1
|
Poggi G, Treccani G, von der Bey M, Tanti A, Schmeisser MJ, Müller M. Canonical and non-canonical roles of oligodendrocyte precursor cells in mental disorders. NPJ MENTAL HEALTH RESEARCH 2025; 4:19. [PMID: 40374740 DOI: 10.1038/s44184-025-00133-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/29/2025] [Indexed: 05/18/2025]
Abstract
Psychiatric research has shifted from a neuroncentric view to understanding mental disorders as disturbances of heterogeneous brain networks. Oligodendrocyte precursor cells (OPCs)- actively involved in the modulation of neuronal functions - are altered in psychiatric patients, but the extent and related consequences are unclear. This review explores canonical and non-canonical OPC-related pathways in schizophrenia, bipolar disorder, post-traumatic stress disorder, and depression in humans, highlighting potential mechanisms shared across diagnostic entities.
Collapse
Affiliation(s)
- Giulia Poggi
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| | - Giulia Treccani
- Department of Systemic Neuroscience Institute of Anatomy and Cell Biology, Philipps Universität Marburg, Marburg, Germany
| | - Martina von der Bey
- Molecular and Translational Neuroscience, Department of Neurology, University Hospital Ulm, Ulm, Germany
| | - Arnaud Tanti
- Inserm, UMR 1253, iBrain, Université de Tours, Tours, France
| | - Michael J Schmeisser
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Focus Program Translational Neurosciences, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Marianne Müller
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Leibniz Institute for Resilience Research, Mainz, Germany
| |
Collapse
|
2
|
Perrotta C, Carnovale C, Pozzi M, De Palma C, Cervia D, Nobile M, Clementi E. Antipsychotics and dietary interventions: Pharmacodynamics, pharmacokinetics, and synergisms in therapy. Pharmacol Rev 2025; 77:100061. [PMID: 40412008 DOI: 10.1016/j.pharmr.2025.100061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/18/2025] [Accepted: 04/19/2025] [Indexed: 05/27/2025] Open
Abstract
Antipsychotic (AP) medications are the primary treatment for severe mental illnesses, including schizophrenia and severe mood disorders. APs are currently categorized into typical or first-generation APs and atypical or second-generation APs. Although both first-generation and second-generation APs are considered effective in treating psychotic symptoms in severe mental disorders, they differ in their mechanisms, treatment strategies, and side effect profiles. Because of their potential motor and metabolic side effects, which often compromise patient adherence and clinical outcomes, whether and how to use APs remains controversial. The use of dietary interventions in combination with APs is emerging as a viable strategy to reduce AP adverse effects while maintaining their efficacy and enhance patient adherence to treatment. In contrast to drugs that possess a well defined molecular mechanism of action, dietary interventions act in pleiotropic ways by nature. While providing a holistic approach to patient care this pleiotropy needs to be analyzed and systematized to enhance the efficacy and safety of the combination of them with APs. Guidelines for this type of treatment are still needed. In this review, we explore the pharmacological properties, therapeutic applications, and limitations of APs, and discuss the potential benefits and limitations of those dietary interventions that are employed to improve the efficacy and counteract side effects of APs discussing also their mechanisms of action. Finally, we critically discuss the main results of clinical studies combining APs and dietary interventions and provide a view on future directions in terms of research and clinical use of these combinations. SIGNIFICANCE STATEMENT: Antipsychotic drugs are useful in a variety of psychiatric conditions, yet their use is hampered by issues of efficacy and safety. An important step toward therapy optimization is their use in combination with dietary interventions (ie, dietary supplements and nutraceuticals) that have shown promising results in clinical trials.
Collapse
Affiliation(s)
- Cristiana Perrotta
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milano, Italy
| | - Carla Carnovale
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milano, Italy
| | - Marco Pozzi
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Clara De Palma
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano, Segrate, Italy
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Maria Nobile
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy.
| | - Emilio Clementi
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy; Department of Biomedical and Clinical Sciences (DIBIC), ASST Fatebenefratelli-Sacco Hospital, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
3
|
Shao X, Volk L. PICK1 links KIBRA and AMPA receptor subunit GluA2 in coiled-coil-driven supramolecular complexes. J Biol Chem 2025; 301:108397. [PMID: 40074086 DOI: 10.1016/j.jbc.2025.108397] [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: 11/08/2023] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
The human memory-associated protein KIBRA regulates synaptic plasticity and trafficking of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type glutamate receptors, and is implicated in multiple neuropsychiatric and cognitive disorders. How KIBRA forms complexes with and regulates AMPA receptors remains unclear. Here, we show that KIBRA does not interact directly with the AMPA receptor subunit GluA2, but that protein interacting with C kinase 1 (PICK1), a key regulator of AMPA receptor trafficking, can serve as a bridge between KIBRA and GluA2. In contrast, KIBRA can form a complex with GluA1 independent of PICK1. We identified structural determinants of KIBRA-PICK1-AMPAR complexes by investigating interactions and cellular expression patterns of different combinations of KIBRA and PICK1 domain mutants. We find that the PICK1 BAR domain, a coiled-coil structure, is sufficient for interaction with KIBRA, whereas mutation of the PICK1 BAR domain disrupts KIBRA-PICK1-GluA2 complex formation. In addition, KIBRA recruits PICK1 into large supramolecular complexes, a process which requires KIBRA coiled-coil domains. These findings reveal molecular mechanisms by which KIBRA can organize key synaptic signaling complexes.
Collapse
Affiliation(s)
- Xin Shao
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Lenora Volk
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas, USA; Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, Texas, USA; Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas, USA; Peter O'Donnell Jr Brain Institute Investigator, UT Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
4
|
Bernstein HG, Nussbaumer M, Vasilevska V, Dobrowolny H, Nickl-Jockschat T, Guest PC, Steiner J. Glial cell deficits are a key feature of schizophrenia: implications for neuronal circuit maintenance and histological differentiation from classical neurodegeneration. Mol Psychiatry 2025; 30:1102-1116. [PMID: 39639174 PMCID: PMC11835740 DOI: 10.1038/s41380-024-02861-6] [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: 07/13/2024] [Revised: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
Dysfunctional glial cells play a pre-eminent role in schizophrenia pathophysiology. Post-mortem studies have provided evidence for significantly decreased glial cell numbers in different brain regions of individuals with schizophrenia. Reduced glial cell numbers are most pronounced in oligodendroglia, but reduced astrocyte cell densities have also been reported. This review highlights that oligo- and astroglial deficits are a key histopathological feature in schizophrenia, distinct from typical changes seen in neurodegenerative disorders. Significant deficits of oligodendrocytes in schizophrenia may arise in two ways: (i) demise of mature functionally compromised oligodendrocytes; and (ii) lack of mature oligodendrocytes due to failed maturation of progenitor cells. We also analyse in detail the controversy regarding deficits of astrocytes. Regardless of their origin, glial cell deficits have several pathophysiological consequences. Among these, myelination deficits due to a reduced number of oligodendrocytes may be the most important factor, resulting in the disconnectivity between neurons and different brain regions observed in schizophrenia. When glial cells die, it appears to be through degeneration, a process which is basically reversible. Thus, therapeutic interventions that (i) help rescue glial cells (ii) or improve their maturation might be a viable option. Since antipsychotic treatment alone does not seem to prevent glial cell loss or maturation deficits, there is intense search for new therapeutic options. Current proposals range from the application of antidepressants and other chemical agents as well as physical exercise to engrafting healthy glial cells into brains of schizophrenia patients.
Collapse
Affiliation(s)
- Hans-Gert Bernstein
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Madeleine Nussbaumer
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Veronika Vasilevska
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Department of Radiotherapy, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Henrik Dobrowolny
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Thomas Nickl-Jockschat
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa, IA, USA
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- German Center for Mental Health (DZPG), Partner Site Halle-Jena-Magdeburg, Magdeburg, Germany
| | - Paul C Guest
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Johann Steiner
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.
- German Center for Mental Health (DZPG), Partner Site Halle-Jena-Magdeburg, Magdeburg, Germany.
| |
Collapse
|
5
|
D'Addario C, Di Bartolomeo M. Epigenetic Control in Schizophrenia. Subcell Biochem 2025; 108:191-215. [PMID: 39820863 DOI: 10.1007/978-3-031-75980-2_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Schizophrenia is a severe and complex psychiatric condition ranking among the top 15 leading causes of disability worldwide. Despite the well-established heritability component, a complex interplay between genetic and environmental risk factors plays a key role in the development of schizophrenia and psychotic disorders in general. This chapter covers all the clinical evidence showing how the analysis of the epigenetic modulation in schizophrenia might be relevant to understand the pathogenesis of schizophrenia as well as potentially useful to develop new pharmacotherapies.
Collapse
Affiliation(s)
- Claudio D'Addario
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy.
| | - Martina Di Bartolomeo
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| |
Collapse
|
6
|
Tang W, Wang Q, Sun M, Liu C, Huang Y, Zhou M, Zhang X, Meng Z, Zhang J. The gut microbiota-oligodendrocyte axis: A promising pathway for modulating oligodendrocyte homeostasis and demyelination-associated disorders. Life Sci 2024; 354:122952. [PMID: 39127317 DOI: 10.1016/j.lfs.2024.122952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
The bidirectional regulation between the gut microbiota and brain, known as gut-brain axis, has received significant attention. The myelin sheath, produced by oligodendrocytes or Schwann cells, is essential for efficient nervous signal transmission and the maintenance of brain function. Growing evidence shows that both oligodendrogenesis and myelination are modulated by gut microbiota and its metabolites, and when dysbiosis occurs, changes in the microbiota composition and/or associated metabolites may impact developmental myelination and the occurrence of neurodevelopmental disabilities. Although the link between the microbiota and demyelinating disease such as multiple sclerosis has been extensively studied, our knowledge about the role of the microbiota in other myelin-related disorders, such as neurodegenerative diseases, is limited. Mechanistically, the microbiota-oligodendrocyte axis is primarily mediated by factors such as inflammation, the vagus nerve, endocrine hormones, and microbiota metabolites as evidenced by metagenomics, metabolomics, vagotomy, and morphological and molecular approaches. Treatments targeting this axis include probiotics, prebiotics, microbial metabolites, herbal bioactive compounds, and specific dietary management. In addition to the commonly used approaches, viral vector-mediated tracing and gene manipulation, integrated multiomics and multicenter clinical trials will greatly promote the mechanistic and interventional studies and ultimately, the development of new preventive and therapeutic strategies against gut-oligodendrocyte axis-mediated brain impairments. Interestingly, recent findings showed that microbiota dysbiosis can be induced by hippocampal myelin damage and is reversible by myelin-targeted drugs, which provides new insights into understanding how hippocampus-based functional impairment (such as in neurodegenerative Alzheimer's disease) regulates the peripheral homeostasis of microbiota and associated systemic disorders.
Collapse
Affiliation(s)
- Wen Tang
- Department of Gastroenterology, Chongqing Western Hospital, Chongqing 400052, China
| | - Qi Wang
- Department of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Mingguang Sun
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China; Department of Neurology, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, Beijing University of Chinese Medicine, Beijing 100853, China
| | - Chang''e Liu
- Department of Nutrition, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Yonghua Huang
- Department of Neurology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Maohu Zhou
- Department of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Xuan Zhang
- Department of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Zhaoyou Meng
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China.
| | - Jiqiang Zhang
- Department of Neurobiology, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
7
|
Bergstrom JJD, Fu MM. Dysregulation of myelination-related genes in schizophrenia. J Neurochem 2024; 168:2227-2242. [PMID: 39086020 PMCID: PMC11449665 DOI: 10.1111/jnc.16152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 08/02/2024]
Abstract
Schizophrenic individuals display disrupted myelination patterns, altered oligodendrocyte distribution, and abnormal oligodendrocyte morphology. Schizophrenia is linked with dysregulation of a variety of genes involved in oligodendrocyte function and myelin production. Single-nucleotide polymorphisms (SNPs) and rare mutations in myelination-related genes are observed in certain schizophrenic populations, representing potential genetic risk factors. Downregulation of myelination-related RNAs and proteins, particularly in frontal and limbic regions, is consistently associated with the disorder across multiple studies. These findings support the notion that disruptions in myelination may contribute to the cognitive and behavioral impairments experienced in schizophrenia, although further evidence of causation is needed.
Collapse
Affiliation(s)
| | - Meng-Meng Fu
- Department of Molecular and Cell Biology, UC Berkeley, Berkeley, California, USA
| |
Collapse
|
8
|
Jørgensen KN, Nerland S, Slapø NB, Norbom LB, Mørch-Johnsen L, Wortinger LA, Barth C, Andreou D, Maximov II, Geier OM, Andreassen OA, Jönsson EG, Agartz I. Assessing regional intracortical myelination in schizophrenia spectrum and bipolar disorders using the optimized T1w/T2w-ratio. Psychol Med 2024; 54:2369-2379. [PMID: 38563302 DOI: 10.1017/s0033291724000503] [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: 04/04/2024]
Abstract
BACKGROUND Dysmyelination could be part of the pathophysiology of schizophrenia spectrum (SCZ) and bipolar disorders (BPD), yet few studies have examined myelination of the cerebral cortex. The ratio of T1- and T2-weighted magnetic resonance images (MRI) correlates with intracortical myelin. We investigated the T1w/T2w-ratio and its age trajectories in patients and healthy controls (CTR) and explored associations with antipsychotic medication use and psychotic symptoms. METHODS Patients with SCZ (n = 64; mean age = 30.4 years, s.d. = 9.8), BPD (n = 91; mean age 31.0 years, s.d. = 10.2), and CTR (n = 155; mean age = 31.9 years, s.d. = 9.1) who participated in the TOP study (NORMENT, University of Oslo, Norway) were clinically assessed and scanned using a General Electric 3 T MRI system. T1w/T2w-ratio images were computed using an optimized pipeline with intensity normalization and field inhomogeneity correction. Vertex-wise regression models were used to compare groups and examine group × age interactions. In regions showing significant differences, we explored associations with antipsychotic medication use and psychotic symptoms. RESULTS No main effect of diagnosis was found. However, age slopes of the T1w/T2w-ratio differed significantly between SCZ and CTR, predominantly in frontal and temporal lobe regions: Lower T1w/T2w-ratio values with higher age were found in CTR, but not in SCZ. Follow-up analyses revealed a more positive age slope in patients who were using antipsychotics and patients using higher chlorpromazine-equivalent doses. CONCLUSIONS While we found no evidence of reduced intracortical myelin in SCZ or BPD relative to CTR, different regional age trajectories in SCZ may suggest a promyelinating effect of antipsychotic medication.
Collapse
Affiliation(s)
- Kjetil Nordbø Jørgensen
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry, Telemark Hospital, Skien, Norway
| | - Stener Nerland
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Nora Berz Slapø
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Linn B Norbom
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Department of Psychology, PROMENTA Research Center, University of Oslo, Oslo, Norway
| | - Lynn Mørch-Johnsen
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry & Department of Clinical Research, Østfold Hospital, Grålum, Norway
| | - Laura Anne Wortinger
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Claudia Barth
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Dimitrios Andreou
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden
| | - Ivan I Maximov
- Department of Psychology, University of Oslo, Oslo, Norway
- The Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Health and Functioning, Western Norway University of Applied Sciences, Bergen, Norway
| | - Oliver M Geier
- Department of Physics and Computational Radiology, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Ole A Andreassen
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- The Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Erik G Jönsson
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden
| | - Ingrid Agartz
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden
| |
Collapse
|
9
|
Oliver D, Chesney E, Cullen AE, Davies C, Englund A, Gifford G, Kerins S, Lalousis PA, Logeswaran Y, Merritt K, Zahid U, Crossley NA, McCutcheon RA, McGuire P, Fusar-Poli P. Exploring causal mechanisms of psychosis risk. Neurosci Biobehav Rev 2024; 162:105699. [PMID: 38710421 PMCID: PMC11250118 DOI: 10.1016/j.neubiorev.2024.105699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/17/2024] [Accepted: 04/28/2024] [Indexed: 05/08/2024]
Abstract
Robust epidemiological evidence of risk and protective factors for psychosis is essential to inform preventive interventions. Previous evidence syntheses have classified these risk and protective factors according to their strength of association with psychosis. In this critical review we appraise the distinct and overlapping mechanisms of 25 key environmental risk factors for psychosis, and link these to mechanistic pathways that may contribute to neurochemical alterations hypothesised to underlie psychotic symptoms. We then discuss the implications of our findings for future research, specifically considering interactions between factors, exploring universal and subgroup-specific factors, improving understanding of temporality and risk dynamics, standardising operationalisation and measurement of risk and protective factors, and developing preventive interventions targeting risk and protective factors.
Collapse
Affiliation(s)
- Dominic Oliver
- Department of Psychiatry, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Oxford, UK; OPEN Early Detection Service, Oxford Health NHS Foundation Trust, Oxford, UK; Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
| | - Edward Chesney
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Addictions Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 4 Windsor Walk, London SE5 8AF, UK
| | - Alexis E Cullen
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Clinical Neuroscience, Karolinska Institutet, Sweden
| | - Cathy Davies
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Amir Englund
- Addictions Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 4 Windsor Walk, London SE5 8AF, UK
| | - George Gifford
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Sarah Kerins
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Paris Alexandros Lalousis
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Yanakan Logeswaran
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Biostatistics & Health Informatics, King's College London, London, UK
| | - Kate Merritt
- Division of Psychiatry, Institute of Mental Health, UCL, London, UK
| | - Uzma Zahid
- Department of Psychology, King's College London, London, UK
| | - Nicolas A Crossley
- Department of Psychiatry, University of Oxford, Oxford, UK; Department of Psychiatry, School of Medicine, Pontificia Universidad Católica de Chile, Chile
| | - Robert A McCutcheon
- Department of Psychiatry, University of Oxford, Oxford, UK; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Oxford Health NHS Foundation Trust, Oxford, UK
| | - Philip McGuire
- Department of Psychiatry, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Oxford, UK; OPEN Early Detection Service, Oxford Health NHS Foundation Trust, Oxford, UK
| | - Paolo Fusar-Poli
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; OASIS Service, South London and Maudsley NHS Foundation Trust, London SE11 5DL, UK
| |
Collapse
|
10
|
Chandra J. The potential role of the p75 receptor in schizophrenia: neuroimmunomodulation and making life or death decisions. Brain Behav Immun Health 2024; 38:100796. [PMID: 38813083 PMCID: PMC11134531 DOI: 10.1016/j.bbih.2024.100796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/06/2024] [Accepted: 05/12/2024] [Indexed: 05/31/2024] Open
Abstract
The nerve growth factor receptor, also referred to as tumour necrosis factor II and the p75 neurotrophin receptor (p75), serves pleiotropic functions in both the peripheral and central nervous system, involving modulation of immune responses, cell survival and cell death signalling in response to multiple ligands including cytokines such as TNFα, as well as proneurotrophins and mature neurotrophins. Whilst in vitro and in vivo studies have characterised various responses of the p75 receptor in isolated conditions, it remains unclear whether the p75 receptor serves to provide neuroprotection or contributes to neurotoxicity in neuroinflammatory and neurotrophin-deficit conditions, such as those presenting in schizophrenia. The purpose of this mini-review is to characterise the potential signalling mechanisms of the p75 receptor respective to neuropathological changes prevailing in schizophrenia to ultimately propose how specific functions of the receptor may underlie altered levels of p75 in specific cell types. On the basis of this evaluation, this mini-review aims to promote avenues for future research in utilising the therapeutic potential of ligands for the p75 receptor in psychiatric disorders, whereby heightened inflammation and reductions in trophic signalling mechanisms coalesce in the brain, potentially resulting in tissue damage.
Collapse
Affiliation(s)
- Jessica Chandra
- Neuroscience Research Australia, University of New South Wales, Sydney, Australia
| |
Collapse
|
11
|
Singh S, Sutkus L, Li Z, Baker S, Bear J, Dilger RN, Miller DJ. Standardization of a silver stain to reveal mesoscale myelin in histological preparations of the mammalian brain. J Neurosci Methods 2024; 407:110139. [PMID: 38626852 DOI: 10.1016/j.jneumeth.2024.110139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/26/2024] [Accepted: 04/12/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND The brain is built of neurons supported by myelin, a fatty substance that improves cellular communication. Noninvasive magnetic resonance imaging (MRI) is now able to measure brain structure like myelin and requires histological validation. NEW METHOD Here we present work in small and large biomedical model mammals to standardize a silver impregnation method as a high-throughput histological myelin visualization procedure. Specifically, we built a new staining well plate to increase batch size, and then systematically varied the staining and clearing cycles to describe the staining response curve across taxa and conditions. We compared tissues fixed by immersion or perfusion, mounted versus free-floating, and cut as thicker or thinner slices, with two-weeks of post-fixation. RESULTS The staining response curves show optimal staining with a single exposure across taxa when incubation and clearing epochs are held to within 3-9 min. We show that clearing was slower in mounted vs free-floating tissue, and that staining was faster and caused fracturing earlier in thinner sliced and smaller volumes of tissue. COMPARISON WITH EXISTING METHODS We developed a batch processing approach to increase throughput while ensuring reproducibility and demonstrate the optimal conditions for fine myelinated fiber morphology visualization with short cycles (<9 minutes). CONCLUSIONS We present our optimized protocol to reveal mesoscale neuroanatomical myelin content in histology across mammals. This standard staining procedure will facilitate multiscale analyses of myelin content across development as well as in the presence of injury or disease.
Collapse
Affiliation(s)
- S Singh
- Department of Evolution, Ecology, and Behavior, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - L Sutkus
- Neuroscience Program, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - Z Li
- Neuroscience Program, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - S Baker
- Machine Shop, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - J Bear
- Machine Shop, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - R N Dilger
- Department of Animal Sciences, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America; Neuroscience Program, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America; Beckman Institute for Advanced Science and Technology, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - D J Miller
- Department of Evolution, Ecology, and Behavior, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America; Neuroscience Program, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America; Beckman Institute for Advanced Science and Technology, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America.
| |
Collapse
|
12
|
Song J, Saglam A, Zuchero JB, Buch VP. Translating Molecular Approaches to Oligodendrocyte-Mediated Neurological Circuit Modulation. Brain Sci 2024; 14:648. [PMID: 39061389 PMCID: PMC11275066 DOI: 10.3390/brainsci14070648] [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: 06/14/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
The central nervous system (CNS) exhibits remarkable adaptability throughout life, enabled by intricate interactions between neurons and glial cells, in particular, oligodendrocytes (OLs) and oligodendrocyte precursor cells (OPCs). This adaptability is pivotal for learning and memory, with OLs and OPCs playing a crucial role in neural circuit development, synaptic modulation, and myelination dynamics. Myelination by OLs not only supports axonal conduction but also undergoes adaptive modifications in response to neuronal activity, which is vital for cognitive processing and memory functions. This review discusses how these cellular interactions and myelin dynamics are implicated in various neurocircuit diseases and disorders such as epilepsy, gliomas, and psychiatric conditions, focusing on how maladaptive changes contribute to disease pathology and influence clinical outcomes. It also covers the potential for new diagnostics and therapeutic approaches, including pharmacological strategies and emerging biomarkers in oligodendrocyte functions and myelination processes. The evidence supports a fundamental role for myelin plasticity and oligodendrocyte functionality in synchronizing neural activity and high-level cognitive functions, offering promising avenues for targeted interventions in CNS disorders.
Collapse
Affiliation(s)
- Jingwei Song
- Medical Scientist Training Program, School of Medicine, Stanford University, Stanford, CA 94305, USA;
| | - Aybike Saglam
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; (A.S.); (J.B.Z.)
| | - J. Bradley Zuchero
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; (A.S.); (J.B.Z.)
| | - Vivek P. Buch
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; (A.S.); (J.B.Z.)
| |
Collapse
|
13
|
Hill MD, Gill SS, Le-Niculescu H, MacKie O, Bhagar R, Roseberry K, Murray OK, Dainton HD, Wolf SK, Shekhar A, Kurian SM, Niculescu AB. Precision medicine for psychotic disorders: objective assessment, risk prediction, and pharmacogenomics. Mol Psychiatry 2024; 29:1528-1549. [PMID: 38326562 DOI: 10.1038/s41380-024-02433-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/16/2023] [Accepted: 01/15/2024] [Indexed: 02/09/2024]
Abstract
Psychosis occurs inside the brain, but may have external manifestations (peripheral molecular biomarkers, behaviors) that can be objectively and quantitatively measured. Blood biomarkers that track core psychotic manifestations such as hallucinations and delusions could provide a window into the biology of psychosis, as well as help with diagnosis and treatment. We endeavored to identify objective blood gene expression biomarkers for hallucinations and delusions, using a stepwise discovery, prioritization, validation, and testing in independent cohorts design. We were successful in identifying biomarkers that were predictive of high hallucinations and of high delusions states, and of future psychiatric hospitalizations related to them, more so when personalized by gender and diagnosis. Top biomarkers for hallucinations that survived discovery, prioritization, validation and testing include PPP3CB, DLG1, ENPP2, ZEB2, and RTN4. Top biomarkers for delusions include AUTS2, MACROD2, NR4A2, PDE4D, PDP1, and RORA. The top biological pathways uncovered by our work are glutamatergic synapse for hallucinations, as well as Rap1 signaling for delusions. Some of the biomarkers are targets of existing drugs, of potential utility in pharmacogenomics approaches (matching patients to medications, monitoring response to treatment). The top biomarkers gene expression signatures through bioinformatic analyses suggested a prioritization of existing medications such as clozapine and risperidone, as well as of lithium, fluoxetine, valproate, and the nutraceuticals omega-3 fatty acids and magnesium. Finally, we provide an example of how a personalized laboratory report for doctors would look. Overall, our work provides advances for the improved diagnosis and treatment for schizophrenia and other psychotic disorders.
Collapse
Affiliation(s)
- M D Hill
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - S S Gill
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - H Le-Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - O MacKie
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - R Bhagar
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - K Roseberry
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - O K Murray
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - H D Dainton
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurology, Medical University of South Carolina, Charleston, SC, USA
| | - S K Wolf
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurology, Ohio State University Medical Center, Columbus, OH, USA
| | - A Shekhar
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Office of the Dean, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - A B Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indianapolis VA Medical Center, Indianapolis, IN, USA.
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
14
|
Vieira R, Mariani JN, Huynh NPT, Stephensen HJT, Solly R, Tate A, Schanz S, Cotrupi N, Mousaei M, Sporring J, Benraiss A, Goldman SA. Young glial progenitor cells competitively replace aged and diseased human glia in the adult chimeric mouse brain. Nat Biotechnol 2024; 42:719-730. [PMID: 37460676 PMCID: PMC11098747 DOI: 10.1038/s41587-023-01798-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 04/20/2023] [Indexed: 08/26/2023]
Abstract
Competition among adult brain cells has not been extensively researched. To investigate whether healthy glia can outcompete diseased human glia in the adult forebrain, we engrafted wild-type (WT) human glial progenitor cells (hGPCs) produced from human embryonic stem cells into the striata of adult mice that had been neonatally chimerized with mutant Huntingtin (mHTT)-expressing hGPCs. The WT hGPCs outcompeted and ultimately eliminated their human Huntington's disease (HD) counterparts, repopulating the host striata with healthy glia. Single-cell RNA sequencing revealed that WT hGPCs acquired a YAP1/MYC/E2F-defined dominant competitor phenotype upon interaction with the host HD glia. WT hGPCs also outcompeted older resident isogenic WT cells that had been transplanted neonatally, suggesting that competitive success depended primarily on the relative ages of competing populations, rather than on the presence of mHTT. These data indicate that aged and diseased human glia may be broadly replaced in adult brain by younger healthy hGPCs, suggesting a therapeutic strategy for the replacement of aged and diseased human glia.
Collapse
Affiliation(s)
- Ricardo Vieira
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - John N Mariani
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Nguyen P T Huynh
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
- Sana Biotechnology, Inc, Cambridge, MA, USA
| | - Hans J T Stephensen
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
- Department of Computer Science, University of Copenhagen Faculty of Science, Copenhagen, Denmark
| | - Renee Solly
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
- Sana Biotechnology, Inc, Cambridge, MA, USA
| | - Ashley Tate
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
- Sana Biotechnology, Inc, Cambridge, MA, USA
| | - Steven Schanz
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Natasha Cotrupi
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Marzieh Mousaei
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Jon Sporring
- Department of Computer Science, University of Copenhagen Faculty of Science, Copenhagen, Denmark
| | - Abdellatif Benraiss
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark.
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA.
- Sana Biotechnology, Inc, Cambridge, MA, USA.
| |
Collapse
|
15
|
Rivera AD, Normanton JR, Butt AM, Azim K. The Genomic Intersection of Oligodendrocyte Dynamics in Schizophrenia and Aging Unravels Novel Pathological Mechanisms and Therapeutic Potentials. Int J Mol Sci 2024; 25:4452. [PMID: 38674040 PMCID: PMC11050044 DOI: 10.3390/ijms25084452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/28/2024] Open
Abstract
Schizophrenia is a significant worldwide health concern, affecting over 20 million individuals and contributing to a potential reduction in life expectancy by up to 14.5 years. Despite its profound impact, the precise pathological mechanisms underlying schizophrenia continue to remain enigmatic, with previous research yielding diverse and occasionally conflicting findings. Nonetheless, one consistently observed phenomenon in brain imaging studies of schizophrenia patients is the disruption of white matter, the bundles of myelinated axons that provide connectivity and rapid signalling between brain regions. Myelin is produced by specialised glial cells known as oligodendrocytes, which have been shown to be disrupted in post-mortem analyses of schizophrenia patients. Oligodendrocytes are generated throughout life by a major population of oligodendrocyte progenitor cells (OPC), which are essential for white matter health and plasticity. Notably, a decline in a specific subpopulation of OPC has been identified as a principal factor in oligodendrocyte disruption and white matter loss in the aging brain, suggesting this may also be a factor in schizophrenia. In this review, we analysed genomic databases to pinpoint intersections between aging and schizophrenia and identify shared mechanisms of white matter disruption and cognitive dysfunction.
Collapse
Affiliation(s)
- Andrea D. Rivera
- Department of Neuroscience, Institute of Human Anatomy, University of Padova, Via A. Gabelli 65, 35127 Padua, Italy;
| | - John R. Normanton
- GliaGenesis Limited, Orchard Lea, Horns Lane, Oxfordshire, Witney OX29 8NH, UK; (J.R.N.); (K.A.)
| | - Arthur M. Butt
- GliaGenesis Limited, Orchard Lea, Horns Lane, Oxfordshire, Witney OX29 8NH, UK; (J.R.N.); (K.A.)
- School of Pharmacy and Biomedical Science, University of Portsmouth, Hampshire PO1 2UP, UK
| | - Kasum Azim
- GliaGenesis Limited, Orchard Lea, Horns Lane, Oxfordshire, Witney OX29 8NH, UK; (J.R.N.); (K.A.)
- Independent Data Lab UG, Frauenmantelanger 31, 80937 Munich, Germany
| |
Collapse
|
16
|
Shao X, Volk L. PICK1 links KIBRA and AMPA receptors in coiled-coil-driven supramolecular complexes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584494. [PMID: 38558978 PMCID: PMC10980033 DOI: 10.1101/2024.03.12.584494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The human memory-associated protein KIBRA regulates synaptic plasticity and trafficking of AMPA-type glutamate receptors, and is implicated in multiple neuropsychiatric and cognitive disorders. How KIBRA forms complexes with and regulates AMPA receptors remains unclear. Here, we show that KIBRA does not interact directly with the AMPA receptor subunit GluA2, but that PICK1, a key regulator of AMPA receptor trafficking, can serve as a bridge between KIBRA and GluA2. We identified structural determinants of KIBRA-PICK1-AMPAR complexes by investigating interactions and cellular expression patterns of different combinations of KIBRA and PICK1 domain mutants. We find that the PICK1 BAR domain, a coiled-coil structure, is sufficient for interaction with KIBRA, whereas mutation of the BAR domain disrupts KIBRA-PICK1-GluA2 complex formation. In addition, KIBRA recruits PICK1 into large supramolecular complexes, a process which requires KIBRA coiled-coil domains. These findings reveal molecular mechanisms by which KIBRA can organize key synaptic signaling complexes.
Collapse
|
17
|
Lv Y, Wen L, Hu WJ, Deng C, Ren HW, Bao YN, Su BW, Gao P, Man ZY, Luo YY, Li CJ, Xiang ZX, Wang B, Luan ZL. Schizophrenia in the genetic era: a review from development history, clinical features and genomic research approaches to insights of susceptibility genes. Metab Brain Dis 2024; 39:147-171. [PMID: 37542622 DOI: 10.1007/s11011-023-01271-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/27/2023] [Indexed: 08/07/2023]
Abstract
Schizophrenia is a devastating neuropsychiatric disorder affecting 1% of the world population and ranks as one of the disorders providing the most severe burden for society. Schizophrenia etiology remains obscure involving multi-risk factors, such as genetic, environmental, nutritional, and developmental factors. Complex interactions of genetic and environmental factors have been implicated in the etiology of schizophrenia. This review provides an overview of the historical origins, pathophysiological mechanisms, diagnosis, clinical symptoms and corresponding treatment of schizophrenia. In addition, as schizophrenia is a polygenic, genetic disorder caused by the combined action of multiple micro-effective genes, we further detail several approaches, such as candidate gene association study (CGAS) and genome-wide association study (GWAS), which are commonly used in schizophrenia genomics studies. A number of GWASs about schizophrenia have been performed with the hope to identify novel, consistent and influential risk genetic factors. Finally, some schizophrenia susceptibility genes have been identified and reported in recent years and their biological functions are also listed. This review may serve as a summary of past research on schizophrenia genomics and susceptibility genes (NRG1, DISC1, RELN, BDNF, MSI2), which may point the way to future schizophrenia genetics research. In addition, depending on the above discovery of susceptibility genes and their exact function, the development and application of antipsychotic drugs will be promoted in the future.
Collapse
Affiliation(s)
- Ye Lv
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Lin Wen
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Wen-Juan Hu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Chong Deng
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China
| | - Hui-Wen Ren
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Ya-Nan Bao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Bo-Wei Su
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Ping Gao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Zi-Yue Man
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Yi-Yang Luo
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Cheng-Jie Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Zhi-Xin Xiang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Bing Wang
- Department of Endocrinology and Metabolism, The Central hospital of Dalian University of Technology, Dalian, 116000, China.
| | - Zhi-Lin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
18
|
Sahay S, Devine EA, McCullumsmith RE, O’Donovan SM. Adenosine Receptor mRNA Expression in Frontal Cortical Neurons in Schizophrenia. Cells 2023; 13:32. [PMID: 38201235 PMCID: PMC10778287 DOI: 10.3390/cells13010032] [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: 11/01/2023] [Revised: 12/14/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Schizophrenia is a devastating neuropsychiatric disorder associated with the dysregulation of glutamate and dopamine neurotransmitter systems. The adenosine system is an important neuroregulatory system in the brain that modulates glutamate and dopamine signaling via the ubiquitously expressed adenosine receptors; however, adenosine A1 and A2A receptor (A1R and A2AR) mRNA expression is poorly understood in specific cell subtypes in the frontal cortical brain regions implicated in this disorder. In this study, we assayed A1R and A2AR mRNA expression via qPCR in enriched populations of pyramidal neurons, which were isolated from postmortem anterior cingulate cortex (ACC) tissue from schizophrenia (n = 20) and control (n = 20) subjects using laser microdissection (LMD). A1R expression was significantly increased in female schizophrenia subjects compared to female control subjects (t(13) = -4.008, p = 0.001). A1R expression was also significantly decreased in female control subjects compared to male control subjects, suggesting sex differences in basal A1R expression (t(17) = 2.137, p = 0.047). A significant, positive association was found between dementia severity (clinical dementia rating (CDR) scores) and A2AR mRNA expression (Spearman's r = 0.424, p = 0.009). A2AR mRNA expression was significantly increased in unmedicated schizophrenia subjects, suggesting that A2AR expression may be normalized by chronic antipsychotic treatment (F(1,14) = 9.259, p = 0.009). Together, these results provide novel insights into the neuronal expression of adenosine receptors in the ACC in schizophrenia and suggest that receptor expression changes may be sex-dependent and associated with cognitive decline in these subjects.
Collapse
Affiliation(s)
- Smita Sahay
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.); (R.E.M.)
| | - Emily A. Devine
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | - Robert E. McCullumsmith
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.); (R.E.M.)
- Neuroscience Institute Promedica, Toledo, OH 43606, USA
| | - Sinead M. O’Donovan
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.); (R.E.M.)
| |
Collapse
|
19
|
Lan H, Suo X, Zuo C, Ni W, Wang S, Kemp GJ, Gong Q. Shared and distinct abnormalities of brain magnetization transfer ratio in schizophrenia and major depressive disorder: a comparative voxel-based meta-analysis. Chin Med J (Engl) 2023; 136:2824-2833. [PMID: 37697951 PMCID: PMC10686600 DOI: 10.1097/cm9.0000000000002538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND Patients with schizophrenia (SCZ) and major depressive disorder (MDD) share significant clinical overlap, although it remains unknown to what extent this overlap reflects shared neural profiles. To identify the shared and specific abnormalities in SCZ and MDD, we performed a whole-brain voxel-based meta-analysis using magnetization transfer imaging, a technique that characterizes the macromolecular structural integrity of brain tissue in terms of the magnetization transfer ratio (MTR). METHODS A systematic search based on Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines was conducted in PubMed, EMBASE, International Scientific Index (ISI) Web of Science, and MEDLINE for relevant studies up to March 2022. Two researchers independently screened the articles. Rigorous scrutiny and data extraction were performed for the studies that met the inclusion criteria. Voxel-wise meta-analyses were conducted using anisotropic effect size-signed differential mapping with a unified template. Meta-regression was used to explore the potential effects of demographic and clinical characteristics. RESULTS A total of 15 studies with 17 datasets describing 365 SCZ patients, 224 MDD patients, and 550 healthy controls (HCs) were identified. The conjunction analysis showed that both disorders shared higher MTR than HC in the left cerebellum ( P =0.0006) and left fusiform gyrus ( P =0.0004). Additionally, SCZ patients showed disorder-specific lower MTR in the anterior cingulate/paracingulate gyrus, right superior temporal gyrus, and right superior frontal gyrus, and higher MTR in the left thalamus, precuneus/cuneus, posterior cingulate gyrus, and paracentral lobule; and MDD patients showed higher MTR in the left middle occipital region. Meta-regression showed no statistical significance in either group. CONCLUSIONS The results revealed a structural neural basis shared between SCZ and MDD patients, emphasizing the importance of shared neural substrates across psychopathology. Meanwhile, distinct disease-specific characteristics could have implications for future differential diagnosis and targeted treatment.
Collapse
Affiliation(s)
- Huan Lan
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Xueling Suo
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian 361000, China
| | - Chao Zuo
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan 610041, China
| | - Weishi Ni
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Song Wang
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Graham J. Kemp
- Liverpool Magnetic Resonance Imaging Centre (LiMRIC) and Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L693BX, United Kingdom
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian 361000, China
| |
Collapse
|
20
|
Falkai P, Rossner MJ, Raabe FJ, Wagner E, Keeser D, Maurus I, Roell L, Chang E, Seitz-Holland J, Schulze TG, Schmitt A. Disturbed Oligodendroglial Maturation Causes Cognitive Dysfunction in Schizophrenia: A New Hypothesis. Schizophr Bull 2023; 49:1614-1624. [PMID: 37163675 PMCID: PMC10686333 DOI: 10.1093/schbul/sbad065] [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: 05/12/2023]
Abstract
BACKGROUND AND HYPOTHESIS Cognitive impairment is a hallmark of schizophrenia, but no effective treatment is available to date. The underlying pathophysiology includes disconnectivity between hippocampal and prefrontal brain regions. Supporting evidence comes from diffusion-weighted imaging studies that suggest abnormal organization of frontotemporal white matter pathways in schizophrenia. STUDY DESIGN Here, we hypothesize that in schizophrenia, deficient maturation of oligodendrocyte precursor cells (OPCs) into mature oligodendrocytes substantially contributes to abnormal frontotemporal macro- and micro-connectivity and subsequent cognitive deficits. STUDY RESULTS Our postmortem studies indicate a reduced oligodendrocyte number in the cornu ammonis 4 (CA4) subregion of the hippocampus, and others have reported the same histopathological finding in the dorsolateral prefrontal cortex. Our series of studies on aerobic exercise training showed a volume increase in the hippocampus, specifically in the CA4 region, and improved cognition in individuals with schizophrenia. The cognitive effects were subsequently confirmed by meta-analyses. Cell-specific schizophrenia polygenic risk scores showed that exercise-induced CA4 volume increase significantly correlates with OPCs. From animal models, it is evident that early life stress and oligodendrocyte-related gene variants lead to schizophrenia-related behavior, cognitive deficits, impaired oligodendrocyte maturation, and reduced myelin thickness. CONCLUSIONS Based on these findings, we propose that pro-myelinating drugs (e.g., the histamine blocker clemastine) combined with aerobic exercise training may foster the regeneration of myelin plasticity as a basis for restoring frontotemporal connectivity and cognition in schizophrenia.
Collapse
Affiliation(s)
- Peter Falkai
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilian University, Munich, Germany
- Max-Planck-Institute of Psychiatry, Munich, Germany
| | - Moritz J Rossner
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilian University, Munich, Germany
| | - Florian J Raabe
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilian University, Munich, Germany
| | - Elias Wagner
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilian University, Munich, Germany
| | - Daniel Keeser
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilian University, Munich, Germany
- NeuroImaging Core Unit Munich (NICUM), University Hospital, Ludwig-Maximilian University, Munich, Germany
| | - Isabel Maurus
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilian University, Munich, Germany
| | - Lukas Roell
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilian University, Munich, Germany
- NeuroImaging Core Unit Munich (NICUM), University Hospital, Ludwig-Maximilian University, Munich, Germany
| | - Emily Chang
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilian University, Munich, Germany
| | - Johanna Seitz-Holland
- Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas G Schulze
- Institute for Psychiatric Phenomic and Genomic (IPPG), Munich, Germany
| | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilian University, Munich, Germany
- Laboratory of Neuroscience (LIM27), Institute of Psychiatry, University of São Paulo (USP), São Paulo-SP, Brazil
| |
Collapse
|
21
|
Wu Q, Ren Q, Meng J, Gao WJ, Chang YZ. Brain Iron Homeostasis and Mental Disorders. Antioxidants (Basel) 2023; 12:1997. [PMID: 38001850 PMCID: PMC10669508 DOI: 10.3390/antiox12111997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/30/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Iron plays an essential role in various physiological processes. A disruption in iron homeostasis can lead to severe consequences, including impaired neurodevelopment, neurodegenerative disorders, stroke, and cancer. Interestingly, the link between mental health disorders and iron homeostasis has not received significant attention. Therefore, our understanding of iron metabolism in the context of psychological diseases is incomplete. In this review, we aim to discuss the pathologies and potential mechanisms that relate to iron homeostasis in associated mental disorders. We propose the hypothesis that maintaining brain iron homeostasis can support neuronal physiological functions by impacting key enzymatic activities during neurotransmission, redox balance, and myelination. In conclusion, our review highlights the importance of investigating the relationship between trace element nutrition and the pathological process of mental disorders, focusing on iron. This nutritional perspective can offer valuable insights for the clinical treatment of mental disorders.
Collapse
Affiliation(s)
- Qiong Wu
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Hebei University of Chinese Medicine, Shijiazhuang 050200, China;
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan’erhuan Eastern Road, Shijiazhuang 050024, China; (Q.R.); (J.M.)
| | - Qiuyang Ren
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan’erhuan Eastern Road, Shijiazhuang 050024, China; (Q.R.); (J.M.)
| | - Jingsi Meng
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan’erhuan Eastern Road, Shijiazhuang 050024, China; (Q.R.); (J.M.)
| | - Wei-Juan Gao
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Hebei University of Chinese Medicine, Shijiazhuang 050200, China;
| | - Yan-Zhong Chang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan’erhuan Eastern Road, Shijiazhuang 050024, China; (Q.R.); (J.M.)
| |
Collapse
|
22
|
Kato D, Aoyama Y, Nishida K, Takahashi Y, Sakamoto T, Takeda I, Tatematsu T, Go S, Saito Y, Kunishima S, Cheng J, Hou L, Tachibana Y, Sugio S, Kondo R, Eto F, Sato S, Moorhouse AJ, Yao I, Kadomatsu K, Setou M, Wake H. Regulation of lipid synthesis in myelin modulates neural activity and is required for motor learning. Glia 2023; 71:2591-2608. [PMID: 37475643 DOI: 10.1002/glia.24441] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 06/11/2023] [Accepted: 07/03/2023] [Indexed: 07/22/2023]
Abstract
Brain function relies on both rapid electrical communication in neural circuitry and appropriate patterns or synchrony of neural activity. Rapid communication between neurons is facilitated by wrapping nerve axons with insulation by a myelin sheath composed largely of different lipids. Recent evidence has indicated that the extent of myelination of nerve axons can adapt based on neural activity levels and this adaptive myelination is associated with improved learning of motor tasks, suggesting such plasticity may enhance effective learning. In this study, we examined whether another aspect of myelin plasticity-changes in myelin lipid synthesis and composition-may also be associated with motor learning. We combined a motor learning task in mice with in vivo two-photon imaging of neural activity in the primary motor cortex (M1) to distinguish early and late stages of learning and then probed levels of some key myelin lipids using mass spectrometry analysis. Sphingomyelin levels were elevated in the early stage of motor learning while galactosylceramide levels were elevated in the middle and late stages of motor learning, and these changes were correlated across individual mice with both learning performance and neural activity changes. Targeted inhibition of oligodendrocyte-specific galactosyltransferase expression, the enzyme that synthesizes myelin galactosylceramide, impaired motor learning. Our results suggest regulation of myelin lipid composition could be a novel facet of myelin adaptations associated with learning.
Collapse
Affiliation(s)
- Daisuke Kato
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
| | - Yuki Aoyama
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuki Nishida
- Division of System Neuroscience, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yutaka Takahashi
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takumi Sakamoto
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Ikuko Takeda
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
| | - Tsuyako Tatematsu
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shiori Go
- Institute for Glyco-core Research, Nagoya University, Nagoya, Japan
| | - Yutaro Saito
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shiho Kunishima
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jinlei Cheng
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Lingnan Hou
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihisa Tachibana
- Division of System Neuroscience, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shouta Sugio
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Reon Kondo
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumihiro Eto
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Sanda, Japan
| | - Shumpei Sato
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Andrew J Moorhouse
- School of Medical Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Ikuko Yao
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Sanda, Japan
| | - Kenji Kadomatsu
- Institute for Glyco-core Research, Nagoya University, Nagoya, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hiroaki Wake
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Center of Optical Scattering Image Science, Kobe University, Kobe, Japan
- Department of Physiological Sciences, Graduate University for Advanced Studies, SOKENDAI, Hayama, Japan
- Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama, Japan
| |
Collapse
|
23
|
Parker N, Cheng W, Hindley GFL, Parekh P, Shadrin AA, Maximov II, Smeland OB, Djurovic S, Dale AM, Westlye LT, Frei O, Andreassen OA. Psychiatric disorders and brain white matter exhibit genetic overlap implicating developmental and neural cell biology. Mol Psychiatry 2023; 28:4924-4932. [PMID: 37759039 PMCID: PMC11771104 DOI: 10.1038/s41380-023-02264-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023]
Abstract
Improved understanding of the shared genetic architecture between psychiatric disorders and brain white matter may provide mechanistic insights for observed phenotypic associations. Our objective is to characterize the shared genetic architecture of bipolar disorder (BD), major depression (MD), and schizophrenia (SZ) with white matter fractional anisotropy (FA) and identify shared genetic loci to uncover biological underpinnings. We used genome-wide association study (GWAS) summary statistics for BD (n = 413,466), MD (n = 420,359), SZ (n = 320,404), and white matter FA (n = 33,292) to uncover the genetic architecture (i.e., polygenicity and discoverability) of each phenotype and their genetic overlap (i.e., genetic correlations, overlapping trait-influencing variants, and shared loci). This revealed that BD, MD, and SZ are at least 7-times more polygenic and less genetically discoverable than average FA. Even in the presence of weak genetic correlations (range = -0.05 to -0.09), average FA shared an estimated 42.5%, 43.0%, and 90.7% of trait-influencing variants as well as 12, 4, and 28 shared loci with BD, MD, and SZ, respectively. Shared variants were mapped to genes and tested for enrichment among gene-sets which implicated neurodevelopmental expression, neural cell types, myelin, and cell adhesion molecules. For BD and SZ, case vs control tract-level differences in FA associated with genetic correlations between those same tracts and the respective disorder (rBD = 0.83, p = 4.99e-7 and rSZ = 0.65, p = 5.79e-4). Genetic overlap at the tract-level was consistent with average FA results. Overall, these findings suggest a genetic basis for the involvement of brain white matter aberrations in the pathophysiology of psychiatric disorders.
Collapse
Affiliation(s)
- Nadine Parker
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Weiqiu Cheng
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Guy F L Hindley
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Psychosis Studies, Institute of Psychiatry, Psychology and Neurosciences, King's College London, London, UK
| | - Pravesh Parekh
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alexey A Shadrin
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway
| | - Ivan I Maximov
- Department of Health and Functioning, Western Norway University of Applied Sciences, Bergen, Norway
| | - Olav B Smeland
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Anders M Dale
- Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Department of Radiology, University of California San Diego, La Jolla, CA, USA
| | - Lars T Westlye
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Oleksandr Frei
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Center for Bioinformatics, Department of Informatics, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
24
|
Israel-Elgali I, Pan H, Oved K, Pillar N, Levy G, Barak B, Carneiro A, Gurwitz D, Shomron N. Impaired myelin ultrastructure is reversed by citalopram treatment in a mouse model for major depressive disorder. J Psychiatr Res 2023; 166:100-114. [PMID: 37757703 DOI: 10.1016/j.jpsychires.2023.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/24/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023]
Abstract
Major depressive disorder (MDD) is the most common and widespread mental disorder. Selective serotonin reuptake inhibitors (SSRIs) are the first-line treatment for MDD. The relation between the inhibition of serotonin reuptake in the central nervous system and remission from MDD remains controversial, as reuptake inhibition occurs rapidly, but remission from MDD takes weeks to months. Myelination-related deficits and white matter abnormalities were shown to be involved in psychiatric disorders such as MDD. This may explain the delay in remission following SSRI administration. The raphe nuclei (RN), located in the brain stem, consist of clusters of serotonergic (5-HT) neurons that project to almost all regions of the brain. Thus, the RN are an intriguing area for research of the potential effect of SSRI on myelination, and their involvement in MDD. MicroRNAs (miRNAs) regulate many biological features that might be altered by antidepressants. Two cohorts of chronic unpredictable stress (CUS) mouse model for depression underwent behavioral tests for evaluating stress, anxiety, and depression levels. Following application of the CUS protocol and treatment with the SSRI, citalopram, 48 mice of the second cohort were tested via magnetic resonance imaging and diffusion tensor imaging for differences in brain white matter tracts. RN and superior colliculus were excised from both cohorts and measured for changes in miRNAs, mRNA, and protein levels of candidate genes. Using MRI-DTI scans we found lower fractional anisotropy and axial diffusivity in brains of stressed mice. Moreover, both miR-30b-5p and miR-101a-3p were found to be downregulated in the RN following CUS, and upregulated following CUS and citalopram treatment. The direct binding of these miRNAs to Qki, and the subsequent effects on mRNA and protein levels of myelin basic protein (Mbp), indicated involvement of these miRNAs in myelination ultrastructure processes in the RN, in response to CUS followed by SSRI treatment. We suggest that SSRIs are implicated in repairing myelin deficits resulting from chronic stress that leads to depression.
Collapse
Affiliation(s)
- Ifat Israel-Elgali
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hope Pan
- Department of Pharmacology, Center for Molecular Neuroscience, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Keren Oved
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nir Pillar
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gilad Levy
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv, Israel
| | - Boaz Barak
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv, Israel; Faculty of Social Sciences, School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ana Carneiro
- Department of Pharmacology, Center for Molecular Neuroscience, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - David Gurwitz
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Noam Shomron
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Edmond J Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, Israel; Tel Aviv University Innovation Laboratories (TILabs), Tel Aviv, Israel.
| |
Collapse
|
25
|
Adraoui FW, Douw L, Martens GJM, Maas DA. Connecting Neurobiological Features with Interregional Dysconnectivity in Social-Cognitive Impairments of Schizophrenia. Int J Mol Sci 2023; 24:ijms24097680. [PMID: 37175387 PMCID: PMC10177877 DOI: 10.3390/ijms24097680] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Schizophrenia (SZ) is a devastating psychiatric disorder affecting about 1% of the world's population. Social-cognitive impairments in SZ prevent positive social interactions and lead to progressive social withdrawal. The neurobiological underpinnings of social-cognitive symptoms remain poorly understood, which hinders the development of novel treatments. At the whole-brain level, an abnormal activation of social brain regions and interregional dysconnectivity within social-cognitive brain networks have been identified as major contributors to these symptoms. At the cellular and subcellular levels, an interplay between oxidative stress, neuroinflammation and N-methyl-D-aspartate receptor hypofunction is thought to underly SZ pathology. However, it is not clear how these molecular processes are linked with interregional dysconnectivity in the genesis of social-cognitive symptoms. Here, we aim to bridge the gap between macroscale (connectivity analyses) and microscale (molecular and cellular mechanistic) knowledge by proposing impaired myelination and the disinhibition of local microcircuits as possible causative biological pathways leading to dysconnectivity and abnormal activity of the social brain. Furthermore, we recommend electroencephalography as a promising translational technique that can foster pre-clinical drug development and discuss attractive drug targets for the treatment of social-cognitive symptoms in SZ.
Collapse
Affiliation(s)
- Florian W Adraoui
- Biotrial, Preclinical Pharmacology Department, 7-9 rue Jean-Louis Bertrand, 35000 Rennes, France
| | - Linda Douw
- Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan, 1081 HZ Amsterdam, The Netherlands
| | - Gerard J M Martens
- Donders Centre for Neuroscience (DCN), Department of Molecular Animal Physiology, Faculty of Science, Donders Institute for Brain, Cognition and Behavior, Radboud University, 6525 GA Nijmegen, The Netherlands
- NeuroDrug Research Ltd., 6525 ED Nijmegen, The Netherlands
| | - Dorien A Maas
- Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
26
|
Sano F, Kikushima K, Benner S, Xu L, Kahyo T, Yamasue H, Setou M. Associations between prefrontal PI (16:0/20:4) lipid, TNC mRNA, and APOA1 protein in schizophrenia: A trans-omics analysis in post-mortem brain. Front Psychiatry 2023; 14:1145437. [PMID: 37143779 PMCID: PMC10151580 DOI: 10.3389/fpsyt.2023.1145437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/24/2023] [Indexed: 05/06/2023] Open
Abstract
Background Though various mechanisms have been proposed for the pathophysiology of schizophrenia, the full extent of these mechanisms remains unclear, and little is known about the relationships among them. We carried out trans-omics analyses by comparing the results of the previously reported lipidomics, transcriptomics, and proteomics analyses; all of these studies used common post-mortem brain samples. Methods We collected the data from three aforementioned omics studies on 6 common post-mortem samples (3 schizophrenia patients and 3 controls), and analyzed them as a whole group sample. Three correlation analyses were performed for each of the two of three omics studies in these samples. In order to discuss the strength of the correlations in a limited sample size, the p-values of each correlation coefficient were confirmed using the Student's t-test. In addition, partial correlation analysis was also performed for some correlations, to verify the strength of the impact of each factor on the correlations. Results The following three factors were strongly correlated with each other: the lipid level of phosphatidylinositol (PI) (16:0/20:4), the amount of TNC mRNA, and the quantitative signal intensity of APOA1 protein. PI (16:0/20:4) and TNC showed a positive correlation, while PI (16:0/20:4) and APOA1, and TNC and APOA1 showed negative correlations. All of these correlations reached at p < 0.01. PI (16:0/20:4) and TNC were decreased in the prefrontal cortex of schizophrenia samples, while APOA1 was increased. Partial correlation analyses among them suggested that PI (16:0/20:4) and TNC have no direct correlation, but their relationships are mediated by APOA1. Conclusion The current results suggest that these three factors may provide new clues to elucidate the relationships among the candidate mechanisms of schizophrenia, and support the potential of trans-omics analyses as a new analytical method.
Collapse
Affiliation(s)
- Fumito Sano
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kenji Kikushima
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Seico Benner
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Lili Xu
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomoaki Kahyo
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hidenori Yamasue
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Systems Molecular Anatomy, Institute for Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
27
|
Kwon HJ, Hahn KR, Kang MS, Choi JH, Moon SM, Yoon YS, Hwang IK, Kim DW. Tat-malate dehydrogenase fusion protein protects neurons from oxidative and ischemic damage by reduction of reactive oxygen species and modulation of glutathione redox system. Sci Rep 2023; 13:5653. [PMID: 37024665 PMCID: PMC10079925 DOI: 10.1038/s41598-023-32812-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
Malate dehydrogenase (MDH) plays an important role in the conversion of malate to oxaloacetate during the tricarboxylic acid cycle. In this study, we examined the role of cytoplasmic MDH (MDH1) in hydrogen peroxide (H2O2)-induced oxidative stress in HT22 cells and ischemia-induced neuronal damage in the gerbil hippocampus. The Tat-MDH1 fusion protein was constructed to enable the delivery of MDH1 into the intracellular space and penetration of the blood-brain barrier. Tat-MDH1, but not MDH1 control protein, showed significant cellular delivery in HT22 cells in a concentration- and time-dependent manner and gradual intracellular degradation in HT22 cells. Treatment with 4 μM Tat-MDH1 significantly ameliorated 200 μM H2O2-induced cell death, DNA fragmentation, and reactive oxygen species formation in HT22 cells. Transient increases in MDH1 immunoreactivity were detected in the hippocampal CA1 region 6-12 h after ischemia, but MDH1 activity significantly decreased 2 days after ischemia. Supplementation of Tat-MDH1 immediately after ischemia alleviated ischemia-induced hyperlocomotion and neuronal damage 1 and 4 days after ischemia. In addition, treatment with Tat-MDH1 significantly ameliorated the increases in hydroperoxides, lipid peroxidation, and reactive oxygen species 2 days after ischemia. Tat-MDH1 treatment maintained the redox status of the glutathione system in the hippocampus 2 days after ischemia. These results suggest that Tat-MDH1 exerts neuroprotective effects by reducing oxidative stress and maintaining glutathione redox system in the hippocampus.
Collapse
Affiliation(s)
- Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea
- Department of Biomedical Sciences, and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Kyu Ri Hahn
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Min Soo Kang
- Department of Anatomy, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, South Korea
| | - Jung Hoon Choi
- Department of Anatomy, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, South Korea
| | - Seung Myung Moon
- Department of Neurosurgery, Kangnam Sacred Heart Hospital, College of Medicine, Hallym University, Seoul, 07441, South Korea
- Research Institute for Complementary & Alternative Medicine, Hallym University, Chuncheon, 24253, South Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea.
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea.
| |
Collapse
|
28
|
Zakaria WNA, Sasongko TH, Al-Rahbi B, Al-Sowayan N, Ahmad AH, Zakaria R, Ahmi A, Othman Z. Gene and schizophrenia in the pregenome and postgenome-wide association studies era: a bibliometric analysis and network visualization. Psychiatr Genet 2023; 33:37-49. [PMID: 36825838 DOI: 10.1097/ypg.0000000000000336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
This study aimed to perform a bibliometric analysis on genetic studies in schizophrenia in the pregenome-wide association studies (GWAS) and post-GWAS era. We searched the literature on genes and schizophrenia using the Scopus database. The documents increased with time, especially after the human genome project and International HapMap Project, with the highest citation in 2008. The top occurrence author keywords were discovered to be different in the pre-GWAS and post-GWAS eras, reflecting the progress of genetic studies connected to schizophrenia. Emerging keywords highlighted a trend towards an application of precision medicine, showing an interplay of environmental exposures as well as genetic factors in schizophrenia pathogenesis, progression, and response to therapy. In conclusion, the gene and schizophrenia literature has grown rapidly after the human genome project, and the temporal variation in the author keywords pattern reflects the trend of genetic studies related to schizophrenia in the pre-GWAS and post-GWAS era.
Collapse
Affiliation(s)
- Wan Nur Amalina Zakaria
- Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Kota Bharu, Kelantan, Malaysia
| | - Teguh Haryo Sasongko
- Department of Physiology, School of Medicine, and Institute for Research, Development and Innovation, International Medical University, Kuala Lumpur, Malaysia
| | | | - Noorah Al-Sowayan
- Department of Biology, College of Science, Qassim University, Saudi Arabia
| | - Asma Hayati Ahmad
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Rahimah Zakaria
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Aidi Ahmi
- Tunku Puteri Intan Safinaz School of Accountancy, Universiti Utara Malaysia 06010 UUM Sintok, Kedah
| | - Zahiruddin Othman
- Department of Psychiatry, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
29
|
Dong D, Yao D, Wang Y, Hong SJ, Genon S, Xin F, Jung K, He H, Chang X, Duan M, Bernhardt BC, Margulies DS, Sepulcre J, Eickhoff SB, Luo C. Compressed sensorimotor-to-transmodal hierarchical organization in schizophrenia. Psychol Med 2023; 53:771-784. [PMID: 34100349 DOI: 10.1017/s0033291721002129] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Schizophrenia has been primarily conceptualized as a disorder of high-order cognitive functions with deficits in executive brain regions. Yet due to the increasing reports of early sensory processing deficit, recent models focus more on the developmental effects of impaired sensory process on high-order functions. The present study examined whether this pathological interaction relates to an overarching system-level imbalance, specifically a disruption in macroscale hierarchy affecting integration and segregation of unimodal and transmodal networks. METHODS We applied a novel combination of connectome gradient and stepwise connectivity analysis to resting-state fMRI to characterize the sensorimotor-to-transmodal cortical hierarchy organization (96 patients v. 122 controls). RESULTS We demonstrated compression of the cortical hierarchy organization in schizophrenia, with a prominent compression from the sensorimotor region and a less prominent compression from the frontal-parietal region, resulting in a diminished separation between sensory and fronto-parietal cognitive systems. Further analyses suggested reduced differentiation related to atypical functional connectome transition from unimodal to transmodal brain areas. Specifically, we found hypo-connectivity within unimodal regions and hyper-connectivity between unimodal regions and fronto-parietal and ventral attention regions along the classical sensation-to-cognition continuum (voxel-level corrected, p < 0.05). CONCLUSIONS The compression of cortical hierarchy organization represents a novel and integrative system-level substrate underlying the pathological interaction of early sensory and cognitive function in schizophrenia. This abnormal cortical hierarchy organization suggests cascading impairments from the disruption of the somatosensory-motor system and inefficient integration of bottom-up sensory information with attentional demands and executive control processes partially account for high-level cognitive deficits characteristic of schizophrenia.
Collapse
Affiliation(s)
- Debo Dong
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Life Science and Technology, University of Electronic Science and Technology of China, China
| | - Dezhong Yao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Life Science and Technology, University of Electronic Science and Technology of China, China
- Research Unit of NeuroInformation, Chinese Academy of Medical Sciences, 2019RU035, Chengdu, China
| | - Yulin Wang
- Faculty of Psychological and Educational Sciences, Department of Experimental and Applied Psychology, Vrije Universiteit Brussel, Belgium
- Faculty of Psychology and Educational Sciences, Department of Data Analysis, Ghent University, Belgium
| | - Seok-Jun Hong
- Center for the Developing Brain, Child Mind Institute, NY, USA
- Department of Biomedical Engineering, Center for Neuroscience Imaging Research, Institute for Basic Science, Sungkyunkwan University, South Korea
| | - Sarah Genon
- Institute for Systems Neuroscience, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
| | - Fei Xin
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Life Science and Technology, University of Electronic Science and Technology of China, China
| | - Kyesam Jung
- Institute for Systems Neuroscience, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
| | - Hui He
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Life Science and Technology, University of Electronic Science and Technology of China, China
- Department of Psychiatry, The Fourth People's Hospital of Chengdu, Chengdu, China
| | - Xuebin Chang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Life Science and Technology, University of Electronic Science and Technology of China, China
| | - Mingjun Duan
- Department of Psychiatry, The Fourth People's Hospital of Chengdu, Chengdu, China
| | - Boris C Bernhardt
- Multimodal Imaging and Connectome Analysis Lab, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Daniel S Margulies
- Centre National de la Recherche Scientifique (CNRS) UMR 7225, Institut du Cerveau et de la Moelle épinière, Paris, France
| | - Jorge Sepulcre
- Department of Radiology, Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Simon B Eickhoff
- Institute for Systems Neuroscience, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
| | - Cheng Luo
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Life Science and Technology, University of Electronic Science and Technology of China, China
- Department of Neurology, Brain Disorders and Brain Function Key Laboratory, First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
30
|
Gene Expression and Epigenetic Regulation in the Prefrontal Cortex of Schizophrenia. Genes (Basel) 2023; 14:genes14020243. [PMID: 36833173 PMCID: PMC9957055 DOI: 10.3390/genes14020243] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/03/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Schizophrenia pathogenesis remains challenging to define; however, there is strong evidence that the interaction of genetic and environmental factors causes the disorder. This paper focuses on transcriptional abnormalities in the prefrontal cortex (PFC), a key anatomical structure that determines functional outcomes in schizophrenia. This review summarises genetic and epigenetic data from human studies to understand the etiological and clinical heterogeneity of schizophrenia. Gene expression studies using microarray and sequencing technologies reported the aberrant transcription of numerous genes in the PFC in patients with schizophrenia. Altered gene expression in schizophrenia is related to several biological pathways and networks (synaptic function, neurotransmission, signalling, myelination, immune/inflammatory mechanisms, energy production and response to oxidative stress). Studies investigating mechanisms driving these transcriptional abnormalities focused on alternations in transcription factors, gene promoter elements, DNA methylation, posttranslational histone modifications or posttranscriptional regulation of gene expression mediated by non-coding RNAs.
Collapse
|
31
|
Ishihara K, Takata K, Mizutani KI. Involvement of an Aberrant Vascular System in Neurodevelopmental, Neuropsychiatric, and Neuro-Degenerative Diseases. LIFE (BASEL, SWITZERLAND) 2023; 13:life13010221. [PMID: 36676170 PMCID: PMC9866034 DOI: 10.3390/life13010221] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
The vascular system of the prenatal brain is crucial for the development of the central nervous system. Communication between vessels and neural cells is bidirectional, and dysfunctional communication can lead to neurodevelopmental diseases. In the present review, we introduce neurodevelopmental and neuropsychiatric diseases potentially caused by disturbances in the neurovascular system and discuss candidate genes responsible for neurovascular system impairments. In contrast to diseases that can manifest during the developing stage, we have also summarized the disturbances of the neurovascular system in neurodegenerative diseases including Alzheimer's disease and Parkinson's disease. Furthermore, we discussed the role of abnormal vascularization and dysfunctional vessels in the development of neurovascular-related diseases.
Collapse
Affiliation(s)
- Keiichi Ishihara
- Department of Pathological Biochemistry, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
- Correspondence: ; Tel.: +81-75-595-4656
| | - Kazuyuki Takata
- Division of Integrated Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Ken-ichi Mizutani
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe 650-8586, Japan
| |
Collapse
|
32
|
Singh B, Doborjeh M, Doborjeh Z, Budhraja S, Tan S, Sumich A, Goh W, Lee J, Lai E, Kasabov N. Constrained neuro fuzzy inference methodology for explainable personalised modelling with applications on gene expression data. Sci Rep 2023; 13:456. [PMID: 36624117 PMCID: PMC9829920 DOI: 10.1038/s41598-022-27132-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/26/2022] [Indexed: 01/11/2023] Open
Abstract
Interpretable machine learning models for gene expression datasets are important for understanding the decision-making process of a classifier and gaining insights on the underlying molecular processes of genetic conditions. Interpretable models can potentially support early diagnosis before full disease manifestation. This is particularly important yet, challenging for mental health. We hypothesise this is due to extreme heterogeneity issues which may be overcome and explained by personalised modelling techniques. Thus far, most machine learning methods applied to gene expression datasets, including deep neural networks, lack personalised interpretability. This paper proposes a new methodology named personalised constrained neuro fuzzy inference (PCNFI) for learning personalised rules from high dimensional datasets which are structurally and semantically interpretable. Case studies on two mental health related datasets (schizophrenia and bipolar disorders) have shown that the relatively short and simple personalised fuzzy rules provided enhanced interpretability as well as better classification performance compared to other commonly used machine learning methods. Performance test on a cancer dataset also showed that PCNFI matches previous benchmarks. Insights from our approach also indicated the importance of two genes (ATRX and TSPAN2) as possible biomarkers for early differentiation of ultra-high risk, bipolar and healthy individuals. These genes are linked to cognitive ability and impulsive behaviour. Our findings suggest a significant starting point for further research into the biological role of cognitive and impulsivity-related differences. With potential applications across bio-medical research, the proposed PCNFI method is promising for diagnosis, prognosis, and the design of personalised treatment plans for better outcomes in the future.
Collapse
Affiliation(s)
- Balkaran Singh
- Knowledge Engineering and Discovery Research Innovation (KEDRI), School of Engineering Computer and Mathematical Sciences, Auckland University of Technology, Auckland, New Zealand.
| | - Maryam Doborjeh
- Knowledge Engineering and Discovery Research Innovation (KEDRI), School of Engineering Computer and Mathematical Sciences, Auckland University of Technology, Auckland, New Zealand.
| | - Zohreh Doborjeh
- School of Population Health, The University of Auckland, Auckland, New Zealand
- School of Psychology, The University of Waikato, Hamilton, New Zealand
| | - Sugam Budhraja
- Knowledge Engineering and Discovery Research Innovation (KEDRI), School of Engineering Computer and Mathematical Sciences, Auckland University of Technology, Auckland, New Zealand
| | - Samuel Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore, Singapore
| | - Alexander Sumich
- Department of Psychology, Nottingham Trent University, Nottingham, UK
| | - Wilson Goh
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore, Singapore
- Center for Biomedical Informatics, Nanyang Technological University (NTU), Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University (NTU), Singapore, Singapore
| | - Jimmy Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore, Singapore
- Institute for Mental Health, Singapore, Singapore
| | - Edmund Lai
- Knowledge Engineering and Discovery Research Innovation (KEDRI), School of Engineering Computer and Mathematical Sciences, Auckland University of Technology, Auckland, New Zealand
| | - Nikola Kasabov
- Knowledge Engineering and Discovery Research Innovation (KEDRI), School of Engineering Computer and Mathematical Sciences, Auckland University of Technology, Auckland, New Zealand
- Intelligent Systems Research Center, Ulster University, Derry, UK
- Institute for Information and Communication Technologies, Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
33
|
Mendoza ML, Quigley LD, Dunham T, Volk LJ. KIBRA regulates activity-induced AMPA receptor expression and synaptic plasticity in an age-dependent manner. iScience 2022; 25:105623. [PMID: 36465112 PMCID: PMC9713372 DOI: 10.1016/j.isci.2022.105623] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 10/14/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022] Open
Abstract
A growing body of human literature implicates KIBRA in memory and neurodevelopmental disorders. Memory and the cellular substrates supporting adaptive cognition change across development. Using an inducible KIBRA knockout mouse, we demonstrate that adult-onset deletion of KIBRA in forebrain neurons impairs long-term spatial memory and long-term potentiation (LTP). These LTP deficits correlate with adult-selective decreases in extrasynaptic AMPA receptors under basal conditions, and we identify a role for KIBRA in LTP-induced AMPAR upregulation. In contrast, juvenile-onset deletion of KIBRA in forebrain neurons did not affect LTP and had minimal effects on basal AMPAR expression. LTP did not increase AMPAR protein expression in juvenile WT mice, providing a potential explanation for juvenile resilience to KIBRA deletion. These data suggest that KIBRA serves a unique role in adult hippocampal function through regulation of basal and activity-dependent AMPAR proteostasis that supports synaptic plasticity.
Collapse
Affiliation(s)
- Matthew L. Mendoza
- Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lilyana D. Quigley
- Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Thomas Dunham
- Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lenora J. Volk
- Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Peter O’ Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
34
|
Leo H, Kipp M. Remyelination in Multiple Sclerosis: Findings in the Cuprizone Model. Int J Mol Sci 2022; 23:ijms232416093. [PMID: 36555733 PMCID: PMC9783537 DOI: 10.3390/ijms232416093] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Remyelination therapies, which are currently under development, have a great potential to delay, prevent or even reverse disability in multiple sclerosis patients. Several models are available to study the effectiveness of novel compounds in vivo, among which is the cuprizone model. This model is characterized by toxin-induced demyelination, followed by endogenous remyelination after cessation of the intoxication. Due to its high reproducibility and ease of use, this model enjoys high popularity among various research and industrial groups. In this review article, we will summarize recent findings using this model and discuss the potential of some of the identified compounds to promote remyelination in multiple sclerosis patients.
Collapse
Affiliation(s)
| | - Markus Kipp
- Correspondence: ; Tel.: +49-(0)-381-494-8400
| |
Collapse
|
35
|
Roychaudhuri R, Gadalla MM, West T, Snyder SH. A Novel Stereospecific Bioluminescent Assay for Detection of Endogenous d-Cysteine. ACS Chem Neurosci 2022; 13:3257-3262. [PMID: 36403160 DOI: 10.1021/acschemneuro.2c00528] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The presence of endogenous d-stereoisomers of amino acids in mammals dispels a long-standing dogma about their existence. d-Serine and d-aspartate function as novel neurotransmitters in mammals. However, the stereoisomer with the fastest, spontaneous in vitro racemization rate, d-cysteine, has not been reported. We utilized a novel, stereospecific, bioluminescent assay to identify endogenous d-cysteine in substantial amounts in the eye, brain, and pancreas of mice. d-Cysteine is enriched in mice embryonic brains at day E9.5 (4.5 mM) and decreases progressively with development (μM levels). d-Cysteine is also present in significantly higher amounts in the human brain white matter compared with gray matter. In the luciferase assay, d-cysteine conjugates with cyano hydroxy benzothiazole in the presence of a base and reducing agent to form d-luciferin. d-Luciferin, subsequently, in the presence of firefly luciferase and ATP, emits bioluminescence proportional to the concentration of d-cysteine. The assay is stereospecific and allows the quantitative estimation of endogenous d-cysteine in tissues in addition to its specificity for d-cysteine. Future efforts aimed at bioluminescent in vivo imaging of d-cysteine may allow a more noninvasive means of its detection, thereby elucidating its function.
Collapse
Affiliation(s)
- Robin Roychaudhuri
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Moataz M Gadalla
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Timothy West
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Solomon H Snyder
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| |
Collapse
|
36
|
Chen P, Wang D, Xiu M, Chen D, Lackey B, Wu HE, Wang L, Zhang X. Association of Transferrin Gene Polymorphism with Cognitive Deficits and Psychiatric Symptoms in Patients with Chronic Schizophrenia. J Clin Med 2022; 11:jcm11216414. [PMID: 36362642 PMCID: PMC9654946 DOI: 10.3390/jcm11216414] [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: 10/04/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 11/16/2022] Open
Abstract
A large amount of recent literature has focused on impaired iron homeostasis in the pathophysiology of schizophrenia. Specifically, microarray analysis has illustrated associations between the transferrin locus and schizophrenia. To elaborate on the effects of transferrin on schizophrenia and its psychiatric phenotypes, our study aimed to investigate whether transferrin gene polymorphism was correlated with cognitive deficits and clinical symptoms in schizophrenia. We recruited 564 patients with chronic schizophrenia and 422 healthy controls (HCs) in a Han Chinese population, collected phenotypic data, and genotyped the rs3811655 polymorphism of the transferrin gene. Our results showed that the rs3811655 polymorphism was related to cognitive performance in both patients and HCs, as well as negative symptoms in patients (all p < 0.05), and patients carrying at least one G-allele showed worsened cognition/severe negative symptoms (all p < 0.05). Further analyses also found that the rs3811655 polymorphism in combination with cognition may exert small but significant contributions to the negative (β = −0.10, t = −2.48, p < 0.05) or total psychiatric symptoms (β = −0.08, t = −1.92, p < 0.05) in patients. Our findings indicated that the rs3811655 polymorphism may be implicated in the cognitive deficits of schizophrenia and HCs as well as psychiatric symptoms in patients, which suggested the possible iron regulatory mechanism in the pathology of schizophrenia.
Collapse
Affiliation(s)
- Pinhong Chen
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Dongmei Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meihong Xiu
- Beijing Huilongguan Hospital, Peking University, Beijing 100871, China
| | - Dachun Chen
- Beijing Huilongguan Hospital, Peking University, Beijing 100871, China
| | - Blake Lackey
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Hanjing E. Wu
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Lubin Wang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Xiangyang Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China
- Correspondence:
| |
Collapse
|
37
|
Heller C, Kimmig ACS, Kubicki MR, Derntl B, Kikinis Z. Imaging the human brain on oral contraceptives: A review of structural imaging methods and implications for future research goals. Front Neuroendocrinol 2022; 67:101031. [PMID: 35998859 DOI: 10.1016/j.yfrne.2022.101031] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/30/2022] [Accepted: 08/15/2022] [Indexed: 12/21/2022]
Abstract
Worldwide over 150 million women use oral contraceptives (OCs), which are the most prescribed form of contraception in both the United States and in European countries. Sex hormones, such as estradiol and progesterone, are important endogenous hormones known for shaping the brain across the life span. Synthetic hormones, which are present in OCs, interfere with the natural hormonal balance by reducing the endogenous hormone levels. Little is known how this affects the brain, especially during the most vulnerable times of brain maturation. Here, we review studies that investigate differences in brain gray and white matter in women using OCs in comparison to naturally cycling women. We focus on two neuroimaging methods used to quantify structural gray and white matter changes, namely structural MRI and diffusion MRI. Finally, we discuss the potential of these imaging techniques to advance knowledge about the effects of OCs on the brain and wellbeing in women.
Collapse
Affiliation(s)
- Carina Heller
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry and Psychotherapy, Jena University Hospital, Germany; Department of Clinical Psychology, Friedrich Schiller University Jena, Germany.
| | - Ann-Christin S Kimmig
- Department of Psychiatry and Psychotherapy, Innovative Neuroimaging, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, Germany; Graduate Training Centre of Neuroscience, International Max Planck Research School, University of Tübingen, Tübingen, Germany
| | - Marek R Kubicki
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Birgit Derntl
- Department of Psychiatry and Psychotherapy, Innovative Neuroimaging, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, Germany; Lead Graduate School, University of Tübingen, Tübingen, Germany
| | - Zora Kikinis
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
38
|
Caprariello AV, Adams DJ. The landscape of targets and lead molecules for remyelination. Nat Chem Biol 2022; 18:925-933. [PMID: 35995862 PMCID: PMC9773298 DOI: 10.1038/s41589-022-01115-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 07/18/2022] [Indexed: 12/24/2022]
Abstract
Remyelination, or the restoration of myelin sheaths around axons in the central nervous system, is a multi-stage repair process that remains a major need for millions of patients with multiple sclerosis and other diseases of myelin. Even into adulthood, rodents and humans can generate new myelin-producing oligodendrocytes, leading to the therapeutic hypothesis that enhancing remyelination could lessen disease burden in multiple sclerosis. Multiple labs have used phenotypic screening to identify dozens of drugs that enhance oligodendrocyte formation, and several hit molecules have now advanced to clinical evaluation. Target identification studies have revealed that a large majority of these hits share the ability to inhibit a narrow range of cholesterol pathway enzymes and thereby induce cellular accumulation of specific sterol precursors to cholesterol. This Perspective surveys the recent fruitful intersection of chemical biology and remyelination and suggests multiple approaches toward new targets and lead molecules to promote remyelination.
Collapse
Affiliation(s)
| | - Drew J Adams
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
39
|
Zakutansky PM, Feng Y. The Long Non-Coding RNA GOMAFU in Schizophrenia: Function, Disease Risk, and Beyond. Cells 2022; 11:1949. [PMID: 35741078 PMCID: PMC9221589 DOI: 10.3390/cells11121949] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 02/05/2023] Open
Abstract
Neuropsychiatric diseases are among the most common brain developmental disorders, represented by schizophrenia (SZ). The complex multifactorial etiology of SZ remains poorly understood, which reflects genetic vulnerabilities and environmental risks that affect numerous genes and biological pathways. Besides the dysregulation of protein-coding genes, recent discoveries demonstrate that abnormalities associated with non-coding RNAs, including microRNAs and long non-coding RNAs (lncRNAs), also contribute to the pathogenesis of SZ. lncRNAs are an actively evolving family of non-coding RNAs that harbor greater than 200 nucleotides but do not encode for proteins. In general, lncRNA genes are poorly conserved. The large number of lncRNAs specifically expressed in the human brain, together with the genetic alterations and dysregulation of lncRNA genes in the SZ brain, suggests a critical role in normal cognitive function and the pathogenesis of neuropsychiatric diseases. A particular lncRNA of interest is GOMAFU, also known as MIAT and RNCR2. Growing evidence suggests the function of GOMAFU in governing neuronal development and its potential roles as a risk factor and biomarker for SZ, which will be reviewed in this article. Moreover, we discuss the potential mechanisms through which GOMAFU regulates molecular pathways, including its subcellular localization and interaction with RNA-binding proteins, and how interruption to GOMAFU pathways may contribute to the pathogenesis of SZ.
Collapse
Affiliation(s)
- Paul M. Zakutansky
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University, Atlanta, GA 30322, USA;
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yue Feng
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
40
|
Loss of RNA binding protein HuD facilitates the production of the senescence-associated secretory phenotype. Cell Death Dis 2022; 13:329. [PMID: 35411051 PMCID: PMC9001635 DOI: 10.1038/s41419-022-04792-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 02/08/2022] [Accepted: 02/16/2022] [Indexed: 02/06/2023]
Abstract
HuD, an RNA binding protein, plays a role in the regulation of gene expression in certain types of cells, including neuronal cells and pancreatic β-cells, via RNA metabolism. Its aberrant expression is associated with the pathogenesis of several human diseases. To explore HuD-mediated gene regulation, stable cells expressing short hairpin RNA against HuD were established using mouse neuroblastoma Neuro2a (N2a) cells, which displayed enhanced phenotypic characteristics of cellular senescence. Two approaches, RNA immunoprecipitation (RNA IP)-NanoString profiling and cytokine array, were used to subsequently identify a subset of putative HuD targets that act as senescence-associated secretory phenotype (SASP), including C-C motif ligand 2 (CCL2), CCL20, C-X-C motif chemokine ligand 2 (CXCL2), and interleukin-6 (IL-6). Here, we further demonstrated that HuD regulates the expression of CCL2, a SASP candidate upregulated in cells following HuD knockdown, by binding to the 3′-untranslated region (UTR) of Ccl2 mRNA. Downregulation of HuD increased the level of CCL2 in N2a cells and the brain tissues of HuD knockout (KO) mice. Exposure to γ-irradiation induced cellular senescence in N2a cells and HuD knockdown facilitated stress-induced cellular senescence. Our results reveal that HuD acts as a novel regulator of CCL2 expression, and its aberrant expression may contribute to cellular senescence by regulating SASP production.
Collapse
|
41
|
Waszczuk K, Tyburski E, Rek-Owodziń K, Plichta P, Rudkowski K, Podwalski P, Bielecki M, Mak M, Bober A, Misiak B, Sagan L, Michalczyk A, Kucharska-Mazur J, Samochowiec J. Relationship between White Matter Alterations and Pathophysiological Symptoms in Patients with Ultra-High Risk of Psychosis, First-Episode, and Chronic Schizophrenia. Brain Sci 2022; 12:brainsci12030354. [PMID: 35326310 PMCID: PMC8946295 DOI: 10.3390/brainsci12030354] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/20/2022] [Accepted: 03/03/2022] [Indexed: 12/03/2022] Open
Abstract
Some symptoms of schizophrenia might be present before full-blown psychosis, so white matter changes must be studied both in individuals with emerging psychosis and chronic schizophrenia. A total of 86 patients—12 ultra-high risk of psychosis (UHR), 20 first episode psychosis (FEP), 54 chronic schizophrenia (CS), and 33 healthy controls (HC)—underwent psychiatric examination and diffusion tensor imaging (DTI) in a 3-Tesla MRI scanner. We assessed fractional anisotropy (FA) and mean diffusivity (MD) of the superior longitudinal fasciculus (SLF) and inferior longitudinal fasciculus (ILS). We found that CS patients had lower FA than FEP patients (p = 0.025) and HC (p = 0.088), and higher MD than HC (p = 0.037) in the right SLF. In the CS group, we found positive correlations of MD in both right ILF (rho = 0.39, p < 0.05) and SLF (rho = 0.43, p < 0.01) with disorganization symptoms, as well as negative correlation of FA in the right ILF with disorganization symptoms (rho = −0.43, p < 0.05). Among UHR individuals, we found significant negative correlations between MD in the left ILF and negative (r = −0.74, p < 0.05) and general symptoms (r = −0.77, p < 0.05). However promising, these findings should be treated as preliminary, and further research must verify whether they can be treated as potential biomarkers of psychosis.
Collapse
Affiliation(s)
- Katarzyna Waszczuk
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Broniewskiego 26 Street, 71-460 Szczecin, Poland
| | - Ernest Tyburski
- Department of Health Psychology, Pomeranian Medical University in Szczecin, Broniewskiego 26 Street, 71-460 Szczecin, Poland
| | - Katarzyna Rek-Owodziń
- Department of Health Psychology, Pomeranian Medical University in Szczecin, Broniewskiego 26 Street, 71-460 Szczecin, Poland
| | - Piotr Plichta
- Department of Health Psychology, Pomeranian Medical University in Szczecin, Broniewskiego 26 Street, 71-460 Szczecin, Poland
| | - Krzysztof Rudkowski
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Broniewskiego 26 Street, 71-460 Szczecin, Poland
| | - Piotr Podwalski
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Broniewskiego 26 Street, 71-460 Szczecin, Poland
| | - Maksymilian Bielecki
- Department of Health Psychology, Pomeranian Medical University in Szczecin, Broniewskiego 26 Street, 71-460 Szczecin, Poland
| | - Monika Mak
- Department of Health Psychology, Pomeranian Medical University in Szczecin, Broniewskiego 26 Street, 71-460 Szczecin, Poland
| | - Adrianna Bober
- Institute of Psychology, University of Szczecin, Krakowska 69 Street, 71-017 Szczecin, Poland
| | - Błażej Misiak
- Department of Psychiatry, Division of Consultation Psychiatry and Neuroscience, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Leszek Sagan
- Department of Neurosurgery, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1 Street, 71-252 Szczecin, Poland
| | - Anna Michalczyk
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Broniewskiego 26 Street, 71-460 Szczecin, Poland
| | - Jolanta Kucharska-Mazur
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Broniewskiego 26 Street, 71-460 Szczecin, Poland
| | - Jerzy Samochowiec
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Broniewskiego 26 Street, 71-460 Szczecin, Poland
| |
Collapse
|
42
|
Godinez A, Rajput R, Chitranshi N, Gupta V, Basavarajappa D, Sharma S, You Y, Pushpitha K, Dhiman K, Mirzaei M, Graham S, Gupta V. Neuroserpin, a crucial regulator for axogenesis, synaptic modelling and cell-cell interactions in the pathophysiology of neurological disease. Cell Mol Life Sci 2022; 79:172. [PMID: 35244780 PMCID: PMC8897380 DOI: 10.1007/s00018-022-04185-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 01/31/2023]
Abstract
Neuroserpin is an axonally secreted serpin that is involved in regulating plasminogen and its enzyme activators, such as tissue plasminogen activator (tPA). The protein has been increasingly shown to play key roles in neuronal development, plasticity, maturation and synaptic refinement. The proteinase inhibitor may function both independently and through tPA-dependent mechanisms. Herein, we discuss the recent evidence regarding the role of neuroserpin in healthy and diseased conditions and highlight the participation of the serpin in various cellular signalling pathways. Several polymorphisms and mutations have also been identified in the protein that may affect the serpin conformation, leading to polymer formation and its intracellular accumulation. The current understanding of the involvement of neuroserpin in Alzheimer's disease, cancer, glaucoma, stroke, neuropsychiatric disorders and familial encephalopathy with neuroserpin inclusion bodies (FENIB) is presented. To truly understand the detrimental consequences of neuroserpin dysfunction and the effective therapeutic targeting of this molecule in pathological conditions, a cross-disciplinary understanding of neuroserpin alterations and its cellular signaling networks is essential.
Collapse
Affiliation(s)
- Angela Godinez
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| | - Rashi Rajput
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| | - Nitin Chitranshi
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia.
| | - Veer Gupta
- School of Medicine, Deakin University, Melbourne, VIC, Australia
| | - Devaraj Basavarajappa
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| | - Samridhi Sharma
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| | - Yuyi You
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| | - Kanishka Pushpitha
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| | - Kunal Dhiman
- School of Medicine, Deakin University, Melbourne, VIC, Australia
| | - Mehdi Mirzaei
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| | - Stuart Graham
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
- Save Sight Institute, University of Sydney, Sydney, NSW, Australia
| | - Vivek Gupta
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia.
| |
Collapse
|
43
|
White-Matter Integrity and Working Memory: Links to Aging and Dopamine-Related Genes. eNeuro 2022; 9:ENEURO.0413-21.2022. [PMID: 35346961 PMCID: PMC9014983 DOI: 10.1523/eneuro.0413-21.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/22/2022] [Accepted: 02/07/2022] [Indexed: 11/21/2022] Open
Abstract
Working memory, a core function underlying many higher-level cognitive processes, requires cooperation of multiple brain regions. White matter refers to myelinated axons, which are critical to interregional brain communication. Past studies on the association between white-matter integrity and working memory have yielded mixed findings. Using voxelwise tract-based spatial statistics analysis, we investigated this relationship in a sample of 328 healthy adults from 25 to 80 years of age. Given the important role of dopamine (DA) in working-memory functioning and white matter, we also analyzed the effects of dopamine-related genes on them. There were associations between white-matter integrity and working memory in multiple tracts, indicating that working-memory functioning relies on global connections between different brain areas across the adult life span. Moreover, a mediation analysis suggested that white-matter integrity contributes to age-related differences in working memory. Finally, there was an effect of the COMT Val158Met polymorphism on white-matter integrity, such that Val/Val carriers had lower fractional anisotropy values than any Met carriers in the internal capsule, corona radiata, and posterior thalamic radiation. As this polymorphism has been associated with dopaminergic tone in the prefrontal cortex, this result provides evidence for a link between DA neurotransmission and white matter. Together, the results support a link between white-matter integrity and working memory, and provide evidence for its interplay with age- and DA-related genes.
Collapse
|
44
|
El Nagar Z, El Shahawi HH, Effat SM, El Sheikh MM, Adel A, Ibrahim YA, Aufa OM. Single episode brief psychotic disorder versus bipolar disorder: A diffusion tensor imaging and executive functions study. Schizophr Res Cogn 2022; 27:100214. [PMID: 34557386 PMCID: PMC8446778 DOI: 10.1016/j.scog.2021.100214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/21/2021] [Accepted: 08/23/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Despite fast progress in neuroscientific approaches, the neurobiological continuum links psychotic spectrum, and affective disorder is obscure. White matter WM abnormalities found utilizing Diffusion Tensor Imaging (DTI) showing impaired communication in both disorders have been consistently demonstrated; however, direct comparisons of findings between them are scarce. This study aims to study WM abnormalities in single episode bipolar I disorder, and single episode brief psychotic disorder related to healthy control with the association of executive function. METHODS A cross-sectional case-control study was used to assess 60 subjects divided into 20 patients with single episode bipolar I disorder, 20 individuals with single episode brief psychotic disorder (both groups of patients were in remission), and 20 healthy controls. The present study examined the superior longitudinal fasciculus (SLF), and cingulum bundle fractional anisotropy (FA) determined from DTI images symmetrically and connected these results with cognitive functions as assessed by the trail making test (TMT) and Wisconsin card sorting test (WCST). RESULTS DTI data indicated that the psychotic group had a significant decrease in FA of the right SLF (p-value less than 0.001), left SLF (p-value less than 0.001), and left cingulum (p-value less than 0.001) than the bipolar I group. In terms of executive functioning, the psychotic group performed significantly worse than the bipolar I group on the TMT part B (p-value less than 0.001), the WCST (number of classifications fulfilled) (p-value less than 0.001), and perseverative errors (p-value less than 0.001). CONCLUSION Even after clinical remission, individuals with single episode brief psychotic disorder had more pronounced white matter impairments and executive function deficiencies than individuals with single episode bipolar I disorder.
Collapse
Affiliation(s)
- Zeinab El Nagar
- Institute of Psychiatry, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Heba H. El Shahawi
- Institute of Psychiatry, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Safeya M. Effat
- Institute of Psychiatry, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mona M. El Sheikh
- Institute of Psychiatry, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ahmed Adel
- Institute of Psychiatry, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Yosra A. Ibrahim
- Radiology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ola M. Aufa
- Institute of Psychiatry, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
45
|
Valdés-Tovar M, Rodríguez-Ramírez AM, Rodríguez-Cárdenas L, Sotelo-Ramírez CE, Camarena B, Sanabrais-Jiménez MA, Solís-Chagoyán H, Argueta J, López-Riquelme GO. Insights into myelin dysfunction in schizophrenia and bipolar disorder. World J Psychiatry 2022; 12:264-285. [PMID: 35317338 PMCID: PMC8900585 DOI: 10.5498/wjp.v12.i2.264] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/10/2021] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
Schizophrenia and bipolar disorder are disabling psychiatric disorders with a worldwide prevalence of approximately 1%. Both disorders present chronic and deteriorating prognoses that impose a large burden, not only on patients but also on society and health systems. These mental illnesses share several clinical and neurobiological traits; of these traits, oligodendroglial dysfunction and alterations to white matter (WM) tracts could underlie the disconnection between brain regions related to their symptomatic domains. WM is mainly composed of heavily myelinated axons and glial cells. Myelin internodes are discrete axon-wrapping membrane sheaths formed by oligodendrocyte processes. Myelin ensheathment allows fast and efficient conduction of nerve impulses through the nodes of Ranvier, improving the overall function of neuronal circuits. Rapid and precisely synchronized nerve impulse conduction through fibers that connect distant brain structures is crucial for higher-level functions, such as cognition, memory, mood, and language. Several cellular and subcellular anomalies related to myelin and oligodendrocytes have been found in postmortem samples from patients with schizophrenia or bipolar disorder, and neuroimaging techniques have revealed consistent alterations at the macroscale connectomic level in both disorders. In this work, evidence regarding these multilevel alterations in oligodendrocytes and myelinated tracts is discussed, and the involvement of proteins in key functions of the oligodendroglial lineage, such as oligodendrogenesis and myelination, is highlighted. The molecular components of the axo-myelin unit could be important targets for novel therapeutic approaches to schizophrenia and bipolar disorder.
Collapse
Affiliation(s)
- Marcela Valdés-Tovar
- Departamento de Farmacogenética, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | | | - Leslye Rodríguez-Cárdenas
- Departamento de Farmacogenética, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Carlo E Sotelo-Ramírez
- Departamento de Farmacogenética, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
- Doctorado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico
| | - Beatriz Camarena
- Departamento de Farmacogenética, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | | | - Héctor Solís-Chagoyán
- Laboratorio de Neurofarmacología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Jesús Argueta
- Doctorado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico
- Laboratorio de Neurofarmacología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Germán Octavio López-Riquelme
- Laboratorio de Socioneurobiología, Centro de Investigación en Ciencias Cognitivas, Universidad del Estado de Morelos, Cuernavaca 62209, Morelos, Mexico
| |
Collapse
|
46
|
What Can We Learn from Animal Models to Study Schizophrenia? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1400:15-33. [DOI: 10.1007/978-3-030-97182-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
47
|
Cai J, Wei W, Zhao L, Li M, Li X, Liang S, Deng W, Du XD, Wang Q, Guo WJ, Ma X, Sham PC, Li T. Abnormal Brain Structure Morphology in Early-Onset Schizophrenia. Front Psychiatry 2022; 13:925204. [PMID: 35873260 PMCID: PMC9301254 DOI: 10.3389/fpsyt.2022.925204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/31/2022] [Indexed: 11/25/2022] Open
Abstract
With less exposure to environmental and medication influences, individuals with early-onset schizophrenia (EOS) may provide valuable evidence to study the pathogenesis and phenotypic pattern of schizophrenia.T1-weighted magnetic resonance images were collected in 60 individuals with EOS and 40 healthy controls. Voxel-based morphometry and surface-based morphometry analyzes were performed. Gray matter volume, cortical thickness and cortical surface area were compared between the EOS and healthy controls and among schizophrenia subgroups (with or without family history of schizophrenia). Compared with healthy controls, the EOS group had reduced gray matter volume in the bilateral middle temporal gyrus and reduced cortical thickness in several brain regions. The sporadic early onset schizophrenia and the familial early onset schizophrenia showed different brain structure morphology. These findings suggest that abnormal brain structure morphology, especially in the temporal and frontal lobes, may be an important pathophysiological feature of EOS.
Collapse
Affiliation(s)
- Jia Cai
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, China
| | - Wei Wei
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, China
| | - Liansheng Zhao
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, China
| | - Mingli Li
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, China
| | - Xiaojing Li
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, China
| | - Sugai Liang
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Deng
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiang Dong Du
- Suzhou Psychiatry Hospital, Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Qiang Wang
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, China
| | - Wan-Jun Guo
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, China
| | - Xiaohong Ma
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, China
| | - Pak C Sham
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.,Center for PanorOmic Sciences, The University of Hong Kong, Hong Kong SAR, China.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Tao Li
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, China.,Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
48
|
Osuch E, Ursano R, Li H, Webster M, Hough C, Fullerton C, Leskin G. Brain Environment Interactions: Stress, Posttraumatic Stress Disorder, and the Need for a Postmortem Brain Collection. Psychiatry 2022; 85:113-145. [PMID: 35588486 DOI: 10.1080/00332747.2022.2068916] [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] [Indexed: 10/18/2022]
Abstract
Stress, especially the extreme stress of traumatic events, can alter both neurobiology and behavior. Such extreme environmental situations provide a useful model for understanding environmental influences on human biology and behavior. This paper will review some of the evidence of brain alterations that occur with exposure to environmental stress. This will include recent studies using neuroimaging and will address the need for histological confirmation of imaging study results. We will review the current scientific approaches to understanding brain environment interactions, and then make the case for the collection and study of postmortem brain tissue for the advancement of our understanding of the effects of environment on the brain.Creating a brain tissue collection specifically for the investigation of the effects of extreme environmental stressors fills a gap in the current research; it will provide another of the important pieces to the puzzle that constitutes the scientific investigation of negative effects of environmental exposures. Such a resource will facilitate new discoveries related to the psychiatric illnesses of acute stress disorder and posttraumatic stress disorder, and can enable scientists to correlate structural and functional imaging findings with tissue abnormalities, which is essential to validate the results of recent imaging studies.
Collapse
|
49
|
Elevated endogenous GDNF induces altered dopamine signalling in mice and correlates with clinical severity in schizophrenia. Mol Psychiatry 2022; 27:3247-3261. [PMID: 35618883 PMCID: PMC9708553 DOI: 10.1038/s41380-022-01554-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/08/2022]
Abstract
Presynaptic increase in striatal dopamine is the primary dopaminergic abnormality in schizophrenia, but the underlying mechanisms are not understood. Here, we hypothesized that increased expression of endogenous GDNF could induce dopaminergic abnormalities that resemble those seen in schizophrenia. To test the impact of GDNF elevation, without inducing adverse effects caused by ectopic overexpression, we developed a novel in vivo approach to conditionally increase endogenous GDNF expression. We found that a 2-3-fold increase in endogenous GDNF in the brain was sufficient to induce molecular, cellular, and functional changes in dopamine signalling in the striatum and prefrontal cortex, including increased striatal presynaptic dopamine levels and reduction of dopamine in prefrontal cortex. Mechanistically, we identified adenosine A2a receptor (A2AR), a G-protein coupled receptor that modulates dopaminergic signalling, as a possible mediator of GDNF-driven dopaminergic abnormalities. We further showed that pharmacological inhibition of A2AR with istradefylline partially normalised striatal GDNF and striatal and cortical dopamine levels in mice. Lastly, we found that GDNF levels are increased in the cerebrospinal fluid of first episode psychosis patients, and in post-mortem striatum of schizophrenia patients. Our results reveal a possible contributor for increased striatal dopamine signalling in a subgroup of schizophrenia patients and suggest that GDNF-A2AR crosstalk may regulate dopamine function in a therapeutically targetable manner.
Collapse
|
50
|
Förster A, Model V, Gos T, Frodl T, Schiltz K, Dobrowolny H, Meyer-Lotz G, Guest PC, Mawrin C, Bernstein HG, Bogerts B, Schlaaff K, Steiner J. Reduced GABAergic neuropil and interneuron profiles in schizophrenia: Complementary analysis of disease course-related differences. J Psychiatr Res 2021; 145:50-59. [PMID: 34864489 DOI: 10.1016/j.jpsychires.2021.11.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 10/12/2021] [Accepted: 11/17/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND GABAergic interneuron dysfunction has been implicated in the pathophysiology of schizophrenia. Expression of glutamic acid decarboxylase (GAD), a key enzyme in GABA synthesis, may also be altered. Here, we have simultaneously evaluated GAD-immunoreactive (GAD-ir) neuropil and cell profiles in schizophrenia-relevant brain regions, and analysed disease-course related differences. METHODS GAD65/67 immunoreactivity was quantified in specific brain regions for profiles of fibres and cell bodies of interneurons by automated digital image analysis in post-mortem brains of 16 schizophrenia patients from paranoid (n = 10) and residual (n = 6) diagnostic subgroups and 16 matched controls. Regions of interest were superior temporal gyrus (STG) layers III and V, mediodorsal (MD) and laterodorsal (LD) thalamus, and hippocampal CA1 and dentate gyrus (DG) regions. RESULTS A reduction in GAD-ir neuropil profiles (p < 0.001), particularly in STG layer V (p = 0.012) and MD (p = 0.001), paralleled decreased GAD-ir cell profiles (p = 0.029) in schizophrenia patients compared to controls. Paranoid schizophrenia patients had lower GAD-ir neuron cell profiles in STG layers III (p = 0.007) and V (p = 0.001), MD (p = 0.002), CA1 (p = 0.001) and DG (p = 0.043) than residual patients. There was no difference in GAD-ir neuropil profiles between paranoid and residual subgroups (p = 0.369). CONCLUSIONS These results support the hypothesis of GABAergic dysfunction in schizophrenia. They show a more prominent reduction of GAD-ir interneurons in paranoid versus residual patients, suggestive of more pronounced GABAergic dysfunction in the former. Fully automated analyses of histological sections represent a step towards user-independent assessment of brain structure.
Collapse
Affiliation(s)
- Antonia Förster
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; German Center for Mental Health (DZP), Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Vera Model
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; German Center for Mental Health (DZP), Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Tomasz Gos
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Department of Forensic Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Thomas Frodl
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; German Center for Mental Health (DZP), Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Kolja Schiltz
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany; Department of Forensic Psychiatry, Mental Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Henrik Dobrowolny
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; German Center for Mental Health (DZP), Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Gabriela Meyer-Lotz
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; German Center for Mental Health (DZP), Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Christian Mawrin
- Center for Behavioral Brain Sciences, Magdeburg, Germany; Department of Neuropathology, University of Magdeburg, Magdeburg, Germany
| | - Hans-Gert Bernstein
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Bernhard Bogerts
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany; Salus Institute, Magdeburg, Germany
| | - Konstantin Schlaaff
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; German Center for Mental Health (DZP), Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Johann Steiner
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; German Center for Mental Health (DZP), Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany.
| |
Collapse
|