1
|
Furia F, Levy AM, Theunis M, Bamshad MJ, Bartos MN, Bijlsma EK, Brancati F, Cejudo L, Chong JX, De Luca C, Dean SJ, Egense A, Goel H, Guenzel AJ, Hüffmeier U, Legius E, Mancini GMS, Marcos-Alcalde I, Niclass T, Planes M, Redon S, Ros-Pardo D, Rouault K, Schot R, Schuhmann S, Shen JJ, Tao AM, Thiffault I, Van Esch H, Wentzensen IM, Barakat TS, Møller RS, Gomez-Puertas P, Chung WK, Gardella E, Tümer Z. The phenotypic and genotypic spectrum of individuals with mono- or biallelic ANK3 variants. Clin Genet 2024; 106:574-584. [PMID: 38988293 PMCID: PMC11444875 DOI: 10.1111/cge.14587] [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: 05/03/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 07/12/2024]
Abstract
ANK3 encodes ankyrin-G, a protein involved in neuronal development and signaling. Alternative splicing gives rise to three ankyrin-G isoforms comprising different domains with distinct expression patterns. Mono- or biallelic ANK3 variants are associated with non-specific syndromic intellectual disability in 14 individuals (seven with monoallelic and seven with biallelic variants). In this study, we describe the clinical features of 13 additional individuals and review the data on a total of 27 individuals (16 individuals with monoallelic and 11 with biallelic ANK3 variants) and demonstrate that the phenotype for biallelic variants is more severe. The phenotypic features include language delay (92%), autism spectrum disorder (76%), intellectual disability (78%), hypotonia (65%), motor delay (68%), attention deficit disorder (ADD) or attention deficit hyperactivity disorder (ADHD) (57%), sleep disturbances (50%), aggressivity/self-injury (37.5%), and epilepsy (35%). A notable phenotypic difference was presence of ataxia in three individuals with biallelic variants, but in none of the individuals with monoallelic variants. While the majority of the monoallelic variants are predicted to result in a truncated protein, biallelic variants are almost exclusively missense. Moreover, mono- and biallelic variants appear to be localized differently across the three different ankyrin-G isoforms, suggesting isoform-specific pathological mechanisms.
Collapse
Affiliation(s)
- Francesca Furia
- Department of Epilepsy Genetics and Personalized Treatment, The Danish Epilepsy Centre, Dianalund, Denmark
- Faculty of Health Science, University of Southern Denmark (SDU), Odense, Denmark
| | - Amanda M Levy
- Department of Clinical Genetics, Kennedy Center, Copenhagen University Hospital, Copenhagen, Denmark
| | - Miel Theunis
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Michael J Bamshad
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, Washington, USA
- Brotman-Baty Institute for Precision Medicine, University of Washington, Seattle, Washington, USA
- Department of Pediatrics, Division of Genetic Medicine, Seattle Children's Hospital, Seattle, Washington, USA
| | - Meghan N Bartos
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Emilia K Bijlsma
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Francesco Brancati
- Human Genetics, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
- Human Functional Genetics Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Roma, Rome, Italy
| | - Lucile Cejudo
- CHU de Poitiers, Service de Génétique, Poitiers, France
| | - Jessica X Chong
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, Washington, USA
- Brotman-Baty Institute for Precision Medicine, University of Washington, Seattle, Washington, USA
| | - Chiara De Luca
- Human Genetics, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Sarah Joy Dean
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Alena Egense
- Division of Genomic Medicine, Department of Pediatrics, University of California Davis, Sacramento, California, USA
| | - Himanshu Goel
- General Genetics Service, Hunter Genetics, Waratah, New South Wales, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
| | | | - Ulrike Hüffmeier
- Institute of Human Genetics, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Eric Legius
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Grazia M S Mancini
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Iñigo Marcos-Alcalde
- Molecular Modeling Group, Centro de Biología Molecular Severo Ochoa (CBM, CSIC-UAM), Madrid, Spain
| | | | - Marc Planes
- Service de Génétique Clinique, CHRU de Brest, Brest, France
| | - Sylvia Redon
- Service de Génétique Médicale et Biologie de la Reproduction, CHU de Brest, Brest, France
- Université de Brest, INSERM, Etablissement Français du Sang, UMR 1078, Brest, France
| | - David Ros-Pardo
- Molecular Modeling Group, Centro de Biología Molecular Severo Ochoa (CBM, CSIC-UAM), Madrid, Spain
| | - Karen Rouault
- Service de Génétique Médicale et Biologie de la Reproduction, CHU de Brest, Brest, France
- Université de Brest, INSERM, Etablissement Français du Sang, UMR 1078, Brest, France
| | - Rachel Schot
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Discovery Unit, Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Sarah Schuhmann
- Institute of Human Genetics, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Joseph J Shen
- Division of Genomic Medicine, Department of Pediatrics, University of California Davis, Sacramento, California, USA
| | - Alice M Tao
- Vagelos School of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Isabelle Thiffault
- Department of Pathology, Children's Mercy Kansas City, Kansas City, Missouri, USA
- Genomic Medicine Center, Children's Mercy Kansas City, Kansas City, Missouri, USA
| | - Hilde Van Esch
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
- Laboratory for the Genetics of Cognition, KU Leuven, Leuven, Belgium
| | | | - Tahsin Stefan Barakat
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Discovery Unit, Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Rikke S Møller
- Department of Epilepsy Genetics and Personalized Treatment, The Danish Epilepsy Centre, Dianalund, Denmark
- Faculty of Health Science, University of Southern Denmark (SDU), Odense, Denmark
| | - Paulino Gomez-Puertas
- Molecular Modeling Group, Centro de Biología Molecular Severo Ochoa (CBM, CSIC-UAM), Madrid, Spain
| | - Wendy K Chung
- Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Elena Gardella
- Department of Epilepsy Genetics and Personalized Treatment, The Danish Epilepsy Centre, Dianalund, Denmark
- Faculty of Health Science, University of Southern Denmark (SDU), Odense, Denmark
- Department of Neurophysiology, The Danish Epilepsy Centre, Dianalund, Denmark
| | - Zeynep Tümer
- Department of Clinical Genetics, Kennedy Center, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Zenge C, Ordureau A. Ubiquitin system mutations in neurological diseases. Trends Biochem Sci 2024; 49:875-887. [PMID: 38972780 PMCID: PMC11455613 DOI: 10.1016/j.tibs.2024.06.011] [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: 03/20/2024] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 07/09/2024]
Abstract
Neuronal ubiquitin balance impacts the fate of countless cellular proteins, and its disruption is associated with various neurological disorders. The ubiquitin system is critical for proper neuronal cell state transitions and the clearance of misfolded or aggregated proteins that threaten cellular integrity. This article reviews the state of and recent advancements in our understanding of the disruptions to components of the ubiquitin system, in particular E3 ligases and deubiquitylases, in neurodevelopmental and neurodegenerative diseases. Specific focus is on enzymes with recent progress in their characterization, including identifying enzyme-substrate pairs, the use of stem cell and animal models, and the development of therapeutics for ubiquitin-related diseases.
Collapse
Affiliation(s)
- Colin Zenge
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Alban Ordureau
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
3
|
Togawa S, Usui N, Doi M, Kobayashi Y, Koyama Y, Nakamura Y, Shinoda K, Kobayashi H, Shimada S. Neuroprotective effects of Si-based hydrogen-producing agent on 6-hydroxydopamine-induced neurotoxicity in juvenile mouse model. Behav Brain Res 2024; 468:115040. [PMID: 38723675 DOI: 10.1016/j.bbr.2024.115040] [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: 01/18/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024]
Abstract
Neurotoxins have been extensively investigated, particularly in the field of neuroscience. They induce toxic damage, oxidative stress, and inflammation on neurons, triggering neuronal dysfunction and neurodegenerative diseases. Here we demonstrate the neuroprotective effect of a silicon (Si)-based hydrogen-producing agent (Si-based agent) in a juvenile neurotoxic mouse model induced by 6-hydroxydopamine (6-OHDA). The Si-based agent produces hydrogen in bowels and functions as an antioxidant and anti-inflammatory agent. However, the effects of the Si-based agent on neural degeneration in areas other than the lesion and behavioral alterations caused by it are largely unknown. Moreover, the neuroprotective effects of Si-based agent in the context of lactation and use during infancy have not been explored in prior studies. In this study, we show the neuroprotective effect of the Si-based agent on 6-OHDA during lactation period and infancy using the mouse model. The Si-based agent safeguards against the degradation and neuronal cell death of dopaminergic neurons and loss of dopaminergic fibers in the striatum (STR) and ventral tegmental area (VTA) caused by 6-OHDA. Furthermore, the Si-based agent exhibits a neuroprotective effect on the length of axon initial segment (AIS) in the layer 2/3 (L2/3) neurons of the medial prefrontal cortex (mPFC). As a result, the Si-based agent mitigates hyperactive behavior in a juvenile neurotoxic mouse model induced by 6-OHDA. These results suggest that the Si-based agent serves as an effective neuroprotectant and antioxidant against neurotoxic effects in the brain, offering the possibility of the Si-based agent as a neuroprotectant for nervous system diseases.
Collapse
Affiliation(s)
- Shogo Togawa
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan
| | - Noriyoshi Usui
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; Omics Center, Center of Medical Innovation and Translational Research, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; United Graduate School of Child Development, Osaka University, Suita 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Suita 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan.
| | - Miyuki Doi
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
| | - Yuki Kobayashi
- SANKEN (Institute of Scientific and Industrial Research), Osaka University, Ibaraki, 567-0047, Japan
| | - Yoshihisa Koyama
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
| | - Yukiko Nakamura
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
| | - Koh Shinoda
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan
| | - Hikaru Kobayashi
- SANKEN (Institute of Scientific and Industrial Research), Osaka University, Ibaraki, 567-0047, Japan
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; United Graduate School of Child Development, Osaka University, Suita 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Suita 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
| |
Collapse
|
4
|
Oliva M, Sarkar MK, March ME, Saeidian AH, Mentch FD, Hsieh CL, Tang F, Uppala R, Patrick MT, Li Q, Bogle R, Kahlenberg JM, Watson D, Glessner JT, Tsoi LC, Hakonarson H, Gudjonsson JE, Smith KM, Riley-Gillis B. Multi-ancestry Genome-Wide Association Meta-Analysis Identifies Novel Loci in Atopic Dermatitis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.17.24308897. [PMID: 38946956 PMCID: PMC11213042 DOI: 10.1101/2024.06.17.24308897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Atopic dermatitis (AD) is a highly heritable and common inflammatory skin condition affecting children and adults worldwide. Multi-ancestry approaches to AD genetic association studies are poised to boost power to detect genetic signal and identify ancestry-specific loci contributing to AD risk. Here, we present a multi-ancestry GWAS meta-analysis of twelve AD cohorts from five ancestral populations totaling 56,146 cases and 602,280 controls. We report 101 genomic loci associated with AD, including 15 loci that have not been previously associated with AD or eczema. Fine-mapping, QTL colocalization, and cell-type enrichment analyses identified genes and cell types implicated in AD pathophysiology. Functional analyses in keratinocytes provide evidence for genes that could play a role in AD through epidermal barrier function. Our study provides new insights into the etiology of AD by harnessing multiple genetic and functional approaches to unveil the mechanisms by which AD-associated variants impact genes and cell types.
Collapse
Affiliation(s)
- Meritxell Oliva
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, USA
| | | | | | | | | | - Chen-Lin Hsieh
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, USA
| | - Fanying Tang
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, USA
| | | | | | - Qinmengge Li
- University of Michigan, Ann Arbor, Michigan 48109
| | | | | | - Deborah Watson
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | | | - Lam C. Tsoi
- University of Michigan, Ann Arbor, Michigan 48109
| | | | | | | | | |
Collapse
|
5
|
Ding X, Wu Y, Rodriguez V, Ricco E, Okoh JT, Liu Y, Kraushaar DC, Rasband MN. Age-dependent regulation of axoglial interactions and behavior by oligodendrocyte AnkyrinG. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.01.587609. [PMID: 38617359 PMCID: PMC11014615 DOI: 10.1101/2024.04.01.587609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The bipolar disorder (BD) risk gene ANK3 encodes the scaffolding protein AnkyrinG (AnkG). In neurons, AnkG regulates polarity and ion channel clustering at axon initial segments and nodes of Ranvier. Disruption of neuronal AnkG causes BD-like phenotypes in mice. During development, AnkG is also expressed at comparable levels in oligodendrocytes and facilitates the efficient assembly of paranodal junctions. However, the physiological roles of glial AnkG in the mature nervous system, and its contributions to BD-like phenotypes, remain unexplored. Here, we generated oligodendroglia-specific AnkG conditional knockout mice and observed the destabilization of axoglial interactions in aged but not young adult mice. In addition, these mice exhibited profound histological, electrophysiological, and behavioral pathophysiologies. Unbiased translatomic profiling revealed potential compensatory machineries. These results highlight the critical functions of glial AnkG in maintaining proper axoglial interactions throughout aging and suggests a previously unrecognized contribution of oligodendroglial AnkG to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Xiaoyun Ding
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Yu Wu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Victoria Rodriguez
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX 77030
| | - Emily Ricco
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX 77030
| | - James T. Okoh
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Yanhong Liu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Daniel C. Kraushaar
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX 77030
| | - Matthew N. Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
6
|
Huyghebaert J, Mateiu L, Elinck E, Van Rossem KE, Christiaenssen B, D'Incal CP, McCormack MK, Lazzarini A, Vandeweyer G, Kooy RF. Identification of a DLG3 stop mutation in the MRX20 family. Eur J Hum Genet 2024; 32:317-323. [PMID: 38273165 PMCID: PMC10923781 DOI: 10.1038/s41431-024-01537-7] [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: 06/30/2023] [Revised: 12/22/2023] [Accepted: 01/09/2024] [Indexed: 01/27/2024] Open
Abstract
Here, we identified the causal mutation in the MRX20 family, one of the larger X-linked pedigrees that have been described in which no gene had been identified up till now. In 1995, the putative disease gene had been mapped to the pericentromeric region on the X chromosome, but no follow-up studies were performed. Here, whole exome sequencing (WES) on two affected and one unaffected family member revealed the c.195del/p.(Thr66ProfsTer55) mutation in the DLG3 gene (NM_021120.4) that segregated with the affected individuals in the family. DLG3 mutations have been consequently associated with intellectual disability and are a plausible explanation for the clinical abnormalities observed in this family. In addition, we identified two other variants co-segregating with the phenotype: a stop gain mutation in SSX1 (c.358G>T/p.(Glu120Ter)) (NM_001278691.2) and a nonsynonymous SNV in USP27X (c.56 A>G/p.(Gln19Arg)) (NM_001145073.3). RNA sequencing revealed 14 differentially expressed genes (p value < 0.1) in 7 affected males compared to 4 unaffected males of the family, including four genes known to be associated with neurological disorders. Thus, in this paper we identified the c.195del/p.(Thr66ProfsTer55) mutation in the DLG3 gene (NM_021120.4) as likely responsible for the phenotype observed in the MRX20 family.
Collapse
Affiliation(s)
| | - Ligia Mateiu
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Ellen Elinck
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | | | | | | | - Michael K McCormack
- Department of Psychiatry, Rutgers University-Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
- Department of Cell Biology and Neurosciences, Virtua Health College of Medicine and Life Sciences of Rowan University, Stratford, NJ, 08084, USA
| | - Alice Lazzarini
- Department of Neurology, Rutgers University-Robert Wood Johnson Medical School, New Brunswick, NJ, 08903, USA
| | - Geert Vandeweyer
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - R Frank Kooy
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
7
|
Iwahashi M, Yoshimura T, Harigai W, Takuma K, Hashimoto H, Katayama T, Hayata-Takano A. Pituitary adenylate cyclase-activating polypeptide deficient mice show length abnormalities of the axon initial segment. J Pharmacol Sci 2023; 153:175-182. [PMID: 37770159 DOI: 10.1016/j.jphs.2023.08.006] [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: 06/30/2023] [Revised: 08/08/2023] [Accepted: 08/25/2023] [Indexed: 10/03/2023] Open
Abstract
We previously found that pituitary adenylate cyclase-activating polypeptide (PACAP)-deficient (PACAP-/-) mice exhibit dendritic spine morphology impairment and neurodevelopmental disorder (NDD)-like behaviors such as hyperactivity, increased novelty-seeking behavior, and deficient pre-pulse inhibition. Recent studies have indicated that rodent models of NDDs (e.g., attention-deficit hyperactivity disorder (ADHD) and autism spectrum disorder) show abnormalities in the axon initial segment (AIS). Here, we revealed that PACAP-/- mice exhibited a longer AIS length in layer 2/3 pyramidal neurons of the primary somatosensory barrel field compared with wild-type control mice. Further, we previously showed that a single injection of atomoxetine, an ADHD drug, improved hyperactivity in PACAP-/- mice. In this study, we found that repeated treatments of atomoxetine significantly improved AIS abnormality along with hyperactivity in PACAP-/- mice. These results suggest that AIS abnormalities are associated with NDDs-like behaviors in PACAP-/- mice. Thus, improvement in AIS abnormalities will be a novel drug therapy for NDDs.
Collapse
Affiliation(s)
- Misaki Iwahashi
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takeshi Yoshimura
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Wakana Harigai
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kazuhiro Takuma
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan; Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hitoshi Hashimoto
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Division of Bioscience, Institute for Datability Science, Osaka University, 2-8 Yamadaoka, Suita, Osaka, 565-0871, Japan; Department of Molecular Pharmaceutical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Taiichi Katayama
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Atsuko Hayata-Takano
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan; Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
8
|
Li X, Zhao Y, Kong H, Song C, Liu J, Xia J. Identification of region-specific splicing QTLs in human hippocampal tissue and its distinctive role in brain disorders. iScience 2023; 26:107958. [PMID: 37810239 PMCID: PMC10558811 DOI: 10.1016/j.isci.2023.107958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/28/2023] [Accepted: 09/14/2023] [Indexed: 10/10/2023] Open
Abstract
Alternative splicing (AS) regulation has an essential role in complex diseases. However, the AS profiles in the hippocampal (HIPPO) region of human brain are underexplored. Here, we investigated cis-acting sQTLs of HIPPO region in 264 samples and identified thousands of significant sQTLs. By enrichment analysis and functional characterization of these sQTLs, we found that the HIPPO sQTLs were enriched among histone-marked regions, transcription factors binding sites, RNA binding proteins sites, and brain disorders-associated loci. Comparative analyses with the dorsolateral prefrontal cortex revealed the importance of AS regulation in HIPPO (rg = 0.87). Furthermore, we performed a transcriptome-wide association study of Alzheimer's disease and identified 16 significant genes whose genetically regulated splicing levels may have a causal role in Alzheimer. Overall, our study improves our knowledge of the transcriptome gene regulation in the HIPPO region and provides novel insights into elucidating the pathogenesis of potential genes associated with brain disorders.
Collapse
Affiliation(s)
- Xiaoyan Li
- Information Materials and Intelligent Sensing Laboratory of Anhui Province and Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| | - Yiran Zhao
- Information Materials and Intelligent Sensing Laboratory of Anhui Province and Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| | - Hui Kong
- Information Materials and Intelligent Sensing Laboratory of Anhui Province and Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| | - Chengcheng Song
- Information Materials and Intelligent Sensing Laboratory of Anhui Province and Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| | - Jie Liu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province and Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| | - Junfeng Xia
- Information Materials and Intelligent Sensing Laboratory of Anhui Province and Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| |
Collapse
|
9
|
Rusina E, Simonti M, Duprat F, Cestèle S, Mantegazza M. Voltage-gated sodium channels in genetic epilepsy: up and down of excitability. J Neurochem 2023. [PMID: 37654020 DOI: 10.1111/jnc.15947] [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: 07/16/2023] [Revised: 08/11/2023] [Accepted: 08/13/2023] [Indexed: 09/02/2023]
Abstract
The past two decades have witnessed a wide range of studies investigating genetic variants of voltage-gated sodium (NaV ) channels, which are involved in a broad spectrum of diseases, including several types of epilepsy. We have reviewed here phenotypes and pathological mechanisms of genetic epilepsies caused by variants in NaV α and β subunits, as well as of some relevant interacting proteins (FGF12/FHF1, PRRT2, and Ankyrin-G). Notably, variants of all these genes can induce either gain- or loss-of-function of NaV leading to either neuronal hyperexcitability or hypoexcitability. We present the results of functional studies obtained with different experimental models, highlighting that they should be interpreted considering the features of the experimental system used. These systems are models, but they have allowed us to better understand pathophysiological issues, ameliorate diagnostics, orientate genetic counseling, and select/develop therapies within a precision medicine framework. These studies have also allowed us to gain insights into the physiological roles of different NaV channels and of the cells that express them. Overall, our review shows the progress that has been made, but also the need for further studies on aspects that have not yet been clarified. Finally, we conclude by highlighting some significant themes of general interest that can be gleaned from the results of the work of the last two decades.
Collapse
Affiliation(s)
- Evgeniia Rusina
- University Cote d'Azur, Valbonne-Sophia Antipolis, France
- CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France
| | - Martina Simonti
- University Cote d'Azur, Valbonne-Sophia Antipolis, France
- CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France
| | - Fabrice Duprat
- University Cote d'Azur, Valbonne-Sophia Antipolis, France
- CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France
- Inserm, Valbonne-Sophia Antipolis, France
| | - Sandrine Cestèle
- University Cote d'Azur, Valbonne-Sophia Antipolis, France
- CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France
| | - Massimo Mantegazza
- University Cote d'Azur, Valbonne-Sophia Antipolis, France
- CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France
- Inserm, Valbonne-Sophia Antipolis, France
| |
Collapse
|
10
|
Kouchi Z, Kojima M. A Structural Network Analysis of Neuronal ArhGAP21/23 Interactors by Computational Modeling. ACS OMEGA 2023; 8:19249-19264. [PMID: 37305272 PMCID: PMC10249030 DOI: 10.1021/acsomega.2c08054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 05/05/2023] [Indexed: 06/13/2023]
Abstract
RhoGTPase-activating proteins (RhoGAPs) play multiple roles in neuronal development; however, details of their substrate recognition system remain elusive. ArhGAP21 and ArhGAP23 are RhoGAPs that contain N-terminal PDZ and pleckstrin homology domains. In the present study, the RhoGAP domain of these ArhGAPs was computationally modeled by template-based methods and the AlphaFold2 software program, and their intrinsic RhoGTPase recognition mechanism was analyzed from the domain structures using the protein docking programs HADDOCK and HDOCK. ArhGAP21 was predicted to preferentially catalyze Cdc42, RhoA, RhoB, RhoC, and RhoG and to downregulate RhoD and Tc10 activities. Regarding ArhGAP23, RhoA and Cdc42 were deduced to be its substrates, whereas RhoD downregulation was predicted to be less efficient. The PDZ domains of ArhGAP21/23 possess the FTLRXXXVY sequence, and similar globular folding consists of antiparalleled β-sheets and two α-helices that are conserved with PDZ domains of MAST-family proteins. A peptide docking analysis revealed the specific interaction of the ArhGAP23 PDZ domain with the PTEN C-terminus. The pleckstrin homology domain structure of ArhGAP23 was also predicted, and the functional selectivity for the interactors regulated by the folding and disordered domains in ArhGAP21 and ArhGAP23 was examined by an in silico analysis. An interaction analysis of these RhoGAPs revealed the existence of mammalian ArhGAP21/23-specific type I and type III Arf- and RhoGTPase-regulated signaling. Multiple recognition systems of RhoGTPase substrates and selective Arf-dependent localization of ArhGAP21/23 may form the basis of the functional core signaling necessary for synaptic homeostasis and axon/dendritic transport regulated by RhoGAP localization and activities.
Collapse
Affiliation(s)
- Zen Kouchi
- Department
of Genetics, Institute for Developmental
Research, Aichi Developmental Disability Center, 713-8 Kamiya-cho, Kasugai-city 480-0392 Aichi, Japan
| | - Masaki Kojima
- Laboratory
of Bioinformatics, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji 192-0392, Japan
| |
Collapse
|
11
|
Wang J, Jia C, Gao Q, Zhang J, Gu X. iASPP regulates neurite development by interacting with Spectrin proteins. Front Mol Neurosci 2023; 16:1154770. [PMID: 37284462 PMCID: PMC10240065 DOI: 10.3389/fnmol.2023.1154770] [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: 01/31/2023] [Accepted: 04/24/2023] [Indexed: 06/08/2023] Open
Abstract
Introduction Since its discovery in 1999, a substantial body of research has shown that iASPP is highly expressed in various kinds of tumors, interacts with p53, and promotes cancer cell survival by antagonizing the apoptotic activity of p53. However, its role in neurodevelopment is still unknown. Methods We studied the role of iASPP in neuronal differentiation through different neuronal differentiation cellular models, combined with immunohistochemistry, RNA interference and gene overexpression, and studied the molecular mechanism involved in the regulation of neuronal development by iASPP through coimmunoprecipitation coupled with mass spectrometry (CoIP-MS) and coimmunoprecipitation (CoIP). Results In this study, we found that the expression of iASPP gradually decreased during neuronal development. iASPP silencing promotes neuronal differentiation, while its overexpression inhibited neurite differentiation in a variety of neuronal differentiation cellular models. iASPP associated with the cytoskeleton-related protein Sptan1 and dephosphorylated the serine residues in the last spectrin repeat domain of Sptan1 by recruiting PP1. The non-phosphorylated and phosphomimetic mutant form of Sptbn1 inhibited and promoted neuronal cell development respectively. Conclusion Overall, we demonstrate that iASPP suppressed neurite development by inhibiting phosphorylation of Sptbn1.
Collapse
Affiliation(s)
- Junhao Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Chunhong Jia
- Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qiong Gao
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Jiwen Zhang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xi Gu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
12
|
Schwartz S, Wilson SJ, Hale TK, Fitzsimons HL. Ankyrin2 is essential for neuronal morphogenesis and long-term courtship memory in Drosophila. Mol Brain 2023; 16:42. [PMID: 37194019 DOI: 10.1186/s13041-023-01026-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 04/13/2023] [Indexed: 05/18/2023] Open
Abstract
Dysregulation of HDAC4 expression and/or nucleocytoplasmic shuttling results in impaired neuronal morphogenesis and long-term memory in Drosophila melanogaster. A recent genetic screen for genes that interact in the same molecular pathway as HDAC4 identified the cytoskeletal adapter Ankyrin2 (Ank2). Here we sought to investigate the role of Ank2 in neuronal morphogenesis, learning and memory. We found that Ank2 is expressed widely throughout the Drosophila brain where it localizes predominantly to axon tracts. Pan-neuronal knockdown of Ank2 in the mushroom body, a region critical for memory formation, resulted in defects in axon morphogenesis. Similarly, reduction of Ank2 in lobular plate tangential neurons of the optic lobe disrupted dendritic branching and arborization. Conditional knockdown of Ank2 in the mushroom body of adult Drosophila significantly impaired long-term memory (LTM) of courtship suppression, and its expression was essential in the γ neurons of the mushroom body for normal LTM. In summary, we provide the first characterization of the expression pattern of Ank2 in the adult Drosophila brain and demonstrate that Ank2 is critical for morphogenesis of the mushroom body and for the molecular processes required in the adult brain for the formation of long-term memories.
Collapse
Affiliation(s)
- Silvia Schwartz
- School of Natural Sciences, Massey University, Palmerston North, New Zealand
- Current Address: Istituto Italiano di Tecnologia, Center for Life NanoScience, Rome, Italy
| | - Sarah J Wilson
- School of Natural Sciences, Massey University, Palmerston North, New Zealand
| | - Tracy K Hale
- School of Natural Sciences, Massey University, Palmerston North, New Zealand
| | - Helen L Fitzsimons
- School of Natural Sciences, Massey University, Palmerston North, New Zealand.
| |
Collapse
|
13
|
Garrido JJ. Contribution of Axon Initial Segment Structure and Channels to Brain Pathology. Cells 2023; 12:cells12081210. [PMID: 37190119 DOI: 10.3390/cells12081210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 05/17/2023] Open
Abstract
Brain channelopathies are a group of neurological disorders that result from genetic mutations affecting ion channels in the brain. Ion channels are specialized proteins that play a crucial role in the electrical activity of nerve cells by controlling the flow of ions such as sodium, potassium, and calcium. When these channels are not functioning properly, they can cause a wide range of neurological symptoms such as seizures, movement disorders, and cognitive impairment. In this context, the axon initial segment (AIS) is the site of action potential initiation in most neurons. This region is characterized by a high density of voltage-gated sodium channels (VGSCs), which are responsible for the rapid depolarization that occurs when the neuron is stimulated. The AIS is also enriched in other ion channels, such as potassium channels, that play a role in shaping the action potential waveform and determining the firing frequency of the neuron. In addition to ion channels, the AIS contains a complex cytoskeletal structure that helps to anchor the channels in place and regulate their function. Therefore, alterations in this complex structure of ion channels, scaffold proteins, and specialized cytoskeleton may also cause brain channelopathies not necessarily associated with ion channel mutations. This review will focus on how the AISs structure, plasticity, and composition alterations may generate changes in action potentials and neuronal dysfunction leading to brain diseases. AIS function alterations may be the consequence of voltage-gated ion channel mutations, but also may be due to ligand-activated channels and receptors and AIS structural and membrane proteins that support the function of voltage-gated ion channels.
Collapse
Affiliation(s)
- Juan José Garrido
- Instituto Cajal, CSIC, 28002 Madrid, Spain
- Alzheimer's Disease and Other Degenerative Dementias, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28002 Madrid, Spain
| |
Collapse
|
14
|
Piguel NH, Sanders SS, De Simone FI, Martin-de-Saavedra MD, McCoig E, Dionisio LE, Smith KR, Thomas GM, Penzes P. Palmitoylation controls the stability of 190 kDa ankyrin-G in dendritic spines and is regulated by ZDHHC8 and lithium. Front Mol Neurosci 2023; 16:1144066. [PMID: 36969554 PMCID: PMC10031057 DOI: 10.3389/fnmol.2023.1144066] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/14/2023] [Indexed: 03/10/2023] Open
Abstract
Introduction AnkG, encoded by the ANK3 gene, is a multifunctional scaffold protein with complex isoform expression: the 480 and 270 kDa isoforms have roles at the axon initial segment and node of Ranvier, whereas the 190 kDa isoform (AnkG-190) has an emerging role in the dendritic shaft and spine heads. All isoforms of AnkG undergo palmitoylation, a post-translational modification regulating protein attachment to lipid membranes. However, palmitoylation of AnkG-190 has not been investigated in dendritic spines. The ANK3 gene and altered expression of AnkG proteins are associated with a variety of neuropsychiatric and neurodevelopmental disorders including bipolar disorder and are implicated in the lithium response, a commonly used mood stabilizer for bipolar disorder patients, although the precise mechanisms involved are unknown. Result Here, we showed that Cys70 palmitoylation stabilizes the localization of AnkG-190 in spine heads and at dendritic plasma membrane nanodomains. Mutation of Cys70 impairs AnkG-190 function in dendritic spines and alters PSD-95 scaffolding. Interestingly, we find that lithium reduces AnkG-190 palmitoylation thereby increasing its mobility in dendritic spines. Finally, we demonstrate that the palmitoyl acyl transferase ZDHHC8, but not ZDHHC5, increases AnkG-190 stability in spine heads and is inhibited by lithium. Discussion Together, our data reveal that palmitoylation is critical for AnkG-190 localization and function and a potential ZDHHC8/AnkG-190 mechanism linking AnkG-190 mobility to the neuronal effects of lithium.
Collapse
Affiliation(s)
- Nicolas H. Piguel
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Shaun S. Sanders
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Francesca I. De Simone
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Maria D. Martin-de-Saavedra
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Instituto Universitario de Investigación en Neuroquímica, Complutense University of Madrid, Madrid, Spain
| | - Emmarose McCoig
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Leonardo E. Dionisio
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Katharine R. Smith
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Gareth M. Thomas
- Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Peter Penzes
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Northwestern University Center for Autism and Neurodevelopment, Chicago, IL, United States
| |
Collapse
|
15
|
Impaired OTUD7A-dependent Ankyrin regulation mediates neuronal dysfunction in mouse and human models of the 15q13.3 microdeletion syndrome. Mol Psychiatry 2023; 28:1747-1769. [PMID: 36604605 DOI: 10.1038/s41380-022-01937-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023]
Abstract
Copy number variations (CNVs) are associated with psychiatric and neurodevelopmental disorders (NDDs), and most, including the recurrent 15q13.3 microdeletion disorder, have unknown disease mechanisms. We used a heterozygous 15q13.3 microdeletion mouse model and patient iPSC-derived neurons to reveal developmental defects in neuronal maturation and network activity. To identify the underlying molecular dysfunction, we developed a neuron-specific proximity-labeling proteomics (BioID2) pipeline, combined with patient mutations, to target the 15q13.3 CNV genetic driver OTUD7A. OTUD7A is an emerging independent NDD risk gene with no known function in the brain, but has putative deubiquitinase function. The OTUD7A protein-protein interaction network included synaptic, axonal, and cytoskeletal proteins and was enriched for ASD and epilepsy risk genes (Ank3, Ank2, SPTAN1, SPTBN1). The interactions between OTUD7A and Ankyrin-G (Ank3) and Ankyrin-B (Ank2) were disrupted by an epilepsy-associated OTUD7A L233F variant. Further investigation of Ankyrin-G in mouse and human 15q13.3 microdeletion and OTUD7AL233F/L233F models revealed protein instability, increased polyubiquitination, and decreased levels in the axon initial segment, while structured illumination microscopy identified reduced Ankyrin-G nanodomains in dendritic spines. Functional analysis of human 15q13.3 microdeletion and OTUD7AL233F/L233F models revealed shared and distinct impairments to axonal growth and intrinsic excitability. Importantly, restoring OTUD7A or Ankyrin-G expression in 15q13.3 microdeletion neurons led to a reversal of abnormalities. These data reveal a critical OTUD7A-Ankyrin pathway in neuronal development, which is impaired in the 15q13.3 microdeletion syndrome, leading to neuronal dysfunction. Furthermore, our study highlights the utility of targeting CNV genes using cell type-specific proteomics to identify shared and unexplored disease mechanisms across NDDs.
Collapse
|
16
|
Kim Y, Jeon SJ, Gonzales EL, Shin D, Remonde CG, Ahn T, Shin CY. Pirenperone relieves the symptoms of fragile X syndrome in Fmr1 knockout mice. Sci Rep 2022; 12:20966. [PMID: 36470953 PMCID: PMC9723111 DOI: 10.1038/s41598-022-25582-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder that is caused by the loss of Fragile X-linked mental retardation protein (FMRP), an RNA binding protein that can bind and recognize different RNA structures and regulate the target mRNAs' translation involved in neuronal synaptic plasticity. Perturbations of this gene expression network have been related to abnormal behavioral symptoms such as hyperactivity, and impulsivity. Considering the roles of FMRP in the modulation of mRNA translation, we investigated the differentially expressed genes which might be targeted to revert to normal and ameliorate behavioral symptoms. Gene expression data was analyzed and used the connectivity map (CMap) to understand the changes in gene expression in FXS and predict the effective drug candidates. We analyzed the GSE7329 dataset that had 15 control and 8 FXS patients' lymphoblastoid samples. Among 924 genes, 42 genes were selected as signatures for CMap analysis, and 24 associated drugs were found. Pirenperone was selected as a potential drug candidate for FXS for its possible antipsychotic effect. Treatment of pirenperone increased the expression level of Fmr1 gene. Moreover, pirenperone rescued the behavioral deficits in Fmr1 KO mice including hyperactivity, spatial memory, and impulsivity. These results suggest that pirenperone is a new drug candidate for FXS, which should be verified in future studies.
Collapse
Affiliation(s)
- Yujeong Kim
- grid.258676.80000 0004 0532 8339Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul, 05029 Republic of Korea
| | - Se Jin Jeon
- grid.412357.60000 0004 0533 2063Department of Integrative Biotechnology, College of Science and Technology, Sahmyook University, Seoul, 01795 Republic of Korea
| | - Edson Luck Gonzales
- grid.258676.80000 0004 0532 8339Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul, 05029 Republic of Korea
| | - Dongpil Shin
- grid.258676.80000 0004 0532 8339Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul, 05029 Republic of Korea
| | - Chilly Gay Remonde
- grid.258676.80000 0004 0532 8339Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul, 05029 Republic of Korea
| | - TaeJin Ahn
- grid.411957.f0000 0004 0647 2543Department of Life Science, Handong Global University, Nehemiah 36, Handong-ro 558, Pohang, 37554 Republic of Korea
| | - Chan Young Shin
- grid.258676.80000 0004 0532 8339Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul, 05029 Republic of Korea
| |
Collapse
|
17
|
Ochneva A, Zorkina Y, Abramova O, Pavlova O, Ushakova V, Morozova A, Zubkov E, Pavlov K, Gurina O, Chekhonin V. Protein Misfolding and Aggregation in the Brain: Common Pathogenetic Pathways in Neurodegenerative and Mental Disorders. Int J Mol Sci 2022; 23:14498. [PMID: 36430976 PMCID: PMC9695177 DOI: 10.3390/ijms232214498] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/07/2022] [Accepted: 11/19/2022] [Indexed: 11/23/2022] Open
Abstract
Mental disorders represent common brain diseases characterized by substantial impairments of social and cognitive functions. The neurobiological causes and mechanisms of psychopathologies still have not been definitively determined. Various forms of brain proteinopathies, which include a disruption of protein conformations and the formation of protein aggregates in brain tissues, may be a possible cause behind the development of psychiatric disorders. Proteinopathies are known to be the main cause of neurodegeneration, but much less attention is given to the role of protein impairments in psychiatric disorders' pathogenesis, such as depression and schizophrenia. For this reason, the aim of this review was to discuss the potential contribution of protein illnesses in the development of psychopathologies. The first part of the review describes the possible mechanisms of disruption to protein folding and aggregation in the cell: endoplasmic reticulum stress, dysfunction of chaperone proteins, altered mitochondrial function, and impaired autophagy processes. The second part of the review addresses the known proteins whose aggregation in brain tissue has been observed in psychiatric disorders (amyloid, tau protein, α-synuclein, DISC-1, disbindin-1, CRMP1, SNAP25, TRIOBP, NPAS3, GluA1, FABP, and ankyrin-G).
Collapse
Affiliation(s)
- Aleksandra Ochneva
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
| | - Yana Zorkina
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
| | - Olga Abramova
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
| | - Olga Pavlova
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
| | - Valeriya Ushakova
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
- Department of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Anna Morozova
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
| | - Eugene Zubkov
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
| | - Konstantin Pavlov
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
| | - Olga Gurina
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
| | - Vladimir Chekhonin
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- National University of Science and Technology “MISiS”, Leninskiy Avenue 4, 119049 Moscow, Russia
| |
Collapse
|
18
|
Pinson A, Sevrin E, Chatzi C, Le Gac B, Thiry M, Westbrook GL, Parent AS. Induction of Oxidative Stress and Alteration of Synaptic Gene Expression in Newborn Hippocampal Granule Cells after Developmental Exposure to Aroclor 1254. Neuroendocrinology 2022; 113:1248-1261. [PMID: 36257292 PMCID: PMC10110769 DOI: 10.1159/000527576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/10/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Hippocampal newborn neurons integrate into functional circuits where they play an important role in learning and memory. We previously showed that perinatal exposure to Aroclor 1254, a commercial mixture of polychlorinated biphenyls (PCBs) associated with alterations of cognitive function in children, disrupted the normal maturation of excitatory synapses in the dentate gyrus. We hypothesized that hippocampal immature neurons underlie some of the cognitive effects of PCBs. METHODS We used newly generated neurons to examine the effects of PCBs in mice following maternal exposure. Newborn dentate granule cells were tagged with enhanced green fluorescent protein using a transgenic mouse line. The transcriptome of the newly generated granule cells was assessed using RNA sequencing. RESULTS Gestational and lactational exposure to 6 mg/kg/day of Aroclor 1254 disrupted the mRNA expression of 1,308 genes in newborn granule cells. Genes involved in mitochondrial functions were highly enriched with 154 genes significantly increased in exposed compared to control mice. The upregulation of genes involved in oxidative phosphorylation was accompanied by signs of endoplasmic reticulum stress and an increase in lipid peroxidation, a marker of oxidative stress, in the subgranular zone of the dentate gyrus but not in mature granule cells in the granular zone. Aroclor 1254 exposure also disrupted the expression of synaptic genes. Using laser-captured subgranular and granular zones, this effect was restricted to the subgranular zone, where newborn neurons are located. CONCLUSION Our data suggest that gene expression in newborn granule cells is disrupted by Aroclor 1254 and provide clues to the effects of endocrine-disrupting chemicals on the brain.
Collapse
Affiliation(s)
- Anneline Pinson
- Neuroendocrinology Unit, GIGA-Neurosciences, University of Liège, Liège, Belgium
| | - Elena Sevrin
- Neuroendocrinology Unit, GIGA-Neurosciences, University of Liège, Liège, Belgium
| | - Christina Chatzi
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Benjamin Le Gac
- Neuroendocrinology Unit, GIGA-Neurosciences, University of Liège, Liège, Belgium
| | - Marc Thiry
- Cellular and tissular biology, GIGA-Neurosciences, University of Liège, Liège, Belgium
| | - Gary L Westbrook
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Anne-Simone Parent
- Neuroendocrinology Unit, GIGA-Neurosciences, University of Liège, Liège, Belgium
| |
Collapse
|
19
|
Perfilyeva A, Bespalova K, Perfilyeva Y, Skvortsova L, Musralina L, Zhunussova G, Khussainova E, Iskakova U, Bekmanov B, Djansugurova L. Integrative Functional Genomic Analysis in Multiplex Autism Families from Kazakhstan. DISEASE MARKERS 2022; 2022:1509994. [PMID: 36199823 PMCID: PMC9529466 DOI: 10.1155/2022/1509994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/21/2022] [Accepted: 09/06/2022] [Indexed: 12/14/2022]
Abstract
The study of extended pedigrees containing autism spectrum disorder- (ASD-) related broader autism phenotypes (BAP) offers a promising approach to the search for ASD candidate variants. Here, a total of 650,000 genetic markers were tested in four Kazakhstani multiplex families with ASD and BAP to obtain data on de novo mutations (DNMs), common, and rare inherited variants that may contribute to the genetic risk for developing autistic traits. The variants were analyzed in the context of gene networks and pathways. Several previously well-described enriched pathways were identified, including ion channel activity, regulation of synaptic function, and membrane depolarization. Perhaps these pathways are crucial not only for the development of ASD but also for ВАР. The results also point to several additional biological pathways (circadian entrainment, NCAM and BTN family interactions, and interaction between L1 and Ankyrins) and hub genes (CFTR, NOD2, PPP2R2B, and TTR). The obtained results suggest that further exploration of PPI networks combining ASD and BAP risk genes can be used to identify novel or overlooked ASD molecular mechanisms.
Collapse
Affiliation(s)
| | - Kira Bespalova
- Institute of Genetics and Physiology, 93 Al-Farabi Ave., Almaty 050060, Kazakhstan
- Al-Farabi Kazakh National University, 71 Al-Farabi Ave., Almaty 050040, Kazakhstan
| | - Yuliya Perfilyeva
- M.A. Aitkhozhin's Institute of Molecular Biology and Biochemistry, 86 Dosmukhamedov St., Almaty 050012, Kazakhstan
- Branch of the National Center for Biotechnology, 14 Zhahanger St., Almaty 050054, Kazakhstan
| | - Liliya Skvortsova
- Institute of Genetics and Physiology, 93 Al-Farabi Ave., Almaty 050060, Kazakhstan
| | - Lyazzat Musralina
- Institute of Genetics and Physiology, 93 Al-Farabi Ave., Almaty 050060, Kazakhstan
| | - Gulnur Zhunussova
- Institute of Genetics and Physiology, 93 Al-Farabi Ave., Almaty 050060, Kazakhstan
| | - Elmira Khussainova
- Institute of Genetics and Physiology, 93 Al-Farabi Ave., Almaty 050060, Kazakhstan
| | - Ulzhan Iskakova
- Kazakh National Medical University, 94 Tole Bi St., Almaty 050000, Kazakhstan
| | - Bakhytzhan Bekmanov
- Institute of Genetics and Physiology, 93 Al-Farabi Ave., Almaty 050060, Kazakhstan
| | - Leyla Djansugurova
- Institute of Genetics and Physiology, 93 Al-Farabi Ave., Almaty 050060, Kazakhstan
| |
Collapse
|
20
|
Huang Y, Wei L, Huang Y, Wen S, Liu T, Duan X, Wang Y, Zhang H, Fan B, Hu B. Identification of distinct genomic features reveals frequent somatic AHNAK and PTEN mutations predominantly in primary malignant melanoma presenting in the ureter. Jpn J Clin Oncol 2022; 52:930-943. [PMID: 35578896 DOI: 10.1093/jjco/hyac061] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 04/07/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Primary malignant melanoma of the ureter is extremely rare. Genetic variants to the increased risk of developing the disease have not yet been investigated. METHODS Tumour mutation profiling for primary malignant melanoma of the ureter was performed by whole-exome sequencing. Immunohistochemistry was performed to verify histopathological features and the variants of predisposing genes and driver mutation genes. Furthermore, we conducted a literature review and Surveillance, Epidemiology and End Result-based study by searching public databases. RESULTS We identified 38 somatic single nucleotide variants and 9 somatic insertions and deletions (INDELs) in tumour specimens. After filtering with the Cancer Gene Census database, seven predisposing genes and two driver mutation genes were identified. Moreover, the immunohistochemical profile showed that tumour cells were positive for Melan-A, melanoma gp100 human melanoma black 45 (HMB45), S100 beta and P53. The expression levels of two driver mutation genes (phosphatase and tensin homolog (PTEN) and desmoyokin (AHNAK) and five predisposing genes (AT-rich interaction domain 1B (ARID1B), catalase, eukaryotic translation initiation factor 4 gamma 3 (EIF4G3), ANK3 and collagen type I) were significantly downregulated in tumour tissues compared to paracancerous tissues. In the literature review and Surveillance, Epidemiology and End Results-based study, patients with primary malignant melanoma of the urinary tract had worse clinical outcomes than patients with primary urothelial carcinoma after 1:2 propensity score matching (P = 0.010). Additionally, Cox multivariate analysis for patients with primary malignant melanoma of the urinary tract indicated that distant metastasis (hazard ratio = 1.185; P = 0.044) was an independent predictor for overall survival, and tumour focality (hazard ratio = 0.602; P = 0.017) and non-surgery (hazard ratio = 0.434; P = 0.003) were independent factors for tumour progression. CONCLUSIONS Our study is the first to provide evidence that the distinct phenotypes of primary malignant melanoma of the ureter may be due to different genetic variations. The prognosis of primary malignant melanoma of the urinary tract was poorer than that of primary urothelial carcinoma of the urinary tract.
Collapse
Affiliation(s)
- Yan Huang
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Lai Wei
- Department of Radiology, Sichuan Province Orthopedic Hospital, Chengdu, Sichuan, China
| | - Yuanbin Huang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Shuang Wen
- Department of Pathology, Dalian Friendship Hospital, Dalian, Liaoning, China
| | - Tianqing Liu
- Department of Pathology, Dalian Friendship Hospital, Dalian, Liaoning, China
| | - Xu Duan
- First Clinical College, Dalian Medical University, Dalian, Liaoning, China
| | - Yutong Wang
- First Clinical College, Dalian Medical University, Dalian, Liaoning, China
| | - Hongshuo Zhang
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, Dalian, Liaoning, China
| | - Bo Fan
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bin Hu
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| |
Collapse
|
21
|
Eichel K, Shen K. The function of the axon initial segment in neuronal polarity. Dev Biol 2022; 489:47-54. [DOI: 10.1016/j.ydbio.2022.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 05/09/2022] [Accepted: 05/23/2022] [Indexed: 11/25/2022]
|
22
|
Dashtban S, Haj-Nasrolah-Fard F, Kosari Z, Ghamari R, Forouzesh F, Alizadeh F. ANK3 and ZNF804A intronic variants increase risk of schizophrenia in Iranian population: An association study. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
23
|
Abstract
BACKGROUND To date, besides genome-wide association studies, a variety of other genetic analyses (e.g. polygenic risk scores, whole-exome sequencing and whole-genome sequencing) have been conducted, and a large amount of data has been gathered for investigating the involvement of common, rare and very rare types of DNA sequence variants in bipolar disorder. Also, non-invasive neuroimaging methods can be used to quantify changes in brain structure and function in patients with bipolar disorder. AIMS To provide a comprehensive assessment of genetic findings associated with bipolar disorder, based on the evaluation of different genomic approaches and neuroimaging studies. METHOD We conducted a PubMed search of all relevant literatures from the beginning to the present, by querying related search strings. RESULTS ANK3, CACNA1C, SYNE1, ODZ4 and TRANK1 are five genes that have been replicated as key gene candidates in bipolar disorder pathophysiology, through the investigated studies. The percentage of phenotypic variance explained by the identified variants is small (approximately 4.7%). Bipolar disorder polygenic risk scores are associated with other psychiatric phenotypes. The ENIGMA-BD studies show a replicable pattern of lower cortical thickness, altered white matter integrity and smaller subcortical volumes in bipolar disorder. CONCLUSIONS The low amount of explained phenotypic variance highlights the need for further large-scale investigations, especially among non-European populations, to achieve a more complete understanding of the genetic architecture of bipolar disorder and the missing heritability. Combining neuroimaging data with genetic data in large-scale studies might help researchers acquire a better knowledge of the engaged brain regions in bipolar disorder.
Collapse
Affiliation(s)
- Mojtaba Oraki Kohshour
- Institute of Psychiatric Phenomics and Genomics, University Hospital LMU Munich, Germany; and Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Iran
| | - Sergi Papiol
- Institute of Psychiatric Phenomics and Genomics, University Hospital LMU Munich, Germany; and Department of Psychiatry and Psychotherapy, University Hospital LMU Munich, Germany
| | - Christopher R K Ching
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, USA
| | - Thomas G Schulze
- Institute of Psychiatric Phenomics and Genomics, University Hospital LMU Munich, Germany; and Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, USA
| |
Collapse
|
24
|
Moon S, Zhao YT. Recursive splicing is a rare event in the mouse brain. PLoS One 2022; 17:e0263082. [PMID: 35089962 PMCID: PMC8797253 DOI: 10.1371/journal.pone.0263082] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/11/2022] [Indexed: 11/18/2022] Open
Abstract
Recursive splicing (RS) is a splicing mechanism to remove long introns from messenger RNA precursors of long genes. Compared to the hundreds of RS events identified in humans and drosophila, only ten RS events have been reported in mice. To further investigate RS in mice, we analyzed RS in the mouse brain, a tissue that is enriched in the expression of long genes. We found that nuclear total RNA sequencing is an efficient approach to investigate RS events. We analyzed 1.15 billion uniquely mapped reads from the nuclear total RNA sequencing data in the mouse cerebral cortex. Unexpectedly, we only identified 20 RS sites, suggesting that RS is a rare event in the mouse brain. We also identified that RS is constitutive between excitatory and inhibitory neurons and between sexes in the mouse cerebral cortex. In addition, we found that the primary sequence context is associated with RS splicing intermediates and distinguishes RS AGGT site from non-RS AGGT sites, indicating the importance of the primary sequence context in RS sites. Moreover, we discovered that cryptic exons may use an RS-like mechanism for splicing. Overall, we provide novel findings about RS in long genes in the mouse brain.
Collapse
Affiliation(s)
- Sohyun Moon
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, United States of America
| | - Ying-Tao Zhao
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, United States of America
- * E-mail:
| |
Collapse
|
25
|
Usui N, Tian X, Harigai W, Togawa S, Utsunomiya R, Doi T, Miyoshi K, Shinoda K, Tanaka J, Shimada S, Katayama T, Yoshimura T. Length impairments of the axon initial segment in rodent models of attention-deficit hyperactivity disorder and autism spectrum disorder. Neurochem Int 2021; 153:105273. [PMID: 34971749 DOI: 10.1016/j.neuint.2021.105273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 11/25/2022]
Abstract
The axon initial segment (AIS) is a structural neuronal compartment of the proximal axon that plays key roles in sodium channel clustering, action potential initiation, and signal propagation of neuronal outputs. Mutations in constitutive genes of the AIS, such as ANK3, have been identified in patients with neurodevelopmental disorders. Nevertheless, morphological changes in the AIS in neurodevelopmental disorders have not been characterized. In this study, we investigated the length of the AIS in rodent models of attention-deficit hyperactivity disorder (ADHD) and autism spectrum disorder (ASD). We observed abnormalities in AIS length in both animal models. In ADHD model rodents, we observed shorter AIS length in layer 2/3 (L2/3) neurons of the medial prefrontal cortex (mPFC) and primary somatosensory barrel field (S1BF). Further, we observed shorter AIS length in S1BF L5 neurons. In ASD model mice, we observed shorter AIS length in L2/3 and L5 neurons of the S1BF. These results suggest that impairments in AIS length are common phenomena in neurodevelopmental disorders such as ADHD and ASD and may be conserved across species. Our findings provide novel insight into the potential contribution of the AIS to the pathophysiology and pathogenesis of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Noriyoshi Usui
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Suita, 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Hirakata, 573-0022, Japan.
| | - Xiaoye Tian
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan
| | - Wakana Harigai
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan
| | - Shogo Togawa
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, 755-8505, Japan
| | - Ryo Utsunomiya
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Ehime, 791-0295, Japan
| | - Tomomi Doi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Ehime, 791-0295, Japan
| | - Ko Miyoshi
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan
| | - Koh Shinoda
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, 755-8505, Japan
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Ehime, 791-0295, Japan
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Hirakata, 573-0022, Japan
| | - Taiichi Katayama
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan
| | - Takeshi Yoshimura
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan.
| |
Collapse
|
26
|
Wong JC, Butler KM, Shapiro L, Thelin JT, Mattison KA, Garber KB, Goldenberg PC, Kubendran S, Schaefer GB, Escayg A. Pathogenic in-Frame Variants in SCN8A: Expanding the Genetic Landscape of SCN8A-Associated Disease. Front Pharmacol 2021; 12:748415. [PMID: 34867351 PMCID: PMC8635767 DOI: 10.3389/fphar.2021.748415] [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] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/21/2021] [Indexed: 01/11/2023] Open
Abstract
Numerous SCN8A mutations have been identified, of which, the majority are de novo missense variants. Most mutations result in epileptic encephalopathy; however, some are associated with less severe phenotypes. Mouse models generated by knock-in of human missense SCN8A mutations exhibit seizures and a range of behavioral abnormalities. To date, there are only a few Scn8a mouse models with in-frame deletions or insertions, and notably, none of these mouse lines exhibit increased seizure susceptibility. In the current study, we report the generation and characterization of two Scn8a mouse models (ΔIRL/+ and ΔVIR/+) carrying overlapping in-frame deletions within the voltage sensor of domain 4 (DIVS4). Both mouse lines show increased seizure susceptibility and infrequent spontaneous seizures. We also describe two unrelated patients with the same in-frame SCN8A deletion in the DIV S5-S6 pore region, highlighting the clinical relevance of this class of mutations.
Collapse
Affiliation(s)
- Jennifer C Wong
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | - Kameryn M Butler
- Department of Human Genetics, Emory University, Atlanta, GA, United States.,Greenwood Genetic Center, Greenwood, SC, United States
| | - Lindsey Shapiro
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | - Jacquelyn T Thelin
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | - Kari A Mattison
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | - Kathryn B Garber
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | - Paula C Goldenberg
- Department of Pediatrics and Medical Genetics, Harvard Medical School, Boston, MA, United States
| | - Shobana Kubendran
- Department of Pediatrics, Kansas University School of Medicine-Wichita, Wichita, KS, United States
| | - G Bradley Schaefer
- University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Andrew Escayg
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| |
Collapse
|
27
|
Mitani T, Isikay S, Gezdirici A, Gulec EY, Punetha J, Fatih JM, Herman I, Akay G, Du H, Calame DG, Ayaz A, Tos T, Yesil G, Aydin H, Geckinli B, Elcioglu N, Candan S, Sezer O, Erdem HB, Gul D, Demiral E, Elmas M, Yesilbas O, Kilic B, Gungor S, Ceylan AC, Bozdogan S, Ozalp O, Cicek S, Aslan H, Yalcintepe S, Topcu V, Bayram Y, Grochowski CM, Jolly A, Dawood M, Duan R, Jhangiani SN, Doddapaneni H, Hu J, Muzny DM, Marafi D, Akdemir ZC, Karaca E, Carvalho CMB, Gibbs RA, Posey JE, Lupski JR, Pehlivan D. High prevalence of multilocus pathogenic variation in neurodevelopmental disorders in the Turkish population. Am J Hum Genet 2021; 108:1981-2005. [PMID: 34582790 PMCID: PMC8546040 DOI: 10.1016/j.ajhg.2021.08.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/20/2021] [Indexed: 02/06/2023] Open
Abstract
Neurodevelopmental disorders (NDDs) are clinically and genetically heterogenous; many such disorders are secondary to perturbation in brain development and/or function. The prevalence of NDDs is > 3%, resulting in significant sociocultural and economic challenges to society. With recent advances in family-based genomics, rare-variant analyses, and further exploration of the Clan Genomics hypothesis, there has been a logarithmic explosion in neurogenetic "disease-associated genes" molecular etiology and biology of NDDs; however, the majority of NDDs remain molecularly undiagnosed. We applied genome-wide screening technologies, including exome sequencing (ES) and whole-genome sequencing (WGS), to identify the molecular etiology of 234 newly enrolled subjects and 20 previously unsolved Turkish NDD families. In 176 of the 234 studied families (75.2%), a plausible and genetically parsimonious molecular etiology was identified. Out of 176 solved families, deleterious variants were identified in 218 distinct genes, further documenting the enormous genetic heterogeneity and diverse perturbations in human biology underlying NDDs. We propose 86 candidate disease-trait-associated genes for an NDD phenotype. Importantly, on the basis of objective and internally established variant prioritization criteria, we identified 51 families (51/176 = 28.9%) with multilocus pathogenic variation (MPV), mostly driven by runs of homozygosity (ROHs) - reflecting genomic segments/haplotypes that are identical-by-descent. Furthermore, with the use of additional bioinformatic tools and expansion of ES to additional family members, we established a molecular diagnosis in 5 out of 20 families (25%) who remained undiagnosed in our previously studied NDD cohort emanating from Turkey.
Collapse
Affiliation(s)
- Tadahiro Mitani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sedat Isikay
- Department of Pediatric Neurology, Faculty of Medicine, University of Gaziantep, Gaziantep 27310, Turkey
| | - Alper Gezdirici
- Department of Medical Genetics, Basaksehir Cam and Sakura City Hospital, Istanbul 34480, Turkey
| | - Elif Yilmaz Gulec
- Department of Medical Genetics, Kanuni Sultan Suleyman Training and Research Hospital, 34303 Istanbul, Turkey
| | - Jaya Punetha
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jawid M Fatih
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Isabella Herman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Gulsen Akay
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Haowei Du
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Daniel G Calame
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Akif Ayaz
- Department of Medical Genetics, Adana City Training and Research Hospital, Adana 01170, Turkey; Departments of Medical Genetics, School of Medicine, Istanbul Medipol University, Istanbul 34810, Turkey
| | - Tulay Tos
- University of Health Sciences Zubeyde Hanim Research and Training Hospital of Women's Health and Diseases, Department of Medical Genetics, Ankara 06080, Turkey
| | - Gozde Yesil
- Istanbul Faculty of Medicine, Department of Medical Genetics, Istanbul University, Istanbul 34093, Turkey
| | - Hatip Aydin
- Centre of Genetics Diagnosis, Zeynep Kamil Maternity and Children's Training and Research Hospital, Istanbul, Turkey; Private Reyap Istanbul Hospital, Istanbul 34515, Turkey
| | - Bilgen Geckinli
- Centre of Genetics Diagnosis, Zeynep Kamil Maternity and Children's Training and Research Hospital, Istanbul, Turkey; Department of Medical Genetics, School of Medicine, Marmara University, Istanbul 34722, Turkey
| | - Nursel Elcioglu
- Department of Pediatric Genetics, School of Medicine, Marmara University, Istanbul 34722, Turkey; Eastern Mediterranean University Medical School, Magosa, Mersin 10, Turkey
| | - Sukru Candan
- Medical Genetics Section, Balikesir Ataturk Public Hospital, Balikesir 10100, Turkey
| | - Ozlem Sezer
- Department of Medical Genetics, Samsun Education and Research Hospital, Samsun 55100, Turkey
| | - Haktan Bagis Erdem
- Department of Medical Genetics, University of Health Sciences, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara 06110, Turkey
| | - Davut Gul
- Department of Medical Genetics, Gulhane Military Medical School, Ankara 06010, Turkey
| | - Emine Demiral
- Department of Medical Genetics, School of Medicine, University of Inonu, Malatya 44280, Turkey
| | - Muhsin Elmas
- Department of Medical Genetics, Afyon Kocatepe University, School of Medicine, Afyon 03218, Turkey
| | - Osman Yesilbas
- Division of Critical Care Medicine, Department of Pediatrics, School of Medicine, Bezmialem Foundation University, Istanbul 34093, Turkey; Department of Pediatrics, Division of Pediatric Critical Care Medicine, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Betul Kilic
- Department of Pediatrics and Pediatric Neurology, Faculty of Medicine, Inonu University, Malatya 34218, Turkey
| | - Serdal Gungor
- Department of Pediatrics and Pediatric Neurology, Faculty of Medicine, Inonu University, Malatya 34218, Turkey
| | - Ahmet C Ceylan
- Department of Medical Genetics, University of Health Sciences, Ankara Training and Research Hospital, Ankara 06110, Turkey
| | - Sevcan Bozdogan
- Department of Medical Genetics, Cukurova University Faculty of Medicine, Adana 01330, Turkey
| | - Ozge Ozalp
- Department of Medical Genetics, Adana City Training and Research Hospital, Adana 01170, Turkey
| | - Salih Cicek
- Department of Medical Genetics, Konya Training and Research Hospital, Konya 42250, Turkey
| | - Huseyin Aslan
- Department of Medical Genetics, Adana City Training and Research Hospital, Adana 01170, Turkey
| | - Sinem Yalcintepe
- Department of Medical Genetics, School of Medicine, Trakya University, Edirne 22130, Turkey
| | - Vehap Topcu
- Department of Medical Genetics, Ankara City Hospital, Ankara 06800, Turkey
| | - Yavuz Bayram
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Angad Jolly
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Moez Dawood
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ruizhi Duan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shalini N Jhangiani
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Harsha Doddapaneni
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jianhong Hu
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Donna M Muzny
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dana Marafi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zeynep Coban Akdemir
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ender Karaca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Claudia M B Carvalho
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Richard A Gibbs
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jennifer E Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Hospital, Houston, TX 77030, USA.
| | - Davut Pehlivan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA.
| |
Collapse
|
28
|
Avazzadeh S, Quinlan LR, Reilly J, McDonagh K, Jalali A, Wang Y, McInerney V, Krawczyk J, Ding Y, Fitzgerald J, O'Sullivan M, Forman EB, Lynch SA, Ennis S, Feerick N, Reilly R, Li W, Shen X, Yang G, Lu Y, Peeters H, Dockery P, O'Brien T, Shen S, Gallagher L. NRXN1α +/- is associated with increased excitability in ASD iPSC-derived neurons. BMC Neurosci 2021; 22:56. [PMID: 34525970 PMCID: PMC8442436 DOI: 10.1186/s12868-021-00661-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 09/01/2021] [Indexed: 12/14/2022] Open
Abstract
Background NRXN1 deletions are identified as one of major rare risk factors for autism spectrum disorder (ASD) and other neurodevelopmental disorders. ASD has 30% co-morbidity with epilepsy, and the latter is associated with excessive neuronal firing. NRXN1 encodes hundreds of presynaptic neuro-adhesion proteins categorized as NRXN1α/β/γ. Previous studies on cultured cells show that the short NRXN1β primarily exerts excitation effect, whereas the long NRXN1α which is more commonly deleted in patients involves in both excitation and inhibition. However, patient-derived models are essential for understanding functional consequences of NRXN1α deletions in human neurons. We recently derived induced pluripotent stem cells (iPSCs) from five controls and three ASD patients carrying NRXN1α+/- and showed increased calcium transients in patient neurons. Methods In this study we investigated the electrophysiological properties of iPSC-derived cortical neurons in control and ASD patients carrying NRXN1α+/- using patch clamping. Whole genome RNA sequencing was carried out to further understand the potential underlying molecular mechanism. Results NRXN1α+/- cortical neurons were shown to display larger sodium currents, higher AP amplitude and accelerated depolarization time. RNASeq analyses revealed transcriptomic changes with significant upregulation glutamatergic synapse and ion channels/transporter activity including voltage-gated potassium channels (GRIN1, GRIN3B, SLC17A6, CACNG3, CACNA1A, SHANK1), which are likely to couple with the increased excitability in NRXN1α+/- cortical neurons. Conclusions Together with recent evidence of increased calcium transients, our results showed that human NRXN1α+/- isoform deletions altered neuronal excitability and non-synaptic function, and NRXN1α+/- patient iPSCs may be used as an ASD model for therapeutic development with calcium transients and excitability as readouts. Supplementary Information The online version contains supplementary material available at 10.1186/s12868-021-00661-0.
Collapse
Affiliation(s)
- Sahar Avazzadeh
- School of Medicine, Regenerative Medicine Institute, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Ireland
| | - Leo R Quinlan
- Physiology and Cellular Physiology Research Laboratory, School of Medicine, CÚRAM SFI Centre for Research in Medical Devices, National University of Ireland (NUI), Galway, Ireland
| | - Jamie Reilly
- School of Medicine, Regenerative Medicine Institute, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Ireland
| | - Katya McDonagh
- School of Medicine, Regenerative Medicine Institute, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Ireland
| | | | - Yanqin Wang
- School of Medicine, Regenerative Medicine Institute, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Ireland.,Department of Physiology, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Veronica McInerney
- HRB Clinical Research Facility, National University of Ireland (NUI), Galway, Ireland
| | - Janusz Krawczyk
- Department of Haematology, Galway University Hospital, Galway, Ireland
| | - Yicheng Ding
- School of Medicine, Regenerative Medicine Institute, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Ireland
| | | | - Matthew O'Sullivan
- Trinity Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Eva B Forman
- Children's University Hospital, Temple Street, Dublin, Ireland
| | - Sally A Lynch
- Children's University Hospital, Temple Street, Dublin, Ireland.,Department of Clinical Genetics, OLCHC, Dublin 12, Ireland
| | - Sean Ennis
- School of Medicine and Medical Science, UCD Academic Centre On Rare Diseases, University College Dublin, Dublin, Ireland
| | - Niamh Feerick
- Centre for Bioengineering, Trinity College Institute of Neuroscience, School of Medicine, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Richard Reilly
- Centre for Bioengineering, Trinity College Institute of Neuroscience, School of Medicine, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Weidong Li
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Xu Shen
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China
| | - Guangming Yang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yin Lu
- College of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Hilde Peeters
- Centre for Human Genetics, University Hospital Leuven, KU Leuven, 3000, Leuven, Belgium
| | - Peter Dockery
- Centre for Microscopy and Imaging, Anatomy, School of Medicine, National University of Ireland (NUI), Galway, Ireland
| | - Timothy O'Brien
- School of Medicine, Regenerative Medicine Institute, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Ireland
| | - Sanbing Shen
- School of Medicine, Regenerative Medicine Institute, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Ireland. .,FutureNeuro Research Centre, Royal College of Surgeons in Ireland, Dublin, D02, Ireland.
| | - Louise Gallagher
- Trinity Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
29
|
Tang L, Liu J, Zhu Y, Duan J, Chen Y, Wei Y, Gong X, Wang F, Tang Y. ANK3 Gene Polymorphism Rs10994336 Influences Executive Functions by Modulating Methylation in Patients With Bipolar Disorder. Front Neurosci 2021; 15:682873. [PMID: 34421516 PMCID: PMC8371237 DOI: 10.3389/fnins.2021.682873] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/31/2021] [Indexed: 01/11/2023] Open
Abstract
Background: A large body of evidence suggests that epigenetic modification including DNA methylation plays a critical role in BD's pathogenesis while the identification of methylation quantitative trait loci (meQTLs) shed light on the interpretation of the function of genetic variants in non-coding regions. The intronic single nucleotide polymorphism (SNP) rs10994336 within the ANK3 has emerged as one of the most replicated risk variants for bipolar disorder (BD) in genome-wide association studies. Whether rs10994336 functions as a meQTL to mediate the association between genotype and phenotype remains unclear. Method: A total of 154 patients with BD and 181 healthy controls (HC) were recruited. The genotypes of rs10994336 and methylation levels of CpG sites within ANK3 were tested. Executive functions were assessed using a computerized version of the Wisconsin Card Sorting Test (WCST). Results: Bipolar disorder patients with the risk-T allele of rs10994336 scored lower on tests of executive function compared to homozygous CC carriers, after controlling for age, gender, and education level. No significant difference was found in HC individuals. The risk-T allele is associated with a lower methylation level of CpG site cg02172182 in HC after multiple corrections and replicated in the BD group in the same direction. Further mediation analysis revealed that the cg02172182 methylation significantly mediated the association between the polymorphism rs10994336 and PE index of WCST in patients with BD. Conclusion: Our study suggests that BD-related genetic variant rs10994336 in ANK3 impacts executive functions by modulating ANK3 methylation, supporting the theory that methylation acts as a mediator between genotype and phenotype.
Collapse
Affiliation(s)
- Lili Tang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Juan Liu
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yue Zhu
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jia Duan
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yifan Chen
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yange Wei
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaohong Gong
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, China
| | - Fei Wang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China.,Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Yanqing Tang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
30
|
Fujitani M, Otani Y, Miyajima H. Pathophysiological Roles of Abnormal Axon Initial Segments in Neurodevelopmental Disorders. Cells 2021; 10:2110. [PMID: 34440880 PMCID: PMC8392614 DOI: 10.3390/cells10082110] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/13/2021] [Accepted: 08/15/2021] [Indexed: 11/17/2022] Open
Abstract
The 20-60 μm axon initial segment (AIS) is proximally located at the interface between the axon and cell body. AIS has characteristic molecular and structural properties regulated by the crucial protein, ankyrin-G. The AIS contains a high density of Na+ channels relative to the cell body, which allows low thresholds for the initiation of action potential (AP). Molecular and physiological studies have shown that the AIS is also a key domain for the control of neuronal excitability by homeostatic mechanisms. The AIS has high plasticity in normal developmental processes and pathological activities, such as injury, neurodegeneration, and neurodevelopmental disorders (NDDs). In the first half of this review, we provide an overview of the molecular, structural, and ion-channel characteristics of AIS, AIS regulation through axo-axonic synapses, and axo-glial interactions. In the second half, to understand the relationship between NDDs and AIS, we discuss the activity-dependent plasticity of AIS, the human mutation of AIS regulatory genes, and the pathophysiological role of an abnormal AIS in NDD model animals and patients. We propose that the AIS may provide a potentially valuable structural biomarker in response to abnormal network activity in vivo as well as a new treatment concept at the neural circuit level.
Collapse
Affiliation(s)
- Masashi Fujitani
- Department of Anatomy and Neuroscience, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-shi 693-8501, Shimane, Japan; (Y.O.); (H.M.)
| | | | | |
Collapse
|
31
|
ANK3 related neurodevelopmental disorders: expanding the spectrum of heterozygous loss-of-function variants. Neurogenetics 2021; 22:263-269. [PMID: 34218362 PMCID: PMC8426245 DOI: 10.1007/s10048-021-00655-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/20/2021] [Indexed: 11/03/2022]
Abstract
ANK3 encodes multiple isoforms of ankyrin-G, resulting in variegated tissue expression and function, especially regarding its role in neuronal development. Based on the zygosity, location, and type, ANK3 variants result in different neurodevelopmental phenotypes. Autism spectrum disorder has been associated with heterozygous missense variants in ANK3, whereas a more severe neurodevelopmental phenotype is caused by isoform-dependent, autosomal-dominant, or autosomal-recessive loss-of-function variants. Here, we present four individuals affected by a variable neurodevelopmental phenotype harboring a heterozygous frameshift or nonsense variant affecting all ANK3 transcripts. Thus, we provide further evidence of an isoform-based phenotypic continuum underlying ANK3-associated pathologies and expand its phenotypic spectrum.
Collapse
|
32
|
Yoon S, Parnell E, Penzes P. TGF-β-Induced Phosphorylation of Usp9X Stabilizes Ankyrin-G and Regulates Dendritic Spine Development and Maintenance. Cell Rep 2021; 31:107685. [PMID: 32460012 PMCID: PMC7324065 DOI: 10.1016/j.celrep.2020.107685] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 04/02/2020] [Accepted: 05/04/2020] [Indexed: 12/30/2022] Open
Abstract
Signaling by the cytokine transforming growth factor β (TGF-β) has been implicated in a multitude of biological functions; however, TGF-β signaling, particularly in the CNS, remains largely unexplored. ANK3 variants (encoding ankyrin-G) are associated with bipolar disorder, intellectual disability, and autism spectrum disorder, while mutations in USP9X, which encodes a deubiquitinase, are associated with X-linked intellectual disability and autism in humans. Here, we show that TGF-β signaling promotes Usp9X phosphorylation, which enhances its interaction with ankyrin-G and stabilizes ankyrin-G in spines, leading to spine enlargement. Using in situ proximity ligation combined with structured illumination superresolution microscopy, we characterize the postsynaptic spatial organization of phosphorylation-dependent regulation of Usp9X/ankyrin-G interactions in dendrites and its quantitative relationship with spine morphology and number. These data reveal a cytokine-mediated mechanism regulating protein stability in spines and suggest a role for deubiquitination and TGF-β signaling in neurodevelopmental disorder pathogenesis and treatment. Yoon et al. show that phosphorylation of a deubiquitinating enzyme by a cytokine enhances the stabilization of synaptic scaffolding protein during dendritic spine development, and its alterations result in deficient synaptic structural maintenance, with relevance for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Sehyoun Yoon
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Euan Parnell
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Northwestern University, Center for Autism and Neurodevelopment, Chicago, IL 60611, USA.
| |
Collapse
|
33
|
Juvale IIA, Che Has AT. The Potential Role of miRNAs as Predictive Biomarkers in Neurodevelopmental Disorders. J Mol Neurosci 2021; 71:1338-1355. [PMID: 33774758 DOI: 10.1007/s12031-021-01825-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/02/2021] [Indexed: 12/22/2022]
Abstract
Neurodevelopmental disorders are defined as a set of abnormal brain developmental conditions marked by the early childhood onset of cognitive, behavioral, and functional deficits leading to memory and learning problems, emotional instability, and impulsivity. Autism spectrum disorder, attention-deficit/hyperactivity disorder, Tourette syndrome, fragile X syndrome, and Down's syndrome are a few known examples of neurodevelopmental disorders. Although they are relatively common in both developed and developing countries, very little is currently known about their underlying molecular mechanisms. Both genetic and environmental factors are known to increase the risk of neurodevelopmental disorders. Current diagnostic and screening tests for neurodevelopmental disorders are not reliable; hence, individuals with neurodevelopmental disorders are often diagnosed in the later stages. This negatively affects their prognosis and quality of life, prompting the need for a better diagnostic biomarker. Recent studies on microRNAs and their altered regulation in diseases have shed some light on the possible role they could play in the development of the central nervous system. This review attempts to elucidate our current understanding of the role that microRNAs play in neurodevelopmental disorders with the hope of utilizing them as potential biomarkers in the future.
Collapse
Affiliation(s)
- Iman Imtiyaz Ahmed Juvale
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Ahmad Tarmizi Che Has
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
34
|
Karimian SS, Akbari MT, Sadr SS, Javadi G. Association of Candidate Single Nucleotide Polymorphisms Related to Candidate Genes in Patients With Schizophrenia. Basic Clin Neurosci 2021; 11:595-608. [PMID: 33643553 PMCID: PMC7878058 DOI: 10.32598/bcn.9.10.470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 06/10/2018] [Accepted: 04/06/2019] [Indexed: 11/20/2022] Open
Abstract
Introduction: Schizophrenia is a chronic heterogenic neurodevelopment disorder. Many genes interfere in the development of SCZ. All four genes, NrCAM, PRODH, ANK3, and ANKK1, which were evaluated in this study, were previously reported to be associated with Schizophrenia. The NrCAM contributes to creating cognitive deficiencies through the CAM’s signaling pathway. PRODH plays a vital role in creating SCZ negative symptoms through the signaling pathway of glutamatergic and NMDA receptors. ANK3 affects ion channel and molecular adhesion in Ranvier and initial segments of axons, leading to mental retardation, sleep disorder, and SCZ. ANKK1 encodes a protein kinase and was reported to be associated with alcohol addiction, Attention Deficit Hyperactivity Disorder (ADHD), and SCZ. Methods: The subjects were selected from Schizophrenic patients referring to the Psychiatric Ward of Imam-Hussein Hospital and Schizophrenic Patients Support Institution (AHEBBA). 95 (30 Schizoaffective patients, 57 Paranoid patients, and 8 disorganized) patients were recruited as the subjects in the present case-control association study. 120 healthy subjects were recruited from the Tehran Medical Genetics Laboratory staff and a group of students from the Islamic Azad University of Science and Research in Tehran. The genotypes were determined with molecular genotyping techniques of PCR-RFLP, ARMS-PCR, and Cycle sequencing. Results were analyzed by the Chi-Square test using SPSS V. 24 and R, SNP STATE Package to investigate significant differences between cases and controls. Results: The incidence of schizophrenia was 68% and 32% among men and women, respectively. The evaluation of the allelic association between schizophrenia and all the candidate SNPs showed a significant association between NrCAM’s SNP rs10235968 and SCZ (P=0.001). Haplotype T, T, C in rs10235968, rs6967368, rs3763463, respectively, within the NrCAM gene, showed significant association with schizophrenia disorder (P=0.0001). Conclusion: No association was found between other candidate SNPs and SCZ among the subjects.
Collapse
Affiliation(s)
- Seyedeh Sara Karimian
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Taghi Akbari
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modarres University, Tehran, Iran.,Tehran Medical Genetics Laboratory, Tehran, Iran
| | - Seyed Saeed Sadr
- Department of Psychiatry, Imam Hussein Hospital, Shahid Beheshti University, Tehran, Iran
| | - Gholamreza Javadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
35
|
Toma C, Shaw AD, Heath A, Pierce KD, Mitchell PB, Schofield PR, Fullerton JM. A linkage and exome study of multiplex families with bipolar disorder implicates rare coding variants of ANK3 and additional rare alleles at 10q11-q21. J Psychiatry Neurosci 2021; 46:E247-E257. [PMID: 33729739 PMCID: PMC8061732 DOI: 10.1503/jpn.200083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Bipolar disorder is a highly heritable psychiatric condition for which specific genetic factors remain largely unknown. In the present study, we used combined whole-exome sequencing and linkage analysis to identify risk loci and dissect the contribution of common and rare variants in families with a high density of illness. METHODS Overall, 117 participants from 15 Australian extended families with bipolar disorder (72 with affective disorder, including 50 with bipolar disorder type I or II, 13 with schizoaffective disorder-manic type and 9 with recurrent unipolar disorder) underwent whole-exome sequencing. We performed genome-wide linkage analysis using MERLIN and conditional linkage analysis using LAMP. We assessed the contribution of potentially functional rare variants using a genebased segregation test. RESULTS We identified a significant linkage peak on chromosome 10q11-q21 (maximal single nucleotide polymorphism = rs10761725; exponential logarithm of the odds [LODexp] = 3.03; empirical p = 0.046). The linkage interval spanned 36 protein-coding genes, including a gene associated with bipolar disorder, ankyrin 3 (ANK3). Conditional linkage analysis showed that common ANK3 risk variants previously identified in genome-wide association studies - or variants in linkage disequilibrium with those variants - did not explain the linkage signal (rs10994397 LOD = 0.63; rs9804190 LOD = 0.04). A family-based segregation test with 34 rare variants from 14 genes under the linkage interval suggested rare variant contributions of 3 brain-expressed genes: NRBF2 (p = 0.005), PCDH15 (p = 0.002) and ANK3 (p = 0.014). LIMITATIONS We did not examine non-coding variants, but they may explain the remaining linkage signal. CONCLUSION Combining family-based linkage analysis with next-generation sequencing data is effective for identifying putative disease genes and specific risk variants in complex disorders. We identified rare missense variants in ANK3, PCDH15 and NRBF2 that could confer disease risk, providing valuable targets for functional characterization.
Collapse
Affiliation(s)
- Claudio Toma
- From Neuroscience Research Australia, Sydney, Australia (Toma, Shaw, Heath, Pierce, Schofield); the School of Medical Sciences, University of New South Wales, Sydney, Australia (Toma, Shaw, Schofield, Fullerton); the Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid/CSIC, Madrid, Spain (Toma); the School of Psychiatry, University of New South Wales, Sydney, Australia (Mitchell); and the Black Dog Institute, Prince of Wales Hospital, Sydney, Australia (Mitchell)
| | - Alex D Shaw
- From Neuroscience Research Australia, Sydney, Australia (Toma, Shaw, Heath, Pierce, Schofield); the School of Medical Sciences, University of New South Wales, Sydney, Australia (Toma, Shaw, Schofield, Fullerton); the Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid/CSIC, Madrid, Spain (Toma); the School of Psychiatry, University of New South Wales, Sydney, Australia (Mitchell); and the Black Dog Institute, Prince of Wales Hospital, Sydney, Australia (Mitchell)
| | - Anna Heath
- From Neuroscience Research Australia, Sydney, Australia (Toma, Shaw, Heath, Pierce, Schofield); the School of Medical Sciences, University of New South Wales, Sydney, Australia (Toma, Shaw, Schofield, Fullerton); the Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid/CSIC, Madrid, Spain (Toma); the School of Psychiatry, University of New South Wales, Sydney, Australia (Mitchell); and the Black Dog Institute, Prince of Wales Hospital, Sydney, Australia (Mitchell)
| | - Kerrie D Pierce
- From Neuroscience Research Australia, Sydney, Australia (Toma, Shaw, Heath, Pierce, Schofield); the School of Medical Sciences, University of New South Wales, Sydney, Australia (Toma, Shaw, Schofield, Fullerton); the Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid/CSIC, Madrid, Spain (Toma); the School of Psychiatry, University of New South Wales, Sydney, Australia (Mitchell); and the Black Dog Institute, Prince of Wales Hospital, Sydney, Australia (Mitchell)
| | - Philip B Mitchell
- From Neuroscience Research Australia, Sydney, Australia (Toma, Shaw, Heath, Pierce, Schofield); the School of Medical Sciences, University of New South Wales, Sydney, Australia (Toma, Shaw, Schofield, Fullerton); the Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid/CSIC, Madrid, Spain (Toma); the School of Psychiatry, University of New South Wales, Sydney, Australia (Mitchell); and the Black Dog Institute, Prince of Wales Hospital, Sydney, Australia (Mitchell)
| | - Peter R Schofield
- From Neuroscience Research Australia, Sydney, Australia (Toma, Shaw, Heath, Pierce, Schofield); the School of Medical Sciences, University of New South Wales, Sydney, Australia (Toma, Shaw, Schofield, Fullerton); the Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid/CSIC, Madrid, Spain (Toma); the School of Psychiatry, University of New South Wales, Sydney, Australia (Mitchell); and the Black Dog Institute, Prince of Wales Hospital, Sydney, Australia (Mitchell)
| | - Janice M Fullerton
- From Neuroscience Research Australia, Sydney, Australia (Toma, Shaw, Heath, Pierce, Schofield); the School of Medical Sciences, University of New South Wales, Sydney, Australia (Toma, Shaw, Schofield, Fullerton); the Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid/CSIC, Madrid, Spain (Toma); the School of Psychiatry, University of New South Wales, Sydney, Australia (Mitchell); and the Black Dog Institute, Prince of Wales Hospital, Sydney, Australia (Mitchell)
| |
Collapse
|
36
|
Basar MA, Beck DB, Werner A. Deubiquitylases in developmental ubiquitin signaling and congenital diseases. Cell Death Differ 2021; 28:538-556. [PMID: 33335288 PMCID: PMC7862630 DOI: 10.1038/s41418-020-00697-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023] Open
Abstract
Metazoan development from a one-cell zygote to a fully formed organism requires complex cellular differentiation and communication pathways. To coordinate these processes, embryos frequently encode signaling information with the small protein modifier ubiquitin, which is typically attached to lysine residues within substrates. During ubiquitin signaling, a three-step enzymatic cascade modifies specific substrates with topologically unique ubiquitin modifications, which mediate changes in the substrate's stability, activity, localization, or interacting proteins. Ubiquitin signaling is critically regulated by deubiquitylases (DUBs), a class of ~100 human enzymes that oppose the conjugation of ubiquitin. DUBs control many essential cellular functions and various aspects of human physiology and development. Recent genetic studies have identified mutations in several DUBs that cause developmental disorders. Here we review principles controlling DUB activity and substrate recruitment that allow these enzymes to regulate ubiquitin signaling during development. We summarize key mechanisms of how DUBs control embryonic and postnatal differentiation processes, highlight developmental disorders that are caused by mutations in particular DUB members, and describe our current understanding of how these mutations disrupt development. Finally, we discuss how emerging tools from human disease genetics will enable the identification and study of novel congenital disease-causing DUBs.
Collapse
Affiliation(s)
- Mohammed A Basar
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - David B Beck
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
- Metabolic, Cardiovascular and Inflammatory Disease Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Achim Werner
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
37
|
Rathje M, Waxman H, Benoit M, Tammineni P, Leu C, Loebrich S, Nedivi E. Genetic variants in the bipolar disorder risk locus SYNE1 that affect CPG2 expression and protein function. Mol Psychiatry 2021; 26:508-523. [PMID: 30610203 PMCID: PMC6609516 DOI: 10.1038/s41380-018-0314-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 11/14/2018] [Accepted: 11/15/2018] [Indexed: 12/28/2022]
Abstract
Bipolar disorder (BD) is a common mood disorder characterized by recurrent episodes of mania and depression. Both genetic and environmental factors have been implicated in BD etiology, but the biological underpinnings remain elusive. Recently, genome-wide association studies (GWAS) of neuropsychiatric disorders have identified a risk locus for BD containing the SYNE1 gene, a large gene encoding multiple proteins. The BD association signal spans, almost exclusively, the part of SYNE1 encoding CPG2, a brain-specific protein localized to excitatory postsynaptic sites, where it regulates glutamate receptor internalization. Here we show that CPG2 protein levels are significantly decreased in postmortem brain tissue from BD patients, as compared to control subjects, as well as schizophrenia and depression patients. We identify genetic variants within the postmortem brains that map to the CPG2 promoter region, and show that they negatively affect gene expression. We also identify missense single nucleotide polymorphisms (SNPs) in CPG2 coding regions that affect CPG2 expression, localization, and synaptic function. Our findings link genetic variation in the CPG2 region of SYNE1 with a mechanism for glutamatergic synapse dysfunction that could underlie susceptibility to BD in some individuals. Few GWAS hits in human genetics for neuropsychiatric disorders to date have afforded such mechanistic clues. Further, the potential for genetic distinction of susceptibility to BD from other neuropsychiatric disorders with overlapping clinical traits holds promise for improved diagnostics and treatment of this devastating illness.
Collapse
Affiliation(s)
- Mette Rathje
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hannah Waxman
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marc Benoit
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Prasad Tammineni
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Costin Leu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA,Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA, USA,Institute of Neurology, University College London, London, United Kingdom
| | - Sven Loebrich
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Elly Nedivi
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
38
|
Stevens SR, Rasband MN. Ankyrins and neurological disease. Curr Opin Neurobiol 2021; 69:51-57. [PMID: 33485190 DOI: 10.1016/j.conb.2021.01.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/11/2022]
Abstract
Ankyrins are scaffolding proteins widely expressed throughout the nervous system. Ankyrins recruit diverse membrane proteins, including ion channels and cell adhesion molecules, into specialized subcellular membrane domains. These domains are stabilized by ankyrins interacting with the spectrin cytoskeleton. Ankyrin genes are highly associated with a number of neurological disorders, including Alzheimer's disease, schizophrenia, autism spectrum disorders, and bipolar disorder. Here, we discuss ankyrin function and their role in neurological disease. We propose mutations in ankyrins contribute to disease through two primary mechanisms: 1) altered neuronal excitability by disrupting ion channel clustering at key excitable domains, and 2) altered neuronal connectivity via impaired stabilization of membrane proteins.
Collapse
Affiliation(s)
- Sharon R Stevens
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
39
|
Structural Insights into Ankyrin Repeat-Containing Proteins and Their Influence in Ubiquitylation. Int J Mol Sci 2021; 22:ijms22020609. [PMID: 33435370 PMCID: PMC7826745 DOI: 10.3390/ijms22020609] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
Ankyrin repeat (AR) domains are considered the most abundant repeat motif found in eukaryotic proteins. AR domains are predominantly known to mediate specific protein-protein interactions (PPIs) without necessarily recognizing specific primary sequences, nor requiring strict conformity within its own primary sequence. This promiscuity allows for one AR domain to recognize and bind to a variety of intracellular substrates, suggesting that AR-containing proteins may be involved in a wide array of functions. Many AR-containing proteins serve a critical role in biological processes including the ubiquitylation signaling pathway (USP). There is also strong evidence that AR-containing protein malfunction are associated with several neurological diseases and disorders. In this review, the structure and mechanism of key AR-containing proteins are discussed to suggest and/or identify how each protein utilizes their AR domains to support ubiquitylation and the cascading pathways that follow upon substrate modification.
Collapse
|
40
|
Yoon S, Piguel NH, Khalatyan N, Dionisio LE, Savas JN, Penzes P. Homer1 promotes dendritic spine growth through ankyrin-G and its loss reshapes the synaptic proteome. Mol Psychiatry 2021; 26:1775-1789. [PMID: 33398084 PMCID: PMC8254828 DOI: 10.1038/s41380-020-00991-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 11/24/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022]
Abstract
Homer1 is a synaptic scaffold protein that regulates glutamatergic synapses and spine morphogenesis. HOMER1 knockout (KO) mice show behavioral abnormalities related to psychiatric disorders, and HOMER1 has been associated with psychiatric disorders such as addiction, autism disorder (ASD), schizophrenia (SZ), and depression. However, the mechanisms by which it promotes spine stability and its global function in maintaining the synaptic proteome has not yet been fully investigated. Here, we used computational approaches to identify global functions for proteins containing the Homer1-interacting PPXXF motif within the postsynaptic compartment. Ankyrin-G was one of the most topologically important nodes in the postsynaptic peripheral membrane subnetwork, and we show that one of the PPXXF motifs, present in the postsynaptically-enriched 190 kDa isoform of ankyrin-G (ankyrin-G 190), is recognized by the EVH1 domain of Homer1. We use proximity ligation combined with super-resolution microscopy to map the interaction of ankyrin-G and Homer1 to distinct nanodomains within the spine head and correlate them with spine head size. This interaction motif is critical for ankyrin-G 190's ability to increase spine head size, and for the maintenance of a stable ankyrin-G pool in spines. Intriguingly, lack of Homer1 significantly upregulated the abundance of ankyrin-G, but downregulated Shank3 in cortical crude plasma membrane fractions. In addition, proteomic analysis of the cortex in HOMER1 KO and wild-type (WT) mice revealed a global reshaping of the postsynaptic proteome, surprisingly characterized by extensive upregulation of synaptic proteins. Taken together, we show that Homer1 and its protein interaction motif have broad global functions within synaptic protein-protein interaction networks. Enrichment of disease risk factors within these networks has important implications for neurodevelopmental disorders including bipolar disorder, ASD, and SZ.
Collapse
Affiliation(s)
- Sehyoun Yoon
- grid.16753.360000 0001 2299 3507Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Nicolas H. Piguel
- grid.16753.360000 0001 2299 3507Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Natalia Khalatyan
- grid.16753.360000 0001 2299 3507Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Leonardo E. Dionisio
- grid.16753.360000 0001 2299 3507Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA ,grid.19006.3e0000 0000 9632 6718Present Address: Neuroscience Interdepartmental Program, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Jeffrey N. Savas
- grid.16753.360000 0001 2299 3507Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA. .,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA. .,Northwestern University, Center for Autism and Neurodevelopment, Chicago, IL, 60611, USA.
| |
Collapse
|
41
|
Solé L, Tamkun MM. Trafficking mechanisms underlying Na v channel subcellular localization in neurons. Channels (Austin) 2020; 14:1-17. [PMID: 31841065 PMCID: PMC7039628 DOI: 10.1080/19336950.2019.1700082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 11/13/2019] [Indexed: 01/06/2023] Open
Abstract
Voltage gated sodium channels (Nav) play a crucial role in action potential initiation and propagation. Although the discovery of Nav channels dates back more than 65 years, and great advances in understanding their localization, biophysical properties, and links to disease have been made, there are still many questions to be answered regarding the cellular and molecular mechanisms involved in Nav channel trafficking, localization and regulation. This review summarizes the different trafficking mechanisms underlying the polarized Nav channel localization in neurons, with an emphasis on the axon initial segment (AIS), as well as discussing the latest advances regarding how neurons regulate their excitability by modifying AIS length and location. The importance of Nav channel localization is emphasized by the relationship between mutations, impaired trafficking and disease. While this review focuses on Nav1.6, other Nav isoforms are also discussed.
Collapse
Affiliation(s)
- Laura Solé
- Molecular, Cellular and Integrative Neurosciences Graduate Program, Colorado State University, Fort Collins, CO, USA
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Michael M. Tamkun
- Molecular, Cellular and Integrative Neurosciences Graduate Program, Colorado State University, Fort Collins, CO, USA
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
42
|
Dirkx N, Miceli F, Taglialatela M, Weckhuysen S. The Role of Kv7.2 in Neurodevelopment: Insights and Gaps in Our Understanding. Front Physiol 2020; 11:570588. [PMID: 33192566 PMCID: PMC7657400 DOI: 10.3389/fphys.2020.570588] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/07/2020] [Indexed: 11/13/2022] Open
Abstract
Kv7.2 subunits encoded by the KCNQ2 gene constitute a critical molecular component of the M-current, a subthreshold voltage-gated potassium current controlling neuronal excitability by dampening repetitive action potential firing. Pathogenic loss-of-function variants in KCNQ2 have been linked to epilepsy since 1998, and there is ample functional evidence showing that dysfunction of the channel indeed results in neuronal hyperexcitability. The recent description of individuals with severe developmental delay with or without seizures due to pathogenic variants in KCNQ2 (KCNQ2-encephalopathy) reveals that Kv7.2 channels also have an important role in neurodevelopment. Kv7.2 channels are expressed already very early in the developing brain when key developmental processes such as proliferation, differentiation, and synaptogenesis play a crucial role in brain morphogenesis and maturation. In this review, we will discuss the available evidence for a role of Kv7.2 channels in these neurodevelopmental processes, focusing in particular on insights derived from KCNQ2-related human phenotypes, from the spatio-temporal expression of Kv7.2 and other Kv7 family member, and from cellular and rodent models, highlighting critical gaps and research strategies to be implemented in the future. Lastly, we propose a model which divides the M-current activity in three different developmental stages, correlating with the cell characteristics during these particular periods in neuronal development, and how this can be linked with KCNQ2-related disorders. Understanding these mechanisms can create opportunities for new targeted therapies for KCNQ2-encephalopathy.
Collapse
Affiliation(s)
- Nina Dirkx
- Applied and Translational Neurogenomics Group, VIB Center for Molecular Neurology, Vlaams Instituut voor Biotechnologie, Antwerp, Belgium
| | - Francesco Miceli
- Section of Pharmacology, Department of Neuroscience, University of Naples Federico II, Naples, Italy
| | - Maurizio Taglialatela
- Section of Pharmacology, Department of Neuroscience, University of Naples Federico II, Naples, Italy
| | - Sarah Weckhuysen
- Applied and Translational Neurogenomics Group, VIB Center for Molecular Neurology, Vlaams Instituut voor Biotechnologie, Antwerp, Belgium.,Department of Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Department of Neurology, Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
43
|
Mahmood T, El-Asrag ME, Poulter JA, Cardno AG, Tomlinson A, Ahmed S, Al-Amri A, Nazari J, Neill J, Chamali RS, Kiwan N, Ghuloum S, Alhaj HA, Randerson Moor J, Khan S, Al-Amin H, Johnson CA, Woodruff P, Wilkinson ID, Ali M, Clapcote SJ, Inglehearn CF. A Recessively Inherited Risk Locus on Chromosome 13q22-31 Conferring Susceptibility to Schizophrenia. Schizophr Bull 2020; 47:796-802. [PMID: 33159203 PMCID: PMC8084434 DOI: 10.1093/schbul/sbaa161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We report a consanguineous family in which schizophrenia segregates in a manner consistent with recessive inheritance of a rare, partial-penetrance susceptibility allele. From 4 marriages between 2 sets of siblings who are half first cousins, 6 offspring have diagnoses of psychotic disorder. Homozygosity mapping revealed a 6.1-Mb homozygous region on chromosome 13q22.2-31.1 shared by all affected individuals, containing 13 protein-coding genes. Microsatellite analysis confirmed homozygosity for the affected haplotype in 12 further apparently unaffected members of the family. Psychiatric reports suggested an endophenotype of milder psychiatric illness in 4 of these individuals. Exome and genome sequencing revealed no potentially pathogenic coding or structural variants within the risk haplotype. Filtering for noncoding variants with a minor allele frequency of <0.05 identified 17 variants predicted to have significant effects, the 2 most significant being within or adjacent to the SCEL gene. RNA sequencing of blood from an affected homozygote showed the upregulation of transcription from NDFIP2 and SCEL. NDFIP2 is highly expressed in brain, unlike SCEL, and is involved in determining T helper (Th) cell type 1 and Th2 phenotypes, which have previously been implicated with schizophrenia.
Collapse
Affiliation(s)
- Tariq Mahmood
- Becklin Centre, Leeds and York Partnership NHS Foundation Trust, Leeds, UK
| | - Mohammed E El-Asrag
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
- Department of Zoology, Faculty of Science, Benha University, Benha, Egypt
- Division of Cardiovascular Sciences, School of Medicine, University of Manchester, Manchester, UK
| | - James A Poulter
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | | | - Anneka Tomlinson
- Becklin Centre, Leeds and York Partnership NHS Foundation Trust, Leeds, UK
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Sophia Ahmed
- NIHR-Sheffield Biomedical Research Centre, University of Sheffield, Sheffield, UK
| | - Ahmed Al-Amri
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
- National Genetic Centre, Royal Hospital, Muscat, Oman
| | - Jamshid Nazari
- Becklin Centre, Leeds and York Partnership NHS Foundation Trust, Leeds, UK
| | - Joanna Neill
- Division of Pharmacy and Optometry, University of Manchester, Manchester, UK
| | - Rifka S Chamali
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Nancy Kiwan
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Suhaila Ghuloum
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
- Psychiatry Department, Hamad Medical Corporation, Doha, Qatar
| | - Hamid A Alhaj
- Sheffield Health and Social Care NHS Foundation Trust, Sheffield, UK
| | | | - Shabana Khan
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Hassen Al-Amin
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Colin A Johnson
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Peter Woodruff
- NIHR-Sheffield Biomedical Research Centre, University of Sheffield, Sheffield, UK
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
- Psychiatry Department, Hamad Medical Corporation, Doha, Qatar
- Sheffield Health and Social Care NHS Foundation Trust, Sheffield, UK
| | - Iain D Wilkinson
- NIHR-Sheffield Biomedical Research Centre, University of Sheffield, Sheffield, UK
| | - Manir Ali
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | | | - Chris F Inglehearn
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
- To whom correspondence should be addressed; Beckett Street, Leeds, LS9 7TF, UK; tel: 44-(0)113-343-8646, e-mail:
| |
Collapse
|
44
|
Nelson AD, Caballero-Florán RN, Rodríguez Díaz JC, Hull JM, Yuan Y, Li J, Chen K, Walder KK, Lopez-Santiago LF, Bennett V, McInnis MG, Isom LL, Wang C, Zhang M, Jones KS, Jenkins PM. Ankyrin-G regulates forebrain connectivity and network synchronization via interaction with GABARAP. Mol Psychiatry 2020; 25:2800-2817. [PMID: 30504823 PMCID: PMC6542726 DOI: 10.1038/s41380-018-0308-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/17/2018] [Accepted: 11/19/2018] [Indexed: 12/16/2022]
Abstract
GABAergic circuits are critical for the synchronization and higher order function of brain networks. Defects in this circuitry are linked to neuropsychiatric diseases, including bipolar disorder, schizophrenia, and autism. Work in cultured neurons has shown that ankyrin-G plays a key role in the regulation of GABAergic synapses on the axon initial segment and somatodendritic domain of pyramidal neurons, where it interacts directly with the GABAA receptor-associated protein (GABARAP) to stabilize cell surface GABAA receptors. Here, we generated a knock-in mouse model expressing a mutation that abolishes the ankyrin-G/GABARAP interaction (Ank3 W1989R) to understand how ankyrin-G and GABARAP regulate GABAergic circuitry in vivo. We found that Ank3 W1989R mice exhibit a striking reduction in forebrain GABAergic synapses resulting in pyramidal cell hyperexcitability and disruptions in network synchronization. In addition, we identified changes in pyramidal cell dendritic spines and axon initial segments consistent with compensation for hyperexcitability. Finally, we identified the ANK3 W1989R variant in a family with bipolar disorder, suggesting a potential role of this variant in disease. Our results highlight the importance of ankyrin-G in regulating forebrain circuitry and provide novel insights into how ANK3 loss-of-function variants may contribute to human disease.
Collapse
Affiliation(s)
- A D Nelson
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - R N Caballero-Florán
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - J C Rodríguez Díaz
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - J M Hull
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Y Yuan
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - J Li
- Division of Life Sciences, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - K Chen
- Division of Life Sciences, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - K K Walder
- Department of Cell Biology, Duke University Medical Center, Durham, NC, 27710, USA
| | - L F Lopez-Santiago
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - V Bennett
- Department of Cell Biology, Duke University Medical Center, Durham, NC, 27710, USA
- Departments of Biochemistry, Neurobiology, and Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC, 27710, USA
| | - M G McInnis
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - L L Isom
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - C Wang
- Division of Life Sciences, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - M Zhang
- Division of Life Sciences, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - K S Jones
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - P M Jenkins
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
45
|
Overrepresentation of genetic variation in the AnkyrinG interactome is related to a range of neurodevelopmental disorders. Eur J Hum Genet 2020; 28:1726-1733. [PMID: 32651551 DOI: 10.1038/s41431-020-0682-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 05/28/2020] [Accepted: 06/15/2020] [Indexed: 12/30/2022] Open
Abstract
Upon the discovery of numerous genes involved in the pathogenesis of neurodevelopmental disorders, several studies showed that a significant proportion of these genes converge on common pathways and protein networks. Here, we used a reversed approach, by screening the AnkyrinG protein-protein interaction network for genetic variation in a large cohort of 1009 cases with neurodevelopmental disorders. We identified a significant enrichment of de novo potentially disease-causing variants in this network, confirming that this protein network plays an important role in the emergence of several neurodevelopmental disorders.
Collapse
|
46
|
Li J, Chen K, Zhu R, Zhang M. Structural Basis Underlying Strong Interactions between Ankyrins and Spectrins. J Mol Biol 2020; 432:3838-3850. [DOI: 10.1016/j.jmb.2020.04.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/18/2020] [Accepted: 04/23/2020] [Indexed: 01/06/2023]
|
47
|
Vitsios D, Petrovski S. Mantis-ml: Disease-Agnostic Gene Prioritization from High-Throughput Genomic Screens by Stochastic Semi-supervised Learning. Am J Hum Genet 2020; 106:659-678. [PMID: 32386536 PMCID: PMC7212270 DOI: 10.1016/j.ajhg.2020.03.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/26/2020] [Indexed: 12/22/2022] Open
Abstract
Access to large-scale genomics datasets has increased the utility of hypothesis-free genome-wide analyses. However, gene signals are often insufficiently powered to reach experiment-wide significance, triggering a process of laborious triaging of genomic-association-study results. We introduce mantis-ml, a multi-dimensional, multi-step machine-learning framework that allows objective assessment of the biological relevance of genes to disease studies. Mantis-ml is an automated machine-learning framework that follows a multi-model approach of stochastic semi-supervised learning to rank disease-associated genes through iterative learning sessions on random balanced datasets across the protein-coding exome. When applied to a range of human diseases, including chronic kidney disease (CKD), epilepsy, and amyotrophic lateral sclerosis (ALS), mantis-ml achieved an average area under curve (AUC) prediction performance of 0.81-0.89. Critically, to prove its value as a tool that can be used to interpret exome-wide association studies, we overlapped mantis-ml predictions with data from published cohort-level association studies. We found a statistically significant enrichment of high mantis-ml predictions among the highest-ranked genes from hypothesis-free cohort-level statistics, indicating a substantial improvement over the performance of current state-of-the-art methods and pointing to the capture of true prioritization signals for disease-associated genes. Finally, we introduce a generic mantis-ml score (GMS) trained with over 1,200 features as a generic-disease-likelihood estimator, outperforming published gene-level scores. In addition to our tool, we provide a gene prioritization atlas that includes mantis-ml's predictions across ten disease areas and empowers researchers to interactively navigate through the gene-triaging framework. Mantis-ml is an intuitive tool that supports the objective triaging of large-scale genomic discovery studies and enhances our understanding of complex genotype-phenotype associations.
Collapse
Affiliation(s)
- Dimitrios Vitsios
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, 1 Francis Crick Avenue, CB2 0RE Cambridge, UK.
| | - Slavé Petrovski
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, 1 Francis Crick Avenue, CB2 0RE Cambridge, UK.
| |
Collapse
|
48
|
Mechanisms and Alterations of Cardiac Ion Channels Leading to Disease: Role of Ankyrin-B in Cardiac Function. Biomolecules 2020; 10:biom10020211. [PMID: 32023981 PMCID: PMC7072516 DOI: 10.3390/biom10020211] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 01/24/2020] [Accepted: 01/28/2020] [Indexed: 01/08/2023] Open
Abstract
Ankyrin-B (encoded by ANK2), originally identified as a key cytoskeletal-associated protein in the brain, is highly expressed in the heart and plays critical roles in cardiac physiology and cell biology. In the heart, ankyrin-B plays key roles in the targeting and localization of key ion channels and transporters, structural proteins, and signaling molecules. The role of ankyrin-B in normal cardiac function is illustrated in animal models lacking ankyrin-B expression, which display significant electrical and structural phenotypes and life-threatening arrhythmias. Further, ankyrin-B dysfunction has been associated with cardiac phenotypes in humans (now referred to as “ankyrin-B syndrome”) including sinus node dysfunction, heart rate variability, atrial fibrillation, conduction block, arrhythmogenic cardiomyopathy, structural remodeling, and sudden cardiac death. Here, we review the diverse roles of ankyrin-B in the vertebrate heart with a significant focus on ankyrin-B-linked cell- and molecular-pathways and disease.
Collapse
|
49
|
Hamdan H, Lim BC, Torii T, Joshi A, Konning M, Smith C, Palmer DJ, Ng P, Leterrier C, Oses-Prieto JA, Burlingame AL, Rasband MN. Mapping axon initial segment structure and function by multiplexed proximity biotinylation. Nat Commun 2020; 11:100. [PMID: 31900387 PMCID: PMC6941957 DOI: 10.1038/s41467-019-13658-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 11/15/2019] [Indexed: 12/19/2022] Open
Abstract
Axon initial segments (AISs) generate action potentials and regulate the polarized distribution of proteins, lipids, and organelles in neurons. While the mechanisms of AIS Na+ and K+ channel clustering are understood, the molecular mechanisms that stabilize the AIS and control neuronal polarity remain obscure. Here, we use proximity biotinylation and mass spectrometry to identify the AIS proteome. We target the biotin-ligase BirA* to the AIS by generating fusion proteins of BirA* with NF186, Ndel1, and Trim46; these chimeras map the molecular organization of AIS intracellular membrane, cytosolic, and microtubule compartments. Our experiments reveal a diverse set of biotinylated proteins not previously reported at the AIS. We show many are located at the AIS, interact with known AIS proteins, and their loss disrupts AIS structure and function. Our results provide conceptual insights and a resource for AIS molecular organization, the mechanisms of AIS stability, and polarized trafficking in neurons.
Collapse
Affiliation(s)
- Hamdan Hamdan
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.,Department of Physiology, College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia
| | - Brian C Lim
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Tomohiro Torii
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Abhijeet Joshi
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Matthias Konning
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Cameron Smith
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Donna J Palmer
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Philip Ng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | | | - Juan A Oses-Prieto
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
50
|
Chagula DB, Rechciński T, Rudnicka K, Chmiela M. Ankyrins in human health and disease - an update of recent experimental findings. Arch Med Sci 2020; 16:715-726. [PMID: 32542072 PMCID: PMC7286341 DOI: 10.5114/aoms.2019.89836] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/25/2018] [Indexed: 12/17/2022] Open
Abstract
Ankyrins are adaptor molecules that in eukaryotic cells form complexes with ion channel proteins, cell adhesion and signalling molecules and components of the cytoskeleton. They play a pivotal role as scaffolding proteins, in the structural anchoring to the muscle membrane, in muscle development, neurogenesis and synapse formation. Dysfunction of ankyrins is implicated in numerous diseases such as hereditary spherocytosis, neurodegeneration of Purkinje cells, cardiac arrhythmia, Brugada syndrome, bipolar disorders and schizophrenia, congenital myopathies and congenital heart disease as well as cancers. Detecting either down- or over-expression of ankyrins and ergo their use as biomarkers can provide a new paradigm in the diagnosis of these diseases. This paper provides an outline of knowledge about the structure of ankyrins, and by making use of recent experimental research studies critically discusses their role in several health disorders. Moreover, therapeutic options utilizing engineered ankyrins, designed ankyrin repeat proteins (DARPins), are discussed.
Collapse
Affiliation(s)
- Damian B. Chagula
- Laboratory of Gastroimmunology, Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Tomasz Rechciński
- Department of Cardiology, Bieganski Regional Speciality Hospital, Medical University of Lodz, Lodz, Poland
| | - Karolina Rudnicka
- Laboratory of Gastroimmunology, Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Magdalena Chmiela
- Laboratory of Gastroimmunology, Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
- Corresponding author: Prof. Magdalena Chmiela Laboratory of Gastroimmunology, Department of Immmunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St, 90-237 Lodz, Poland, E-mail:
| |
Collapse
|