1
|
Khan MS, Qureshi N, Khan R, Son YO, Maqbool T. CRISPR/Cas9-Based therapeutics as a promising strategy for management of Alzheimer's disease: progress and prospects. Front Cell Neurosci 2025; 19:1578138. [PMID: 40260080 PMCID: PMC12009953 DOI: 10.3389/fncel.2025.1578138] [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: 02/17/2025] [Accepted: 03/20/2025] [Indexed: 04/23/2025] Open
Abstract
CRISPR/Cas9 technology has revolutionized genetic and biomedical research in recent years. It enables editing and modulation of gene function with an unparalleled precision and effectiveness. Among the various applications and prospects of this technology, the opportunities it offers in unraveling the molecular underpinnings of a myriad of central nervous system diseases, including neurodegenerative disorders, psychiatric conditions, and developmental abnormalities, are unprecedented. In this review, we highlight the applications of CRISPR/Cas9-based therapeutics as a promising strategy for management of Alzheimer's disease and transformative impact of this technology on AD research. Further, we emphasize the role of CRISPR/Cas9 in generating accurate AD models for identification of novel therapeutic targets, besides the role of CRISPR-based therapies aimed at correcting AD-associated mutations and modulating the neurodegenerative processes. Furthermore, various delivery systems are reviewed and potential of the non-viral nanotechnology-based carriers for overcoming the critical limitations of effective delivery systems for CRISPR/Cas9 is discussed. Overall, this review highlights the promise and prospects of CRISPR/Cas9 technology for unraveling the intricate molecular processes underlying the development of AD, discusses its limitations, ethical concerns and several challenges including efficient delivery across the BBB, ensuring specificity, avoiding off-target effects. This article can be helpful in better understanding the applications of CRISPR/Cas9 based therapeutic approaches and the way forward utilizing enormous potential of this technology in targeted, gene-specific treatments that could change the trajectory of this debilitating and incurable illness.
Collapse
Affiliation(s)
- Mohamad Sultan Khan
- Laboratory of Nanotherapeutics and Regenerative Medicine, Department of Nanotechnology, University of Kashmir, Srinagar, India
| | - Nousheen Qureshi
- Department of Higher Education, Government of Jammu and Kashmir, Srinagar, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Mohali, Punjab, India
| | - Young-Ok Son
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences and Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, Republic of Korea
| | - Tariq Maqbool
- Laboratory of Nanotherapeutics and Regenerative Medicine, Department of Nanotechnology, University of Kashmir, Srinagar, India
| |
Collapse
|
2
|
Tolmacheva EN, Kashevarova AA, Fonova EA, Salyukova OA, Seitova GN, Nazarenko LP, Agafonova AA, Minaycheva LI, Ravzhaeva EG, Petrova VV, Lopatkina ME, Belyaeva EO, Vovk SL, Fedotov DA, Vasilyeva OY, Skryabin NA, Lebedev IN. Prevalence of CNVs on the X chromosome in patients with neurodevelopmental disorders. Mol Cytogenet 2025; 18:3. [PMID: 39985054 PMCID: PMC11846235 DOI: 10.1186/s13039-025-00703-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 01/19/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND The X chromosome is enriched with genes related to brain development, and the hemizygous state of these genes in men causes some difficulties in the clinical interpretation of copy number variations (CNVs). In this study, we present data on the frequency and spectrum of CNVs on the X chromosome in a cohort of patients with neurodevelopmental disorders (NDDs). METHODS Chromosomal microarray analysis was performed for 1175 patients with NDDs. CNVs were confirmed by real-time quantitative PCR. X chromosome inactivation was analysed by methyl-sensitive PCR. To determine the pathogenic significance of the CNVs, several criteria, including the origin (inherited or de novo), variant type (microdeletion or microduplication), and X chromosome inactivation pattern in asymptomatic and symptomatic carriers, were considered. Additionally, the spectrum, size and molecular bases of copy number changes in genes or gene regions involved in the development of the pathological phenotype in each patient were considered. RESULTS CNVs on the X chromosome were identified in 33 patients (2.8%). Duplications and triplications (27 cases) were four times more common than deletions (6 cases). In 74% of patients, CNVs were of maternal origin; in 10% they were of paternal origin; and in 16% they arose de novo. The frequency of skewed X inactivation among family members who were healthy carriers of pathogenic and likely pathogenic CNVs and variants of uncertain significance (VUSs) on the X chromosome was 23%. For the first time, we reported several CNVs, including a pathogenic microdeletion at Xq26.1q26.2 involving the ARHGAP36 gene and a microduplication at Xp22.2 involving the OFD1 gene, CONCLUSIONS: This study expands on the frequency and spectrum of CNVs in patients with NDDs. Pathogenic variants on the X chromosome were present in 15% of cases, LP in 12%, VUS in 57%, and LB in 16% of cases. Previously unreported CNVs aid in the identification of new structural variants and genes associated with X-linked intellectual disability. We propose to consider the X-chromosome inactivation status when assessing the pathogenetic significance of CNVs using the ACMG algorithm (American College of Medical Genetics).
Collapse
Affiliation(s)
- Ekaterina N Tolmacheva
- Laboratory of Cytogenetics, Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Ushaika Street 10, Tomsk, 634050, Russia.
| | - Anna A Kashevarova
- Laboratory of Cytogenetics, Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Ushaika Street 10, Tomsk, 634050, Russia
| | - Elizaveta A Fonova
- Laboratory of Genomics of Orphan Diseases, Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Olga A Salyukova
- Medical Genetic Center (Genetic Clinic), Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Gulnara N Seitova
- Medical Genetic Center (Genetic Clinic), Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Lyudmila P Nazarenko
- Medical Genetic Center (Genetic Clinic), Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Anna A Agafonova
- Medical Genetic Center (Genetic Clinic), Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Larisa I Minaycheva
- Medical Genetic Center (Genetic Clinic), Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Ekaterina G Ravzhaeva
- Medical Genetic Center (Genetic Clinic), Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Valeria V Petrova
- Medical Genetic Center (Genetic Clinic), Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Maria E Lopatkina
- Laboratory of Ontogenetics, Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Elena O Belyaeva
- Laboratory of Ontogenetics, Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Svetlana L Vovk
- Medical Genetic Center (Genetic Clinic), Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Dmitry A Fedotov
- Laboratory of Ontogenetics, Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Oksana Y Vasilyeva
- Laboratory of Cytogenetics, Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Ushaika Street 10, Tomsk, 634050, Russia
| | - Nikolay A Skryabin
- Laboratory of Genomics of Orphan Diseases, Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Igor N Lebedev
- Laboratory of Ontogenetics, Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, 634050, Russia
| |
Collapse
|
3
|
Ali NMH, Chawner SJRA, Kushan-Wells L, Bearden CE, Mulle JG, Pollak RM, Gur RE, Chung WK, Owen MJ, van den Bree MBM. Comparison of autism domains across thirty rare variant genotypes. EBioMedicine 2025; 112:105521. [PMID: 39891993 PMCID: PMC11835590 DOI: 10.1016/j.ebiom.2024.105521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND A number of Neurodevelopmental risk Copy Number Variants (ND-CNVs) and Single Gene Variants (SGVs) are strongly linked to elevated likelihood of autism. However, few studies have examined the impact on autism phenotypes across a wide range of rare variant genotypes. METHODS This study compared Social Communication Questionnaire (SCQ) scores (total and subdomains: social, communication, repetitive behaviour) in 1314 young people with one of thirty rare variant genotypes (15 ND-CNVs; n = 1005, 9.2 ± 3.5 years and 15 SGVs; n = 309, 8.3 ± 4.0 years). Comparisons were also conducted with young people without known genetic conditions (controls; n = 460, 10.6 ± 3.4 years) and with idiopathic autism (n = 480, 8.6 ± 3.2 years). FINDINGS The prevalence of indicative autism (SCQ ≥ 22) was higher in those with a rare variant genotype compared to controls (32% vs 2%; OR = 43.1, CI = 6.6-282.2, p < 0.001) and in those with SGVs compared to ND-CNVs (53% vs 25%; OR = 4.00, CI = 2.2-7.3, p = 0.002). The prevalence of indicative autism varied considerably across the 30 rare variant genotypes (range 10-85%). SGVs were associated with greater impairment in total, social, communication and repetitive behaviour subdomains than ND-CNVs. However, genotype explained limited variation in these scores (η2 between 11.8 and 21.4%), indicating more convergence than divergence in autism phenotype across rare variant genotypes. Comparisons with young people with idiopathic autism indicated no differences compared to those with ND-CNVs, whereas those with SGVs showed greater communication and less repetitive behaviour. INTERPRETATION The likelihood of autism was higher across all rare variant genotypes, with individuals with SGVs showing higher prevalence and greater impairment compared to those with ND-CNVs. Despite subdomain-specific patterns, there was no strong evidence for specific genotype-phenotype associations. This suggests that rare variant genotypes alone may have limited predictive value for autism phenotypes and that other factors like polygenic risk and the environment are likely to play a role. Further research is needed in order to understand these influences, improve risk prediction and inform genetic counselling and interventions. FUNDING This work was funded by the Tackling Multimorbidity at Scale Strategic Priorities Fund programme (MR/W014416/1) (van den Bree) delivered by the Medical Research Council and the National Institute for Health Research in partnership with the Economic and Social Research Council and in collaboration with the Engineering and Physical Sciences Research Council. NIMH U01 MH119738-01 (van den Bree), IMAGINE study (Medical Research Council UK: MR/T033045/1; MR/N022572/1; and MR/L011166/1) (van den Bree) and Medical Research Council UK Centre Grant (MR/L010305/1) (Owen). SJRAC is funded by a Medical Research Foundation Fellowship (MRF-058-0015-F-CHAW). We would also like to acknowledge NIH 1R01MH110701-01A1 (PI Mulle), U01MH119736 (CEB), R21MH116473 (CEB), and R01MH085953 (CEB), and the Simons Foundation (SFARI Explorer Award to CEB).
Collapse
Affiliation(s)
- Nabila M H Ali
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| | - Samuel J R A Chawner
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| | - Leila Kushan-Wells
- Departments of Psychiatry and Behavioral Sciences and Psychology, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, USA
| | - Carrie E Bearden
- Departments of Psychiatry and Behavioral Sciences and Psychology, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, USA
| | - Jennifer Gladys Mulle
- Department of Psychiatry, Robert Wood Johnson School of Medicine, Rutgers University, USA; Center for Advanced Biotechnology and Medicine, Rutgers University, USA
| | - Rebecca M Pollak
- Department of Psychiatry, Robert Wood Johnson School of Medicine, Rutgers University, USA; Center for Advanced Biotechnology and Medicine, Rutgers University, USA
| | - Raquel E Gur
- Department of Psychiatry, Neurodevelopment & Psychosis Section, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wendy K Chung
- Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael J Owen
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| | - Marianne B M van den Bree
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK.
| |
Collapse
|
4
|
Berardelli I, Cifrodelli M, Sarubbi S, Giuliani C, Antonelli G, Schirripa F, Putotto C, Pulvirenti F, Innamorati M, Pompili M. Investigating psychiatric morbidity, hopelessness and suicide risk in patients with 22q11.2 deletion syndrome: a case-control study. Int J Psychiatry Clin Pract 2024; 28:198-203. [PMID: 39556145 DOI: 10.1080/13651501.2024.2427624] [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: 09/03/2024] [Revised: 10/03/2024] [Accepted: 11/04/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND The psychiatric phenotype of the 22q11.2 deletion syndrome (22q11DS) has been largely described. OBJECTIVES With a case-control study design, we now compared a sample of 22q11DS patients with a psychiatric diagnosis with a sample of psychiatric patients without 22q11DS to investigate possible differences between groups for depression severity, hopelessness, and suicide. Patients with 22q11DS were divided into two groups according to the levels of hopelessness to evaluate the relationship between hopelessness and the severity of the 22q11DS, the level of disability, functional impairment, physical frailty, and autonomy level. RESULTS Results showed that suicide risk evaluated with the C-SSRS was similar in the two groups of patients and that a diagnosis of 22q11DS does not appear to be a risk factor for suicide; however, 22q11DS patients had more severe hopelessness. Patients with a more severe clinical presentation and worse overall functioning have higher levels of depressive symptoms and hopelessness. CONCLUSIONS The results suggest the need to assess and monitor psychiatric symptoms in patients with 22q11DS.
Collapse
Affiliation(s)
- Isabella Berardelli
- Department of Neurosciences, Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Suicide Prevention Centre, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Mariarosaria Cifrodelli
- Psychiatry Residency Training Program, Psychiatry Unit, Faculty of Medicine and Psychology, Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Salvatore Sarubbi
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Carlotta Giuliani
- Psychiatry Residency Training Program, Psychiatry Unit, Faculty of Medicine and Psychology, Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Giulia Antonelli
- Psychiatry Residency Training Program, Psychiatry Unit, Faculty of Medicine and Psychology, Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Fabrizio Schirripa
- Psychiatry Residency Training Program, Psychiatry Unit, Faculty of Medicine and Psychology, Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Carolina Putotto
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Federica Pulvirenti
- Centre for Primary Immune Deficiencies, Azienda Ospedaliera Universitaria Policlinico Umberto I, Rome, Italy
| | - Marco Innamorati
- Department of Human Sciences, European University of Rome, Rome, Italy
| | - Maurizio Pompili
- Department of Neurosciences, Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Suicide Prevention Centre, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
5
|
Forsyth JK, Zhu J, Chavannes AS, Trevorrow ZH, Hyat M, Sievertsen SA, Ferreira-Ianone S, Conomos MP, Nuechterlein KH, Asarnow RF, Green MF, Karlsgodt KH, Perkins DO, Cannon TD, Addington JM, Cadenhead KS, Cornblatt BA, Keshavan MS, Mathalon DH, Stone WS, Tsuang MT, Walker EF, Woods SW, Narr KL, McEwen SC, Schleifer CH, Yee CM, Diehl CK, Guha A, Miller GA, Alexander-Bloch AF, Seidlitz J, Bethlehem RAI, Ophoff RA, Bearden CE. Fetal Gene Regulatory Gene Deletions are Associated with Poor Cognition and Altered Cortical Morphology in Schizophrenia and Community-Based Samples. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.02.24311302. [PMID: 39211869 PMCID: PMC11361264 DOI: 10.1101/2024.08.02.24311302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Schizophrenia spectrum disorders (SSDs) are characterized by substantial clinical and genetic heterogeneity. Multiple recurrent copy number variants (CNVs) increase risk for SSDs; however, how known risk CNVs and broader genome-wide CNVs influence clinical variability is unclear. The current study examined associations between borderline intellectual functioning or childhood-onset psychosis, known risk CNVs, and burden of deletions affecting genes in 18 previously validated neurodevelopmental gene-sets in 618 SSD individuals. CNV associations were assessed for replication in 235 SSD relatives and 583 controls, and 9,930 youth from the Adolescent Brain Cognitive Development (ABCD) Study. Known SSD- and neurodevelopmental disorder (NDD)-risk CNVs were associated with borderline intellectual functioning in SSD cases (odds ratios (OR) = 7.09 and 4.57, respectively); NDD-risk deletions were nominally associated with childhood-onset psychosis (OR = 4.34). Furthermore, deletion of genes involved in regulating gene expression during fetal brain development was associated with borderline intellectual functioning across SSD cases and non-cases (OR = 2.58), with partial replication in the ABCD cohort. Exploratory analyses of cortical morphology showed associations between fetal gene regulatory gene deletions and altered gray matter volume and cortical thickness across cohorts. Results highlight contributions of known risk CNVs to phenotypic variability in SSD and the utility of a neurodevelopmental framework for identifying mechanisms that influence phenotypic variability in SSDs, as well as the broader population, with implications for personalized medicine approaches to care.
Collapse
|
6
|
Zhou B, Purmann C, Guo H, Shin G, Huang Y, Pattni R, Meng Q, Greer SU, Roychowdhury T, Wood RN, Ho M, zu Dohna H, Abyzov A, Hallmayer JF, Wong WH, Ji HP, Urban AE. Resolving the 22q11.2 deletion using CTLR-Seq reveals chromosomal rearrangement mechanisms and individual variance in breakpoints. Proc Natl Acad Sci U S A 2024; 121:e2322834121. [PMID: 39042694 PMCID: PMC11295037 DOI: 10.1073/pnas.2322834121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 06/15/2024] [Indexed: 07/25/2024] Open
Abstract
We developed a generally applicable method, CRISPR/Cas9-targeted long-read sequencing (CTLR-Seq), to resolve, haplotype-specifically, the large and complex regions in the human genome that had been previously impenetrable to sequencing analysis, such as large segmental duplications (SegDups) and their associated genome rearrangements. CTLR-Seq combines in vitro Cas9-mediated cutting of the genome and pulse-field gel electrophoresis to isolate intact large (i.e., up to 2,000 kb) genomic regions that encompass previously unresolvable genomic sequences. These targets are then sequenced (amplification-free) at high on-target coverage using long-read sequencing, allowing for their complete sequence assembly. We applied CTLR-Seq to the SegDup-mediated rearrangements that constitute the boundaries of, and give rise to, the 22q11.2 Deletion Syndrome (22q11DS), the most common human microdeletion disorder. We then performed de novo assembly to resolve, at base-pair resolution, the full sequence rearrangements and exact chromosomal breakpoints of 22q11.2DS (including all common subtypes). Across multiple patients, we found a high degree of variability for both the rearranged SegDup sequences and the exact chromosomal breakpoint locations, which coincide with various transposons within the 22q11.2 SegDups, suggesting that 22q11DS can be driven by transposon-mediated genome recombination. Guided by CTLR-Seq results from two 22q11DS patients, we performed three-dimensional chromosomal folding analysis for the 22q11.2 SegDups from patient-derived neurons and astrocytes and found chromosome interactions anchored within the SegDups to be both cell type-specific and patient-specific. Lastly, we demonstrated that CTLR-Seq enables cell-type specific analysis of DNA methylation patterns within the deletion haplotype of 22q11DS.
Collapse
Affiliation(s)
- Bo Zhou
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA94305
- Stanford Maternal and Child Health Research Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Carolin Purmann
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA94305
- Stanford Maternal and Child Health Research Institute, Stanford University School of Medicine, Stanford, CA94305
- Department of Genetics, Stanford University School of Medicine, Stanford, CA94305
| | - Hanmin Guo
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA94305
- Stanford Maternal and Child Health Research Institute, Stanford University School of Medicine, Stanford, CA94305
- Department of Genetics, Stanford University School of Medicine, Stanford, CA94305
- Department of Statistics, Stanford University, Stanford, CA94305
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA 94305
| | - GiWon Shin
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Yiling Huang
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA94305
- Department of Genetics, Stanford University School of Medicine, Stanford, CA94305
| | - Reenal Pattni
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA94305
- Department of Genetics, Stanford University School of Medicine, Stanford, CA94305
| | - Qingxi Meng
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Stephanie U. Greer
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Tanmoy Roychowdhury
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN55905
| | - Raegan N. Wood
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Marcus Ho
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA94305
- Department of Genetics, Stanford University School of Medicine, Stanford, CA94305
| | - Heinrich zu Dohna
- Department of Biology, American University of Beirut, Beirut1107 2020, Lebanon
| | - Alexej Abyzov
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN55905
| | - Joachim F. Hallmayer
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA94305
| | - Wing H. Wong
- Department of Statistics, Stanford University, Stanford, CA94305
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA 94305
| | - Hanlee P. Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Alexander E. Urban
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA94305
- Stanford Maternal and Child Health Research Institute, Stanford University School of Medicine, Stanford, CA94305
- Department of Genetics, Stanford University School of Medicine, Stanford, CA94305
- Program on Genetics of Brain Function, Stanford Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
7
|
Zaks N, Mahjani B, Reichenberg A, Birnbaum R. CLINICAL AND COGNITIVE PHENOTYPING OF COPY NUMBER VARIANTS PATHOGENIC FOR NEURODEVELOPMENTAL DISORDERS FROM A MULTI-ANCESTRY BIOBANK. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.16.24310489. [PMID: 39072027 PMCID: PMC11275656 DOI: 10.1101/2024.07.16.24310489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Background Rare copy number variants (CNVs) are pathogenic for neurodevelopmental disorders (NDDs) and effect neurocognitive impairment. In aggregate, NDD CNVs may present in up to 2% of population cohorts with implications for neuropsychiatric disease risk and cognitive health. However, analyses of NDD CNVs in biobanks or population cohorts have been hindered by limited clinical or cognitive phenotypes, and a lack of ancestral diversity. In the current proof-of-concept study, NDD CNV carriers were recontacted from BioMe, a multi-ancestry biobank derived from the Mount Sinai healthcare system, to enable 'deep phenotyping' beyond electronic health record outcomes. Methods From BioMe biobank, 892 adult participants were recontacted, including 335 harboring NDD CNVs, 217 with schizophrenia and 340 neurotypical controls as comparators. Clinical and cognitive assessments were administered to each recruited participant. Results Seventy-three participants completed study assessments (mean age=48.8 years; 66% female; 36% African, 26% European, 34% Hispanic), or 8% of the recontacted subset, including 30 NDD CNV carriers across 15 loci. Among NDD CNV carriers, assessments indicated 40% with mood and anxiety disorders, 30% with learning disorders, and 13% with a history of special education. NDD CNV carriers were significantly cognitively impaired compared to controls on digit span backwards (Beta=-1.76, FDR=0.04) and digit span sequencing (Beta=-2.01, FDR=0.04). Conclusions Feasibility of "recall-by-genotype" from a multi-ancestry biobank was established for NDD CNV carriers, along with comparator groups. The current study corroborated past reports of NDD CNVs effects of cognitive impairment, while elucidating clinical phenotypes for recalled individuals. Future "recall-by-genotype" studies may further facilitate clinical characterization of disease-relevant genomic variants.
Collapse
Affiliation(s)
- Nina Zaks
- Department of Child and Adolescent Psychiatry, NYU Langone Health
| | - Behrang Mahjani
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai
| | - Abraham Reichenberg
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai
| | - Rebecca Birnbaum
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai
- Department of Genetic and Genomics Sciences, Icahn School of Medicine at Mount Sinai
| |
Collapse
|
8
|
Leone R, Zuglian C, Brambilla R, Morella I. Understanding copy number variations through their genes: a molecular view on 16p11.2 deletion and duplication syndromes. Front Pharmacol 2024; 15:1407865. [PMID: 38948459 PMCID: PMC11211608 DOI: 10.3389/fphar.2024.1407865] [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: 03/27/2024] [Accepted: 05/16/2024] [Indexed: 07/02/2024] Open
Abstract
Neurodevelopmental disorders (NDDs) include a broad spectrum of pathological conditions that affect >4% of children worldwide, share common features and present a variegated genetic origin. They include clinically defined diseases, such as autism spectrum disorders (ASD), attention-deficit/hyperactivity disorder (ADHD), motor disorders such as Tics and Tourette's syndromes, but also much more heterogeneous conditions like intellectual disability (ID) and epilepsy. Schizophrenia (SCZ) has also recently been proposed to belong to NDDs. Relatively common causes of NDDs are copy number variations (CNVs), characterised by the gain or the loss of a portion of a chromosome. In this review, we focus on deletions and duplications at the 16p11.2 chromosomal region, associated with NDDs, ID, ASD but also epilepsy and SCZ. Some of the core phenotypes presented by human carriers could be recapitulated in animal and cellular models, which also highlighted prominent neurophysiological and signalling alterations underpinning 16p11.2 CNVs-associated phenotypes. In this review, we also provide an overview of the genes within the 16p11.2 locus, including those with partially known or unknown function as well as non-coding RNAs. A particularly interesting interplay was observed between MVP and MAPK3 in modulating some of the pathological phenotypes associated with the 16p11.2 deletion. Elucidating their role in intracellular signalling and their functional links will be a key step to devise novel therapeutic strategies for 16p11.2 CNVs-related syndromes.
Collapse
Affiliation(s)
- Roberta Leone
- Università di Pavia, Dipartimento di Biologia e Biotecnologie “Lazzaro Spallanzani”, Pavia, Italy
| | - Cecilia Zuglian
- Università di Pavia, Dipartimento di Biologia e Biotecnologie “Lazzaro Spallanzani”, Pavia, Italy
| | - Riccardo Brambilla
- Università di Pavia, Dipartimento di Biologia e Biotecnologie “Lazzaro Spallanzani”, Pavia, Italy
- Cardiff University, School of Biosciences, Neuroscience and Mental Health Innovation Institute, Cardiff, United Kingdom
| | - Ilaria Morella
- Cardiff University, School of Biosciences, Neuroscience and Mental Health Innovation Institute, Cardiff, United Kingdom
| |
Collapse
|
9
|
Bamford RA, Zuko A, Eve M, Sprengers JJ, Post H, Taggenbrock RLRE, Fäβler D, Mehr A, Jones OJR, Kudzinskas A, Gandawijaya J, Müller UC, Kas MJH, Burbach JPH, Oguro-Ando A. CNTN4 modulates neural elongation through interplay with APP. Open Biol 2024; 14:240018. [PMID: 38745463 PMCID: PMC11293442 DOI: 10.1098/rsob.240018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 05/16/2024] Open
Abstract
The neuronal cell adhesion molecule contactin-4 (CNTN4) is genetically associated with autism spectrum disorder (ASD) and other psychiatric disorders. Cntn4-deficient mouse models have previously shown that CNTN4 plays important roles in axon guidance and synaptic plasticity in the hippocampus. However, the pathogenesis and functional role of CNTN4 in the cortex has not yet been investigated. Our study found a reduction in cortical thickness in the motor cortex of Cntn4 -/- mice, but cortical cell migration and differentiation were unaffected. Significant morphological changes were observed in neurons in the M1 region of the motor cortex, indicating that CNTN4 is also involved in the morphology and spine density of neurons in the motor cortex. Furthermore, mass spectrometry analysis identified an interaction partner for CNTN4, confirming an interaction between CNTN4 and amyloid-precursor protein (APP). Knockout human cells for CNTN4 and/or APP revealed a relationship between CNTN4 and APP. This study demonstrates that CNTN4 contributes to cortical development and that binding and interplay with APP controls neural elongation. This is an important finding for understanding the physiological function of APP, a key protein for Alzheimer's disease. The binding between CNTN4 and APP, which is involved in neurodevelopment, is essential for healthy nerve outgrowth.
Collapse
Affiliation(s)
- Rosemary A. Bamford
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
| | - Amila Zuko
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Madeline Eve
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
| | - Jan J. Sprengers
- Department of Translational Neuroscience, UMC Utrecht Brain Center, UMC Utrecht, Utrecht3508 AB, The Netherlands
| | - Harm Post
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht, Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Netherlands Proteomics Centre, Utrecht, The Netherlands
| | - Renske L. R. E. Taggenbrock
- Department of Translational Neuroscience, UMC Utrecht Brain Center, UMC Utrecht, Utrecht3508 AB, The Netherlands
| | - Dominique Fäβler
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Functional Genomics, University of Heidelberg, Heidelberg69120, Germany
| | - Annika Mehr
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Functional Genomics, University of Heidelberg, Heidelberg69120, Germany
| | - Owen J. R. Jones
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
| | - Aurimas Kudzinskas
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
| | - Josan Gandawijaya
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
| | - Ulrike C. Müller
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Functional Genomics, University of Heidelberg, Heidelberg69120, Germany
| | - Martien J. H. Kas
- Department of Translational Neuroscience, UMC Utrecht Brain Center, UMC Utrecht, Utrecht3508 AB, The Netherlands
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - J. Peter H. Burbach
- Department of Translational Neuroscience, UMC Utrecht Brain Center, UMC Utrecht, Utrecht3508 AB, The Netherlands
| | - Asami Oguro-Ando
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
- Research Institute for Science and Technology, Tokyo University of Science, Tokyo, Japan
| |
Collapse
|
10
|
Hu X, Wang W, Luo K, Dai J, Zhang Y, Wan Z, He W, Zhang S, Yang L, Tan Q, Li W, Zhang Q, Gong F, Lu G, Tan YQ, Lin G, Du J. Extended application of PGT-M strategies for small pathogenic CNVs. J Assist Reprod Genet 2024; 41:739-750. [PMID: 38263474 PMCID: PMC10957852 DOI: 10.1007/s10815-024-03028-6] [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: 09/28/2023] [Accepted: 01/04/2024] [Indexed: 01/25/2024] Open
Abstract
PURPOSE The preimplantation genetic testing for aneuploidy (PGT-A) platform is not currently available for small copy-number variants (CNVs), especially those < 1 Mb. Through strategies used in PGT for monogenic disease (PGT-M), this study intended to perform PGT for families with small pathogenic CNVs. METHODS Couples who carried small pathogenic CNVs and underwent PGT at the Reproductive and Genetic Hospital of CITIC-Xiangya (Hunan, China) between November 2019 and April 2023 were included in this study. Haplotype analysis was performed through two platforms (targeted sequencing and whole-genome arrays) to identify the unaffected embryos, which were subjected to transplantation. Prenatal diagnosis using amniotic fluid was performed during 18-20 weeks of pregnancy. RESULTS PGT was successfully performed for 20 small CNVs (15 microdeletions and 5 microduplications) in 20 families. These CNVs distributed on chromosomes 1, 2, 6, 7, 13, 15, 16, and X with sizes ranging from 57 to 2120 kb. Three haplotyping-based PGT-M strategies were applied. A total of 89 embryos were identified in 25 PGT cycles for the 20 families. The diagnostic yield was 98.9% (88/89). Nineteen transfers were performed for 17 women, resulting in a 78.9% (15/19) clinical pregnancy rate after each transplantation. Of the nine women who had healthy babies, eight accepted prenatal diagnosis and the results showed no related pathogenic CNVs. CONCLUSION Our results show that the extended haplotyping-based PGT-M strategy application for small pathogenic CNVs compensated for the insufficient resolution of PGT-A. These three PGT-M strategies could be applied to couples with small pathogenic CNVs.
Collapse
Affiliation(s)
- Xiao Hu
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
| | - Weili Wang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
| | - Keli Luo
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
| | - Jing Dai
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
| | - Yi Zhang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
| | - Zhenxing Wan
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
| | - Wenbin He
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
- College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Shuoping Zhang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
| | - Lanlin Yang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
| | - Qin Tan
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
| | - Wen Li
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- College of Life Science, Hunan Normal University, Changsha, 410081, China
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, 410000, China
| | - Qianjun Zhang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
- College of Life Science, Hunan Normal University, Changsha, 410081, China
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, 410000, China
| | - Fei Gong
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- College of Life Science, Hunan Normal University, Changsha, 410081, China
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, 410000, China
| | - Guangxiu Lu
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, 410000, China
| | - Yue-Qiu Tan
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
- College of Life Science, Hunan Normal University, Changsha, 410081, China
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, 410000, China
| | - Ge Lin
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China.
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, 410078, China.
- College of Life Science, Hunan Normal University, Changsha, 410081, China.
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, 410000, China.
| | - Juan Du
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China.
- College of Life Science, Hunan Normal University, Changsha, 410081, China.
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, 410000, China.
| |
Collapse
|
11
|
Colijn MA, Smith CS, Thomas MA. Maternal 15q11.2-q13.1 duplication syndrome-associated psychosis and mania: a new case and review of the literature. Psychiatr Genet 2024; 34:1-7. [PMID: 38019137 DOI: 10.1097/ypg.0000000000000354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Maternal 15q11.2-q13.1 duplication syndrome is associated with a variety of developmental and neuropsychiatric abnormalities. Although schizophrenia-like presentations have been reported, details pertaining to the nature of the corresponding psychotic symptoms and their response to treatment have only been described in a few cases, and no reviews summarizing the literature currently exist. As such, we describe a new case of 15q11.2-q13.1 duplication syndrome-associated schizoaffective disorder and also performed a systematic review of the literature. Our patient's presentation is somewhat unique as she experienced visual hallucinations in the absence of auditory hallucinations. This is also the first report to describe full symptomatic remission in response to relatively low-dose atypical antipsychotic therapy.
Collapse
Affiliation(s)
- Mark Ainsley Colijn
- Department of Psychiatry, Hotchkiss Brain Institute, Mathison Centre for Mental Health Research and Education, University of Calgary
| | - Christopher S Smith
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary
| | - Mary Ann Thomas
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
12
|
Liharska L, Charney A. Transcriptomics : Approaches to Quantifying Gene Expression and Their Application to Studying the Human Brain. Curr Top Behav Neurosci 2024; 68:129-176. [PMID: 38972894 DOI: 10.1007/7854_2024_466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
To date, the field of transcriptomics has been characterized by rapid methods development and technological advancement, with new technologies continuously rendering older ones obsolete.This chapter traces the evolution of approaches to quantifying gene expression and provides an overall view of the current state of the field of transcriptomics, its applications to the study of the human brain, and its place in the broader emerging multiomics landscape.
Collapse
Affiliation(s)
- Lora Liharska
- Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | | |
Collapse
|
13
|
Ormond C, Ryan NM, Heron EA, Gill M, Byerley W, Corvin A. Ultrarare Missense Variants Implicated in Utah Pedigrees Multiply Affected With Schizophrenia. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:797-802. [PMID: 37881554 PMCID: PMC10593875 DOI: 10.1016/j.bpsgos.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 02/17/2023] Open
Abstract
Background Recent work from the Schizophrenia Exome Sequencing Meta-analysis (SCHEMA) consortium showed significant enrichment of ultrarare variants in schizophrenia cases. Family-based studies offer a unique opportunity to evaluate rare variants because risk in multiplex pedigrees is more likely to be influenced by the same collection of variants than an unrelated cohort. Methods Here, we examine whole genome sequencing data from 35 individuals across 6 pedigrees multiply affected by schizophrenia. We applied a rigorous filtering pipeline to search for classes of protein-coding variants that cosegregated with disease status, and we examined these for evidence of enrichment in the SCHEMA dataset. Additionally, we applied a family-based consensus approach to call copy number variants and screen against a list of schizophrenia-associated risk variants. Results We identified deleterious missense variants in 3 genes (ATP2B2, SLC25A28, and GSK3A) that cosegregated with disease in 3 of the pedigrees. In the SCHEMA, the gene ATP2B2 shows highly suggestive evidence for deleterious missense variants in schizophrenia cases (p = .000072). ATP2B2 is involved in intracellular calcium homeostasis, expressed in multiple brain tissue types, and predicted to be intolerant to loss-of-function and missense variants. Conclusions We have identified genes that are likely to increase schizophrenia risk in 3 of the 6 pedigrees examined, the strongest evidence being for a gene involved in calcium homeostasis. Further work is required to examine other classes of variants that may be contributing to disease burden.
Collapse
Affiliation(s)
- Cathal Ormond
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Trinity College Dublin, Dublin, Ireland
| | - Niamh M. Ryan
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Trinity College Dublin, Dublin, Ireland
| | - Elizabeth A. Heron
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Trinity College Dublin, Dublin, Ireland
| | - Michael Gill
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Trinity College Dublin, Dublin, Ireland
| | - William Byerley
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, California
| | - Aiden Corvin
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
14
|
Molloy CJ, Cooke J, Gatford NJF, Rivera-Olvera A, Avazzadeh S, Homberg JR, Grandjean J, Fernandes C, Shen S, Loth E, Srivastava DP, Gallagher L. Bridging the translational gap: what can synaptopathies tell us about autism? Front Mol Neurosci 2023; 16:1191323. [PMID: 37441676 PMCID: PMC10333541 DOI: 10.3389/fnmol.2023.1191323] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/24/2023] [Indexed: 07/15/2023] Open
Abstract
Multiple molecular pathways and cellular processes have been implicated in the neurobiology of autism and other neurodevelopmental conditions. There is a current focus on synaptic gene conditions, or synaptopathies, which refer to clinical conditions associated with rare genetic variants disrupting genes involved in synaptic biology. Synaptopathies are commonly associated with autism and developmental delay and may be associated with a range of other neuropsychiatric outcomes. Altered synaptic biology is suggested by both preclinical and clinical studies in autism based on evidence of differences in early brain structural development and altered glutamatergic and GABAergic neurotransmission potentially perturbing excitatory and inhibitory balance. This review focusses on the NRXN-NLGN-SHANK pathway, which is implicated in the synaptic assembly, trans-synaptic signalling, and synaptic functioning. We provide an overview of the insights from preclinical molecular studies of the pathway. Concentrating on NRXN1 deletion and SHANK3 mutations, we discuss emerging understanding of cellular processes and electrophysiology from induced pluripotent stem cells (iPSC) models derived from individuals with synaptopathies, neuroimaging and behavioural findings in animal models of Nrxn1 and Shank3 synaptic gene conditions, and key findings regarding autism features, brain and behavioural phenotypes from human clinical studies of synaptopathies. The identification of molecular-based biomarkers from preclinical models aims to advance the development of targeted therapeutic treatments. However, it remains challenging to translate preclinical animal models and iPSC studies to interpret human brain development and autism features. We discuss the existing challenges in preclinical and clinical synaptopathy research, and potential solutions to align methodologies across preclinical and clinical research. Bridging the translational gap between preclinical and clinical studies will be necessary to understand biological mechanisms, to identify targeted therapies, and ultimately to progress towards personalised approaches for complex neurodevelopmental conditions such as autism.
Collapse
Affiliation(s)
- Ciara J. Molloy
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Jennifer Cooke
- Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Nicholas J. F. Gatford
- Kavli Institute for Nanoscience Discovery, Nuffield Department of Clinical Neurosciences, University of Oxford, Medical Sciences Division, Oxford, United Kingdom
| | - Alejandro Rivera-Olvera
- Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Sahar Avazzadeh
- Physiology and Cellular Physiology Research Laboratory, CÚRAM SFI Centre for Research in Medical Devices, School of Medicine, Human Biology Building, University of Galway, Galway, Ireland
| | - Judith R. Homberg
- Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Joanes Grandjean
- Physiology and Cellular Physiology Research Laboratory, CÚRAM SFI Centre for Research in Medical Devices, School of Medicine, Human Biology Building, University of Galway, Galway, Ireland
- Department of Medical Imaging, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Cathy Fernandes
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Sanbing Shen
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
- FutureNeuro, The SFI Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons, Dublin, Ireland
| | - Eva Loth
- Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Deepak P. Srivastava
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Louise Gallagher
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
- The Hospital for SickKids, Toronto, ON, Canada
- The Peter Gilgan Centre for Research and Learning, SickKids Research Institute, Toronto, ON, Canada
- The Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
15
|
Molloy CJ, Quigley C, McNicholas Á, Lisanti L, Gallagher L. A review of the cognitive impact of neurodevelopmental and neuropsychiatric associated copy number variants. Transl Psychiatry 2023; 13:116. [PMID: 37031194 PMCID: PMC10082763 DOI: 10.1038/s41398-023-02421-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/10/2023] Open
Abstract
The heritability of intelligence or general cognitive ability is estimated at 41% and 66% in children and adults respectively. Many rare copy number variants are associated with neurodevelopmental and neuropsychiatric conditions (ND-CNV), including schizophrenia and autism spectrum disorders, and may contribute to the observed variability in cognitive ability. Here, we reviewed studies of intelligence quotient or cognitive function in ND-CNV carriers, from both general population and clinical cohorts, to understand the cognitive impact of ND-CNV in both contexts and identify potential genotype-specific cognitive phenotypes. We reviewed aggregate studies of sets ND-CNV broadly linked to neurodevelopmental and neuropsychiatric conditions, and genotype-first studies of a subset of 12 ND-CNV robustly associated with schizophrenia and autism. Cognitive impacts were observed across ND-CNV in both general population and clinical cohorts, with reports of phenotypic heterogeneity. Evidence for ND-CNV-specific impacts were limited by a small number of studies and samples sizes. A comprehensive understanding of the cognitive impact of ND-CNVs would be clinically informative and could identify potential educational needs for ND-CNV carriers. This could improve genetic counselling for families impacted by ND-CNV, and clinical outcomes for those with complex needs.
Collapse
Affiliation(s)
- Ciara J Molloy
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland.
- Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland.
| | - Ciara Quigley
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland
| | - Áine McNicholas
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland
| | - Linda Lisanti
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland
| | - Louise Gallagher
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland
- The Hospital for SickKids, Toronto, ON, Canada
- The Peter Gilgan Centre for Research and Learning, SickKids Research Institute, SickKids Research Institute, Toronto, ON, Canada
- The Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
16
|
Mollon J, Almasy L, Jacquemont S, Glahn DC. The contribution of copy number variants to psychiatric symptoms and cognitive ability. Mol Psychiatry 2023; 28:1480-1493. [PMID: 36737482 PMCID: PMC10213133 DOI: 10.1038/s41380-023-01978-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 02/05/2023]
Abstract
Copy number variants (CNVs) are deletions and duplications of DNA sequence. The most frequently studied CNVs, which are described in this review, are recurrent CNVs that occur in the same locations on the genome. These CNVs have been strongly implicated in neurodevelopmental disorders, namely autism spectrum disorder (ASD), intellectual disability (ID), and developmental delay (DD), but also in schizophrenia. More recent work has also shown that CNVs increase risk for other psychiatric disorders, namely, depression, bipolar disorder, and post-traumatic stress disorder. Many of the same CNVs are implicated across all of these disorders, and these neuropsychiatric CNVs are also associated with cognitive ability in the general population, as well as with structural and functional brain alterations. Neuropsychiatric CNVs also show incomplete penetrance, such that carriers do not always develop any psychiatric disorder, and may show only mild symptoms, if any. Variable expressivity, whereby the same CNVs are associated with many different phenotypes of varied severity, also points to highly complex mechanisms underlying disease risk in CNV carriers. Comprehensive and longitudinal phenotyping studies of individual CNVs have provided initial insights into these mechanisms. However, more work is needed to estimate and predict the effect of non-recurrent, ultra-rare CNVs, which also contribute to psychiatric and cognitive outcomes. Moreover, delineating the broader phenotypic landscape of neuropsychiatric CNVs in both clinical and general population cohorts may also offer important mechanistic insights.
Collapse
Affiliation(s)
- Josephine Mollon
- Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Laura Almasy
- Department of Genetics, Perelman School of Medicine, Penn-CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Sebastien Jacquemont
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
- Center Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada
| | - David C Glahn
- Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Olin Neuropsychiatry Research Center, Institute of Living, Hartford, CT, USA
| |
Collapse
|
17
|
Wahbeh MH, Peng X, Bacharaki S, Hatzimanolis A, Dimitrakopoulos S, Wohler E, Yang X, Yovo C, Maher BJ, Sobreira N, Stefanis NC, Avramopoulos D. A Missense Variant in CASKIN1's Proline-Rich Region Segregates with Psychosis in a Three-Generation Family. Genes (Basel) 2023; 14:177. [PMID: 36672919 PMCID: PMC9859343 DOI: 10.3390/genes14010177] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
The polygenic nature of schizophrenia (SCZ) implicates many variants in disease development. Rare variants of high penetrance have been shown to contribute to the disease prevalence. Whole-exome sequencing of a large three-generation family with SCZ and bipolar disorder identified a single segregating novel, rare, non-synonymous variant in the gene CASKIN1. The variant D1204N is absent from all databases, and CASKIN1 has a gnomAD missense score Z = 1.79 and pLI = 1, indicating its strong intolerance to variation. We find that introducing variants in the proline-rich region where the D1204N resides results in significant cellular changes in iPSC-derived neurons, consistent with CASKIN1’s known functions. We observe significant transcriptomic changes in 368 genes (padj < 0.05) involved in neuronal differentiation and nervous system development. We also observed nominally significant changes in the frequency of action potentials during differentiation, where the speed at which the edited and unedited cells reach the same level of activity differs. Our results suggest that CASKIN1 is an excellent gene candidate for psychosis development with high penetrance in this family.
Collapse
Affiliation(s)
- Marah H. Wahbeh
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Predoctoral Training Program in Human Genetics and Molecular Biology, Johns Hopkins School of Medicine, Baltimore, MD 21201, USA
| | - Xi Peng
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Sofia Bacharaki
- Department of Psychiatry, General Hospital of Syros, 84100 Cyclades, Greece
| | - Alexandros Hatzimanolis
- Department of Psychiatry, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, 15772 Athens, Greece
| | - Stefanos Dimitrakopoulos
- Department of Psychiatry, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, 15772 Athens, Greece
| | - Elizabeth Wohler
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Xue Yang
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Christian Yovo
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Brady J. Maher
- Lieber Institute for Brain Development, Baltimore, MD 21205, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Nara Sobreira
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Nikos C. Stefanis
- Department of Psychiatry, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, 15772 Athens, Greece
| | - Dimitrios Avramopoulos
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
18
|
White LK, Crowley TB, Finucane B, McClellan EJ, Donoghue S, Garcia-Minaur S, Repetto GM, Fischer M, Jacquemont S, Gur RE, Maillard AM, Donald KA, Bassett AS, Swillen A, McDonald-McGinn DM. Gathering the Stakeholder's Perspective: Experiences and Opportunities in Rare Genetic Disease Research. Genes (Basel) 2023; 14:169. [PMID: 36672911 PMCID: PMC9859499 DOI: 10.3390/genes14010169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/22/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Research participant feedback is rarely collected; therefore, investigators have limited understanding regarding stakeholders' (affected individuals/caregivers) motivation to participate. Members of the Genes to Mental Health Network (G2MH) surveyed stakeholders affected by copy number variants (CNVs) regarding perceived incentives for study participation, opinions concerning research priorities, and the necessity for future funding. Respondents were also asked about feelings of preparedness, research burden, and satisfaction with research study participation. METHODS Modified validated surveys were used to assess stakeholders´ views across three domains: (1) Research Study Enrollment, Retainment, Withdrawal, and Future Participation; (2) Overall Research Experience, Burden, and Preparedness; (3) Research Priorities and Obstacles. Top box score analyses were performed. RESULTS A total of 704 stakeholders´ responded from 29 countries representing 55 CNVs. The top reasons for initial participation in the research included reasons related to education and altruism. The top reasons for leaving a research study included treatment risks and side effects. The importance of sharing research findings and laboratory results with stakeholders was underscored by participants. Most stakeholders reported positive research experiences. CONCLUSIONS This study provides important insight into how individuals and families affected with a rare CNV feel toward research participation and their overall experience in rare disease research. There are clear targets for areas of improvement for study teams, although many stakeholders reported positive research experiences. Key findings from this international survey may help advance collaborative research and improve the experience of participants, investigators, and other stakeholders moving forward.
Collapse
Affiliation(s)
- Lauren K. White
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | - Emily J. McClellan
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah Donoghue
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sixto Garcia-Minaur
- Institute of Medical and Molecular Genetics (INGEMM), La Paz University Hospital, 28046 Madrid, Spain
| | | | - Matthias Fischer
- Clinic and Policlinic for Psychiatry and Psychotherapy, University of Rostock, 18147 Rostock, Germany
- Sigma-Zentrum, 79713 Bad Säckingen, Germany
| | - Sebastien Jacquemont
- Sainte Justine Research Center, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Raquel E. Gur
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Kirsten A. Donald
- Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, Rondebosch, Cape Town 7700, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town 7935, South Africa
| | - Anne S. Bassett
- The Dalglish Family 22q Clinic, University Health Network, Toronto, ON M5G 2C4, Canada
- Clinical Genetics Research Program and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, and Department of Psychiatry, University of Toronto, Toronto, ON M5S 2S1, Canada
- Division of Cardiology, Department of Medicine, and Centre for Mental Health, and Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2N2, Canada
| | - Ann Swillen
- Center for Human Genetics, University Hospital UZ Leuven, and Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Donna M. McDonald-McGinn
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Human Biology and Medical Genetics, Sapienza University, 00185 Roma, Italy
| |
Collapse
|
19
|
Brownstein CA, Douard E, Mollon J, Smith R, Hojlo MA, Das A, Goldman M, Garvey E, Cabral K, Li J, Bowen J, Rao AS, Genetti C, Carroll D, Knowles EEM, Deaso E, Agrawal PB, Beggs AH, D'Angelo E, Almasy L, Alexander-Bloch A, Saci Z, Moreau CA, Huguet G, Deo AJ, Jacquemont S, Glahn DC, Gonzalez-Heydrich J. Similar Rates of Deleterious Copy Number Variants in Early-Onset Psychosis and Autism Spectrum Disorder. Am J Psychiatry 2022; 179:853-861. [PMID: 36000218 PMCID: PMC9633349 DOI: 10.1176/appi.ajp.21111175] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Copy number variants (CNVs) are strongly associated with neurodevelopmental and psychotic disorders. Early-onset psychosis (EOP), where symptoms appear before 18 years of age, is thought to be more strongly influenced by genetic factors than adult-onset psychotic disorders. However, the prevalence and effect of CNVs in EOP is unclear. METHODS The authors documented the prevalence of recurrent CNVs and the functional impact of deletions and duplications genome-wide in 137 children and adolescents with EOP compared with 5,540 individuals with autism spectrum disorder (ASD) and 16,504 population control subjects. Specifically, the frequency of 47 recurrent CNVs previously associated with neurodevelopmental and neuropsychiatric illnesses in each cohort were compared. Next, CNV risk scores (CRSs), indices reflecting the dosage sensitivity for any gene across the genome that is encapsulated in a deletion or duplication separately, were compared between groups. RESULTS The prevalence of recurrent CNVs was significantly higher in the EOP group than in the ASD (odds ratio=2.30) and control (odds ratio=5.06) groups. However, the difference between the EOP and ASD groups was attenuated when EOP participants with co-occurring ASD were excluded. CRS was significantly higher in the EOP group compared with the control group for both deletions (odds ratio=1.30) and duplications (odds ratio=1.09). In contrast, the EOP and ASD groups did not differ significantly in terms of CRS. CONCLUSIONS Given the high frequency of recurrent CNVs in the EOP group and comparable CRSs in the EOP and ASD groups, the findings suggest that all children and adolescents with a psychotic diagnosis should undergo genetic screening, as is recommended in ASD.
Collapse
Affiliation(s)
- Catherine A Brownstein
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Elise Douard
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Josephine Mollon
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Richard Smith
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Margaret A Hojlo
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Ananth Das
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Maria Goldman
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Emily Garvey
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Kristin Cabral
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Jianqiao Li
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Joshua Bowen
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Abhijit S Rao
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Casie Genetti
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Devon Carroll
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Emma E M Knowles
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Emma Deaso
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Pankaj B Agrawal
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Alan H Beggs
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Eugene D'Angelo
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Laura Almasy
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Aaron Alexander-Bloch
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Zohra Saci
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Clara A Moreau
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Guillaume Huguet
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Anthony J Deo
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Sébastien Jacquemont
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - David C Glahn
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| | - Joseph Gonzalez-Heydrich
- Early Psychosis Investigation Center (Brownstein, Mollon, Smith, Hojlo, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Division of Genetics and Genomics (Brownstein, Smith, Cabral, Li, Bowen, Rao, Genetti, Agrawal, Beggs, Glahn), Manton Center for Orphan Disease Research (Brownstein, Smith, Cabral, Li, Bowen, Genetti, Agrawal, Beggs, Glahn), Department of Psychiatry and Behavioral Sciences (Mollon, Hojlo, Das, Goldman, Garvey, Carroll, Knowles, Deaso, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Tommy Fuss Center for Neuropsychiatric Disease Research (Deo, Glahn, Gonzalez-Heydrich), and Division of Newborn Medicine (Agrawal), Boston Children's Hospital, Boston; Department of Pediatrics (Brownstein, Smith, Genetti, Agrawal, Beggs, Deo) and Department of Psychiatry (Mollon, Carroll, Knowles, D'Angelo, Deo, Glahn, Gonzalez-Heydrich), Harvard Medical School, Boston; Department of Pediatrics (Jacquemont) and Department of Neuroscience (Douard, Moreau), Université de Montréal, Montreal; Sainte-Justine Hospital Research Center, Montreal (Douard, Saci, Moreau, Huguet, Jacquemont); Department of Biomedical and Health Informatics (Almasy) and Department of Psychiatry (Alexander-Bloch), Children's Hospital of Philadelphia, Philadelphia; Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, N.J. (Deo); Rutgers University Behavioral Health Care, Piscataway, N.J. (Deo). Lifespan Brain Institute, Children's Hospital of Philadelphia, Philadelphia (Almasy); Department of Genetics, University of Pennsylvania, Philadelphia (Almasy); Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago (Smith)
| |
Collapse
|
20
|
The Roles of Par3, Par6, and aPKC Polarity Proteins in Normal Neurodevelopment and in Neurodegenerative and Neuropsychiatric Disorders. J Neurosci 2022; 42:4774-4793. [PMID: 35705493 DOI: 10.1523/jneurosci.0059-22.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/30/2022] [Accepted: 05/02/2022] [Indexed: 11/21/2022] Open
Abstract
Normal neural circuits and functions depend on proper neuronal differentiation, migration, synaptic plasticity, and maintenance. Abnormalities in these processes underlie various neurodevelopmental, neuropsychiatric, and neurodegenerative disorders. Neural development and maintenance are regulated by many proteins. Among them are Par3, Par6 (partitioning defective 3 and 6), and aPKC (atypical protein kinase C) families of evolutionarily conserved polarity proteins. These proteins perform versatile functions by forming tripartite or other combinations of protein complexes, which hereafter are collectively referred to as "Par complexes." In this review, we summarize the major findings on their biophysical and biochemical properties in cell polarization and signaling pathways. We next summarize their expression and localization in the nervous system as well as their versatile functions in various aspects of neurodevelopment, including neuroepithelial polarity, neurogenesis, neuronal migration, neurite differentiation, synaptic plasticity, and memory. These versatile functions rely on the fundamental roles of Par complexes in cell polarity in distinct cellular contexts. We also discuss how cell polarization may correlate with subcellular polarization in neurons. Finally, we review the involvement of Par complexes in neuropsychiatric and neurodegenerative disorders, such as schizophrenia and Alzheimer's disease. While emerging evidence indicates that Par complexes are essential for proper neural development and maintenance, many questions on their in vivo functions have yet to be answered. Thus, Par3, Par6, and aPKC continue to be important research topics to advance neuroscience.
Collapse
|
21
|
Murray RM, Bora E, Modinos G, Vernon A. Schizophrenia: A developmental disorder with a risk of non-specific but avoidable decline. Schizophr Res 2022; 243:181-186. [PMID: 35390609 DOI: 10.1016/j.schres.2022.03.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 12/31/2022]
Abstract
The onset of schizophrenia is determined by biological and social risk factors operating predominantly during development. These result in subtle deviations in brain structure and cognitive function. Striatal dopamine dysregulation follows, causing abnormal salience and resultant psychotic symptoms. Most people diagnosed as having schizophrenia do not progressively deteriorate; many improve or recover. However, poor care can allow a cycle of deterioration to be established, stress increasing dopamine dysregulation, leading to more stress consequent on continuing psychotic experiences, and so further dopamine release. Additionally, long-term antipsychotics can induce dopamine supersensitivity with resultant relapse and eventually treatment resistance. Some patients suffer loss of social and cognitive function, but this is a consequence of the hazards that afflict the person with schizophrenia, not a direct consequence of genetic predisposition. Thus, brain health and cognition can be further impaired by chronic medication effects, cardiovascular and cerebrovascular events, obesity, poor diet, and lack of exercise; drug use, especially of tobacco and cannabis, are likely to contribute. Poverty, homelessness and poor nutrition which become the lot of some people with schizophrenia, can also affect cognition. Regrettably, the model of progressive deterioration provides psychiatry and its funders with an alibi for the effects of poor care.
Collapse
Affiliation(s)
- R M Murray
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom.
| | - E Bora
- Dokuz Eylül Üniversitesi, Izmir, Izmir, Turkey
| | - G Modinos
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom
| | - A Vernon
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom
| |
Collapse
|
22
|
The Kraepelian concept of schizophrenia: Dying but not yet dead. Schizophr Res 2022; 242:102-105. [PMID: 34952779 DOI: 10.1016/j.schres.2021.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/05/2021] [Accepted: 12/06/2021] [Indexed: 12/16/2022]
|
23
|
White LK, Crowley TB, Finucane B, Garcia-Minaur S, Repetto GM, van den Bree M, Fischer M, Jacquemont S, Barzilay R, Maillard AM, Donald KA, Gur RE, Bassett AS, Swillen A, McDonald-McGinn DM. The COVID-19 pandemic's impact on worry and medical disruptions reported by individuals with chromosome 22q11.2 copy number variants and their caregivers. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2022; 66:313-322. [PMID: 35191118 PMCID: PMC9725107 DOI: 10.1111/jir.12918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 01/04/2022] [Accepted: 01/09/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND The world has suffered immeasurably during the COVID-19 pandemic. Increased distress and mental and medical health concerns are collateral consequences to the disease itself. The Genes to Mental Health (G2MH) Network consortium sought to understand how individuals affected by the rare copy number variations of 22q11.2 deletion and duplication syndrome, associated with neurodevelopmental/neuropsychiatric conditions, were coping. The article focuses on worry and disruptions in medical care caused by the pandemic. METHODS The University of Pennsylvania COVID-19 Stressor List and care disruption questions were circulated by 22 advocacy groups in English and 11 other languages. RESULTS A total of 512 people from 23 countries completed the survey; most were caregivers of affected individuals. Worry about family members acquiring COVID-19 had the highest average endorsed worry, whilst currently having COVID-19 had the lowest rated worry. Total COVID-19 worries were higher in individuals completing the survey towards the end of the study (later pandemic wave); 36% (n = 186) of the sample reported a significant effect on health due to care interruption during the pandemic; 44% of individuals (n = 111) receiving care for their genetic syndrome in a hospital setting reported delaying appointments due to COVID-19 fears; 12% (n = 59) of the sample reported disruptions to treatments; and of those reporting no current disruptions, 59% (n = 269) worried about future disruptions if the pandemic continued. Higher levels of care disruptions were related to higher COVID-19 worries (Ps < 0.005). Minimal differences by respondent type or copy number variation type emerged. CONCLUSIONS Widespread medical care disruptions and pandemic-related worries were reported by individuals with 22q11.2 syndrome and their family members. Reported worries were broadly consistent with research results from prior reports in the general population. The long-term effects of COVID-19 worries, interruptions to care and hospital avoidance require further study.
Collapse
Affiliation(s)
- L K White
- Lifespan Brain Institute, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - T B Crowley
- Lifespan Brain Institute, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - B Finucane
- Geisinger Autism & Developmental Medicine Institute, Geisinger Health System, Lewisburg, PA, USA
| | - S Garcia-Minaur
- Instituto de Genética Médica y Molecular, Hospital Universitario La Paz, Madrid, Spain
| | - G M Repetto
- Center for Genetics and Genomics, Facultad de Medicina Clínica Alemana - Universidad del Desarrollo, Santiago, Chile
| | - M van den Bree
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - M Fischer
- Clinic and Policlinic for Psychiatry and Psychotherapy, University of Rostock, Rostock, Germany
| | - S Jacquemont
- Sainte Justine Research Center, University of Montreal, Montreal, Canada
| | - R Barzilay
- Lifespan Brain Institute, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - A M Maillard
- Service des Troubles du Spectre de l'Autisme (STSA), Lausanne University Hospital, Lausanne, Switzerland
| | - K A Donald
- Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa
| | - R E Gur
- Lifespan Brain Institute, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - A S Bassett
- Centre for Addiction and Mental Health, University Health Network and Department of Psychiatry, University of Toronto, Toronto, Canada
| | - A Swillen
- Center for Human Genetics, University Hospital Leuven and Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - D M McDonald-McGinn
- Lifespan Brain Institute, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
24
|
Hayashi Y, Kushima I, Aleksic B, Senaha T, Ozaki N. Variable psychiatric manifestations in patients with 16p11.2 duplication: a case series of 4 patients. Psychiatry Clin Neurosci 2022; 76:86-88. [PMID: 34940990 DOI: 10.1111/pcn.13324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/13/2021] [Accepted: 12/13/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Yu Hayashi
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Itaru Kushima
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Medical Genomics Center, Nagoya University Hospital, Nagoya, Japan
| | - Branko Aleksic
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsu Senaha
- Department of Psychiatry, KACHI Memorial Hospital, Toyohashi, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
25
|
Birnbaum R, Mahjani B, Loos RJF, Sharp AJ. Clinical Characterization of Copy Number Variants Associated With Neurodevelopmental Disorders in a Large-scale Multiancestry Biobank. JAMA Psychiatry 2022; 79:250-259. [PMID: 35080590 PMCID: PMC8792794 DOI: 10.1001/jamapsychiatry.2021.4080] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/30/2021] [Indexed: 01/28/2023]
Abstract
IMPORTANCE Past studies identified rare copy number variants (CNVs) as risk factors for neurodevelopmental disorders (NDDs), including autism spectrum disorder and schizophrenia. However, the clinical characterization of NDD CNVs is understudied in population cohorts unselected for neuropsychiatric disorders and in cohorts of diverse ancestry. OBJECTIVE To identify individuals harboring NDD CNVs in a multiancestry biobank and to query their enrichment for select neuropsychiatric disorders as well as association with multiple medical disorders. DESIGN, SETTINGS, AND PARTICIPANTS In a series of phenotypic enrichment and association analyses, NDD CNVs were clinically characterized among 24 877 participants in the BioMe biobank, an electronic health record-linked biobank derived from the Mount Sinai Health System, New York, New York. Participants were recruited into the biobank since September 2007 across diverse ancestry and medical and neuropsychiatric specialties. For the current analyses, electronic health record data were analyzed from May 2004 through May 2019. MAIN OUTCOMES AND MEASURES NDD CNVs were identified using a consensus of 2 CNV calling algorithms, based on whole-exome sequencing and genotype array data, followed by novel in-silico clinical assessments. RESULTS Of 24 877 participants, 14 586 (58.7%) were female; self-reported ancestry categories included 5965 (24.0%) who were of African ancestry, 7892 (31.7%) who were of European ancestry, and 8536 (34.3%) who were of Hispanic ancestry; and the mean (SD) age was 50.5 (17.3) years. Among 24 877 individuals, the prevalence of 64 NDD CNVs was 2.5% (n = 627), with prevalence varying by locus, corroborating the presence of some relatively highly prevalent NDD CNVs (eg, 15q11.2 deletion/duplication). An aggregate set of NDD CNVs were enriched for congenital disorders (odds ratio, 2.0; 95% CI, 1.1-3.5; P = .01) and major depressive disorder (odds ratio, 1.5; 95% CI, 1.1-2.0; P = .01). In a meta-analysis of medical diagnoses (n = 195 hierarchically clustered diagnostic codes), NDD CNVs were significantly associated with several medical outcomes, including essential hypertension (z score = 3.6; P = 2.8 × 10-4), kidney failure (z score = 3.3; P = 1.1 × 10-3), and obstructive sleep apnea (z score = 3.4; P = 8.1 × 10-4) and, in another analysis, morbid obesity (z score = 3.8; P = 1.3 × 10-4). Further, NDD CNVs were associated with increased body mass index in a multiancestry analysis (β = 0.19; 95% CI, 0.10-0.31; P = .003). For 36 common serum tests, there was no association with NDD CNVs. CONCLUSIONS AND RELEVANCE Clinical features of individuals harboring NDD CNVs were elucidated in a large-scale, multiancestry biobank, identifying enrichments for congenital disorders and major depressive disorder as well as associations with several medical outcomes, including hypertension, kidney failure, and obesity and obesity-related phenotypes, specifically obstructive sleep apnea and increased body mass index. The association between NDD CNVs and obesity outcomes indicate further potential pleiotropy of NDD CNVs beyond neurodevelopmental outcomes previously reported. Future clinical genetic investigations may lead to insights of at-risk individuals and therapeutic strategies targeting specific genetic variants. The importance of diverse inclusion within biobanks and considering the effect of rare genetic variants in a multiancestry context is evident.
Collapse
Affiliation(s)
- Rebecca Birnbaum
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Behrang Mahjani
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ruth J. F. Loos
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- NovoNordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Andrew J. Sharp
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
26
|
Using induced pluripotent stem cells to investigate human neuronal phenotypes in 1q21.1 deletion and duplication syndrome. Mol Psychiatry 2022; 27:819-830. [PMID: 34112971 PMCID: PMC9054650 DOI: 10.1038/s41380-021-01182-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/17/2021] [Accepted: 05/27/2021] [Indexed: 01/08/2023]
Abstract
Copy Number Variation (CNV) at the 1q21.1 locus is associated with a range of neurodevelopmental and psychiatric disorders in humans, including abnormalities in head size and motor deficits. Yet, the functional consequences of these CNVs (both deletion and duplication) on neuronal development remain unknown. To determine the impact of CNV at the 1q21.1 locus on neuronal development, we generated induced pluripotent stem cells from individuals harbouring 1q21.1 deletion or duplication and differentiated them into functional cortical neurons. We show that neurons with 1q21.1 deletion or duplication display reciprocal phenotype with respect to proliferation, differentiation potential, neuronal maturation, synaptic density and functional activity. Deletion of the 1q21.1 locus was also associated with an increased expression of lower cortical layer markers. This difference was conserved in the mouse model of 1q21.1 deletion, which displayed altered corticogenesis. Importantly, we show that neurons with 1q21.1 deletion and duplication are associated with differential expression of calcium channels and demonstrate that physiological deficits in neurons with 1q21.1 deletion or duplication can be pharmacologically modulated by targeting Ca2+ channel activity. These findings provide biological insight into the neuropathological mechanism underlying 1q21.1 associated brain disorder and indicate a potential target for therapeutic interventions.
Collapse
|
27
|
Calle Sánchez X, Helenius D, Bybjerg-Grauholm J, Pedersen C, Hougaard DM, Børglum AD, Nordentoft M, Mors O, Mortensen PB, Geschwind DH, Montalbano S, Raznahan A, Thompson WK, Ingason A, Werge T. Comparing Copy Number Variations in a Danish Case Cohort of Individuals With Psychiatric Disorders. JAMA Psychiatry 2022; 79:59-69. [PMID: 34817560 PMCID: PMC8733851 DOI: 10.1001/jamapsychiatry.2021.3392] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Importance Although the association between several recurrent genomic copy number variants (CNVs) and mental disorders has been studied for more than a decade, unbiased, population-based estimates of the prevalence, disease risks and trajectories, fertility, and mortality to contrast chromosomal abnormalities and advance precision health care are lacking. Objective To generate unbiased, population-based estimates of prevalence, disease risks and trajectories, fertility, and mortality of CNVs implicated in neuropsychiatric disorders. Design, Setting, and Participants In a population-based case-cohort study, using the Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH) 2012 database, individuals born between May 1, 1981, and December 31, 2005, and followed up until December 31, 2012, were analyzed. All individuals (n = 57 377) with attention-deficit/hyperactivity disorder (ADHD), major depressive disorder (MDD), schizophrenia (SCZ), autism spectrum disorder (ASD), or bipolar disorder (BPD) were included, as well as 30 000 individuals randomly drawn from the database. Data analysis was conducted from July 1, 2017, to September 7, 2021. Exposures Copy number variants at 6 genomic loci (1q21.1, 15q11.2, 15q13.3, 16p11.2, 17p12, and 17q12). Main Outcomes and Measures Population-unbiased hazard ratio (HR) and survival estimates of CNV associations with the 5 ascertained psychiatric disorders, epilepsy, intellectual disability, selected somatic disorders, fertility, and mortality. Results Participants' age ranged from 1 to 32 years (mean, 12.0 [IQR, 6.9] years) during follow-up, and 38 662 were male (52.3%). Copy number variants broadly associated with an increased risk of autism spectrum disorder and ADHD, whereas risk estimates of SCZ for most CNVs were lower than previously reported. Comparison with previous studies suggests that the lower risk estimates are associated with a higher CNV prevalence in the general population than in control samples of most case-control studies. Significant risk of major depressive disorder (HR, 5.8; 95% CI, 1.5-22.2) and sex-specific risk of bipolar disorder (HR, 17; 95% CI, 1.5-189.3, in men only) were noted for the 1q21.1 deletion. Although CNVs at 1q21.1 and 15q13.3 were associated with increased risk across most diagnoses, the 17p12 deletion consistently conferred less risk of psychiatric disorders (HR 0.4-0.8), although none of the estimates differed significantly from the general population. Trajectory analyses noted that, although diagnostic risk profiles differed across loci, they were similar for deletions and duplications within each locus. Sex-stratified analyses suggest that pathogenicity of many CNVs may be modulated by sex. Conclusions and Relevance The findings of this study suggest that the iPSYCH population case cohort reveals broad disease risk for some of the studied CNVs and narrower risk for others, in addition to sex differential liability. This finding on genomic risk variants at the level of a population may be important for health care planning and clinical decision making, and thus the advancement of precision health care.
Collapse
Affiliation(s)
- Xabier Calle Sánchez
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen and Aarhus, Denmark
| | - Dorte Helenius
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen and Aarhus, Denmark
| | - Jonas Bybjerg-Grauholm
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen and Aarhus, Denmark
- Section of Neonatal Screening and Hormones, Department of Clinical Biochemistry, Immunology and Genetics, Statens Serum Institut, Copenhagen, Denmark
| | - Carsten Pedersen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen and Aarhus, Denmark
- National Centre for Register-based Research, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-based Research at Aarhus University, Aarhus, Denmark
- Big Data Centre for Environment and Health, Aarhus University, Aarhus, Denmark
| | - David M. Hougaard
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen and Aarhus, Denmark
- Section of Neonatal Screening and Hormones, Department of Clinical Biochemistry, Immunology and Genetics, Statens Serum Institut, Copenhagen, Denmark
| | - Anders D. Børglum
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen and Aarhus, Denmark
- Department of Biomedicine and iSEQ-Centre for Integrative Sequencing, Aarhus University, Denmark
- Center for Genomics and Personalized Medicine, Aarhus University, Denmark
| | - Merete Nordentoft
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen and Aarhus, Denmark
- Copenhagen Research Centre for Mental Health, Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ole Mors
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen and Aarhus, Denmark
- Psychosis Research Unit, Aarhus University Hospital, Risskov, Denmark
| | - Preben B. Mortensen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen and Aarhus, Denmark
- National Centre for Register-based Research, Aarhus University, Aarhus, Denmark
- Department of Biomedicine and iSEQ-Centre for Integrative Sequencing, Aarhus University, Denmark
| | - Daniel H. Geschwind
- Department of Neurology, University of California, Los Angeles
- Department of Human Genetics, University of California, Los Angeles
- Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine at UCLA, University of California, Los Angeles
- Center for Human Development, University of California, San Diego
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine at UCLA, University of California Los Angeles
| | - Simone Montalbano
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen and Aarhus, Denmark
| | - Armin Raznahan
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland
| | - Wesley K. Thompson
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen and Aarhus, Denmark
- Herbert Wertheim School of Public Health and Human Longevity, University of California, San Diego, La Jolla, California
| | - Andrés Ingason
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen and Aarhus, Denmark
- Lundbeck Foundation Center for GeoGenetics, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Werge
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen and Aarhus, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Lundbeck Foundation Center for GeoGenetics, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Wang J, Zhou S, He F, Zhang X, Lu J, Zhang J, Zhang F, Xu X, Yang F, Xiong F. Familial Translocation t(2;4) (q37.3;p16.3), Resulting in a Partial Trisomy of 2q (or 4p) and a Partial Monosomy of 4p (or 2q), Causes Dysplasia. Front Genet 2021; 12:741607. [PMID: 34887900 PMCID: PMC8650131 DOI: 10.3389/fgene.2021.741607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/27/2021] [Indexed: 11/25/2022] Open
Abstract
Background: Wolf-Hirschhorn syndrome, a well-known contiguous microdeletion syndrome, is caused by deletions on chromosome 4p. While the clinical symptoms and the critical region for this disorder have been identified based on genotype-phenotype correlations, duplications in this region have been infrequently reported. Conclusion: Our case report shows that both deletions and duplications of the Wolf-Hirshhorn critical region cause intellectual disability/developmental delay and multiple congenital anomalies.
Collapse
Affiliation(s)
- Jian Wang
- Department of Fetal Medicine and Prenatal Diagnosis, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shiyuan Zhou
- Henan Provincial Research Institute for Population and Family Planning Zhengzhou China, Zhengzhou, China
| | - Fei He
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xuelian Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jianqi Lu
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), State Key Laboratory of Genetic Engineering at School of Life Sciences, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), State Key Laboratory of Genetic Engineering at School of Life Sciences, Fudan University, Shanghai, China
| | - Xiangmin Xu
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Fang Yang
- Department of Fetal Medicine and Prenatal Diagnosis, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Fu Xiong
- Department of Fetal Medicine and Prenatal Diagnosis, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| |
Collapse
|
29
|
Abstract
In the last 40 years, there has been a huge increase in autism genetics research and a rapidly growing number of discoveries. We now know autism is one of the most highly heritable disorders with negligible shared environmental contributions. Recent discoveries also show that rare variants of large effect size as well as small effect common gene variants all contribute to autism risk. These discoveries challenge traditional diagnostic boundaries and highlight huge heterogeneity in autism. In this review, we consider some of the key findings that are shaping current understanding of autism and what these discoveries mean for clinicians.
Collapse
Affiliation(s)
- Anita Thapar
- Division of Psychological Medicine and Clinical Neurosciences and MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Hadyn Ellis Building, Cardiff, Maindy Road, Wales, CF24 4HQ, UK.
| | - Michael Rutter
- Social, Genetic and Developmental Psychiatry Centre, Kings College London, London, UK
| |
Collapse
|
30
|
Finucane B, Oetjens MT, Johns A, Myers SM, Fisher C, Habegger L, Maxwell EK, Reid JG, Ledbetter DH, Kirchner HL, Martin CL. Medical manifestations and health care utilization among adult MyCode participants with neurodevelopmental psychiatric copy number variants. Genet Med 2021; 24:703-711. [PMID: 34906480 DOI: 10.1016/j.gim.2021.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/07/2021] [Accepted: 11/12/2021] [Indexed: 10/19/2022] Open
Abstract
PURPOSE Recurrent pathogenic copy number variants (pCNVs) have large-effect impacts on brain function and represent important etiologies of neurodevelopmental psychiatric disorders (NPDs), including autism and schizophrenia. Patterns of health care utilization in adults with pCNVs have gone largely unstudied and are likely to differ in significant ways from those of children. METHODS We compared the prevalence of NPDs and electronic health record-based medical conditions in 928 adults with 26 pCNVs to a demographically-matched cohort of pCNV-negative controls from >135,000 patient-participants in Geisinger's MyCode Community Health Initiative. We also evaluated 3 quantitative health care utilization measures (outpatient, inpatient, and emergency department visits) in both groups. RESULTS Adults with pCNVs (24.9%) were more likely than controls (16.0%) to have a documented NPD. They had significantly higher rates of several chronic diseases, including diabetes (29.3% in participants with pCNVs vs 20.4% in participants without pCNVs) and dementia (2.2% in participants with pCNVs vs 1.0% participants without pCNVs), and twice as many annual emergency department visits. CONCLUSION These findings highlight the potential for genetic information-specifically, pCNVs-to inform the study of health care outcomes and utilization in adults. If, as our findings suggest, adults with pCNVs have poorer health and require disproportionate health care resources, early genetic diagnosis paired with patient-centered interventions may help to anticipate problems, improve outcomes, and reduce the associated economic burden.
Collapse
Affiliation(s)
- Brenda Finucane
- Geisinger, Autism & Developmental Medicine Institute, Lewisburg, PA.
| | | | - Alicia Johns
- Geisinger, Department of Population Health Sciences, Danville, PA
| | - Scott M Myers
- Geisinger, Autism & Developmental Medicine Institute, Lewisburg, PA
| | - Ciaran Fisher
- Geisinger, Autism & Developmental Medicine Institute, Lewisburg, PA
| | | | | | | | | | | | | |
Collapse
|
31
|
O'Connell KS, Coombes BJ. Genetic contributions to bipolar disorder: current status and future directions. Psychol Med 2021; 51:2156-2167. [PMID: 33879273 PMCID: PMC8477227 DOI: 10.1017/s0033291721001252] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 03/12/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022]
Abstract
Bipolar disorder (BD) is a highly heritable mental disorder and is estimated to affect about 50 million people worldwide. Our understanding of the genetic etiology of BD has greatly increased in recent years with advances in technology and methodology as well as the adoption of international consortiums and large population-based biobanks. It is clear that BD is also highly heterogeneous and polygenic and shows substantial genetic overlap with other psychiatric disorders. Genetic studies of BD suggest that the number of associated loci is expected to substantially increase in larger future studies and with it, improved genetic prediction of the disorder. Still, a number of challenges remain to fully characterize the genetic architecture of BD. First among these is the need to incorporate ancestrally-diverse samples to move research away from a Eurocentric bias that has the potential to exacerbate health disparities already seen in BD. Furthermore, incorporation of population biobanks, registry data, and electronic health records will be required to increase the sample size necessary for continued genetic discovery, while increased deep phenotyping is necessary to elucidate subtypes within BD. Lastly, the role of rare variation in BD remains to be determined. Meeting these challenges will enable improved identification of causal variants for the disorder and also allow for equitable future clinical applications of both genetic risk prediction and therapeutic interventions.
Collapse
Affiliation(s)
- Kevin S. O'Connell
- Division of Mental Health and Addiction, NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo University Hospital, 0407Oslo, Norway
| | - Brandon J. Coombes
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
32
|
Silva AI, Kirov G, Kendall KM, Bracher-Smith M, Wilkinson LS, Hall J, Ulfarsson MO, Walters GB, Stefansson H, Stefansson K, Linden DEJ, Caseras X. Analysis of Diffusion Tensor Imaging Data From the UK Biobank Confirms Dosage Effect of 15q11.2 Copy Number Variation on White Matter and Shows Association With Cognition. Biol Psychiatry 2021; 90:307-316. [PMID: 33931204 PMCID: PMC8343146 DOI: 10.1016/j.biopsych.2021.02.969] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND Copy number variations at the 15q11.2 BP1-BP2 locus are present in 0.5%-1.0% of the population, and the deletion is associated with several neurodevelopmental disorders. Previously, we showed a reciprocal effect of 15q11.2 copy number variation on fractional anisotropy, with widespread increases in deletion carriers. We aim to expand these findings using a larger sample of participants (N = 29,166) and higher resolution imaging and by examining the implications for cognitive performance. METHODS Diffusion tensor imaging measures from participants with no neurological or psychiatric diagnoses were obtained from the UK Biobank database. We compared 15q11.2 BP1-BP2 deletion (n = 102) and duplication (n = 113) carriers to a large cohort of control individuals with no neuropsychiatric copy number variants (n = 28,951). Additionally, we assessed how changes in white matter mediated the association between carrier status and cognitive performance. RESULTS Deletion carriers showed increases in fractional anisotropy in the internal capsule and cingulum and decreases in the posterior thalamic radiation compared with both duplication carriers and control subjects (who had intermediate values). Compared with control subjects, deletion carriers had lower scores across cognitive tasks, which were partly influenced by white matter. Reduced fractional anisotropy in the posterior thalamic radiation partially contributed to worse cognitive performance in deletion carriers. CONCLUSIONS These results, together with our previous findings, provide convergent evidence for an effect of 15q11.2 BP1-BP2 on white matter microstructure, this being more pronounced in deletion carriers. Additionally, changes in white matter were found to partially mediate cognitive ability in deletion carriers, providing a link between white matter changes in 15q11.2 BP1-BP2 carriers and cognitive function.
Collapse
Affiliation(s)
- Ana I Silva
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff, United Kingdom; Cardiff University Brain Research Imaging Centre School of Psychology, Cardiff University, Cardiff, United Kingdom; School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands.
| | - George Kirov
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff, United Kingdom
| | - Kimberley M Kendall
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff, United Kingdom
| | - Mathew Bracher-Smith
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff, United Kingdom
| | - Lawrence S Wilkinson
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff, United Kingdom; Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom; School of Psychology, Cardiff University, Cardiff, United Kingdom
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff, United Kingdom; Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Magnus O Ulfarsson
- deCODE genetics/Amgen, Reykjavik, Iceland; Faculty of Electrical and Computer Engineering, University of Iceland, Reykjavik, Iceland
| | - G Bragi Walters
- deCODE genetics/Amgen, Reykjavik, Iceland; Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | | | - Kari Stefansson
- deCODE genetics/Amgen, Reykjavik, Iceland; Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - David E J Linden
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff, United Kingdom; School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Xavier Caseras
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff, United Kingdom.
| |
Collapse
|
33
|
Gokhale A, Lee CE, Zlatic SA, Freeman AAH, Shearing N, Hartwig C, Ogunbona O, Bassell JL, Wynne ME, Werner E, Xu C, Wen Z, Duong D, Seyfried NT, Bearden CE, Oláh VJ, Rowan MJM, Glausier JR, Lewis DA, Faundez V. Mitochondrial Proteostasis Requires Genes Encoded in a Neurodevelopmental Syndrome Locus. J Neurosci 2021; 41:6596-6616. [PMID: 34261699 PMCID: PMC8336702 DOI: 10.1523/jneurosci.2197-20.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 06/23/2021] [Accepted: 06/26/2021] [Indexed: 02/08/2023] Open
Abstract
Eukaryotic cells maintain proteostasis through mechanisms that require cytoplasmic and mitochondrial translation. Genetic defects affecting cytoplasmic translation perturb synapse development, neurotransmission, and are causative of neurodevelopmental disorders, such as Fragile X syndrome. In contrast, there is little indication that mitochondrial proteostasis, either in the form of mitochondrial protein translation and/or degradation, is required for synapse development and function. Here we focus on two genes deleted in a recurrent copy number variation causing neurodevelopmental disorders, the 22q11.2 microdeletion syndrome. We demonstrate that SLC25A1 and MRPL40, two genes present in the microdeleted segment and whose products localize to mitochondria, interact and are necessary for mitochondrial ribosomal integrity and proteostasis. Our Drosophila studies show that mitochondrial ribosome function is necessary for synapse neurodevelopment, function, and behavior. We propose that mitochondrial proteostasis perturbations, either by genetic or environmental factors, are a pathogenic mechanism for neurodevelopmental disorders.SIGNIFICANCE STATEMENT The balance between cytoplasmic protein synthesis and degradation, or cytoplasmic proteostasis, is required for normal synapse function and neurodevelopment. Cytoplasmic and mitochondrial ribosomes are necessary for two compartmentalized, yet interdependent, forms of proteostasis. Proteostasis dependent on cytoplasmic ribosomes is a well-established target of genetic defects that cause neurodevelopmental disorders, such as autism. Here we show that the mitochondrial ribosome is a neurodevelopmentally regulated organelle whose function is required for synapse development and function. We propose that defective mitochondrial proteostasis is a mechanism with the potential to contribute to neurodevelopmental disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Zhexing Wen
- Departments of Cell Biology
- Psychiatry and Behavioral Sciences
| | - Duc Duong
- and Biochemistry, Emory University, Atlanta, Georgia 30322
| | | | - Carrie E Bearden
- Semel Institute for Neuroscience and Human Behavior Department of Psychology, UCLA, Los Angeles, California 90095
| | | | | | - Jill R Glausier
- Departments of Psychiatry and Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | - David A Lewis
- Departments of Psychiatry and Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | | |
Collapse
|
34
|
Dinneen TJ, Ghrálaigh FN, Walsh R, Lopez LM, Gallagher L. How does genetic variation modify ND-CNV phenotypes? Trends Genet 2021; 38:140-151. [PMID: 34364706 DOI: 10.1016/j.tig.2021.07.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/30/2021] [Accepted: 07/06/2021] [Indexed: 02/05/2023]
Abstract
Rare copy-number variants (CNVs) associated with neurodevelopmental disorders (NDDs), i.e., ND-CNVs, provide an insight into the neurobiology of NDDs and, potentially, a link between biology and clinical outcomes. However, ND-CNVs are characterised by incomplete penetrance resulting in heterogeneous carrier phenotypes, ranging from non-affected to multimorbid psychiatric, neurological, and physical phenotypes. Recent evidence indicates that other variants in the genome, or 'other hits', may partially explain the variable expressivity of ND-CNVs. These may be other rare variants or the aggregated effects of common variants that modify NDD risk. Here we discuss the recent findings, current questions, and future challenges relating to other hits research in the context of ND-CNVs and their potential for improved clinical diagnostics and therapeutics for ND-CNV carriers.
Collapse
Affiliation(s)
- Thomas J Dinneen
- Department of Psychiatry, Trinity College Dublin, Dublin, Ireland.
| | - Fiana Ní Ghrálaigh
- Department of Psychiatry, Trinity College Dublin, Dublin, Ireland; Department of Biology, National University of Ireland Maynooth, Maynooth, Ireland
| | - Ruth Walsh
- Department of Psychiatry, Trinity College Dublin, Dublin, Ireland
| | - Lorna M Lopez
- Department of Psychiatry, Trinity College Dublin, Dublin, Ireland; Department of Biology, National University of Ireland Maynooth, Maynooth, Ireland
| | - Louise Gallagher
- Department of Psychiatry, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
35
|
Contextualizing genetic risk score for disease screening and rare variant discovery. Nat Commun 2021; 12:4418. [PMID: 34285202 PMCID: PMC8292385 DOI: 10.1038/s41467-021-24387-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 06/07/2021] [Indexed: 11/08/2022] Open
Abstract
Studies of the genetic basis of complex traits have demonstrated a substantial role for common, small-effect variant polygenic burden (PB) as well as large-effect variants (LEV, primarily rare). We identify sufficient conditions in which GWAS-derived PB may be used for well-powered rare pathogenic variant discovery or as a sample prioritization tool for whole-genome or exome sequencing. Through extensive simulations of genetic architectures and generative models of disease liability with parameters informed by empirical data, we quantify the power to detect, among cases, a lower PB in LEV carriers than in non-carriers. Furthermore, we uncover clinically useful conditions wherein the risk derived from the PB is comparable to the LEV-derived risk. The resulting summary-statistics-based methodology (with publicly available software, PB-LEV-SCAN) makes predictions on PB-based LEV screening for 36 complex traits, which we confirm in several disease datasets with available LEV information in the UK Biobank, with important implications on clinical decision-making.
Collapse
|
36
|
Dowden L, Tucker D, Morgan S, Uzun O, Syed YA. Contribution of Congenital Heart Disorders Associated With Copy Number Variants in Mediating Risk for Brain Developmental Disorders: Evidence From 20-Year Retrospective Cohort Study. Front Cardiovasc Med 2021; 8:655463. [PMID: 34336942 PMCID: PMC8319541 DOI: 10.3389/fcvm.2021.655463] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 06/15/2021] [Indexed: 11/25/2022] Open
Abstract
Rare pathogenic copy number variants (CNVs) are genetic rearrangements that have been associated with an increased risk for congenital heart disorders (CHDs). However, the association of CNVs with atypical brain development, leading to neurodevelopmental disorders (NDDs), in the presence of CHDs remains unclear. We attempted to explore this association by establishing the prevalence and burden of CNVs associated with CHD in a Welsh population and by studying the effect of rare CNVs associated with CHDs in mediating the risk of NDDs. Toward this goal, we analyzed data from the Congenital Anomaly Register for Wales (CARIS), referred from hospitals in Wales between 1998 and 2018, which included 1,113 subjects in total. Of these, 785 subjects were included in the study following application of the exclusion criteria, and a total of 28 rare CNVs associated with CHD were analyzed. The findings from this cohort study identified 22q11.2 deletion as the most prominent CNV across the cohort. Our data demonstrates that the survival rate of the cohort after 3 years was 99.9%, and mortality fell significantly between 1 and 2 years and between 2 and 3 years [F(1,27) = 10, p = 0.0027; F(1,27) = 5.8, p = 0.0222]. Importantly, the data set revealed a positive correlation between the incidence of congenital heart disease and the incidence of neurodevelopmental abnormalities in patients with CNVs across the whole cohort [95% CI (0.4062, 0.8449), p < 0.0001, r = 0.6829]. Additionally, we identified significant CNVs that result in the co-morbidity of CHD and NDD and show that septal defects and global developmental delay are major congenital defects. Further research should identify a common molecular mechanism leading to the phenotypic comorbidity of CHDs and NDDs, arising from a common CNV, which can have an implication for improving risk classification and for fetal neuroprotection strategies in the affected children and in precision medicine.
Collapse
Affiliation(s)
- Luke Dowden
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom.,School of Bioscience, Cardiff University, Cardiff, United Kingdom
| | - David Tucker
- Knowledge Directorate, Public Health Wales, Swansea, United Kingdom
| | - Sian Morgan
- Department of Medical Genetics, University Hospital of Wales, Cardiff, United Kingdom
| | - Orhan Uzun
- Department of Fetal Cardiology, University Hospital of Wales, Cardiff, United Kingdom
| | - Yasir Ahmed Syed
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom.,School of Bioscience, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
37
|
Rare Copy Number Variants Are Associated With Poorer Cognition in Schizophrenia. Biol Psychiatry 2021; 90:28-34. [PMID: 33678419 DOI: 10.1016/j.biopsych.2020.11.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 10/09/2020] [Accepted: 11/15/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Cognitive impairment in schizophrenia is a major contributor to poor outcomes, yet its causes are poorly understood. Some rare copy number variants (CNVs) are associated with schizophrenia risk and affect cognition in healthy populations, but their contribution to cognitive impairment in schizophrenia has not been investigated. We examined the effect of 12 schizophrenia CNVs on cognition in those with schizophrenia. METHODS General cognitive ability was measured using the Measurement and Treatment Research to Improve Cognition in Schizophrenia composite z score in 875 patients with schizophrenia and in a replication sample of 519 patients with schizophrenia using Wechsler Adult Intelligence Scale Full Scale IQ. Using linear regression, we tested for association between cognition and schizophrenia CNV status, covarying for age and sex. In addition, we tested whether CNVs hitting genes in schizophrenia-enriched gene sets (loss-of-function intolerant and synaptic gene sets) were associated with cognitive impairment. RESULTS A total of 23 schizophrenia CNV carriers were identified. Schizophrenia CNV carriers had lower general cognitive ability than nonschizophrenia CNV carriers in discovery (β = -0.66, 95% confidence interval [CI] = -1.31 to -0.01) and replication samples (β = -0.91, 95% CI = -1.71 to -0.11) and after meta-analysis (β = -0.76, 95% CI = -1.26 to -0.25, p = .003). CNVs hitting loss-of-function intolerant genes were associated with lower cognition (β = -0.15, 95% CI = -0.29 to -0.001, p = .048). CONCLUSIONS In those with schizophrenia, cognitive ability in schizophrenia CNV carriers is 0.5-1.0 standard deviations below non-CNV carriers, which may have implications for clinical assessment and management. We also demonstrate that rare CNVs hitting genes intolerant to loss-of-function variation lead to more severe cognitive impairment, above and beyond the effect of known schizophrenia CNVs.
Collapse
|
38
|
Cross-platform validation of neurotransmitter release impairments in schizophrenia patient-derived NRXN1-mutant neurons. Proc Natl Acad Sci U S A 2021; 118:2025598118. [PMID: 34035170 DOI: 10.1073/pnas.2025598118] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Heterozygous NRXN1 deletions constitute the most prevalent currently known single-gene mutation associated with schizophrenia, and additionally predispose to multiple other neurodevelopmental disorders. Engineered heterozygous NRXN1 deletions impaired neurotransmitter release in human neurons, suggesting a synaptic pathophysiological mechanism. Utilizing this observation for drug discovery, however, requires confidence in its robustness and validity. Here, we describe a multicenter effort to test the generality of this pivotal observation, using independent analyses at two laboratories of patient-derived and newly engineered human neurons with heterozygous NRXN1 deletions. Using neurons transdifferentiated from induced pluripotent stem cells that were derived from schizophrenia patients carrying heterozygous NRXN1 deletions, we observed the same synaptic impairment as in engineered NRXN1-deficient neurons. This impairment manifested as a large decrease in spontaneous synaptic events, in evoked synaptic responses, and in synaptic paired-pulse depression. Nrxn1-deficient mouse neurons generated from embryonic stem cells by the same method as human neurons did not exhibit impaired neurotransmitter release, suggesting a human-specific phenotype. Human NRXN1 deletions produced a reproducible increase in the levels of CASK, an intracellular NRXN1-binding protein, and were associated with characteristic gene-expression changes. Thus, heterozygous NRXN1 deletions robustly impair synaptic function in human neurons regardless of genetic background, enabling future drug discovery efforts.
Collapse
|
39
|
Wilson KE, Fish AM, Mankiw C, Xenophontos A, Warling A, Whitman E, Clasen L, Torres E, Blumenthal J, Raznahan A. Modeling familial predictors of proband outcomes in neurogenetic disorders: initial application in XYY syndrome. J Neurodev Disord 2021; 13:12. [PMID: 33752588 PMCID: PMC7986517 DOI: 10.1186/s11689-021-09360-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 03/08/2021] [Indexed: 10/24/2022] Open
Abstract
BACKGROUND Disorders of gene dosage can significantly increase risk for psychopathology, but outcomes vary greatly amongst carriers of any given chromosomal aneuploidy or sub-chromosomal copy number variation (CNV). One potential path to advance precision medicine for neurogenetic disorders is modeling penetrance in probands relative to observed phenotypes in their non-carrier relatives. Here, we seek to advance this general analytic framework by developing new methods in application to XYY syndrome-a sex chromosome aneuploidy that is known to increase risk for psychopathology. METHODS We analyzed a range of cognitive and behavioral domains in XYY probands and their non-carrier family members (n = 58 families), including general cognitive ability (FSIQ), as well as continuous measures of traits related to autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD). Proband and relative scores were compared using covariance, regression and cluster analysis. Comparisons were made both within and across traits. RESULTS Proband scores were shifted away from family scores with effect sizes varying between 0.9 and 2.4 across traits. Only FSIQ and vocabulary scores showed a significant positive correlation between probands and their non-carrier relatives across families (R2 ~ 0.4). Variability in family FSIQ also cross-predicted variability in proband ASD trait severity. Cluster analysis across all trait-relative pairings revealed that variability in parental psychopathology was more weakly coupled to their XYY versus their euploid offspring. CONCLUSIONS We present a suite of generalizable methods for modeling variable penetrance in aneuploidy and CNV carriers using family data. These methods update estimates of phenotypic penetrance for XYY and suggest that the predictive utility of family data is likely to vary for different traits and different gene dosage disorders. TRIAL REGISTRATIONS ClinicalTrials.gov NCT00001246 , "89-M-0006: Brain Imaging of Childhood Onset Psychiatric Disorders, Endocrine Disorders and Healthy Controls." Date of registry: 01 October 1989.
Collapse
Affiliation(s)
- Kathleen E Wilson
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Building 10 Room 4N242 MSC 1367, 10 Center Drive, Bethesda, MD, 20892-1367, USA
| | - Ari M Fish
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Building 10 Room 4N242 MSC 1367, 10 Center Drive, Bethesda, MD, 20892-1367, USA
| | - Catherine Mankiw
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Building 10 Room 4N242 MSC 1367, 10 Center Drive, Bethesda, MD, 20892-1367, USA
| | - Anastasia Xenophontos
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Building 10 Room 4N242 MSC 1367, 10 Center Drive, Bethesda, MD, 20892-1367, USA
| | - Allysa Warling
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Building 10 Room 4N242 MSC 1367, 10 Center Drive, Bethesda, MD, 20892-1367, USA
| | - Ethan Whitman
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Building 10 Room 4N242 MSC 1367, 10 Center Drive, Bethesda, MD, 20892-1367, USA
| | - Liv Clasen
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Building 10 Room 4N242 MSC 1367, 10 Center Drive, Bethesda, MD, 20892-1367, USA
| | - Erin Torres
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Building 10 Room 4N242 MSC 1367, 10 Center Drive, Bethesda, MD, 20892-1367, USA
| | - Jonathan Blumenthal
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Building 10 Room 4N242 MSC 1367, 10 Center Drive, Bethesda, MD, 20892-1367, USA
| | - Armin Raznahan
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Building 10 Room 4N242 MSC 1367, 10 Center Drive, Bethesda, MD, 20892-1367, USA.
| |
Collapse
|
40
|
Lencz T, Yu J, Khan RR, Flaherty E, Carmi S, Lam M, Ben-Avraham D, Barzilai N, Bressman S, Darvasi A, Cho JH, Clark LN, Gümüş ZH, Vijai J, Klein RJ, Lipkin S, Offit K, Ostrer H, Ozelius LJ, Peter I, Malhotra AK, Maniatis T, Atzmon G, Pe'er I. Novel ultra-rare exonic variants identified in a founder population implicate cadherins in schizophrenia. Neuron 2021; 109:1465-1478.e4. [PMID: 33756103 DOI: 10.1016/j.neuron.2021.03.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/16/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022]
Abstract
The identification of rare variants associated with schizophrenia has proven challenging due to genetic heterogeneity, which is reduced in founder populations. In samples from the Ashkenazi Jewish population, we report that schizophrenia cases had a greater frequency of novel missense or loss of function (MisLoF) ultra-rare variants (URVs) compared to controls, and the MisLoF URV burden was inversely correlated with polygenic risk scores in cases. Characterizing 141 "case-only" genes (MisLoF URVs in ≥3 cases with none in controls), the cadherin gene set was associated with schizophrenia. We report a recurrent case mutation in PCDHA3 that results in the formation of cytoplasmic aggregates and failure to engage in homophilic interactions on the plasma membrane in cultured cells. Modeling purifying selection, we demonstrate that deleterious URVs are greatly overrepresented in the Ashkenazi population, yielding enhanced power for association studies. Identification of the cadherin/protocadherin family as risk genes helps specify the synaptic abnormalities central to schizophrenia.
Collapse
Affiliation(s)
- Todd Lencz
- Departments of Psychiatry and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11550, USA; Department of Psychiatry, Division of Research, The Zucker Hillside Hospital Division of Northwell Health, Glen Oaks, NY 11004, USA; Institute for Behavioral Science, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA.
| | - Jin Yu
- Department of Psychiatry, Division of Research, The Zucker Hillside Hospital Division of Northwell Health, Glen Oaks, NY 11004, USA; Institute for Behavioral Science, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Raiyan Rashid Khan
- Department of Computer Science, Columbia University, New York, NY 10027, USA
| | - Erin Flaherty
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Shai Carmi
- Braun School of Public Health and Community Medicine, Faculty of Medicine, Hebrew University of Jerusalem, Ein Kerem, Jerusalem 9112102, Israel
| | - Max Lam
- Department of Psychiatry, Division of Research, The Zucker Hillside Hospital Division of Northwell Health, Glen Oaks, NY 11004, USA; Institute for Behavioral Science, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Danny Ben-Avraham
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Nir Barzilai
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Susan Bressman
- Department of Neurology, Beth Israel Medical Center, New York, NY 10003, USA
| | - Ariel Darvasi
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of Jerusalem, Givat Ram, Jerusalem 91904, Israel
| | - Judy H Cho
- Institute for Personalized Medicine, 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
| | - Lorraine N Clark
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA; Taub Institute for Research of Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY 10032, USA
| | - Zeynep H Gümüş
- Department of Genetics and Genomic Sciences, 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
| | - Joseph Vijai
- Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Robert J Klein
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Taub Institute for Research of Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY 10032, USA
| | - Steven Lipkin
- Departments of Medicine, Genetic Medicine and Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Kenneth Offit
- Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Harry Ostrer
- Departments of Pathology and Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Laurie J Ozelius
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Inga Peter
- Department of Genetics and Genomic Sciences, 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
| | - Anil K Malhotra
- Departments of Psychiatry and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11550, USA; Department of Psychiatry, Division of Research, The Zucker Hillside Hospital Division of Northwell Health, Glen Oaks, NY 11004, USA; Institute for Behavioral Science, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Tom Maniatis
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY 10027, USA; New York Genome Center, New York, NY 10013, USA
| | - Gil Atzmon
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Human Biology, Haifa University, Haifa, Israel
| | - Itsik Pe'er
- Department of Computer Science, Columbia University, New York, NY 10027, USA; Center for Computational Biology and Bioinformatics, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
41
|
Mezinska S, Gallagher L, Verbrugge M, Bunnik EM. Ethical issues in genomics research on neurodevelopmental disorders: a critical interpretive review. Hum Genomics 2021; 15:16. [PMID: 33712057 PMCID: PMC7953558 DOI: 10.1186/s40246-021-00317-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/04/2021] [Indexed: 11/16/2022] Open
Abstract
Background Genomic research on neurodevelopmental disorders (NDDs), particularly involving minors, combines and amplifies existing research ethics issues for biomedical research. We performed a review of the literature on the ethical issues associated with genomic research involving children affected by NDDs as an aid to researchers to better anticipate and address ethical concerns. Results Qualitative thematic analysis of the included articles revealed themes in three main areas: research design and ethics review, inclusion of research participants, and communication of research results. Ethical issues known to be associated with genomic research in general, such as privacy risks and informed consent/assent, seem especially pressing for NDD participants because of their potentially decreased cognitive abilities, increased vulnerability, and stigma associated with mental health problems. Additionally, there are informational risks: learning genetic information about NDD may have psychological and social impact, not only for the research participant but also for family members. However, there are potential benefits associated with research participation, too: by enrolling in research, the participants may access genetic testing and thus increase their chances of receiving a (genetic) diagnosis for their neurodevelopmental symptoms, prognostic or predictive information about disease progression or the risk of concurrent future disorders. Based on the results of our review, we developed an ethics checklist for genomic research involving children affected by NDDs. Conclusions In setting up and designing genomic research efforts in NDD, researchers should partner with communities of persons with NDDs. Particular attention should be paid to preventing disproportional burdens of research participation of children with NDDs and their siblings, parents and other family members. Researchers should carefully tailor the information and informed consent procedures to avoid therapeutic and diagnostic misconception in NDD research. To better anticipate and address ethical issues in specific NDD studies, we suggest researchers to use the ethics checklist for genomic research involving children affected by NDDs presented in this paper. Supplementary Information The online version contains supplementary material available at 10.1186/s40246-021-00317-4.
Collapse
Affiliation(s)
- S Mezinska
- Faculty of Medicine and Institute of Clinical and Preventive Medicine, University of Latvia, Jelgavas Str.3, Riga, LV-1004, Latvia.
| | - L Gallagher
- Discipline of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, St. James Hospital, Dublin 8, Ireland
| | - M Verbrugge
- Department of Medical Ethics, Philosophy and History of Medicine, Erasmus MC, University Medical Centre Rotterdam, PO Box 2400, Rotterdam, 3000, CA, The Netherlands
| | - E M Bunnik
- Department of Medical Ethics, Philosophy and History of Medicine, Erasmus MC, University Medical Centre Rotterdam, PO Box 2400, Rotterdam, 3000, CA, The Netherlands
| |
Collapse
|
42
|
Al-Absi AR, Qvist P, Glerup S, Sanchez C, Nyengaard JR. Df(h15q13)/+ Mouse Model Reveals Loss of Astrocytes and Synaptic-Related Changes of the Excitatory and Inhibitory Circuits in the Medial Prefrontal Cortex. Cereb Cortex 2021; 31:1609-1621. [PMID: 33123721 DOI: 10.1093/cercor/bhaa313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 08/19/2020] [Accepted: 09/20/2020] [Indexed: 11/13/2022] Open
Abstract
The 15q13.3 deletion is associated with multiple neurodevelopmental disorders including epilepsy, schizophrenia, and autism. The Df(h15q13)/+ mouse model was recently generated that recapitulates several phenotypic features of the human 15q13.3 deletion syndrome (DS). However, the biological substrates underlying these phenotypes in Df(h15q13)/+ mice have not yet been fully characterized. RNA sequencing followed by real-time quantitative PCR, western blotting, liquid chromatography-mass spectrometry, and stereological analysis were employed to dissect the molecular, structural, and neurochemical phenotypes of the medial prefrontal cortex (mPFC) circuits in Df(h15q13)/+ mouse model. Transcriptomic profiling revealed enrichment for astrocyte-specific genes among differentially expressed genes, translated by a decrease in the number of glial fibrillary acidic protein positive cells in mPFC of Df(h15q13)/+ mice compared with wild-type mice. mPFC in Df(h15q13)/+ mice also showed a deficit of the inhibitory presynaptic marker GAD65, in addition to a reduction in dendritic arborization and spine density of pyramidal neurons from layers II/III. mPFC levels of GABA and glutamate neurotransmitters were not different between genotypes. Our results suggest that the 15q13.3 deletion modulates nonneuronal circuits in mPFC and confers molecular and morphometric alterations in the inhibitory and excitatory neurocircuits, respectively. These alterations potentially contribute to the phenotypes accompanied with the 15q13.3DS.
Collapse
Affiliation(s)
- Abdel-Rahman Al-Absi
- Center for Molecular Morphology, Section for Stereology and Microscopy, Center for Stochastic Geometry and Advanced Bioimaging, Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Per Qvist
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark.,The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, 8210 Aarhus, Denmark.,Centre for Integrative Sequencing, iSEQ, Aarhus University, 8000 Aarhus, Denmark.,Center for Genomics and Personalized Medicine, CGPM, Aarhus University, 8000 Aarhus, Denmark
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Connie Sanchez
- Translational Neuropsychiatry Unit, Aarhus University, 8000 Aarhus, Denmark
| | - Jens R Nyengaard
- Center for Molecular Morphology, Section for Stereology and Microscopy, Center for Stochastic Geometry and Advanced Bioimaging, Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
43
|
Cntn4, a risk gene for neuropsychiatric disorders, modulates hippocampal synaptic plasticity and behavior. Transl Psychiatry 2021; 11:106. [PMID: 33542194 PMCID: PMC7862349 DOI: 10.1038/s41398-021-01223-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 01/05/2021] [Accepted: 01/18/2021] [Indexed: 12/27/2022] Open
Abstract
Neurodevelopmental and neuropsychiatric disorders, such as autism spectrum disorders (ASD), anorexia nervosa (AN), Alzheimer's disease (AD), and schizophrenia (SZ), are heterogeneous brain disorders with unknown etiology. Genome wide studies have revealed a wide variety of risk genes for these disorders, indicating a biological link between genetic signaling pathways and brain pathology. A unique risk gene is Contactin 4 (Cntn4), an Ig cell adhesion molecule (IgCAM) gene, which has been associated with several neuropsychiatric disorders including ASD, AN, AD, and SZ. Here, we investigated the Cntn4 gene knockout (KO) mouse model to determine whether memory dysfunction and altered brain plasticity, common neuropsychiatric symptoms, are affected by Cntn4 genetic disruption. For that purpose, we tested if Cntn4 genetic disruption affects CA1 synaptic transmission and the ability to induce LTP in hippocampal slices. Stimulation in CA1 striatum radiatum significantly decreased synaptic potentiation in slices of Cntn4 KO mice. Neuroanatomical analyses showed abnormal dendritic arborization and spines of hippocampal CA1 neurons. Short- and long-term recognition memory, spatial memory, and fear conditioning responses were also assessed. These behavioral studies showed increased contextual fear conditioning in heterozygous and homozygous KO mice, quantified by a gene-dose dependent increase in freezing response. In comparison to wild-type mice, Cntn4-deficient animals froze significantly longer and groomed more, indicative of increased stress responsiveness under these test conditions. Our electrophysiological, neuro-anatomical, and behavioral results in Cntn4 KO mice suggest that Cntn4 has important functions related to fear memory possibly in association with the neuronal morphological and synaptic plasticity changes in hippocampus CA1 neurons.
Collapse
|
44
|
Gualtieri CT. Genomic Variation, Evolvability, and the Paradox of Mental Illness. Front Psychiatry 2021; 11:593233. [PMID: 33551865 PMCID: PMC7859268 DOI: 10.3389/fpsyt.2020.593233] [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: 08/10/2020] [Accepted: 11/27/2020] [Indexed: 12/30/2022] Open
Abstract
Twentieth-century genetics was hard put to explain the irregular behavior of neuropsychiatric disorders. Autism and schizophrenia defy a principle of natural selection; they are highly heritable but associated with low reproductive success. Nevertheless, they persist. The genetic origins of such conditions are confounded by the problem of variable expression, that is, when a given genetic aberration can lead to any one of several distinct disorders. Also, autism and schizophrenia occur on a spectrum of severity, from mild and subclinical cases to the overt and disabling. Such irregularities reflect the problem of missing heritability; although hundreds of genes may be associated with autism or schizophrenia, together they account for only a small proportion of cases. Techniques for higher resolution, genomewide analysis have begun to illuminate the irregular and unpredictable behavior of the human genome. Thus, the origins of neuropsychiatric disorders in particular and complex disease in general have been illuminated. The human genome is characterized by a high degree of structural and behavioral variability: DNA content variation, epistasis, stochasticity in gene expression, and epigenetic changes. These elements have grown more complex as evolution scaled the phylogenetic tree. They are especially pertinent to brain development and function. Genomic variability is a window on the origins of complex disease, neuropsychiatric disorders, and neurodevelopmental disorders in particular. Genomic variability, as it happens, is also the fuel of evolvability. The genomic events that presided over the evolution of the primate and hominid lineages are over-represented in patients with autism and schizophrenia, as well as intellectual disability and epilepsy. That the special qualities of the human genome that drove evolution might, in some way, contribute to neuropsychiatric disorders is a matter of no little interest.
Collapse
|
45
|
Finucane BM, Ledbetter DH, Vorstman JA. Diagnostic genetic testing for neurodevelopmental psychiatric disorders: closing the gap between recommendation and clinical implementation. Curr Opin Genet Dev 2021; 68:1-8. [PMID: 33434711 DOI: 10.1016/j.gde.2020.12.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/09/2020] [Accepted: 12/22/2020] [Indexed: 12/19/2022]
Abstract
Advances in laboratory testing have significantly increased the detection of rare genetic etiologies of neurodevelopmental psychiatric disorders (NPD), particularly developmental delay/ intellectual disability, autism spectrum disorder, and schizophrenia. Establishing a genetic diagnosis has important medical and personal utility for individuals with these conditions. Diagnostic genetic tests for NPD are clinically available but underutilized outside of medical genetics settings. Without clear multidisciplinary consensus recommendations, active involvement of medical specialists working with NPD patients, and practical education and training, the implementation of genetic testing for NPD will continue to lag behind other areas of medicine. In the long-term, collaborative efforts to address educational, logistical, and workforce obstacles will improve patient care and pave the way for targeted, effective NPD treatments.
Collapse
Affiliation(s)
- Brenda M Finucane
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA USA.
| | - David H Ledbetter
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA USA
| | - Jacob As Vorstman
- Department of Psychiatry, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
46
|
Saarentaus EC, Havulinna AS, Mars N, Ahola-Olli A, Kiiskinen TTJ, Partanen J, Ruotsalainen S, Kurki M, Urpa LM, Chen L, Perola M, Salomaa V, Veijola J, Männikkö M, Hall IM, Pietiläinen O, Kaprio J, Ripatti S, Daly M, Palotie A. Polygenic burden has broader impact on health, cognition, and socioeconomic outcomes than most rare and high-risk copy number variants. Mol Psychiatry 2021; 26:4884-4895. [PMID: 33526825 PMCID: PMC8589645 DOI: 10.1038/s41380-021-01026-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 12/18/2020] [Accepted: 01/11/2021] [Indexed: 12/29/2022]
Abstract
Copy number variants (CNVs) are associated with syndromic and severe neurological and psychiatric disorders (SNPDs), such as intellectual disability, epilepsy, schizophrenia, and bipolar disorder. Although considered high-impact, CNVs are also observed in the general population. This presents a diagnostic challenge in evaluating their clinical significance. To estimate the phenotypic differences between CNV carriers and non-carriers regarding general health and well-being, we compared the impact of SNPD-associated CNVs on health, cognition, and socioeconomic phenotypes to the impact of three genome-wide polygenic risk score (PRS) in two Finnish cohorts (FINRISK, n = 23,053 and NFBC1966, n = 4895). The focus was on CNV carriers and PRS extremes who do not have an SNPD diagnosis. We identified high-risk CNVs (DECIPHER CNVs, risk gene deletions, or large [>1 Mb] CNVs) in 744 study participants (2.66%), 36 (4.8%) of whom had a diagnosed SNPD. In the remaining 708 unaffected carriers, we observed lower educational attainment (EA; OR = 0.77 [95% CI 0.66-0.89]) and lower household income (OR = 0.77 [0.66-0.89]). Income-associated CNVs also lowered household income (OR = 0.50 [0.38-0.66]), and CNVs with medical consequences lowered subjective health (OR = 0.48 [0.32-0.72]). The impact of PRSs was broader. At the lowest extreme of PRS for EA, we observed lower EA (OR = 0.31 [0.26-0.37]), lower-income (OR = 0.66 [0.57-0.77]), lower subjective health (OR = 0.72 [0.61-0.83]), and increased mortality (Cox's HR = 1.55 [1.21-1.98]). PRS for intelligence had a similar impact, whereas PRS for schizophrenia did not affect these traits. We conclude that the majority of working-age individuals carrying high-risk CNVs without SNPD diagnosis have a modest impact on morbidity and mortality, as well as the limited impact on income and educational attainment, compared to individuals at the extreme end of common genetic variation. Our findings highlight that the contribution of traditional high-risk variants such as CNVs should be analyzed in a broader genetic context, rather than evaluated in isolation.
Collapse
Affiliation(s)
- Elmo Christian Saarentaus
- grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Aki Samuli Havulinna
- grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland ,grid.14758.3f0000 0001 1013 0499Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Nina Mars
- grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Ari Ahola-Olli
- grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland ,grid.66859.34Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA USA ,grid.32224.350000 0004 0386 9924Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, USA
| | | | - Juulia Partanen
- grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Sanni Ruotsalainen
- grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Mitja Kurki
- grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland ,grid.66859.34Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA USA ,grid.32224.350000 0004 0386 9924Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, USA
| | - Lea Martta Urpa
- grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Lei Chen
- grid.47100.320000000419368710Department of Genetics, Yale School of Medicine, New Haven, CT USA ,grid.4367.60000 0001 2355 7002McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO USA
| | - Markus Perola
- grid.14758.3f0000 0001 1013 0499Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Veikko Salomaa
- grid.14758.3f0000 0001 1013 0499Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Juha Veijola
- grid.10858.340000 0001 0941 4873Research Unit of Clinical Neuroscience, University of Oulu & Oulu University Hospital, Oulu, Finland
| | - Minna Männikkö
- grid.10858.340000 0001 0941 4873Northern Finland Birth Cohorts, Infrastructure for Population Studies, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Ira M. Hall
- grid.47100.320000000419368710Department of Genetics, Yale School of Medicine, New Haven, CT USA ,grid.4367.60000 0001 2355 7002McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO USA
| | - Olli Pietiläinen
- grid.66859.34Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA USA ,grid.38142.3c000000041936754XStem Cell and Regenerative Biology, Harvard University, Cambridge, USA ,grid.7737.40000 0004 0410 2071Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Jaakko Kaprio
- grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland ,grid.7737.40000 0004 0410 2071Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Samuli Ripatti
- grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland ,grid.66859.34Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA USA ,grid.32224.350000 0004 0386 9924Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, USA ,grid.7737.40000 0004 0410 2071Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Mark Daly
- grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland ,grid.66859.34Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA USA ,grid.32224.350000 0004 0386 9924Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, USA
| | - Aarno Palotie
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland. .,Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA, USA. .,Analytic and Translational Genetics Unit, Department of Medicine, Department of Neurology and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
47
|
Kimura H, Mori D, Aleksic B, Ozaki N. Elucidation of molecular pathogenesis and drug development for psychiatric disorders from rare disease-susceptibility variants. Neurosci Res 2020; 170:24-31. [PMID: 33316300 DOI: 10.1016/j.neures.2020.11.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/19/2020] [Accepted: 11/26/2020] [Indexed: 10/22/2022]
Abstract
Recent rapid progress in genome analysis and large-scale consortia has made it possible to discover variants with a variety of allele frequencies and effect sizes associated with psychiatric disorders. Among psychiatric disorder-susceptibility variants, rare variants with large effect sizes detected by sequencing analysis or array comparative genomic hybridization would be particularly useful for elucidating pathophysiology by developing disease models, such as genome-edited mouse or induced pluripotent stem cells. In the last decade, investigations of rare variants with large effect size have revealed an important role of neurodevelopment in the pathogenesis of psychiatric disorders. In future research, integration of recent evidence concerning the contribution of the immune system or gut microbiota will enhance our understanding of psychiatric disorders and facilitate novel drug development.
Collapse
Affiliation(s)
- Hiroki Kimura
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Daisuke Mori
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan; Brain & Mind Research Center, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Branko Aleksic
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan; Brain & Mind Research Center, Nagoya University Graduate School of Medicine, Nagoya, Japan; Medical Genomics Center, Nagoya University Hospital, Nagoya, Japan
| |
Collapse
|
48
|
Martin CL, Wain KE, Oetjens MT, Tolwinski K, Palen E, Hare-Harris A, Habegger L, Maxwell EK, Reid JG, Walsh LK, Myers SM, Ledbetter DH. Identification of Neuropsychiatric Copy Number Variants in a Health Care System Population. JAMA Psychiatry 2020; 77:1276-1285. [PMID: 32697297 PMCID: PMC7376464 DOI: 10.1001/jamapsychiatry.2020.2159] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPORTANCE Population screening for medically relevant genomic variants that cause diseases such as hereditary cancer and cardiovascular disorders is increasing to facilitate early disease detection or prevention. Neuropsychiatric disorders (NPDs) are common, complex disorders with clear genetic causes; yet, access to genetic diagnosis is limited. We explored whether inclusion of NPD in population-based genomic screening programs is warranted by assessing 3 key factors: prevalence, penetrance, and personal utility. OBJECTIVE To evaluate the suitability of including pathogenic copy number variants (CNVs) associated with NPD in population screening by determining their prevalence and penetrance and exploring the personal utility of disclosing results. DESIGN, SETTING, AND PARTICIPANTS In this cohort study, the frequency of 31 NPD CNVs was determined in patient-participants via exome data. Associated clinical phenotypes were assessed using linked electronic health records. Nine CNVs were selected for disclosure by licensed genetic counselors, and participants' psychosocial reactions were evaluated using a mixed-methods approach. A primarily adult population receiving medical care at Geisinger, a large integrated health care system in the United States with the only population-based genomic screening program approved for medically relevant results disclosure, was included. The cohort was identified from the Geisinger MyCode Community Health Initiative. Exome and linked electronic health record data were available for this cohort, which was recruited from February 2007 to April 2017. Data were collected for the qualitative analysis April 2017 through February 2018. Analysis began February 2018 and ended December 2019. MAIN OUTCOMES AND MEASURES The planned outcomes of this study include (1) prevalence estimate of NPD-associated CNVs in an unselected health care system population; (2) penetrance estimate of NPD diagnoses in CNV-positive individuals; and (3) qualitative themes that describe participants' responses to receiving NPD-associated genomic results. RESULTS Of 90 595 participants with CNV data, a pathogenic CNV was identified in 708 (0.8%; 436 women [61.6%]; mean [SD] age, 50.04 [18.74] years). Seventy percent (n = 494) had at least 1 associated clinical symptom. Of these, 28.8% (204) of CNV-positive individuals had an NPD code in their electronic health record, compared with 13.3% (11 835 of 89 887) of CNV-negative individuals (odds ratio, 2.21; 95% CI, 1.86-2.61; P < .001); 66.4% (470) of CNV-positive individuals had a history of depression and anxiety compared with 54.6% (49 118 of 89 887) of CNV-negative individuals (odds ratio, 1.53; 95% CI, 1.31-1.80; P < .001). 16p13.11 (71 [0.078%]) and 22q11.2 (108 [0.119%]) were the most prevalent deletions and duplications, respectively. Only 5.8% of individuals (41 of 708) had a previously known genetic diagnosis. Results disclosure was completed for 141 individuals. Positive participant responses included poignant reactions to learning a medical reason for lifelong cognitive and psychiatric disabilities. CONCLUSIONS AND RELEVANCE This study informs critical factors central to the development of population-based genomic screening programs and supports the inclusion of NPD in future designs to promote equitable access to clinically useful genomic information.
Collapse
Affiliation(s)
- Christa Lese Martin
- Autism & Developmental Medicine Institute, Geisinger, Danville, Pennsylvania
| | - Karen E. Wain
- Autism & Developmental Medicine Institute, Geisinger, Danville, Pennsylvania
| | - Matthew T. Oetjens
- Autism & Developmental Medicine Institute, Geisinger, Danville, Pennsylvania
| | - Kasia Tolwinski
- Autism & Developmental Medicine Institute, Geisinger, Danville, Pennsylvania,Biomedical Ethics Unit, McGill University, Montreal, Quebec, Canada
| | - Emily Palen
- Autism & Developmental Medicine Institute, Geisinger, Danville, Pennsylvania
| | | | | | | | | | | | - Scott M. Myers
- Autism & Developmental Medicine Institute, Geisinger, Danville, Pennsylvania
| | - David H. Ledbetter
- Autism & Developmental Medicine Institute, Geisinger, Danville, Pennsylvania
| |
Collapse
|
49
|
Watson CJ, Thomas RH, Solomon T, Michael BD, Nicholson TR, Pollak TA. COVID-19 and psychosis risk: Real or delusional concern? Neurosci Lett 2020; 741:135491. [PMID: 33220366 DOI: 10.1016/j.neulet.2020.135491] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023]
Abstract
Historical epidemiological perspectives from past pandemics and recent neurobiological evidence link infections and psychoses, leading to concerns that COVID-19 will present a significant risk for the development of psychosis. But are these concerns justified, or mere sensationalism? In this article we review the historical associations between viral infection and the immune system more broadly in the development of psychosis, before critically evaluating the current evidence pertaining to SARS-CoV-2 and risk of psychosis as an acute or post-infectious manifestation of COVID-19. We review the 42 cases of psychosis reported in infected patients to date, and discuss the potential implications of in utero infection on subsequent neurodevelopment and psychiatric risk. Finally, in the context of the wider neurological and psychiatric manifestations of COVID-19 and our current understanding of the aetiology of psychotic disorders, we evaluate possible neurobiological and psychosocial mechanisms as well as the numerous challenges in ascribing a causal pathogenic role to the infection.
Collapse
Affiliation(s)
- Cameron J Watson
- Preventive Neurology Unit, Wolfson Institute of Preventive Medicine, Queen Mary University London, UK.
| | - Rhys H Thomas
- Translational and Clinical Research Institute, University of Newcastle, UK; Royal Victoria Infirmary, Newcastle, UK
| | - Tom Solomon
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, UK; National Institute for Health Research Health Protection Research Unit in Emerging and Zoonotic Infection, University of Liverpool, UK; Clinical Infection Microbiology and Immunology, Institute of Infection, Veterinary, and Zoological Science, University of Liverpool, UK
| | - Benedict Daniel Michael
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, UK; National Institute for Health Research Health Protection Research Unit in Emerging and Zoonotic Infection, University of Liverpool, UK; Clinical Infection Microbiology and Immunology, Institute of Infection, Veterinary, and Zoological Science, University of Liverpool, UK
| | - Timothy R Nicholson
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Thomas A Pollak
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| |
Collapse
|
50
|
Implications of germline copy-number variations in psychiatric disorders: review of large-scale genetic studies. J Hum Genet 2020; 66:25-37. [PMID: 32958875 DOI: 10.1038/s10038-020-00838-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/28/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023]
Abstract
Copy number variants (CNVs), defined as genome sequences of ≥50 bp that differ in copy number from that in a reference genome, are a common form of structural variation. Germline CNVs account for some of the missing heritability that single nucleotide polymorphisms could not account for. Recent technological advances have had a huge impact on CNV research. Microarray technology enables relatively low-cost, high-throughput, genome-wide measurements, and short-read sequencing technology enables the detection of short CNVs that cannot be detected by microarrays. As a result, large-scale genetic studies have been able to identify a variety of common and rare germline CNVs and their associations with diseases. Rare germline CNVs have been reported to be associated with neuropsychiatric disorders. In this review, we focused on germline CNVs and briefly described their functional characteristics, formation mechanisms, detection methods, related databases, and the latest findings. Finally, we introduced recent large-scale genetic studies to assess associations of CNVs with diseases, especially psychiatric disorders, and discussed the use of CNV-based animal models to investigate the molecular and cellular mechanisms underlying these disorders. The development and implementation of improved detection methods, such as long-read single-molecule sequencing, are expected to provide additional insight into the molecular basis of psychiatric disorders and other complex diseases, thus facilitating basic and clinical research on CNVs.
Collapse
|