1
|
Hisaoka T, Komori T, Fujimoto K, Kitamura T, Morikawa Y. Comprehensive expression pattern of kin of irregular chiasm-like 3 in the adult mouse brain. Biochem Biophys Res Commun 2021; 563:66-72. [PMID: 34062388 DOI: 10.1016/j.bbrc.2021.05.063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 05/18/2021] [Indexed: 11/16/2022]
Abstract
Kin of irregular chiasm-like 3 (Kirrel3), a member of the immunoglobulin superfamily, is expressed in the central nervous system during development and in adulthood. It has been reported that Kirrel3 is involved in the axonal fasciculation in the olfactory bulb, the neuronal migration in the pontine nucleus, and the synapse formation in the hippocampal neurons in mice. Although KIRREL3 mutations have been implicated in autism spectrum disorder and intellectual disability in humans, the comprehensive expression pattern of Kirrel3 in the adult brain is not fully understood. To better visualize Kirrel3 expression pattern and to gain insight into the role of Kirrel3 in the brain, we investigated the expression of Kirrel3 in the adult brain of Kirrel3-heterozygous (Kirrel3+/-) mice, in which Kirrel3-expressing cells could be identified by the expression of β-galactosidase (β-gal) in the nucleus of cells. The strong expression of β-gal was observed in the hippocampus, cerebral cortex, olfactory bulb, amygdala, thalamus, and cerebellum. In the hippocampus, β-gal was detected in the dentate gyrus and in the ventral parts of CA1 and CA3, which are known to be involved in the social recognition memory. Within the cerebral cortex, many cells with β-gal expression were observed in the olfactory area and auditory area. In the striatum, neurons with β-gal expression were mainly observed in the ventral striatum. Expression of β-gal was observed in all layers in the cerebellum and olfactory bulb, except for the olfactory nerve layer. Double-immunofluorescence staining of β-galactosidase with neuronal markers revealed that β-gal expression was exclusively detected in neurons. These results suggest that Kirrel3 may be involved in the maintenance of neuronal networks, such as the maintenance of synaptic connectivity and plasticity in the motor, sensory, and cognitive circuits of adult brain.
Collapse
Affiliation(s)
- Tomoko Hisaoka
- Department of Anatomy and Neurobiology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Tadasuke Komori
- Department of Anatomy and Neurobiology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Kohta Fujimoto
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Toshio Kitamura
- Division of Cellular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Yoshihiro Morikawa
- Department of Anatomy and Neurobiology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan.
| |
Collapse
|
2
|
Völker LA, Maar BA, Pulido Guevara BA, Bilkei-Gorzo A, Zimmer A, Brönneke H, Dafinger C, Bertsch S, Wagener JR, Schweizer H, Schermer B, Benzing T, Hoehne M. Neph2/Kirrel3 regulates sensory input, motor coordination, and home-cage activity in rodents. GENES BRAIN AND BEHAVIOR 2018; 17:e12516. [DOI: 10.1111/gbb.12516] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 07/22/2018] [Accepted: 08/17/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Linus A. Völker
- Department II of Internal Medicine and Center for Molecular Medicine Cologne; University of Cologne; Cologne Germany
| | - Barbara A. Maar
- Department II of Internal Medicine and Center for Molecular Medicine Cologne; University of Cologne; Cologne Germany
| | - Barbara A. Pulido Guevara
- Department II of Internal Medicine and Center for Molecular Medicine Cologne; University of Cologne; Cologne Germany
| | - Andras Bilkei-Gorzo
- Institute of Molecular Psychiatry; Medical Faculty of the University of Bonn; Bonn Germany
| | - Andreas Zimmer
- Institute of Molecular Psychiatry; Medical Faculty of the University of Bonn; Bonn Germany
| | - Hella Brönneke
- Mouse Phenotyping Core Facility; Cologne Excellence Cluster on Cellular Stress Responses (CECAD); 50931 Cologne Germany
| | - Claudia Dafinger
- Department II of Internal Medicine and Center for Molecular Medicine Cologne; University of Cologne; Cologne Germany
| | - Sabine Bertsch
- Department II of Internal Medicine and Center for Molecular Medicine Cologne; University of Cologne; Cologne Germany
| | - Jan-Robin Wagener
- Institute for Neuroanatomy, Universitätsmedizin Göttingen; Georg-August-University Göttingen; Göttingen Germany
| | - Heiko Schweizer
- Renal Division; University Hospital Freiburg; Freiburg Germany
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne; University of Cologne; Cologne Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD); University of Cologne; Cologne Germany
- Systems Biology of Ageing Cologne (Sybacol); University of Cologne; Cologne Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne; University of Cologne; Cologne Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD); University of Cologne; Cologne Germany
- Systems Biology of Ageing Cologne (Sybacol); University of Cologne; Cologne Germany
| | - Martin Hoehne
- Department II of Internal Medicine and Center for Molecular Medicine Cologne; University of Cologne; Cologne Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD); University of Cologne; Cologne Germany
- Systems Biology of Ageing Cologne (Sybacol); University of Cologne; Cologne Germany
| |
Collapse
|
3
|
Hisaoka T, Komori T, Kitamura T, Morikawa Y. Abnormal behaviours relevant to neurodevelopmental disorders in Kirrel3-knockout mice. Sci Rep 2018; 8:1408. [PMID: 29362445 PMCID: PMC5780462 DOI: 10.1038/s41598-018-19844-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 01/09/2018] [Indexed: 12/21/2022] Open
Abstract
In the nervous system, Kirrel3 is involved in neuronal migration, axonal fasciculation, and synapse formation. Recently, genetic links have been reported between mutations in the KIRREL3 gene and increased risk of neurodevelopmental disorders, including autism spectrum disorder (ASD) and intellectual disability. To elucidate the causal relationship between KIRREL3 deficiency and behavioural abnormalities relevant to neurodevelopmental disorders, we generated global Kirrel3-knockout (Kirrel3−/−) mice and investigated the detailed behavioural phenotypes. In the three-chambered social approach test, Kirrel3−/− mice displayed a significant preference for a mouse over a non-social object but no significant preference for a stranger mouse over a familiar mouse. Ultrasonic communications, including pup-to-mother calls, male-female courtship vocalisation and resident responses to intruder, were significantly impaired in Kirrel3−/− mice. Significant increases in locomotor activity and repetitive rearing were also observed in Kirrel3−/− mice. Furthermore, the performance of Kirrel3−/− mice in the rotarod test was significantly better than that of wild-type mice. In the acoustic startle test, Kirrel3−/− mice were significantly hypersensitive to acoustic stimuli. Anxiety-related behaviours and spatial or fear memory acquisition were normal in Kirrel3−/− mice. These findings suggest that Kirrel3−/− mice exhibit autistic-like behaviours, including social and communicative deficits, repetitive behaviours, and sensory abnormalities, as well as hyperactivity.
Collapse
Affiliation(s)
- Tomoko Hisaoka
- Department of Anatomy and Neurobiology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Tadasuke Komori
- Department of Anatomy and Neurobiology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Toshio Kitamura
- Division of Cellular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Yoshihiro Morikawa
- Department of Anatomy and Neurobiology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan.
| |
Collapse
|
4
|
Baig DN, Yanagawa T, Tabuchi K. Distortion of the normal function of synaptic cell adhesion molecules by genetic variants as a risk for autism spectrum disorders. Brain Res Bull 2016; 129:82-90. [PMID: 27743928 DOI: 10.1016/j.brainresbull.2016.10.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 10/08/2016] [Accepted: 10/10/2016] [Indexed: 12/15/2022]
Abstract
Synaptic cell adhesion molecules (SCAMs) are a functional category of cell adhesion molecules that connect pre- and postsynapses by the protein-protein interaction via their extracellular cell adhesion domains. Countless numbers of common genetic variants and rare mutations in SCAMs have been identified in the patients with autism spectrum disorders (ASDs). Among these, NRXN and NLGN family proteins cooperatively function at synaptic terminals both of which genes are strongly implicated as risk genes for ASDs. Knock-in mice carrying a single rare point mutation of NLGN3 (NLGN3 R451C) discovered in the patients with ASDs display a deficit in social interaction and an enhancement of spatial learning and memory ability reminiscent of the clinical phenotype of ASDs. NLGN4 knockout (KO) and NRXN2α KO mice also show a deficit in sociability as well as some specific neuropsychiatric behaviors. In this review, we selected NRXNs/NLGNs, CNTNAP2/CNTNAP4, CNTN4, ITGB3, and KIRREL3 as strong ASD risk genes based on SFARI score and summarize the protein structures, functions at synapses, representative discoveries in human genetic studies, and phenotypes of the mutant model mice in light of the pathophysiology of ASDs.
Collapse
Affiliation(s)
- Deeba Noreen Baig
- Department of Biological Sciences, Forman Christian College, Zahoor Elahi Rd, Lahore, 54600, Pakistan
| | - Toru Yanagawa
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Katsuhiko Tabuchi
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan; Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, 390-8621, Japan; PRESTO, JST, Saitama, 332-0012, Japan.
| |
Collapse
|
5
|
Saleh M, Jürchott K, Oberland S, Neuhaus EM, Kramer A, Abraham U. Genome-Wide Screen Reveals Rhythmic Regulation of Genes Involved in Odor Processing in the Olfactory Epithelium. J Biol Rhythms 2015; 30:506-18. [PMID: 26482709 DOI: 10.1177/0748730415610197] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Odor discrimination behavior displays circadian fluctuations in mice, indicating that mammalian olfactory function is under control of the circadian system. This is further supported by the facts that odor discrimination rhythms depend on the presence of clock genes and that olfactory tissues contain autonomous circadian clocks. However, the molecular link between circadian function and olfactory processing is still unknown. To elucidate the molecular mechanisms underlying this link, we focused on the olfactory epithelium (OE), the primary target of odors and the site of the initial events in olfactory processing. We asked whether olfactory sensory neurons (OSNs) within the OE possess an autonomous circadian clock and whether olfactory pathways are under circadian control. Employing clock gene-driven bioluminescence reporter assays and time-dependent immunohistochemistry on OE samples, we found robust circadian rhythms of core clock genes and their proteins in OSNs, suggesting that the OE indeed contains an autonomous circadian clock. Furthermore, we performed a circadian transcriptome analysis and identified several OSN-specific components that are under circadian control, including those with putative roles in circadian olfactory processing, such as KIRREL2-an established factor involved in short-term OSN activation. The spatiotemporal expression patterns of our candidate proteins suggest that they are involved in short-term anabolic processes to rhythmically prepare the cell for peak performances and to promote circadian function of OSNs.
Collapse
Affiliation(s)
- Manjana Saleh
- Laboratory of Chronobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Karsten Jürchott
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sonja Oberland
- Cluster of Excellence NeuroCure, Charité-Universtitätsmedizin Berlin, Berlin, Germany Department of Pharmacology and Toxicology, University Hospital Jena, Jena, Germany
| | - Eva M Neuhaus
- Department of Pharmacology and Toxicology, University Hospital Jena, Jena, Germany
| | - Achim Kramer
- Laboratory of Chronobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ute Abraham
- Laboratory of Chronobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
6
|
Linneweber GA, Winking M, Fischbach KF. The Cell Adhesion Molecules Roughest, Hibris, Kin of Irre and Sticks and Stones Are Required for Long Range Spacing of the Drosophila Wing Disc Sensory Sensilla. PLoS One 2015; 10:e0128490. [PMID: 26053791 PMCID: PMC4459997 DOI: 10.1371/journal.pone.0128490] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 04/27/2015] [Indexed: 12/18/2022] Open
Abstract
Most animal tissues and organ systems are comprised of highly ordered arrays of varying cell types. The development of external sensory organs requires complex cell-cell communication in order to give each cell a specific identity and to ensure a regular distributed pattern of the sensory bristles. This involves both long and short range signaling mediated by either diffusible or cell anchored factors. In a variety of processes the heterophilic Irre Cell Recognition Module, consisting of the Neph-like proteins: Roughest, Kin of irre and of the Nephrin-like proteins: Sticks and Stones, Hibris, plays key roles in the recognition events of different cell types throughout development. In the present study these proteins are apically expressed in the adhesive belt of epithelial cells participating in sense organ development in a partially exclusive and asymmetric manner. Using mutant analysis the GAL4/UAS system, RNAi and gain of function we found an involvement of all four Irre Cell Recognition Module-proteins in the development of a highly structured array of sensory organs in the wing disc. The proteins secure the regular spacing of sensory organs showing partial redundancy and may function in early lateral inhibition events as well as in cell sorting processes. Comparisons with other systems suggest that the Irre Cell Recognition module is a key organizer of highly repetitive structures.
Collapse
Affiliation(s)
- Gerit Arne Linneweber
- Department of Neurobiology, Albert-Ludwigs-University Freiburg, Schänzlestr. 1, D-79104, Freiburg, Germany
| | - Mathis Winking
- Department of Neurobiology, Albert-Ludwigs-University Freiburg, Schänzlestr. 1, D-79104, Freiburg, Germany
| | - Karl-Friedrich Fischbach
- Department of Neurobiology, Albert-Ludwigs-University Freiburg, Schänzlestr. 1, D-79104, Freiburg, Germany
| |
Collapse
|
7
|
Autism and Intellectual Disability-Associated KIRREL3 Interacts with Neuronal Proteins MAP1B and MYO16 with Potential Roles in Neurodevelopment. PLoS One 2015; 10:e0123106. [PMID: 25902260 PMCID: PMC4406691 DOI: 10.1371/journal.pone.0123106] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 02/27/2015] [Indexed: 11/19/2022] Open
Abstract
Cell-adhesion molecules of the immunoglobulin superfamily play critical roles in brain development, as well as in maintaining synaptic plasticity, the dysfunction of which is known to cause cognitive impairment. Recently dysfunction of KIRREL3, a synaptic molecule of the immunoglobulin superfamily, has been implicated in several neurodevelopmental conditions including intellectual disability, autism spectrum disorder, and in the neurocognitive delay associated with Jacobsen syndrome. However, the molecular mechanisms of its physiological actions remain largely unknown. Using a yeast two-hybrid screen, we found that the KIRREL3 extracellular domain interacts with brain expressed proteins MAP1B and MYO16 and its intracellular domain can potentially interact with ATP1B1, UFC1, and SHMT2. The interactions were confirmed by co-immunoprecipitation and colocalization analyses of proteins expressed in human embryonic kidney cells, mouse neuronal cells, and rat primary neuronal cells. Furthermore, we show KIRREL3 colocalization with the marker for the Golgi apparatus and synaptic vesicles. Previously, we have shown that KIRREL3 interacts with the X-linked intellectual disability associated synaptic scaffolding protein CASK through its cytoplasmic domain. In addition, we found a genomic deletion encompassing MAP1B in one patient with intellectual disability, microcephaly and seizures and deletions encompassing MYO16 in two unrelated patients with intellectual disability, autism and microcephaly. MAP1B has been previously implicated in synaptogenesis and is involved in the development of the actin-based membrane skeleton. MYO16 is expressed in hippocampal neurons and also indirectly affects actin cytoskeleton through its interaction with WAVE1 complex. We speculate KIRREL3 interacting proteins are potential candidates for intellectual disability and autism spectrum disorder. Moreover, our findings provide further insight into understanding the molecular mechanisms underlying the physiological action of KIRREL3 and its role in neurodevelopment.
Collapse
|
8
|
Identification of novel Kirrel3 gene splice variants in adult human skeletal muscle. BMC PHYSIOLOGY 2014; 14:11. [PMID: 25488023 PMCID: PMC4269076 DOI: 10.1186/s12899-014-0011-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 11/19/2014] [Indexed: 01/08/2023]
Abstract
Background Multiple cell types including trophoblasts, osteoclasts and myoblasts require somatic cell fusion events as part of their physiological functions. In Drosophila Melanogaster the paralogus type 1 transmembrane receptors and members of the immunoglobulin superfamily Kin of Irre (Kirre) and roughest (Rst) regulate myoblast fusion during embryonic development. Present within the human genome are three homologs to Kirre termed Kin of Irre like (Kirrel) 1, 2 and 3. Currently it is unknown if Kirrel3 is expressed in adult human skeletal muscle. Results We investigated (using PCR and Western blot) Kirrel3 in adult human skeletal muscle samples taken at rest and after mild exercise induced muscle damage. Kirrel3 mRNA expression was verified by sequencing and protein presence via blotting with 2 different anti-Kirrel3 protein antibodies. Evidence for three alternatively spliced Kirrel3 mRNA transcripts in adult human skeletal muscle was obtained. Kirrel3 mRNA in adult human skeletal muscle was detected at low or moderate levels, or not at all. This sporadic expression suggests that Kirrel3 is expressed in a pulsatile manner. Several anti Kirrel3 immunoreactive proteins were detected in all adult human skeletal muscle samples analysed and results suggest the presence of different isoforms or posttranslational modification, or both. Conclusion The results presented here demonstrate for the first time that there are at least 3 splice variants of Kirrel3 expressed in adult human skeletal muscle, two of which have never previously been identified in human muscle. Importantly, mRNA of all splice variants was not always present, a finding with potential physiological relevance. These initial discoveries highlight the need for more molecular and functional studies to understand the role of Kirrel3 in human skeletal muscle.
Collapse
|
9
|
Durcan PJ, Al-Shanti N, Stewart CE. Identification and characterization of novel Kirrel isoform during myogenesis. Physiol Rep 2013; 1:e00044. [PMID: 24303129 PMCID: PMC3835000 DOI: 10.1002/phy2.44] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 07/03/2013] [Indexed: 12/31/2022] Open
Abstract
Somatic cell fusion is an essential component of skeletal muscle development and growth and repair from injury. Additional cell types such as trophoblasts and osteoclasts also require somatic cell fusion events to perform their physiological functions. Currently we have rudimentary knowledge on molecular mechanisms regulating somatic cell fusion events in mammals. We therefore investigated during in vitro murine myogenesis a mammalian homolog, Kirrel, of the Drosophila Melanogaster genes Roughest (Rst) and Kin of Irre (Kirre) which regulate somatic muscle cell fusion during embryonic development. Our results demonstrate the presence of a novel murine Kirrel isoform containing a truncated cytoplasmic domain which we term Kirrel B. Protein expression levels of Kirrel B are inverse to the occurrence of cell fusion events during in vitro myogenesis which is in stark contrast to the expression profile of Rst and Kirre during myogenesis in Drosophila. Furthermore, chemical inhibition of cell fusion confirmed the inverse expression pattern of Kirrel B protein levels in relation to cell fusion events. The discovery of a novel Kirrel B protein isoform during myogenesis highlights the need for more thorough investigation of the similarities and potential differences between fly and mammals with regards to the muscle cell fusion process.
Collapse
Affiliation(s)
- Peter J Durcan
- Department of Physiological Sciences, Stellenbosch University Merriman avenue, Stellenbosch, 7600, Western Cape, South Africa ; Institute for Biomedical Research into Human movement, School of Healthcare Science, Manchester Metropolitan University Oxford road, M1 5GD, Manchester, U.K
| | | | | |
Collapse
|
10
|
Kong L, Choi RC, Tsim KW, Jing N, Nakayama DK, Wang Z. Distribution and expression of Kirre, an IgSF molecule, during postnatal development of rat cerebellum. Neurosci Lett 2013; 543:22-6. [DOI: 10.1016/j.neulet.2013.03.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 03/11/2013] [Accepted: 03/17/2013] [Indexed: 11/24/2022]
|
11
|
Talkowski ME, Rosenfeld JA, Blumenthal I, Pillalamarri V, Chiang C, Heilbut A, Ernst C, Hanscom C, Rossin E, Lindgren A, Pereira S, Ruderfer D, Kirby A, Ripke S, Harris D, Lee JH, Ha K, Kim HG, Solomon BD, Gropman AL, Lucente D, Sims K, Ohsumi TK, Borowsky ML, Loranger S, Quade B, Lage K, Miles J, Wu BL, Shen Y, Neale B, Shaffer LG, Daly MJ, Morton CC, Gusella JF. Sequencing chromosomal abnormalities reveals neurodevelopmental loci that confer risk across diagnostic boundaries. Cell 2012; 149:525-37. [PMID: 22521361 PMCID: PMC3340505 DOI: 10.1016/j.cell.2012.03.028] [Citation(s) in RCA: 434] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 02/27/2012] [Accepted: 03/28/2012] [Indexed: 01/18/2023]
Abstract
Balanced chromosomal abnormalities (BCAs) represent a relatively untapped reservoir of single-gene disruptions in neurodevelopmental disorders (NDDs). We sequenced BCAs in patients with autism or related NDDs, revealing disruption of 33 loci in four general categories: (1) genes previously associated with abnormal neurodevelopment (e.g., AUTS2, FOXP1, and CDKL5), (2) single-gene contributors to microdeletion syndromes (MBD5, SATB2, EHMT1, and SNURF-SNRPN), (3) novel risk loci (e.g., CHD8, KIRREL3, and ZNF507), and (4) genes associated with later-onset psychiatric disorders (e.g., TCF4, ZNF804A, PDE10A, GRIN2B, and ANK3). We also discovered among neurodevelopmental cases a profoundly increased burden of copy-number variants from these 33 loci and a significant enrichment of polygenic risk alleles from genome-wide association studies of autism and schizophrenia. Our findings suggest a polygenic risk model of autism and reveal that some neurodevelopmental genes are sensitive to perturbation by multiple mutational mechanisms, leading to variable phenotypic outcomes that manifest at different life stages.
Collapse
Affiliation(s)
- Michael E. Talkowski
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
- Department of Neurology, Harvard Medical School, Boston, MA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA
| | | | - Ian Blumenthal
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
| | - Vamsee Pillalamarri
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
| | - Colby Chiang
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
| | - Adrian Heilbut
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
| | - Carl Ernst
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
| | - Carrie Hanscom
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
| | - Elizabeth Rossin
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA
| | - Amelia Lindgren
- Departments of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women’s Hospital, Boston, MA
| | - Shahrin Pereira
- Departments of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women’s Hospital, Boston, MA
| | - Douglas Ruderfer
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA
| | - Andrew Kirby
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA
| | - Stephan Ripke
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA
| | - David Harris
- Division of Clinical Genetics, Children’s Hospital of Boston, Boston, MA
| | - Ji-Hyun Lee
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
| | - Kyungsoo Ha
- Cancer Research Center, Georgia Health Sciences University, Augusta, GA
| | - Hyung-Goo Kim
- Department of OB/GYN, IMMAG, Georgia Health Sciences University, Augusta, GA
| | - Benjamin D. Solomon
- Medical Genetics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Andrea L. Gropman
- Department of Neurology, Children’s National Medical Center, Washington, DC, USA
- Department of Neurology, George Washington University of Health Sciences, Washington, DC, USA
| | - Diane Lucente
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
| | - Katherine Sims
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
| | - Toshiro K. Ohsumi
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA
| | - Mark L. Borowsky
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA
| | | | - Bradley Quade
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Kasper Lage
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA
- Pediatric Surgical Research Laboratories, MassGeneral Hospital for Children, Massachusetts General Hospital, Boston, MA, USA
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
- Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Judith Miles
- Departments of Pediatrics, Medical Genetics & Pathology, The Thompson Center for Autism & Neurodevelopmental Disorders, University of Missouri Hospitals and Clinics, Columbia, MO
| | - Bai-Lin Wu
- Department of Pathology, Massachusetts General Hospital, Boston, MA
- Department of Laboratory Medicine, Children’s Hospital Boston, Boston, MA
- Children’s Hospital and Institutes of Biomedical Science, Fudan University, Shanghai, China
| | - Yiping Shen
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Boston, MA
- Department of Laboratory Medicine, Children’s Hospital Boston, Boston, MA
- Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Benjamin Neale
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA
| | - Lisa G. Shaffer
- Signature Genomic Laboratories, PerkinElmer, Inc., Spokane, WA
| | - Mark J. Daly
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA
- Autism Consortium of Boston, Boston, MA
| | - Cynthia C. Morton
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA
- Departments of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women’s Hospital, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - James F. Gusella
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA
- Autism Consortium of Boston, Boston, MA
- Department of Genetics, Harvard Medical School, Boston, MA
| |
Collapse
|
12
|
Expression of the IgSF protein Kirre in the rat central nervous system. Life Sci 2011; 88:590-7. [DOI: 10.1016/j.lfs.2011.01.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Revised: 12/17/2010] [Accepted: 01/14/2011] [Indexed: 11/23/2022]
|
13
|
Nishida K, Nakayama K, Yoshimura S, Murakami F. Role of Neph2 in pontine nuclei formation in the developing hindbrain. Mol Cell Neurosci 2011; 46:662-70. [DOI: 10.1016/j.mcn.2011.01.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Revised: 01/07/2011] [Accepted: 01/07/2011] [Indexed: 11/29/2022] Open
|
14
|
Sugie A, Umetsu D, Yasugi T, Fischbach KF, Tabata T. Recognition of pre- and postsynaptic neurons via nephrin/NEPH1 homologs is a basis for the formation of the Drosophila retinotopic map. Development 2010; 137:3303-13. [PMID: 20724453 DOI: 10.1242/dev.047332] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Topographic maps, which maintain the spatial order of neurons in the order of their axonal connections, are found in many parts of the nervous system. Here, we focus on the communication between retinal axons and their postsynaptic partners, lamina neurons, in the first ganglion of the Drosophila visual system, as a model for the formation of topographic maps. Post-mitotic lamina precursor cells differentiate upon receiving Hedgehog signals delivered through newly arriving retinal axons and, before maturing to extend neurites, extend short processes toward retinal axons to create the lamina column. The lamina column provides the cellular basis for establishing stereotypic synapses between retinal axons and lamina neurons. In this study, we identified two cell-adhesion molecules: Hibris, which is expressed in post-mitotic lamina precursor cells; and Roughest, which is expressed on retinal axons. Both proteins belong to the nephrin/NEPH1 family. We provide evidence that recognition between post-mitotic lamina precursor cells and retinal axons is mediated by interactions between Hibris and Roughest. These findings revealed mechanisms by which axons of presynaptic neurons deliver signals to induce the development of postsynaptic partners at the target area. Postsynaptic partners then recognize the presynaptic axons to make ensembles, thus establishing a topographic map along the anterior/posterior axis.
Collapse
Affiliation(s)
- Atsushi Sugie
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | |
Collapse
|
15
|
Nishida K, Hoshino M, Kawaguchi Y, Murakami F. Ptf1a directly controls expression of immunoglobulin superfamily molecules Nephrin and Neph3 in the developing central nervous system. J Biol Chem 2009; 285:373-80. [PMID: 19887377 DOI: 10.1074/jbc.m109.060657] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ptf1a, a basic helix-loop-helix transcription factor, plays an indispensable role for cell fate specification of subsets of neurons in the developing central nervous system. However, downstream molecules induced by Ptf1a during neural development have not been well characterized. In the present study, we identified immunoglobulin superfamily molecules, Nephrin and Neph3, as direct downstream targets of Ptf1a. First, the expression domains of Nephrin and Neph3 closely resembled those of Ptf1a in the developing retina, hypothalamus, cerebellum, hindbrain, and spinal cord. Second, Ptf1a bound directly to a PTF-binding motif in the 5'-flanking region of Nephrin and Neph3 genes. Third, Ptf1a activated transcription driven by the 5'-flanking region of these genes. Finally, the expression of Nephrin and Neph3 was lost in Ptf1a-null mice, whereas ectopic expression of Nephrin and Neph3 was induced by forced expression of Ptf1a. We provided further evidence that Nephrin and Neph3 could interact homophilically and heterophilically, suggesting that Nephrin and Neph3 might regulate certain developmental aspects of Ptf1a-positive neurons as homo- or heterooligomers.
Collapse
Affiliation(s)
- Kazuhiko Nishida
- Graduate School of Frontier Biosciences, Osaka University, Yamadaoka 1-3, Suita, Osaka 565-0871, Japan.
| | | | | | | |
Collapse
|
16
|
Shibata F, Goto-Koshino Y, Morikawa Y, Komori T, Ito M, Fukuchi Y, Houchins JP, Tsang M, Li DY, Kitamura T, Nakajima H. Roundabout 4 is expressed on hematopoietic stem cells and potentially involved in the niche-mediated regulation of the side population phenotype. Stem Cells 2009; 27:183-90. [PMID: 18927479 DOI: 10.1634/stemcells.2008-0292] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Roundabout (Robo) family proteins are immunoglobulin-type cell surface receptors that are expressed predominantly in the nervous system. The fourth member of this family, Robo4, is distinct from the other family members in that it is expressed specifically in endothelial cells. In this study, we examined the expression of Robo4 in hematopoietic stem cells (HSCs) and its possible role in HSC regulation. Robo4 mRNA was specifically expressed in murine HSCs and the immature progenitor cell fraction but not in lineage-positive cells or differentiated progenitors. Moreover, flow cytometry showed a correlation between higher expression of Robo4 and immature phenotypes of hematopoietic cells. Robo4(high) hematopoietic stem/progenitor cells presented higher clonogenic activity or long-term repopulating activity by colony assays or transplantation assays, respectively. A ligand for Robo4, Slit2, is specifically expressed in bone marrow stromal cells, and its expression was induced in osteoblasts in response to myelosuppressive stress. Interestingly, overexpression of Robo4 or Slit2 in HSCs resulted in their decreased residence in the c-Kit(+)Sca-1(+)Lineage(-)-side population fraction. These results indicate that Robo4 is expressed in HSCs, and Robo4/Slit2 signaling may play a role in HSC homeostasis in the bone marrow niche.
Collapse
Affiliation(s)
- Fumi Shibata
- Advanced Clinical Research Center, University of Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Fischbach KF, Linneweber GA, Andlauer TFM, Hertenstein A, Bonengel B, Chaudhary K. The irre cell recognition module (IRM) proteins. J Neurogenet 2009; 23:48-67. [PMID: 19132596 DOI: 10.1080/01677060802471668] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
One of the most challenging problems in developmental neurosciences is to understand the establishment and maintenance of specific membrane contacts between axonal, dendritic, and glial processes in the neuropils, which eventually secure neuronal connectivity. However, underlying cell recognition events are pivotal in other tissues as well. This brief review focuses on the pleiotropic functions of a small, evolutionarily conserved group of proteins of the immunoglobulin superfamily involved in cell recognition. In Drosophila, this protein family comprises Irregular chiasm C/Roughest (IrreC/Rst), Kin of irre (Kirre), and their interacting protein partners, Sticks and stones (SNS) and Hibris (Hbs). For simplicity, we propose to name this ensemble of proteins the irre cell recognition module (IRM) after the first identified member of this family. Here, we summarize evidence that the IRM proteins function together in various cellular interactions, including myoblast fusion, cell sorting, axonal pathfinding, and target recognition in the optic neuropils of Drosophila. Understanding IRM protein function will help to unravel the epigenetic rules by which the intricate neurite networks in sensory neuropils are formed.
Collapse
Affiliation(s)
- Karl-Friedrich Fischbach
- Department of Neurobiology, Institute for Biology III, Faculty of Biology, Albert-Ludwigs-University Freiburg, Germany.
| | | | | | | | | | | |
Collapse
|
18
|
Bhalla K, Luo Y, Buchan T, Beachem MA, Guzauskas GF, Ladd S, Bratcher SJ, Schroer RJ, Balsamo J, DuPont BR, Lilien J, Srivastava AK. Alterations in CDH15 and KIRREL3 in patients with mild to severe intellectual disability. Am J Hum Genet 2008; 83:703-13. [PMID: 19012874 DOI: 10.1016/j.ajhg.2008.10.020] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2008] [Revised: 10/18/2008] [Accepted: 10/24/2008] [Indexed: 11/17/2022] Open
Abstract
Cell-adhesion molecules play critical roles in brain development, as well as maintaining synaptic structure, function, and plasticity. Here we have found the disruption of two genes encoding putative cell-adhesion molecules, CDH15 (cadherin superfamily) and KIRREL3 (immunoglobulin superfamily), by a chromosomal translocation t(11;16) in a female patient with intellectual disability (ID). We screened coding regions of these two genes in a cohort of patients with ID and controls and identified four nonsynonymous CDH15 variants and three nonsynonymous KIRREL3 variants that appear rare and unique to ID. These variations altered highly conserved residues and were absent in more than 600 unrelated patients with ID and 800 control individuals. Furthermore, in vivo expression studies showed that three of the CDH15 variations adversely altered its ability to mediate cell-cell adhesion. We also show that in neuronal cells, human KIRREL3 colocalizes and interacts with the synaptic scaffolding protein, CASK, recently implicated in X-linked brain malformation and ID. Taken together, our data suggest that alterations in CDH15 and KIRREL3, either alone or in combination with other factors, could play a role in phenotypic expression of ID in some patients.
Collapse
MESH Headings
- Cadherins/chemistry
- Cadherins/genetics
- Cadherins/metabolism
- Carrier Proteins/chemistry
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Case-Control Studies
- Cell Adhesion
- Cell Adhesion Molecules, Neuronal/chemistry
- Cell Adhesion Molecules, Neuronal/genetics
- Cell Adhesion Molecules, Neuronal/metabolism
- Chromosomes, Human, Pair 11
- Chromosomes, Human, Pair 16
- Cohort Studies
- Female
- Genetic Variation
- Humans
- Intellectual Disability/genetics
- Membrane Proteins/chemistry
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Middle Aged
- Models, Biological
- Protein Structure, Tertiary
- Translocation, Genetic
Collapse
Affiliation(s)
- Kavita Bhalla
- J.C. Self Research Institute of Human Genetics, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Komori T, Gyobu H, Ueno H, Kitamura T, Senba E, Morikawa Y. Expression of kin of irregular chiasm-like 3/mKirre in proprioceptive neurons of the dorsal root ganglia and its interaction with nephrin in muscle spindles. J Comp Neurol 2008; 511:92-108. [DOI: 10.1002/cne.21838] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
20
|
Expression of mKirre in the developing sensory pathways: Its close apposition to nephrin-expressing cells. Neuroscience 2007; 150:880-6. [DOI: 10.1016/j.neuroscience.2007.10.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Revised: 10/15/2007] [Accepted: 10/19/2007] [Indexed: 11/18/2022]
|
21
|
Gerke P, Benzing T, Höhne M, Kispert A, Frotscher M, Walz G, Kretz O. Neuronal expression and interaction with the synaptic protein CASK suggest a role for Neph1 and Neph2 in synaptogenesis. J Comp Neurol 2006; 498:466-75. [PMID: 16874800 DOI: 10.1002/cne.21064] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Formation, differentiation, and plasticity of synapses require interactions between pre- and postsynaptic partners. Recently, it was shown that the transmembrane immunoglobulin superfamily protein SYG-1 is required for providing synaptic specificity in C. elegans. However, it is unclear whether the mammalian orthologs of SYG-1 are also involved in local cell interactions to determine specificity during synapse formation. We used in situ hybridization, immunohistochemistry, and immunogold electron microscopy to study the temporal and spatial expression of Neph1 and Neph2 in the developing and adult mouse brain. Both proteins show similar patterns with neuronal expression starting around embryonic days 12 and 11, respectively. Expression is strongest in areas of high migratory activity. In the adult brain, Neph1 and Neph2 are predominantly seen in the olfactory nerve layer and the glomerular layer of the olfactory bulb, in the hippocampus, and in Purkinje cells of the cerebellum. At the ultrastructural level, Neph1 and Neph2 are detectable within the dendritic shafts of pyramidal neurons. To a lesser extent, there is also synaptic localization of Neph1 within the stratum pyramidale of the hippocampal CA1 and CA3 region on both pre- and postsynaptic sites. Here it colocalizes with the synaptic scaffolder calmodulin-associated serin/threonin kinase (CASK), and both Neph1 and Neph2 interact with the PDZ domain of CASK via their cytoplasmic tail. Our results show that Neph proteins are expressed in the developing nervous system of mammals and suggest that these proteins may have a conserved function in synapse formation or neurogenesis.
Collapse
Affiliation(s)
- Peter Gerke
- Renal Division, University of Freiburg, D-79104 Freiburg, Germany
| | | | | | | | | | | | | |
Collapse
|