1
|
Jaspers Faijer-Westerink H, von Scheibler ENMM, van Rossum EFC, van Haelst MM, Vingerhoets C, van Amelsvoort TAMJ, van Eeghen AM, Boot E. Obesity and metabolic syndrome in adults with a 22q11.2 microdeletion. Int J Obes (Lond) 2025; 49:642-648. [PMID: 39616274 PMCID: PMC11999860 DOI: 10.1038/s41366-024-01685-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 04/17/2025]
Abstract
OBJECTIVE Copy number variations (CNVs) may contribute to medical conditions. However, research on the impact of individual CNVs on endocrine disease is limited. This study aimed to provide new data on obesity and metabolic syndrome (MetS) in adults with microdeletion 22q11.2, the pathogenic CNV associated with 22q11.2 deletion syndrome. METHODS We examined prevalence rates of obesity and MetS in 103 adults with a typical 22q11.2 deletion (45.2% male, at median age 30.0 (range 17-71) years) and compared these rates with population-based data. Generalized obesity was defined by a body mass index (BMI) ≥ 30 kg/m2, abdominal obesity by a waist circumference (WC) of ≥102 cm in males and ≥88 cm in females, and MetS by standard Joint Interim Statement criteria. General linear models were used to examine the independent associations of age, sex, congenital heart defect, smoking, and antipsychotic use with BMI, WC, and the presence of MetS. RESULTS Prevalence rates of generalized obesity (32.0%), abdominal obesity (51.5%), and MetS (33.0%) were significantly higher compared to a population-based cohort (15.7% (P < 0.0001), 36.1% (P = 0.002), and 15.2% (P < 0.0001), respectively). In antipsychotic naïve subjects, significant correlations were observed between age and BMI (r = 0.54, P < 0.001), and age and WC (r = 0.60, P < 0.001). These correlations were not present in individuals taking antipsychotic medication. The models predicting BMI (F(5, 97) = 3.083, R2 = 0.137, P = 0.01) and WC (F(5, 92) = 5.985, R2 = 0.245, P < 0.001) were significant. Only age was individually predictive of outcomes (P < 0.05 and P < 0.001). The model predicting MetS was also significant (P < 0.001), with higher age being the only factor associated with MetS (OR = 1.07, 95% CI = 1.03-1.12, P < 0.001). CONCLUSIONS Generalized and abdominal obesity, as well as MetS, appear to be common in adults with 22q11.2 deletion syndrome, emphasizing the importance of careful monitoring from a young age. These findings contribute to the limited knowledge about the association between pathogenic CNVs, obesity, and MetS.
Collapse
Affiliation(s)
| | - Emma N M M von Scheibler
- Koraal, Maastricht, The Netherlands
- Department of Psychiatry and Neuropsychology, MHeNs, Maastricht University, Maastricht, The Netherlands
| | - Elisabeth F C van Rossum
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Obesity Center CGG, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Mieke M van Haelst
- Department of Human Genetics, Amsterdam Reproduction & Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Emma Children's Hospital, University of Amsterdam, Amsterdam, The Netherlands
| | - Claudia Vingerhoets
- Advisium, 's Heeren Loo Zorggroep, Amersfoort, The Netherlands
- Department of Psychiatry and Neuropsychology, MHeNs, Maastricht University, Maastricht, The Netherlands
| | | | - Agnies M van Eeghen
- Advisium, 's Heeren Loo Zorggroep, Amersfoort, The Netherlands
- Emma Children's Hospital, University of Amsterdam, Amsterdam, The Netherlands
| | - Erik Boot
- Advisium, 's Heeren Loo Zorggroep, Amersfoort, The Netherlands.
- Department of Psychiatry and Neuropsychology, MHeNs, Maastricht University, Maastricht, The Netherlands.
- The Dalglish Family 22q Clinic, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
2
|
Van L, Heung T, Reyes NGD, Boot E, Chow EWC, Corral M, Bassett AS. Real-World Treatment of Schizophrenia in Adults With a 22q11.2 Microdeletion: Traitement dans le monde réel de la schizophrénie chez des adultes atteints du syndrome de microdélétion 22q11.2. CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 2025; 70:160-170. [PMID: 39641288 PMCID: PMC11624517 DOI: 10.1177/07067437241293983] [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] [Indexed: 12/07/2024]
Abstract
OBJECTIVE One in every 4 individuals born with a 22q11.2 microdeletion will develop schizophrenia. Thirty years of clinical genetic testing capability have enabled detection of this major molecular susceptibility for psychotic illness. However, there is limited literature on the treatment of schizophrenia in individuals with a 22q11.2 microdeletion, particularly regarding the issue of treatment resistance. METHODS From a large, well-characterized adult cohort with a typical 22q11.2 microdeletion followed for up to 25 years at a specialty clinic, we studied all 107 adults (49 females, 45.8%) meeting the criteria for schizophrenia or schizoaffective disorder. We performed a comprehensive review of lifetime (1,801 patient-years) psychiatric records to determine treatments used and the prevalence of treatment-resistant schizophrenia (TRS). We used Clinical Global Impression-Improvement (CGI-I) scores to compare within-individual responses to clozapine and nonclozapine antipsychotics. For a subgroup with contemporary data (n = 88, 82.2%), we examined antipsychotics and dosage at the last follow-up. RESULTS Lifetime treatments involved on average 4 different antipsychotic medications per individual. Sixty-three (58.9%) individuals met the study criteria for TRS, a significantly greater proportion than for a community-based comparison (42.9%; χ2 = 10.38, df = 1, p < 0.01). The non-TRS group was enriched for individuals with genetic diagnosis before schizophrenia diagnosis. Within-person treatment response in TRS was significantly better for clozapine than for nonclozapine antipsychotics (p < 0.0001). At the last follow-up, clozapine was the most common antipsychotic prescribed, followed by olanzapine, risperidone, and paliperidone. Total antipsychotic chlorpromazine equivalent dosages were in typical clinical ranges (median: 450 mg; interquartile range: 300, 750 mg). CONCLUSION The results for this large sample indicate that patients with 22q11.2 microdeletion have an increased propensity to treatment resistance. The findings provide evidence about how genetic diagnosis can inform clinical psychiatric management and could help reduce treatment delays. Further research is needed to shed light on the pathophysiology of antipsychotic response and on strategies to optimize outcomes. PLAIN LANGUAGE SUMMARY TITLE Real-world treatment of schizophrenia in adults with a 22q11.2 microdeletion.
Collapse
Affiliation(s)
- Lily Van
- The Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Tracy Heung
- The Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- The Dalglish Family 22q Clinic, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Nikolai Gil D. Reyes
- The Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- The Dalglish Family 22q Clinic, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Erik Boot
- The Dalglish Family 22q Clinic, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
- Advisium, 's Heeren Loo Zorggroep, Amersfoort, the Netherlands
- Department of Psychiatry and Neuropsychology, MHeNs, Maastricht University, Maastricht, the Netherlands
| | - Eva W. C. Chow
- The Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Maria Corral
- The Dalglish Family 22q Clinic, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Anne S. Bassett
- The Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- The Dalglish Family 22q Clinic, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
- Toronto Congenital Cardiac Centre for Adults, and Division of Cardiology, Department of Medicine, University Health Network, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute and Campbell Family Mental Health Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Doyle AE, Bearden CE, Gur RE, Ledbetter DH, Martin CL, McCoy TH, Pasaniuc B, Perlis RH, Smoller JW, Davis LK. Advancing Mental Health Research Through Strategic Integration of Transdiagnostic Dimensions and Genomics. Biol Psychiatry 2025; 97:450-460. [PMID: 39424167 DOI: 10.1016/j.biopsych.2024.10.006] [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: 12/18/2023] [Revised: 09/11/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
Genome-wide studies are yielding a growing catalog of common and rare variants that confer risk for psychopathology. However, despite representing unprecedented progress, emerging data also indicate that the full promise of psychiatric genetics-including understanding pathophysiology and improving personalized care-will not be fully realized by targeting traditional dichotomous diagnostic categories. The current article provides reflections on themes that emerged from a 2021 National Institute of Mental Health-sponsored conference convened to address strategies for the evolving field of psychiatric genetics. As anticipated by the National Institute of Mental Health's Research Domain Criteria framework, multilevel investigations of dimensional and transdiagnostic phenotypes, particularly when integrated with biobanks and big data, will be critical to advancing knowledge. The path forward will also require more diverse representation in source studies. Additionally, progress will be catalyzed by a range of converging approaches, including capitalizing on computational methods, pursuing biological insights, working within a developmental framework, and engaging health care systems and patient communities.
Collapse
Affiliation(s)
- Alysa E Doyle
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts.
| | - Carrie E Bearden
- Departments of Psychiatry and Biobehavioral Sciences & Psychology, University of California at Los Angeles, Los Angeles, California
| | - Raquel E Gur
- Departments of Psychiatry, Neurology and Radiology, Perelman School of Medicine, University of Pennsylvania, and the Lifespan Brain Institute of Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, Pennsylvania
| | - David H Ledbetter
- Departments of Pediatrics and Psychiatry, University of Florida College of Medicine, Jacksonville, Florida
| | - Christa L Martin
- Geisinger Autism & Developmental Medicine Institute, Lewisburg, Pennsylvania
| | - Thomas H McCoy
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Bogdan Pasaniuc
- Departments of Computational Medicine, Pathology and Laboratory Medicine, and Human Genetics, University of California at Los Angeles, Los Angeles, California
| | - Roy H Perlis
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jordan W Smoller
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Lea K Davis
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
4
|
Kushima I, Nakatochi M, Ozaki N. Copy Number Variations and Human Well-Being: Integrating Psychiatric, Physical, and Socioeconomic Perspectives. Biol Psychiatry 2024:S0006-3223(24)01788-8. [PMID: 39643102 DOI: 10.1016/j.biopsych.2024.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 11/12/2024] [Accepted: 11/30/2024] [Indexed: 12/09/2024]
Abstract
Copy number variations (CNVs) have emerged as crucial genetic factors that influence a wide spectrum of human health outcomes, with particularly strong associations to psychiatric disorders. In this review, we present a synthesis of diverse impacts of psychiatric disorder-associated CNVs on neurodevelopment, brain function, and physical health across the lifespan. Large-scale studies have revealed that CNV carriers exhibit an increased risk for psychiatric disorders, cognitive deficits, sleep disturbances, neurological disorders, and other physical conditions, including cardiovascular diseases, diabetes, and renal disease, highlighting the wide-ranging impact of CNVs beyond the brain. Neuroimaging studies have revealed substantial CNV effects on brain structure, from cortical and subcortical alterations to white matter microstructure, with effect sizes often exceeding those observed in idiopathic psychiatric disorders. Cellular and animal models have begun to elucidate dynamic CNV effects on neurodevelopment, neuronal function, and cellular energy metabolism, while revealing complex CNV-environment interactions and cell type-specific responses, particularly in studies of 22q11.2 deletion syndrome. This review also explores the complex interplay between psychiatric and physical health conditions in CNV carriers and how these interactions contribute to adverse socioeconomic outcomes, including reduced educational attainment and income levels, creating a feedback loop that further impacts health outcomes. Finally, in this review, we also highlight research limitations and propose key priorities for clinical implementation, including the need for longitudinal studies, standardized guidelines for CNV result reporting and genetic counseling, and integrated care networks to provide a foundation for advancing the field of precision psychiatry.
Collapse
Affiliation(s)
- Itaru Kushima
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan; Medical Genomics Center, Nagoya University Hospital, Nagoya, Japan.
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norio Ozaki
- Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Japan; Institute for Glyco-core Research, Nagoya University, Nagoya, Japan
| |
Collapse
|
5
|
Choudhary A, Peles D, Nayak R, Mizrahi L, Stern S. Current progress in understanding schizophrenia using genomics and pluripotent stem cells: A meta-analytical overview. Schizophr Res 2024; 273:24-38. [PMID: 36443183 DOI: 10.1016/j.schres.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/16/2022] [Accepted: 11/01/2022] [Indexed: 11/27/2022]
Abstract
Schizophrenia (SCZ) is a complex, heritable and polygenic neuropsychiatric disease, which disables the patients as well as decreases their life expectancy and quality of life. Common and rare variants studies on SCZ subjects have provided >100 genomic loci that hold importance in the context of SCZ pathophysiology. Transcriptomic studies from clinical samples have informed about the differentially expressed genes (DEGs) and non-coding RNAs in SCZ patients. Despite these advancements, no causative genes for SCZ were found and hence SCZ is difficult to recapitulate in animal models. In the last decade, induced Pluripotent Stem Cells (iPSCs)-based models have helped in understanding the neural phenotypes of SCZ by studying patient iPSC-derived 2D neuronal cultures and 3D brain organoids. Here, we have aimed to provide a simplistic overview of the current progress and advancements after synthesizing the enormous literature on SCZ genetics and SCZ iPSC-based models. Although further understanding of SCZ genetics and pathophysiological mechanisms using these technological advancements is required, the recent approaches have allowed to delineate important cellular mechanisms and biological pathways affected in SCZ.
Collapse
Affiliation(s)
- Ashwani Choudhary
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - David Peles
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Ritu Nayak
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Liron Mizrahi
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel.
| |
Collapse
|
6
|
Muhtaseb AW, Duan J. Modeling common and rare genetic risk factors of neuropsychiatric disorders in human induced pluripotent stem cells. Schizophr Res 2024; 273:39-61. [PMID: 35459617 PMCID: PMC9735430 DOI: 10.1016/j.schres.2022.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 12/13/2022]
Abstract
Recent genome-wide association studies (GWAS) and whole-exome sequencing of neuropsychiatric disorders, especially schizophrenia, have identified a plethora of common and rare disease risk variants/genes. Translating the mounting human genetic discoveries into novel disease biology and more tailored clinical treatments is tied to our ability to causally connect genetic risk variants to molecular and cellular phenotypes. When combined with the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated (Cas) nuclease-mediated genome editing system, human induced pluripotent stem cell (hiPSC)-derived neural cultures (both 2D and 3D organoids) provide a promising tractable cellular model for bridging the gap between genetic findings and disease biology. In this review, we first conceptualize the advances in understanding the disease polygenicity and convergence from the past decade of iPSC modeling of different types of genetic risk factors of neuropsychiatric disorders. We then discuss the major cell types and cellular phenotypes that are most relevant to neuropsychiatric disorders in iPSC modeling. Finally, we critically review the limitations of iPSC modeling of neuropsychiatric disorders and outline the need for implementing and developing novel methods to scale up the number of iPSC lines and disease risk variants in a systematic manner. Sufficiently scaled-up iPSC modeling and a better functional interpretation of genetic risk variants, in combination with cutting-edge CRISPR/Cas9 gene editing and single-cell multi-omics methods, will enable the field to identify the specific and convergent molecular and cellular phenotypes in precision for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Abdurrahman W Muhtaseb
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, United States of America; Department of Human Genetics, The University of Chicago, Chicago, IL 60637, United States of America
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, United States of America; Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL 60637, United States of America.
| |
Collapse
|
7
|
Jensen M, Smolen C, Tyryshkina A, Pizzo L, Banerjee D, Oetjens M, Shimelis H, Taylor CM, Pounraja VK, Song H, Rohan L, Huber E, El Khattabi L, van de Laar I, Tadros R, Bezzina C, van Slegtenhorst M, Kammeraad J, Prontera P, Caberg JH, Fraser H, Banka S, Van Dijck A, Schwartz C, Voorhoeve E, Callier P, Mosca-Boidron AL, Marle N, Lefebvre M, Pope K, Snell P, Boys A, Lockhart PJ, Ashfaq M, McCready E, Nowacyzk M, Castiglia L, Galesi O, Avola E, Mattina T, Fichera M, Bruccheri MG, Mandarà GML, Mari F, Privitera F, Longo I, Curró A, Renieri A, Keren B, Charles P, Cuinat S, Nizon M, Pichon O, Bénéteau C, Stoeva R, Martin-Coignard D, Blesson S, Le Caignec C, Mercier S, Vincent M, Martin C, Mannik K, Reymond A, Faivre L, Sistermans E, Kooy RF, Amor DJ, Romano C, Andrieux J, Girirajan S. Genetic modifiers and ascertainment drive variable expressivity of complex disorders. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.27.24312158. [PMID: 39252907 PMCID: PMC11383473 DOI: 10.1101/2024.08.27.24312158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Variable expressivity of disease-associated variants implies a role for secondary variants that modify clinical features. We assessed the effects of modifier variants towards clinical outcomes of 2,252 individuals with primary variants. Among 132 families with the 16p12.1 deletion, distinct rare and common variant classes conferred risk for specific developmental features, including short tandem repeats for neurological defects and SNVs for microcephaly, while additional disease-associated variants conferred multiple genetic diagnoses. Within disease and population cohorts of 773 individuals with the 16p12.1 deletion, we found opposing effects of secondary variants towards clinical features across ascertainments. Additional analysis of 1,479 probands with other primary variants, such as 16p11.2 deletion and CHD8 variants, and 1,084 without primary variants, showed that phenotypic associations differed by primary variant context and were influenced by synergistic interactions between primary and secondary variants. Our study provides a paradigm to dissect the genomic architecture of complex disorders towards personalized treatment.
Collapse
Affiliation(s)
- Matthew Jensen
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- Bioinformatics and Genomics Graduate program, Pennsylvania State University, University Park, PA 16802, USA
| | - Corrine Smolen
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- Bioinformatics and Genomics Graduate program, Pennsylvania State University, University Park, PA 16802, USA
| | - Anastasia Tyryshkina
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Lucilla Pizzo
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Deepro Banerjee
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Matthew Oetjens
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA 17837, USA
| | - Hermela Shimelis
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA 17837, USA
| | - Cora M. Taylor
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA 17837, USA
| | - Vijay Kumar Pounraja
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- Bioinformatics and Genomics Graduate program, Pennsylvania State University, University Park, PA 16802, USA
| | - Hyebin Song
- Department of Statistics, Pennsylvania State University, University Park, PA 16802, USA
| | - Laura Rohan
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Emily Huber
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Laila El Khattabi
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris Cité, CARPEM, Paris, France
| | - Ingrid van de Laar
- Department of Clinical Genetics, Erasmus MC, Univ. Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rafik Tadros
- Department of Clinical Genetics, Erasmus MC, Univ. Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Connie Bezzina
- Department of Clinical Genetics, Erasmus MC, Univ. Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marjon van Slegtenhorst
- Department of Clinical Genetics, Erasmus MC, Univ. Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Janneke Kammeraad
- Department of Clinical Genetics, Erasmus MC, Univ. Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Paolo Prontera
- Medical Genetics Unit, Hospital Santa Maria della Misericordia, Perugia, Italy
| | - Jean-Hubert Caberg
- Centre Hospitalier Universitaire de Liège. Domaine Universitaire du Sart Tilman, Liège, Belgium
| | - Harry Fraser
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Siddhartha Banka
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, St. Mary’s Hospital, Central Manchester University Hospitals, NHS Foundation Trust Manchester Academic Health Sciences Centre, Manchester, UK
| | - Anke Van Dijck
- Department of Medical Genetics, University and University Hospital Antwerp, Antwerp, Belgium
| | | | - Els Voorhoeve
- Department of Clinical Genetics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Patrick Callier
- Center for Rare Diseases and Reference Developmental Anomalies and Malformation Syndromes, CHU Dijon, Dijon, France
| | - Anne-Laure Mosca-Boidron
- Center for Rare Diseases and Reference Developmental Anomalies and Malformation Syndromes, CHU Dijon, Dijon, France
| | - Nathalie Marle
- Center for Rare Diseases and Reference Developmental Anomalies and Malformation Syndromes, CHU Dijon, Dijon, France
| | - Mathilde Lefebvre
- Laboratoire de Genetique Chromosomique et Moleculaire, CHU Dijon, France
| | - Kate Pope
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Melbourne, Australia
| | - Penny Snell
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Melbourne, Australia
| | - Amber Boys
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Melbourne, Australia
| | - Paul J. Lockhart
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Myla Ashfaq
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Elizabeth McCready
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Margaret Nowacyzk
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Lucia Castiglia
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
| | - Ornella Galesi
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
| | - Emanuela Avola
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
| | - Teresa Mattina
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
| | - Marco Fichera
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
- Section of Clinical Biochemistry and Medical Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania School of Medicine, Catania, Italy
| | - Maria Grazia Bruccheri
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
| | | | - Francesca Mari
- Laboratory of Clinical Molecular Genetics and Cytogenetics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Flavia Privitera
- Laboratory of Clinical Molecular Genetics and Cytogenetics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ilaria Longo
- Laboratory of Clinical Molecular Genetics and Cytogenetics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Aurora Curró
- Laboratory of Clinical Molecular Genetics and Cytogenetics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Renieri
- Laboratory of Clinical Molecular Genetics and Cytogenetics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Boris Keren
- Département de Génétique, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, 75019 Paris, France
| | - Perrine Charles
- Département de Génétique, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, 75019 Paris, France
| | | | | | | | | | - Radka Stoeva
- CHU Nantes, Medical Genetics Department, Nantes, France
| | | | - Sophia Blesson
- Department of Genetics, Bretonneau University Hospital, Tours, France
| | - Cedric Le Caignec
- CHU Toulouse, Department of Medical Genetics, Toulouse, France
- Toulouse Neuro Imaging, Center, Inserm, UPS, Université de Toulouse, Toulouse, France
| | - Sandra Mercier
- Department of Genetics, Bretonneau University Hospital, Tours, France
| | - Marie Vincent
- Department of Genetics, Bretonneau University Hospital, Tours, France
| | - Christa Martin
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA 17837, USA
| | - Katrin Mannik
- Institute of Genomics, University of Tartu, Estonia
- Health2030 Genome Center, Fondation Campus Biotech, Geneva, Switzerland
| | - Alexandre Reymond
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Switzerland
| | - Laurence Faivre
- Center for Rare Diseases and Reference Developmental Anomalies and Malformation Syndromes, CHU Dijon, Dijon, France
- Laboratoire de Genetique Chromosomique et Moleculaire, CHU Dijon, France
| | - Erik Sistermans
- Department of Clinical Genetics, Amsterdam UMC, Amsterdam, The Netherlands
| | - R. Frank Kooy
- Department of Medical Genetics, University and University Hospital Antwerp, Antwerp, Belgium
| | - David J. Amor
- Department of Clinical Genetics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Corrado Romano
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
- Section of Clinical Biochemistry and Medical Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania School of Medicine, Catania, Italy
| | - Joris Andrieux
- Institut de Genetique Medicale, Hopital Jeanne de Flandre, CHRU de Lille, Lille, France
| | - Santhosh Girirajan
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- Bioinformatics and Genomics Graduate program, Pennsylvania State University, University Park, PA 16802, USA
- Department of Anthropology, Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
8
|
Sun Y, Zhang Y, Lu Z, Liao Y, Feng Q, Yu M, Chen Y, Kang Z, Feng X, Zhao G, Sun J, Yang Y, Guo L, Zhang D, Bi W, Huang H, Yue W. Contribution of copy number variants on antipsychotic treatment response in Han Chinese patients with schizophrenia. EBioMedicine 2024; 105:105195. [PMID: 38870545 PMCID: PMC11225184 DOI: 10.1016/j.ebiom.2024.105195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 05/17/2024] [Accepted: 05/28/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Response to antipsychotic drugs (APD) varies greatly among individuals and is affected by genetic factors. This study aims to demonstrate genome-wide associations between copy number variants (CNVs) and response to APD in patients with schizophrenia. METHODS A total of 3030 patients of Han Chinese ethnicity randomly received APD (aripiprazole, olanzapine, quetiapine, risperidone, ziprasidone, haloperidol and perphenazine) treatment for six weeks. This study is a secondary data analysis. Percentage change on the Positive and Negative Syndrome Scale (PANSS) reduction was used to assess APD efficacy, and more than 50% change was considered as APD response. Associations between CNV burden, gene set, CNV loci and CNV break-point and APD efficacy were analysed. FINDINGS Higher CNV losses burden decreased the odds of 6-week APD response (OR = 0.66 [0.44, 0.98]). CNV losses in synaptic pathway involved in neurotransmitters were associated with 2-week PANSS reduction rate. CNV involved in sialylation (1p31.1 losses) and cellular metabolism (19q13.32 gains) associated with 6-week PANSS reduction rate at genome-wide significant level. Additional 36 CNVs associated with PANSS factors improvement. The OR of protective CNVs for 6-week APD response was 3.10 (95% CI: 1.33-7.19) and risk CNVs was 8.47 (95% CI: 1.92-37.43). CNV interacted with genetic risk score on APD efficacy (Beta = -1.53, SE = 0.66, P = 0.021). The area under curve to differ 6-week APD response attained 80.45% (95% CI: 78.07%-82.82%). INTERPRETATION Copy number variants contributed to poor APD efficacy and synaptic pathway involved in neurotransmitter was highlighted. FUNDING National Natural Science Foundation of China, National Key R&D Program of China, China Postdoctoral Science Foundation.
Collapse
Affiliation(s)
- Yaoyao Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Yuyanan Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Zhe Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Yundan Liao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Qidi Feng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mingrui Yu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yu Chen
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Zhewei Kang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Xiaoyang Feng
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Guorui Zhao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Junyuan Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Yang Yang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Liangkun Guo
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Dai Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China; Chinese Institute for Brain Research, Beijing 102206, China
| | - Wenjian Bi
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Hailiang Huang
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Weihua Yue
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| |
Collapse
|
9
|
Enokida T, Yoshida N, Tatsumi M, Hidese S, Goto YI, Hoshino M, Kunugi H, Hattori K. Neuronal autoantibodies in the cerebrospinal fluid of 148 patients with schizophrenia and 151 healthy controls. Heliyon 2024; 10:e30695. [PMID: 38770306 PMCID: PMC11103440 DOI: 10.1016/j.heliyon.2024.e30695] [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: 01/18/2024] [Revised: 04/24/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024] Open
Abstract
Schizophrenia is a syndrome with multiple etiologies, one of which is the potential for an autoimmune disease of the brain such as N-methyl-d-aspartate receptor (NMDAR) encephalitis, which can induce psychosis resembling schizophrenia. Here, we examined anti-neuronal autoantibodies related to psychosis using both cell- (CBA) and tissue-based assays (TBA) in the cerebrospinal fluid (CSF) of patients with chronic schizophrenia and control participants. First, we screened for the antibodies against leucine-rich glioma-inactivated 1 (LGI1), γ-aminobutyric acid B receptor (GABABR), dipeptidyl aminopeptidase-like protein 6 (DPPX), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR1/R2), and contactin-associated protein-like 2 (CASPR2) in 148 patients with schizophrenia. No antibodies were detected. Next, we performed CBA for NMDAR antibodies in 148 patients with schizophrenia and 151 age- and sex-matched controls. Although we detected relatively weak immunoreactivity for NMDAR in the CSFs of two patients with schizophrenia and three controls, no samples were positive when strict criteria were applied. For TBA in the rat hippocampus and cerebellum, we detected positive signals in the CSFs of 13 patients with schizophrenia and eight controls. Positive samples were analyzed for paraneoplastic syndrome and antinuclear antibodies using immunoblotting. The CSFs of nine patients and six controls were positive for dense fine speckle 70 (DFS70) antibodies. Additionally, antibodies against centromere protein (CENP)-A and CENP-B were detected in patients with schizophrenia. Our results suggest that autoantibodies against NMDAR, LG1, GABABR, DPPX, AMPAR1/R2, and CASPR2 are not associated with the pathogenesis of chronic schizophrenia. Moreover, we emphasize the importance of considering the effect of anti-DFS70 antibodies when analyzing autoantibodies in CSF samples. Conclusively, we obtained no evidence suggesting that the most frequent neuronal autoantibodies in the CSF play a role in the pathogenesis of schizophrenia, even in our sample.
Collapse
Affiliation(s)
- Takako Enokida
- Department of Bioresources, Medical Genome Center, National Center of Neurology and Psychiatry, 4-1-1, Ogawahigashi, Kodaira, Tokyo, 187-8502, Japan
- Department of NCNP Brain Physiology and Pathology, Cognitive and Behavioral Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Nanako Yoshida
- Department of Bioresources, Medical Genome Center, National Center of Neurology and Psychiatry, 4-1-1, Ogawahigashi, Kodaira, Tokyo, 187-8502, Japan
| | - Megumi Tatsumi
- Department of Bioresources, Medical Genome Center, National Center of Neurology and Psychiatry, 4-1-1, Ogawahigashi, Kodaira, Tokyo, 187-8502, Japan
| | - Shinsuke Hidese
- Department of Psychiatry, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Yu-ichi Goto
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1, Ogawahigashi, Kodaira, Tokyo, 187-8502, Japan
| | - Mikio Hoshino
- Department of NCNP Brain Physiology and Pathology, Cognitive and Behavioral Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1, Ogawahigashi, Kodaira, Tokyo, 187-8502, Japan
| | - Hiroshi Kunugi
- Department of Psychiatry, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-Ku, Tokyo, 173-8605, Japan
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1, Ogawahigashi, Kodaira, Tokyo, 187-8502, Japan
| | - Kotaro Hattori
- Department of Bioresources, Medical Genome Center, National Center of Neurology and Psychiatry, 4-1-1, Ogawahigashi, Kodaira, Tokyo, 187-8502, Japan
| |
Collapse
|
10
|
Besterman AD. A genetics-guided approach to the clinical management of schizophrenia. Schizophr Res 2024; 267:462-469. [PMID: 37813777 DOI: 10.1016/j.schres.2023.09.042] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 09/27/2023] [Accepted: 09/29/2023] [Indexed: 10/11/2023]
Abstract
Schizophrenia is a highly heritable, severe mental illness characterized by hallucinations, delusions, social withdrawal, and cognitive dysfunction present in ∼1% of populations across cultures. There have been recent major advancements in our understanding of the genetic architecture of schizophrenia. Both rare, highly penetrant genetic variants as well as common, low-penetrant genetic variants can predispose individuals to schizophrenia and can impact the way people metabolize psychoactive medications used to treat schizophrenia. However, the impact of these findings on the clinical management of schizophrenia remains limited. This review highlights the few places where genetics currently informs schizophrenia management strategies, discusses major limitations, and reviews promising areas of genetics research that are most likely to impact future schizophrenia care. Specifically, I focuss on psychiatric genetic counseling, genetic testing strategies, pharmacogenetics, polygenic risk, and genetics-guided treatment. Lastly, I emphasize important ethical considerations in the clinical use of genetics for schizophrenia management, including the exacerbation of healthcare inequalities and unintended consequences of new genetic technologies.
Collapse
Affiliation(s)
- Aaron D Besterman
- University of California San Diego, Department of Psychiatry, San Diego, CA, USA; Rady Children's Hospital San Diego, Division of Behavioral Health Services, San Diego, CA, USA; Rady Children's Institute for Genomic Medicine, San Diego, CA, USA.
| |
Collapse
|
11
|
Deng X, Lu S, Li Y, Fang X, Zhang R, Shen X, Du J, Xie S. Association between increased BMI and cognitive function in first-episode drug-naïve male schizophrenia. Front Psychiatry 2024; 15:1362674. [PMID: 38505798 PMCID: PMC10948420 DOI: 10.3389/fpsyt.2024.1362674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 02/21/2024] [Indexed: 03/21/2024] Open
Abstract
Objective Although the adverse effects of obesity in schizophrenia are documented, there is limited research exists on the implications for untreated initial schizophrenia. Our investigation aimed to explore the connections between BMI and cognitive function in first-episode drug-naïve (FEDN)schizophrenia. Methods We enrolled 143 FEDN schizophrenia patients, and collected data on their body mass index, fasting blood glucose and lipid levels. Cognitive function was measured with the MATRICS Consensus Cognitive Battery (MCCB). Using correlation and regression analysis to assess the relationship between BMI and cognitive performance. Results The prevalence rate of overweight plus obesity in FEDN schizophrenia patients was 33.57%. Patients with FEDN schizophrenia exhibited extensive cognitive impairment, and those who were overweight/obesity demonstrated more severe impairments in working memory and visual learning when compared to normal/under weight counterparts. Correlation analysis indicated a negative association between working memory and BMI and TG, as well as a link between visual learning and BMI and LDL-C. Multiple linear regression analysis revealed that a higher BMI predicted a decrease in working memory in FEDN schizophrenia patients. Conclusion Our results indicate that the rate of overweight plus obesity is high in FEDN schizophrenia patients, and there is an association between BMI and cognitive function in schizophrenia, particularly in relation to working memory.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jinglun Du
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Shiping Xie
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
12
|
Racedo SE, Liu Y, Shi L, Zheng D, Morrow BE. Dgcr8 functions in the secondary heart field for outflow tract and right ventricle development in mammals. Dev Biol 2024; 506:72-84. [PMID: 38110169 PMCID: PMC10793380 DOI: 10.1016/j.ydbio.2023.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/28/2023] [Accepted: 12/11/2023] [Indexed: 12/20/2023]
Abstract
The DGCR8 gene, encoding a critical miRNA processing protein, maps within the hemizygous region in patients with 22q11.2 deletion syndrome. Most patients have malformations of the cardiac outflow tract that is derived in part from the anterior second heart field (aSHF) mesoderm. To understand the function of Dgcr8 in the aSHF, we inactivated it in mice using Mef2c-AHF-Cre. Inactivation resulted in a fully penetrant persistent truncus arteriosus and a hypoplastic right ventricle leading to lethality by E14.5. To understand the molecular mechanism for this phenotype, we performed gene expression profiling of the aSHF and the cardiac outflow tract with right ventricle in conditional null versus normal mouse littermates at stage E9.5 prior to morphology changes. We identified dysregulation of mRNA gene expression, of which some are relevant to cardiogenesis. Many pri-miRNA genes were strongly increased in expression in mutant embryos along with reduced expression of mature miRNA genes. We further examined the individual, mature miRNAs that were decreased in expression along with pri-miRNAs that were accumulated that could be direct effects due to loss of Dgcr8. Among these genes, were miR-1a, miR-133a, miR-134, miR143 and miR145a, which have known functions in heart development. These early mRNA and miRNA changes may in part, explain the first steps that lead to the resulting phenotype in Dgcr8 aSHF conditional mutant embryos.
Collapse
Affiliation(s)
- Silvia E Racedo
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yang Liu
- Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Bell Buckle, TN, USA
| | - Lijie Shi
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bernice E Morrow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA; Departments of Pediatrics and Ob/Gyn & Population Health, USA.
| |
Collapse
|
13
|
Tandon R, Nasrallah H, Akbarian S, Carpenter WT, DeLisi LE, Gaebel W, Green MF, Gur RE, Heckers S, Kane JM, Malaspina D, Meyer-Lindenberg A, Murray R, Owen M, Smoller JW, Yassin W, Keshavan M. The schizophrenia syndrome, circa 2024: What we know and how that informs its nature. Schizophr Res 2024; 264:1-28. [PMID: 38086109 DOI: 10.1016/j.schres.2023.11.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/23/2023] [Accepted: 11/29/2023] [Indexed: 03/01/2024]
Abstract
With new data about different aspects of schizophrenia being continually generated, it becomes necessary to periodically revisit exactly what we know. Along with a need to review what we currently know about schizophrenia, there is an equal imperative to evaluate the construct itself. With these objectives, we undertook an iterative, multi-phase process involving fifty international experts in the field, with each step building on learnings from the prior one. This review assembles currently established findings about schizophrenia (construct, etiology, pathophysiology, clinical expression, treatment) and posits what they reveal about its nature. Schizophrenia is a heritable, complex, multi-dimensional syndrome with varying degrees of psychotic, negative, cognitive, mood, and motor manifestations. The illness exhibits a remitting and relapsing course, with varying degrees of recovery among affected individuals with most experiencing significant social and functional impairment. Genetic risk factors likely include thousands of common genetic variants that each have a small impact on an individual's risk and a plethora of rare gene variants that have a larger individual impact on risk. Their biological effects are concentrated in the brain and many of the same variants also increase the risk of other psychiatric disorders such as bipolar disorder, autism, and other neurodevelopmental conditions. Environmental risk factors include but are not limited to urban residence in childhood, migration, older paternal age at birth, cannabis use, childhood trauma, antenatal maternal infection, and perinatal hypoxia. Structural, functional, and neurochemical brain alterations implicate multiple regions and functional circuits. Dopamine D-2 receptor antagonists and partial agonists improve psychotic symptoms and reduce risk of relapse. Certain psychological and psychosocial interventions are beneficial. Early intervention can reduce treatment delay and improve outcomes. Schizophrenia is increasingly considered to be a heterogeneous syndrome and not a singular disease entity. There is no necessary or sufficient etiology, pathology, set of clinical features, or treatment that fully circumscribes this syndrome. A single, common pathophysiological pathway appears unlikely. The boundaries of schizophrenia remain fuzzy, suggesting the absence of a categorical fit and need to reconceptualize it as a broader, multi-dimensional and/or spectrum construct.
Collapse
Affiliation(s)
- Rajiv Tandon
- Department of Psychiatry, WMU Homer Stryker School of Medicine, Kalamazoo, MI 49008, United States of America.
| | - Henry Nasrallah
- Department of Psychiatry, University of Cincinnati College of Medicine Cincinnati, OH 45267, United States of America
| | - Schahram Akbarian
- Department of Psychiatry, Icahn School of Medicine at Mt. Sinai, New York, NY 10029, United States of America
| | - William T Carpenter
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| | - Lynn E DeLisi
- Department of Psychiatry, Cambridge Health Alliance and Harvard Medical School, Cambridge, MA 02139, United States of America
| | - Wolfgang Gaebel
- Department of Psychiatry and Psychotherapy, LVR-Klinikum Dusseldorf, Heinrich-Heine University, Dusseldorf, Germany
| | - Michael F Green
- Department of Psychiatry and Biobehavioral Sciences, Jane and Terry Semel Institute of Neuroscience and Human Behavior, UCLA, Los Angeles, CA 90024, United States of America; Greater Los Angeles Veterans' Administration Healthcare System, United States of America
| | - Raquel E Gur
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States of America
| | - Stephan Heckers
- Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN 37232, United States of America
| | - John M Kane
- Department of Psychiatry, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Glen Oaks, NY 11004, United States of America
| | - Dolores Malaspina
- Department of Psychiatry, Neuroscience, Genetics, and Genomics, Icahn School of Medicine at Mt. Sinai, New York, NY 10029, United States of America
| | - Andreas Meyer-Lindenberg
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Mannhein/Heidelberg University, Mannheim, Germany
| | - Robin Murray
- Institute of Psychiatry, Psychology, and Neuroscience, Kings College, London, UK
| | - Michael Owen
- Centre for Neuropsychiatric Genetics and Genomics, and Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Jordan W Smoller
- Center for Precision Psychiatry, Department of Psychiatry, Psychiatric and Neurodevelopmental Unit, Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States of America
| | - Walid Yassin
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, United States of America
| | - Matcheri Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, United States of America
| |
Collapse
|
14
|
Stefanski A, Pérez-Palma E, Brünger T, Montanucci L, Gati C, Klöckner C, Johannesen KM, Goodspeed K, Macnee M, Deng AT, Aledo-Serrano Á, Borovikov A, Kava M, Bouman AM, Hajianpour MJ, Pal DK, Engelen M, Hagebeuk EEO, Shinawi M, Heidlebaugh AR, Oetjens K, Hoffman TL, Striano P, Freed AS, Futtrup L, Balslev T, Abulí A, Danvoye L, Lederer D, Balci T, Nouri MN, Butler E, Drewes S, van Engelen K, Howell KB, Khoury J, May P, Trinidad M, Froelich S, Lemke JR, Tiller J, Freed AN, Kang JQ, Wuster A, Møller RS, Lal D. SLC6A1 variant pathogenicity, molecular function and phenotype: a genetic and clinical analysis. Brain 2023; 146:5198-5208. [PMID: 37647852 PMCID: PMC10689929 DOI: 10.1093/brain/awad292] [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: 12/16/2022] [Revised: 06/05/2023] [Accepted: 07/08/2023] [Indexed: 09/01/2023] Open
Abstract
Genetic variants in the SLC6A1 gene can cause a broad phenotypic disease spectrum by altering the protein function. Thus, systematically curated clinically relevant genotype-phenotype associations are needed to understand the disease mechanism and improve therapeutic decision-making. We aggregated genetic and clinical data from 172 individuals with likely pathogenic/pathogenic (lp/p) SLC6A1 variants and functional data for 184 variants (14.1% lp/p). Clinical and functional data were available for a subset of 126 individuals. We explored the potential associations of variant positions on the GAT1 3D structure with variant pathogenicity, altered molecular function and phenotype severity using bioinformatic approaches. The GAT1 transmembrane domains 1, 6 and extracellular loop 4 (EL4) were enriched for patient over population variants. Across functionally tested missense variants (n = 156), the spatial proximity from the ligand was associated with loss-of-function in the GAT1 transporter activity. For variants with complete loss of in vitro GABA uptake, we found a 4.6-fold enrichment in patients having severe disease versus non-severe disease (P = 2.9 × 10-3, 95% confidence interval: 1.5-15.3). In summary, we delineated associations between the 3D structure and variant pathogenicity, variant function and phenotype in SLC6A1-related disorders. This knowledge supports biology-informed variant interpretation and research on GAT1 function. All our data can be interactively explored in the SLC6A1 portal (https://slc6a1-portal.broadinstitute.org/).
Collapse
Affiliation(s)
- Arthur Stefanski
- Genomic Medicine Institute and Epilepsy Center, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Eduardo Pérez-Palma
- Universidad del Desarrollo, Centro de Genética y Genómica, Facultad de Medicina Clínica Alemana, Santiago de Chile 7610658, Chile
| | - Tobias Brünger
- Cologne Center for Genomics (CCG), Medical Faculty of the University of Cologne, University Hospital of Cologne, Cologne 50931, Germany
| | - Ludovica Montanucci
- Genomic Medicine Institute and Epilepsy Center, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Cornelius Gati
- Department of Biological Sciences, Bridge Institute, USC Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA
| | - Chiara Klöckner
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig 04103, Germany
| | - Katrine M Johannesen
- Department of Epilepsy Genetics and Personalized Medicine, The Danish Epilepsy Centre, Dianalund 4293, Denmark
- Department of Genetics, University Hospital of Copenhagen, Rigshispitalet, Copenhagen 2100, Denmark
| | - Kimberly Goodspeed
- Children’s Health, Medical Center, Dallas, TX 75235, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Marie Macnee
- Cologne Center for Genomics (CCG), Medical Faculty of the University of Cologne, University Hospital of Cologne, Cologne 50931, Germany
| | - Alexander T Deng
- Clinical Genetics, Guys and St Thomas NHS Trust, London SE19RT, UK
| | - Ángel Aledo-Serrano
- Epilepsy Program, Neurology Department, Hospital Ruber Internacional, Madrid 28034, Spain
| | - Artem Borovikov
- Research and Counseling Department, Research Centre for Medical Genetics, Moscow 115478, Russia
| | - Maina Kava
- Department of Neurology and Metabolic Medicine, Perth Children’s Hospital, Perth 6009, Australia
- School of Paediatrics and Child Health, UWA Medical School, University of Western Australia, Perth 6009, Australia
| | - Arjan M Bouman
- Department of Clinical Genetics, Erasmus MC, University Medical Center, Rotterdam 3015GD, The Netherlands
| | - M J Hajianpour
- Department of Pediatrics, Division of Medical Genetics and Genomics, Albany Medical College, Albany Med Health System, Albany, NY 12208, USA
| | - Deb K Pal
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College, London SE58AF, UK
- Department of Basic and Clinical Neurosciences, King’s College Hospital, London SE59RS, UK
| | - Marc Engelen
- Department of Pediatric Neurology, Amsterdam Public Health, Amsterdam University Medical Center, Amsterdam 1081HV, The Netherlands
| | - Eveline E O Hagebeuk
- Department of Pediatric Neurology, Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede and Zwolle 2103SW, The Netherlands
| | - Marwan Shinawi
- Division of Genetics and Genomic Medicine, Department of Pediatrics, St.Louis Children’s Hospital, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Kathryn Oetjens
- Autism and Developmental Medicine Institute, Geisinger, Danville, PA 17837, USA
| | - Trevor L Hoffman
- Department of Regional Genetics, Anaheim, Southern California Kaiser Permanente Medical Group, CA 92806, USA
| | - Pasquale Striano
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, Genoa 16147, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa 16132, Italy
| | - Amanda S Freed
- Department of Clinical Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA 91101, USA
| | - Line Futtrup
- Department of Paediatrics, Regional Hospital of Central Jutland, Viborg 8800, Denmark
| | - Thomas Balslev
- Department of Paediatrics, Regional Hospital of Central Jutland, Viborg 8800, Denmark
- Centre for Educational Development, Aarhus University, Aarhus 8200, Denmark
| | - Anna Abulí
- Department of Clinical and Molecular Genetics and Medicine Genetics Group, VHIR, University Hospital Vall d’Hebron, Barcelona 08035, Spain
| | - Leslie Danvoye
- Department of Neurology, Université catholique de Louvain, Cliniques universitaires Saint-Luc, Brussels 1200, Belgium
| | - Damien Lederer
- Centre for Human Genetics, Institute for Pathology and Genetics, Gosselies 6041, Belgium
| | - Tugce Balci
- Department of Pediatrics, Division of Medical Genetics, Western University, London, ON N6A3K7, Canada
- Medical Genetics Program of Southwestern Ontario, London Health Sciences Centre and Children's Health Research Institute, London, ON N6A5A5, Canada
| | - Maryam Nabavi Nouri
- Department of Paediatrics, Division of Pediatric Neurology, London Health Sciences Centre, London, ON N6A5W9, Canada
| | | | - Sarah Drewes
- Department of Medical Genetics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Kalene van Engelen
- Medical Genetics Program of Southwestern Ontario, London Health Sciences Centre, London, ON N6A5W9, Canada
| | - Katherine B Howell
- Department of Neurology, Royal Children’s Hospital, Melbourne, VIC 3052, Australia
- Department of Pediatrics, University of Melbourne, Melbourne, VIC 3052, Australia
- Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
| | - Jean Khoury
- Genomic Medicine Institute and Epilepsy Center, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Patrick May
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette 4362, Luxembourg
| | - Marena Trinidad
- Translational Genomics, BioMarin Pharmaceutical Inc., Novato, CA 94949, USA
| | - Steven Froelich
- Translational Genomics, BioMarin Pharmaceutical Inc., Novato, CA 94949, USA
| | - Johannes R Lemke
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig 04103, Germany
- Center for Rare Diseases, University of Leipzig Medical Center, Leipzig 04103, Germany
| | | | | | - Jing-Qiong Kang
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37240, USA
- Neuroscience Graduate Program, Vanderbilt University, Nashville, TN 37235, USA
- Department of Neurology, Vanderbilt Brain Institute, Nashville, TN 37235, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Kennedy Center of Human Development, Nashville, TN 37203, USA
| | - Arthur Wuster
- Translational Genomics, BioMarin Pharmaceutical Inc., Novato, CA 94949, USA
| | - Rikke S Møller
- Department of Epilepsy Genetics and Personalized Medicine, The Danish Epilepsy Centre, Dianalund 4293, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense 5000, Denmark
| | - Dennis Lal
- Genomic Medicine Institute and Epilepsy Center, Cleveland Clinic, Cleveland, OH 44195, USA
- Stanley Center of Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Neurology, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
15
|
Kolar D, Krajcovic B, Kleteckova L, Kuncicka D, Vales K, Brozka H. Review: Genes Involved in Mitochondrial Physiology Within 22q11.2 Deleted Region and Their Relevance to Schizophrenia. Schizophr Bull 2023; 49:1637-1653. [PMID: 37379469 PMCID: PMC10686339 DOI: 10.1093/schbul/sbad066] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
BACKGROUND AND HYPOTHESIS Schizophrenia is associated with altered energy metabolism, but the cause and potential impact of these metabolic changes remain unknown. 22q11.2 deletion syndrome (22q11.2DS) represents a genetic risk factor for schizophrenia, which is associated with the loss of several genes involved in mitochondrial physiology. Here we examine how the haploinsufficiency of these genes could contribute to the emergence of schizophrenia in 22q11.2DS. STUDY DESIGN We characterize changes in neuronal mitochondrial function caused by haploinsufficiency of mitochondria-associated genes within the 22q11.2 region (PRODH, MRPL40, TANGO2, ZDHHC8, SLC25A1, TXNRD2, UFD1, and DGCR8). For that purpose, we combine data from 22q11.2DS carriers and schizophrenia patients, in vivo (animal models) and in vitro (induced pluripotent stem cells, IPSCs) studies. We also review the current knowledge about seven non-coding microRNA molecules located in the 22q11.2 region that may be indirectly involved in energy metabolism by acting as regulatory factors. STUDY RESULTS We found that the haploinsufficiency of genes of interest is mainly associated with increased oxidative stress, altered energy metabolism, and calcium homeostasis in animal models. Studies on IPSCs from 22q11.2DS carriers corroborate findings of deficits in the brain energy metabolism, implying a causal role between impaired mitochondrial function and the development of schizophrenia in 22q11.2DS. CONCLUSIONS The haploinsufficiency of genes within the 22q11.2 region leads to multifaceted mitochondrial dysfunction with consequences to neuronal function, viability, and wiring. Overlap between in vitro and in vivo studies implies a causal role between impaired mitochondrial function and the development of schizophrenia in 22q11.2DS.
Collapse
Affiliation(s)
- David Kolar
- National Institute of Mental Health, Klecany, Czech Republic
| | - Branislav Krajcovic
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Daniela Kuncicka
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Karel Vales
- National Institute of Mental Health, Klecany, Czech Republic
| | - Hana Brozka
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
16
|
Ying S, Heung T, Thiruvahindrapuram B, Engchuan W, Yin Y, Blagojevic C, Zhang Z, Hegele RA, Yuen RKC, Bassett AS. Polygenic risk for triglyceride levels in the presence of a high impact rare variant. BMC Med Genomics 2023; 16:281. [PMID: 37940981 PMCID: PMC10634078 DOI: 10.1186/s12920-023-01717-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 10/25/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Elevated triglyceride (TG) levels are a heritable and modifiable risk factor for cardiovascular disease and have well-established associations with common genetic variation captured in a polygenic risk score (PRS). In young adulthood, the 22q11.2 microdeletion conveys a 2-fold increased risk for mild-moderate hypertriglyceridemia. This study aimed to assess the role of the TG-PRS in individuals with this elevated baseline risk for mild-moderate hypertriglyceridemia. METHODS We studied a deeply phenotyped cohort of adults (n = 157, median age 34 years) with a 22q11.2 microdeletion and available genome sequencing, lipid level, and other clinical data. The association between a previously developed TG-PRS and TG levels was assessed using a multivariable regression model adjusting for effects of sex, BMI, and other covariates. We also constructed receiver operating characteristic (ROC) curves using logistic regression models to assess the ability of TG-PRS and significant clinical variables to predict mild-moderate hypertriglyceridemia status. RESULTS The TG-PRS was a significant predictor of TG-levels (p = 1.52E-04), along with male sex and BMI, in a multivariable model (pmodel = 7.26E-05). The effect of TG-PRS appeared to be slightly stronger in individuals with obesity (BMI ≥ 30) (beta = 0.4617) than without (beta = 0.1778), in a model unadjusted for other covariates (p-interaction = 0.045). Among ROC curves constructed, the inclusion of TG-PRS, sex, and BMI as predictor variables produced the greatest area under the curve (0.749) for classifying those with mild-moderate hypertriglyceridemia, achieving an optimal sensitivity and specificity of 0.746 and 0.707, respectively. CONCLUSIONS These results demonstrate that in addition to significant effects of sex and BMI, genome-wide common variation captured in a PRS also contributes to the variable expression of the 22q11.2 microdeletion with respect to elevated TG levels.
Collapse
Affiliation(s)
- Shengjie Ying
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Tracy Heung
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, ON, Canada
- The Dalglish Family 22Q Clinic, University Health Network, Toronto, ON, Canada
| | | | - Worrawat Engchuan
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Yue Yin
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Christina Blagojevic
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Zhaolei Zhang
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Robert A Hegele
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Ryan K C Yuen
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Anne S Bassett
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- The Dalglish Family 22Q Clinic, University Health Network, Toronto, ON, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
- Toronto General Hospital Research Institute and Campbell Family Mental Health Research Institute, Toronto, ON, Canada.
| |
Collapse
|
17
|
Modasi J, Khachadourian V, O'Hora K, Kushan L, Slavich GM, Shields GS, Velthorst E, Bearden CE. Associations between acute and chronic lifetime stressors and psychosis-risk symptoms in individuals with 22q11.2 copy number variants. Psychol Med 2023; 53:7222-7231. [PMID: 37078394 PMCID: PMC10719673 DOI: 10.1017/s0033291723000740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 02/28/2023] [Accepted: 03/06/2023] [Indexed: 04/21/2023]
Abstract
BACKGROUND The 22q11.2 deletion (22q11Del) is among the strongest known genetic risk factors for psychosis. Stress, a known risk factor for psychosis in the general population, has seldom been studied in 22q11Del. We investigated how lifetime stressors related to symptomatic outcomes in patients with 22q11Del. We also explored this association in individuals with 22q11.2 duplications (22q11Dup), which may be potentially protective against psychosis. METHOD One hundred individuals (46 with 22q11Del, 30 with 22q11Dup, and 24 healthy controls; Mage = 17.30 years±10.15) were included. Logistic models were used to examine cross-sectional associations between lifetime acute and chronic stressors (severity and count) and the presence (score ⩾3) of positive, negative, and general symptoms, assessed via the Structured Interview for Psychosis-risk Syndromes (SIPS). RESULTS The 22q11Dup group reported the greatest number and severity of acute lifetime stressors, but did not differ from 22q11Del in chronic stressor count or severity. Lifetime chronic and acute stressors were uniquely associated with positive symptoms in 22q11Del (chronic count: odds ratio [OR] = 2.35, p = 0.02; chronic severity: OR = 1.88, p = 0.03; acute count: OR = 1.78, p = 0.03), but not with negative or general symptoms (ps > 0.05). CONCLUSION Findings suggest that stress may play a role in psychotic symptoms in 22q1Del, while the 22q11Dup CNV appears protective against psychotic symptoms despite higher rates of stressors. Interventions that mitigate effects of stressors in 22qDel may reduce the odds of psychosis in this group. Prospective longitudinal research is needed to replicate these findings.
Collapse
Affiliation(s)
- Jasmine Modasi
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vahe Khachadourian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kathleen O'Hora
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
- Interdepartmental Program for Neuroscience, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Leila Kushan
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - George M. Slavich
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Grant S. Shields
- Department of Psychological Science, University of Arkansas, Fayetteville, AR, USA
| | - Eva Velthorst
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carrie E. Bearden
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
- Department of Psychology, University of California, Los Angeles, CA, USA
| |
Collapse
|
18
|
Forsyth JK, Bearden CE. Rethinking the First Episode of Schizophrenia: Identifying Convergent Mechanisms During Development and Moving Toward Prediction. Am J Psychiatry 2023; 180:792-804. [PMID: 37908094 DOI: 10.1176/appi.ajp.20230736] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Affiliation(s)
- Jennifer K Forsyth
- Department of Psychology, University of Washington, Seattle (Forsyth); Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Behavioral Sciences, and Department of Psychology, University of California, Los Angeles (Bearden)
| | - Carrie E Bearden
- Department of Psychology, University of Washington, Seattle (Forsyth); Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Behavioral Sciences, and Department of Psychology, University of California, Los Angeles (Bearden)
| |
Collapse
|
19
|
Stein A, Zhu C, Du F, Öngür D. Magnetic Resonance Spectroscopy Studies of Brain Energy Metabolism in Schizophrenia: Progression from Prodrome to Chronic Psychosis. Curr Psychiatry Rep 2023; 25:659-669. [PMID: 37812338 DOI: 10.1007/s11920-023-01457-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 10/10/2023]
Abstract
PURPOSE OF REVIEW Schizophrenia (SZ) is a debilitating mental illness; existing treatments are partially effective and associated with significant side effect burden, largely due to our limited understanding of disease mechanisms and the trajectory of disease progression. Accumulating evidence suggests that metabolic changes associated with glucose metabolism, mitochondrial dysfunction, and redox imbalance play an important role in the pathophysiology of schizophrenia. However, the molecular mechanisms associated with these abnormalities in the brains of schizophrenia patients and the ways in which they change over time remain unclear. This paper aims to review the current literature on molecular mechanisms and in vivo magnetic resonance spectroscopy (MRS) studies of impaired energy metabolism in patients at clinical high risk for psychosis, with first-episode SZ, and with chronic SZ. Our review covers research related to high-energy phosphate metabolism, lactate, intracellular pH, redox ratio, and the antioxidant glutathione. RECENT FINDINGS Both first-episode and chronic SZ patients display a significant reduction in creatine kinase reaction activity and redox (NAD + /NADH) ratio in the prefrontal cortex. Chronic, but not first-episode, SZ patients also show a trend toward increased lactate levels and decreased pH value. These findings suggest a progressive shift from oxidative phosphorylation to glycolysis for energy production over the course of SZ, which is associated with redox imbalance and mitochondrial dysfunction. Accumulating evidence indicates that aberrant brain energy metabolism associated with mitochondrial dysfunction and redox imbalance plays a critical role in SZ and will be a promising target for future treatments.
Collapse
Affiliation(s)
- Abigail Stein
- Psychotic Disorders Division, McLean Hospital, Belmont, 02478, USA
- McLean Imaging Center, McLean Hospital, Belmont, 02478, USA
| | - Chenyanwen Zhu
- Psychotic Disorders Division, McLean Hospital, Belmont, 02478, USA
- McLean Imaging Center, McLean Hospital, Belmont, 02478, USA
| | - Fei Du
- Psychotic Disorders Division, McLean Hospital, Belmont, 02478, USA.
- McLean Imaging Center, McLean Hospital, Belmont, 02478, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| | - Dost Öngür
- Psychotic Disorders Division, McLean Hospital, Belmont, 02478, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
20
|
Mollon J, Schultz LM, Huguet G, Knowles EEM, Mathias SR, Rodrigue A, Alexander-Bloch A, Saci Z, Jean-Louis M, Kumar K, Douard E, Almasy L, Jacquemont S, Glahn DC. Impact of Copy Number Variants and Polygenic Risk Scores on Psychopathology in the UK Biobank. Biol Psychiatry 2023; 94:591-600. [PMID: 36764568 PMCID: PMC10409883 DOI: 10.1016/j.biopsych.2023.01.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/11/2023]
Abstract
BACKGROUND Our understanding of the impact of copy number variants (CNVs) on psychopathology and their joint influence with polygenic risk scores (PRSs) remains limited. METHODS The UK Biobank recruited 502,534 individuals ages 37 to 73 years living in the United Kingdom between 2006 and 2010. After quality control, genotype data from 459,855 individuals were available for CNV calling. A total of 61 commonly studied recurrent neuropsychiatric CNVs were selected for analyses and examined individually and in aggregate (any CNV, deletion, or duplication). CNV risk scores were used to quantify intolerance of CNVs to haploinsufficiency. Major depressive disorder and generalized anxiety disorder PRSs were generated for White British individuals (N = 408,870). Mood/anxiety factor scores were generated using item-level questionnaire data (N = 501,289). RESULTS CNV carriers showed higher mood/anxiety scores than noncarriers, with the largest effects seen for intolerant deletions. A total of 11 individual deletions and 8 duplications were associated with higher mood/anxiety. Carriers of the 9p24.3 (DMRT1) duplication showed lower mood/anxiety. Associations remained significant for most CNVs when excluding individuals with psychiatric diagnoses. Nominally significant CNV × PRS interactions provided preliminary evidence that associations between select individual CNVs, but not CNVs in aggregate, and mood/anxiety may be modulated by PRSs. CONCLUSIONS CNVs associated with risk for psychiatric disorders showed small to large effects on dimensional mood/anxiety scores in a general population cohort, even when excluding individuals with psychiatric diagnoses. CNV × PRS interactions showed that associations between select CNVs and mood/anxiety may be modulated by PRSs.
Collapse
Affiliation(s)
- Josephine Mollon
- Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Laura M Schultz
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Guillaume Huguet
- Department of Pediatrics, Université de Montréal, Montreal, Quebec, Canada; Department of Pediatrics, Center Hospitalier Universitaire Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - Emma E M Knowles
- Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Samuel R Mathias
- Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Amanda Rodrigue
- Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Aaron Alexander-Bloch
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Neurodevelopment and Psychosis Section, Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Zohra Saci
- Department of Pediatrics, Center Hospitalier Universitaire Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - Martineau Jean-Louis
- Department of Pediatrics, Center Hospitalier Universitaire Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - Kuldeep Kumar
- Department of Pediatrics, Center Hospitalier Universitaire Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - Elise Douard
- Department of Pediatrics, Université de Montréal, Montreal, Quebec, Canada; Department of Pediatrics, Center Hospitalier Universitaire Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - Laura Almasy
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Genetics, Perelman School of Medicine, Penn-CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sebastien Jacquemont
- Department of Pediatrics, Université de Montréal, Montreal, Quebec, Canada; Department of Pediatrics, Center Hospitalier Universitaire Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - David C Glahn
- Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; Olin Neuropsychiatry Research Center, Institute of Living, Hartford, Connecticut
| |
Collapse
|
21
|
Owen MJ, Legge SE, Rees E, Walters JTR, O'Donovan MC. Genomic findings in schizophrenia and their implications. Mol Psychiatry 2023; 28:3638-3647. [PMID: 37853064 PMCID: PMC10730422 DOI: 10.1038/s41380-023-02293-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/20/2023]
Abstract
There has been substantial progress in understanding the genetics of schizophrenia over the past 15 years. This has revealed a highly polygenic condition with the majority of the currently explained heritability coming from common alleles of small effect but with additional contributions from rare copy number and coding variants. Many specific genes and loci have been implicated that provide a firm basis upon which mechanistic research can proceed. These point to disturbances in neuronal, and particularly synaptic, functions that are not confined to a small number of brain regions and circuits. Genetic findings have also revealed the nature of schizophrenia's close relationship to other conditions, particularly bipolar disorder and childhood neurodevelopmental disorders, and provided an explanation for how common risk alleles persist in the population in the face of reduced fecundity. Current genomic approaches only potentially explain around 40% of heritability, but only a small proportion of this is attributable to robustly identified loci. The extreme polygenicity poses challenges for understanding biological mechanisms. The high degree of pleiotropy points to the need for more transdiagnostic research and the shortcomings of current diagnostic criteria as means of delineating biologically distinct strata. It also poses challenges for inferring causality in observational and experimental studies in both humans and model systems. Finally, the Eurocentric bias of genomic studies needs to be rectified to maximise benefits and ensure these are felt across diverse communities. Further advances are likely to come through the application of new and emerging technologies, such as whole-genome and long-read sequencing, to large and diverse samples. Substantive progress in biological understanding will require parallel advances in functional genomics and proteomics applied to the brain across developmental stages. For these efforts to succeed in identifying disease mechanisms and defining novel strata they will need to be combined with sufficiently granular phenotypic data.
Collapse
Affiliation(s)
- Michael J Owen
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK.
| | - Sophie E Legge
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Elliott Rees
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - James T R Walters
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Michael C O'Donovan
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK.
| |
Collapse
|
22
|
Montanucci L, Lewis-Smith D, Collins RL, Niestroj LM, Parthasarathy S, Xian J, Ganesan S, Macnee M, Brünger T, Thomas RH, Talkowski M, Helbig I, Leu C, Lal D. Genome-wide identification and phenotypic characterization of seizure-associated copy number variations in 741,075 individuals. Nat Commun 2023; 14:4392. [PMID: 37474567 PMCID: PMC10359300 DOI: 10.1038/s41467-023-39539-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 06/16/2023] [Indexed: 07/22/2023] Open
Abstract
Copy number variants (CNV) are established risk factors for neurodevelopmental disorders with seizures or epilepsy. With the hypothesis that seizure disorders share genetic risk factors, we pooled CNV data from 10,590 individuals with seizure disorders, 16,109 individuals with clinically validated epilepsy, and 492,324 population controls and identified 25 genome-wide significant loci, 22 of which are novel for seizure disorders, such as deletions at 1p36.33, 1q44, 2p21-p16.3, 3q29, 8p23.3-p23.2, 9p24.3, 10q26.3, 15q11.2, 15q12-q13.1, 16p12.2, 17q21.31, duplications at 2q13, 9q34.3, 16p13.3, 17q12, 19p13.3, 20q13.33, and reciprocal CNVs at 16p11.2, and 22q11.21. Using genetic data from additional 248,751 individuals with 23 neuropsychiatric phenotypes, we explored the pleiotropy of these 25 loci. Finally, in a subset of individuals with epilepsy and detailed clinical data available, we performed phenome-wide association analyses between individual CNVs and clinical annotations categorized through the Human Phenotype Ontology (HPO). For six CNVs, we identified 19 significant associations with specific HPO terms and generated, for all CNVs, phenotype signatures across 17 clinical categories relevant for epileptologists. This is the most comprehensive investigation of CNVs in epilepsy and related seizure disorders, with potential implications for clinical practice.
Collapse
Affiliation(s)
- Ludovica Montanucci
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, USA
| | - David Lewis-Smith
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Clinical Neurosciences, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- The Epilepsy NeuroGenetics Initiative, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ryan L Collins
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, USA
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (M.I.T.) and Harvard, Cambridge, USA
| | | | - Shridhar Parthasarathy
- The Epilepsy NeuroGenetics Initiative, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Julie Xian
- The Epilepsy NeuroGenetics Initiative, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Shiva Ganesan
- The Epilepsy NeuroGenetics Initiative, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Marie Macnee
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Tobias Brünger
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Rhys H Thomas
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Clinical Neurosciences, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Michael Talkowski
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, USA
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (M.I.T.) and Harvard, Cambridge, USA
| | - Ingo Helbig
- The Epilepsy NeuroGenetics Initiative, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Costin Leu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, USA.
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London, UK.
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and M.I.T, Cambridge, MA, USA.
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, US.
| | - Dennis Lal
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, USA.
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (M.I.T.) and Harvard, Cambridge, USA.
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and M.I.T, Cambridge, MA, USA.
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, US.
| |
Collapse
|
23
|
Sharo AG, Zou Y, Adhikari AN, Brenner SE. ClinVar and HGMD genomic variant classification accuracy has improved over time, as measured by implied disease burden. Genome Med 2023; 15:51. [PMID: 37443081 PMCID: PMC10347827 DOI: 10.1186/s13073-023-01199-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 05/31/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Curated databases of genetic variants assist clinicians and researchers in interpreting genetic variation. Yet, these databases contain some misclassified variants. It is unclear whether variant misclassification is abating as these databases rapidly grow and implement new guidelines. METHODS Using archives of ClinVar and HGMD, we investigated how variant misclassification has changed over 6 years, across different ancestry groups. We considered inborn errors of metabolism (IEMs) screened in newborns as a model system because these disorders are often highly penetrant with neonatal phenotypes. We used samples from the 1000 Genomes Project (1KGP) to identify individuals with genotypes that were classified by the databases as pathogenic. Due to the rarity of IEMs, nearly all such classified pathogenic genotypes indicate likely variant misclassification in ClinVar or HGMD. RESULTS While the false-positive rates of both ClinVar and HGMD have improved over time, HGMD variants currently imply two orders of magnitude more affected individuals in 1KGP than ClinVar variants. We observed that African ancestry individuals have a significantly increased chance of being incorrectly indicated to be affected by a screened IEM when HGMD variants are used. However, this bias affecting genomes of African ancestry was no longer significant once common variants were removed in accordance with recent variant classification guidelines. We discovered that ClinVar variants classified as Pathogenic or Likely Pathogenic are reclassified sixfold more often than DM or DM? variants in HGMD, which has likely resulted in ClinVar's lower false-positive rate. CONCLUSIONS Considering misclassified variants that have since been reclassified reveals our increasing understanding of rare genetic variation. We found that variant classification guidelines and allele frequency databases comprising genetically diverse samples are important factors in reclassification. We also discovered that ClinVar variants common in European and South Asian individuals were more likely to be reclassified to a lower confidence category, perhaps due to an increased chance of these variants being classified by multiple submitters. We discuss features for variant classification databases that would support their continued improvement.
Collapse
Affiliation(s)
- Andrew G. Sharo
- Biophysics Graduate Group, University of California, Berkeley, CA 94720 USA
- Center for Computational Biology, University of California, Berkeley, CA 94720 USA
- Department of Ecology and Evolutionary Biology, University of California, 124 Biomed Building, 1156 High St., Santa Cruz, CA 95064 USA
| | - Yangyun Zou
- Center for Computational Biology, University of California, Berkeley, CA 94720 USA
- Department of Plant and Microbial Biology, University of California, 461 Koshland Hall, Berkeley, CA 94720 USA
- Currently at: Department of Clinical Research, Yikon Genomics Company, Ltd., Shanghai, China
| | - Aashish N. Adhikari
- Center for Computational Biology, University of California, Berkeley, CA 94720 USA
- Department of Plant and Microbial Biology, University of California, 461 Koshland Hall, Berkeley, CA 94720 USA
- Currently at: Illumina, Foster City, CA 94404 USA
| | - Steven E. Brenner
- Biophysics Graduate Group, University of California, Berkeley, CA 94720 USA
- Center for Computational Biology, University of California, Berkeley, CA 94720 USA
- Department of Plant and Microbial Biology, University of California, 461 Koshland Hall, Berkeley, CA 94720 USA
| |
Collapse
|
24
|
Bassett AS, McDonald-McGinn DM, Boot E, Óskarsdóttir S, Yuen RKC. Approaches to studying the impact of 22q11.2 copy number variants. Am J Hum Genet 2023; 110:1216-1218. [PMID: 37419092 PMCID: PMC10357417 DOI: 10.1016/j.ajhg.2023.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/02/2023] [Accepted: 05/19/2023] [Indexed: 07/09/2023] Open
Affiliation(s)
- Anne S Bassett
- The Dalglish Family 22q Clinic for Adults, and Department of Psychiatry, University Health Network, Toronto, ON, Canada; Toronto General Research Institute and Division of Cardiology, Department of Medicine, University Health Network, Toronto, ON, Canada; Clinical Genetics Research Program and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| | - Donna M McDonald-McGinn
- 22q and You Center, Clinical Genetics Center, and Section of Genetic Counseling, Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA; Department of Human Biology and Medical Genetics, Sapienza University, Rome, Italy
| | - Erik Boot
- The Dalglish Family 22q Clinic for Adults, and Department of Psychiatry, University Health Network, Toronto, ON, Canada; Advisium, 's Heeren Loo Zorggroep, Amersfoort, the Netherlands; Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands
| | - Sólveig Óskarsdóttir
- Department of Pediatric Rheumatology and Immunology, Queen Silvia Children's Hospital, SahlgrenskaUniversity Hospital, Gothenburg, Sweden; Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ryan K C Yuen
- Genetics & Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
25
|
Szczawińska-Popłonyk A, Schwartzmann E, Chmara Z, Głukowska A, Krysa T, Majchrzycki M, Olejnicki M, Ostrowska P, Babik J. Chromosome 22q11.2 Deletion Syndrome: A Comprehensive Review of Molecular Genetics in the Context of Multidisciplinary Clinical Approach. Int J Mol Sci 2023; 24:ijms24098317. [PMID: 37176024 PMCID: PMC10179617 DOI: 10.3390/ijms24098317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 04/30/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
The 22q11.2 deletion syndrome is a multisystemic disorder characterized by a marked variability of phenotypic features, making the diagnosis challenging for clinicians. The wide spectrum of clinical manifestations includes congenital heart defects-most frequently conotruncal cardiac anomalies-thymic hypoplasia and predominating cellular immune deficiency, laryngeal developmental defects, midline anomalies with cleft palate and velar insufficiency, structural airway defects, facial dysmorphism, parathyroid and thyroid gland hormonal dysfunctions, speech delay, developmental delay, and neurocognitive and psychiatric disorders. Significant progress has been made in understanding the complex molecular genetic etiology of 22q11.2 deletion syndrome underpinning the heterogeneity of clinical manifestations. The deletion is caused by chromosomal rearrangements in meiosis and is mediated by non-allelic homologous recombination events between low copy repeats or segmental duplications in the 22q11.2 region. A range of genetic modifiers and environmental factors, as well as the impact of hemizygosity on the remaining allele, contribute to the intricate genotype-phenotype relationships. This comprehensive review has been aimed at highlighting the molecular genetic background of 22q11.2 deletion syndrome in correlation with a clinical multidisciplinary approach.
Collapse
Affiliation(s)
- Aleksandra Szczawińska-Popłonyk
- Department of Pediatric Pneumonology, Allergy and Clinical Immunology, Institute of Pediatrics, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
| | - Eyal Schwartzmann
- Medical Student Scientific Society, English Division, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
| | - Zuzanna Chmara
- Medical Student Scientific Society, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
| | - Antonina Głukowska
- Medical Student Scientific Society, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
| | - Tomasz Krysa
- Medical Student Scientific Society, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
| | - Maksymilian Majchrzycki
- Medical Student Scientific Society, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
| | - Maurycy Olejnicki
- Medical Student Scientific Society, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
| | - Paulina Ostrowska
- Medical Student Scientific Society, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
| | - Joanna Babik
- Gynecology and Obstetrics with Pregnancy Pathology Unit, Franciszek Raszeja Municipal Hospital, 60-834 Poznań, Poland
| |
Collapse
|
26
|
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
|
27
|
Neuroinflammation and Oxidative Stress in Individuals Affected by DiGeorge Syndrome. Int J Mol Sci 2023; 24:ijms24044242. [PMID: 36835652 PMCID: PMC9965448 DOI: 10.3390/ijms24044242] [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: 11/01/2022] [Revised: 02/07/2023] [Accepted: 02/18/2023] [Indexed: 02/25/2023] Open
Abstract
DiGeorge syndrome (DGS) is a rare genetic disease caused by microdeletions of the 22q11.2 region (DGS1). A haploinsufficiency at 10p level has been proposed also as a DGS cause (DGS2). Clinical manifestations are variable. The most frequent features are thymic hypoplasia or aplasia with consequent immune deficiency, cardiac malformations, hypoparathyroidism, facial and palatine abnormalities, variable degrees of cognitive impairment and psychiatric disorders. The specific aim of this descriptive report is to discuss the correlation between oxidative stress and neuroinflammation in DGS patients with microdeletions of the 22q11.2 region. The deleted chromosomic region maps various genes involved in mitochondrial metabolisms, such as DGCR8 and TXNRD2, that could lead to reactive oxygen species (ROS) increased production and antioxidant depletion. Furthermore, increased levels of ROS in mitochondria would lead to the destruction of the projection neurons in the cerebral cortex with consequent neurocognitive impairment. Finally, the increase in modified protein belonging to the family of sulfoxide compounds and hexoses, acting as inhibitors of the IV and V mitochondria complex, could result in direct ROS overproduction. Neuroinflammation in DGS individuals could be directly related to the development of the syndrome's characteristic psychiatric and cognitive disorders. In patients with psychotic disorders, the most frequent psychiatric manifestation in DGS, Th-17, Th-1 and Th-2 cells are increased with consequent elevation of proinflammatory cytokine IL-6 and IL1β. In patients with anxiety disorders, both CD3 and CD4 are increased. Some patients with autism spectrum disorders (ASDs) have an augmented level of proinflammatory cytokines IL-12, IL-6 and IL-1β, while IFNγ and the anti-inflammatory cytokine IL-10 seem to be reduced. Other data proposed that altered synaptic plasticity could be directly involved in DGS cognitive disorders. In conclusion, the use of antioxidants for restoring mitochondrial functionality in DGS could be a useful tool to protect cortical connectivity and cognitive behavior.
Collapse
|
28
|
Impaired OTUD7A-dependent Ankyrin regulation mediates neuronal dysfunction in mouse and human models of the 15q13.3 microdeletion syndrome. Mol Psychiatry 2023; 28:1747-1769. [PMID: 36604605 DOI: 10.1038/s41380-022-01937-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023]
Abstract
Copy number variations (CNVs) are associated with psychiatric and neurodevelopmental disorders (NDDs), and most, including the recurrent 15q13.3 microdeletion disorder, have unknown disease mechanisms. We used a heterozygous 15q13.3 microdeletion mouse model and patient iPSC-derived neurons to reveal developmental defects in neuronal maturation and network activity. To identify the underlying molecular dysfunction, we developed a neuron-specific proximity-labeling proteomics (BioID2) pipeline, combined with patient mutations, to target the 15q13.3 CNV genetic driver OTUD7A. OTUD7A is an emerging independent NDD risk gene with no known function in the brain, but has putative deubiquitinase function. The OTUD7A protein-protein interaction network included synaptic, axonal, and cytoskeletal proteins and was enriched for ASD and epilepsy risk genes (Ank3, Ank2, SPTAN1, SPTBN1). The interactions between OTUD7A and Ankyrin-G (Ank3) and Ankyrin-B (Ank2) were disrupted by an epilepsy-associated OTUD7A L233F variant. Further investigation of Ankyrin-G in mouse and human 15q13.3 microdeletion and OTUD7AL233F/L233F models revealed protein instability, increased polyubiquitination, and decreased levels in the axon initial segment, while structured illumination microscopy identified reduced Ankyrin-G nanodomains in dendritic spines. Functional analysis of human 15q13.3 microdeletion and OTUD7AL233F/L233F models revealed shared and distinct impairments to axonal growth and intrinsic excitability. Importantly, restoring OTUD7A or Ankyrin-G expression in 15q13.3 microdeletion neurons led to a reversal of abnormalities. These data reveal a critical OTUD7A-Ankyrin pathway in neuronal development, which is impaired in the 15q13.3 microdeletion syndrome, leading to neuronal dysfunction. Furthermore, our study highlights the utility of targeting CNV genes using cell type-specific proteomics to identify shared and unexplored disease mechanisms across NDDs.
Collapse
|
29
|
Bassett AS. Clinical genetics of schizophrenia and related neuropsychiatric disorders. Psychiatry Res 2023; 319:114992. [PMID: 36463725 DOI: 10.1016/j.psychres.2022.114992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/22/2022] [Accepted: 11/27/2022] [Indexed: 11/29/2022]
Abstract
Rare structural variants have turned out to be the long sought for genetic variants of (relatively) high effect size for schizophrenia. Delineating the 22q11.2 microdeletion as the first molecular subtype of schizophrenia was a milestone in schizophrenia research, foreshadowing a more general role for rare copy number variation (CNV) in schizophrenia. The 22q11.2 microdeletion has a high effect size - one in every four individuals born with this deletion develops schizophrenia - and a relatively high prevalence for a rare condition. Discovery of this human genetic high-risk model for schizophrenia has shown how genetics can change clinical management, and also provide new opportunities for animal and cellular models. Further new findings indicate a role for tandem repeat expansion, other less complex rare variants, and collective background effects of common variants in the genetics of schizophrenia. Thus, the genetic architecture of schizophrenia is taking shape, with further advances on the horizon.
Collapse
Affiliation(s)
- Anne S Bassett
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Clinical Genetics Research Program, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; The Dalglish Family 22q Clinic, Department of Psychiatry and Division of Cardiology, Department of Medicine, and Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
30
|
Taylor CM, Finucane BM, Moreno-De-Luca A, Walsh LK, Martin CL, Ledbetter DH. Phenotypic shift in copy number variants: Evidence in 16p11.2 duplication syndrome. Genet Med 2023; 25:151-154. [PMID: 36609147 PMCID: PMC10068678 DOI: 10.1016/j.gim.2022.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/07/2022] Open
Abstract
PURPOSE Recurrent 16p11.2 duplications produce a wide range of clinical outcomes with varying effects on cognition and social functioning. Family-based studies of copy number variants (CNVs) have revealed significant contributions of genomic background on variable expressivity. In this study, we measured the phenotypic effect of 16p11.2 duplications and quantified the modulating effect of familial background on cognitive and social outcomes. METHODS Genomic and clinical data were ascertained from 41 probands with a 16p11.2 duplication and their first-degree relatives. Paired comparisons were completed to determine the duplication's effect on expected vs actual performance on standardized tests of intelligence (IQ) and social functioning (Social Responsiveness Scale-2). Intraclass correlations between relatives and probands were also calculated. RESULTS Cognitive and social functioning were significantly lower among individuals with 16p11.2 duplications than their CNV-negative relatives, whereas intraclass correlations between the groups remained high for full-scale IQ and Social Responsiveness Scale-2 scores. CONCLUSION The 16p11.2 duplication confers deleterious effects on cognition and social functioning, whereas familial background significantly influences phenotypic expression of these traits. Understanding variable expressivity in CNV disorders has implications for anticipatory clinical care, particularly for individuals who receive a genetic diagnosis at an early age, long before the full scope of manifestations becomes evident.
Collapse
Affiliation(s)
- Cora M Taylor
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA.
| | | | | | - Lauren K Walsh
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA
| | | | - David H Ledbetter
- Department of Psychiatry, College of Medicine, University of Florida, Gainesville, FL
| |
Collapse
|
31
|
Adult Height, 22q11.2 Deletion Extent, and Short Stature in 22q11.2 Deletion Syndrome. Genes (Basel) 2022; 13:genes13112038. [DOI: 10.3390/genes13112038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/20/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022] Open
Abstract
The 22q11.2 deletion syndrome (22q11.2DS) manifests as a wide range of medical conditions across a number of systems. Pediatric growth deficiency with some catch-up growth is reported, but there are few studies of final adult height. We aimed to investigate how final adult height in 22q11.2DS compared with general population norms, and to examine predictors of short stature in in a cohort of 397 adults with 22q11.2DS (aged 17.6–76.3 years) with confirmed typical 22q11.2 microdeletion (overlapping the LCR22A to LCR22B region). We defined short stature as <3rd percentile using population norms. For the subset (n = 314, 79.1%) with 22q11.2 deletion extent, we used a binomial logistic regression model to predict short stature in 22q11.2DS, accounting for effects of sex, age, ancestry, major congenital heart disease (CHD), moderate-to-severe intellectual disability (ID), and 22q11.2 deletion extent. Adult height in 22q11.2DS showed a normal distribution but with a shift to the left, compared with population norms. Those with short stature represented 22.7% of the 22q11.2DS sample, 7.6-fold greater than population expectations (p < 0.0001). In the regression model, moderate-to-severe ID, major CHD, and the common LCR22A-LCR22D (A-D) deletion were significant independent risk factors for short stature while accounting for other factors (model p = 0.0004). The results suggest that the 22q11.2 microdeletion has a significant effect on final adult height distribution, and on short stature with effects appearing to arise from reduced gene dosage involving both the proximal and distal sub-regions of the A-D region. Future studies involving larger sample sizes with proximal nested 22q11.2 deletions, longitudinal lifetime data, parental heights, and genotype data will be valuable.
Collapse
|
32
|
Snihirova Y, Linden DEJ, van Amelsvoort T, van der Meer D. Environmental Influences on the Relation between the 22q11.2 Deletion Syndrome and Mental Health: A Literature Review. Genes (Basel) 2022; 13:2003. [PMID: 36360240 PMCID: PMC9690390 DOI: 10.3390/genes13112003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/21/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
22q11.2 deletion syndrome (22q11DS) is a clinically heterogeneous genetic syndrome, associated with a wide array of neuropsychiatric symptoms. The clinical presentation is likely to be influenced by environmental factors, yet little is known about this. Here, we review the available research literature on the role of the environment in 22q11DS. We find that within-patient design studies have mainly investigated the role of parental factors, stress, and substance use, reporting significant effects of these factors on the clinical profile. Case-control studies have been less successful, with almost no reports of significant moderating effects of the environment. We go on to hypothesize which specific environmental measures are most likely to interact with the 22q11 deletion, based on the genes in this region and their involvement in molecular pathways. We end by discussing potential reasons for the limited findings so far, including modest sample sizes and limited availability of environmental measures, and make recommendations how to move forward.
Collapse
Affiliation(s)
- Yelyzaveta Snihirova
- School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - David E. J. Linden
- School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Therese van Amelsvoort
- School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Dennis van der Meer
- School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
| |
Collapse
|
33
|
Affiliation(s)
- Cathryn M Lewis
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London (Lewis, Vassos); Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London (Lewis)
| | - Evangelos Vassos
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London (Lewis, Vassos); Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London (Lewis)
| |
Collapse
|
34
|
Fiksinski AM, Heung T, Corral M, Breetvelt EJ, Costain G, Marshall CR, Kahn RS, Vorstman JA, Bassett AS. Within-family influences on dimensional neurobehavioral traits in a high-risk genetic model. Psychol Med 2022; 52:3184-3192. [PMID: 33443009 PMCID: PMC9693655 DOI: 10.1017/s0033291720005279] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/08/2020] [Accepted: 12/17/2020] [Indexed: 01/05/2023]
Abstract
BACKGROUND Genotype-first and within-family studies can elucidate factors that contribute to psychiatric illness. Combining these approaches, we investigated the patterns of influence of parental scores, a high-impact variant, and schizophrenia on dimensional neurobehavioral phenotypes implicated in major psychiatric disorders. METHODS We quantitatively assessed cognitive (FSIQ, VIQ, PIQ), social, and motor functioning in 82 adult individuals with a de novo 22q11.2 deletion (22 with schizophrenia), and 148 of their unaffected parents. We calculated within-family correlations and effect sizes of the 22q11.2 deletion and schizophrenia, and used linear regressions to assess contributions to neurobehavioral measures. RESULTS Proband-parent intra-class correlations (ICC) were significant for cognitive measures (e.g. FSIQ ICC = 0.549, p < 0.0001), but not for social or motor measures. Compared to biparental scores, the 22q11.2 deletion conferred significant impairments for all phenotypes assessed (effect sizes -1.39 to -2.07 s.d.), strongest for PIQ. There were further decrements in those with schizophrenia. Regression models explained up to 37.7% of the variance in IQ and indicated that for proband IQ, parental IQ had larger effects than schizophrenia. CONCLUSIONS This study, for the first time, disentangles the impact of a high-impact variant from the modifying effects of parental scores and schizophrenia on relevant neurobehavioral phenotypes. The robust proband-parent correlations for cognitive measures, independent of the impact of the 22q11.2 deletion and of schizophrenia, suggest that, for certain phenotypes, shared genetic variation plays a significant role in expression. Molecular genetic and predictor studies are needed to elucidate shared factors and their contribution to psychiatric illness in this and other high-risk groups.
Collapse
Affiliation(s)
- Ania M. Fiksinski
- Department of Psychiatry, Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- The Dalglish Family 22q Clinic for 22q11.2 Deletion Syndrome, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Tracy Heung
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- The Dalglish Family 22q Clinic for 22q11.2 Deletion Syndrome, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Maria Corral
- The Dalglish Family 22q Clinic for 22q11.2 Deletion Syndrome, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Elemi J. Breetvelt
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Gregory Costain
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Christian R. Marshall
- Genome Diagnostics, Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Rene S. Kahn
- Department of Psychiatry, Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jacob A.S. Vorstman
- Department of Psychiatry, Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Anne S. Bassett
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- The Dalglish Family 22q Clinic for 22q11.2 Deletion Syndrome, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute and Campbell Family Mental Health Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
35
|
Belyaeva EO, Lebedev IN. Interloci CNV Interactions in Variability of the Phenotypes of Neurodevelopmental Disorders. RUSS J GENET+ 2022. [DOI: 10.1134/s1022795422100027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
36
|
Mancini V, Rochas V, Seeber M, Grent-'t-Jong T, Rihs TA, Latrèche C, Uhlhaas PJ, Michel CM, Eliez S. Oscillatory Neural Signatures of Visual Perception Across Developmental Stages in Individuals With 22q11.2 Deletion Syndrome. Biol Psychiatry 2022; 92:407-418. [PMID: 35550793 DOI: 10.1016/j.biopsych.2022.02.961] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND Numerous behavioral studies have highlighted the contribution of visual perceptual deficits to the nonverbal cognitive profile of individuals with 22q11.2 deletion syndrome. However, the neurobiological processes underlying these widespread behavioral alterations are yet to be fully understood. Thus, in this paper, we investigated the role of neural oscillations toward visuoperceptual deficits to elucidate the neurobiology of sensory impairments in deletion carriers. METHODS We acquired 125 high-density electroencephalography recordings during a visual grating task in a group of 62 deletion carriers and 63 control subjects. Stimulus-elicited oscillatory responses were analyzed with 1) time-frequency analysis using wavelets decomposition at sensor and source level, 2) intertrial phase coherence, and 3) Granger causality connectivity in source space. Additional analyses examined the development of neural oscillations across age bins. RESULTS Deletion carriers had decreased theta-band (4-8 Hz) and gamma-band (58-68 Hz) spectral power compared with control subjects in response to the visual stimuli, with an absence of age-related increase of theta- and gamma-band responses. Moreover, adult deletion carriers had decreased gamma- and theta-band responses but increased alpha/beta desynchronization (10-25 Hz) that correlated with behavioral performance. Granger causality estimates reflected an increased frontal-occipital connectivity in the beta range (22-40 Hz). CONCLUSIONS Deletion carriers exhibited decreased theta- and gamma-band responses to visual stimuli, while alpha/beta desynchronization was preserved. Overall, the lack of age-related changes in deletion carriers implicates developmental impairments in circuit mechanisms underlying neural oscillations. The dissociation between the maturation of theta/gamma- and alpha/beta-band responses may indicate a selective impairment in supragranular cortical layers, leading to compensatory top-down connectivity.
Collapse
Affiliation(s)
- Valentina Mancini
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland.
| | - Vincent Rochas
- Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland; Human Neuroscience Platform, Fondation Campus Biotech Geneva, Geneva, Switzerland
| | - Martin Seeber
- Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Tineke Grent-'t-Jong
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland; Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin, Germany
| | - Tonia A Rihs
- Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Caren Latrèche
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland
| | - Peter J Uhlhaas
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland; Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Charité Universitätsmedizin, Berlin, Germany
| | - Christoph M Michel
- Functional Brain Mapping Laboratory, Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland; Center for Biomedical Imaging, Lausanne, Switzerland
| | - Stephan Eliez
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland; Department of Genetic Medicine and Development, University of Geneva School of Medicine, Geneva, Switzerland
| |
Collapse
|
37
|
Nagai Y, Nishioka M, Tanaka T, Shimano T, Kirino E, Suzuki T, Kato T. Identification of 22q11.2 deletion in a patient with schizophrenia and clinically diagnosed Rubinstein-Taybi syndrome. PCN REPORTS : PSYCHIATRY AND CLINICAL NEUROSCIENCES 2022; 1:e34. [PMID: 38868697 PMCID: PMC11114328 DOI: 10.1002/pcn5.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 06/22/2022] [Accepted: 07/07/2022] [Indexed: 06/14/2024]
Abstract
Background Rubinstein-Taybi syndrome (RTS) is a rare autosomal-dominant disease. Almost all cases are sporadic and attributed to de novo variant. Psychotic symptoms in RTS are rare and have been reported in only a few published cases. On the other hand, 22q11.2 deletion syndrome is the most common chromosomal microdeletion in humans. The 22q11.2 deletion is well recognized as a risk factor for schizophrenia. Here, we present a schizophrenic psychosis case clinically diagnosed as RTS but resolved as carrying 22q11.2 deletion by genomic analysis. Case presentation A 38-year-old Japanese male was admitted to our hospital due to psychotic symptoms. He had been diagnosed with RTS based on physical characteristics at the age of 9 months. On admission, we performed whole exome sequencing. He had no pathogenic variant in CREBBP or EP300. We detected 2.5 Mb deletion on 22q11.2 and one rare loss-of-function variant in a loss-of-function-constrained gene (MTSS1) and three rare missense variants in missense-constrained genes (CELSR3, HERC1, and TLN1). Psychotic symptoms were ameliorated by the treatment of risperidone. Conclusion The psychiatric manifestation and genomic analysis may be a clue to detecting 22q11.2 deletion syndrome in undiagnosed patients. The reason for similarity in physical characteristics in 22q11.2 deletion syndrome and RTS remains unresolved. The 22q11.2 deletion and HERC1 contribute to the patient's phenotype.
Collapse
Affiliation(s)
- Yasuhito Nagai
- Department of PsychiatryJuntendo University School of MedicineTokyoJapan
- Department of PsychiatryJuntendo Tokyo Koto Geriatric Medical CenterTokyoJapan
| | - Masaki Nishioka
- Department of PsychiatryJuntendo University School of MedicineTokyoJapan
- Department of PsychiatryJuntendo University HospitalTokyoJapan
| | - Tatsuki Tanaka
- Department of PsychiatryJuntendo University School of MedicineTokyoJapan
- Department of PsychiatryJuntendo University Koshigaya HospitalKoshigayaJapan
| | | | - Eiji Kirino
- Department of PsychiatryJuntendo University School of MedicineTokyoJapan
- Department of PsychiatryJuntendo University Shizuoka HospitalIzunokuniJapan
| | - Toshihito Suzuki
- Department of PsychiatryJuntendo University School of MedicineTokyoJapan
- Department of PsychiatryJuntendo University Koshigaya HospitalKoshigayaJapan
| | - Tadafumi Kato
- Department of PsychiatryJuntendo University School of MedicineTokyoJapan
- Department of PsychiatryJuntendo University HospitalTokyoJapan
| |
Collapse
|
38
|
Schizophrenia: A Narrative Review of Etiopathogenetic, Diagnostic and Treatment Aspects. J Clin Med 2022; 11:jcm11175040. [PMID: 36078967 PMCID: PMC9457502 DOI: 10.3390/jcm11175040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/25/2022] Open
Abstract
Although schizophrenia is currently conceptualized as being characterized as a syndrome that includes a collection of signs and symptoms, there is strong evidence of heterogeneous and complex underpinned etiological, etiopathogenetic, and psychopathological mechanisms, which are still under investigation. Therefore, the present viewpoint review is aimed at providing some insights into the recently investigated schizophrenia research fields in order to discuss the potential future research directions in schizophrenia research. The traditional schizophrenia construct and diagnosis were progressively revised and revisited, based on the recently emerging neurobiological, genetic, and epidemiological research. Moreover, innovative diagnostic and therapeutic approaches are pointed to build a new construct, allowing the development of better clinical and treatment outcomes and characterization for schizophrenic individuals, considering a more patient-centered, personalized, and tailored-based dimensional approach. Further translational studies are needed in order to integrate neurobiological, genetic, and environmental studies into clinical practice and to help clinicians and researchers to understand how to redesign a new schizophrenia construct.
Collapse
|
39
|
Kozlova A, Zhang S, Kotlar AV, Jamison B, Zhang H, Shi S, Forrest MP, McDaid J, Cutler DJ, Epstein MP, Zwick ME, Pang ZP, Sanders AR, Warren ST, Gejman PV, Mulle JG, Duan J. Loss of function of OTUD7A in the schizophrenia- associated 15q13.3 deletion impairs synapse development and function in human neurons. Am J Hum Genet 2022; 109:1500-1519. [PMID: 35931052 PMCID: PMC9388388 DOI: 10.1016/j.ajhg.2022.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 06/27/2022] [Indexed: 02/06/2023] Open
Abstract
Identifying causative gene(s) within disease-associated large genomic regions of copy-number variants (CNVs) is challenging. Here, by targeted sequencing of genes within schizophrenia (SZ)-associated CNVs in 1,779 SZ cases and 1,418 controls, we identified three rare putative loss-of-function (LoF) mutations in OTU deubiquitinase 7A (OTUD7A) within the 15q13.3 deletion in cases but none in controls. To tie OTUD7A LoF with any SZ-relevant cellular phenotypes, we modeled the OTUD7A LoF mutation, rs757148409, in human induced pluripotent stem cell (hiPSC)-derived induced excitatory neurons (iNs) by CRISPR-Cas9 engineering. The mutant iNs showed a ∼50% decrease in OTUD7A expression without undergoing nonsense-mediated mRNA decay. The mutant iNs also exhibited marked reduction of dendritic complexity, density of synaptic proteins GluA1 and PSD-95, and neuronal network activity. Congruent with the neuronal phenotypes in mutant iNs, our transcriptomic analysis showed that the set of OTUD7A LoF-downregulated genes was enriched for those relating to synapse development and function and was associated with SZ and other neuropsychiatric disorders. These results suggest that OTUD7A LoF impairs synapse development and neuronal function in human neurons, providing mechanistic insight into the possible role of OTUD7A in driving neuropsychiatric phenotypes associated with the 15q13.3 deletion.
Collapse
Affiliation(s)
- Alena Kozlova
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Siwei Zhang
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA; Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Alex V Kotlar
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; Pillar Biosciences Inc., Natick, MA 01760, USA
| | - Brendan Jamison
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Hanwen Zhang
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Serena Shi
- Winston Churchill High School, Potomac, MD 20854, USA
| | - Marc P Forrest
- Department of Neuroscience, Northwestern University, Chicago, IL 60611, USA; Center for Autism and Neurodevelopment, Northwestern University, Chicago, IL 60611, USA
| | - John McDaid
- Department of Neurosurgery, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - David J Cutler
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael P Epstein
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael E Zwick
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; Senior Vice President for Research, Rutgers University, New Brunswick, NJ 08901, USA
| | - Zhiping P Pang
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Alan R Sanders
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA; Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Stephen T Warren
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Pablo V Gejman
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA; Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Jennifer G Mulle
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA; Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
40
|
Nehme R, Pietiläinen O, Artomov M, Tegtmeyer M, Valakh V, Lehtonen L, Bell C, Singh T, Trehan A, Sherwood J, Manning D, Peirent E, Malik R, Guss EJ, Hawes D, Beccard A, Bara AM, Hazelbaker DZ, Zuccaro E, Genovese G, Loboda AA, Neumann A, Lilliehook C, Kuismin O, Hamalainen E, Kurki M, Hultman CM, Kähler AK, Paulo JA, Ganna A, Madison J, Cohen B, McPhie D, Adolfsson R, Perlis R, Dolmetsch R, Farhi S, McCarroll S, Hyman S, Neale B, Barrett LE, Harper W, Palotie A, Daly M, Eggan K. The 22q11.2 region regulates presynaptic gene-products linked to schizophrenia. Nat Commun 2022; 13:3690. [PMID: 35760976 PMCID: PMC9237031 DOI: 10.1038/s41467-022-31436-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 06/08/2022] [Indexed: 12/30/2022] Open
Abstract
It is unclear how the 22q11.2 deletion predisposes to psychiatric disease. To study this, we generated induced pluripotent stem cells from deletion carriers and controls and utilized CRISPR/Cas9 to introduce the heterozygous deletion into a control cell line. Here, we show that upon differentiation into neural progenitor cells, the deletion acted in trans to alter the abundance of transcripts associated with risk for neurodevelopmental disorders including autism. In excitatory neurons, altered transcripts encoded presynaptic factors and were associated with genetic risk for schizophrenia, including common and rare variants. To understand how the deletion contributed to these changes, we defined the minimal protein-protein interaction network that best explains gene expression alterations. We found that many genes in 22q11.2 interact in presynaptic, proteasome, and JUN/FOS transcriptional pathways. Our findings suggest that the 22q11.2 deletion impacts genes that may converge with psychiatric risk loci to influence disease manifestation in each deletion carrier.
Collapse
Affiliation(s)
- Ralda Nehme
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA.
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA.
| | - Olli Pietiläinen
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA.
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA.
| | - Mykyta Artomov
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Matthew Tegtmeyer
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Vera Valakh
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Leevi Lehtonen
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00014, Helsinki, Finland
| | - Christina Bell
- Department of Cell Biology, Blavatnik Institute of Harvard Medical School, Boston, MA, USA
| | - Tarjinder Singh
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Aditi Trehan
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - John Sherwood
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Danielle Manning
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Emily Peirent
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Rhea Malik
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Ellen J Guss
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Derek Hawes
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Amanda Beccard
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Anne M Bara
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Dane Z Hazelbaker
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Emanuela Zuccaro
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Giulio Genovese
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Alexander A Loboda
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- ITMO University, St. Petersburg, Russia
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - Anna Neumann
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Christina Lilliehook
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Outi Kuismin
- Psychiatric & Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
- PEDEGO Research Unit, University of Oulu, FI-90014, Oulu, Finland
- Medical Research Center, Oulu University Hospital, FI-90014, Oulu, Finland
- Department of Clinical Genetics, Oulu University Hospital, 90220, Oulu, Finland
| | - Eija Hamalainen
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00014, Helsinki, Finland
| | - Mitja Kurki
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00014, Helsinki, Finland
- Psychiatric & Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Christina M Hultman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Anna K Kähler
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Joao A Paulo
- Department of Cell Biology, Blavatnik Institute of Harvard Medical School, Boston, MA, USA
| | - Andrea Ganna
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Jon Madison
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Bruce Cohen
- Department of Psychiatry, McLean Hospital, Belmont, MA, 02478, USA
| | - Donna McPhie
- Department of Psychiatry, McLean Hospital, Belmont, MA, 02478, USA
| | - Rolf Adolfsson
- Umea University, Faculty of Medicine, Department of Clinical Sciences, Psychiatry, 901 85, Umea, Sweden
| | - Roy Perlis
- Psychiatry Dept., Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Ricardo Dolmetsch
- Novartis Institutes for Biomedical Research, Novartis, Cambridge, MA, 02139, USA
| | - Samouil Farhi
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Steven McCarroll
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Steven Hyman
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Ben Neale
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Lindy E Barrett
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Wade Harper
- Department of Cell Biology, Blavatnik Institute of Harvard Medical School, Boston, MA, USA
| | - Aarno Palotie
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00014, Helsinki, Finland
- Psychiatric & Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Mark Daly
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00014, Helsinki, Finland
- Psychiatric & Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Kevin Eggan
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA.
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA.
- BioMarin Pharmaceutical, San Rafael, CA, 94901, USA.
| |
Collapse
|
41
|
Fiksinski AM, Bearden CE, Bassett AS, Kahn RS, Zinkstok JR, Hooper SR, Tempelaar W, McDonald-McGinn D, Swillen A, Emanuel B, Morrow B, Gur R, Chow E, van den Bree M, Vermeesch J, Warren S, Owen M, van Amelsvoort T, Eliez S, Gothelf D, Arango C, Kates W, Simon T, Murphy K, Repetto G, Suner DH, Vicari S, Cubells J, Armando M, Philip N, Campbell L, Garcia-Minaur S, Schneider M, Shashi V, Vorstman J, Breetvelt EJ. A normative chart for cognitive development in a genetically selected population. Neuropsychopharmacology 2022; 47:1379-1386. [PMID: 33782512 PMCID: PMC9117666 DOI: 10.1038/s41386-021-00988-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 01/12/2021] [Accepted: 01/30/2021] [Indexed: 02/03/2023]
Abstract
Certain pathogenic genetic variants impact neurodevelopment and cause deviations from typical cognitive trajectories. Understanding variant-specific cognitive trajectories is clinically important for informed monitoring and identifying patients at risk for comorbid conditions. Here, we demonstrate a variant-specific normative chart for cognitive development for individuals with 22q11.2 deletion syndrome (22q11DS). We used IQ data from 1365 individuals with 22q11DS to construct variant-specific normative charts for cognitive development (Full Scale, Verbal, and Performance IQ). This allowed us to calculate Z-scores for each IQ datapoint. Then, we calculated the change between first and last available IQ assessments (delta Z-IQ-scores) for each individual with longitudinal IQ data (n = 708). We subsequently investigated whether using the variant-specific IQ-Z-scores would decrease required sample size to detect an effect with schizophrenia risk, as compared to standard IQ-scores. The mean Z-IQ-scores for FSIQ, VIQ, and PIQ were close to 0, indicating that participants had IQ-scores as predicted by the normative chart. The mean delta-Z-IQ-scores were equally close to 0, demonstrating a good fit of the normative chart and indicating that, as a group, individuals with 22q11DS show a decline in IQ-scores as they grow into adulthood. Using variant-specific IQ-Z-scores resulted in 30% decrease of required sample size, as compared to the standard IQ-based approach, to detect the association between IQ-decline and schizophrenia (p < 0.01). Our findings suggest that using variant-specific normative IQ data significantly reduces required sample size in a research context, and may facilitate a more clinically informative interpretation of IQ data. This approach allows identification of individuals that deviate from their expected, variant-specific, trajectory. This group may be at increased risk for comorbid conditions, such as schizophrenia in the case of 22q11DS.
Collapse
Affiliation(s)
- Ania M Fiksinski
- Wilhelmina Children's Hospital & University Medical Center Utrecht, Brain Center, Utrecht, The Netherlands.
- Centre for Addiction and Mental Health, Toronto, ON, Canada.
- The Dalglish Family 22q Clinic for 22q11.2 Deletion Syndrome, Toronto General Hospital, University Health Network, Toronto, ON, Canada.
- Department of Psychiatry and Neuropsychology, Division of Mental Health, MHeNS, Maastricht University, Maastricht, The Netherlands.
| | - Carrie E Bearden
- Departments of Psychiatry and Biobehavioral Sciences and Psychology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, USA
| | - Anne S Bassett
- The Dalglish Family 22q Clinic, Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, Canada
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - René S Kahn
- Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Janneke R Zinkstok
- Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Stephen R Hooper
- Department of Allied Health Sciences, School of Medicine, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Wanda Tempelaar
- Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Donna McDonald-McGinn
- Division of Human Genetics, 22q and You Center, Clinical Genetics Center, and Section of Genetic Counseling, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA
| | - Ann Swillen
- Center for Human Genetics, University Hospital Gasthuisberg, Leuven, Belgium
- Department of Human Genetics KU Leuven, Leuven, Belgium
| | - Beverly Emanuel
- Division of Human Genetics, 22q and You Center, Clinical Genetics Center, and Section of Genetic Counseling, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Bernice Morrow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Raquel Gur
- Department of Psychiatry and Lifespan Brain Institute, Penn Medicine-CHOP, University of Pennsylvania, Philadelphia, PA, USA
| | - Eva Chow
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Marianne van den Bree
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Joris Vermeesch
- Center for Human Genetics, University Hospital Gasthuisberg, Leuven, Belgium
| | - Stephen Warren
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Therese van Amelsvoort
- Department of Psychiatry and Neuropsychology, Division of Mental Health, MHeNS, Maastricht University, Maastricht, The Netherlands
| | - Stephan Eliez
- Developmental Imaging and Psychopathology, Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Doron Gothelf
- The Child Psychiatry Division, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
- Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Celso Arango
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, IiSGM, CIBERSAM, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Wendy Kates
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Tony Simon
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis, Sacramento, CA, USA
| | - Kieran Murphy
- Department of Psychiatry, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Gabriela Repetto
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Damian Heine Suner
- Genomics of Health Group and Molecular Diagnostics and Clinical Genetics Unit (UDMGC), Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitari Son Espases, Palma de Mallorca, Spain
| | - Stefano Vicari
- Department of Life Sciences and Public Health, Catholic University; Child and Adolescent Psychiatry Unit, Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | - Joseph Cubells
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
- Emory Autism Center, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Marco Armando
- Developmental Imaging and Psychopathology, Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Nicole Philip
- Département de Génétique Médicale, APHM, CHU Timone Enfants, Marseille, France
- Aix Marseille Université, MMG, INSERM, Marseille, France
| | - Linda Campbell
- School of Psychology, University of Newcastle, Newcastle, Australia
| | - Sixto Garcia-Minaur
- Institute of Medical and Molecular Genetics (INGEMM), La Paz University Hospital, Madrid, Spain
| | - Maude Schneider
- Developmental Imaging and Psychopathology, Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Vandana Shashi
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Jacob Vorstman
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Psychiatry, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Elemi J Breetvelt
- Department of Psychiatry, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
42
|
Blagojevic C, Heung T, Malecki S, Ying S, Cancelliere S, Hegele RA, Bassett AS. Hypertriglyceridemia in young adults with a 22q11.2 microdeletion. Eur J Endocrinol 2022; 187:91-99. [PMID: 35521712 DOI: 10.1530/eje-21-1104] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 04/27/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Mild to moderate hypertriglyceridemia is a condition often associated with obesity and diabetes, with as yet incomplete knowledge of underlying genetic architecture. The 22q11.2 microdeletion is associated with multimorbidity, including increased risk of obesity and diabetes. In this study, we sought to investigate whether the 22q11.2 microdeletion was associated with mild to moderate hypertriglyceridemia (1.7-10 mmol/L). DESIGN This was a cohort study comparing 6793 population-based adults and 267 with a 22q11.2 microdeletion aged 17-69 years, excluding those with diabetes or on statins. METHODS We used binomial logistic regression modeling to identify predictors of hypertriglyceridemia, accounting for the 22q11.2 microdeletion, male sex, BMI, ethnicity, age, and antipsychotic medications. RESULTS The 22q11.2 microdeletion was a significant independent predictor of mild to moderate hypertriglyceridemia (odds ratio (OR): 2.35, 95% CI: 1.70-3.26). All other factors examined were also significant predictors (OR: 1.23-2.10), except for antipsychotic medication use. Within the 22q11.2 microdeletion subgroup, only male sex (OR: 3.10, 95% CI: 1.77-5.44) and BMI (OR: 1.63, 95% CI: 1.14-1.98) were significant predictors of hypertriglyceridemia, evident at mean age 31.2 years. CONCLUSIONS The 22q11.2 microdeletion is associated with hypertriglyceridemia even when accounting for other known risk factors for elevated triglycerides. This effect is seen in young adulthood (76.6% were <40 years), in the absence of diabetes, and irrespective of antipsychotics, suggesting that the 22q11.2 microdeletion may represent an unrecognized genetic risk factor for hypertriglyceridemia, providing novel opportunities for animal and cellular models. Early dyslipidemia screening and management strategies would appear prudent for individuals with 22q11.2 microdeletions.
Collapse
Affiliation(s)
- Christina Blagojevic
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- The Dalglish Family 22q Clinic, University Health Network, Toronto, Ontario, Canada
| | - Tracy Heung
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- The Dalglish Family 22q Clinic, University Health Network, Toronto, Ontario, Canada
| | - Sarah Malecki
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- The Dalglish Family 22q Clinic, University Health Network, Toronto, Ontario, Canada
| | - Shengjie Ying
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- The Dalglish Family 22q Clinic, University Health Network, Toronto, Ontario, Canada
| | - Sabrina Cancelliere
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Robert A Hegele
- Departments of Medicine and Biochemistry, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Anne S Bassett
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- The Dalglish Family 22q Clinic, University Health Network, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto General Hospital Research Institute, and Campbell Family Mental Health Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
43
|
Breetvelt EJ, Smit KC, van Setten J, Merico D, Wang X, Vaartjes I, Bassett AS, Boks MPM, Szatmari P, Scherer SW, Kahn RS, Vorstman JAS. A Regional Burden of Sequence-Level Variation in the 22q11.2 Region Influences Schizophrenia Risk and Educational Attainment. Biol Psychiatry 2022; 91:718-726. [PMID: 35063188 DOI: 10.1016/j.biopsych.2021.11.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 10/25/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Genomic loci where recurrent pathogenic copy number variants are associated with psychiatric phenotypes in the population may also be sensitive to the collective impact of multiple functional low-frequency single nucleotide variants (SNVs). METHODS We examined the cumulative impact of low-frequency, functional SNVs within the 22q11.2 region on schizophrenia risk in a discovery cohort and an independent replication cohort (N = 1933 and N = 11,128, respectively), as well as the impact on educational attainment (EA) in a third, independent, general population cohort (N = 2081). In the discovery and EA cohorts, SNVs were identified using genotyping arrays; in the replication cohort, whole-exome sequencing was available. For verification, we compared the regional SNV count for schizophrenia cases in the discovery cohort with a normative count distribution derived from a large population dataset (N = 26,500) using bootstrap procedures. RESULTS In both schizophrenia cohorts, an increased regional SNV burden (≥4 low-frequency SNVs) in the 22q11.2 region was associated with schizophrenia (discovery cohort: odds ratio = 7.48, p = .039; replication cohort: odds ratio = 1.92, p = .004). In the EA cohort, an increased regional SNV burden at 22q11.2 was associated with decreased EA (odds ratio = 4.65, p = .049). Comparing the SNV count for schizophrenia cases with a normative distribution confirmed the unique nature of the distribution for schizophrenia cases (p = .002). CONCLUSIONS In the general population, an increased burden of low-frequency, functional SNVs in the 22q11.2 region is associated with schizophrenia risk and a decrease in EA. These findings suggest that in addition to structural variation, a cumulative regional burden of low-frequency, functional SNVs in the 22q11.2 region can also have a relevant phenotypic impact.
Collapse
Affiliation(s)
- Elemi J Breetvelt
- Department of Psychiatry, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| | - Karel C Smit
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht, The Netherlands; Department of Medical Oncology, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands; Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Jessica van Setten
- Department of Cardiology, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Daniele Merico
- Center for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada; Deep Genomics Inc., Toronto, Ontario, Canada
| | - Xiao Wang
- Center for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ilonca Vaartjes
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Anne S Bassett
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Dalglish Family 22q Clinic for Adults with 22q11.2 Deletion Syndrome, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario, Canada; Medical Genetics and Genomics Residency Training Program, University of Toronto, Toronto, Ontario, Canada; Toronto General Research Institute, Toronto, Ontario, Canada; Campbell Family Mental Health Research Institute, Toronto, Ontario, Canada
| | - Marco P M Boks
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht, The Netherlands
| | - Peter Szatmari
- Department of Psychiatry, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Stephen W Scherer
- Center for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada; McLaughlin Centre and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - René S Kahn
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht, The Netherlands; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NewYork, New York
| | - Jacob A S Vorstman
- Department of Psychiatry, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht, The Netherlands
| |
Collapse
|
44
|
Vermeesch JR. The Hunt for the Chromosome 22q11.2 Deletion Syndrome Schizophrenia Genes. Biol Psychiatry 2022; 91:692-693. [PMID: 35331390 DOI: 10.1016/j.biopsych.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 11/24/2022]
Affiliation(s)
- Joris Robert Vermeesch
- Laboratory of Cytogenetics and Genome Research, Department of Human Genetics, KU Leuven, Leuven, Belgium.
| |
Collapse
|
45
|
Ying S, Heung T, Zhang Z, Yuen RKC, Bassett AS. Schizophrenia Risk Mediated by microRNA Target Genes Overlapped by Genome-Wide Rare Copy Number Variation in 22q11.2 Deletion Syndrome. Front Genet 2022; 13:812183. [PMID: 35495153 PMCID: PMC9053669 DOI: 10.3389/fgene.2022.812183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
The 22q11.2 deletion is associated with >20-fold increased risk for schizophrenia. The presence of gene DGCR8 in the 22q11.2 deletion region has suggested microRNA (miRNA) dysregulation as possibly contributing to this risk. We therefore investigated the role of miRNA target genes in the context of previously identified genome-wide risk for schizophrenia conveyed by additional copy number variation (CNV) in 22q11.2 deletion syndrome (22q11.2DS). Using a cohort of individuals with 22q11.2DS and documented additional rare CNVs overlapping protein coding genes, we compared those with schizophrenia (n = 100) to those with no psychotic illness (n = 118), assessing for rare CNVs that overlapped experimentally supported miRNA target genes. We further characterized the contributing miRNA target genes using gene set enrichment analyses and identified the miRNAs most implicated. Consistent with our hypothesis, we found a significantly higher proportion of individuals in the schizophrenia than in the non-psychotic group to have an additional rare CNV that overlapped one or more miRNA target genes (odds ratio = 2.12, p = 0.0138). Gene set analyses identified an enrichment of FMRP targets and genes involved in nervous system development and postsynaptic density amongst these miRNA target genes in the schizophrenia group. The miRNAs most implicated included miR-17-5p, miR-34a-5p and miR-124-3p. These results provide initial correlational evidence in support of a possible role for miRNA perturbation involving genes affected by rare genome-wide CNVs in the elevated risk for schizophrenia in 22q11.2DS, consistent with the multi-hit and multi-layered genetic mechanisms implicated in this and other forms of schizophrenia.
Collapse
Affiliation(s)
- Shengjie Ying
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Tracy Heung
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, ON, Canada
- The Dalglish Family 22q Clinic, University Health Network, Toronto, ON, Canada
| | - Zhaolei Zhang
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Ryan K. C. Yuen
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Anne S. Bassett
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, Toronto, ON, Canada
- The Dalglish Family 22q Clinic, University Health Network, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute and Campbell Family Mental Health Research Institute, Toronto, ON, Canada
- *Correspondence: Anne S. Bassett,
| |
Collapse
|
46
|
Sánchez-Hidalgo AC, Martín-Cuevas C, Crespo-Facorro B, Garrido-Torres N. Reelin Alterations, Behavioral Phenotypes, and Brain Anomalies in Schizophrenia: A Systematic Review of Insights From Rodent Models. Front Neuroanat 2022; 16:844737. [PMID: 35401125 PMCID: PMC8986979 DOI: 10.3389/fnana.2022.844737] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/14/2022] [Indexed: 12/09/2022] Open
Abstract
Reelin is an extracellular matrix glycoprotein reduced in brain regions (the prefrontal cortex and the hippocampus) of patients with schizophrenia. There are diverse rodent models of schizophrenia that mimic patient symptoms based on various causal theories; however, likely shared reelin alterations have not yet been systematically assessed in those models. A systematic review of the literature was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) model. Articles focused on psychotic disorders or schizophrenia and their relationship with reelin in rodent models were selected. Data (first author, publication year, results, both open field and prepulse inhibition test results, and type of reelin alteration) were extracted in duplicate by two independent reviewers. The 37 reviewed articles reported about various schizophrenia models and their reelin alterations, brain morphology, and behavioral defects. We conclude that reelin is an altered preclinical biomarker common to all models included, mainly prenatal or genetic models, and a key protein in schizophrenia disease, making the reelin signaling pathway in prenatal stages a target of special interest for future preclinical and clinical studies. All models presented at least one of the four described reelin alteration types. Systematic Review Registration: [https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021210568], identifier [CRD42021210568].
Collapse
Affiliation(s)
- Ana C. Sánchez-Hidalgo
- Spanish Network for Research in Mental Health (CIBERSAM), Madrid, Spain
- Seville Biomedical Research Centre (IBiS), Seville, Spain
| | - Celia Martín-Cuevas
- Spanish Network for Research in Mental Health (CIBERSAM), Madrid, Spain
- Seville Biomedical Research Centre (IBiS), Seville, Spain
| | - Benedicto Crespo-Facorro
- Spanish Network for Research in Mental Health (CIBERSAM), Madrid, Spain
- Seville Biomedical Research Centre (IBiS), Seville, Spain
- Department of Psychiatry, School of Medicine, University Hospital Virgen del Rocío-IBiS, Seville, Spain
- *Correspondence: Benedicto Crespo-Facorro,
| | - Nathalia Garrido-Torres
- Spanish Network for Research in Mental Health (CIBERSAM), Madrid, Spain
- Seville Biomedical Research Centre (IBiS), Seville, Spain
- Department of Psychiatry, School of Medicine, University Hospital Virgen del Rocío-IBiS, Seville, Spain
| |
Collapse
|
47
|
Jacquemont S, Huguet G, Klein M, Chawner SJRA, Donald KA, van den Bree MBM, Sebat J, Ledbetter DH, Constantino JN, Earl RK, McDonald-McGinn DM, van Amelsvoort T, Swillen A, O'Donnell-Luria AH, Glahn DC, Almasy L, Eichler EE, Scherer SW, Robinson E, Bassett AS, Martin CL, Finucane B, Vorstman JAS, Bearden CE, Gur RE. Genes To Mental Health (G2MH): A Framework to Map the Combined Effects of Rare and Common Variants on Dimensions of Cognition and Psychopathology. Am J Psychiatry 2022; 179:189-203. [PMID: 35236119 PMCID: PMC9345000 DOI: 10.1176/appi.ajp.2021.21040432] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Rare genomic disorders (RGDs) confer elevated risk for neurodevelopmental psychiatric disorders. In this era of intense genomics discoveries, the landscape of RGDs is rapidly evolving. However, there has not been comparable progress to date in scalable, harmonized phenotyping methods. As a result, beyond associations with categorical diagnoses, the effects on dimensional traits remain unclear for many RGDs. The nature and specificity of RGD effects on cognitive and behavioral traits is an area of intense investigation: RGDs are frequently associated with more than one psychiatric condition, and those studied to date affect, to varying degrees, a broad range of developmental and cognitive functions. Although many RGDs have large effects, phenotypic expression is typically influenced by additional genomic and environmental factors. There is emerging evidence that using polygenic risk scores in individuals with RGDs offers opportunities to refine prediction, thus allowing for the identification of those at greatest risk of psychiatric illness. However, translation into the clinic is hindered by roadblocks, which include limited genetic testing in clinical psychiatry, and the lack of guidelines for following individuals with RGDs, who are at high risk of developing psychiatric symptoms. The Genes to Mental Health Network (G2MH) is a newly funded National Institute of Mental Health initiative that will collect, share, and analyze large-scale data sets combining genomics and dimensional measures of psychopathology spanning diverse populations and geography. The authors present here the most recent understanding of the effects of RGDs on dimensional behavioral traits and risk for psychiatric conditions and discuss strategies that will be pursued within the G2MH network, as well as how expected results can be translated into clinical practice to improve patient outcomes.
Collapse
Affiliation(s)
- Sébastien Jacquemont
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Guillaume Huguet
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Marieke Klein
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Samuel J R A Chawner
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Kirsten A Donald
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Marianne B M van den Bree
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Jonathan Sebat
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - David H Ledbetter
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - John N Constantino
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Rachel K Earl
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Donna M McDonald-McGinn
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Therese van Amelsvoort
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Ann Swillen
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Anne H O'Donnell-Luria
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - David C Glahn
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Laura Almasy
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Evan E Eichler
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Stephen W Scherer
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Elise Robinson
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Anne S Bassett
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Christa Lese Martin
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Brenda Finucane
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Jacob A S Vorstman
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Carrie E Bearden
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | - Raquel E Gur
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| | -
- Department of Pediatrics, University of Montreal, Montreal (Jacquemont, Huguet); Sainte Justine Hospital Research Center, Montreal (Jacquemont, Huguet); Department of Psychiatry, University of California San Diego, La Jolla (Klein, Sebat); Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom (Chawner, van den Bree); Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa (Donald); Neuroscience Institute, University of Cape Town, Cape Town, South Africa (Donald); Autism and Developmental Medicine Institute, Geisinger, Danville, Pa. (Ledbetter, Martin, Finucane); Department of Psychiatry, Washington University School of Medicine, St. Louis (Constantino); Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle (Earl); Division of Human Genetics, 22q and You Center, Section of Clinical Genetics and Genetic Counseling, Children's Hospital of Philadelphia and Department of Pediatrics, Philadelphia (McDonald-McGinn); Perelman School of Medicine, University of Pennsylvania, Philadelphia (McDonald-McGinn); Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands (van Amelsvoort); Center for Human Genetics, University Hospital UZ Leuven, Leuven, Belgium (Swillen); Department of Human Genetics, KU Leuven, Leuven, Belgium (Swillen); Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston (O'Donnell-Luria); Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (O'Donnell-Luria); Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston (Glahn); Department of Psychiatry, Harvard Medical School, Boston (Glahn); Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Almasy); Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Lifespan Brain Institute, University of Pennsylvania, Philadelphia (Almasy); Department of Genome Sciences, University of Washington School of Medicine, Seattle (Eichler); Howard Hughes Medical Institute, University of Washington, Seattle (Eichler); Center for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Scherer); McLaughlin Center and Department of Molecular Genetics, University of Toronto, Toronto (Scherer); Harvard T.H. Chan School of Public Health and Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Mass. (Robinson); Department of Psychiatry, Dalglish Family 22q Clinic, University Health Network, Toronto (Bassett); Department of Psychiatry, Toronto General Hospital Research Institute, University Health Network, Toronto (Bassett); Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto Clinical Genetics Research Program, Center for Addiction and Mental Health, Toronto (Bassett); Department of Psychiatry, University of Toronto, Toronto (Vorstman); Centre for Applied Genomics and Program in Genetics and Genomic Biology, Hospital for Sick Children, Toronto (Vorstman);Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (Bearden); Department of Psychology, University of California, Los Angeles (Bearden); Lifespan Brain Institute, Penn Medicine, and Children's Hospital of Philadelphia, Philadelphia (Gur); Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Gur); Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia (Gur)
| |
Collapse
|
48
|
Wang R, Chopra N, Nho K, Maloney B, Obukhov AG, Nelson PT, Counts SE, Lahiri DK. Human microRNA (miR-20b-5p) modulates Alzheimer's disease pathways and neuronal function, and a specific polymorphism close to the MIR20B gene influences Alzheimer's biomarkers. Mol Psychiatry 2022; 27:1256-1273. [PMID: 35087196 PMCID: PMC9054681 DOI: 10.1038/s41380-021-01351-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder with loss of cognitive, executive, and other mental functions, and is the most common form of age-related dementia. Amyloid-β peptide (Aβ) contributes to the etiology and progression of the disease. Aβ is derived from the amyloid-β precursor protein (APP). Multiple microRNA (miRNA) species are also implicated in AD. We report that human hsa-miR20b-5p (miR-20b), produced from the MIR20B gene on Chromosome X, may play complex roles in AD pathogenesis, including Aβ regulation. Specifically, miR-20b-5p miRNA levels were altered in association with disease progression in three regions of the human brain: temporal neocortex, cerebellum, and posterior cingulate cortex. In cultured human neuronal cells, miR-20b-5p treatment interfered with calcium homeostasis, neurite outgrowth, and branchpoints. A single-nucleotide polymorphism (SNP) upstream of the MIR20B gene (rs13897515) associated with differences in levels of cerebrospinal fluid (CSF) Aβ1-42 and thickness of the entorhinal cortex. We located a miR-20b-5p binding site in the APP mRNA 3'-untranslated region (UTR), and treatment with miR-20b-5p reduced APP mRNA and protein levels. Network analysis of protein-protein interactions and gene coexpression revealed other important potential miR-20b-5p targets among AD-related proteins/genes. MiR-20b-5p, a miRNA that downregulated APP, was paradoxically associated with an increased risk for AD. However, miR-20b-5p also reduced, and the blockade of APP by siRNA likewise reduced calcium influx. As APP plays vital roles in neuronal health and does not exist solely to be the source of "pathogenic" Aβ, the molecular etiology of AD is likely to not just be a disease of "excess" but a disruption of delicate homeostasis.
Collapse
Affiliation(s)
- Ruizhi Wang
- Laboratory of Molecular Neurogenetics, Department of Psychiatry, Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Nipun Chopra
- Laboratory of Molecular Neurogenetics, Department of Psychiatry, Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- DePauw University, Greencastle, IN, 46135, USA
| | - Kwangsik Nho
- Radiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Bryan Maloney
- Laboratory of Molecular Neurogenetics, Department of Psychiatry, Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Alexander G Obukhov
- Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Kentucky Alzheimer's Disease Research Center, Lexington, KY, 40536, USA
| | - Scott E Counts
- Departments of Translational Neuroscience & Family Medicine, Michigan State University, Grand Rapids, and Michigan Alzheimer's Disease Research Center, Ann Arbor, MI, USA
| | - Debomoy K Lahiri
- Laboratory of Molecular Neurogenetics, Department of Psychiatry, Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
49
|
Barbagallo D, Palermo CI, Barbagallo C, Battaglia R, Caponnetto A, Spina V, Ragusa M, Di Pietro C, Scalia G, Purrello M. Competing endogenous RNA network mediated by circ_3205 in SARS-CoV-2 infected cells. Cell Mol Life Sci 2022; 79:75. [PMID: 35039944 PMCID: PMC8763136 DOI: 10.1007/s00018-021-04119-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a new member of the Betacoronaviridae family, responsible for the recent pandemic outbreak of COVID-19. To start exploring the molecular events that follow host cell infection, we queried VirusCircBase and identified a circular RNA (circRNA) predicted to be synthesized by SARS-CoV-2, circ_3205, which we used to probe: (i) a training cohort comprised of two pools of cells from three nasopharyngeal swabs of SARS-CoV-2 infected (positive) or uninfected (negative, UCs) individuals; (ii) a validation cohort made up of 12 positive and 3 negative samples. The expression of circRNAs, miRNAs and miRNA targets was assayed through real-time PCR. CircRNA-miRNA interactions were predicted by TarpMiR, Analysis of Common Targets for circular RNAs (ACT), and STarMir tools. Enrichment of the biological processes and the list of predicted miRNA targets were retrieved from DIANA miRPath v3.0. Our results showed that the predicted SARS-CoV-2 circ_3205 was expressed only in positive samples and its amount positively correlated with that of SARS-CoV-2 Spike (S) mRNA and the viral load (r values = 0.80952 and 0.84867, Spearman's correlation test, respectively). Human (hsa) miR-298 was predicted to interact with circ_3205 by all three predictive tools. KCNMB4 and PRKCE were predicted as hsa-miR-298 targets. Interestingly, the function of both is correlated with blood coagulation and immune response. KCNMB4 and PRKCE mRNAs were upregulated in positive samples as compared to UCs (6 and 8.1-fold, p values = 0.049 and 0.02, Student's t test, respectively) and their expression positively correlated with that of circ_3205 (r values = 0.6 and 0.25, Spearman's correlation test, respectively). We propose that our results convincingly suggest that circ_3205 is a circRNA synthesized by SARS-CoV-2 upon host cell infection and that it may behave as a competitive endogenous RNA (ceRNA), sponging hsa-miR-298 and contributing to the upregulation of KCNMB4 and PRKCE mRNAs.
Collapse
Affiliation(s)
- Davide Barbagallo
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics Giovanni Sichel, University of Catania, 95123, Catania, Italy.
| | - Concetta Ilenia Palermo
- U.O.C. Laboratory Analysis Unit, A.O.U. Policlinico‑Vittorio Emanuele, 95123, Catania, Italy
| | - Cristina Barbagallo
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics Giovanni Sichel, University of Catania, 95123, Catania, Italy
| | - Rosalia Battaglia
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics Giovanni Sichel, University of Catania, 95123, Catania, Italy
| | - Angela Caponnetto
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics Giovanni Sichel, University of Catania, 95123, Catania, Italy
| | - Vittoria Spina
- Department of Biomedical and Biotechnological Sciences, Section of Medical Biochemistry, University of Catania, 95123, Catania, Italy
| | - Marco Ragusa
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics Giovanni Sichel, University of Catania, 95123, Catania, Italy
| | - Cinzia Di Pietro
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics Giovanni Sichel, University of Catania, 95123, Catania, Italy
| | - Guido Scalia
- Department of Biomedical and Biotechnological Sciences, Section of Microbiology, University of Catania, 95123, Catania, Italy
| | - Michele Purrello
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics Giovanni Sichel, University of Catania, 95123, Catania, Italy
| |
Collapse
|
50
|
Contribution of schizophrenia polygenic burden to longitudinal phenotypic variance in 22q11.2 deletion syndrome. Mol Psychiatry 2022; 27:4191-4200. [PMID: 35768638 PMCID: PMC9718680 DOI: 10.1038/s41380-022-01674-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/01/2022] [Accepted: 06/10/2022] [Indexed: 02/07/2023]
Abstract
While the recurrent 22q11.2 deletion is one of the strongest genetic risk factors for schizophrenia (SCZ), variability of its associated neuropsychiatric endophenotypes reflects its incomplete penetrance for psychosis development. To assess whether this phenotypic variability is linked to common variants associated with SCZ, we studied the association between SCZ polygenic risk score (PRS) and longitudinally acquired phenotypic information of the Swiss 22q11.2DS cohort (n = 97, 50% females, mean age 17.7 yr, mean visit interval 3.8 yr). The SCZ PRS with the best predictive performance was ascertained in the Estonian Biobank (n = 201,146) with LDpred. The infinitesimal SCZ PRS model showed the strongest capacity in discriminating SCZ cases from controls with one SD difference in SCZ PRS corresponding to an odds ratio (OR) of 1.73 (95% CI 1.57-1.90, P = 1.47 × 10-29). In 22q11.2 patients, random-effects ordinal regression modelling using longitudinal data showed SCZ PRS to have the strongest effect on social anhedonia (OR = 2.09, P = 0.0002), and occupational functioning (OR = 1.82, P = 0.0003) within the negative symptoms course, and dysphoric mood (OR = 2.00, P = 0.002) and stress intolerance (OR = 1.76, P = 0.0002) within the general symptoms course. Genetic liability for SCZ was additionally associated with full scale cognitive decline (β = -0.25, P = 0.02) and with longitudinal volumetric reduction of the right and left hippocampi (β = -0.28, P = 0.005; β = -0.23, P = 0.02, respectively). Our results indicate that the polygenic contribution to SCZ acts upon the threshold-lowering first hit (i.e., the deletion). It modifies the endophenotypes of 22q11.2DS and augments the derailment of developmental trajectories of negative and general symptoms, cognition, and hippocampal volume.
Collapse
|