1
|
Hafeman DM, Uher R, Merranko J, Zwicker A, Goldstein B, Goldstein TR, Axelson D, Monk K, Sakolsky D, Iyengar S, Diler R, Nimgaonkar V, Birmaher B. Person-level contributions of bipolar polygenic risk score to the prediction of new-onset bipolar disorder in at-risk offspring. J Affect Disord 2025; 368:359-365. [PMID: 39299598 DOI: 10.1016/j.jad.2024.09.107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/12/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Previous work indicates that polygenic risk scores (PRS) for bipolar disorder (BD) are elevated in adults and youth with BD, but whether BD-PRS can inform person-level diagnostic prediction is unknown. Here, we test whether BD-PRS improves performance of a previously published risk calculator (RC) for BD. METHODS 156 parents with BD-I/II and their offspring ages 6-18 were recruited and evaluated with standardized diagnostic assessments every two years for >12 years. DNA was extracted from saliva samples, genotyping performed, and BD-PRS calculated based on a 2021 meta-analysis. Using a bootstrapped and cross-validated penalized Cox regression, we assessed whether BD-PRS (alone and interacting with clinical variables) improved RC performance. RESULTS Of 227 offspring, 38 developed BD during follow-up. The penalized regression selected BD-PRS and interactions between BD-PRS and parental age at mood disorder onset (AAO), depression, and anxiety. The resulting RC discriminated offspring who developed BD (vs. those that did not) with good accuracy (AUC = 0.81); removing BD-PRS and its interaction terms was associated with a significant decrement to the AUC (decrement = 0.07, p = 0.039). Further exploration of selected interaction terms indicated that all were significant (p-values<0.02), indicating that BD-PRS has a larger effect on the outcome in offspring with depression and anxiety, whose affected parent had a younger AAO. CONCLUSIONS The addition of BD-PRS to clinical/demographic predictors in the RC significantly improved its accuracy. BD-PRS predicted BD on the person-level, particularly in offspring of parents with earlier AAO who already had symptoms of anxiety and depression at intake.
Collapse
Affiliation(s)
- Danella M Hafeman
- University of Pittsburgh School of Medicine, Department of Psychiatry, Western Psychiatric Hospital, University of Pittsburgh Medical Center, United States of America.
| | - Rudolf Uher
- Dalhousie University, Department of Psychiatry, Canada
| | - John Merranko
- University of Pittsburgh School of Medicine, Department of Psychiatry, Western Psychiatric Hospital, University of Pittsburgh Medical Center, United States of America
| | | | - Benjamin Goldstein
- Center for Addiction and Mental Health, University of Toronto Faculty of Medicine, Canada
| | - Tina R Goldstein
- University of Pittsburgh School of Medicine, Department of Psychiatry, Western Psychiatric Hospital, University of Pittsburgh Medical Center, United States of America
| | - David Axelson
- Nationwide Children's Hospital and The Ohio State College of Medicine, United States of America
| | - Kelly Monk
- University of Pittsburgh School of Medicine, Department of Psychiatry, Western Psychiatric Hospital, University of Pittsburgh Medical Center, United States of America
| | - Dara Sakolsky
- University of Pittsburgh School of Medicine, Department of Psychiatry, Western Psychiatric Hospital, University of Pittsburgh Medical Center, United States of America
| | - Satish Iyengar
- University of Pittsburgh, Department of Statistics, United States of America
| | - Rasim Diler
- University of Pittsburgh School of Medicine, Department of Psychiatry, Western Psychiatric Hospital, University of Pittsburgh Medical Center, United States of America
| | - Vishwajit Nimgaonkar
- University of Pittsburgh School of Medicine, Department of Psychiatry, Western Psychiatric Hospital, University of Pittsburgh Medical Center, United States of America
| | - Boris Birmaher
- University of Pittsburgh School of Medicine, Department of Psychiatry, Western Psychiatric Hospital, University of Pittsburgh Medical Center, United States of America
| |
Collapse
|
2
|
Hartwell EE, Jinwala Z, Milone J, Ramirez S, Gelernter J, Kranzler HR, Kember RL. Application of polygenic scores to a deeply phenotyped sample enriched for substance use disorders reveals extensive pleiotropy with psychiatric and somatic traits. Neuropsychopharmacology 2024; 49:1958-1967. [PMID: 39043921 PMCID: PMC11480112 DOI: 10.1038/s41386-024-01922-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/07/2024] [Accepted: 06/28/2024] [Indexed: 07/25/2024]
Abstract
Co-occurring psychiatric, medical, and substance use disorders (SUDs) are common, but the complex pathways leading to such comorbidities are poorly understood. A greater understanding of genetic influences on this phenomenon could inform precision medicine efforts. We used the Yale-Penn dataset, a cross-sectional sample enriched for individuals with SUDs, to examine pleiotropic effects of genetic liability for psychiatric and somatic traits. Participants completed an in-depth interview that provides information on demographics, environment, medical illnesses, and psychiatric and SUDs. Polygenic scores (PGS) for psychiatric disorders and somatic traits were calculated in European-ancestry (EUR; n = 5691) participants and, when discovery datasets were available, for African-ancestry (AFR; n = 4918) participants. Phenome-wide association studies (PheWAS) were then conducted. In AFR participants, the only PGS with significant associations was bipolar disorder (BD), all of which were with substance use phenotypes. In EUR participants, PGS for major depressive disorder (MDD), generalized anxiety disorder (GAD), post-traumatic stress disorder (PTSD), schizophrenia (SCZ), body mass index (BMI), coronary artery disease (CAD), and type 2 diabetes (T2D) all showed significant associations, the majority of which were with phenotypes in the substance use categories. For instance, PGSMDD was associated with over 200 phenotypes, 15 of which were depression-related (e.g., depression criterion count), 55 of which were other psychiatric phenotypes, and 126 of which were substance use phenotypes; and PGSBMI was associated with 138 phenotypes, 105 of which were substance related. Genetic liability for psychiatric and somatic traits is associated with numerous phenotypes across multiple categories, indicative of the broad genetic liability of these traits.
Collapse
Affiliation(s)
- Emily E Hartwell
- Crescenz VA Medical Center, Philadelphia, PA, USA
- University of Pennsylvania, Philadelphia, PA, USA
| | - Zeal Jinwala
- Crescenz VA Medical Center, Philadelphia, PA, USA
- University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Joel Gelernter
- West Haven VA Medical Center, West Haven, CT, USA
- Yale University, New Haven, CT, USA
| | - Henry R Kranzler
- Crescenz VA Medical Center, Philadelphia, PA, USA
- University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel L Kember
- Crescenz VA Medical Center, Philadelphia, PA, USA.
- University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Stern S, Zhang L, Wang M, Wright R, Rosh I, Hussein Y, Stern T, Choudhary A, Tripathi U, Reed P, Sadis H, Nayak R, Shemen A, Agarwal K, Cordeiro D, Peles D, Hang Y, Mendes APD, Baul TD, Roth JG, Coorapati S, Boks MP, McCombie WR, Hulshoff Pol H, Brennand KJ, Réthelyi JM, Kahn RS, Marchetto MC, Gage FH. Monozygotic twins discordant for schizophrenia differ in maturation and synaptic transmission. Mol Psychiatry 2024; 29:3208-3222. [PMID: 38704507 PMCID: PMC11449799 DOI: 10.1038/s41380-024-02561-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 04/01/2024] [Accepted: 04/12/2024] [Indexed: 05/06/2024]
Abstract
Schizophrenia affects approximately 1% of the world population. Genetics, epigenetics, and environmental factors are known to play a role in this psychiatric disorder. While there is a high concordance in monozygotic twins, about half of twin pairs are discordant for schizophrenia. To address the question of how and when concordance in monozygotic twins occur, we have obtained fibroblasts from two pairs of schizophrenia discordant twins (one sibling with schizophrenia while the second one is unaffected by schizophrenia) and three pairs of healthy twins (both of the siblings are healthy). We have prepared iPSC models for these 3 groups of patients with schizophrenia, unaffected co-twins, and the healthy twins. When the study started the co-twins were considered healthy and unaffected but both the co-twins were later diagnosed with a depressive disorder. The reprogrammed iPSCs were differentiated into hippocampal neurons to measure the neurophysiological abnormalities in the patients. We found that the neurons derived from the schizophrenia patients were less arborized, were hypoexcitable with immature spike features, and exhibited a significant reduction in synaptic activity with dysregulation in synapse-related genes. Interestingly, the neurons derived from the co-twin siblings who did not have schizophrenia formed another distinct group that was different from the neurons in the group of the affected twin siblings but also different from the neurons in the group of the control twins. Importantly, their synaptic activity was not affected. Our measurements that were obtained from schizophrenia patients and their monozygotic twin and compared also to control healthy twins point to hippocampal synaptic deficits as a central mechanism in schizophrenia.
Collapse
Affiliation(s)
- Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel.
| | - Lei Zhang
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Meiyan Wang
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Rebecca Wright
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Idan Rosh
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Yara Hussein
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Tchelet Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Ashwani Choudhary
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Utkarsh Tripathi
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Patrick Reed
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Hagit Sadis
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Ritu Nayak
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Aviram Shemen
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Karishma Agarwal
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Diogo Cordeiro
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - David Peles
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Yuqing Hang
- Razavi Newman Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ana P D Mendes
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Tithi D Baul
- Department of Psychiatry at the Boston Medical Center, Boston, MA, USA
| | - Julien G Roth
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Shashank Coorapati
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Marco P Boks
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
| | | | - Hilleke Hulshoff Pol
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
- Department of Experimental Psychology, Utrecht University, Heidelberglaan 1, 3584CS, Utrecht, The Netherlands
| | - Kristen J Brennand
- Nash Family Department of Neuroscience, Friedman Brain Institute, Pamela Sklar Division of Psychiatric Genomics, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Department of Genetics, Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - János M Réthelyi
- Molecular Psychiatry Research Group and Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| | - René S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, New York, NY, USA
| | - Maria C Marchetto
- Department of Anthropology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
4
|
Gnatowski ER, Jurmain JL, Dozmorov MG, Wolstenholme JT, Miles MF. Ninein, a candidate gene for ethanol anxiolysis, shows complex exon-specific expression and alternative splicing differences between C57BL/6J and DBA/2J mice. Front Genet 2024; 15:1455616. [PMID: 39323865 PMCID: PMC11422218 DOI: 10.3389/fgene.2024.1455616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/29/2024] [Indexed: 09/27/2024] Open
Abstract
Ethanol's anxiolytic actions contribute to increased consumption and the development of Alcohol Use Disorder (AUD). Our laboratory previously identified genetic loci contributing to the anxiolytic-like properties of ethanol in BXD recombinant inbred mice, derived from C57BL/6J (B6) and DBA/2J (D2) progenitor strains. That work identified Ninein (Nin) as a candidate gene underlying ethanol's acute anxiolytic-like properties in BXD mice. Nin has a complex exonic content with known alternative splicing events that alter cellular distribution of the NIN protein. We hypothesize that strain-specific differences in Nin alternative splicing contribute to changes in Nin gene expression and B6/D2 strain differences in ethanol anxiolysis. Using quantitative reverse-transcriptase PCR to target specific Nin splice variants, we identified isoform-specific exon expression differences between B6 and D2 mice in prefrontal cortex, nucleus accumbens and amygdala. We extended this analysis using deep RNA sequencing in B6 and D2 nucleus accumbens samples and found that total Nin expression was significantly higher in D2 mice. Furthermore, exon utilization and alternative splicing analyses identified eight differentially utilized exons and significant exon-skipping events between the strains, including three novel splicing events in the 3' end of the Nin gene that were specific to the D2 strain. Additionally, we document multiple single nucleotide polymorphisms in D2 Nin exons that are predicted to have deleterious effects on protein function. Our studies provide the first in-depth analysis of Nin alternative splicing in brain and identify a potential genetic mechanism altering Nin expression and function between B6 and D2 mice, thus possibly contributing to differences in the anxiolytic-like properties of ethanol between these strains. This work adds novel information to our understanding of genetic differences modulating ethanol actions on anxiety that may contribute to the risk for alcohol use disorder.
Collapse
Affiliation(s)
- E. R. Gnatowski
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, United States
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, United States
| | - J. L. Jurmain
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, United States
| | - M. G. Dozmorov
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, United States
- Department of Biostatistics, Virginia Commonwealth University, Richmond, United States
| | - J. T. Wolstenholme
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, United States
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, United States
| | - M. F. Miles
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, United States
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, United States
| |
Collapse
|
5
|
Puerta R, de Rojas I, García-González P, Olivé C, Sotolongo-Grau O, García-Sánchez A, García-Gutiérrez F, Montrreal L, Pablo Tartari J, Sanabria Á, Pytel V, Lage C, Quintela I, Aguilera N, Rodriguez-Rodriguez E, Alarcón-Martín E, Orellana A, Pastor P, Pérez-Tur J, Piñol-Ripoll G, de Munian AL, García-Alberca JM, Royo JL, Bullido MJ, Álvarez V, Real LM, Anchuelo AC, Gómez-Garre D, Larrad MTM, Franco-Macías E, Mir P, Medina M, Sánchez-Valle R, Dols-Icardo O, Sáez ME, Carracedo Á, Tárraga L, Alegret M, Valero S, Marquié M, Boada M, Juan PS, Cavazos JE, Cabrera A, Cano A. Connecting genomic and proteomic signatures of amyloid burden in the brain. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.06.24313124. [PMID: 39281766 PMCID: PMC11398581 DOI: 10.1101/2024.09.06.24313124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Background Alzheimer's disease (AD) has a high heritable component characteristic of complex diseases, yet many of the genetic risk factors remain unknown. We combined genome-wide association studies (GWAS) on amyloid endophenotypes measured in cerebrospinal fluid (CSF) and positron emission tomography (PET) as surrogates of amyloid pathology, which may be helpful to understand the underlying biology of the disease. Methods We performed a meta-analysis of GWAS of CSF Aβ42 and PET measures combining six independent cohorts (n=2,076). Due to the opposite effect direction of Aβ phenotypes in CSF and PET measures, only genetic signals in the opposite direction were considered for analysis (n=376,599). Polygenic risk scores (PRS) were calculated and evaluated for AD status and amyloid endophenotypes. We then searched the CSF proteome signature of brain amyloidosis using SOMAscan proteomic data (Ace cohort, n=1,008) and connected it with GWAS results of loci modulating amyloidosis. Finally, we compared our results with a large meta-analysis using publicly available datasets in CSF (n=13,409) and PET (n=13,116). This combined approach enabled the identification of overlapping genes and proteins associated with amyloid burden and the assessment of their biological significance using enrichment analyses. Results After filtering the meta-GWAS, we observed genome-wide significance in the rs429358-APOE locus and nine suggestive hits were annotated. We replicated the APOE loci using the large CSF-PET meta-GWAS and identified multiple AD-associated genes as well as the novel GADL1 locus. Additionally, we found a significant association between the AD PRS and amyloid levels, whereas no significant association was found between any Aβ PRS with AD risk. CSF SOMAscan analysis identified 1,387 FDR-significant proteins associated with CSF Aβ42 levels. The overlap among GWAS loci and proteins associated with amyloid burden was very poor (n=35). The enrichment analysis of overlapping hits strongly suggested several signalling pathways connecting amyloidosis with the anchored component of the plasma membrane, synapse physiology and mental disorders that were replicated in the large CSF-PET meta-analysis. Conclusions The strategy of combining CSF and PET amyloid endophenotypes GWAS with CSF proteome analyses might be effective for identifying signals associated with the AD pathological process and elucidate causative molecular mechanisms behind the amyloid mobilization in AD.
Collapse
Affiliation(s)
- Raquel Puerta
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- Universitat de Barcelona (UB)
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pablo García-González
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Clàudia Olivé
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | | | | | | | - Laura Montrreal
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Juan Pablo Tartari
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Ángela Sanabria
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Vanesa Pytel
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Carmen Lage
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | - Inés Quintela
- Grupo de Medicina Xenómica, Centro Nacional de Genotipado (CEGEN-PRB3-ISCIII). Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Nuria Aguilera
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Eloy Rodriguez-Rodriguez
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | | | - Adelina Orellana
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pau Pastor
- Unit of Neurodegenerative diseases, Department of Neurology, University Hospital Germans Trias i Pujol, Badalona, Barcelona, Spain
- The Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Jordi Pérez-Tur
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Unitat de Genètica Molecular, Institut de Biomedicina de València-CSIC, Valencia, Spain
- Unidad Mixta de Neurologia Genètica, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Gerard Piñol-Ripoll
- Unitat Trastorns Cognitius, Hospital Universitari Santa Maria de Lleida, Lleida, Spain
- Institut de Recerca Biomedica de Lleida (IRBLLeida), Lleida, Spain
| | - Adolfo López de Munian
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Department of Neurology. Hospital Universitario Donostia. San Sebastian, Spain
- Department of Neurosciences. Faculty of Medicine and Nursery. University of the Basque Country, San Sebastián, Spain
- Neurosciences Area. Instituto Biodonostia. San Sebastian, Spain
| | - Jose María García-Alberca
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Alzheimer Research Center & Memory Clinic, Andalusian Institute for Neuroscience, Málaga, Spain
| | - Jose Luís Royo
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología. School of Medicine. University of Malaga. Málaga, Spain
| | - María Jesús Bullido
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC)
- Instituto de Investigacion Sanitaria ‘Hospital la Paz’ (IdIPaz), Madrid, Spain
- Universidad Autónoma de Madrid
| | - Victoria Álvarez
- Laboratorio de Genética. Hospital Universitario Central de Asturias, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)
| | - Luis Miguel Real
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología. School of Medicine. University of Malaga. Málaga, Spain
- Unidad Clínica de Enfermedades Infecciosas y Microbiología.Hospital Universitario de Valme, Sevilla, Spain
| | - Arturo Corbatón Anchuelo
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
| | - Dulcenombre Gómez-Garre
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
- Laboratorio de Riesgo Cardiovascular y Microbiota, Hospital Clínico San Carlos; Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid (UCM)
- Biomedical Research Networking Center in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - María Teresa Martínez Larrad
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)
| | - Emilio Franco-Macías
- Dementia Unit, Department of Neurology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS), Sevilla, Spain
| | - Pablo Mir
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología. Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Miguel Medina
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- CIEN Foundation/Queen Sofia Foundation Alzheimer Center
| | - Raquel Sánchez-Valle
- Alzheimer’s disease and other cognitive disorders unit. Service of Neurology. Hospital Clínic of Barcelona. Institut d’Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - Oriol Dols-Icardo
- Department of Neurology, Sant Pau Memory Unit, Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Ángel Carracedo
- Grupo de Medicina Xenómica, Centro Nacional de Genotipado (CEGEN-PRB3-ISCIII). Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica – CIBERER-IDIS, Santiago de Compostela, Spain
| | - Lluís Tárraga
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Montse Alegret
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Sergi Valero
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Marta Marquié
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pascual Sánchez Juan
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | - Jose Enrique Cavazos
- South Texas Medical Science Training Program, University of Texas Health San Antonio, San Antonio
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 USA
| | - Alfredo Cabrera
- Neuroscience Therapeutic Area, Janssen Research & Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Amanda Cano
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Alzheimer’s Disease Neuroimaging Initiative.
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 USA
| |
Collapse
|
6
|
Bigdeli TB, Chatzinakos C, Bendl J, Barr PB, Venkatesh S, Gorman BR, Clarence T, Genovese G, Iyegbe CO, Peterson RE, Kolokotronis SO, Burstein D, Meyers JL, Li Y, Rajeevan N, Sayward F, Cheung KH, DeLisi LE, Kosten TR, Zhao H, Achtyes E, Buckley P, Malaspina D, Lehrer D, Rapaport MH, Braff DL, Pato MT, Fanous AH, Pato CN, Huang GD, Muralidhar S, Michael Gaziano J, Pyarajan S, Girdhar K, Lee D, Hoffman GE, Aslan M, Fullard JF, Voloudakis G, Harvey PD, Roussos P. Biological Insights from Schizophrenia-associated Loci in Ancestral Populations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.27.24312631. [PMID: 39252912 PMCID: PMC11383513 DOI: 10.1101/2024.08.27.24312631] [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
Large-scale genome-wide association studies of schizophrenia have uncovered hundreds of associated loci but with extremely limited representation of African diaspora populations. We surveyed electronic health records of 200,000 individuals of African ancestry in the Million Veteran and All of Us Research Programs, and, coupled with genotype-level data from four case-control studies, realized a combined sample size of 13,012 affected and 54,266 unaffected persons. Three genome-wide significant signals - near PLXNA4, PMAIP1, and TRPA1 - are the first to be independently identified in populations of predominantly African ancestry. Joint analyses of African, European, and East Asian ancestries across 86,981 cases and 303,771 controls, yielded 376 distinct autosomal loci, which were refined to 708 putatively causal variants via multi-ancestry fine-mapping. Utilizing single-cell functional genomic data from human brain tissue and two complementary approaches, transcriptome-wide association studies and enhancer-promoter contact mapping, we identified a consensus set of 94 genes across ancestries and pinpointed the specific cell types in which they act. We identified reproducible associations of schizophrenia polygenic risk scores with schizophrenia diagnoses and a range of other mental and physical health problems. Our study addresses a longstanding gap in the generalizability of research findings for schizophrenia across ancestral populations, underlining shared biological underpinnings of schizophrenia across global populations in the presence of broadly divergent risk allele frequencies.
Collapse
Affiliation(s)
- Tim B. Bigdeli
- VA New York Harbor Healthcare System, Brooklyn, NY
- Department of Psychiatry and Behavioral Sciences and SUNY Downstate Health Sciences University, Brooklyn, NY
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Chris Chatzinakos
- Department of Psychiatry and Behavioral Sciences and SUNY Downstate Health Sciences University, Brooklyn, NY
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Jaroslav Bendl
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Peter B. Barr
- VA New York Harbor Healthcare System, Brooklyn, NY
- Department of Psychiatry and Behavioral Sciences and SUNY Downstate Health Sciences University, Brooklyn, NY
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Sanan Venkatesh
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Bryan R. Gorman
- Massachusetts Area Veterans Epidemiology, Research, and Information Center (MAVERIC), Jamaica Plain, MA
| | - Tereza Clarence
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Giulio Genovese
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - Conrad O. Iyegbe
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
| | - Roseann E. Peterson
- VA New York Harbor Healthcare System, Brooklyn, NY
- Department of Psychiatry and Behavioral Sciences and SUNY Downstate Health Sciences University, Brooklyn, NY
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Sergios-Orestis Kolokotronis
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY
- Division of Infectious Diseases, Department of Medicine, College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY
- Department of Cell Biology, College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY
| | - David Burstein
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
- Mental Illness Research, Education and Clinical Center VISN2, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Jacquelyn L. Meyers
- Department of Psychiatry and Behavioral Sciences and SUNY Downstate Health Sciences University, Brooklyn, NY
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Yuli Li
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - Nallakkandi Rajeevan
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - Frederick Sayward
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - Kei-Hoi Cheung
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | | | | | | | - Lynn E. DeLisi
- Department of Psychiatry, Cambridge Health Alliance, Cambridge, MA
| | - Thomas R. Kosten
- Michael E. DeBakey VA Medical Center, Houston, TX
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX
| | - Hongyu Zhao
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - Eric Achtyes
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI
| | - Peter Buckley
- University of Tennessee Health Science Center in Memphis, TN
| | - Dolores Malaspina
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Douglas Lehrer
- Department of Psychiatry, Wright State University, Dayton, OH
| | - Mark H. Rapaport
- Huntsman Mental Health Institute, Department of Psychiatry, University of Utah, Salt Lake City, UT
| | - David L. Braff
- Department of Psychiatry, University of California, San Diego, CA
- VA San Diego Healthcare System, San Diego, CA
| | - Michele T. Pato
- Department of Psychiatry, Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Ayman H. Fanous
- Department of Psychiatry, University of Arizona College of Medicine-Phoenix, Phoenix, AZ
- Department of Psychiatry, VA Phoenix Healthcare System, Phoenix, AZ
| | - Carlos N. Pato
- Department of Psychiatry, Robert Wood Johnson Medical School, New Brunswick, NJ
| | | | | | | | - Grant D. Huang
- Office of Research and Development, Veterans Health Administration, Washington, DC
| | - Sumitra Muralidhar
- Office of Research and Development, Veterans Health Administration, Washington, DC
| | - J. Michael Gaziano
- Massachusetts Area Veterans Epidemiology, Research, and Information Center (MAVERIC), Jamaica Plain, MA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Saiju Pyarajan
- Massachusetts Area Veterans Epidemiology, Research, and Information Center (MAVERIC), Jamaica Plain, MA
| | - Kiran Girdhar
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Donghoon Lee
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Gabriel E. Hoffman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
- Mental Illness Research, Education and Clinical Center VISN2, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Mihaela Aslan
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - John F. Fullard
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Georgios Voloudakis
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
- Mental Illness Research, Education and Clinical Center VISN2, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Philip D. Harvey
- Bruce W. Carter Miami Veterans Affairs (VA) Medical Center, Miami, FL
- University of Miami School of Medicine, Miami, FL
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
- Mental Illness Research, Education and Clinical Center VISN2, James J. Peters VA Medical Center, Bronx, NY, USA
| |
Collapse
|
7
|
Chen M, Zhang Y, Shi W, Song X, Yang Y, Hou G, Ding H, Chen S, Yang W, Shen N, Cui Y, Zuo X, Tang Y. SPATS2L is a positive feedback regulator of the type I interferon signaling pathway and plays a vital role in lupus. Acta Biochim Biophys Sin (Shanghai) 2024. [PMID: 39099414 DOI: 10.3724/abbs.2024132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/06/2024] Open
Abstract
Through genome-wide association studies (GWAS) and integrated expression quantitative trait locus (eQTL) analyses, numerous susceptibility genes ("eGenes", whose expressions are significantly associated with common variants) associated with systemic lupus erythematosus (SLE) have been identified. Notably, a subset of these eGenes is correlated with disease activity. However, the precise mechanisms through which these genes contribute to the initiation and progression of the disease remain to be fully elucidated. In this investigation, we initially identify SPATS2L as an SLE eGene correlated with disease activity. eSignaling and transcriptomic analyses suggest its involvement in the type I interferon (IFN) pathway. We observe a significant increase in SPATS2L expression following type I IFN stimulation, and the expression levels are dependent on both the concentration and duration of stimulation. Furthermore, through dual-luciferase reporter assays, western blot analysis, and imaging flow cytometry, we confirm that SPATS2L positively modulates the type I IFN pathway, acting as a positive feedback regulator. Notably, siRNA-mediated intervention targeting SPATS2L, an interferon-inducible gene, in peripheral blood mononuclear cells (PBMCs) from patients with SLE reverses the activation of the interferon pathway. In conclusion, our research highlights the pivotal role of SPATS2L as a positive-feedback regulatory molecule within the type I IFN pathway. Our findings suggest that SPATS2L plays a critical role in the onset and progression of SLE and may serve as a promising target for disease activity assessment and intervention strategies.
Collapse
Affiliation(s)
- Mengke Chen
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200001, China
| | - Yutong Zhang
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200001, China
| | - Weiwen Shi
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200001, China
| | - Xuejiao Song
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yue Yang
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| | - Guojun Hou
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200001, China
| | - Huihua Ding
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200001, China
| | - Sheng Chen
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200001, China
| | - Wanling Yang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Nan Shen
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200001, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200032, China
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH 45229, USA
| | - Yong Cui
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xianbo Zuo
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yuanjia Tang
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200001, China
| |
Collapse
|
8
|
Kranzler HR, Davis CN, Feinn R, Jinwala Z, Khan Y, Oikonomou A, Silva-Lopez D, Burton I, Dixon M, Milone J, Ramirez S, Shifman N, Levey D, Gelernter J, Hartwell EE, Kember RL. Gene × environment effects and mediation involving adverse childhood events, mood and anxiety disorders, and substance dependence. Nat Hum Behav 2024; 8:1616-1627. [PMID: 38834750 DOI: 10.1038/s41562-024-01885-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 04/10/2024] [Indexed: 06/06/2024]
Abstract
Adverse childhood events (ACEs) contribute to the development of mood and anxiety disorders and substance dependence. However, the extent to which these effects are direct or indirect and whether genetic risk moderates them is unclear. We examined associations among ACEs, mood/anxiety disorders and substance dependence in 12,668 individuals (44.9% female, 42.5% African American/Black, 42.1% European American/white). Using latent variables for each phenotype, we modelled direct and indirect associations of ACEs with substance dependence, mediated by mood/anxiety disorders (the forward or 'self-medication' model) and of ACEs with mood/anxiety disorders, mediated by substance dependence (the reverse or 'substance-induced' model). In a subsample, we tested polygenic scores for the substance dependence and mood/anxiety disorder factors as moderators in the mediation models. Although there were significant indirect paths in both directions, mediation by mood/anxiety disorders (the forward model) was greater than that by substance dependence (the reverse model). Greater genetic risk for substance use disorders was associated with a weaker direct association between ACEs and substance dependence in both ancestry groups (reflecting gene × environment interactions) and a weaker indirect association in European-ancestry individuals (reflecting moderated mediation). We found greater evidence that substance dependence reflects self-medication of mood/anxiety disorders than that mood/anxiety disorders are substance induced. Among individuals at higher genetic risk for substance dependence, ACEs were less associated with that outcome. Following exposure to ACEs, multiple pathways appear to underlie the associations between mood/anxiety disorders and substance dependence. Specification of these pathways could inform individually targeted prevention and treatment approaches.
Collapse
Affiliation(s)
- Henry R Kranzler
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA.
| | - Christal N Davis
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
| | - Richard Feinn
- Department of Medical Sciences, Frank H. Netter School of Medicine at Quinnipiac University, North Haven, CT, USA
| | - Zeal Jinwala
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
| | - Yousef Khan
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ariadni Oikonomou
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Damaris Silva-Lopez
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Isabel Burton
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Morgan Dixon
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jackson Milone
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah Ramirez
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Naomi Shifman
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel Levey
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- VA CT Healthcare Center, West Haven, CT, USA
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Departments of Genetics and Neurobiology, Yale University School of Medicine, New Haven, CT, USA
- VA CT Healthcare Center, West Haven, CT, USA
| | - Emily E Hartwell
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
| | - Rachel L Kember
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
| |
Collapse
|
9
|
Monson ET, Colbert SMC, Andreassen OA, Ayinde OO, Bejan CA, Ceja Z, Coon H, DiBlasi E, Izotova A, Kaufman EA, Koromina M, Myung W, Nurnberger JI, Serretti A, Smoller JW, Stein MB, Zai CC, Aslan M, Barr PB, Bigdeli TB, Harvey PD, Kimbrel NA, Patel PR, Ruderfer D, Docherty AR, Mullins N, Mann JJ. Defining Suicidal Thought and Behavior Phenotypes for Genetic Studies. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.27.24311110. [PMID: 39132474 PMCID: PMC11312669 DOI: 10.1101/2024.07.27.24311110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Background Standardized definitions of suicidality phenotypes, including suicidal ideation (SI), attempt (SA), and death (SD) are a critical step towards improving understanding and comparison of results in suicide research. The complexity of suicidality contributes to heterogeneity in phenotype definitions, impeding evaluation of clinical and genetic risk factors across studies and efforts to combine samples within consortia. Here, we present expert and data-supported recommendations for defining suicidality and control phenotypes to facilitate merging current/legacy samples with definition variability and aid future sample creation. Methods A subgroup of clinician researchers and experts from the Suicide Workgroup of the Psychiatric Genomics Consortium (PGC) reviewed existing PGC definitions for SI, SA, SD, and control groups and generated preliminary consensus guidelines for instrument-derived and international classification of disease (ICD) data. ICD lists were validated in two independent datasets (N = 9,151 and 12,394). Results Recommendations are provided for evaluated instruments for SA and SI, emphasizing selection of lifetime measures phenotype-specific wording. Recommendations are also provided for defining SI and SD from ICD data. As the SA ICD definition is complex, SA code list recommendations were validated against instrument results with sensitivity (range = 15.4% to 80.6%), specificity (range = 67.6% to 97.4%), and positive predictive values (range = 0.59-0.93) reported. Conclusions Best-practice guidelines are presented for the use of existing information to define SI/SA/SD in consortia research. These proposed definitions are expected to facilitate more homogeneous data aggregation for genetic and multisite studies. Future research should involve refinement, improved generalizability, and validation in diverse populations.
Collapse
Affiliation(s)
- Eric T. Monson
- Department of Psychiatry, University of Utah Spencer Fox Eccles School of Medicine
- Huntsman Mental Health Institute
| | - Sarah M. C. Colbert
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai
| | - Ole A. Andreassen
- Division of Mental Health and Addiction, Oslo University Hospital
- NORMENT Centre, University of Oslo
| | | | - Cosmin A. Bejan
- Department of Biomedical Informatics, Vanderbilt University Medical Center
| | - Zuriel Ceja
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland
| | - Hilary Coon
- Department of Psychiatry, University of Utah Spencer Fox Eccles School of Medicine
- Huntsman Mental Health Institute
| | - Emily DiBlasi
- Department of Psychiatry, University of Utah Spencer Fox Eccles School of Medicine
- Huntsman Mental Health Institute
| | - Anastasia Izotova
- Nic Waals Institute, Lovisenberg Diaconal Hospital
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health
- Department of Psychology, University of Oslo
| | - Erin A. Kaufman
- Department of Psychiatry, University of Utah Spencer Fox Eccles School of Medicine
- Huntsman Mental Health Institute
| | - Maria Koromina
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai
| | - Woojae Myung
- Department of Neuropsychiatry, Seoul National University Bundang Hospital
- Department of Psychiatry, Seoul National University College of Medicine
| | - John I. Nurnberger
- Department of Psychiatry, Indiana University School of Medicine
- Department of Medical & Molecular Genetics, Indiana University
| | | | - Jordan W. Smoller
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital
- Stanley Center for Psychiatric Research, Broad Institute
- Psychiatric and Neurodevelopmental Genetics Unit (PNGU), Massachusetts General Hospital
| | - Murray B. Stein
- Department of Psychiatry and School of Public Health, University of California San Diego
| | - Clement C. Zai
- Stanley Center for Psychiatric Research, Broad Institute
- Department of Psychiatry, University of Toronto
- Institute of Medical Science, University of Toronto
- Molecular Brain Science, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health
- Laboratory Medicine and Pathobiology, University of Toronto
| | | | - Mihaela Aslan
- Cooperative Studies Program Clinical Epidemiology Research Center (CSP-CERC), VA Connecticut Healthcare System
- Department of Internal Medicine, Yale University School of Medicine
| | - Peter B. Barr
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University
- VA New York Harbor Healthcare System
- Institute for Genomics in Health, SUNY Downstate Health Sciences University
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University
| | - Tim B. Bigdeli
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University
- VA New York Harbor Healthcare System
- Institute for Genomics in Health, SUNY Downstate Health Sciences University
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University
| | - Philip D. Harvey
- Bruce W. Carter Miami Veterans Affairs (VA) Medical Center
- University of Miami School of Medicine
| | - Nathan A. Kimbrel
- Durham VA Health Care System
- VA Health Services Research and Development Center of Innovation to Accelerate Discovery and Practice Transformation
- VISN 6 Mid-Atlantic Mental Illness Research, Education, and Clinical Center
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine
| | - Pujan R. Patel
- Durham VA Health Care System
- VA Health Services Research and Development Center of Innovation to Accelerate Discovery and Practice Transformation
| | | | - Douglas Ruderfer
- Department of Biomedical Informatics, Vanderbilt University Medical Center
- Vanderbilt Genetics Institute, Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center
| | - Anna R. Docherty
- Department of Psychiatry, University of Utah Spencer Fox Eccles School of Medicine
- Huntsman Mental Health Institute
- Clinical and Translational Science Institute & the Center for Genomic Medicine, University of Utah
| | - Niamh Mullins
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai
| | - J. John Mann
- Departments of Psychiatry and Radiology, Columbia University
| |
Collapse
|
10
|
Vellucci L, De Simone G, Morley-Fletcher S, Buonaguro EF, Avagliano C, Barone A, Maccari S, Iasevoli F, de Bartolomeis A. Perinatal stress modulates glutamatergic functional connectivity: A post-synaptic density immediate early gene-based network analysis. Prog Neuropsychopharmacol Biol Psychiatry 2024; 133:111032. [PMID: 38762163 DOI: 10.1016/j.pnpbp.2024.111032] [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: 01/10/2024] [Revised: 04/29/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
Early life stress may induce synaptic changes within brain regions associated with behavioral disorders. Here, we investigated glutamatergic functional connectivity by a postsynaptic density immediate-early gene-based network analysis. Pregnant female Sprague-Dawley rats were randomly divided into two experimental groups: one exposed to stress sessions and the other serving as a stress-free control group. Homer1 expression was evaluated by in situ hybridization technique in eighty-eight brain regions of interest of male rat offspring. Differences between the perinatal stress exposed group (PRS) (n = 5) and the control group (CTR) (n = 5) were assessed by performing the Student's t-test via SPSS 28.0.1.0 with Bonferroni correction. Additionally, all possible pairwise Spearman's correlations were computed as well as correlation matrices and networks for each experimental group were generated via RStudio and Cytoscape. Perinatal stress exposure was associated with Homer1a reduction in several cortical, thalamic, and striatal regions. Furthermore, it was found to affect functional connectivity between: the lateral septal nucleus, the central medial thalamic nucleus, the anterior part of the paraventricular thalamic nucleus, and both retrosplenial granular b cortex and hippocampal regions; the orbitofrontal cortex, amygdaloid nuclei, and hippocampal regions; and lastly, among regions involved in limbic system. Finally, the PRS networks showed a significant reduction in multiple connections for the ventrolateral part of the anteroventral thalamic nucleus after perinatal stress exposure, as well as a decrease in the centrality of ventral anterior thalamic and amygdaloid nuclei suggestive of putative reduced cortical control over these regions. Within the present preclinical setting, perinatal stress exposure is a modifier of glutamatergic early gene-based functional connectivity in neuronal circuits involved in behaviors relevant to model neurodevelopmental disorders.
Collapse
Affiliation(s)
- Licia Vellucci
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", Naples, Italy; Department of Translational Medical Sciences, University of Naples "Federico II", Via S. Pansini 5, 80131 Naples, Italy
| | - Giuseppe De Simone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", Naples, Italy
| | - Sara Morley-Fletcher
- Unité de Glycobiologie Structurale et Fonctionnelle, University of Lille, CNRS, UMR 8576, UGSF, F-59000 Lille, France; International Associated Laboratory (LIA) "Perinatal Stress and Neurodegenerative Diseases", Sapienza University of Rome - IRCCS, Neuromed, Rome, Italy and University of Lille - CNRS, UMR 8576, Lille, France
| | - Elisabetta Filomena Buonaguro
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", Naples, Italy
| | - Camilla Avagliano
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", Naples, Italy
| | - Annarita Barone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", Naples, Italy
| | - Stefania Maccari
- Unité de Glycobiologie Structurale et Fonctionnelle, University of Lille, CNRS, UMR 8576, UGSF, F-59000 Lille, France; International Associated Laboratory (LIA) "Perinatal Stress and Neurodegenerative Diseases", Sapienza University of Rome - IRCCS, Neuromed, Rome, Italy and University of Lille - CNRS, UMR 8576, Lille, France; Department of Science and Medical-Surgical Biotechnology, Sapienza University of Rome, Rome, Italy
| | - Felice Iasevoli
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", Naples, Italy
| | - Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", Naples, Italy.
| |
Collapse
|
11
|
Arena G, Landoulsi Z, Grossmann D, Payne T, Vitali A, Delcambre S, Baron A, Antony P, Boussaad I, Bobbili DR, Sreelatha AAK, Pavelka L, J Diederich N, Klein C, Seibler P, Glaab E, Foltynie T, Bandmann O, Sharma M, Krüger R, May P, Grünewald A. Polygenic Risk Scores Validated in Patient-Derived Cells Stratify for Mitochondrial Subtypes of Parkinson's Disease. Ann Neurol 2024; 96:133-149. [PMID: 38767023 DOI: 10.1002/ana.26949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 04/25/2024] [Accepted: 04/28/2024] [Indexed: 05/22/2024]
Abstract
OBJECTIVE The aim of our study is to better understand the genetic architecture and pathological mechanisms underlying neurodegeneration in idiopathic Parkinson's disease (iPD). We hypothesized that a fraction of iPD patients may harbor a combination of common variants in nuclear-encoded mitochondrial genes ultimately resulting in neurodegeneration. METHODS We used mitochondria-specific polygenic risk scores (mitoPRSs) and created pathway-specific mitoPRSs using genotype data from different iPD case-control datasets worldwide, including the Luxembourg Parkinson's Study (412 iPD patients and 576 healthy controls) and COURAGE-PD cohorts (7,270 iPD cases and 6,819 healthy controls). Cellular models from individuals stratified according to the most significant mitoPRS were subsequently used to characterize different aspects of mitochondrial function. RESULTS Common variants in genes regulating Oxidative Phosphorylation (OXPHOS-PRS) were significantly associated with a higher PD risk in independent cohorts (Luxembourg Parkinson's Study odds ratio, OR = 1.31[1.14-1.50], p-value = 5.4e-04; COURAGE-PD OR = 1.23[1.18-1.27], p-value = 1.5e-29). Functional analyses in fibroblasts and induced pluripotent stem cells-derived neuronal progenitors revealed significant differences in mitochondrial respiration between iPD patients with high or low OXPHOS-PRS (p-values < 0.05). Clinically, iPD patients with high OXPHOS-PRS have a significantly earlier age at disease onset compared to low-risk patients (false discovery rate [FDR]-adj p-value = 0.015), similar to prototypic monogenic forms of PD. Finally, iPD patients with high OXPHOS-PRS responded more effectively to treatment with mitochondrially active ursodeoxycholic acid. INTERPRETATION OXPHOS-PRS may provide a precision medicine tool to stratify iPD patients into a pathogenic subgroup genetically defined by specific mitochondrial impairment, making these individuals eligible for future intelligent clinical trial designs. ANN NEUROL 2024;96:133-149.
Collapse
Affiliation(s)
- Giuseppe Arena
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Zied Landoulsi
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Dajana Grossmann
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Translational Neurodegeneration Section "Albrecht-Kossel", Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Thomas Payne
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Armelle Vitali
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Sylvie Delcambre
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Alexandre Baron
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Paul Antony
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Ibrahim Boussaad
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Dheeraj Reddy Bobbili
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Ashwin Ashok Kumar Sreelatha
- Centre for Genetic Epidemiology, Institute for Clinical Epidemiology and Applied Biometry, University of Tübingen, Tübingen, Germany
| | - Lukas Pavelka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, Strassen, Luxembourg
- Parkinson Research Clinic, Centre Hospitalier du Luxembourg, Luxembourg, Luxembourg
| | - Nico J Diederich
- Department of Neurosciences, Centre Hospitalier de Luxembourg, Strassen, Luxembourg
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Philip Seibler
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Thomas Foltynie
- Department of Clinical and Movement Neurosciences, Institute of Neurology, University College London, London, UK
| | - Oliver Bandmann
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Manu Sharma
- Centre for Genetic Epidemiology, Institute for Clinical Epidemiology and Applied Biometry, University of Tübingen, Tübingen, Germany
| | - Rejko Krüger
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, Strassen, Luxembourg
- Parkinson Research Clinic, Centre Hospitalier du Luxembourg, Luxembourg, Luxembourg
| | - Patrick May
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Anne Grünewald
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
12
|
Pisanu C, Congiu D, Meloni A, Paribello P, Patrinos GP, Severino G, Ardau R, Chillotti C, Manchia M, Squassina A. Dissecting the genetic overlap between severe mental disorders and markers of cellular aging: Identification of pleiotropic genes and druggable targets. Neuropsychopharmacology 2024; 49:1033-1041. [PMID: 38402365 PMCID: PMC11039620 DOI: 10.1038/s41386-024-01822-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/17/2024] [Accepted: 02/04/2024] [Indexed: 02/26/2024]
Abstract
Patients with severe mental disorders such as bipolar disorder (BD), schizophrenia (SCZ) and major depressive disorder (MDD) show a substantial reduction in life expectancy, increased incidence of comorbid medical conditions commonly observed with advanced age and alterations of aging hallmarks. While severe mental disorders are heritable, the extent to which genetic predisposition might contribute to accelerated cellular aging is not known. We used bivariate causal mixture models to quantify the trait-specific and shared architecture of mental disorders and 2 aging hallmarks (leukocyte telomere length [LTL] and mitochondrial DNA copy number), and the conjunctional false discovery rate method to detect shared genetic loci. We integrated gene expression data from brain regions from GTEx and used different tools to functionally annotate identified loci and investigate their druggability. Aging hallmarks showed low polygenicity compared with severe mental disorders. We observed a significant negative global genetic correlation between MDD and LTL (rg = -0.14, p = 6.5E-10), and no significant results for other severe mental disorders or for mtDNA-cn. However, conditional QQ plots and bivariate causal mixture models pointed to significant pleiotropy among all severe mental disorders and aging hallmarks. We identified genetic variants significantly shared between LTL and BD (n = 17), SCZ (n = 55) or MDD (n = 19), or mtDNA-cn and BD (n = 4), SCZ (n = 12) or MDD (n = 1), with mixed direction of effects. The exonic rs7909129 variant in the SORCS3 gene, encoding a member of the retromer complex involved in protein trafficking and intracellular/intercellular signaling, was associated with shorter LTL and increased predisposition to all severe mental disorders. Genetic variants underlying risk of SCZ or MDD and shorter LTL modulate expression of several druggable genes in different brain regions. Genistein, a phytoestrogen with anti-inflammatory and antioxidant effects, was an upstream regulator of 2 genes modulated by variants associated with risk of MDD and shorter LTL. While our results suggest that shared heritability might play a limited role in contributing to accelerated cellular aging in severe mental disorders, we identified shared genetic determinants and prioritized different druggable targets and compounds.
Collapse
Affiliation(s)
- Claudia Pisanu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy.
| | - Donatella Congiu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Anna Meloni
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Pasquale Paribello
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy
| | - George P Patrinos
- Laboratory of Pharmacogenomics and Individualized Therapy, School of Health Sciences, Department of Pharmacy, University of Patras, Patras, Greece
- College of Medicine and Health Sciences, Department of Genetics and Genomics, United Arab Emirates University, Al‑Ain, Abu Dhabi, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, Al‑Ain, Abu Dhabi, UAE
| | - Giovanni Severino
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Raffaella Ardau
- Unit of Clinical Pharmacology, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Caterina Chillotti
- Unit of Clinical Pharmacology, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Alessio Squassina
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
13
|
Barr P, Neale Z, Chatzinakos C, Schulman J, Mullins N, Zhang J, Chorlian D, Kamarajan C, Kinreich S, Pandey A, Pandey G, de Viteri SS, Acion L, Bauer L, Bucholz K, Chan G, Dick D, Edenberg H, Foroud T, Goate A, Hesselbrock V, Johnson E, Kramer J, Lai D, Plawecki M, Salvatore J, Wetherill L, Agrawal A, Porjesz B, Meyers J. Clinical, genomic, and neurophysiological correlates of lifetime suicide attempts among individuals with alcohol dependence. RESEARCH SQUARE 2024:rs.3.rs-3894892. [PMID: 38405959 PMCID: PMC10889049 DOI: 10.21203/rs.3.rs-3894892/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Research has identified clinical, genomic, and neurophysiological markers associated with suicide attempts (SA) among individuals with psychiatric illness. However, there is limited research among those with an alcohol use disorder (AUD), despite their disproportionately higher rates of SA. We examined lifetime SA in 4,068 individuals with DSM-IV alcohol dependence from the Collaborative Study on the Genetics of Alcoholism (23% lifetime suicide attempt; 53% female; 17% Admixed African American ancestries; mean age: 38). We 1) conducted a genome-wide association study (GWAS) of SA and performed downstream analyses to determine whether we could identify specific biological pathways of risk, and 2) explored risk in aggregate across other clinical conditions, polygenic scores (PGS) for comorbid psychiatric problems, and neurocognitive functioning between those with AD who have and have not reported a lifetime suicide attempt. The GWAS and downstream analyses did not produce any significant associations. Participants with an AUD who had attempted suicide had greater rates of trauma exposure, major depressive disorder, post-traumatic stress disorder, and other substance use disorders compared to those who had not attempted suicide. Polygenic scores for suicide attempt, depression, and PTSD were associated with reporting a suicide attempt (ORs = 1.22-1.44). Participants who reported a SA also had decreased right hemispheric frontal-parietal theta and decreased interhemispheric temporal-parietal alpha electroencephalogram resting-state coherences relative to those who did not, but differences were small. Overall, individuals with alcohol dependence who report SA appear to experience a variety of severe comorbidities and elevated polygenic risk for SA. Our results demonstrate the need to further investigate suicide attempts in the presence of substance use disorders.
Collapse
Affiliation(s)
- Peter Barr
- SUNY Downstate Health Sciences University
| | - Zoe Neale
- SUNY Downstate Health Sciences University
| | | | | | | | | | | | | | | | - Ashwini Pandey
- State University of New York Downstate Health Sciences University
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jacquelyn Meyers
- State University of New York (SUNY) Downstate Health Sciences University
| |
Collapse
|
14
|
Hartwell EE, Jinwala Z, Milone J, Ramirez S, Gelernter J, Kranzler HR, Kember RL. Application of polygenic scores to a deeply phenotyped sample enriched for substance use disorders reveals extensive pleiotropy with psychiatric and medical traits. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.22.24301615. [PMID: 38343859 PMCID: PMC10854354 DOI: 10.1101/2024.01.22.24301615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Co-occurring psychiatric, medical, and substance use disorders (SUDs) are common, but the complex pathways leading to such comorbidities are poorly understood. A greater understanding of genetic influences on this phenomenon could inform precision medicine efforts. We used the Yale-Penn dataset, a cross-sectional sample enriched for individuals with SUDs, to examine pleiotropic effects of genetic liability for psychiatric and medical traits. Participants completed an in-depth interview that provides information on demographics, environment, medical illnesses, and psychiatric and SUDs. Polygenic scores (PGS) for psychiatric disorders and medical traits were calculated in European-ancestry (EUR; n=5,691) participants and, when discovery datasets were available, for African-ancestry (AFR; n=4,918) participants. Phenome-wide association studies (PheWAS) were then conducted. In AFR participants, the only PGS with significant associations was bipolar disorder (BD), all of which were with substance use phenotypes. In EUR participants, PGS for major depressive disorder (MDD), generalized anxiety disorder (GAD), post-traumatic stress disorder (PTSD), schizophrenia (SCZ), body mass index (BMI), coronary artery disease (CAD), and type 2 diabetes (T2D) all showed significant associations, the majority of which were with phenotypes in the substance use categories. For instance, PGS MDD was associated with over 200 phenotypes, 15 of which were depression-related (e.g., depression criterion count), 55 of which were other psychiatric phenotypes, and 126 of which were substance use phenotypes; and PGS BMI was associated with 138 phenotypes, 105 of which were substance related. Genetic liability for psychiatric and medical traits is associated with numerous phenotypes across multiple categories, indicative of the broad genetic liability of these traits.
Collapse
|
15
|
Lin F, Jun Li, Ziqi Wang, Zhang T, Lu T, Jiang M, Yang K, Jia M, Zhang D, Wang L. Replication of previous autism-GWAS hits suggests the association between NAA1, SORCS3, and GSDME and autism in the Han Chinese population. Heliyon 2024; 10:e23677. [PMID: 38234914 PMCID: PMC10792458 DOI: 10.1016/j.heliyon.2023.e23677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/25/2023] [Accepted: 12/09/2023] [Indexed: 01/19/2024] Open
Abstract
Background Autism is a severe neurodevelopmental disorder characterized by social interaction deficits, impairments in communication, and restricted and repetitive stereotyped behavior and activities. Family and twin studies suggested an essential role of genetic factors in the etiology of autism spectrum disorder (ASD). Also, other studies found SORCS3 and GSDME (DFNA5) might be involved in brain development and susceptible to ASD. Methods In this study, 17 genome-wide significant SNPs reported in previous ASD genome-wide association studies (GWAS) and 7 SNPs in strong linkage disequilibrium with known ASD GWAS hits were selected to investigate the association between these SNPs and autism in the Han Chinese population. Then, 10 tagSNPs in SORCS3 and 11 tagSNPs in GSDME were selected to analyze the association between these SNPs and autism. The selected 24 SNPs and tagSNPs were genotyped using the Agena MassARRAY SNP genotyping assay in 757 Han Chinese autism trios. Results Rs1484144 in NAA11 was significantly associated with autism; significance remained after the Bonferroni correction (P < 0.0022). Also, rs79879286, rs12154597, and rs12540919 near GSDME, as well as rs9787523 and rs3750261 in SORCS3, were nominally associated with autism. Conclusion Our study suggests that rs1484144 in NAA11 is a significant SNP for autism in the Han Chinese population, while SORCS3 and GSDME might be the susceptibility genes for autism in this population.
Collapse
Affiliation(s)
- Fen Lin
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
| | - Jun Li
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
| | - Ziqi Wang
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
| | - Tian Zhang
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
| | - Tianlan Lu
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
| | - Miaomiao Jiang
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
| | - Kang Yang
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
| | - Meixiang Jia
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
| | - Dai Zhang
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- Guangdong Key Laboratory of Mental Health and Cognitive Science, Institute for Brain Research and Rehabilitation (IBRR), South China Normal University, Guangzhou, China
| | - Lifang Wang
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
| |
Collapse
|
16
|
Levine Z, Kalka I, Kolobkov D, Rossman H, Godneva A, Shilo S, Keshet A, Weissglas-Volkov D, Shor T, Diament A, Talmor-Barkan Y, Aviv Y, Sharon T, Weinberger A, Segal E. Genome-wide association studies and polygenic risk score phenome-wide association studies across complex phenotypes in the human phenotype project. MED 2024; 5:90-101.e4. [PMID: 38157848 DOI: 10.1016/j.medj.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 09/29/2023] [Accepted: 12/03/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Genome-wide association studies (GWASs) associate phenotypes and genetic variants across a study cohort. GWASs require large-scale cohorts with both phenotype and genetic sequencing data, limiting studied phenotypes. The Human Phenotype Project is a longitudinal study that has measured a wide range of clinical and biomolecular features from a self-assignment cohort over 5 years. The phenotypes collected are quantitative traits, providing higher-resolution insights into the genetics of complex phenotypes. METHODS We present the results of GWASs and polygenic risk score phenome-wide association studies with 729 clinical phenotypes and 4,043 molecular features from the Human Phenotype Project. This includes clinical traits that have not been previously associated with genetics, including measures from continuous sleep monitoring, continuous glucose monitoring, liver ultrasound, hormonal status, and fundus imaging. FINDINGS In GWAS of 8,706 individuals, we found significant associations between 169 clinical traits and 1,184 single-nucleotide polymorphisms. We found genes associated with both glycemic control and mental disorders, and we quantify the strength of genetic signals in serum metabolites. In polygenic risk score phenome-wide association studies for clinical traits, we found 16,047 significant associations. CONCLUSIONS The entire set of findings, which we disseminate publicly, provides newfound resolution into the genetic architecture of complex human phenotypes. FUNDING E.S. is supported by the Minerva foundation with funding from the Federal German Ministry for Education and Research and by the European Research Council and the Israel Science Foundation.
Collapse
Affiliation(s)
- Zachary Levine
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Iris Kalka
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Dmitry Kolobkov
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Hagai Rossman
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel; Pheno.AI, Tel-Aviv, Israel
| | - Anastasia Godneva
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Smadar Shilo
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ayya Keshet
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel; Pheno.AI, Tel-Aviv, Israel
| | - Daphna Weissglas-Volkov
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel; Pheno.AI, Tel-Aviv, Israel
| | - Tal Shor
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel; Pheno.AI, Tel-Aviv, Israel
| | - Alon Diament
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel; Pheno.AI, Tel-Aviv, Israel
| | - Yeela Talmor-Barkan
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel; Department of Cardiology, Rabin Medical Center, Petah-Tikva 49100, Israel
| | - Yaron Aviv
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel; Department of Cardiology, Rabin Medical Center, Petah-Tikva 49100, Israel
| | - Tom Sharon
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Adina Weinberger
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel; Pheno.AI, Tel-Aviv, Israel
| | - Eran Segal
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
17
|
Stauffer EM, Bethlehem RAI, Dorfschmidt L, Won H, Warrier V, Bullmore ET. The genetic relationships between brain structure and schizophrenia. Nat Commun 2023; 14:7820. [PMID: 38016951 PMCID: PMC10684873 DOI: 10.1038/s41467-023-43567-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 11/14/2023] [Indexed: 11/30/2023] Open
Abstract
Genetic risks for schizophrenia are theoretically mediated by genetic effects on brain structure but it has been unclear which genes are associated with both schizophrenia and cortical phenotypes. We accessed genome-wide association studies (GWAS) of schizophrenia (N = 69,369 cases; 236,642 controls), and of three magnetic resonance imaging (MRI) metrics (surface area, cortical thickness, neurite density index) measured at 180 cortical areas (N = 36,843, UK Biobank). Using Hi-C-coupled MAGMA, 61 genes were significantly associated with both schizophrenia and one or more MRI metrics. Whole genome analysis with partial least squares demonstrated significant genetic covariation between schizophrenia and area or thickness of most cortical regions. Genetic similarity between cortical areas was strongly coupled to their phenotypic covariance, and genetic covariation between schizophrenia and brain phenotypes was strongest in the hubs of structural covariance networks. Pleiotropically associated genes were enriched for neurodevelopmental processes and positionally concentrated in chromosomes 3p21, 17q21 and 11p11. Mendelian randomization analysis indicated that genetically determined variation in a posterior cingulate cortical area could be causal for schizophrenia. Parallel analyses of GWAS on bipolar disorder, Alzheimer's disease and height showed that pleiotropic association with MRI metrics was stronger for schizophrenia compared to other disorders.
Collapse
Affiliation(s)
| | - Richard A I Bethlehem
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Lena Dorfschmidt
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Hyejung Won
- Department of Genetics and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Varun Warrier
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Edward T Bullmore
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire & Peterborough NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
18
|
Kranzler H, Davis C, Feinn R, Jinwala Z, Khan Y, Oikonomou A, Silva-Lopez D, Burton I, Dixon M, Milone J, Ramirez S, Shifman N, Levey D, Gelernter J, Hartwell E, Kember R. Adverse Childhood Events, Mood and Anxiety Disorders, and Substance Dependence: Gene x Environment Effects and Moderated Mediation. RESEARCH SQUARE 2023:rs.3.rs-3483320. [PMID: 37961429 PMCID: PMC10635374 DOI: 10.21203/rs.3.rs-3483320/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Adverse childhood events (ACEs) contribute to the development of mood and anxiety disorders and substance dependence. However, the extent to which these effects are direct or indirect and whether genetic risk moderates them is unclear. Methods We examined associations among ACEs, mood/anxiety disorders, and substance dependence in 12,668 individuals (44.9% female, 42.5% African American/Black, 42.1% European American/White). We generated latent variables for each phenotype and modeled direct and indirect effects of ACEs on substance dependence, mediated by mood/anxiety disorders (forward or "self-medication" model) and of ACEs on mood/anxiety disorders, mediated by substance dependence (reverse or "substance-induced" model). In a sub-sample, we also generated polygenic scores for substance dependence and mood/anxiety disorder factors, which we tested as moderators in the mediation models. Results Although there were significant indirect effects in both directions, mediation by mood/anxiety disorders (forward model) was greater than by substance dependence (reverse model). Greater genetic risk for substance dependence was associated with a weaker direct effect of ACEs on substance dependence in both the African- and European-ancestry groups (i.e., gene-environment interaction) and a weaker indirect effect in European-ancestry individuals (i.e., moderated mediation). Conclusion We found greater evidence that substance dependence results from self-medication of mood/anxiety disorders than that mood/anxiety disorders are substance induced. Among individuals at higher genetic risk for substance dependence who are more likely to develop a dependence diagnosis, ACEs exert less of an effect in promoting that outcome. Following exposure to ACEs, multiple pathways lead to mood/anxiety disorders and substance dependence. Specification of these pathways could inform individually targeted prevention and treatment approaches.
Collapse
Affiliation(s)
| | | | | | - Zeal Jinwala
- University of Pennsylvania Perelman School of Medicine
| | - Yousef Khan
- University of Pennsylvania Perelman School of Medicine
| | | | | | - Isabel Burton
- University of Pennsylvania Perelman School of Medicine
| | - Morgan Dixon
- University of Pennsylvania Perelman School of Medicine
| | | | - Sarah Ramirez
- University of Pennsylvania Perelman School of Medicine
| | | | | | | | | | | |
Collapse
|
19
|
Kranzler HR, Davis CN, Feinn R, Jinwala Z, Khan Y, Oikonomou A, Silva-Lopez D, Burton I, Dixon M, Milone J, Ramirez S, Shifman N, Levey D, Gelernter J, Hartwell EE, Kember RL. Adverse Childhood Events, Mood and Anxiety Disorders, and Substance Dependence: Gene X Environment Effects and Moderated Mediation. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.10.24.23297419. [PMID: 37961309 PMCID: PMC10635185 DOI: 10.1101/2023.10.24.23297419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Adverse childhood events (ACEs) contribute to the development of mood and anxiety disorders and substance dependence. However, the extent to which these effects are direct or indirect and whether genetic risk moderates them is unclear. Methods We examined associations among ACEs, mood/anxiety disorders, and substance dependence in 12,668 individuals (44.9% female, 42.5% African American/Black, 42.1% European American/White). We generated latent variables for each phenotype and modeled direct and indirect effects of ACEs on substance dependence, mediated by mood/anxiety disorders (forward or "self-medication" model) and of ACEs on mood/anxiety disorders, mediated by substance dependence (reverse or "substance-induced" model). In a sub-sample, we also generated polygenic scores for substance dependence and mood/anxiety disorder factors, which we tested as moderators in the mediation models. Results Although there were significant indirect effects in both directions, mediation by mood/anxiety disorders (forward model) was greater than by substance dependence (reverse model). Greater genetic risk for substance dependence was associated with a weaker direct effect of ACEs on substance dependence in both the African- and European-ancestry groups (i.e., gene-environment interaction) and a weaker indirect effect in European-ancestry individuals (i.e., moderated mediation). Conclusion We found greater evidence that substance dependence results from self-medication of mood/anxiety disorders than that mood/anxiety disorders are substance induced. Among individuals at higher genetic risk for substance dependence who are more likely to develop a dependence diagnosis, ACEs exert less of an effect in promoting that outcome. Following exposure to ACEs, multiple pathways lead to mood/anxiety disorders and substance dependence. Specification of these pathways could inform individually targeted prevention and treatment approaches.
Collapse
Affiliation(s)
- Henry R. Kranzler
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104
| | - Christal N. Davis
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104
| | - Richard Feinn
- Department of Medical Sciences, Frank H. Netter School of Medicine at Quinnipiac University, North Haven, CT 06473
| | - Zeal Jinwala
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104
| | - Yousef Khan
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Ariadni Oikonomou
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Damaris Silva-Lopez
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Isabel Burton
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Morgan Dixon
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Jackson Milone
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Sarah Ramirez
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Naomi Shifman
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Daniel Levey
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT and VA CT Healthcare Center, 950 Campbell Avenue, West Haven, CT 06516, USA
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT and VA CT Healthcare Center, 950 Campbell Avenue, West Haven, CT 06516, USA
- Departments of Genetics and Neurobiology, Yale University School of Medicine, New Haven, CT and VA CT Healthcare Center, 950 Campbell Avenue, West Haven, CT 06516, USA
| | - Emily E. Hartwell
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104
| | - Rachel L. Kember
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104
| |
Collapse
|
20
|
Cuellar-Barboza AB, Prieto ML, Coombes BJ, Gardea-Resendez M, Núñez N, Winham SJ, Romo-Nava F, González S, McElroy SL, Frye MA, Biernacka JM. Polygenic prediction of bipolar disorder in a Latin American sample. Am J Med Genet B Neuropsychiatr Genet 2023; 192:139-146. [PMID: 36919637 DOI: 10.1002/ajmg.b.32936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 01/31/2023] [Accepted: 03/01/2023] [Indexed: 03/16/2023]
Abstract
To date, bipolar disorder (BD) genetic studies and polygenic risk scores (PRSs) for BD are based primarily on populations of European descent (EUR) and lack representation from other ancestries including Latin American (LAT). Here, we describe a new LAT cohort from the Mayo Clinic Bipolar Biobank (MCBB), a multisite collaboration with recruitment sites in the United States (EUR; 1,443 cases and 777 controls) and Mexico and Chile (LAT; 211 cases and 161 controls) and use the sample to explore the performance of a BD-PRS in a LAT population. Using results from the largest genome-wide association study of BD in EUR individuals, PRSice2 and LDpred2 were used to compute BD-PRSs in the LAT and EUR samples from the MCBB. PRSs explained up to 1.4% (PRSice) and 4% (LDpred2) of the phenotypic variance on the liability scale in the LAT sample compared to 3.8% (PRSice2) and 3.4% (LDpred2) in the EUR samples. Future larger studies should further explore the differential performance of different PRS approaches across ancestries. International multisite studies, such as this one, have the potential to address diversity-related limitations of prior genomic studies and ultimately contribute to the reduction of health disparities.
Collapse
Affiliation(s)
- Alfredo B Cuellar-Barboza
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Psychiatry, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | - Miguel L Prieto
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Psychiatry, Universidad de los Andes, Santiago, Chile
- Mental Health Service, Clinica Universidad de los Andes, Santiago, Chile
| | - Brandon J Coombes
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Nicolás Núñez
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | - Stacey J Winham
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Sarai González
- Department of Psychiatry, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | - Susan L McElroy
- Lindner Center of HOPE/University of Cincinnati, Cincinnati, Ohio, USA
| | - Mark A Frye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | - Joanna M Biernacka
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| |
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: 13] [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
|
Murlanova K, Pletnikov MV. Modeling psychotic disorders: Environment x environment interaction. Neurosci Biobehav Rev 2023; 152:105310. [PMID: 37437753 DOI: 10.1016/j.neubiorev.2023.105310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 06/26/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023]
Abstract
Schizophrenia is a major psychotic disorder with multifactorial etiology that includes interactions between genetic vulnerability and environmental risk factors. In addition, interplay of multiple environmental adversities affects neurodevelopment and may increase the individual risk of developing schizophrenia. Consistent with the two-hit hypothesis of schizophrenia, we review rodent models that combine maternal immune activation as the first hit with other adverse environmental exposures as the second hit. We discuss the strengths and pitfalls of the current animal models of environment x environment interplay and propose some future directions to advance the field.
Collapse
Affiliation(s)
- Kateryna Murlanova
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Mikhail V Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
23
|
D'Sa K, Guelfi S, Vandrovcova J, Reynolds RH, Zhang D, Hardy J, Botía JA, Weale ME, Taliun SAG, Small KS, Ryten M. Analysis of subcellular RNA fractions demonstrates significant genetic regulation of gene expression in human brain post-transcriptionally. Sci Rep 2023; 13:13874. [PMID: 37620324 PMCID: PMC10449874 DOI: 10.1038/s41598-023-40324-0] [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: 10/07/2022] [Accepted: 08/08/2023] [Indexed: 08/26/2023] Open
Abstract
Gaining insight into the genetic regulation of gene expression in human brain is key to the interpretation of genome-wide association studies for major neurological and neuropsychiatric diseases. Expression quantitative trait loci (eQTL) analyses have largely been used to achieve this, providing valuable insights into the genetic regulation of steady-state RNA in human brain, but not distinguishing between molecular processes regulating transcription and stability. RNA quantification within cellular fractions can disentangle these processes in cell types and tissues which are challenging to model in vitro. We investigated the underlying molecular processes driving the genetic regulation of gene expression specific to a cellular fraction using allele-specific expression (ASE). Applying ASE analysis to genomic and transcriptomic data from paired nuclear and cytoplasmic fractions of anterior prefrontal cortex, cerebellar cortex and putamen tissues from 4 post-mortem neuropathologically-confirmed control human brains, we demonstrate that a significant proportion of genetic regulation of gene expression occurs post-transcriptionally in the cytoplasm, with genes undergoing this form of regulation more likely to be synaptic. These findings have implications for understanding the structure of gene expression regulation in human brain, and importantly the interpretation of rapidly growing single-nucleus brain RNA-sequencing and eQTL datasets, where cytoplasm-specific regulatory events could be missed.
Collapse
Affiliation(s)
- Karishma D'Sa
- Department of Neurodegenerative Disease, University College London, London, WC1N 3BG, UK
- Department of Medical & Molecular Genetics, School of Medical Sciences, King's College London, Guy's Hospital, London, SE1 1UL, UK
- Department of Clinical and Movement Neurosciences, University College London, London, WC1N 3BG, UK
| | - Sebastian Guelfi
- Department of Neurodegenerative Disease, University College London, London, WC1N 3BG, UK
- Verge Genomics, Tower Pl, South San Francisco, CA, 94080, USA
| | - Jana Vandrovcova
- Dept of Neuromuscular Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Regina H Reynolds
- Great Ormond Street Institute of Child Health, Genetics and Genomic Medicine, University College London, London, WC1N 1EH, UK
| | - David Zhang
- Great Ormond Street Institute of Child Health, Genetics and Genomic Medicine, University College London, London, WC1N 1EH, UK
| | - John Hardy
- Department of Neurodegenerative Disease, University College London, London, WC1N 3BG, UK
- UK Dementia Research Institute at University College London, London, WC1N 3BG, UK
| | - Juan A Botía
- Great Ormond Street Institute of Child Health, Genetics and Genomic Medicine, University College London, London, WC1N 1EH, UK
- Departamento de Ingeniería de la Información y las Comunicaciones, Universidad de Murcia, 30100, Murcia, Spain
| | - Michael E Weale
- Department of Medical & Molecular Genetics, School of Medical Sciences, King's College London, Guy's Hospital, London, SE1 1UL, UK
- Genomics Plc, Oxford, OX1 1JD, UK
| | - Sarah A Gagliano Taliun
- Department of Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Montréal Heart Institute, Montréal, QC, H1T 1C8, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Kerrin S Small
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK
| | - Mina Ryten
- Great Ormond Street Institute of Child Health, Genetics and Genomic Medicine, University College London, London, WC1N 1EH, UK.
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, WC1N 3JH, UK.
| |
Collapse
|
24
|
Johnson EC, Colbert SMC, Jeffries PW, Tillman R, Bigdeli TB, Karcher NR, Chan G, Kuperman S, Meyers JL, Nurnberger JI, Plawecki MH, Degenhardt L, Martin NG, Kamarajan C, Schuckit MA, Murray RM, Dick DM, Edenberg HJ, D’Souza DC, Di Forti M, Porjesz B, Nelson EC, Agrawal A. Associations Between Cannabis Use, Polygenic Liability for Schizophrenia, and Cannabis-related Experiences in a Sample of Cannabis Users. Schizophr Bull 2023; 49:778-787. [PMID: 36545904 PMCID: PMC10154717 DOI: 10.1093/schbul/sbac196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND HYPOTHESIS Risk for cannabis use and schizophrenia is influenced in part by genetic factors, and there is evidence that genetic risk for schizophrenia is associated with subclinical psychotic-like experiences (PLEs). Few studies to date have examined whether genetic risk for schizophrenia is associated with cannabis-related PLEs. STUDY DESIGN We tested whether measures of cannabis involvement and polygenic risk scores (PRS) for schizophrenia were associated with self-reported cannabis-related experiences in a sample ascertained for alcohol use disorders (AUDs), the Collaborative Study on the Genetics of Alcoholism (COGA). We analyzed 4832 subjects (3128 of European ancestry and 1704 of African ancestry; 42% female; 74% meeting lifetime criteria for an AUD). STUDY RESULTS Cannabis use disorder (CUD) was prevalent in this analytic sample (70%), with 40% classified as mild, 25% as moderate, and 35% as severe. Polygenic risk for schizophrenia was positively associated with cannabis-related paranoia, feeling depressed or anhedonia, social withdrawal, and cognitive difficulties, even when controlling for duration of daily cannabis use, CUD, and age at first cannabis use. The schizophrenia PRS was most robustly associated with cannabis-related cognitive difficulties (β = 0.22, SE = 0.04, P = 5.2e-7). In an independent replication sample (N = 1446), associations between the schizophrenia PRS and cannabis-related experiences were in the expected direction and not statistically different in magnitude from those in the COGA sample. CONCLUSIONS Among individuals who regularly use cannabis, genetic liability for schizophrenia-even in those without clinical features-may increase the likelihood of reporting unusual experiences related to cannabis use.
Collapse
Affiliation(s)
- Emma C Johnson
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Sarah M C Colbert
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Paul W Jeffries
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Rebecca Tillman
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Tim B Bigdeli
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Henri Begleiter Neurodynamics Lab, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Nicole R Karcher
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Grace Chan
- Department of Psychiatry, University of Connecticut, Farmington, CT, USA
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Samuel Kuperman
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Jacquelyn L Meyers
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Henri Begleiter Neurodynamics Lab, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - John I Nurnberger
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Martin H Plawecki
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Louisa Degenhardt
- National Drug and Alcohol Research Centre, UNSW Sydney, Sydney, New South Wales, Australia
| | - Nicholas G Martin
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Chella Kamarajan
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Henri Begleiter Neurodynamics Lab, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Marc A Schuckit
- Department of Psychiatry, University of California San Diego Medical School, San Diego, CA, USA
| | - Robin M Murray
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Danielle M Dick
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Howard J Edenberg
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Deepak Cyril D’Souza
- Department of Psychiatry, Yale University, New Haven, CT, USA
- Psychiatry Service, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Marta Di Forti
- Department of Social Genetics and Developmental Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- South London and Maudsley NHS Mental Health Foundation Trust, London, UK
| | - Bernice Porjesz
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Henri Begleiter Neurodynamics Lab, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Elliot C Nelson
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Arpana Agrawal
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
25
|
Pine JG, Paul SE, Johnson E, Bogdan R, Kandala S, Barch DM. Polygenic Risk for Schizophrenia, Major Depression, and Post-traumatic Stress Disorder and Hippocampal Subregion Volumes in Middle Childhood. Behav Genet 2023; 53:279-291. [PMID: 36720770 PMCID: PMC10875985 DOI: 10.1007/s10519-023-10134-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/17/2023] [Indexed: 02/02/2023]
Abstract
Studies demonstrate that individuals with diagnoses for Major Depressive Disorder (MDD), Post-traumatic Stress Disorder (PTSD), and Schizophrenia (SCZ) may exhibit smaller hippocampal gray matter relative to otherwise healthy controls, although the effect sizes vary in each disorder. Existing work suggests that hippocampal abnormalities in each disorder may be attributable to genetic liability and/or environmental variables. The following study uses baseline data from the Adolescent Brain and Cognitive Development[Formula: see text] Study (ABCD Study[Formula: see text]) to address three open questions regarding the relationship between genetic risk for each disorder and hippocampal volume reductions: (a) whether polygenic risk scores (PGRS) for MDD, PTSD, and SCZ are related to hippocampal volume; (b) whether PGRS for MDD, PTSD, and SCZ are differentially related to specific hippocampal subregions along the longitudinal axis; and (c) whether the association between PGRS for MDD, PTSD, and SCZ and hippocampal volume is moderated by sex and/or environmental adversity. In short, we did not find associations between PGRS for MDD, PTSD, and SCZ to be significantly related to any hippocampal subregion volumes. Furthermore, neither sex nor enviornmental adversity significantly moderated these associations. Our study provides an important null finding on the relationship genetic risk for MDD, PTSD, and SCZ to measures of hippocampal volume.
Collapse
Affiliation(s)
- Jacob G Pine
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, 63130, USA.
| | - Sarah E Paul
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Emma Johnson
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ryan Bogdan
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Sridhar Kandala
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Deanna M Barch
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, 63130, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
26
|
van den Oord EJCG, Xie LY, Zhao M, Campbell TL, Turecki G, Kähler AK, Dean B, Mors O, Hultman CM, Staunstrup NH, Aberg KA. Genes implicated by a methylome-wide schizophrenia study in neonatal blood show differential expression in adult brain samples. Mol Psychiatry 2023; 28:2088-2094. [PMID: 37106120 DOI: 10.1038/s41380-023-02080-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023]
Abstract
Schizophrenia is a disabling disorder involving genetic predisposition in combination with environmental influences that likely act via dynamic alterations of the epigenome and the transcriptome but its detailed pathophysiology is largely unknown. We performed cell-type specific methylome-wide association study of neonatal blood (N = 333) from individuals who later in life developed schizophrenia and controls. Suggestively significant associations (P < 1.0 × 10-6) were detected in all cell-types and in whole blood with methylome-wide significant associations in monocytes (P = 2.85 × 10-9-4.87 × 10-9), natural killer cells (P = 1.72 × 10-9-7.82 × 10-9) and B cells (P = 3.8 × 10-9). Validation of methylation findings in post-mortem brains (N = 596) from independent schizophrenia cases and controls showed significant enrichment of transcriptional differences (enrichment ratio = 1.98-3.23, P = 2.3 × 10-3-1.0 × 10-5), with specific highly significant differential expression for, for example, BDNF (t = -6.11, P = 1.90 × 10-9). In addition, expression difference in brain significantly predicted schizophrenia (multiple correlation = 0.15-0.22, P = 3.6 × 10-4-4.5 × 10-8). In summary, using a unique design combining pre-disease onset (neonatal) blood methylomic data and post-disease onset (post-mortem) brain transcriptional data, we have identified genes of likely functional relevance that are associated with schizophrenia susceptibility, rather than confounding disease associated artifacts. The identified loci may be of clinical value as a methylation-based biomarker for early detection of increased schizophrenia susceptibility.
Collapse
Affiliation(s)
- Edwin J C G van den Oord
- Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Lin Y Xie
- Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Min Zhao
- Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Thomas L Campbell
- Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Gustavo Turecki
- Douglas Mental Health University Institute and McGill University, Montréal, Québec, Canada
| | - Anna K Kähler
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Brian Dean
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Ole Mors
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Psychosis Research Unit, Aarhus University Hospital - Psychiatry, Risskov, Denmark
- Center for Genomics and Personalized Medicine, University of Aarhus, Aarhus, Denmark
| | - Christina M Hultman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Nicklas H Staunstrup
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, University of Aarhus, Aarhus, Denmark
- Department of Biomedicine, University of Aarhus, Aarhus C, Denmark
| | - Karolina A Aberg
- Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
27
|
Sano F, Kikushima K, Benner S, Xu L, Kahyo T, Yamasue H, Setou M. Associations between prefrontal PI (16:0/20:4) lipid, TNC mRNA, and APOA1 protein in schizophrenia: A trans-omics analysis in post-mortem brain. Front Psychiatry 2023; 14:1145437. [PMID: 37143779 PMCID: PMC10151580 DOI: 10.3389/fpsyt.2023.1145437] [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: 01/16/2023] [Accepted: 03/24/2023] [Indexed: 05/06/2023] Open
Abstract
Background Though various mechanisms have been proposed for the pathophysiology of schizophrenia, the full extent of these mechanisms remains unclear, and little is known about the relationships among them. We carried out trans-omics analyses by comparing the results of the previously reported lipidomics, transcriptomics, and proteomics analyses; all of these studies used common post-mortem brain samples. Methods We collected the data from three aforementioned omics studies on 6 common post-mortem samples (3 schizophrenia patients and 3 controls), and analyzed them as a whole group sample. Three correlation analyses were performed for each of the two of three omics studies in these samples. In order to discuss the strength of the correlations in a limited sample size, the p-values of each correlation coefficient were confirmed using the Student's t-test. In addition, partial correlation analysis was also performed for some correlations, to verify the strength of the impact of each factor on the correlations. Results The following three factors were strongly correlated with each other: the lipid level of phosphatidylinositol (PI) (16:0/20:4), the amount of TNC mRNA, and the quantitative signal intensity of APOA1 protein. PI (16:0/20:4) and TNC showed a positive correlation, while PI (16:0/20:4) and APOA1, and TNC and APOA1 showed negative correlations. All of these correlations reached at p < 0.01. PI (16:0/20:4) and TNC were decreased in the prefrontal cortex of schizophrenia samples, while APOA1 was increased. Partial correlation analyses among them suggested that PI (16:0/20:4) and TNC have no direct correlation, but their relationships are mediated by APOA1. Conclusion The current results suggest that these three factors may provide new clues to elucidate the relationships among the candidate mechanisms of schizophrenia, and support the potential of trans-omics analyses as a new analytical method.
Collapse
Affiliation(s)
- Fumito Sano
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kenji Kikushima
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Seico Benner
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Lili Xu
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomoaki Kahyo
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hidenori Yamasue
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Systems Molecular Anatomy, Institute for Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
28
|
Zhang W, Du JL, Fang XY, Ni LY, Zhu YY, Yan W, Lu SP, Zhang RR, Xie SP. Shared and distinct structural brain alterations and cognitive features in drug-naïve schizophrenia and bipolar disorder. Asian J Psychiatr 2023; 82:103513. [PMID: 36827938 DOI: 10.1016/j.ajp.2023.103513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/21/2023] [Accepted: 02/08/2023] [Indexed: 02/13/2023]
Abstract
Our study aimed to examine the shared and distinct structural brain alterations, including cortical thickness(CT) and local gyrification index(LGI), and cognitive impairments between the early course stage of drug-naïve schizophrenia(SZ) and bipolar disorder(BD) patients when compared to healthy controls(HCs), and to further explore the correlation between altered brain structure and cognitive impairments. We included 72 SZ patients, 35 BD patients and 43 HCs. The cognitive function was assessed using the MATRICS Consensus Cognitive Battery. Cerebral cortex analyses were performed with FreeSurfer. Furthermore, any structural aberrations related to cognition impairments were examined. Cognitive impairments existed in SZ and BD patients and were much more severe and widespread in SZ patients, compared to HCs. There were no significant differences in LGI among three groups. Compared to HCs, SZ had thicker cortex in left pars triangularis, and BD showed thinner CT in left postcentral gyrus. In addition, BD showed thinner cortex in left pars triangularis, left pars opercularis, left insula and right fusiform gyrus compared to SZ. Moreover, our results indicated that CT in many brain areas were significantly correlated with cognitive function in HCs, but only CT of left pars triangularis was correlated with impaired social cognition found in SZ. The findings suggest that changes of CT in the left pars triangularis and left postcentral gyrus may be potential pathophysiological mechanisms of the cognition impairments in SZ and BD, respectively, and the divergent CT of partly brain areas in BD vs. SZ may help distinguish them in early phases.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.
| | - Jing-Lun Du
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.
| | - Xing-Yu Fang
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.
| | - Long-Yan Ni
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.
| | - Yuan-Yuan Zhu
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.
| | - Wei Yan
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.
| | - Shui-Ping Lu
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.
| | - Rong-Rong Zhang
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.
| | - Shi-Ping Xie
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
29
|
Privitera F, Trusso MA, Valentino F, Doddato G, Fallerini C, Brunelli G, D'Aurizio R, Furini S, Goracci A, Fagiolini A, Mari F, Renieri A, Ariani F. Heterozygosity for neuronal ceroid lipofuscinosis predisposes to bipolar disorder. REVISTA BRASILEIRA DE PSIQUIATRIA (SAO PAULO, BRAZIL : 1999) 2023; 45:11-19. [PMID: 35881528 PMCID: PMC9976914 DOI: 10.47626/1516-4446-2022-2650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 07/13/2022] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Bipolar disorder is a heritable chronic mental disorder that causes psychosocial impairment through depressive/manic episodes. Familial transmission of bipolar disorder does not follow simple Mendelian patterns of inheritance. The aim of this study was to describe a large family with 12 members affected by bipolar disorder. Whole-exome sequencing was performed for eight members, three of whom were diagnosed with bipolar disorder, and another reported as "borderline." METHODS Whole-exome sequencing data allowed us to select variants that the affected members had in common, including and excluding the "borderline" individual with moderate anxiety and obsessive-compulsive traits. RESULTS The results favored designating certain genes as predispositional to bipolar disorder: a heterozygous missense variant in CLN6 resulted in a "borderline" phenotype that, if combined with a heterozygous missense variant in ZNF92, is responsible for the more severe bipolar disorder phenotype. Both rare missense changes are predicted to disrupt protein function. CONCLUSIONS Loss of both alleles in CLN6 causes neuronal ceroid lipofuscinosis, a severe progressive childhood neurological disorder. Our results indicate that heterozygous CLN6 carriers, previously reported as healthy, may be susceptible to bipolar disorder later in life if associated with additional variants in ZNF92.
Collapse
Affiliation(s)
- Flavia Privitera
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Maria A Trusso
- Department of Molecular Medicine and Development, University of Siena, Siena, Italy
| | - Floriana Valentino
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Gabriella Doddato
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Chiara Fallerini
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Giulia Brunelli
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Romina D'Aurizio
- Institute of Informatics and Telematics, National Research Council, Pisa, Italy
| | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Arianna Goracci
- Department of Molecular Medicine and Development, University of Siena, Siena, Italy. Department of Mental Health; Psychiatry Unit, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Andrea Fagiolini
- Department of Molecular Medicine and Development, University of Siena, Siena, Italy
| | - Francesca Mari
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy. Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Alessandra Renieri
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy. Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Francesca Ariani
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy. Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| |
Collapse
|
30
|
Liu D, Meyer D, Fennessy B, Feng C, Cheng E, Johnson JS, Park YJ, Rieder MK, Ascolillo S, de Pins A, Dobbyn A, Lebovitch D, Moya E, Nguyen TH, Wilkins L, Hassan A, Burdick KE, Buxbaum JD, Domenici E, Frangou S, Hartmann AM, Laurent-Levinson C, Malhotra D, Pato CN, Pato MT, Ressler K, Roussos P, Rujescu D, Arango C, Bertolino A, Blasi G, Bocchio-Chiavetto L, Campion D, Carr V, Fullerton JM, Gennarelli M, González-Peñas J, Levinson DF, Mowry B, Nimgaokar VL, Pergola G, Rampino A, Cervilla JA, Rivera M, Schwab SG, Wildenauer DB, Daly M, Neale B, Singh T, O'Donovan MC, Owen MJ, Walters JT, Ayub M, Malhotra AK, Lencz T, Sullivan PF, Sklar P, Stahl EA, Huckins LM, Charney AW. Schizophrenia risk conferred by rare protein-truncating variants is conserved across diverse human populations. Nat Genet 2023; 55:369-376. [PMID: 36914870 PMCID: PMC10011128 DOI: 10.1038/s41588-023-01305-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/23/2023] [Indexed: 03/14/2023]
Abstract
Schizophrenia (SCZ) is a chronic mental illness and among the most debilitating conditions encountered in medical practice. A recent landmark SCZ study of the protein-coding regions of the genome identified a causal role for ten genes and a concentration of rare variant signals in evolutionarily constrained genes1. This recent study-and most other large-scale human genetics studies-was mainly composed of individuals of European (EUR) ancestry, and the generalizability of the findings in non-EUR populations remains unclear. To address this gap, we designed a custom sequencing panel of 161 genes selected based on the current knowledge of SCZ genetics and sequenced a new cohort of 11,580 SCZ cases and 10,555 controls of diverse ancestries. Replicating earlier work, we found that cases carried a significantly higher burden of rare protein-truncating variants (PTVs) among evolutionarily constrained genes (odds ratio = 1.48; P = 5.4 × 10-6). In meta-analyses with existing datasets totaling up to 35,828 cases and 107,877 controls, this excess burden was largely consistent across five ancestral populations. Two genes (SRRM2 and AKAP11) were newly implicated as SCZ risk genes, and one gene (PCLO) was identified as shared by individuals with SCZ and those with autism. Overall, our results lend robust support to the rare allelic spectrum of the genetic architecture of SCZ being conserved across diverse human populations.
Collapse
Affiliation(s)
- Dongjing Liu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Dara Meyer
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian Fennessy
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Claudia Feng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Wellcome Sanger Institute, Hinxton, UK
| | - Esther Cheng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica S Johnson
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - You Jeong Park
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marysia-Kolbe Rieder
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven Ascolillo
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Agathe de Pins
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amanda Dobbyn
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dannielle Lebovitch
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily Moya
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tan-Hoang Nguyen
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - Lillian Wilkins
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Katherine E Burdick
- Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Joseph D Buxbaum
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Enrico Domenici
- Centre for Computational and Systems Biology, Fondazione The Microsoft Research - University of Trento, Rovereto, Italy
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Sophia Frangou
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Annette M Hartmann
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Claudine Laurent-Levinson
- Faculté de Médecine Sorbonne Université, Groupe de Recherche Clinique n°15-Troubles Psychiatriques et Développement, Department of Child and Adolescent Psychiatry, Hôpital Universitaire de la Pitié-Salpêtrière, Paris, France
- Centre de Référence des Maladies Rares à Expression Psychiatrique, Department of Child and Adolescent Psychiatry, AP-HP Sorbonne Université, Hôpital Universitaire de la Pitié-Salpêtrière, Paris, France
| | - Dheeraj Malhotra
- Department of Neuroscience and Rare Diseases, Roche Pharma Research and Early Development, F. Hoffmann-La Roche, Basel, Switzerland
| | - Carlos N Pato
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate College of Medicine, New York, NY, USA
| | - Michele T Pato
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate College of Medicine, New York, NY, USA
| | - Kerry Ressler
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, MA, USA
| | - Panos Roussos
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education, and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, New York, NY, USA
| | - Dan Rujescu
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Celso Arango
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Madrid, Spain
| | - Alessandro Bertolino
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Blasi
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Luisella Bocchio-Chiavetto
- Department of Theoretical and Applied Sciences, eCampus University, Novedrate, Italy
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Dominique Campion
- INSERM U1245, Rouen, France
- Centre Hospitalier du Rouvray, Rouen, France
| | - Vaughan Carr
- Neuroscience Research Australia, Sydney, New South Wales, Australia
- School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
- Department of Psychiatry, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Janice M Fullerton
- Neuroscience Research Australia, Sydney, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Massimo Gennarelli
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Javier González-Peñas
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Madrid, Spain
| | | | - Bryan Mowry
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
- Queensland Centre for Mental Health Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Vishwajit L Nimgaokar
- Department of Psychiatry, University of Pittsburgh School of Medicine, Western Psychiatric Hospital, Pittsburgh, PA, USA
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Giulio Pergola
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Antonio Rampino
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Jorge A Cervilla
- Institute of Neurosciences, Biomedical Research Centre, University of Granada, Granada, Spain
- Department of Psychiatry, San Cecilio University Hospital, University of Granada, Granada, Spain
| | - Margarita Rivera
- Institute of Neurosciences, Biomedical Research Centre, University of Granada, Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain
| | - Sibylle G Schwab
- Molecular Horizons, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, New South Wales, Australia
| | | | - Mark Daly
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Benjamin Neale
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Tarjinder Singh
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael C O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Michael J Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - James T Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Muhammad Ayub
- University College London, London, UK
- Department of Psychiatry, Queen's University, Kingston, Ontario, Canada
| | - Anil K Malhotra
- Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Institute for Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Division of Psychiatry Research, The Zucker Hillside Hospital, Northwell Health, New York, NY, USA
| | - Todd Lencz
- Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Institute for Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Division of Psychiatry Research, The Zucker Hillside Hospital, Northwell Health, New York, NY, USA
| | - Patrick F Sullivan
- Departments of Genetics and Psychiatry, University of North Carolina, Chapel Hill, NC, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Pamela Sklar
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eli A Stahl
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Laura M Huckins
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Alexander W Charney
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
31
|
Kamran M, Laighneach A, Bibi F, Donohoe G, Ahmed N, Rehman AU, Morris DW. Independent Associated SNPs at SORCS3 and Its Protein Interactors for Multiple Brain-Related Disorders and Traits. Genes (Basel) 2023; 14:482. [PMID: 36833409 PMCID: PMC9956385 DOI: 10.3390/genes14020482] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/26/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023] Open
Abstract
Sortilin-related vacuolar protein sorting 10 (VPS10) domain containing receptor 3 (SORCS3) is a neuron-specific transmembrane protein involved in the trafficking of proteins between intracellular vesicles and the plasma membrane. Genetic variation at SORCS3 is associated with multiple neuropsychiatric disorders and behavioural phenotypes. Here, we undertake a systematic search of published genome-wide association studies to identify and catalogue associations between SORCS3 and brain-related disorders and traits. We also generate a SORCS3 gene-set based on protein-protein interactions and investigate the contribution of this gene-set to the heritability of these phenotypes and its overlap with synaptic biology. Analysis of association signals at SORSC3 showed individual SNPs to be associated with multiple neuropsychiatric and neurodevelopmental brain-related disorders and traits that have an impact on the experience of feeling, emotion or mood or cognitive function, while multiple LD-independent SNPs were associated with the same phenotypes. Across these SNPs, alleles associated with the more favourable outcomes for each phenotype (e.g., decreased risk of neuropsychiatric illness) were associated with increased expression of the SORCS3 gene. The SORCS3 gene-set was enriched for heritability contributing to schizophrenia (SCZ), bipolar disorder (BPD), intelligence (IQ) and education attainment (EA). Eleven genes from the SORCS3 gene-set were associated with more than one of these phenotypes at the genome-wide level, with RBFOX1 associated with SCZ, IQ and EA. Functional annotation revealed that the SORCS3 gene-set is enriched for multiple ontologies related to the structure and function of synapses. Overall, we find many independent association signals at SORCS3 with brain-related disorders and traits, with the effect possibly mediated by reduced gene expression, resulting in a negative impact on synaptic function.
Collapse
Affiliation(s)
- Muhammad Kamran
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, H91 CF50 Galway, Ireland
| | - Aodán Laighneach
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, H91 CF50 Galway, Ireland
| | - Farhana Bibi
- Department of Biosciences, Grand Asian University, Sialkot 51040, Pakistan
| | - Gary Donohoe
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, H91 CF50 Galway, Ireland
| | - Naveed Ahmed
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Asim Ur Rehman
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Derek W. Morris
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, H91 CF50 Galway, Ireland
| |
Collapse
|
32
|
Comparison of Demographic and Clinical Features of Bipolar Disorder in Persons of African and European Ancestry. J Racial Ethn Health Disparities 2023; 10:367-372. [PMID: 35064520 DOI: 10.1007/s40615-022-01228-3] [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: 07/19/2021] [Revised: 11/24/2021] [Accepted: 01/04/2022] [Indexed: 02/03/2023]
Abstract
AIM This study quantified and compared demographic and clinical features of bipolar disorder (BD) in persons of African ancestry (AA) and European ancestry (EUR). METHODS Participants enrolled in the Mayo Clinic Bipolar Biobank from 2009 to 2015. The structured clinical interview for DSM-IV was used to confirm the diagnosis of BD, and a questionnaire was developed to collect data on the clinical course of illness. Descriptive statistics and bivariate analyses were completed to compare AA versus EUR participants. Subsequently, clinical outcomes were compared between AA and EUR participants using linear regression for continuous outcomes or logistic regression for binary outcomes while controlling for differences in age, sex, and recruitment site. RESULTS Of 1865 participants enrolled in the bipolar biobank, 65 (3.5%) self-identified as AA. The clinical phenotype for AA participants, in comparison to EUR participants, was more likely to include a history of PTSD (39.7% vs. 26.2%), cocaine use disorder (24.2% vs. 11.9%), and tardive dyskinesia (7.1% vs. 3%). CONCLUSION The low rate of AA enrollment is consistent with other genetic studies. While clinical features of bipolar disorder are largely similar, this study identified differences in rates of trauma, substance use, and tardive dyskinesia that may represent health disparities in bipolar patients of African ancestry. Future bipolar biomarker studies with larger sample sizes focused on underrepresented populations will provide greater ancestry diversity in genomic medicine with greater applicability to diverse patient populations, serving to inform health care policies to address disparities in bipolar disorder.
Collapse
|
33
|
Genetic substrates of bipolar disorder risk in Latino families. Mol Psychiatry 2023; 28:154-167. [PMID: 35948660 DOI: 10.1038/s41380-022-01705-5] [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: 04/30/2021] [Revised: 06/22/2022] [Accepted: 07/07/2022] [Indexed: 01/07/2023]
Abstract
Genetic studies of bipolar disorder (BP) have been conducted in the Latin American population, to date, in several countries, including Mexico, the United States, Costa Rica, Colombia, and, to a lesser extent, Brazil. These studies focused primarily on linkage-based designs utilizing families with multiplex cases of BP. Significant BP loci were identified on Chromosomes 18, 5 and 8, and fine mapping suggested several genes of interest underlying these linkage peaks. More recently, studies in these same pedigrees yielded significant linkage loci for BP endophenotypes, including measures of activity, sleep cycles, and personality traits. Building from findings in other populations, candidate gene association analyses in Latinos from Mexican and Central American ancestry confirmed the role of several genes (including CACNA1C and ANK3) in conferring BP risk. Although GWAS, methylation, and deep sequencing studies have only begun in these populations, there is evidence that CNVs and rare SNPs both play a role in BP risk of these populations. Large segments of the Latino populations in the Americas remain largely unstudied regarding BP genetics, but evidence to date has shown that this type of research can be successfully conducted in these populations and that the genetic underpinnings of BP in these cohorts share at least some characteristics with risk genes identified in European and other populations.
Collapse
|
34
|
Gu X, Dou M, Su W, Jiang Z, Duan Q, Cao B, Chen Y. Identifying novel proteins underlying schizophrenia via integrating pQTLs of the plasma, CSF, and brain with GWAS summary data. BMC Med 2022; 20:474. [PMID: 36482464 PMCID: PMC9730613 DOI: 10.1186/s12916-022-02679-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Schizophrenia (SCZ) is a chronic and severe mental illness with no cure so far. Mendelian randomization (MR) is a genetic method widely used to explore etiologies of complex traits. In the current study, we aimed to identify novel proteins underlying SCZ with a systematic analytical approach. METHODS We integrated protein quantitative trait loci (pQTLs) of the brain, cerebrospinal fluid (CSF), and plasma with the latest and largest SCZ genome-wide association study (GWAS) via a systematic analytical framework, including two-sample MR analysis, Steiger filtering analysis, and Bayesian colocalization analysis. RESULTS The genetically determined protein level of C4A/C4B (OR = 0.70, p = 1.66E-07) in the brain and ACP5 (OR = 0.42, p = 3.73E-05), CNTN2 (OR = 0.62, p = 2.57E-04), and PLA2G7 (OR = 0.71, p = 1.48E-04) in the CSF was associated with a lower risk of SCZ, while the genetically determined protein level of TIE1 (OR = 3.46, p = 4.76E-05), BCL6 (OR = 3.63, p = 1.59E-07), and MICB (OR = 4.49, p = 2.31E-11) in the CSF were associated with an increased risk for SCZ. Pathway enrichment analysis indicated that genetically determined proteins suggestively associated with SCZ were enriched in the biological process of the immune response. CONCLUSION In conclusion, we identified one protein in the brain and six proteins in the CSF that showed supporting evidence of being potentially associated with SCZ, which could provide insights into future mechanistic studies to find new treatments for the disease. Our results also supported the important role of neuroinflammation in the pathogenesis of SCZ.
Collapse
Affiliation(s)
- Xiaojing Gu
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Meng Dou
- Chengdu Institute of Computer Application, Chinese Academy of Sciences, Chengdu, 610041, Sichuan, China
| | - Weiming Su
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.,Lab of Neurodegenerative Disorders, Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.,Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zheng Jiang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.,Lab of Neurodegenerative Disorders, Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.,Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qingqing Duan
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.,Lab of Neurodegenerative Disorders, Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.,Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Bei Cao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.,Lab of Neurodegenerative Disorders, Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.,Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yongping Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China. .,Lab of Neurodegenerative Disorders, Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China. .,Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
35
|
Barr PB, Driver MN, Kuo SIC, Stephenson M, Aliev F, Linnér RK, Marks J, Anokhin AP, Bucholz K, Chan G, Edenberg HJ, Edwards AC, Francis MW, Hancock DB, Harden KP, Kamarajan C, Kaprio J, Kinreich S, Kramer JR, Kuperman S, Latvala A, Meyers JL, Palmer AA, Plawecki MH, Porjesz B, Rose RJ, Schuckit MA, Salvatore JE, Dick DM. Clinical, environmental, and genetic risk factors for substance use disorders: characterizing combined effects across multiple cohorts. Mol Psychiatry 2022; 27:4633-4641. [PMID: 36195638 PMCID: PMC9938102 DOI: 10.1038/s41380-022-01801-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Substance use disorders (SUDs) incur serious social and personal costs. The risk for SUDs is complex, with risk factors ranging from social conditions to individual genetic variation. We examined whether models that include a clinical/environmental risk index (CERI) and polygenic scores (PGS) are able to identify individuals at increased risk of SUD in young adulthood across four longitudinal cohorts for a combined sample of N = 15,134. Our analyses included participants of European (NEUR = 12,659) and African (NAFR = 2475) ancestries. SUD outcomes included: (1) alcohol dependence, (2) nicotine dependence; (3) drug dependence, and (4) any substance dependence. In the models containing the PGS and CERI, the CERI was associated with all three outcomes (ORs = 01.37-1.67). PGS for problematic alcohol use, externalizing, and smoking quantity were associated with alcohol dependence, drug dependence, and nicotine dependence, respectively (OR = 1.11-1.33). PGS for problematic alcohol use and externalizing were also associated with any substance dependence (ORs = 1.09-1.18). The full model explained 6-13% of the variance in SUDs. Those in the top 10% of CERI and PGS had relative risk ratios of 3.86-8.04 for each SUD relative to the bottom 90%. Overall, the combined measures of clinical, environmental, and genetic risk demonstrated modest ability to distinguish between affected and unaffected individuals in young adulthood. PGS were significant but added little in addition to the clinical/environmental risk index. Results from our analysis demonstrate there is still considerable work to be done before tools such as these are ready for clinical applications.
Collapse
Affiliation(s)
- Peter B Barr
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA.
- VA New York Harbor Healthcare System, Brooklyn, NY, USA.
| | - Morgan N Driver
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Sally I-Chun Kuo
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Mallory Stephenson
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - Fazil Aliev
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
- Rutgers Addiction Research Center, Rutgers University, Piscataway, NJ, USA
| | | | - Jesse Marks
- Biostatistics and Epidemiology Division, RTI International, Research Triangle Park, Durham, NC, USA
| | - Andrey P Anokhin
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St Louis, MO, USA
| | - Kathleen Bucholz
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St Louis, MO, USA
| | - Grace Chan
- Department of Psychiatry, School of Medicine, University of Connecticut, Farmington, CT, USA
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Howard J Edenberg
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Alexis C Edwards
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - Meredith W Francis
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St Louis, MO, USA
| | - Dana B Hancock
- Biostatistics and Epidemiology Division, RTI International, Research Triangle Park, Durham, NC, USA
| | - K Paige Harden
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
- Population Research Center, University of Texas at Austin, Austin, TX, USA
| | - Chella Kamarajan
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Sivan Kinreich
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - John R Kramer
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Samuel Kuperman
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Antti Latvala
- Institute of Criminology and Legal Policy, University of Helsinki, Helsinki, Finland
| | - Jacquelyn L Meyers
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- VA New York Harbor Healthcare System, Brooklyn, NY, USA
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Martin H Plawecki
- Department of Psychiatry, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Bernice Porjesz
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Richard J Rose
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Marc A Schuckit
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Jessica E Salvatore
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Danielle M Dick
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
- Rutgers Addiction Research Center, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
36
|
Malaspina D. A sister's search for the seeds of psychosis. Psychiatry Res 2022; 317:114846. [PMID: 36244157 DOI: 10.1016/j.psychres.2022.114846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/07/2022] [Accepted: 09/11/2022] [Indexed: 01/04/2023]
Abstract
Dr. Dolores Malaspina sought a better way to understand the origins of psychosis than a schizophrenogenic mother, as her family had been informed upon her sisters illness. She moved her attention from environmental biology and zoology, to medical science and assembled knowledge on the multilevel components purported to underpin severe mental illness. Her studies cross levels to consider connections among exposures and genetic etiologies, intrinsic homeostatic mechanisms, stimuli perception and clinical illness features. Original contributions include associating later paternal age with increasing risk for schizophrenia in offspring and proposing that de novo mutations with shorter cell cycles explained the association, showing increased resting hippocampal blood flow in psychosis and that it was associated with inflammation, and that autonomic nervous system dysfunction was related to hippocampal inflammation, plausibly reflecting vascular abnormalities. She has been a professor of psychiatry in medical schools at Columbia University, New York University and at Mount Sinai in New York, USA.
Collapse
Affiliation(s)
- Dolores Malaspina
- Department of Psychiatry, Neuroscience, Genetic & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
37
|
Pergola G, Penzel N, Sportelli L, Bertolino A. Lessons Learned From Parsing Genetic Risk for Schizophrenia Into Biological Pathways. Biol Psychiatry 2022:S0006-3223(22)01701-2. [PMID: 36740470 DOI: 10.1016/j.biopsych.2022.10.009] [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: 04/16/2022] [Revised: 09/10/2022] [Accepted: 10/06/2022] [Indexed: 02/07/2023]
Abstract
The clinically heterogeneous presentation of schizophrenia is compounded by the heterogeneity of risk factors and neurobiological correlates of the disorder. Genome-wide association studies in schizophrenia have uncovered a remarkably high number of genetic variants, but the biological pathways they impact upon remain largely unidentified. Among the diverse methodological approaches employed to provide a more granular understanding of genetic risk for schizophrenia, the use of biological labels, such as gene ontologies, regulome approaches, and gene coexpression have all provided novel perspectives into how genetic risk translates into the neurobiology of schizophrenia. Here, we review the salient aspects of parsing polygenic risk for schizophrenia into biological pathways. We argue that parsed scores, compared to standard polygenic risk scores, may afford a more biologically plausible and accurate physiological modeling of the different dimensions involved in translating genetic risk into brain mechanisms, including multiple brain regions, cell types, and maturation stages. We discuss caveats, opportunities, and pitfalls inherent in the parsed risk approach.
Collapse
Affiliation(s)
- Giulio Pergola
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy.
| | - Nora Penzel
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Leonardo Sportelli
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Alessandro Bertolino
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
38
|
Fusion of Clinical and Lived Experiences of Psychosis: Lessons Learned and Implications for Future Clinical Teaching. PSYCHIATRY INTERNATIONAL 2022. [DOI: 10.3390/psychiatryint3040023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Psychosis is a clinical syndrome that can cause significant distress leading to hospitalisation/long term stays in psychiatric services. However, limited academic evidence is available examining the lived experience of psychosis. Additionally, no evidence is available looking to combine both learned and experiential knowledge as it pertains to psychosis. As such this article was created to combine both knowledge subsets in order to provide a more complete interpretation of the syndrome itself. This was achieved through academic input from a psychiatrist’s perspective as well as a reflective, autoethnographic input from a service user who has experienced psychosis. Following this collaboration, several recommendations were made to support health professionals to engage appropriately with service users with psychosis. However, the lived experiences of psychosis itself requires further investigation to identify commonalities in experiences that can support clinicians in the diagnosis and co-production of treatment regimens for these service users.
Collapse
|
39
|
Bigdeli TB, Voloudakis G, Barr PB, Gorman BR, Genovese G, Peterson RE, Burstein DE, Velicu VI, Li Y, Gupta R, Mattheisen M, Tomasi S, Rajeevan N, Sayward F, Radhakrishnan K, Natarajan S, Malhotra AK, Shi Y, Zhao H, Kosten TR, Concato J, O’Leary TJ, Przygodzki R, Gleason T, Pyarajan S, Brophy M, Huang GD, Muralidhar S, Gaziano JM, Aslan M, Fanous AH, Harvey PD, Roussos P. Penetrance and Pleiotropy of Polygenic Risk Scores for Schizophrenia, Bipolar Disorder, and Depression Among Adults in the US Veterans Affairs Health Care System. JAMA Psychiatry 2022; 79:2796413. [PMID: 36103194 PMCID: PMC9475441 DOI: 10.1001/jamapsychiatry.2022.2742] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 07/20/2022] [Indexed: 12/21/2022]
Abstract
Importance Serious mental illnesses, including schizophrenia, bipolar disorder, and depression, are heritable, highly multifactorial disorders and major causes of disability worldwide. Objective To benchmark the penetrance of current neuropsychiatric polygenic risk scores (PRSs) in the Veterans Health Administration health care system and to explore associations between PRS and broad categories of human disease via phenome-wide association studies. Design, Setting, and Participants Extensive Veterans Health Administration's electronic health records were assessed from October 1999 to January 2021, and an embedded cohort of 9378 individuals with confirmed diagnoses of schizophrenia or bipolar 1 disorder were found. The performance of schizophrenia, bipolar disorder, and major depression PRSs were compared in participants of African or European ancestry in the Million Veteran Program (approximately 400 000 individuals), and associations between PRSs and 1650 disease categories based on ICD-9/10 billing codes were explored. Last, genomic structural equation modeling was applied to derive novel PRSs indexing common and disorder-specific genetic factors. Analysis took place from January 2021 to January 2022. Main Outcomes and Measures Diagnoses based on in-person structured clinical interviews were compared with ICD-9/10 billing codes. PRSs were constructed using summary statistics from genome-wide association studies of schizophrenia, bipolar disorder, and major depression. Results Of 707 299 enrolled study participants, 459 667 were genotyped at the time of writing; 84 806 were of broadly African ancestry (mean [SD] age, 58 [12.1] years) and 314 909 were of broadly European ancestry (mean [SD] age, 66.4 [13.5] years). Among 9378 individuals with confirmed diagnoses of schizophrenia or bipolar 1 disorder, 8962 (95.6%) were correctly identified using ICD-9/10 codes (2 or more). Among those of European ancestry, PRSs were robustly associated with having received a diagnosis of schizophrenia (odds ratio [OR], 1.81 [95% CI, 1.76-1.87]; P < 10-257) or bipolar disorder (OR, 1.42 [95% CI, 1.39-1.44]; P < 10-295). Corresponding effect sizes in participants of African ancestry were considerably smaller for schizophrenia (OR, 1.35 [95% CI, 1.29-1.42]; P < 10-38) and bipolar disorder (OR, 1.16 [95% CI, 1.11-1.12]; P < 10-10). Neuropsychiatric PRSs were associated with increased risk for a range of psychiatric and physical health problems. Conclusions and Relevance Using diagnoses confirmed by in-person structured clinical interviews and current neuropsychiatric PRSs, the validity of an electronic health records-based phenotyping approach in US veterans was demonstrated, highlighting the potential of PRSs for disentangling biological and mediated pleiotropy.
Collapse
Affiliation(s)
- Tim B. Bigdeli
- VA New York Harbor Healthcare System, Brooklyn
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, New York
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York
| | - Georgios Voloudakis
- James J. Peters Veterans Affairs Medical Center, Bronx, New York
- Departments of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York
- Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Peter B. Barr
- VA New York Harbor Healthcare System, Brooklyn
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, New York
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York
| | - Bryan R. Gorman
- Massachusetts Area Veterans Epidemiology, Research, and Information Center (MAVERIC), Jamaica Plain
| | - Giulio Genovese
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Roseann E. Peterson
- VA New York Harbor Healthcare System, Brooklyn
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, New York
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York
- Department of Psychiatry, Virginia Commonwealth University, Richmond
| | - David E. Burstein
- James J. Peters Veterans Affairs Medical Center, Bronx, New York
- Departments of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York
- Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Vlad I. Velicu
- James J. Peters Veterans Affairs Medical Center, Bronx, New York
- Departments of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York
- Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yuli Li
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut
- Yale University School of Medicine, New Haven, Connecticut
| | - Rishab Gupta
- Department of Psychiatry, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Manuel Mattheisen
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Community Health, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Epidemiology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Simone Tomasi
- James J. Peters Veterans Affairs Medical Center, Bronx, New York
- Departments of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York
- Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nallakkandi Rajeevan
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut
- Yale University School of Medicine, New Haven, Connecticut
| | - Frederick Sayward
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut
- Yale University School of Medicine, New Haven, Connecticut
| | - Krishnan Radhakrishnan
- National Mental Health and Substance Use Policy Laboratory, Substance Abuse and Mental Health Services Administration, Rockville, Maryland
| | | | - Anil K. Malhotra
- Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, New York
- Division of Psychiatry Research, The Zucker Hillside Hospital, Northwell Health, Glen Oaks, New York
- Department of Psychiatry, Hofstra Northwell School of Medicine, Hempstead, New York
| | - Yunling Shi
- Massachusetts Area Veterans Epidemiology, Research, and Information Center (MAVERIC), Jamaica Plain
| | - Hongyu Zhao
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut
- Yale University School of Medicine, New Haven, Connecticut
| | - Thomas R. Kosten
- Michael E. DeBakey VA Medical Center, Houston, Texas
- Department of Psychiatry, Neuroscience, Pharmacology, and Immunology and Rheumatology, Baylor College of Medicine, Houston, Texas
| | - John Concato
- Yale University School of Medicine, New Haven, Connecticut
- Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Timothy J. O’Leary
- Office of Research and Development, Veterans Health Administration, Washington, DC
| | - Ronald Przygodzki
- Office of Research and Development, Veterans Health Administration, Washington, DC
| | - Theresa Gleason
- Office of Research and Development, Veterans Health Administration, Washington, DC
| | - Saiju Pyarajan
- Massachusetts Area Veterans Epidemiology, Research, and Information Center (MAVERIC), Jamaica Plain
| | - Mary Brophy
- Massachusetts Area Veterans Epidemiology, Research, and Information Center (MAVERIC), Jamaica Plain
- Boston University School of Medicine, Boston, Massachusetts
| | - Grant D. Huang
- Office of Research and Development, Veterans Health Administration, Washington, DC
| | - Sumitra Muralidhar
- Office of Research and Development, Veterans Health Administration, Washington, DC
| | - J. Michael Gaziano
- Massachusetts Area Veterans Epidemiology, Research, and Information Center (MAVERIC), Jamaica Plain
- Harvard Medical School, Boston, Massachusetts
| | - Mihaela Aslan
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut
- Yale University School of Medicine, New Haven, Connecticut
| | - Ayman H. Fanous
- Department of Psychiatry, University of Arizona College of Medicine-Phoenix, Phoenix
- Carl T. Hayden Veterans Affairs Medical Center, Phoenix, Arizona
| | - Philip D. Harvey
- Bruce W. Carter Miami Veterans Affairs (VA) Medical Center, Miami, Florida
- University of Miami Miller School of Medicine, Miami, Florida
| | - Panos Roussos
- James J. Peters Veterans Affairs Medical Center, Bronx, New York
- Departments of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York
- Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
40
|
Yang Q, Li Y, Li B, Gong Y. A novel multi-class classification model for schizophrenia, bipolar disorder and healthy controls using comprehensive transcriptomic data. Comput Biol Med 2022; 148:105956. [PMID: 35981456 DOI: 10.1016/j.compbiomed.2022.105956] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/30/2022] [Accepted: 08/06/2022] [Indexed: 01/01/2023]
Abstract
Two common psychiatric disorders, schizophrenia (SCZ) and bipolar disorder (BP), confer lifelong disability and collectively affect 2% of the world population. Because the diagnosis of psychiatry is based only on symptoms, developing more effective methods for the diagnosis of psychiatric disorders is a major international public health priority. Furthermore, SCZ and BP overlap considerably in terms of symptoms and risk genes. Therefore, the clarity of the underlying etiology and pathology remains lacking for these two disorders. Although many studies have been conducted, a classification model with higher accuracy and consistency was found to still be necessary for accurate diagnoses of SCZ and BP. In this study, a comprehensive dataset was combined from five independent transcriptomic studies. This dataset comprised 120 patients with SCZ, 101 patients with BP, and 149 healthy subjects. The partial least squares discriminant analysis (PLS-DA) method was applied to identify the gene signature among multiple groups, and 341 differentially expressed genes (DEGs) were identified. Then, the disease relevance of these DEGs was systematically performed, including (α) the great disease relevance of the identified signature, (β) the hub genes of the protein-protein interaction network playing a key role in psychiatric disorders, and (γ) gene ontology terms and enriched pathways playing a key role in psychiatric disorders. Finally, a popular multi-class classifier, support vector machine (SVM), was applied to construct a novel multi-class classification model using the identified signature for SCZ and BP. Using the independent test sets, the classification capacity of this multi-class model was assessed, which showed this model had a strong classification ability.
Collapse
Affiliation(s)
- Qingxia Yang
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China.
| | - Yi Li
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Bo Li
- College of Life Sciences, Chongqing Normal University, Chongqing, Chongqing, 401331, China
| | - Yaguo Gong
- School of Pharmacy, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
41
|
Kolarcik CL, Bledsoe MJ, O'Leary TJ. Returning Individual Research Results to Vulnerable Individuals. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1218-1229. [PMID: 35750259 DOI: 10.1016/j.ajpath.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
Although issues associated with returning individual research results to study participants have been well explored, these issues have been less thoroughly investigated in vulnerable individuals and populations. Considerations regarding return of research results to these individuals and populations, including how best to ensure truly informed consent, how to minimize the risks and benefits of the return of research results, and how best to ensure justice may differ from those of the population at large. This article discusses the issues and challenges associated with the return of individual research results (such as genomic, proteomic, or other biomarker data) to potentially vulnerable individuals and populations, including those who may be vulnerable for cognitive, communicative, institutional, social, deferential, medical, economic, or social reasons. It explores factors that should be considered in the design, conduct, and oversight of ethically responsible research involving the return of research results to vulnerable individuals and populations and discuss recommendations for those engaged in this work.
Collapse
Affiliation(s)
- Christi L Kolarcik
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Timothy J O'Leary
- Office of Research and Development, Veterans Health Administration, Washington, District of Columbia; Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
42
|
Wainberg M, Jacobs GR, Voineskos AN, Tripathy SJ. Neurobiological, familial and genetic risk factors for dimensional psychopathology in the Adolescent Brain Cognitive Development study. Mol Psychiatry 2022; 27:2731-2741. [PMID: 35361904 DOI: 10.1038/s41380-022-01522-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/27/2022] [Accepted: 03/10/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Adolescence is a key period for brain development and the emergence of psychopathology. The Adolescent Brain Cognitive Development (ABCD) study was created to study the biopsychosocial factors underlying healthy and pathological brain development during this period, and comprises the world's largest youth cohort with neuroimaging, family history and genetic data. METHODS We examined 9856 unrelated 9-to-10-year-old participants in the ABCD study drawn from 21 sites across the United States, of which 7662 had multimodal magnetic resonance imaging scans passing quality control, and 4447 were non-Hispanic white and used for polygenic risk score analyses. Using data available at baseline, we associated eight 'syndrome scale scores' from the Child Behavior Checklist-summarizing anxious/depressed symptoms, withdrawn/depressed symptoms, somatic complaints, social problems, thought problems, attention problems, rule-breaking behavior, and aggressive behavior-with resting-state functional and structural brain magnetic resonance imaging measures; eight indicators of family history of psychopathology; and polygenic risk scores for major depression, bipolar disorder, schizophrenia, attention deficit hyperactivity disorder (ADHD) and anorexia nervosa. As a sensitivity analysis, we excluded participants with clinically significant (>97th percentile) or borderline (93rd-97th percentile) scores for each dimension. RESULTS Most Child Behavior Checklist dimensions were associated with reduced functional connectivity within one or more of four large-scale brain networks-default mode, cingulo-parietal, dorsal attention, and retrosplenial-temporal. Several dimensions were also associated with increased functional connectivity between the default mode, dorsal attention, ventral attention and cingulo-opercular networks. Conversely, almost no global or regional brain structural measures were associated with any of the dimensions. Every family history indicator was associated with every dimension. Major depression polygenic risk was associated with six of the eight dimensions, whereas ADHD polygenic risk was exclusively associated with attention problems and externalizing behavior (rule-breaking and aggressive behavior). Bipolar disorder, schizophrenia and anorexia nervosa polygenic risk were not associated with any of the dimensions. Many associations remained statistically significant even after excluding participants with clinically significant or borderline psychopathology, suggesting that the same risk factors that contribute to clinically significant psychopathology also contribute to continuous variation within the clinically normal range. CONCLUSIONS This study codifies neurobiological, familial and genetic risk factors for dimensional psychopathology across a population-scale cohort of community-dwelling preadolescents. Future efforts are needed to understand how these multiple modalities of risk intersect to influence trajectories of psychopathology into late adolescence and adulthood.
Collapse
Affiliation(s)
- Michael Wainberg
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Grace R Jacobs
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Aristotle N Voineskos
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Shreejoy J Tripathy
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada. .,Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada. .,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada. .,Department of Physiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
43
|
Karpov D, Golimbet V. Cellular and supracellular models in the study of molecular mechanisms associated with schizophrenia. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:46-50. [DOI: 10.17116/jnevro202212211146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
44
|
Daskalakis NP, Schultz LM, Visoki E, Moore TM, Argabright ST, Harnett NG, DiDomenico GE, Warrier V, Almasy L, Barzilay R. Contributions of PTSD polygenic risk and environmental stress to suicidality in preadolescents. Neurobiol Stress 2021; 15:100411. [PMID: 34765698 PMCID: PMC8569631 DOI: 10.1016/j.ynstr.2021.100411] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 09/17/2021] [Accepted: 10/23/2021] [Indexed: 11/19/2022] Open
Abstract
Suicidal ideation and attempts (i.e., suicidality) are complex behaviors driven by environmental stress, genetic susceptibility, and their interaction. Preadolescent suicidality is a major health problem with rising rates, yet its underlying biology is understudied. Here we studied effects of genetic stress susceptibility, approximated by the polygenic risk score (PRS) for post-traumatic-stress-disorder (PTSD), on preadolescent suicidality in participants from the Adolescent Brain Cognitive Development (ABCD) Study®. We further evaluated PTSD-PRS effects on suicidality in the presence of environmental stressors that are established suicide risk factors. Analyses included both European and African ancestry participants using PRS calculated based on summary statistics from ancestry-specific genome-wide association studies. In European ancestry participants (N = 4,619, n = 378 suicidal), PTSD-PRS was associated with preadolescent suicidality (odds ratio [OR] = 1.12, 95%CI 1-1.25, p = 0.038). Results in African ancestry participants (N = 1,334, n = 130 suicidal) showed a similar direction but were not statistically significant (OR = 1.21, 95%CI 0.93-1.57, p = 0.153). Sensitivity analyses using non-psychiatric polygenic score for height and using cross-ancestry PTSD-PRS did not reveal any association with suicidality, supporting the specificity of the association of ancestry-specific PTSD-PRS with suicidality. Environmental stressors were robustly associated with suicidality across ancestries with moderate effect size for negative life events and family conflict (OR 1.27-1.6); and with large effect size (OR ∼ 4) for sexual-orientation discrimination. When combined with environmental factors, PTSD-PRS showed marginal additive effects in explaining variability in suicidality, with no evidence for G × E interaction. Results support use of cross-phenotype PRS, specifically stress-susceptibility, as a genetic marker for suicidality risk early in the lifespan.
Collapse
Affiliation(s)
- Nikolaos P. Daskalakis
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Laura M. Schultz
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
- Lifespan Brain Institute, CHOP and Penn Medicine, Philadelphia, PA, USA
| | - Elina Visoki
- Lifespan Brain Institute, CHOP and Penn Medicine, Philadelphia, PA, USA
| | - Tyler M. Moore
- Lifespan Brain Institute, CHOP and Penn Medicine, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Nathaniel G. Harnett
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | | | - Varun Warrier
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Laura Almasy
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
- Lifespan Brain Institute, CHOP and Penn Medicine, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ran Barzilay
- Lifespan Brain Institute, CHOP and Penn Medicine, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Child and Adolescent Psychiatry and Behavioral Sciences, CHOP, Philadelphia, PA, USA
| |
Collapse
|
45
|
Lancaster TM, Dimitriadis SI, Perry G, Zammit S, O’Donovan MC, Linden DE. Morphometric Analysis of Structural MRI Using Schizophrenia Meta-analytic Priors Distinguish Patients from Controls in Two Independent Samples and in a Sample of Individuals With High Polygenic Risk. Schizophr Bull 2021; 48:524-532. [PMID: 34662406 PMCID: PMC8886591 DOI: 10.1093/schbul/sbab125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Schizophrenia (SCZ) is associated with structural brain changes, with considerable variation in the extent to which these cortical regions are influenced. We present a novel metric that summarises individual structural variation across the brain, while considering prior effect sizes, established via meta-analysis. We determine individual participant deviation from a within-sample-norm across structural MRI regions of interest (ROIs). For each participant, we weight the normalised deviation of each ROI by the effect size (Cohen's d) of the difference between SCZ/control for the corresponding ROI from the SCZ Enhancing Neuroimaging Genomics through Meta-Analysis working group. We generate a morphometric risk score (MRS) representing the average of these weighted deviations. We investigate if SCZ-MRS is elevated in a SCZ case/control sample (NCASE = 50; NCONTROL = 125), a replication sample (NCASE = 23; NCONTROL = 20) and a sample of asymptomatic young adults with extreme SCZ polygenic risk (NHIGH-SCZ-PRS = 95; NLOW-SCZ-PRS = 94). SCZ cases had higher SCZ-MRS than healthy controls in both samples (Study 1: β = 0.62, P < 0.001; Study 2: β = 0.81, P = 0.018). The high liability SCZ-PRS group also had a higher SCZ-MRS (Study 3: β = 0.29, P = 0.044). Furthermore, the SCZ-MRS was uniquely associated with SCZ status, but not attention-deficit hyperactivity disorder (ADHD), whereas an ADHD-MRS was linked to ADHD status, but not SCZ. This approach provides a promising solution when considering individual heterogeneity in SCZ-related brain alterations by identifying individual's patterns of structural brain-wide alterations.
Collapse
Affiliation(s)
- Thomas M Lancaster
- Department of Psychology, Bath University, Bath, UK,Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK,To whom correspondence should be addressed; Department of Psychology, Bath University, Bath, UK, tel.: +44-1225-384658, e-mail:
| | - Stavros I Dimitriadis
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK,MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff School of Medicine, Cardiff University, Cardiff, UK
| | - Gavin Perry
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| | - Stan Zammit
- MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff School of Medicine, Cardiff University, Cardiff, UK,Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Michael C O’Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff School of Medicine, Cardiff University, Cardiff, UK,Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
| | - David E Linden
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK,MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff School of Medicine, Cardiff University, Cardiff, UK,Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK,School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
46
|
Altaf-Ul-Amin M, Hirose K, Nani JV, Porta LC, Tasic L, Hossain SF, Huang M, Ono N, Hayashi MAF, Kanaya S. A system biology approach based on metabolic biomarkers and protein-protein interactions for identifying pathways underlying schizophrenia and bipolar disorder. Sci Rep 2021; 11:14450. [PMID: 34262063 PMCID: PMC8280132 DOI: 10.1038/s41598-021-93653-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 06/28/2021] [Indexed: 11/10/2022] Open
Abstract
Mental disorders (MDs), including schizophrenia (SCZ) and bipolar disorder (BD), have attracted special attention from scientists due to their high prevalence and significantly debilitating clinical features. The diagnosis of MDs is still essentially based on clinical interviews, and intensive efforts to introduce biochemical based diagnostic methods have faced several difficulties for implementation in clinics, due to the complexity and still limited knowledge in MDs. In this context, aiming for improving the knowledge in etiology and pathophysiology, many authors have reported several alterations in metabolites in MDs and other brain diseases. After potentially fishing all metabolite biomarkers reported up to now for SCZ and BD, we investigated here the proteins related to these metabolites in order to construct a protein-protein interaction (PPI) network associated with these diseases. We determined the statistically significant clusters in this PPI network and, based on these clusters, we identified 28 significant pathways for SCZ and BDs that essentially compose three groups representing three major systems, namely stress response, energy and neuron systems. By characterizing new pathways with potential to innovate the diagnosis and treatment of psychiatric diseases, the present data may also contribute to the proposal of new intervention for the treatment of still unmet aspects in MDs.
Collapse
Affiliation(s)
- Md Altaf-Ul-Amin
- Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan.
| | - Kazuhisa Hirose
- Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - João V Nani
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
- National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil
| | - Lucas C Porta
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Ljubica Tasic
- Chemical Biology Laboratory, Department of Organic Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (Unicamp), Campinas, SP, Brazil
| | | | - Ming Huang
- Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Naoaki Ono
- Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Mirian A F Hayashi
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
- National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil.
| | - Shigehiko Kanaya
- Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| |
Collapse
|
47
|
Harvey PD, Bigdeli TB, Fanous AH, Li Y, Rajeevan N, Sayward F, Radhakrishnan K, Huang G, Aslan M. Cooperative Studies Program (CSP) #572: A Study of Serious Mental Illness in Veterans as a Pathway to personalized medicine in Schizophrenia and Bipolar Illness. PERSONALIZED MEDICINE IN PSYCHIATRY 2021; 27-28:10.1016/j.pmip.2021.100078. [PMID: 34222732 PMCID: PMC8247126 DOI: 10.1016/j.pmip.2021.100078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Personalization of psychiatric treatment includes treatment of symptoms, cognition and functional deficits, suicide, and medical co-morbidities. VA Collaborative Study 572 examined a large sample of male and female veterans with schizophrenia (n=3,942) and with bipolar disorder (n=5,414) with phenotyping and genomic analyses. We present the results to date and future directions. METHODS All veterans received a structured diagnostic interview and assessments of suicidal ideation and behavior, PTSD, and health. Veterans with schizophrenia were assessed for negative symptoms and lifetime depression. All were assessed with a cognitive and functional capacity assessment. Data for genome wide association studies were collected. Controls came from the VA Million Veteran Program. RESULTS Suicidal ideation or behavior was present in 66%. Cognitive and functional deficits were consistent with previous studies. 40% of the veterans with schizophrenia had a lifetime major depressive episode and PTSD was present in over 30%. Polygenic risk score (PRS) analyses indicated that cognitive and functional deficits overlapped with PRS for cognition, education, and intelligence in the general population and PRS for suicidal ideation and behavior correlated with previous PRS for depression and suicidal ideation and behavior, as did the PRS for PTSD. DISCUSSION Results to date provide directions for personalization of treatment in SMI, veterans with SMI, and veterans in general. The results of the genomic analyses suggest that cognitive deficits in SMI may be associated with general population features. Upcoming genomic analyses will reexamine the issues above, as well as genomic factors associated with smoking, substance abuse, negative symptoms, and treatment response.
Collapse
Affiliation(s)
- Philip D. Harvey
- Bruce W. Carter Miami Veterans Affairs (VA) Medical Center, Miami, FL
- University of Miami School of Medicine, Miami, FL
| | - Tim B. Bigdeli
- VA New York Harbor Healthcare System, Brooklyn, NY
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Medical Center, Brooklyn, NY
| | - Ayman H. Fanous
- VA New York Harbor Healthcare System, Brooklyn, NY
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Medical Center, Brooklyn, NY
| | - Yuli Li
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - Nallakkandi Rajeevan
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - Frederick Sayward
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - Krishnan Radhakrishnan
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Center for Behavioral Health Statistics and Quality, Substance Abuse and Mental Health Services Administration
- University of Kentucky School of Medicine, Lexington, KY
| | - Grant Huang
- Office of Research and Development, Veterans Health Administration, Washington, DC
| | - Mihaela Aslan
- VA New York Harbor Healthcare System, Brooklyn, NY
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Medical Center, Brooklyn, NY
| |
Collapse
|