1
|
Dibaj M, Haghi M, Safaralizadeh R, Saberi A. The role of EZH2 and its regulatory lncRNAs as a serum-based biomarker in Alzheimer's disease. Mol Biol Rep 2024; 51:866. [PMID: 39073683 DOI: 10.1007/s11033-024-09802-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) have become a hot topic in the human nervous system. Moreover, circulating lncRNAs have been suggested as possible biomarkers for central nervous system processes and neurodegenerative diseases. The present research aimed to highlight the role of plasma lncRNAs TUG1, FEZF1-AS1, and EZH2 gene as diagnostic biomarkers in Alzheimer's disease (AD). METHODS Plasma samples for the study were provided by 100 AD patients and 100 matched controls. Real-time quantitative reverse transcriptase PCR was used to determine the plasma level of the aforementioned lncRNAs. Furthermore, the plasma level of EZH2 protein in the participants' blood was determined using the ELISA technique. RESULTS In contrast to controls, down-regulation of the EZH2 gene and protein was reported in the plasma of patients with AD. Additionally, plasma samples from AD patients showed up-and-down-regulation of the lncRNAs TUG1 and FEZF1-AS1, respectively. CONCLUSION Our new findings suggest that the EZH2 gene, plasma lncRNA TUG1, and FEZF1-AS1 may contribute, as valuable biomarkers, to AD diagnosis.
Collapse
Affiliation(s)
- Mohsen Dibaj
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mehdi Haghi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Alia Saberi
- Neurology Department, School of Medicine, Poursina Hospital, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
2
|
Bhuiyan P, Sun Z, Khan MA, Hossain MA, Rahman MH, Qian Y. System biology approaches to identify hub genes linked with ECM organization and inflammatory signaling pathways in schizophrenia pathogenesis. Heliyon 2024; 10:e25191. [PMID: 38322840 PMCID: PMC10844262 DOI: 10.1016/j.heliyon.2024.e25191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 12/18/2023] [Accepted: 01/22/2024] [Indexed: 02/08/2024] Open
Abstract
Schizophrenia (SZ) is a chronic and devastating mental illness that affects around 20 million individuals worldwide. Cognitive deficits and structural and functional changes of the brain, abnormalities of brain ECM components, chronic neuroinflammation, and devastating clinical manifestation during SZ are likely etiological factors shown by affected individuals. However, the pathophysiological events associated with multiple regulatory pathways involved in the brain of this complex disorder are still unclear. This study aimed to develop a pipeline based on bioinformatics and systems biology approaches for identifying potential therapeutic targets involving possible biological mechanisms from SZ patients and healthy volunteers. About 420 overlapping differentially expressed genes (DEGs) from three RNA-seq datasets were identified. Gene ontology (GO), and pathways analysis showed several biological mechanisms enriched by the commonly shared DEGs, including extracellular matrix organization (ECM) organization, collagen fibril organization, integrin signaling pathway, inflammation mediated by chemokines and cytokines signaling pathway, and GABA-B receptor II and IL4 mediated signaling. Besides, 15 hub genes (FN1, COL1A1, COL3A1, COL1A2, COL5A1, COL2A1, COL6A2, COL6A3, MMP2, THBS1, DCN, LUM, HLA-A, HLA-C, and FBN1) were discovered by comprehensive analysis, which was mainly involved in the ECM organization and inflammatory signaling pathway. Furthermore, the miRNA target of the hub genes was analyzed with the random-forest-based approach software miRTarBase. In addition, the transcriptional factors and protein kinases regulating overlapping DEGs in SZ, namely, SUZ12, EZH2, TRIM28, TP53, EGR1, CSNK2A1, GSK3B, CDK1, and MAPK14, were also identified. The results point to a new understanding that the hub genes (fibronectin 1, collagen, matrix metalloproteinase-2, and lumican) in the ECM organization and inflammatory signaling pathways may be involved in the SZ occurrence and pathogenesis.
Collapse
Affiliation(s)
- Piplu Bhuiyan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China
- Department of Biotechnology and Genetic Engineering, Faculty of Life Science, University of Development Alternative, Dhaka, 1209, Bangladesh
| | - Zhaochu Sun
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China
| | - Md Arif Khan
- Department of Biotechnology and Genetic Engineering, Faculty of Life Science, University of Development Alternative, Dhaka, 1209, Bangladesh
- Bio-Bio-1 Bioinformatics Research Foundation, Dhaka, Bangladesh
| | - Md Arju Hossain
- Department of Microbiology, Primeasia University, Banani, Dhaka 1213, Bangladesh
| | - Md Habibur Rahman
- Department of Computer Science and Engineering, Faculty of Engineering and Technology, Islamic University, Kushtia-7003, Bangladesh
| | - Yanning Qian
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China
| |
Collapse
|
3
|
Baker MR, Lee AS, Rajadhyaksha AM. L-type calcium channels and neuropsychiatric diseases: Insights into genetic risk variant-associated genomic regulation and impact on brain development. Channels (Austin) 2023; 17:2176984. [PMID: 36803254 PMCID: PMC9980663 DOI: 10.1080/19336950.2023.2176984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 02/01/2023] [Indexed: 02/21/2023] Open
Abstract
Recent human genetic studies have linked a variety of genetic variants in the CACNA1C and CACNA1D genes to neuropsychiatric and neurodevelopmental disorders. This is not surprising given the work from multiple laboratories using cell and animal models that have established that Cav1.2 and Cav1.3 L-type calcium channels (LTCCs), encoded by CACNA1C and CACNA1D, respectively, play a key role in various neuronal processes that are essential for normal brain development, connectivity, and experience-dependent plasticity. Of the multiple genetic aberrations reported, genome-wide association studies (GWASs) have identified multiple single nucleotide polymorphisms (SNPs) in CACNA1C and CACNA1D that are present within introns, in accordance with the growing body of literature establishing that large numbers of SNPs associated with complex diseases, including neuropsychiatric disorders, are present within non-coding regions. How these intronic SNPs affect gene expression has remained a question. Here, we review recent studies that are beginning to shed light on how neuropsychiatric-linked non-coding genetic variants can impact gene expression via regulation at the genomic and chromatin levels. We additionally review recent studies that are uncovering how altered calcium signaling through LTCCs impact some of the neuronal developmental processes, such as neurogenesis, neuron migration, and neuron differentiation. Together, the described changes in genomic regulation and disruptions in neurodevelopment provide possible mechanisms by which genetic variants of LTCC genes contribute to neuropsychiatric and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Madelyn R. Baker
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, USA
| | - Andrew S. Lee
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, USA
- Developmental Biology Program, Sloan Kettering Institute, New York, USA
| | - Anjali M. Rajadhyaksha
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, USA
- Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA
- Weill Cornell Autism Research Program, Weill Cornell Medicine, New York, USA
| |
Collapse
|
4
|
Mäki-Marttunen T, Blackwell KT, Akkouh I, Shadrin A, Valstad M, Elvsåshagen T, Linne ML, Djurovic S, Einevoll GT, Andreassen OA. Genetic mechanisms for impaired synaptic plasticity in schizophrenia revealed by computational modelling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.14.544920. [PMID: 37398070 PMCID: PMC10312778 DOI: 10.1101/2023.06.14.544920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Schizophrenia phenotypes are suggestive of impaired cortical plasticity in the disease, but the mechanisms of these deficits are unknown. Genomic association studies have implicated a large number of genes that regulate neuromodulation and plasticity, indicating that the plasticity deficits have a genetic origin. Here, we used biochemically detailed computational modelling of post-synaptic plasticity to investigate how schizophrenia-associated genes regulate long-term potentiation (LTP) and depression (LTD). We combined our model with data from post-mortem mRNA expression studies (CommonMind gene-expression datasets) to assess the consequences of altered expression of plasticity-regulating genes for the amplitude of LTP and LTD. Our results show that the expression alterations observed post mortem, especially those in anterior cingulate cortex, lead to impaired PKA-pathway-mediated LTP in synapses containing GluR1 receptors. We validated these findings using a genotyped EEG dataset where polygenic risk scores for synaptic and ion channel-encoding genes as well as modulation of visual evoked potentials (VEP) were determined for 286 healthy controls. Our results provide a possible genetic mechanism for plasticity impairments in schizophrenia, which can lead to improved understanding and, ultimately, treatment of the disorder.
Collapse
Affiliation(s)
- Tuomo Mäki-Marttunen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Kim T Blackwell
- The Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA
| | - Ibrahim Akkouh
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Alexey Shadrin
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Mathias Valstad
- Department of Mental Disorders, Norwegian Institute of Public Health, Oslo, Norway
| | - Tobjørn Elvsåshagen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Norway
| | - Marja-Leena Linne
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Srdjan Djurovic
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Gaute T Einevoll
- Department of Physics, Norwegian University of Life Sciences, Ås, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
5
|
Filippini L, Ortner NJ, Kaserer T, Striessnig J. Ca v 1.3-selective inhibitors of voltage-gated L-type Ca 2+ channels: Fact or (still) fiction? Br J Pharmacol 2023; 180:1289-1303. [PMID: 36788128 PMCID: PMC10953394 DOI: 10.1111/bph.16060] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/17/2022] [Accepted: 01/29/2023] [Indexed: 02/16/2023] Open
Abstract
Voltage-gated L-type Ca2+ -channels (LTCCs) are the target of Ca2+ -channel blockers (CCBs), which are in clinical use for the evidence-based treatment of hypertension and angina. Their cardiovascular effects are largely mediated by the Cav 1.2-subtype. However, based on our current understanding of their physiological and pathophysiological roles, Cav 1.3 LTCCs also appear as attractive drug targets for the therapy of various diseases, including treatment-resistant hypertension, spasticity after spinal cord injury and neuroprotection in Parkinson's disease. Since CCBs inhibit both Cav 1.2 and Cav 1.3, Cav 1.3-selective inhibitors would be valuable tools to validate the therapeutic potential of Cav 1.3 channel inhibition in preclinical models. Despite a number of publications reporting the discovery of Cav 1.3-selective blockers, their selectivity remains controversial. We conclude that at present no pharmacological tools exist that are suitable to confirm or refute a role of Cav 1.3 channels in cellular responses. We also suggest essential criteria for a small molecule to be considered Cav 1.3-selective.
Collapse
Affiliation(s)
- Ludovica Filippini
- Department of Pharmacology and Toxicology and Center of Molecular BiosciencesUniversity of InnsbruckInnsbruckAustria
- Department of Pharmaceutical Chemistry, Institute of PharmacyUniversity of InnsbruckInnsbruckAustria
| | - Nadine J. Ortner
- Department of Pharmacology and Toxicology and Center of Molecular BiosciencesUniversity of InnsbruckInnsbruckAustria
| | - Teresa Kaserer
- Department of Pharmaceutical Chemistry, Institute of PharmacyUniversity of InnsbruckInnsbruckAustria
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology and Center of Molecular BiosciencesUniversity of InnsbruckInnsbruckAustria
| |
Collapse
|
6
|
Doke M, McLaughlin JP, Cai JJ, Pendyala G, Kashanchi F, Khan MA, Samikkannu T. HIV-1 Tat and cocaine impact astrocytic energy reservoirs and epigenetic regulation by influencing the LINC01133-hsa-miR-4726-5p-NDUFA9 axis. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 29:243-258. [PMID: 35892093 PMCID: PMC9307901 DOI: 10.1016/j.omtn.2022.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/04/2022] [Indexed: 06/15/2023]
Abstract
Clinical research has proven that HIV-positive (HIV+) individuals with cocaine abuse show behavioral and neurocognitive disorders. Noncoding RNAs (ncRNAs), such as long ncRNAs (lncRNAs) and microRNAs (miRNAs), are known to regulate gene expression in the contexts of HIV infection and drug abuse. However, there are no specific lncRNA or miRNA biomarkers associated with HIV-1 Transactivator of transcription protein (Tat) and cocaine coexposure. In the central nervous system (CNS), astrocytes are the primary regulators of energy metabolism, and impairment of the astrocytic energy supply can trigger neurodegeneration. The aim of this study was to uncover the roles of lncRNAs and miRNAs in the regulation of messenger RNA (mRNA) targets affected by HIV infection and cocaine abuse. Integrative bioinformatics analysis revealed altered expression of 10 lncRNAs, 10 miRNAs, and 4 mRNA/gene targets in human primary astrocytes treated with cocaine and HIV-1 Tat. We assessed the alterations in the expression of two miRNAs, hsa-miR-2355 and hsa-miR-4726-5p; four lncRNAs, LINC01133, H19, HHIP-AS1, and NOP14-AS1; and four genes, NDUFA9, KYNU, HKDC1, and LIPG. The results revealed interactions in the LINC01133-hsa-miR-4726-5p-NDUFA9 axis that may eventually help us understand cocaine- and HIV-1 Tat-induced astrocyte dysfunction that may ultimately result in neurodegeneration.
Collapse
Affiliation(s)
- Mayur Doke
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, TX 78363, USA
| | - Jay P. McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| | - James J. Cai
- Veterinary Integrative Biosciences, Texas A&M University, TAMU 4458, College Station, TX 77845, USA
| | - Gurudutt Pendyala
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA
| | - Fatah Kashanchi
- National Center for Biodefense and Infectious Disease, Laboratory of Molecular Virology, George Mason University, Manassas, VA 20110, USA
| | - Mansoor A. Khan
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, TX 78363, USA
| | - Thangavel Samikkannu
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, TX 78363, USA
| |
Collapse
|
7
|
Peedicayil J. The relevance of polycomb group proteins to the development of psychiatric disorders. Front Cell Dev Biol 2022; 10:927833. [PMID: 35938156 PMCID: PMC9354779 DOI: 10.3389/fcell.2022.927833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/28/2022] [Indexed: 11/18/2022] Open
|
8
|
Clifton NE, Collado-Torres L, Burke EE, Pardiñas AF, Harwood JC, Di Florio A, Walters JTR, Owen MJ, O'Donovan MC, Weinberger DR, Holmans PA, Jaffe AE, Hall J. Developmental Profile of Psychiatric Risk Associated With Voltage-Gated Cation Channel Activity. Biol Psychiatry 2021; 90:399-408. [PMID: 33965196 PMCID: PMC8375582 DOI: 10.1016/j.biopsych.2021.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 02/26/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Recent breakthroughs in psychiatric genetics have implicated biological pathways onto which genetic risk for psychiatric disorders converges. However, these studies do not reveal the developmental time point(s) at which these pathways are relevant. METHODS We aimed to determine the relationship between psychiatric risk and developmental gene expression relating to discrete biological pathways. We used postmortem RNA sequencing data (BrainSeq and BrainSpan) from brain tissue at multiple prenatal and postnatal time points, with summary statistics from recent genome-wide association studies of schizophrenia, bipolar disorder, and major depressive disorder. We prioritized gene sets for overall enrichment of association with each disorder and then tested the relationship between the association of their constituent genes with their relative expression at each developmental stage. RESULTS We observed relationships between the expression of genes involved in voltage-gated cation channel activity during early midfetal, adolescence, and early adulthood time points and association with schizophrenia and bipolar disorder, such that genes more strongly associated with these disorders had relatively low expression during early midfetal development and higher expression during adolescence and early adulthood. The relationship with schizophrenia was strongest for the subset of genes related to calcium channel activity, while for bipolar disorder, the relationship was distributed between calcium and potassium channel activity genes. CONCLUSIONS Our results indicate periods during development when biological pathways related to the activity of calcium and potassium channels may be most vulnerable to the effects of genetic variants conferring risk for psychiatric disorders. Furthermore, they indicate key time points and potential targets for disorder-specific therapeutic interventions.
Collapse
Affiliation(s)
- Nicholas E Clifton
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom; MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom.
| | - Leonardo Collado-Torres
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland; Centre for Computational Biology, Johns Hopkins University Medical Campus, Baltimore, Maryland
| | - Emily E Burke
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland
| | - Antonio F Pardiñas
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Janet C Harwood
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Arianna Di Florio
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - James T R Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Michael J Owen
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom; MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Michael C O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland; Departments of Psychiatry, Neurology, Neuroscience and Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Peter A Holmans
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Andrew E Jaffe
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland; Centre for Computational Biology, Johns Hopkins University Medical Campus, Baltimore, Maryland; Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom; MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
9
|
Abashkin DA, Kurishev AO, Karpov DS, Golimbet VE. Cellular Models in Schizophrenia Research. Int J Mol Sci 2021; 22:ijms22168518. [PMID: 34445221 PMCID: PMC8395162 DOI: 10.3390/ijms22168518] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia (SZ) is a prevalent functional psychosis characterized by clinical behavioural symptoms and underlying abnormalities in brain function. Genome-wide association studies (GWAS) of schizophrenia have revealed many loci that do not directly identify processes disturbed in the disease. For this reason, the development of cellular models containing SZ-associated variations has become a focus in the post-GWAS research era. The application of revolutionary clustered regularly interspaced palindromic repeats CRISPR/Cas9 gene-editing tools, along with recently developed technologies for cultivating brain organoids in vitro, have opened new perspectives for the construction of these models. In general, cellular models are intended to unravel particular biological phenomena. They can provide the missing link between schizophrenia-related phenotypic features (such as transcriptional dysregulation, oxidative stress and synaptic dysregulation) and data from pathomorphological, electrophysiological and behavioural studies. The objectives of this review are the systematization and classification of cellular models of schizophrenia, based on their complexity and validity for understanding schizophrenia-related phenotypes.
Collapse
Affiliation(s)
- Dmitrii A. Abashkin
- Mental Health Research Center, Clinical Genetics Laboratory, Kashirskoe Sh. 34, 115522 Moscow, Russia; (D.A.A.); (A.O.K.); (D.S.K.)
| | - Artemii O. Kurishev
- Mental Health Research Center, Clinical Genetics Laboratory, Kashirskoe Sh. 34, 115522 Moscow, Russia; (D.A.A.); (A.O.K.); (D.S.K.)
| | - Dmitry S. Karpov
- Mental Health Research Center, Clinical Genetics Laboratory, Kashirskoe Sh. 34, 115522 Moscow, Russia; (D.A.A.); (A.O.K.); (D.S.K.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, 119991 Moscow, Russia
| | - Vera E. Golimbet
- Mental Health Research Center, Clinical Genetics Laboratory, Kashirskoe Sh. 34, 115522 Moscow, Russia; (D.A.A.); (A.O.K.); (D.S.K.)
- Correspondence:
| |
Collapse
|
10
|
Shulman ED, Elkon R. Genetic mapping of developmental trajectories for complex traits and diseases. Comput Struct Biotechnol J 2021; 19:3458-3469. [PMID: 34194671 PMCID: PMC8220172 DOI: 10.1016/j.csbj.2021.05.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/30/2021] [Accepted: 05/30/2021] [Indexed: 11/04/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified numerous common genetic variants associated with complex human traits and diseases. However, the translation of GWAS discoveries into biological and clinical insights is highly challenging. In this study, we present a novel bioinformatics approach for enhancing the functional interpretation of GWAS signals, based on their integration with single-cell (sc)RNA-seq datasets that examine developmental processes. Our approach performs three tasks: (1) Identification of links between cell differentiation trajectories and traits; (2) Elucidation of biological processes and molecular pathways that underlie such trajectory-trait links; and (3) Prioritization of target genes that carry the links between trajectories, pathways and traits. We applied our method to a set of 11 traits of various pathologies, and 12 scRNA-seq datasets of diverse developmental processes, and it readily detected well-established biological connections, including those between the maturation of cortical inhibitory interneurons and schizophrenia, hepatocytes and cholesterol levels, and pancreatic beta-islet cells and type-2 diabetes. For each of these associations, our method pinpointed top candidate genes that are strongly associated with both the kinetics of the differentiation trajectory and the disease's genetic risk. By the identification of trajectory-disease links, molecular pathways that underlie them and prioritizing candidate risk genes, our method improves the understanding of the etiology of complex diseases, and thus holds promise for enhancing rational drug development that is aimed at targeting specific biological processes that mediate the genetic predisposition to diseases.
Collapse
Affiliation(s)
- Eldad David Shulman
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ran Elkon
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
11
|
Mishra P, Kumar S. Association of lncRNA with regulatory molecular factors in brain and their role in the pathophysiology of schizophrenia. Metab Brain Dis 2021; 36:849-858. [PMID: 33608830 DOI: 10.1007/s11011-021-00692-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/11/2021] [Indexed: 01/12/2023]
Abstract
Schizophrenia is one of the most agonizing neurodegenerative diseases of the brain. Research undertaken to understand the molecular mechanism of this disease has undergone a transition and currently more emphasis is put on long noncoding RNA (lncRNA). High expression level of lncRNA in the brain contributes to several molecular pathways essential for the proper functioning of neurons, neurotransmitters, and synapses, that are often found dysfunctional in Schizophrenia. Recently, the association of lncRNA with various molecular factors in the brain has been explored to a considerably large extent. This review comprehends the significance of lncRNA in causing profound regulatory effect in the brain and how any alterations to the association of lncRNA with regulatory proteins, enzymes and other noncoding RNA could contribute to the aetiology of Schizophrenia.
Collapse
Affiliation(s)
- Parinita Mishra
- Life Science Department, National Institute of Technology, Rourkela, Odisha, 769008, India
| | - Santosh Kumar
- Life Science Department, National Institute of Technology, Rourkela, Odisha, 769008, India.
| |
Collapse
|
12
|
Matsuda VDV, Tejada MB, Motta-Teixeira LC, Ikebara JM, Cardoso DS, Machado-Nils AV, Lee VY, Diccini I, Arruda BP, Martins PP, Dias NMM, Tessarotto RP, Raeisossadati R, Bruno M, Takase LF, Kihara AH, Nogueira MI, Xavier GF, Takada SH. Impact of neonatal anoxia and hypothermic treatment on development and memory of rats. Exp Neurol 2021; 340:113691. [PMID: 33713657 DOI: 10.1016/j.expneurol.2021.113691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 02/18/2021] [Accepted: 03/07/2021] [Indexed: 01/02/2023]
Abstract
Therapeutic hypothermia (TH) is well established as a standard treatment for term and near-term infants. However, therapeutic effects of hypothermia following neonatal anoxia in very premature babies remains inconclusive. The present rodent model of preterm neonatal anoxia has been shown to alter developmental milestones and hippocampal neurogenesis, and to disrupt spatial learning and memory in adulthood. These effects seem to be reduced by post-insult hypothermia. Epigenetic-related mechanisms have been postulated as valuable tools for developing new therapies. Dentate gyrus neurogenesis is regulated by epigenetic factors. This study evaluated whether TH effects in a rodent model of preterm oxygen deprivation are based on epigenetic alterations. The effects of TH on both developmental features (somatic growth, maturation of physical characteristics and early neurological reflexes) and performance of behavioral tasks at adulthood (spatial reference and working memory, and fear conditioning) were investigated in association with the possible involvement of the epigenetic operator Enhancer of zeste homolog 2 (Ezh2), possibly related to long-lasting effects on hippocampal neurogenesis. Results showed that TH reduced both anoxia-induced hippocampal neurodegeneration and anoxia-induced impairments on risk assessment behavior, acquisition of spatial memory, and extinction of auditory and contextual fear conditioning. In contrast, TH did not prevent developmental alterations caused by neonatal anoxia and did not restore hippocampal neurogenesis or cause changes in EZH2 levels. In conclusion, despite the beneficial effects of TH in hippocampal neurodegeneration and in reversing disruption of performance of behavioral tasks following oxygen deprivation in prematurity, these effects seem not related to developmental alterations and hippocampal neurogenesis and, apparently, is not caused by Ezh2-mediated epigenetic alteration.
Collapse
Affiliation(s)
- Victor Daniel Vasquez Matsuda
- Neuroscience and Behaviour Laboratory, Department of Physiology, Institute of Biosciences, Universidade de São Paulo, São Paulo, SP, Brazil; Neuroscience Laboratory, Department of Anatomy, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Martin Bustelo Tejada
- Department of Pediatrics, Maastricht University Medical Center (MUMC), Maastricht, the Netherlands; Experimental Neuropathology Laboratory, Institute of Cellular Biology and Neuroscience "Prof. E. De Robertis" (IBCN), Faculty of Medicine, University of Buenos Aires, CONICET, Buenos Aires, Argentina; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, the Netherlands; Biomedical Sciences Institute, Faculty of Medical Sciences, Catholic University of Cuyo, San Juan, Argentina; Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Lívia Clemente Motta-Teixeira
- Neuroscience and Behaviour Laboratory, Department of Physiology, Institute of Biosciences, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Juliane Midori Ikebara
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | | | - Aline Vilar Machado-Nils
- Neuroscience and Behaviour Laboratory, Department of Physiology, Institute of Biosciences, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Vitor Yonamine Lee
- Neuroscience Laboratory, Department of Anatomy, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Isabelle Diccini
- Neuroscience Laboratory, Department of Anatomy, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Bruna Petrucelli Arruda
- Neuroscience Laboratory, Department of Anatomy, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, SP, Brazil; Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | | | | | | | - Reza Raeisossadati
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Martin Bruno
- Biomedical Sciences Institute, Faculty of Medical Sciences, Catholic University of Cuyo, San Juan, Argentina; National Council of Scientific and Technical Research (CONICET), Argentina
| | - Luiz Fernando Takase
- Department of Morphology and Pathology, Biological Sciences and Health Center, Universidade Federal de São Carlos, São Carlos, Brazil
| | | | - Maria Inês Nogueira
- Neuroscience Laboratory, Department of Anatomy, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Gilberto Fernando Xavier
- Neuroscience and Behaviour Laboratory, Department of Physiology, Institute of Biosciences, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Silvia Honda Takada
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil.
| |
Collapse
|
13
|
Han JW, Kang C, Kim Y, Lee MG, Kim JY. Isoproterenol-induced hypertrophy of neonatal cardiac myocytes and H9c2 cell is dependent on TRPC3-regulated Ca V1.2 expression. Cell Calcium 2020; 92:102305. [PMID: 33069962 DOI: 10.1016/j.ceca.2020.102305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023]
Abstract
CaV1.2 and transient receptor potential canonical channel 3 (TRPC3) are two proteins known to have important roles in pathological cardiac hypertrophy; however, such roles still remain unclear. A better understanding of these roles is important for furthering the clinical understanding of heart failure. We previously reported that Trpc3-knockout (KO) mice are resistant to pathologic hypertrophy and that their CaV1.2 protein expression is reduced. In this study, we aimed to examine the relationship between these two proteins and characterize their role in neonatal cardiomyocytes. We measured CaV1.2 expression in the hearts of wild-type (WT) and Trpc3-/- mice, and examined the effects of Trpc3 knockdown and overexpression in the rat cell line H9c2. We also compared the hypertrophic responses of neonatal cardiomyocytes cultured from Trpc3-/- mice to a representative hypertrophy-causing drug, isoproterenol (ISO), and measured the activity of nuclear factor of activated T cells 3 (NFAT3) in neonatal cardiomyocytes (NCMCs). We inhibited the L-type current with nifedipine, and measured the intracellular calcium concentration using Fura-2 with 1-oleoyl-2-acetyl-sn-glycerol (OAG)-induced Ba2+ influx. When using the Trpc3-mediated Ca2+ influx, both intracellular calcium concentration and calcium influx were reduced in Trpc3-KO myocytes. Not only was the expression of CaV1.2 greatly reduced in Trpc3-KO cardiac lysate, but the size of the CaV1.2 currents in NCMCs was also greatly reduced. When NCMCs were treated with Trpc3 siRNA, it was confirmed that the expression of CaV1.2 and the intracellular nuclear transfer activity of NFAT decreased. In H9c2 cells, the ISO activated- and verapamil inhibited- Ca2+ influxes were dramatically attenuated by Trpc3 siRNA treatment. In addition, it was confirmed that both the expression of CaV1.2 and the size of H9c2 cells were regulated according to the expression and activation level of TRPC3. We found that after stimulation with ISO, cell hypertrophy occurred in WT myocytes, while the increase in size of Trpc3-KO myocytes was greatly reduced. These results suggest that not only the cell hypertrophy process in neonatal cardiac myocytes and H9c2 cells were regulated according to the expression level of CaV1.2, but also that the expression level of CaV1.2 was regulated by TRPC3 through the activation of NFAT.
Collapse
Affiliation(s)
- Jung Woo Han
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, South Korea.
| | - Choeun Kang
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, South Korea.
| | - Yonjung Kim
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, South Korea.
| | - Min Goo Lee
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, South Korea.
| | - Joo Young Kim
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, South Korea.
| |
Collapse
|
14
|
Analysis of transcript levels of a few schizophrenia candidate genes in neurons from a transgenic mouse embryonic stem cell model overexpressing DNMT1. Gene 2020; 757:144934. [PMID: 32640307 DOI: 10.1016/j.gene.2020.144934] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/23/2020] [Accepted: 07/01/2020] [Indexed: 02/08/2023]
Abstract
Overexpression of DNA Methyltransferase I (DNMT1) is considered as one of the etiological factors for schizophrenia (SZ). However, information on genes subjected to dysregulation because of DNMT1 overexpression is limited. To test whether a larger group of SZ-associated genes are affected, we selected 15 genes reported to be dysregulated in patients (Gad1, Reln, Ank3, Cacna1c, Dkk3, As3mt, Ppp1r11, Smad5, Syn1, Wnt1, Pdgfra, Gsk3b, Cxcl12, Tcf4 and Fez1). Transcript levels of these genes were compared between neurons derived from Dnmt1tet/tet (Tet/Tet) mouse embryonic stem cells (ESCs) that overexpress DNMT1 with R1 (wild-type) neurons. Transcript levels of thirteen genes were significantly altered in Tet/Tet neurons of which, the dysregulation patterns of 11 were similar to patients. Transcript levels of eight out of these eleven were also significantly altered in Tet/Tet ESCs, but the dysregulation patterns of only five were similar to neurons. Comparative analyses among ESCs, embryoid bodies and neurons divided the 15 genes into four distinct groups with a majority showing developmental stage-specific patterns of dysregulation. Reduced Representational Bisulfite Sequencing data from neurons did not show any altered promoter DNA methylation for the dysregulated genes. Doxycycline treatment of Tet/Tet ESCs that eliminated DNMT1, reversed the direction of dysregulation of only four genes (Gad1, Dkk3, As3mt and Syn1). These results suggest that 1. Increased DNMT1 affected the levels of a majority of the transcripts studied, 2. Dysregulation appears to be independent of promoter methylation, 3. Effects of increased DNMT1 levels were reversible for only a subset of the genes studied, and 4. Increased DNMT1 levels may affect transcript levels of multiple schizophrenia-associated genes.
Collapse
|
15
|
Gardea-Resendez M, Kucuker MU, Blacker CJ, Ho AMC, Croarkin PE, Frye MA, Veldic M. Dissecting the Epigenetic Changes Induced by Non-Antipsychotic Mood Stabilizers on Schizophrenia and Affective Disorders: A Systematic Review. Front Pharmacol 2020; 11:467. [PMID: 32390836 PMCID: PMC7189731 DOI: 10.3389/fphar.2020.00467] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/25/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Epimutations secondary to gene-environment interactions have a key role in the pathophysiology of major psychiatric disorders. In vivo and in vitro evidence suggest that mood stabilizers can potentially reverse epigenetic deregulations found in patients with schizophrenia or mood disorders through mechanisms that are not yet fully understood. However, their activity on epigenetic processes has made them a research target for therapeutic approaches. METHODS We conducted a comprehensive literature search of PubMed and EMBASE for studies investigating the specific epigenetic changes induced by non-antipsychotic mood stabilizers (valproate, lithium, lamotrigine, and carbamazepine) in animal models, human cell lines, or patients with schizophrenia, bipolar disorder, or major depressive disorder. Each paper was reviewed for the nature of research, the species and tissue examined, sample size, mood stabilizer, targeted gene, epigenetic changes found, and associated psychiatric disorder. Every article was appraised for quality using a modified published process and those who met a quality score of moderate or high were included. RESULTS A total of 2,429 records were identified; 1,956 records remained after duplicates were removed and were screened via title, abstract and keywords; 129 records were selected for full-text screening and a remaining of 38 articles were included in the qualitative synthesis. Valproate and lithium were found to induce broader epigenetic changes through different mechanisms, mainly DNA demethylation and histones acetylation. There was less literature and hence smaller effects attributable to lamotrigine and carbamazepine could be associated overall with the small number of studies on these agents. Findings were congruent across sample types. CONCLUSIONS An advanced understanding of the specific epigenetic changes induced by classic mood stabilizers in patients with major psychiatric disorders will facilitate personalized interventions. Further related drug discovery should target the induction of selective chromatin remodeling and gene-specific expression effects.
Collapse
Affiliation(s)
| | - Mehmet Utku Kucuker
- Department of Psychiatry and Psychology, Mayo Clinic Depression Center, Mayo Clinic, Rochester, MN, United States
| | - Caren J. Blacker
- Department of Psychiatry and Psychology, Mayo Clinic Depression Center, Mayo Clinic, Rochester, MN, United States
| | - Ada M.-C. Ho
- Department of Psychiatry and Psychology, Mayo Clinic Depression Center, Mayo Clinic, Rochester, MN, United States
| | - Paul E. Croarkin
- Department of Psychiatry and Psychology, Mayo Clinic Depression Center, Mayo Clinic, Rochester, MN, United States
| | - Mark A. Frye
- Department of Psychiatry and Psychology, Mayo Clinic Depression Center, Mayo Clinic, Rochester, MN, United States
| | - Marin Veldic
- Department of Psychiatry and Psychology, Mayo Clinic Depression Center, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
16
|
Osborne AJ, Pearson JF, Noble AJ, Gemmell NJ, Horwood LJ, Boden JM, Benton MC, Macartney-Coxson DP, Kennedy MA. Genome-wide DNA methylation analysis of heavy cannabis exposure in a New Zealand longitudinal cohort. Transl Psychiatry 2020; 10:114. [PMID: 32321915 PMCID: PMC7176736 DOI: 10.1038/s41398-020-0800-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/11/2020] [Accepted: 03/20/2020] [Indexed: 12/24/2022] Open
Abstract
Cannabis use is of increasing public health interest globally. Here we examined the effect of heavy cannabis use, with and without tobacco, on genome-wide DNA methylation in a longitudinal birth cohort (Christchurch Health and Development Study, CHDS). A total of 48 heavy cannabis users were selected from the CHDS cohort, on the basis of their adult exposure to cannabis and tobacco, and DNA methylation assessed from whole blood samples, collected at approximately age 28. Methylation in heavy cannabis users was assessed, relative to non-users (n = 48 controls) via the Illumina Infinium® MethylationEPIC BeadChip. We found the most differentially methylated sites in cannabis with tobacco users were in the AHRR and F2RL3 genes, replicating previous studies on the effects of tobacco. Cannabis-only users had no evidence of differential methylation in these genes, or at any other loci at the epigenome-wide significance level (P < 10-7). However, there were 521 sites differentially methylated at P < 0.001 which were enriched for genes involved in neuronal signalling (glutamatergic synapse and long-term potentiation) and cardiomyopathy. Further, the most differentially methylated loci were associated with genes with reported roles in brain function (e.g. TMEM190, MUC3L, CDC20 and SP9). We conclude that the effects of cannabis use on the mature human blood methylome differ from, and are less pronounced than, the effects of tobacco use, and that larger sample sizes are required to investigate this further.
Collapse
Affiliation(s)
- Amy J. Osborne
- grid.21006.350000 0001 2179 4063School of Biological Sciences, University of Canterbury, Christchurch, 8041 New Zealand
| | - John F. Pearson
- grid.29980.3a0000 0004 1936 7830Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, 8011 New Zealand
| | - Alexandra J. Noble
- grid.21006.350000 0001 2179 4063School of Biological Sciences, University of Canterbury, Christchurch, 8041 New Zealand
| | - Neil J. Gemmell
- grid.29980.3a0000 0004 1936 7830Department of Anatomy, Otago School of Medical Sciences, University of Otago, Dunedin, 9054 New Zealand
| | - L. John Horwood
- grid.29980.3a0000 0004 1936 7830Department of Psychological Medicine, University of Otago Christchurch, Christchurch, 8011 New Zealand
| | - Joseph M. Boden
- grid.29980.3a0000 0004 1936 7830Department of Psychological Medicine, University of Otago Christchurch, Christchurch, 8011 New Zealand
| | - Miles C. Benton
- grid.419706.d0000 0001 2234 622XHuman Genomics, Institute of Environmental Science and Research, Kenepuru Science Centre, Porirua, 5240 New Zealand
| | - Donia P. Macartney-Coxson
- grid.419706.d0000 0001 2234 622XHuman Genomics, Institute of Environmental Science and Research, Kenepuru Science Centre, Porirua, 5240 New Zealand
| | - Martin A. Kennedy
- grid.29980.3a0000 0004 1936 7830Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, 8011 New Zealand
| |
Collapse
|
17
|
van Loo KMJ, Becker AJ. Transcriptional Regulation of Channelopathies in Genetic and Acquired Epilepsies. Front Cell Neurosci 2020; 13:587. [PMID: 31992970 PMCID: PMC6971179 DOI: 10.3389/fncel.2019.00587] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/23/2019] [Indexed: 01/03/2023] Open
Abstract
Epilepsy is a common neurological disorder characterized by recurrent uncontrolled seizures and has an idiopathic “genetic” etiology or a symptomatic “acquired” component. Genetic studies have revealed that many epilepsy susceptibility genes encode ion channels, including voltage-gated sodium, potassium and calcium channels. The high prevalence of ion channels in epilepsy pathogenesis led to the causative concept of “ion channelopathies,” which can be elicited by specific mutations in the coding or promoter regions of genes in genetic epilepsies. Intriguingly, expression changes of the same ion channel genes by augmentation of specific transcription factors (TFs) early after an insult can underlie acquired epilepsies. In this study, we review how the transcriptional regulation of ion channels in both genetic and acquired epilepsies can be controlled, and compare these epilepsy “ion channelopathies” with other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Karen M J van Loo
- Department of Neuropathology, Section for Translational Epilepsy Research, University of Bonn Medical Center, Bonn, Germany
| | - Albert J Becker
- Department of Neuropathology, Section for Translational Epilepsy Research, University of Bonn Medical Center, Bonn, Germany
| |
Collapse
|
18
|
Fallah H, Azari I, Neishabouri SM, Oskooei VK, Taheri M, Ghafouri-Fard S. Sex-specific up-regulation of lncRNAs in peripheral blood of patients with schizophrenia. Sci Rep 2019; 9:12737. [PMID: 31484957 PMCID: PMC6726592 DOI: 10.1038/s41598-019-49265-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/22/2019] [Indexed: 02/06/2023] Open
Abstract
Schizophrenia as a common disabling psychiatric disorder has been associated with dysregulation of several genes and pathways among them are those being regulated by long non-coding RNAs (lncRNAs). Based on the acknowledged roles of lncRNAs in neurodevelopment, in the current study, we assessed expression of six lncRNAs namely HOXA-AS2, Linc-ROR, MALAT1, MEG3, SPRY4-IT1 and UCA1 in peripheral blood of 60 patients with schizophrenia and 60 healthy subjects. HOXA-AS2, Linc-ROR, MEG3, SPRY4-IT1 and UCA1 levels were significantly higher in total patients compared with total controls. However, when evaluating expression of genes in sex-based subgroups, the differences in the expression of these lncRNAs were significant only among females. Assessment of partial correlation between expression of lncRNAs and age of study participants after controlling the effect of sex, revealed significant correlations for HOXA-AS2, MALAT1 and UCA1 in both patients and controls. Besides, expressions of Linc-ROR and SPRY4-IT1 were correlated with age only in patients. Significant pairwise correlations were recognized between expression levels of lncRNAs in both patients with schizophrenia and controls. Based on the area under curve (AUC) values, SPRY4-IT1 had the best performance in differentiation of female patients with schizophrenia from female controls (AUC = 0.85, P < 0.0001). Combination of Linc-ROR, MEG3, SPRY4-IT1 and UCA1 expression levels could differentiate female patients with 95.2% sensitivity, 76.9% specificity and diagnostic power of 0.88 (P < 0.0001). The current study suggests the presence of a sex-based dysregulation of lncRNAs in patients with schizophrenia and their possible application as diagnostic biomarkers.
Collapse
Affiliation(s)
- Hamid Fallah
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Iman Azari
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Vahid Kholghi Oskooei
- Department of Laboratory Sciences, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
- Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Quinn JP, Savage AL, Bubb VJ. Non-coding genetic variation shaping mental health. Curr Opin Psychol 2018; 27:18-24. [PMID: 30099302 PMCID: PMC6624474 DOI: 10.1016/j.copsyc.2018.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 07/16/2018] [Indexed: 12/12/2022]
Abstract
Gene expression determined by the genome mediating a response to cell environment. Genetic variation results in distinct individual response in gene expression. Non-coding DNA is an important site for such functional genetic variation. Gene expression is a major modulator of brain chemistry and thus behavior.
Over 98% of our genome is non-coding and is now recognised to have a major role in orchestrating the tissue specific and stimulus inducible gene expression pattern which underpins our wellbeing and mental health. The non-coding genome responds functionally to our environment at all levels, encompassing the span from psychological to physiological challenge. The gene expression pattern, termed the transcriptome, ultimately gives us our neurochemistry. Therefore a major modulator of mental wellbeing is how our genes are regulated in response to life experiences. Superimposed on the aforementioned non-coding DNA framework is a vast body of genetic variation in the elements that control response to challenges. These differences, termed polymorphisms, allow for a differential response from a specific DNA element to the same challenge thus potentially allowing ‘individuality’ in the modulation of our transcriptome. This review will focus on a fundamental mechanism defining our psychological and psychiatric wellbeing, namely how genetic variation can be correlated with differential gene expression in response to specific challenges, thus resulting in altered neurochemistry which consequently may shape behaviour.
Collapse
Affiliation(s)
- John P Quinn
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, The University of Liverpool, Liverpool L69 3BX, UK.
| | - Abigail L Savage
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, The University of Liverpool, Liverpool L69 3BX, UK
| | - Vivien J Bubb
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, The University of Liverpool, Liverpool L69 3BX, UK
| |
Collapse
|