1
|
Fan S, Liu J, Chofflet N, Bailey AO, Russell WK, Zhang Z, Takahashi H, Ren G, Rudenko G. Molecular mechanism of contactin 2 homophilic interaction. Structure 2024; 32:1652-1666.e8. [PMID: 38968938 PMCID: PMC11455609 DOI: 10.1016/j.str.2024.06.004] [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: 04/12/2024] [Revised: 05/23/2024] [Accepted: 06/10/2024] [Indexed: 07/07/2024]
Abstract
Contactin 2 (CNTN2) is a cell adhesion molecule involved in axon guidance, neuronal migration, and fasciculation. The ectodomains of CNTN1-CNTN6 are composed of six Ig domains (Ig1-Ig6) and four FN domains. Here, we show that CNTN2 forms transient homophilic interactions (KD ∼200 nM). Cryo-EM structures of full-length CNTN2 and CNTN2_Ig1-Ig6 reveal a T-shaped homodimer formed by intertwined, parallel monomers. Unexpectedly, the horseshoe-shaped Ig1-Ig4 headpieces extend their Ig2-Ig3 tips outwards on either side of the homodimer, while Ig4, Ig5, Ig6, and the FN domains form a central stalk. Cross-linking mass spectrometry and cell-based binding assays confirm the 3D assembly of the CNTN2 homodimer. The interface mediating homodimer formation differs between CNTNs, as do the homophilic versus heterophilic interaction mechanisms. The CNTN family thus encodes a versatile molecular platform that supports a very diverse portfolio of protein interactions and that can be leveraged to strategically guide neural circuit development.
Collapse
Affiliation(s)
- Shanghua Fan
- Department of Pharmacology and Toxicology; University of Texas Medical Branch, Galveston, TX 77555, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jianfang Liu
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| | - Nicolas Chofflet
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 2B2, Canada
| | - Aaron O Bailey
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - William K Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ziqi Zhang
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada
| | - Hideto Takahashi
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada; Division of Experimental Medicine, McGill University, Montreal, QC H3A 0G4, Canada.
| | - Gang Ren
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| | - Gabby Rudenko
- Department of Pharmacology and Toxicology; University of Texas Medical Branch, Galveston, TX 77555, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
2
|
Taki S, Boron WF, Moss FJ. Novel RPTPγ and RPTPζ splice variants from mixed neuron-astrocyte hippocampal cultures as well as from the hippocampi of newborn and adult mice. Front Physiol 2024; 15:1406448. [PMID: 38952869 PMCID: PMC11215419 DOI: 10.3389/fphys.2024.1406448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/06/2024] [Indexed: 07/03/2024] Open
Abstract
Receptor protein tyrosine phosphatases γ and ζ (RPTPγ and RPTPζ) are transmembrane signaling proteins with extracellular carbonic anhydrase-like domains that play vital roles in the development and functioning of the central nervous system (CNS) and are implicated in tumor suppression, neurodegeneration, and sensing of extracellular [CO2] and [HCO3 -]. RPTPγ expresses throughout the body, whereas RPTPζ preferentially expresses in the CNS. Here, we investigate differential RPTPγ-RPTPζ expression in three sources derived from a wild-type laboratory strain of C57BL/6 mice: (a) mixed neuron-astrocyte hippocampal (HC) cultures 14 days post isolation from P0-P2 pups; (b) P0-P2 pup hippocampi; and (c) 9- to 12-week-old adult hippocampi. Regarding RPTPγ, we detect the Ptprg variant-1 (V1) transcript, representing canonical exons 1-30. Moreover, we newly validate the hypothetical assembly [XM_006517956] (propose name, Ptprg-V3), which lacks exon 14. Both transcripts are in all three HC sources. Regarding RPTPζ, we confirm the expression of Ptprz1-V1, detecting it in pups and adults but not in cultures, and Ptprz1-V3 through Ptprz1-V7 in all three preparations. We newly validate hypothetical assemblies Ptprz1-X1 (in cultures and pups), Ptprz1-X2 (in all three), and Ptprz1-X5 (in pups and adults) and propose to re-designate them as Ptprz1-V0, Ptprz1-V2, and Ptprz1-V8, respectively. The diversity of RPTPγ and RPTPζ splice variants likely corresponds to distinct signaling functions, in different cellular compartments, during development vs later life. In contrast to previous studies that report divergent RPTPγ and RPTPζ protein expressions in neurons and sometimes in the glia, we observe that RPTPγ and RPTPζ co-express in the somata and processes of almost all HC neurons but not in astrocytes, in all three HC preparations.
Collapse
Affiliation(s)
- Sara Taki
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Walter F. Boron
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Fraser J. Moss
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
3
|
Bamford RA, Zuko A, Eve M, Sprengers JJ, Post H, Taggenbrock RLRE, Fäβler D, Mehr A, Jones OJR, Kudzinskas A, Gandawijaya J, Müller UC, Kas MJH, Burbach JPH, Oguro-Ando A. CNTN4 modulates neural elongation through interplay with APP. Open Biol 2024; 14:240018. [PMID: 38745463 PMCID: PMC11293442 DOI: 10.1098/rsob.240018] [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: 01/23/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 05/16/2024] Open
Abstract
The neuronal cell adhesion molecule contactin-4 (CNTN4) is genetically associated with autism spectrum disorder (ASD) and other psychiatric disorders. Cntn4-deficient mouse models have previously shown that CNTN4 plays important roles in axon guidance and synaptic plasticity in the hippocampus. However, the pathogenesis and functional role of CNTN4 in the cortex has not yet been investigated. Our study found a reduction in cortical thickness in the motor cortex of Cntn4 -/- mice, but cortical cell migration and differentiation were unaffected. Significant morphological changes were observed in neurons in the M1 region of the motor cortex, indicating that CNTN4 is also involved in the morphology and spine density of neurons in the motor cortex. Furthermore, mass spectrometry analysis identified an interaction partner for CNTN4, confirming an interaction between CNTN4 and amyloid-precursor protein (APP). Knockout human cells for CNTN4 and/or APP revealed a relationship between CNTN4 and APP. This study demonstrates that CNTN4 contributes to cortical development and that binding and interplay with APP controls neural elongation. This is an important finding for understanding the physiological function of APP, a key protein for Alzheimer's disease. The binding between CNTN4 and APP, which is involved in neurodevelopment, is essential for healthy nerve outgrowth.
Collapse
Affiliation(s)
- Rosemary A. Bamford
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
| | - Amila Zuko
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Madeline Eve
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
| | - Jan J. Sprengers
- Department of Translational Neuroscience, UMC Utrecht Brain Center, UMC Utrecht, Utrecht3508 AB, The Netherlands
| | - Harm Post
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht, Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Netherlands Proteomics Centre, Utrecht, The Netherlands
| | - Renske L. R. E. Taggenbrock
- Department of Translational Neuroscience, UMC Utrecht Brain Center, UMC Utrecht, Utrecht3508 AB, The Netherlands
| | - Dominique Fäβler
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Functional Genomics, University of Heidelberg, Heidelberg69120, Germany
| | - Annika Mehr
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Functional Genomics, University of Heidelberg, Heidelberg69120, Germany
| | - Owen J. R. Jones
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
| | - Aurimas Kudzinskas
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
| | - Josan Gandawijaya
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
| | - Ulrike C. Müller
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Functional Genomics, University of Heidelberg, Heidelberg69120, Germany
| | - Martien J. H. Kas
- Department of Translational Neuroscience, UMC Utrecht Brain Center, UMC Utrecht, Utrecht3508 AB, The Netherlands
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - J. Peter H. Burbach
- Department of Translational Neuroscience, UMC Utrecht Brain Center, UMC Utrecht, Utrecht3508 AB, The Netherlands
| | - Asami Oguro-Ando
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
- Research Institute for Science and Technology, Tokyo University of Science, Tokyo, Japan
| |
Collapse
|
4
|
Dauar MT, Picard C, Labonté A, Breitner J, Rosa-Neto P, Villeneuve S, Poirier J. Contactin 5 and Apolipoproteins Interplay in Alzheimer's Disease. J Alzheimers Dis 2024; 98:1361-1375. [PMID: 38578887 DOI: 10.3233/jad-231003] [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] [Indexed: 04/07/2024]
Abstract
Background Apolipoproteins and contactin 5 are proteins associated with Alzheimer's disease (AD) pathophysiology. Apolipoproteins act on transport and clearance of cholesterol and phospholipids during synaptic turnover and terminal proliferation. Contactin 5 is a neuronal membrane protein involved in key processes of neurodevelopment. Objective To investigate the interactions between contactin 5 and apolipoproteins in AD, and the role of these proteins in response to neuronal damage. Methods Apolipoproteins (measured by Luminex), contactin 5 (measured by Olink's proximity extension assay), and cholesterol (measured by liquid chromatography mass spectrometry) were assessed in the cerebrospinal fluid (CSF) and plasma of cognitively unimpaired participants (n = 93). Gene expression was measured using polymerase chain reaction in the frontal cortex of autopsied-confirmed AD (n = 57) and control subjects (n = 31) and in the hippocampi of mice following entorhinal cortex lesions. Results Contactin 5 positively correlated with apolipoproteins B (p = 5.4×10-8), D (p = 1.86×10-4), E (p = 2.92×10-9), J (p = 2.65×10-9), and with cholesterol (p = 0.0096) in the CSF, and with cholesterol (p = 0.02), HDL (p = 0.0143), and LDL (p = 0.0121) in the plasma. Negative correlations were seen between CNTN5, APOB (p = 0.034) and APOE (p = 0.015) mRNA levels in the brains of control subjects. In the mouse model, apoe and apoj gene expression increased during the reinnervation phase (p < 0.05), while apob (p = 0.023) and apod (p = 0.006) increased in the deafferentation stage. Conclusions Extensive interactions were observed between contactin 5 and apolipoproteins and cholesterol, possibly due to neuronal damage. The alterations in gene expression of apolipoproteins suggest a role in axonal, terminal, and synaptic remodeling in response to entorhinal cortex damage.
Collapse
Affiliation(s)
- Marina Tedeschi Dauar
- Douglas Mental Health University Institute, Montréal, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Canada
- McGill University, Montreal, Canada
- CAPES Foundation, Ministry of Education of Brazil, Brasília, Brazil
| | - Cynthia Picard
- Douglas Mental Health University Institute, Montréal, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Canada
| | - Anne Labonté
- Douglas Mental Health University Institute, Montréal, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Canada
| | - John Breitner
- Douglas Mental Health University Institute, Montréal, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Canada
- McGill University, Montreal, Canada
- Department of Psychiatry, McGill University, Montreal, Canada
| | - Pedro Rosa-Neto
- McGill University, Montreal, Canada
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Verdun, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Canada
| | - Sylvia Villeneuve
- Douglas Mental Health University Institute, Montréal, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Canada
- McGill University, Montreal, Canada
- Department of Psychiatry, McGill University, Montreal, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute, Montréal, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Canada
- McGill University, Montreal, Canada
- Department of Psychiatry, McGill University, Montreal, Canada
| |
Collapse
|
5
|
Garofalo M, Vansenne F, Sival DA, Verbeek DS. Pathogenetic Insights into Developmental Coordination Disorder Reveal Substantial Overlap with Movement Disorders. Brain Sci 2023; 13:1625. [PMID: 38137073 PMCID: PMC10741651 DOI: 10.3390/brainsci13121625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/09/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Developmental Coordination Disorder (DCD) is a neurodevelopmental condition characterized by non-progressive central motor impairments. Mild movement disorder features have been observed in DCD. Until now, the etiology of DCD has been unclear. Recent studies suggested a genetic substrate in some patients with DCD, but comprehensive knowledge about associated genes and underlying pathogenetic mechanisms is still lacking. In this study, we first identified genes described in the literature in patients with a diagnosis of DCD according to the official diagnostic criteria. Second, we exposed the underlying pathogenetic mechanisms of DCD, by investigating tissue- and temporal gene expression patterns and brain-specific biological mechanisms. Third, we explored putative shared pathogenetic mechanisms between DCD and frequent movement disorders with a known genetic component, including ataxia, chorea, dystonia, and myoclonus. We identified 12 genes associated with DCD in the literature, which are ubiquitously expressed in the central nervous system throughout brain development. These genes are involved in cellular processes, neural signaling, and nervous system development. There was a remarkable overlap (62%) in pathogenetic mechanisms between DCD-associated genes and genes linked with movement disorders. Our findings suggest that some patients might have a genetic etiology of DCD, which could be considered part of a pathogenetic movement disorder spectrum.
Collapse
Affiliation(s)
- Martinica Garofalo
- Department of Pediatric Neurology, Beatrix Children’s Hospital, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (M.G.); (D.A.S.)
- Expertise Center Movement Disorders Groningen, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands;
| | - Fleur Vansenne
- Expertise Center Movement Disorders Groningen, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands;
- Department of Genetics, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Deborah A. Sival
- Department of Pediatric Neurology, Beatrix Children’s Hospital, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (M.G.); (D.A.S.)
- Expertise Center Movement Disorders Groningen, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands;
| | - Dineke S. Verbeek
- Expertise Center Movement Disorders Groningen, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands;
- Department of Genetics, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
6
|
Yu S, Wang Z, Nan J, Li A, Yang X, Tang X. Potential Schizophrenia Disease-Related Genes Prediction Using Metagraph Representations Based on a Protein-Protein Interaction Keyword Network: Framework Development and Validation. JMIR Form Res 2023; 7:e50998. [PMID: 37966892 PMCID: PMC10687686 DOI: 10.2196/50998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/28/2023] [Accepted: 10/27/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Schizophrenia is a serious mental disease. With increased research funding for this disease, schizophrenia has become one of the key areas of focus in the medical field. Searching for associations between diseases and genes is an effective approach to study complex diseases, which may enhance research on schizophrenia pathology and lead to the identification of new treatment targets. OBJECTIVE The aim of this study was to identify potential schizophrenia risk genes by employing machine learning methods to extract topological characteristics of proteins and their functional roles in a protein-protein interaction (PPI)-keywords (PPIK) network and understand the complex disease-causing property. Consequently, a PPIK-based metagraph representation approach is proposed. METHODS To enrich the PPI network, we integrated keywords describing protein properties and constructed a PPIK network. We extracted features that describe the topology of this network through metagraphs. We further transformed these metagraphs into vectors and represented proteins with a series of vectors. We then trained and optimized our model using random forest (RF), extreme gradient boosting, light gradient boosting machine, and logistic regression models. RESULTS Comprehensive experiments demonstrated the good performance of our proposed method with an area under the receiver operating characteristic curve (AUC) value between 0.72 and 0.76. Our model also outperformed baseline methods for overall disease protein prediction, including the random walk with restart, average commute time, and Katz models. Compared with the PPI network constructed from the baseline models, complementation of keywords in the PPIK network improved the performance (AUC) by 0.08 on average, and the metagraph-based method improved the AUC by 0.30 on average compared with that of the baseline methods. According to the comprehensive performance of the four models, RF was selected as the best model for disease protein prediction, with precision, recall, F1-score, and AUC values of 0.76, 0.73, 0.72, and 0.76, respectively. We transformed these proteins to their encoding gene IDs and identified the top 20 genes as the most probable schizophrenia-risk genes, including the EYA3, CNTN4, HSPA8, LRRK2, and AFP genes. We further validated these outcomes against metagraph features and evidence from the literature, performed a features analysis, and exploited evidence from the literature to interpret the correlation between the predicted genes and diseases. CONCLUSIONS The metagraph representation based on the PPIK network framework was found to be effective for potential schizophrenia risk genes identification. The results are quite reliable as evidence can be found in the literature to support our prediction. Our approach can provide more biological insights into the pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Shirui Yu
- Institute of Medical Information, Chinese Academy of Medical Sciences, Beijing, China
| | - Ziyang Wang
- Institute of Medical Information, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiale Nan
- Institute of Medical Information, Chinese Academy of Medical Sciences, Beijing, China
| | - Aihua Li
- Institute of Medical Information, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuemei Yang
- Institute of Medical Information, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoli Tang
- Institute of Medical Information, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
7
|
Cho Y, Lin K, Lee SH, Yu C, Valle DS, Avery D, Lv J, Jung K, Li L, Smith GD, China Kadoorie Biobank Collaborative Group, Sun D, Chen Z, Millwood IY, Hemani G, Walters RG. Genetic influences on alcohol flushing in East Asian populations. BMC Genomics 2023; 24:638. [PMID: 37875790 PMCID: PMC10594868 DOI: 10.1186/s12864-023-09721-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 10/06/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Although it is known that variation in the aldehyde dehydrogenase 2 (ALDH2) gene family influences the East Asian alcohol flushing response, knowledge about other genetic variants that affect flushing symptoms is limited. METHODS We performed a genome-wide association study meta-analysis and heritability analysis of alcohol flushing in 15,105 males of East Asian ancestry (Koreans and Chinese) to identify genetic associations with alcohol flushing. We also evaluated whether self-reported flushing can be used as an instrumental variable for alcohol intake. RESULTS We identified variants in the region of ALDH2 strongly associated with alcohol flushing, replicating previous studies conducted in East Asian populations. Additionally, we identified variants in the alcohol dehydrogenase 1B (ADH1B) gene region associated with alcohol flushing. Several novel variants were identified after adjustment for the lead variants (ALDH2-rs671 and ADH1B-rs1229984), which need to be confirmed in larger studies. The estimated SNP-heritability on the liability scale was 13% (S.E. = 4%) for flushing, but the heritability estimate decreased to 6% (S.E. = 4%) when the effects of the lead variants were controlled for. Genetic instrumentation of higher alcohol intake using these variants recapitulated known associations of alcohol intake with hypertension. Using self-reported alcohol flushing as an instrument gave a similar association pattern of higher alcohol intake and cardiovascular disease-related traits (e.g. stroke). CONCLUSION This study confirms that ALDH2-rs671 and ADH1B-rs1229984 are associated with alcohol flushing in East Asian populations. Our findings also suggest that self-reported alcohol flushing can be used as an instrumental variable in future studies of alcohol consumption.
Collapse
Affiliation(s)
- Yoonsu Cho
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Barley House, Oakfield Grove, Bristol, UK
| | - Kuang Lin
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Su-Hyun Lee
- Department of Epidemiology and Health Promotion, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, South Korea
| | - Canqing Yu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, 100191, China
| | - Dan Schmidt Valle
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Daniel Avery
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Jun Lv
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, 100191, China
| | - Keumji Jung
- Department of Epidemiology and Health Promotion, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, South Korea
| | - Liming Li
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, 100191, China
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Barley House, Oakfield Grove, Bristol, UK
| | | | - Dianjianyi Sun
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, 100191, China
| | - Zhengming Chen
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, University of Oxford, Oxford, UK
| | - Iona Y Millwood
- Nuffield Department of Population Health, University of Oxford, Oxford, UK.
- MRC Population Health Research Unit, University of Oxford, Oxford, UK.
| | - Gibran Hemani
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
- Population Health Sciences, Bristol Medical School, University of Bristol, Barley House, Oakfield Grove, Bristol, UK.
| | - Robin G Walters
- Nuffield Department of Population Health, University of Oxford, Oxford, UK.
- MRC Population Health Research Unit, University of Oxford, Oxford, UK.
| |
Collapse
|
8
|
Arbeev KG, Ukraintseva S, Bagley O, Duan H, Wu D, Akushevich I, Stallard E, Kulminski A, Christensen K, Feitosa MF, O’Connell JR, Parker D, Whitson H, Yashin AI. Interactions between genes involved in physiological dysregulation and axon guidance: role in Alzheimer's disease. Front Genet 2023; 14:1236509. [PMID: 37719713 PMCID: PMC10500346 DOI: 10.3389/fgene.2023.1236509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/17/2023] [Indexed: 09/19/2023] Open
Abstract
Dysregulation of physiological processes may contribute to Alzheimer's disease (AD) development. We previously found that an increase in the level of physiological dysregulation (PD) in the aging body is associated with declining resilience and robustness to major diseases. Also, our genome-wide association study found that genes associated with the age-related increase in PD frequently represented pathways implicated in axon guidance and synaptic function, which in turn were linked to AD and related traits (e.g., amyloid, tau, neurodegeneration) in the literature. Here, we tested the hypothesis that genes involved in PD and axon guidance/synapse function may jointly influence onset of AD. We assessed the impact of interactions between SNPs in such genes on AD onset in the Long Life Family Study and sought to replicate the findings in the Health and Retirement Study. We found significant interactions between SNPs in the UNC5C and CNTN6, and PLXNA4 and EPHB2 genes that influenced AD onset in both datasets. Associations with individual SNPs were not statistically significant. Our findings, thus, support a major role of genetic interactions in the heterogeneity of AD and suggest the joint contribution of genes involved in PD and axon guidance/synapse function (essential for the maintenance of complex neural networks) to AD development.
Collapse
Affiliation(s)
- Konstantin G. Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Olivia Bagley
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Hongzhe Duan
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Deqing Wu
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Igor Akushevich
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Eric Stallard
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Alexander Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Kaare Christensen
- Danish Aging Research Center, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Jeffrey R. O’Connell
- Division of Endocrinology, Diabetes and Nutrition and Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Daniel Parker
- Duke Center for the Study of Aging and Human Development, Duke University, Durham, NC, United States
| | - Heather Whitson
- Duke Center for the Study of Aging and Human Development, Duke University, Durham, NC, United States
- Durham VA Geriatrics Research Education and Clinical Center, Durham, NC, United States
| | - Anatoliy I. Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| |
Collapse
|
9
|
Zhang W, Huang H, Gui A, Mu D, Zhao T, Li H, Watanabe K, Xiao Z, Ye H, Xu Y. Contactin-6-deficient male mice exhibit the abnormal function of the accessory olfactory system and impaired reproductive behavior. Brain Behav 2023; 13:e2893. [PMID: 36860170 PMCID: PMC10097056 DOI: 10.1002/brb3.2893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 12/21/2022] [Accepted: 01/05/2023] [Indexed: 03/03/2023] Open
Abstract
INTRODUCTION Contactin-6 (CNTN6), also known as NB-3, is a neural recognition molecule and a member of the contactin subgroup of the immunoglobulin superfamily. Gene encoding CNTN6 is expressed in many regions of the neural system, including the accessory olfactory bulb (AOB) in mice. We aim to determine the effect of CNTN6 deficiency on the function of the accessory olfactory system (AOS). METHODS We examined the effect of CNTN6 deficiency on the reproductive behavior of male mice through behavioral experiments such as urine sniffing and mate preference tests. Staining and electron microscopy were used to observe the gross structure and the circuitry activity of the AOS. RESULTS Cntn6 is highly expressed in the vomeronasal organ (VNO) and the AOB, and sparsely expressed in the medial amygdala (MeA) and the medial preoptic area (MPOA), which receive direct and/or indirect projections from the AOB. Behavioral tests to examine reproductive function in mice, which is mostly controlled by the AOS, revealed that Cntn6-/- adult male mice showed less interest and reduced mating attempts toward estrous female mice in comparison with their Cntn6+/+ littermates. Although Cntn6-/- adult male mice displayed no obvious changes in the gross structure of the VNO or AOB, we observed the increased activation of granule cells in the AOB and the lower activation of neurons in the MeA and the MPOA as compared with Cntn6+/+ adult male mice. Moreover, there were an increased number of synapses between mitral cells and granule cells in the AOB of Cntn6-/- adult male mice as compared with wild-type controls. CONCLUSION These results indicate that CNTN6 deficiency affects the reproductive behavior of male mice, suggesting that CNTN6 participated in normal function of the AOS and its ablation was involved in synapse formation between mitral and granule cells in the AOB, rather than affecting the gross structure of the AOS.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Huiling Huang
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Ailing Gui
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Di Mu
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Tian Zhao
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Hongtao Li
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing, China
| | - Kazutada Watanabe
- Department of Bioengineering, Nagaoka University of Technology, Nagaoka, Niigata, Japan
| | - Zhicheng Xiao
- The Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, China.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne, Australia
| | - Haihong Ye
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Yiliang Xu
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Bauleo A, Pace V, Montesanto A, De Stefano L, Brando R, Puntorieri D, Cento L, Genuardi M, Falcone E. 3q29 microduplication syndrome: New evidence for the refinement of the critical region. Mol Genet Genomic Med 2023; 11:e2130. [PMID: 36691815 PMCID: PMC10094080 DOI: 10.1002/mgg3.2130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 11/18/2022] [Accepted: 12/21/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The 3q29 microduplication syndrome is a rare genomic disorder characterized by an extremely variable neurodevelopmental phenotype usually involving a genomic region ranging from 1.6 to 1.76 Mb. A small microduplication of 448.8 Kb containing only two genes was recently described in a patient with a 3q29 microduplication that was proposed as the minimal critical region of overlap of this syndrome. METHODS Molecular karyotyping (array-CGH) was performed on DNA extracted from peripheral blood samples using Agilent-California USA Human Genome CGH Microarray 4 × 180 K. The proband and his younger brother were further tested with a next generation sequencing (NGS) panel including genes implicated in autism spectrum disorder and in neurodevelopmental disorders. Quantitative real-time PCR was applied to verify the abnormal array-CGH findings. RESULTS Here, we report on a family with two males with neurodevelopmental disorders and an unaffected sibling with a small 3q29 microduplication (432.8 Kb) inherited from an unaffected mother that involves only two genes: DGL1 and BDH1. The proband had an additional intragenic duplication inherited from the unaffected father. Further testing was negative for Fragile X syndrome and for genes implicated in autism spectrum disorder and in neurodevelopmental disorders. CONCLUSION To the best of our knowledge, one of the family members here analyzed is the second reported case of a patient carrying a small 3q29 microduplication including only DGL1 and BDH1 genes and without any additional genetic aberration. The recognition of the clinical spectrum in patients with the critical region of overlap associated with the 3q29 duplication syndrome should prove valuable for predicting outcomes and providing more informed genetic counseling to patients with duplications in this region.
Collapse
Affiliation(s)
- Alessia Bauleo
- BIOGENET, Medical and Forensic Genetics Laboratory, Cosenza, Italy
| | - Vincenza Pace
- BIOGENET, Medical and Forensic Genetics Laboratory, Cosenza, Italy
| | - Alberto Montesanto
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Laura De Stefano
- BIOGENET, Medical and Forensic Genetics Laboratory, Cosenza, Italy
| | - Rossella Brando
- BIOGENET, Medical and Forensic Genetics Laboratory, Cosenza, Italy
| | - Domenica Puntorieri
- Dipartimento Materno Infantile Neuropsichiatria Infanzia e Adolescenza Rossano - Cariati, Azienda Sanitaria Provinciale di Cosenza, Cosenza, Italy
| | - Luca Cento
- Dipartimento Materno Infantile Neuropsichiatria Infanzia e Adolescenza Rossano - Cariati, Azienda Sanitaria Provinciale di Cosenza, Cosenza, Italy
| | - Maurizio Genuardi
- UOC Genetica Medica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Elena Falcone
- BIOGENET, Medical and Forensic Genetics Laboratory, Cosenza, Italy
| |
Collapse
|
11
|
Bizzoca A, Jirillo E, Flace P, Gennarini G. Overall Role of Contactins Expression in Neurodevelopmental Events and Contribution to Neurological Disorders. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:1176-1193. [PMID: 36515028 DOI: 10.2174/1871527322666221212160048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/21/2022] [Accepted: 10/28/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Neurodegenerative disorders may depend upon a misregulation of the pathways which sustain neurodevelopmental control. In this context, this review article focuses on Friedreich ataxia (FA), a neurodegenerative disorder resulting from mutations within the gene encoding the Frataxin protein, which is involved in the control of mitochondrial function and oxidative metabolism. OBJECTIVE The specific aim of the present study concerns the FA molecular and cellular substrates, for which available transgenic mice models are proposed, including mutants undergoing misexpression of adhesive/morphoregulatory proteins, in particular belonging to the Contactin subset of the immunoglobulin supergene family. METHODS In both mutant and control mice, neurogenesis was explored by morphological/morphometric analysis through the expression of cell type-specific markers, including b-tubulin, the Contactin-1 axonal adhesive glycoprotein, as well as the Glial Fibrillary Acidic Protein (GFAP). RESULTS Specific consequences were found to arise from the chosen misexpression approach, consisting of a neuronal developmental delay associated with glial upregulation. Protective effects against the arising phenotype resulted from antioxidants (essentially epigallocatechin gallate (EGCG)) administration, which was demonstrated through the profiles of neuronal (b-tubulin and Contactin 1) as well as glial (GFAP) markers, in turn indicating the concomitant activation of neurodegeneration and neuro repair processes. The latter also implied activation of the Notch-1 signaling. CONCLUSION Overall, this study supports the significance of changes in morphoregulatory proteins expression in the FA pathogenesis and of antioxidant administration in counteracting it, which, in turn, allows to devise potential therapeutic approaches.
Collapse
Affiliation(s)
- Antonella Bizzoca
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Medical School, University of Bari, Piazza Giulio Cesare, 11. Bari I-70124, Italy
| | - Emilio Jirillo
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Medical School, University of Bari, Piazza Giulio Cesare, 11. Bari I-70124, Italy
| | - Paolo Flace
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Medical School, University of Bari, Piazza Giulio Cesare, 11. Bari I-70124, Italy
| | - Gianfranco Gennarini
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Medical School, University of Bari, Piazza Giulio Cesare, 11. Bari I-70124, Italy
| |
Collapse
|
12
|
Dauar MT, Labonté A, Picard C, Miron J, Rosa-Neto P, Zetterberg H, Blennow K, Villeneuve S, Poirier J. Characterization of the contactin 5 protein and its risk-associated polymorphic variant throughout the Alzheimer's disease spectrum. Alzheimers Dement 2022. [PMID: 36583624 DOI: 10.1002/alz.12868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 10/04/2022] [Accepted: 10/17/2022] [Indexed: 12/31/2022]
Abstract
INTRODUCTION We investigate the CNTN5 rs1461684 G variant and the contactin 5 protein in sporadic Alzheimer's disease (sAD). METHODS Contactin 5, sAD biomarkers, and synaptic markers were measured in the cerebrospinal fluid (CSF). Amyloid and tau deposition were assessed using positron emission tomography. Contactin 5 protein and mRNA levels were measured in brain tissue. RESULTS CSF contactin 5 increases progressively in cognitively unimpaired individuals and is decreased in mild cognitive impairment and sAD. CSF contactin 5 correlates with sAD biomarkers and with synaptic markers. The rs1461684 G variant associates with faster disease progression in cognitively unimpaired subjects. Cortical full-length and isoform 3 CNTN5 mRNAs are decreased in the presence of the G allele and as a function of Consortium to Establish a Registry for Alzheimer's Disease stages. DISCUSSION The newly identified rs1461684 G variant associates with sAD risk, rate of disease progression, and gene expression. Contactin 5 protein and mRNA are affected particularly in the early stages of the disease.
Collapse
Affiliation(s)
- Marina Tedeschi Dauar
- Douglas Mental Health University Institute, Montréal, Canada.,Centre for the Studies in the Prevention of Alzheimer's, Douglas Mental Health University Institute, Montréal, Canada.,McGill University, Montréal, Canada.,CAPES Foundation, Ministry of Education of Brazil, Brasília, Brazil
| | - Anne Labonté
- Douglas Mental Health University Institute, Montréal, Canada.,Centre for the Studies in the Prevention of Alzheimer's, Douglas Mental Health University Institute, Montréal, Canada
| | - Cynthia Picard
- Douglas Mental Health University Institute, Montréal, Canada.,Centre for the Studies in the Prevention of Alzheimer's, Douglas Mental Health University Institute, Montréal, Canada
| | - Justin Miron
- Douglas Mental Health University Institute, Montréal, Canada.,Centre for the Studies in the Prevention of Alzheimer's, Douglas Mental Health University Institute, Montréal, Canada.,McGill University, Montréal, Canada
| | - Pedro Rosa-Neto
- Douglas Mental Health University Institute, Montréal, Canada.,Centre for the Studies in the Prevention of Alzheimer's, Douglas Mental Health University Institute, Montréal, Canada.,McGill University, Montréal, Canada.,Department of Psychiatry, McGill University, Montréal, Canada
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute at UCL, London, UK.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Sylvia Villeneuve
- Douglas Mental Health University Institute, Montréal, Canada.,Centre for the Studies in the Prevention of Alzheimer's, Douglas Mental Health University Institute, Montréal, Canada.,McGill University, Montréal, Canada.,Department of Psychiatry, McGill University, Montréal, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute, Montréal, Canada.,Centre for the Studies in the Prevention of Alzheimer's, Douglas Mental Health University Institute, Montréal, Canada.,McGill University, Montréal, Canada.,Department of Psychiatry, McGill University, Montréal, Canada
| |
Collapse
|
13
|
Luo W, Cruz-Ochoa NA, Seng C, Egger M, Lukacsovich D, Lukacsovich T, Földy C. Pcdh11x controls target specification of mossy fiber sprouting. Front Neurosci 2022; 16:888362. [PMID: 36117624 PMCID: PMC9475199 DOI: 10.3389/fnins.2022.888362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Circuit formation is a defining characteristic of the developing brain. However, multiple lines of evidence suggest that circuit formation can also take place in adults, the mechanisms of which remain poorly understood. Here, we investigated the epilepsy-associated mossy fiber (MF) sprouting in the adult hippocampus and asked which cell surface molecules define its target specificity. Using single-cell RNAseq data, we found lack and expression of Pcdh11x in non-sprouting and sprouting neurons respectively. Subsequently, we used CRISPR/Cas9 genome editing to disrupt the Pcdh11x gene and characterized its consequences on sprouting. Although MF sprouting still developed, its target specificity was altered. New synapses were frequently formed on granule cell somata in addition to dendrites. Our findings shed light onto a key molecular determinant of target specificity in MF sprouting and contribute to understanding the molecular mechanism of adult brain rewiring.
Collapse
|
14
|
Cukier HN, Kim H, Griswold AJ, Codreanu SG, Prince LM, Sherrod SD, McLean JA, Dykxhoorn DM, Ess KC, Hedera P, Bowman AB, Neely MD. Genomic, transcriptomic, and metabolomic profiles of hiPSC-derived dopamine neurons from clinically discordant brothers with identical PRKN deletions. NPJ Parkinsons Dis 2022; 8:84. [PMID: 35768426 PMCID: PMC9243035 DOI: 10.1038/s41531-022-00346-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 05/27/2022] [Indexed: 11/25/2022] Open
Abstract
We previously reported on two brothers who carry identical compound heterozygous PRKN mutations yet present with significantly different Parkinson's Disease (PD) clinical phenotypes. Juvenile cases demonstrate that PD is not necessarily an aging-associated disease. Indeed, evidence for a developmental component to PD pathogenesis is accumulating. Thus, we hypothesized that the presence of additional genetic modifiers, including genetic loci relevant to mesencephalic dopamine neuron development, could potentially contribute to the different clinical manifestations of the two brothers. We differentiated human-induced pluripotent stem cells (hiPSCs) derived from the two brothers into mesencephalic neural precursor cells and early postmitotic dopaminergic neurons and performed wholeexome sequencing and transcriptomic and metabolomic analyses. No significant differences in the expression of canonical dopamine neuron differentiation markers were observed. Yet our transcriptomic analysis revealed a significant downregulation of the expression of three neurodevelopmentally relevant cell adhesion molecules, CNTN6, CNTN4 and CHL1, in the cultures of the more severely affected brother. In addition, several HLA genes, known to play a role in neurodevelopment, were differentially regulated. The expression of EN2, a transcription factor crucial for mesencephalic dopamine neuron development, was also differentially regulated. We further identified differences in cellular processes relevant to dopamine metabolism. Lastly, wholeexome sequencing, transcriptomics and metabolomics data all revealed differences in glutathione (GSH) homeostasis, the dysregulation of which has been previously associated with PD. In summary, we identified genetic differences which could potentially, at least partially, contribute to the discordant clinical PD presentation of the two brothers.
Collapse
Affiliation(s)
- Holly N Cukier
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Hyunjin Kim
- School of Health Sciences, Purdue University, West Lafayette, Indiana, IN, USA
| | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Simona G Codreanu
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Lisa M Prince
- School of Health Sciences, Purdue University, West Lafayette, Indiana, IN, USA
| | - Stacy D Sherrod
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - John A McLean
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Derek M Dykxhoorn
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kevin C Ess
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Peter Hedera
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Neurology, University of Louisville, Louisville, KY, USA
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, Indiana, IN, USA.
| | - M Diana Neely
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
15
|
Remberk B, Niwiński P, Brzóska-Konkol E, Borowska A, Papasz-Siemieniuk A, Brągoszewska J, Bażyńska AK, Szostakiewicz Ł, Herman A. Ectodermal disturbance in development shared by anorexia and schizophrenia may reflect neurodevelopmental abnormalities. Brain Behav 2021; 11:e2281. [PMID: 34510800 PMCID: PMC8553323 DOI: 10.1002/brb3.2281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/25/2021] [Accepted: 06/27/2021] [Indexed: 12/25/2022] Open
Abstract
Minor physical abnormalities (MPA) are subtle dysmorphic features of bodily structures that have little or no impact on function. Most MPA develop during the first gestational trimester and are considered as important indicators of neuroectodermal deficiencies emerging during early brain development. A higher frequency of MPA was confirmed in schizophrenia patients and their relatives, when compared to controls. These findings are consistent with the neurodevelopmental model of schizophrenia. A neurodevelopmental component amongst other risk factors has also been recently proposed for anorexia nervosa (AN). The current study aimed to assess MPA frequency in adolescent inpatients with either schizophrenia spectrum disorders (SSD) or AN as compared to healthy controls (HC). The Waldrop Scale was used for assessing MPA. The mean MPA total score and mean head subscore was significantly higher in both test groups than in HC. There were no statistically significant differences between SSD and AN groups. The MPA profile (not frequency) was similar in all three groups. This finding is consistent both with widely acknowledged neurodevelopmental schizophrenia hypothesis as well as with more recent neurodevelopmental model of AN. Nevertheless, the findings should not be overgeneralized and further studies are warranted.
Collapse
Affiliation(s)
| | - Piotr Niwiński
- Psychological and Pedagogical Counselling Centre no 7, Warsaw, Poland
| | | | - Anna Borowska
- Institute of Psychiatry and Neurology, Warsaw, Poland
| | | | | | | | | | - Anna Herman
- Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
16
|
Zhang J, Qiu W, Hu F, Zhang X, Deng Y, Nie H, Xu R. The rs2619566, rs10260404, and rs79609816 Polymorphisms Are Associated With Sporadic Amyotrophic Lateral Sclerosis in Individuals of Han Ancestry From Mainland China. Front Genet 2021; 12:679204. [PMID: 34421992 PMCID: PMC8378233 DOI: 10.3389/fgene.2021.679204] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/25/2021] [Indexed: 11/21/2022] Open
Abstract
The pathogenesis of sporadic amyotrophic lateral sclerosis (sALS) remains unknown; however, recent research suggests that genetic factors may play an important role. This study aimed at investigating possible genetic risk factors for the pathogenesis of sALS. In our previous study, we conducted a genome-wide association study (GWAS) in 250 sALS patients and 250 control participants of Han ancestry from mainland China (HACM) and retrospectively analyzed the previously reported candidate loci related with sALS including our GWAS investigated results. In this study, twenty-seven candidate loci that were most likely associated with sALS were selected for further analysis in an independent case/control population of 239 sALS patients and 261 control subjects of HACM ethnicity using sequenom massARRAY methodology and DNA sequencing. We discovered that the polymorphism rs2619566 located within the contactin-4 (CNTN4) gene, rs10260404 in the dipeptidyl-peptidase 6 (DPP6) gene, and rs79609816 in the inositol polyphosphate-5-phosphatase B (INPP5B) gene were strongly associated with sALS in subjects of HACM ethnicity. Subjects harboring the minor C allele of rs2619566 and the minor T allele of rs79609816 exhibited an increased risk for sALS development, while carriers of the minor C allele of rs10260404 showed a decreased risk of sALS development compared to the subjects of other genotypes. The polymorphisms of rs2619566, rs10260404, and rs79609816 may change or affect the splicing, transcription, and translation of CNTN4, DPP6, and INPP5B genes and may play roles in the pathogenesis of sALS roles in the pathogenesis of sALS.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Weiwen Qiu
- Department of Neurology, The Affiliated People's Hospital of Nanchang University, The First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Fan Hu
- Department of Neurology, The Affiliated People's Hospital of Nanchang University, The First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Xiong Zhang
- Department of Neurology, Maoming People's Hospital, Maoming, China
| | - Youqing Deng
- Department of Neurology, The Third Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hongbing Nie
- Department of Neurology, The Affiliated People's Hospital of Nanchang University, The First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Renshi Xu
- Department of Neurology, The Affiliated People's Hospital of Nanchang University, The First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People's Hospital, Nanchang, China
| |
Collapse
|
17
|
Aranda S, Jiménez E, Martorell L, Muntané G, Vieta E, Vilella E. A systematic review on genome-wide association studies exploring comorbidity in bipolar disorder. JOURNAL OF AFFECTIVE DISORDERS REPORTS 2021. [DOI: 10.1016/j.jadr.2021.100130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
18
|
Tillotson R, Cholewa-Waclaw J, Chhatbar K, Connelly JC, Kirschner SA, Webb S, Koerner MV, Selfridge J, Kelly DA, De Sousa D, Brown K, Lyst MJ, Kriaucionis S, Bird A. Neuronal non-CG methylation is an essential target for MeCP2 function. Mol Cell 2021; 81:1260-1275.e12. [PMID: 33561390 PMCID: PMC7980222 DOI: 10.1016/j.molcel.2021.01.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/17/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023]
Abstract
DNA methylation is implicated in neuronal biology via the protein MeCP2, the mutation of which causes Rett syndrome. MeCP2 recruits the NCOR1/2 co-repressor complexes to methylated cytosine in the CG dinucleotide, but also to sites of non-CG methylation, which are abundant in neurons. To test the biological significance of the dual-binding specificity of MeCP2, we replaced its DNA binding domain with an orthologous domain from MBD2, which can only bind mCG motifs. Knockin mice expressing the domain-swap protein displayed severe Rett-syndrome-like phenotypes, indicating that normal brain function requires the interaction of MeCP2 with sites of non-CG methylation, specifically mCAC. The results support the notion that the delayed onset of Rett syndrome is due to the simultaneous post-natal accumulation of mCAC and its reader MeCP2. Intriguingly, genes dysregulated in both Mecp2 null and domain-swap mice are implicated in other neurological disorders, potentially highlighting targets of relevance to the Rett syndrome phenotype.
Collapse
Affiliation(s)
- Rebekah Tillotson
- Wellcome Centre for Cell Biology, University of Edinburgh, The Michael Swann Building, King's Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Justyna Cholewa-Waclaw
- Wellcome Centre for Cell Biology, University of Edinburgh, The Michael Swann Building, King's Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Kashyap Chhatbar
- Wellcome Centre for Cell Biology, University of Edinburgh, The Michael Swann Building, King's Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - John C Connelly
- Wellcome Centre for Cell Biology, University of Edinburgh, The Michael Swann Building, King's Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Sophie A Kirschner
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Shaun Webb
- Wellcome Centre for Cell Biology, University of Edinburgh, The Michael Swann Building, King's Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Martha V Koerner
- Wellcome Centre for Cell Biology, University of Edinburgh, The Michael Swann Building, King's Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Jim Selfridge
- Wellcome Centre for Cell Biology, University of Edinburgh, The Michael Swann Building, King's Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - David A Kelly
- Wellcome Centre for Cell Biology, University of Edinburgh, The Michael Swann Building, King's Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Dina De Sousa
- Wellcome Centre for Cell Biology, University of Edinburgh, The Michael Swann Building, King's Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Kyla Brown
- Wellcome Centre for Cell Biology, University of Edinburgh, The Michael Swann Building, King's Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Matthew J Lyst
- Wellcome Centre for Cell Biology, University of Edinburgh, The Michael Swann Building, King's Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Skirmantas Kriaucionis
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Adrian Bird
- Wellcome Centre for Cell Biology, University of Edinburgh, The Michael Swann Building, King's Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK.
| |
Collapse
|
19
|
Cntn4, a risk gene for neuropsychiatric disorders, modulates hippocampal synaptic plasticity and behavior. Transl Psychiatry 2021; 11:106. [PMID: 33542194 PMCID: PMC7862349 DOI: 10.1038/s41398-021-01223-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 01/05/2021] [Accepted: 01/18/2021] [Indexed: 12/27/2022] Open
Abstract
Neurodevelopmental and neuropsychiatric disorders, such as autism spectrum disorders (ASD), anorexia nervosa (AN), Alzheimer's disease (AD), and schizophrenia (SZ), are heterogeneous brain disorders with unknown etiology. Genome wide studies have revealed a wide variety of risk genes for these disorders, indicating a biological link between genetic signaling pathways and brain pathology. A unique risk gene is Contactin 4 (Cntn4), an Ig cell adhesion molecule (IgCAM) gene, which has been associated with several neuropsychiatric disorders including ASD, AN, AD, and SZ. Here, we investigated the Cntn4 gene knockout (KO) mouse model to determine whether memory dysfunction and altered brain plasticity, common neuropsychiatric symptoms, are affected by Cntn4 genetic disruption. For that purpose, we tested if Cntn4 genetic disruption affects CA1 synaptic transmission and the ability to induce LTP in hippocampal slices. Stimulation in CA1 striatum radiatum significantly decreased synaptic potentiation in slices of Cntn4 KO mice. Neuroanatomical analyses showed abnormal dendritic arborization and spines of hippocampal CA1 neurons. Short- and long-term recognition memory, spatial memory, and fear conditioning responses were also assessed. These behavioral studies showed increased contextual fear conditioning in heterozygous and homozygous KO mice, quantified by a gene-dose dependent increase in freezing response. In comparison to wild-type mice, Cntn4-deficient animals froze significantly longer and groomed more, indicative of increased stress responsiveness under these test conditions. Our electrophysiological, neuro-anatomical, and behavioral results in Cntn4 KO mice suggest that Cntn4 has important functions related to fear memory possibly in association with the neuronal morphological and synaptic plasticity changes in hippocampus CA1 neurons.
Collapse
|
20
|
Kalafatakis I, Savvaki M, Velona T, Karagogeos D. Implication of Contactins in Demyelinating Pathologies. Life (Basel) 2021; 11:life11010051. [PMID: 33451101 PMCID: PMC7828632 DOI: 10.3390/life11010051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/09/2021] [Accepted: 01/11/2021] [Indexed: 12/19/2022] Open
Abstract
Demyelinating pathologies comprise of a variety of conditions where either central or peripheral myelin is attacked, resulting in white matter lesions and neurodegeneration. Myelinated axons are organized into molecularly distinct domains, and this segregation is crucial for their proper function. These defined domains are differentially affected at the different stages of demyelination as well as at the lesion and perilesion sites. Among the main players in myelinated axon organization are proteins of the contactin (CNTN) group of the immunoglobulin superfamily (IgSF) of cell adhesion molecules, namely Contactin-1 and Contactin-2 (CNTN1, CNTN2). The two contactins perform their functions through intermolecular interactions, which are crucial for myelinated axon integrity and functionality. In this review, we focus on the implication of these two molecules as well as their interactors in demyelinating pathologies in humans. At first, we describe the organization and function of myelinated axons in the central (CNS) and the peripheral (PNS) nervous system, further analyzing the role of CNTN1 and CNTN2 as well as their interactors in myelination. In the last section, studies showing the correlation of the two contactins with demyelinating pathologies are reviewed, highlighting the importance of these recognition molecules in shaping the function of the nervous system in multiple ways.
Collapse
|
21
|
Gandawijaya J, Bamford RA, Burbach JPH, Oguro-Ando A. Cell Adhesion Molecules Involved in Neurodevelopmental Pathways Implicated in 3p-Deletion Syndrome and Autism Spectrum Disorder. Front Cell Neurosci 2021; 14:611379. [PMID: 33519384 PMCID: PMC7838543 DOI: 10.3389/fncel.2020.611379] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/15/2020] [Indexed: 01/06/2023] Open
Abstract
Autism spectrum disorder (ASD) is characterized by impaired social interaction, language delay and repetitive or restrictive behaviors. With increasing prevalence, ASD is currently estimated to affect 0.5–2.0% of the global population. However, its etiology remains unclear due to high genetic and phenotypic heterogeneity. Copy number variations (CNVs) are implicated in several forms of syndromic ASD and have been demonstrated to contribute toward ASD development by altering gene dosage and expression. Increasing evidence points toward the p-arm of chromosome 3 (chromosome 3p) as an ASD risk locus. Deletions occurring at chromosome 3p result in 3p-deletion syndrome (Del3p), a rare genetic disorder characterized by developmental delay, intellectual disability, facial dysmorphisms and often, ASD or ASD-associated behaviors. Therefore, we hypothesize that overlapping molecular mechanisms underlie the pathogenesis of Del3p and ASD. To investigate which genes encoded in chromosome 3p could contribute toward Del3p and ASD, we performed a comprehensive literature review and collated reports investigating the phenotypes of individuals with chromosome 3p CNVs. We observe that high frequencies of CNVs occur in the 3p26.3 region, the terminal cytoband of chromosome 3p. This suggests that CNVs disrupting genes encoded within the 3p26.3 region are likely to contribute toward the neurodevelopmental phenotypes observed in individuals affected by Del3p. The 3p26.3 region contains three consecutive genes encoding closely related neuronal immunoglobulin cell adhesion molecules (IgCAMs): Close Homolog of L1 (CHL1), Contactin-6 (CNTN6), and Contactin-4 (CNTN4). CNVs disrupting these neuronal IgCAMs may contribute toward ASD phenotypes as they have been associated with key roles in neurodevelopment. CHL1, CNTN6, and CNTN4 have been observed to promote neurogenesis and neuronal survival, and regulate neuritogenesis and synaptic function. Furthermore, there is evidence that these neuronal IgCAMs possess overlapping interactomes and participate in common signaling pathways regulating axon guidance. Notably, mouse models deficient for these neuronal IgCAMs do not display strong deficits in axonal migration or behavioral phenotypes, which is in contrast to the pronounced defects in neuritogenesis and axon guidance observed in vitro. This suggests that when CHL1, CNTN6, or CNTN4 function is disrupted by CNVs, other neuronal IgCAMs may suppress behavioral phenotypes by compensating for the loss of function.
Collapse
Affiliation(s)
- Josan Gandawijaya
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Rosemary A Bamford
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - J Peter H Burbach
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Asami Oguro-Ando
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
22
|
Bizzoca A, Caracciolo M, Corsi P, Magrone T, Jirillo E, Gennarini G. Molecular and Cellular Substrates for the Friedreich Ataxia. Significance of Contactin Expression and of Antioxidant Administration. Molecules 2020; 25:E4085. [PMID: 32906751 PMCID: PMC7570916 DOI: 10.3390/molecules25184085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/28/2020] [Accepted: 09/02/2020] [Indexed: 11/16/2022] Open
Abstract
In this study, the neural phenotype is explored in rodent models of the spinocerebellar disorder known as the Friedreich Ataxia (FA), which results from mutations within the gene encoding the Frataxin mitochondrial protein. For this, the M12 line, bearing a targeted mutation, which disrupts the Frataxin gene exon 4 was used, together with the M02 line, which, in addition, is hemizygous for the human Frataxin gene mutation (Pook transgene), implying the occurrence of 82-190 GAA repeats within its first intron. The mutant mice phenotype was compared to the one of wild type littermates in regions undergoing differential profiles of neurogenesis, including the cerebellar cortex and the spinal cord by using neuronal (β-tubulin) and glial (Glial Fibrillary Acidic Protein) markers as well as the Contactin 1 axonal glycoprotein, involved in neurite growth control. Morphological/morphometric analyses revealed that while in Frataxin mutant mice the neuronal phenotype was significantly counteracted, a glial upregulation occurred at the same time. Furthermore, Contactin 1 downregulation suggested that changes in the underlying gene contributed to the disorder pathogenesis. Therefore, the FA phenotype implies an alteration of the developmental profile of neuronal and glial precursors. Finally, epigallocatechin gallate polyphenol administration counteracted the disorder, indicating protective effects of antioxidant administration.
Collapse
Affiliation(s)
| | | | | | | | | | - Gianfranco Gennarini
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari, Piazza Giulio Cesare, 11. I-70124 Bari, Italy; (A.B.); (M.C.); (P.C.); (T.M.); (E.J.)
| |
Collapse
|
23
|
Kukharsky MS, Skvortsova VI, Bachurin SO, Buchman VL. In a search for efficient treatment for amyotrophic lateral sclerosis: Old drugs for new approaches. Med Res Rev 2020; 41:2804-2822. [DOI: 10.1002/med.21725] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/23/2020] [Accepted: 08/08/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Michail S. Kukharsky
- Faculty of Medical Biology Pirogov Russian National Research Medical University Moscow Russian Federation
- Institute of Physiologically Active Compounds Russian Academy of Sciences Moscow Region Russian Federation
| | - Veronika I. Skvortsova
- Faculty of Medical Biology Pirogov Russian National Research Medical University Moscow Russian Federation
| | - Sergey O. Bachurin
- Institute of Physiologically Active Compounds Russian Academy of Sciences Moscow Region Russian Federation
| | - Vladimir L. Buchman
- Institute of Physiologically Active Compounds Russian Academy of Sciences Moscow Region Russian Federation
- School of Biosciences Cardiff University Cardiff United Kingdom
| |
Collapse
|
24
|
Maccarini S, Cipani A, Bertini V, Skripac J, Salvi A, Borsani G, Marchina E. Inherited duplication of the pseudoautosomal region Xq28 in a subject with Gilles de la Tourette syndrome and intellectual disability: a case report. Mol Cytogenet 2020; 13:23. [PMID: 32582378 PMCID: PMC7310047 DOI: 10.1186/s13039-020-00493-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/10/2020] [Indexed: 12/22/2022] Open
Abstract
Background Tourette syndrome (TS) is a complex neurodevelopmental disorder (NDD) characterized by multiple chronic involuntary motor and vocal tics with onset during childhood or adolescence. Most TS patients present with additional comorbidities, typically attention deficit hyperactivity disorder (ADHD), obsessive- compulsive disorder (OCD), autism spectrum disorder (ASD) and intellectual disability (ID). Both TS and ID are genetically complex disorders that likely occur as a result of the effects of multiple genes interacting with other environmental factors. In addition to single gene mutations and chromosomal disorders, copy number variations (CNVs) are implicated across many NDDs and ID and contribute to their shared genetic etiology. Screening of CNVs using microarray-based Comparative Genomic Hybridization (aCGH) is now routinely performed in all subjects with NDD and ID. Case presentation We report a case of a 12-year-old girl diagnosed with Gilles de la Tourette Syndrome associated to behavior disorders and intellectual disability in particular with regard to language. Array-CGH analysis showed a CNV of a subtelomeric region Xq28 (gain of 260 kb) inherited from the healthy father. The duplication contains two genes, VAMP7 and SPRY3 of the PAR2 pseudoautosomal region. FISH analysis revealed that the duplicated segment is located on the short arm of a chromosome 13, resulting in a trisomy of the region. In the proband the expression levels of the genes evaluated in the peripheral blood sample are comparable both those of the mother and to those of female control subjects. Conclusions Although the trisomy of the 260 kb region from Xq28 identified in proband is also shared by the healthy father, it is tantalizing to speculate that, together with genetic risk factors inherited from the mother, it may play a role in the development of a form of Tourette syndrome with intellectual disability. This hypothesis is also supported by the fact that both genes present in the duplicated region (VAMP7 and SPRY3) are expressed in the CNS and are implicated in neurotransmission and neurite growth and branching. In addition, similar CNVs have been identified in individuals whose phenotype is associated with autism spectrum disorders or intellectual disability.
Collapse
Affiliation(s)
- Stefania Maccarini
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Annamaria Cipani
- Unit of Child and Adolescent Neuropsychiatry, ASST of Garda, Brescia, Italy
| | - Valeria Bertini
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Jelena Skripac
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Alessandro Salvi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giuseppe Borsani
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Eleonora Marchina
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
25
|
Mangum KD, Farber MA. Genetic and epigenetic regulation of abdominal aortic aneurysms. Clin Genet 2020; 97:815-826. [PMID: 31957007 DOI: 10.1111/cge.13705] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/22/2019] [Accepted: 01/11/2020] [Indexed: 12/11/2022]
Abstract
Abdominal aortic aneurysms (AAAs) are focal dilations of the aorta that develop from degenerative changes in the media and adventitia of the vessel. Ruptured AAAs have a mortality of up to 85%, thus it is important to identify patients with AAA at increased risk for rupture who would benefit from increased surveillance and/or surgical repair. Although the exact genetic and epigenetic mechanisms regulating AAA formation are not completely understood, Mendelian cases of AAA, which result from pathologic variants in a single gene, have helped provide a basic understanding of AAA pathophysiology. More recently, genome wide associated studies (GWAS) have identified additional variants, termed single nucleotide polymorphisms, in humans that may be associated with AAAs. While some variants may be associated with AAAs and play causal roles in aneurysm pathogenesis, it should be emphasized that the majority of SNPs do not actually cause disease. In addition to GWAS, other studies have uncovered epigenetic causes of disease that regulate expression of genes known to be important in AAA pathogenesis. This review describes many of these genetic and epigenetic contributors of AAAs, which altogether provide a deeper insight into AAA pathogenesis.
Collapse
Affiliation(s)
- Kevin D Mangum
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Mark A Farber
- Division of Vascular Surgery, UNC Department of Surgery, Chapel Hill, North Carolina
| |
Collapse
|
26
|
The Interaction Between Contactin and Amyloid Precursor Protein and Its Role in Alzheimer’s Disease. Neuroscience 2020; 424:184-202. [DOI: 10.1016/j.neuroscience.2019.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 01/06/2023]
|
27
|
The Reeler Mouse: A Translational Model of Human Neurological Conditions, or Simply a Good Tool for Better Understanding Neurodevelopment? J Clin Med 2019; 8:jcm8122088. [PMID: 31805691 PMCID: PMC6947477 DOI: 10.3390/jcm8122088] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/26/2019] [Accepted: 11/28/2019] [Indexed: 12/25/2022] Open
Abstract
The first description of the Reeler mutation in mouse dates to more than fifty years ago, and later, its causative gene (reln) was discovered in mouse, and its human orthologue (RELN) was demonstrated to be causative of lissencephaly 2 (LIS2) and about 20% of the cases of autosomal-dominant lateral temporal epilepsy (ADLTE). In both human and mice, the gene encodes for a glycoprotein referred to as reelin (Reln) that plays a primary function in neuronal migration during development and synaptic stabilization in adulthood. Besides LIS2 and ADLTE, RELN and/or other genes coding for the proteins of the Reln intracellular cascade have been associated substantially to other conditions such as spinocerebellar ataxia type 7 and 37, VLDLR-associated cerebellar hypoplasia, PAFAH1B1-associated lissencephaly, autism, and schizophrenia. According to their modalities of inheritances and with significant differences among each other, these neuropsychiatric disorders can be modeled in the homozygous (reln−/−) or heterozygous (reln+/−) Reeler mouse. The worth of these mice as translational models is discussed, with focus on their construct and face validity. Description of face validity, i.e., the resemblance of phenotypes between the two species, centers onto the histological, neurochemical, and functional observations in the cerebral cortex, hippocampus, and cerebellum of Reeler mice and their human counterparts.
Collapse
|
28
|
Kastriti ME, Stratigi A, Mariatos D, Theodosiou M, Savvaki M, Kavkova M, Theodorakis K, Vidaki M, Zikmund T, Kaiser J, Adameyko I, Karagogeos D. Ablation of CNTN2+ Pyramidal Neurons During Development Results in Defects in Neocortical Size and Axonal Tract Formation. Front Cell Neurosci 2019; 13:454. [PMID: 31749685 PMCID: PMC6844266 DOI: 10.3389/fncel.2019.00454] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/23/2019] [Indexed: 01/22/2023] Open
Abstract
Corticothalamic axons express Contactin-2 (CNTN2/TAG-1), a neuronal recognition molecule of the immunoglobulin superfamily involved in neurogenesis, neurite outgrowth, and fasciculation. TAG-1, which is expressed transiently by cortical pyramidal neurons during embryonic development, has been shown to be fundamental for axonal recognition, cellular migration, and neuronal proliferation in the developing cortex. Although Tag-1−/− mice do not exhibit any obvious defects in the corticofugal system, the role of TAG-1+ neurons during the development of the cortex remains elusive. We have generated a mouse model expressing EGFP under the Tag-1 promoter and encompassing the coding sequence of Diptheria Toxin subunit A (DTA) under quiescence with no effect on the expression of endogenous Tag-1. We show that while the line recapitulates the expression pattern of the molecule, it highlights an extended expression in the forebrain, including multiple axonal tracts and neuronal populations, both spatially and temporally. Crossing these mice to the Emx1-Cre strain, we ablated the vast majority of TAG-1+ cortical neurons. Among the observed defects were a significantly smaller cortex, a reduction of corticothalamic axons as well as callosal and commissural defects. Such defects are common in neurodevelopmental disorders, thus this mouse could serve as a useful model to study physiological and pathophysiological cortical development.
Collapse
Affiliation(s)
- Maria Eleni Kastriti
- Department of Basic Science, Faculty of Medicine, University of Crete, Heraklion, Greece.,Foundation for Research and Technology, Institute of Molecular Biology and Biotechnology, Heraklion, Greece.,Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Aikaterini Stratigi
- Department of Basic Science, Faculty of Medicine, University of Crete, Heraklion, Greece.,Foundation for Research and Technology, Institute of Molecular Biology and Biotechnology, Heraklion, Greece.,Laboratory of Neurophysiology, Université Libre de Bruxelles, UNI, Brussels, Belgium
| | - Dimitris Mariatos
- Department of Basic Science, Faculty of Medicine, University of Crete, Heraklion, Greece.,Foundation for Research and Technology, Institute of Molecular Biology and Biotechnology, Heraklion, Greece
| | - Marina Theodosiou
- Department of Basic Science, Faculty of Medicine, University of Crete, Heraklion, Greece.,Foundation for Research and Technology, Institute of Molecular Biology and Biotechnology, Heraklion, Greece.,Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Maria Savvaki
- Department of Basic Science, Faculty of Medicine, University of Crete, Heraklion, Greece.,Foundation for Research and Technology, Institute of Molecular Biology and Biotechnology, Heraklion, Greece
| | - Michaela Kavkova
- CEITEC-Central European Institute of Technology, Brno University of Technology, Brno, Czechia
| | - Kostas Theodorakis
- Department of Basic Science, Faculty of Medicine, University of Crete, Heraklion, Greece.,Foundation for Research and Technology, Institute of Molecular Biology and Biotechnology, Heraklion, Greece
| | - Marina Vidaki
- Department of Basic Science, Faculty of Medicine, University of Crete, Heraklion, Greece.,Foundation for Research and Technology, Institute of Molecular Biology and Biotechnology, Heraklion, Greece.,The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Tomas Zikmund
- CEITEC-Central European Institute of Technology, Brno University of Technology, Brno, Czechia
| | - Jozef Kaiser
- CEITEC-Central European Institute of Technology, Brno University of Technology, Brno, Czechia
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Domna Karagogeos
- Department of Basic Science, Faculty of Medicine, University of Crete, Heraklion, Greece.,Foundation for Research and Technology, Institute of Molecular Biology and Biotechnology, Heraklion, Greece
| |
Collapse
|
29
|
Peripheral Brain-Derived Neurotrophic Factor and Contactin-1 Levels in Patients with Attention-Deficit/Hyperactivity Disorder. J Clin Med 2019; 8:jcm8091366. [PMID: 31480710 PMCID: PMC6780884 DOI: 10.3390/jcm8091366] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/20/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) facilitates neuronal growth and plasticity, and is crucial for learning and memory. Contactin-1 (CNTN1) is a member of the subfamily of neural immunoglobulin and is involved in the formation of axon connections in the developing nervous system. This cross-sectional study investigates whether BDNF and CNTN1 affect susceptibility to attention deficit/hyperactivity disorder (ADHD). A total of 136 drug-naïve patients with ADHD (108 boys and 28 girls) and 71 healthy controls (45 boys and 26 girls) were recruited. Blood samples were obtained to measure the plasma levels of BDNF and CNTN1 in each child. We found that BDNF levels in the ADHD boys exceeded those in the control boys, but BDNF levels in the ADHD girls were lower than those in the control girls. Boys who had higher BDNF levels performed worse on the Wechsler Intelligence Scale for Children—Fourth Edition, but girls who had higher BDNF levels made fewer omission errors in the Conners’ Continuous Performance Test. However, CNTN1 level did not differ significantly between patients and controls, and were not correlated to ADHD characteristics, regardless of gender. The findings suggest BDNF may influence sex-specific susceptibility to ADHD, but CNTN1 was not associated with ADHD pathophysiology.
Collapse
|
30
|
Zhang SQ, Fleischer J, Al-Kateb H, Mito Y, Amarillo I, Shinawi M. Intragenic CNTN4 copy number variants associated with a spectrum of neurobehavioral phenotypes. Eur J Med Genet 2019; 63:103736. [PMID: 31422286 DOI: 10.1016/j.ejmg.2019.103736] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 06/26/2019] [Accepted: 08/11/2019] [Indexed: 12/12/2022]
Abstract
Deletions and duplications involving the CNTN4 gene, which encodes for the contactin 4 protein, have been reported in children with autism spectrum disorder (ASD) and other neurodevelopmental phenotypes. In this study, we performed clinical and genetic characterization of three individuals from unrelated families with copy number variants (CNV) (one deletion and two duplications) within CNTN4. The patients exhibited cognitive delay (3/3), growth restriction (3/3), motor delay (2/3), and febrile seizure/epilepsy (2/3). In contrast to previous reports, all probands presented with speech apraxia or delay with no diagnosis of ASD. Parental studies for the proband with the deletion and one of the 2 probands with the duplication revealed paternal origin of the CNTN4 CNV. Interestingly, previously documented CNV involving this gene were mostly inherited from unaffected fathers, raising questions regarding reduced penetrance and potential parent-of-origin effect. Our findings are compared with previously reported patients and patients in the DECIPHER database. The speech impairment in the three probands suggests a role for CNTN4 in language development. We discuss potential factors contributing to phenotypic heterogeneity and reduced penetrance and attempt to find possible genotype-phenotype correlation. Larger cohorts are needed for comprehensive and unbiased phenotyping and molecular characterization that may lead to better understanding of the underlying mechanisms of reduced penetrance, variable expressivity, and potential parent-of-origin effect of copy number variants encompassing CNTN4.
Collapse
Affiliation(s)
| | - Julie Fleischer
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA; Southern Illinois University, Springfield, IL, USA
| | - Hussam Al-Kateb
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yoshiko Mito
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ina Amarillo
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Marwan Shinawi
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
31
|
Castagna C, Merighi A, Lossi L. Decreased Expression of Synaptophysin 1 (SYP1 Major Synaptic Vesicle Protein p38) and Contactin 6 (CNTN6/NB3) in the Cerebellar Vermis of reln Haplodeficient Mice. Cell Mol Neurobiol 2019; 39:833-856. [PMID: 31098770 DOI: 10.1007/s10571-019-00683-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 05/02/2019] [Indexed: 01/17/2023]
Abstract
Reeler heterozygous mice (reln+/-) are seemingly normal but haplodeficient in reln, a gene implicated in autism. Structural/neurochemical alterations in the reln+/- brain are subtle and difficult to demonstrate. Therefore, the usefulness of these mice in translational research is still debated. As evidence implicated several synapse-related genes in autism and the cerebellar vermis is structurally altered in the condition, we have investigated the expression of synaptophysin 1 (SYP1) and contactin 6 (CNTN6) within the vermis of reln+/- mice. Semi-thin plastic sections of the vermis from adult mice of both sexes and different genotypes (reln+/- and reln+/+) were processed with an indirect immunofluorescence protocol. Immunofluorescence was quantified on binary images and statistically analyzed. Reln+/- males displayed a statistically significant reduction of 11.89% in the expression of SYP1 compared to sex-matched wild-type animals, whereas no differences were observed between reln+/+ and reln+/- females. In reln+/- male mice, reductions were particularly evident in the molecular layer: 10.23% less SYP1 than reln+/+ males and 5.84% < reln+/+ females. In reln+/- females, decrease was 9.84% versus reln+/+ males and 5.43% versus reln+/+ females. Both reln+/- males and females showed a stronger decrease in CNTN6 expression throughout all the three cortical layers of the vermis: 17-23% in the granular layer, 24-26% in the Purkinje cell layer, and 9-14% in the molecular layer. Altogether, decrease of vermian SYP1 and CNTN6 in reln+/- mice displayed patterns compatible with the structural modifications of the autistic cerebellum. Therefore, these mice may be a good model in translational studies.
Collapse
Affiliation(s)
- Claudia Castagna
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, I-10095, Turin, Grugliasco (TO), Italy.
| | - Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, I-10095, Turin, Grugliasco (TO), Italy
| | - Laura Lossi
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, I-10095, Turin, Grugliasco (TO), Italy
| |
Collapse
|
32
|
Peng X, Williams J, Smallwood PM, Nathans J. Defining the binding interface of Amyloid Precursor Protein (APP) and Contactin3 (CNTN3) by site-directed mutagenesis. PLoS One 2019; 14:e0219384. [PMID: 31318883 PMCID: PMC6638891 DOI: 10.1371/journal.pone.0219384] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 06/22/2019] [Indexed: 11/19/2022] Open
Abstract
The Amyloid Precursor Protein (APP) and Contactin (CNTN) families of cell-surface proteins have been intensively studied in the context of neural development and neuropsychiatric diseases. Earlier studies demonstrated both genetic and biochemical interactions between the extracellular domains of APP and CNTN3, but their precise binding interfaces were not defined. In the present study, we have used binding assays between APP-alkaline phosphatase (AP) fusion proteins and CNTN-Fc fusion proteins, together with alanine substitution mutagenesis, to show that: (i) the second Fibronectin domain (Fn(2)) in CNTN3 mediates APP binding; (ii) the copper binding domain (CuBD) in APP mediates CNTN3 binding; and (iii) the most important amino acids for APP-CNTN3 binding reside on one face of CNTN3-Fn(2) and on one face of APP-CuBD. These experiments define the regions of direct contact that mediate the binding interaction between APP and CNTN3.
Collapse
Affiliation(s)
- Xi Peng
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - John Williams
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Philip M. Smallwood
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
33
|
Burbach JPH, Meijer DH. Latrophilin's Social Protein Network. Front Neurosci 2019; 13:643. [PMID: 31297045 PMCID: PMC6608557 DOI: 10.3389/fnins.2019.00643] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/05/2019] [Indexed: 01/06/2023] Open
Abstract
Latrophilins (LPHNs) are adhesion GPCRs that are originally discovered as spider's toxin receptors, but are now known to be involved in brain development and linked to several neuronal and non-neuronal disorders. Latrophilins act in conjunction with other cell adhesion molecules and may play a leading role in its network organization. Here, we focus on the main protein partners of latrophilins, namely teneurins, FLRTs and contactins and summarize their respective temporal and spatial expression patterns, links to neurodevelopmental disorders as well as their structural characteristics. We discuss how more recent insights into the separate cell biological functions of these proteins shed light on the central role of latrophilins in this network. We postulate that latrophilins control the refinement of synaptic properties of specific subtypes of neurons, requiring discrete combinations of proteins.
Collapse
Affiliation(s)
- J Peter H Burbach
- Department of Translational Neuroscience, UMCU Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Dimphna H Meijer
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, Netherlands
| |
Collapse
|
34
|
García-Ortiz JE, Zarazúa-Niño AI, Hernández-Orozco AA, Reyes-Oliva EA, Pérez-Ávila CE, Becerra-Solano LE, Galán-Huerta KA, Rivas-Estilla AM, Córdova-Fletes C. Case Report: Whole Exome Sequencing Unveils an Inherited Truncating Variant in CNTN6 (p.Ser189Ter) in a Mexican Child with Autism Spectrum Disorder. J Autism Dev Disord 2019; 50:2247-2251. [PMID: 30826922 DOI: 10.1007/s10803-019-03951-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- José E García-Ortiz
- División de Genética, Centro de Investigación Biomédica de Occidente, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico
| | - Ana I Zarazúa-Niño
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Fco. I. Madero s/n, Mitras Centro, Monterrey, Nuevo León, Mexico
| | - Angélica A Hernández-Orozco
- División de Genética, Centro de Investigación Biomédica de Occidente, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico
| | - Edwin A Reyes-Oliva
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Fco. I. Madero s/n, Mitras Centro, Monterrey, Nuevo León, Mexico
| | - Carlos E Pérez-Ávila
- Unidad Médica de Alta Especialidad Hospital de Gineco-obstetricia, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico
| | - Luis E Becerra-Solano
- Unidad de Investigación Médica, Unidad Médica de Alta Especialidad, Hospital de Pediatría, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico
| | - Kame A Galán-Huerta
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Fco. I. Madero s/n, Mitras Centro, Monterrey, Nuevo León, Mexico
| | - Ana M Rivas-Estilla
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Fco. I. Madero s/n, Mitras Centro, Monterrey, Nuevo León, Mexico
| | - Carlos Córdova-Fletes
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Fco. I. Madero s/n, Mitras Centro, Monterrey, Nuevo León, Mexico.
| |
Collapse
|
35
|
Strawbridge RJ, Ward J, Ferguson A, Graham N, Shaw RJ, Cullen B, Pearsall R, Lyall LM, Johnston KJA, Niedzwiedz CL, Pell JP, Mackay D, Martin JL, Lyall DM, Bailey MES, Smith DJ. Identification of novel genome-wide associations for suicidality in UK Biobank, genetic correlation with psychiatric disorders and polygenic association with completed suicide. EBioMedicine 2019; 41:517-525. [PMID: 30745170 PMCID: PMC6442001 DOI: 10.1016/j.ebiom.2019.02.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/04/2019] [Accepted: 02/04/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Suicide is a major issue for global public health. Suicidality describes a broad spectrum of thoughts and behaviours, some of which are common in the general population. Although suicide results from a complex interaction of multiple social and psychological factors, predisposition to suicidality is at least partly genetic. METHODS Ordinal genome-wide association study of suicidality in the UK Biobank cohort comparing: 'no suicidality' controls (N = 83,557); 'thoughts that life was not worth living' (N = 21,063); 'ever contemplated self-harm' (N = 13,038); 'act of deliberate self-harm in the past' (N = 2498); and 'previous suicide attempt' (N = 2666). OUTCOMES We identified three novel genome-wide significant loci for suicidality (on chromosomes nine, 11 and 13) and moderate-to-strong genetic correlations between suicidality and a range of psychiatric disorders, most notably depression (rg 0·81). INTERPRETATION These findings provide new information about genetic variants relating to increased risk of suicidal thoughts and behaviours. Future work should assess the extent to which polygenic risk scores for suicidality, in combination with non-genetic risk factors, may be useful for stratified approaches to suicide prevention at a population level. FUND: UKRI Innovation-HDR-UK Fellowship (MR/S003061/1). MRC Mental Health Data Pathfinder Award (MC_PC_17217). MRC Doctoral Training Programme Studentship at the University of Glasgow (MR/K501335/1). MRC Doctoral Training Programme Studentship at the Universities of Glasgow and Edinburgh. UKRI Innovation Fellowship (MR/R024774/1).
Collapse
Affiliation(s)
- Rona J Strawbridge
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK; Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Joey Ward
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Amy Ferguson
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Nicholas Graham
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Richard J Shaw
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Breda Cullen
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Robert Pearsall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Laura M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Keira J A Johnston
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK; Division of Psychiatry, College of Medicine, University of Edinburgh, UK; School of Life Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | | - Jill P Pell
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Daniel Mackay
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | | | - Donald M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Mark E S Bailey
- School of Life Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Daniel J Smith
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK.
| |
Collapse
|
36
|
Chatterjee M, Schild D, Teunissen CE. Contactins in the central nervous system: role in health and disease. Neural Regen Res 2019; 14:206-216. [PMID: 30530999 PMCID: PMC6301169 DOI: 10.4103/1673-5374.244776] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/17/2018] [Indexed: 01/06/2023] Open
Abstract
Contactins are a group of cell adhesion molecules that are mainly expressed in the brain and play pivotal roles in the organization of axonal domains, axonal guidance, neuritogenesis, neuronal development, synapse formation and plasticity, axo-glia interactions and neural regeneration. Contactins comprise a family of six members. Their absence leads to malformed axons and impaired nerve conduction. Contactin mediated protein complex formation is critical for the organization of the axon in early central nervous system development. Mutations and differential expression of contactins have been identified in neuro-developmental or neurological disorders. Taken together, contactins are extensively studied in the context of nervous system development. This review summarizes the physiological roles of all six members of the Contactin family in neurodevelopment as well as their involvement in neurological/neurodevelopmental disorders.
Collapse
Affiliation(s)
- Madhurima Chatterjee
- Amsterdam UMC, VU University Medical Center, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Detlev Schild
- Institute of Neurophysiology and Cellular Biophysics, University of Göttingen, Göttingen, Germany
- DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University of Göttingen, Göttingen, Germany
- DFG Excellence Cluster 171, University of Göttingen, Göttingen, Germany
| | - Charlotte E. Teunissen
- Amsterdam UMC, VU University Medical Center, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
37
|
Wang Y, Gray DR, Robbins AK, Crowgey EL, Chanock SJ, Greene MH, McGlynn KA, Nathanson K, Turnbull C, Wang Z, Devoto M, Barthold JS. Subphenotype meta-analysis of testicular cancer genome-wide association study data suggests a role for RBFOX family genes in cryptorchidism susceptibility. Hum Reprod 2019; 33:967-977. [PMID: 29618007 DOI: 10.1093/humrep/dey066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 03/09/2018] [Indexed: 12/25/2022] Open
Abstract
STUDY QUESTION Can subphenotype analysis of genome-wide association study (GWAS) data from subjects with testicular germ cell tumor (TGCT) provide insight into cryptorchidism (undescended testis, UDT) susceptibility? SUMMARY ANSWER Suggestive intragenic GWAS signals common to UDT, TGCT case-case and TGCT case-control analyses occur in genes encoding RBFOX RNA-binding proteins (RBPs) and their neurodevelopmental targets. WHAT IS KNOWN ALREADY UDT is a strong risk factor for TGCT, but while genetic risk factors for TGCT are well-known, genetic susceptibility to UDT is poorly understood and appears to be more complex. STUDY DESIGN, SIZE, DURATION We performed a secondary subphenotype analysis of existing GWAS data from the Testicular Cancer Consortium (TECAC) and compared these results with our previously published UDT GWAS data, and with data previously acquired from studies of the fetal rat gubernaculum. PARTICIPANTS/MATERIALS, SETTING, METHODS Studies from the National Cancer Institute (NCI), United Kingdom (UK) and University of Pennsylvania (Penn) that enrolled white subjects were the source of the TGCT GWAS data. We completed UDT subphenotype case-case (TGCT/UDT vs TGCT/non-UDT) and case-control (TGCT/UDT vs control), collectively referred to as 'TECAC' analyses, followed by a meta-analysis comprising 129 TGCT/UDT cases, 1771 TGCT/non-UDT cases, and 3967 unaffected controls. We reanalyzed our UDT GWAS results comprising 844 cases and 2718 controls by mapping suggestive UDT and TECAC signals (defined as P < 0.001) to genes using Ingenuity Pathway Analysis (IPA®). We compared associated pathways and enriched gene categories common to all analyses after Benjamini-Hochberg multiple testing correction, and analyzed transcript levels and protein expression using qRT-PCR and rat fetal gubernaculum confocal imaging, respectively. MAIN RESULTS AND THE ROLE OF CHANCE We found suggestive signals within 19 genes common to all three analyses, including RBFOX1 and RBFOX3, neurodevelopmental paralogs that encode RBPs targeting (U)GCATG-containing transcripts. Ten of the 19 genes participate in neurodevelopment and/or contribute to risk of neurodevelopmental disorders. Experimentally predicted RBFOX gene targets were strongly overrepresented among suggestive intragenic signals for the UDT (117 of 628 (19%), P = 3.5 × 10-24), TECAC case-case (129 of 711 (18%), P = 2.5 × 10-27) and TECAC case-control (117 of 679 (17%), P = 2 × 10-21) analyses, and a majority of the genes common to all three analyses (12 of 19 (63%), P = 3 × 10-9) are predicted RBFOX targets. Rbfox1, Rbfox2 and their encoded proteins are expressed in the rat fetal gubernaculum. Predicted RBFOX targets are also enriched among transcripts differentially regulated in the fetal gubernaculum during normal development (P = 3 × 10-31), in response to in vitro hormonal stimulation (P = 5 × 10-45) and in the cryptorchid LE/orl rat (P = 2 × 10-42). LARGE SCALE DATA GWAS data included in this study are available in the database of Genotypes and Phenotypes (dbGaP accession numbers phs000986.v1.p1 and phs001349.v1p1). LIMITATIONS, REASONS FOR CAUTION These GWAS data did not reach genome-wide significance for any individual analysis. UDT appears to have a complex etiology that also includes environmental factors, and such complexity may require much larger sample sizes than are currently available. The current methodology may also introduce bias that favors false discovery of larger genes. WIDER IMPLICATIONS OF THE FINDINGS Common suggestive intragenic GWAS signals suggest that RBFOX paralogs and other neurodevelopmental genes are potential UDT risk candidates, and potential TGCT susceptibility modifiers. Enrichment of predicted RBFOX targets among differentially expressed transcripts in the fetal gubernaculum additionally suggests a role for this RBP family in regulation of testicular descent. As RBFOX proteins regulate alternative splicing of Calca to generate calcitonin gene-related peptide, a protein linked to development and function of the gubernaculum, additional studies that address the role of these proteins in UDT are warranted. STUDY FUNDING/COMPETING INTEREST(S) The Eunice Kennedy Shriver National Institute for Child Health and Human Development (R01HD060769); National Center for Research Resources (P20RR20173), National Institute of General Medical Sciences (P20GM103464), Nemours Biomedical Research, the Testicular Cancer Consortium (U01CA164947), the Intramural Research Program of the NCI, a support services contract HHSN26120130003C with IMS, Inc., the Abramson Cancer Center at Penn, National Cancer Institute (CA114478), the Institute of Cancer Research, UK and the Wellcome Trust Case-Control Consortium (WTCCC) 2. None of the authors reports a conflict of interest.
Collapse
Affiliation(s)
- Yanping Wang
- Nemours Biomedical Research/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Dione R Gray
- Nemours Biomedical Research/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Alan K Robbins
- Nemours Biomedical Research/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Erin L Crowgey
- Nemours Biomedical Research/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, Department of Health and Human Services, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Mark H Greene
- Division of Cancer Epidemiology and Genetics, Department of Health and Human Services, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Katherine A McGlynn
- Division of Cancer Epidemiology and Genetics, Department of Health and Human Services, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Katherine Nathanson
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Clare Turnbull
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Zhaoming Wang
- St. Jude Children's Research Hospital, Department of Computational Biology, Memphis, TN, USA
| | - Marcella Devoto
- Division of Genetics, Children's Hospital of Philadelphia and Departments of Biostatistics and Epidemiology, and Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Molecular Medicine, Sapienza University, Rome, Italy
| | | | | |
Collapse
|
38
|
Tassano E, Uccella S, Giacomini T, Severino M, Fiorio P, Gimelli G, Ronchetto P. Clinical and Molecular Characterization of Two Patients with CNTN6 Copy Number Variations. Cytogenet Genome Res 2018; 156:144-149. [PMID: 30508811 DOI: 10.1159/000494152] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2018] [Indexed: 12/15/2022] Open
Abstract
Submicroscopic chromosomal alterations usually involve different protein-coding genes and regulatory elements that are responsible for rare contiguous gene disorders, which complicate the understanding of genotype-phenotype correlations. Chromosome band 3p26.3 contains 3 genes encoding neuronal cell adhesion molecules: CHL1, CNTN6, and CNTN4. We describe 2 boys aged 8 years and 11 years mainly affected by intellectual disability and autism spectrum disorder, who harbor a paternally inherited 3p26.3 microdeletion and a 3p26.3 microduplication, respectively. Both anomalies involved only the CNTN6 gene, which encodes contactin 6, a member of the contactin family (MIM 607220). Contactins show pronounced brain expression and function. Interestingly, phenotypes in reciprocal microdeletions and microduplications of CNTN6 are very similar. In conclusion, our data, added to those reported in the literature, are particularly significant for understanding the pathogenic effect of single gene dosage alterations. As for other recurrent syndromes with variable phenotype, these findings are challenging in genetic counselling because of an evident variable penetrance.
Collapse
|
39
|
Proliferative Cells From Kaposiform Lymphangiomatosis Lesions Resemble Mesenchyme Stem Cell-like Pericytes Defective in Vessel Formation. J Pediatr Hematol Oncol 2018; 40:e495-e504. [PMID: 30256265 DOI: 10.1097/mph.0000000000001284] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Kaposiform lymphangiomatosis (KLA) is a vascular anomaly featuring lymphatic expansion. It has no known cause, no effective treatment, and is associated with high morbidity. Proliferative cells from 3 KLA patient lesions were characterized relative to adiopose-derived mesenchyme stem cells (ADSCs) and cells derived from a patient with the related disease kaposiform hemangioendothelioma (KHE). KLA cells variably expressed markers of mesenchyme stem cells (CD73, CD90, CD105, CD146) and lacked endothelial cell markers (CD31, CD34) as determined by flow cytometry. They expressed markers of vascular pericytes (neural/glial antigen 2, alpha-smooth muscle actin, platelet-derived growth factor-beta receptor, and CXCL12) as determined by quantitative reverse transcription polymerase chain reaction. Lesion cells transcribed vascular markers VEGFC and VEGFD, as well as VCAM-1, the latter of which was confirmed by flow cytometry, consistent with angiogenic MSC-like pericytes. Furthermore, conditioned medium from each was shown to promote the proliferation of growth factor-starved lymphatic endothelial cells. Unlike kaposiform hemangioendothelioma-derived MSC-like pericytes and ADSCs, KLA isolates were defective in support of vascular network formation in co-cultures with either vascular or lymphatic endothelial cells. Genetic analysis by whole exome sequencing revealed novel variant alleles in 2 populations of KLA cells (BAD, TSC1) that may bear on aberrant pericyte growth and function.
Collapse
|
40
|
Deng Y, Zhu W, Zhou X. Immune Regulatory Genes Are Major Genetic Factors to Behcet Disease: Systematic Review. Open Rheumatol J 2018; 12:70-85. [PMID: 30069262 PMCID: PMC6040213 DOI: 10.2174/1874312901812010070] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/04/2018] [Accepted: 06/04/2018] [Indexed: 12/15/2022] Open
Abstract
Behcet's disease (BD) is a chronic refractory multi-system autoimmune disorder that occurs in a genetically susceptible host. Multiple genetic factors have been identified that may contribute to the pathogenesis of BD. The major genes with polymorphisms associated with BD include HLA-B and -A, CIITA, ERAP1, MICA, IL10, IL12A, IL12RB2, IL23R, MEFV, IRF8, TNFAIP3, REL, TLR4, NOD1,2, CCR1,CCR3, GIMAP1,2,4, KLRC4, STAT4, NCOA5, FOXP3, PSORS1C1, FUT2, UBAC2, SUMO4, ADO-EGR2, CEBPB-PTPN1, and JPKL-CNTN5. These genes encode proteins involved mainly in immune regulation and inflammation, and some in transcription and post-translational modification. A complete view of these BD-associated genes may provide a clue to this complex disease in terms of its pathogenesis and exploring potentially targeted therapies for BD.
Collapse
Affiliation(s)
- Yan Deng
- The Second Affiliated Hospital of Nanchang University, Nanchangine>, China.,Department of Internal Medicine/Rheumatology, University of Texas Health Science Center at Houston McGovern Medical School, USA
| | - Weifeng Zhu
- Department of Internal Medicine/Rheumatology, University of Texas Health Science Center at Houston McGovern Medical School, USA.,College of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Xiaodong Zhou
- Department of Internal Medicine/Rheumatology, University of Texas Health Science Center at Houston McGovern Medical School, USA
| |
Collapse
|
41
|
Heise C, Preuss JM, Schroeder JC, Battaglia CR, Kolibius J, Schmid R, Kreutz MR, Kas MJH, Burbach JPH, Boeckers TM. Heterogeneity of Cell Surface Glutamate and GABA Receptor Expression in Shank and CNTN4 Autism Mouse Models. Front Mol Neurosci 2018; 11:212. [PMID: 29970989 PMCID: PMC6018460 DOI: 10.3389/fnmol.2018.00212] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/30/2018] [Indexed: 12/21/2022] Open
Abstract
Autism spectrum disorder (ASD) refers to a large set of neurodevelopmental disorders, which have in common both repetitive behavior and abnormalities in social interactions and communication. Interestingly, most forms of ASD have a strong genetic contribution. However, the molecular underpinnings of this disorder remain elusive. The SHANK3 gene (and to a lesser degree SHANK2) which encode for the postsynaptic density (PSD) proteins SHANK3/SHANK2 and the CONTACTIN 4 gene which encodes for the neuronal glycoprotein CONTACTIN4 (CNTN4) exhibit mutated variants which are associated with ASD. Like many of the other genes associated with ASD, both SHANKs and CNTN4 affect synapse formation and function and are therefore related to the proper development and signaling capability of excitatory and inhibitory neuronal networks in the adult mammal brain. In this study, we used mutant/knock-out mice of Shank2 (Shank2−/−), Shank3 (Shank3αβ−/−), and Cntn4 (Cntn4−/−) as ASD-models to explore whether these mice share a molecular signature in glutamatergic and GABAergic synaptic transmission in ASD-related brain regions. Using a biotinylation assay and subsequent western blotting we focused our analysis on cell surface expression of several ionotropic glutamate and GABA receptor subunits: GluA1, GluA2, and GluN1 were analyzed for excitatory synaptic transmission, and the α1 subunit of the GABAA receptor was analyzed for inhibitory synaptic transmission. We found that both Shank2−/− and Shank3αβ−/− mice exhibit reduced levels of several cell surface glutamate receptors in the analyzed brain regions—especially in the striatum and thalamus—when compared to wildtype controls. Interestingly, even though Cntn4−/− mice also show reduced levels of some cell surface glutamate receptors in the cortex and hippocampus, increased levels of cell surface glutamate receptors were found in the striatum. Moreover, Cntn4−/− mice do not only show brain region-specific alterations in cell surface glutamate receptors but also a downregulation of cell surface GABA receptors in several of the analyzed brain regions. The results of this study suggest that even though mutations in defined genes can be associated with ASD this does not necessarily result in a common molecular phenotype in surface expression of glutamatergic and GABAergic receptor subunits in defined brain regions.
Collapse
Affiliation(s)
- Christopher Heise
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany.,RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Jonathan M Preuss
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Jan C Schroeder
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | | | - Jonas Kolibius
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Rebecca Schmid
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Martien J H Kas
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands.,Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - J Peter H Burbach
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| |
Collapse
|
42
|
Mu D, Xu Y, Zhao T, Watanabe K, Xiao Z, Ye H. Cntn6 deficiency impairs allocentric navigation in mice. Brain Behav 2018; 8:e00969. [PMID: 30106251 PMCID: PMC5991572 DOI: 10.1002/brb3.969] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 03/12/2018] [Accepted: 03/13/2018] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION CNTN6 is an immunoglobulin domain-containing cell adhesion molecule that belongs to the contactin family. It is involved in the development of the nervous system. We aim to determine the effect of Cntn6 deficiency on the allocentric navigation in mice. METHODS We recorded the travel distance and escape time of wild-type and Cntn6 mutant male and female mice in the Morris water maze task according to the protocol. RESULTS There was hardly any Cntn6 expression in the hippocampus of postnatal day 0 (P0) mice, while obvious Cntn6 expression was present in the hippocampal CA1 region of the P7 mice. During the acquisition period of Morris water maze task (Day 1 to 4), Cntn6-/- male mice failed to shorten the escape time to reach platform on the third day, while the travel distance to platform was not significantly different. There was no significant difference in both escape time and travel distance to the platform among all female subjects. In the probe trial test (Day 5), spatial memory of the female mutant mice was mildly affected, while Cntn6-/- male mice were normal. In the spatial relearning test (Day 7 to 10), Cntn6-/- male mice showed no difference in escape time to the platform compared to the wild-type male mice, while Cntn6 deficient female mice required shorter escape time to travel to the platform on day 7, day 8, and day 10. CONCLUSIONS Cntn6 is expressed in the developing hippocampus in mice. Cntn6 deficiency affects spatial learning and memory, indicating that Cntn6 plays a role in the development of hippocampus and affects allocentric navigation of the animals.
Collapse
Affiliation(s)
- Di Mu
- Department of Medical Genetics and Developmental BiologySchool of Basic Medical SciencesBeijing Institute for Brain DisordersCenter of SchizophreniaCapital Medical UniversityBeijingChina
| | - Yiliang Xu
- Department of Medical Genetics and Developmental BiologySchool of Basic Medical SciencesBeijing Institute for Brain DisordersCenter of SchizophreniaCapital Medical UniversityBeijingChina
| | - Tian Zhao
- Department of Medical Genetics and Developmental BiologySchool of Basic Medical SciencesBeijing Institute for Brain DisordersCenter of SchizophreniaCapital Medical UniversityBeijingChina
| | - Kazutada Watanabe
- Department of BioengineeringNagaoka University of TechnologyNagaokaNiigataJapan
| | - Zhi‐Cheng Xiao
- The Key Laboratory of Stem Cell and Regenerative MedicineInstitute of Molecular and Clinical MedicineKunming Medical UniversityKunmingChina
- Department of Anatomy and Developmental BiologyMonash UniversityClaytonMELAustralia
| | - Haihong Ye
- Department of Medical Genetics and Developmental BiologySchool of Basic Medical SciencesBeijing Institute for Brain DisordersCenter of SchizophreniaCapital Medical UniversityBeijingChina
| |
Collapse
|
43
|
Thygesen JH, Wolfe K, McQuillin A, Viñas-Jornet M, Baena N, Brison N, D'Haenens G, Esteba-Castillo S, Gabau E, Ribas-Vidal N, Ruiz A, Vermeesch J, Weyts E, Novell R, Buggenhout GV, Strydom A, Bass N, Guitart M, Vogels A. Neurodevelopmental risk copy number variants in adults with intellectual disabilities and comorbid psychiatric disorders. Br J Psychiatry 2018; 212:287-294. [PMID: 29693535 PMCID: PMC7083594 DOI: 10.1192/bjp.2017.65] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Copy number variants (CNVs) are established risk factors for neurodevelopmental disorders. To date the study of CNVs in psychiatric illness has focused on single disorder populations. The role of CNVs in individuals with intellectual disabilities and psychiatric comorbidities are less well characterised.AimsTo determine the type and frequency of CNVs in adults with intellectual disabilities and comorbid psychiatric disorders. METHOD A chromosomal microarray analysis of 599 adults recruited from intellectual disabilities psychiatry services at three European sites. RESULTS The yield of pathogenic CNVs was high - 13%. Focusing on established neurodevelopmental disorder risk loci we find a significantly higher frequency in individuals with intellectual disabilities and comorbid psychiatric disorder (10%) compared with healthy controls (1.2%, P<0.0001), schizophrenia (3.1%, P<0.0001) and intellectual disability/autism spectrum disorder (6.5%, P < 0.00084) populations. CONCLUSIONS In the largest sample of adults with intellectual disabilities and comorbid psychiatric disorders to date, we find a high rate of pathogenic CNVs. This has clinical implications for the use of genetic investigations in intellectual disability psychiatry.Declaration of interestNone.
Collapse
Affiliation(s)
| | - Kate Wolfe
- Division of Psychiatry, University College London, London, UK
| | | | - Marina Viñas-Jornet
- Genetics Laboratory, UDIAT-Centre Diagnostic, Hospital de Sabadell, Parc Taulí, Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Neus Baena
- Genetics Laboratory, UDIAT-Centre Diagnostic, Hospital de Sabadell, Parc Taulí, Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Nathalie Brison
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | | | - Susanna Esteba-Castillo
- Mental Health and Intellectual Disability Specialized Service, Institut Assistència Sanitària (IAS), Parc Hospitalari Martí i Julià, Girona, Spain
| | - Elisabeth Gabau
- Genetics Laboratory, UDIAT-Centre Diagnostic, Hospital de Sabadell, Parc Taulí, Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Núria Ribas-Vidal
- Mental Health and Intellectual Disability Specialized Service, Institut Assistència Sanitària (IAS), Parc Hospitalari Martí i Julià, Girona, Spain
| | - Anna Ruiz
- Genetics Laboratory, UDIAT-Centre Diagnostic, Hospital de Sabadell, Parc Taulí, Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Joris Vermeesch
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Eddy Weyts
- St Camillus Psychiatric Hospital, Bierbeek, Belgium
| | - Ramon Novell
- Mental Health and Intellectual Disability Specialized Service, Institut Assistència Sanitària (IAS), Parc Hospitalari Martí i Julià, Girona, Spain
| | - Griet Van Buggenhout
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - André Strydom
- Division of Psychiatry, University College London and Department of Forensic and Neurodevelopmental Science, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Nick Bass
- Division of Psychiatry, University College London, London, UK
| | - Miriam Guitart
- Genetics Laboratory, UDIAT-Centre Diagnostic, Hospital de Sabadell, Parc Taulí, Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Annick Vogels
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, Belgium,Correspondence: Annick Vogels, Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, O&N I Herestraat 49 - Box 602, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
44
|
Abstract
Genome-wide association studies (GWAS) have identified more than 100 loci that show robust association with schizophrenia risk. However, due to the complexity of linkage disequilibrium and gene regulatory, it is challenging to pinpoint the causal genes at the risk loci and translate the genetic findings from GWAS into disease mechanism and clinical treatment. Here we systematically predicted the plausible candidate causal genes for schizophrenia at genome-wide level. We utilized different approaches and strategies to predict causal genes for schizophrenia, including Sherlock, SMR, DAPPLE, Prix Fixe, NetWAS, and DEPICT. By integrating the results from different prediction approaches, we identified six top candidates that represent promising causal genes for schizophrenia, including CNTN4, GATAD2A, GPM6A, MMP16, PSMA4, and TCF4. Besides, we also identified 35 additional high-confidence causal genes for schizophrenia. The identified causal genes showed distinct spatio-temporal expression patterns in developing and adult human brain. Cell-type-specific expression analysis indicated that the expression level of the predicted causal genes was significantly higher in neurons compared with oligodendrocytes and microglia (P < 0.05). We found that synaptic transmission-related genes were significantly enriched among the identified causal genes (P < 0.05), providing further support for the dysregulation of synaptic transmission in schizophrenia. Finally, we showed that the top six causal genes are dysregulated in schizophrenia cases compared with controls and knockdown of these genes impaired the proliferation of neuronal cells. Our study depicts the landscape of plausible schizophrenia causal genes for the first time. Further genetic and functional validation of these genes will provide mechanistic insights into schizophrenia pathogenesis and may facilitate to provide potential targets for future therapeutics and diagnostics.
Collapse
Affiliation(s)
- Changguo Ma
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223 China
| | - Chunjie Gu
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223 China
| | - Yongxia Huo
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223 China
| | - Xiaoyan Li
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223 China
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China. .,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China.
| |
Collapse
|
45
|
DNA sequence-level analyses reveal potential phenotypic modifiers in a large family with psychiatric disorders. Mol Psychiatry 2018; 23:2254-2265. [PMID: 29880880 PMCID: PMC6294736 DOI: 10.1038/s41380-018-0087-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 03/30/2018] [Accepted: 04/09/2018] [Indexed: 02/07/2023]
Abstract
Psychiatric disorders are a group of genetically related diseases with highly polygenic architectures. Genome-wide association analyses have made substantial progress towards understanding the genetic architecture of these disorders. More recently, exome- and whole-genome sequencing of cases and families have identified rare, high penetrant variants that provide direct functional insight. There remains, however, a gap in the heritability explained by these complementary approaches. To understand how multiple genetic variants combine to modify both severity and penetrance of a highly penetrant variant, we sequenced 48 whole genomes from a family with a high loading of psychiatric disorder linked to a balanced chromosomal translocation. The (1;11)(q42;q14.3) translocation directly disrupts three genes: DISC1, DISC2, DISC1FP and has been linked to multiple brain imaging and neurocognitive outcomes in the family. Using DNA sequence-level linkage analysis, functional annotation and population-based association, we identified common and rare variants in GRM5 (minor allele frequency (MAF) > 0.05), PDE4D (MAF > 0.2) and CNTN5 (MAF < 0.01) that may help explain the individual differences in phenotypic expression in the family. We suggest that whole-genome sequencing in large families will improve the understanding of the combined effects of the rare and common sequence variation underlying psychiatric phenotypes.
Collapse
|
46
|
Smirnov AV, Kontsevaya GV, Feofanova NA, Anisimova MV, Serova IA, Gerlinskaya LA, Battulin NR, Moshkin MP, Serov OL. Unexpected phenotypic effects of a transgene integration causing a knockout of the endogenous Contactin-5 gene in mice. Transgenic Res 2017; 27:1-13. [PMID: 29264679 DOI: 10.1007/s11248-017-0053-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/01/2017] [Indexed: 01/06/2023]
Abstract
Contactins (Cntn1-6) are a family of neuronal membrane proteins expressed in the brain. They are required for establishing cell-to-cell contacts between neurons and for the growth and maturation of the axons. In humans, structural genomic variations in the Contactin genes are implicated in neurodevelopmental disorders. In addition, population genetic studies associate Contactins loci with obesity and hypertension. Cntn5 knockout mice were first described in 2003, but showed no gross physiological or behavioral abnormalities (just minor auditory defects). We report a novel Cntn5 knockout mouse line generated by a random transgene integration as an outcome of pronuclear microinjection. Investigation of the transgene integration site revealed that the 6Kbp transgene construct coding for the human granulocyte-macrophage colony-stimulating factor (hGMCSF) replaced 170 Kbp of the Cntn5 gene, including four exons. Reverse transcription PCR analysis of the Cntn5 transcripts in the wild-type and transgenic mouse lines showed that splicing of the transgene leads to a set of chimeric hGMCSF-Cntn5 transcript variants, none of which encode functional Cntn5 protein due to introduction of stop codons. Although Cntn5 knockout animals displayed no abnormalities in behavior, we noted that they were leaner, with less body mass and fat percentage than wild-type animals. Their cardiovascular parameters (heart rate, blood pressure and blood flow speed) were elevated compared to controls. These findings link Cntn5 deficiency to obesity and hypertension.
Collapse
Affiliation(s)
- Alexander V Smirnov
- Federal Research Center Institute of Cytology and Genetics, Siberian Division of the Russian Academy of Sciences, Novosibirsk, Russia.
| | - Galina V Kontsevaya
- Federal Research Center Institute of Cytology and Genetics, Siberian Division of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Natalia A Feofanova
- Federal Research Center Institute of Cytology and Genetics, Siberian Division of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Margarita V Anisimova
- Federal Research Center Institute of Cytology and Genetics, Siberian Division of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Irina A Serova
- Federal Research Center Institute of Cytology and Genetics, Siberian Division of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Lyudmila A Gerlinskaya
- Federal Research Center Institute of Cytology and Genetics, Siberian Division of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Nariman R Battulin
- Federal Research Center Institute of Cytology and Genetics, Siberian Division of the Russian Academy of Sciences, Novosibirsk, Russia.,Novosibirsk State University, Novosibirsk, Russia
| | - Mikhail P Moshkin
- Federal Research Center Institute of Cytology and Genetics, Siberian Division of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Oleg L Serov
- Federal Research Center Institute of Cytology and Genetics, Siberian Division of the Russian Academy of Sciences, Novosibirsk, Russia. .,Novosibirsk State University, Novosibirsk, Russia.
| |
Collapse
|
47
|
Chen Y, Wang J, Yang S, Utturkar S, Crodian J, Cummings S, Thimmapuram J, San Miguel P, Kuang S, Gribskov M, Plaut K, Casey T. Effect of high-fat diet on secreted milk transcriptome in midlactation mice. Physiol Genomics 2017; 49:747-762. [PMID: 29093195 DOI: 10.1152/physiolgenomics.00080.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
High-fat diet (HFD) during lactation alters milk composition and is associated with development of metabolic diseases in the offspring. We hypothesized that HFD affects milk microRNA (miRNA) and mRNA content, which potentially impact offspring development. Our objective was to determine the effect of maternal HFD on secreted milk transcriptome. To meet this objective, 4 wk old female ICR mice were divided into two treatments: control diet containing 10% kcal fat and HFD containing 60% kcal fat. After 4 wk on CD or HFD, mice were bred while continuously fed the same diets. On postnatal day 2 (P2), litters were normalized to 10 pups, and half the pups in each litter were cross-fostered between treatments. Milk was collected from dams on P10 and P12. Total RNA was isolated from milk fat fraction of P10 samples and used for mRNA-Seq and small RNA-Seq. P12 milk was used to determine macronutrient composition. After 4 wk of prepregnancy feeding HFD mice weighed significantly more than did the control mice. Lactose and fat concentration were significantly ( P < 0.05) higher in milk of HFD dams. Pup weight was significantly greater ( P < 0.05) in groups suckled by HFD vs. control dams. There were 25 miRNA and over 1,500 mRNA differentially expressed (DE) in milk of HFD vs. control dams. DE mRNA and target genes of DE miRNA enriched categories that were primarily related to multicellular organismal development. Maternal HFD impacts mRNA and miRNA content of milk, if bioactive nucleic acids are absorbed by neonate differences may affect development.
Collapse
Affiliation(s)
- Y. Chen
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana
| | - J. Wang
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana
| | - S. Yang
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana
| | - S. Utturkar
- Bioinformatics Core, Purdue University, West Lafayette, Indiana
| | - J. Crodian
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana
| | - S. Cummings
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana
| | - J. Thimmapuram
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana
| | - P. San Miguel
- Genomics Core at Purdue University, West Lafayette, Indiana
| | - S. Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana
| | - M. Gribskov
- Bioinformatics Core, Purdue University, West Lafayette, Indiana
| | - K. Plaut
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana
| | - T. Casey
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana
| |
Collapse
|
48
|
Lowther C, Merico D, Costain G, Waserman J, Boyd K, Noor A, Speevak M, Stavropoulos DJ, Wei J, Lionel AC, Marshall CR, Scherer SW, Bassett AS. Impact of IQ on the diagnostic yield of chromosomal microarray in a community sample of adults with schizophrenia. Genome Med 2017; 9:105. [PMID: 29187259 PMCID: PMC5708103 DOI: 10.1186/s13073-017-0488-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/01/2017] [Indexed: 11/10/2022] Open
Abstract
Background Schizophrenia is a severe psychiatric disorder associated with IQ deficits. Rare copy number variations (CNVs) have been established to play an important role in the etiology of schizophrenia. Several of the large rare CNVs associated with schizophrenia have been shown to negatively affect IQ in population-based controls where no major neuropsychiatric disorder is reported. The aim of this study was to examine the diagnostic yield of microarray testing and the functional impact of genome-wide rare CNVs in a community ascertained cohort of adults with schizophrenia and low (< 85) or average (≥ 85) IQ. Methods We recruited 546 adults of European ancestry with schizophrenia from six community psychiatric clinics in Canada. Each individual was assigned to the low or average IQ group based on standardized tests and/or educational attainment. We used rigorous methods to detect genome-wide rare CNVs from high-resolution microarray data. We compared the burden of rare CNVs classified as pathogenic or as a variant of unknown significance (VUS) between each of the IQ groups and the genome-wide burden and functional impact of rare CNVs after excluding individuals with a pathogenic CNV. Results There were 39/546 (7.1%; 95% confidence interval [CI] = 5.2–9.7%) schizophrenia participants with at least one pathogenic CNV detected, significantly more of whom were from the low IQ group (odds ratio [OR] = 5.01 [2.28–11.03], p = 0.0001). Secondary analyses revealed that individuals with schizophrenia and average IQ had the lowest yield of pathogenic CNVs (n = 9/325; 2.8%), followed by those with borderline intellectual functioning (n = 9/130; 6.9%), non-verbal learning disability (n = 6/29; 20.7%), and co-morbid intellectual disability (n = 15/62; 24.2%). There was no significant difference in the burden of rare CNVs classified as a VUS between any of the IQ subgroups. There was a significantly (p=0.002) increased burden of rare genic duplications in individuals with schizophrenia and low IQ that persisted after excluding individuals with a pathogenic CNV. Conclusions Using high-resolution microarrays we were able to demonstrate for the first time that the burden of pathogenic CNVs in schizophrenia differs significantly between IQ subgroups. The results of this study have implications for clinical practice and may help inform future rare variant studies of schizophrenia using next-generation sequencing technologies. Electronic supplementary material The online version of this article (doi:10.1186/s13073-017-0488-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chelsea Lowther
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, 33 Russell Street, Room 1100, Toronto, ON, Canada, M5S 2S1.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Daniele Merico
- Deep Genomics Inc, Toronto, ON, Canada.,The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Gregory Costain
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, 33 Russell Street, Room 1100, Toronto, ON, Canada, M5S 2S1.,Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, ON, Canada
| | | | - Kerry Boyd
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Abdul Noor
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Marsha Speevak
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | | | - John Wei
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Anath C Lionel
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Christian R Marshall
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.,Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Genome Diagnostics, Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Stephen W Scherer
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,McLaughlin Centre, University of Toronto, Toronto, ON, Canada
| | - Anne S Bassett
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, 33 Russell Street, Room 1100, Toronto, ON, Canada, M5S 2S1. .,Institute of Medical Science, University of Toronto, Toronto, ON, Canada. .,Toronto General Research Institute, University Health Network, Toronto, ON, Canada. .,Cambell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada. .,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
49
|
Schormair B, Zhao C, Bell S, Tilch E, Salminen AV, Pütz B, Dauvilliers Y, Stefani A, Högl B, Poewe W, Kemlink D, Sonka K, Bachmann CG, Paulus W, Trenkwalder C, Oertel WH, Hornyak M, Teder-Laving M, Metspalu A, Hadjigeorgiou GM, Polo O, Fietze I, Ross OA, Wszolek Z, Butterworth AS, Soranzo N, Ouwehand WH, Roberts DJ, Danesh J, Allen RP, Earley CJ, Ondo WG, Xiong L, Montplaisir J, Gan-Or Z, Perola M, Vodicka P, Dina C, Franke A, Tittmann L, Stewart AFR, Shah SH, Gieger C, Peters A, Rouleau GA, Berger K, Oexle K, Di Angelantonio E, Hinds DA, Müller-Myhsok B, Winkelmann J. Identification of novel risk loci for restless legs syndrome in genome-wide association studies in individuals of European ancestry: a meta-analysis. Lancet Neurol 2017; 16:898-907. [PMID: 29029846 PMCID: PMC5755468 DOI: 10.1016/s1474-4422(17)30327-7] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 08/10/2017] [Accepted: 08/17/2017] [Indexed: 02/02/2023]
Abstract
BACKGROUND Restless legs syndrome is a prevalent chronic neurological disorder with potentially severe mental and physical health consequences. Clearer understanding of the underlying pathophysiology is needed to improve treatment options. We did a meta-analysis of genome-wide association studies (GWASs) to identify potential molecular targets. METHODS In the discovery stage, we combined three GWAS datasets (EU-RLS GENE, INTERVAL, and 23andMe) with diagnosis data collected from 2003 to 2017, in face-to-face interviews or via questionnaires, and involving 15 126 cases and 95 725 controls of European ancestry. We identified common variants by fixed-effect inverse-variance meta-analysis. Significant genome-wide signals (p≤5 × 10-8) were tested for replication in an independent GWAS of 30 770 cases and 286 913 controls, followed by a joint analysis of the discovery and replication stages. We did gene annotation, pathway, and gene-set-enrichment analyses and studied the genetic correlations between restless legs syndrome and traits of interest. FINDINGS We identified and replicated 13 new risk loci for restless legs syndrome and confirmed the previously identified six risk loci. MEIS1 was confirmed as the strongest genetic risk factor for restless legs syndrome (odds ratio 1·92, 95% CI 1·85-1·99). Gene prioritisation, enrichment, and genetic correlation analyses showed that identified pathways were related to neurodevelopment and highlighted genes linked to axon guidance (associated with SEMA6D), synapse formation (NTNG1), and neuronal specification (HOXB cluster family and MYT1). INTERPRETATION Identification of new candidate genes and associated pathways will inform future functional research. Advances in understanding of the molecular mechanisms that underlie restless legs syndrome could lead to new treatment options. We focused on common variants; thus, additional studies are needed to dissect the roles of rare and structural variations. FUNDING Deutsche Forschungsgemeinschaft, Helmholtz Zentrum München-Deutsches Forschungszentrum für Gesundheit und Umwelt, National Research Institutions, NHS Blood and Transplant, National Institute for Health Research, British Heart Foundation, European Commission, European Research Council, National Institutes of Health, National Institute of Neurological Disorders and Stroke, NIH Research Cambridge Biomedical Research Centre, and UK Medical Research Council.
Collapse
Affiliation(s)
- Barbara Schormair
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany
| | - Chen Zhao
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany
| | - Steven Bell
- National Institute for Health Research Blood and Transplant Unit in Donor Health and Genomics at the University of Cambridge, Strangeways Research Laboratory, University of Cambridge, Cambridge, UK; MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Cambridge, UK; National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Erik Tilch
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany
| | - Aaro V Salminen
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany
| | - Benno Pütz
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Yves Dauvilliers
- Sleep-Wake Disorders Centre, Department of Neurology, Hôpital Gui-de-Chauliac, INSERM U1061, CHU Montpellier, France
| | - Ambra Stefani
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Birgit Högl
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Werner Poewe
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - David Kemlink
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine and General University Hospital in Prague, Charles University, Prague, Czech Republic
| | - Karel Sonka
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine and General University Hospital in Prague, Charles University, Prague, Czech Republic
| | | | - Walter Paulus
- Department of Clinical Neurophysiology, University Medical Centre, Georg August University Göttingen, Göttingen, Germany
| | - Claudia Trenkwalder
- Clinic for Neurosurgery, University Medical Centre, Georg August University Göttingen, Göttingen, Germany; Paracelsus-Elena Hospital, Centre of Parkinsonism and Movement Disorders, Kassel, Germany
| | - Wolfgang H Oertel
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany; Department of Neurology, Philipps University Marburg, Marburg, Germany
| | - Magdolna Hornyak
- Department of Neurology, University of Ulm, Ulm, Germany; Neuropsychiatry Centre Erding/München, Erding, Germany
| | - Maris Teder-Laving
- Estonian Genome Centre, University of Tartu and Estonian Biocentre, Tartu, Estonia
| | - Andres Metspalu
- Estonian Genome Centre, University of Tartu and Estonian Biocentre, Tartu, Estonia
| | - Georgios M Hadjigeorgiou
- Laboratory of Neurogenetics, Department of Neurology, Faculty of Medicine, University of Thessaly, University Hospital of Larissa, Biopolis, Larissa, Greece
| | - Olli Polo
- Unesta Research Centre, Tampere, Finland; Department of Pulmonary Diseases, Tampere University Hospital, Tampere, Finland
| | - Ingo Fietze
- Department of Cardiology and Angiology, Centre of Sleep Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Adam S Butterworth
- National Institute for Health Research Blood and Transplant Unit in Donor Health and Genomics at the University of Cambridge, Strangeways Research Laboratory, University of Cambridge, Cambridge, UK; MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Cambridge, UK; National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK; British Heart Foundation Centre of Excellence, Division of Cardiovascular Medicine, Addenbrooke's Hospital, Cambridge, UK
| | - Nicole Soranzo
- National Institute for Health Research Blood and Transplant Unit in Donor Health and Genomics at the University of Cambridge, Strangeways Research Laboratory, University of Cambridge, Cambridge, UK; Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; Department of Human Genetics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Willem H Ouwehand
- National Institute for Health Research Blood and Transplant Unit in Donor Health and Genomics at the University of Cambridge, Strangeways Research Laboratory, University of Cambridge, Cambridge, UK; Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; NHS Blood and Transplant, Cambridge, UK; British Heart Foundation Centre of Excellence, Division of Cardiovascular Medicine, Addenbrooke's Hospital, Cambridge, UK; Department of Human Genetics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - David J Roberts
- NHS Blood and Transplant, Oxford, UK; Radcliffe Department of Medicine, BRC Haematology Theme and NHS Blood and Transplant, John Radcliffe Hospital, Headington, Oxford, UK; Department of Haematology and BRC Haematology Theme, Churchill Hospital, Oxford, UK
| | - John Danesh
- National Institute for Health Research Blood and Transplant Unit in Donor Health and Genomics at the University of Cambridge, Strangeways Research Laboratory, University of Cambridge, Cambridge, UK; MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Cambridge, UK; National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK; British Heart Foundation Centre of Excellence, Division of Cardiovascular Medicine, Addenbrooke's Hospital, Cambridge, UK; Department of Human Genetics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Richard P Allen
- Center for Restless Legs Study, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Christopher J Earley
- Center for Restless Legs Study, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - William G Ondo
- Department of Neurology, Methodist Neurological Institute, Houston, TX, USA
| | - Lan Xiong
- Laboratoire de Neurogénétique, Centre de Recherche, Institut Universitaire en Santé Mentale de Montréal, Montréal, QC, Canada; Département de Psychiatrie, Université de Montréal, Montréal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Jacques Montplaisir
- Département de Psychiatrie, Université de Montréal, Montréal, QC, Canada; Hôpital du Sacré-Coeur de Montréal, 67120, Center for Advanced Research in Sleep Medicine, Montréal, QC, Canada
| | - Ziv Gan-Or
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada; Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Markus Perola
- Department of Health, National Institute for Health and Welfare, Helsinki, Finland; Institute of Molecular Medicine FIMM, University of Helsinki, Helsinki, Finland
| | - Pavel Vodicka
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine, Academy of Science of Czech Republic, Prague, Czech Republic; Biomedical Centre, Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| | - Christian Dina
- Inserm UMR1087, CNRS UMR 6291, Institut du Thorax, Nantes, France; Centre Hospitalier Universitaire (CHU) Nantes, Université de Nantes, France
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Lukas Tittmann
- PopGen Biobank and Institute of Epidemiology, Christian Albrechts University Kiel, Kiel, Germany
| | - Alexandre F R Stewart
- John and Jennifer Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Svati H Shah
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA; Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Christian Gieger
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany; Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany; German Centre for Diabetes Research (DZD), Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany; German Centre for Diabetes Research (DZD), Neuherberg, Germany; German Centre for Cardiovascular Disease Research (DZHK), Berlin, Germany
| | - Guy A Rouleau
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada; Department of Human Genetics, McGill University, Montréal, QC, Canada; Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Klaus Berger
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - Konrad Oexle
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany
| | - Emanuele Di Angelantonio
- National Institute for Health Research Blood and Transplant Unit in Donor Health and Genomics at the University of Cambridge, Strangeways Research Laboratory, University of Cambridge, Cambridge, UK; MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Cambridge, UK; NHS Blood and Transplant, Cambridge, UK; National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK; British Heart Foundation Centre of Excellence, Division of Cardiovascular Medicine, Addenbrooke's Hospital, Cambridge, UK
| | | | - Bertram Müller-Myhsok
- Max Planck Institute of Psychiatry, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Juliane Winkelmann
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Institute of Human Genetics, Technische Universität München, Munich, Germany; Neurologische Klinik und Poliklinik, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
| |
Collapse
|
50
|
Abstract
In the current issue of Neuron, Huang et al. (2017) provide new insights from a consortium study of Tourette syndrome pinpointing copy number variations that are involved in the genomic architecture and implicate genes of interest.
Collapse
Affiliation(s)
- Anne S Bassett
- Clinical Genetics Research Program and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Division of Cardiology, Department of Medicine, Toronto General Research Institute, University Health Network, Toronto, ON, Canada; The Dalglish 22q Clinic for Adults with 22q11.2 Deletion Syndrome, and Department of Psychiatry, University Health Network, Toronto, ON, Canada.
| | - Stephen W Scherer
- Institute of Medical Science, University of Toronto, ON, Canada; The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada; McLaughlin Centre and Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|