1
|
Ebrahim N, Kondratyev N, Artyuhov A, Timofeev A, Gurskaya N, Andrianov A, Izrailov R, Volchkov E, Dyuzheva T, Kopantseva E, Kiseleva E, Golimbet V, Dashinimaev E. Human pancreatic islet-derived stromal cells reveal combined features of mesenchymal stromal cells and pancreatic stellate cells. Stem Cell Res Ther 2024; 15:351. [PMID: 39380125 PMCID: PMC11463112 DOI: 10.1186/s13287-024-03963-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) are recognized for their potential in regenerative medicine, attributed to their multipotent differentiation capabilities and immunomodulatory properties. Despite this potential, the classification and detailed characterization of MSCs, especially those derived from specific tissues like the pancreas, remains challenging leading to a proliferation of terminology in the literature. This study aims to address these challenges by providing a thorough characterization of human pancreatic islets-derived mesenchymal stromal cells (hPD-MSCs). METHODS hPD-MSCs were isolated from donor islets using enzymatic digestion, immortalized through lentiviral transduction of human telomerase reverse transcriptase (hTERT). Cells were characterized by immunostaining, flow cytometry and multilineage differentiation potential into adipogenic and osteogenic lineages. Further a transcriptomic analysis was done to compare the gene expression profiles of hPD-MSCs with other mesenchymal cells. RESULTS We show that hPD-MSCs express the classical MSC features, including morphological characteristics, surface markers expression (CD90, CD73, CD105, CD44, and CD106) and the ability to differentiate into both adipogenic and osteogenic lineages. Furthermore, transcriptomic analysis revealed distinct gene expression profiles, showing notable similarities between hPD-MSCs and pancreatic stellate cells (PSCs). The study also identified specific genes that distinguish hPD-MSCs from MSCs of other origins, including genes associated with pancreatic function (e.g., ISL1) and neural development (e.g., NPTX1, ZNF804A). A novel gene with an unknown function (ENSG00000286190) was also discovered. CONCLUSIONS This study enhances the understanding of hPD-MSCs, demonstrating their unique characteristics and potential applications in therapeutic strategies. The identification of specific gene expression profiles differentiates hPD-MSCs from other mesenchymal cells and opens new avenues for research into their role in pancreatic function and neural development.
Collapse
Affiliation(s)
- Nour Ebrahim
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia, 117997
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia, 141701
| | | | - Alexander Artyuhov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia, 117997
- Research Institute of Molecular and Cellular Medicine, RUDN University, Moscow, Russia, 117198
| | - Alexei Timofeev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia, 117997
| | - Nadya Gurskaya
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia, 117997
| | - Alexey Andrianov
- Loginov Moscow Clinical Scientific Center, Moscow, Russia, 111123
| | - Roman Izrailov
- Loginov Moscow Clinical Scientific Center, Moscow, Russia, 111123
| | - Egor Volchkov
- Research Institute of Molecular and Cellular Medicine, RUDN University, Moscow, Russia, 117198
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology (D. Rogachev, NMRCPHOI) of Ministry of Healthcare of the Russian Federation, 1, Samory Mashela St, Moscow, Russia, 117997
| | - Tatyana Dyuzheva
- Department of Hospital Surgery, Sklifosovsky Institute for Clinical Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia, 119435
| | - Elena Kopantseva
- Research Institute of Molecular and Cellular Medicine, RUDN University, Moscow, Russia, 117198
| | - Ekaterina Kiseleva
- Research Institute for Systems Biology and Medicine, Moscow, Russia, 117246
| | - Vera Golimbet
- Mental Health Research Center, Moscow, Russia, 115522
| | - Erdem Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia, 117997.
- Research Institute of Molecular and Cellular Medicine, RUDN University, Moscow, Russia, 117198.
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia, 141701.
- Institute of Medicine, Banzarov Buryat State University, Ulan-Ude, Russia, 670000.
| |
Collapse
|
2
|
Hu YQ, Liu WT, Wu Y, Hu ZB, Tao YC, Zhang Q, Chen JY, Li M, Hu L, Ding YQ. DCC in the cerebral cortex is required for cognitive functions in mouse. Brain Pathol 2024:e13306. [PMID: 39293934 DOI: 10.1111/bpa.13306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/26/2024] [Indexed: 09/20/2024] Open
Abstract
Schizophrenia (SZ) is a highly heritable mental disorder, and genome-wide association studies have identified the association between deleted in colorectal cancer (DCC) and SZ. Previous study has shown a lowered expression of DCC in the cerebral cortex of SZ patient. In this study, we identified novel single nucleotide polymorphisms (SNPs) of DCC statistically correlated with SZ. Based on these, we generated DCC conditional knockout (CKO) mice and explored behavioral phenotypes in these mice. We observed that deletion of DCC in cortical layer VI but not layer V led to deficits in fear and spatial memory, as well as defective sensorimotor gating revealed by the prepulse inhibition test (PPI). Critically, the defective sensorimotor gating could be restored by olanzapine, an antipsychotic drug. Furthermore, we found that the levels of p-AKT and p-GSK3α/β were decreased, which was responsible for impaired PPI in the DCC-deficient mice. Finally, the DCC-deficient mice also displayed reduced spine density of pyramidal neurons and disturbed delta-oscillations. Our data, for the first time, identified and explored downstream substrates and signaling pathway of DCC which supports the hypothesis that DCC is a SZ-related risky gene and when defective, may promote SZ-like pathogenesis and behavioral phenotypes in mice.
Collapse
Affiliation(s)
- Yun-Qing Hu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Wei-Tang Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Yong Wu
- Research Center for Mental Health and Neuroscience, Wuhan Mental Health Center, Jianghan University, Wuhan, China
| | - Zhi-Bin Hu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yun-Chao Tao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Qiong Zhang
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Jia-Yin Chen
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Ling Hu
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Yu-Qiang Ding
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Ziemka-Nalecz M, Pawelec P, Ziabska K, Zalewska T. Sex Differences in Brain Disorders. Int J Mol Sci 2023; 24:14571. [PMID: 37834018 PMCID: PMC10572175 DOI: 10.3390/ijms241914571] [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: 09/06/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
A remarkable feature of the brain is its sexual dimorphism. Sexual dimorphism in brain structure and function is associated with clinical implications documented previously in healthy individuals but also in those who suffer from various brain disorders. Sex-based differences concerning some features such as the risk, prevalence, age of onset, and symptomatology have been confirmed in a range of neurological and neuropsychiatric diseases. The mechanisms responsible for the establishment of sex-based differences between men and women are not fully understood. The present paper provides up-to-date data on sex-related dissimilarities observed in brain disorders and highlights the most relevant features that differ between males and females. The topic is very important as the recognition of disparities between the sexes might allow for the identification of therapeutic targets and pharmacological approaches for intractable neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
| | | | | | - Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5, A. Pawinskiego Str., 02-106 Warsaw, Poland; (M.Z.-N.); (P.P.); (K.Z.)
| |
Collapse
|
4
|
Kember RL, Vickers-Smith R, Zhou H, Xu H, Jennings M, Dao C, Davis L, Sanchez-Roige S, Justice AC, Gelernter J, Vujkovic M, Kranzler HR. Genetic Underpinnings of the Transition From Alcohol Consumption to Alcohol Use Disorder: Shared and Unique Genetic Architectures in a Cross-Ancestry Sample. Am J Psychiatry 2023; 180:584-593. [PMID: 37282553 PMCID: PMC10731616 DOI: 10.1176/appi.ajp.21090892] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
OBJECTIVE Recent genome-wide association studies (GWASs) of alcohol-related phenotypes have uncovered key differences in the underlying genetic architectures of alcohol consumption and alcohol use disorder (AUD), with the two traits having opposite genetic correlations with psychiatric disorders. Understanding the genetic factors that underlie the transition from heavy drinking to AUD has important theoretical and clinical implications. METHODS The authors used longitudinal data from the cross-ancestry Million Veteran Program sample to identify 1) novel loci associated with AUD and alcohol consumption (measured by the score on the consumption subscale of the Alcohol Use Disorders Identification Test [AUDIT-C]), 2) the impact of phenotypic variation on genetic discovery, and 3) genetic variants with direct effects on AUD that are not mediated through alcohol consumption. RESULTS The authors identified 26 loci associated with AUD and 22 loci associated with AUDIT-C score, including ancestry-specific and novel loci. In secondary GWASs that excluded individuals who report abstinence, the authors identified seven additional loci for AUD and eight additional loci for AUDIT-C score. Although the heterogeneity of the abstinent group biases the GWAS findings, unique variance between alcohol consumption and disorder remained after the abstinent group was excluded. Finally, using mediation analysis, the authors identified a set of variants with effects on AUD that are not mediated through alcohol consumption. CONCLUSIONS Differences in genetic architecture between alcohol consumption and AUD are consistent with their having different biological contributions. Genetic variants with direct effects on AUD are potentially relevant to understanding the transition from heavy alcohol consumption to AUD and may be targets for translational prevention and treatment efforts.
Collapse
Affiliation(s)
- Rachel L Kember
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Rachel Vickers-Smith
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Hang Zhou
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Heng Xu
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Mariela Jennings
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Cecilia Dao
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Lea Davis
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Sandra Sanchez-Roige
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Amy C Justice
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Joel Gelernter
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Marijana Vujkovic
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Henry R Kranzler
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| |
Collapse
|
5
|
Maity S, Munisamy M, Sagar R, Udupa N, Puluturu Shilpa V, Subbiah V. Psychosis susceptibility zinc finger protein 804A (ZNF804A) gene polymorphism in schizophrenia patients treated with olanzapine in North Indian population. Int J Neurosci 2023; 133:467-472. [PMID: 33866919 DOI: 10.1080/00207454.2021.1916742] [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: 10/21/2022]
Abstract
PURPOSE The Zinc finger protein 804A (ZNF804A) is a potential schizophrenia candidate gene that has emerged from genome-wide association studies. The aim of the study is to investigate whether this gene variant influences the response of positive or negative symptoms to antipsychotic drug olanzapine in North Indian schizophrenia patients. MATERIALS AND METHODS Our study involved 184 unrelated schizophrenia cases (114 males and 70 females; mean age: 52.8 ± 11.6 years) and 300 normal controls (168 males and 132 females; mean age: 54.9 ± 6.9 years). At the start of treatment and after four weeks, we assessed the response of positive and negative symptoms by positive and negative syndrome scale (PANSS). Olanzapine drug level was estimated using HPLC Method and Genotyping was performed using PCR-Snap Shot technique. RESULTS Significant differences were observed in the genotype distribution (χ2 = 6.10, d.f. = 2, p = 0.04) and allele frequencies (χ2 = 5.14, d.f. = 1, p = 0.02; odds ratio = 0.57, 95% confidence interval =1.09-3.48) between schizophrenia patients and controls group. The improvement of positive and negative schizophrenia symptoms after 4 weeks of treatment with olanzapine was assessed. Patients homozygous for the ZNF804A risk allele for AA show poorer improvement of positive symptoms compared to patients with a protective allele. CONCLUSIONS Our findings indicate that ZNF804A gene polymorphism plays a significant role in the treatment of schizophrenia, suggesting that ZNF804A may be an effective marker for schizophrenia treatment.
Collapse
Affiliation(s)
- Sayantika Maity
- Department of Neurobiochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Murali Munisamy
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal, Karnataka, India
| | - Rajesh Sagar
- Department of Psychiatry, All India Institute of Medical Sciences, New Delhi, India
| | - Nayanabhirama Udupa
- Directorate of Research, Department of Pharmacy Management, Manipal College of Pharmaceutical Sciences, Manipal, India
| | | | - Vivekanandhan Subbiah
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh, India
| |
Collapse
|
6
|
Ding YC, Adamson AW, Bakhtiari M, Patrick C, Park J, Laitman Y, Weitzel JN, Bafna V, Friedman E, Neuhausen SL. Variable number tandem repeats (VNTRs) as modifiers of breast cancer risk in carriers of BRCA1 185delAG. Eur J Hum Genet 2023; 31:216-222. [PMID: 36434258 PMCID: PMC9905572 DOI: 10.1038/s41431-022-01238-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 10/10/2022] [Accepted: 11/08/2022] [Indexed: 11/27/2022] Open
Abstract
Despite substantial efforts in identifying both rare and common variants affecting disease risk, in the majority of diseases, a large proportion of unexplained genetic risk remains. We propose that variable number tandem repeats (VNTRs) may explain a proportion of the missing genetic risk. Herein, in a pilot study with a retrospective cohort design, we tested whether VNTRs are causal modifiers of breast cancer risk in 347 female carriers of the BRCA1 185delAG pathogenic variant, an important group given their high risk of developing breast cancer. We performed targeted-capture to sequence VNTRs, called genotypes with adVNTR, tested the association of VNTRs and breast cancer risk using Cox regression models, and estimated the effect size using a retrospective likelihood approach. Of 303 VNTRs that passed quality control checks, 4 VNTRs were significantly associated with risk to develop breast cancer at false discovery rate [FDR] < 0.05 and an additional 4 VNTRs had FDR < 0.25. After determining the specific risk alleles, there was a significantly earlier age at diagnosis of breast cancer in carriers of the risk alleles compared to those without the risk alleles for seven of eight VNTRs. One example is a VNTR in exon 2 of LINC01973 with a per-allele hazard ratio of 1.58 (1.07-2.33) and 5.28 (2.79-9.99) for the homozygous risk-allele genotype. Results from this first systematic study of VNTRs demonstrate that VNTRs may explain a proportion of the unexplained genetic risk for breast cancer.
Collapse
Affiliation(s)
- Yuan Chun Ding
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Aaron W Adamson
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Mehrdad Bakhtiari
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Carmina Patrick
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Jonghun Park
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Yael Laitman
- Oncogenetics Unit, Institute of Human Genetics, Sheba Medical Center, Ramat Gan, Israel
| | - Jeffrey N Weitzel
- Latin American School of Oncology, Tuxla Gutierrez, Chiapas, MX and Natera, San Carlos, CA, USA
| | - Vineet Bafna
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Eitan Friedman
- Oncogenetics Unit, Institute of Human Genetics, Sheba Medical Center, Ramat Gan, Israel
- The Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Center for Preventive Personalized Medicine, Assuta Medical Center, Tel Aviv, Israel
| | - Susan L Neuhausen
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA, USA.
| |
Collapse
|
7
|
Hegarty CE, Ianni AM, Kohn PD, Kolachana B, Gregory M, Masdeu JC, Eisenberg DP, Berman KF. Polymorphism in the ZNF804A Gene and Variation in D 1 and D 2/D 3 Dopamine Receptor Availability in the Healthy Human Brain: A Dual Positron Emission Tomography Study. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2023; 8:121-128. [PMID: 33712377 PMCID: PMC10501410 DOI: 10.1016/j.bpsc.2020.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/03/2020] [Accepted: 12/15/2020] [Indexed: 01/11/2023]
Abstract
BACKGROUND The rs1344706 single nucleotide polymorphism in the ZNF804A gene has been associated with risk for psychosis in multiple genome-wide association studies, yet mechanisms underlying this association are not known. Given preclinical work suggesting an impact of ZNF804A on dopamine receptor gene transcription and clinical studies establishing dopaminergic dysfunction in patients with schizophrenia, we hypothesized that the ZNF804A risk single nucleotide polymorphism would be associated with variation in dopamine receptor availability in the human brain. METHODS In this study, 72 healthy individuals genotyped for rs1344706 completed both [18F]fallypride and [11C]NNC-112 positron emission tomography scans to measure D2/D3 and D1 receptor availability, respectively. Genetic effects on estimates of binding potential for each ligand were tested first with canonical subject-specific striatal regions of interest analyses, followed by exploratory whole-brain voxelwise analyses to test for more localized striatal signals and for extrastriatal effects. RESULTS Region of interest analyses revealed significantly less D2/D3 receptor availability in risk-allele homozygotes (TT) compared with non-risk allele carriers (G-allele carrier group: TG and GG) in the associative striatum and sensorimotor striatum, but no significant differences in striatal D1 receptor availability. CONCLUSIONS These data suggest that ZNF804A genotype may be meaningfully linked to dopaminergic function in the human brain. The results also may provide information to guide future studies of ZNF804A-related mechanisms of schizophrenia risk.
Collapse
Affiliation(s)
- Catherine E Hegarty
- Clinical and Translational Neuroscience Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland; Neuroscience Graduate Program, Brown University, Providence, Rhode Island
| | - Angela M Ianni
- Clinical and Translational Neuroscience Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Philip D Kohn
- Clinical and Translational Neuroscience Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Bhaskar Kolachana
- Human Brain Collection Core, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Michael Gregory
- Clinical and Translational Neuroscience Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Joseph C Masdeu
- Clinical and Translational Neuroscience Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Daniel P Eisenberg
- Clinical and Translational Neuroscience Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Karen F Berman
- Clinical and Translational Neuroscience Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland.
| |
Collapse
|
8
|
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
|
9
|
Vouga Ribeiro N, Tavares V, Bramon E, Toulopoulou T, Valli I, Shergill S, Murray R, Prata D. Effects of psychosis-associated genetic markers on brain volumetry: a systematic review of replicated findings and an independent validation. Psychol Med 2022; 52:1-16. [PMID: 36168994 PMCID: PMC9811278 DOI: 10.1017/s0033291722002896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 08/13/2022] [Accepted: 08/24/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Given psychotic illnesses' high heritability and associations with brain structure, numerous neuroimaging-genetics findings have been reported in the last two decades. However, few findings have been replicated. In the present independent sample we aimed to replicate any psychosis-implicated SNPs (single nucleotide polymorphisms), which had previously shown at least two main effects on brain volume. METHODS A systematic review for SNPs showing a replicated effect on brain volume yielded 25 studies implicating seven SNPs in five genes. Their effect was then tested in 113 subjects with either schizophrenia, bipolar disorder, 'at risk mental state' or healthy state, for whole-brain and region-of-interest (ROI) associations with grey and white matter volume changes, using voxel-based morphometry. RESULTS We found FWER-corrected (Family-wise error rate) (i.e. statistically significant) associations of: (1) CACNA1C-rs769087-A with larger bilateral hippocampus and thalamus white matter, across the whole brain; and (2) CACNA1C-rs769087-A with larger superior frontal gyrus, as ROI. Higher replication concordance with existing literature was found, in decreasing order, for: (1) CACNA1C-rs769087-A, with larger dorsolateral-prefrontal/superior frontal gyrus and hippocampi (both with anatomical and directional concordance); (2) ZNF804A-rs11681373-A, with smaller angular gyrus grey matter and rectus gyri white matter (both with anatomical and directional concordance); and (3) BDNF-rs6265-T with superior frontal and middle cingulate gyri volume change (with anatomical and allelic concordance). CONCLUSIONS Most literature findings were not herein replicated. Nevertheless, high degree/likelihood of replication was found for two genome-wide association studies- and one candidate-implicated SNPs, supporting their involvement in psychosis and brain structure.
Collapse
Affiliation(s)
- Nuno Vouga Ribeiro
- Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Vânia Tavares
- Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Elvira Bramon
- Division of Psychiatry, University College London, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’ College London, London, UK
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - Timothea Toulopoulou
- Department of Psychology & National Magnetic Resonance Research Center (UMRAM), Aysel Sabuncu Brain Research Centre (ASBAM), Bilkent University, Ankara, Turkey
| | - Isabel Valli
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’ College London, London, UK
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Sukhi Shergill
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’ College London, London, UK
| | - Robin Murray
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’ College London, London, UK
| | - Diana Prata
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| |
Collapse
|
10
|
de Bartolomeis A, Barone A, Vellucci L, Mazza B, Austin MC, Iasevoli F, Ciccarelli M. Linking Inflammation, Aberrant Glutamate-Dopamine Interaction, and Post-synaptic Changes: Translational Relevance for Schizophrenia and Antipsychotic Treatment: a Systematic Review. Mol Neurobiol 2022; 59:6460-6501. [PMID: 35963926 PMCID: PMC9463235 DOI: 10.1007/s12035-022-02976-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/24/2022] [Indexed: 12/16/2022]
Abstract
Evidence from clinical, preclinical, and post-mortem studies supports the inflammatory/immune hypothesis of schizophrenia pathogenesis. Less evident is the link between the inflammatory background and two well-recognized functional and structural findings of schizophrenia pathophysiology: the dopamine-glutamate aberrant interaction and the alteration of dendritic spines architecture, both believed to be the “quantal” elements of cortical-subcortical dysfunctional network. In this systematic review, we tried to capture the major findings linking inflammation, aberrant glutamate-dopamine interaction, and post-synaptic changes under a direct and inverse translational perspective, a paramount picture that at present is lacking. The inflammatory effects on dopaminergic function appear to be bidirectional: the inflammation influences dopamine release, and dopamine acts as a regulator of discrete inflammatory processes involved in schizophrenia such as dysregulated interleukin and kynurenine pathways. Furthermore, the link between inflammation and glutamate is strongly supported by clinical studies aimed at exploring overactive microglia in schizophrenia patients and maternal immune activation models, indicating impaired glutamate regulation and reduced N-methyl-D-aspartate receptor (NMDAR) function. In addition, an inflammatory/immune-induced alteration of post-synaptic density scaffold proteins, crucial for downstream NMDAR signaling and synaptic efficacy, has been demonstrated. According to these findings, a significant increase in plasma inflammatory markers has been found in schizophrenia patients compared to healthy controls, associated with reduced cortical integrity and functional connectivity, relevant to the cognitive deficit of schizophrenia. Finally, the link between altered inflammatory/immune responses raises relevant questions regarding potential new therapeutic strategies specifically for those forms of schizophrenia that are resistant to canonical antipsychotics or unresponsive to clozapine.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy. .,Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy.
| | - Annarita Barone
- Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy.,Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy
| | - Licia Vellucci
- Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy.,Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy
| | - Benedetta Mazza
- Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy
| | - Mark C Austin
- Clinical Psychopharmacology Program, College of Pharmacy, Idaho State University (ISU), Pocatello, ID, USA
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy.,Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy
| | - Mariateresa Ciccarelli
- Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy.,Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy
| |
Collapse
|
11
|
Harrison PJ, Mould A, Tunbridge EM. New drug targets in psychiatry: Neurobiological considerations in the genomics era. Neurosci Biobehav Rev 2022; 139:104763. [PMID: 35787892 DOI: 10.1016/j.neubiorev.2022.104763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 05/15/2022] [Accepted: 06/14/2022] [Indexed: 01/11/2023]
Abstract
After a period of withdrawal, pharmaceutical companies have begun to reinvest in neuropsychiatric disorders, due to improvements in our understanding of these disorders, stimulated in part by genomic studies. However, translating this information into disease insights and ultimately into tractable therapeutic targets is a major challenge. Here we consider how different sources of information might be integrated to guide this process. We review how an understanding of neurobiology has been used to advance therapeutic candidates identified in the pre-genomic era, using catechol-O-methyltransferase (COMT) as an exemplar. We then contrast with ZNF804A, the first genome-wide significant schizophrenia gene, and draw on some of the lessons that these and other examples provide. We highlight that, at least in the short term, the translation of potential targets for which there is orthogonal neurobiological support is likely to be more straightforward and productive than that those relying solely on genomic information. Although we focus here on information from genomic studies of schizophrenia, the points are broadly applicable across major psychiatric disorders and their symptoms.
Collapse
Affiliation(s)
- Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health NHS Foundation Trust, Oxford, UK
| | - Arne Mould
- Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health NHS Foundation Trust, Oxford, UK
| | - Elizabeth M Tunbridge
- Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
12
|
Münch-Anguiano L, Camarena B, Nieto-Quinto J, de la Torre P, Pedro Laclette J, Hirata-Hernández H, Hernández-Muñoz S, Aguilar-García A, Becerra-Palars C, Gutiérrez-Mora D, Ortega-Ortiz H, Escamilla-Orozco R, Saracco-Álvarez R, Bustos-Jaimes I. Genetic analysis of the ZNF804A gene in Mexican patients with schizophrenia, schizoaffective disorder and bipolar disorder. Gene 2022; 829:146508. [PMID: 35447233 DOI: 10.1016/j.gene.2022.146508] [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: 07/02/2021] [Revised: 03/09/2022] [Accepted: 04/14/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Evidence suggests that schizophrenia (SCZ), schizoaffective disorder (SAD) and bipolar disorder (BPD) share genetic risk variants. ZNF804A gene has been associated with these disorders in different populations. GWAS and candidate gene studies have reported association between the rs1344706 A allele with SCZ, SAD and BPD in European and Asian populations. In Mexican patients, no studies have specifically analyzed ZNF804A gene variants with these disorders. The aim of the study was to analyze the rs1344706 and identify common and rare variants in a targeted region of the ZNF804A gene in Mexican patients with SCZ, BPD and SAD compared with a control group. METHODS We genotyped the rs1344706 in 228 Mexican patients diagnosed with SCZ, SAD and BPD, and 295 controls. Also, an additional sample of 167 patients with these disorders and 170 controls was analyzed to identify rare and common variants using the Sanger-sequence analysis of a targeted region of ZNF804A gene. RESULTS Association analysis of rs1344706 observed a higher frequency of A allele in the patients compared with the control group; however, did not show statistical differences after Bonferronís correction (χ2 = 5.3, p = 0.0208). In the sequence analysis, we did not identify rare variants; however, we identified three common variants: rs3046266, rs1366842 and rs12477430. A comparison of the three identified variants between patients and controls did not show statistical differences (p > 0.0125). Finally, haplotype analysis did not show statistical differences between SCZ, SAD and BPD and controls. CONCLUSIONS Our findings did not support the evidence suggesting that ZNF804A gene participates in the etiology of SCZ, SAD and BPD. Future studies are needed in a larger sample size to identify the effect of this gene in psychiatric disorders.
Collapse
Affiliation(s)
- Lucía Münch-Anguiano
- Dirección de Servicios Clínicos, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico; Programa de Maestría y Doctorado en Ciencias Médicas y Odontológicas de la Salud, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Beatriz Camarena
- Departamento de Farmacogenética, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico.
| | - Jesica Nieto-Quinto
- Departamento de Farmacogenética, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Patricia de la Torre
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Juan Pedro Laclette
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Harumi Hirata-Hernández
- Dirección de Servicios Clínicos, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Sandra Hernández-Muñoz
- Departamento de Farmacogenética, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Alejandro Aguilar-García
- Departamento de Farmacogenética, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Claudia Becerra-Palars
- Dirección de Servicios Clínicos, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Doris Gutiérrez-Mora
- Dirección de Servicios Clínicos, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Hiram Ortega-Ortiz
- Dirección de Servicios Clínicos, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Raúl Escamilla-Orozco
- Dirección de Servicios Clínicos, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Ricardo Saracco-Álvarez
- Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Ismael Bustos-Jaimes
- Laboratorio de Fisicoquímica e Ingeniería de Proteínas, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, Mexico
| |
Collapse
|
13
|
Dashtban S, Haj-Nasrolah-Fard F, Kosari Z, Ghamari R, Forouzesh F, Alizadeh F. ANK3 and ZNF804A intronic variants increase risk of schizophrenia in Iranian population: An association study. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
14
|
Zhang CY, Xiao X, Zhang Z, Hu Z, Li M. An alternative splicing hypothesis for neuropathology of schizophrenia: evidence from studies on historical candidate genes and multi-omics data. Mol Psychiatry 2022; 27:95-112. [PMID: 33686213 DOI: 10.1038/s41380-021-01037-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 01/08/2021] [Accepted: 01/22/2021] [Indexed: 01/31/2023]
Abstract
Alternative splicing of schizophrenia risk genes, such as DRD2, GRM3, and DISC1, has been extensively described. Nevertheless, the alternative splicing characteristics of the growing number of schizophrenia risk genes identified through genetic analyses remain relatively opaque. Recently, transcriptomic analyses in human brains based on short-read RNA-sequencing have discovered many "local splicing" events (e.g., exon skipping junctions) associated with genetic risk of schizophrenia, and further molecular characterizations have identified novel spliced isoforms, such as AS3MTd2d3 and ZNF804AE3E4. In addition, long-read sequencing analyses of schizophrenia risk genes (e.g., CACNA1C and NRXN1) have revealed multiple previously unannotated brain-abundant isoforms with therapeutic potentials, and functional analyses of KCNH2-3.1 and Ube3a1 have provided examples for investigating such spliced isoforms in vitro and in vivo. These findings suggest that alternative splicing may be an essential molecular mechanism underlying genetic risk of schizophrenia, however, the incomplete annotations of human brain transcriptomes might have limited our understanding of schizophrenia pathogenesis, and further efforts to elucidate these transcriptional characteristics are urgently needed to gain insights into the illness-correlated brain physiology and pathology as well as to translate genetic discoveries into novel therapeutic targets.
Collapse
Affiliation(s)
- Chu-Yi Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Zhuohua Zhang
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Zhonghua Hu
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China. .,Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China. .,Eye Center of Xiangya Hospital and Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, Hunan, China. .,National Clinical Research Center on Mental Disorders, Changsha, Hunan, China.
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China. .,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China. .,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
15
|
Li W, Zhang CY, Liu J, Guan F, Shao M, Zhang L, Liu Q, Yang Y, Su X, Zhang Y, Xiao X, Luo XJ, Li M, Lv L. Identification of a Risk Locus at 7p22.3 for Schizophrenia and Bipolar Disorder in East Asian Populations. Front Genet 2021; 12:789512. [PMID: 34976021 PMCID: PMC8719163 DOI: 10.3389/fgene.2021.789512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/23/2021] [Indexed: 12/03/2022] Open
Abstract
Background: Shared psychopathological features and mechanisms have been observed between schizophrenia (SZ) and bipolar disorder (BD), but their common risk genes and full genetic architectures remain to be fully characterized. The genome-wide association study (GWAS) datasets offer the opportunity to explore this scientific question using combined genetic data from enormous samples, ultimately allowing a better understanding of the onset and development of these illnesses. Methods: We have herein performed a genome-wide meta-analysis in two GWAS datasets of SZ and BD respectively (24,600 cases and 40,012 controls in total, discovery sample), followed by replication analyses in an independent sample of 4,918 SZ cases and 5,506 controls of Han Chinese origin (replication sample). The risk SNPs were then explored for their correlations with mRNA expression of nearby genes in multiple expression quantitative trait loci (eQTL) datasets. Results: The single nucleotide polymorphisms (SNPs) rs1637749 and rs3800908 at 7p22.3 region were significant in both discovery and replication samples, and exhibited genome-wide significant associations when combining all East Asian SZ and BD samples (29,518 cases and 45,518 controls). The risk SNPs were also significant in GWAS of SZ and BD among Europeans. Both risk SNPs significantly predicted lower expression of MRM2 in the whole blood and brain samples in multiple datasets, which was consistent with its reduced mRNA level in the brains of SZ patients compared with normal controls. The risk SNPs were also associated with MAD1L1 expression in the whole blood sample. Discussion: We have identified a novel genome-wide risk locus associated with SZ and BD in East Asians, adding further support for the putative common genetic risk of the two illnesses. Our study also highlights the necessity and importance of mining public datasets to explore risk genes for complex psychiatric diseases.
Collapse
Affiliation(s)
- Wenqiang Li
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Chu-Yi Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Jiewei Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Fanglin Guan
- Department of Forensic Psychiatry, School of Medicine and Forensics, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Minglong Shao
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Luwen Zhang
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Qing Liu
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Yongfeng Yang
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Xi Su
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Yan Zhang
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- *Correspondence: Ming Li, ; Luxian Lv,
| | - Luxian Lv
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
- Henan Province People’s Hospital, Zhengzhou, China
- *Correspondence: Ming Li, ; Luxian Lv,
| |
Collapse
|
16
|
Bartsch U, Corbin LJ, Hellmich C, Taylor M, Easey KE, Durant C, Marston HM, Timpson NJ, Jones MW. Schizophrenia-associated variation at ZNF804A correlates with altered experience-dependent dynamics of sleep slow waves and spindles in healthy young adults. Sleep 2021; 44:zsab191. [PMID: 34329479 PMCID: PMC8664578 DOI: 10.1093/sleep/zsab191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
The rs1344706 polymorphism in ZNF804A is robustly associated with schizophrenia and schizophrenia is, in turn, associated with abnormal non-rapid eye movement (NREM) sleep neurophysiology. To examine whether rs1344706 is associated with intermediate neurophysiological traits in the absence of disease, we assessed the relationship between genotype, sleep neurophysiology, and sleep-dependent memory consolidation in healthy participants. We recruited healthy adult males with no history of psychiatric disorder from the Avon Longitudinal Study of Parents and Children (ALSPAC) birth cohort. Participants were homozygous for either the schizophrenia-associated 'A' allele (N = 22) or the alternative 'C' allele (N = 18) at rs1344706. Actigraphy, polysomnography (PSG) and a motor sequence task (MST) were used to characterize daily activity patterns, sleep neurophysiology and sleep-dependent memory consolidation. Average MST learning and sleep-dependent performance improvements were similar across genotype groups, albeit more variable in the AA group. During sleep after learning, CC participants showed increased slow-wave (SW) and spindle amplitudes, plus augmented coupling of SW activity across recording electrodes. SW and spindles in those with the AA genotype were insensitive to learning, whilst SW coherence decreased following MST training. Accordingly, NREM neurophysiology robustly predicted the degree of overnight motor memory consolidation in CC carriers, but not in AA carriers. We describe evidence that rs1344706 polymorphism in ZNF804A is associated with changes in the coordinated neural network activity that supports offline information processing during sleep in a healthy population. These findings highlight the utility of sleep neurophysiology in mapping the impacts of schizophrenia-associated common genetic variants on neural circuit oscillations and function.
Collapse
Affiliation(s)
- Ullrich Bartsch
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
- Translational Neuroscience, Eli Lilly & Co Ltd UK, Erl Wood Manor, Windlesham, UK
- UK DRI Health Care & Technology at Imperial College London and the University of Surrey, Surrey Sleep Research Centre, University of Surrey, Clinical Research Building, Egerton Road, Guildford, Surrey, UK
| | - Laura J Corbin
- MRC Integrative Epidemiology Unit at University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Charlotte Hellmich
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Michelle Taylor
- MRC Integrative Epidemiology Unit at University of Bristol, Bristol, UK
| | - Kayleigh E Easey
- MRC Integrative Epidemiology Unit at University of Bristol, Bristol, UK
- UK Centre for Tobacco and Alcohol Studies, School of Psychological Science, University of Bristol, Bristol, UK
| | - Claire Durant
- Clinical Research and Imaging Centre (CRIC), University of Bristol, Bristol, UK
| | - Hugh M Marston
- Translational Neuroscience, Eli Lilly & Co Ltd UK, Erl Wood Manor, Windlesham, UK
- Böhringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit at University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Matthew W Jones
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| |
Collapse
|
17
|
Matveeva N, Titov B, Bazyleva E, Pevzner A, Favorova O. Towards Understanding the Genetic Nature of Vasovagal Syncope. Int J Mol Sci 2021; 22:10316. [PMID: 34638656 PMCID: PMC8508958 DOI: 10.3390/ijms221910316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 11/17/2022] Open
Abstract
Syncope, defined as a transient loss of consciousness caused by transient global cerebral hypoperfusion, affects 30-40% of humans during their lifetime. Vasovagal syncope (VVS) is the most common cause of syncope, the etiology of which is still unclear. This review summarizes data on the genetics of VVS, describing the inheritance pattern of the disorder, candidate gene association studies and genome-wide studies. According to this evidence, VVS is a complex disorder, which can be caused by the interplay between genetic factors, whose contribution varies from monogenic Mendelian inheritance to polygenic inherited predisposition, and external factors affecting the monogenic (resulting in incomplete penetrance) and polygenic syncope types.
Collapse
Affiliation(s)
- Natalia Matveeva
- National Medical Research Center for Cardiology, 121552 Moscow, Russia; (N.M.); (B.T.); (E.B.); (A.P.)
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Boris Titov
- National Medical Research Center for Cardiology, 121552 Moscow, Russia; (N.M.); (B.T.); (E.B.); (A.P.)
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Elizabeth Bazyleva
- National Medical Research Center for Cardiology, 121552 Moscow, Russia; (N.M.); (B.T.); (E.B.); (A.P.)
| | - Alexander Pevzner
- National Medical Research Center for Cardiology, 121552 Moscow, Russia; (N.M.); (B.T.); (E.B.); (A.P.)
| | - Olga Favorova
- National Medical Research Center for Cardiology, 121552 Moscow, Russia; (N.M.); (B.T.); (E.B.); (A.P.)
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
18
|
Ethnicity-dependent effects of Zinc finger 804A variant on schizophrenia: a systematic review and meta-analysis. Psychiatr Genet 2021; 31:21-28. [PMID: 33395218 DOI: 10.1097/ypg.0000000000000275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Previous studies and meta-analysis indicated that rs1344706 was associated with schizophrenia in European population, whereas the conclusions in other populations were disputed. To further explore whether the allele A of rs1344706 would increase the risk of schizophrenia in different populations and update the original meta-analysis, we conducted a systematic review and meta-analysis worldwide. METHODS A literature search was performed in PubMed, Embase, Cochrane Library, PsycINFO and Web of Science (up to 10 July 2019) according to the inclusion criteria. RESULTS A total of 27 articles were included. Our meta-analysis showed an association between rs1344706 and schizophrenia in total populations [P = 0.000; odds ratio (OR) = 1.105; 95% confidence interval (CI), 1.048-1.165], Europe population (P = 0.025; OR = 1.108; 95% CI, 1.013-1.222) and Asian population(P = 0.005; OR = 1.094; 95% CI, 1.027-1.164). CONCLUSIONS Our findings suggested that the risk of single nucleotide polymorphism rs1344706 A-allele may increase the risk of schizophrenia worldwide. Also, this ethnicity-dependent effects of ZNF804A variant on schizophrenia may be related to the opposite allele direction. But to elucidate the underlying biological mechanism, further studies with large participant populations are needed.
Collapse
|
19
|
Klockmeier K, Silva Ramos E, Raskó T, Martí Pastor A, Wanker EE. Schizophrenia risk candidate protein ZNF804A interacts with STAT2 and influences interferon-mediated gene transcription in mammalian cells. J Mol Biol 2021; 433:167184. [PMID: 34364876 DOI: 10.1016/j.jmb.2021.167184] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/23/2021] [Accepted: 07/30/2021] [Indexed: 11/29/2022]
Abstract
Previously evidence was presented that the single-nucleotide polymorphism rs1344706 located in an intronic region of the ZNF804A gene is associated with reduced transcript levels in fetal brains. This genetic variation in the gene encoding the zinc-finger protein ZNF804A is associated with schizophrenia (SZ) and bipolar disorder. Currently, the molecular and cellular function of ZNF804A is unclear. Here, we generated a high-confidence protein-protein interaction (PPI) network for ZNF804A using a combination of yeast two-hybrid and bioluminescence-based PPI detection assays, directly linking 12 proteins to the disease-associated target protein. Among the top hits was the signal transducer and activator of transcription 2 (STAT2), an interferon-regulated transcription factor. Detailed mechanistic studies revealed that STAT2 binds to the unstructured N-terminus of ZNF804A. This interaction is mediated by multiple short amino acid motifs in ZNF804A but not by the conserved C2H2 zinc-finger domain, which is also located at the N-terminus. Interestingly, investigations in HEK293 cells demonstrated that ZNF804A and STAT2 both co-translocate from the cytoplasm into the nucleus upon interferon (IFN) treatment. Furthermore, a concentration-dependent effect of ZNF804A overproduction on STAT2-mediated gene expression was observed using a luciferase reporter, which is under the control of an IFN-stimulated response element (ISRE). Together these results indicate the formation of ZNF804A:STAT2 protein complex and its translocation from the cytoplasm into the nucleus upon IFN stimulation, suggesting that it may function as a signal transducer that activates IFN-mediated gene expression programs.
Collapse
Affiliation(s)
- Konrad Klockmeier
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), L aboratory for Neuroproteomics, Berlin, Germany
| | - Eduardo Silva Ramos
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), L aboratory for Neuroproteomics, Berlin, Germany
| | - Tamás Raskó
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Laboratory for Mobile DNA, Berlin, Germany
| | - Adrián Martí Pastor
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), L aboratory for Neuroproteomics, Berlin, Germany
| | - Erich E Wanker
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), L aboratory for Neuroproteomics, Berlin, Germany.
| |
Collapse
|
20
|
Dehghani N, Guven G, Kun-Rodrigues C, Gouveia C, Foster K, Hanagasi H, Lohmann E, Samanci B, Gurvit H, Bilgic B, Bras J, Guerreiro R. A comprehensive analysis of copy number variation in a Turkish dementia cohort. Hum Genomics 2021; 15:48. [PMID: 34321086 PMCID: PMC8317312 DOI: 10.1186/s40246-021-00346-z] [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: 04/15/2021] [Accepted: 07/09/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Copy number variants (CNVs) include deletions or multiplications spanning genomic regions. These regions vary in size and may span genes known to play a role in human diseases. As examples, duplications and triplications of SNCA have been shown to cause forms of Parkinson's disease, while duplications of APP cause early onset Alzheimer's disease (AD). RESULTS Here, we performed a systematic analysis of CNVs in a Turkish dementia cohort in order to further characterize the genetic causes of dementia in this population. One hundred twenty-four Turkish individuals, either at risk of dementia due to family history, diagnosed with mild cognitive impairment, AD, or frontotemporal dementia, were whole-genome genotyped and CNVs were detected. We integrated family analysis with a comprehensive assessment of potentially disease-associated CNVs in this Turkish dementia cohort. We also utilized both dementia and non-dementia individuals from the UK Biobank in order to further elucidate the potential role of the identified CNVs in neurodegenerative diseases. We report CNVs overlapping the previously implicated genes ZNF804A, SNORA70B, USP34, XPO1, and a locus on chromosome 9 which includes a cluster of olfactory receptors and ABCA1. Additionally, we also describe novel CNVs potentially associated with dementia, overlapping the genes AFG1L, SNX3, VWDE, and BC039545. CONCLUSIONS Genotyping data from understudied populations can be utilized to identify copy number variation which may contribute to dementia.
Collapse
Affiliation(s)
- Nadia Dehghani
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Gamze Guven
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Celia Kun-Rodrigues
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Catarina Gouveia
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Kalina Foster
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA
- Neuroscience Department, Michigan State University College of Natural Science, East Lansing, MI, USA
| | - Hasmet Hanagasi
- Behavioural Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ebba Lohmann
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- DZNE, German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Bedia Samanci
- Behavioural Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Hakan Gurvit
- Behavioural Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Basar Bilgic
- Behavioural Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Jose Bras
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA
- Division of Psychiatry and Behavioral Medicine, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - Rita Guerreiro
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA.
- Division of Psychiatry and Behavioral Medicine, Michigan State University College of Human Medicine, Grand Rapids, MI, USA.
| |
Collapse
|
21
|
Xu K, Zhang Y, Li J. Expression and function of circular RNAs in the mammalian brain. Cell Mol Life Sci 2021; 78:4189-4200. [PMID: 33558994 PMCID: PMC11071837 DOI: 10.1007/s00018-021-03780-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/07/2021] [Accepted: 01/27/2021] [Indexed: 01/19/2023]
Abstract
Mammalian brain presents extraordinary complexity reflected in the structure, function, and dynamic changes in the biological and physiological processes of development, maturity, and aging. Recent transcriptomic profiles from the brain tissues of distinct species have described a novel class of transcripts with a covalently closed-loop structure, called circular RNAs (circRNAs), which are produced by alternative back-splicing and derived from genes associated with synaptogenesis and neural activities. Brain is a tightly regulated and largely unexplored organ where circRNAs are highly enriched and expressed in the cell type-, spatiotemporal-specific, sex-biased, and age-related manner. Although the biological functions of most of the circRNAs in the brain remain elusive, increased evidence suggests that dynamic changes in circRNA expression are critical for brain function and the maintenance of physiological homeostasis in the brain. Here, we review the latest immense progresses in the understanding of circRNA expression and function in the mammalian brain. We also discuss possibly biological functions of circRNAs in the brain, which may provide new sights of understanding brain development and aging, as well as the pathogenesis of mental diseases.
Collapse
Affiliation(s)
- Kaiyu Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Ying Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jiali Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- National Institute on Drug Dependence, Peking University, Beijing, China.
- PKU/McGovern Institute for Brain Research, Peking University, Beijing, China.
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
22
|
Abstract
Introduction: Most medical diagnoses present somewhat differently in men and women, more so at specific periods of life. Treatment effects may also differ. This is true for schizophrenia, where premorbid effects are experienced earlier in life in boys than in girls, and where symptoms and outcomes differ.Areas covered: This review does not cover all the differences that have been reported between men and women but, instead, focuses on the ones that carry important implications for clinical care: effective antipsychotic doses, medication side effects, symptom fluctuation due to hormonal levels, comorbidities, and women's requirements for prenatal, obstetric, postpartum, and parenting support.Expert opinion: Of consequence to schizophrenia, sex-biased genes, epigenetic modifications, and sex steroids all impact the structure and function of the brain. Furthermore, life experiences and social roles exert major sex-specific influences. The co-morbidities that accompany schizophrenia also affect men and women to different degrees. This review offers several examples of sex-specific intervention and concludes that gold standard treatment must look beyond symptoms and address all the physiologic, psychologic, and social role needs of men and women suffering from this psychiatric disorder.
Collapse
Affiliation(s)
- Mary V Seeman
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Corsi-Zuelli F, Deakin B. Impaired regulatory T cell control of astroglial overdrive and microglial pruning in schizophrenia. Neurosci Biobehav Rev 2021; 125:637-653. [PMID: 33713699 DOI: 10.1016/j.neubiorev.2021.03.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/16/2021] [Accepted: 03/06/2021] [Indexed: 02/07/2023]
Abstract
It is widely held that schizophrenia involves an active process of peripheral inflammation that induces or reflects brain inflammation with activation of microglia, the brain's resident immune cells. However, recent in vivo radioligand binding studies and large-scale transcriptomics in post-mortem brain report reduced markers of microglial inflammation. The findings suggest a contrary hypothesis; that microglia are diverted into their non-inflammatory synaptic remodelling phenotype that interferes with neurodevelopment and perhaps contributes to the relapsing nature of schizophrenia. Recent discoveries on the regulatory interactions between micro- and astroglial cells and immune regulatory T cells (Tregs) cohere with clinical omics data to suggest that: i) disinhibited astrocytes mediate the shift in microglial phenotype via the production of transforming growth factor-beta, which also contributes to the disturbances of dopamine and GABA function in schizophrenia, and ii) systemically impaired functioning of Treg cells contributes to the dysregulation of glial function, the low-grade peripheral inflammation, and the hitherto unexplained predisposition to auto-immunity and reduced life-expectancy in schizophrenia, including greater COVID-19 mortality.
Collapse
Affiliation(s)
- Fabiana Corsi-Zuelli
- Department of Neuroscience and Behaviour, Division of Psychiatry, Ribeirão Preto Medical School, University of São Paulo, 14048-900, Ribeirão Preto, São Paulo, Brazil
| | - Bill Deakin
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK.
| |
Collapse
|
24
|
Liu X, Han T, Xie H, Fu Z, Yao Q, Lin Z, Zhu H, Zhan D. Evaluation of the relationship between VRK2, rs4380187 polymorphisms, and genetic susceptibility to schizophrenia in the Chinese Han population. J Gene Med 2021; 23:e3313. [PMID: 33522046 DOI: 10.1002/jgm.3313] [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: 10/10/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Schizophrenia (SZ) is a serious hereditary mental disease with a low recovery rate, especially due to the lack of understanding about the cause of the disease. VRK2 is considered to be related to the pathogenesis of schizophrenia. In this study, we analyzed the correlation between VRK2, rs4380187 single-nucleotide polymorphism (SNP), and schizophrenia. METHODS Peripheral blood DNA was extracted using a genomic DNA extraction kit. The DNA samples were genotyped using the Agena MassARRAY platform, and four genetic models were applied to compute the odds ratios (ORs) and 95% confidence intervals (CIs) using unconditional logistic regression. The p value was obtained by the chi-square and t test for independent samples. RESULTS The C allele of rs4380187 SNP was significantly (p = 0.008) associated with decreased risk of SZ. The AA genotype of rs4380187 showed significantly (p = 0.009) lower frequency in cases with SZ than in controls and was associated with decreased risk of the disease. The frequency of the CA genotype of rs4380187 correlated with a 0.73-fold decreased risk of SZ (p = 0.033). In the co-dominant genetic model, the genotype of rs4380187 was associated with a decreased risk of SZ (p = 0.010). We also found that the log-additive model of rs4380187 significantly reduced the risk of SZ disease (p = 0.007). CONCLUSION This study provides further evidence that rs4380187 SNP is associated with SZ. This genotype variation could be associated with the psychopathology and cognitive function in SZ.
Collapse
Affiliation(s)
- Xianglai Liu
- Institute of Mental Health, Anning Hospital, Hainan Province, China
| | - Tianming Han
- Institute of Mental Health, Anning Hospital, Hainan Province, China
| | - Hailing Xie
- Institute of Mental Health, Anning Hospital, Hainan Province, China.,The Third Department of Psychiatry, Anning Hospital, Hainan Province, China
| | - Zejuan Fu
- Institute of Mental Health, Anning Hospital, Hainan Province, China
| | - Qiankun Yao
- Institute of Mental Health, Anning Hospital, Hainan Province, China
| | - Zhan Lin
- Institute of Mental Health, Anning Hospital, Hainan Province, China
| | - Hong Zhu
- Institute of Mental Health, Anning Hospital, Hainan Province, China
| | - Dafei Zhan
- Institute of Mental Health, Anning Hospital, Hainan Province, China
| |
Collapse
|
25
|
ZFP804A mutant mice display sex-dependent schizophrenia-like behaviors. Mol Psychiatry 2021; 26:2514-2532. [PMID: 33303946 PMCID: PMC8440220 DOI: 10.1038/s41380-020-00972-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 10/20/2020] [Accepted: 11/24/2020] [Indexed: 12/22/2022]
Abstract
Genome-wide association studies uncovered the association of ZNF804A (Zinc-finger protein 804A) with schizophrenia (SZ). In vitro data have indicated that ZNF804A might exert its biological roles by regulating spine and neurite morphogenesis. However, no in vivo data are available for the role of ZNF804A in psychiatric disorders in general, SZ in particular. We generated ZFP804A mutant mice, and they showed deficits in contextual fear and spatial memory. We also observed the sensorimotor gating impairment, as revealed by the prepulse inhibition test, but only in female ZFP804A mutant mice from the age of 6 months. Notably, the PPI difference between the female mutant and control mice was no longer existed with the administration of Clozapine or after the ovariectomy. Hippocampal long-term potentiation was normal in both genders of the mutant mice. Long-term depression was absent in male mutants, but facilitated in the female mutants. Protein levels of hippocampal serotonin-6 receptor and GABAB1 receptor were increased, while those of cortical dopamine 2 receptor were decreased in the female mutants with no obvious changes in the male mutants. Moreover, the spine density was reduced in the cerebral cortex and hippocampus of the mutant mice. Knockdown of ZFP804A impaired the neurite morphogenesis of cortical and hippocampal neurons, while its overexpression enhanced neurite morphogenesis only in the cortical neurons in vitro. Our data collectively support the idea that ZFP804A/ZNF804A plays important roles in the cognitive functions and sensorimotor gating, and its dysfunction may contribute to SZ, particularly in the female patients.
Collapse
|
26
|
Zhang C, Xiao X, Li T, Li M. Translational genomics and beyond in bipolar disorder. Mol Psychiatry 2021; 26:186-202. [PMID: 32424235 DOI: 10.1038/s41380-020-0782-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 02/08/2023]
Abstract
Genome-wide association studies (GWAS) have revealed multiple genomic loci conferring risk of bipolar disorder (BD), providing hints for its underlying pathobiology. However, there are still remaining questions to answer. For example, discordance exists between BD heritability estimated with earlier epidemiological evidence and that calculated based on common GWAS variations. Where is the "missing heritability"? How can we explain the biology of the disease based on genetic findings? In this review, we summarize the accomplishments and limitations of current BD GWAS, and discuss potential reasons for the "missing heritability." In addition, progresses of research for the biological mechanisms underlying BD genetic risk using brain tissues, reprogrammed cells, and model animals are reviewed. While our knowledge of BD genetic basis is significantly promoted by these efforts, the complexities of gene regulation in the genome, the spatial-temporal heterogeneity during brain development, and the limitations of different experimental models should always be considered. Notably, several genes have been widely studied given their relatively well-characterized involvement in BD (e.g., CACAN1C and ANK3), and findings of these genes are summarized to both outline possible biological mechanisms of BD and describe examples of translating GWAS discoveries into the pathophysiology.
Collapse
Affiliation(s)
- Chen Zhang
- Division of Mood Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Tao Li
- Mental Health Center and Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China. .,West China Brain Research Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
27
|
Yang L, Xu F, He Y, Li Y, Chen Z, Wang S. Association Between ZNF804A Gene rs1344706 Polymorphism and Brain Functions in Healthy Individuals: A Systematic Review and Voxel-Based Meta-Analysis. Neuropsychiatr Dis Treat 2021; 17:2925-2935. [PMID: 34548792 PMCID: PMC8449690 DOI: 10.2147/ndt.s322114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/02/2021] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Zinc finger protein 804A (ZNF804A) protein participates in embryonic neural repair and development. The single nucleotide polymorphism rs1344706 in ZNF804A gene is closely related to functional abnormalities of the human brain. However, these results are inconsistent. This association was verified by meta-analysis in this study. METHODS Fifteen studies on functional magnetic resonance imaging involving 1710 healthy individuals were included in the systematic review and meta-analysis used by Anisotropic Effect-Size Signed Differential Mapping software. RESULTS Functional connectivity of the right dorsolateral prefrontal cortex (rDLPFC)-left hippocampus in the rs1344706 risk allele carrier was significantly increased (z = 2.066, p < 0.001), while those in the rDLPFC-left middle frontal gyrus (z = -1.420, p < 0.001) and rDLPFC-right middle frontal gyrus (z = -1.298, p < 0.001) were significantly decreased. Neural activity of the left anterior cingulate gyrus in the rs1344706 risk allele carrier was significantly decreased (z = -2.525, p < 0.001). Sensitivity analysis was almost stable, and no publication bias was found. CONCLUSION The changes in brain function have a clear correlation with ZNF804A gene in healthy individuals, which indicate the contribution of genetic variants on brain dysfunction. REGISTRATION NUMBER This meta-analysis is registered in PROSPERO (No. CRD42016051331).
Collapse
Affiliation(s)
- Liqiong Yang
- Department of Pharmacy, Chengdu Medical College, Chengdu, 610500, People's Republic of China
| | - Fan Xu
- Department of Public Health, Chengdu Medical College, Chengdu, 610500, People's Republic of China
| | - Yi He
- Department of Medicine, National Engineering and Research Center for Natural Medicines, Chengdu, 610400, People's Republic of China
| | - Yanzhang Li
- Department of Psychology, Chengdu Medical College, Chengdu, 610500, People's Republic of China
| | - Zi Chen
- Department of Psychology, Chengdu Medical College, Chengdu, 610500, People's Republic of China
| | - Shuai Wang
- Department of Psychology, Chengdu Medical College, Chengdu, 610500, People's Republic of China
| |
Collapse
|
28
|
Identification and prioritization of gene sets associated with schizophrenia risk by co-expression network analysis in human brain. Mol Psychiatry 2020; 25:791-804. [PMID: 30478419 DOI: 10.1038/s41380-018-0304-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 08/07/2018] [Accepted: 10/29/2018] [Indexed: 12/13/2022]
Abstract
Schizophrenia polygenic risk is plausibly manifested by complex transcriptional dysregulation in the brain, involving networks of co-expressed and functionally related genes. The main purpose of this study was to identify and prioritize co-expressed gene sets in a hierarchical manner, based on the strength of the relationships with clinical diagnosis and with polygenic risk for schizophrenia. Weighted Gene Co-expression Network Analysis (WGCNA) was applied to RNA-quality-adjusted DLPFC RNA-Seq data from the LIBD Postmortem Human Brain Repository (90 controls, 74 schizophrenia cases; all Caucasians) to construct co-expression networks and detect "modules" of co-expressed genes. After multiple internal and external validation procedures, modules of selected interest were tested for enrichment in biological ontologies, for association with schizophrenia polygenic risk scores (PRSs) and with diagnosis, and also for enrichment in genes within the significant GWAS loci reported by the Psychiatric Genomic Consortium (PGC2). The association between schizophrenia genetic signals and modules of co-expression converged on one module showing not only a significant association with both diagnosis and PRS but also significant overlap with 36 PGC2 loci genes, deemed the strongest candidates for drug targets. This module contained many genes involved in synaptic signaling and neuroplasticity. Fifty-three PGC2 genes were in modules associated only with diagnosis and 59 in modules unrelated to diagnosis or PRS. Our study highlights complex relationships between gene co-expression networks in the brain and clinical state and polygenic risk for SCZ and provides a strategy for using this information in selecting and prioritizing potentially targetable gene sets for therapeutic drug development.
Collapse
|
29
|
Zhou D, Xiao X, Li M. The schizophrenia risk isoform ZNF804A E3E4 affects dendritic spine. Schizophr Res 2020; 218:324-325. [PMID: 31956006 DOI: 10.1016/j.schres.2019.12.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 12/31/2019] [Indexed: 11/17/2022]
Affiliation(s)
- Danyang Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
30
|
Li HJ, Qu N, Hui L, Cai X, Zhang CY, Zhong BL, Zhang SF, Chen J, Xia B, Wang L, Jia QF, Li W, Chang H, Xiao X, Li M, Li Y. Further confirmation of netrin 1 receptor (DCC) as a depression risk gene via integrations of multi-omics data. Transl Psychiatry 2020; 10:98. [PMID: 32184385 PMCID: PMC7078234 DOI: 10.1038/s41398-020-0777-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/21/2020] [Accepted: 03/03/2020] [Indexed: 12/15/2022] Open
Abstract
Genome-wide association studies (GWAS) of major depression and its relevant biological phenotypes have been extensively conducted in large samples, and transcriptome-wide analyses in the tissues of brain regions relevant to pathogenesis of depression, e.g., dorsolateral prefrontal cortex (DLPFC), have also been widely performed recently. Integrating these multi-omics data will enable unveiling of depression risk genes and even underlying pathological mechanisms. Here, we employ summary data-based Mendelian randomization (SMR) and integrative risk gene selector (iRIGS) approaches to integrate multi-omics data from GWAS, DLPFC expression quantitative trait loci (eQTL) analyses and enhancer-promoter physical link studies to prioritize high-confidence risk genes for depression, followed by independent replications across distinct populations. These integrative analyses identify multiple high-confidence depression risk genes, and numerous lines of evidence supporting pivotal roles of the netrin 1 receptor (DCC) gene in this illness across different populations. Our subsequent explorative analyses further suggest that DCC significantly predicts neuroticism, well-being spectrum, cognitive function and putamen structure in general populations. Gene expression correlation and pathway analyses in DLPFC further show that DCC potentially participates in the biological processes and pathways underlying synaptic plasticity, axon guidance, circadian entrainment, as well as learning and long-term potentiation. These results are in agreement with the recent findings of this gene in neurodevelopment and psychiatric disorders, and we thus further confirm that DCC is an important susceptibility gene for depression, and might be a potential target for new antidepressants.
Collapse
Affiliation(s)
- Hui-Juan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Na Qu
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China
| | - Li Hui
- Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xin Cai
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chu-Yi Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Bao-Liang Zhong
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China
| | - Shu-Fang Zhang
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China
| | - Jing Chen
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China
| | - Bin Xia
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China
| | - Lu Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Qiu-Fang Jia
- Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Wei Li
- Department of Blood Transfusion, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hong Chang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Yi Li
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China.
| |
Collapse
|
31
|
Al-Naama N, Mackeh R, Kino T. C 2H 2-Type Zinc Finger Proteins in Brain Development, Neurodevelopmental, and Other Neuropsychiatric Disorders: Systematic Literature-Based Analysis. Front Neurol 2020; 11:32. [PMID: 32117005 PMCID: PMC7034409 DOI: 10.3389/fneur.2020.00032] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/10/2020] [Indexed: 12/15/2022] Open
Abstract
Neurodevelopmental disorders (NDDs) are multifaceted pathologic conditions manifested with intellectual disability, autistic features, psychiatric problems, motor dysfunction, and/or genetic/chromosomal abnormalities. They are associated with skewed neurogenesis and brain development, in part through dysfunction of the neural stem cells (NSCs) where abnormal transcriptional regulation on key genes play significant roles. Recent accumulated evidence highlights C2H2-type zinc finger proteins (C2H2-ZNFs), the largest transcription factor family in humans, as important targets for the pathologic processes associated with NDDs. In this review, we identified their significant accumulation (74 C2H2-ZNFs: ~10% of all human member proteins) in brain physiology and pathology. Specifically, we discuss their physiologic contribution to brain development, particularly focusing on their actions in NSCs. We then explain their pathologic implications in various forms of NDDs, such as morphological brain abnormalities, intellectual disabilities, and psychiatric disorders. We found an important tendency that poly-ZNFs and KRAB-ZNFs tend to be involved in the diseases that compromise gross brain structure and human-specific higher-order functions, respectively. This may be consistent with their characteristic appearance in the course of species evolution and corresponding contribution to these brain activities.
Collapse
Affiliation(s)
- Njoud Al-Naama
- Laboratory of Molecular and Genomic Endocrinology, Division of Translational Medicine, Sidra Medicine, Doha, Qatar
| | - Rafah Mackeh
- Laboratory of Molecular and Genomic Endocrinology, Division of Translational Medicine, Sidra Medicine, Doha, Qatar
| | - Tomoshige Kino
- Laboratory of Molecular and Genomic Endocrinology, Division of Translational Medicine, Sidra Medicine, Doha, Qatar
| |
Collapse
|
32
|
The genome-wide risk alleles for psychiatric disorders at 3p21.1 show convergent effects on mRNA expression, cognitive function, and mushroom dendritic spine. Mol Psychiatry 2020; 25:48-66. [PMID: 31723243 DOI: 10.1038/s41380-019-0592-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 12/13/2022]
Abstract
Schizophrenia and bipolar disorder (BPD) are believed to share clinical features, etiological factors, and disease pathologies (such as impaired cognitive functions and dendritic spine pathology). Meanwhile, there is growing evidence of shared genetic risk between schizophrenia and BPD, despite that our knowledge of the functional risk variations and biological mechanisms is still limited. Here, we conduct summary data-based Mendelian randomization (SMR) analyses through combining the statistical data from genome-wide association studies (GWAS) of both schizophrenia and BPD and multiple expression quantitative trait loci (eQTL) datasets of the human brain dorsolateral prefrontal cortex (DLPFC) tissues. These integrative investigations identify a lead risk locus at the chromosome 3p21.1 region, which contains numerous single-nucleotide polymorphisms (SNPs) in varied linkage disequilibrium (LD) and encompasses more than 20 genes. Further analyses suggest that many SNPs at 3p21.1 are significantly associated with both schizophrenia and BPD, and even depression, and the psychiatric risk alleles at 3p21.1 are correlated with mRNA expression of multiple genes such as NEK4, GNL3, and PBRM1. We also identify a 335-bp functional Alu polymorphism rs71052682 in significant LD with the psychiatric GWAS risk SNP rs2251219, and confirm the regulatory effects of this Alu polymorphism on transcription activities. We then explore the involvement of the 3p21.1 locus in the common clinical features and etiology of these illnesses. We reveal that psychiatric risk alleles at 3p21.1 in low-to-high LD consistently predict worse cognitive functions in humans, and manipulating the gene expression (NEK4, GNL3, and PBRM1) linked with higher genetic risk could reduce the density of mushroom dendritic spines in rat primary cortical neurons, mirroring the spine pathology in the prefrontal cortex of psychiatric patients. Our results find that, although the risk alleles at 3p21.1 are in low-to-moderate LD spanning a large genomic area, their underlying biological mechanisms in psychiatric disorders likely converge. These results provide essential insights into the neural mechanisms underlying the chromosome 3p21.1 risk locus in the shared pathological and etiological features of both schizophrenia and BPD.
Collapse
|
33
|
Hadji-Turdeghal K, Andreasen L, Hagen CM, Ahlberg G, Ghouse J, Bækvad-Hansen M, Bybjerg-Grauholm J, Hougaard DM, Hedley P, Haunsø S, Svendsen JH, Kanters JK, Jepps TA, Skov MW, Christiansen M, Olesen MS. Genome-wide association study identifies locus at chromosome 2q32.1 associated with syncope and collapse. Cardiovasc Res 2020; 116:138-148. [PMID: 31049583 PMCID: PMC6918066 DOI: 10.1093/cvr/cvz106] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 04/25/2019] [Indexed: 12/11/2022] Open
Abstract
AIMS Syncope is a common condition associated with frequent hospitalization or visits to the emergency department. Family aggregation and twin studies have shown that syncope has a heritable component. We investigated whether common genetic variants predispose to syncope and collapse. METHODS AND RESULTS We used genome-wide association data on syncope on 408 961 individuals with European ancestry from the UK Biobank study. In a replication study, we used the Integrative Psychiatric Research Consortium (iPSYCH) cohort (n = 86 189), to investigate the risk of incident syncope stratified by genotype carrier status. We report on a genome-wide significant locus located on chromosome 2q32.1 [odds ratio = 1.13, 95% confidence interval (CI) 1.10-1.17, P = 5.8 × 10-15], with lead single nucleotide polymorphism rs12465214 in proximity to the gene zinc finger protein 804a (ZNF804A). This association was also shown in the iPSYCH cohort, where homozygous carriers of the C allele conferred an increased hazard ratio (1.30, 95% CI 1.15-1.46, P = 1.68 × 10-5) of incident syncope. Quantitative polymerase chain reaction analysis showed ZNF804A to be expressed most abundantly in brain tissue. CONCLUSION We identified a genome-wide significant locus (rs12465214) associated with syncope and collapse. The association was replicated in an independent cohort. This is the first genome-wide association study to associate a locus with syncope and collapse.
Collapse
Affiliation(s)
- Katra Hadji-Turdeghal
- Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Rigshospitalet (Copenhagen University Hospital), Copenhagen, Denmark
- Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Laura Andreasen
- Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Rigshospitalet (Copenhagen University Hospital), Copenhagen, Denmark
- Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian M Hagen
- Department of Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Copenhagen, Denmark
| | - Gustav Ahlberg
- Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Rigshospitalet (Copenhagen University Hospital), Copenhagen, Denmark
- Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas Ghouse
- Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Rigshospitalet (Copenhagen University Hospital), Copenhagen, Denmark
- Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marie Bækvad-Hansen
- Department of Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Copenhagen, Denmark
| | - Jonas Bybjerg-Grauholm
- Department of Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Copenhagen, Denmark
| | - David M Hougaard
- Department of Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Copenhagen, Denmark
| | - Paula Hedley
- Department of Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Copenhagen, Denmark
| | - Stig Haunsø
- Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Rigshospitalet (Copenhagen University Hospital), Copenhagen, Denmark
- Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper H Svendsen
- Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Rigshospitalet (Copenhagen University Hospital), Copenhagen, Denmark
- Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen K Kanters
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas A Jepps
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten W Skov
- Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Rigshospitalet (Copenhagen University Hospital), Copenhagen, Denmark
- Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Christiansen
- Department of Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Copenhagen, Denmark
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten S Olesen
- Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Rigshospitalet (Copenhagen University Hospital), Copenhagen, Denmark
- Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
34
|
Liu W, Yan H, Zhou D, Cai X, Zhang Y, Li S, Li H, Li S, Zhou DS, Li X, Zhang C, Sun Y, Dai JP, Zhong J, Yao YG, Luo XJ, Fang Y, Zhang D, Ma Y, Yue W, Li M, Xiao X. The depression GWAS risk allele predicts smaller cerebellar gray matter volume and reduced SIRT1 mRNA expression in Chinese population. Transl Psychiatry 2019; 9:333. [PMID: 31819045 PMCID: PMC6901563 DOI: 10.1038/s41398-019-0675-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/19/2019] [Accepted: 11/27/2019] [Indexed: 12/28/2022] Open
Abstract
Major depressive disorder (MDD) is recognized as a primary cause of disability worldwide, and effective management of this illness has been a great challenge. While genetic component is supposed to play pivotal roles in MDD pathogenesis, the genetic and phenotypic heterogeneity of the illness has hampered the discovery of its genetic determinants. In this study, in an independent Han Chinese sample (1824 MDD cases and 3031 controls), we conducted replication analyses of two genetic loci highlighted in a previous Chinese MDD genome-wide association study (GWAS), and confirmed the significant association of a single nucleotide polymorphism (SNP) rs12415800 near SIRT1. Subsequently, using hypothesis-free whole-brain analysis in two independent Han Chinese imaging samples, we found that individuals carrying the MDD risk allele of rs12415800 exhibited aberrant gray matter volume in the left posterior cerebellar lobe compared with those carrying the non-risk allele. Besides, in independent Han Chinese postmortem brain and peripheral blood samples, the MDD risk allele of rs12415800 predicted lower SIRT1 mRNA levels, which was consistent with the reduced expression of this gene in MDD patients compared with healthy subjects. These results provide further evidence for the involvement of SIRT1 in MDD, and suggest that this gene might participate in the illness via affecting the development of cerebellum, a brain region that is potentially underestimated in previous MDD studies.
Collapse
Affiliation(s)
- Weipeng Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Hao Yan
- Peking University Sixth Hospital/Institute of Mental Health, Beijing, China
- NHC Key Laboratory of Mental Health (Peking University) and National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Danyang Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xin Cai
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yuyanan Zhang
- Peking University Sixth Hospital/Institute of Mental Health, Beijing, China
- NHC Key Laboratory of Mental Health (Peking University) and National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Shiyi Li
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Huijuan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Shiwu Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Dong-Sheng Zhou
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang, China
| | - Xingxing Li
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang, China
| | - Chen Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Sun
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, Hubei, China
- Chinese Brain Bank Center, Wuhan, Hubei, China
| | - Jia-Pei Dai
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, Hubei, China
- Chinese Brain Bank Center, Wuhan, Hubei, China
| | - Jingmei Zhong
- Psychiatry Department, The first people's hospital of Yunnan province, Kunming, Yunnan, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, Yunnan, China
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, Yunnan, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yiru Fang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Dai Zhang
- Peking University Sixth Hospital/Institute of Mental Health, Beijing, China
- NHC Key Laboratory of Mental Health (Peking University) and National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
- Peking-Tsinghua Joint Center for Life Sciences and PKU IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Yina Ma
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Weihua Yue
- Peking University Sixth Hospital/Institute of Mental Health, Beijing, China.
- NHC Key Laboratory of Mental Health (Peking University) and National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China.
- Peking-Tsinghua Joint Center for Life Sciences and PKU IDG/McGovern Institute for Brain Research, Peking University, Beijing, China.
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China.
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
35
|
Yin L, Chau CKL, Sham PC, So HC. Integrating Clinical Data and Imputed Transcriptome from GWAS to Uncover Complex Disease Subtypes: Applications in Psychiatry and Cardiology. Am J Hum Genet 2019; 105:1193-1212. [PMID: 31785786 PMCID: PMC6904812 DOI: 10.1016/j.ajhg.2019.10.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 10/22/2019] [Indexed: 12/19/2022] Open
Abstract
Classifying subjects into clinically and biologically homogeneous subgroups will facilitate the understanding of disease pathophysiology and development of targeted prevention and intervention strategies. Traditionally, disease subtyping is based on clinical characteristics alone, but subtypes identified by such an approach may not conform exactly to the underlying biological mechanisms. Very few studies have integrated genomic profiles (e.g., those from GWASs) with clinical symptoms for disease subtyping. Here we proposed an analytic framework capable of finding complex diseases subgroups by leveraging both GWAS-predicted gene expression levels and clinical data by a multi-view bicluster analysis. This approach connects SNPs to genes via their effects on expression, so the analysis is more biologically relevant and interpretable than a pure SNP-based analysis. Transcriptome of different tissues can also be readily modeled. We also proposed various evaluation metrics for assessing clustering performance. Our framework was able to subtype schizophrenia subjects into diverse subgroups with different prognosis and treatment response. We also applied the framework to the Northern Finland Birth Cohort (NFBC) 1966 dataset and identified high and low cardiometabolic risk subgroups in a gender-stratified analysis. The prediction strength by cross-validation was generally greater than 80%, suggesting good stability of the clustering model. Our results suggest a more data-driven and biologically informed approach to defining metabolic syndrome and subtyping psychiatric disorders. Moreover, we found that the genes "blindly" selected by the algorithm are significantly enriched for known susceptibility genes discovered in GWASs of schizophrenia or cardiovascular diseases. The proposed framework opens up an approach to subject stratification.
Collapse
Affiliation(s)
- Liangying Yin
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Carlos K L Chau
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Pak-Chung Sham
- Centre for Genomic Sciences, University of Hong Kong, Hong Kong SAR, China; Department of Psychiatry, University of Hong Kong, Hong Kong SAR, China; State Key Laboratory for Cognitive and Brain Sciences, University of Hong Kong, Hong Kong SAR, China
| | - Hon-Cheong So
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Zoology Institute of Zoology and The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Psychiatry, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K.L. Cheung Research Centre for Management of Parkinsonism, The Chinese University of Hong Kong, Hong Kong SAR, China; Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518000, China.
| |
Collapse
|
36
|
Chapman RM, Tinsley CL, Hill MJ, Forrest MP, Tansey KE, Pardiñas AF, Rees E, Doyle AM, Wilkinson LS, Owen MJ, O’Donovan MC, Blake DJ. Convergent Evidence That ZNF804A Is a Regulator of Pre-messenger RNA Processing and Gene Expression. Schizophr Bull 2019; 45:1267-1278. [PMID: 30597088 PMCID: PMC6811834 DOI: 10.1093/schbul/sby183] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Genome-wide association studies have linked common variation in ZNF804A with an increased risk of schizophrenia. However, little is known about the biology of ZNF804A and its role in schizophrenia. Here, we investigate the function of ZNF804A using a variety of complementary molecular techniques. We show that ZNF804A is a nuclear protein that interacts with neuronal RNA splicing factors and RNA-binding proteins including RBFOX1, which is also associated with schizophrenia, CELF3/4, components of the ubiquitin-proteasome system and the ZNF804A paralog, GPATCH8. GPATCH8 also interacts with splicing factors and is localized to nuclear speckles indicative of a role in pre-messenger RNA (mRNA) processing. Sequence analysis showed that GPATCH8 contains ultraconserved, alternatively spliced poison exons that are also regulated by RBFOX proteins. ZNF804A knockdown in SH-SY5Y cells resulted in robust changes in gene expression and pre-mRNA splicing converging on pathways associated with nervous system development, synaptic contact, and cell adhesion. We observed enrichment (P = 1.66 × 10-9) for differentially spliced genes in ZNF804A-depleted cells among genes that contain RBFOX-dependent alternatively spliced exons. Differentially spliced genes in ZNF804A-depleted cells were also enriched for genes harboring de novo loss of function mutations in autism spectrum disorder (P = 6.25 × 10-7, enrichment 2.16) and common variant alleles associated with schizophrenia (P = .014), bipolar disorder and schizophrenia (P = .003), and autism spectrum disorder (P = .005). These data suggest that ZNF804A and its paralogs may interact with neuronal-splicing factors and RNA-binding proteins to regulate the expression of a subset of synaptic and neurodevelopmental genes.
Collapse
Affiliation(s)
- Ria M Chapman
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Caroline L Tinsley
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Matthew J Hill
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Marc P Forrest
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Katherine E Tansey
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
- College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Antonio F Pardiñas
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Elliott Rees
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - A Michelle Doyle
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Lawrence S Wilkinson
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
- School of Psychology, Cardiff University, Cardiff, UK
| | - Michael J Owen
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Michael C O’Donovan
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Derek J Blake
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
37
|
Squassina A, Meloni A, Chillotti C, Pisanu C. Zinc finger proteins in psychiatric disorders and response to psychotropic medications. Psychiatr Genet 2019; 29:132-141. [PMID: 31464994 DOI: 10.1097/ypg.0000000000000231] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Zinc finger proteins are a large family of abundantly expressed small motifs that play a crucial role in a wide range of physiological and pathophysiological mechanisms. Findings published so far support an involvement of zinc fingers in psychiatric disorders. Most of the evidence has been provided for the zinc finger protein 804A (ZNF804A) gene, which has been suggested to be implicated in schizophrenia and bipolar disorder. This evidence has been corroborated by a wide range of functional studies showing that ZNF804A regulates the expression of genes involved in cell adhesion and plays a crucial role in neurite formation and maintenance of dendritic spines. On the other hand, far less is known on other zinc finger proteins and their involvement in psychiatric disorders. In this review, we discussed studies exploring the role of zinc finger proteins in schizophrenia, bipolar disorder, and major depressive disorder as well as in pharmacogenetics of psychotropic drugs.
Collapse
Affiliation(s)
- Alessio Squassina
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari Unit of Clinical Pharmacology, University Hospital of Cagliari, Cagliari, Italy Department of Psychiatry, Dalhousie University, Halifax, NS, Canada Department of Neuroscience, Unit of Functional Pharmacology, Uppsala University, Uppsala, Sweden
| | | | | | | |
Collapse
|
38
|
Dashti HS, Merino J, Lane JM, Song Y, Smith CE, Tanaka T, McKeown NM, Tucker C, Sun D, Bartz TM, Li-Gao R, Nisa H, Reutrakul S, Lemaitre RN, Alshehri TM, de Mutsert R, Bazzano L, Qi L, Knutson KL, Psaty BM, Mook-Kanamori DO, Perica VB, Neuhouser ML, Scheer FAJL, Rutter MK, Garaulet M, Saxena R. Genome-wide association study of breakfast skipping links clock regulation with food timing. Am J Clin Nutr 2019; 110:473-484. [PMID: 31190057 PMCID: PMC6669061 DOI: 10.1093/ajcn/nqz076] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 04/08/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Little is known about the contribution of genetic variation to food timing, and breakfast has been determined to exhibit the most heritable meal timing. As breakfast timing and skipping are not routinely measured in large cohort studies, alternative approaches include analyses of correlated traits. OBJECTIVES The aim of this study was to elucidate breakfast skipping genetic variants through a proxy-phenotype genome-wide association study (GWAS) for breakfast cereal skipping, a commonly assessed correlated trait. METHODS We leveraged the statistical power of the UK Biobank (n = 193,860) to identify genetic variants related to breakfast cereal skipping as a proxy-phenotype for breakfast skipping and applied several in silico approaches to investigate mechanistic functions and links to traits/diseases. Next, we attempted validation of our approach in smaller breakfast skipping GWAS from the TwinUK (n = 2,006) and the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) consortium (n = 11,963). RESULTS In the UK Biobank, we identified 6 independent GWAS variants, including those implicated for caffeine (ARID3B/CYP1A1), carbohydrate metabolism (FGF21), schizophrenia (ZNF804A), and encoding enzymes important for N6-methyladenosine RNA transmethylation (METTL4, YWHAB, and YTHDF3), which regulates the pace of the circadian clock. Expression of identified genes was enriched in the cerebellum. Genome-wide correlation analyses indicated positive correlations with anthropometric traits. Through Mendelian randomization (MR), we observed causal links between genetically determined breakfast skipping and higher body mass index, more depressive symptoms, and smoking. In bidirectional MR, we demonstrated a causal link between being an evening person and skipping breakfast, but not vice versa. We observed association of our signals in an independent breakfast skipping GWAS in another British cohort (P = 0.032), TwinUK, but not in a meta-analysis of non-British cohorts from the CHARGE consortium (P = 0.095). CONCLUSIONS Our proxy-phenotype GWAS identified 6 genetic variants for breakfast skipping, linking clock regulation with food timing and suggesting a possible beneficial role of regular breakfast intake as part of a healthy lifestyle.
Collapse
Affiliation(s)
- Hassan S Dashti
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA,Program in Medical and Population Genetics, Broad Institute, Cambridge, MA,Department of Anesthesia, Critical Care, and Pain Medicine,Address correspondence to HSD (e-mail:
| | - Jordi Merino
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA,Program in Medical and Population Genetics, Broad Institute, Cambridge, MA,Diabetes Unit, Massachusetts General Hospital, and Department of Medicine, Harvard Medical School, Boston, MA
| | - Jacqueline M Lane
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA,Program in Medical and Population Genetics, Broad Institute, Cambridge, MA,Department of Anesthesia, Critical Care, and Pain Medicine
| | - Yanwei Song
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA,Program in Medical and Population Genetics, Broad Institute, Cambridge, MA
| | | | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD
| | - Nicola M McKeown
- Nutritional Epidemiology Program, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA
| | - Chandler Tucker
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
| | - Dianjianyi Sun
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Departments of Biostatistics and Medicine, University of Washington, Seattle, WA
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Hoirun Nisa
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA
| | - Sirimon Reutrakul
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Rozenn N Lemaitre
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - Tahani M Alshehri
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Lydia Bazzano
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA,Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Kristen L Knutson
- Center for Circadian and Sleep Medicine, Department of Neurology, Northwestern University, Chicago, IL
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Epidemiology, Medicine, and Health Services, University of Washington, Seattle, WA,Kaiser Permanente Washington Health Research Institute, Seattle, WA
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands,Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, Netherlands
| | - Vesna Boraska Perica
- Department for Medical Biology, University of Split School of Medicine, Split, Croatia
| | - Marian L Neuhouser
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Frank A J L Scheer
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA,Division of Sleep Medicine, Harvard Medical School, Boston, MA,Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA
| | - Martin K Rutter
- Division of Endocrinology, Diabetes, and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom,Manchester Diabetes Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Marta Garaulet
- Department of Physiology, University of Murcia, Murcia, Spain,IMIB-Arrixaca, Murcia, Spain
| | - Richa Saxena
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA,Program in Medical and Population Genetics, Broad Institute, Cambridge, MA,Department of Anesthesia, Critical Care, and Pain Medicine,E-mail: )
| |
Collapse
|
39
|
Li W, Yang Y, Luo B, Zhang Y, Song X, Li M, Lv L. Association of SYNE1 locus with bipolar disorder in Chinese population. Hereditas 2019; 156:19. [PMID: 31236099 PMCID: PMC6580462 DOI: 10.1186/s41065-019-0095-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 06/11/2019] [Indexed: 01/08/2023] Open
Abstract
Objectives Genome-wide association studies (GWAS) suggest that rs9371601 in the SYNE1 gene is a risk SNP for bipolar disorder (BPD) in populations of European ancestry, but further replication analyses across distinct populations are needed. Methods We analyzed the association between rs9371601 and BPD in a Han Chinese sample of 1315 BPD cases and 1956 controls. Results We observed a significant association between rs9371601 and BPD in Han Chinese (p = 0.0121, OR = 0.859). However, further examinations revealed that the Europeans and Chinese subjects had different BPD risk alleles at the locus. We then found that rs9371601 had different “minor alleles” and distinct linkage disequilibrium (LD) patterns surrounding itself in Europeans and Han Chinese, which might be the explanation of the observed inconsistent association signals for this locus in different populations. Our explorative analyses of the biological impact of rs9371601 suggested that this SNP was significantly associated with the methylation of a CpG site (cg01844274, p = 5.05⨯10− 6) within SYNE1 in human dorsal lateral prefrontal cortex (DLPFC) tissues. Conclusions Our data confirms the association between rs9371601 and BPD, but the underlying biological mechanism remains to be fully elucidated in further studies. Electronic supplementary material The online version of this article (10.1186/s41065-019-0095-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wenqiang Li
- 1Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan China.,2Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan China
| | - Yongfeng Yang
- 1Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan China.,2Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan China
| | - Binbin Luo
- 1Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan China.,2Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan China
| | - Yan Zhang
- 1Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan China.,2Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan China
| | - Xueqin Song
- 3The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan China
| | - Ming Li
- 4Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan China
| | - Luxian Lv
- 1Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan China.,2Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan China.,5Henan Province People's Hospital, Zhengzhou, Henan China
| |
Collapse
|
40
|
Meller T, Schmitt S, Stein F, Brosch K, Mosebach J, Yüksel D, Zaremba D, Grotegerd D, Dohm K, Meinert S, Förster K, Redlich R, Opel N, Repple J, Hahn T, Jansen A, Andlauer TFM, Forstner AJ, Heilmann-Heimbach S, Streit F, Witt SH, Rietschel M, Müller-Myhsok B, Nöthen MM, Dannlowski U, Krug A, Kircher T, Nenadić I. Associations of schizophrenia risk genes ZNF804A and CACNA1C with schizotypy and modulation of attention in healthy subjects. Schizophr Res 2019; 208:67-75. [PMID: 31076262 DOI: 10.1016/j.schres.2019.04.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 03/05/2019] [Accepted: 04/20/2019] [Indexed: 12/30/2022]
Abstract
Schizotypy is a multidimensional risk phenotype distributed in the general population, constituting of subclinical, psychotic-like symptoms. It is associated with psychosis proneness, and several risk genes for psychosis are associated with schizotypy in non-clinical populations. Schizotypy might also modulate cognitive abilities as it is associated with attentional deficits in healthy subjects. In this study, we tested the hypothesis that established genetic risk variants ZNF804A rs1344706 and CACNA1C rs1006737 are associated with psychometric schizotypy and that schizotypy mediates their effect on attention or vice versa. In 615 healthy subjects from the FOR2107 cohort study, we analysed the genetic risk variants ZNF804A rs1344706 and CACNA1C rs1006737, psychometric schizotypy (schizotypal personality questionnaire-brief SPQB), and a neuropsychological measure of sustained and selective attention (d2 test). ZNF804A rs1344706 C (non-risk) alleles were significantly associated with higher SPQ-B Cognitive-Perceptual subscores in women and with attention deficits in both sexes. This schizotypy dimension also mediated the effect of ZNF804A on attention in women, but not in men. CACNA1C rs1006737-A showed a significant sex-modulated negative association with Interpersonal schizotypy only in men, and no effect on attention. Our multivariate model demonstrates differential genetic contributions of two psychosis risk genes to dimensions of schizotypy and, partly, to attention. This supports a model of shared genetic influence between schizotypy and cognitive functions impaired in schizophrenia.
Collapse
Affiliation(s)
- Tina Meller
- Department of Psychiatry and Psychotherapy, Philipps-Universität Marburg and University Hospital Marburg, UKGM, Rudolf-Bultmann-Str. 8, 35039 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), Hans-Meerwein-Str. 6, 35032 Marburg, Germany.
| | - Simon Schmitt
- Department of Psychiatry and Psychotherapy, Philipps-Universität Marburg and University Hospital Marburg, UKGM, Rudolf-Bultmann-Str. 8, 35039 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), Hans-Meerwein-Str. 6, 35032 Marburg, Germany
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, Philipps-Universität Marburg and University Hospital Marburg, UKGM, Rudolf-Bultmann-Str. 8, 35039 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), Hans-Meerwein-Str. 6, 35032 Marburg, Germany
| | - Katharina Brosch
- Department of Psychiatry and Psychotherapy, Philipps-Universität Marburg and University Hospital Marburg, UKGM, Rudolf-Bultmann-Str. 8, 35039 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), Hans-Meerwein-Str. 6, 35032 Marburg, Germany
| | - Johannes Mosebach
- Department of Psychiatry and Psychotherapy, Philipps-Universität Marburg and University Hospital Marburg, UKGM, Rudolf-Bultmann-Str. 8, 35039 Marburg, Germany
| | - Dilara Yüksel
- Department of Psychiatry and Psychotherapy, Philipps-Universität Marburg and University Hospital Marburg, UKGM, Rudolf-Bultmann-Str. 8, 35039 Marburg, Germany; SRI International, Center for Health Sciences, Bioscience Division, 333 Ravenswood Avenue, 94025 Menlo Park, CA, USA
| | - Dario Zaremba
- Department of Psychiatry and Psychotherapy, Westfälische Wilhelms-Universität Münster, Albert-Schweitzer-Campus 1, Building A9, 48149 Münster, Germany
| | - Dominik Grotegerd
- Department of Psychiatry and Psychotherapy, Westfälische Wilhelms-Universität Münster, Albert-Schweitzer-Campus 1, Building A9, 48149 Münster, Germany
| | - Katharina Dohm
- Department of Psychiatry and Psychotherapy, Westfälische Wilhelms-Universität Münster, Albert-Schweitzer-Campus 1, Building A9, 48149 Münster, Germany
| | - Susanne Meinert
- Department of Psychiatry and Psychotherapy, Westfälische Wilhelms-Universität Münster, Albert-Schweitzer-Campus 1, Building A9, 48149 Münster, Germany
| | - Katharina Förster
- Department of Psychiatry and Psychotherapy, Westfälische Wilhelms-Universität Münster, Albert-Schweitzer-Campus 1, Building A9, 48149 Münster, Germany
| | - Ronny Redlich
- Department of Psychiatry and Psychotherapy, Westfälische Wilhelms-Universität Münster, Albert-Schweitzer-Campus 1, Building A9, 48149 Münster, Germany
| | - Nils Opel
- Department of Psychiatry and Psychotherapy, Westfälische Wilhelms-Universität Münster, Albert-Schweitzer-Campus 1, Building A9, 48149 Münster, Germany
| | - Jonathan Repple
- Department of Psychiatry and Psychotherapy, Westfälische Wilhelms-Universität Münster, Albert-Schweitzer-Campus 1, Building A9, 48149 Münster, Germany
| | - Tim Hahn
- Department of Psychiatry and Psychotherapy, Westfälische Wilhelms-Universität Münster, Albert-Schweitzer-Campus 1, Building A9, 48149 Münster, Germany
| | - Andreas Jansen
- Department of Psychiatry and Psychotherapy, Philipps-Universität Marburg and University Hospital Marburg, UKGM, Rudolf-Bultmann-Str. 8, 35039 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), Hans-Meerwein-Str. 6, 35032 Marburg, Germany; Core-Facility BrainImaging, Faculty of Medicine, Philipps-Universität Marburg, Rudolf-Bultmann-Str. 8, 35039 Marburg, Germany
| | - Till F M Andlauer
- Max-Planck-Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany; Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Straße 22, 81675 Munich, Germany
| | - Andreas J Forstner
- Institute of Human Genetics, University of Bonn School of Medicine & University Hospital Bonn, Sigmund-Freud-Straße 25, 53127 Bonn, Germany; Institute of Human Genetics, Philipps-Universität Marburg, Baldingerstraße, 35033 Marburg, Germany; Department of Biomedicine, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland; Institute of Medical Genetics and Pathology, University Hospital Basel, Schönbeinstr. 40, 4056 Basel, Switzerland
| | - Stefanie Heilmann-Heimbach
- Institute of Human Genetics, University of Bonn School of Medicine & University Hospital Bonn, Sigmund-Freud-Straße 25, 53127 Bonn, Germany
| | - Fabian Streit
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J5, 68159 Mannheim, Germany
| | - Stephanie H Witt
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J5, 68159 Mannheim, Germany
| | - Marcella Rietschel
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J5, 68159 Mannheim, Germany
| | - Bertram Müller-Myhsok
- Max-Planck-Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377 Munich, Germany; Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn School of Medicine & University Hospital Bonn, Sigmund-Freud-Straße 25, 53127 Bonn, Germany
| | - Udo Dannlowski
- Department of Psychiatry and Psychotherapy, Westfälische Wilhelms-Universität Münster, Albert-Schweitzer-Campus 1, Building A9, 48149 Münster, Germany
| | - Axel Krug
- Department of Psychiatry and Psychotherapy, Philipps-Universität Marburg and University Hospital Marburg, UKGM, Rudolf-Bultmann-Str. 8, 35039 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), Hans-Meerwein-Str. 6, 35032 Marburg, Germany
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, Philipps-Universität Marburg and University Hospital Marburg, UKGM, Rudolf-Bultmann-Str. 8, 35039 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), Hans-Meerwein-Str. 6, 35032 Marburg, Germany
| | - Igor Nenadić
- Department of Psychiatry and Psychotherapy, Philipps-Universität Marburg and University Hospital Marburg, UKGM, Rudolf-Bultmann-Str. 8, 35039 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), Hans-Meerwein-Str. 6, 35032 Marburg, Germany
| |
Collapse
|
41
|
Wu Y, Bi R, Zeng C, Ma C, Sun C, Li J, Xiao X, Li M, Zhang DF, Zheng P, Sheng N, Luo XJ, Yao YG. Identification of the primate-specific gene BTN3A2 as an additional schizophrenia risk gene in the MHC loci. EBioMedicine 2019; 44:530-541. [PMID: 31133542 PMCID: PMC6603853 DOI: 10.1016/j.ebiom.2019.05.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 04/26/2019] [Accepted: 05/03/2019] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Schizophrenia is a complex mental disorder resulting in poor life quality and high social and economic burden. Despite the fact that genome-wide association studies (GWASs) have successfully identified a number of risk loci for schizophrenia, identifying the causal genes at the risk loci and elucidating their roles in disease pathogenesis remain major challenges. METHODS The summary data-based Mendelian randomization analysis (SMR) was used to integrate a large-scale GWAS of schizophrenia with brain expression quantitative trait loci (eQTL) data and brain methylation expression quantitative trait loci (meQTL) data, to identify novel risk gene(s) for schizophrenia. We then analyzed the mRNA expression and methylation statuses of the gene hit BTN3A2 during the early brain development. Electrophysiological analyses of CA1 pyramidal neurons were performed to evaluate the excitatory and inhibitory synaptic activity after overexpression of BTN3A2 in rat hippocampal slices. Cell surface binding assay was used to test the interaction of BTN3A2 and neurexins. FINDINGS We identified BTN3A2 as a potential risk gene for schizophrenia. The mRNA expression and methylation data showed that BTN3A2 expression in human brain is highest post-natally. Further electrophysiological analyses of rat hippocampal slices showed that BTN3A2 overexpression specifically suppressed the excitatory synaptic activity onto CA1 pyramidal neurons, most likely through its interaction with the presynaptic adhesion molecule neurexins. INTERPRETATION Increased expression of BTN3A2 might confer risk for schizophrenia by altering excitatory synaptic function. Our result constitutes a paradigm for distilling risk gene using an integrative analysis and functional characterization in the post-GWAS era. FUND: This study was supported by the Strategic Priority Research Program (B) of the Chinese Academy of Sciences (XDB02020003 to Y-GY), the National Natural Science Foundation of China (31730037 to Y-GY), and the Bureau of Frontier Sciences and Education, Chinese Academy of Sciences (QYZDJ-SSW-SMC005 to Y-GY).
Collapse
Affiliation(s)
- Yong Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Rui Bi
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| | - Chunhua Zeng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Changguo Ma
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Chunli Sun
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China; State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Jingzheng Li
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China; State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Deng-Feng Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| | - Ping Zheng
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China; State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Nengyin Sheng
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China; State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.
| |
Collapse
|
42
|
A Bayesian framework that integrates multi-omics data and gene networks predicts risk genes from schizophrenia GWAS data. Nat Neurosci 2019; 22:691-699. [PMID: 30988527 PMCID: PMC6646046 DOI: 10.1038/s41593-019-0382-7] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 03/13/2019] [Indexed: 12/17/2022]
Abstract
Genome-wide association studies (GWAS) have identified >100 schizophrenia (SCZ)-associated loci, but using these findings to illuminate disease biology remains a challenge. Here, we present integrative RIsk Gene Selector (iRIGS), a Bayesian framework that integrates multi-omics data and gene networks to infer risk genes in GWAS loci. By applying iRIGS to SCZ GWAS data, we predicted a set of high-confidence risk genes (HRGs), most of which are not the nearest genes to the GWAS index variants. HRGs account for a significantly enriched heritability estimated by stratified LD-score regression. Moreover, HRGs are predominantly expressed in brain tissues, especially prenatally, and are enriched for targets of approved drugs, suggesting opportunities to reposition existing drugs for SCZ. Thus, iRIGS can leverage accumulating functional genomics and GWAS data to advance understanding of SCZ etiology and potential therapeutics.
Collapse
|
43
|
The interaction between the ZNF804A gene and cannabis use on the risk of psychosis in a non-clinical sample. Prog Neuropsychopharmacol Biol Psychiatry 2019; 89:174-180. [PMID: 30118824 DOI: 10.1016/j.pnpbp.2018.08.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 07/26/2018] [Accepted: 08/13/2018] [Indexed: 12/19/2022]
Abstract
The ZNF804A gene and cannabis use are risk factors for psychosis and both have also been associated with schizotypal traits. This study aimed to investigate: i) the association of lifetime cannabis use (and its dose effect) with schizotypal personality traits, and ii) whether the genetic variability at ZNF804A gene modulates that association. Our sample consisted of 385 Spanish non-clinical subjects (43.1% males, mean age = 21.11(2.19)). Schizotypy was evaluated using the three factors of the Schizotypal Personality Questionnaire-Brief (SPQ-B): Cognitive-Perceptual (SPQ-CP), Interpersonal (SPQ-I) and Disorganized (SPQ-D). Subjects were classified according to their frequency of cannabis consumption, and dichotomized as users or non-users. The effects of a genetic variant of ZNF804A (rs1344706) and cannabis use, as well as their interaction, on each of the three SPQ-B factors were assessed using linear models and permutation tests. Sex, SCL anxiety scores and use of other drugs were included as covariates. Our analysis showed a significant relationship between ZNF804A and SPQ-I: AA genotype was associated with higher scores (β = 0.885 pFDR = .018). An interaction between the AA genotype and lifetime cannabis use was found in SPQ-CP (β = 1.297 pFDR = 0.018). This interaction showed a dose-effect pattern among AA subjects: schizotypy scores increased with increasing frequency of cannabis use (sporadic users: β = 0.746 pFDR = 0.208; monthly users: β = 1.688 pFDR = 0.091; intense users: β = 1.623 pFDR = 0.038). These results add evidence on that the ZNF804A gene is associated with schizotypy and suggest that the interaction between cannabis use and ZNF804A genotype could modulate psychosis proneness.
Collapse
|
44
|
Zhang L, Qin Y, Gong X, Peng R, Cai C, Zheng Y, Du Y, Wang H. A promoter variant in ZNF804A decreasing its expression increases the risk of autism spectrum disorder in the Han Chinese population. Transl Psychiatry 2019; 9:31. [PMID: 30670685 PMCID: PMC6342935 DOI: 10.1038/s41398-019-0369-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 12/10/2018] [Accepted: 01/02/2019] [Indexed: 12/15/2022] Open
Abstract
Synaptic pathology may be one of the cellular substrates underlying autism spectrum disorder (ASD). ZNF804A is a transcription factor that can affect or regulate the expression of many candidate genes involved in ASD. It also localizes at synapses and regulates neuronal and synaptic morphology. So far, few reports have addressed possible associations between ZNF804A polymorphisms and ASD. This study aimed to investigate whether ZNF804A genetic variants contribute to ASD susceptibility and its possible pathological role in the disorder. We analyzed the relationship of two polymorphisms (rs10497655 and rs34714481) in ZNF804A promoter region with ASD in 854 cases versus 926 controls. The functional analyses of rs10497655 were then performed using real-time quantitative polymerase chain reaction, electrophoretic mobility shift assays, chromatin immunoprecipitation and dual-luciferase assays. The variant rs10497655 was significantly associated with ASD (P = 0.007851), which had a significant effect on ZNF804A expression, with the T risk allele homozygotes related with reduced ZNF804A expression in human fetal brains. HSF2 acted as a suppressor by down-regulating ZNF804A expression and had a stronger binding affinity for the T allele of rs10497655 than for the C allele. This was the first experiment to elucidate the process in which a disease-associated SNP affects the level of ZNF804A expression by binding with the upstream regulation factor HSF2. This result indicates that the rs10497655 allelic expression difference of ZNF804A during the critical period of brain development may have an effect on postnatal phenotypes of ASD. It reveals new roles of ZNF804A polymorphisms in the pathogenesis of psychiatric disorders.
Collapse
Affiliation(s)
- Linna Zhang
- Department of Child & Adolescent Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yue Qin
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China
- Ministry of Education (MOE) Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Xiaohong Gong
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China
- Ministry of Education (MOE) Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Rui Peng
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China
- Ministry of Education (MOE) Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Chunquan Cai
- Department of Neurosurgery, Tianjin Children's Hospital, Tianjin, 300134, China
| | - Yufang Zheng
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China
| | - Yasong Du
- Department of Child & Adolescent Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Hongyan Wang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China.
- Key Laboratory of Reproduction Regulation of NPFPC, Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, 200032, China.
- Children's Hospital of Fudan University, Shanghai, 201102, China.
| |
Collapse
|
45
|
Xiao X, Zhang C, Grigoroiu-Serbanescu M, Wang L, Li L, Zhou D, Yuan TF, Wang C, Chang H, Wu Y, Li Y, Wu DD, Yao YG, Li M. The cAMP responsive element-binding (CREB)-1 gene increases risk of major psychiatric disorders. Mol Psychiatry 2018; 23:1957-1967. [PMID: 29158582 DOI: 10.1038/mp.2017.243] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/25/2017] [Accepted: 09/14/2017] [Indexed: 12/11/2022]
Abstract
Bipolar disorder (BPD), schizophrenia (SCZ) and unipolar major depressive disorder (MDD) are primary psychiatric disorders sharing substantial genetic risk factors. We previously reported that two single-nucleotide polymorphisms (SNPs) rs2709370 and rs6785 in the cAMP responsive element-binding (CREB)-1 gene (CREB1) were associated with the risk of BPD and abnormal hippocampal function in populations of European ancestry. In the present study, we further expanded our analyses of rs2709370 and rs6785 in multiple BPD, SCZ and MDD data sets, including the published Psychiatric Genomics Consortium (PGC) genome-wide association study, the samples used in our previous CREB1 study, and six additional cohorts (three new BPD samples, two new SCZ samples and one new MDD sample). Although the associations of both CREB1 SNPs with each illness were not replicated in the new cohorts (BPD analysis in 871 cases and 1089 controls (rs2709370, P=0.0611; rs6785, P=0.0544); SCZ analysis in 1273 cases and 1072 controls (rs2709370, P=0.230; rs6785, P=0.661); and MDD analysis in 129 cases and 100 controls (rs2709370, P=0.114; rs6785, P=0.188)), an overall meta-analysis of all included samples suggested that both SNPs were significantly associated with increased risk of BPD (11 105 cases and 51 331 controls; rs2709370, P=2.33 × 10-4; rs6785, P=6.33 × 10-5), SCZ (34 913 cases and 44 528 controls; rs2709370, P=3.96 × 10-5; rs6785, P=2.44 × 10-5) and MDD (9369 cases and 9619 controls; rs2709370, P=0.0144; rs6785, P=0.0314), with the same direction of allelic effects across diagnostic categories. We then examined the impact of diagnostic status on CREB1 mRNA expression using data obtained from independent brain tissue samples, and observed that the mRNA expression of CREB1 was significantly downregulated in psychiatric patients compared with healthy controls. The protein-protein interaction analyses showed that the protein encoded by CREB1 directly interacted with several risk genes of psychiatric disorders identified by GWAS. In conclusion, the current study suggests that CREB1 might be a common risk gene for major psychiatric disorders, and further investigations are necessary.
Collapse
Affiliation(s)
- X Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - C Zhang
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - M Grigoroiu-Serbanescu
- Biometric Psychiatric Genetics Research Unit, Alexandru Obregia Clinical Psychiatric Hospital, Bucharest, Romania.
| | - L Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - L Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - D Zhou
- Ningbo Kangning Hospital, Ningbo, China
| | - T-F Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - C Wang
- Department of Pharmacology, and Provincial Key Laboratory of Pathophysiology in Ningbo University School of Medicine, Ningbo, China
| | - H Chang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - Y Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - Y Li
- Laboratory for Conservation and Utilization of Bio-Resource, Yunnan University, Kunming, China
| | - D-D Wu
- State Key Laboratory of Genetic Resources and Evolution, Chinese Academy of Sciences, Kunming, China
| | - Y-G Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - M Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China. .,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
46
|
ZNF804A Variation May Affect Hippocampal-Prefrontal Resting-State Functional Connectivity in Schizophrenic and Healthy Individuals. Neurosci Bull 2018; 34:507-516. [PMID: 29611035 DOI: 10.1007/s12264-018-0221-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 02/05/2018] [Indexed: 01/05/2023] Open
Abstract
The ZNF804A variant rs1344706 has consistently been associated with schizophrenia and plays a role in hippocampal-prefrontal functional connectivity during working memory. Whether the effect exists in the resting state and in patients with schizophrenia remains unclear. In this study, we investigated the ZNF804A polymorphism at rs1344706 in 92 schizophrenic patients and 99 healthy controls of Han Chinese descent, and used resting-state functional magnetic resonance imaging to explore the functional connectivity in the participants. We found a significant main effect of genotype on the resting-state functional connectivity (RSFC) between the hippocampus and the dorsolateral prefrontal cortex (DLPFC) in both schizophrenic patients and healthy controls. The homozygous ZNF804A rs1344706 genotype (AA) conferred a high risk of schizophrenia, and also exhibited significantly decreased resting functional coupling between the left hippocampus and right DLPFC (F(2,165) = 13.43, P < 0.001). The RSFC strength was also correlated with cognitive performance and the severity of psychosis in schizophrenia. The current findings identified the neural impact of the ZNF804A rs1344706 on hippocampal-prefrontal RSFC associated with schizophrenia.
Collapse
|
47
|
de Castro-Catala M, Mora-Solano A, Kwapil TR, Cristóbal-Narváez P, Sheinbaum T, Racioppi A, Barrantes-Vidal N, Rosa A. The genome-wide associated candidate gene ZNF804A and psychosis-proneness: Evidence of sex-modulated association. PLoS One 2017; 12:e0185072. [PMID: 28931092 PMCID: PMC5607189 DOI: 10.1371/journal.pone.0185072] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 09/06/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The Zinc finger protein 804A (ZNF804A) is a promising candidate gene for schizophrenia and the broader psychosis phenotype that emerged from genome-wide association studies. It is related to neurodevelopment and associated to severe symptoms of schizophrenia and alterations in brain structure, as well as positive schizotypal personality traits in non-clinical samples. Moreover, a female-specific association has been observed between ZNF804A and schizophrenia. AIM The present study examined the association of two ZNF804A polymorphisms (rs1344706 and rs7597593) with the positive dimension of schizotypy and psychotic-like experiences in a sample of 808 non-clinical subjects. Additionally, we wanted to explore whether the sexual differences reported in schizophrenia are also present in psychosis-proneness. RESULTS Our results showed an association between rs7597593 and both schizotypy and psychotic-like experiences. These associations were driven by females, such those carrying the C allele had higher scores in the positive dimension of both variables compared to TT allele homozygotes. CONCLUSION The findings of the present study support the inclusion of ZNF804 variability in studies of the vulnerability for the development of psychopathology in non-clinical samples and consideration of sex as a moderator of this association.
Collapse
Affiliation(s)
- Marta de Castro-Catala
- Secció de Zoologia i Antropologia Biològica, Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain
| | - Aurea Mora-Solano
- Secció de Zoologia i Antropologia Biològica, Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona, Spain
| | - Thomas R. Kwapil
- Department of Psychology, University of North Carolina at Greensboro, Greensboro, North Carolina, United States of America
- Department of Psychology, University of Illinois at Champaign-Urbana, Champaign, Illinois, United States of America
| | - Paula Cristóbal-Narváez
- Departament de Psicologia Clínica i de la Salut, Facultat de Psicologia, Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain
| | - Tamara Sheinbaum
- Departament de Psicologia Clínica i de la Salut, Facultat de Psicologia, Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain
| | - Anna Racioppi
- Departament de Psicologia Clínica i de la Salut, Facultat de Psicologia, Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain
| | - Neus Barrantes-Vidal
- Department of Psychology, University of North Carolina at Greensboro, Greensboro, North Carolina, United States of America
- Departament de Psicologia Clínica i de la Salut, Facultat de Psicologia, Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain
- Sant Pere Claver - Fundació Sanitària, Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Araceli Rosa
- Secció de Zoologia i Antropologia Biològica, Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Salud Mental (CIBERSAM), Madrid, Spain
| |
Collapse
|
48
|
Harrison PJ. ZNF804A: Insights From the First Genome-wide Significant Schizophrenia Gene. Biol Psychiatry 2017; 82:6-7. [PMID: 28619252 DOI: 10.1016/j.biopsych.2017.03.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 03/31/2017] [Indexed: 10/19/2022]
Affiliation(s)
- Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom; Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, United Kingdom.
| |
Collapse
|