1
|
Bergstrom JJD, Fu MM. Dysregulation of myelination-related genes in schizophrenia. J Neurochem 2024; 168:2227-2242. [PMID: 39086020 PMCID: PMC11449665 DOI: 10.1111/jnc.16152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 08/02/2024]
Abstract
Schizophrenic individuals display disrupted myelination patterns, altered oligodendrocyte distribution, and abnormal oligodendrocyte morphology. Schizophrenia is linked with dysregulation of a variety of genes involved in oligodendrocyte function and myelin production. Single-nucleotide polymorphisms (SNPs) and rare mutations in myelination-related genes are observed in certain schizophrenic populations, representing potential genetic risk factors. Downregulation of myelination-related RNAs and proteins, particularly in frontal and limbic regions, is consistently associated with the disorder across multiple studies. These findings support the notion that disruptions in myelination may contribute to the cognitive and behavioral impairments experienced in schizophrenia, although further evidence of causation is needed.
Collapse
Affiliation(s)
| | - Meng-Meng Fu
- Department of Molecular and Cell Biology, UC Berkeley, Berkeley, California, USA
| |
Collapse
|
2
|
Li X, Fernandes BS, Liu A, Chen J, Chen X, Zhao Z, Dai Y. GRPa-PRS: A risk stratification method to identify genetically-regulated pathways in polygenic diseases. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.06.19.23291621. [PMID: 37425929 PMCID: PMC10327215 DOI: 10.1101/2023.06.19.23291621] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Background Polygenic risk scores (PRS) are tools used to evaluate an individual's susceptibility to polygenic diseases based on their genetic profile. A considerable proportion of people carry a high genetic risk but evade the disease. On the other hand, some individuals with a low risk of eventually developing the disease. We hypothesized that unknown counterfactors might be involved in reversing the PRS prediction, which might provide new insights into the pathogenesis, prevention, and early intervention of diseases. Methods We built a novel computational framework to identify genetically-regulated pathways (GRPas) using PRS-based stratification for each cohort. We curated two AD cohorts with genotyping data; the discovery (disc) and the replication (rep) datasets include 2722 and 2854 individuals, respectively. First, we calculated the optimized PRS model based on the three recent AD GWAS summary statistics for each cohort. Then, we stratified the individuals by their PRS and clinical diagnosis into six biologically meaningful PRS strata, such as AD cases with low/high risk and cognitively normal (CN) with low/high risk. Lastly, we imputed individual genetically-regulated expression (GReX) and identified differential GReX and GRPas between risk strata using gene-set enrichment and variational analyses in two models, with and without APOE effects. An orthogonality test was further conducted to verify those GRPas are independent of PRS risk. To verify the generalizability of other polygenic diseases, we further applied a default model of GRPa-PRS for schizophrenia (SCZ). Results For each stratum, we conducted the same procedures in both the disc and rep datasets for comparison. In AD, we identified several well-known AD-related pathways, including amyloid-beta clearance, tau protein binding, and astrocyte response to oxidative stress. Additionally, we discovered resilience-related GRPs that are orthogonal to AD PRS, such as the calcium signaling pathway and divalent inorganic cation homeostasis. In SCZ, pathways related to mitochondrial function and muscle development were highlighted. Finally, our GRPa-PRS method identified more consistent differential pathways compared to another variant-based pathway PRS method. Conclusions We developed a framework, GRPa-PRS, to systematically explore the differential GReX and GRPas among individuals stratified by their estimated PRS. The GReX-level comparison among those strata unveiled new insights into the pathways associated with disease risk and resilience. Our framework is extendable to other polygenic complex diseases.
Collapse
Affiliation(s)
- Xiaoyang Li
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Brisa S. Fernandes
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Andi Liu
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jingchun Chen
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA
| | - Xiangning Chen
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhongming Zhao
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yulin Dai
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
3
|
Jørgensen KN, Nerland S, Slapø NB, Norbom LB, Mørch-Johnsen L, Wortinger LA, Barth C, Andreou D, Maximov II, Geier OM, Andreassen OA, Jönsson EG, Agartz I. Assessing regional intracortical myelination in schizophrenia spectrum and bipolar disorders using the optimized T1w/T2w-ratio. Psychol Med 2024; 54:2369-2379. [PMID: 38563302 DOI: 10.1017/s0033291724000503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
BACKGROUND Dysmyelination could be part of the pathophysiology of schizophrenia spectrum (SCZ) and bipolar disorders (BPD), yet few studies have examined myelination of the cerebral cortex. The ratio of T1- and T2-weighted magnetic resonance images (MRI) correlates with intracortical myelin. We investigated the T1w/T2w-ratio and its age trajectories in patients and healthy controls (CTR) and explored associations with antipsychotic medication use and psychotic symptoms. METHODS Patients with SCZ (n = 64; mean age = 30.4 years, s.d. = 9.8), BPD (n = 91; mean age 31.0 years, s.d. = 10.2), and CTR (n = 155; mean age = 31.9 years, s.d. = 9.1) who participated in the TOP study (NORMENT, University of Oslo, Norway) were clinically assessed and scanned using a General Electric 3 T MRI system. T1w/T2w-ratio images were computed using an optimized pipeline with intensity normalization and field inhomogeneity correction. Vertex-wise regression models were used to compare groups and examine group × age interactions. In regions showing significant differences, we explored associations with antipsychotic medication use and psychotic symptoms. RESULTS No main effect of diagnosis was found. However, age slopes of the T1w/T2w-ratio differed significantly between SCZ and CTR, predominantly in frontal and temporal lobe regions: Lower T1w/T2w-ratio values with higher age were found in CTR, but not in SCZ. Follow-up analyses revealed a more positive age slope in patients who were using antipsychotics and patients using higher chlorpromazine-equivalent doses. CONCLUSIONS While we found no evidence of reduced intracortical myelin in SCZ or BPD relative to CTR, different regional age trajectories in SCZ may suggest a promyelinating effect of antipsychotic medication.
Collapse
Affiliation(s)
- Kjetil Nordbø Jørgensen
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry, Telemark Hospital, Skien, Norway
| | - Stener Nerland
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Nora Berz Slapø
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Linn B Norbom
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Department of Psychology, PROMENTA Research Center, University of Oslo, Oslo, Norway
| | - Lynn Mørch-Johnsen
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry & Department of Clinical Research, Østfold Hospital, Grålum, Norway
| | - Laura Anne Wortinger
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Claudia Barth
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Dimitrios Andreou
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden
| | - Ivan I Maximov
- Department of Psychology, University of Oslo, Oslo, Norway
- The Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Health and Functioning, Western Norway University of Applied Sciences, Bergen, Norway
| | - Oliver M Geier
- Department of Physics and Computational Radiology, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Ole A Andreassen
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- The Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Erik G Jönsson
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden
| | - Ingrid Agartz
- The Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden
| |
Collapse
|
4
|
Jenkins AK, Ketchesin KD, Becker-Krail DD, McClung CA. Molecular Rhythmicity in Glia: Importance for Brain Health and Relevance to Psychiatric Disease. Biol Psychiatry 2024; 96:S0006-3223(24)01298-8. [PMID: 38735357 PMCID: PMC11550267 DOI: 10.1016/j.biopsych.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/05/2024] [Accepted: 05/03/2024] [Indexed: 05/14/2024]
Abstract
Circadian rhythms are approximate 24-hour rhythms present in nearly all aspects of human physiology, including proper brain function. These rhythms are produced at the cellular level through a transcriptional-translational feedback loop known as the molecular clock. Diurnal variation in gene expression has been demonstrated in brain tissue from multiple species, including humans, in both cortical and subcortical regions. Interestingly, these rhythms in gene expression have been shown to be disrupted across psychiatric disorders and may be implicated in their underlying pathophysiology. However, little is known regarding molecular rhythms in specific cell types in the brain and how they might be involved in psychiatric disease. Although glial cells (e.g., astrocytes, microglia, and oligodendrocytes) have been historically understudied compared to neurons, evidence of the molecular clock is found within each of these cell subtypes. Here, we review the current literature, which suggests that molecular rhythmicity is essential to functional physiologic outputs from each glial subtype. Furthermore, disrupted molecular rhythms within these cells and the resultant functional deficits may be relevant to specific phenotypes across psychiatric illnesses. Given that circadian rhythm disruptions have been so integrally tied to psychiatric disease, the molecular mechanisms governing these associations could represent exciting new avenues for future research and potential novel pharmacologic targets for treatment.
Collapse
Affiliation(s)
- Aaron K Jenkins
- Translational Neuroscience Program, Department of Psychiatry, and Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kyle D Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, and Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Darius D Becker-Krail
- Translational Neuroscience Program, Department of Psychiatry, and Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Colleen A McClung
- Translational Neuroscience Program, Department of Psychiatry, and Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
5
|
Kobayashi H, Sasabayashi D, Takahashi T, Furuichi A, Kido M, Takayanagi Y, Noguchi K, Suzuki M. The relationship between gray/white matter contrast and cognitive performance in first-episode schizophrenia. Cereb Cortex 2024; 34:bhae009. [PMID: 38265871 DOI: 10.1093/cercor/bhae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 01/26/2024] Open
Abstract
Previous postmortem brain studies have revealed disturbed myelination in the intracortical regions in patients with schizophrenia, possibly reflecting anomalous brain maturational processes. However, it currently remains unclear whether this anomalous myelination is already present in early illness stages and/or progresses during the course of the illness. In this magnetic resonance imaging study, we examined gray/white matter contrast (GWC) as a potential marker of intracortical myelination in 63 first-episode schizophrenia (FESz) patients and 77 healthy controls (HC). Furthermore, we investigated the relationships between GWC findings and clinical/cognitive variables in FESz patients. GWC in the bilateral temporal, parietal, occipital, and insular regions was significantly higher in FESz patients than in HC, which was partly associated with the durations of illness and medication, the onset age, and lower executive and verbal learning performances. Because higher GWC implicates lower myelin in the deeper layers of the cortex, these results suggest that schizophrenia patients have less intracortical myelin at the time of their first psychotic episode, which underlies lower cognitive performance in early illness stages.
Collapse
Affiliation(s)
- Haruko Kobayashi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, 2630 Sugitani, Toyama 930-0194, Japan
- Research Center for idling Brain Science, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Daiki Sasabayashi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, 2630 Sugitani, Toyama 930-0194, Japan
- Research Center for idling Brain Science, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Tsutomu Takahashi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, 2630 Sugitani, Toyama 930-0194, Japan
- Research Center for idling Brain Science, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Atsushi Furuichi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, 2630 Sugitani, Toyama 930-0194, Japan
- Research Center for idling Brain Science, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Mikio Kido
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, 2630 Sugitani, Toyama 930-0194, Japan
- Kido Clinic, 244 Honoki, Imizu City, Toyama, 934-0053, Japan
| | - Yoichiro Takayanagi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, 2630 Sugitani, Toyama 930-0194, Japan
- Arisawabashi Hospital, 5-5 Hane-Shin, Fuchu-Machi, Toyama, 939-2704, Japan
| | - Kyo Noguchi
- Department of Radiology, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Michio Suzuki
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, 2630 Sugitani, Toyama 930-0194, Japan
- Research Center for idling Brain Science, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| |
Collapse
|
6
|
Bhuvaneshwar K, Gusev Y. Translational bioinformatics and data science for biomarker discovery in mental health: an analytical review. Brief Bioinform 2024; 25:bbae098. [PMID: 38493340 PMCID: PMC10944574 DOI: 10.1093/bib/bbae098] [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: 06/21/2023] [Revised: 01/23/2024] [Accepted: 02/18/2024] [Indexed: 03/18/2024] Open
Abstract
Translational bioinformatics and data science play a crucial role in biomarker discovery as it enables translational research and helps to bridge the gap between the bench research and the bedside clinical applications. Thanks to newer and faster molecular profiling technologies and reducing costs, there are many opportunities for researchers to explore the molecular and physiological mechanisms of diseases. Biomarker discovery enables researchers to better characterize patients, enables early detection and intervention/prevention and predicts treatment responses. Due to increasing prevalence and rising treatment costs, mental health (MH) disorders have become an important venue for biomarker discovery with the goal of improved patient diagnostics, treatment and care. Exploration of underlying biological mechanisms is the key to the understanding of pathogenesis and pathophysiology of MH disorders. In an effort to better understand the underlying mechanisms of MH disorders, we reviewed the major accomplishments in the MH space from a bioinformatics and data science perspective, summarized existing knowledge derived from molecular and cellular data and described challenges and areas of opportunities in this space.
Collapse
Affiliation(s)
- Krithika Bhuvaneshwar
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University, Washington DC, 20007, USA
| | - Yuriy Gusev
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University, Washington DC, 20007, USA
| |
Collapse
|
7
|
Farsi Z, Nicolella A, Simmons SK, Aryal S, Shepard N, Brenner K, Lin S, Herzog L, Moran SP, Stalnaker KJ, Shin W, Gazestani V, Song BJ, Bonanno K, Keshishian H, Carr SA, Pan JQ, Macosko EZ, Datta SR, Dejanovic B, Kim E, Levin JZ, Sheng M. Brain-region-specific changes in neurons and glia and dysregulation of dopamine signaling in Grin2a mutant mice. Neuron 2023; 111:3378-3396.e9. [PMID: 37657442 DOI: 10.1016/j.neuron.2023.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 05/19/2023] [Accepted: 08/04/2023] [Indexed: 09/03/2023]
Abstract
A genetically valid animal model could transform our understanding of schizophrenia (SCZ) disease mechanisms. Rare heterozygous loss-of-function (LoF) mutations in GRIN2A, encoding a subunit of the NMDA receptor, greatly increase the risk of SCZ. By transcriptomic, proteomic, and behavioral analyses, we report that heterozygous Grin2a mutant mice show (1) large-scale gene expression changes across multiple brain regions and in neuronal (excitatory and inhibitory) and non-neuronal cells (astrocytes and oligodendrocytes), (2) evidence of hypoactivity in the prefrontal cortex (PFC) and hyperactivity in the hippocampus and striatum, (3) an elevated dopamine signaling in the striatum and hypersensitivity to amphetamine-induced hyperlocomotion (AIH), (4) altered cholesterol biosynthesis in astrocytes, (5) a reduction in glutamatergic receptor signaling proteins in the synapse, and (6) an aberrant locomotor pattern opposite of that induced by antipsychotic drugs. These findings reveal potential pathophysiologic mechanisms, provide support for both the "hypo-glutamate" and "hyper-dopamine" hypotheses of SCZ, and underscore the utility of Grin2a-deficient mice as a genetic model of SCZ.
Collapse
Affiliation(s)
- Zohreh Farsi
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Ally Nicolella
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sean K Simmons
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sameer Aryal
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nate Shepard
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kira Brenner
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sherry Lin
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Linnea Herzog
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sean P Moran
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Katherine J Stalnaker
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Wangyong Shin
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea
| | - Vahid Gazestani
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bryan J Song
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kevin Bonanno
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hasmik Keshishian
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Steven A Carr
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jen Q Pan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Evan Z Macosko
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Massachusetts General Hospital, Department of Psychiatry, Boston, MA, USA
| | | | - Borislav Dejanovic
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea; Department of Biological Sciences, Korea Advanced Institute for Science and Technology, Daejeon, South Korea
| | - Joshua Z Levin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Morgan Sheng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
8
|
Pietiläinen O, Trehan A, Meyer D, Mitchell J, Tegtmeyer M, Valakh V, Gebre H, Chen T, Vartiainen E, Farhi SL, Eggan K, McCarroll SA, Nehme R. Astrocytic cell adhesion genes linked to schizophrenia correlate with synaptic programs in neurons. Cell Rep 2023; 42:111988. [PMID: 36640364 PMCID: PMC10721115 DOI: 10.1016/j.celrep.2022.111988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 11/16/2022] [Accepted: 12/23/2022] [Indexed: 01/15/2023] Open
Abstract
The maturation of neurons and the development of synapses, although emblematic of neurons, also relies on interactions with astrocytes and other glia. Here, to study the role of glia-neuron interactions, we analyze the transcriptomes of human pluripotent stem cell (hPSC)-derived neurons, from 80 human donors, that were cultured with or without contact with glial cells. We find that the presence of astrocytes enhances synaptic gene-expression programs in neurons when in physical contact with astrocytes. These changes in neurons correlate with increased expression, in the cocultured glia, of genes that encode synaptic cell adhesion molecules. Both the neuronal and astrocyte gene-expression programs are enriched for genes associated with schizophrenia risk. Our results suggest that astrocyte-expressed genes with synaptic functions are associated with stronger expression of synaptic genetic programs in neurons, and they suggest a potential role for astrocyte-neuron interactions in schizophrenia.
Collapse
Affiliation(s)
- Olli Pietiläinen
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA 02138, USA; Neuroscience Center, Helsinki Institute for Life Science, University of Helsinki, 00290 Helsinki, Finland.
| | - Aditi Trehan
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA 02138, USA
| | - Daniel Meyer
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Jana Mitchell
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA 02138, USA
| | - Matthew Tegtmeyer
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA 02138, USA; Centre for Gene Therapy and Regenerative Medicine, King's College, London WC2R 2LS, UK
| | - Vera Valakh
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Hilena Gebre
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Theresa Chen
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Emilia Vartiainen
- Neuroscience Center, Helsinki Institute for Life Science, University of Helsinki, 00290 Helsinki, Finland
| | - Samouil L Farhi
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kevin Eggan
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA 02138, USA
| | - Steven A McCarroll
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Ralda Nehme
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
9
|
Carrier M, Dolhan K, Bobotis BC, Desjardins M, Tremblay MÈ. The implication of a diversity of non-neuronal cells in disorders affecting brain networks. Front Cell Neurosci 2022; 16:1015556. [PMID: 36439206 PMCID: PMC9693782 DOI: 10.3389/fncel.2022.1015556] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/07/2022] [Indexed: 11/13/2022] Open
Abstract
In the central nervous system (CNS) neurons are classically considered the functional unit of the brain. Analysis of the physical connections and co-activation of neurons, referred to as structural and functional connectivity, respectively, is a metric used to understand their interplay at a higher level. A myriad of glial cell types throughout the brain composed of microglia, astrocytes and oligodendrocytes are key players in the maintenance and regulation of neuronal network dynamics. Microglia are the central immune cells of the CNS, able to affect neuronal populations in number and connectivity, allowing for maturation and plasticity of the CNS. Microglia and astrocytes are part of the neurovascular unit, and together they are essential to protect and supply nutrients to the CNS. Oligodendrocytes are known for their canonical role in axonal myelination, but also contribute, with microglia and astrocytes, to CNS energy metabolism. Glial cells can achieve this variety of roles because of their heterogeneous populations comprised of different states. The neuroglial relationship can be compromised in various manners in case of pathologies affecting development and plasticity of the CNS, but also consciousness and mood. This review covers structural and functional connectivity alterations in schizophrenia, major depressive disorder, and disorder of consciousness, as well as their correlation with vascular connectivity. These networks are further explored at the cellular scale by integrating the role of glial cell diversity across the CNS to explain how these networks are affected in pathology.
Collapse
Affiliation(s)
- Micaël Carrier
- Neurosciences Axis, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Kira Dolhan
- Department of Psychology, University of Victoria, Victoria, BC, Canada
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | | | - Michèle Desjardins
- Department of Physics, Physical Engineering and Optics, Université Laval, Québec City, QC, Canada
- Oncology Axis, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Neurosciences Axis, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Marie-Ève Tremblay,
| |
Collapse
|
10
|
Puvogel S, Alsema A, Kracht L, Webster MJ, Weickert CS, Sommer IEC, Eggen BJL. Single-nucleus RNA sequencing of midbrain blood-brain barrier cells in schizophrenia reveals subtle transcriptional changes with overall preservation of cellular proportions and phenotypes. Mol Psychiatry 2022; 27:4731-4740. [PMID: 36192459 PMCID: PMC9734060 DOI: 10.1038/s41380-022-01796-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 12/14/2022]
Abstract
The midbrain is an extensively studied brain region in schizophrenia, in view of its reported dopamine pathophysiology and neuroimmune changes associated with this disease. Besides the dopaminergic system, the midbrain contains other cell types that may be involved in schizophrenia pathophysiology. The neurovascular hypothesis of schizophrenia postulates that both the neurovasculature structure and the functioning of the blood-brain barrier (BBB) are compromised in schizophrenia. In the present study, potential alteration in the BBB of patients with schizophrenia was investigated by single-nucleus RNA sequencing of post-mortem midbrain tissue (15 schizophrenia cases and 14 matched controls). We did not identify changes in the relative abundance of the major BBB cell types, nor in the sub-populations, associated with schizophrenia. However, we identified 14 differentially expressed genes in the cells of the BBB in schizophrenia as compared to controls, including genes that have previously been related to schizophrenia, such as FOXP2 and PDE4D. These transcriptional changes were limited to the ependymal cells and pericytes, suggesting that the cells of the BBB are not broadly affected in schizophrenia.
Collapse
Affiliation(s)
- Sofía Puvogel
- Department of Biomedical Sciences of Cells and Systems, section Cognitive Neuroscience, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
- Department of Biomedical Sciences of Cells and Systems, section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Astrid Alsema
- Department of Biomedical Sciences of Cells and Systems, section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Laura Kracht
- Department of Biomedical Sciences of Cells and Systems, section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maree J Webster
- Laboratory of Brain Research, Stanley Medical Research Institute, Rockville, MD, USA
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, NSW, Australia
- School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, USA
| | - Iris E C Sommer
- Department of Biomedical Sciences of Cells and Systems, section Cognitive Neuroscience, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Bart J L Eggen
- Department of Biomedical Sciences of Cells and Systems, section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
11
|
Brisch R, Wojtylak S, Saniotis A, Steiner J, Gos T, Kumaratilake J, Henneberg M, Wolf R. The role of microglia in neuropsychiatric disorders and suicide. Eur Arch Psychiatry Clin Neurosci 2022; 272:929-945. [PMID: 34595576 PMCID: PMC9388452 DOI: 10.1007/s00406-021-01334-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 09/09/2021] [Indexed: 02/08/2023]
Abstract
This narrative review examines the possible role of microglial cells, first, in neuroinflammation and, second, in schizophrenia, depression, and suicide. Recent research on the interactions between microglia, astrocytes and neurons and their involvement in pathophysiological processes of neuropsychiatric disorders is presented. This review focuses on results from postmortem, positron emission tomography (PET) imaging studies, and animal models of schizophrenia and depression. Third, the effects of antipsychotic and antidepressant drug therapy, and of electroconvulsive therapy on microglial cells are explored and the upcoming development of therapeutic drugs targeting microglia is described. Finally, there is a discussion on the role of microglia in the evolutionary progression of human lineage. This view may contribute to a new understanding of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Ralf Brisch
- Department of Forensic Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Szymon Wojtylak
- Department of Pathomorphology, Medical University of Gdańsk, Gdańsk, Poland
| | - Arthur Saniotis
- Department of Anthropology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
- Department of Pharmacy, Knowledge University, Erbil, Kurdistan Region, Iraq
| | - Johann Steiner
- Department of Psychiatry and Psychotherapy, Otto-von-Guericke-University, Magdeburg, Germany
| | - Tomasz Gos
- Department of Forensic Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Jaliya Kumaratilake
- Biological Anthropology and Comparative Anatomy Research Unit, Medical School, The University of Adelaide, Adelaide, Australia
| | - Maciej Henneberg
- Biological Anthropology and Comparative Anatomy Research Unit, Medical School, The University of Adelaide, Adelaide, Australia
- Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland
| | - Rainer Wolf
- Department of Nursing and Health, Hochschule Fulda, University of Applied Sciences, Fulda, Germany.
| |
Collapse
|
12
|
Avery SN, Huang AS, Sheffield JM, Rogers BP, Vandekar S, Anticevic A, Woodward ND. Development of Thalamocortical Structural Connectivity in Typically Developing and Psychosis Spectrum Youths. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:782-792. [PMID: 34655804 PMCID: PMC9008075 DOI: 10.1016/j.bpsc.2021.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Thalamocortical white matter connectivity is disrupted in psychosis and is hypothesized to play a role in its etiology and associated cognitive impairment. Attenuated cognitive symptoms often begin in adolescence, during a critical phase of white matter and cognitive development. However, little is known about the development of thalamocortical white matter connectivity and its association with cognition. METHODS This study characterized effects of age, sex, psychosis symptomatology, and cognition in thalamocortical networks in a large sample of youths (N = 1144, ages 8-22 years, 46% male) from the Philadelphia Neurodevelopmental Cohort, which included 316 typically developing youths, 330 youths on the psychosis spectrum, and 498 youths with other psychopathology. Probabilistic tractography was used to quantify percent total connectivity between the thalamus and six cortical regions and assess microstructural properties (i.e., fractional anisotropy) of thalamocortical white matter tracts. RESULTS Overall, percent total connectivity of the thalamus was weakly associated with age and was not associated with psychopathology or cognition. In contrast, fractional anisotropy of all thalamocortical tracts increased significantly with age, was generally higher in males than females, and was lowest in youths on the psychosis spectrum. Fractional anisotropy of tracts linking the thalamus to prefrontal and posterior parietal cortices was related to better cognitive function across subjects. CONCLUSIONS By characterizing the pattern of typical development and alterations in those at risk for psychotic disorders, this study provides a foundation for further conceptualization of thalamocortical white matter microstructure as a marker of neurodevelopment supporting cognition and an important risk marker for psychosis.
Collapse
Affiliation(s)
- Suzanne N Avery
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee.
| | - Anna S Huang
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Julia M Sheffield
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Baxter P Rogers
- Vanderbilt University Institute of Imaging Sciences, Nashville, Tennessee
| | - Simon Vandekar
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Alan Anticevic
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Neil D Woodward
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
13
|
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: 17] [Impact Index Per Article: 8.5] [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
|
14
|
Prats C, Fatjó-Vilas M, Penzol MJ, Kebir O, Pina-Camacho L, Demontis D, Crespo-Facorro B, Peralta V, González-Pinto A, Pomarol-Clotet E, Papiol S, Parellada M, Krebs MO, Fañanás L. Association and epistatic analysis of white matter related genes across the continuum schizophrenia and autism spectrum disorders: The joint effect of NRG1-ErbB genes. World J Biol Psychiatry 2022; 23:208-218. [PMID: 34338147 DOI: 10.1080/15622975.2021.1939155] [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: 10/20/2022]
Abstract
BACKGROUND Schizophrenia-spectrum disorders (SSD) and Autism spectrum disorders (ASD) are neurodevelopmental disorders that share clinical, cognitive, and genetic characteristics, as well as particular white matter (WM) abnormalities. In this study, we aimed to investigate the role of a set of oligodendrocyte/myelin-related (OMR) genes and their epistatic effect on the risk for SSD and ASD. METHODS We examined 108 SNPs in a set of 22 OMR genes in 1749 subjects divided into three independent samples (187 SSD trios, 915 SSD cases/control, and 91 ASD trios). Genetic association and gene-gene interaction analyses were conducted with PLINK and MB-MDR, and permutation procedures were implemented in both. RESULTS Some OMR genes showed an association trend with SSD, while after correction, the ones that remained significantly associated were MBP, ERBB3, and AKT1. Significant gene-gene interactions were found between (i) NRG1*MBP (perm p-value = 0.002) in the SSD trios sample, (ii) ERBB3*AKT1 (perm p-value = 0.001) in the SSD case-control sample, and (iii) ERBB3*QKI (perm p-value = 0.0006) in the ASD trios sample. DISCUSSION Our results suggest the implication of OMR genes in the risk for both SSD and ASD and highlight the role of NRG1 and ERBB genes. These findings are in line with the previous evidence and may suggest pathophysiological mechanisms related to NRG1/ERBBs signalling in these disorders.
Collapse
Affiliation(s)
- C Prats
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain.,Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Institut d'Investigació Biomèdica de Bellvitge, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
| | - M Fatjó-Vilas
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain.,Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain
| | - M J Penzol
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense, IiSGM, Madrid, Spain
| | - O Kebir
- INSERM, U1266, Laboratory "Pathophysiology of psychiatric disorders", Institute of psychiatry and neurosciences of Paris, Paris, France.,GHU Psychiatrie et Neurosciences de Paris, Paris, France
| | - L Pina-Camacho
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense, IiSGM, Madrid, Spain
| | - D Demontis
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research iPSYCH, Aarhus, Denmark
| | - B Crespo-Facorro
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,University Hospital Virgen del Rocio, IbiS Department of Psychiatry, School of Medicine, University of Sevilla, Sevilla, Spain
| | - V Peralta
- Gerencia de Salud Mental, Servicio Navarro de Salud-Osasunbidea, Pamplona, Navarra, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNa), Pamplona, Navarra, Spain
| | - A González-Pinto
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Psychiatry Service, University Hospital of Alava-Santiago, EMBREC, EHU/UPV University of the Basque Country, Kronikgune, Vitoria, Spain
| | - E Pomarol-Clotet
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain
| | - S Papiol
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany.,Department of Psychiatry, University Hospital, Ludwig Maximilian University, Munich, Germany
| | - M Parellada
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense, IiSGM, Madrid, Spain
| | - M O Krebs
- INSERM, U1266, Laboratory "Pathophysiology of psychiatric disorders", Institute of psychiatry and neurosciences of Paris, Paris, France.,University Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Paris Descartes, Service Hospitalo-Universitaire, Centre Hospitalier Sainte-Anne, Paris, France
| | - L Fañanás
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain.,Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| |
Collapse
|
15
|
Valdés-Tovar M, Rodríguez-Ramírez AM, Rodríguez-Cárdenas L, Sotelo-Ramírez CE, Camarena B, Sanabrais-Jiménez MA, Solís-Chagoyán H, Argueta J, López-Riquelme GO. Insights into myelin dysfunction in schizophrenia and bipolar disorder. World J Psychiatry 2022; 12:264-285. [PMID: 35317338 PMCID: PMC8900585 DOI: 10.5498/wjp.v12.i2.264] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/10/2021] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
Schizophrenia and bipolar disorder are disabling psychiatric disorders with a worldwide prevalence of approximately 1%. Both disorders present chronic and deteriorating prognoses that impose a large burden, not only on patients but also on society and health systems. These mental illnesses share several clinical and neurobiological traits; of these traits, oligodendroglial dysfunction and alterations to white matter (WM) tracts could underlie the disconnection between brain regions related to their symptomatic domains. WM is mainly composed of heavily myelinated axons and glial cells. Myelin internodes are discrete axon-wrapping membrane sheaths formed by oligodendrocyte processes. Myelin ensheathment allows fast and efficient conduction of nerve impulses through the nodes of Ranvier, improving the overall function of neuronal circuits. Rapid and precisely synchronized nerve impulse conduction through fibers that connect distant brain structures is crucial for higher-level functions, such as cognition, memory, mood, and language. Several cellular and subcellular anomalies related to myelin and oligodendrocytes have been found in postmortem samples from patients with schizophrenia or bipolar disorder, and neuroimaging techniques have revealed consistent alterations at the macroscale connectomic level in both disorders. In this work, evidence regarding these multilevel alterations in oligodendrocytes and myelinated tracts is discussed, and the involvement of proteins in key functions of the oligodendroglial lineage, such as oligodendrogenesis and myelination, is highlighted. The molecular components of the axo-myelin unit could be important targets for novel therapeutic approaches to schizophrenia and bipolar disorder.
Collapse
Affiliation(s)
- Marcela Valdés-Tovar
- Departamento de Farmacogenética, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | | | - Leslye Rodríguez-Cárdenas
- Departamento de Farmacogenética, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Carlo E Sotelo-Ramírez
- Departamento de Farmacogenética, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
- Doctorado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico
| | - Beatriz Camarena
- Departamento de Farmacogenética, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | | | - Héctor Solís-Chagoyán
- Laboratorio de Neurofarmacología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Jesús Argueta
- Doctorado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico
- Laboratorio de Neurofarmacología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Germán Octavio López-Riquelme
- Laboratorio de Socioneurobiología, Centro de Investigación en Ciencias Cognitivas, Universidad del Estado de Morelos, Cuernavaca 62209, Morelos, Mexico
| |
Collapse
|
16
|
Francisco RD, Fernando V, Norma E, Madai ME, Marcelo B. Glial changes in schizophrenia: Genetic and epigenetic approach. Indian J Psychiatry 2022; 64:3-12. [PMID: 35400734 PMCID: PMC8992743 DOI: 10.4103/indianjpsychiatry.indianjpsychiatry_104_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 10/24/2021] [Accepted: 12/23/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Schizophrenia (SCZ) is a severe mental illness that affects one percent of the population, affecting how people think, feel, and behave. Evidence suggests glial cell alteration and some researchers have found genetic risk loci and epigenetic marks that may regulate glia-related genes implicated in SCZ. AIM The aim of this study is to identify genetic and epigenetic changes that have been reported in glial cells or glial-associated genes in SCZ. MATERIALS AND METHODS We searched the articles from PubMed, PubMed Central, Medline, Medscape, and Embase databases up to December 2020 to identify relevant peer-reviewed articles in English. The titles and abstracts were screened to eliminate irrelevant citations. RESULTS Twenty-four original articles were included in the review. Studies were categorized into the following four thematic via: (1) oligodendrocytes, (2) microglia, (3) astrocytes, and (4) perspectives. CONCLUSION This study is the first of its kind to review research on genetic variants and epigenetic modifications associated with glia-related genes implicated in SCZ. Epigenetic evidence is considerably less than genetic evidence in this field. Understanding the pathways of some risk genes and their genetic and epigenetic regulation allows us to understand and find potential targets for future interventions in this mental illness.
Collapse
Affiliation(s)
- Ramos Daniel Francisco
- Faculty of Chemical Sciences, Juarez University of the State of Durango, Durango, Mexico
| | - Vazquez Fernando
- Faculty of Chemical Sciences, Juarez University of the State of Durango, Durango, Mexico.,Research Unit, General Hospital 450, Durango, Mexico
| | - Estrada Norma
- Faculty of Chemical Sciences, Juarez University of the State of Durango, Durango, Mexico
| | - Méndez Edna Madai
- Scientific Research Institute, Juarez University of the State of Durango, Durango, Mexico
| | - Barraza Marcelo
- Faculty of Chemical Sciences, Juarez University of the State of Durango, Durango, Mexico
| |
Collapse
|
17
|
Long KLP, Chao LL, Kazama Y, An A, Hu KY, Peretz L, Muller DCY, Roan VD, Misra R, Toth CE, Breton JM, Casazza W, Mostafavi S, Huber BR, Woodward SH, Neylan TC, Kaufer D. Regional gray matter oligodendrocyte- and myelin-related measures are associated with differential susceptibility to stress-induced behavior in rats and humans. Transl Psychiatry 2021; 11:631. [PMID: 34903726 PMCID: PMC8668977 DOI: 10.1038/s41398-021-01745-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/30/2021] [Accepted: 11/23/2021] [Indexed: 12/23/2022] Open
Abstract
Individual reactions to traumatic stress vary dramatically, yet the biological basis of this variation remains poorly understood. Recent studies demonstrate the surprising plasticity of oligodendrocytes and myelin with stress and experience, providing a potential mechanism by which trauma induces aberrant structural and functional changes in the adult brain. In this study, we utilized a translational approach to test the hypothesis that gray matter oligodendrocytes contribute to traumatic-stress-induced behavioral variation in both rats and humans. We exposed adult, male rats to a single, severe stressor and used a multimodal approach to characterize avoidance, startle, and fear-learning behavior, as well as oligodendrocyte and myelin basic protein (MBP) content in multiple brain areas. We found that oligodendrocyte cell density and MBP were correlated with behavioral outcomes in a region-specific manner. Specifically, stress-induced avoidance positively correlated with hippocampal dentate gyrus oligodendrocytes and MBP. Viral overexpression of the oligodendrogenic factor Olig1 in the dentate gyrus was sufficient to induce an anxiety-like behavioral phenotype. In contrast, contextual fear learning positively correlated with MBP in the amygdala and spatial-processing regions of the hippocampus. In a group of trauma-exposed US veterans, T1-/T2-weighted magnetic resonance imaging estimates of hippocampal and amygdala myelin associated with symptom profiles in a region-specific manner that mirrored the findings in rats. These results demonstrate a species-independent relationship between region-specific, gray matter oligodendrocytes and differential behavioral phenotypes following traumatic stress exposure. This study suggests a novel mechanism for brain plasticity that underlies individual variance in sensitivity to traumatic stress.
Collapse
Affiliation(s)
- Kimberly L P Long
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, 94720, USA
- Department of Psychiatry and Behavioral Sciences, University of California, SanFrancisco, San Francisco, CA, 94143, USA
| | - Linda L Chao
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94143, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Yurika Kazama
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Anjile An
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, 94720, USA
- Division of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Kelsey Y Hu
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Lior Peretz
- Department of Molecular, Cellular, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Dyana C Y Muller
- Department of Computer Science, University of Arizona, Tucson, AZ, 85721, USA
| | - Vivian D Roan
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Rhea Misra
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Claire E Toth
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Jocelyn M Breton
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, 94720, USA
- Department of Psychiatry, Columbia University, New York, NY, 10027, USA
| | - William Casazza
- Department of Statistics and Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Sara Mostafavi
- Department of Statistics and Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
- Canadian Institute for Advanced Research, Toronto, ON, M5G 1M1, Canada
| | - Bertrand R Huber
- Department of Neurology, Boston University, Boston, MA, 02215, USA
- National Center for PTSD, VA New England Health Care System, Boston, MA, 02130, USA
| | - Steven H Woodward
- National Center for PTSD, VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Thomas C Neylan
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, 94143, USA
- San Francisco VA Health Care System, San Francisco, CA, 94121, USA
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, 94720, USA.
- Canadian Institute for Advanced Research, Toronto, ON, M5G 1M1, Canada.
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, 94720, USA.
| |
Collapse
|
18
|
Sui YV, Bertisch H, Lee HH, Storey P, Babb JS, Goff DC, Samsonov A, Lazar M. Quantitative Macromolecular Proton Fraction Mapping Reveals Altered Cortical Myelin Profile in Schizophrenia Spectrum Disorders. Cereb Cortex Commun 2021; 2:tgab015. [PMID: 34296161 PMCID: PMC8271044 DOI: 10.1093/texcom/tgab015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/15/2021] [Accepted: 02/19/2021] [Indexed: 01/12/2023] Open
Abstract
Myelin abnormalities have been reported in schizophrenia spectrum disorders (SSD) in white matter. However, in vivo examinations of cortical myeloarchitecture in SSD, especially those using quantitative measures, are limited. Here, we employed macromolecular proton fraction (MPF) obtained from quantitative magnetization transfer imaging to characterize intracortical myelin organization in 30 SSD patients versus 34 healthy control (HC) participants. We constructed cortical myelin profiles by extracting MPF values at various cortical depths and quantified their shape using a nonlinearity index (NLI). To delineate the association of illness duration with myelin changes, SSD patients were further divided into 3 duration groups. Between-group comparisons revealed reduced NLI in the SSD group with the longest illness duration (>5.5 years) compared with HC predominantly in bilateral prefrontal areas. Within the SSD group, cortical NLI decreased with disease duration and was positively associated with a measure of spatial working memory capacity as well as with cortical thickness (CT). Layer-specific analyses suggested that NLI decreases in the long-duration SSD group may arise in part from significantly increased MPF values in the midcortical layers. The current study reveals cortical myelin profile changes in SSD with illness progression, which may reflect an abnormal compensatory mechanism of the disorder.
Collapse
Affiliation(s)
- Yu Veronica Sui
- Department of Radiology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Hilary Bertisch
- Department of Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Hong-Hsi Lee
- Department of Radiology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Pippa Storey
- Department of Radiology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - James S Babb
- Department of Radiology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Donald C Goff
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Alexey Samsonov
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Mariana Lazar
- Department of Radiology, NYU Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
19
|
Katrinli S, Smith AK. Immune system regulation and role of the human leukocyte antigen in posttraumatic stress disorder. Neurobiol Stress 2021; 15:100366. [PMID: 34355049 PMCID: PMC8322450 DOI: 10.1016/j.ynstr.2021.100366] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 05/28/2021] [Accepted: 07/10/2021] [Indexed: 11/01/2022] Open
Abstract
Posttraumatic stress disorder (PTSD) is a debilitating condition that adversely affect mental and physical health. Recent studies have increasingly explored the role of the immune system in risk for PTSD and its related symptoms. Dysregulation of the immune system may lead to central nervous system tissue damage and impair learning and memory processes. Individuals with PTSD often have comorbid inflammatory or auto-immune disorders. Evidence shows associations between PTSD and multiple genes that are involved in immune-related or inflammatory pathways. In this review, we will summarize the evidence of immune dysregulation in PTSD, outlining the contributions of distinct cell types, genes, and biological pathways. We use the Human Leukocyte Antigen (HLA) locus to illustrate the contribution of genetic variation to function in different tissues that contribute to PTSD etiology, severity, and comorbidities.
Collapse
Affiliation(s)
- Seyma Katrinli
- Emory University, Department of Gynecology and Obstetrics, Atlanta, GA, USA
| | - Alicia K Smith
- Emory University, Department of Gynecology and Obstetrics, Atlanta, GA, USA.,Emory University School of Medicine, Department of Psychiatry and Behavioral Sciences, Atlanta, GA, USA
| |
Collapse
|
20
|
Liu A, Dai Y, Mendez EF, Hu R, Fries GR, Najera KE, Jiang S, Meyer TD, Stertz L, Jia P, Walss-Bass C, Zhao Z. Genome-Wide Correlation of DNA Methylation and Gene Expression in Postmortem Brain Tissues of Opioid Use Disorder Patients. Int J Neuropsychopharmacol 2021; 24:879-891. [PMID: 34214162 PMCID: PMC8598308 DOI: 10.1093/ijnp/pyab043] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/01/2021] [Accepted: 06/29/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Opioid use disorder (OUD) affects millions of people, causing nearly 50 000 deaths annually in the United States. While opioid exposure and OUD are known to cause widespread transcriptomic and epigenetic changes, few studies in human samples have been conducted. Understanding how OUD affects the brain at the molecular level could help decipher disease pathogenesis and shed light on OUD treatment. METHODS We generated genome-wide transcriptomic and DNA methylation profiles of 22 OUD subjects and 19 non-psychiatric controls. We applied weighted gene co-expression network analysis to identify genetic markers consistently associated with OUD at both transcriptomic and methylomic levels. We then performed functional enrichment for biological interpretation. We employed cross-omics analysis to uncover OUD-specific regulatory networks. RESULTS We found 6 OUD-associated co-expression gene modules and 6 co-methylation modules (false discovery rate <0.1). Genes in these modules are involved in astrocyte and glial cell differentiation, gliogenesis, response to organic substance, and response to cytokine (false discovery rate <0.05). Cross-omics analysis revealed immune-related transcription regulators, suggesting the role of transcription factor-targeted regulatory networks in OUD pathogenesis. CONCLUSIONS Our integrative analysis of multi-omics data in OUD postmortem brain samples suggested complex gene regulatory mechanisms involved in OUD-associated expression patterns. Candidate genes and their upstream regulators revealed in astrocyte, and glial cells could provide new insights into OUD treatment development.
Collapse
Affiliation(s)
- Andi Liu
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yulin Dai
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX,USA
| | - Emily F Mendez
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX,USA
| | - Ruifeng Hu
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX,USA
| | - Gabriel R Fries
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX,USA,Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX,USA
| | - Katherine E Najera
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX,USA
| | - Shan Jiang
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX,USA
| | - Thomas D Meyer
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX,USA
| | - Laura Stertz
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX,USA
| | - Peilin Jia
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX,USA
| | - Consuelo Walss-Bass
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX,USA,Correspondence: Zhongming Zhao, PhD, Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, 7000 Fannin St #600, Houston, TX, USA () and Consuelo Walss-Bass, PhD, Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA ()
| | - Zhongming Zhao
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA,Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX,USA,Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX,USA,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA,Correspondence: Zhongming Zhao, PhD, Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, 7000 Fannin St #600, Houston, TX, USA () and Consuelo Walss-Bass, PhD, Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA ()
| |
Collapse
|
21
|
Abstract
Schizophrenia is a severe and clinically heterogenous mental disorder
affecting approximately 1% of the population worldwide. Despite
tremendous achievements in the field of schizophrenia research, its
precise aetiology remains elusive. Besides dysfunctional neuronal
signalling, the pathophysiology of schizophrenia appears to involve
molecular and functional abnormalities in glial cells, including
astrocytes. This article provides a concise overview of the current
evidence supporting altered astrocyte activity in schizophrenia, which
ranges from findings obtained from post-mortem immunohistochemical
analyses, genetic association studies and transcriptomic
investigations, as well as from experimental investigations of
astrocyte functions in animal models. Integrating the existing data
from these research areas strongly suggests that astrocytes have the
capacity to critically affect key neurodevelopmental and homeostatic
processes pertaining to schizophrenia pathogenesis, including
glutamatergic signalling, synaptogenesis, synaptic pruning and
myelination. The further elucidation of astrocytes functions in health
and disease may, therefore, offer new insights into how these glial
cells contribute to abnormal brain development and functioning
underlying this debilitating mental disorder.
Collapse
Affiliation(s)
- Tina Notter
- Tina Notter, Institute of
Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich,
Switzerland. Emails: ;
| |
Collapse
|
22
|
Tsetsos F, Yu D, Sul JH, Huang AY, Illmann C, Osiecki L, Darrow SM, Hirschtritt ME, Greenberg E, Muller-Vahl KR, Stuhrmann M, Dion Y, Rouleau GA, Aschauer H, Stamenkovic M, Schlögelhofer M, Sandor P, Barr CL, Grados MA, Singer HS, Nöthen MM, Hebebrand J, Hinney A, King RA, Fernandez TV, Barta C, Tarnok Z, Nagy P, Depienne C, Worbe Y, Hartmann A, Budman CL, Rizzo R, Lyon GJ, McMahon WM, Batterson JR, Cath DC, Malaty IA, Okun MS, Berlin C, Woods DW, Lee PC, Jankovic J, Robertson MM, Gilbert DL, Brown LW, Coffey BJ, Dietrich A, Hoekstra PJ, Kuperman S, Zinner SH, Wagner M, Knowles JA, Jeremy Willsey A, Tischfield JA, Heiman GA, Cox NJ, Freimer NB, Neale BM, Davis LK, Coppola G, Mathews CA, Scharf JM, Paschou P, Barr CL, Batterson JR, Berlin C, Budman CL, Cath DC, Coppola G, Cox NJ, Darrow S, Davis LK, Dion Y, Freimer NB, Grados MA, Greenberg E, Hirschtritt ME, Huang AY, Illmann C, King RA, Kurlan R, Leckman JF, Lyon GJ, Malaty IA, Mathews CA, McMahon WM, Neale BM, Okun MS, Osiecki L, Robertson MM, Rouleau GA, Sandor P, Scharf JM, Singer HS, Smit JH, Sul JH, Yu D, Aschauer HAH, Barta C, Budman CL, Cath DC, Depienne C, Hartmann A, Hebebrand J, Konstantinidis A, Mathews CA, Müller-Vahl K, Nagy P, Nöthen MM, Paschou P, Rizzo R, Rouleau GA, Sandor P, Scharf JM, Schlögelhofer M, Stamenkovic M, Stuhrmann M, Tsetsos F, Tarnok Z, Wolanczyk T, Worbe Y, Brown L, Cheon KA, Coffey BJ, Dietrich A, Fernandez TV, Garcia-Delgar B, Gilbert D, Grice DE, Hagstrøm J, Hedderly T, Heiman GA, Heyman I, Hoekstra PJ, Huyser C, Kim YK, Kim YS, King RA, Koh YJ, Kook S, Kuperman S, Leventhal BL, Madruga-Garrido M, Mir P, Morer A, Münchau A, Plessen KJ, Roessner V, Shin EY, Song DH, Song J, Tischfield JA, Willsey AJ, Zinner S, Aschauer H, Barr CL, Barta C, Batterson JR, Berlin C, Brown L, Budman CL, Cath DC, Coffey BJ, Coppola G, Cox NJ, Darrow S, Davis LK, Depienne C, Dietrich A, Dion Y, Fernandez T, Freimer NB, Gilbert D, Grados MA, Greenberg E, Hartmann A, Hebebrand J, Heiman G, Hirschtritt ME, Hoekstra P, Huang AY, Illmann C, Jankovic J, King RA, Kuperman S, Lee PC, Lyon GJ, Malaty IA, Mathews CA, McMahon WM, Müller-Vahl K, Nagy P, Neale BM, Nöthen MM, Okun MS, Osiecki L, Paschou P, Rizzo R, Robertson MM, Rouleau GA, Sandor P, Scharf JM, Schlögelhofer M, Singer HS, Stamenkovic M, Stuhrmann M, Sul JH, Tarnok Z, Tischfield J, Tsetsos F, Willsey AJ, Woods D, Worbe Y, Yu D, Zinner S. Synaptic processes and immune-related pathways implicated in Tourette syndrome. Transl Psychiatry 2021; 11:56. [PMID: 33462189 PMCID: PMC7814139 DOI: 10.1038/s41398-020-01082-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/18/2020] [Accepted: 10/21/2020] [Indexed: 12/23/2022] Open
Abstract
Tourette syndrome (TS) is a neuropsychiatric disorder of complex genetic architecture involving multiple interacting genes. Here, we sought to elucidate the pathways that underlie the neurobiology of the disorder through genome-wide analysis. We analyzed genome-wide genotypic data of 3581 individuals with TS and 7682 ancestry-matched controls and investigated associations of TS with sets of genes that are expressed in particular cell types and operate in specific neuronal and glial functions. We employed a self-contained, set-based association method (SBA) as well as a competitive gene set method (MAGMA) using individual-level genotype data to perform a comprehensive investigation of the biological background of TS. Our SBA analysis identified three significant gene sets after Bonferroni correction, implicating ligand-gated ion channel signaling, lymphocytic, and cell adhesion and transsynaptic signaling processes. MAGMA analysis further supported the involvement of the cell adhesion and trans-synaptic signaling gene set. The lymphocytic gene set was driven by variants in FLT3, raising an intriguing hypothesis for the involvement of a neuroinflammatory element in TS pathogenesis. The indications of involvement of ligand-gated ion channel signaling reinforce the role of GABA in TS, while the association of cell adhesion and trans-synaptic signaling gene set provides additional support for the role of adhesion molecules in neuropsychiatric disorders. This study reinforces previous findings but also provides new insights into the neurobiology of TS.
Collapse
Grants
- R01 NS102371 NINDS NIH HHS
- R01 NS096207 NINDS NIH HHS
- R01 NS096008 NINDS NIH HHS
- R01 MH115958 NIMH NIH HHS
- K08 MH099424 NIMH NIH HHS
- U24 NS095914 NINDS NIH HHS
- K02 NS085048 NINDS NIH HHS
- R01 MH115963 NIMH NIH HHS
- U01 HG009086 NHGRI NIH HHS
- R56 MH120736 NIMH NIH HHS
- U54 MD010722 NIMHD NIH HHS
- UL1 TR001863 NCATS NIH HHS
- R01 DC016977 NIDCD NIH HHS
- R01 NS105746 NINDS NIH HHS
- R01 MH118233 NIMH NIH HHS
- DP2 HD098859 NICHD NIH HHS
- R01 MH115961 NIMH NIH HHS
- U24 MH068457 NIMH NIH HHS
- R25 NS108939 NINDS NIH HHS
- R01 MH114927 NIMH NIH HHS
- R01 NR014852 NINR NIH HHS
- R21 HG010652 NHGRI NIH HHS
- R01 MH113362 NIMH NIH HHS
- RM1 HG009034 NHGRI NIH HHS
- FT is co-financed by Greece and the European Union (European Social Fund- ESF) through the Operational Programme «Human Resources Development, Education and Lifelong Learning» in the context of the project “Reinforcement of Postdoctoral Researchers - 2nd Cycle” (MIS-5033021), implemented by the State Scholarships Foundation (IKY)
- KMV has received financial or material research support from the EU (FP7-HEALTH-2011 No. 278367, FP7-PEOPLE-2012-ITN No. 316978), the German Research Foundation (DFG: GZ MU 1527/3-1), the German Ministry of Education and Research (BMBF: 01KG1421), the National Institute of Mental Health (NIMH), the Tourette Gesellschaft Deutschland e.V., the Else-Kroner-Fresenius-Stiftung, and GW, Almirall, Abide Therapeutics, and Therapix Biosiences and has received consultant’s honoraria from Abide Therapeutics, Tilray, Resalo Vertrieb GmbH, and Wayland Group, speaker’s fees from Tilray and Cogitando GmbH, and royalties from Medizinisch Wissenschaftliche Verlagsgesellschaft Berlin, Elsevier, and Kohlhammer; and is a consultant for Nuvelution TS Pharma Inc., Zynerba Pharmaceuticals, Resalo Vertrieb GmbH, CannaXan GmbH, Therapix Biosiences, Syqe, Nomovo Pharma, and Columbia Care.
- MMN has received fees for memberships in Scientific Advisory Boards from the Lundbeck Foundation and the Robert-Bosch-Stiftung, and for membership in the Medical-Scientific Editorial Office of the Deutsches Ärzteblatt. MMN was reimbursed travel expenses for a conference participation by Shire Deutschland GmbH. MMN receives salary payments from Life & Brain GmbH and holds shares in Life & Brain GmbH. All this concerned activities outside the submitted work.
- IM has participated in research funded by the Parkinson Foundation, Tourette Association, Dystonia Coalition, AbbVie, Biogen, Boston Scientific, Eli Lilly, Impax, Neuroderm, Prilenia, Revance, Teva but has no owner interest in any pharmaceutical company. She has received travel compensation or honoraria from the Tourette Association of America, Parkinson Foundation, International Association of Parkinsonism and Related Disorders, Medscape, and Cleveland Clinic, and royalties for writing a book with Robert rose publishers.
- MSO serves as a consultant for the Parkinson’s Foundation, and has received research grants from NIH, Parkinson’s Foundation, the Michael J. Fox Foundation, the Parkinson Alliance, Smallwood Foundation, the Bachmann-Strauss Foundation, the Tourette Syndrome Association, and the UF Foundation. MSO’s DBS research is supported by: NIH R01 NR014852 and R01NS096008. MSO is PI of the NIH R25NS108939 Training Grant. MSO has received royalties for publications with Demos, Manson, Amazon, Smashwords, Books4Patients, Perseus, Robert Rose, Oxford and Cambridge (movement disorders books). MSO is an associate editor for New England Journal of Medicine Journal Watch Neurology. MSO has participated in CME and educational activities on movement disorders sponsored by the Academy for Healthcare Learning, PeerView, Prime, QuantiaMD, WebMD/Medscape, Medicus, MedNet, Einstein, MedNet, Henry Stewart, American Academy of Neurology, Movement Disorders Society and by Vanderbilt University. The institution and not MSO receives grants from Medtronic, Abbvie, Boston Scientific, Abbott and Allergan and the PI has no financial interest in these grants. MSO has participated as a site PI and/or co-I for several NIH, foundation, and industry sponsored trials over the years but has not received honoraria. Research projects at the University of Florida receive device and drug donations.
- DW receives royalties for books on Tourette Syndrome with Guilford Press, Oxford University Press, and Springer Press.
- BMN is a member of the scientific advisory board at Deep Genomics and consultant for Camp4 Therapeutics, Takeda Pharmaceutical and Biogen.
Collapse
Affiliation(s)
- Fotis Tsetsos
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Dongmei Yu
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jae Hoon Sul
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Alden Y Huang
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Cornelia Illmann
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Lisa Osiecki
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Sabrina M Darrow
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Matthew E Hirschtritt
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Erica Greenberg
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Kirsten R Muller-Vahl
- Clinic of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Manfred Stuhrmann
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Yves Dion
- McGill University Health Center, University of Montreal, McGill University Health Centre, Montreal, Canada
| | - Guy A Rouleau
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
| | - Harald Aschauer
- Department of Psychiatry and Psychotherapy, Medical University Vienna, Vienna, Austria
- Biopsychosocial Corporation, Vienna, Austria
| | - Mara Stamenkovic
- Department of Psychiatry and Psychotherapy, Medical University Vienna, Vienna, Austria
| | | | - Paul Sandor
- University Health Network, Youthdale Treatment Centres, and University of Toronto, Toronto, Canada
| | - Cathy L Barr
- Krembil Research Institute, University Health Network, Hospital for Sick Children, and University of Toronto, Toronto, Canada
| | - Marco A Grados
- Johns Hopkins University School of Medicine and the Kennedy Krieger Institute, Baltimore, MD, USA
| | - Harvey S Singer
- Johns Hopkins University School of Medicine and the Kennedy Krieger Institute, Baltimore, MD, USA
| | - Markus M Nöthen
- Institute of Human Genetics, University Hospital Bonn, University of Bonn Medical School, Bonn, Germany
| | - Johannes Hebebrand
- Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Anke Hinney
- Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Robert A King
- Yale Child Study Center and the Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Thomas V Fernandez
- Yale Child Study Center and the Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Csaba Barta
- Institute of Medical Chemistry, Molecular Biology, and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Zsanett Tarnok
- Vadaskert Child and Adolescent Psychiatric Hospital, Budapest, Hungary
| | - Peter Nagy
- Vadaskert Child and Adolescent Psychiatric Hospital, Budapest, Hungary
| | - Christel Depienne
- Institute of Human Genetics, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1127, CNRS UMR 7225, ICM, Paris, France
| | - Yulia Worbe
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1127, CNRS UMR 7225, ICM, Paris, France
- French Reference Centre for Gilles de la Tourette Syndrome, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
- Assistance Publique-Hôpitaux de Paris, Department of Neurology, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
- Assistance Publique Hôpitaux de Paris, Hopital Saint Antoine, Paris, France
| | - Andreas Hartmann
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1127, CNRS UMR 7225, ICM, Paris, France
- French Reference Centre for Gilles de la Tourette Syndrome, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
- Assistance Publique-Hôpitaux de Paris, Department of Neurology, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Cathy L Budman
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Renata Rizzo
- Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Gholson J Lyon
- Jervis Clinic, NYS Institute for Basic Research in Developmental Disabilities (IBR), Staten Island, NY, USA
| | - William M McMahon
- Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
| | | | - Danielle C Cath
- Department of Psychiatry, University Medical Center Groningen and Rijksuniversity Groningen, and Drenthe Mental Health Center, Groningen, the Netherlands
| | - Irene A Malaty
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, USA
| | - Michael S Okun
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, USA
| | - Cheston Berlin
- Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Douglas W Woods
- Marquette University and University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Paul C Lee
- Tripler Army Medical Center and University of Hawaii John A. Burns School of Medicine, Honolulu, HI, USA
| | - Joseph Jankovic
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Mary M Robertson
- Division of Psychiatry, Department of Neuropsychiatry, University College London, London, UK
| | - Donald L Gilbert
- Division of Pediatric Neurology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, Cincinnati, USA
| | | | - Barbara J Coffey
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrea Dietrich
- Department of Child and Adolescent Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Pieter J Hoekstra
- Department of Child and Adolescent Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Samuel Kuperman
- University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Samuel H Zinner
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Michael Wagner
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | | | - A Jeremy Willsey
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Jay A Tischfield
- Department of Genetics and the Human Genetics Institute of New Jersey, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Gary A Heiman
- Department of Genetics and the Human Genetics Institute of New Jersey, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Nancy J Cox
- Division of Genetic Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nelson B Freimer
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Benjamin M Neale
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Lea K Davis
- Division of Genetic Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Giovanni Coppola
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Carol A Mathews
- Department of Psychiatry, Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Jeremiah M Scharf
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Neurology, Brigham and Women's Hospital, and the Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Peristera Paschou
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.
| | - Cathy L Barr
- Krembil Research Institute, University Health Network, Hospital for Sick Children, and University of Toronto, Toronto, Canada
| | | | - Cheston Berlin
- Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Cathy L Budman
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Danielle C Cath
- Department of Psychiatry, University Medical Center Groningen and Rijksuniversity Groningen, and Drenthe Mental Health Center, Groningen, the Netherlands
| | - Giovanni Coppola
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Nancy J Cox
- Division of Genetic Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sabrina Darrow
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Lea K Davis
- Division of Genetic Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yves Dion
- McGill University Health Center, University of Montreal, McGill University Health Centre, Montreal, Canada
| | - Nelson B Freimer
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Marco A Grados
- Johns Hopkins University School of Medicine and the Kennedy Krieger Institute, Baltimore, MD, USA
| | - Erica Greenberg
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Matthew E Hirschtritt
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Alden Y Huang
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Cornelia Illmann
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Robert A King
- Yale Child Study Center and the Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Roger Kurlan
- Atlantic Neuroscience Institute, Overlook Hospital, Summit, NJ, USA
| | - James F Leckman
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, USA
| | - Gholson J Lyon
- Jervis Clinic, NYS Institute for Basic Research in Developmental Disabilities (IBR), Staten Island, NY, USA
| | - Irene A Malaty
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, USA
| | - Carol A Mathews
- Department of Psychiatry, Genetics Institute, University of Florida, Gainesville, FL, USA
| | - William M McMahon
- Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
| | - Benjamin M Neale
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Michael S Okun
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, USA
| | - Lisa Osiecki
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Mary M Robertson
- Division of Psychiatry, Department of Neuropsychiatry, University College London, London, UK
| | - Guy A Rouleau
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
| | - Paul Sandor
- University Health Network, Youthdale Treatment Centres, and University of Toronto, Toronto, Canada
| | - Jeremiah M Scharf
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Neurology, Brigham and Women's Hospital, and the Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Harvey S Singer
- Johns Hopkins University School of Medicine and the Kennedy Krieger Institute, Baltimore, MD, USA
| | - Jan H Smit
- Department of Psychiatry, VU UniversityMedical Center, Amsterdam, The Netherlands
| | - Jae Hoon Sul
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Dongmei Yu
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Harald Aschauer Harald Aschauer
- Department of Psychiatry and Psychotherapy, Medical University Vienna, Vienna, Austria
- Biopsychosocial Corporation, Vienna, Austria
| | - Csaba Barta
- Institute of Medical Chemistry, Molecular Biology, and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Cathy L Budman
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Danielle C Cath
- Department of Psychiatry, University Medical Center Groningen and Rijksuniversity Groningen, and Drenthe Mental Health Center, Groningen, the Netherlands
| | - Christel Depienne
- Institute of Human Genetics, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1127, CNRS UMR 7225, ICM, Paris, France
| | - Andreas Hartmann
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1127, CNRS UMR 7225, ICM, Paris, France
- French Reference Centre for Gilles de la Tourette Syndrome, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
- Assistance Publique-Hôpitaux de Paris, Department of Neurology, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Johannes Hebebrand
- Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Anastasios Konstantinidis
- Department of Psychiatry and Psychotherapy, Medical University Vienna, Vienna, Austria
- Center for Mental Health Muldenstrasse, BBRZMed, Linz, Austria
| | - Carol A Mathews
- Department of Psychiatry, Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Kirsten Müller-Vahl
- Clinic of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Peter Nagy
- Vadaskert Child and Adolescent Psychiatric Hospital, Budapest, Hungary
| | - Markus M Nöthen
- Institute of Human Genetics, University Hospital Bonn, University of Bonn Medical School, Bonn, Germany
| | - Peristera Paschou
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Renata Rizzo
- Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Guy A Rouleau
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
| | - Paul Sandor
- University Health Network, Youthdale Treatment Centres, and University of Toronto, Toronto, Canada
| | - Jeremiah M Scharf
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Neurology, Brigham and Women's Hospital, and the Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Mara Stamenkovic
- Department of Psychiatry and Psychotherapy, Medical University Vienna, Vienna, Austria
| | - Manfred Stuhrmann
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Fotis Tsetsos
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Zsanett Tarnok
- Vadaskert Child and Adolescent Psychiatric Hospital, Budapest, Hungary
| | - Tomasz Wolanczyk
- Department of Child Psychiatry, Medical University of Warsaw, 00-001, Warsaw, Poland
| | - Yulia Worbe
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1127, CNRS UMR 7225, ICM, Paris, France
- French Reference Centre for Gilles de la Tourette Syndrome, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
- Assistance Publique-Hôpitaux de Paris, Department of Neurology, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
- Assistance Publique Hôpitaux de Paris, Hopital Saint Antoine, Paris, France
| | - Lawrence Brown
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Keun-Ah Cheon
- Yonsei University College of Medicine, Yonsei Yoo & Kim Mental Health Clinic, Seoul, South Korea
| | - Barbara J Coffey
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrea Dietrich
- Department of Child and Adolescent Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Thomas V Fernandez
- Yale Child Study Center and the Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Blanca Garcia-Delgar
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clinic Universitari, Barcelona, Spain
| | - Donald Gilbert
- Division of Pediatric Neurology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, Cincinnati, USA
| | - Dorothy E Grice
- Department of Psychiatry, Friedman Brain Institute, Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julie Hagstrøm
- Child and Adolescent Mental Health Center, Mental Health Services, Capital Region of Denmark and University of Copenhagen, Copenhagen, Denmark
| | - Tammy Hedderly
- Tic and Neurodevelopmental Movements Service (TANDeM), Evelina Children's Hospital, Guys and St Thomas' NHS Foundation Trust, London, UK
- Paediatric Neurosciences, Kings College London, London, UK
| | - Gary A Heiman
- Department of Genetics and the Human Genetics Institute of New Jersey, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Isobel Heyman
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- Psychological and Mental Health Services, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Pieter J Hoekstra
- Department of Child and Adolescent Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Chaim Huyser
- De Bascule, Academic Centre for Child and Adolescent Psychiatry, Amsterdam, The Netherlands
| | | | - Young-Shin Kim
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - Robert A King
- Yale Child Study Center and the Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Yun-Joo Koh
- The Korea Institute for Children's Social Development, Rudolph Child Research Center, Seoul, South Korea
| | - Sodahm Kook
- Kangbuk Samsung Hospital, Seoul, South Korea
| | - Samuel Kuperman
- University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Bennett L Leventhal
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - Marcos Madruga-Garrido
- Sección de Neuropediatría, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Pablo Mir
- Hospital Universitario Virgen del Rocío, Sevilla, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Astrid Morer
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clínic Universitari, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en red de Salud Mental (CIBERSAM), Barcelona, Spain
| | - Alexander Münchau
- Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
| | - Kerstin J Plessen
- Child and Adolescent Mental Health Centre, Mental Health Services, Capital Region of Denmark, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Service of Child and Adolescent Psychiatry, Department of Psychiatry, University Medical Center, University of Lausanne, Lausanne, Switzerland
| | - Veit Roessner
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, University Hospital Carl Gustav CarusTU Dresden, Dresden, Germany
| | - Eun-Young Shin
- Yonsei University College of Medicine, Yonsei Yoo & Kim Mental Health Clinic, Seoul, South Korea
| | - Dong-Ho Song
- Yonsei University College of Medicine, Yonsei Yoo & Kim Mental Health Clinic, Seoul, South Korea
| | - Jungeun Song
- National Health Insurance Service Ilsan Hospital, Goyang-Si, South Korea
| | - Jay A Tischfield
- Department of Genetics and the Human Genetics Institute of New Jersey, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - A Jeremy Willsey
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Samuel Zinner
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Harald Aschauer
- Department of Psychiatry and Psychotherapy, Medical University Vienna, Vienna, Austria
- Biopsychosocial Corporation, Vienna, Austria
| | - Cathy L Barr
- Krembil Research Institute, University Health Network, Hospital for Sick Children, and University of Toronto, Toronto, Canada
| | - Csaba Barta
- Institute of Medical Chemistry, Molecular Biology, and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | | | - Cheston Berlin
- Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Lawrence Brown
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Cathy L Budman
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Danielle C Cath
- Department of Psychiatry, University Medical Center Groningen and Rijksuniversity Groningen, and Drenthe Mental Health Center, Groningen, the Netherlands
| | - Barbara J Coffey
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Giovanni Coppola
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Nancy J Cox
- Division of Genetic Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sabrina Darrow
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Lea K Davis
- Division of Genetic Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christel Depienne
- Institute of Human Genetics, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1127, CNRS UMR 7225, ICM, Paris, France
| | - Andrea Dietrich
- Department of Child and Adolescent Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Yves Dion
- McGill University Health Center, University of Montreal, McGill University Health Centre, Montreal, Canada
| | - Thomas Fernandez
- Yale Child Study Center and the Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Nelson B Freimer
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Donald Gilbert
- Division of Pediatric Neurology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, Cincinnati, USA
| | - Marco A Grados
- Johns Hopkins University School of Medicine and the Kennedy Krieger Institute, Baltimore, MD, USA
| | - Erica Greenberg
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Andreas Hartmann
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1127, CNRS UMR 7225, ICM, Paris, France
- French Reference Centre for Gilles de la Tourette Syndrome, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
- Assistance Publique-Hôpitaux de Paris, Department of Neurology, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Johannes Hebebrand
- Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Gary Heiman
- Department of Genetics and the Human Genetics Institute of New Jersey, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Matthew E Hirschtritt
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Pieter Hoekstra
- Department of Child and Adolescent Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Alden Y Huang
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Cornelia Illmann
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Joseph Jankovic
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Robert A King
- Yale Child Study Center and the Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Samuel Kuperman
- University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Paul C Lee
- Tripler Army Medical Center and University of Hawaii John A. Burns School of Medicine, Honolulu, HI, USA
| | - Gholson J Lyon
- Jervis Clinic, NYS Institute for Basic Research in Developmental Disabilities (IBR), Staten Island, NY, USA
| | - Irene A Malaty
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, USA
| | - Carol A Mathews
- Department of Psychiatry, Genetics Institute, University of Florida, Gainesville, FL, USA
| | - William M McMahon
- Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
| | - Kirsten Müller-Vahl
- Clinic of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Peter Nagy
- Vadaskert Child and Adolescent Psychiatric Hospital, Budapest, Hungary
| | - Benjamin M Neale
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Markus M Nöthen
- Institute of Human Genetics, University Hospital Bonn, University of Bonn Medical School, Bonn, Germany
| | - Michael S Okun
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, USA
| | - Lisa Osiecki
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Peristera Paschou
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Renata Rizzo
- Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Mary M Robertson
- Division of Psychiatry, Department of Neuropsychiatry, University College London, London, UK
| | - Guy A Rouleau
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
| | - Paul Sandor
- University Health Network, Youthdale Treatment Centres, and University of Toronto, Toronto, Canada
| | - Jeremiah M Scharf
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Neurology, Brigham and Women's Hospital, and the Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Harvey S Singer
- Johns Hopkins University School of Medicine and the Kennedy Krieger Institute, Baltimore, MD, USA
| | - Mara Stamenkovic
- Department of Psychiatry and Psychotherapy, Medical University Vienna, Vienna, Austria
| | - Manfred Stuhrmann
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Jae Hoon Sul
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Zsanett Tarnok
- Vadaskert Child and Adolescent Psychiatric Hospital, Budapest, Hungary
| | - Jay Tischfield
- Department of Genetics and the Human Genetics Institute of New Jersey, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Fotis Tsetsos
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - A Jeremy Willsey
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Douglas Woods
- Marquette University and University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Yulia Worbe
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1127, CNRS UMR 7225, ICM, Paris, France
- French Reference Centre for Gilles de la Tourette Syndrome, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
- Assistance Publique-Hôpitaux de Paris, Department of Neurology, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
- Assistance Publique Hôpitaux de Paris, Hopital Saint Antoine, Paris, France
| | - Dongmei Yu
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Samuel Zinner
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| |
Collapse
|
23
|
Yang A, Chen J, Zhao XM. nMAGMA: a network-enhanced method for inferring risk genes from GWAS summary statistics and its application to schizophrenia. Brief Bioinform 2020; 22:5998843. [PMID: 33230537 DOI: 10.1093/bib/bbaa298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/21/2020] [Accepted: 10/07/2020] [Indexed: 12/21/2022] Open
Abstract
MOTIVATION Annotating genetic variants from summary statistics of genome-wide association studies (GWAS) is crucial for predicting risk genes of various disorders. The multimarker analysis of genomic annotation (MAGMA) is one of the most popular tools for this purpose, where MAGMA aggregates signals of single nucleotide polymorphisms (SNPs) to their nearby genes. In biology, SNPs may also affect genes that are far away in the genome, thus missed by MAGMA. Although different upgrades of MAGMA have been proposed to extend gene-wise variant annotations with more information (e.g. Hi-C or eQTL), the regulatory relationships among genes and the tissue specificity of signals have not been taken into account. RESULTS We propose a new approach, namely network-enhanced MAGMA (nMAGMA), for gene-wise annotation of variants from GWAS summary statistics. Compared with MAGMA and H-MAGMA, nMAGMA significantly extends the lists of genes that can be annotated to SNPs by integrating local signals, long-range regulation signals (i.e. interactions between distal DNA elements), and tissue-specific gene networks. When applied to schizophrenia (SCZ), nMAGMA is able to detect more risk genes (217% more than MAGMA and 57% more than H-MAGMA) that are involved in SCZ compared with MAGMA and H-MAGMA, and more of nMAGMA results can be validated with known SCZ risk genes. Some disease-related functions (e.g. the ATPase pathway in Cortex) are also uncovered in nMAGMA but not in MAGMA or H-MAGMA. Moreover, nMAGMA provides tissue-specific risk signals, which are useful for understanding disorders with multitissue origins.
Collapse
Affiliation(s)
- Anyi Yang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, China
| | - Jingqi Chen
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, China
| | | |
Collapse
|
24
|
Genetic Variation in CNS Myelination and Functional Brain Connectivity in Recombinant Inbred Mice. Cells 2020; 9:cells9092119. [PMID: 32961889 PMCID: PMC7564997 DOI: 10.3390/cells9092119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 01/29/2023] Open
Abstract
Myelination greatly increases the speed of action potential propagation of neurons, thereby enhancing the efficacy of inter-neuronal communication and hence, potentially, optimizing the brain’s signal processing capability. The impact of genetic variation on the extent of axonal myelination and its consequences for brain functioning remain to be determined. Here we investigated this question using a genetic reference panel (GRP) of mouse BXD recombinant inbred (RI) strains, which partly model genetic diversity as observed in human populations, and which show substantial genetic differences in a variety of behaviors, including learning, memory and anxiety. We found coherent differences in the expression of myelin genes in brain tissue of RI strains of the BXD panel, with the largest differences in the hippocampus. The parental C57BL/6J (C57) and DBA/2J (DBA) strains were on opposite ends of the expression spectrum, with C57 showing higher myelin transcript expression compared with DBA. Our experiments showed accompanying differences between C57 and DBA in myelin protein composition, total myelin content, and white matter conduction velocity. Finally, the hippocampal myelin gene expression of the BXD strains correlated significantly with behavioral traits involving anxiety and/or activity. Taken together, our data indicate that genetic variation in myelin gene expression translates to differences observed in myelination, axonal conduction speed, and possibly in anxiety/activity related behaviors.
Collapse
|
25
|
Sokolowski M, Wasserman D. Genetic origins of suicidality? A synopsis of genes in suicidal behaviours, with regard to evidence diversity, disorder specificity and neurodevelopmental brain transcriptomics. Eur Neuropsychopharmacol 2020; 37:1-11. [PMID: 32636053 DOI: 10.1016/j.euroneuro.2020.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 06/08/2020] [Indexed: 12/17/2022]
Abstract
With regard to suicidal behavior (SB) genetics, many novel genes have been implicated over the years, in particular by a variety of hypothesis-free genomic methods (e.g. GWAS and exome sequencing). In addition, many novel SB gene findings appear enigmatic in their biological relevance and have weak statistical support, e.g. lack direct replications. Adding to this is the comorbidity between psychiatric disorders and SB. Here we provide a synopsis of SB genes, by prioritization of 106 (out of ~2500) genes based on their highest level of evidence diversity across mainly five genetic evidence types (candidate/GWAS SNP, CNV, linkage and whole exome sequencing), supplemented by three functional categories. This is a representative set of both old and new SB gene candidates, implicated by all kinds of evidence. Furthermore, we define a subset of 40 SB "specific" genes, which are not found among ~3900 genes implicated in other neuropsychiatric disorders, e.g. Autism spectrum disorders (ASD) or Schizophrenia. Biological research of suicidality contains a major developmental focus, e.g. with regard to the gene-environment interactions and epigenetic effects during childhood. Less is known about early (fetal) development and SB genes. Inspired by huge efforts to understand the role early (fetal) neurodevelopment in e.g. ASD by using brain transcriptomic data, we here also characterize the 106 SB genes. We find interesting spatiotemporal expression differences and similarities between SB specific and non-specific genes during brain neurodevelopment. These aspects are of interest to investigate further, to better understand and counteract the genetic origins suicidality.
Collapse
Affiliation(s)
- Marcus Sokolowski
- National Centre for Suicide Research and Prevention of Mental Ill-Health (NASP), Karolinska Institute (KI), Stockholm, Sweden.
| | - Danuta Wasserman
- National Centre for Suicide Research and Prevention of Mental Ill-Health (NASP), Karolinska Institute (KI), Stockholm, Sweden; WHO collaborating Centre for research, methods, development and training in suicide prevention, Sweden
| |
Collapse
|
26
|
Dysregulated Glial Differentiation in Schizophrenia May Be Relieved by Suppression of SMAD4- and REST-Dependent Signaling. Cell Rep 2020; 27:3832-3843.e6. [PMID: 31242417 DOI: 10.1016/j.celrep.2019.05.088] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 04/04/2019] [Accepted: 05/22/2019] [Indexed: 12/22/2022] Open
Abstract
Astrocytic differentiation is developmentally impaired in patients with childhood-onset schizophrenia (SCZ). To determine why, we used genetic gain- and loss-of-function studies to establish the contributions of differentially expressed transcriptional regulators to the defective differentiation of glial progenitor cells (GPCs) produced from SCZ patient-derived induced pluripotent cells (iPSCs). Negative regulators of the bone morphogenetic protein (BMP) pathway were upregulated in SCZ GPCs, including BAMBI, FST, and GREM1, whose overexpression retained SCZ GPCs at the progenitor stage. SMAD4 knockdown (KD) suppressed the production of these BMP inhibitors by SCZ GPCs and rescued normal astrocytic differentiation. In addition, the BMP-regulated transcriptional repressor REST was upregulated in SCZ GPCs, and its KD similarly restored normal glial differentiation. REST KD also rescued potassium-transport-associated gene expression and K+ uptake, which were otherwise deficient in SCZ glia. These data suggest that the glial differentiation defect in childhood-onset SCZ, and its attendant disruption in K+ homeostasis, may be rescued by targeting BMP/SMAD4- and REST-dependent transcription.
Collapse
|
27
|
Roman C, Egert L, Di Benedetto B. Astrocytic-neuronal crosstalk gets jammed: Alternative perspectives on the onset of neuropsychiatric disorders. Eur J Neurosci 2020; 54:5717-5729. [PMID: 32644273 DOI: 10.1111/ejn.14900] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 06/09/2020] [Accepted: 07/03/2020] [Indexed: 12/12/2022]
Abstract
Investigating interactions of glia cells and synapses during development and in adulthood is the focus of several research programmes which aim at understanding the neurobiology of brain physiological and pathological processes. Both glia-specific released and membrane-bound proteins play essential roles in the development, maintenance and functionality of synaptic connections. Alterations in synaptic contacts in specific brain areas are hallmarks of several brain diseases, such as major depressive disorder, autism spectrum disorder and schizophrenia. Thus, a deeper knowledge about putative astrocyte dysfunctions which might affect the synaptic compartment is warranted to improve treatment options. Here, we present the latest advances about the role of glia cells in orchestrating the arrangement of synapses and neuronal networks in physiological and pathological states. We specifically focus on the role of astrocytes in the phagocytosis of neuronal synapses as a novel mechanism which drives the refinement of neuronal circuits and might be affected in pathological conditions. Finally, we propose this astrocyte-dependent mechanism as a putative alternative target of pharmacological interventions for the treatment of brain disorders.
Collapse
Affiliation(s)
- Celia Roman
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Luisa Egert
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Barbara Di Benedetto
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany.,Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| |
Collapse
|
28
|
Goldman SA. Glial evolution as a determinant of human behavior and its disorders. Ann N Y Acad Sci 2020; 1471:72-85. [PMID: 32449961 DOI: 10.1111/nyas.14372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 04/24/2020] [Accepted: 04/24/2020] [Indexed: 01/08/2023]
Abstract
Astroglial complexity and pleomorphism have increased significantly with hominid evolution. This suggests a potential association between glial evolution and the development of human cognition, as well as between glial evolution and the advent of human-selective neurodegenerative and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York.,Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Science, Copenhagen N, Denmark.,Neuroscience Center, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
29
|
Rodrigues-Amorim D, Rivera-Baltanás T, Vallejo-Curto MDC, Rodriguez-Jamardo C, de las Heras E, Barreiro-Villar C, Blanco-Formoso M, Fernández-Palleiro P, Álvarez-Ariza M, López M, García-Caballero A, Olivares JM, Spuch C. Proteomics in Schizophrenia: A Gateway to Discover Potential Biomarkers of Psychoneuroimmune Pathways. Front Psychiatry 2019; 10:885. [PMID: 31849731 PMCID: PMC6897280 DOI: 10.3389/fpsyt.2019.00885] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 11/11/2019] [Indexed: 12/17/2022] Open
Abstract
Schizophrenia is a severe and disabling psychiatric disorder with a complex and multifactorial etiology. The lack of consensus regarding the multifaceted dysfunction of this ailment has increased the need to explore new research lines. This research makes use of proteomics data to discover possible analytes associated with psychoneuroimmune signaling pathways in schizophrenia. Thus, we analyze plasma of 45 patients [10 patients with first-episode schizophrenia (FES) and 35 patients with chronic schizophrenia] and 43 healthy subjects by label-free liquid chromatography-tandem mass spectrometry. The analysis revealed a significant reduction in the levels of glia maturation factor beta (GMF-β), the brain-derived neurotrophic factor (BDNF), and the 115-kDa isoform of the Rab3 GTPase-activating protein catalytic subunit (RAB3GAP1) in patients with schizophrenia as compared to healthy volunteers. In conclusion, GMF-β, BDNF, and 115-kDa isoform of RAB3GAP1 showed significantly reduced levels in plasma of patients with schizophrenia, thus making them potential biomarkers in schizophrenia.
Collapse
Affiliation(s)
- Daniela Rodrigues-Amorim
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Tania Rivera-Baltanás
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - María del Carmen Vallejo-Curto
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Cynthia Rodriguez-Jamardo
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Elena de las Heras
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Carolina Barreiro-Villar
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - María Blanco-Formoso
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Patricia Fernández-Palleiro
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - María Álvarez-Ariza
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Marta López
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Alejandro García-Caballero
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
- Department of Psychiatry, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - José Manuel Olivares
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Carlos Spuch
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| |
Collapse
|
30
|
Chen X, Wang F, Gan J, Zhang Z, Liang X, Li T, Huang N, Zhao X, Mei F, Xiao L. Myelin Deficits Caused by Olig2 Deficiency Lead to Cognitive Dysfunction and Increase Vulnerability to Social Withdrawal in Adult Mice. Neurosci Bull 2019; 36:419-426. [PMID: 31758330 DOI: 10.1007/s12264-019-00449-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/27/2019] [Indexed: 01/25/2023] Open
Abstract
Oligodendrocyte (OL) and myelin development are crucial for network integration and are associated with higher brain functions. Accumulating evidence has demonstrated structural and functional impairment of OLs and myelin in serious mental illnesses. However, whether these deficits contribute to the brain dysfunction or pathogenesis of such diseases still lacks direct evidence. In this study, we conditionally deleted Olig2 in oligodendroglial lineage cells (Olig2 cKO) and screened the behavioral changes in adult mice. We found that Olig2 ablation impaired myelin development, which further resulted in severe hypomyelination in the anterior cingulate cortex. Strikingly, Olig2 cKO mice exhibited an anxious phenotype, aberrant responses to stress, and cognitive deficits. Moreover, Olig2 cKO mice showed increased vulnerability to social avoidance under the mild stress of social isolation. Together, these results indicate that developmental deficits in OL and myelin lead to cognitive impairment and increase the risk of phenotypes reminiscent of mental illnesses.
Collapse
Affiliation(s)
- Xianjun Chen
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University (Third Military Medical University), Chongqing, 400038, China.,Department of Psychiatry, Mental Diseases Prevention and Treatment Institute of The People's Liberation Army (PLA), 988 Hospital of Joint Logistic Support Force of PLA, Jiaozuo, 454003, China
| | - Fei Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Jingli Gan
- Department of Psychiatry, Mental Diseases Prevention and Treatment Institute of The People's Liberation Army (PLA), 988 Hospital of Joint Logistic Support Force of PLA, Jiaozuo, 454003, China
| | - Zhonghua Zhang
- Department of Psychiatry, Mental Diseases Prevention and Treatment Institute of The People's Liberation Army (PLA), 988 Hospital of Joint Logistic Support Force of PLA, Jiaozuo, 454003, China
| | - Xuejun Liang
- Department of Psychiatry, Mental Diseases Prevention and Treatment Institute of The People's Liberation Army (PLA), 988 Hospital of Joint Logistic Support Force of PLA, Jiaozuo, 454003, China
| | - Tao Li
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Nanxin Huang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xiaofeng Zhao
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 310036, China
| | - Feng Mei
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Lan Xiao
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
31
|
Palaniyappan L, Al-Radaideh A, Mougin O, Das T, Gowland P, Liddle PF. Aberrant myelination of the cingulum and Schneiderian delusions in schizophrenia: a 7T magnetization transfer study. Psychol Med 2019; 49:1890-1896. [PMID: 30229713 DOI: 10.1017/s0033291718002647] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND The structural integrity of the anterior cingulum has been repeatedly observed to be abnormal in patients with schizophrenia. More recently, aberrant myelination of frontal fasciculi, especially, cingulum has been proposed to underlie delayed corollary discharges that can affect sense of agency and contribute to delusions of control (Schneiderian delusions). Using the magnetization transfer phenomenon at an ultra-high field 7T MRI, we investigated the putative myelin content of cingulum bundle in patients with schizophrenia. METHODS Seventeen clinically stable patients with schizophrenia and 20 controls were recruited for this 7T MRI study. We used a region-of-interest method and extracted magnetization transfer ratio (MTR) from left and right dorsal cingulum bundles and estimated patients v. controls differences. We also related the cingulum MTR values to the severity of Schneiderian delusions. RESULTS Patients had a significant reduction in the MTR, indicating reduced myelin content, in the cingulum bundle (right cingulum Hedges' g = 0.91; left cingulum g = 0.03). The reduced MTR of left cingulum was associated with higher severity of Schneiderian delusions (τ = -0.45, p = 0.026) but no such relationship was seen for the right cingulum MTR (τ = -0.136, p = 0.50) among patients. The association between the left cingulum MTR and Schneiderian delusions was not explained by the presence of other delusions, hallucinations, disorganization or negative symptoms. CONCLUSIONS Dysmyelination of the cingulum bundle is seen in a subgroup of patients with schizophrenia and may be involved in the mechanism of Schneiderian delusions.
Collapse
Affiliation(s)
- Lena Palaniyappan
- Robarts Research Institute, University of Western Ontario,London, Ontario,Canada
| | - Ali Al-Radaideh
- Department of Medical Imaging, Faculty of Allied Health Sciences,The Hashemite University,Zarqa,Jordan
| | - Olivier Mougin
- Sir Peter Mansfield Imaging Centre (SPMIC), School of Physics and Astronomy, University of Nottingham,Nottingham,UK
| | - Tushar Das
- Robarts Research Institute, University of Western Ontario,London, Ontario,Canada
| | - Penny Gowland
- Sir Peter Mansfield Imaging Centre (SPMIC), School of Physics and Astronomy, University of Nottingham,Nottingham,UK
| | - Peter F Liddle
- Translational Neuroimaging for Mental Health, Division of Psychiatry and Applied Psychology,University of Nottingham,Nottingham,UK
| |
Collapse
|
32
|
Rapid-Onset Psychotic Symptoms After Interferon-β-1a Treatment of Multiple Sclerosis. Am J Ther 2019; 27:e631-e632. [PMID: 31232706 DOI: 10.1097/mjt.0000000000001017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
33
|
de Vrij FM, Bouwkamp CG, Gunhanlar N, Shpak G, Lendemeijer B, Baghdadi M, Gopalakrishna S, Ghazvini M, Li TM, Quadri M, Olgiati S, Breedveld GJ, Coesmans M, Mientjes E, de Wit T, Verheijen FW, Beverloo HB, Cohen D, Kok RM, Bakker PR, Nijburg A, Spijker AT, Haffmans PMJ, Hoencamp E, Bergink V, Vorstman JA, Wu T, Olde Loohuis LM, Amin N, Langen CD, Hofman A, Hoogendijk WJ, van Duijn CM, Ikram MA, Vernooij MW, Tiemeier H, Uitterlinden AG, Elgersma Y, Distel B, Gribnau J, White T, Bonifati V, Kushner SA. Candidate CSPG4 mutations and induced pluripotent stem cell modeling implicate oligodendrocyte progenitor cell dysfunction in familial schizophrenia. Mol Psychiatry 2019; 24:757-771. [PMID: 29302076 PMCID: PMC6755981 DOI: 10.1038/s41380-017-0004-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 09/24/2017] [Accepted: 11/01/2017] [Indexed: 01/09/2023]
Abstract
Schizophrenia is highly heritable, yet its underlying pathophysiology remains largely unknown. Among the most well-replicated findings in neurobiological studies of schizophrenia are deficits in myelination and white matter integrity; however, direct etiological genetic and cellular evidence has thus far been lacking. Here, we implement a family-based approach for genetic discovery in schizophrenia combined with functional analysis using induced pluripotent stem cells (iPSCs). We observed familial segregation of two rare missense mutations in Chondroitin Sulfate Proteoglycan 4 (CSPG4) (c.391G > A [p.A131T], MAF 7.79 × 10-5 and c.2702T > G [p.V901G], MAF 2.51 × 10-3). The CSPG4A131T mutation was absent from the Swedish Schizophrenia Exome Sequencing Study (2536 cases, 2543 controls), while the CSPG4V901G mutation was nominally enriched in cases (11 cases vs. 3 controls, P = 0.026, OR 3.77, 95% CI 1.05-13.52). CSPG4/NG2 is a hallmark protein of oligodendrocyte progenitor cells (OPCs). iPSC-derived OPCs from CSPG4A131T mutation carriers exhibited abnormal post-translational processing (P = 0.029), subcellular localization of mutant NG2 (P = 0.007), as well as aberrant cellular morphology (P = 3.0 × 10-8), viability (P = 8.9 × 10-7), and myelination potential (P = 0.038). Moreover, transfection of healthy non-carrier sibling OPCs confirmed a pathogenic effect on cell survival of both the CSPG4A131T (P = 0.006) and CSPG4V901G (P = 3.4 × 10-4) mutations. Finally, in vivo diffusion tensor imaging of CSPG4A131T mutation carriers demonstrated a reduction of brain white matter integrity compared to unaffected sibling and matched general population controls (P = 2.2 × 10-5). Together, our findings provide a convergence of genetic and functional evidence to implicate OPC dysfunction as a candidate pathophysiological mechanism of familial schizophrenia.
Collapse
Affiliation(s)
- Femke M de Vrij
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Christian G Bouwkamp
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Nilhan Gunhanlar
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Guy Shpak
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Bas Lendemeijer
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Maarouf Baghdadi
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Mehrnaz Ghazvini
- Department of Developmental Biology, and Erasmus MC iPS Facility, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Tracy M Li
- Department of Developmental Biology, and Erasmus MC iPS Facility, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marialuisa Quadri
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Simone Olgiati
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Guido J Breedveld
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Michiel Coesmans
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
- Delta Psychiatric Center, Poortugaal, The Netherlands
| | - Edwin Mientjes
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ton de Wit
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Frans W Verheijen
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - H Berna Beverloo
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Dan Cohen
- Mental Health Care Organization North-Holland North, Heerhugowaard, The Netherlands
| | - Rob M Kok
- Parnassia Psychiatric Institute, The Hague, The Netherlands
| | - P Roberto Bakker
- Department of Psychiatry and Psychology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
- Psychiatric Center GGZ Centraal, Amersfoort, The Netherlands
| | - Aviva Nijburg
- Parnassia Psychiatric Institute, The Hague, The Netherlands
| | | | - P M Judith Haffmans
- Faculty of Social and Behavioral Sciences Clinical, Health and Neuro Psychology, Department of Affective Disorders, PsyQ, Leiden University, Leiden, The Netherlands
| | - Erik Hoencamp
- Parnassia Psychiatric Institute, The Hague, The Netherlands
- Institute of Psychology, Leiden University, Leiden, The Netherlands
| | - Veerle Bergink
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jacob A Vorstman
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Psychiatry, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Timothy Wu
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Loes M Olde Loohuis
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Najaf Amin
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Carolyn D Langen
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
- Biomedical Imaging Group Rotterdam, Departments of Radiology & Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Witte J Hoogendijk
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - M Arfan Ikram
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Henning Tiemeier
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ype Elgersma
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ben Distel
- Department of Medical Biochemistry, Academic Medical Centre, Amsterdam, The Netherlands
| | - Joost Gribnau
- Department of Developmental Biology, and Erasmus MC iPS Facility, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Tonya White
- Department of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Vincenzo Bonifati
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Steven A Kushner
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
34
|
Lu X, Zhang D, Shoji H, Duan C, Zhang G, Isaji T, Wang Y, Fukuda T, Gu J. Deficiency of α1,6-fucosyltransferase promotes neuroinflammation by increasing the sensitivity of glial cells to inflammatory mediators. Biochim Biophys Acta Gen Subj 2019; 1863:598-608. [DOI: 10.1016/j.bbagen.2018.12.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/05/2018] [Accepted: 12/13/2018] [Indexed: 10/27/2022]
|
35
|
Effects on Glial Cell Glycolysis in Schizophrenia: An Advanced Aging Phenotype? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1178:25-38. [DOI: 10.1007/978-3-030-25650-0_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
36
|
Chen X, Liu H, Gan J, Wang X, Yu G, Li T, Liang X, Yu B, Xiao L. Quetiapine Modulates Histone Methylation Status in Oligodendroglia and Rescues Adolescent Behavioral Alterations of Socially Isolated Mice. Front Psychiatry 2019; 10:984. [PMID: 32082195 PMCID: PMC7005666 DOI: 10.3389/fpsyt.2019.00984] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 12/11/2019] [Indexed: 01/08/2023] Open
Abstract
Epigenetic alterations and impaired oligodendroglial myelination in the prefrontal cortex have been shown to correlate with behavioral and cognitive dysfunctions in social deprivation. Our previous study demonstrated that quetiapine, an atypical antipsychotic, could promote oligodendroglial differentiation and myelination. However, whether and how quetiapine could be beneficial in modulating aberrant epigenetic alterations in oligodendroglial cells and relieving behavioral alterations from social isolation is unknown. In this study, quetiapine was orally administered in adolescent mice undergoing mild stress of social isolation. We firstly confirmed that social isolation during a novel adolescent period could impair sociability, but not locomotive behaviors in mice. Moreover, quetiapine alleviated myelin deficits, and increased levels of histone methylation (H3K9me3) in mature oligodendroglia in the prefrontal cortex of socially isolated mice. Strikingly, quetiapine treatment significantly increased locomotive activity, and successfully reversed social avoidance behavior of the socially isolated mice. Taken together, our data suggest that quetiapine may rescue behavioral changes from social isolation through modulating epigenetic status toward the beneficial direction for oligodendroglial maturation, providing new insights into the pharmacological mechanism of quetiapine for mental illnesses.
Collapse
Affiliation(s)
- Xianjun Chen
- Department of Histology and Embryology, Institute of Brain and Intelligence, Army Medical University (Third Military Medical University), Chongqing, China.,Department of Psychiatry, Mental Diseases Prevention and Treatment Institute of PLA, No.988 Hospital of Joint Logistic Support Force of PLA, Jiaozuo, China
| | - Hao Liu
- Department of Histology and Embryology, Institute of Brain and Intelligence, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jingli Gan
- Department of Psychiatry, Mental Diseases Prevention and Treatment Institute of PLA, No.988 Hospital of Joint Logistic Support Force of PLA, Jiaozuo, China
| | - Xiaorui Wang
- Department of Histology and Embryology, Institute of Brain and Intelligence, Army Medical University (Third Military Medical University), Chongqing, China
| | - Guangdan Yu
- Department of Histology and Embryology, Institute of Brain and Intelligence, Army Medical University (Third Military Medical University), Chongqing, China
| | - Tao Li
- Department of Histology and Embryology, Institute of Brain and Intelligence, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xuejun Liang
- Department of Psychiatry, Mental Diseases Prevention and Treatment Institute of PLA, No.988 Hospital of Joint Logistic Support Force of PLA, Jiaozuo, China
| | - Bin Yu
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Lan Xiao
- Department of Histology and Embryology, Institute of Brain and Intelligence, Army Medical University (Third Military Medical University), Chongqing, China.,Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
37
|
Ultrastructural pathology of oligodendrocytes adjacent to microglia in prefrontal white matter in schizophrenia. NPJ SCHIZOPHRENIA 2018; 4:26. [PMID: 30546020 PMCID: PMC6292874 DOI: 10.1038/s41537-018-0068-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 11/21/2018] [Indexed: 12/13/2022]
Abstract
Microglial activation has been proposed to be involved in the pathophysiology of schizophrenia (SCZ). We hypothesized that dystrophic alterations of oligodendrocytes previously reported in the prefrontal white matter in SCZ might be associated with microglial activation in the acute state of SCZ. White matter of the prefrontal cortex (BA10) was studied in post-mortem brain tissue from 21 SCZ cases and 20 normal controls. The SCZ group included 12 subjects with predominantly positive symptoms and 9 subjects with predominantly negative symptoms. Electron microscopy was applied to estimate cell density, size, volume fraction (Vv) and the number (N) of organelles in oligodendrocytes adjacent to microglia and in oligodendrocytes adjacent to myelin, neurons and capillaries and not adjacent to microglia. Cell density of oligodendrocytes was not changed in the SCZ group as compared to controls. Vv and N of mitochondria were significantly decreased, while Vv of vacuoles of endoplasmic reticulum and lipofuscin granules were significantly increased in oligodendrocytes adjacent to either microglia or myelin in the SCZ group and in patients displaying predominantly positive symptoms as compared to the control group. There were no significant differences between oligodendrocytes adjacent to microglia and to myelin. Vv and N of lipofuscin were also increased in peri-capillary oligodendrocytes. There was no effect of clinical subgroups on the parameters of peri-capillary and peri-neuronal oligodendrocytes. Though many ameboid and dystrophic microglia adjacent to oligodendrocytes were found in the SCZ samples, we provide no quantitative evidence that oligodendrocyte dystrophy is associated with microglial activation in white matter in SCZ.
Collapse
|
38
|
Chen X, Duan H, Xiao L, Gan J. Genetic and Epigenetic Alterations Underlie Oligodendroglia Susceptibility and White Matter Etiology in Psychiatric Disorders. Front Genet 2018; 9:565. [PMID: 30524471 PMCID: PMC6262033 DOI: 10.3389/fgene.2018.00565] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/06/2018] [Indexed: 12/19/2022] Open
Abstract
Numerous genetic risk loci are found to associate with major neuropsychiatric disorders represented by schizophrenia. The pathogenic roles of genetic risk loci in psychiatric diseases are further complicated by the association with cell lineage- and/or developmental stage-specific epigenetic alterations. Besides aberrant assembly and malfunction of neuronal circuitry, an increasing volume of discoveries clearly demonstrate impairment of oligodendroglia and disruption of white matter integrity in psychiatric diseases. Nonetheless, whether and how genetic risk factors and epigenetic dysregulations for neuronal susceptibility may affect oligodendroglia is largely unknown. In this mini-review, we will discuss emerging evidence regarding the functional interplay between genetic risk loci and epigenetic factors, which may underlie compromised oligodendroglia and myelin development in neuropsychiatric disorders. Transcriptional and epigenetic factors are the major aspects affected in oligodendroglia. Moreover, multiple disease susceptibility genes are connected by epigenetically modulated transcriptional and post-transcriptional mechanisms. Oligodendroglia specific complex molecular orchestra may explain how distinct risk factors lead to the common clinical expression of white matter pathology of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Xianjun Chen
- Department of Psychiatry, Mental Diseases Prevention and Treatment Institute of PLA, PLA 91st Central Hospital, Jiaozuo, China
| | - Huifeng Duan
- Department of Psychiatry, Mental Diseases Prevention and Treatment Institute of PLA, PLA 91st Central Hospital, Jiaozuo, China
| | - Lan Xiao
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jingli Gan
- Department of Psychiatry, Mental Diseases Prevention and Treatment Institute of PLA, PLA 91st Central Hospital, Jiaozuo, China
| |
Collapse
|
39
|
Fitzgerald E, Boardman JP, Drake AJ. Preterm Birth and the Risk of Neurodevelopmental Disorders - Is There a Role for Epigenetic Dysregulation? Curr Genomics 2018; 19:507-521. [PMID: 30386170 PMCID: PMC6158617 DOI: 10.2174/1389202919666171229144807] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 04/06/2017] [Accepted: 12/17/2017] [Indexed: 12/29/2022] Open
Abstract
Preterm Birth (PTB) accounts for approximately 11% of all births worldwide each year and is a profound physiological stressor in early life. The burden of neuropsychiatric and developmental impairment is high, with severity and prevalence correlated with gestational age at delivery. PTB is a major risk factor for the development of cerebral palsy, lower educational attainment and deficits in cognitive functioning, and individuals born preterm have higher rates of schizophrenia, autistic spectrum disorder and attention deficit/hyperactivity disorder. Factors such as gestational age at birth, systemic inflammation, respiratory morbidity, sub-optimal nutrition, and genetic vulnerability are associated with poor outcome after preterm birth, but the mechanisms linking these factors to adverse long term outcome are poorly understood. One potential mechanism linking PTB with neurodevelopmental effects is changes in the epigenome. Epigenetic processes can be defined as those leading to altered gene expression in the absence of a change in the underlying DNA sequence and include DNA methylation/hydroxymethylation and histone modifications. Such epigenetic modifications may be susceptible to environmental stimuli, and changes may persist long after the stimulus has ceased, providing a mechanism to explain the long-term consequences of acute exposures in early life. Many factors such as inflammation, fluctuating oxygenation and excitotoxicity which are known factors in PTB related brain injury, have also been implicated in epigenetic dysfunction. In this review, we will discuss the potential role of epigenetic dysregulation in mediating the effects of PTB on neurodevelopmental outcome, with specific emphasis on DNA methylation and the α-ketoglutarate dependent dioxygenase family of enzymes.
Collapse
Affiliation(s)
| | | | - Amanda J. Drake
- Address correspondence to this author at the University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh EH16 4TJ, UK; Tel: 44 131 2426748; Fax: 44 131 2426779; E-mail:
| |
Collapse
|
40
|
McPhie DL, Nehme R, Ravichandran C, Babb SM, Ghosh SD, Staskus A, Kalinowski A, Kaur R, Douvaras P, Du F, Ongur D, Fossati V, Eggan K, Cohen BM. Oligodendrocyte differentiation of induced pluripotent stem cells derived from subjects with schizophrenias implicate abnormalities in development. Transl Psychiatry 2018; 8:230. [PMID: 30352993 PMCID: PMC6199264 DOI: 10.1038/s41398-018-0284-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 09/26/2018] [Indexed: 12/22/2022] Open
Abstract
Abnormalities of brain connectivity and signal transduction are consistently observed in individuals with schizophrenias (SZ). Underlying these anomalies, convergent in vivo, post mortem, and genomic evidence suggest abnormal oligodendrocyte (OL) development and function and lower myelination in SZ. Our primary hypothesis was that there would be abnormalities in the number of induced pluripotent stem (iPS) cell-derived OLs from subjects with SZ. Our secondary hypothesis was that these in vitro abnormalities would correlate with measures of white matter (WM) integrity and myelination in the same subjects in vivo, estimated from magnetic resonance imaging. Six healthy control (HC) and six SZ iPS cell lines, derived from skin fibroblasts from well-characterized subjects, were differentiated into OLs. FACS analysis of the oligodendrocyte-specific surface, glycoprotein O4, was performed at three time points of development (days 65, 75, and 85) to quantify the number of late oligodendrocyte progenitor cells (OPCs) and OLs in each line. Significantly fewer O4-positive cells developed from SZ versus HC lines (95% CI 1.0: 8.6, F1,10 = 8.06, p = 0.02). The difference was greater when corrected for age (95% CI 5.4:10.4, F1,8 = 53.6, p < 0.001). A correlation between myelin content in WM in vivo, estimated by magnetization transfer ratio (MTR) and number of O4-positive cells in vitro was also observed across all time points (F1,9 = 4.3, p = 0.07), reaching significance for mature OLs at day 85 in culture (r = 0.70, p < 0.02). Low production of OPCs may be a contributing mechanism underlying WM reduction in SZ.
Collapse
Affiliation(s)
- Donna L. McPhie
- 000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| | - Ralda Nehme
- grid.66859.34Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142 USA ,000000041936754Xgrid.38142.3cDepartment of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138 USA
| | - Caitlin Ravichandran
- 000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| | - Suzann M. Babb
- 000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| | - Sulagna Dia Ghosh
- grid.66859.34Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142 USA ,000000041936754Xgrid.38142.3cDepartment of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138 USA
| | - Alexandra Staskus
- 0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| | - Amy Kalinowski
- 0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| | - Rupinderjit Kaur
- 0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| | - Panagiotis Douvaras
- Blue Rock Therapeutics, Alexandria Center for Life Science, 450 E 29th Street, Suite 504, New York, NY 10016 USA
| | - Fei Du
- 000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| | - Dost Ongur
- 000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| | - Valentina Fossati
- 0000 0004 5906 3313grid.430819.7The New York Stem Cell Foundation Research Institute, 619 West 54th Street, 3rd Floor, New York, NY 10019 USA
| | - Kevin Eggan
- 000000041936754Xgrid.38142.3cDepartment of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138 USA ,grid.66859.34Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142 USA
| | - Bruce M. Cohen
- 000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2McLean Hospital, 115 Mill St., Belmont, MA 02478 USA
| |
Collapse
|
41
|
de Almeida V, Martins-de-Souza D. Cannabinoids and glial cells: possible mechanism to understand schizophrenia. Eur Arch Psychiatry Clin Neurosci 2018; 268:727-737. [PMID: 29392440 DOI: 10.1007/s00406-018-0874-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/24/2018] [Indexed: 01/03/2023]
Abstract
Clinical and neurobiological findings have reported the involvement of endocannabinoid signaling in the pathophysiology of schizophrenia. This system modulates dopaminergic and glutamatergic neurotransmission that is associated with positive, negative, and cognitive symptoms of schizophrenia. Despite neurotransmitter impairments, increasing evidence points to a role of glial cells in schizophrenia pathobiology. Glial cells encompass three main groups: oligodendrocytes, microglia, and astrocytes. These cells promote several neurobiological functions, such as myelination of axons, metabolic and structural support, and immune response in the central nervous system. Impairments in glial cells lead to disruptions in communication and in the homeostasis of neurons that play role in pathobiology of disorders such as schizophrenia. Therefore, data suggest that glial cells may be a potential pharmacological tool to treat schizophrenia and other brain disorders. In this regard, glial cells express cannabinoid receptors and synthesize endocannabinoids, and cannabinoid drugs affect some functions of these cells that can be implicated in schizophrenia pathobiology. Thus, the aim of this review is to provide data about the glial changes observed in schizophrenia, and how cannabinoids could modulate these alterations.
Collapse
Affiliation(s)
- Valéria de Almeida
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato 255, Campinas, SP, 13083-862, Brazil.
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato 255, Campinas, SP, 13083-862, Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil
| |
Collapse
|
42
|
Skene NG, Bryois J, Bakken TE, Breen G, Crowley JJ, Gaspar HA, Giusti-Rodriguez P, Hodge RD, Miller JA, Muñoz-Manchado AB, O'Donovan MC, Owen MJ, Pardiñas AF, Ryge J, Walters JTR, Linnarsson S, Lein ES, Sullivan PF, Hjerling-Leffler J. Genetic identification of brain cell types underlying schizophrenia. Nat Genet 2018; 50:825-833. [PMID: 29785013 PMCID: PMC6477180 DOI: 10.1038/s41588-018-0129-5] [Citation(s) in RCA: 379] [Impact Index Per Article: 63.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 04/03/2018] [Indexed: 12/17/2022]
Abstract
With few exceptions, the marked advances in knowledge about the genetic basis of schizophrenia have not converged on findings that can be confidently used for precise experimental modeling. By applying knowledge of the cellular taxonomy of the brain from single-cell RNA sequencing, we evaluated whether the genomic loci implicated in schizophrenia map onto specific brain cell types. We found that the common-variant genomic results consistently mapped to pyramidal cells, medium spiny neurons (MSNs) and certain interneurons, but far less consistently to embryonic, progenitor or glial cells. These enrichments were due to sets of genes that were specifically expressed in each of these cell types. We also found that many of the diverse gene sets previously associated with schizophrenia (genes involved in synaptic function, those encoding mRNAs that interact with FMRP, antipsychotic targets, etc.) generally implicated the same brain cell types. Our results suggest a parsimonious explanation: the common-variant genetic results for schizophrenia point at a limited set of neurons, and the gene sets point to the same cells. The genetic risk associated with MSNs did not overlap with that of glutamatergic pyramidal cells and interneurons, suggesting that different cell types have biologically distinct roles in schizophrenia.
Collapse
Affiliation(s)
- Nathan G Skene
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- UCL Institute of Neurology, Queen Square, London, UK
| | - Julien Bryois
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | | | - Gerome Breen
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, UK
- National Institute for Health Research Biomedical Research Centre, South London and Maudsley National Health Service Trust, London, UK
| | - James J Crowley
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Héléna A Gaspar
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, UK
- National Institute for Health Research Biomedical Research Centre, South London and Maudsley National Health Service Trust, London, UK
| | | | | | | | - Ana B Muñoz-Manchado
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Michael C O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Michael J Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Antonio F Pardiñas
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Jesper Ryge
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - James T R Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Sten Linnarsson
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Patrick F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.
| | - Jens Hjerling-Leffler
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
43
|
Genetic variation in 117 myelination-related genes in schizophrenia: Replication of association to lipid biosynthesis genes. Sci Rep 2018; 8:6915. [PMID: 29720671 PMCID: PMC5931982 DOI: 10.1038/s41598-018-25280-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 04/10/2018] [Indexed: 01/18/2023] Open
Abstract
Schizophrenia is a serious psychotic disorder with high heritability. Several common genetic variants, rare copy number variants and ultra-rare gene-disrupting mutations have been linked to disease susceptibility, but there is still a large gap between the estimated and explained heritability. Since several studies have indicated brain myelination abnormalities in schizophrenia, we aimed to examine whether variants in myelination-related genes could be associated with risk for schizophrenia. We established a set of 117 myelination genes by database searches and manual curation. We used a combination of GWAS (SCZ_N = 35,476; CTRL_N = 46,839), exome chip (SCZ_N = 269; CTRL_N = 336) and exome sequencing data (SCZ_N = 2,527; CTRL_N = 2,536) from schizophrenia cases and healthy controls to examine common and rare variants. We found that a subset of lipid-related genes was nominally associated with schizophrenia (p = 0.037), but this signal did not survive multiple testing correction (FWER = 0.16) and was mainly driven by the SREBF1 and SREBF2 genes that have already been linked to schizophrenia. Further analysis demonstrated that the lowest nominal p-values were p = 0.0018 for a single common variant (rs8539) and p = 0.012 for burden of rare variants (LRP1 gene), but none of them survived multiple testing correction. Our findings suggest that variation in myelination-related genes is not a major risk factor for schizophrenia.
Collapse
|
44
|
Cao T, Zhen XC. Dysregulation of miRNA and its potential therapeutic application in schizophrenia. CNS Neurosci Ther 2018. [PMID: 29529357 DOI: 10.1111/cns.12840] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although it is generally believed that genetic and developmental factors play critical roles in pathogenesis of schizophrenia, however, the precise etiological mechanism of schizophrenia remains largely unknown. Over past decades, miRNAs have emerged as an essential post-transcriptional regulator in gene expression regulation. The importance of miRNA in brain development and neuroplasticity has been well-established. Abnormal expression and dysfunction of miRNAs are known to involve in the pathophysiology of many neuropsychiatric diseases including schizophrenia. In this review, we summarized the recent findings in the schizophrenia-associated dysregulation of miRNA and functional roles in the development and pathogenesis of schizophrenia. We also discussed the potential therapeutic implications of miRNA regulation in the illness.
Collapse
Affiliation(s)
- Ting Cao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China.,The Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China
| | - Xue-Chu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China.,The Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China
| |
Collapse
|
45
|
Sakamoto K, Crowley JJ. A comprehensive review of the genetic and biological evidence supports a role for MicroRNA-137 in the etiology of schizophrenia. Am J Med Genet B Neuropsychiatr Genet 2018; 177:242-256. [PMID: 29442441 PMCID: PMC5815396 DOI: 10.1002/ajmg.b.32554] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/05/2017] [Indexed: 01/06/2023]
Abstract
Since it was first associated with schizophrenia (SCZ) in a 2011 genome-wide association study (GWAS), there have been over 100 publications focused on MIR137, the gene encoding microRNA-137. These studies have examined everything from its fundamental role in the development of mice, flies, and fish to the intriguing enrichment of its target gene network in SCZ. Indeed, much of the excitement surrounding MIR137 is due to the distinct possibility that it could regulate a gene network involved in SCZ etiology, a disease which we now recognize is highly polygenic. Here we comprehensively review, to the best of our ability, all published genetic and biological evidence that could support or refute a role for MIR137 in the etiology of SCZ. Through a careful consideration of the literature, we conclude that the data gathered to date continues to strongly support the involvement of MIR137 and its target gene network in neuropsychiatric traits, including SCZ risk. There remain, however, more unanswered than answered questions regarding the mechanisms linking MIR137 genetic variation with behavior. These questions need answers before we can determine whether there are opportunities for diagnostic or therapeutic interventions based on MIR137. We conclude with a number of suggestions for future research on MIR137 that could help to provide answers and hope for a greater understanding of this devastating disorder.
Collapse
Affiliation(s)
- Kensuke Sakamoto
- Department of Genetics, University of North Carolina at Chapel Hill, NC, USA
| | - James J. Crowley
- Department of Genetics, University of North Carolina at Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, NC, USA
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
46
|
Gonzalez DM, Gregory J, Brennand KJ. The Importance of Non-neuronal Cell Types in hiPSC-Based Disease Modeling and Drug Screening. Front Cell Dev Biol 2017; 5:117. [PMID: 29312938 PMCID: PMC5742170 DOI: 10.3389/fcell.2017.00117] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/08/2017] [Indexed: 12/13/2022] Open
Abstract
Current applications of human induced pluripotent stem cell (hiPSC) technologies in patient-specific models of neurodegenerative and neuropsychiatric disorders tend to focus on neuronal phenotypes. Here, we review recent efforts toward advancing hiPSCs toward non-neuronal cell types of the central nervous system (CNS) and highlight their potential use for the development of more complex in vitro models of neurodevelopment and disease. We present evidence from previous works in both rodents and humans of the importance of these cell types (oligodendrocytes, microglia, astrocytes) in neurological disease and highlight new hiPSC-based models that have sought to explore these relationships in vitro. Lastly, we summarize efforts toward conducting high-throughput screening experiments with hiPSCs and propose methods by which new screening platforms could be designed to better capture complex relationships between neural cell populations in health and disease.
Collapse
Affiliation(s)
- David M Gonzalez
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Developmental and Stem Cell Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jill Gregory
- Instructional Technology Group, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kristen J Brennand
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
47
|
Ersland KM, Skrede S, Stansberg C, Steen VM. Subchronic olanzapine exposure leads to increased expression of myelination-related genes in rat fronto-medial cortex. Transl Psychiatry 2017; 7:1262. [PMID: 29187753 PMCID: PMC5802494 DOI: 10.1038/s41398-017-0008-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 07/14/2017] [Indexed: 01/02/2023] Open
Abstract
Schizophrenia is a psychotic disorder with severe and disabling symptoms, such as hallucinations, delusions, blunted affect and social withdrawal. The neuropathology remains elusive, but disturbances in immunity-related processes, neuronal connectivity and myelination have consistently been linked to schizophrenia. Antipsychotic drugs can be efficient in reducing symptoms, acting primarily on the dopamine system, but additional biological targets are likely to exist. Here we have screened for novel mechanisms of action in an animal model, using adult rats exposed to long-acting olanzapine, achieving stable and clinically relevant antipsychotic drug concentrations. By microarray-based examination of global gene expression in the fronto-medial cortex, at the single gene- and gene-set level, we observed downregulation of two neuropeptide-encoding genes, Vgf and Cort (fold change -1,25 and -1,48, respectively) in response to olanzapine exposure. Furthermore, we demonstrated significant upregulation of five out of ~2000 GO predefined gene sets after olanzapine exposure. Strikingly, all were linked to myelination and oligodendrocyte development; "Ensheathment of neurons", "Axon ensheathment", "Myelination", "Myelin sheath" and "Oligodendrocyte development" (FDR-values < 25). Sixteen of the leading edge genes in these gene sets were analysed independently by qPCR, of which 11 genes displayed significant upregulation, including Plp1, Mal, Mag and Cnp (fold change: 1,30, 1,50, 1,30 and 1,15, respectively). Several of the upregulated genes (e.g. MAG, MAL and CNP) have previously been reported as downregulated in post-mortem brain samples from schizophrenia patients. Although caution needs to be taken when extrapolating results from animal studies to humans, the data suggest a role for olanzapine in alleviating myelination-related dysfunction in schizophrenia.
Collapse
Affiliation(s)
- Kari M. Ersland
- 0000 0000 9753 1393grid.412008.fDr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, 5021 Norway ,0000 0004 1936 7443grid.7914.bThe Norwegian Centre for Mental Disorders Research (NORMENT) and the K.G. Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Silje Skrede
- 0000 0000 9753 1393grid.412008.fDr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, 5021 Norway ,0000 0004 1936 7443grid.7914.bThe Norwegian Centre for Mental Disorders Research (NORMENT) and the K.G. Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Christine Stansberg
- 0000 0000 9753 1393grid.412008.fDr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, 5021 Norway ,0000 0004 1936 7443grid.7914.bThe Norwegian Centre for Mental Disorders Research (NORMENT) and the K.G. Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Vidar M. Steen
- 0000 0000 9753 1393grid.412008.fDr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, 5021 Norway ,0000 0004 1936 7443grid.7914.bThe Norwegian Centre for Mental Disorders Research (NORMENT) and the K.G. Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
48
|
Prytkova I, Brennand KJ. Prospects for Modeling Abnormal Neuronal Function in Schizophrenia Using Human Induced Pluripotent Stem Cells. Front Cell Neurosci 2017; 11:360. [PMID: 29217999 PMCID: PMC5703699 DOI: 10.3389/fncel.2017.00360] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/03/2017] [Indexed: 01/21/2023] Open
Abstract
Excitatory dopaminergic neurons, inhibitory GABAergic neurons, microglia, and oligodendrocytes have all been implicated in schizophrenia (SZ) network pathology. Still, SZ has been a difficult disorder to study, not only because of the limitations of animal models in capturing the complexity of the human mind, but also because it is greatly polygenic, with high rates of variability across the population. The advent of patient-derived pluripotent stem cells and induced neural and glial cultures has brought hope for modeling the molecular dysfunction underlying SZ pathology in a patient-specific manner. Here I review the successes of the patient-specific induced cultures in generating different cell types for the study of SZ, with special emphasis on the utility of co-culture techniques, both two- and three-dimensional, for modeling network dysfunction in disease.
Collapse
Affiliation(s)
- Iya Prytkova
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, line>New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kristen J Brennand
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, line>New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
49
|
Gibson EM, Geraghty AC, Monje M. Bad wrap: Myelin and myelin plasticity in health and disease. Dev Neurobiol 2017; 78:123-135. [PMID: 28986960 DOI: 10.1002/dneu.22541] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/31/2017] [Accepted: 10/03/2017] [Indexed: 12/21/2022]
Abstract
Human central nervous system myelin development extends well into the fourth decade of life, and this protracted period underscores the potential for experience to modulate myelination. The concept of myelin plasticity implies adaptability in myelin structure and function in response to experiences during development and beyond. Mounting evidence supports this concept of neuronal activity-regulated changes in myelin-forming cells, including oligodendrocyte precursor cell proliferation, oligodendrogenesis and modulation of myelin microstructure. In healthy individuals, myelin plasticity in associative white matter structures of the brain is implicated in learning and motor function in both rodents and humans. Activity-dependent changes in myelin-forming cells may influence the function of neural networks that depend on the convergence of numerous neural signals on both a temporal and spatial scale. However, dysregulation of myelin plasticity can disadvantageously alter myelin microstructure and result in aberrant circuit function or contribute to pathological cell proliferation. Emerging roles for myelin plasticity in normal neurological function and in disease are discussed. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 123-135, 2018.
Collapse
Affiliation(s)
- Erin M Gibson
- Department of Neurology, Stanford University School of Medicine, Stanford, California, 94305
| | - Anna C Geraghty
- Department of Neurology, Stanford University School of Medicine, Stanford, California, 94305
| | - Michelle Monje
- Department of Neurology, Stanford University School of Medicine, Stanford, California, 94305
| |
Collapse
|
50
|
Palaniyappan L, Das T, Dempster K. The neurobiology of transition to psychosis: clearing the cache. J Psychiatry Neurosci 2017; 42:294-299. [PMID: 28834527 PMCID: PMC5573571 DOI: 10.1503/jpn.170137] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The prepsychotic phase of schizophrenia is not only important for indicated prevention strategies, but also crucial for developing mechanistic models of the emergence of frank psychosis (transition). This commentary highlights the work of Dukart and colleagues, published in this issue of the Journal of Psychiatry and Neurosicence, who sought to identify MRI-based anatomic endophenotypes of psychosis in a well-characterized sample of patients with at-risk mental state (ARMS) and first-episode psychosis (FEP). Conceptual and translational challenges in clarifying the neurobiology of transitional prepsychotic states are discussed. A role of intracortical myelin in the neurobiology of transition is proposed. Transition may not be an outcome of "progressive structural deficits"; it may occur due to inadequate compensatory responses in the predisposed. The need to revise our current "deficit-oriented" models of neurobiology of psychosis in the wake of burgeoning evidence indicating a dynamic process of cortical reorganization is emphasized.
Collapse
Affiliation(s)
- Lena Palaniyappan
- Correspondence to: L. Palaniyappan, Prevention & Early Intervention Program for Psychoses (PEPP), A2-636, LHSC-VH, 800 Commissioners Road, London, Ont., Canada N6A 5W9;
| | | | | |
Collapse
|