1
|
Yang Q, Zhang X, Zhang L, Cheng C, Zhao J. Exploring the influence of the DRD2 gene on mathematical ability: perspectives of gene association and gene-environment interaction. BMC Psychol 2024; 12:572. [PMID: 39425204 PMCID: PMC11488083 DOI: 10.1186/s40359-024-01997-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 09/11/2024] [Indexed: 10/21/2024] Open
Abstract
Mathematical ability is influenced by genes and environment. This study focused on the effect of DRD2, a candidate gene for working memory, on mathematical ability. The results in child participants revealed associations between the DRD2 gene and mathematical ability. It was found that individual's mathematical ability was influenced by Single Nucleotide Polymorphisms (SNPs) in DRD2, both in the form of haplotypes and in the way of interaction with parental education. These findings suggest that dopaminergic genes are linked to mathematical ability. This study provides evidence for the genetic basis of mathematical ability and offers guidance for personalized intervention in mathematical education.
Collapse
Affiliation(s)
- Qing Yang
- School of Psychology, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, China
| | - Ximiao Zhang
- School of Psychology, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, China
| | - Liming Zhang
- School of Psychology, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, China
| | - Chen Cheng
- School of Psychology, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, China
| | - Jingjing Zhao
- Department of Psychology, The Chinese University of Hong Kong, Shatin, Hong Kong.
| |
Collapse
|
2
|
Spencer S, Harker SA, Barry F, Beauchemin J, Braden BB, Burton P, D'sa V, Koinis-Mitchell D, Mennenga SE, Deoni SCL, Lewis CR. The peripheral epigenome predicts white matter volume contingent on developmental stage: An ECHO study. RESEARCH SQUARE 2024:rs.3.rs-4139933. [PMID: 38699338 PMCID: PMC11065062 DOI: 10.21203/rs.3.rs-4139933/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Epigenetic processes, including DNA methylation, are emerging as key areas of interest for their potential roles as biomarkers and contributors to the risk of neurodevelopmental, psychiatric, and other brain-based disorders. Despite this growing focus, there remains a notable gap in our understanding of how DNA methylation correlates with individual variations in brain function and structure. Additionally, the dynamics of these relationships during developmental periods, which are critical windows during which many disorders first appear, are still largely unexplored. The current study extends the field by examining if peripheral DNA methylation of myelination-related genes predicts white matter volume in a healthy pediatric population [N = 250; females = 113; age range 2 months-14 years; Mage = 5.14, SDage = 3.60]. We assessed if DNA methylation of 17 myelin-related genes predict white matter volume and if age moderates these relationships. Results highlight low variability in myelin-related epigenetic variance at birth, which rapidly increases non-linearly with age, and that DNA methylation, measured at both the level of a CpG site or gene, is highly predictive of white matter volume, in early childhood but not late childhood. These novel findings propel the field forward by establishing that DNA methylation of myelin-related genes from a peripheral tissue is a predictive marker of white matter volume in children and is influenced by developmental stage. The research underscores the significance of peripheral epigenetic patterns as a proxy for investigating the effects of environmental factors, behaviors, and disorders associated with white matter.
Collapse
|
3
|
Navarro L, Gómez-Carballa A, Pischedda S, Montoto-Louzao J, Viz-Lasheras S, Camino-Mera A, Hinault T, Martinón-Torres F, Salas A. Sensogenomics of music and Alzheimer's disease: An interdisciplinary view from neuroscience, transcriptomics, and epigenomics. Front Aging Neurosci 2023; 15:1063536. [PMID: 36819725 PMCID: PMC9935844 DOI: 10.3389/fnagi.2023.1063536] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/11/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction The relationship between music and Alzheimer's disease (AD) has been approached by different disciplines, but most of our outstanding comes from neuroscience. Methods First, we systematically reviewed the state-of-the-art of neuroscience and cognitive sciences research on music and AD (>100 studies), and the progress made on the therapeutic impact of music stimuli in memory. Next, we meta-analyzed transcriptomic and epigenomic data of AD patients to search for commonalities with genes and pathways previously connected to music in genome association, epigenetic, and gene expression studies. Results Our findings indicate that >93% of the neuroscience/ cognitive sciences studies indicate at least one beneficial effect of music on patients with neurodegenerative diseases, being improvements on memory and cognition the most frequent outcomes; other common benefits were on social behavior, mood and emotion, anxiety and agitation, quality of life, and depression. Out of the 334 music-related genes, 127 (38%) were found to be linked to epigenome/transcriptome analysis in AD (vs. healthy controls); some of them (SNCA, SLC6A4, ASCC2, FTH1, PLAUR and ARHGAP26) have been reported to be associated e.g. with musical aptitude and music effect on the transcriptome. Other music-related genes (GMPR, SELENBP1 and ADIPOR1) associated to neuropsychiatric, neurodegenerative diseases and music performance, emerged as hub genes in consensus co-expression modules detected between AD and music estimulated transcriptomes. In addition, we found connections between music, AD and dopamine related genes, with SCNA being the most remarkable - a gene previously associated with learning and memory, and neurodegenerative disorders (e.g., Parkinson's disease and AD). Discussion The present study indicate that the vast majority of neuroscientific studies unambiguously show that music has a beneficial effect on health, being the most common benefits relevant to Alzheimer's disease. These findings illuminate a new roadmap for genetic research in neurosciences, and musical interventions in AD and other neurodegenerative conditions.
Collapse
Affiliation(s)
- Laura Navarro
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain,Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Santiago de Compostela, Galicia, Spain,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Alberto Gómez-Carballa
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain,Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Santiago de Compostela, Galicia, Spain,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Sara Pischedda
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain,Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Santiago de Compostela, Galicia, Spain,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Julián Montoto-Louzao
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain,Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Santiago de Compostela, Galicia, Spain,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Sandra Viz-Lasheras
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain,Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Santiago de Compostela, Galicia, Spain,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Alba Camino-Mera
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain,Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Santiago de Compostela, Galicia, Spain,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Thomas Hinault
- Normandie Université, UNICAEN, PSL Université Paris, EPHE, Inserm, U1077, CHU de Caen, Centre Cyceron, Neuropsychologie et Imagerie de la Mémoire Humaine, Caen, France
| | - Federico Martinón-Torres
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Antonio Salas
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain,Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Santiago de Compostela, Galicia, Spain,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain,*Correspondence: Antonio Salas, ✉
| |
Collapse
|
4
|
Smederevac S, Delgado-Cruzata L, Mitrović D, Dinić BM, Bravo TAT, Delgado M, Bugarski Ignjatović V, Sadiković S, Milovanović I, Vučinić N, Branovački B, Prinz M, Budimlija Z, Kušić‐Tišma J, Nikolašević Ž. Differences in MB-COMT DNA methylation in monozygotic twins on phenotypic indicators of impulsivity. Front Genet 2023; 13:1067276. [PMID: 36685886 PMCID: PMC9852709 DOI: 10.3389/fgene.2022.1067276] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/28/2022] [Indexed: 01/09/2023] Open
Abstract
Epigenetic modifications of the membrane bound catechol-O-methyltransferase (MB-COMT) gene may affect the enzymatic degradation of dopamine, and consequently, human behavior. This study investigated the association between membrane bound catechol-O-methyltransferase DNA methylation (DNAm) differences in 92 monozygotic (MZ) twins with phenotypic manifestations of cognitive, behavioral, and personality indicators associated with reward-related behaviors and lack of control. We used pyrosequencing to determine DNAm of the regulatory region of membrane bound catechol-O-methyltransferase in saliva DNA. Results of intrapair differences in the percentage of membrane bound catechol-O-methyltransferase DNAm at each of five CpG sites show that there are associations between phenotypic indicators of lack of control and membrane bound catechol-O-methyltransferase DNAm differences on CpG1, CpG2 and CpG4, suggesting the common epigenetic patterns for personality traits, cognitive functions, and risk behaviors.
Collapse
Affiliation(s)
- Snežana Smederevac
- Department of Psychology, Faculty of Philosophy, University of Novi Sad, Novi Sad, Serbia,*Correspondence: Selka Sadiković, ; Snežana Smederevac,
| | | | - Dušanka Mitrović
- Department of Psychology, Faculty of Philosophy, University of Novi Sad, Novi Sad, Serbia
| | - Bojana M. Dinić
- Department of Psychology, Faculty of Philosophy, University of Novi Sad, Novi Sad, Serbia
| | | | - Maria Delgado
- Mount Holyoke College, South Hadley, MA, United States
| | | | - Selka Sadiković
- Department of Psychology, Faculty of Philosophy, University of Novi Sad, Novi Sad, Serbia,*Correspondence: Selka Sadiković, ; Snežana Smederevac,
| | - Ilija Milovanović
- Department of Psychology, Faculty of Philosophy, University of Novi Sad, Novi Sad, Serbia
| | - Nataša Vučinić
- Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Bojan Branovački
- Department of Psychology, Faculty of Philosophy, University of Novi Sad, Novi Sad, Serbia
| | - Mechthild Prinz
- John Jay College of Criminal Justice, New York, NY, United States
| | - Zoran Budimlija
- Department of Neurology, School of Medicine, New York University, New York City, NY, United States
| | - Jelena Kušić‐Tišma
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | | |
Collapse
|
5
|
Marzilli E, Cerniglia L, Tambelli R, Cimino S. Children’s ADHD and Dysregulation Problems, DAT1 Genotype and Methylation, and their Interplay with Family Environment. CHILD & YOUTH CARE FORUM 2022. [DOI: 10.1007/s10566-022-09687-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Abstract
Background
International literature has underlined the complex interplay between genetic and environmental variables in shaping children’s emotional-behavioral functioning.
Objective
This study aimed to explore the dynamic relationship between children’s Dopamine Transporter (DAT1) genotype and methylation, and maternal and paternal affective environment, on children’s Attention Deficit Hyperactivity Disorder (ADHD) problems and dysregulation problems.
Method
In a community sample of 76 families with school-aged children, we assessed children’s DAT1 genotype and methylation, their own ADHD problems and dysregulation profile (CBCL 6–18 DP), and maternal and paternal psychopathological risk, parenting stress, and marital adjustment. Hierarchical regressions were carried out to verify the possible moderation of children’s genotype on the relationship between children’s methylation and psychopathological risk, parental environment and children’s methylation, and parental environment and children’s psychopathological risk.
Results
The levels of methylation at M1 CpG significantly predicted ADHD problems among children with 10/10 genotype, whereas high levels of methylation at M6 CpG predicted low ADHD problems for children with 9/x genotype. High levels of methylation at M3 CpG were associated with high scores of CBCL DP. DAT1 genotype moderated the relationship between maternal and paternal variables with children’s methylation and psychopathological risk. The scores of maternal and paternal Dyadic Adjustment Scale showed indirect effects on children’s methylation and psychopathological risk in relation to those exerted by risk factors.
Conclusion
Our study has supported the emerging evidence on the complex nature of children’s emotional-behavioral functioning and the associated risk and protective factors, with important implications for the planning of preventive programs.
Collapse
|
6
|
Tu R, Zhang C, Feng L, Wang H, Wang W, Li P. Impact of selenium on cerebellar injury and mRNA expression in offspring of rat exposed to methylmercury. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 223:112584. [PMID: 34365210 DOI: 10.1016/j.ecoenv.2021.112584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/20/2021] [Accepted: 07/29/2021] [Indexed: 06/13/2023]
Abstract
During the fetal development stage, the Central Nervous System (CNS) is particularly sensitive to methylmercury (MeHg). However, the mechanism underlying the antagonistic effect of selenium (Se) on MeHg toxicity is still not fully understood. In this study, female rat models with MeHg and Se co-exposure were developed. Pathological changes in the cerebellum and differential mRNA expression profiles in offspring rats were studied. In the MeHg-exposed group, a large number of Purkinje cells showed pathological changes and mitochondria were significantly swollen; co-exposure with Se significantly improved the structure and organization of the cerebellum. In total, 378 differentially expressed genes (DEGs) (including 284 up-regulated genes and 94 down-regulated genes) in the cerebellum of the MeHg-exposed group and 210 DEGs (including 84 up-regulated genes and 126 down-regulated genes) in the cerebellum of the MeHg+Se co-exposed group were identified. The genes involved in neurotransmitter synthesis and release and calcium ion balance in the cerebellum were significantly up-regulated in the MeHg-exposed group. These genes in the MeHg+Se co-exposed group were not changed or down-regulated. These findings demonstrate that the neurotoxicity caused by MeHg exposure is related to the up-regulation of multiple genes in the nerve signal transduction and calcium ion signal pathways, which are closely related to impairments in cell apoptosis and learning and memory. Supplementation with Se can mitigate the changes to related genes and protect neurons in the mammalian brain (especially the developing cerebellum) from MeHg toxicity. Se provides a potential intervention strategy for MeHg toxicity.
Collapse
Affiliation(s)
- Rui Tu
- State Key Laboratory of Environmental Geochemistry, Institute of Geochemistry, Chinese Academy of Sciences, Guiyang 550081, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control/School of Public Health, Guizhou Medical University, Guiyang 550025, China; Division of Infection Management, Guiyang First People's Hospital, Guiyang 550000, China
| | - Chanchan Zhang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control/School of Public Health, Guizhou Medical University, Guiyang 550025, China
| | - Ling Feng
- Key Laboratory of Environmental Pollution Monitoring and Disease Control/School of Public Health, Guizhou Medical University, Guiyang 550025, China
| | - Huiqun Wang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control/School of Public Health, Guizhou Medical University, Guiyang 550025, China
| | - Wenjuan Wang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control/School of Public Health, Guizhou Medical University, Guiyang 550025, China.
| | - Ping Li
- State Key Laboratory of Environmental Geochemistry, Institute of Geochemistry, Chinese Academy of Sciences, Guiyang 550081, China.
| |
Collapse
|
7
|
Genomic selection signatures in autism spectrum disorder identifies cognitive genomic tradeoff and its relevance in paradoxical phenotypes of deficits versus potentialities. Sci Rep 2021; 11:10245. [PMID: 33986442 PMCID: PMC8119484 DOI: 10.1038/s41598-021-89798-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/26/2021] [Indexed: 11/18/2022] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental disorder characterized by paradoxical phenotypes of deficits as well as gain in brain function. To address this a genomic tradeoff hypothesis was tested and followed up with the biological interaction and evolutionary significance of positively selected ASD risk genes. SFARI database was used to retrieve the ASD risk genes while for population datasets 1000 genome data was used. Common risk SNPs were subjected to machine learning as well as independent tests for selection, followed by Bayesian analysis to identify the cumulative effect of selection on risk SNPs. Functional implication of these positively selected risk SNPs was assessed and subjected to ontology analysis, pertaining to their interaction and enrichment of biological and cellular functions. This was followed by comparative analysis with the ancient genomes to identify their evolutionary patterns. Our results identified significant positive selection signals in 18 ASD risk SNPs. Functional and ontology analysis indicate the role of biological and cellular processes associated with various brain functions. The core of the biological interaction network constitutes genes for cognition and learning while genes in the periphery of the network had direct or indirect impact on brain function. Ancient genome analysis identified de novo and conserved evolutionary selection clusters. The de-novo evolutionary cluster represented genes involved in cognitive function. Relative enrichment of the ASD risk SNPs from the respective evolutionary cluster or biological interaction networks may help in addressing the phenotypic diversity in ASD. This cognitive genomic tradeoff signatures impacting the biological networks can explain the paradoxical phenotypes in ASD.
Collapse
|
8
|
Lewis CR, Breitenstein RS, Henderson A, Sowards HA, Piras IS, Huentelman MJ, Doane LD, Lemery-Chalfant K. Harsh Parenting Predicts Novel HPA Receptor Gene Methylation and NR3C1 Methylation Predicts Cortisol Daily Slope in Middle Childhood. Cell Mol Neurobiol 2021; 41:783-793. [PMID: 32472381 DOI: 10.1007/s10571-020-00885-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/23/2020] [Indexed: 01/16/2023]
Abstract
Adverse experiences in childhood are associated with altered hypothalamic-pituitary-adrenal (HPA) axis function and negative health outcomes throughout life. It is now commonly accepted that abuse and neglect can alter epigenetic regulation of HPA genes. Accumulated evidence suggests harsh parenting practices such as spanking are also strong predictors of negative health outcomes. We predicted harsh parenting at 2.5 years old would predict HPA gene DNA methylation similarly to abuse and neglect, and cortisol output at 8.5 years old. Saliva samples were collected three times a day across 3 days to estimate cortisol diurnal slopes. Methylation was quantified using the Illumina Infinium MethylationEPIC array BeadChip (850 K) with DNA collected from buccal cells. We used principal components analysis to compute a summary statistic for CpG sites across candidate genes. The first and second components were used as outcome variables in mixed linear regression analyses with harsh parenting as a predictor variable. We found harsh parenting significantly predicted methylation of several HPA axis genes, including novel gene associations with AVPRB1, CRHR1, CRHR2, and MC2R (FDR corrected p < 0.05). Further, we found NR3C1 methylation predicted a steeper diurnal cortisol slope. Our results extend the current literature by demonstrating harsh parenting may influence DNA methylation similarly to more extreme early life experiences such as abuse and neglect. Further, we show NR3C1 methylation is associated with diurnal HPA function. Elucidating the molecular consequences of harsh parenting on health can inform best parenting practices and provide potential treatment targets for common complex disorders.
Collapse
Affiliation(s)
- Candace R Lewis
- Neurogenomics, Translational Genomics Research Institute, 445 N 5th St., Phoenix, AZ, 85004, USA.
- Psychology, Arizona State University, Tempe, AZ, USA.
| | | | - Adrienne Henderson
- Neurogenomics, Translational Genomics Research Institute, 445 N 5th St., Phoenix, AZ, 85004, USA
| | | | - Ignazio S Piras
- Neurogenomics, Translational Genomics Research Institute, 445 N 5th St., Phoenix, AZ, 85004, USA
| | - Matthew J Huentelman
- Neurogenomics, Translational Genomics Research Institute, 445 N 5th St., Phoenix, AZ, 85004, USA
| | - Leah D Doane
- Psychology, Arizona State University, Tempe, AZ, USA
| | | |
Collapse
|
9
|
Antonijoan RM, Ferrero-Cafiero JM, Coimbra J, Puntes M, Martínez-Colomer J, Arévalo MI, Mascaró C, Molinero C, Buesa C, Maes T. First-in-Human Randomized Trial to Assess Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of the KDM1A Inhibitor Vafidemstat. CNS Drugs 2021; 35:331-344. [PMID: 33755924 PMCID: PMC7985749 DOI: 10.1007/s40263-021-00797-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/12/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Vafidemstat, an inhibitor of the histone lysine-specific demethylase KDM1A, corrects cognition deficits and behavior alterations in rodent models. Here, we report the results from the first-in-human trial of vafidemstat in healthy young and older adult volunteers. A total of 110 volunteers participated: 87 were treated with vafidemstat and 23 with placebo. OBJECTIVES The study aimed to determine the safety and tolerability of vafidemstat, to characterize its pharmacokinetic and pharmacodynamic profiles, to assess its central nervous system (CNS) exposure, and to acquire the necessary data to select the appropriate doses for long-term treatment of patients with CNS disease in phase II trials. METHODS This single-center, randomized, double-blind, placebo-controlled phase I trial included a single and 5-day repeated dose-escalation and open-label CNS penetration substudy. Primary outcomes were safety and tolerability; secondary outcomes included analysis of the pharmacokinetics and pharmacodynamics, including chemoprobe-based immune analysis of KDM1A target engagement (TE) in peripheral blood mononuclear cells (PBMCs) and platelet monoamine oxidase B (MAOB) inhibition. CNS and cognitive function were also evaluated. RESULTS No severe adverse events (AEs) were reported in the dose-escalation stage. AEs were reported at all dose levels; none were dose dependent, and no significant differences were observed between active treatment and placebo. Biochemistry, urinalysis, vital signs, electrocardiogram, and hematology did not change significantly with dose escalation, with the exception of a transient reduction of platelet counts in an extra dose level incorporated for that purpose. Vafidemstat exhibits rapid oral absorption, approximate dose-proportional exposures, and moderate systemic accumulation after 5 days of treatment. The cerebrospinal fluid-to-plasma unbound ratio demonstrated CNS penetration. Vafidemstat bound KDM1A in PBMCs in a dose-dependent manner. No MAOB inhibition was detected. Vafidemstat did not affect the CNS or cognitive function. CONCLUSIONS Vafidemstat displayed good safety and tolerability. This phase I trial confirmed KDM1A TE and CNS penetration and permitted characterization of platelet dynamics and selection of phase IIa doses. TRIAL REGISTRATION EUDRACT No. 2015-003721-33, filed 30 October 2015.
Collapse
Affiliation(s)
- Rosa María Antonijoan
- Centre d'Investigació del Medicament, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau (IIB-Sant Pau), Barcelona, Spain
- Pharmacology and Therapeutics Department, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Juan Manuel Ferrero-Cafiero
- Centre d'Investigació del Medicament, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Jimena Coimbra
- Centre d'Investigació del Medicament, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Montse Puntes
- Centre d'Investigació del Medicament, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Joan Martínez-Colomer
- Centre d'Investigació del Medicament, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - María Isabel Arévalo
- Oryzon Genomics S.A. Carrer Sant Ferran 74, Cornellà de Llobregat, 08940, Barcelona, Spain
| | - Cristina Mascaró
- Oryzon Genomics S.A. Carrer Sant Ferran 74, Cornellà de Llobregat, 08940, Barcelona, Spain
| | - Cesar Molinero
- Oryzon Genomics S.A. Carrer Sant Ferran 74, Cornellà de Llobregat, 08940, Barcelona, Spain
| | - Carlos Buesa
- Oryzon Genomics S.A. Carrer Sant Ferran 74, Cornellà de Llobregat, 08940, Barcelona, Spain
| | - Tamara Maes
- Oryzon Genomics S.A. Carrer Sant Ferran 74, Cornellà de Llobregat, 08940, Barcelona, Spain.
| |
Collapse
|
10
|
Mustafin RN, Kazantseva AV, Malykh SB, Khusnutdinova EK. Genetic Mechanisms of Cognitive Development. RUSS J GENET+ 2020. [DOI: 10.1134/s102279542007011x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
11
|
Chen J, Zang Z, Braun U, Schwarz K, Harneit A, Kremer T, Ma R, Schweiger J, Moessnang C, Geiger L, Cao H, Degenhardt F, Nöthen MM, Tost H, Meyer-Lindenberg A, Schwarz E. Association of a Reproducible Epigenetic Risk Profile for Schizophrenia With Brain Methylation and Function. JAMA Psychiatry 2020; 77:628-636. [PMID: 32049268 PMCID: PMC7042900 DOI: 10.1001/jamapsychiatry.2019.4792] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
IMPORTANCE Schizophrenia is a severe mental disorder in which epigenetic mechanisms may contribute to illness risk. Epigenetic profiles can be derived from blood cells, but to our knowledge, it is unknown whether these predict established brain alterations associated with schizophrenia. OBJECTIVE To identify an epigenetic signature (quantified as polymethylation score [PMS]) of schizophrenia using machine learning applied to genome-wide blood DNA-methylation data; evaluate whether differences in blood-derived PMS are mirrored in data from postmortem brain samples; test whether the PMS is associated with alterations of dorsolateral prefrontal cortex hippocampal (DLPFC-HC) connectivity during working memory in healthy controls (HC); explore the association between interactions between polygenic and epigenetic risk with DLPFC-HC connectivity; and test the specificity of the signature compared with other serious psychiatric disorders. DESIGN, SETTING, AND PARTICIPANTS In this case-control study conducted from 2008 to 2018 in sites in Germany, the United Kingdom, the United States, and Australia, blood DNA-methylation data from 2230 whole-blood samples from 6 independent cohorts comprising HC (1238 [55.5%]) and participants with schizophrenia (803 [36.0%]), bipolar disorder (39 [1.7%]), major depressive disorder 35 [1.6%]), and autism (27 [1.2%]), and first-degree relatives of all patient groups (88 [3.9%]) were analyzed. DNA-methylation data were further explored from 244 postmortem DLPFC samples from 136 HC and 108 patients with schizophrenia. Neuroimaging and genome-wide association data were available for 393 HC. The latter data was used to calculate a polygenic risk score (PRS) for schizophrenia. The data were analyzed in 2019. MAIN OUTCOMES AND MEASURES The accuracy of schizophrenia control classification based on machine learning using epigenetic data; association of schizophrenia PMS scores with DLPFC-HC connectivity; and association of the interaction between PRS and PMS with DLPFC-HC connectivity. RESULTS This study included 7488 participants (4395 men [58.7%]), of whom 3158 (2230 men [70.6%]) received a diagnosis of schizophrenia. The PMS signature was associated with schizophrenia across 3 independent data sets (area under the curve [AUC] from 0.69 to 0.78; P value from 0.049 to 1.24 × 10-7) and data from postmortem DLPFC samples (AUC = 0.63; P = 1.42 × 10-4), but not with major depressive disorder (AUC = 0.51; P = .16), autism (AUC = 0.53; P = .66), or bipolar disorder (AUC = 0.58; P = .21). Pathways contributing most to the classification included synaptic processes. Healthy controls with schizophrenia-like PMS showed significantly altered DLPFC-HC connectivity (validation methylation/magnetic resonance imaging, t < -3.81; P for familywise error, <.04; validation magnetic resonance imaging, t < -3.54; P for familywise error, <.02), mirroring the lack of functional decoupling in schizophrenia. There was no significant association of the interaction between PMS and PRS with DLPFC-HC connectivity (P > .19). CONCLUSIONS AND RELEVANCE We identified a reproducible blood DNA-methylation signature specific for schizophrenia that was correlated with altered functional DLPFC-HC coupling during working memory and mapped to methylation differences found in DLPFC postmortem samples. This indicates a possible epigenetic contribution to a schizophrenia intermediate phenotype and suggests that PMS could be of interest to be studied in the context of multimodal biomarkers for disease stratification and treatment personalization.
Collapse
Affiliation(s)
- Junfang Chen
- Central Institute of Mental Health, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Zhenxiang Zang
- Central Institute of Mental Health, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Urs Braun
- Central Institute of Mental Health, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kristina Schwarz
- Central Institute of Mental Health, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Anais Harneit
- Central Institute of Mental Health, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Thomas Kremer
- Central Institute of Mental Health, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ren Ma
- Central Institute of Mental Health, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Janina Schweiger
- Central Institute of Mental Health, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Carolin Moessnang
- Central Institute of Mental Health, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lena Geiger
- Central Institute of Mental Health, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Han Cao
- Central Institute of Mental Health, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Franziska Degenhardt
- School of Medicine & University Hospital Bonn, Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Markus M. Nöthen
- School of Medicine & University Hospital Bonn, Institute of Human Genetics, University of Bonn, Bonn, Germany,Life & Brain Center, Department of Genomics, University of Bonn, Bonn, Germany
| | - Heike Tost
- Central Institute of Mental Health, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Meyer-Lindenberg
- Central Institute of Mental Health, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Emanuel Schwarz
- Central Institute of Mental Health, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
12
|
Lewis CR, Sowards HA, Huentelman MJ, Doane LD, Lemery-Chalfant K. Epigenetic differences in inflammation genes of monozygotic twins are related to parent-child emotional availability and health. Brain Behav Immun Health 2020; 5:100084. [PMID: 34589859 PMCID: PMC8474531 DOI: 10.1016/j.bbih.2020.100084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/16/2020] [Indexed: 01/03/2023] Open
Abstract
The inflammatory response is an immune defense engaged immediately after injury or infection. Chronic inflammation can be deleterious for various health outcomes and is characterized by high levels of pro-inflammatory markers such as C-reactive protein (CRP), interleukin 6 (IL-6), and tumor necrosis factor alpha (TNF-α). A large body of research demonstrates these inflammatory markers are responsive to stress and quality of social relationships throughout the lifespan. For example, the quality of the early parental bond predicts various health outcomes and may be driven by changes in immune function. Epigenetic processes, such as DNA methylation, may be one mechanism by which early social experiences shape immune functioning. The present study used a monozygotic twin difference design to assess if mother-reported emotional availability at 1 year and 2.5 years predicted immune gene methylation at 8 years of age. Further, we assessed if inflammation gene methylation was related to general health problems (e.g. infections, allergies, etc.). We found that mother-reported emotional availability at 1 year, but not 2.5 years, was related to methylation of various immune genes in monozygotic twins. Furthermore, twin pairs discordant in health problems have more difference in immune gene methylation compared to twin pairs concordant for health problems, suggesting that methylation of immune genes may have functional consequences for general health. These results suggest that the emotional component of attachment quality during infancy contributes to immune epigenetic profiles in childhood, which may influence general health.
Collapse
Affiliation(s)
- Candace R Lewis
- Translational Genomics Research Institute, Neurogenomics Division, United States.,Arizona State University, Psychology Department, United States
| | | | - Matthew J Huentelman
- Translational Genomics Research Institute, Neurogenomics Division, United States
| | - Leah D Doane
- Arizona State University, Psychology Department, United States
| | | |
Collapse
|
13
|
Abstract
In the last decade, advances in neuroimaging technologies have given rise to a large number of research studies that investigate the neural underpinnings of executive function (EF). EF has long been associated with the prefrontal cortex (PFC) and involves both a unified, general element, as well as the distinct, separable elements of working memory, inhibitory control and set shifting. We will highlight the value of utilising advances in neuroimaging techniques to uncover answers to some of the most pressing questions in the field of early EF development. First, this review will explore the development and neural substrates of each element of EF. Second, the structural, anatomical and biochemical changes that occur in the PFC during infancy and throughout childhood will be examined, in order to address the importance of these changes for the development of EF. Third, the importance of connectivity between regions of the PFC and other brain areas in EF development is reviewed. Finally, throughout this review more recent developments in neuroimaging techniques will be addressed, alongside the implications for further elucidating the neural substrates of early EF development in the future.
Collapse
Affiliation(s)
- Abigail Fiske
- School of Psychology, University of Nottingham, Nottingham, United Kingdom
| | - Karla Holmboe
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|