JT. D-Serine: A Cross Species Review of Safety. Front Psychiatry 2021; 12:726365. [PMID: 34447324 PMCID: PMC8384137 DOI: 10.3389/fpsyt.2021.726365] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 07/13/2021] [Indexed: 12/18/2022] Open
Abstract
Background:D-Serine, a direct, full agonist at the D-serine/glycine modulatory site of the N-methyl-D-aspartate-type glutamate receptors (NMDAR), has been assessed as a treatment for multiple psychiatric and neurological conditions. Based on studies in rats, concerns of nephrotoxicity have limited D-serine research in humans, particularly using high doses. A review of D-serine's safety is timely and pertinent, as D-serine remains under active study for schizophrenia, both directly (R61 MH116093) and indirectly through D-amino acid oxidase (DAAO) inhibitors. The principal focus is on nephrotoxicity, but safety in other physiologic and pathophysiologic systems are also reviewed. Methods: Using the search terms "D-serine," "D-serine and schizophrenia," "D-serine and safety," "D-serine and nephrotoxicity" in PubMed, we conducted a systematic review on D-serine safety. D-serine physiology, dose-response and efficacy in clinical studies and dAAO inhibitor safety is also discussed. Results: When D-serine doses >500 mg/kg are used in rats, nephrotoxicity, manifesting as an acute tubular necrosis syndrome, seen within hours of administration is highly common, if not universal. In other species, however, D-serine induced nephrotoxicity has not been reported, even in other rodent species such as mice and rabbits. Even in rats, D--serine related toxicity is dose dependent and reversible; and does not appear to be present in rats at doses producing an acute Cmax of <2,000 nmol/mL. For comparison, the Cmax of D-serine 120 mg/kg, the highest dose tested in humans, is ~500 nmol/mL in acute dosing. Across all published human studies, only one subject has been reported to have abnormal renal values related to D-serine treatment. This abnormality did not clearly map on to the acute tubular necrosis syndrome seen in rats, and fully resolved within a few days of stopping treatment. DAAO inhibitors may be nephroprotective. D-Serine may have a physiologic role in metabolic, extra-pyramidal, cardiac and other systems, but no other clinically significant safety concerns are revealed in the literature. Conclusions: Even before considering human to rat differences in renal physiology, using current FDA guided monitoring paradigms, D-serine appears safe at currently studied maximal doses, with potential safety in combination with DAAO inhibitors.
Collapse
Affiliation(s)
- Amir Meftah
- College of Physicians and Surgeons, Columbia University, New York City, NY, United States
- New York State Psychiatric Institute, New York City, NY, United States
| | - Hiroshi Hasegawa
- Department of Pathophysiology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Joshua T. Kantrowitz
- College of Physicians and Surgeons, Columbia University, New York City, NY, United States
- New York State Psychiatric Institute, New York City, NY, United States
- Nathan Kline Institute, Orangeburg, NY, United States
| |
Collapse
|
49
|
Cruz G, Grent-'t-Jong T, Krishnadas R, Palva JM, Palva S, Uhlhaas PJ. Long range temporal correlations (LRTCs) in MEG-data during emerging psychosis: Relationship to symptoms, medication-status and clinical trajectory. Neuroimage Clin 2021; 31:102722. [PMID: 34130193 PMCID: PMC8209846 DOI: 10.1016/j.nicl.2021.102722] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/30/2021] [Accepted: 06/04/2021] [Indexed: 12/24/2022]
Abstract
Long-Range Temporal Correlations (LRTCs) index the capacity of the brain to optimally process information. Previous research has shown that patients with chronic schizophrenia present altered LRTCs at alpha and beta oscillations. However, it is currently unclear at which stage of schizophrenia aberrant LRTCs emerge. To address this question, we investigated LRTCs in resting-state magnetoencephalographic (MEG) recordings obtained from patients with affective disorders and substance abuse (clinically at low-risk of psychosis, CHR-N), patients at clinical high-risk of psychosis (CHR-P) (n = 115), as well as patients with a first episode (FEP) (n = 25). Matched healthy controls (n = 47) served as comparison group. LRTCs were obtained for frequencies from 4 to 40 Hz and correlated with clinical and neuropsychological data. In addition, we examined the relationship between LRTCs and transition to psychosis in CHR-P participants, and the relationship between LRTC and antipsychotic medication in FEP participants. Our results show that participants from the clinical groups have similar LRTCs to controls. In addition, LRTCs did not correlate with clinical and neurocognitive variables across participants nor did LRTCs predict transition to psychosis. Therefore, impaired LRTCs do not reflect a feature in the clinical trajectory of psychosis. Nevertheless, reduced LRTCs in the beta-band over posterior sensors of medicated FEP participants indicate that altered LRTCs may appear at the onset of the illness. Future studies are needed to elucidate the role of anti-psychotic medication in altered LRTCs.
Collapse
Affiliation(s)
- Gabriela Cruz
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, United Kingdom.
| | - Tineke Grent-'t-Jong
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, United Kingdom; Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany
| | - Rajeev Krishnadas
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, United Kingdom
| | - J Matias Palva
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, United Kingdom; Neuroscience Centre, Helsinki Institute of Life Science, University of Helsinki, Finland; Department of Neuroscience and Biomedical Engineering, Aalto University, Finland
| | - Satu Palva
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, United Kingdom; Neuroscience Centre, Helsinki Institute of Life Science, University of Helsinki, Finland
| | - Peter J Uhlhaas
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, United Kingdom; Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany
| |
Collapse
|
50
|
Blacker KJ, Seech TR, Funke ME, Kinney MJ. Deficits in Visual Processing During Hypoxia as Evidenced by Visual Mismatch Negativity. Aerosp Med Hum Perform 2021; 92:326-332. [PMID: 33875065 DOI: 10.3357/amhp.5735.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
INTRODUCTION: Hypoxia is an ever-present threat in tactical aviation and gained recent attention due to its putative role in physiological episodes. Previous work has demonstrated that hypoxia negatively impacts a variety of sensory, cognitive, and motor systems. In particular, the visual system is one of the earliest systems affected by hypoxia. While the majority of previous studies have relied on self-report and behavioral testing, the use of event-related potentials as a novel tool to monitor responses to low oxygen in humans has recently been investigated. Specifically, ERP components that are evoked passively in response to unattended changes in background sensory stimulation have been explored.METHOD: Subjects (N 28) completed a continuous visuomotor tracking task while EEG was recorded. During the tracking task, a series of standard color checkerboard patterns were presented in the periphery while occasionally a deviant color checkerboard was presented. The visual mismatch negativity (MMN) component was assessed in response to the deviant compared to the standard stimuli. Subjects completed two sessions in counterbalanced order that only differed by the oxygen concentration breathed (10.6% vs. 20.4%).RESULTS: Results demonstrated a significant reduction in the amplitude of the visual MMN under hypoxic compared to normoxic conditions, showing a 50% reduction in amplitude during hypoxia. Our results suggest that during low-oxygen exposure the ability to detect environmental changes and process sensory information is impaired.DISCUSSION: The visual MMN may represent an early and reliable predictor of sensory and cognitive deficits during hypoxia exposure, which may be of great use to the aviation community.Blacker KJ, Seech TR, Funke ME, Kinney MJ. Deficits in visual processing during hypoxia as evidenced by visual mismatch negativity. Aerosp Med Hum Perform. 2021; 92(5):326332.
Collapse
|