1
|
Sun X, Xia M. Schizophrenia and Neurodevelopment: Insights From Connectome Perspective. Schizophr Bull 2025; 51:309-324. [PMID: 39209793 PMCID: PMC11908871 DOI: 10.1093/schbul/sbae148] [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: 09/04/2024]
Abstract
BACKGROUND Schizophrenia is conceptualized as a brain connectome disorder that can emerge as early as late childhood and adolescence. However, the underlying neurodevelopmental basis remains unclear. Recent interest has grown in children and adolescent patients who experience symptom onset during critical brain development periods. Inspired by advanced methodological theories and large patient cohorts, Chinese researchers have made significant original contributions to understanding altered brain connectome development in early-onset schizophrenia (EOS). STUDY DESIGN We conducted a search of PubMed and Web of Science for studies on brain connectomes in schizophrenia and neurodevelopment. In this selective review, we first address the latest theories of brain structural and functional development. Subsequently, we synthesize Chinese findings regarding mechanisms of brain structural and functional abnormalities in EOS. Finally, we highlight several pivotal challenges and issues in this field. STUDY RESULTS Typical neurodevelopment follows a trajectory characterized by gray matter volume pruning, enhanced structural and functional connectivity, improved structural connectome efficiency, and differentiated modules in the functional connectome during late childhood and adolescence. Conversely, EOS deviates with excessive gray matter volume decline, cortical thinning, reduced information processing efficiency in the structural brain network, and dysregulated maturation of the functional brain network. Additionally, common functional connectome disruptions of default mode regions were found in early- and adult-onset patients. CONCLUSIONS Chinese research on brain connectomes of EOS provides crucial evidence for understanding pathological mechanisms. Further studies, utilizing standardized analyses based on large-sample multicenter datasets, have the potential to offer objective markers for early intervention and disease treatment.
Collapse
Affiliation(s)
- Xiaoyi Sun
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- School of Systems Science, Beijing Normal University, Beijing, China
| | - Mingrui Xia
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| |
Collapse
|
2
|
D'Amico A, Sung H, Arbona-Lampaya A, Freifeld A, Hosey K, Garcia J, Lacbawan L, Besançon E, Kassem L, Akula N, Knowles EEM, Dickinson D, McMahon FJ. Independent inheritance of cognition and bipolar disorder in a family sample. Am J Med Genet B Neuropsychiatr Genet 2025; 198:e33001. [PMID: 39011872 DOI: 10.1002/ajmg.b.33001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/28/2024] [Accepted: 06/17/2024] [Indexed: 07/17/2024]
Abstract
Cognitive deficits in people with bipolar disorder (BD) may be the result of the illness or its treatment, but they could also reflect genetic risk factors shared between BD and cognition. We investigated this question using empirical genetic relationships within a sample of patients with BD and their unaffected relatives. Participants with bipolar I, II, or schizoaffective disorder ("narrow" BD, n = 69), related mood disorders ("broad" BD, n = 135), and their clinically unaffected relatives (n = 227) completed five cognitive tests. General cognitive function (g) was quantified via principal components analysis (PCA). Heritability and genetic correlations were estimated with SOLAR-Eclipse. Participants with "narrow" or "broad" diagnoses showed deficits in g, although affect recognition was unimpaired. Cognitive performance was significantly heritable (h2 = 0.322 for g, p < 0.005). Coheritability between psychopathology and g was small (0.0184 for narrow and 0.0327 for broad) and healthy relatives of those with BD were cognitively unimpaired. In this family sample, cognitive deficits were present in participants with BD but were not explained by substantial overlaps in genetic determinants of mood and cognition. These findings support the view that cognitive deficits in BD are largely the result of the illness or its treatment.
Collapse
Affiliation(s)
- Alexander D'Amico
- Intramural Research Program, National Institute of Mental Health, NIH, DHHS, Bethesda, Maryland, USA
| | - Heejong Sung
- Intramural Research Program, National Institute of Mental Health, NIH, DHHS, Bethesda, Maryland, USA
| | - Alejandro Arbona-Lampaya
- Intramural Research Program, National Institute of Mental Health, NIH, DHHS, Bethesda, Maryland, USA
| | - Ally Freifeld
- Intramural Research Program, National Institute of Mental Health, NIH, DHHS, Bethesda, Maryland, USA
| | - Katie Hosey
- Intramural Research Program, National Institute of Mental Health, NIH, DHHS, Bethesda, Maryland, USA
| | - Joshua Garcia
- Intramural Research Program, National Institute of Mental Health, NIH, DHHS, Bethesda, Maryland, USA
| | - Ley Lacbawan
- Intramural Research Program, National Institute of Mental Health, NIH, DHHS, Bethesda, Maryland, USA
| | - Emily Besançon
- Intramural Research Program, National Institute of Mental Health, NIH, DHHS, Bethesda, Maryland, USA
| | - Layla Kassem
- Intramural Research Program, National Institute of Mental Health, NIH, DHHS, Bethesda, Maryland, USA
| | - Nirmala Akula
- Intramural Research Program, National Institute of Mental Health, NIH, DHHS, Bethesda, Maryland, USA
| | | | - Dwight Dickinson
- Intramural Research Program, National Institute of Mental Health, NIH, DHHS, Bethesda, Maryland, USA
| | - Francis J McMahon
- Intramural Research Program, National Institute of Mental Health, NIH, DHHS, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Clocchiatti-Tuozzo S, Rivier CA, Misra S, Zelano J, Mazumder R, Sansing LH, de Havenon A, Hirsch LJ, Liebeskind DS, Gilmore EJ, Sheth KN, Kim JA, Worrall BB, Falcone GJ, Mishra NK. Polygenic Risk of Epilepsy and Poststroke Epilepsy. Stroke 2024; 55:2835-2843. [PMID: 39502073 PMCID: PMC11653790 DOI: 10.1161/strokeaha.124.047459] [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: 04/11/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Epilepsy is highly heritable, with numerous known genetic risk loci. However, the genetic predisposition's role in poststroke epilepsy (PSE) remains understudied. This study assesses whether a higher genetic predisposition to epilepsy raises poststroke survivor's risk of PSE. METHODS We conducted a case-control genetic association study nested within the UK Biobank, a large UK-based prospective cohort. Our exposures of interest were 2 distinct polygenic risk scores-generalized and focal epilepsy-modeled as deciles and constructed using genetic variants identified in the latest International League Against Epilepsy genome-wide association study meta-analysis. We aimed to evaluate the association between these polygenic risk scores and their corresponding subtype of PSE-generalized and focal. In sensitivity analyses, we evaluated participants of European ancestry separately and considered focal and generalized epilepsy outcomes in participants without a history of stroke. In secondary analyses, we evaluated the polygenic risk of PSE by stroke subtype (ischemic, hemorrhagic, or any stroke). Multivariable logistic regression models were fitted, adjusting for age, sex, genetic ancestry, and the first 5 principal genetic components. RESULTS Among 17 549 UK Biobank stroke survivors with available genetic information (mean age, 61; 43% female), 185 (1%) developed generalized PSE, while 124 (0.7%) developed focal PSE. Multivariable logistic regression results showed that, when compared against the lowest decile, participants within the highest PRS decile for generalized PSE had 5-fold higher odds of developing generalized PSE (OR, 5.05 [95% CI, 2.37-12.5]; P trend<0.001). Similarly, when compared against the lowest decile, participants within the highest polygenic risk score decile for focal PSE had 3-fold higher odds of developing focal PSE (OR, 3.20; [5% CI, 1.25-9.82]; P trend=0.024). Sensitivity analyses among participants of European ancestry yielded similar results. CONCLUSIONS Our findings suggest that, like other forms of epilepsy, genetic predisposition plays an essential role in PSE. These results underscore the need for future studies to elucidate the mechanisms underlying PSE development and to identify novel therapeutic avenues.
Collapse
Affiliation(s)
- Santiago Clocchiatti-Tuozzo
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Department of Internal Medicine, Geriatrics, Yale School of Medicine, New Haven, CT, USA
| | - Cyprien A. Rivier
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Shubham Misra
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Johan Zelano
- Department of Clinical Neuroscience, University of Gothenburg, Göteborg, Sweden
| | - Rajarshi Mazumder
- Department of Neurology, The University of California, Los Angeles, USA
| | - Lauren H Sansing
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Adam de Havenon
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | | | | | - Emily J. Gilmore
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Kevin N. Sheth
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Jennifer A. Kim
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Bradford B. Worrall
- Departments of Neurology and Public Health Sciences, University of Virginia, Charlottesville, USA
| | - Guido J Falcone
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Nishant K. Mishra
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Department of Neurology, West Haven VA Medical Center, West Haven, USA
| |
Collapse
|
4
|
Poletti M, Raballo A. Schizophrenia spectrum disorders in children and adolescents: Clinical, phenomenological, diagnostic, and prognostic features across subtypes. Schizophr Res 2024; 274:189-198. [PMID: 39341098 DOI: 10.1016/j.schres.2024.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024]
Abstract
Schizophrenia spectrum disorders (SSD) typically have a diagnostically recognizable onset in young adulthood, yet it is not unusual that help-seeking due to initial SSD-related clinical manifestations emerge in earlier developmental phases, such as childhood and adolescence. Varieties of SSD manifestations in children and adolescents can be distinguished according to variations in clinical expressivity, severity and timing (i.e. developmental stage). Some individuals may reach the full clinical threshold for a diagnosis of schizophrenia according to the same descriptive diagnostic criteria used for adults, and in this case, it's possible to distinguish a pre-pubertal onset in childhood (aka Very Early Onset Schizophrenia, VEOS) and a post-pubertal onset in adolescence (aka Early Onset Schizophrenia, EOS). Other individuals may not reach such clinically overt diagnostic threshold but nonetheless present Childhood Schizotypal Disorder (CSD) or a Clinical High-Risk for Psychosis (CHRP). While EOS is clinically more similar to the canonical adult-onset presentation, the other 3 subgroups (i.e. VEOS, CSD, CHRP) present more nuances and specific clinical characteristics, which require ad-hoc developmental and phenomenological considerations for appropriate differential diagnosis and prognosis. Therefore, current scoping review intends to saturate such knowledge gap with respect to early SSD-phenotypes.
Collapse
Affiliation(s)
- Michele Poletti
- Department of Mental Health and Pathological Addiction, Child and Adolescent Neuropsychiatry Service, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy.
| | - Andrea Raballo
- Chair of Psychiatry, Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Lugano, Switzerland; Public Health Division, Department of Health and Social Care, Cantonal Socio-Psychiatric Organization, Repubblica e Cantone Ticino, Mendrisio, Switzerland
| |
Collapse
|
5
|
Chen J, Wei Y, Xue K, Gao X, Zhang M, Han S, Wen B, Wu G, Cheng J. Static and temporal dynamic changes of intrinsic brain activity in early-onset and adult-onset schizophrenia: a fMRI study of interaction effects. Front Neurol 2024; 15:1445599. [PMID: 39655163 PMCID: PMC11625647 DOI: 10.3389/fneur.2024.1445599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 10/28/2024] [Indexed: 12/12/2024] Open
Abstract
Background Schizophrenia is characterized by altered static and dynamic spontaneous brain activity. However, the conclusions regarding this are inconsistent. Evidence has revealed that this inconsistency could be due to mixed effects of age of onset. Methods We enrolled 66/84 drug-naïve first-episode patients with early-onset/adult-onset schizophrenia (EOS/AOS) and matched normal controls (NCs) (46 adolescents, 73 adults), undergoing resting-state functional magnetic resonance imaging. Two-way ANOVA was used to determine the amplitude of low-frequency fluctuation (ALFF) and dynamic ALFF (dALFF) among the four groups. Result Compared to NCs, EOS had a higher ALFF in inferior frontal gyrus bilateral triangular part (IFG-tri), left opercular part (IFG-oper), left orbital part (IFG-orb), and left middle frontal gyrus (MFG). The AOS had a lower ALFF in left IFG-tri, IFG-oper, and lower dALFF in left IFG-tri. Compared to AOS, EOS had a higher ALFF in the left IFG-orb, and MFG, and higher dALFF in IFG-tri. Adult NCs had higher ALFF and dALFF in the prefrontal cortex (PFC) than adolescent NCs. The main effects of diagnosis were found in the PFC, medial temporal structures, cerebrum, visual and sensorimotor networks, the main effects of age were found in the visual and motor networks of ALFF and PFC of dALFF. Conclusion Our findings unveil the static and dynamic neural activity mechanisms involved in the interaction between disorder and age in schizophrenia. Our results underscore age-related abnormalities in the neural activity of the PFC, shedding new light on the neurobiological mechanisms underlying the development of schizophrenia. This insight may offer valuable perspectives for the specific treatment of EOS in clinical settings.
Collapse
Affiliation(s)
- Jingli Chen
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou, China
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yarui Wei
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou, China
| | - Kangkang Xue
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou, China
| | - Xinyu Gao
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou, China
| | - Mengzhe Zhang
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou, China
| | - Shaoqiang Han
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou, China
| | - Baohong Wen
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou, China
| | - Guangyu Wu
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou, China
| |
Collapse
|
6
|
Garcia MF, Retallick-Townsley K, Pruitt A, Davidson E, Dai Y, Fitzpatrick SE, Sen A, Cohen S, Livoti O, Khan S, Dossou G, Cheung J, Deans PJM, Wang Z, Huckins L, Hoffman E, Brennand K. Dynamic convergence of autism disorder risk genes across neurodevelopment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609190. [PMID: 39229156 PMCID: PMC11370590 DOI: 10.1101/2024.08.23.609190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Over a hundred risk genes underlie risk for autism spectrum disorder (ASD) but the extent to which they converge on shared downstream targets to increase ASD risk is unknown. To test the hypothesis that cellular context impacts the nature of convergence, here we apply a pooled CRISPR approach to target 29 ASD loss-of-function genes in human induced pluripotent stem cell (hiPSC)-derived neural progenitor cells, glutamatergic neurons, and GABAergic neurons. Two distinct approaches (gene-level and network-level analyses) demonstrate that convergence is greatest in mature glutamatergic neurons. Convergent effects are dynamic, varying in strength, composition, and biological role between cell types, increasing with functional similarity of the ASD genes examined, and driven by cell-type-specific gene co-expression patterns. Stratification of ASD genes yield targeted drug predictions capable of reversing gene-specific convergent signatures in human cells and ASD-related behaviors in zebrafish. Altogether, convergent networks downstream of ASD risk genes represent novel points of individualized therapeutic intervention.
Collapse
Affiliation(s)
- Meilin Fernandez Garcia
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
| | - Kayla Retallick-Townsley
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - April Pruitt
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06511
| | - Elizabeth Davidson
- Child Study Center, Yale University School of Medicine, New Haven, CT 06511
| | - Yi Dai
- Child Study Center, Yale University School of Medicine, New Haven, CT 06511
| | - Sarah E Fitzpatrick
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06511
| | - Annabel Sen
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
| | - Sophie Cohen
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
| | - Olivia Livoti
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
| | - Suha Khan
- Child Study Center, Yale University School of Medicine, New Haven, CT 06511
| | - Grace Dossou
- Child Study Center, Yale University School of Medicine, New Haven, CT 06511
| | - Jen Cheung
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
| | - P J Michael Deans
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
| | - Zuoheng Wang
- Child Study Center, Yale University School of Medicine, New Haven, CT 06511
| | - Laura Huckins
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06511
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ellen Hoffman
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06511
- Child Study Center, Yale University School of Medicine, New Haven, CT 06511
| | - Kristen Brennand
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06511
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
7
|
Kong L, Chen Y, Shen Y, Zhang D, Wei C, Lai J, Hu S. Progress and Implications from Genetic Studies of Bipolar Disorder. Neurosci Bull 2024; 40:1160-1172. [PMID: 38206551 PMCID: PMC11306703 DOI: 10.1007/s12264-023-01169-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 10/05/2023] [Indexed: 01/12/2024] Open
Abstract
With the advancements in gene sequencing technologies, including genome-wide association studies, polygenetic risk scores, and high-throughput sequencing, there has been a tremendous advantage in mapping a detailed blueprint for the genetic model of bipolar disorder (BD). To date, intriguing genetic clues have been identified to explain the development of BD, as well as the genetic association that might be applied for the development of susceptibility prediction and pharmacogenetic intervention. Risk genes of BD, such as CACNA1C, ANK3, TRANK1, and CLOCK, have been found to be involved in various pathophysiological processes correlated with BD. Although the specific roles of these genes have yet to be determined, genetic research on BD will help improve the prevention, therapeutics, and prognosis in clinical practice. The latest preclinical and clinical studies, and reviews of the genetics of BD, are analyzed in this review, aiming to summarize the progress in this intriguing field and to provide perspectives for individualized, precise, and effective clinical practice.
Collapse
Affiliation(s)
- Lingzhuo Kong
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yiqing Chen
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuting Shen
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Danhua Zhang
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Chen Wei
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jianbo Lai
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, 310003, China.
- Brain Research Institute of Zhejiang University, Hangzhou, 310003, China.
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, 310003, China.
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and MOE Frontier Science Center for Brain Science and Brain-machine Integration, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Shaohua Hu
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, 310003, China.
- Brain Research Institute of Zhejiang University, Hangzhou, 310003, China.
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, 310003, China.
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and MOE Frontier Science Center for Brain Science and Brain-machine Integration, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
8
|
Fortea A, van Eijndhoven P, Calvet-Mirabent A, Ilzarbe D, Batalla A, de la Serna E, Puig O, Castro-Fornieles J, Dolz M, Tor J, Parrilla S, Via E, Stephan-Otto C, Baeza I, Sugranyes G. Age-related change in cortical thickness in adolescents at clinical high risk for psychosis: a longitudinal study. Eur Child Adolesc Psychiatry 2024; 33:1837-1846. [PMID: 37644217 DOI: 10.1007/s00787-023-02278-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/03/2023] [Indexed: 08/31/2023]
Abstract
Progression to psychosis has been associated with increased cortical thinning in the frontal, temporal and parietal lobes in individuals at clinical high risk for the disorder (CHR-P). The timing and spatial extent of these changes are thought to be influenced by age. However, most evidence so far stems from adult samples. Longitudinal studies are essential to understanding the neuroanatomical changes associated to transition to psychosis during adolescence, and their relationship with age. We conducted a longitudinal, multisite study including adolescents at CHR-P and healthy controls (HC), aged 10-17 years. Structural images were acquired at baseline and at 18-month follow-up. Images were processed with the longitudinal pipeline in FreeSurfer. We used a longitudinal two-stage model to compute the regional cortical thickness (CT) change, and analyze between-group differences controlling for age, sex and scan, and corrected for multiple comparisons. Linear regression was used to study the effect of age at baseline. A total of 103 individuals (49 CHR-P and 54 HC) were included in the analysis. During follow-up, the 13 CHR-P participants who transitioned to psychosis exhibited greater CT decrease over time in the right parietal cortex compared to those who did not transition to psychosis and to HC. Age at baseline correlated with longitudinal changes in CT, with younger individuals showing greater cortical thinning in this region. The emergence of psychosis during early adolescence may have an impact on typical neuromaturational processes. This study provides new insights on the cortical changes taking place prior to illness onset.
Collapse
Affiliation(s)
- Adriana Fortea
- Psychiatry and Psychology Department, Institute Clinic of Neurosciences, Hospital Clínic of Barcelona, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Fundació Clínic per a la Recerca Biomèdica (FCRB), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Barcelona, Spain
| | - Philip van Eijndhoven
- Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain Cognition and Behavior, Nijmegen, The Netherlands
| | - Angels Calvet-Mirabent
- Institut d'Investigacions Biomèdiques Agustí Pi i Sunyer (IDIBAPS), C/Rosselló 149-153, 08036, Barcelona, Spain
| | - Daniel Ilzarbe
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Barcelona, Spain
- Institut d'Investigacions Biomèdiques Agustí Pi i Sunyer (IDIBAPS), C/Rosselló 149-153, 08036, Barcelona, Spain
- Child and Adolescent Psychiatry and Psychology Department, 2021SGR01319, Institute Clinic of Neurosciences, Hospital Clínic of Barcelona, C/Villarroel 170, 08036, Barcelona, Spain
| | - Albert Batalla
- UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Elena de la Serna
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Barcelona, Spain
- Child and Adolescent Psychiatry and Psychology Department, 2021SGR01319, Institute Clinic of Neurosciences, Hospital Clínic of Barcelona, C/Villarroel 170, 08036, Barcelona, Spain
| | - Olga Puig
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Barcelona, Spain
- Child and Adolescent Psychiatry and Psychology Department, 2021SGR01319, Institute Clinic of Neurosciences, Hospital Clínic of Barcelona, C/Villarroel 170, 08036, Barcelona, Spain
| | - Josefina Castro-Fornieles
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Barcelona, Spain
- Institut d'Investigacions Biomèdiques Agustí Pi i Sunyer (IDIBAPS), C/Rosselló 149-153, 08036, Barcelona, Spain
- Child and Adolescent Psychiatry and Psychology Department, 2021SGR01319, Institute Clinic of Neurosciences, Hospital Clínic of Barcelona, C/Villarroel 170, 08036, Barcelona, Spain
| | - Montserrat Dolz
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Barcelona, Spain
- Child and Adolescent Mental Health Research Group, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Child and Adolescent Psychiatry and Psychology Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Jordina Tor
- Child and Adolescent Mental Health Research Group, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Child and Adolescent Psychiatry and Psychology Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Sara Parrilla
- Child and Adolescent Mental Health Research Group, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Esther Via
- Child and Adolescent Mental Health Research Group, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Child and Adolescent Psychiatry and Psychology Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Christian Stephan-Otto
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Barcelona, Spain
- Child and Adolescent Mental Health Research Group, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Pediatric Computational Imaging Group (PeCIC), Hospital Sant Joan de Déu, Barcelona, Spain
| | - Inmaculada Baeza
- Department of Medicine, University of Barcelona, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Barcelona, Spain.
- Institut d'Investigacions Biomèdiques Agustí Pi i Sunyer (IDIBAPS), C/Rosselló 149-153, 08036, Barcelona, Spain.
- Child and Adolescent Psychiatry and Psychology Department, 2021SGR01319, Institute Clinic of Neurosciences, Hospital Clínic of Barcelona, C/Villarroel 170, 08036, Barcelona, Spain.
| | - Gisela Sugranyes
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Barcelona, Spain.
- Institut d'Investigacions Biomèdiques Agustí Pi i Sunyer (IDIBAPS), C/Rosselló 149-153, 08036, Barcelona, Spain.
- Child and Adolescent Psychiatry and Psychology Department, 2021SGR01319, Institute Clinic of Neurosciences, Hospital Clínic of Barcelona, C/Villarroel 170, 08036, Barcelona, Spain.
| |
Collapse
|
9
|
Ma Y, Bendl J, Hartley BJ, Fullard JF, Abdelaal R, Ho SM, Kosoy R, Gochman P, Rapoport J, Hoffman GE, Brennand KJ, Roussos P. Activity-Dependent Transcriptional Program in NGN2+ Neurons Enriched for Genetic Risk for Brain-Related Disorders. Biol Psychiatry 2024; 95:187-198. [PMID: 37454787 PMCID: PMC10787819 DOI: 10.1016/j.biopsych.2023.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 06/07/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Converging evidence from large-scale genetic and postmortem studies highlights the role of aberrant neurotransmission and genetic regulation in brain-related disorders. However, identifying neuronal activity-regulated transcriptional programs in the human brain and understanding how changes contribute to disease remain challenging. METHODS To better understand how the activity-dependent regulome contributes to risk for brain-related disorders, we profiled the transcriptomic and epigenomic changes following neuronal depolarization in human induced pluripotent stem cell-derived glutamatergic neurons (NGN2) from 6 patients with schizophrenia and 5 control participants. RESULTS Multiomic data integration associated global patterns of chromatin accessibility with gene expression and identified enhancer-promoter interactions in glutamatergic neurons. Within 1 hour of potassium chloride-induced depolarization, independent of diagnosis, glutamatergic neurons displayed substantial activity-dependent changes in the expression of genes regulating synaptic function. Depolarization-induced changes in the regulome revealed significant heritability enrichment for schizophrenia and Parkinson's disease, adding to mounting evidence that sequence variation within activation-dependent regulatory elements contributes to the genetic risk for brain-related disorders. Gene coexpression network analysis elucidated interactions among activity-dependent and disease-associated genes and pointed to a key driver (NAV3) that interacted with multiple genes involved in axon guidance. CONCLUSIONS Overall, we demonstrated that deciphering the activity-dependent regulome in glutamatergic neurons reveals novel targets for advanced diagnosis and therapy.
Collapse
Affiliation(s)
- Yixuan Ma
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, New York; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jaroslav Bendl
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Brigham J Hartley
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Black Family Stem Cell Institute, New York, New York
| | - John F Fullard
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Rawan Abdelaal
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Black Family Stem Cell Institute, New York, New York
| | - Seok-Man Ho
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Black Family Stem Cell Institute, New York, New York
| | - Roman Kosoy
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Peter Gochman
- Childhood Psychiatry Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Judith Rapoport
- Childhood Psychiatry Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Gabriel E Hoffman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, New York; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kristen J Brennand
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Black Family Stem Cell Institute, New York, New York.
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Dementia Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, New York; Mental Illness Research Education and Clinical Center, James J. Peters VA Medical Center, Bronx, New York.
| |
Collapse
|
10
|
Zhong Y, Tubbs JD, Leung PBM, Zhan N, Hui TCK, Ho KKY, Hung KSY, Cheung EFC, So HC, Lui SSY, Sham PC. Early-onset schizophrenia is associated with immune-related rare variants in a Chinese sample. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.21.23298115. [PMID: 38045317 PMCID: PMC10690336 DOI: 10.1101/2023.11.21.23298115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Background Rare variants are likely to contribute to schizophrenia (SCZ), given the large discrepancy between the heritability estimated from twin and GWAS studies. Furthermore, the nature of the rare-variant contribution to SCZ may vary with the "age-at-onset" (AAO), since early-onset has been suggested as being indicative of neurodevelopment deviance. Objective To examine the association of rare deleterious coding variants in early- and adult-onset SCZ in a Chinese sample. Method Exome sequencing was performed on DNA from 197 patients with SCZ spectrum disorder and 82 healthy controls (HC) of Chinese ancestry recruited in Hong Kong. We also gathered AAO information in the majority of SCZ samples. Patients were classified into early-onset (EOS, AAO<18) and adult-onset (AOS, AAO>18). We collapsed the rare variants to improve statistical power and examined the overall association of rare variants in SCZ versus HC, EOS versus HC, and AOS versus HC at the gene and gene-set levels by Sequence Kernel Association Test. The quantitative rare-variant association test of AAO was also conducted. We focused on variants which were predicted to have a medium or high impact on the protein-encoding process as defined by Ensembl. We applied a 100000-time permutation test to obtain empirical p-values, with significance threshold set at p < 1e -3 to control family-wise error rates. Moreover, we compared the burden of targeted rare variants in significant risk genes and gene sets in cases and controls. Results Based on several binary-trait association tests (i.e., SCZ vs HC, EOS vs HC and AOS vs HC), we identified 7 candidate risk genes and 20 gene ontology biological processes (GOBP) terms, which exhibited higher burdens in SCZ than in controls. Based on quantitative rare-variant association tests, we found that alterations in 5 candidate risk genes and 7 GOBP pathways were significantly correlated with AAO. Based on biological and functional profiles of the candidate risk genes and gene sets, our findings suggested that, in addition to the involvement of perturbations in neural systems in SCZ in general, altered immune responses may be specifically implicated in EOS. Conclusion Disrupted immune responses may exacerbate abnormal perturbations during neurodevelopment and trigger the early onset of SCZ. We provided evidence of rare variants increasing SCZ risk in the Chinese population.
Collapse
|
11
|
Patterson EM, Lim J, Fuchs P, Smith JR, Moussa-Tooks A, Ward HB. Use of First-Generation Antipsychotics in an Adolescent Male with Catatonic Schizophrenia. Harv Rev Psychiatry 2023; 31:267-273. [PMID: 37823777 PMCID: PMC11530942 DOI: 10.1097/hrp.0000000000000381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Affiliation(s)
- Emmy Masur Patterson
- From Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN (Drs. Patterson, Moussa-Tooks, and Ward); Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN (Dr. Lim); Sheppard Pratt Hospital, Baltimore, MD (Dr. Fuchs); Division of Child and Adolescent Psychiatry, Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN (Dr. Smith); Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN (Dr. Smith)
| | | | | | | | | | | |
Collapse
|
12
|
Rivier CA, Clocchiatti-Tuozzo S, Misra S, Zelano J, Mazumder R, Sansing LH, de Havenon A, Hirsch LJ, Liebeskind DS, Gilmore EJ, Sheth KN, Kim JA, Worrall BB, Falcone G, Mishra NK. Polygenic Risk of Epilepsy and Post-Stroke Epilepsy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.18.23295739. [PMID: 37790357 PMCID: PMC10543238 DOI: 10.1101/2023.09.18.23295739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Background and Aims Epilepsy is highly heritable, with numerous known genetic risk loci. However, the genetic predisposition's role in post-acute brain injury epilepsy remains understudied. This study assesses whether a higher genetic predisposition to epilepsy raises post-stroke or Transient Ischemic Attack (TIA) survivor's risk of Post-Stroke Epilepsy (PSE). Methods We conducted a three-stage genetic analysis. First, we identified independent epilepsy-associated ( p <5x10 -8 ) genetic variants from public data. Second, we estimated PSE-specific variant weights in stroke/TIA survivors from the UK Biobank. Third, we tested for an association between a polygenic risk score (PRS) and PSE risk in stroke/TIA survivors from the All of Us Research Program. Primary analysis included all ancestries, while a secondary analysis was restricted to European ancestry only. A sensitivity analysis excluded TIA survivors. Association testing was conducted via multivariable logistic regression, adjusting for age, sex, and genetic ancestry. Results Among 19,708 UK Biobank participants with stroke/TIA, 805 (4.1%) developed PSE. Likewise, among 12,251 All of Us participants with stroke/TIA, 394 (3.2%) developed PSE. After establishing PSE-specific weights for 39 epilepsy-linked genetic variants in the UK Biobank, the resultant PRS was associated with elevated odds of PSE development in All of Us (OR:1.16[1.02-1.32]). A similar result was obtained when restricting to participants of European ancestry (OR:1.23[1.02-1.49]) and when excluding participants with a TIA history (OR:1.18[1.02-1.38]). Conclusions Our findings suggest that akin to other forms of epilepsy, genetic predisposition plays an essential role in PSE. Because the PSE data were sparse, our results should be interpreted cautiously.
Collapse
|
13
|
Gupta N, Gupta M, Esang M. Lost in Translation: Challenges in the Diagnosis and Treatment of Early-Onset Schizophrenia. Cureus 2023; 15:e39488. [PMID: 37362509 PMCID: PMC10290525 DOI: 10.7759/cureus.39488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
Early-onset schizophrenia (EOS) is a heterogeneous condition that has a serious, insidious clinical course and poor long-term mental health outcomes. The clinical presentations are highly complex due to the overlapping symptomatology with other illnesses, which contributes to a delay in the diagnosis. The objective of the review is to study if an earlier age of onset (AAO) of EOS has poor clinical outcomes, the diagnostic challenges of EOS, and effective treatment strategies. The review provides a comprehensive literature search of 5966 articles and summarizes 126 selected for empirical evidence to methodically consider challenges in diagnosing and treating EOS for practicing clinicians. The risk factors of EOS are unique but have been shared with many other neuropsychiatric illnesses. Most of the risk factors, including genetics and obstetric complications, are nonmodifiable. The role of early diagnosis in reducing the duration of untreated psychosis (DUP) remains critical to reducing overall morbidity. Many specific issues contribute to the risk and clinical outcomes. Therefore, issues around diagnostic ambiguity, treatment resistance, nonadherence, and rehospitalizations further extend the DUP. There is hesitancy to initiate clozapine early, even though the empirical evidence strongly supports its use. There is a growing body of research that suggests the use of long-acting injectables to address nonadherence, and these measures are largely underutilized in acute settings. The clinical presentations of EOS are complex. In addition to the presence of specific risk factors, patients with an early onset of illness are also at a higher risk for treatment resistance. While there is a need to develop tools for early diagnosis, established evidence-based measures to address nonadherence, psychoeducation, and resistance must be incorporated into the treatment planning.
Collapse
Affiliation(s)
- Nihit Gupta
- Psychiatry, Dayton Children's Hospital, Dayton, USA
| | - Mayank Gupta
- Psychiatry and Behavioral Sciences, Southwood Psychiatric Hospital, Pittsburgh, USA
| | - Michael Esang
- Psychiatry and Behavioral Sciences, Clarion Psychiatric Center, Clarion, USA
| |
Collapse
|
14
|
Chen J, Wei Y, Xue K, Han S, Wang C, Wen B, Cheng J. The interaction between first-episode drug-naïve schizophrenia and age based on gray matter volume and its molecular analysis: a multimodal magnetic resonance imaging study. Psychopharmacology (Berl) 2023; 240:813-826. [PMID: 36719459 DOI: 10.1007/s00213-023-06323-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/19/2023] [Indexed: 02/01/2023]
Abstract
OBJECTIVES Schizophrenia is a neurodevelopmental disorder characterized by progressive and widespread gray matter (GM) atrophy. Studies have shown that normal brain development has an impact on schizophrenia-induced GM alterations. However, the neuropathology and underlying molecular mechanisms of interaction between age and schizophrenia are unclear. METHODS This study enrolled 66/84 first-episode drug-naïve patients with early-onset/adult-onset schizophrenia ((EOS)/(AOS)) and matched normal controls (NC) (46 adolescents/73 adults), undergoing T1-weighted high-resolution magnetic resonance imaging. Gray matter volume (GMV) in four groups was detected using 2-way analyses of variance with diagnosis and age as factors. Then, factors-related volume maps and neurotransmitter maps were spatially correlated using JuSpace to determine the relationship to molecular structure. RESULTS Compared to AOS, EOS and adult NC had larger GMV in right middle frontal gyrus. Compared to adolescent NC, EOS and adult NC had smaller GMV in right lingual gyrus, right fusiform gyrus, and right cerebellum_6. Disease-induced GMV reductions were mainly distributed in frontal, parietal, thalamus, visual, motor cortex, and medial temporal lobe structures. Age-induced GMV alterations were mainly distributed in visual and motor cortex. The changed GMV induced by schizophrenia, age, and their interaction was related to dopaminergic and serotonergic receptors. Age is also related to glutamate receptors, and schizophrenia is also associated with GABAaergic and noradrenergic receptors. CONCLUSIONS Our results revealed the multimodal neural mechanism of interaction between disease and age. We emphasized age-related GM abnormalities of ventral stream of visual perceptual pathways and high-level cognitive brain in EOS, which may be affected by imbalance of excitatory and inhibitory neurotransmitters.
Collapse
Affiliation(s)
- Jingli Chen
- Department of Magnetic Resonance Imaging, Two Seven District, The First Affiliated Hospital of Zhengzhou University, 1St Construction of E Rd, Zhengzhou, 450052, China
- Key Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou, China
- Engineering Technology Research Center for Detection and Application of Brain Function of Henan Province, Zhengzhou, China
- Engineering Research Center of Medical Imaging Intelligent Diagnosis and Treatment of Henan Province, Zhengzhou, China
- Key Laboratory of Magnetic Resonance and Brain Function of Henan Province, Zhengzhou, China
- Key Laboratory of Brain Function and Cognitive Magnetic Resonance Imaging of Zhengzhou, Zhengzhou, China
- Key Laboratory of Imaging Intelligence Research Medicine of Henan Province, Zhengzhou, China
| | - Yarui Wei
- Department of Magnetic Resonance Imaging, Two Seven District, The First Affiliated Hospital of Zhengzhou University, 1St Construction of E Rd, Zhengzhou, 450052, China
- Key Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou, China
- Engineering Technology Research Center for Detection and Application of Brain Function of Henan Province, Zhengzhou, China
- Engineering Research Center of Medical Imaging Intelligent Diagnosis and Treatment of Henan Province, Zhengzhou, China
- Key Laboratory of Magnetic Resonance and Brain Function of Henan Province, Zhengzhou, China
- Key Laboratory of Brain Function and Cognitive Magnetic Resonance Imaging of Zhengzhou, Zhengzhou, China
- Key Laboratory of Imaging Intelligence Research Medicine of Henan Province, Zhengzhou, China
| | - Kangkang Xue
- Department of Magnetic Resonance Imaging, Two Seven District, The First Affiliated Hospital of Zhengzhou University, 1St Construction of E Rd, Zhengzhou, 450052, China
- Key Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou, China
- Engineering Technology Research Center for Detection and Application of Brain Function of Henan Province, Zhengzhou, China
- Engineering Research Center of Medical Imaging Intelligent Diagnosis and Treatment of Henan Province, Zhengzhou, China
- Key Laboratory of Magnetic Resonance and Brain Function of Henan Province, Zhengzhou, China
- Key Laboratory of Brain Function and Cognitive Magnetic Resonance Imaging of Zhengzhou, Zhengzhou, China
- Key Laboratory of Imaging Intelligence Research Medicine of Henan Province, Zhengzhou, China
| | - Shaoqiang Han
- Department of Magnetic Resonance Imaging, Two Seven District, The First Affiliated Hospital of Zhengzhou University, 1St Construction of E Rd, Zhengzhou, 450052, China
- Key Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou, China
- Engineering Technology Research Center for Detection and Application of Brain Function of Henan Province, Zhengzhou, China
- Engineering Research Center of Medical Imaging Intelligent Diagnosis and Treatment of Henan Province, Zhengzhou, China
- Key Laboratory of Magnetic Resonance and Brain Function of Henan Province, Zhengzhou, China
- Key Laboratory of Brain Function and Cognitive Magnetic Resonance Imaging of Zhengzhou, Zhengzhou, China
- Key Laboratory of Imaging Intelligence Research Medicine of Henan Province, Zhengzhou, China
| | - Caihong Wang
- Department of Magnetic Resonance Imaging, Two Seven District, The First Affiliated Hospital of Zhengzhou University, 1St Construction of E Rd, Zhengzhou, 450052, China
- Key Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou, China
- Engineering Technology Research Center for Detection and Application of Brain Function of Henan Province, Zhengzhou, China
- Engineering Research Center of Medical Imaging Intelligent Diagnosis and Treatment of Henan Province, Zhengzhou, China
- Key Laboratory of Magnetic Resonance and Brain Function of Henan Province, Zhengzhou, China
- Key Laboratory of Brain Function and Cognitive Magnetic Resonance Imaging of Zhengzhou, Zhengzhou, China
- Key Laboratory of Imaging Intelligence Research Medicine of Henan Province, Zhengzhou, China
| | - Baohong Wen
- Department of Magnetic Resonance Imaging, Two Seven District, The First Affiliated Hospital of Zhengzhou University, 1St Construction of E Rd, Zhengzhou, 450052, China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, Two Seven District, The First Affiliated Hospital of Zhengzhou University, 1St Construction of E Rd, Zhengzhou, 450052, China.
- Key Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou, China.
- Engineering Technology Research Center for Detection and Application of Brain Function of Henan Province, Zhengzhou, China.
- Engineering Research Center of Medical Imaging Intelligent Diagnosis and Treatment of Henan Province, Zhengzhou, China.
- Key Laboratory of Magnetic Resonance and Brain Function of Henan Province, Zhengzhou, China.
- Key Laboratory of Brain Function and Cognitive Magnetic Resonance Imaging of Zhengzhou, Zhengzhou, China.
- Key Laboratory of Imaging Intelligence Research Medicine of Henan Province, Zhengzhou, China.
| |
Collapse
|
15
|
Alkelai A, Greenbaum L, Shohat S, Povysil G, Malakar A, Ren Z, Motelow JE, Schechter T, Draiman B, Chitrit-Raveh E, Hughes D, Jobanputra V, Shifman S, Goldstein DB, Kohn Y. Genetic insights into childhood-onset schizophrenia: The yield of clinical exome sequencing. Schizophr Res 2023; 252:138-145. [PMID: 36645932 DOI: 10.1016/j.schres.2022.12.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 12/19/2022] [Accepted: 12/26/2022] [Indexed: 01/15/2023]
Abstract
Childhood-onset schizophrenia (COS) is a rare form of schizophrenia with an onset prior to 13 years of age. Although genetic factors play a role in COS etiology, only a few causal variants have been reported to date. This study presents a diagnostic exome sequencing (ES) in 37 Israeli Jewish families with a proband diagnosed with COS. By implementing a trio/duo ES approach and applying a well-established diagnostic pipeline, we detected clinically significant variants in 7 probands (19 %). These single nucleotide variants and indels were mostly inherited. The implicated genes were ANKRD11, GRIA2, CHD2, CLCN3, CLTC, IGF1R and MICU1. In a secondary analysis that compared COS patients to 4721 healthy controls, we observed that patients had a significant enrichment of rare loss of function (LoF) variants in LoF intolerant genes associated with developmental diseases. Taken together, ES could be considered as a valuable tool in the genetic workup for COS patients.
Collapse
Affiliation(s)
- Anna Alkelai
- Institute for Genomic Medicine, Columbia University Medical Center, New York, USA; Regeneron Genetics Center, Tarrytown, NY, USA.
| | - Lior Greenbaum
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Tel Hashomer, Israel; The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel; Sackler Faculty of Medicine, Tal Aviv University, Tel Aviv, Israel
| | - Shahar Shohat
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gundula Povysil
- Institute for Genomic Medicine, Columbia University Medical Center, New York, USA
| | - Ayan Malakar
- Institute for Genomic Medicine, Columbia University Medical Center, New York, USA
| | - Zhong Ren
- Institute for Genomic Medicine, Columbia University Medical Center, New York, USA
| | - Joshua E Motelow
- Institute for Genomic Medicine, Columbia University Medical Center, New York, USA; Department of Pediatrics, Division of Critical Care and Hospital Medicine, Columbia University Irving Medical Center, New York-Presbyterian Morgan Stanley Children's Hospital of New York, New York, NY, USA
| | - Tanya Schechter
- Department of Child and Adolescent Psychiatry, Jerusalem Mental Health Center, Eitanim Psychiatric Hospital, Israel
| | - Benjamin Draiman
- Department of Child and Adolescent Psychiatry, Jerusalem Mental Health Center, Eitanim Psychiatric Hospital, Israel
| | - Eti Chitrit-Raveh
- Department of Child and Adolescent Psychiatry, Jerusalem Mental Health Center, Eitanim Psychiatric Hospital, Israel
| | - Daniel Hughes
- Institute for Genomic Medicine, Columbia University Medical Center, New York, USA
| | - Vaidehi Jobanputra
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA; New York Genome Center, New York, NY, USA
| | - Sagiv Shifman
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - David B Goldstein
- Institute for Genomic Medicine, Columbia University Medical Center, New York, USA
| | - Yoav Kohn
- Department of Child and Adolescent Psychiatry, Jerusalem Mental Health Center, Eitanim Psychiatric Hospital, Israel; Hadassah-Hebrew University School of Medicine, Jerusalem, Israel
| |
Collapse
|
16
|
Impact on the Risk and Severity of Childhood Onset Schizophrenia of Schizophrenia Risk Genetic Variants at the DRD2 and ZNF804A Loci. Child Psychiatry Hum Dev 2023; 54:241-247. [PMID: 34524581 DOI: 10.1007/s10578-021-01245-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 01/25/2023]
Abstract
The study explored whether schizophrenia risk alleles of the DRD2 rs2514218 and ZNF804A rs1344706 polymorphisms also influenced the risk and severity of childhood-onset schizophrenia (COS) and differentiated it from autism spectrum disorders (ASD). We compared 75 children with COS to 75 children with ASD, 150 patients with adult-onset schizophrenia and 150 healthy individuals. Frequency of the DRD2 T-allele, assumed to be protective against schizophrenia overall, was higher in COS compared to adult-onset schizophrenia and healthy controls. The risk allele A of ZNF804A was associated with greater severity of negative symptoms in COS. The latter result is consistent with the involvement of ZNF804A in the development of severe forms of schizophrenia. The findings regarding DRD2 suggest that the same genetic variants may play different roles in schizophrenia with childhood and adult onset. This warrants further research, since D2 receptor blockade is a general pharmacodynamic property of antipsychotics.
Collapse
|
17
|
Fortea A, Pinzón-Espinosa J, Ilzarbe D, Espinosa L, Lázaro L, Calvo RM, Castro-Fornieles J, de la Serna E, Bargalló N, Baeza I, Sugranyes G. Radiological findings in brain MRI scans in youth with early-onset psychosis: A controlled study. J Psychiatr Res 2022; 156:151-158. [PMID: 36252344 DOI: 10.1016/j.jpsychires.2022.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/08/2022] [Accepted: 10/03/2022] [Indexed: 12/12/2022]
Abstract
There is a lack of consensus on whether routine brain magnetic resonance imaging (MRI) should be recommended as part of the initial assessment in patients with psychosis. No study so far has qualitatively assessed brain MRI in patients with early-onset psychosis (EOP), in whom neurodevelopmental factors may play a stronger role. We aimed to determine the prevalence of brain MRI findings in patients with EOP compared to healthy controls, and assess whether these findings were clinically relevant. Retrospective clinical chart review of all patients with EOP in whom a brain MRI scan was acquired during admission to an inpatient child and adolescent psychiatry unit during January 2013-December 2017, compared to age and biologically assigned gender matched healthy controls. Between group analyses tested differences in rates of qualitatively abnormal MRI scans and changes in clinical management as a result of radiological findings. A total of 256 individuals were included (128 patients with EOP and 128 healthy controls). Patients with EOP presented with a significantly higher rate of abnormal MRI scans relative to healthy controls (21.9% vs 11.7%, p = .030; OR = 2.11, [95% CI:1.06-4.17]). Radiological findings in the EOP group triggered clinical referral for further evaluation or management more often than in the healthy control group (7.0% vs 1.6%, p = .030; OR = 4.76, [95% CI:1.01-22.50]). MRI scans in youth with EOP may be characterized by an increased number of radiological abnormalities than in controls. The rates of MRI findings requiring clinical referral suggests that routine MRI acquisition may need to be considered in patients with EOP.
Collapse
Affiliation(s)
- Adriana Fortea
- Department of Child and Adolescent Psychiatry and Psychology, 2017SGR-881 Institute of Neuroscience, Hospital Clínic of Barcelona, 170 Villarroel St., Barcelona, 08036, Spain; Department of Medicine, School of Medicine, University of Barcelona, 143 Casanova St Barcelona 08036, Spain; Fundació Clínic per a La Recerca Biomèdica (FCRB), 153 Rosselló St., Barcelona, 08036, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 153 Rosselló St., Barcelona, 08036, Spain; Centro de Investigación Biomédica en Red Salud Mental (CIBERSAM), 3-5, Monforte de Lemos St., Madrid, 28029, Spain
| | - Justo Pinzón-Espinosa
- Department of Medicine, School of Medicine, University of Barcelona, 143 Casanova St Barcelona 08036, Spain; Department of Psychiatry and Psychology, Institute of Neuroscience, Hospital Clínic of Barcelona, 170 Villarroel St., Barcelona, 08036, Spain; Department of Mental Health, Parc Tauli University Hospital, Sabadell, 1 Parc Taulí, Sabadell, 08208, Barcelona, Spain; Department of Clinical Psychiatry, School of Medicine, University of Panama, Ave. Octavio Méndez Pereira, Panamá, XFJ8+V2Q, Panama
| | - Daniel Ilzarbe
- Department of Child and Adolescent Psychiatry and Psychology, 2017SGR-881 Institute of Neuroscience, Hospital Clínic of Barcelona, 170 Villarroel St., Barcelona, 08036, Spain; Department of Medicine, School of Medicine, University of Barcelona, 143 Casanova St Barcelona 08036, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 153 Rosselló St., Barcelona, 08036, Spain; Centro de Investigación Biomédica en Red Salud Mental (CIBERSAM), 3-5, Monforte de Lemos St., Madrid, 28029, Spain
| | - Laura Espinosa
- Department of Child and Adolescent Psychiatry and Psychology, 2017SGR-881 Institute of Neuroscience, Hospital Clínic of Barcelona, 170 Villarroel St., Barcelona, 08036, Spain; Department of Child and Adolescent Psychiatry, Hospital Sant Joan de Deu, Fundació Althaia, 1-3 Dr. Joan Soler St., Manresa, 08243, Barcelona, Spain
| | - Luisa Lázaro
- Department of Child and Adolescent Psychiatry and Psychology, 2017SGR-881 Institute of Neuroscience, Hospital Clínic of Barcelona, 170 Villarroel St., Barcelona, 08036, Spain; Department of Medicine, School of Medicine, University of Barcelona, 143 Casanova St Barcelona 08036, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 153 Rosselló St., Barcelona, 08036, Spain; Centro de Investigación Biomédica en Red Salud Mental (CIBERSAM), 3-5, Monforte de Lemos St., Madrid, 28029, Spain
| | - Rosa M Calvo
- Department of Child and Adolescent Psychiatry and Psychology, 2017SGR-881 Institute of Neuroscience, Hospital Clínic of Barcelona, 170 Villarroel St., Barcelona, 08036, Spain; Department of Medicine, School of Medicine, University of Barcelona, 143 Casanova St Barcelona 08036, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 153 Rosselló St., Barcelona, 08036, Spain; Centro de Investigación Biomédica en Red Salud Mental (CIBERSAM), 3-5, Monforte de Lemos St., Madrid, 28029, Spain
| | - Josefina Castro-Fornieles
- Department of Child and Adolescent Psychiatry and Psychology, 2017SGR-881 Institute of Neuroscience, Hospital Clínic of Barcelona, 170 Villarroel St., Barcelona, 08036, Spain; Department of Medicine, School of Medicine, University of Barcelona, 143 Casanova St Barcelona 08036, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 153 Rosselló St., Barcelona, 08036, Spain; Centro de Investigación Biomédica en Red Salud Mental (CIBERSAM), 3-5, Monforte de Lemos St., Madrid, 28029, Spain
| | - Elena de la Serna
- Department of Child and Adolescent Psychiatry and Psychology, 2017SGR-881 Institute of Neuroscience, Hospital Clínic of Barcelona, 170 Villarroel St., Barcelona, 08036, Spain; Centro de Investigación Biomédica en Red Salud Mental (CIBERSAM), 3-5, Monforte de Lemos St., Madrid, 28029, Spain
| | - Nuria Bargalló
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 153 Rosselló St., Barcelona, 08036, Spain; Department of Radiology, Imaging Diagnosis Center, Hospital Clínic of Barcelona, 170 Villarroel St., Barcelona, 08036, Spain
| | - Inmaculada Baeza
- Department of Child and Adolescent Psychiatry and Psychology, 2017SGR-881 Institute of Neuroscience, Hospital Clínic of Barcelona, 170 Villarroel St., Barcelona, 08036, Spain; Department of Medicine, School of Medicine, University of Barcelona, 143 Casanova St Barcelona 08036, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 153 Rosselló St., Barcelona, 08036, Spain; Centro de Investigación Biomédica en Red Salud Mental (CIBERSAM), 3-5, Monforte de Lemos St., Madrid, 28029, Spain
| | - Gisela Sugranyes
- Department of Child and Adolescent Psychiatry and Psychology, 2017SGR-881 Institute of Neuroscience, Hospital Clínic of Barcelona, 170 Villarroel St., Barcelona, 08036, Spain; Fundació Clínic per a La Recerca Biomèdica (FCRB), 153 Rosselló St., Barcelona, 08036, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 153 Rosselló St., Barcelona, 08036, Spain; Centro de Investigación Biomédica en Red Salud Mental (CIBERSAM), 3-5, Monforte de Lemos St., Madrid, 28029, Spain.
| |
Collapse
|
18
|
Almodóvar-Payá C, Guardiola-Ripoll M, Giralt-López M, Gallego C, Salgado-Pineda P, Miret S, Salvador R, Muñoz MJ, Lázaro L, Guerrero-Pedraza A, Parellada M, Carrión MI, Cuesta MJ, Maristany T, Sarró S, Fañanás L, Callado LF, Arias B, Pomarol-Clotet E, Fatjó-Vilas M. NRN1 Gene as a Potential Marker of Early-Onset Schizophrenia: Evidence from Genetic and Neuroimaging Approaches. Int J Mol Sci 2022; 23:ijms23137456. [PMID: 35806464 PMCID: PMC9267632 DOI: 10.3390/ijms23137456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 12/10/2022] Open
Abstract
Included in the neurotrophins family, the Neuritin 1 gene (NRN1) has emerged as an attractive candidate gene for schizophrenia (SZ) since it has been associated with the risk for the disorder and general cognitive performance. In this work, we aimed to further investigate the association of NRN1 with SZ by exploring its role on age at onset and its brain activity correlates. First, we developed two genetic association analyses using a family-based sample (80 early-onset (EO) trios (offspring onset ≤ 18 years) and 71 adult-onset (AO) trios) and an independent case–control sample (120 healthy subjects (HS), 87 EO and 138 AO patients). Second, we explored the effect of NRN1 on brain activity during a working memory task (N-back task; 39 HS, 39 EO and 39 AO; matched by age, sex and estimated IQ). Different haplotypes encompassing the same three Single Nucleotide Polymorphisms(SNPs, rs3763180–rs10484320–rs4960155) were associated with EO in the two samples (GCT, TCC and GTT). Besides, the GTT haplotype was associated with worse N-back task performance in EO and was linked to an inefficient dorsolateral prefrontal cortex activity in subjects with EO compared to HS. Our results show convergent evidence on the NRN1 association with EO both from genetic and neuroimaging approaches, highlighting the role of neurotrophins in the pathophysiology of SZ.
Collapse
Affiliation(s)
- Carmen Almodóvar-Payá
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Barcelona, Spain; (C.A.-P.); (M.G.-R.); (P.S.-P.); (R.S.); (A.G.-P.); (S.S.)
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
| | - Maria Guardiola-Ripoll
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Barcelona, Spain; (C.A.-P.); (M.G.-R.); (P.S.-P.); (R.S.); (A.G.-P.); (S.S.)
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
| | - Maria Giralt-López
- Departament de Psiquiatria, Hospital Universitari Germans Trias i Pujol (HUGTP), 08916 Badalona, Barcelona, Spain;
- Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Barcelona, Spain
| | - Carme Gallego
- Department of Cell Biology, Molecular Biology Institute of Barcelona (IBMB-CSIC), 08028 Barcelona, Barcelona, Spain;
| | - Pilar Salgado-Pineda
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Barcelona, Spain; (C.A.-P.); (M.G.-R.); (P.S.-P.); (R.S.); (A.G.-P.); (S.S.)
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
| | - Salvador Miret
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
- Centre de Salut Mental d’Adults de Lleida, Servei de Psiquiatria, Salut Mental i Addiccions, Hospital Universitari Santa Maria de Lleida, 25198 Lleida, Lleida, Spain
- Institut de Recerca Biomèdica (IRB), 25198 Lleida, Lleida, Spain
| | - Raymond Salvador
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Barcelona, Spain; (C.A.-P.); (M.G.-R.); (P.S.-P.); (R.S.); (A.G.-P.); (S.S.)
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
| | - María J. Muñoz
- Complex Assistencial en Salut Mental Benito Menni, 08830 Sant Boi de Llobregat, Barcelona, Spain;
| | - Luisa Lázaro
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clinic de Barcelona, 08036 Barcelona, Barcelona, Spain
- Departament de Medicina, Universitat de Barcelona (UB), 08036 Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Barcelona, Spain
| | - Amalia Guerrero-Pedraza
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Barcelona, Spain; (C.A.-P.); (M.G.-R.); (P.S.-P.); (R.S.); (A.G.-P.); (S.S.)
- Complex Assistencial en Salut Mental Benito Menni, 08830 Sant Boi de Llobregat, Barcelona, Spain;
| | - Mara Parellada
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
- Servicio de Psiquiatría del Niño y del Adolescente, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Gregorio Marañón (IiSGM), 28007 Madrid, Madrid, Spain
- Departamento de Psiquiatría, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Madrid, Spain
| | | | - Manuel J. Cuesta
- Servicio de Psiquiatría, Hospital Universitario de Navarra, 31008 Pamplona, Navarra, Spain;
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Navarra, Spain
| | - Teresa Maristany
- Departament de Diagnòstic per la Imatge, Hospital Sant Joan de Déu Fundació de Recerca, 08950 Esplugues de Llobregat, Barcelona, Spain;
| | - Salvador Sarró
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Barcelona, Spain; (C.A.-P.); (M.G.-R.); (P.S.-P.); (R.S.); (A.G.-P.); (S.S.)
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
| | - Lourdes Fañanás
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
- Departament de Biologia Evolutiva, Ecología i Ciències Ambientals, Universitat de Barcelona (UB), 08028 Barcelona, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Barcelona, Spain
| | - Luis F. Callado
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
- Department of Pharmacology, University of the Basque Country, UPV/EHU, 48940 Leioa, Bizkaia, Spain
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
| | - Bárbara Arias
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
- Departament de Biologia Evolutiva, Ecología i Ciències Ambientals, Universitat de Barcelona (UB), 08028 Barcelona, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Barcelona, Spain
| | - Edith Pomarol-Clotet
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Barcelona, Spain; (C.A.-P.); (M.G.-R.); (P.S.-P.); (R.S.); (A.G.-P.); (S.S.)
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
- Correspondence: (E.P.-C.); (M.F.-V.)
| | - Mar Fatjó-Vilas
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Barcelona, Spain; (C.A.-P.); (M.G.-R.); (P.S.-P.); (R.S.); (A.G.-P.); (S.S.)
- Instituto de Salud Carlos III, Biomedical Research Network in Mental Health (CIBERSAM), 28029 Madrid, Madrid, Spain; (S.M.); (L.L.); (M.P.); (L.F.); (L.F.C.); (B.A.)
- Departament de Biologia Evolutiva, Ecología i Ciències Ambientals, Universitat de Barcelona (UB), 08028 Barcelona, Barcelona, Spain
- Correspondence: (E.P.-C.); (M.F.-V.)
| |
Collapse
|
19
|
Iasevoli F, Razzino E, Altavilla B, Avagliano C, Barone A, Ciccarelli M, D'Ambrosio L, Matrone M, Milandri F, Notar Francesco D, Fornaro M, de Bartolomeis A. Relationships between early age at onset of psychotic symptoms and treatment resistant schizophrenia. Early Interv Psychiatry 2022; 16:352-362. [PMID: 33998142 PMCID: PMC9291026 DOI: 10.1111/eip.13174] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 04/21/2021] [Accepted: 05/05/2021] [Indexed: 11/30/2022]
Abstract
AIM Early age at schizophrenia onset (EOS) has been associated with a worse clinical course, although previous studies reported substantial heterogeneity. Despite the relevance of the subject, the relationship between the age of onset and treatment resistant schizophrenia (TRS) is less clear. METHODS We screened 197 non-affective psychotic patients. Of these, 99 suffered from schizophrenia and were putative TRS and were included in a prospective 4-to-8-week trial to assess their response to antipsychotics. According to status (TRS/nonTRS) and age-at-onset (early: ≤18 years, EOS; adult: >18 years, adult onset schizophrenia [AOS]) patients were subdivided in EOS-TRS, EOS-nonTRS, AOS-TRS, AOS-nonTRS. Multiple clinical variables were measured and compared by analysis of covariance (ANCOVA), using age as a covariate. Two-way analysis of variance (ANOVA) was used to assess whether significant differences were attributable to TRS status or age-at-onset. RESULTS The rate of TRS patients was significantly higher in EOS compared to AOS. At the ANCOVA, EOS-TRS had significantly worse clinical, cognitive, and psychosocial outcomes compared to the other groups. Overall, EOS-TRS were more impaired than EOS-nonTRS, while significant differences with AOS-TRS were less consistent, albeit appreciable. Two-way ANOVA demonstrated that, in the majority of the investigated variables, the significant differences among groups were attributable to the TRS status effect rather than to age-at-onset or combined effects. CONCLUSIONS These results suggest that refractoriness to antipsychotics may be strongly linked to the early onset of psychotic symptoms, possibly as a result of common neurobiology.
Collapse
Affiliation(s)
- Felice Iasevoli
- Section of Psychiatry - Unit on Treatment Resistant Psychosis, and Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine Federico II, Naples, Italy
| | - Eugenio Razzino
- Section of Psychiatry - Unit on Treatment Resistant Psychosis, and Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine Federico II, Naples, Italy
| | - Benedetta Altavilla
- Section of Psychiatry - Unit on Treatment Resistant Psychosis, and Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine Federico II, Naples, Italy
| | - Camilla Avagliano
- Section of Psychiatry - Unit on Treatment Resistant Psychosis, and Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine Federico II, Naples, Italy
| | - Annarita Barone
- Section of Psychiatry - Unit on Treatment Resistant Psychosis, and Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine Federico II, Naples, Italy
| | - Mariateresa Ciccarelli
- Section of Psychiatry - Unit on Treatment Resistant Psychosis, and Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine Federico II, Naples, Italy
| | - Luigi D'Ambrosio
- Section of Psychiatry - Unit on Treatment Resistant Psychosis, and Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine Federico II, Naples, Italy
| | - Marta Matrone
- Section of Psychiatry - Unit on Treatment Resistant Psychosis, and Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine Federico II, Naples, Italy
| | - Federica Milandri
- Section of Psychiatry - Unit on Treatment Resistant Psychosis, and Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine Federico II, Naples, Italy
| | - Danilo Notar Francesco
- Section of Psychiatry - Unit on Treatment Resistant Psychosis, and Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine Federico II, Naples, Italy
| | - Michele Fornaro
- Section of Psychiatry - Unit on Treatment Resistant Psychosis, and Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine Federico II, Naples, Italy
| | - Andrea de Bartolomeis
- Section of Psychiatry - Unit on Treatment Resistant Psychosis, and Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine Federico II, Naples, Italy
| |
Collapse
|
20
|
Akingbuwa WA, Hammerschlag AR, Bartels M, Middeldorp CM. Systematic Review: Molecular Studies of Common Genetic Variation in Child and Adolescent Psychiatric Disorders. J Am Acad Child Adolesc Psychiatry 2022; 61:227-242. [PMID: 33932494 DOI: 10.1016/j.jaac.2021.03.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 03/08/2021] [Accepted: 03/19/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVE A systematic review of studies using molecular genetics and statistical approaches to investigate the role of common genetic variation in the development, persistence, and comorbidity of childhood psychiatric traits was conducted. METHOD A literature review was performed using the PubMed database, following PRISMA guidelines. There were 131 studies meeting inclusion criteria, having investigated at least one type of childhood-onset or childhood-measured psychiatric disorder or trait with the aim of identifying trait-associated common genetic variants, estimating the contribution of single nucleotide polymorphisms (SNPs) to the amount of variance explained (SNP-based heritability), investigating genetic overlap between psychiatric traits, or investigating whether the stability in traits or the association with adult traits is explained by genetic factors. RESULTS The first robustly associated genetic variants have started to be identified for childhood psychiatric traits. There were substantial contributions of common genetic variants to many traits, with variation in single nucleotide polymorphism heritability estimates depending on age and raters. Moreover, genetic variants also appeared to explain comorbidity as well as stability across a range of psychiatric traits in childhood and across the life span. CONCLUSION Common genetic variation plays a substantial role in childhood psychiatric traits. Increased sample sizes will lead to increased power to identify genetic variants and to understand genetic architecture, which will ultimately be beneficial to targeted and prevention strategies. This can be achieved by harmonizing phenotype measurements, as is already proposed by large international consortia and by including the collection of genetic material in every study.
Collapse
Affiliation(s)
- Wonuola A Akingbuwa
- Ms. Akingbuwa, Dr. Hammerschlag, and Profs. Bartels and Middeldorp are with Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Ms. Akingbuwa, Dr. Hammerschlag, and Prof. Bartels are also with Amsterdam Public Health Research Institute, Amsterdam University Medical Centres, Amsterdam, The Netherlands.
| | - Anke R Hammerschlag
- Ms. Akingbuwa, Dr. Hammerschlag, and Profs. Bartels and Middeldorp are with Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Ms. Akingbuwa, Dr. Hammerschlag, and Prof. Bartels are also with Amsterdam Public Health Research Institute, Amsterdam University Medical Centres, Amsterdam, The Netherlands; Dr. Hammerschlag and Prof. Middeldorp are also with the Child Health Research Centre, the University of Queensland, Brisbane, Queensland, Australia
| | - Meike Bartels
- Ms. Akingbuwa, Dr. Hammerschlag, and Profs. Bartels and Middeldorp are with Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Ms. Akingbuwa, Dr. Hammerschlag, and Prof. Bartels are also with Amsterdam Public Health Research Institute, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Christel M Middeldorp
- Ms. Akingbuwa, Dr. Hammerschlag, and Profs. Bartels and Middeldorp are with Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Dr. Hammerschlag and Prof. Middeldorp are also with the Child Health Research Centre, the University of Queensland, Brisbane, Queensland, Australia; Prof. Middeldorp is also with the Child and Youth Mental Health Service, Children's Health Queensland Hospital and Health Services, Brisbane, Queensland, Australia
| |
Collapse
|
21
|
Hoffman GE, Roussos P. Dream: powerful differential expression analysis for repeated measures designs. Bioinformatics 2021; 37:192-201. [PMID: 32730587 DOI: 10.1093/bioinformatics/btaa687] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/13/2020] [Accepted: 07/23/2020] [Indexed: 01/08/2023] Open
Abstract
SUMMARY Large-scale transcriptome studies with multiple samples per individual are widely used to study disease biology. Yet, current methods for differential expression are inadequate for cross-individual testing for these repeated measures designs. Most problematic, we observe across multiple datasets that current methods can give reproducible false-positive findings that are driven by genetic regulation of gene expression, yet are unrelated to the trait of interest. Here, we introduce a statistical software package, dream, that increases power, controls the false positive rate, enables multiple types of hypothesis tests, and integrates with standard workflows. In 12 analyses in 6 independent datasets, dream yields biological insight not found with existing software while addressing the issue of reproducible false-positive findings. AVAILABILITY AND IMPLEMENTATION Dream is available within the variancePartition Bioconductor package at http://bioconductor.org/packages/variancePartition. CONTACT gabriel.hoffman@mssm.edu. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Gabriel E Hoffman
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Panos Roussos
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Mental Illness Research, Education, and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY 10468, USA
| |
Collapse
|
22
|
Fernandez A, Drozd M, Thümmler S, Bardoni B, Askenazy F, Capovilla M. A novel microduplication in INPP5A segregates with schizophrenia spectrum disorder in the family of a patient with both childhood onset schizophrenia and autism spectrum disorder. Am J Med Genet A 2021; 185:1841-1847. [PMID: 33720513 DOI: 10.1002/ajmg.a.62155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/14/2021] [Indexed: 01/15/2023]
Abstract
Childhood-Onset Schizophrenia (COS) is a very rare and severe psychiatric disorder defined by adult schizophrenia symptoms occurring before the age of 13. We report a microduplication in the 10q26.3 region including part of the Inositol Polyphosphate-5-Phosphatase A (INPP5A) gene that segregates with Schizophrenia Spectrum Disorders (SSDs) in the family of a female patient affected by both COS and Autism Spectrum Disorder (ASD). Phenotyping and genotyping (including CGH-array) were performed for mother, healthy sister, and affected child according to the GenAuDiss protocol (NCT02565524). The duplication size is 324 kb and is present in a patient with COS and in her mother with SSD, but not in the patient's healthy sister. INPP5A encodes a membrane-associated 43 kDa type I inositol 1,4,5-trisphosphate (InsP3) 5-phosphatase. This protein is found both in mouse and human brains and we found that its Drosophila homologue 5PtaseI is specifically expressed in the central nervous system. Hydrolyzed products from InsP3 5-phosphatases mobilize intracellular calcium, which is relevant for dendritic spine morphogenesis in neurons and altered in both schizophrenia and ASD. These may constitute arguments in favor of this gene alteration in the pathophysiology of COS.
Collapse
Affiliation(s)
- Arnaud Fernandez
- Département de Psychiatrie de l'Enfant et de l'Adolescent, Hôpital de NICE CHU-Lenval, Nice, France.,CoBTek, FRIS, Université Côte d'Azur, Nice, France.,Université Côte d'Azur, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Małgorzata Drozd
- Université Côte d'Azur, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Susanne Thümmler
- Département de Psychiatrie de l'Enfant et de l'Adolescent, Hôpital de NICE CHU-Lenval, Nice, France.,CoBTek, FRIS, Université Côte d'Azur, Nice, France
| | - Barbara Bardoni
- Université Côte d'Azur, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France.,Université Côte d'Azur, INSERM, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Florence Askenazy
- Département de Psychiatrie de l'Enfant et de l'Adolescent, Hôpital de NICE CHU-Lenval, Nice, France.,CoBTek, FRIS, Université Côte d'Azur, Nice, France
| | - Maria Capovilla
- Université Côte d'Azur, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| |
Collapse
|
23
|
Tromp A, Mowry B, Giacomotto J. Neurexins in autism and schizophrenia-a review of patient mutations, mouse models and potential future directions. Mol Psychiatry 2021; 26:747-760. [PMID: 33191396 DOI: 10.1038/s41380-020-00944-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/15/2020] [Accepted: 10/26/2020] [Indexed: 01/29/2023]
Abstract
Mutations in the family of neurexins (NRXN1, NRXN2 and NRXN3) have been repeatedly identified in patients with autism spectrum disorder (ASD) and schizophrenia (SCZ). However, it remains unclear how these DNA variants affect neurexin functions and thereby predispose to these neurodevelopmental disorders. Understanding both the wild-type and pathologic roles of these genes in the brain could help unveil biological mechanisms underlying mental disorders. In this regard, numerous studies have focused on generating relevant loss-of-function (LOF) mammalian models. Although this has increased our knowledge about their normal functions, the potential pathologic role(s) of these human variants remains elusive. Indeed, after reviewing the literature, it seems apparent that a traditional LOF-genetic approach based on complete LOF might not be sufficient to unveil the role of these human mutations. First, these genes present a very complex transcriptome and total-LOF of all isoforms may not be the cause of toxicity in patients, particularly given evidence that causative variants act through haploinsufficiency. Moreover, human DNA variants may not all lead to LOF but potentially to intricate transcriptome changes that could also include the generation of aberrant isoforms acting as a gain-of-function (GOF). Furthermore, their transcriptomic complexity most likely renders them prone to genetic compensation when one tries to manipulate them using traditional site-directed mutagenesis approaches, and this could act differently from model to model leading to heterogeneous and conflicting phenotypes. This review compiles the relevant literature on variants identified in human studies and on the mouse models currently deployed, and offers suggestions for future research.
Collapse
Affiliation(s)
- Alisha Tromp
- Queensland Brain Institute, University of Queensland, St Lucia, QLD, Australia
| | - Bryan Mowry
- Queensland Brain Institute, University of Queensland, St Lucia, QLD, Australia.
- Queensland Centre for Mental Health Research, Brisbane, QLD, Australia.
| | - Jean Giacomotto
- Queensland Brain Institute, University of Queensland, St Lucia, QLD, Australia.
- Queensland Centre for Mental Health Research, Brisbane, QLD, Australia.
| |
Collapse
|
24
|
De Berardis D, De Filippis S, Masi G, Vicari S, Zuddas A. A Neurodevelopment Approach for a Transitional Model of Early Onset Schizophrenia. Brain Sci 2021; 11:brainsci11020275. [PMID: 33672396 PMCID: PMC7926620 DOI: 10.3390/brainsci11020275] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/16/2022] Open
Abstract
In the last decades, the conceptualization of schizophrenia has dramatically changed, moving from a neurodegenerative process occurring in early adult life to a neurodevelopmental disorder starting be-fore birth, showing a variety of premorbid and prodromal symptoms and, in relatively few cases, evolving in the full-blown psychotic syndrome. High rates of co-occurring different neurodevelopmental disorders such as Autism spectrum disorder and ADHD, predating the onset of SCZ, and neurobio-logical underpinning with significant similarities, support the notion of a pan-developmental disturbance consisting of impairments in neuromotor, receptive language, social and cognitive development. Con-sidering that many SCZ risk factors may be similar to symptoms of other neurodevelopmental psychi-atric disorders, transition processes from child & adolescent to adult systems of care should include both high risk people as well as subject with other neurodevelopmental psychiatric disorders with different levels of severity. This descriptive mini-review discuss the need of innovative clinical approaches, re-considering specific diagnostic categories, stimulating a careful analysis of risk factors and promoting the appropriate use of new and safer medications.
Collapse
Affiliation(s)
- Domenico De Berardis
- Department of Mental Health, Psychiatric Service of Diagnosis and Treatment, Hospital “G. Mazzini,” National Health Service (NHS), 64100 ASL 4 Teramo, Italy
- Department of Neurosciences and Imaging, University “G. D’Annunzio”, 66100 Chieti, Italy
- Correspondence:
| | - Sergio De Filippis
- Department of Neuropsychiatry, Villa von Siebenthal Neuropsychiatric Hospital and Clinic, Genzano di Roma, 100045 Rome, Italy;
| | - Gabriele Masi
- IRCCS Stella Maris, Scientific Institute of Child Neurology and Psychiatry, Calambrone, 56128 Pisa, Italy;
| | - Stefano Vicari
- Department of Life Sciences and Publich Health, Catholic University, 00135 Rome, Italy;
- Child & Adolescent Psychiatry, Bambino Gesù Children’s Hospital, 00168 Rome, Italy
| | - Alessandro Zuddas
- Child and Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, University of Cagliari and “A Cao” Paediatric Hospital, “G Brotzu” Hospital Trust, 109134 Cagliari, Italy;
| |
Collapse
|
25
|
Mørch-Johnsen L, Smelror RE, Andreou D, Barth C, Johannessen C, Wedervang-Resell K, Wortinger LA, Díaz R, Victoria G, Ueland T, Andreassen OA, Myhre AM, Rund BR, Ulloa RE, Agartz I. Negative Symptom Domains Are Associated With Verbal Learning in Adolescents With Early Onset Psychosis. Front Psychiatry 2021; 12:825681. [PMID: 35069300 PMCID: PMC8777217 DOI: 10.3389/fpsyt.2021.825681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/16/2021] [Indexed: 01/09/2023] Open
Abstract
Background: Early-onset psychosis (EOP) is among the leading causes of disease burden in adolescents. Negative symptoms and cognitive deficits predicts poorer functional outcome. A better understanding of the association between negative symptoms and cognitive impairment may inform theories on underlying mechanisms and elucidate targets for development of new treatments. Two domains of negative symptoms have been described in adult patients with schizophrenia: apathy and diminished expression, however, the factorial structure of negative symptoms has not been investigated in EOP. We aimed to explore the factorial structure of negative symptoms and investigate associations between cognitive performance and negative symptom domains in adolescents with EOP. We hypothesized that (1) two negative symptom factors would be identifiable, and that (2) diminished expression would be more strongly associated with cognitive performance, similar to adult psychosis patients. Methods: Adolescent patients with non-affective EOP (n = 169) were included from three cohorts: Youth-TOP, Norway (n = 45), Early-Onset Study, Norway (n = 27) and Adolescent Schizophrenia Study, Mexico (n = 97). An exploratory factor analysis was performed to investigate the underlying structure of negative symptoms (measured with the Positive and Negative Syndrome Scale (PANSS)). Factor-models were further assessed using confirmatory factor analyses. Associations between negative symptom domains and six cognitive domains were assessed using multiple linear regression models controlling for age, sex and cohort. The neurocognitive domains from the MATRICS Consensus Cognitive Battery included: speed of processing, attention, working memory, verbal learning, visual learning, and reasoning and problem solving. Results: The exploratory factor analysis of PANSS negative symptoms suggested retaining only a single factor, but a forced two factor solution corroborated previously described factors of apathy and diminished expression in adult-onset schizophrenia. Results from confirmatory factor analysis indicated a better fit for the two-factor model than for the one-factor model. For both negative symptom domains, negative symptom scores were inversely associated with verbal learning scores. Conclusion: The results support the presence of two domains of negative symptoms in EOP; apathy and diminished expression. Future studies on negative symptoms in EOP should examine putative differential effects of these symptom domains. For both domains, negative symptom scores were significantly inversely associated with verbal learning.
Collapse
Affiliation(s)
- Lynn Mørch-Johnsen
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Psychiatry and Department of Clinical Research, Østfold Hospital, Grålum, Norway
| | - Runar Elle Smelror
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Dimitrios Andreou
- Norwegian Centre for Mental Disorders Research, 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 and Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - Claudia Barth
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Cecilie Johannessen
- Department of Neurohabilitation, Oslo University Hospital Ullevål, Oslo, Norway
| | - Kirsten Wedervang-Resell
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Laura A Wortinger
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Ricardo Díaz
- Research Department, Arete Proyectos y Administración, Mexico City, Mexico
| | - Gamaliel Victoria
- Planning of Prevention Programs in the Directorate of Integral Attention to Girls, Boys and Adolescents, System for the Integral Development of the Family, Mexico City, Mexico
| | - Torill Ueland
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, Oslo University Hospital, Oslo, Norway.,Department of Psychology, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, Oslo University Hospital, Oslo, Norway
| | - Anne M Myhre
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bjørn Rishovd Rund
- Department of Psychology, University of Oslo, Oslo, Norway.,Research Department, Vestre Viken Hospital Trust, Drammen, Norway
| | - Rosa Elena Ulloa
- Developmental Psychopharmacology at the Research Division, Child Psychiatric Hospital, Mexico City, Mexico
| | - Ingrid Agartz
- Norwegian Centre for Mental Disorders Research, 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 and Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden.,Institute of Clinical Medicine, K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| |
Collapse
|
26
|
Reduced levels of circulating adhesion molecules in adolescents with early-onset psychosis. NPJ SCHIZOPHRENIA 2020; 6:20. [PMID: 32811840 PMCID: PMC7434772 DOI: 10.1038/s41537-020-00112-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
Abstract
It is suggested that neurodevelopmental abnormalities are involved in the disease mechanisms of psychotic disorders. Although cellular adhesion molecules (CAMs) participate in neurodevelopment, modulate blood–brain barrier permeability, and facilitate leukocyte migration, findings concerning their systemic levels in adults with psychosis are inconsistent. We examined plasma levels and mRNA expression in peripheral blood mononuclear cells (PBMCs) of selected CAMs in adolescents with early-onset psychosis (EOP) aged 12–18 years (n = 37) and age-matched healthy controls (HC) (n = 68). EOP patients exhibited significantly lower circulating levels of soluble platelet selectin (~−22%) and soluble vascular cell adhesion molecule-1 (~−14%) than HC. We found no significant associations with symptom severity. PSEL mRNA expression was increased in PBMCs of patients and significantly negatively correlated to duration of illness. These findings suggest a role for CAMs in the pathophysiology of psychotic disorders.
Collapse
|
27
|
Ohi K, Nishizawa D, Shimada T, Kataoka Y, Hasegawa J, Shioiri T, Kawasaki Y, Hashimoto R, Ikeda K. Polygenetic Risk Scores for Major Psychiatric Disorders Among Schizophrenia Patients, Their First-Degree Relatives, and Healthy Participants. Int J Neuropsychopharmacol 2020; 23:157-164. [PMID: 31900488 PMCID: PMC7171929 DOI: 10.1093/ijnp/pyz073] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/17/2019] [Accepted: 01/01/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The genetic etiology of schizophrenia (SCZ) overlaps with that of other major psychiatric disorders in samples of European ancestry. The present study investigated transethnic polygenetic features shared between Japanese SCZ or their unaffected first-degree relatives and European patients with major psychiatric disorders by conducting polygenic risk score (PRS) analyses. METHODS To calculate PRSs for 5 psychiatric disorders (SCZ, bipolar disorder [BIP], major depressive disorder, autism spectrum disorder, and attention-deficit/hyperactivity disorder) and PRSs differentiating SCZ from BIP, we utilized large-scale European genome-wide association study (GWAS) datasets as discovery samples. PRSs derived from these GWASs were calculated for 335 Japanese target participants [SCZ patients, FRs, and healthy controls (HCs)]. We took these PRSs based on GWASs of European psychiatric disorders and investigated their effect on risk in Japanese SCZ patients and unaffected first-degree relatives. RESULTS The PRSs obtained from European SCZ and BIP patients were higher in Japanese SCZ patients than in HCs. Furthermore, PRSs differentiating SCZ patients from European BIP patients were higher in Japanese SCZ patients than in HCs. Interestingly, PRSs related to European autism spectrum disorder were lower in Japanese first-degree relatives than in HCs or SCZ patients. The PRSs of autism spectrum disorder were positively correlated with a young onset age of SCZ. CONCLUSIONS These findings suggest that polygenic factors related to European SCZ and BIP and the polygenic components differentiating SCZ from BIP can transethnically contribute to SCZ risk in Japanese people. Furthermore, we suggest that reduced levels of an ASD-related genetic factor in unaffected first-degree relatives may help protect against SCZ development.
Collapse
Affiliation(s)
- Kazutaka Ohi
- Department of Neuropsychiatry, Kanazawa Medical University, Ishikawa, Japan
- Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
- Department of General Internal Medicine, Kanazawa Medical University, Ishikawa, Japan
- Department of Psychiatry and Psychotherapy, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Daisuke Nishizawa
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takamitsu Shimada
- Department of Neuropsychiatry, Kanazawa Medical University, Ishikawa, Japan
| | - Yuzuru Kataoka
- Department of Neuropsychiatry, Kanazawa Medical University, Ishikawa, Japan
| | - Junko Hasegawa
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Toshiki Shioiri
- Department of Psychiatry and Psychotherapy, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yasuhiro Kawasaki
- Department of Neuropsychiatry, Kanazawa Medical University, Ishikawa, Japan
| | - Ryota Hashimoto
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
- Molecular Research Center for Children’s Mental Development, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
28
|
Moreau C, Rébillard RM, Wolking S, Michaud J, Tremblay F, Girard A, Bouchard J, Minassian B, Laprise C, Cossette P, Girard SL. Polygenic risk scores of several subtypes of epilepsies in a founder population. NEUROLOGY-GENETICS 2020; 6:e416. [PMID: 32337343 PMCID: PMC7164970 DOI: 10.1212/nxg.0000000000000416] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/13/2020] [Indexed: 02/04/2023]
Abstract
Objective Polygenic risk scores (PRSs) are used to quantify the cumulative effects of a number of genetic variants, which may individually have a very small effect on susceptibility to a disease; we used PRSs to better understand the genetic contribution to common epilepsy and its subtypes. Methods We first replicated previous single associations using 373 unrelated patients. We then calculated PRSs in the same French Canadian patients with epilepsy divided into 7 epilepsy subtypes and population-based controls. We fitted a logistic mixed model to calculate the variance explained by the PRS using pseudo-R2 statistics. Results We show that the PRS explains more of the variance in idiopathic generalized epilepsy than in patients with nonacquired focal epilepsy. We also demonstrate that the variance explained is different within each epilepsy subtype. Conclusions Globally, we support the notion that PRSs provide a reliable measure to rightfully estimate the contribution of genetic factors to the pathophysiologic mechanism of epilepsies, but further studies are needed on PRSs before they can be used clinically.
Collapse
Affiliation(s)
- Claudia Moreau
- Centre Intersectoriel en Santé Durable (C.M., F.T., A.G., J.B., C.L., S.L.G.), Université du Québec à Chicoutimi, Saguenay; Axe Neurosciences (R.-M.R., S.W., P.C.), Centre de recherche de l'Université de Montréal, Université de Montréal; Centre de recherche du CHU Ste-Justine (J.M.), Université de Montréal, Canada; and Department of Pediatrics and Neurology and Neurotherapeutics (B.M.), UT Southwestern Medical Center, TX
| | - Rose-Marie Rébillard
- Centre Intersectoriel en Santé Durable (C.M., F.T., A.G., J.B., C.L., S.L.G.), Université du Québec à Chicoutimi, Saguenay; Axe Neurosciences (R.-M.R., S.W., P.C.), Centre de recherche de l'Université de Montréal, Université de Montréal; Centre de recherche du CHU Ste-Justine (J.M.), Université de Montréal, Canada; and Department of Pediatrics and Neurology and Neurotherapeutics (B.M.), UT Southwestern Medical Center, TX
| | - Stefan Wolking
- Centre Intersectoriel en Santé Durable (C.M., F.T., A.G., J.B., C.L., S.L.G.), Université du Québec à Chicoutimi, Saguenay; Axe Neurosciences (R.-M.R., S.W., P.C.), Centre de recherche de l'Université de Montréal, Université de Montréal; Centre de recherche du CHU Ste-Justine (J.M.), Université de Montréal, Canada; and Department of Pediatrics and Neurology and Neurotherapeutics (B.M.), UT Southwestern Medical Center, TX
| | - Jacques Michaud
- Centre Intersectoriel en Santé Durable (C.M., F.T., A.G., J.B., C.L., S.L.G.), Université du Québec à Chicoutimi, Saguenay; Axe Neurosciences (R.-M.R., S.W., P.C.), Centre de recherche de l'Université de Montréal, Université de Montréal; Centre de recherche du CHU Ste-Justine (J.M.), Université de Montréal, Canada; and Department of Pediatrics and Neurology and Neurotherapeutics (B.M.), UT Southwestern Medical Center, TX
| | - Frédérique Tremblay
- Centre Intersectoriel en Santé Durable (C.M., F.T., A.G., J.B., C.L., S.L.G.), Université du Québec à Chicoutimi, Saguenay; Axe Neurosciences (R.-M.R., S.W., P.C.), Centre de recherche de l'Université de Montréal, Université de Montréal; Centre de recherche du CHU Ste-Justine (J.M.), Université de Montréal, Canada; and Department of Pediatrics and Neurology and Neurotherapeutics (B.M.), UT Southwestern Medical Center, TX
| | - Alexandre Girard
- Centre Intersectoriel en Santé Durable (C.M., F.T., A.G., J.B., C.L., S.L.G.), Université du Québec à Chicoutimi, Saguenay; Axe Neurosciences (R.-M.R., S.W., P.C.), Centre de recherche de l'Université de Montréal, Université de Montréal; Centre de recherche du CHU Ste-Justine (J.M.), Université de Montréal, Canada; and Department of Pediatrics and Neurology and Neurotherapeutics (B.M.), UT Southwestern Medical Center, TX
| | - Joanie Bouchard
- Centre Intersectoriel en Santé Durable (C.M., F.T., A.G., J.B., C.L., S.L.G.), Université du Québec à Chicoutimi, Saguenay; Axe Neurosciences (R.-M.R., S.W., P.C.), Centre de recherche de l'Université de Montréal, Université de Montréal; Centre de recherche du CHU Ste-Justine (J.M.), Université de Montréal, Canada; and Department of Pediatrics and Neurology and Neurotherapeutics (B.M.), UT Southwestern Medical Center, TX
| | - Berge Minassian
- Centre Intersectoriel en Santé Durable (C.M., F.T., A.G., J.B., C.L., S.L.G.), Université du Québec à Chicoutimi, Saguenay; Axe Neurosciences (R.-M.R., S.W., P.C.), Centre de recherche de l'Université de Montréal, Université de Montréal; Centre de recherche du CHU Ste-Justine (J.M.), Université de Montréal, Canada; and Department of Pediatrics and Neurology and Neurotherapeutics (B.M.), UT Southwestern Medical Center, TX
| | - Catherine Laprise
- Centre Intersectoriel en Santé Durable (C.M., F.T., A.G., J.B., C.L., S.L.G.), Université du Québec à Chicoutimi, Saguenay; Axe Neurosciences (R.-M.R., S.W., P.C.), Centre de recherche de l'Université de Montréal, Université de Montréal; Centre de recherche du CHU Ste-Justine (J.M.), Université de Montréal, Canada; and Department of Pediatrics and Neurology and Neurotherapeutics (B.M.), UT Southwestern Medical Center, TX
| | - Patrick Cossette
- Centre Intersectoriel en Santé Durable (C.M., F.T., A.G., J.B., C.L., S.L.G.), Université du Québec à Chicoutimi, Saguenay; Axe Neurosciences (R.-M.R., S.W., P.C.), Centre de recherche de l'Université de Montréal, Université de Montréal; Centre de recherche du CHU Ste-Justine (J.M.), Université de Montréal, Canada; and Department of Pediatrics and Neurology and Neurotherapeutics (B.M.), UT Southwestern Medical Center, TX
| | - Simon L Girard
- Centre Intersectoriel en Santé Durable (C.M., F.T., A.G., J.B., C.L., S.L.G.), Université du Québec à Chicoutimi, Saguenay; Axe Neurosciences (R.-M.R., S.W., P.C.), Centre de recherche de l'Université de Montréal, Université de Montréal; Centre de recherche du CHU Ste-Justine (J.M.), Université de Montréal, Canada; and Department of Pediatrics and Neurology and Neurotherapeutics (B.M.), UT Southwestern Medical Center, TX
| |
Collapse
|
29
|
Gordovez FJA, McMahon FJ. The genetics of bipolar disorder. Mol Psychiatry 2020; 25:544-559. [PMID: 31907381 DOI: 10.1038/s41380-019-0634-7] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 11/22/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022]
Abstract
Bipolar disorder (BD) is one of the most heritable mental illnesses, but the elucidation of its genetic basis has proven to be a very challenging endeavor. Genome-Wide Association Studies (GWAS) have transformed our understanding of BD, providing the first reproducible evidence of specific genetic markers and a highly polygenic architecture that overlaps with that of schizophrenia, major depression, and other disorders. Individual GWAS markers appear to confer little risk, but common variants together account for about 25% of the heritability of BD. A few higher-risk associations have also been identified, such as a rare copy number variant on chromosome 16p11.2. Large scale next-generation sequencing studies are actively searching for other alleles that confer substantial risk. As our understanding of the genetics of BD improves, there is growing optimism that some clear biological pathways will emerge, providing a basis for future studies aimed at molecular diagnosis and novel therapeutics.
Collapse
Affiliation(s)
- Francis James A Gordovez
- Human Genetics Branch, National Institute of Mental Health Intramural Research Program, Department of Health and Human Services, National Institutes of Health, Bethesda, MD, USA.,College of Medicine, University of the Philippines Manila, 1000, Ermita, Manila, Philippines
| | - Francis J McMahon
- Human Genetics Branch, National Institute of Mental Health Intramural Research Program, Department of Health and Human Services, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
30
|
Coulon N, Godin O, Bulzacka E, Dubertret C, Mallet J, Fond G, Brunel L, Andrianarisoa M, Anderson G, Chereau I, Denizot H, Rey R, Dorey JM, Lançon C, Faget C, Roux P, Passerieux C, Dubreucq J, Leignier S, Capdevielle D, André M, Aouizerate B, Misdrahi D, Berna F, Vidailhet P, Leboyer M, Schürhoff F. Early and very early-onset schizophrenia compared with adult-onset schizophrenia: French FACE-SZ database. Brain Behav 2020; 10:e01495. [PMID: 31908151 PMCID: PMC7010576 DOI: 10.1002/brb3.1495] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 11/06/2019] [Accepted: 11/11/2019] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To compare the clinical symptomatology in patients with Early-Onset Schizophrenia (EOS, N = 176), especially the subgroup Very Early Onset Schizophrenia (VEOS) and Adult Onset Schizophrenia (AOS, N = 551). METHOD In a large French multicentric sample, 727 stable schizophrenia patients, classified by age at onset of the disorder, were assessed using standardized and extensive clinical and neuropsychological batteries: AOS with onset ≥ 18 years and EOS with onset < 18 years (including 22 VEOS < 13 years). RESULTS The importance of better diagnosing EOS group, and in particularly VEOS, appeared in a longer DUP Duration of Untreated Psychosis (respectively, 2.6 years ± 4.1 and 8.1 years ± 5.7 vs. 1.0 years ± 2.5), more severe symptomatology (PANSS Positive And Negative Syndrome Scale scores), and lower educational level than the AOS group. In addition, the VEOS subgroup had a more frequent childhood history of learning disabilities and lower prevalence of right-handedness quotient than the AOS. CONCLUSION The study demonstrates the existence of an increased gradient of clinical severity from AOS to VEOS. In order to improve the prognosis of the early forms of schizophrenia and to reduce the DUP, clinicians need to pay attention to the prodromal manifestations of the disease.
Collapse
Affiliation(s)
- Nathalie Coulon
- Fondation FondaMental, Créteil, France.,INSERM U955, Translational Psychiatry laboratory, AP-HP, DHU Pe-PSY, Centre Expert Schizophrénie, Pôle de Psychiatrie et d'Addictologie des Hôpitaux Universitaires Henri Mondor, Université Paris Est Créteil, Créteil, France.,INSERM U894, AP-HP, Department of Psychiatry, Louis Mourier Hospital, Paris Diderot University, Sorbonne Paris Cité, Faculté de médecine, Colombes, France
| | - Ophélia Godin
- Fondation FondaMental, Créteil, France.,INSERM U955, Translational Psychiatry laboratory, AP-HP, DHU Pe-PSY, Centre Expert Schizophrénie, Pôle de Psychiatrie et d'Addictologie des Hôpitaux Universitaires Henri Mondor, Université Paris Est Créteil, Créteil, France
| | - Ewa Bulzacka
- Fondation FondaMental, Créteil, France.,INSERM U955, Translational Psychiatry laboratory, AP-HP, DHU Pe-PSY, Centre Expert Schizophrénie, Pôle de Psychiatrie et d'Addictologie des Hôpitaux Universitaires Henri Mondor, Université Paris Est Créteil, Créteil, France
| | - Caroline Dubertret
- Fondation FondaMental, Créteil, France.,INSERM U894, AP-HP, Department of Psychiatry, Louis Mourier Hospital, Paris Diderot University, Sorbonne Paris Cité, Faculté de médecine, Colombes, France
| | - Jasmina Mallet
- Fondation FondaMental, Créteil, France.,INSERM U894, AP-HP, Department of Psychiatry, Louis Mourier Hospital, Paris Diderot University, Sorbonne Paris Cité, Faculté de médecine, Colombes, France
| | - Guillaume Fond
- Fondation FondaMental, Créteil, France.,EA 3279 : CEReSS -Centre d'Etude et de Recherche sur les Services de Santé et la Qualité de vie, Aix-Marseille Univ, Faculté de Médecine, Marseille, France
| | - Lore Brunel
- Fondation FondaMental, Créteil, France.,INSERM U955, Translational Psychiatry laboratory, AP-HP, DHU Pe-PSY, Centre Expert Schizophrénie, Pôle de Psychiatrie et d'Addictologie des Hôpitaux Universitaires Henri Mondor, Université Paris Est Créteil, Créteil, France
| | - Méja Andrianarisoa
- Fondation FondaMental, Créteil, France.,INSERM U955, Translational Psychiatry laboratory, AP-HP, DHU Pe-PSY, Centre Expert Schizophrénie, Pôle de Psychiatrie et d'Addictologie des Hôpitaux Universitaires Henri Mondor, Université Paris Est Créteil, Créteil, France
| | | | - Isabelle Chereau
- Fondation FondaMental, Créteil, France.,Clermont-Ferrand University Hospital, EA 7280 Auvergne University, BP 69, Clermont-Ferrand, France
| | - Hélène Denizot
- Fondation FondaMental, Créteil, France.,Clermont-Ferrand University Hospital, EA 7280 Auvergne University, BP 69, Clermont-Ferrand, France
| | - Romain Rey
- Fondation FondaMental, Créteil, France.,INSERM U1028, CNRS UMR 5292, Centre de Recherche en Neurosciences de Lyon, Equipe PSYR2, Centre Hospitalier Le Vinatier, Pole Est, Claude Bernard Lyon 1 University, Bron Cedex, France
| | - Jean-Michel Dorey
- Fondation FondaMental, Créteil, France.,INSERM U1028, CNRS UMR 5292, Centre de Recherche en Neurosciences de Lyon, Equipe PSYR2, Centre Hospitalier Le Vinatier, Pole Est, Claude Bernard Lyon 1 University, Bron Cedex, France
| | - Christophe Lançon
- Fondation FondaMental, Créteil, France.,Department of Psychiatry (AP-HM), Sainte-Marguerite University Hospital, Marseille, France
| | - Catherine Faget
- Fondation FondaMental, Créteil, France.,Department of Psychiatry (AP-HM), Sainte-Marguerite University Hospital, Marseille, France
| | - Paul Roux
- Fondation FondaMental, Créteil, France.,Department of Adult Psychiatry, Versailles Hospital, Le Chesnay, France
| | - Christine Passerieux
- Fondation FondaMental, Créteil, France.,Department of Adult Psychiatry, Versailles Hospital, Le Chesnay, France
| | - Julien Dubreucq
- Fondation FondaMental, Créteil, France.,Psychosocial Rehabilitation Reference Center, Alpes Isère Hospital, Grenoble, France
| | - Sylvain Leignier
- Fondation FondaMental, Créteil, France.,Psychosocial Rehabilitation Reference Center, Alpes Isère Hospital, Grenoble, France
| | - Delphine Capdevielle
- Fondation FondaMental, Créteil, France.,INSERM 1061, University Department of Adult Psychiatry, La Colombiere Hospital, CHU Montpellier, University of Montpellier 1, Montpellier, France
| | - Myrtille André
- Fondation FondaMental, Créteil, France.,INSERM 1061, University Department of Adult Psychiatry, La Colombiere Hospital, CHU Montpellier, University of Montpellier 1, Montpellier, France
| | - Bruno Aouizerate
- Fondation FondaMental, Créteil, France.,Department of Adult Psychiatry, Charles Perrens Hospital, University of Bordeaux, Bordeaux, France
| | - David Misdrahi
- Fondation FondaMental, Créteil, France.,Department of Adult Psychiatry, Charles Perrens Hospital, University of Bordeaux, Bordeaux, France
| | - Fabrice Berna
- Fondation FondaMental, Créteil, France.,INSERM U1114, Strasbourg University Hospital, University of Strasbourg, Federation of Translational Psychiatry, Strasbourg, France
| | - Pierre Vidailhet
- Fondation FondaMental, Créteil, France.,INSERM U1114, Strasbourg University Hospital, University of Strasbourg, Federation of Translational Psychiatry, Strasbourg, France
| | - Marion Leboyer
- Fondation FondaMental, Créteil, France.,INSERM U955, Translational Psychiatry laboratory, AP-HP, DHU Pe-PSY, Centre Expert Schizophrénie, Pôle de Psychiatrie et d'Addictologie des Hôpitaux Universitaires Henri Mondor, Université Paris Est Créteil, Créteil, France
| | - Franck Schürhoff
- Fondation FondaMental, Créteil, France.,INSERM U955, Translational Psychiatry laboratory, AP-HP, DHU Pe-PSY, Centre Expert Schizophrénie, Pôle de Psychiatrie et d'Addictologie des Hôpitaux Universitaires Henri Mondor, Université Paris Est Créteil, Créteil, France
| |
Collapse
|
31
|
Poisson A, Chatron N, Labalme A, Fourneret P, Ville D, Mathieu ML, Sanlaville D, Demily C, Lesca G. Chromatin remodeling dysfunction extends the etiological spectrum of schizophrenia: a case report. BMC MEDICAL GENETICS 2020; 21:10. [PMID: 31914951 PMCID: PMC6950831 DOI: 10.1186/s12881-019-0946-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 12/29/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND The role of deleterious copy number variations in schizophrenia is well established while data regarding pathogenic variations remain scarce. We report for the first time a case of schizophrenia in a child with a pathogenic mutation of the chromodomain helicase DNA binding protein 2 (CHD2) gene. CASE PRESENTATION The proband was the second child of unrelated parents. Anxiety and sleep disorders appeared at the age of 10 months. He presented febrile seizures and, at the age of 8, two generalized tonic-clonic seizures. At the age of 10, emotional withdrawal emerged, along with a flat affect, disorganization and paranoid ideation, without seizures. He began to talk and giggle with self. Eventually, the patient presented daily auditory and visual hallucinations. The diagnosis of childhood onset schizophrenia (DSM V) was then evoked. Brain imaging was unremarkable. Wakefulness electroencephalography showed a normal background and some bilateral spike-wave discharges that did not explain the psychosis features. A comparative genomic hybridization array (180 K, Agilent, Santa Clara, CA, USA) revealed an 867-kb 16p13.3 duplication, interpreted as a variant of unknown significance confirmed by a quantitative PCR that also showed its maternal inheritance. Risperidone (1,5 mg per day), led to clinical improvement. At the age of 11, an explosive relapse of epilepsy occurred with daily seizures of various types. The sequencing of a panel for monogenic epileptic disorders and Sanger sequencing revealed a de novo pathogenic heterozygous transition in CHD2 (NM_001271.3: c.4003G > T). CONCLUSIONS This case underlines that schizophrenia may be, sometimes, underpinned by a Mendelian disease. It addresses the question of systematic genetic investigations in the presence of warning signs such as a childhood onset of the schizophrenia or a resistant epilepsy. It points that, in the absence of pathogenic copy number variation, the investigations should also include a search for pathogenic variations, which means that some of the patients with schizophrenia should benefit from Next Generation Sequencing tools. Last but not least, CHD2 encodes a member of the chromodomain helicase DNA-binding (CHD) family involved in chromatin remodeling. This observation adds schizophrenia to the phenotypic spectrum of chromodomain remodeling disorders, which may lead to innovative therapeutic approaches.
Collapse
Affiliation(s)
- Alice Poisson
- GénoPsy, Reference Center for Diagnosis and Management of Genetic Psychiatric Disorders, Centre Hospitalier le Vinatier and EDR-Psy Q19 Team (Centre National de la Recherche Scientifique & Lyon 1 Claude Bernard University), le Vinatier, 69500, Bron, CH, France.
| | - Nicolas Chatron
- Institut Neuromyogène, métabolisme énergétique et développement durable, CNRS UMR 5310, INSERM U1217, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Audrey Labalme
- Institut Neuromyogène, métabolisme énergétique et développement durable, CNRS UMR 5310, INSERM U1217, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Pierre Fourneret
- Service de psychopathologie du développement, hôpital Femme-Mère-Enfant, hospices civils de Lyon, 69677, Bron cedex, France.,Institut des sciences cognitives CNRS UMR, 530467 boulevard Pinel, 69675, Bron cedex, France.,Faculté de médecine Lyon-Est, université Claude-Bernard - Lyon 1, 69003, Lyon, France
| | - Dorothée Ville
- Département de Neurologie Pédiatrique et Centre de Référence des Epilepsies Rares, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Centre Hospitalier Universitaire de Lyon, Lyon, France
| | - Marie Laure Mathieu
- Neuropaediatrics Department, Femme Mère Enfant Hospital, Lyon, France.,Claude Bernard Lyon 1 University, Lyon, France
| | - Damien Sanlaville
- Institut Neuromyogène, métabolisme énergétique et développement durable, CNRS UMR 5310, INSERM U1217, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Caroline Demily
- GénoPsy, Reference Center for Diagnosis and Management of Genetic Psychiatric Disorders, Centre Hospitalier le Vinatier and EDR-Psy Q19 Team (Centre National de la Recherche Scientifique & Lyon 1 Claude Bernard University), le Vinatier, 69500, Bron, CH, France
| | - Gaëtan Lesca
- Institut Neuromyogène, métabolisme énergétique et développement durable, CNRS UMR 5310, INSERM U1217, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
32
|
Driver DI, Thomas S, Gogtay N, Rapoport JL. Childhood-Onset Schizophrenia and Early-onset Schizophrenia Spectrum Disorders: An Update. Child Adolesc Psychiatr Clin N Am 2020; 29:71-90. [PMID: 31708054 DOI: 10.1016/j.chc.2019.08.017] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The clinical severity, impact on development, and poor prognosis of childhood-onset schizophrenia may represent a more homogeneous group. Positive symptoms in children are necessary for the diagnosis, and hallucinations are more often multimodal. In healthy children and children with a variety of other psychiatric illnesses, hallucinations are not uncommon and diagnosis should not be based on these alone. Childhood-onset schizophrenia is an extraordinarily rare illness that is poorly understood but seems continuous with the adult-onset disorder. Once a diagnosis is confirmed, aggressive medication treatment combined with family education and individual counseling may prevent further deterioration.
Collapse
Affiliation(s)
- David I Driver
- Child Psychiatry Branch, National Institutes of Mental Health (NIMH), National Institutes Health (NIH), Building 10, Room 4N313C, 10 Center Drive, Bethesda, MD 20814, USA.
| | - Shari Thomas
- Healthy Foundations Group, 4350 East West Highway, Suite 200, Bethesda, Maryland 20814, USA
| | - Nitin Gogtay
- National Institutes Health (NIH), NSC Building, Room 6104, 6001 Executive Boulevard, Rockville, MD 20852, USA
| | - Judith L Rapoport
- National Institutes Health (NIH), Building 10-CRC, Room 6-5332, 10 Center Drive, Bethesda, MD 20814, USA
| |
Collapse
|
33
|
Abstract
The genetic architecture of schizophrenia is complex and highly polygenic. This article discusses key findings from genetic studies of childhood-onset schizophrenia (COS) and the more common adult-onset schizophrenia (AOS), including studies of familial aggregation and common, rare, and copy number variants. Extant literature suggests that COS is a rare variant of AOS involving greater familial aggregation of schizophrenia spectrum disorders and a potentially higher occurrence of pathogenic copy number variants. The direct utility of genetics to clinical practice for COS is currently limited; however, identifying common pathways through which risk genes affect brain function offers promise for novel interventions.
Collapse
Affiliation(s)
- Jennifer K Forsyth
- Department of Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90095, USA.
| | - Robert F Asarnow
- Department of Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90095, USA; Department of Psychology, University of California, Los Angeles, 502 Portola Plaza Los Angeles, CA 90095, USA; Brain Research Institute, University of California, Los Angeles, 695 Charles E Young Dr S, Los Angeles, CA 90095, USA.
| |
Collapse
|
34
|
Fernandez A, Drozd MM, Thümmler S, Dor E, Capovilla M, Askenazy F, Bardoni B. Childhood-Onset Schizophrenia: A Systematic Overview of Its Genetic Heterogeneity From Classical Studies to the Genomic Era. Front Genet 2019; 10:1137. [PMID: 31921276 PMCID: PMC6930680 DOI: 10.3389/fgene.2019.01137] [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: 06/20/2019] [Accepted: 10/21/2019] [Indexed: 12/18/2022] Open
Abstract
Childhood-onset schizophrenia (COS), a very rare and severe chronic psychiatric condition, is defined by an onset of positive symptoms (delusions, hallucinations and disorganized speech or behavior) before the age of 13. COS is associated with other neurodevelopmental disorders such as autism spectrum disorder (ASD) and attention deficit and hyperactivity disorder. Copy number variations (CNVs) represent well documented neurodevelopmental disorder risk factors and, recently, de novo single nucleotide variations (SNVs) in genes involved in brain development have also been implicated in the complex genetic architecture of COS. Here, we aim to review the genetic changes (CNVs and SNVs) reported for COS, going from previous studies to the whole genome sequencing era. We carried out a systematic review search in PubMed using the keywords “childhood(early)-onset schizophrenia(psychosis)” and “genetic(s) or gene(s) or genomic(s)” without language and date limitations. The main inclusion criteria are COS (onset before 13 years old) and all changes/variations at the DNA level (CNVs or SNVs). Thirty-six studies out of 205 met the inclusion criteria. Cytogenetic abnormalities (n = 72, including 66 CNVs) were identified in 16 autosomes and 2 sex chromosomes (X, Y), some with a higher frequency and clinical significance than others (e.g., 2p16.3, 3q29, 15q13.3, 22q11.21 deletions; 2p25.3, 3p25.3 and 16p11.2 duplications). Thirty-one single nucleotide mutations in genes principally involved in brain development and/or function have been found in 12 autosomes and one sex chromosome (X). We also describe five SNVs in X-linked genes inherited from a healthy mother, arguing for the X-linked recessive inheritance hypothesis. Moreover, ATP1A3 (19q13.2) is the only gene carrying more than one SNV in more than one patient, making it a strong candidate for COS. Mutations were distributed in various chromosomes illustrating the genetic heterogeneity of COS. More than 90% of CNVs involved in COS are also involved in ASD, supporting the idea that there may be genetic overlap between these disorders. Different mutations associated with COS are probably still unknown, and pathogenesis might also be explained by the association of different genetic variations (two or more CNVs or CNVs and SNVs) as well as association with early acquired brain lesions such as infection, hypoxia, or early childhood trauma.
Collapse
Affiliation(s)
- Arnaud Fernandez
- University Department of Child and Adolescent Psychiatry, Children's, Hospitals of NICE CHU-Lenval, Nice, France.,CoBTek, Université Côte d'Azur, Nice, France.,Université Côte d'Azur, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Malgorzata Marta Drozd
- Université Côte d'Azur, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Susanne Thümmler
- University Department of Child and Adolescent Psychiatry, Children's, Hospitals of NICE CHU-Lenval, Nice, France.,CoBTek, Université Côte d'Azur, Nice, France
| | - Emmanuelle Dor
- University Department of Child and Adolescent Psychiatry, Children's, Hospitals of NICE CHU-Lenval, Nice, France.,CoBTek, Université Côte d'Azur, Nice, France
| | - Maria Capovilla
- Université Côte d'Azur, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Florence Askenazy
- University Department of Child and Adolescent Psychiatry, Children's, Hospitals of NICE CHU-Lenval, Nice, France.,CoBTek, Université Côte d'Azur, Nice, France
| | - Barbara Bardoni
- Université Côte d'Azur, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France.,Université Côte d'Azur, INSERM, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| |
Collapse
|
35
|
Neuronal impact of patient-specific aberrant NRXN1α splicing. Nat Genet 2019; 51:1679-1690. [PMID: 31784728 PMCID: PMC7451045 DOI: 10.1038/s41588-019-0539-z] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 10/25/2019] [Indexed: 02/08/2023]
Abstract
NRXN1 undergoes extensive alternative splicing, and non-recurrent heterozygous deletions in NRXN1 are strongly associated with neuropsychiatric disorders. We establish that human induced pluripotent stem cell (hiPSC)-derived neurons represent well the diversity of NRXN1α alternative splicing observed in the human brain, cataloguing 123 high-confidence in-frame human NRXN1α isoforms. Patient-derived NRXN1+/− hiPSC-neurons show greater than two-fold reduction of half of the wild-type NRXN1α isoforms and express dozens of novel isoforms expressed from the mutant allele. Reduced neuronal activity in patient-derived NRXN1+/− hiPSC-neurons is ameliorated by overexpression of individual control isoforms in a genotype-dependent manner, whereas individual mutant isoforms decrease neuronal activity levels in control hiPSC-neurons. In a genotype-dependent manner, the phenotypic impact of patient-specific NRXN1+/− mutations can occur through a reduction in wild-type NRXN1α isoform levels as well as the presence of mutant NRXN1α isoforms.
Collapse
|
36
|
Wang SH, Hsiao PC, Yeh LL, Liu CM, Liu CC, Hwang TJ, Hsieh MH, Chien YL, Lin YT, Huang YT, Chen CY, Chandler SD, Faraone SV, Neale B, Glatt SJ, Tsuang MT, Hwu HG, Chen WJ. Advanced Paternal Age and Early Onset of Schizophrenia in Sporadic Cases: Not Confounded by Parental Polygenic Risk for Schizophrenia. Biol Psychiatry 2019; 86:56-64. [PMID: 30926130 DOI: 10.1016/j.biopsych.2019.01.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 01/24/2019] [Accepted: 01/24/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND Whether paternal age effect on schizophrenia is a causation or just an association due to confounding by selection into late parenthood is still debated. We investigated the association between paternal age and early onset of schizophrenia in offspring, controlling for both paternal and maternal predisposition to schizophrenia as empirically estimated using polygenic risk score (PRS) derived from the Psychiatric Genomics Consortium. METHODS Among 2923 sporadic schizophrenia cases selected from the Schizophrenia Trio Genomic Research in Taiwan project, 1649 had parents' genotyping data. The relationships of paternal schizophrenia PRS to paternal age at first birth (AFB) and of maternal schizophrenia PRS to maternal AFB were examined. A logistic regression model of patients' early onset of schizophrenia (≤18 years old) on paternal age was conducted. RESULTS Advanced paternal age over 20 years exhibited a trend of an increasing proportion of early onset of schizophrenia (odds ratio per 10-year increase in paternal age = 1.28, p = .007) after adjusting for maternal age, sex, and age. Older paternal AFB also exhibited an increasing trend of paternal schizophrenia PRS. Additionally, a U-shaped relationship between maternal AFB and maternal schizophrenia PRS was observed. After adjusting for both paternal and maternal schizophrenia PRS, the association of paternal age with patients' early onset of schizophrenia remained (odds ratio = 1.29, p = .04). CONCLUSIONS The association between paternal age and early onset of schizophrenia was not confounded by parental PRS for schizophrenia, which partially captures parental genetic vulnerability to schizophrenia. Our findings support an independent role of paternal age per se in increased risk of early onset of schizophrenia in offspring.
Collapse
Affiliation(s)
- Shi-Heng Wang
- Departments of Public Health and Occupational Safety and Health, College of Public Health, China Medical University, Taichung, Taiwan
| | - Po-Chang Hsiao
- Institute of Epidemiology and Preventive Medicine, College of Public Health, Taipei, Taiwan
| | - Ling-Ling Yeh
- Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Chih-Min Liu
- Department of Psychiatry, College of Medicine and National Taiwan University Hospital, Taipei, Taiwan
| | - Chen-Chung Liu
- Department of Psychiatry, College of Medicine and National Taiwan University Hospital, Taipei, Taiwan
| | - Tzung-Jeng Hwang
- Department of Psychiatry, College of Medicine and National Taiwan University Hospital, Taipei, Taiwan
| | - Ming H Hsieh
- Department of Psychiatry, College of Medicine and National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Ling Chien
- Department of Psychiatry, College of Medicine and National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Ting Lin
- Department of Psychiatry, College of Medicine and National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Tsung Huang
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Chia-Yen Chen
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Sharon D Chandler
- Center for Behavioral Genomics, Department of Psychiatry and Institute for Genomic Medicine, University of California San Diego, La Jolla, California
| | - Stephen V Faraone
- Departments of Psychiatry and Behavioral Sciences and Neuroscience and Physiology, Medical Genetics Research Center, State University of New York Upstate Medical University, Syracuse, New York
| | - Benjamin Neale
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Stephen J Glatt
- Departments of Psychiatry and Behavioral Sciences and Neuroscience and Physiology, Medical Genetics Research Center, State University of New York Upstate Medical University, Syracuse, New York
| | - Ming T Tsuang
- Center for Behavioral Genomics, Department of Psychiatry and Institute for Genomic Medicine, University of California San Diego, La Jolla, California
| | - Hai-Gwo Hwu
- Department of Psychiatry, College of Medicine and National Taiwan University Hospital, Taipei, Taiwan; Institute of Brain and Mind Sciences, College of Medicine, Taipei, Taiwan
| | - Wei J Chen
- Institute of Epidemiology and Preventive Medicine, College of Public Health, Taipei, Taiwan; Department of Public Health, College of Public Health, Taipei, Taiwan; Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
37
|
Feczko E, Miranda-Dominguez O, Marr M, Graham AM, Nigg JT, Fair DA. The Heterogeneity Problem: Approaches to Identify Psychiatric Subtypes. Trends Cogn Sci 2019; 23:584-601. [PMID: 31153774 PMCID: PMC6821457 DOI: 10.1016/j.tics.2019.03.009] [Citation(s) in RCA: 239] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 03/28/2019] [Accepted: 03/29/2019] [Indexed: 12/12/2022]
Abstract
The imprecise nature of psychiatric nosology restricts progress towards characterizing and treating mental health disorders. One issue is the 'heterogeneity problem': different causal mechanisms may relate to the same disorder, and multiple outcomes of interest can occur within one individual. Our review tackles this heterogeneity problem, providing considerations, concepts, and approaches for investigators examining human cognition and mental health. We highlight the difficulty of pure dimensional approaches due to 'the curse of dimensionality'. Computationally, we consider supervised and unsupervised statistical approaches to identify putative subtypes within a population. However, we emphasize that subtype identification should be linked to a particular outcome or question. We conclude with novel hybrid approaches that can identify subtypes tied to outcomes, and may help advance precision diagnostic and treatment tools.
Collapse
Affiliation(s)
- Eric Feczko
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; Department of Medical Informatics and Clinical Epidemiology Oregon Health & Science University, Portland, OR 97239, USA.
| | - Oscar Miranda-Dominguez
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA
| | - Mollie Marr
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA
| | - Alice M Graham
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; Department of Psychiatry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Joel T Nigg
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; Department of Psychiatry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Damien A Fair
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; Department of Psychiatry, Oregon Health & Science University, Portland, OR 97239, USA; Advanced Imaging Research Center Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
38
|
de Jong S, Diniz MJA, Saloma A, Gadelha A, Santoro ML, Ota VK, Noto C, Curtis C, Newhouse SJ, Patel H, Hall LS, O Reilly PF, Belangero SI, Bressan RA, Breen G. Applying polygenic risk scoring for psychiatric disorders to a large family with bipolar disorder and major depressive disorder. Commun Biol 2018; 1:163. [PMID: 30320231 PMCID: PMC6175827 DOI: 10.1038/s42003-018-0155-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 08/06/2018] [Indexed: 01/03/2023] Open
Abstract
Psychiatric disorders are thought to have a complex genetic pathology consisting of interplay of common and rare variation. Traditionally, pedigrees are used to shed light on the latter only, while here we discuss the application of polygenic risk scores to also highlight patterns of common genetic risk. We analyze polygenic risk scores for psychiatric disorders in a large pedigree (n ~ 260) in which 30% of family members suffer from major depressive disorder or bipolar disorder. Studying patterns of assortative mating and anticipation, it appears increased polygenic risk is contributed by affected individuals who married into the family, resulting in an increasing genetic risk over generations. This may explain the observation of anticipation in mood disorders, whereby onset is earlier and the severity increases over the generations of a family. Joint analyses of rare and common variation may be a powerful way to understand the familial genetics of psychiatric disorders.
Collapse
Grants
- U01 MH109536 NIMH NIH HHS
- R01 MH085548 NIMH NIH HHS
- Wellcome Trust
- G0401207 Medical Research Council
- G0200243 Medical Research Council
- MR/K006584/1 Medical Research Council
- SJN is also supported by the National Institute for Health Research (NIHR) University College London Hospitals Biomedical Research Centre, and by awards establishing the Farr Institute of Health Informatics Research at UCLPartners, from the Medical Research Council, Arthritis Research UK, British Heart Foundation, Cancer Research UK, Chief Scientist Office, Economic and Social Research Council, Engineering and Physical Sciences Research Council, National Institute for Health Research, National Institute for Social Care and Health Research, and Wellcome Trust (grant MR/K006584/1).
- This paper represents independent research part-funded by FAPESP (2014/50830-2; 2010/08968-6), the Marie Curie International Research Staff Exchange (FP7-PEOPLE-2011-IRSES/295192), and the National Institute for Health Research (NIHR) Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London. SDJ is funded by the European Union’s Horizon 2020 research and innovation programme under Marie Skłodowska-Curie grant IF 658195.
Collapse
Affiliation(s)
- Simone de Jong
- MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
- National Institute of Health Research Biomedical Research Centre for Mental Health, Maudsley Hospital and Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Mateus Jose Abdalla Diniz
- Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, 04021-001, Brazil
- Pax Instituto de Psiquiatria, BR153, km 505, Villa Sul V, Aparecida de Goiânia, 74911-516, Brazil
| | - Andiara Saloma
- Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, 04021-001, Brazil
- Pax Instituto de Psiquiatria, BR153, km 505, Villa Sul V, Aparecida de Goiânia, 74911-516, Brazil
| | - Ary Gadelha
- Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, 04021-001, Brazil
| | - Marcos L Santoro
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, 04021-001, Brazil
| | - Vanessa K Ota
- Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, 04021-001, Brazil
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, 04021-001, Brazil
| | - Cristiano Noto
- Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, 04021-001, Brazil
| | - Charles Curtis
- MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
- National Institute of Health Research Biomedical Research Centre for Mental Health, Maudsley Hospital and Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Stephen J Newhouse
- National Institute of Health Research Biomedical Research Centre for Mental Health, Maudsley Hospital and Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
- Farr Institute of Health Informatics Research, UCL Institute of Health Informatics, University College London, London, NW1 2DA, UK
| | - Hamel Patel
- National Institute of Health Research Biomedical Research Centre for Mental Health, Maudsley Hospital and Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Lynsey S Hall
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, CF10 3AT, UK
| | - Paul F O Reilly
- MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Sintia I Belangero
- Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, 04021-001, Brazil
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, 04021-001, Brazil
| | - Rodrigo A Bressan
- Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, 04021-001, Brazil
| | - Gerome Breen
- MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry Psychology and Neuroscience, King's College London, London, SE5 8AF, UK.
- National Institute of Health Research Biomedical Research Centre for Mental Health, Maudsley Hospital and Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK.
| |
Collapse
|
39
|
Abstract
Schizophrenia and other types of psychosis incur suffering, high health care costs and loss of human potential, due to the combination of early onset and poor response to treatment. Our ability to prevent or cure psychosis depends on knowledge of causal mechanisms. Molecular genetic studies show that thousands of common and rare variants contribute to the genetic risk for psychosis. Epidemiological studies have identified many environmental factors associated with increased risk of psychosis. However, no single genetic or environmental factor is sufficient to cause psychosis on its own. The risk of developing psychosis increases with the accumulation of many genetic risk variants and exposures to multiple adverse environmental factors. Additionally, the impact of environmental exposures likely depends on genetic factors, through gene-environment interactions. Only a few specific gene-environment combinations that lead to increased risk of psychosis have been identified to date. An example of replicable gene-environment interaction is a common polymorphism in the AKT1 gene that makes its carriers sensitive to developing psychosis with regular cannabis use. A synthesis of results from twin studies, molecular genetics, and epidemiological research outlines the many genetic and environmental factors contributing to psychosis. The interplay between these factors needs to be considered to draw a complete picture of etiology. To reach a more complete explanation of psychosis that can inform preventive strategies, future research should focus on longitudinal assessments of multiple environmental exposures within large, genotyped cohorts beginning early in life.
Collapse
Affiliation(s)
- Alyson Zwicker
- Department of Pathology,Dalhousie University,Halifax,NS,Canada
| | | | - Rudolf Uher
- Department of Pathology,Dalhousie University,Halifax,NS,Canada
| |
Collapse
|
40
|
Bogdan R, Baranger DAA, Agrawal A. Polygenic Risk Scores in Clinical Psychology: Bridging Genomic Risk to Individual Differences. Annu Rev Clin Psychol 2018; 14:119-157. [PMID: 29579395 PMCID: PMC7772939 DOI: 10.1146/annurev-clinpsy-050817-084847] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genomewide association studies (GWASs) across psychiatric phenotypes have shown that common genetic variants generally confer risk with small effect sizes (odds ratio < 1.1) that additively contribute to polygenic risk. Summary statistics derived from large discovery GWASs can be used to generate polygenic risk scores (PRS) in independent, target data sets to examine correlates of polygenic disorder liability (e.g., does genetic liability to schizophrenia predict cognition?). The intuitive appeal and generalizability of PRS have led to their widespread use and new insights into mechanisms of polygenic liability. However, when currently applied across traits they account for small amounts of variance (<3%), are relatively uninformative for clinical treatment, and, in isolation, provide no insight into molecular mechanisms. Larger GWASs are needed to increase the precision of PRS, and novel approaches integrating various data sources (e.g., multitrait analysis of GWASs) may improve the utility of current PRS.
Collapse
Affiliation(s)
- Ryan Bogdan
- BRAINLab, Department of Psychological and Brain Sciences, and Division of Biology and Biomedical Sciences, Washington University in St. Louis, St. Louis, Missouri 63110, USA;
| | - David A A Baranger
- BRAINLab, Department of Psychological and Brain Sciences, and Division of Biology and Biomedical Sciences, Washington University in St. Louis, St. Louis, Missouri 63110, USA;
| | - Arpana Agrawal
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
41
|
Boies S, Mérette C, Paccalet T, Maziade M, Bureau A. Polygenic risk scores distinguish patients from non-affected adult relatives and from normal controls in schizophrenia and bipolar disorder multi-affected kindreds. Am J Med Genet B Neuropsychiatr Genet 2018; 177:329-336. [PMID: 29193655 DOI: 10.1002/ajmg.b.32614] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 11/13/2017] [Indexed: 11/09/2022]
Abstract
Recent studies have used results on SNP association with schizophrenia (SZ) and bipolar disorder (BD) to create polygenic risk scores (PRS) discriminating non-familial unrelated patients from controls. Little is known about the role of PRS in densely affected multigenerational families. We tested PRS differences between affected SZ and BD family members from their non-affected adult relatives (NAARs) in Eastern Quebec Kindreds and from controls. We examined 1227 subjects: from 17 SZ and BD kindreds, we studied 153 patients (57 SZ, 13 schizoaffective, and 83 BD) and 180 NAARs, and 894 unrelated controls from the Eastern Quebec population. PRS were derived from published case-control association studies of SZ and BD. We also constructed a combined SZ and BD PRS by using SNPs from both SZ and BD PRS. SZ patients had higher SZ PRS than controls (p = 0.0039, R2 = 0.027) and BD patients had higher BD PRS than controls (p = 0.013, R2 = 0.027). Differences between affected subjects and NAARs and controls were significant with both SZ and BD PRS. Moreover, a combined SZ-BD PRS was also significantly associated with SZ and BD when compared to NAARs (p = 0.0019, R2 = 0.010) and controls (p = 0.0025, R2 = 0.028), revealing a SZ-BD commonality effect in PRS at the diagnosis level. The SZ and the BD PRS, however, showed a degree of specificity regarding thought disorder symptoms. Overall, our report would confirm the usefulness of PRS in capturing the contribution of common genetic variants to the risk of SZ and BD in densely affected families.
Collapse
Affiliation(s)
| | - Chantal Mérette
- Centre de Recherche CERVO, Québec, Canada.,Département de Psychiatrie et Neurosciences, Université Laval, Québec, Canada
| | | | - Michel Maziade
- Centre de Recherche CERVO, Québec, Canada.,Département de Psychiatrie et Neurosciences, Université Laval, Québec, Canada
| | - Alexandre Bureau
- Centre de Recherche CERVO, Québec, Canada.,Département de Médecine Sociale et Préventive, Université Laval, Québec, Canada
| |
Collapse
|
42
|
Hoffman GE, Hartley BJ, Flaherty E, Ladran I, Gochman P, Ruderfer DM, Stahl EA, Rapoport J, Sklar P, Brennand KJ. Transcriptional signatures of schizophrenia in hiPSC-derived NPCs and neurons are concordant with post-mortem adult brains. Nat Commun 2017; 8:2225. [PMID: 29263384 PMCID: PMC5738408 DOI: 10.1038/s41467-017-02330-5] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/20/2017] [Indexed: 12/20/2022] Open
Abstract
The power of human induced pluripotent stem cell (hiPSC)-based studies to resolve the smaller effects of common variants within the size of cohorts that can be realistically assembled remains uncertain. We identified and accounted for a variety of technical and biological sources of variation in a large case/control schizophrenia (SZ) hiPSC-derived cohort of neural progenitor cells and neurons. Reducing the stochastic effects of the differentiation process by correcting for cell type composition boosted the SZ signal and increased the concordance with post-mortem data sets. We predict a growing convergence between hiPSC and post-mortem studies as both approaches expand to larger cohort sizes. For studies of complex genetic disorders, to maximize the power of hiPSC cohorts currently feasible, in most cases and whenever possible, we recommend expanding the number of individuals even at the expense of the number of replicate hiPSC clones.
Collapse
Affiliation(s)
- Gabriel E Hoffman
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Brigham J Hartley
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Erin Flaherty
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ian Ladran
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Peter Gochman
- Childhood Psychiatry Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Douglas M Ruderfer
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Division of Genetic Medicine, Departments of Medicine, Psychiatry and Biomedical Informatics, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Eli A Stahl
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Judith Rapoport
- Childhood Psychiatry Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Pamela Sklar
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kristen J Brennand
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW This is an era where we have significantly advanced the understanding of the genetic architecture of schizophrenia. In this review, we consider how this knowledge may translate into advances that will improve patient care. RECENT FINDINGS Large-scale genome-wide association studies (GWAS) have identified more than a hundred loci each making a small contribution to illness risk. Meta-analysis of copy number variants (CNVs) in the Psychiatric Genomics Consortium (PGC) dataset has confirmed that some variants have a moderate or large impact on risk, although these are rare in the population. Genome sequencing advances allow a much more comprehensive evaluation of genomic variation. We describe the key findings from whole exome studies to date. These studies are happening against a backdrop of growing understanding of the regulation and expression of genes and better functional tools to investigate molecular mechanisms in model systems. We provide an overview of how recent approaches in schizophrenia genetics are converging and consider how they could impact on diagnostics, the development of personalized medicine, and drug discovery.
Collapse
Affiliation(s)
- Claire Foley
- Department of Psychiatry and Neuropsychiatric Genetics Research Group, Trinity College Dublin, Dublin, Ireland
| | - Aiden Corvin
- Department of Psychiatry and Neuropsychiatric Genetics Research Group, Trinity College Dublin, Dublin, Ireland.
| | - Shigeki Nakagome
- Department of Psychiatry and Neuropsychiatric Genetics Research Group, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
44
|
Tang J, Fan Y, Li H, Xiang Q, Zhang DF, Li Z, He Y, Liao Y, Wang Y, He F, Zhang F, Shugart YY, Liu C, Tang Y, Chan RCK, Wang CY, Yao YG, Chen X. Whole-genome sequencing of monozygotic twins discordant for schizophrenia indicates multiple genetic risk factors for schizophrenia. J Genet Genomics 2017; 44:295-306. [PMID: 28645778 DOI: 10.1016/j.jgg.2017.05.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/29/2017] [Accepted: 05/09/2017] [Indexed: 12/18/2022]
Abstract
Schizophrenia is a common disorder with a high heritability, but its genetic architecture is still elusive. We implemented whole-genome sequencing (WGS) analysis of 8 families with monozygotic (MZ) twin pairs discordant for schizophrenia to assess potential association of de novo mutations (DNMs) or inherited variants with susceptibility to schizophrenia. Eight non-synonymous DNMs (including one splicing site) were identified and shared by twins, which were either located in previously reported schizophrenia risk genes (p.V24689I mutation in TTN, p.S2506T mutation in GCN1L1, IVS3+1G > T in DOCK1) or had a benign to damaging effect according to in silico prediction analysis. By searching the inherited rare damaging or loss-of-function (LOF) variants and common susceptible alleles from three classes of schizophrenia candidate genes, we were able to distill genetic alterations in several schizophrenia risk genes, including GAD1, PLXNA2, RELN and FEZ1. Four inherited copy number variations (CNVs; including a large deletion at 16p13.11) implicated for schizophrenia were identified in four families, respectively. Most of families carried both missense DNMs and inherited risk variants, which might suggest that DNMs, inherited rare damaging variants and common risk alleles together conferred to schizophrenia susceptibility. Our results support that schizophrenia is caused by a combination of multiple genetic factors, with each DNM/variant showing a relatively small effect size.
Collapse
Affiliation(s)
- Jinsong Tang
- Institute of Mental Health, National Clinical Research Center for Mental Health Disorders and National Technology Institute of Psychiatry, and Key Laboratory of Psychiatry and Mental Health of Hunan Province, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yu Fan
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
| | - Hong Li
- Institute of Mental Health, National Clinical Research Center for Mental Health Disorders and National Technology Institute of Psychiatry, and Key Laboratory of Psychiatry and Mental Health of Hunan Province, The Second Xiangya Hospital, Central South University, Changsha 410011, China; Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qun Xiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Deng-Feng Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
| | - Zongchang Li
- Institute of Mental Health, National Clinical Research Center for Mental Health Disorders and National Technology Institute of Psychiatry, and Key Laboratory of Psychiatry and Mental Health of Hunan Province, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Ying He
- Institute of Mental Health, National Clinical Research Center for Mental Health Disorders and National Technology Institute of Psychiatry, and Key Laboratory of Psychiatry and Mental Health of Hunan Province, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yanhui Liao
- Institute of Mental Health, National Clinical Research Center for Mental Health Disorders and National Technology Institute of Psychiatry, and Key Laboratory of Psychiatry and Mental Health of Hunan Province, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Ya Wang
- Neuropsychology and Applied Cognitive Neuroscience Laboratory, and CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China
| | - Fan He
- Beijing Key Laboratory of Mental Disorders, Department of Psychiatry, Beijing Anding Hospital, and Center of Schizophrenia, Beijing Institute for Brain Disorders and Laboratory of Brain Disorders of the Ministry of Science and Technology, Capital Medical University, Beijing 100088, China
| | - Fengyu Zhang
- Institute of Mental Health, National Clinical Research Center for Mental Health Disorders and National Technology Institute of Psychiatry, and Key Laboratory of Psychiatry and Mental Health of Hunan Province, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yin Yao Shugart
- Unit on Statistical Genomics, Intramural Research Programs, National Institute of Mental Health, NIH, Bethesda 20892, USA
| | - Chunyu Liu
- Institute of Human Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Yanqing Tang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110122, China.
| | - Raymond C K Chan
- Neuropsychology and Applied Cognitive Neuroscience Laboratory, and CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Chuan-Yue Wang
- Beijing Key Laboratory of Mental Disorders, Department of Psychiatry, Beijing Anding Hospital, and Center of Schizophrenia, Beijing Institute for Brain Disorders and Laboratory of Brain Disorders of the Ministry of Science and Technology, Capital Medical University, Beijing 100088, China.
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Xiaogang Chen
- Institute of Mental Health, National Clinical Research Center for Mental Health Disorders and National Technology Institute of Psychiatry, and Key Laboratory of Psychiatry and Mental Health of Hunan Province, The Second Xiangya Hospital, Central South University, Changsha 410011, China.
| |
Collapse
|
45
|
Flaherty EK, Brennand KJ. Using hiPSCs to model neuropsychiatric copy number variations (CNVs) has potential to reveal underlying disease mechanisms. Brain Res 2017; 1655:283-293. [PMID: 26581337 PMCID: PMC4865445 DOI: 10.1016/j.brainres.2015.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 10/16/2015] [Accepted: 11/03/2015] [Indexed: 12/11/2022]
Abstract
Schizophrenia is a neuropsychological disorder with a strong heritable component; genetic risk for schizophrenia is conferred by both common variants of relatively small effect and rare variants with high penetrance. Genetically engineered mouse models can recapitulate rare variants, displaying some behavioral defects associated with schizophrenia; however, these mouse models cannot recapitulate the full genetic architecture underlying the disorder. Patient-derived human induced pluripotent stem cells (hiPSCs) present an alternative approach for studying rare variants, in the context of all other risk alleles. Genome editing technologies, such as CRISPR-Cas9, enable the generation of isogenic hiPSC lines with which to examine the functional contribution of single variants within any genetic background. Studies of these rare variants using hiPSCs have the potential to identify commonly disrupted pathways in schizophrenia and allow for the identification of new therapeutic targets. This article is part of a Special Issue entitled SI:StemsCellsinPsychiatry.
Collapse
Affiliation(s)
- Erin K Flaherty
- Icahn School of Medicine at Mount Sinai, Department of Psychiatry, 1425 Madison Ave, New York, NY 10029, United States
| | - Kristen J Brennand
- Icahn School of Medicine at Mount Sinai, Department of Psychiatry, 1425 Madison Ave, New York, NY 10029, United States.
| |
Collapse
|
46
|
Kinoshita M, Numata S, Tajima A, Nishi A, Muraki S, Tsuchiya A, Umehara H, Watanabe SY, Imoto I, Ohmori T. Cumulative effect of the plasma total homocysteine-related genetic variants on schizophrenia risk. Psychiatry Res 2016; 246:833-837. [PMID: 27810229 DOI: 10.1016/j.psychres.2016.10.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 08/26/2016] [Accepted: 10/15/2016] [Indexed: 10/20/2022]
Abstract
Previous studies suggest that elevated total homocysteine levels and the methylenetetrahydrofolate reductase (MTHFR) C677T polymorphism, which correlates with plasma total homocysteine levels, are risk factors for schizophrenia (SCZ). Recently, a large genome-wide association study (GWAS) of plasma total homocysteine levels in individuals of European ancestry identified many single-nucleotide polymorphisms (SNPs) (n=13,974). The primary purpose of this study was to examine the association between these plasma total homocysteine-related SNPs and SCZ in the Japanese population. First, we investigated associations between six SNPs and plasma total homocysteine levels in non-psychiatric subjects in the Japanese population (n=1030). Then, we evaluated the cumulative effects of three SNPs on SCZ risk by calculating the Genotype Risk Score (GRS) (1120 cases, 2643 controls). Of the six SNPs examined, we replicated similar associations with the European GWAS at four loci (CENPQ, CPS1, MTHFR, and MUT). GRS based on three SNPs (CENPQ, CPS1, and MTHFR) was significantly associated with SCZ. Our findings suggest that common polygenic variations, which are associated with the plasma total homocysteine levels, may contribute to the risk of SCZ.
Collapse
Affiliation(s)
- Makoto Kinoshita
- Department of Psychiatry, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto, Tokushima 770-8503, Japan
| | - Shusuke Numata
- Department of Psychiatry, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto, Tokushima 770-8503, Japan.
| | - Atsushi Tajima
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical, Sciences, Kanazawa University, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8640, Japan; Department of Human Genetics, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto, Tokushima 770-8503, Japan
| | - Akira Nishi
- Department of Psychiatry, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto, Tokushima 770-8503, Japan
| | - Sho Muraki
- Department of Psychiatry, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto, Tokushima 770-8503, Japan
| | - Atsushi Tsuchiya
- Department of Psychiatry, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto, Tokushima 770-8503, Japan
| | - Hidehiro Umehara
- Department of Psychiatry, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto, Tokushima 770-8503, Japan
| | - Shin-Ya Watanabe
- Department of Psychiatry, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto, Tokushima 770-8503, Japan
| | - Issei Imoto
- Department of Human Genetics, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto, Tokushima 770-8503, Japan
| | - Tetsuro Ohmori
- Department of Psychiatry, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto, Tokushima 770-8503, Japan
| |
Collapse
|
47
|
|
48
|
Lancaster TM, Hall J. Altered intra- and inter-network dynamics reflect symptom dimensions in childhood-onset schizophrenia. Brain 2016; 139:10-2. [DOI: 10.1093/brain/awv330] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
49
|
Abstract
Genes account for a significant proportion of the risk for most common diseases. The genome-wide association scan (GWAS) era of genetic epidemiology has generated a massive amount of data, revolutionized our thinking on the genetic architecture of common diseases and positioned the field to realistically consider risk prediction for common polygenic diseases, such as non-familial cancers, and autoimmune, cardiovascular and psychiatric diseases. Polygenic scoring is an approach that shows promise for understanding the polygenic contribution to common human diseases. This is an approach typically relying on genome-wide SNP data, where a set of SNPs identified in a discovery GWAS are used to construct composite polygenic scores. These scores are then used in additional samples for association testing or risk prediction. This review summarizes the extant literature on the use, power, and accuracy of polygenic scores in studies of the etiology of disease and the promise for disease risk prediction.
Collapse
Affiliation(s)
- Brion S Maher
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
50
|
Berman RA, Gotts SJ, McAdams HM, Greenstein D, Lalonde F, Clasen L, Watsky RE, Shora L, Ordonez AE, Raznahan A, Martin A, Gogtay N, Rapoport J. Disrupted sensorimotor and social-cognitive networks underlie symptoms in childhood-onset schizophrenia. Brain 2015; 139:276-91. [PMID: 26493637 DOI: 10.1093/brain/awv306] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/30/2015] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia is increasingly recognized as a neurodevelopmental disorder with altered connectivity among brain networks. In the current study we examined large-scale network interactions in childhood-onset schizophrenia, a severe form of the disease with salient genetic and neurobiological abnormalities. Using a data-driven analysis of resting-state functional magnetic resonance imaging fluctuations, we characterized data from 19 patients with schizophrenia and 26 typically developing controls, group matched for age, sex, handedness, and magnitude of head motion during scanning. This approach identified 26 regions with decreased functional correlations in schizophrenia compared to controls. These regions were found to organize into two function-related networks, the first with regions associated with social and higher-level cognitive processing, and the second with regions involved in somatosensory and motor processing. Analyses of across- and within-network regional interactions revealed pronounced across-network decreases in functional connectivity in the schizophrenia group, as well as a set of across-network relationships with overall negative coupling indicating competitive or opponent network dynamics. Critically, across-network decreases in functional connectivity in schizophrenia predicted the severity of positive symptoms in the disorder, such as hallucinations and delusions. By contrast, decreases in functional connectivity within the social-cognitive network of regions predicted the severity of negative symptoms, such as impoverished speech and flattened affect. These results point toward the role that abnormal integration of sensorimotor and social-cognitive processing may play in the pathophysiology and symptomatology of schizophrenia.
Collapse
Affiliation(s)
- Rebecca A Berman
- 1 Child Psychiatry Branch, National Institute of Mental Health, NIH, Bethesda MD 20892, USA
| | - Stephen J Gotts
- 2 Laboratory of Brain and Cognition, National Institute of Mental Health, NIH, Bethesda MD 20892, USA
| | - Harrison M McAdams
- 1 Child Psychiatry Branch, National Institute of Mental Health, NIH, Bethesda MD 20892, USA
| | - Dede Greenstein
- 1 Child Psychiatry Branch, National Institute of Mental Health, NIH, Bethesda MD 20892, USA
| | - Francois Lalonde
- 1 Child Psychiatry Branch, National Institute of Mental Health, NIH, Bethesda MD 20892, USA
| | - Liv Clasen
- 1 Child Psychiatry Branch, National Institute of Mental Health, NIH, Bethesda MD 20892, USA
| | - Rebecca E Watsky
- 1 Child Psychiatry Branch, National Institute of Mental Health, NIH, Bethesda MD 20892, USA
| | - Lorie Shora
- 1 Child Psychiatry Branch, National Institute of Mental Health, NIH, Bethesda MD 20892, USA
| | - Anna E Ordonez
- 1 Child Psychiatry Branch, National Institute of Mental Health, NIH, Bethesda MD 20892, USA
| | - Armin Raznahan
- 1 Child Psychiatry Branch, National Institute of Mental Health, NIH, Bethesda MD 20892, USA
| | - Alex Martin
- 2 Laboratory of Brain and Cognition, National Institute of Mental Health, NIH, Bethesda MD 20892, USA
| | - Nitin Gogtay
- 1 Child Psychiatry Branch, National Institute of Mental Health, NIH, Bethesda MD 20892, USA
| | - Judith Rapoport
- 1 Child Psychiatry Branch, National Institute of Mental Health, NIH, Bethesda MD 20892, USA
| |
Collapse
|