1
|
Nisha Aji K, Lalang N, Ramos-Jiménez C, Rahimian R, Mechawar N, Turecki G, Chartrand D, Boileau I, Meyer JH, Rusjan PM, Mizrahi R. Evidence of altered monoamine oxidase B, an astroglia marker, in early psychosis and high-risk state. Mol Psychiatry 2025; 30:2049-2058. [PMID: 39511452 DOI: 10.1038/s41380-024-02816-x] [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: 03/09/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024]
Abstract
A novel radiotracer, [11C]SL25.1188, targets monoamine oxidase-B (MAO-B) enzyme, found primarily in astrocytes, which metabolizes monoamines (including dopamine), particularly in subcortical regions. Altered astrocyte function in schizophrenia is supported by convergent evidence from post-mortem, genetic, transcriptomic, peripheral and preclinical findings. We aimed to test whether levels of MAO-B, an index of astrocyte function are low in the living brains of early psychosis and their high-risk states. Thirty-eight participants including antipsychotic-free/minimally exposed clinical participants with first-episode psychosis (FEP), clinical high-risk (CHR) individuals and healthy volunteers (HVs) underwent a 90-min positron emission tomography (PET) scan with [11C]SL25.1188, to measure MAO-B VT, an index of MAO-B concentration. Participants were excluded if tested positive on urine drug screen (except for cannabis). This study of 14 FEP (mean[SD] age, 25.7[5.7] years; 6 F), 7 CHR (mean[SD] age, 20.9[3.7] years; 4 F) and 17 HV (mean[SD] age, 31.2[13.9] years; 9 F) demonstrated significant group differences in regional MAO-B VT (F(2,37.42) = 4.56, p = 0.02, Cohen's f = 0.49), controlling for tobacco (F (1,37.42) = 5.37, p = 0.03) and cannabis use (F(1,37.42) = 5.11, p = 0.03) with significantly lower MAO-B VT in CHR compared to HV (Cohen's d = 0.99). We report a significant cannabis effect on MAO-B VT (F(1,39.19) = 12.57, p = 0.001, Cohen's f = 0.57), with a significant group-by-cannabis interaction (F(2,37.30) = 3.82, p = 0.03, Cohen's f = 0.45), indicating lower MAO-B VT in cannabis-using clinical groups. Lower MAO-B VT levels were more robust in striatal than cortical regions, in both clinical groups (F(12,46.84) = 2.08, p = 0.04, Cohen's f = 0.73) and in cannabis users (F(6,46.84) = 6.42, p < 0.001, Cohen's f = 0.91). Lower MAO-B concentration supports astrocyte dysfunction in cannabis-using CHR and FEP clinical populations. Lower MAO-B is consistent with replicated striatal dopamine elevation in psychosis, as well as astrocyte dysfunction in schizophrenia.
Collapse
Affiliation(s)
- Kankana Nisha Aji
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
- Clinical and Translational Sciences Lab, Douglas Research Centre, Montreal, QC, Canada
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Nittha Lalang
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
- Vertex Pharmaceuticals, Boston, MA, USA
| | - Christian Ramos-Jiménez
- Clinical and Translational Sciences Lab, Douglas Research Centre, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Reza Rahimian
- Douglas Mental Health University Institute, McGill Group for Suicide Studies, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Naguib Mechawar
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill Group for Suicide Studies, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Gustavo Turecki
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill Group for Suicide Studies, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Daniel Chartrand
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Department of Anesthesia, Montreal Neurological Institute, Montreal, QC, Canada
| | - Isabelle Boileau
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Jeffrey H Meyer
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Pablo M Rusjan
- Clinical and Translational Sciences Lab, Douglas Research Centre, Montreal, QC, Canada.
- Department of Psychiatry, McGill University, Montreal, QC, Canada.
| | - Romina Mizrahi
- Clinical and Translational Sciences Lab, Douglas Research Centre, Montreal, QC, Canada.
- Department of Psychiatry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
2
|
Muszyński J, Bienert A, Elsorady RW, Rybakowski F. New pharmacological approaches in the treatment of schizophrenia. Pharmacol Rep 2025:10.1007/s43440-025-00722-9. [PMID: 40198498 DOI: 10.1007/s43440-025-00722-9] [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: 02/14/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/10/2025]
Abstract
Schizophrenia is a primary health concern, imposing a significant burden on both patients and healthcare systems globally. It is a disease with a complex etiology in which both genetic and environmental factors are involved. Despite numerous studies, the mechanism of its origin is still not fully understood. The hypotheses are synaptic, serotonergic, muscarinic, dopaminergic, microRNA-related, and neurodegenerative theories. Treatment to date is mainly based on antipsychotic drugs that act on the dopaminergic system. Although they are effective in reducing positive symptoms, their effect on negative and cognitive symptoms is limited, and their use is often associated with numerous side effects. A breakthrough in the treatment of schizophrenia came with the approval of the first drug with a non-dopaminergic mechanism of action, which opens up new therapeutic possibilities. As a result, there is intensive research into innovative substances that could increase the effectiveness of treatment and improve the quality of life of patients. In this review, we present the current state of knowledge about schizophrenia, its prevalence, risk factors, and its impact on patients' functioning. We pay special attention to new therapeutic directions, including drugs that affect systems other than the dopaminergic one, which could open up new prospects for treating the condition.
Collapse
Affiliation(s)
- Józef Muszyński
- Dr. Jan Jonston Regional Multispecialty Hospital in Leszno, Leszno, Poland
| | - Agnieszka Bienert
- Department of Pharmacology, Poznan University of Medical Sciences, Rokietnicka 3, Poznań, 60-806, Poland.
| | - Rasha Wafaie Elsorady
- Head of Clinical Pharmacy Departments at Alexandria University Hospitals, Alexandria University, Alexandria, 21523, Egypt
| | - Filip Rybakowski
- Head of Adult Psychiatry Clinic, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
3
|
Byrne RAJ, Nimmo J, Torvell M, Carpanini SM, Daskoulidou N, Hughes TR, Noble LV, Veteleanu A, Watkins LM, Zelek WM, O'Donovan MC, Morgan BP. The schizophrenia-associated gene CSMD1 encodes a complement classical pathway inhibitor predominantly expressed by astrocytes and at synapses in mice and humans. Brain Behav Immun 2025; 127:287-302. [PMID: 40112933 DOI: 10.1016/j.bbi.2025.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025] Open
Abstract
CUB and sushi multiple domains 1 (CSMD1) is predominantly expressed in brain and robustly associated with schizophrenia risk; however, understanding of which cells express CSMD1 in brain and how it impacts risk is lacking. CSMD1 encodes a large transmembrane protein including fifteen tandem short consensus repeats (SCRs), resembling complement C3 convertase regulators. CSMD1 complement regulatory activity has been reported and mapped to SCR17-21. We expressed two SCR domains of CSMD1, SCR17-21 and SCR23-26, and characterised their complement regulatory activity using a panel of functional assays testing convertase and terminal pathway inhibition. Both domains inhibited the classical pathway C3 convertase by acting as factor I cofactors; neither domain caused any inhibition in alternative or terminal pathway assays. Novel anti-CSMD1 monoclonal antibodies cross-reactive with human and mouse CSMD1 were generated that detected endogenous CSMD1 in human and rodent brain; immunostaining showed predominantly astrocyte and synaptic localisation of CSMD1, the latter confirmed using isolated synapses. Using iPSC-derived cells, astrocyte expression was confirmed and expression on cortical neurons demonstrated. We show that CSMD1 is a classical pathway-specific complement regulator expressed predominantly on astrocytes, neurons, and synapses in human and mouse brain. These findings will help reveal the mechanism by which CSMD1 impacts schizophrenia risk.
Collapse
Affiliation(s)
- Robert A J Byrne
- Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK.
| | - Jacqui Nimmo
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Megan Torvell
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Sarah M Carpanini
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Nikoleta Daskoulidou
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Timothy R Hughes
- Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Lucy V Noble
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Aurora Veteleanu
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Lewis M Watkins
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Wioleta M Zelek
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Michael C O'Donovan
- Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK
| | - Bryan Paul Morgan
- Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK.
| |
Collapse
|
4
|
Szabo A, Akkouh I, Osete JR, de Assis DR, Kondratskaya E, Hughes T, Ueland T, Andreassen OA, Djurovic S. NLRP3 inflammasome mediates astroglial dysregulation of innate and adaptive immune responses in schizophrenia. Brain Behav Immun 2025; 124:144-156. [PMID: 39617069 DOI: 10.1016/j.bbi.2024.11.030] [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: 06/22/2024] [Revised: 11/19/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
Mounting evidence indicates the involvement of neuroinflammation in the development of schizophrenia (SCZ), but the potential role of astroglia in this phenomenon remains poorly understood. We assessed the molecular and functional consequences of inflammasome activation using induced pluripotent stem cell (iPSC)-derived astrocytes generated from SCZ patients and healthy controls (CTRL). Screening protein levels in astrocytes at baseline identified lower expression of the NLRP3-ASC complex in SCZ, but increased Caspase-1 activity upon specific NLRP3 stimulation compared to CTRL. Using transcriptional profiling, we found corresponding downregulations of NLRP3 and ASC/PYCARD in both iPSC-derived astrocytes, and in a large (n = 429) brain postmortem case-control sample. Functional analyses following NLRP3 activation revealed an inflammatory phenotype characterized by elevated production of IL-1β/IL-18 and skewed priming of helper T lymphocytes (Th1/Th17) by SCZ astrocytes. This phenotype was rescued by specific inhibition of NLRP3 activation, demonstrating its dependence on the NLRP3 inflammasome. Taken together, SCZ iPSC-astrocytes display unique, NLRP3-dependent inflammatory characteristics that are manifested via various cellular functions, as well as via dysregulated innate and adaptive immune responses.
Collapse
Affiliation(s)
- Attila Szabo
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway.
| | - Ibrahim Akkouh
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Jordi Requena Osete
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Denis Reis de Assis
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Elena Kondratskaya
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Timothy Hughes
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Norway; K.G. Jebsen Thrombosis Research and Expertise Centre, University of Tromsø, Tromsø, Norway
| | - Ole A Andreassen
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
5
|
Antunes ASLM, Reis-de-Oliveira G, Martins-de-Souza D. Molecular overlaps of neurological manifestations of COVID-19 and schizophrenia from a proteomic perspective. Eur Arch Psychiatry Clin Neurosci 2025; 275:109-122. [PMID: 39028452 DOI: 10.1007/s00406-024-01842-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 06/06/2024] [Indexed: 07/20/2024]
Abstract
COVID-19, a complex multisystem disorder affecting the central nervous system, can also have psychiatric sequelae. In addition, clinical evidence indicates that a diagnosis of a schizophrenia spectrum disorder is a risk factor for mortality in patients with COVID-19. In this study, we aimed to explore brain-specific molecular aspects of COVID-19 by using a proteomic approach. We analyzed the brain proteome of fatal COVID-19 cases and compared it with differentially regulated proteins found in postmortem schizophrenia brains. The COVID-19 proteomic dataset revealed a strong enrichment of proteins expressed by glial and neuronal cells and processes related to diseases with a psychiatric and neurodegenerative component. Specifically, the COVID-19 brain proteome enriches processes that are hallmark features of schizophrenia. Furthermore, we identified shared and distinct molecular pathways affected in both conditions. We found that brain ageing processes are likely present in both COVID-19 and schizophrenia, albeit possibly driven by distinct processes. In addition, alterations in brain cell metabolism were observed, with schizophrenia primarily impacting amino acid metabolism and COVID-19 predominantly affecting carbohydrate metabolism. The enrichment of metabolic pathways associated with astrocytic components in both conditions suggests the involvement of this cell type in the pathogenesis. Both COVID-19 and schizophrenia influenced neurotransmitter systems, but with distinct impacts. Future studies exploring the underlying mechanisms linking brain ageing and metabolic dysregulation may provide valuable insights into the complex pathophysiology of these conditions and the increased vulnerability of schizophrenia patients to severe outcomes.
Collapse
Affiliation(s)
- André S L M Antunes
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil.
| | | | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, University of Campinas, Campinas, Brazil.
- D'or Institute for Research and Education, São Paulo, Brazil.
- Experimental Medicine Research Cluster (EMRC), Estate University of Campinas, Campinas, Brazil.
- INCT in Modelling Human Complex Diseases with 3D Platforms (Model3D), INCT in Modelling Human Complex Diseases with 3D Platforms (Model3D), São Paulo, Brazil.
| |
Collapse
|
6
|
Rawani NS, Chan AW, Todd KG, Baker GB, Dursun SM. The Role of Neuroglia in the Development and Progression of Schizophrenia. Biomolecules 2024; 15:10. [PMID: 39858403 PMCID: PMC11761573 DOI: 10.3390/biom15010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/04/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
Schizophrenia is a complex heterogenous disorder thought to be caused by interactions between genetic and environmental factors. The theories developed to explain the etiology of schizophrenia have focused largely on the dysfunction of neurotransmitters such as dopamine, serotonin and glutamate with their receptors, although research in the past several decades has indicated strongly that other factors are also involved and that the role of neuroglial cells in psychotic disorders including schizophrenia should be given more attention. Although glia were originally thought to be present in the brain only to support neurons in a physical, metabolic and nutritional capacity, it has become apparent that these cells have a variety of important physiological roles and that abnormalities in their function may make significant contributions to the symptoms of schizophrenia. In the present paper, we review the interactions of brain microglia, astrocytes and oligodendroglia with aspects such as transmitter dysregulation, neuro-inflammation, oxidative stress, synaptic function, the gut microbiome, myelination and the blood-brain barrier that appear to affect the cause, development and treatment of schizophrenia. We also review crosstalk between microglia, astrocytes and oligodendrocytes and the effects of antipsychotics on neuroglia. Problems associated with studies on specific biomarkers for glia in schizophrenia are discussed.
Collapse
Affiliation(s)
| | | | | | - Glen B. Baker
- Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Department of Psychiatry and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2G3, Canada; (N.S.R.); (A.W.C.); (K.G.T.); (S.M.D.)
| | | |
Collapse
|
7
|
Yao G, Luo J, Li J, Feng K, Liu P, Xu Y. Functional gradient dysfunction in drug-naïve first-episode schizophrenia and its correlation with specific transcriptional patterns and treatment predictions. Psychol Med 2024:1-13. [PMID: 39552400 DOI: 10.1017/s0033291724001739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
BACKGROUND First-episode schizophrenia (FES) is a progressive psychiatric disorder influenced by genetics, environmental factors, and brain function. The functional gradient deficits of drug-naïve FES and its relationship to gene expression profiles and treatment outcomes are unknown. METHODS In this study, we engaged a cohort of 116 FES and 100 healthy controls (HC), aged 7 to 30 years, including 15 FES over an 8-week antipsychotic medication regimen. Our examination focused on primary-to-transmodal alterations in voxel-based connection gradients in FES. Then, we employed network topology, Neurosynth, postmortem gene expression, and support vector regression to evaluate integration and segregation functions, meta-analytic cognitive terms, transcriptional patterns, and treatment predictions. RESULTS FES displayed diminished global connectome gradients (Cohen's d = 0.32-0.57) correlated with compensatory integration and segregation functions (Cohen's d = 0.31-0.36). Predominant alterations were observed in the default (67.6%) and sensorimotor (21.9%) network, related to high-order cognitive functions. Furthermore, we identified notable overlaps between partial least squares (PLS1) weighted genes and dysregulated genes in other psychiatric conditions. Genes linked with gradient alterations were enriched in synaptic signaling, neurodevelopment process, specific astrocytes, cortical layers (layer II and IV), and developmental phases from late/mid fetal to young adulthood. Additionally, the onset age influenced the severity of FES, with discernible differences in connection gradients between minor- and adult-FES. Moreover, the connectivity gradients of FES at baseline significantly predicted treatment outcomes. CONCLUSIONS These results offer significant theoretical foundations for elucidating the intricate interplay between macroscopic functional connection gradient changes and microscopic transcriptional patterns during the onset and progression of FES.
Collapse
Affiliation(s)
- Guanqun Yao
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, 030001, China
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Jing Luo
- School of Medicine, Tsinghua University, Beijing, 100084, China
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Jing Li
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, 030001, China
- College of Humanities and Social Science, Shanxi Medical University, Taiyuan, 030001, China
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Kun Feng
- School of Medicine, Tsinghua University, Beijing, 100084, China
- Department of Psychiatry, Yuquan Hospital, Tsinghua University, Beijing, 100040, China
| | - Pozi Liu
- School of Medicine, Tsinghua University, Beijing, 100084, China
- Department of Psychiatry, Yuquan Hospital, Tsinghua University, Beijing, 100040, China
| | - Yong Xu
- Department of Clinical Psychology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518031, China
| |
Collapse
|
8
|
Santarriaga S, Gerlovin K, Layadi Y, Karmacharya R. Human stem cell-based models to study synaptic dysfunction and cognition in schizophrenia: A narrative review. Schizophr Res 2024; 273:78-97. [PMID: 36925354 PMCID: PMC10500041 DOI: 10.1016/j.schres.2023.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023]
Abstract
Cognitive impairment is the strongest predictor of functional outcomes in schizophrenia and is hypothesized to result from synaptic dysfunction. However, targeting synaptic plasticity and cognitive deficits in patients remains a significant clinical challenge. A comprehensive understanding of synaptic plasticity and the molecular basis of learning and memory in a disease context can provide specific targets for the development of novel therapeutics targeting cognitive impairments in schizophrenia. Here, we describe the role of synaptic plasticity in cognition, summarize evidence for synaptic dysfunction in schizophrenia and demonstrate the use of patient derived induced-pluripotent stem cells for studying synaptic plasticity in vitro. Lastly, we discuss current advances and future technologies for bridging basic science research of synaptic dysfunction with clinical and translational research that can be used to predict treatment response and develop novel therapeutics.
Collapse
Affiliation(s)
- Stephanie Santarriaga
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Kaia Gerlovin
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yasmine Layadi
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chimie ParisTech, Université Paris Sciences et Lettres, Paris, France
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
9
|
Cao H, Fu L, Liu D, Baranova A, Zhang F. Mendelian randomization analysis of causal and druggable circulating inflammatory proteins in schizophrenia. Front Psychiatry 2024; 15:1465291. [PMID: 39544374 PMCID: PMC11560794 DOI: 10.3389/fpsyt.2024.1465291] [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: 07/16/2024] [Accepted: 10/09/2024] [Indexed: 11/17/2024] Open
Abstract
Background Schizophrenia (SZ) is a severe mental disorder with complex origins. Observational studies suggested that inflammatory factors may play a role in the pathophysiology of SZ and we aim to investigate the potential genetic connection between them by examining the causal impact of circulating inflammatory proteins on SZ. Methods We utilized Mendelian randomization (MR) analysis to assess the causal relationship between circulating inflammatory proteins and SZ and the GWAS summary datasets were sourced from public databases. The SZ dataset comprised 74,776 cases and 101,023 controls, while the summary results for 91 plasma proteins in 14,824 participants were obtained through the Olink Target platform. Moreover, to identify and evaluate potential drug targets, we searched the Drug-Gene Interaction Database (DGIdb). Results The results of the MR study confirmed that nine inflammatory proteins had a causal effect on SZ. Among these proteins, IL1A (OR: 0.93), TNFB (OR: 0.94), TNFSF14 (OR: 0.96), and CD40 (OR: 0.95) exhibited protective effects against SZ. Conversely, CCL23 (OR: 1.04), CCL19 (OR: 1.04), 4EBP1 (OR: 1.06), TWEAK (OR: 1.08), and DNER (OR: 1.10) were associated with an increased risk of SZ. The MR-Egger and weighted median methods also supported the direction of these effects. According to the Gene-Drug analysis, LTA, IL1A, CD40, and 4EBP1 can serve as drug targets. Conclusions Our study established causal relationships between circulating inflammatory proteins and SZ. It may be beneficial to personalize the treatment of SZ by incorporating inflammation management into the treatment regimen.
Collapse
Affiliation(s)
- Hongbao Cao
- School of Systems Biology, George Mason University, Manassas, VA, United States
| | - Li Fu
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dongming Liu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Ancha Baranova
- School of Systems Biology, George Mason University, Manassas, VA, United States
- Research Centre for Medical Genetics, Moscow, Russia
| | - Fuquan Zhang
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Institute of Neuropsychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
10
|
Nie FY, Jin RY, Wu SS, Yuan W, Wu YW, Xue SM, Yang XH, Qiao HF. AQP4 is upregulated in schizophrenia and Its inhibition attenuates MK-801-induced schizophrenia-like behaviors in mice. Behav Brain Res 2024; 475:115220. [PMID: 39214422 DOI: 10.1016/j.bbr.2024.115220] [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: 05/13/2024] [Revised: 08/17/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The pathophysiology and molecular mechanisms of schizophrenia (SCZ) remain unclear, and the effective treatment resources are still limited. The goal of this study is to identify the expression of AQP4 in SCZ patients and explore whether AQP4 inhibition could ameliorate schizophrenia-like behaviors and its mechanisms. METHODS Microarray datasets of PFC compared with healthy control were searched in the Gene Expression Omnibus (GEO) database, and differentially expressed genes (DEGs) were analyzed with the GEO2R online tool. The Venny online tool and metascape online software were used to identify common abnormally expressed genes and conduct cell type signature enrichment analysis. SCZ mouse models were induced with MK-801, an NMDA receptor antagonist (intraperitoneal injection, 0.1 mg/kg/day for 7 days), and C6 cell models were treated with 100 μM MK-801. RT-qPCR, Western Blotting, and immunofluorescence were employed to determine the expression of AQP4, proinflammatory cytokines, and GFAP. Open field tests and social interaction tests were performed to evaluate the schizophrenia-like behaviors. RESULTS Bioinformatics analysis identified upregulation of AQP4 in the PFC of SCZ patients compared with healthy controls. Cell type signature enrichment analysis showed that all three DEGs lists were strongly enriched in the FAN EMBRYONIC CTX ASTROCYTE 2 category. Upregulation of AQP4 was also observed in MK-801-treated C6 cells and the PFC of MK-801-induced SCZ mouse model. Moreover, AQP4 inhibition with TGN-020 (an inhibitor of AQP4) improved anxiety-like behavior and social novelty preference defects in MK-801-treated mice. AQP4 inhibition also reduced the expression of IL-1β, IL-6, and TNF-α in MK-801-treated C6 cells and mouse model. CONCLUSIONS AQP4 is upregulated in the PFC of SCZ patients compared with healthy controls. AQP4 inhibition could alleviate the anxiety-like behavior and social novelty defects in MK-801-treated mice, this may be due to the role of AQP4 in the regulation of the expression of proinflammatory cytokines.
Collapse
Affiliation(s)
- Fa-Yi Nie
- Shaanxi Key Laboratory of Acupuncture and Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China; Shaanxi Collaborative Innovation Center of TCM Technologies and Devices, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Ru-Yi Jin
- Shaanxi Key Lab Basic & New Herbal Medicament Research Center, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Shan-Shan Wu
- School of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Wei Yuan
- Shaanxi Key Laboratory of Acupuncture and Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Yu-Wei Wu
- Shaanxi Key Laboratory of Acupuncture and Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Si-Meng Xue
- Shaanxi Key Laboratory of Acupuncture and Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Xiao-Hang Yang
- Shaanxi Collaborative Innovation Center of TCM Technologies and Devices, Shaanxi University of Chinese Medicine, Xianyang 712046, China.
| | - Hai-Fa Qiao
- Shaanxi Key Laboratory of Acupuncture and Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China.
| |
Collapse
|
11
|
Sharkey RJ, Bacon C, Peterson Z, Rootes-Murdy K, Salvador R, Pomarol-Clotet E, Karuk A, Homan P, Ji E, Omlor W, Homan S, Georgiadis F, Kaiser S, Kirschner M, Ehrlich S, Dannlowski U, Grotegerd D, Goltermann J, Meinert S, Kircher T, Stein F, Brosch K, Krug A, Nenadic I, Sim K, Spalletta G, Banaj N, Sponheim SR, Demro C, Ramsay IS, King M, Quidé Y, Green MJ, Nguyen D, Preda A, Calhoun V, Turner J, van Erp T, Nickl-Jockschat T. Differences in the neural correlates of schizophrenia with positive and negative formal thought disorder in patients with schizophrenia in the ENIGMA dataset. Mol Psychiatry 2024; 29:3086-3096. [PMID: 38671214 PMCID: PMC11449795 DOI: 10.1038/s41380-024-02563-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 04/04/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024]
Abstract
Formal thought disorder (FTD) is a clinical key factor in schizophrenia, but the neurobiological underpinnings remain unclear. In particular, the relationship between FTD symptom dimensions and patterns of regional brain volume loss in schizophrenia remains to be established in large cohorts. Even less is known about the cellular basis of FTD. Our study addresses these major obstacles by enrolling a large multi-site cohort acquired by the ENIGMA Schizophrenia Working Group (752 schizophrenia patients and 1256 controls), to unravel the neuroanatomy of FTD in schizophrenia and using virtual histology tools on implicated brain regions to investigate the cellular basis. Based on the findings of previous clinical and neuroimaging studies, we decided to separately explore positive, negative and total formal thought disorder. We used virtual histology tools to relate brain structural changes associated with FTD to cellular distributions in cortical regions. We identified distinct neural networks positive and negative FTD. Both networks encompassed fronto-occipito-amygdalar brain regions, but positive and negative FTD demonstrated a dissociation: negative FTD showed a relative sparing of orbitofrontal cortical thickness, while positive FTD also affected lateral temporal cortices. Virtual histology identified distinct transcriptomic fingerprints associated for both symptom dimensions. Negative FTD was linked to neuronal and astrocyte fingerprints, while positive FTD also showed associations with microglial cell types. These results provide an important step towards linking FTD to brain structural changes and their cellular underpinnings, providing an avenue for a better mechanistic understanding of this syndrome.
Collapse
Affiliation(s)
- Rachel J Sharkey
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Chelsea Bacon
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Zeru Peterson
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | | | - Raymond Salvador
- FIDMAG Germanes Hospitalàries Research Foundation, CIBERSAM ISCIII, Barcelona, Spain
| | - Edith Pomarol-Clotet
- FIDMAG Germanes Hospitalàries Research Foundation, CIBERSAM ISCIII, Barcelona, Spain
| | - Andriana Karuk
- FIDMAG Germanes Hospitalàries Research Foundation, CIBERSAM ISCIII, Barcelona, Spain
| | - Philipp Homan
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich (PUK), Zurich, 8008, Switzerland
| | - Ellen Ji
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich (PUK), Zurich, 8008, Switzerland
| | - Wolfgang Omlor
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich (PUK), Zurich, 8008, Switzerland
| | - Stephanie Homan
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich (PUK), Zurich, 8008, Switzerland
| | - Foivos Georgiadis
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich (PUK), Zurich, 8008, Switzerland
| | - Stefan Kaiser
- Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | - Matthias Kirschner
- Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | - Stefan Ehrlich
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Janik Goltermann
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Katharina Brosch
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Axel Krug
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Igor Nenadic
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Kang Sim
- West Region, Institute of Mental Health, Singapore, Singapore
| | | | - Nerisa Banaj
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Scott R Sponheim
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Caroline Demro
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Ian S Ramsay
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | | | - Yann Quidé
- School of Psychiatry, University of New South Wales (UNSW) Sydney, Sydney, NSW, Australia
| | - Melissa Jane Green
- School of Psychiatry, University of New South Wales (UNSW) Sydney, Sydney, NSW, Australia
| | - Dana Nguyen
- Department of Pediatric Neurology, University of California Irvine, Irvine, CA, USA
| | - Adrian Preda
- Department of Pediatric Neurology, University of California Irvine, Irvine, CA, USA
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, USA
| | - Vince Calhoun
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GE, USA
| | - Jessica Turner
- Department of Psychiatry and Behavioral Medicine, Ohio State University, Columbus, OH, USA
| | - Theo van Erp
- Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA, USA
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, USA
| | - Thomas Nickl-Jockschat
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA.
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Department of Psychiatry and Psychotherapy, Otto-von-Guericke University, Magdeburg, Germany.
- German Center for Mental Health (DZPG), partner site Halle-Jena-Magdeburg, Magdeburg, Germany.
- Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Halle-Jena-Magdeburg, Magdeburg, Germany.
| |
Collapse
|
12
|
Shen HY. Editorial: 15 years of Frontiers in Cellular Neuroscience: the role of glial cells in schizophrenia and other related disorders. Front Cell Neurosci 2024; 18:1471266. [PMID: 39381502 PMCID: PMC11458451 DOI: 10.3389/fncel.2024.1471266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024] Open
Affiliation(s)
- Hai-Ying Shen
- Department of Pediatrics Neurology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
13
|
Vivi E, Di Benedetto B. Brain stars take the lead during critical periods of early postnatal brain development: relevance of astrocytes in health and mental disorders. Mol Psychiatry 2024; 29:2821-2833. [PMID: 38553540 PMCID: PMC11420093 DOI: 10.1038/s41380-024-02534-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 09/25/2024]
Abstract
In the brain, astrocytes regulate shape and functions of the synaptic and vascular compartments through a variety of released factors and membrane-bound proteins. An imbalanced astrocyte activity can therefore have drastic negative impacts on brain development, leading to the onset of severe pathologies. Clinical and pre-clinical studies show alterations in astrocyte cell number, morphology, molecular makeup and astrocyte-dependent processes in different affected brain regions in neurodevelopmental (ND) and neuropsychiatric (NP) disorders. Astrocytes proliferate, differentiate and mature during the critical period of early postnatal brain development, a time window of elevated glia-dependent regulation of a proper balance between synapse formation/elimination, which is pivotal in refining synaptic connectivity. Therefore, any intrinsic and/or extrinsic factors altering these processes during the critical period may result in an aberrant synaptic remodeling and onset of mental disorders. The peculiar bridging position of astrocytes between synaptic and vascular compartments further allows them to "compute" the brain state and consequently secrete factors in the bloodstream, which may serve as diagnostic biomarkers of distinct healthy or disease conditions. Here, we collect recent advancements regarding astrogenesis and astrocyte-mediated regulation of neuronal network remodeling during early postnatal critical periods of brain development, focusing on synapse elimination. We then propose alternative hypotheses for an involvement of aberrancies in these processes in the onset of ND and NP disorders. In light of the well-known differential prevalence of certain brain disorders between males and females, we also discuss putative sex-dependent influences on these neurodevelopmental events. From a translational perspective, understanding age- and sex-dependent astrocyte-specific molecular and functional changes may help to identify biomarkers of distinct cellular (dys)functions in health and disease, favouring the development of diagnostic tools or the selection of tailored treatment options for male/female patients.
Collapse
Affiliation(s)
- Eugenia Vivi
- Laboratory of Neuro-Glia Pharmacology, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Barbara Di Benedetto
- Laboratory of Neuro-Glia Pharmacology, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany.
- Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
14
|
Torres-Carmona E, Nakajima S, Iwata Y, Ueno F, Stefan C, Song J, Abdolizadeh A, Koizumi MT, Kambari Y, Amaev A, Agarwal SM, Mar W, de Luca V, Remington G, Gerretsen P, Graff-Guerrero A. Clozapine treatment and astrocyte activity in treatment resistant schizophrenia: A proton magnetic resonance spectroscopy study. Schizophr Res 2024; 270:152-161. [PMID: 38909486 DOI: 10.1016/j.schres.2024.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 04/17/2024] [Accepted: 06/15/2024] [Indexed: 06/25/2024]
Abstract
Clozapine is the only antipsychotic approved for treating treatment-resistant schizophrenia (TRS), characterized by persistent positive symptoms despite adequate antipsychotic treatment. Unfortunately, clozapine demonstrates clinical efficacy in only ~30-60 % of patients with TRS (clozapine-responders; ClzR+), while the remaining ~40-70 % are left with no pharmacological recourse for improvement (clozapine-resistant; ClzR-). Mechanism(s) underlying clozapine's superior efficacy remain unclear. However, in vitro evidence suggests clozapine may mitigate glutamatergic dysregulations observed in TRS, by modulating astrocyte activity in ClzR+, but not ClzR-. A factor that if proven correct, may help the assessment of treatment response and development of more effective antipsychotics. To explore the presence of clozapine-astrocyte interaction and clinical improvement, we used 3 T proton-magnetic resonance spectroscopy to quantify levels of myo-Inositol, surrogate biomarker of astrocyte activity, in regions related to schizophrenia neurobiology: Dorsal-anterior-cingulate-cortex (dACC), left-dorsolateral-prefrontal-cortex (left-DLPFC), and left-striatum (left-striatum) of 157 participants (ClzR- = 30; ClzR+ = 37; responders = 38; controls = 52). Clozapine treatment was assessed using clozapine to norclozapine plasma levels, 11-12 h after last clozapine dose. Measures for symptom severity (i.e., Positive and Negative Symptoms Scale) and cognition (i.e., Mini-Mental State Examination) were also recorded. Higher levels of myo-Inositol were observed in TRS groups versus responders and controls (dACC (p < 0.001); left-striatum (p = 0.036); left-DLPFC (p = 0.023)). In ClzR+, but not ClzR-, clozapine to norclozapine ratios were positively associated with myo-Inositol levels (dACC (p = 0.004); left-DLPFC (p < 0.001)), and lower positive symptom severity (p < 0.001). Our results support growing in vitro evidence of clozapine-astrocyte interaction in clozapine-responders. Further research may determine the viability of clozapine-astrocyte interactions as an early marker of clozapine response.
Collapse
Affiliation(s)
- Edgardo Torres-Carmona
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Shinichiro Nakajima
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Department of Neuropsychiatry, Keio University, Minato, Tokyo, Japan
| | - Yusuke Iwata
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Fumihiko Ueno
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Cristiana Stefan
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Jianmeng Song
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Ali Abdolizadeh
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | | | - Yasaman Kambari
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Aron Amaev
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Sri Mahavir Agarwal
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada
| | - Wanna Mar
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Vincenzo de Luca
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada
| | - Gary Remington
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada
| | - Philip Gerretsen
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada
| | - Ariel Graff-Guerrero
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada.
| |
Collapse
|
15
|
Rawani NS, Chan AW, Dursun SM, Baker GB. The Underlying Neurobiological Mechanisms of Psychosis: Focus on Neurotransmission Dysregulation, Neuroinflammation, Oxidative Stress, and Mitochondrial Dysfunction. Antioxidants (Basel) 2024; 13:709. [PMID: 38929148 PMCID: PMC11200831 DOI: 10.3390/antiox13060709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/16/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Psychosis, defined as a set of symptoms that results in a distorted sense of reality, is observed in several psychiatric disorders in addition to schizophrenia. This paper reviews the literature relevant to the underlying neurobiology of psychosis. The dopamine hypothesis has been a major influence in the study of the neurochemistry of psychosis and in development of antipsychotic drugs. However, it became clear early on that other factors must be involved in the dysfunction involved in psychosis. In the current review, it is reported how several of these factors, namely dysregulation of neurotransmitters [dopamine, serotonin, glutamate, and γ-aminobutyric acid (GABA)], neuroinflammation, glia (microglia, astrocytes, and oligodendrocytes), the hypothalamic-pituitary-adrenal axis, the gut microbiome, oxidative stress, and mitochondrial dysfunction contribute to psychosis and interact with one another. Research on psychosis has increased knowledge of the complexity of psychotic disorders. Potential new pharmacotherapies, including combinations of drugs (with pre- and probiotics in some cases) affecting several of the factors mentioned above, have been suggested. Similarly, several putative biomarkers, particularly those related to the immune system, have been proposed. Future research on both pharmacotherapy and biomarkers will require better-designed studies conducted on an all stages of psychotic disorders and must consider confounders such as sex differences and comorbidity.
Collapse
Affiliation(s)
| | | | | | - Glen B. Baker
- Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Department of Psychiatry and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2G3, Canada; (N.S.R.); (A.W.C.); (S.M.D.)
| |
Collapse
|
16
|
Del Casale A, Modesti MN, Gentile G, Guariglia C, Ferracuti S, Simmaco M, Borro M. Is the Hedgehog Pathway Involved in the Pathophysiology of Schizophrenia? A Systematic Review of Current Evidence of Neural Molecular Correlates and Perspectives on Drug Development. Curr Issues Mol Biol 2024; 46:5322-5336. [PMID: 38920990 PMCID: PMC11202070 DOI: 10.3390/cimb46060318] [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: 04/12/2024] [Revised: 05/09/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Among the pathophysiological correlates of schizophrenia, recent research suggests a potential role for the Hedgehog (Hh) signalling pathway, which has been traditionally studied in embryonic development and oncology. Its dysregulation may impact brain homeostasis, neuroplasticity, and potential involvement in neural processes. This systematic review provides an overview of the involvement of Hh signalling in the pathophysiology of schizophrenia and antipsychotic responses. We searched the PubMed and Scopus databases to identify peer-reviewed scientific studies focusing on Hh and schizophrenia, following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement, finally including eight studies, including three articles focused on patients with schizophrenia, two animal models of schizophrenia, two animal embryo studies, and one cellular differentiation study. The Hh pathway is crucial in the development of midbrain dopaminergic neurons, neuroplasticity mechanisms, regulating astrocyte phenotype and function, brain-derived neurotrophic factor expression, brain glutamatergic neural transmission, and responses to antipsychotics. Overall, results indicate an involvement of Hh in the pathophysiology of schizophrenia and antipsychotic responses, although an exiguity of studies characterises the literature. The heterogeneity between animal and human studies is another main limitation. Further research can lead to better comprehension and the development of novel personalised drug treatments and therapeutic interventions.
Collapse
Affiliation(s)
- Antonio Del Casale
- Department of Dynamic and Clinical Psychology and Health Studies, Faculty of Medicine and Psychology, Sapienza University of Rome, 00185 Rome, Italy;
- Unit of Psychiatry, Emergency and Admissions Department, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Martina Nicole Modesti
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, 00185 Rome, Italy
- Unit of Psychiatry, Mental Health Department, Santissimo Gonfalone Hospital, Local Health Service Roma 5, Monterotondo, 00015 Rome, Italy
| | - Giovanna Gentile
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
- Unit of Laboratory and Advanced Molecular Diagnostics, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Cecilia Guariglia
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, 00185 Rome, Italy
- Cognitive and Motor Rehabilitation and Neuroimaging Unit, Scientific Institute for Research, Hospitalization and Healthcare Fondazione Santa Lucia, 00179 Rome, Italy
| | - Stefano Ferracuti
- Department of Human Neuroscience, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00185 Rome, Italy;
- Unit of Risk Management, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Maurizio Simmaco
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
- Unit of Laboratory and Advanced Molecular Diagnostics, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Marina Borro
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
- Unit of Laboratory and Advanced Molecular Diagnostics, Sant’Andrea University Hospital, 00189 Rome, Italy
| |
Collapse
|
17
|
Cai M, Ji Y, Zhao Q, Xue H, Sun Z, Wang H, Zhang Y, Chen Y, Zhao Y, Zhang Y, Lei M, Wang C, Zhuo C, Liu N, Liu H, Liu F. Homotopic functional connectivity disruptions in schizophrenia and their associated gene expression. Neuroimage 2024; 289:120551. [PMID: 38382862 DOI: 10.1016/j.neuroimage.2024.120551] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/18/2024] [Accepted: 02/19/2024] [Indexed: 02/23/2024] Open
Abstract
It has been revealed that abnormal voxel-mirrored homotopic connectivity (VMHC) is present in patients with schizophrenia, yet there are inconsistencies in the relevant findings. Moreover, little is known about their association with brain gene expression profiles. In this study, transcription-neuroimaging association analyses using gene expression data from Allen Human Brain Atlas and case-control VMHC differences from both the discovery (meta-analysis, including 9 studies with a total of 386 patients and 357 controls) and replication (separate group-level comparisons within two datasets, including a total of 258 patients and 287 controls) phases were performed to identify genes associated with VMHC alterations. Enrichment analyses were conducted to characterize the biological functions and specific expression of identified genes, and Neurosynth decoding analysis was performed to examine the correlation between cognitive-related processes and VMHC alterations in schizophrenia. In the discovery and replication phases, patients with schizophrenia exhibited consistent VMHC changes compared to controls, which were correlated with a series of cognitive-related processes; meta-regression analysis revealed that illness duration was negatively correlated with VMHC abnormalities in the cerebellum and postcentral/precentral gyrus. The abnormal VMHC patterns were stably correlated with 1287 genes enriched for fundamental biological processes like regulation of cell communication, nervous system development, and cell communication. In addition, these genes were overexpressed in astrocytes and immune cells, enriched in extensive cortical regions and wide developmental time windows. The present findings may contribute to a more comprehensive understanding of the molecular mechanisms underlying VMHC alterations in patients with schizophrenia.
Collapse
Affiliation(s)
- Mengjing Cai
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yuan Ji
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Qiyu Zhao
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hui Xue
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zuhao Sun
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - He Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yijing Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yayuan Chen
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yao Zhao
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yujie Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Minghuan Lei
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chunyang Wang
- Department of Scientific Research, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chuanjun Zhuo
- Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PGNP_Lab), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Nana Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Huaigui Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Feng Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
18
|
Stanca S, Rossetti M, Bokulic Panichi L, Bongioanni P. The Cellular Dysfunction of the Brain-Blood Barrier from Endothelial Cells to Astrocytes: The Pathway towards Neurotransmitter Impairment in Schizophrenia. Int J Mol Sci 2024; 25:1250. [PMID: 38279249 PMCID: PMC10816922 DOI: 10.3390/ijms25021250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Schizophrenia (SCZ) is an articulated psychiatric syndrome characterized by a combination of genetic, epigenetic, and environmental factors. Our intention is to present a pathogenetic model combining SCZ alterations and the main cellular actors of the blood-brain barrier (BBB): endothelial cells (ECs), pericytes, and astrocytes. The homeostasis of the BBB is preserved by the neurovascular unit which is constituted by ECs, astrocytes and microglia, neurons, and the extracellular matrix. The role of the BBB is strictly linked to its ability to preserve the biochemical integrity of brain parenchyma integrity. In SCZ, there is an increased BBB permeability, demonstrated by elevated levels of albumin and immunoglobulins in the cerebrospinal fluid, and this is the result of an intrinsic endothelial impairment. Increased BBB permeability would lead to enhanced concentrations of neurotoxic and neuroactive molecules in the brain. The pathogenetic involvement of astrocytes in SCZ reverberates its consequences on BBB, together with the impact on its permeability and selectivity represented by the EC and pericyte damage occurring in the psychotic picture. Understanding the strict interaction between ECs and astrocytes, and its consequent impact on cognition, is diriment not only for comprehension of neurotransmitter dyshomeostasis in SCZ, but also for focusing on other potential therapeutic targets.
Collapse
Affiliation(s)
- Stefano Stanca
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi 10, 56126 Pisa, Italy
- NeuroCare Onlus, 56100 Pisa, Italy
| | - Martina Rossetti
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi 10, 56126 Pisa, Italy
- NeuroCare Onlus, 56100 Pisa, Italy
| | - Leona Bokulic Panichi
- NeuroCare Onlus, 56100 Pisa, Italy
- Neuroscience Department, Azienda Ospedaliero-Universitaria Pisana, 56100 Pisa, Italy
| | - Paolo Bongioanni
- NeuroCare Onlus, 56100 Pisa, Italy
- Neuroscience Department, Azienda Ospedaliero-Universitaria Pisana, 56100 Pisa, Italy
| |
Collapse
|
19
|
Yao G, Luo J, Zou T, Li J, Hu S, Yang L, Li X, Tian Y, Zhang Y, Feng K, Xu Y, Liu P. Transcriptional patterns of the cortical Morphometric Inverse Divergence in first-episode, treatment-naïve early-onset schizophrenia. Neuroimage 2024; 285:120493. [PMID: 38086496 DOI: 10.1016/j.neuroimage.2023.120493] [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: 09/15/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 12/18/2023] Open
Abstract
Early-onset Schizophrenia (EOS) is a profoundly progressive psychiatric disorder characterized by both positive and negative symptoms, whose pathogenesis is influenced by genes, environment and brain structure development. In this study, the MIND (Morphometric Inverse Divergence) network was employed to explore the relationship between morphological similarity and specific transcriptional expression patterns in EOS patients. This study involved a cohort of 187 participants aged between 7 and 17 years, consisting of 97 EOS patients and 90 healthy controls (HC). Multiple morphological features were used to construct the MIND network for all participants. Furthermore, we explored the associations between MIND network and brain-wide gene expression in EOS patients through partial least squares (PLS) regression, shared genetic predispositions with other psychiatric disorders, functional enrichment of PLS weighted genes, as well as transcriptional signature assessment of cell types, cortical layers, and developmental stages. The MIND showed similarity differences in the orbitofrontal cortex, pericalcarine cortex, lingual gyrus, and multiple networks in EOS patients compared to HC. Moreover, our exploration revealed a significant overlap of PLS2 weighted genes linking to EOS-related MIND differences and the dysregulated genes reported in other psychiatric diseases. Interestingly, genes correlated with MIND changes (PLS2-) exhibited a significant enrichment not only in metabolism-related pathways, but also in specific astrocytes, cortical layers (specifically layer I and III), and posterior developmental stages (late infancy to young adulthood stages). However, PLS2+ genes were primarily enriched in synapses signaling-related pathways and early developmental stages (from early-mid fetal to neonatal early infancy) but not in special cell types or layers. These findings provide a novel perspective on the intricate relationship between macroscopic morphometric structural abnormalities and microscopic transcriptional patterns during the onset and progression of EOS.
Collapse
Affiliation(s)
- Guanqun Yao
- School of Medicine, Tsinghua University, Beijing 100084, China; Department of Psychiatry, Yuquan Hospital, Tsinghua University, Beijing 100040, China; Institute for Precision Medicine, Tsinghua University, Beijing 100084, China
| | - Jing Luo
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Ting Zou
- School of Life Sciences, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Jing Li
- Department of Psychiatry, the First Hospital of Shanxi Medical University, Taiyuan 030001, China; School of Mental Health, Shanxi Medical University, Taiyuan 030001, China
| | - Shuang Hu
- Shanghai Mental Health Center, Shanghai 200030, China
| | - Langxiong Yang
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xinrong Li
- Department of Psychiatry, the First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Yu Tian
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yuqi Zhang
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Kun Feng
- School of Medicine, Tsinghua University, Beijing 100084, China; Department of Psychiatry, Yuquan Hospital, Tsinghua University, Beijing 100040, China; Institute for Precision Medicine, Tsinghua University, Beijing 100084, China.
| | - Yong Xu
- Department of Clinical Psychology, The Eighth Affiliated Hospital, Sun Yat-Sen University, No. 3025, Shennan Middle Road, Futian Street, Futian District, Shenzhen 518031, China.
| | - Pozi Liu
- School of Medicine, Tsinghua University, Beijing 100084, China; Department of Psychiatry, Yuquan Hospital, Tsinghua University, Beijing 100040, China.
| |
Collapse
|
20
|
Guo L, Ma J, Cai M, Zhang M, Xu Q, Wang H, Zhang Y, Yao J, Sun Z, Chen Y, Xue H, Zhang Y, Wang S, Xue K, Zhu D, Liu F. Transcriptional signatures of the whole-brain voxel-wise resting-state functional network centrality alterations in schizophrenia. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2023; 9:87. [PMID: 38104130 PMCID: PMC10725456 DOI: 10.1038/s41537-023-00422-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
Neuroimaging studies have revealed that patients with schizophrenia exhibit disrupted resting-state functional connectivity. However, the inconsistent findings across these studies have hindered our comprehensive understanding of the functional connectivity changes associated with schizophrenia, and the molecular mechanisms associated with these alterations remain largely unclear. A quantitative meta-analysis was first conducted on 21 datasets, involving 1057 patients and 1186 healthy controls, to examine disrupted resting-state functional connectivity in schizophrenia, as measured by whole-brain voxel-wise functional network centrality (FNC). Subsequently, partial least squares regression analysis was employed to investigate the relationship between FNC changes and gene expression profiles obtained from the Allen Human Brain Atlas database. Finally, gene enrichment analysis was performed to unveil the biological significance of the altered FNC-related genes. Compared with healthy controls, patients with schizophrenia show consistently increased FNC in the right inferior parietal cortex extending to the supramarginal gyrus, angular gyrus, bilateral medial prefrontal cortex, and right dorsolateral prefrontal cortex, while decreased FNC in the bilateral insula, bilateral postcentral gyrus, and right inferior temporal gyrus. Meta-regression analysis revealed that increased FNC in the right inferior parietal cortex was positively correlated with clinical score. In addition, these observed functional connectivity changes were found to be spatially associated with the brain-wide expression of specific genes, which were enriched in diverse biological pathways and cell types. These findings highlight the aberrant functional connectivity observed in schizophrenia and its potential molecular underpinnings, providing valuable insights into the neuropathology of dysconnectivity associated with this disorder.
Collapse
Affiliation(s)
- Lining Guo
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Juanwei Ma
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Mengjing Cai
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Minghui Zhang
- Department of Ultrasound, Tianjin Medical University General Hospital Airport Hospital, Tianjin, China
| | - Qiang Xu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - He Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Yijing Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Jia Yao
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Zuhao Sun
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Yayuan Chen
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Hui Xue
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Yujie Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Shaoying Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Kaizhong Xue
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China.
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China.
| | - Dan Zhu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China.
- Department of Radiology, Tianjin Medical University General Hospital Airport Hospital, Tianjin, China.
| | - Feng Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
21
|
Yao G, Zou T, Luo J, Hu S, Yang L, Li J, Li X, Zhang Y, Feng K, Xu Y, Liu P. Cortical structural changes of morphometric similarity network in early-onset schizophrenia correlate with specific transcriptional expression patterns. BMC Med 2023; 21:479. [PMID: 38049797 PMCID: PMC10696871 DOI: 10.1186/s12916-023-03201-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/27/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND This study aimed to investigate the neuroanatomical subtypes among early-onset schizophrenia (EOS) patients by exploring the association between structural alterations and molecular mechanisms using a combined analysis of morphometric similarity network (MSN) changes and specific transcriptional expression patterns. METHODS We recruited 206 subjects aged 7 to 17 years, including 100 EOS patients and 106 healthy controls (HC). Heterogeneity through discriminant analysis (HYDRA) was used to identify the EOS subtypes within the MSN strength. The differences in morphometric similarity between each EOS subtype and HC were compared. Furthermore, we examined the link between morphometric changes and brain-wide gene expression in different EOS subtypes using partial least squares regression (PLS) weight mapping, evaluated genetic commonalities with psychiatric disorders, identified functional enrichments of PLS-weighted genes, and assessed cellular transcriptional signatures. RESULTS Two distinct MSN-based EOS subtypes were identified, each exhibiting different abnormal MSN strength and cognitive functions compared to HC. The PLS1 score mapping demonstrated anterior-posterior gradients of gene expression in EOS1, whereas inverse distributions were observed in EOS2 cohorts. Genetic commonalities were identified in autistic disorder and adult schizophrenia with EOS1 and inflammatory bowel diseases with EOS2 cohorts. The EOS1 PLS1- genes (Z < -5) were significantly enriched in synaptic signaling-related functions, whereas EOS2 demonstrated enrichments in virtual infection-related pathways. Furthermore, the majority of observed associations with EOS1-specific MSN strength differences contributed to specific transcriptional changes in astrocytes and neurons. CONCLUSIONS The findings of this study provide a comprehensive analysis of neuroanatomical subtypes in EOS, shedding light on the intricate relationships between macrostructural and molecular aspects of the EOS disease.
Collapse
Affiliation(s)
- Guanqun Yao
- School of Medicine, Tsinghua University, Beijing, 100084, China
- Department of Psychiatry, Tsinghua University Yuquan Hospital, Shijingshan District, 5 Shijingshan Road, Beijing, China
| | - Ting Zou
- School of Life Sciences, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Jing Luo
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Shuang Hu
- Shanghai Mental Health Center, Shanghai, 200030, China
| | - Langxiong Yang
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Jing Li
- College of Humanities and Social Science, Shanxi Medical University, Taiyuan, 030001, China
- School of Mental Health, Shanxi Medical University, Taiyuan, 030001, China
- Department of Psychiatry, the First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Xinrong Li
- Department of Psychiatry, the First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Yuqi Zhang
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Kun Feng
- School of Medicine, Tsinghua University, Beijing, 100084, China.
- Department of Psychiatry, Tsinghua University Yuquan Hospital, Shijingshan District, 5 Shijingshan Road, Beijing, China.
| | - Yong Xu
- School of Mental Health, Shanxi Medical University, Taiyuan, 030001, China.
- Department of Psychiatry, the First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
- Department of Mental Health, Shanxi Medical University, Taiyuan Central Hospital of Shanxi Medical University, 256 Fen Dongnan Road, Xiaodian District, Taiyuan City, Shanxi Province, China.
| | - Pozi Liu
- School of Medicine, Tsinghua University, Beijing, 100084, China.
- Department of Psychiatry, Tsinghua University Yuquan Hospital, Shijingshan District, 5 Shijingshan Road, Beijing, China.
| |
Collapse
|
22
|
Abstract
Rett syndrome is a neurodevelopmental disorder caused by loss-of-function mutations in the methyl-CpG binding protein-2 (MeCP2) gene that is characterized by epilepsy, intellectual disability, autistic features, speech deficits, and sleep and breathing abnormalities. Neurologically, patients with all three disorders display microcephaly, aberrant dendritic morphology, reduced spine density, and an imbalance of excitatory/inhibitory signaling. Loss-of-function mutations in the cyclin-dependent kinase-like 5 (CDKL5) and FOXG1 genes also cause similar behavioral and neurobiological defects and were referred to as congenital or variant Rett syndrome. The relatively recent realization that CDKL5 deficiency disorder (CDD), FOXG1 syndrome, and Rett syndrome are distinct neurodevelopmental disorders with some distinctive features have resulted in separate focus being placed on each disorder with the assumption that distinct molecular mechanisms underlie their pathogenesis. However, given that many of the core symptoms and neurological features are shared, it is likely that the disorders share some critical molecular underpinnings. This review discusses the possibility that deregulation of common molecules in neurons and astrocytes plays a central role in key behavioral and neurological abnormalities in all three disorders. These include KCC2, a chloride transporter, vGlut1, a vesicular glutamate transporter, GluD1, an orphan-glutamate receptor subunit, and PSD-95, a postsynaptic scaffolding protein. We propose that reduced expression or activity of KCC2, vGlut1, PSD-95, and AKT, along with increased expression of GluD1, is involved in the excitatory/inhibitory that represents a key aspect in all three disorders. In addition, astrocyte-derived brain-derived neurotrophic factor (BDNF), insulin-like growth factor 1 (IGF-1), and inflammatory cytokines likely affect the expression and functioning of these molecules resulting in disease-associated abnormalities.
Collapse
Affiliation(s)
- Santosh R D’Mello
- Department of Biological Sciences, Louisiana State University Shreveport, Shreveport, LA 71104, USA
| |
Collapse
|
23
|
Nickl-Jockschat T, Sharkey R, Bacon C, Peterson Z, Rootes-Murdy K, Salvador R, Pomarol E, Karuk A, Homan P, Ji E, Omlor W, Homan S, Georgiadis F, Kaiser S, Kirschner M, Ehrlich S, Dannlowski U, Grotegerd D, Goltermann J, Meinert S, Kircher T, Stein F, Brosch K, Krug A, Nenadic I, Sim K, Piras F, Banaj N, Sponheim S, Demro C, Ramsay I, King M, Quidé Y, Green M, Nguyen D, Preda A, Calhoun V, Turner J, van Erp T, Spalletta G. Neural Correlates of Positive and Negative Formal Thought Disorder in Individuals with Schizophrenia: An ENIGMA Schizophrenia Working Group Study. RESEARCH SQUARE 2023:rs.3.rs-3179362. [PMID: 37841855 PMCID: PMC10571603 DOI: 10.21203/rs.3.rs-3179362/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Formal thought disorder (FTD) is a key clinical factor in schizophrenia, but the neurobiological underpinnings remain unclear. In particular, relationship between FTD symptom dimensions and patterns of regional brain volume deficiencies in schizophrenia remain to be established in large cohorts. Even less is known about the cellular basis of FTD. Our study addresses these major obstacles based on a large multi-site cohort through the ENIGMA Schizophrenia Working Group (752 individuals with schizophrenia and 1256 controls), to unravel the neuroanatomy of positive, negative and total FTD in schizophrenia and their cellular bases. We used virtual histology tools to relate brain structural changes associated with FTD to cellular distributions in cortical regions. We identified distinct neural networks for positive and negative FTD. Both networks encompassed fronto-occipito-amygdalar brain regions, but negative FTD showed a relative sparing of orbitofrontal cortical thickness, while positive FTD also affected lateral temporal cortices. Virtual histology identified distinct transcriptomic fingerprints associated for both symptom dimensions. Negative FTD was linked to neuronal and astrocyte fingerprints, while positive FTD was also linked to microglial cell types. These findings relate different dimensions of FTD to distinct brain structural changes and their cellular underpinnings, improve our mechanistic understanding of these key psychotic symptoms.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster
| | | | | | | | | | | | | | | | - Igor Nenadic
- Philipps University Marburg / Marburg University Hospital
| | | | | | | | | | | | | | | | | | | | | | | | - Vince Calhoun
- Georgia Institute of Technology, Emory University and Georgia State University
| | | | | | | |
Collapse
|
24
|
Martín-Hernández D, Muñoz-López M, Tendilla-Beltrán H, Caso JR, García-Bueno B, Menchén L, Leza JC. Immune System and Brain/Intestinal Barrier Functions in Psychiatric Diseases: Is Sphingosine-1-Phosphate at the Helm? Int J Mol Sci 2023; 24:12634. [PMID: 37628815 PMCID: PMC10454107 DOI: 10.3390/ijms241612634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Over the past few decades, extensive research has shed light on immune alterations and the significance of dysfunctional biological barriers in psychiatric disorders. The leaky gut phenomenon, intimately linked to the integrity of both brain and intestinal barriers, may play a crucial role in the origin of peripheral and central inflammation in these pathologies. Sphingosine-1-phosphate (S1P) is a bioactive lipid that regulates both the immune response and the permeability of biological barriers. Notably, S1P-based drugs, such as fingolimod and ozanimod, have received approval for treating multiple sclerosis, an autoimmune disease of the central nervous system (CNS), and ulcerative colitis, an inflammatory condition of the colon, respectively. Although the precise mechanisms of action are still under investigation, the effectiveness of S1P-based drugs in treating these pathologies sparks a debate on extending their use in psychiatry. This comprehensive review aims to delve into the molecular mechanisms through which S1P modulates the immune system and brain/intestinal barrier functions. Furthermore, it will specifically focus on psychiatric diseases, with the primary objective of uncovering the potential of innovative therapies based on S1P signaling.
Collapse
Affiliation(s)
- David Martín-Hernández
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Instituto de Investigación Hospital 12 de Octubre (i+12), Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain; (M.M.-L.); (J.R.C.); (B.G.-B.); (J.C.L.)
- Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII), 28029 Madrid, Spain
| | - Marina Muñoz-López
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Instituto de Investigación Hospital 12 de Octubre (i+12), Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain; (M.M.-L.); (J.R.C.); (B.G.-B.); (J.C.L.)
- Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII), 28029 Madrid, Spain
| | - Hiram Tendilla-Beltrán
- Laboratorio de Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), 72570 Puebla, Mexico;
| | - Javier R. Caso
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Instituto de Investigación Hospital 12 de Octubre (i+12), Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain; (M.M.-L.); (J.R.C.); (B.G.-B.); (J.C.L.)
- Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII), 28029 Madrid, Spain
| | - Borja García-Bueno
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Instituto de Investigación Hospital 12 de Octubre (i+12), Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain; (M.M.-L.); (J.R.C.); (B.G.-B.); (J.C.L.)
- Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII), 28029 Madrid, Spain
| | - Luis Menchén
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón, Departamento de Medicina, Universidad Complutense, Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III (CIBEREHD, ISCIII), 28029 Madrid, Spain
| | - Juan C. Leza
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Instituto de Investigación Hospital 12 de Octubre (i+12), Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain; (M.M.-L.); (J.R.C.); (B.G.-B.); (J.C.L.)
- Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII), 28029 Madrid, Spain
| |
Collapse
|
25
|
Śmierciak N, Krzyściak W, Szwajca M, Karcz P, Bryll A, Popiela TJ, Donicz P, Turek A, Aleksandrovych V, Pilecki M. Benefits and Meaning of Lipids Profile in Relation to Oxidative Balance and Brain Morphology in Schizophrenia. Int J Mol Sci 2023; 24:11375. [PMID: 37511134 PMCID: PMC10379229 DOI: 10.3390/ijms241411375] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Schizophrenia is characterized by complex metabolic dysregulations and their consequences. Until now, numerous theories have explained its pathogenesis, using a spectrum of available technologies. We focused our interest on lipid profile-periphery high-density cholesterol level and lipoproteins in the human brain and compared magnetic resonance imaging (MRI) scans of patients with schizophrenia and the healthy group. Detailed analysis of biochemical parameters was performed using magnetic resonance spectroscopy. Our study aimed to reveal correlations between periphery high-density lipoproteins levels and lipoproteins in the brain, depicted in MRI scans, and parameters of peripheral oxidative stress expressed as paraoxonase. Patients with schizophrenia have decreased levels of high-density lipoproteins, low paraoxonase activity, and slightly raised sodium in the blood. Positive significant correlations between serum high-density cholesterol and anterior cingulate cortex, unique brain area for schizophrenia pathophysiology, MR spectroscopy signals, and diffusion have been revealed. To our knowledge, this is the first study to describe the effect of an anterior cingulate disorder on high-density cholesterol levels on the development of schizophrenia.
Collapse
Affiliation(s)
- Natalia Śmierciak
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Jagiellonian University Medical College, 31-501 Krakow, Poland
| | - Wirginia Krzyściak
- Department of Medical Diagnostics, Jagiellonian University Medical College, 30-688 Krakow, Poland
| | - Marta Szwajca
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Jagiellonian University Medical College, 31-501 Krakow, Poland
| | - Paulina Karcz
- Department of Electroradiology, Jagiellonian University Medical College, 31-126 Krakow, Poland
| | - Amira Bryll
- Department of Radiology, Jagiellonian University Medical College, 31-501 Krakow, Poland
| | - Tadeusz J Popiela
- Department of Radiology, Jagiellonian University Medical College, 31-501 Krakow, Poland
| | - Paulina Donicz
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Jagiellonian University Medical College, 31-501 Krakow, Poland
| | - Aleksander Turek
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 31-530 Krakow, Poland
| | - Veronika Aleksandrovych
- Department of Pathophysiology, Jagiellonian University Medical College, 31-121 Krakow, Poland
| | - Maciej Pilecki
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Jagiellonian University Medical College, 31-501 Krakow, Poland
| |
Collapse
|
26
|
Sharkey RJ, Bacon C, Peterson Z, Rootes-Murdy K, Salvador R, Pomarol-Clotet E, Karuk A, Homan P, Ji E, Omlor W, Homan S, Georgiadis F, Kaiser S, Kirschner M, Ehrlich S, Dannlowski U, Grotegerd D, Goltermann J, Meinert S, Kircher T, Stein F, Brosch K, Krug A, Nenadić I, Sim K, Spalletta G, Piras F, Banaj N, Sponheim SR, Demro C, Ramsay IS, King M, Quidé Y, Green MJ, Nguyen D, Preda A, Calhoun VD, Turner JA, van Erp TGM, Nickl-Jockschat T. Neural Correlates of Positive and Negative Formal Thought Disorder in Individuals with Schizophrenia: An ENIGMA Schizophrenia Working Group Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.06.23291034. [PMID: 37333179 PMCID: PMC10274967 DOI: 10.1101/2023.06.06.23291034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Formal thought disorder (FTD) is a key clinical factor in schizophrenia, but the neurobiological underpinnings remain unclear. In particular, relationship between FTD symptom dimensions and patterns of regional brain volume deficiencies in schizophrenia remain to be established in large cohorts. Even less is known about the cellular basis of FTD. Our study addresses these major obstacles based on a large multi-site cohort through the ENIGMA Schizophrenia Working Group (752 individuals with schizophrenia and 1256 controls), to unravel the neuroanatomy of positive, negative and total FTD in schizophrenia and their cellular bases. We used virtual histology tools to relate brain structural changes associated with FTD to cellular distributions in cortical regions. We identified distinct neural networks for positive and negative FTD. Both networks encompassed fronto-occipito-amygdalar brain regions, but negative FTD showed a relative sparing of orbitofrontal cortical thickness, while positive FTD also affected lateral temporal cortices. Virtual histology identified distinct transcriptomic fingerprints associated for both symptom dimensions. Negative FTD was linked to neuronal and astrocyte fingerprints, while positive FTD was also linked to microglial cell types. These findings relate different dimensions of FTD to distinct brain structural changes and their cellular underpinnings, improve our mechanistic understanding of these key psychotic symptoms.
Collapse
|
27
|
Fritschi L, Lenk K. Parameter Inference for an Astrocyte Model using Machine Learning Approaches. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.16.540982. [PMID: 37292854 PMCID: PMC10245674 DOI: 10.1101/2023.05.16.540982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Astrocytes are the largest subset of glial cells and perform structural, metabolic, and regulatory functions. They are directly involved in the communication at neuronal synapses and the maintenance of brain homeostasis. Several disorders, such as Alzheimer's, epilepsy, and schizophrenia, have been associated with astrocyte dysfunction. Computational models on various spatial levels have been proposed to aid in the understanding and research of astrocytes. The difficulty of computational astrocyte models is to fastly and precisely infer parameters. Physics informed neural networks (PINNs) use the underlying physics to infer parameters and, if necessary, dynamics that can not be observed. We have applied PINNs to estimate parameters for a computational model of an astrocytic compartment. The addition of two techniques helped with the gradient pathologies of the PINNS, the dynamic weighting of various loss components and the addition of Transformers. To overcome the issue that the neural network only learned the time dependence but did not know about eventual changes of the input stimulation to the astrocyte model, we followed an adaptation of PINNs from control theory (PINCs). In the end, we were able to infer parameters from artificial, noisy data, with stable results for the computational astrocyte model.
Collapse
Affiliation(s)
| | - Kerstin Lenk
- Institute of Neural Engineering, Graz University of Technology, Graz, Austria
- BioTechMed, 8010 Graz, Austria
| |
Collapse
|
28
|
Brown JS. Comparison of Oncogenes, Tumor Suppressors, and MicroRNAs Between Schizophrenia and Glioma: The Balance of Power. Neurosci Biobehav Rev 2023; 151:105206. [PMID: 37178944 DOI: 10.1016/j.neubiorev.2023.105206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023]
Abstract
The risk of cancer in schizophrenia has been controversial. Confounders of the issue are cigarette smoking in schizophrenia, and antiproliferative effects of antipsychotic medications. The author has previously suggested comparison of a specific cancer like glioma to schizophrenia might help determine a more accurate relationship between cancer and schizophrenia. To accomplish this goal, the author performed three comparisons of data; the first a comparison of conventional tumor suppressors and oncogenes between schizophrenia and cancer including glioma. This comparison determined schizophrenia has both tumor-suppressive and tumor-promoting characteristics. A second, larger comparison between brain-expressed microRNAs in schizophrenia with their expression in glioma was then performed. This identified a core carcinogenic group of miRNAs in schizophrenia offset by a larger group of tumor-suppressive miRNAs. This proposed "balance of power" between oncogenes and tumor suppressors could cause neuroinflammation. This was assessed by a third comparison between schizophrenia, glioma and inflammation in asbestos-related lung cancer and mesothelioma (ALRCM). This revealed that schizophrenia shares more oncogenic similarity to ALRCM than glioma.
Collapse
|
29
|
Inam ME, Fernandes BS, Salagre E, Grande I, Vieta E, Quevedo J, Zhao Z. The kynurenine pathway in major depressive disorder, bipolar disorder, and schizophrenia: a systematic review and meta-analysis of cerebrospinal fluid studies. REVISTA BRASILEIRA DE PSIQUIATRIA (SAO PAULO, BRAZIL : 1999) 2023; 45. [PMID: 37127280 PMCID: PMC10668321 DOI: 10.47626/1516-4446-2022-2973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/18/2023] [Indexed: 05/03/2023]
Abstract
INTRODUCTION The kynurenine pathway has been attracting attention as a relevant pathway in schizophrenia (SZ), bipolar disorder (BD), and major depressive disorder (MDD). We conducted a systematic review and meta-analysis of the kynurenine pathway metabolites from cerebrospinal fluid (CSF) samples in SZ, BD, and MDD. METHODS PubMed and Scopus databases were systematically searched to identify peer-reviewed case-control studies until April 2022 that assessed kynurenine metabolites, namely, tryptophan (TRP), kynurenine (KYN), kynurenic acid (KA), quinolinic acid (QA), and 3- hydroxykynurenine (3-HK) in SZ, BD, or MDD subjects compared with healthy controls (HC). The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed. The random effects model method was selected when comparing the standardized mean differences (SMD) between two groups. RESULTS There were 23 articles that met the inclusion criteria (k=8, k=8, k=11, for SZ, BD, and MDD, respectively). In SZ, KA levels were increased [SMD=2.64, confidence interval (CI) =1.16 to 4.13, p=0.0005, I2=96%, k=6, n=384 subjects]. TRP (k=5) and KYN (k=4) did not differ significantly. In BD, TRP levels (k=7) did not differ significantly. The level of KA was increased in MDD (k=2), but the small number of studies made not possible for statistical significance evaluation. Finally, in MDD, although some studies tended to have an increased level of KYN in those with remission versus decreased levels in those with current depression, no significant difference was found in any of the kynurenine metabolite levels. Similarly, there was an increased level of QA (k=2) but the number of studies (k= 2) was small. CONCLUSION KA, which has possibly neuroprotective effects, is increased in SZ. QA, which has neurotoxic effects, may be increased in MDD. There were no alterations in BD. There may be alterations in this pathway based on population characteristics and mood states. Future studies should explore the utility of these metabolites as biomarkers.
Collapse
Affiliation(s)
- Mehmet Enes Inam
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Brisa S. Fernandes
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Estela Salagre
- Department of Affective Disorders, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Iria Grande
- Bipolar and Depressive Disorders Unit, Hospital Clinic, Institute of Neurosciences, University of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Centro de Investigación Biomédica en Red de Salud Mental, Barcelona, Catalonia, Spain
| | - Eduard Vieta
- Bipolar and Depressive Disorders Unit, Hospital Clinic, Institute of Neurosciences, University of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Centro de Investigación Biomédica en Red de Salud Mental, Barcelona, Catalonia, Spain
| | - João Quevedo
- Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, UTHealth, Houston, TX, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, UTHealth, Houston, TX, USA
- Human Genetics Center, School of Public Health, UTHealth, Houston, TX, USA
| |
Collapse
|
30
|
Veilleux C, Eugenin EA. Mechanisms of Zika astrocyte infection and neuronal toxicity. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2023; 2:5-18. [PMID: 37027343 PMCID: PMC10070016 DOI: 10.1515/nipt-2022-0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/02/2022] [Indexed: 04/08/2023]
Abstract
Objectives Zika virus (ZIKV) has become an epidemic in several countries and was declared a major public health issue by the WHO. Although ZIKV infection is asymptomatic or shows mild fever-related symptoms in most people, the virus can be transmitted from a pregnant mother to the fetus, resulting in severe brain developmental abnormalities, including microcephaly. Multiple groups have identified developmental neuronal and neuronal progenitor compromise during ZIKV infection within the fetal brain, but little is known about whether ZIKV could infect human astrocytes and its effect on the developing brain. Thus, our objective was to determine astrocyte ZiKV infection in a developmental-dependent manner. Methods We analyze infection of pure cultures of astrocytes and mixed cultures of neurons and astrocytes in response to ZIKV using plaque assays, confocal, and electron microscopy to identify infectivity, ZIKV accumulation and intracellular distribution as well as apoptosis and interorganelle dysfunction. Results Here, we demonstrated that ZIKV enters, infects, replicates, and accumulates in large quantities in human fetal astrocytes in a developmental-dependent manner. Astrocyte infection and intracellular viral accumulation resulted in neuronal apoptosis, and we propose astrocytes are a ZIKV reservoir during brain development. Conclusions Our data identify astrocytes in different stages of development as major contributors to the devastating effects of ZIKV in the developing brain.
Collapse
Affiliation(s)
- Courtney Veilleux
- Public Health Research Institute (PHRI), New York, USA
- Deparment of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers the State University of New Jersey, Newark, NJ, USA
| | - Eliseo A. Eugenin
- Public Health Research Institute (PHRI), New York, USA
- Deparment of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers the State University of New Jersey, Newark, NJ, USA
- Department of Neurobiology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| |
Collapse
|
31
|
Abdelfattah AM, Abuelezz SA, Hendawy N, Negm EA, Nawishy SAEK, Khalil AMM. Sonic hedgehog pathway as a new target of atypical antipsychotics: Revisiting of amisulpride and aripiprazole effects in a rat model of schizophrenia. Life Sci 2023; 316:121366. [PMID: 36649751 DOI: 10.1016/j.lfs.2022.121366] [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: 10/12/2022] [Revised: 12/22/2022] [Accepted: 12/31/2022] [Indexed: 01/16/2023]
Abstract
OBJECTIVES Schizophrenia is a chronic mental illness presented by cognitive deficits that precede its positive and negative symptoms. Sonic hedgehog (Shh)-pathway contributes to its pathophysiology. Shh has a role in neurogenesis as it regulates proliferation and survival of neural cells. In this study, effects of the anti-psychotics Amisulpride and/or Aripiprazole on the Shh-pathway and its relation to cognitive functions and neurogenesis in a rat model of schizophrenia were tested. METHODS 60 male Wistar rats were allocated into the following groups: control, socially isolated, amisulpride and/or aripiprazole-treated groups. Rats were then subjected to behavioral, biochemical, and histopathological tests to assess the impact of these drugs on Shh-pathway. KEY FINDINGS Cognitive-dysfunction was evidenced in socially isolated group in novel object, three-chamber, and Morris water maze tests, associated by disorganised Shh-pathway proteins levels concentrations, increased glial fibrillary acidic protein (GFAP)-stained astrocytes. Treated groups favorably reversed these changes evidenced by increased Shh, transmembrane patched-1 and smoothened, glioma-associated-oncogene (GLI)-1 levels, dopamine-1 receptors and brain derived neurotrophic factor, and decreased GLI-3 protein, GFAP immune reaction in astrocytes and inflammatory markers compared to socially isolated group. CONCLUSION Amisulpride and/or aripiprazole have a favorable role in turning on Shh-pathway with subsequent beneficial cognitive and neurogenesis effects.
Collapse
Affiliation(s)
- Ahmed M Abdelfattah
- Clinical Pharmacology Department, Faculty of Medicine, Port Said University, Cairo, Egypt.
| | - Sally A Abuelezz
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Nevien Hendawy
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Eman A Negm
- Histology and Cell Biology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | |
Collapse
|
32
|
Lohr C. Role of P2Y receptors in astrocyte physiology and pathophysiology. Neuropharmacology 2023; 223:109311. [PMID: 36328064 DOI: 10.1016/j.neuropharm.2022.109311] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2022]
Abstract
Astrocytes are active constituents of the brain that manage ion homeostasis and metabolic support of neurons and directly tune synaptic transmission and plasticity. Astrocytes express all known P2Y receptors. These regulate a multitude of physiological functions such as cell proliferation, Ca2+ signalling, gliotransmitter release and neurovascular coupling. In addition, P2Y receptors are fundamental in the transition of astrocytes into reactive astrocytes, as occurring in many brain disorders such as neurodegenerative diseases, neuroinflammation and epilepsy. This review summarizes the current literature addressing the function of P2Y receptors in astrocytes in the healthy brain as well as in brain diseases.
Collapse
Affiliation(s)
- Christian Lohr
- Institute of Cell and Systems Biology of Animals, University of Hamburg, Germany.
| |
Collapse
|
33
|
Fišar Z. Biological hypotheses, risk factors, and biomarkers of schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2023; 120:110626. [PMID: 36055561 DOI: 10.1016/j.pnpbp.2022.110626] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 12/19/2022]
Abstract
Both the discovery of biomarkers of schizophrenia and the verification of biological hypotheses of schizophrenia are an essential part of the process of understanding the etiology of this mental disorder. Schizophrenia has long been considered a neurodevelopmental disease whose symptoms are caused by impaired synaptic signal transduction and brain neuroplasticity. Both the onset and chronic course of schizophrenia are associated with risk factors-induced disruption of brain function and the establishment of a new homeostatic setpoint characterized by biomarkers. Different risk factors and biomarkers can converge to the same symptoms of schizophrenia, suggesting that the primary cause of the disease can be highly individual. Schizophrenia-related biomarkers include measurable biochemical changes induced by stress (elevated allostatic load), mitochondrial dysfunction, neuroinflammation, oxidative and nitrosative stress, and circadian rhythm disturbances. Here is a summary of selected valid biological hypotheses of schizophrenia formulated based on risk factors and biomarkers, neurodevelopment, neuroplasticity, brain chemistry, and antipsychotic medication. The integrative neurodevelopmental-vulnerability-neurochemical model is based on current knowledge of the neurobiology of the onset and progression of the disease and the effects of antipsychotics and psychotomimetics and reflects the complex and multifactorial nature of schizophrenia.
Collapse
Affiliation(s)
- Zdeněk Fišar
- Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Psychiatry, Czech Republic.
| |
Collapse
|
34
|
Casquero-Veiga M, Lamanna-Rama N, Romero-Miguel D, Rojas-Marquez H, Alcaide J, Beltran M, Nacher J, Desco M, Soto-Montenegro ML. The Poly I:C maternal immune stimulation model shows unique patterns of brain metabolism, morphometry, and plasticity in female rats. Front Behav Neurosci 2023; 16:1022622. [PMID: 36733452 PMCID: PMC9888250 DOI: 10.3389/fnbeh.2022.1022622] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction: Prenatal infections are associated with an increased risk of the onset of schizophrenia. Rodent models of maternal immune stimulation (MIS) have been extensively used in preclinical studies. However, many of these studies only include males, omitting pathophysiological features unique to females. The aim of this study is to characterize the MIS model in female rats using positron emission tomography (PET), structural magnetic resonance imaging (MR), and neuroplasticiy studies. Methods: In gestational day 15, Poly I:C (or Saline) was injected into pregnant Wistar rats to induce the MIS model. Imaging studies: [18F]-fluoro-2-deoxy-D-glucose-PET scans of female-offspring were acquired at post-natal day (PND) 35 and PND100. Furthermore, T2-MR brain images were acquired in adulthood. Differences in FDG uptake and morphometry between groups were assessed with SPM12 and Regions of Interest (ROI) analyses. Ex vivo study: The density of parvalbumin expressing interneurons (PV), perineuronal nets (PNN), and parvalbumin expressing interneurons surrounded by perineuronal nets (PV-PNN) were evaluated in the prelimbic cortex and basolateral amygdala using confocal microscopy. ROIs and neuroplasticity data were analyzed by 2-sample T-test and 2-way-ANOVA analyses, respectively. Results: A significant increase in brain metabolism was found in all animals at adulthood compared to adolescence. MIS hardly modified brain glucose metabolism in females, highlighting a significant hypometabolism in the thalamus at adulthood. In addition, MIS induced gray matter (GM) enlargements in the pituitary, hippocampus, substantia nigra, and cingulate cortex, and GM shrinkages in some thalamic nuclei, cerebelar areas, and brainstem. Moreover, MIS induced white matter shrinkages in the cerebellum, brainstem and corpus callosum, along with cerebrospinal fluid enlargements in the lateral and 4th ventricles. Finally, MIS reduced the density of PV, PNN, and PV-PNN in the basolateral amygdala. Conclusion: Our work showed in vivo the differential pattern of functional and morphometric affectation in the MIS model in females, as well as the deficits caused at the synaptic level according to sex. The differences obtained highlight the relevance of including both sexes in psychiatric research in order to consider their pathophysiological particularities and successfully extend the benefits obtained to the entire patient population.
Collapse
Affiliation(s)
- Marta Casquero-Veiga
- Laboratorio de Imagen Médica, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,Cardiovascular Imaging and Population Studies, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Nicolás Lamanna-Rama
- Laboratorio de Imagen Médica, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,Departamento de Bioingeniería e Ingeniería Aeroespacial, Escuela Técnica Superior de Ingeniería, Universidad Carlos III de Madrid, Madrid, Spain
| | - Diego Romero-Miguel
- Laboratorio de Imagen Médica, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,Departamento de Bioingeniería e Ingeniería Aeroespacial, Escuela Técnica Superior de Ingeniería, Universidad Carlos III de Madrid, Madrid, Spain
| | - Henar Rojas-Marquez
- Laboratorio de Imagen Médica, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain,Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Julia Alcaide
- Neurobiology Unit, Cell Biology Departament, BIOTECMED Institute, Universitat de València, Burjassot, Spain,CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| | - Marc Beltran
- Neurobiology Unit, Cell Biology Departament, BIOTECMED Institute, Universitat de València, Burjassot, Spain
| | - Juan Nacher
- Neurobiology Unit, Cell Biology Departament, BIOTECMED Institute, Universitat de València, Burjassot, Spain,CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| | - Manuel Desco
- Laboratorio de Imagen Médica, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Advanced Imaging Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain,Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Campus de Getafe, Madrid, Spain,*Correspondence: Manuel Desco Maria Luisa Soto-Montenegro
| | - Maria Luisa Soto-Montenegro
- Laboratorio de Imagen Médica, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,CIBER de Salud Mental (CIBERSAM), Madrid, Spain,High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut), University Rey Juan Carlos (URJC), Alcorcón, Spain,*Correspondence: Manuel Desco Maria Luisa Soto-Montenegro
| |
Collapse
|
35
|
Fessel J. Formulating treatment of major psychiatric disorders: algorithm targets the dominantly affected brain cell-types. DISCOVER MENTAL HEALTH 2023; 3:3. [PMID: 37861813 PMCID: PMC10501034 DOI: 10.1007/s44192-022-00029-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 12/21/2022] [Indexed: 10/21/2023]
Abstract
BACKGROUND Pharmacotherapy for most psychiatric conditions was developed from serendipitous observations of benefit from drugs prescribed for different reasons. An algorithmic approach to formulating pharmacotherapy is proposed, based upon which combination of changed activities by brain cell-types is dominant for any particular condition, because those cell-types contain and surrogate for genetic, metabolic and environmental information, that has affected their function. The algorithm performs because functions of some or all the affected cell-types benefit from several available drugs: clemastine, dantrolene, erythropoietin, fingolimod, fluoxetine, lithium, memantine, minocycline, pioglitazone, piracetam, and riluzole PROCEDURES/FINDINGS: Bipolar disorder, major depressive disorder, schizophrenia, Alzheimer's disease, and post-traumatic stress disorder, illustrate the algorithm; for them, literature reviews show that no single combination of altered cell-types accounts for all cases; but they identify, for each condition, which combination occurs most frequently, i.e., dominates, as compared with other possible combinations. Knowing the dominant combination of altered cell-types in a particular condition, permits formulation of therapy with combinations of drugs taken from the above list. The percentage of patients who might benefit from that therapy, depends upon the frequency with which the dominant combination occurs in patients with that particular condition. CONCLUSIONS Knowing the dominant combination of changed cell types in psychiatric conditions, permits an algorithmically formulated, rationally-based treatment. Different studies of the same condition often produce discrepant results; all might be correct, because identical clinical phenotypes result from different combinations of impaired cell-types, thus producing different results. Clinical trials would validate both the proposed concept and choice of drugs.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Department of Medicine, University of California, 2069 Filbert Street, San Francisco, CA, 94123, USA.
| |
Collapse
|
36
|
Fujihara K. Beyond the γ-aminobutyric acid hypothesis of schizophrenia. Front Cell Neurosci 2023; 17:1161608. [PMID: 37168420 PMCID: PMC10165250 DOI: 10.3389/fncel.2023.1161608] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 04/04/2023] [Indexed: 05/13/2023] Open
Abstract
Abnormalities in the γ-aminobutyric acid (GABA) system have been reported in the postmortem brains of individuals with schizophrenia. In particular, the reduction of one of the GABA-synthesizing enzymes, the 67-kDa isoform of glutamate decarboxylase (GAD67), has garnered interest among researchers because of its role in the formation of γ-oscillations and its potential involvement in the cognitive dysfunction observed in schizophrenia. Although several animal models have been generated to simulate the alterations observed in postmortem brain studies, they exhibit inconsistent behavioral phenotypes, leading to conflicting views regarding their contributions to the pathogenesis and manifestation of schizophrenia symptoms. For instance, GAD67 knockout rats (also known as Gad1 knockout rats) exhibit marked impairments in spatial working memory, but other model animals do not. In this review, we summarize the phenotypic attributes of these animal models and contemplate the potential for secondary modifications that may arise from the disruption of the GABAergic nervous system.
Collapse
Affiliation(s)
- Kazuyuki Fujihara
- Department of Psychiatry and Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Japan
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Japan
- *Correspondence: Kazuyuki Fujihara,
| |
Collapse
|
37
|
Benchmarking brain organoid recapitulation of fetal corticogenesis. Transl Psychiatry 2022; 12:520. [PMID: 36539399 PMCID: PMC9767930 DOI: 10.1038/s41398-022-02279-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Brain organoids are becoming increasingly relevant to dissect the molecular mechanisms underlying psychiatric and neurological conditions. The in vitro recapitulation of key features of human brain development affords the unique opportunity of investigating the developmental antecedents of neuropsychiatric conditions in the context of the actual patients' genetic backgrounds. Specifically, multiple strategies of brain organoid (BO) differentiation have enabled the investigation of human cerebral corticogenesis in vitro with increasing accuracy. However, the field lacks a systematic investigation of how closely the gene co-expression patterns seen in cultured BO from different protocols match those observed in fetal cortex, a paramount information for ensuring the sensitivity and accuracy of modeling disease trajectories. Here we benchmark BO against fetal corticogenesis by integrating transcriptomes from in-house differentiated cortical BO (CBO), other BO systems, human fetal brain samples processed in-house, and prenatal cortices from the BrainSpan Atlas. We identified co-expression patterns and prioritized hubs of human corticogenesis and CBO differentiation, highlighting both well-preserved and discordant trends across BO protocols. We evaluated the relevance of identified gene modules for neurodevelopmental disorders and psychiatric conditions finding significant enrichment of disease risk genes especially in modules related to neuronal maturation and synapsis development. The longitudinal transcriptomic analysis of CBO revealed a two-step differentiation composed of a fast-evolving phase, corresponding to the appearance of the main cell populations of the cortex, followed by a slow-evolving one characterized by milder transcriptional changes. Finally, we observed heterochronicity of differentiation across BO models compared to fetal cortex. Our approach provides a framework to directly compare the extent of in vivo/in vitro alignment of neurodevelopmentally relevant processes and their attending temporalities, structured as a resource to query for modeling human corticogenesis and the neuropsychiatric outcomes of its alterations.
Collapse
|
38
|
An Integrative Analysis of Identified Schizophrenia-Associated Brain Cell Types and Gene Expression Changes. Int J Mol Sci 2022; 23:ijms231911581. [PMID: 36232882 PMCID: PMC9569514 DOI: 10.3390/ijms231911581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/21/2022] [Accepted: 09/27/2022] [Indexed: 11/23/2022] Open
Abstract
Schizophrenia (SCZ) is a severe mental disorder that may result in hallucinations, delusions, and extremely disordered thinking. How each cell type in the brain contributes to SCZ occurrence is still unclear. Here, we leveraged the human dorsolateral prefrontal cortex bulk RNA-seq data, then used the RNA-seq deconvolution algorithm CIBERSORTx to generate SCZ brain single-cell RNA-seq data for a comprehensive analysis to understand SCZ-associated brain cell types and gene expression changes. Firstly, we observed that the proportions of brain cell types in SCZ differed from normal samples. Among these cell types, astrocyte, pericyte, and PAX6 cells were found to have a higher proportion in SCZ patients (astrocyte: SCZ = 0.163, control = 0.145, P.adj = 4.9 × 10-4, effect size = 0.478; pericyte: SCZ = 0.057, control = 0.066, P.adj = 1.1 × 10-4, effect size = 0.519; PAX6: SCZ = 0.014, control = 0.011, P.adj = 0.014, effect size = 0.377), while the L5/6_IT_CAR3 cells and LAMP5 cells are the exact opposite (L5/6_IT_Car3: SCZ = 0.102, control = 0.108, P.adj = 0.016, effect size = 0.369; LAMP5: SCZ = 0.057, control = 0.066, P.adj = 2.2 × 10-6, effect size = 0.617). Next, we investigated gene expression in cell types and functional pathways in SCZ. We observed chemical synaptic transmission dysregulation in two types of GABAergic neurons (PVALB and LAMP5), and immune reaction involvement in GABAergic neurons (SST) and non-neuronal cell types (endothelial and oligodendrocyte). Furthermore, we observed that some differential expression genes from bulk RNA-seq displayed cell-type-specific abnormalities in the expression of molecules in SCZ. Finally, the cell types with the SCZ-related transcriptomic changes could be considered to belong to the same module since we observed two major similar coordinated transcriptomic changes across these cell types. Together, our results offer novel insights into cellular heterogeneity and the molecular mechanisms underlying SCZ.
Collapse
|
39
|
Xu J, Li Y, Tian B, Liu H, Wu S, Wang W. The effects and mechanism of environmental enrichment on MK-801 induced cognitive impairment in rodents with schizophrenia. Front Cell Neurosci 2022; 16:1024649. [PMID: 36246525 PMCID: PMC9556631 DOI: 10.3389/fncel.2022.1024649] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/13/2022] [Indexed: 12/02/2022] Open
Abstract
Schizophrenia is a severe mental disorder characterized by positive, negative, and cognitive symptoms. Cognitive symptoms are a kind of symptoms with high incidence and great impact on patients. There is no effective treatment in clinical practice. N-methyl-d-aspartic acid (NMDA) receptor hypofunction may be an important cause of cognitive symptoms. MK-801 (also named Dizocilpine), a noncompetitive antagonist of NMDA receptor, is often used to construct a model of NMDA receptor dysfunction. In terms of treatment, environmental enrichment (EE) as an environmental intervention can effectively improve the symptoms of cognitive impairment in rodents. In this paper, we first briefly introduce the background of cognitive symptoms and EE in schizophrenia, and then investigate the manifestations of MK-801 induced cognitive impairment, the improvement of EE on these cognitive impairments based on the MK-801 induced schizophrenia rodent models, and the possible mechanism of EE in improving cognitive symptoms. This article reviews the literature in recent years, which provides an important reference for MK-801 to construct a cognitive symptom model of schizophrenia and the mechanism of EE in improving cognitive symptoms of schizophrenia.
Collapse
|
40
|
Blagburn-Blanco SV, Chappell MS, De Biase LM, DeNardo LA. Synapse-specific roles for microglia in development: New horizons in the prefrontal cortex. Front Mol Neurosci 2022; 15:965756. [PMID: 36003220 PMCID: PMC9394540 DOI: 10.3389/fnmol.2022.965756] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/13/2022] [Indexed: 11/19/2022] Open
Abstract
Dysfunction of both microglia and circuitry in the medial prefrontal cortex (mPFC) have been implicated in numerous neuropsychiatric disorders, but how microglia affect mPFC development in health and disease is not well understood. mPFC circuits undergo a prolonged maturation after birth that is driven by molecular programs and activity-dependent processes. Though this extended development is crucial to acquire mature cognitive abilities, it likely renders mPFC circuitry more susceptible to disruption by genetic and environmental insults that increase the risk of developing mental health disorders. Recent work suggests that microglia directly influence mPFC circuit maturation, though the biological factors underlying this observation remain unclear. In this review, we discuss these recent findings along with new studies on the cellular mechanisms by which microglia shape sensory circuits during postnatal development. We focus on the molecular pathways through which glial cells and immune signals regulate synaptogenesis and activity-dependent synaptic refinement. We further highlight how disruptions in these pathways are implicated in the pathogenesis of neurodevelopmental and psychiatric disorders associated with mPFC dysfunction, including schizophrenia and autism spectrum disorder (ASD). Using these disorders as a framework, we discuss microglial mechanisms that could link environmental risk factors including infections and stress with ongoing genetic programs to aberrantly shape mPFC circuitry.
Collapse
Affiliation(s)
- Sara V. Blagburn-Blanco
- Neuroscience Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, United States
- Medical Scientist Training Program, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Physiology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Megan S. Chappell
- Neuroscience Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Physiology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lindsay M. De Biase
- Department of Physiology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Laura A. DeNardo
- Department of Physiology, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
41
|
Keeping Excitation-Inhibition Ratio in Balance. Int J Mol Sci 2022; 23:ijms23105746. [PMID: 35628556 PMCID: PMC9145842 DOI: 10.3390/ijms23105746] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022] Open
Abstract
Unrelated genetic mutations can lead to convergent manifestations of neurological disorders with similar behavioral phenotypes. Experimental data frequently show a lack of dramatic changes in neuroanatomy, indicating that the key cause of symptoms might arise from impairment in the communication between neurons. A transient imbalance between excitatory (glutamatergic) and inhibitory (GABAergic) synaptic transmission (the E/I balance) during early development is generally considered to underlie the development of several neurological disorders in adults. However, the E/I ratio is a multidimensional variable. Synaptic contacts are highly dynamic and the actual strength of synaptic projections is determined from the balance between synaptogenesis and synaptic elimination. During development, relatively slow postsynaptic receptors are replaced by fast ones that allow for fast stimulus-locked excitation/inhibition. Using the binomial model of synaptic transmission allows for the reassessing of experimental data from different mouse models, showing that a transient E/I shift is frequently counterbalanced by additional pre- and/or postsynaptic changes. Such changes—for instance, the slowing down of postsynaptic currents by means of immature postsynaptic receptors—stabilize the average synaptic strength, but impair the timing of information flow. Compensatory processes and/or astrocytic signaling may represent possible targets for medical treatments of different disorders directed to rescue the proper information processing.
Collapse
|
42
|
Structural and Functional Deviations of the Hippocampus in Schizophrenia and Schizophrenia Animal Models. Int J Mol Sci 2022; 23:ijms23105482. [PMID: 35628292 PMCID: PMC9143100 DOI: 10.3390/ijms23105482] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 01/04/2023] Open
Abstract
Schizophrenia is a grave neuropsychiatric disease which frequently onsets between the end of adolescence and the beginning of adulthood. It is characterized by a variety of neuropsychiatric abnormalities which are categorized into positive, negative and cognitive symptoms. Most therapeutical strategies address the positive symptoms by antagonizing D2-dopamine-receptors (DR). However, negative and cognitive symptoms persist and highly impair the life quality of patients due to their disabling effects. Interestingly, hippocampal deviations are a hallmark of schizophrenia and can be observed in early as well as advanced phases of the disease progression. These alterations are commonly accompanied by a rise in neuronal activity. Therefore, hippocampal formation plays an important role in the manifestation of schizophrenia. Furthermore, studies with animal models revealed a link between environmental risk factors and morphological as well as electrophysiological abnormalities in the hippocampus. Here, we review recent findings on structural and functional hippocampal abnormalities in schizophrenic patients and in schizophrenia animal models, and we give an overview on current experimental approaches that especially target the hippocampus. A better understanding of hippocampal aberrations in schizophrenia might clarify their impact on the manifestation and on the outcome of this severe disease.
Collapse
|
43
|
Śmierciak N, Szwajca M, Popiela TJ, Bryll A, Karcz P, Donicz P, Turek A, Krzyściak W, Pilecki M. Redefining the Cut-Off Ranges for TSH Based on the Clinical Picture, Results of Neuroimaging and Laboratory Tests in Unsupervised Cluster Analysis as Individualized Diagnosis of Early Schizophrenia. J Pers Med 2022; 12:jpm12020247. [PMID: 35207735 PMCID: PMC8874519 DOI: 10.3390/jpm12020247] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 11/16/2022] Open
Abstract
Thyroid abnormalities, including mild forms of hypothyroidism and hyperthyroidism, are reported as risk factors for the development of a number of neuropsychiatric disorders, including schizophrenia. The diagnostic process still takes into account the extreme ranges of the accepted reference values for serum TSH since the concentration of free thyroxine in the serum does not change by definition. TSH mU/L cut-off values in psychiatric patients are currently clinically considered in the case of extremely high serum TSH levels (>4.0 mU/L). The results obtained in this study suggest that the clinically significant value has a lower TSH cut-off point with an upper limit of 2–2.5 mU/L. The criteria for the differential diagnosis of patients with schizophrenia, however, mainly take into account statutory reference ranges without a background related to the history of thyroid diseases in the family. The results indicate the need to lower the upper cut-off values for TSH among patients with early psychosis, which is related to the potential clinical significance of the obtained values both in the field of clinical evaluation and neuroimaging and laboratory evaluation parameters. The cut-off points obtained with the prior available knowledge coincided with the values established in the unsupervised clustering method, which further confirms the legitimacy of their use in the individualized diagnosis strategy of schizophrenia.
Collapse
Affiliation(s)
- Natalia Śmierciak
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 21a, 31-501 Krakow, Poland; (N.Ś.); (M.S.); (P.D.); (A.T.)
| | - Marta Szwajca
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 21a, 31-501 Krakow, Poland; (N.Ś.); (M.S.); (P.D.); (A.T.)
| | - Tadeusz J. Popiela
- Department of Radiology, Jagiellonian University Medical College, Kopernika 19, 31-501 Krakow, Poland;
- Correspondence: (T.J.P.); (W.K.); (M.P.)
| | - Amira Bryll
- Department of Radiology, Jagiellonian University Medical College, Kopernika 19, 31-501 Krakow, Poland;
| | - Paulina Karcz
- Department of Electroradiology, Jagiellonian University Medical College, Michałowskiego 12, 31-126 Krakow, Poland;
| | - Paulina Donicz
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 21a, 31-501 Krakow, Poland; (N.Ś.); (M.S.); (P.D.); (A.T.)
| | - Aleksander Turek
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 21a, 31-501 Krakow, Poland; (N.Ś.); (M.S.); (P.D.); (A.T.)
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, Łazarza 16, 31-530 Krakow, Poland
| | - Wirginia Krzyściak
- Department of Medical Diagnostics, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
- Correspondence: (T.J.P.); (W.K.); (M.P.)
| | - Maciej Pilecki
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 21a, 31-501 Krakow, Poland; (N.Ś.); (M.S.); (P.D.); (A.T.)
- Correspondence: (T.J.P.); (W.K.); (M.P.)
| |
Collapse
|
44
|
Gzielo K, Nikiforuk A. Astroglia in Autism Spectrum Disorder. Int J Mol Sci 2021; 22:11544. [PMID: 34768975 PMCID: PMC8583956 DOI: 10.3390/ijms222111544] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/13/2021] [Accepted: 10/21/2021] [Indexed: 01/12/2023] Open
Abstract
Autism spectrum disorder (ASD) is an umbrella term encompassing several neurodevelopmental disorders such as Asperger syndrome or autism. It is characterised by the occurrence of distinct deficits in social behaviour and communication and repetitive patterns of behaviour. The symptoms may be of different intensity and may vary in types. Risk factors for ASD include disturbed brain homeostasis, genetic predispositions, or inflammation during the prenatal period caused by viruses or bacteria. The number of diagnosed cases is growing, but the main cause and mechanism leading to ASD is still uncertain. Recent findings from animal models and human cases highlight the contribution of glia to the ASD pathophysiology. It is known that glia cells are not only "gluing" neurons together but are key players participating in different processes crucial for proper brain functioning, including neurogenesis, synaptogenesis, inflammation, myelination, proper glutamate processing and many others. Despite the prerequisites for the involvement of glia in the processes related to the onset of autism, there are far too little data regarding the engagement of these cells in the development of ASD.
Collapse
Affiliation(s)
- Kinga Gzielo
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Behavioral Neuroscience and Drug Development, 12 Smętna Street, 31-343 Kraków, Poland;
| | | |
Collapse
|