1
|
Iegiani G, Pallavicini G, Pezzotta A, Brix A, Ferraro A, Gai M, Boda E, Bielas SL, Pistocchi A, Di Cunto F. CITK modulates BRCA1 recruitment at DNA double strand breaks sites through HDAC6. Cell Death Dis 2025; 16:320. [PMID: 40254670 PMCID: PMC12009987 DOI: 10.1038/s41419-025-07655-4] [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: 01/03/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/22/2025]
Abstract
Citron Kinase (CITK) is a protein encoded by the CIT gene, whose pathogenic variants underlie microcephalic phenotypes that characterize MCPH17 syndrome. In neural progenitors, CITK loss leads to microtubule instability, resulting in mitotic spindle positioning defects, cytokinesis failure, and accumulation of DNA double strand breaks (DSBs), ultimately resulting in TP53-dependent senescence and apoptosis. Although DNA damage accumulation has been associated with impaired homologous recombination (HR), the role of CITK in this process and whether microtubule dynamics are involved is still unknown. In this report we show that CITK is required for proper BRCA1 localization at sites of DNA DSBs. We found that CITK's scaffolding, rather than its catalytic activity, is necessary for maintaining BRCA1 interphase levels in progenitor cells during neurodevelopment. CITK regulates the nuclear levels of HDAC6, a modulator of both microtubule stability and DNA damage repair. Targeting HDAC6 in CITK-deficient cells increases microtubule stability and recovers BRCA1 localization defects and DNA damage levels to that detected in controls. In addition, the CIT-HDAC6 axis is functionally relevant in a MCPH17 zebrafish model, as HDAC6 targeting recovers the head size phenotype produced by interfering with the CIT orthologue gene. These data provide novel insights into the functional interplay between HR and microtubule dynamics and into the pathogenesis of CITK based MCPH17, which may be relevant for development of therapeutic strategies.
Collapse
Affiliation(s)
- Giorgia Iegiani
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Torino, Italy
| | - Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Torino, Italy
| | - Alex Pezzotta
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milano, Italy
| | - Alessia Brix
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milano, Italy
| | - Alessia Ferraro
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Torino, Italy
| | - Marta Gai
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Enrica Boda
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Torino, Italy
| | - Stephanie L Bielas
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anna Pistocchi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milano, Italy
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy.
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Torino, Italy.
| |
Collapse
|
2
|
Klimontova M, Zhang H, Campos-Laborie F, Webster N, Andrews B, Kim Chung KC, Hili R, Kouzarides T, Bannister AJ. THUMPD3 regulates alternative splicing of ECM transcripts in human lung cancer cells and promotes proliferation and migration. PLoS One 2024; 19:e0314655. [PMID: 39656728 DOI: 10.1371/journal.pone.0314655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024] Open
Abstract
RNA-modifying enzymes have recently garnered considerable attention due to their relevance in cancer biology, identifying them as potential targets for novel therapeutic intervention. THUMPD3 was recently identified as an RNA methyltransferase catalysing N2-methylguanosine (m2G) within certain tRNAs. In this study, we unveil a novel role for THUMPD3 in lung cancer cells. Depletion of the enzyme from lung cancer cells significantly impairs their fitness, negatively impacting key cellular processes such as proliferation and migration. Notably, exogenous expression of THUMPD3 in normal lung fibroblasts stimulates their proliferation rate. Additionally, transcriptome-wide analyses reveal that depletion of THUMPD3 from lung cancer cells induces substantial changes in the expression of cell surface proteins, including those comprising the extracellular matrix (ECM). We further demonstrate that THUMPD3 maintains expression of an extra-domain B (EDB) containing pro-tumour isoform of Fibronectin-1 mRNA, encoding FN1, an important ECM protein. Crucially, depletion of THUMPD3 promotes an alternative splicing event that removes the EDB-encoding exon from Fibronectin-1. This is consistent with THUMPD3 depletion reducing cellular proliferation and migration. Moreover, depletion of THUMPD3 selectively and preferentially affects the alternative splicing of ECM and cell adhesion molecule encoding transcripts, as well as those encoding neurodevelopmental proteins. Overall, these findings highlight THUMPD3 as an important player in regulating cancer-relevant alternative splicing and they provide a rationale for further investigations into THUMPD3 as a candidate target in anti-cancer therapy.
Collapse
Affiliation(s)
- Marie Klimontova
- The Gurdon Institute and Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- STORM Therapeutics Ltd., Babraham Research Campus, Cambridge, United Kingdom
| | - Han Zhang
- The Gurdon Institute and Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Francisco Campos-Laborie
- The Gurdon Institute and Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Natalie Webster
- STORM Therapeutics Ltd., Babraham Research Campus, Cambridge, United Kingdom
| | - Byron Andrews
- STORM Therapeutics Ltd., Babraham Research Campus, Cambridge, United Kingdom
| | - Kimberley Chung Kim Chung
- Department of Chemistry and Centre for Research on Biomolecular Interactions, York University, Toronto, ON, Canada
| | - Ryan Hili
- Department of Chemistry and Centre for Research on Biomolecular Interactions, York University, Toronto, ON, Canada
| | - Tony Kouzarides
- The Gurdon Institute and Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- Milner Therapeutics Institute, University of Cambridge, Cambridge, United Kingdom
| | - Andrew J Bannister
- The Gurdon Institute and Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
3
|
Pallavicini G, Moccia A, Iegiani G, Parolisi R, Peirent ER, Berto GE, Lorenzati M, Tshuva RY, Ferraro A, Balzac F, Turco E, Salvi SU, Myklebust HF, Wang S, Eisenberg J, Chitale M, Girgla NS, Boda E, Reiner O, Buffo A, Di Cunto F, Bielas SL. Modeling primary microcephaly with human brain organoids reveals fundamental roles of CIT kinase activity. J Clin Invest 2024; 134:e175435. [PMID: 39316437 PMCID: PMC11527453 DOI: 10.1172/jci175435] [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: 09/20/2023] [Accepted: 09/06/2024] [Indexed: 09/26/2024] Open
Abstract
Brain size and cellular heterogeneity are tightly regulated by species-specific proliferation and differentiation of multipotent neural progenitor cells (NPCs). Errors in this process are among the mechanisms of primary hereditary microcephaly (MCPH), a group of disorders characterized by reduced brain size and intellectual disability. Biallelic citron rho-interacting serine/threonine kinase (CIT) missense variants that disrupt kinase function (CITKI/KI) and frameshift loss-of-function variants (CITFS/FS) are the genetic basis for MCPH17; however, the function of CIT catalytic activity in brain development and NPC cytokinesis is unknown. Therefore, we created the CitKI/KI mouse model and found that it did not phenocopy human microcephaly, unlike biallelic CitFS/FS animals. Nevertheless, both Cit models exhibited binucleation, DNA damage, and apoptosis. To investigate human-specific mechanisms of CIT microcephaly, we generated CITKI/KI and CITFS/FS human forebrain organoids. We found that CITKI/KI and CITFS/FS organoids lost cytoarchitectural complexity, transitioning from pseudostratified to simple neuroepithelium. This change was associated with defects that disrupted the polarity of NPC cytokinesis, in addition to elevating apoptosis. Together, our results indicate that both CIT catalytic and scaffolding functions in NPC cytokinesis are critical for human corticogenesis. Species differences in corticogenesis and the dynamic 3D features of NPC mitosis underscore the utility of human forebrain organoid models for understanding human microcephaly.
Collapse
Affiliation(s)
- Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | | | - Giorgia Iegiani
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Roberta Parolisi
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Emily R. Peirent
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Gaia Elena Berto
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Martina Lorenzati
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Rami Y. Tshuva
- Departments of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Alessia Ferraro
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Fiorella Balzac
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | | | | | | | - Julia Eisenberg
- Department of Human Genetics and
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | - Enrica Boda
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Orly Reiner
- Departments of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Annalisa Buffo
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Stephanie L. Bielas
- Department of Human Genetics and
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
Connors CQ, Mauro MS, Wiles JT, Countryman AD, Martin SL, Lacroix B, Shirasu-Hiza M, Dumont J, Kasza KE, Davies TR, Canman JC. Germ fate determinants protect germ precursor cell division by reducing septin and anillin levels at the cell division plane. Mol Biol Cell 2024; 35:ar94. [PMID: 38696255 PMCID: PMC11244169 DOI: 10.1091/mbc.e24-02-0096-t] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 05/04/2024] Open
Abstract
Animal cell cytokinesis, or the physical division of one cell into two, is thought to be driven by constriction of an actomyosin contractile ring at the division plane. The mechanisms underlying cell type-specific differences in cytokinesis remain unknown. Germ cells are totipotent cells that pass genetic information to the next generation. Previously, using formincyk-1(ts) mutant Caenorhabditis elegans 4-cell embryos, we found that the P2 germ precursor cell is protected from cytokinesis failure and can divide with greatly reduced F-actin levels at the cell division plane. Here, we identified two canonical germ fate determinants required for P2-specific cytokinetic protection: PIE-1 and POS-1. Neither has been implicated previously in cytokinesis. These germ fate determinants protect P2 cytokinesis by reducing the accumulation of septinUNC-59 and anillinANI-1 at the division plane, which here act as negative regulators of cytokinesis. These findings may provide insight into the regulation of cytokinesis in other cell types, especially in stem cells with high potency.
Collapse
Affiliation(s)
- Caroline Q. Connors
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
| | - Michael S. Mauro
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
| | - J. Tristian Wiles
- Department of Biological Sciences, Columbia University, New York, NY 10027
| | | | - Sophia L. Martin
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
| | - Benjamin Lacroix
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
- Université de Montpellier, CNRS, Centre de Recherche en Biologie Cellulaire de Montpellier, UMR 5237 Montpellier, France
| | - Mimi Shirasu-Hiza
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032
| | - Julien Dumont
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Karen E. Kasza
- Department of Mechanical Engineering, Columbia University, New York, NY 10027
| | - Timothy R. Davies
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Department of Biosciences, Durham University, Durham DH1 3LE, UK
| | - Julie C. Canman
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
| |
Collapse
|
5
|
Aguila A, Salah S, Kulasekaran G, Shweiki M, Shaul-Lotan N, Mor-Shaked H, Daana M, Harel T, McPherson PS. A neurodevelopmental disorder associated with a loss-of-function missense mutation in RAB35. J Biol Chem 2024; 300:107124. [PMID: 38432637 PMCID: PMC10966776 DOI: 10.1016/j.jbc.2024.107124] [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: 08/01/2023] [Revised: 02/01/2024] [Accepted: 02/18/2024] [Indexed: 03/05/2024] Open
Abstract
Rab35 (Ras-associated binding protein) is a small GTPase that regulates endosomal membrane trafficking and functions in cell polarity, cytokinesis, and growth factor signaling. Altered Rab35 function contributes to progression of glioblastoma, defects in primary cilia formation, and altered cytokinesis. Here, we report a pediatric patient with global developmental delay, hydrocephalus, a Dandy-Walker malformation, axial hypotonia with peripheral hypertonia, visual problems, and conductive hearing impairment. Exome sequencing identified a homozygous missense variant in the GTPase fold of RAB35 (c.80G>A; p.R27H) as the most likely candidate. Functional analysis of the R27H-Rab35 variant protein revealed enhanced interaction with its guanine-nucleotide exchange factor, DENND1A and decreased interaction with a known effector, MICAL1, indicating that the protein is in an inactive conformation. Cellular expression of the variant drives the activation of Arf6, a small GTPase under negative regulatory control of Rab35. Importantly, variant expression leads to delayed cytokinesis and altered length, number, and Arl13b composition of primary cilia, known factors in neurodevelopmental disease. Our findings provide evidence of altered Rab35 function as a causative factor of a neurodevelopmental disorder.
Collapse
Affiliation(s)
- Adriana Aguila
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Somaya Salah
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel
| | - Gopinath Kulasekaran
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Moatasem Shweiki
- Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel
| | - Nava Shaul-Lotan
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel
| | - Hagar Mor-Shaked
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel; Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Muhannad Daana
- Child Development Centers, Clalit Health Care Services, Yokne'am Illit, Israel
| | - Tamar Harel
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel; Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Peter S McPherson
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
6
|
Maw JJ, Coker JA, Arya T, Goins CM, Sonawane D, Han SH, Rees MG, Ronan MM, Roth JA, Wang NS, Heemers HV, Macdonald JD, Stauffer SR. Discovery and Characterization of Selective, First-in-Class Inhibitors of Citron Kinase. J Med Chem 2024; 67:2631-2666. [PMID: 38330278 DOI: 10.1021/acs.jmedchem.3c01807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Citron kinase (CITK) is an AGC-family serine/threonine kinase that regulates cytokinesis. Despite knockdown experiments implicating CITK as an anticancer target, no selective CITK inhibitors exist. We transformed a previously reported kinase inhibitor with weak off-target CITK activity into a first-in-class CITK chemical probe, C3TD879. C3TD879 is a Type I kinase inhibitor which potently inhibits CITK catalytic activity (biochemical IC50 = 12 nM), binds directly to full-length human CITK in cells (NanoBRET Kd < 10 nM), and demonstrates favorable DMPK properties for in vivo evaluation. We engineered exquisite selectivity for CITK (>17-fold versus 373 other human kinases), making C3TD879 the first chemical probe suitable for interrogating the complex biology of CITK. Our small-molecule CITK inhibitors could not phenocopy the effects of CITK knockdown in cell proliferation, cell cycle progression, or cytokinesis assays, providing preliminary evidence that the structural roles of CITK may be more important than its kinase activity.
Collapse
Affiliation(s)
- Joshua J Maw
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Jesse A Coker
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Tarun Arya
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Christopher M Goins
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Dhiraj Sonawane
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Sang Hoon Han
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Matthew G Rees
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge Massachusetts 02142, United States
| | - Melissa M Ronan
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge Massachusetts 02142, United States
| | - Jennifer A Roth
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge Massachusetts 02142, United States
| | - Nancy S Wang
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Hannelore V Heemers
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Jonathan D Macdonald
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Shaun R Stauffer
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| |
Collapse
|
7
|
Connors CQ, Mauro MS, Tristian Wiles J, Countryman AD, Martin SL, Lacroix B, Shirasu-Hiza M, Dumont J, Kasza KE, Davies TR, Canman JC. Germ fate determinants protect germ precursor cell division by restricting septin and anillin levels at the division plane. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.566773. [PMID: 38014027 PMCID: PMC10680835 DOI: 10.1101/2023.11.17.566773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Animal cell cytokinesis, or the physical division of one cell into two, is thought to be driven by constriction of an actomyosin contractile ring at the division plane. The mechanisms underlying cell type-specific differences in cytokinesis remain unknown. Germ cells are totipotent cells that pass genetic information to the next generation. Previously, using formin cyk-1 (ts) mutant C. elegans embryos, we found that the P2 germ precursor cell is protected from cytokinesis failure and can divide without detectable F-actin at the division plane. Here, we identified two canonical germ fate determinants required for P2-specific cytokinetic protection: PIE-1 and POS-1. Neither has been implicated previously in cytokinesis. These germ fate determinants protect P2 cytokinesis by reducing the accumulation of septin UNC-59 and anillin ANI-1 at the division plane, which here act as negative regulators of cytokinesis. These findings may provide insight into cytokinetic regulation in other cell types, especially in stem cells with high potency.
Collapse
|
8
|
Medvedev KE, Schaeffer RD, Pei J, Grishin NV. Pathogenic mutation hotspots in protein kinase domain structure. Protein Sci 2023; 32:e4750. [PMID: 37572333 PMCID: PMC10464295 DOI: 10.1002/pro.4750] [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: 06/12/2023] [Revised: 08/04/2023] [Accepted: 08/08/2023] [Indexed: 08/14/2023]
Abstract
Control of eukaryotic cellular function is heavily reliant on the phosphorylation of proteins at specific amino acid residues, such as serine, threonine, tyrosine, and histidine. Protein kinases that are responsible for this process comprise one of the largest families of evolutionarily related proteins. Dysregulation of protein kinase signaling pathways is a frequent cause of a large variety of human diseases including cancer, autoimmune, neurodegenerative, and cardiovascular disorders. In this study, we mapped all pathogenic mutations in 497 human protein kinase domains from the ClinVar database to the reference structure of Aurora kinase A (AURKA) and grouped them by the relevance to the disease type. Our study revealed that the majority of mutation hotspots associated with cancer are situated within the catalytic and activation loops of the kinase domain, whereas non-cancer-related hotspots tend to be located outside of these regions. Additionally, we identified a hotspot at residue R371 of the AURKA structure that has the highest number of exclusively non-cancer-related pathogenic mutations (21) and has not been previously discussed.
Collapse
Affiliation(s)
- Kirill E. Medvedev
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - R. Dustin Schaeffer
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Jimin Pei
- Eugene McDermott Center for Human Growth and DevelopmentUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Nick V. Grishin
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| |
Collapse
|
9
|
Fong S, Carollo A, Ashour R, Dimitriou D, Gianluca Esposito. Identifying major research themes in the literature on developmental disabilities in Middle Eastern countries: A scientometric review from 1962 to 2023. RESEARCH IN DEVELOPMENTAL DISABILITIES 2023; 140:104551. [PMID: 37473627 DOI: 10.1016/j.ridd.2023.104551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/05/2023] [Accepted: 06/02/2023] [Indexed: 07/22/2023]
Abstract
Developmental disabilities have been widely studied in higher-income countries. However, most individuals with these conditions live in low- and middle-income countries and they are reportedly under-represented in the scientific literature. To tackle this issue, previous research has provided insight into the thematic developments in the research on developmental disabilities in Africa by means of a scientometric approach to reviews. The current work aims to extend the scientometric approach to investigate the main interests in the literature on developmental disabilities conducted in Middle Eastern countries. A total of 1110 documents were retrieved from Scopus and their patterns of co-citation were analysed with the CiteSpace software. Research in Developmental Disabilities emerged to be the main source in the sample of downloaded documents. Furthermore, a total of six main thematic domains and the four most impactful documents in the literature were identified. Results showed that research on developmental disabilities in the Middle East has been mainly focused on uncovering the genetic basis of this group of conditions. The study of clinical profiles, diagnosis, management, and treatment of individuals with developmental disabilities have been so far under-investigated and represents material for future studies.
Collapse
Affiliation(s)
- Seraphina Fong
- Department of Psychology and Cognitive Science, University of Trento, 38068 Rovereto, Italy
| | - Alessandro Carollo
- Department of Psychology and Cognitive Science, University of Trento, 38068 Rovereto, Italy
| | - Rola Ashour
- Sleep Education and Research Laboratory, UCL Institute of Education, London WC1H 0AA, England, UK
| | - Dagmara Dimitriou
- Sleep Education and Research Laboratory, UCL Institute of Education, London WC1H 0AA, England, UK
| | - Gianluca Esposito
- Department of Psychology and Cognitive Science, University of Trento, 38068 Rovereto, Italy.
| |
Collapse
|
10
|
Farcy S, Hachour H, Bahi-Buisson N, Passemard S. Genetic Primary Microcephalies: When Centrosome Dysfunction Dictates Brain and Body Size. Cells 2023; 12:1807. [PMID: 37443841 PMCID: PMC10340463 DOI: 10.3390/cells12131807] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/04/2023] [Accepted: 06/13/2023] [Indexed: 07/15/2023] Open
Abstract
Primary microcephalies (PMs) are defects in brain growth that are detectable at or before birth and are responsible for neurodevelopmental disorders. Most are caused by biallelic or, more rarely, dominant mutations in one of the likely hundreds of genes encoding PM proteins, i.e., ubiquitous centrosome or microtubule-associated proteins required for the division of neural progenitor cells in the embryonic brain. Here, we provide an overview of the different types of PMs, i.e., isolated PMs with or without malformations of cortical development and PMs associated with short stature (microcephalic dwarfism) or sensorineural disorders. We present an overview of the genetic, developmental, neurological, and cognitive aspects characterizing the most representative PMs. The analysis of phenotypic similarities and differences among patients has led scientists to elucidate the roles of these PM proteins in humans. Phenotypic similarities indicate possible redundant functions of a few of these proteins, such as ASPM and WDR62, which play roles only in determining brain size and structure. However, the protein pericentrin (PCNT) is equally required for determining brain and body size. Other PM proteins perform both functions, albeit to different degrees. Finally, by comparing phenotypes, we considered the interrelationships among these proteins.
Collapse
Affiliation(s)
- Sarah Farcy
- UMR144, Institut Curie, 75005 Paris, France;
- Inserm UMR-S 1163, Institut Imagine, 75015 Paris, France
| | - Hassina Hachour
- Service de Neurologie Pédiatrique, DMU INOV-RDB, APHP, Hôpital Robert Debré, 75019 Paris, France;
| | - Nadia Bahi-Buisson
- Service de Neurologie Pédiatrique, DMU MICADO, APHP, Hôpital Necker Enfants Malades, 75015 Paris, France;
- Université Paris Cité, Inserm UMR-S 1163, Institut Imagine, 75015 Paris, France
| | - Sandrine Passemard
- Service de Neurologie Pédiatrique, DMU INOV-RDB, APHP, Hôpital Robert Debré, 75019 Paris, France;
- Université Paris Cité, Inserm UMR 1141, NeuroDiderot, 75019 Paris, France
| |
Collapse
|
11
|
Pallavicini G, Iegiani G, Parolisi R, Ferraro A, Garello F, Bitonto V, Terreno E, Gai M, Di Cunto F. Lestaurtinib inhibits Citron kinase activity and medulloblastoma growth through induction of DNA damage, apoptosis and cytokinesis failure. Front Oncol 2023; 13:1202585. [PMID: 37404750 PMCID: PMC10315473 DOI: 10.3389/fonc.2023.1202585] [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: 04/19/2023] [Accepted: 05/30/2023] [Indexed: 07/06/2023] Open
Abstract
Introduction Medulloblastoma (MB), the most common malignant pediatric brain tumor, is currently treated with surgery followed by radiation and chemotherapy, which is accompanied by severe side effects, raising the need for innovative therapies. Disruption of the microcephaly-related gene Citron kinase (CITK) impairs the expansion of xenograft models as well as spontaneous MB arising in transgenic mice. No specific CITK inhibitors are available. Methods Lestaurtinib, a Staurosporine derivative also known as CEP-701, inhibits CITK with IC50 of 90 nM. We therefore tested the biological effects of this molecule on different MB cell lines, as well as in vivo, injecting the drug in MBs arising in SmoA1 transgenic mice. Results Similar to CITK knockdown, treatment of MB cells with 100 nM Lestaurtinib reduces phospho-INCENP levels at the midbody and leads to late cytokinesis failure. Moreover, Lestaurtinib impairs cell proliferation through CITK-sensitive mechanisms. These phenotypes are accompanied by accumulation of DNA double strand breaks, cell cycle block and TP53 superfamily activation in vitro and in vivo. Lestaurtinib treatment reduces tumor growth and increases mice survival. Discussion Our data indicate that Lestaurtinib produces in MB cells poly-pharmacological effects extending beyond the inhibition of its validated targets, supporting the possibility of repositioning this drug for MB treatment.
Collapse
Affiliation(s)
- Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, Turin, Italy
| | - Giorgia Iegiani
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, Turin, Italy
| | - Roberta Parolisi
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, Turin, Italy
| | - Alessia Ferraro
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, Turin, Italy
| | - Francesca Garello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Valeria Bitonto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Enzo Terreno
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Marta Gai
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, Turin, Italy
| |
Collapse
|
12
|
Dell'Amico C, Angulo Salavarria MM, Takeo Y, Saotome I, Dell'Anno MT, Galimberti M, Pellegrino E, Cattaneo E, Louvi A, Onorati M. Microcephaly-associated protein WDR62 shuttles from the Golgi apparatus to the spindle poles in human neural progenitors. eLife 2023; 12:e81716. [PMID: 37272619 PMCID: PMC10241521 DOI: 10.7554/elife.81716] [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: 07/08/2022] [Accepted: 04/17/2023] [Indexed: 06/06/2023] Open
Abstract
WDR62 is a spindle pole-associated scaffold protein with pleiotropic functions. Recessive mutations in WDR62 cause structural brain abnormalities and account for the second most common cause of autosomal recessive primary microcephaly (MCPH), indicating WDR62 as a critical hub for human brain development. Here, we investigated WDR62 function in corticogenesis through the analysis of a C-terminal truncating mutation (D955AfsX112). Using induced Pluripotent Stem Cells (iPSCs) obtained from a patient and his unaffected parent, as well as isogenic corrected lines, we generated 2D and 3D models of human neurodevelopment, including neuroepithelial stem cells, cerebro-cortical progenitors, terminally differentiated neurons, and cerebral organoids. We report that WDR62 localizes to the Golgi apparatus during interphase in cultured cells and human fetal brain tissue, and translocates to the mitotic spindle poles in a microtubule-dependent manner. Moreover, we demonstrate that WDR62 dysfunction impairs mitotic progression and results in alterations of the neurogenic trajectories of iPSC neuroderivatives. In summary, impairment of WDR62 localization and function results in severe neurodevelopmental abnormalities, thus delineating new mechanisms in the etiology of MCPH.
Collapse
Affiliation(s)
- Claudia Dell'Amico
- Department of Biology, Unit of Cell and Developmental Biology, University of PisaPisaItaly
| | | | - Yutaka Takeo
- Departments of Neurosurgery and Neuroscience, Yale School of MedicineNew HavenUnited States
| | - Ichiko Saotome
- Departments of Neurosurgery and Neuroscience, Yale School of MedicineNew HavenUnited States
| | | | - Maura Galimberti
- Dipartimento di Bioscienze, Università degli Studi di MilanoMilanItaly
- INGM, Istituto Nazionale Genetica MolecolareMilanItaly
| | - Enrica Pellegrino
- Department of Biology, Unit of Cell and Developmental Biology, University of PisaPisaItaly
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Elena Cattaneo
- Dipartimento di Bioscienze, Università degli Studi di MilanoMilanItaly
- INGM, Istituto Nazionale Genetica MolecolareMilanItaly
| | - Angeliki Louvi
- Departments of Neurosurgery and Neuroscience, Yale School of MedicineNew HavenUnited States
| | - Marco Onorati
- Department of Biology, Unit of Cell and Developmental Biology, University of PisaPisaItaly
| |
Collapse
|
13
|
Asif M, Abdullah U, Nürnberg P, Tinschert S, Hussain MS. Congenital Microcephaly: A Debate on Diagnostic Challenges and Etiological Paradigm of the Shift from Isolated/Non-Syndromic to Syndromic Microcephaly. Cells 2023; 12:cells12040642. [PMID: 36831309 PMCID: PMC9954724 DOI: 10.3390/cells12040642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Congenital microcephaly (CM) exhibits broad clinical and genetic heterogeneity and is thus categorized into several subtypes. However, the recent bloom of disease-gene discoveries has revealed more overlaps than differences in the underlying genetic architecture for these clinical sub-categories, complicating the differential diagnosis. Moreover, the mechanism of the paradigm shift from a brain-restricted to a multi-organ phenotype is only vaguely understood. This review article highlights the critical factors considered while defining CM subtypes. It also presents possible arguments on long-standing questions of the brain-specific nature of CM caused by a dysfunction of the ubiquitously expressed proteins. We argue that brain-specific splicing events and organ-restricted protein expression may contribute in part to disparate clinical manifestations. We also highlight the role of genetic modifiers and de novo variants in the multi-organ phenotype of CM and emphasize their consideration in molecular characterization. This review thus attempts to expand our understanding of the phenotypic and etiological variability in CM and invites the development of more comprehensive guidelines.
Collapse
Affiliation(s)
- Maria Asif
- Cologne Center for Genomics (CCG), Faculty of Medicine, University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine, University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Uzma Abdullah
- University Institute of Biochemistry and Biotechnology (UIBB), PMAS-Arid Agriculture University, Rawalpindi, Rawalpindi 46300, Pakistan
| | - Peter Nürnberg
- Cologne Center for Genomics (CCG), Faculty of Medicine, University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine, University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Sigrid Tinschert
- Zentrum Medizinische Genetik, Medizinische Universität, 6020 Innsbruck, Austria
| | - Muhammad Sajid Hussain
- Cologne Center for Genomics (CCG), Faculty of Medicine, University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine, University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
- Correspondence:
| |
Collapse
|
14
|
Farcy S, Albert A, Gressens P, Baffet AD, El Ghouzzi V. Cortical Organoids to Model Microcephaly. Cells 2022; 11:2135. [PMID: 35883578 PMCID: PMC9320662 DOI: 10.3390/cells11142135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/17/2022] [Accepted: 07/05/2022] [Indexed: 02/01/2023] Open
Abstract
How the brain develops and achieves its final size is a fascinating issue that questions cortical evolution across species and man's place in the animal kingdom. Although animal models have so far been highly valuable in understanding the key steps of cortical development, many human specificities call for appropriate models. In particular, microcephaly, a neurodevelopmental disorder that is characterized by a smaller head circumference has been challenging to model in mice, which often do not fully recapitulate the human phenotype. The relatively recent development of brain organoid technology from induced pluripotent stem cells (iPSCs) now makes it possible to model human microcephaly, both due to genetic and environmental origins, and to generate developing cortical tissue from the patients themselves. These 3D tissues rely on iPSCs differentiation into cortical progenitors that self-organize into neuroepithelial rosettes mimicking the earliest stages of human neurogenesis in vitro. Over the last ten years, numerous protocols have been developed to control the identity of the induced brain areas, the reproducibility of the experiments and the longevity of the cultures, allowing analysis of the later stages. In this review, we describe the different approaches that instruct human iPSCs to form cortical organoids, summarize the different microcephalic conditions that have so far been modeled by organoids, and discuss the relevance of this model to decipher the cellular and molecular mechanisms of primary and secondary microcephalies.
Collapse
Affiliation(s)
- Sarah Farcy
- Institut Curie, PSL Research University, CNRS UMR144, F-75005 Paris, France;
| | - Alexandra Albert
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (A.A.); (P.G.)
| | - Pierre Gressens
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (A.A.); (P.G.)
| | - Alexandre D. Baffet
- Institut Curie, PSL Research University, CNRS UMR144, F-75005 Paris, France;
| | - Vincent El Ghouzzi
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (A.A.); (P.G.)
| |
Collapse
|
15
|
Boda E, Lorenzati M, Parolisi R, Harding B, Pallavicini G, Bonfanti L, Moccia A, Bielas S, Di Cunto F, Buffo A. Molecular and functional heterogeneity in dorsal and ventral oligodendrocyte progenitor cells of the mouse forebrain in response to DNA damage. Nat Commun 2022; 13:2331. [PMID: 35484145 PMCID: PMC9051058 DOI: 10.1038/s41467-022-30010-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 04/07/2022] [Indexed: 11/09/2022] Open
Abstract
In the developing mouse forebrain, temporally distinct waves of oligodendrocyte progenitor cells (OPCs) arise from different germinal zones and eventually populate either dorsal or ventral regions, where they present as transcriptionally and functionally equivalent cells. Despite that, developmental heterogeneity influences adult OPC responses upon demyelination. Here we show that accumulation of DNA damage due to ablation of citron-kinase or cisplatin treatment cell-autonomously disrupts OPC fate, resulting in cell death and senescence in the dorsal and ventral subsets, respectively. Such alternative fates are associated with distinct developmental origins of OPCs, and with a different activation of NRF2-mediated anti-oxidant responses. These data indicate that, upon injury, dorsal and ventral OPC subsets show functional and molecular diversity that can make them differentially vulnerable to pathological conditions associated with DNA damage.
Collapse
Affiliation(s)
- Enrica Boda
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy.
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy.
| | - Martina Lorenzati
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
| | - Roberta Parolisi
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
| | - Brian Harding
- Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania and Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Gianmarco Pallavicini
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
| | - Luca Bonfanti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Amanda Moccia
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Stephanie Bielas
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Ferdinando Di Cunto
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
| | - Annalisa Buffo
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
| |
Collapse
|
16
|
González-Mancha N, Rodríguez-Rodríguez C, Alcover A, Merida I. Sorting Nexin 27 Enables MTOC and Secretory Machinery Translocation to the Immune Synapse. Front Immunol 2022; 12:814570. [PMID: 35095913 PMCID: PMC8790036 DOI: 10.3389/fimmu.2021.814570] [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: 11/13/2021] [Accepted: 12/20/2021] [Indexed: 11/24/2022] Open
Abstract
Sorting nexin 27 (SNX27) association to the retromer complex mediates intracellular trafficking of cargoes containing PSD95/Dlg1/ZO-1 (PDZ)-binding C-terminal sequences from endosomes to the cell surface, preventing their lysosomal degradation. Antigen recognition by T lymphocyte leads to the formation of a highly organized structure named the immune synapse (IS), which ensures cell-cell communication and sustained T cell activation. At the neuronal synapse, SNX27 recycles PDZ-binding receptors and its defective expression is associated with synaptic dysfunction and cognitive impairment. In T lymphocytes, SNX27 was found localized at recycling endosomal compartments that polarized to the IS, suggesting a function in polarized traffic to this structure. Proteomic analysis of PDZ-SNX27 interactors during IS formation identify proteins with known functions in cytoskeletal reorganization and lipid regulation, such as diacylglycerol (DAG) kinase (DGK) ζ, as well as components of the retromer and WASH complex. In this study, we investigated the consequences of SNX27 deficiency in cytoskeletal reorganization during IS formation. Our analyses demonstrate that SNX27 controls the polarization towards the cell-cell interface of the PDZ-interacting cargoes DGKζ and the retromer subunit vacuolar protein sorting protein 26, among others. SNX27 silencing abolishes the formation of a DAG gradient at the IS and prevents re-localization of the dynactin complex component dynactin-1/p150Glued, two events that correlate with impaired microtubule organizing center translocation (MTOC). SNX27 silenced cells show marked alteration in cytoskeleton organization including a failure in the organization of the microtubule network and defects in actin clearance at the IS. Reduced SNX27 expression was also found to hinder the arrangement of signaling microclusters at the IS, as well as the polarization of the secretory machinery towards the antigen presenting cells. Our results broaden the knowledge of SNX27 function in T lymphocytes by showing a function in modulating IS organization through regulated trafficking of cargoes.
Collapse
Affiliation(s)
- Natalia González-Mancha
- Department of Immunology and Oncology, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Cristina Rodríguez-Rodríguez
- Department of Immunology and Oncology, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Andrés Alcover
- Institut Pasteur, Université de Paris, Unité Biologie Cellulaire des Lymphocytes, INSERM U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue-2018, Paris, France
| | - Isabel Merida
- Department of Immunology and Oncology, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| |
Collapse
|
17
|
Zaqout S, Kaindl AM. Autosomal Recessive Primary Microcephaly: Not Just a Small Brain. Front Cell Dev Biol 2022; 9:784700. [PMID: 35111754 PMCID: PMC8802810 DOI: 10.3389/fcell.2021.784700] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/01/2021] [Indexed: 02/06/2023] Open
Abstract
Microcephaly or reduced head circumference results from a multitude of abnormal developmental processes affecting brain growth and/or leading to brain atrophy. Autosomal recessive primary microcephaly (MCPH) is the prototype of isolated primary (congenital) microcephaly, affecting predominantly the cerebral cortex. For MCPH, an accelerating number of mutated genes emerge annually, and they are involved in crucial steps of neurogenesis. In this review article, we provide a deeper look into the microcephalic MCPH brain. We explore cytoarchitecture focusing on the cerebral cortex and discuss diverse processes occurring at the level of neural progenitors, early generated and mature neurons, and glial cells. We aim to thereby give an overview of current knowledge in MCPH phenotype and normal brain growth.
Collapse
Affiliation(s)
- Sami Zaqout
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Angela M. Kaindl
- Institute of Cell and Neurobiology, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Center for Chronically Sick Children (Sozialpädiatrisches Zentrum, SPZ), Charité—Universitätsmedizin Berlin, Berlin, Germany
- Department of Pediatric Neurology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
18
|
Kristofova M, Ori A, Wang ZQ. Multifaceted Microcephaly-Related Gene MCPH1. Cells 2022; 11:cells11020275. [PMID: 35053391 PMCID: PMC8774270 DOI: 10.3390/cells11020275] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/19/2022] Open
Abstract
MCPH1, or BRIT1, is often mutated in human primary microcephaly type 1, a neurodevelopmental disorder characterized by a smaller brain size at birth, due to its dysfunction in regulating the proliferation and self-renewal of neuroprogenitor cells. In the last 20 years or so, genetic and cellular studies have identified MCPH1 as a multifaceted protein in various cellular functions, including DNA damage signaling and repair, the regulation of chromosome condensation, cell-cycle progression, centrosome activity and the metabolism. Yet, genetic and animal model studies have revealed an unpredicted essential function of MPCH1 in gonad development and tumorigenesis, although the underlying mechanism remains elusive. These studies have begun to shed light on the role of MPCH1 in controlling various pathobiological processes of the disorder. Here, we summarize the biological functions of MCPH1, and lessons learnt from cellular and mouse models of MCPH1.
Collapse
Affiliation(s)
- Martina Kristofova
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena, Germany; (M.K.); (A.O.)
| | - Alessandro Ori
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena, Germany; (M.K.); (A.O.)
| | - Zhao-Qi Wang
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena, Germany; (M.K.); (A.O.)
- Faculty of Biological Sciences, Friedrich-Schiller University of Jena, Bachstrasse 18k, 07743 Jena, Germany
- Correspondence: ; Tel.: +49-3641-656415; Fax: +49-3641-656335
| |
Collapse
|
19
|
Abstract
In this review, Phan et al. discuss the different models that have been proposed to explain how centrosome dysfunction impairs cortical development, and review the evidence supporting a unified model in which centrosome defects reduce cell proliferation in the developing cortex by prolonging mitosis and activating a mitotic surveillance pathway. Last, they also extend their discussion to centrosome-independent microcephaly mutations, such as those involved in DNA replication and repair Primary microcephaly is a brain growth disorder characterized by a severe reduction of brain size and thinning of the cerebral cortex. Many primary microcephaly mutations occur in genes that encode centrosome proteins, highlighting an important role for centrosomes in cortical development. Centrosomes are microtubule organizing centers that participate in several processes, including controlling polarity, catalyzing spindle assembly in mitosis, and building primary cilia. Understanding which of these processes are altered and how these disruptions contribute to microcephaly pathogenesis is a central unresolved question. In this review, we revisit the different models that have been proposed to explain how centrosome dysfunction impairs cortical development. We review the evidence supporting a unified model in which centrosome defects reduce cell proliferation in the developing cortex by prolonging mitosis and activating a mitotic surveillance pathway. Finally, we also extend our discussion to centrosome-independent microcephaly mutations, such as those involved in DNA replication and repair.
Collapse
|
20
|
Iegiani G, Di Cunto F, Pallavicini G. Inhibiting microcephaly genes as alternative to microtubule targeting agents to treat brain tumors. Cell Death Dis 2021; 12:956. [PMID: 34663805 PMCID: PMC8523548 DOI: 10.1038/s41419-021-04259-6] [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: 07/01/2021] [Revised: 09/10/2021] [Accepted: 09/24/2021] [Indexed: 01/14/2023]
Abstract
Medulloblastoma (MB) and gliomas are the most frequent high-grade brain tumors (HGBT) in children and adulthood, respectively. The general treatment for these tumors consists in surgery, followed by radiotherapy and chemotherapy. Despite the improvement in patient survival, these therapies are only partially effective, and many patients still die. In the last decades, microtubules have emerged as interesting molecular targets for HGBT, as various microtubule targeting agents (MTAs) have been developed and tested pre-clinically and clinically with encouraging results. Nevertheless, these treatments produce relevant side effects since they target microtubules in normal as well as in cancerous cells. A possible strategy to overcome this toxicity could be to target proteins that control microtubule dynamics but are required by HGBT cells much more than in normal cell types. The genes mutated in primary hereditary microcephaly (MCPH) are ubiquitously expressed in proliferating cells, but under normal conditions are selectively required during brain development, in neural progenitors. There is evidence that MB and glioma cells share molecular profiles with progenitors of cerebellar granules and of cortical radial glia cells, in which MCPH gene functions are fundamental. Moreover, several studies indicate that MCPH genes are required for HGBT expansion. Among the 25 known MCPH genes, we focus this review on KNL1, ASPM, CENPE, CITK and KIF14, which have been found to control microtubule stability during cell division. We summarize the current knowledge about the molecular basis of their interaction with microtubules. Moreover, we will discuss data that suggest these genes are promising candidates as HGBT-specific targets.
Collapse
Affiliation(s)
- Giorgia Iegiani
- Neuroscience Institute Cavalieri Ottolenghi, 10043, Orbassano, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, 10126, Turin, Italy
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, 10043, Orbassano, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, 10126, Turin, Italy
| | - Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, 10043, Orbassano, Italy.
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, 10126, Turin, Italy.
| |
Collapse
|
21
|
McNeely KC, Dwyer ND. Cytokinetic Abscission Regulation in Neural Stem Cells and Tissue Development. CURRENT STEM CELL REPORTS 2021; 7:161-173. [PMID: 36303610 PMCID: PMC9603694 DOI: 10.1007/s40778-021-00193-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Purpose of Review How stem cells balance proliferation with differentiation, giving rise to specific daughter cells during development to build an embryo or tissue, remains an open question. Here, we discuss recent evidence that cytokinetic abscission regulation in stem cells, particularly neural stem cells (NSCs), is part of the answer. Abscission is a multi-step process mediated by the midbody, a microtubule-based structure formed in the intercellular bridge between daughter cells after mitosis. Recent Findings Human mutations and mouse knockouts in abscission genes reveal that subtle disruptions of NSC abscission can cause brain malformations. Experiments in several epithelial systems have shown that midbodies serve as scaffolds for apical junction proteins and are positioned near apical membrane fate determinants. Abscission timing is tightly controlled and developmentally regulated in stem cells, with delayed abscission in early embryos and faster abscission later. Midbody remnants (MBRs) contain over 400 proteins and may influence polarity, fate, and ciliogenesis. Summary As NSCs and other stem cells build tissues, they tightly regulate three aspects of abscission: midbody positioning, duration, and MBR handling. Midbody positioning and remnants establish or maintain cell polarity. MBRs are deposited on the apical membranes of epithelia, can be released or internalized by surrounding cells, and may sequester fate determinants or transfer information between cells. Work in cell lines and simpler systems has shown multiple roles for abscission regulation influencing stem cell polarity, potency, and daughter fates during development. Elucidating how the abscission process influences cell fate and tissue growth is important for our continued understanding of brain development and stem cell biology.
Collapse
|
22
|
Lu W, Dong Y, Cui Q, Wang Y, Yang X, Cai X, Zhang M. High Expression of Citron Kinase Contributes to the Development of Esophageal Squamous Cell Carcinoma. Front Genet 2021; 12:628547. [PMID: 34305997 PMCID: PMC8292831 DOI: 10.3389/fgene.2021.628547] [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: 11/12/2020] [Accepted: 06/14/2021] [Indexed: 01/16/2023] Open
Abstract
Objective This study aimed to investigate the role and potential regulatory mechanism of citron kinase (CIT) in esophageal squamous cell carcinoma (ESCC). Methods Citron kinase (CIT) expression in ESCC tissues was analyzed based on the microarray dataset GSE20347, and CIT expression in ESCC cell lines was analyzed. Eca-109 cells were lentivirally transfected with shRNA-CIT (LV-shCIT) to knock down CIT, followed by investigation of cell proliferation and apoptosis. Nude mouse xenograft experiments were performed to evaluate the tumorigenicity of CIT-knockdown Eca-109 cells. Microarray analysis of Eca-109 cells transfected with LV-shCIT or LV-shNC and subsequent Ingenuity Pathway Analysis (IPA) were performed to identify CIT-related differentially expressed genes (DEGs) and signaling pathways. Furthermore, the expression of key DEGs was validated using the clinical samples of ESCC. Results Citron kinase (CIT) was highly expressed in ESCC tissues and cell lines. Knockdown of CIT suppressed Eca-109 cell proliferation and promoted apoptosis in vitro. Moreover, CIT knockdown significantly reduced tumorigenicity of Eca-109 cells in vivo. Microarray and IPA analysis showed that signaling by the Rho family GTPases pathway was significantly activated, and CIT intrinsically interacted with the protein kinase AMP-activated catalytic subunit alpha 1 (PRKAA1), sequestosome 1 (SQSTM1), and interleukin 6 (IL6). Notably, the expression levels of PRKAA1 and SQSTM1 were upregulated in ESCC tissues, while the IL6 expression was downregulated. Conclusion Our findings confirm that CIT functions as an oncogene in ESCC. CIT may contribute to ESCC development by upregulating PRKAA1 and SQSTM1 as well as downregulating IL6. Citron kinase may serve as a promising therapeutic target for ESCC.
Collapse
Affiliation(s)
- Wenfeng Lu
- Department of Integrative Medicine, Zhongshan Hospital and Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yun Dong
- Department of Integrative Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Cui
- Department of Integrative Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yuhan Wang
- Department of Integrative Medicine, Zhongshan Hospital and Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Xiwen Yang
- Department of Integrative Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyue Cai
- Department of Integrative Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Zhang
- Department of Integrative Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
23
|
Van Battum E, Heitz-Marchaland C, Zagar Y, Fouquet S, Kuner R, Chédotal A. Plexin-B2 controls the timing of differentiation and the motility of cerebellar granule neurons. eLife 2021; 10:60554. [PMID: 34100719 PMCID: PMC8211449 DOI: 10.7554/elife.60554] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 06/07/2021] [Indexed: 11/13/2022] Open
Abstract
Plexin-B2 deletion leads to aberrant lamination of cerebellar granule neurons (CGNs) and Purkinje cells. Although in the cerebellum Plexin-B2 is only expressed by proliferating CGN precursors in the outer external granule layer (oEGL), its function in CGN development is still elusive. Here, we used 3D imaging, in vivo electroporation and live-imaging techniques to study CGN development in novel cerebellum-specific Plxnb2 conditional knockout mice. We show that proliferating CGNs in Plxnb2 mutants not only escape the oEGL and mix with newborn postmitotic CGNs. Furthermore, motility of mitotic precursors and early postmitotic CGNs is altered. Together, this leads to the formation of ectopic patches of CGNs at the cerebellar surface and an intermingling of normally time-stamped parallel fibers in the molecular layer (ML), and aberrant arborization of Purkinje cell dendrites. There results suggest that Plexin-B2 restricts CGN motility and might have a function in cytokinesis.
Collapse
Affiliation(s)
- Eljo Van Battum
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Yvrick Zagar
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Stéphane Fouquet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Rohini Kuner
- Pharmacology Institute, Heidelberg University, Heidelberg, Germany
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
24
|
Pallavicini G, Gai M, Iegiani G, Berto GE, Adrait A, Couté Y, Di Cunto F. Goldberg-Shprintzen syndrome protein KIF1BP is a CITK interactor implicated in cytokinesis. J Cell Sci 2021; 134:jcs250902. [PMID: 34100550 DOI: 10.1242/jcs.250902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 05/04/2021] [Indexed: 12/14/2022] Open
Abstract
Goldberg-Shprintzen disease (GOSHS) is a rare microcephaly syndrome accompanied by intellectual disability, dysmorphic facial features, peripheral neuropathy and Hirschsprung disease. It is associated with recessive mutations in the gene encoding kinesin family member 1-binding protein (KIF1BP, also known as KIFBP). The encoded protein regulates axon microtubules dynamics, kinesin attachment and mitochondrial biogenesis, but it is not clear how its loss could lead to microcephaly. We identified KIF1BP in the interactome of citron kinase (CITK, also known as CIT), a protein produced by the primary hereditary microcephaly 17 (MCPH17) gene. KIF1BP and CITK interact under physiological conditions in mitotic cells. Similar to CITK, KIF1BP is enriched at the midbody ring and is required for cytokinesis. The association between KIF1BP and CITK can be influenced by CITK activity, and the two proteins may antagonize each other for their midbody localization. KIF1BP knockdown decreases microtubule stability, increases KIF23 midbody levels and impairs midbody localization of KIF14, as well as of chromosome passenger complex. These data indicate that KIF1BP is a CITK interactor involved in midbody maturation and abscission, and suggest that cytokinesis failure may contribute to the microcephaly phenotype observed in GOSHS.
Collapse
Affiliation(s)
- Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, Turin 10123, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Turin 10126, Italy
| | - Marta Gai
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin 10126, Italy
| | - Giorgia Iegiani
- Neuroscience Institute Cavalieri Ottolenghi, Turin 10123, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Turin 10126, Italy
| | - Gaia Elena Berto
- Neuroscience Institute Cavalieri Ottolenghi, Turin 10123, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Turin 10126, Italy
| | - Annie Adrait
- Univ. Grenoble Alpes, Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut national de la santé et de la recherche médicale (INSERM), Interdisciplinary Research Institute of Grenoble (IRIG), Laboratoire Biologie à Grande Echelle (BGE), 38000 Grenoble, France
| | - Yohann Couté
- Univ. Grenoble Alpes, Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut national de la santé et de la recherche médicale (INSERM), Interdisciplinary Research Institute of Grenoble (IRIG), Laboratoire Biologie à Grande Echelle (BGE), 38000 Grenoble, France
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, Turin 10123, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Turin 10126, Italy
| |
Collapse
|
25
|
Pervaiz N, Kang H, Bao Y, Abbasi AA. Molecular evolutionary analysis of human primary microcephaly genes. BMC Ecol Evol 2021; 21:76. [PMID: 33941077 PMCID: PMC8091745 DOI: 10.1186/s12862-021-01801-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/22/2021] [Indexed: 12/05/2022] Open
Abstract
Background There has been a rapid increase in the brain size relative to body size during mammalian evolutionary history. In particular, the enlarged and globular brain is the most distinctive anatomical feature of modern humans that set us apart from other extinct and extant primate species. Genetic basis of large brain size in modern humans has largely remained enigmatic. Genes associated with the pathological reduction of brain size (primary microcephaly-MCPH) have the characteristics and functions to be considered ideal candidates to unravel the genetic basis of evolutionary enlargement of human brain size. For instance, the brain size of microcephaly patients is similar to the brain size of Pan troglodyte and the very early hominids like the Sahelanthropus tchadensis and Australopithecus afarensis. Results The present study investigates the molecular evolutionary history of subset of autosomal recessive primary microcephaly (MCPH) genes; CEP135, ZNF335, PHC1, SASS6, CDK6, MFSD2A, CIT, and KIF14 across 48 mammalian species. Codon based substitutions site analysis indicated that ZNF335, SASS6, CIT, and KIF14 have experienced positive selection in eutherian evolutionary history. Estimation of divergent selection pressure revealed that almost all of the MCPH genes analyzed in the present study have maintained their functions throughout the history of placental mammals. Contrary to our expectations, human-specific adoptive evolution was not detected for any of the MCPH genes analyzed in the present study. Conclusion Based on these data it can be inferred that protein-coding sequence of MCPH genes might not be the sole determinant of increase in relative brain size during primate evolutionary history. Supplementary Information The online version contains supplementary material available at 10.1186/s12862-021-01801-0.
Collapse
Affiliation(s)
- Nashaiman Pervaiz
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Hongen Kang
- China National Center for Bioinformation and National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yiming Bao
- China National Center for Bioinformation and National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Amir Ali Abbasi
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan.
| |
Collapse
|
26
|
Siskos N, Stylianopoulou E, Skavdis G, Grigoriou ME. Molecular Genetics of Microcephaly Primary Hereditary: An Overview. Brain Sci 2021; 11:brainsci11050581. [PMID: 33946187 PMCID: PMC8145766 DOI: 10.3390/brainsci11050581] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 11/16/2022] Open
Abstract
MicroCephaly Primary Hereditary (MCPH) is a rare congenital neurodevelopmental disorder characterized by a significant reduction of the occipitofrontal head circumference and mild to moderate mental disability. Patients have small brains, though with overall normal architecture; therefore, studying MCPH can reveal not only the pathological mechanisms leading to this condition, but also the mechanisms operating during normal development. MCPH is genetically heterogeneous, with 27 genes listed so far in the Online Mendelian Inheritance in Man (OMIM) database. In this review, we discuss the role of MCPH proteins and delineate the molecular mechanisms and common pathways in which they participate.
Collapse
|
27
|
Loss of Coiled-Coil Protein Cep55 Impairs Neural Stem Cell Abscission and Results in p53-Dependent Apoptosis in Developing Cortex. J Neurosci 2021; 41:3344-3365. [PMID: 33622776 DOI: 10.1523/jneurosci.1955-20.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/24/2020] [Accepted: 02/13/2021] [Indexed: 12/23/2022] Open
Abstract
To build the brain, embryonic neural stem cells (NSCs) tightly regulate their cell divisions, undergoing a polarized form of cytokinesis that is poorly understood. Cytokinetic abscission is mediated by the midbody to sever the daughter cells at the apical membrane. In cell lines, the coiled-coil protein Cep55 was reported to be required for abscission. Mutations of Cep55 in humans cause a variety of cortical malformations. However, its role in the specialized divisions of NSCs is unclear. Here, we elucidate the roles of Cep55 in abscission and brain development. KO of Cep55 in mice causes abscission defects in neural and non-neural cell types, and postnatal lethality. The brain is disproportionately affected, with severe microcephaly at birth. Quantitative analyses of abscission in fixed and live cortical NSCs show that Cep55 acts to increase the speed and success rate of abscission, by facilitating ESCRT recruitment and timely microtubule disassembly. However, most NSCs complete abscission successfully in the absence of Cep55 Those that fail show a tissue-specific response: binucleate NSCs and neurons elevate p53, but binucleate fibroblasts do not. This leads to massive apoptosis in the brain, but not other tissues. Double KO of both p53 and Cep55 blocks apoptosis but only partially rescues Cep55 -/- brain size. This may be because of the persistent NSC cell division defects and p53-independent premature cell cycle exit. This work adds to emerging evidence that abscission regulation and error tolerance vary by cell type and are especially crucial in neural stem cells as they build the brain.SIGNIFICANCE STATEMENT During brain growth, embryonic neural stem cells (NSCs) must divide many times. In the last step of cell division, the daughter cell severs its connection to the mother stem cell, a process called abscission. The protein Cep55 is thought to be essential for recruiting proteins to the mother-daughter cell connection to complete abscission. We find that Cep55 mutants have very small brains with disturbed structure, but almost normal size bodies. NSC abscission can occur, but it is slower than normal, and failures are increased. Furthermore, NSCs that do fail abscission activate a signal for programmed cell death, whereas non-neural cells do not. Blocking this signal only partly restores brain growth, showing that regulation of abscission is crucial for brain development.
Collapse
|
28
|
Jean F, Stuart A, Tarailo-Graovac M. Dissecting the Genetic and Etiological Causes of Primary Microcephaly. Front Neurol 2020; 11:570830. [PMID: 33178111 PMCID: PMC7593518 DOI: 10.3389/fneur.2020.570830] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/09/2020] [Indexed: 12/20/2022] Open
Abstract
Autosomal recessive primary microcephaly (MCPH; “small head syndrome”) is a rare, heterogeneous disease arising from the decreased production of neurons during brain development. As of August 2020, the Online Mendelian Inheritance in Man (OMIM) database lists 25 genes (involved in molecular processes such as centriole biogenesis, microtubule dynamics, spindle positioning, DNA repair, transcriptional regulation, Wnt signaling, and cell cycle checkpoints) that are implicated in causing MCPH. Many of these 25 genes were only discovered in the last 10 years following advances in exome and genome sequencing that have improved our ability to identify disease-causing variants. Despite these advances, many patients still lack a genetic diagnosis. This demonstrates a need to understand in greater detail the molecular mechanisms and genetics underlying MCPH. Here, we briefly review the molecular functions of each MCPH gene and how their loss disrupts the neurogenesis program, ultimately demonstrating that microcephaly arises from cell cycle dysregulation. We also explore the current issues in the genetic basis and clinical presentation of MCPH as additional avenues of improving gene/variant prioritization. Ultimately, we illustrate that the detailed exploration of the etiology and inheritance of MCPH improves the predictive power in identifying previously unknown MCPH candidates and diagnosing microcephalic patients.
Collapse
Affiliation(s)
- Francesca Jean
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Amanda Stuart
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Maja Tarailo-Graovac
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
29
|
Gabriel E, Ramani A, Altinisik N, Gopalakrishnan J. Human Brain Organoids to Decode Mechanisms of Microcephaly. Front Cell Neurosci 2020; 14:115. [PMID: 32457578 PMCID: PMC7225330 DOI: 10.3389/fncel.2020.00115] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022] Open
Abstract
Brain organoids are stem cell-based self-assembling 3D structures that recapitulate early events of human brain development. Recent improvements with patient-specific 3D brain organoids have begun to elucidate unprecedented details of the defective mechanisms that cause neurodevelopmental disorders of congenital and acquired microcephaly. In particular, brain organoids derived from primary microcephaly patients have uncovered mechanisms that deregulate neural stem cell proliferation, maintenance, and differentiation. Not only did brain organoids reveal unknown aspects of neurogenesis but also have illuminated surprising roles of cellular structures of centrosomes and primary cilia in regulating neurogenesis during brain development. Here, we discuss how brain organoids have started contributing to decoding the complexities of microcephaly, which are unlikely to be identified in the existing non-human models. Finally, we discuss the yet unresolved questions and challenges that can be addressed with the use of brain organoids as in vitro models of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Elke Gabriel
- Laboratory for Centrosome and Cytoskeleton Biology, Institute für Humangenetik, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Anand Ramani
- Laboratory for Centrosome and Cytoskeleton Biology, Institute für Humangenetik, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Nazlican Altinisik
- Laboratory for Centrosome and Cytoskeleton Biology, Institute für Humangenetik, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Jay Gopalakrishnan
- Laboratory for Centrosome and Cytoskeleton Biology, Institute für Humangenetik, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| |
Collapse
|
30
|
McNeely KC, Dwyer ND. Cytokinesis and postabscission midbody remnants are regulated during mammalian brain development. Proc Natl Acad Sci U S A 2020; 117:9584-9593. [PMID: 32273386 PMCID: PMC7197019 DOI: 10.1073/pnas.1919658117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Building a brain of the proper size and structure requires neural stem cells (NSCs) to divide with tight temporal and spatial control to produce different daughter cell types in proper numbers and sequence. Mammalian NSCs in the embryonic cortex must maintain their polarized epithelial structure as they undergo both early proliferative divisions and later neurogenic divisions. To do this, they undergo a polarized form of cytokinesis at the apical membrane that is not well understood. Here, we investigate whether polarized furrowing and abscission in mouse NSCs are regulated differently at earlier and later stages and in a cytokinesis mutant, Kif20b This mutant was previously shown to have microcephaly and elevated apoptosis of NSCs. We developed methods to live image furrow ingression and midbody abscission in NSCs within cortical explants. We find that polarized furrow ingression occurs at a steady rate and completes in ∼15 min at two different ages. However, ingression is slower in a subset of Kif20b mutant NSCs. Abscission is usually observed on both sides of the midbody and takes 65 to 75 min to complete. Surprisingly, abscission is accelerated in the Kif20b mutant NSCs. Postabscission midbody remnants are observed at the apical membranes of daughter cells and are much more abundant in early-stage cortices. After NSC divisions in vitro, midbody remnants are more often retained on the daughter cells of early proliferative divisions. Altogether, these results suggest that regulation of abscission timing and midbody remnants in embryonic NSCs may influence proper brain growth and structure.
Collapse
Affiliation(s)
- Katrina C McNeely
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
- Neuroscience Graduate Program, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Noelle D Dwyer
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908;
| |
Collapse
|
31
|
Tedeschi A, Almagro J, Renshaw MJ, Messal HA, Behrens A, Petronczki M. Cep55 promotes cytokinesis of neural progenitors but is dispensable for most mammalian cell divisions. Nat Commun 2020; 11:1746. [PMID: 32269212 PMCID: PMC7142149 DOI: 10.1038/s41467-020-15359-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 02/27/2020] [Indexed: 12/20/2022] Open
Abstract
In mammalian cell lines, the endosomal sorting complex required for transport (ESCRT)-III mediates abscission, the process that physically separates daughter cells and completes cell division. Cep55 protein is regarded as the master regulator of abscission, because it recruits ESCRT-III to the midbody (MB), the site of abscission. However, the importance of this mechanism in a mammalian organism has never been tested. Here we show that Cep55 is dispensable for mouse embryonic development and adult tissue homeostasis. Cep55-knockout offspring show microcephaly and primary neural progenitors require Cep55 and ESCRT for survival and abscission. However, Cep55 is dispensable for cell division in embryonic or adult tissues. In vitro, division of primary fibroblasts occurs without Cep55 and ESCRT-III at the midbody and is not affected by ESCRT depletion. Our work defines Cep55 as an abscission regulator only in specific tissue contexts and necessitates the re-evaluation of an alternative ESCRT-independent cell division mechanism.
Collapse
Affiliation(s)
- Antonio Tedeschi
- Adult Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
- Cell Division and Aneuploidy Laboratory, Clare Hall Laboratories, Cancer Research UK London Research Institute, London, EN6 3LD, UK.
| | - Jorge Almagro
- Adult Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Matthew J Renshaw
- Advanced Light Microscopy, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Hendrik A Messal
- Adult Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Division of Molecular Pathology, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Axel Behrens
- Adult Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Faculty of Life Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Mark Petronczki
- Cell Division and Aneuploidy Laboratory, Clare Hall Laboratories, Cancer Research UK London Research Institute, London, EN6 3LD, UK
- Boehringer Ingelheim RCV GmbH & Co KG, A-1121, Vienna, Austria
| |
Collapse
|
32
|
Moon HM, Hippenmeyer S, Luo L, Wynshaw-Boris A. LIS1 determines cleavage plane positioning by regulating actomyosin-mediated cell membrane contractility. eLife 2020; 9:51512. [PMID: 32159512 PMCID: PMC7112955 DOI: 10.7554/elife.51512] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 03/10/2020] [Indexed: 11/16/2022] Open
Abstract
Heterozygous loss of human PAFAH1B1 (coding for LIS1) results in the disruption of neurogenesis and neuronal migration via dysregulation of microtubule (MT) stability and dynein motor function/localization that alters mitotic spindle orientation, chromosomal segregation, and nuclear migration. Recently, human- induced pluripotent stem cell (iPSC) models revealed an important role for LIS1 in controlling the length of terminal cell divisions of outer radial glial (oRG) progenitors, suggesting cellular functions of LIS1 in regulating neural progenitor cell (NPC) daughter cell separation. Here, we examined the late mitotic stages NPCs in vivo and mouse embryonic fibroblasts (MEFs) in vitro from Pafah1b1-deficient mutants. Pafah1b1-deficient neocortical NPCs and MEFs similarly exhibited cleavage plane displacement with mislocalization of furrow-associated markers, associated with actomyosin dysfunction and cell membrane hyper-contractility. Thus, it suggests LIS1 acts as a key molecular link connecting MTs/dynein and actomyosin, ensuring that cell membrane contractility is tightly controlled to execute proper daughter cell separation.
Collapse
Affiliation(s)
- Hyang Mi Moon
- Department of Pediatrics, Institute for Human Genetics, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, United States
| | - Simon Hippenmeyer
- Howard Hughes Medical Institute and Department of Biology, Stanford University, Stanford, United States
| | - Liqun Luo
- Howard Hughes Medical Institute and Department of Biology, Stanford University, Stanford, United States
| | - Anthony Wynshaw-Boris
- Department of Pediatrics, Institute for Human Genetics, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, United States.,Department of Genetics and Genome Sciences, Case Western Reserve University, School of Medicine, Cleveland, United States
| |
Collapse
|
33
|
CITK Loss Inhibits Growth of Group 3 and Group 4 Medulloblastoma Cells and Sensitizes Them to DNA-Damaging Agents. Cancers (Basel) 2020; 12:cancers12030542. [PMID: 32111106 PMCID: PMC7139701 DOI: 10.3390/cancers12030542] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/15/2020] [Accepted: 02/23/2020] [Indexed: 02/06/2023] Open
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in children, and it is classified into four biological subgroups: WNT, Sonic Hedgehog (SHH), Group 3 and Group 4. The current treatment is surgery, followed by irradiation and chemotherapy. Unfortunately, these therapies are only partially effective. Citron kinase protein (CITK) has been proposed as a promising target for SHH MB, whose inactivation leads to DNA damage and apoptosis. D283 and D341 cell lines (Group 3/Group 4 MB) were silenced with established siRNA sequences against CITK, to assess the direct effects of its loss. Next, D283, D341, ONS-76 and DAOY cells were treated with ionizing radiation (IR) or cisplatin in combination with CITK knockdown. CITK depletion impaired proliferation and induced cytokinesis failure and apoptosis of G3/G4 MB cell lines. Furthermore, CITK knockdown produced an accumulation of DNA damage, with reduced RAD51 nuclear levels. Association of IR or cisplatin with CITK depletion strongly impaired the growth potential of all tested MB cells. These results indicate that CITK inactivation could prevent the expansion of G3/G4 MB and increase their sensitivity to DNA-damaging agents, by impairing homologous recombination. We suggest that CITK inhibition could be broadly associated with IR and adjuvant therapy in MB treatment.
Collapse
|
34
|
Citron Rho-Interacting Serine/Threonine Kinase Promotes HIF1a-CypA Signaling and Growth of Human Pancreatic Adenocarcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9210891. [PMID: 32185224 PMCID: PMC7060418 DOI: 10.1155/2020/9210891] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/19/2020] [Accepted: 01/23/2020] [Indexed: 01/11/2023]
Abstract
In human pancreatic ductal adenocarcinoma (PDAC), the cyclophilin A (CypA) is overexpressed and promotes the development of PDAC. However, the mechanism underlying cyclophilin A expression remains elusive. Here, we reported that the citron Rho-interacting serine/threonine kinase (CIT) promotes the HIF1a-CypA signaling and growth of PDAC cells. CIT expression was higher in PDAC cells compared with the normal epithelial cells, and clinical data showed that CIT was overexpressed in PDAC tissues and high expression of CIT predicted poor overall and disease-free survival. In PDAC cells, knockdown of CIT expression repressed the rate of proliferation and capacity of colony formation, which were accomplished with an increased percentage of apoptotic cells and cell cycle arrest. The knockdown of CIT in PDAC cells reduced the expression of CypA while overexpression of CIT promoted the expression of CypA. We observed that the effects of CIT on the expression of CypA relied on the transcriptional factor HIF1a, which was previously reported to transcriptionally activate the expression of CypA in PDAC cells. Furthermore, the effects of CIT on apoptosis, cell cycle, proliferation, and colony formation of PDAC cells relied on its role in the regulation of CypA expression. Collectively, our data showed that CIT promoted the activation of HIF1-CypA signaling and enhanced the growth of PDAC cells.
Collapse
|
35
|
Johnson CA, Ghashghaei HT. Sp2 regulates late neurogenic but not early expansive divisions of neural stem cells underlying population growth in the mouse cortex. Development 2020; 147:dev186056. [PMID: 32001437 PMCID: PMC7044455 DOI: 10.1242/dev.186056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/23/2020] [Indexed: 12/18/2022]
Abstract
Cellular and molecular mechanisms underlying the switch from self-amplification of cortical stem cells to neuronal and glial generation are incompletely understood, despite their importance for neural development. Here, we have investigated the role of the transcription factor specificity protein 2 (Sp2) in expansive and neurogenic divisions of the developing cerebral cortex by combining conditional genetic deletion with the mosaic analysis with double markers (MADM) system in mice. We find that loss of Sp2 in progenitors undergoing neurogenic divisions results in prolonged mitosis due to extension of early mitotic stages. This disruption is correlated with depletion of the populations of upper layer neurons in the cortex. In contrast, early cortical neural stem cells proliferate and expand normally in the absence of Sp2. These results indicate a stage-specific requirement for Sp2 in neural stem and progenitor cells, and reveal mechanistic differences between the early expansive and later neurogenic periods of cortical development.This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Caroline A Johnson
- Department of Molecular Biomedical Sciences College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | - H Troy Ghashghaei
- Department of Molecular Biomedical Sciences College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| |
Collapse
|
36
|
The Mitotic Apparatus and Kinetochores in Microcephaly and Neurodevelopmental Diseases. Cells 2019; 9:cells9010049. [PMID: 31878213 PMCID: PMC7016623 DOI: 10.3390/cells9010049] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/18/2019] [Accepted: 12/21/2019] [Indexed: 12/15/2022] Open
Abstract
Regulators of mitotic division, when dysfunctional or expressed in a deregulated manner (over- or underexpressed) in somatic cells, cause chromosome instability, which is a predisposing condition to cancer that is associated with unrestricted proliferation. Genes encoding mitotic regulators are growingly implicated in neurodevelopmental diseases. Here, we briefly summarize existing knowledge on how microcephaly-related mitotic genes operate in the control of chromosome segregation during mitosis in somatic cells, with a special focus on the role of kinetochore factors. Then, we review evidence implicating mitotic apparatus- and kinetochore-resident factors in the origin of congenital microcephaly. We discuss data emerging from these works, which suggest a critical role of correct mitotic division in controlling neuronal cell proliferation and shaping the architecture of the central nervous system.
Collapse
|
37
|
Tran THY, Yang DW, Kim M, Lee DH, Gai M, Di Cunto F, Choi KW, Lim DS. Citron kinase interacts with LATS2 and inhibits its activity by occluding its hydrophobic phosphorylation motif. J Mol Cell Biol 2019; 11:1006-1017. [PMID: 30865227 PMCID: PMC6927243 DOI: 10.1093/jmcb/mjz013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 12/24/2018] [Accepted: 03/04/2019] [Indexed: 01/31/2023] Open
Abstract
The inhibitory effect of large tumor suppressor kinase (LATS1/2) on the activity of the oncoprotein yes-associated protein (YAP) is crucial to maintain tissue homeostasis. Proteomic studies have identified several new regulators of this process. Recently, citron kinase (CIT) was listed as a potential binding candidate of Hippo-related components, suggesting a new connection between CIT and the Hippo pathway. Aside from CIT’s role in cytokinesis, the molecular crosstalk between CIT and the Hippo pathway is largely unknown. Here, we demonstrate a role for CIT as a scaffold protein linking LATS2 and YAP. More importantly, CIT interacts with LATS2 to directly suppress LATS2 phosphorylation at the hydrophobic motif—targeted by MST1, leading to LATS2 inactivation and YAP activation. By studying their genetic interactions, we found that Sticky, the CIT homolog in Drosophila melanogaster, functions with Warts to control Drosophila eye development. Together, our study confirms citron kinase as a novel regulator of the Hippo pathway.
Collapse
Affiliation(s)
- Thi Hai Yen Tran
- Department of Biological Sciences, KAIST 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Dae-Wook Yang
- Department of Biological Sciences, KAIST 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Minchul Kim
- Department of Biological Sciences, KAIST 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Da-Hye Lee
- Department of Biological Sciences, KAIST 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Marta Gai
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Ferdinando Di Cunto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Kwang-Wook Choi
- Department of Biological Sciences, KAIST 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Dae-Sik Lim
- Department of Biological Sciences, KAIST 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| |
Collapse
|
38
|
Bressan RB, Pollard SM. Genome Editing in Human Neural Stem and Progenitor Cells. Results Probl Cell Differ 2019; 66:163-182. [PMID: 30209659 DOI: 10.1007/978-3-319-93485-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024]
Abstract
Experimental tools for precise manipulation of mammalian genomes enable reverse genetic approaches to explore biology and disease. Powerful genome editing technologies built upon designer nucleases, such as CRISPR/Cas9, have recently emerged. Parallel progress has been made in methodologies for the expansion and differentiation of human pluripotent and tissue stem cells. Together these innovations provide a remarkable new toolbox for human cellular genetics and are opening up vast opportunities for discoveries and applications across the breadth of life sciences research. In this chapter, we review the emergence of genome editing technologies and how these are being deployed in studies of human neurobiology, neurological disease, and neuro-oncology. We focus our discussion on CRISPR/Cas9 and its application in studies of human neural stem and progenitor cells.
Collapse
Affiliation(s)
- Raul Bardini Bressan
- MRC Centre for Regenerative Medicine and Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh, UK
| | - Steven M Pollard
- MRC Centre for Regenerative Medicine and Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
39
|
Ansar M, Ullah F, Paracha SA, Adams DJ, Lai A, Pais L, Iwaszkiewicz J, Millan F, Sarwar MT, Agha Z, Shah SF, Qaisar AA, Falconnet E, Zoete V, Ranza E, Makrythanasis P, Santoni FA, Ahmed J, Katsanis N, Walsh C, Davis EE, Antonarakis SE. Bi-allelic Variants in DYNC1I2 Cause Syndromic Microcephaly with Intellectual Disability, Cerebral Malformations, and Dysmorphic Facial Features. Am J Hum Genet 2019; 104:1073-1087. [PMID: 31079899 PMCID: PMC6556908 DOI: 10.1016/j.ajhg.2019.04.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/01/2019] [Indexed: 12/30/2022] Open
Abstract
Cargo transport along the cytoplasmic microtubular network is essential for neuronal function, and cytoplasmic dynein-1 is an established molecular motor that is critical for neurogenesis and homeostasis. We performed whole-exome sequencing, homozygosity mapping, and chromosomal microarray studies in five individuals from three independent pedigrees and identified likely-pathogenic variants in DYNC1I2 (Dynein Cytoplasmic 1 Intermediate Chain 2), encoding a component of the cytoplasmic dynein 1 complex. In a consanguineous Pakistani family with three affected individuals presenting with microcephaly, severe intellectual disability, simplification of cerebral gyration, corpus callosum hypoplasia, and dysmorphic facial features, we identified a homozygous splice donor site variant (GenBank: NM_001378.2:c.607+1G>A). We report two additional individuals who have similar neurodevelopmental deficits and craniofacial features and harbor deleterious variants; one individual bears a c.740A>G (p.Tyr247Cys) change in trans with a 374 kb deletion encompassing DYNC1I2, and an unrelated individual harbors the compound-heterozygous variants c.868C>T (p.Gln290∗) and c.740A>G (p.Tyr247Cys). Zebrafish larvae subjected to CRISPR-Cas9 gene disruption or transient suppression of dync1i2a displayed significantly altered craniofacial patterning with concomitant reduction in head size. We monitored cell death and cell cycle progression in dync1i2a zebrafish models and observed significantly increased apoptosis, likely due to prolonged mitosis caused by abnormal spindle morphology, and this finding offers initial insights into the cellular basis of microcephaly. Additionally, complementation studies in zebrafish demonstrate that p.Tyr247Cys attenuates gene function, consistent with protein structural analysis. Our genetic and functional data indicate that DYNC1I2 dysfunction probably causes an autosomal-recessive microcephaly syndrome and highlight further the critical roles of the dynein-1 complex in neurodevelopment.
Collapse
Affiliation(s)
- Muhammad Ansar
- Department of Genetic Medicine and Development, University of Geneva, 1206 Geneva, Switzerland
| | - Farid Ullah
- Center for Human Disease Modeling, Duke University Medical Center, Durham, NC 27701, USA; Human Molecular Genetics Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering, 38000 Faisalabad, Pakistan; Pakistan Institute of Engineering and Applied Sciences, 45650 Islamabad, Pakistan
| | - Sohail A Paracha
- Institute of Basic Medical Sciences, Khyber Medical University, 25100 Peshawar, Pakistan
| | - Darius J Adams
- Atlantic Health System, Goryeb Children's Hospital, Morristown, NJ 07960, USA
| | - Abbe Lai
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA; Departments of Neurology and Pediatrics, Harvard Medical School, Center for Life Sciences, Blackfan Circle, Boston, MA 02115, USA
| | - Lynn Pais
- Medical and Population Genetics Program, Broad Institute of MIT, Cambridge, MA 02142, USA; Center for Mendelian Genomics, Harvard University, Cambridge, MA 02142, USA
| | - Justyna Iwaszkiewicz
- Swiss Institute of Bioinformatics, Molecular Modeling Group, Batiment Genopode, Unil Sorge, 1015 Lausanne, Switzerland
| | | | - Muhammad T Sarwar
- Institute of Basic Medical Sciences, Khyber Medical University, 25100 Peshawar, Pakistan
| | - Zehra Agha
- Department of Biosciences, COMSATS University, 45500 Islamabad, Pakistan
| | - Sayyed Fahim Shah
- Department of Medicine, KMU Institute of Medical Sciences, 26000 Kohat, Pakistan
| | - Azhar Ali Qaisar
- Radiology Department, Lady Reading Hospital, 25000 Peshawar, Pakistan
| | - Emilie Falconnet
- Department of Genetic Medicine and Development, University of Geneva, 1206 Geneva, Switzerland
| | - Vincent Zoete
- Swiss Institute of Bioinformatics, Molecular Modeling Group, Batiment Genopode, Unil Sorge, 1015 Lausanne, Switzerland; Department of Fundamental Oncology, Lausanne University, Ludwig Institute for Cancer Research, Route de la Corniche 9A, 1066 Epalinges, Switzerland
| | - Emmanuelle Ranza
- Department of Genetic Medicine and Development, University of Geneva, 1206 Geneva, Switzerland; Service of Genetic Medicine, University Hospitals of Geneva, 1205 Geneva, Switzerland
| | - Periklis Makrythanasis
- Department of Genetic Medicine and Development, University of Geneva, 1206 Geneva, Switzerland; Biomedical Research Foundation of the Academy of Athens, 115 27 Athens, Greece
| | - Federico A Santoni
- Department of Genetic Medicine and Development, University of Geneva, 1206 Geneva, Switzerland; Department of Endocrinology Diabetes and Metabolism, University Hospital of Lausanne, 1011 Lausanne, Switzerland
| | - Jawad Ahmed
- Institute of Basic Medical Sciences, Khyber Medical University, 25100 Peshawar, Pakistan
| | - Nicholas Katsanis
- Center for Human Disease Modeling, Duke University Medical Center, Durham, NC 27701, USA
| | - Christopher Walsh
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA; Departments of Neurology and Pediatrics, Harvard Medical School, Center for Life Sciences, Blackfan Circle, Boston, MA 02115, USA; Medical and Population Genetics Program, Broad Institute of MIT, Cambridge, MA 02142, USA; Center for Mendelian Genomics, Harvard University, Cambridge, MA 02142, USA
| | - Erica E Davis
- Center for Human Disease Modeling, Duke University Medical Center, Durham, NC 27701, USA.
| | - Stylianos E Antonarakis
- Department of Genetic Medicine and Development, University of Geneva, 1206 Geneva, Switzerland; Service of Genetic Medicine, University Hospitals of Geneva, 1205 Geneva, Switzerland; iGE3 Institute of Genetics and Genomics of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
40
|
Pallavicini G, Berto GE, Di Cunto F. Precision Revisited: Targeting Microcephaly Kinases in Brain Tumors. Int J Mol Sci 2019; 20:ijms20092098. [PMID: 31035417 PMCID: PMC6539168 DOI: 10.3390/ijms20092098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/24/2019] [Accepted: 04/26/2019] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma multiforme and medulloblastoma are the most frequent high-grade brain tumors in adults and children, respectively. Standard therapies for these cancers are mainly based on surgical resection, radiotherapy, and chemotherapy. However, intrinsic or acquired resistance to treatment occurs almost invariably in the first case, and side effects are unacceptable in the second. Therefore, the development of new, effective drugs is a very important unmet medical need. A critical requirement for developing such agents is to identify druggable targets required for the proliferation or survival of tumor cells, but not of other cell types. Under this perspective, genes mutated in congenital microcephaly represent interesting candidates. Congenital microcephaly comprises a heterogeneous group of disorders in which brain volume is reduced, in the absence or presence of variable syndromic features. Genetic studies have clarified that most microcephaly genes encode ubiquitous proteins involved in mitosis and in maintenance of genomic stability, but the effects of their inactivation are particularly strong in neural progenitors. It is therefore conceivable that the inhibition of the function of these genes may specifically affect the proliferation and survival of brain tumor cells. Microcephaly genes encode for a few kinases, including CITK, PLK4, AKT3, DYRK1A, and TRIO. In this review, we summarize the evidence indicating that the inhibition of these molecules could exert beneficial effects on different aspects of brain cancer treatment.
Collapse
Affiliation(s)
- Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, 10126 Turin, Italy.
- Department of Neurosciences, University of Turin, 10126 Turin, Italy.
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy.
| | - Gaia E Berto
- Neuroscience Institute Cavalieri Ottolenghi, 10126 Turin, Italy.
- Department of Neurosciences, University of Turin, 10126 Turin, Italy.
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, 10126 Turin, Italy.
- Department of Neurosciences, University of Turin, 10126 Turin, Italy.
- Neuroscience Institute of Turin (NIT), 10126 Turin, Italy.
| |
Collapse
|
41
|
Li H, Saucedo-Cuevas L, Yuan L, Ross D, Johansen A, Sands D, Stanley V, Guemez-Gamboa A, Gregor A, Evans T, Chen S, Tan L, Molina H, Sheets N, Shiryaev SA, Terskikh AV, Gladfelter AS, Shresta S, Xu Z, Gleeson JG. Zika Virus Protease Cleavage of Host Protein Septin-2 Mediates Mitotic Defects in Neural Progenitors. Neuron 2019; 101:1089-1098.e4. [PMID: 30713029 PMCID: PMC6690588 DOI: 10.1016/j.neuron.2019.01.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/17/2018] [Accepted: 12/31/2018] [Indexed: 01/06/2023]
Abstract
Zika virus (ZIKV) targets neural progenitor cells in the brain, attenuates cell proliferation, and leads to cell death. Here, we describe a role for the ZIKV protease NS2B-NS3 heterodimer in mediating neurotoxicity through cleavage of a host protein required for neurogenesis. Similar to ZIKV infection, NS2B-NS3 expression led to cytokinesis defects and cell death in a protease activity-dependent fashion. Among binding partners, NS2B-NS3 cleaved Septin-2, a cytoskeletal factor involved in cytokinesis. Cleavage of Septin-2 occurred at residue 306 and forced expression of a non-cleavable Septin-2 restored cytokinesis, suggesting a direct mechanism of ZIKV-induced neural toxicity. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Hongda Li
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Laura Saucedo-Cuevas
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Ling Yuan
- Institute of Genetics & Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Danica Ross
- Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Anide Johansen
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Daniel Sands
- Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Valentina Stanley
- Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Alicia Guemez-Gamboa
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Anne Gregor
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lei Tan
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Henrik Molina
- Proteomics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Nicholas Sheets
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Sergey A Shiryaev
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Alexey V Terskikh
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Amy S Gladfelter
- Department of Biology, Howard Hughes Medical Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sujan Shresta
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Zhiheng Xu
- Institute of Genetics & Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Joseph G Gleeson
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
42
|
Li H, Saucedo-Cuevas L, Yuan L, Ross D, Johansen A, Sands D, Stanley V, Guemez-Gamboa A, Gregor A, Evans T, Chen S, Tan L, Molina H, Sheets N, Shiryaev SA, Terskikh AV, Gladfelter AS, Shresta S, Xu Z, Gleeson JG. Zika Virus Protease Cleavage of Host Protein Septin-2 Mediates Mitotic Defects in Neural Progenitors. Neuron 2019; 101:1089-1098.e4. [PMID: 30713029 PMCID: PMC6690588 DOI: 10.1016/j.neuron.2019.01.010#mmc4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/17/2018] [Accepted: 12/31/2018] [Indexed: 10/03/2024]
Abstract
Zika virus (ZIKV) targets neural progenitor cells in the brain, attenuates cell proliferation, and leads to cell death. Here, we describe a role for the ZIKV protease NS2B-NS3 heterodimer in mediating neurotoxicity through cleavage of a host protein required for neurogenesis. Similar to ZIKV infection, NS2B-NS3 expression led to cytokinesis defects and cell death in a protease activity-dependent fashion. Among binding partners, NS2B-NS3 cleaved Septin-2, a cytoskeletal factor involved in cytokinesis. Cleavage of Septin-2 occurred at residue 306 and forced expression of a non-cleavable Septin-2 restored cytokinesis, suggesting a direct mechanism of ZIKV-induced neural toxicity. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Hongda Li
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Laura Saucedo-Cuevas
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Ling Yuan
- Institute of Genetics & Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Danica Ross
- Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Anide Johansen
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Daniel Sands
- Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Valentina Stanley
- Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Alicia Guemez-Gamboa
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Anne Gregor
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lei Tan
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Henrik Molina
- Proteomics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Nicholas Sheets
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Sergey A Shiryaev
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Alexey V Terskikh
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Amy S Gladfelter
- Department of Biology, Howard Hughes Medical Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sujan Shresta
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Zhiheng Xu
- Institute of Genetics & Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Joseph G Gleeson
- Laboratory for Pediatric Brain Disease, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Rady Children's Institute of Genomic Medicine, Department of Neurosciences, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
43
|
Rasika S, Passemard S, Verloes A, Gressens P, El Ghouzzi V. Golgipathies in Neurodevelopment: A New View of Old Defects. Dev Neurosci 2019; 40:396-416. [PMID: 30878996 DOI: 10.1159/000497035] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/16/2019] [Indexed: 11/19/2022] Open
Abstract
The Golgi apparatus (GA) is involved in a whole spectrum of activities, from lipid biosynthesis and membrane secretion to the posttranslational processing and trafficking of most proteins, the control of mitosis, cell polarity, migration and morphogenesis, and diverse processes such as apoptosis, autophagy, and the stress response. In keeping with its versatility, mutations in GA proteins lead to a number of different disorders, including syndromes with multisystem involvement. Intriguingly, however, > 40% of the GA-related genes known to be associated with disease affect the central or peripheral nervous system, highlighting the critical importance of the GA for neural function. We have previously proposed the term "Golgipathies" in relation to a group of disorders in which mutations in GA proteins or their molecular partners lead to consequences for brain development, in particular postnatal-onset microcephaly (POM), white-matter defects, and intellectual disability (ID). Here, taking into account the broader role of the GA in the nervous system, we refine and enlarge this emerging concept to include other disorders whose symptoms may be indicative of altered neurodevelopmental processes, from neurogenesis to neuronal migration and the secretory function critical for the maturation of postmitotic neurons and myelination.
Collapse
Affiliation(s)
- Sowmyalakshmi Rasika
- NeuroDiderot, INSERM UMR1141, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,AP HP, Hôpital Robert Debré, UF de Génétique Clinique, Paris, France
| | - Sandrine Passemard
- NeuroDiderot, INSERM UMR1141, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,AP HP, Hôpital Robert Debré, UF de Génétique Clinique, Paris, France
| | - Alain Verloes
- NeuroDiderot, INSERM UMR1141, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,AP HP, Hôpital Robert Debré, UF de Génétique Clinique, Paris, France
| | - Pierre Gressens
- NeuroDiderot, INSERM UMR1141, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
| | - Vincent El Ghouzzi
- NeuroDiderot, INSERM UMR1141, Université Paris Diderot, Sorbonne Paris Cité, Paris, France,
| |
Collapse
|
44
|
Reilly ML, Stokman MF, Magry V, Jeanpierre C, Alves M, Paydar M, Hellinga J, Delous M, Pouly D, Failler M, Martinovic J, Loeuillet L, Leroy B, Tantau J, Roume J, Gregory-Evans CY, Shan X, Filges I, Allingham JS, Kwok BH, Saunier S, Giles RH, Benmerah A. Loss-of-function mutations in KIF14 cause severe microcephaly and kidney development defects in humans and zebrafish. Hum Mol Genet 2019; 28:778-795. [PMID: 30388224 PMCID: PMC6381319 DOI: 10.1093/hmg/ddy381] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/23/2018] [Accepted: 10/23/2018] [Indexed: 12/28/2022] Open
Abstract
Mutations in KIF14 have previously been associated with either severe, isolated or syndromic microcephaly with renal hypodysplasia (RHD). Syndromic microcephaly-RHD was strongly reminiscent of clinical ciliopathies, relating to defects of the primary cilium, a signalling organelle present on the surface of many quiescent cells. KIF14 encodes a mitotic kinesin, which plays a key role at the midbody during cytokinesis and has not previously been shown to be involved in cilia-related functions. Here, we analysed four families with fetuses presenting with the syndromic form and harbouring biallelic variants in KIF14. Our functional analyses showed that the identified variants severely impact the activity of KIF14 and likely correspond to loss-of-function mutations. Analysis in human fetal tissues further revealed the accumulation of KIF14-positive midbody remnants in the lumen of ureteric bud tips indicating a shared function of KIF14 during brain and kidney development. Subsequently, analysis of a kif14 mutant zebrafish line showed a conserved role for this mitotic kinesin. Interestingly, ciliopathy-associated phenotypes were also present in mutant embryos, supporting a potential direct or indirect role for KIF14 at cilia. However, our in vitro and in vivo analyses did not provide evidence of a direct role for KIF14 in ciliogenesis and suggested that loss of kif14 causes ciliopathy-like phenotypes through an accumulation of mitotic cells in ciliated tissues. Altogether, our results demonstrate that KIF14 mutations result in a severe syndrome associating microcephaly and RHD through its conserved function in cytokinesis during kidney and brain development.
Collapse
Affiliation(s)
- Madeline Louise Reilly
- Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Imagine Institute, Paris, France
- Paris Diderot University, Department of Life Sciences, Paris, France
| | - Marijn F Stokman
- Department of Genetics, University Medical Center Utrecht, Utrecht University, JE Utrecht, Netherlands
| | - Virginie Magry
- Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Cecile Jeanpierre
- Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Marine Alves
- Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Mohammadjavad Paydar
- Institute for Research in Immunology and Cancer, Département de médecine, Université de Montréal, PO Box 6128, Station Centre-Ville, Montréal, QC, Canada
| | - Jacqueline Hellinga
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Marion Delous
- Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Daniel Pouly
- Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Marion Failler
- Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Jelena Martinovic
- Unit of Fetal Pathology, Antoine Béclère Hospital, AP-HP, Clamart, France
- INSERM U-788, Génétique/Neurogénétique, 94270 Le Kremlin-Bicêtre, France
| | - Laurence Loeuillet
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker–Enfants Malades, AP-HP, Paris, France
| | - Brigitte Leroy
- Service d'Anatomie et de Cytologie Pathologiques, Centre hospitalier intercommunal de Poissy, Saint Germain en Laye, France
| | - Julia Tantau
- Service d'Anatomie et de Cytologie Pathologiques, Centre hospitalier intercommunal de Poissy, Saint Germain en Laye, France
| | - Joelle Roume
- Service de Génétique, Centre hospitalier intercommunal de Poissy, 78100 Saint Germain en Laye, France
| | - Cheryl Y Gregory-Evans
- Department of Ophthalmology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Xianghong Shan
- Department of Ophthalmology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Isabel Filges
- Medical Genetics, Institute of Medical Genetics and Pathology, University Hospital of Basel, University of Basel, Basel, Switzerland
- Department of Clinical Research, University Hospital of Basel, University of Basel, Basel, Switzerland
- Department of Genetics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - John S Allingham
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Benjamin H Kwok
- Institute for Research in Immunology and Cancer, Département de médecine, Université de Montréal, PO Box 6128, Station Centre-Ville, Montréal, QC, Canada
| | - Sophie Saunier
- Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Rachel H Giles
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht University, 3512 JE Utrecht, Netherlands
| | - Alexandre Benmerah
- Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Imagine Institute, Paris, France
| |
Collapse
|
45
|
Bowen ME, Attardi LD. The role of p53 in developmental syndromes. J Mol Cell Biol 2019; 11:200-211. [PMID: 30624728 PMCID: PMC6478128 DOI: 10.1093/jmcb/mjy087] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/22/2018] [Accepted: 01/06/2019] [Indexed: 12/17/2022] Open
Abstract
While it is well appreciated that loss of the p53 tumor suppressor protein promotes cancer, growing evidence indicates that increased p53 activity underlies the developmental defects in a wide range of genetic syndromes. The inherited or de novo mutations that cause these syndromes affect diverse cellular processes, such as ribosome biogenesis, DNA repair, and centriole duplication, and analysis of human patient samples and mouse models demonstrates that disrupting these cellular processes can activate the p53 pathway. Importantly, many of the developmental defects in mouse models of these syndromes can be rescued by loss of p53, indicating that inappropriate p53 activation directly contributes to their pathogenesis. A role for p53 in driving developmental defects is further supported by the observation that mouse strains with broad p53 hyperactivation, due to mutations affecting p53 pathway components, display a host of tissue-specific developmental defects, including hematopoietic, neuronal, craniofacial, cardiovascular, and pigmentation defects. Furthermore, germline activating mutations in TP53 were recently identified in two human patients exhibiting bone marrow failure and other developmental defects. Studies in mice suggest that p53 drives developmental defects by inducing apoptosis, restraining proliferation, or modulating other developmental programs in a cell type-dependent manner. Here, we review the growing body of evidence from mouse models that implicates p53 as a driver of tissue-specific developmental defects in diverse genetic syndromes.
Collapse
Affiliation(s)
- Margot E Bowen
- Division of Radiation and Cancer Biology in the Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura D Attardi
- Division of Radiation and Cancer Biology in the Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
46
|
Comprehensive review on the molecular genetics of autosomal recessive primary microcephaly (MCPH). Genet Res (Camb) 2018; 100:e7. [PMID: 30086807 DOI: 10.1017/s0016672318000046] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Primary microcephaly (MCPH) is an autosomal recessive sporadic neurodevelopmental ailment with a trivial head size characteristic that is below 3-4 standard deviations. MCPH is the smaller upshot of an architecturally normal brain; a significant decrease in size is seen in the cerebral cortex. At birth MCPH presents with non-progressive mental retardation, while secondary microcephaly (onset after birth) presents with and without other syndromic features. MCPH is a neurogenic mitotic syndrome nevertheless pretentious patients demonstrate normal neuronal migration, neuronal apoptosis and neural function. Eighteen MCPH loci (MCPH1-MCPH18) have been mapped to date from various populations around the world and contain the following genes: Microcephalin, WDR62, CDK5RAP2, CASC5, ASPM, CENPJ, STIL, CEP135, CEP152, ZNF335, PHC1, CDK6, CENPE, SASS6, MFSD2A, ANKLE2, CIT and WDFY3, clarifying our understanding about the molecular basis of microcephaly genetic disorder. It has previously been reported that phenotype disease is caused by MCB gene mutations and the causes of this phenotype are disarrangement of positions and organization of chromosomes during the cell cycle as a result of mutated DNA, centriole duplication, neurogenesis, neuronal migration, microtubule dynamics, transcriptional control and the cell cycle checkpoint having some invisible centrosomal process that can manage the number of neurons that are produced by neuronal precursor cells. Furthermore, researchers inform us about the clinical management of families that are suffering from MCPH. Establishment of both molecular understanding and genetic advocating may help to decrease the rate of this ailment. This current review study examines newly identified genes along with previously identified genes involved in autosomal recessive MCPH.
Collapse
|
47
|
Davies T, Kim HX, Romano Spica N, Lesea-Pringle BJ, Dumont J, Shirasu-Hiza M, Canman JC. Cell-intrinsic and -extrinsic mechanisms promote cell-type-specific cytokinetic diversity. eLife 2018; 7:36204. [PMID: 30028292 PMCID: PMC6054530 DOI: 10.7554/elife.36204] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 06/10/2018] [Indexed: 01/05/2023] Open
Abstract
Cytokinesis, the physical division of one cell into two, is powered by constriction of an actomyosin contractile ring. It has long been assumed that all animal cells divide by a similar molecular mechanism, but growing evidence suggests that cytokinetic regulation in individual cell types has more variation than previously realized. In the four-cell Caenorhabditis elegans embryo, each blastomere has a distinct cell fate, specified by conserved pathways. Using fast-acting temperature-sensitive mutants and acute drug treatment, we identified cell-type-specific variation in the cytokinetic requirement for a robust forminCYK-1-dependent filamentous-actin (F-actin) cytoskeleton. In one cell (P2), this cytokinetic variation is cell-intrinsically regulated, whereas in another cell (EMS) this variation is cell-extrinsically regulated, dependent on both SrcSRC-1 signaling and direct contact with its neighbor cell, P2. Thus, both cell-intrinsic and -extrinsic mechanisms control cytokinetic variation in individual cell types and can protect against division failure when the contractile ring is weakened. The successful division of one cell into two is essential for all organisms to live, grow and reproduce. For an animal cell, the nucleus – the compartment containing the genetic material – must divide before the surrounding material. The rest of the cell, called the cytoplasm, physically separates later in a process known as cytokinesis. Cytokinesis in animal cells is driven by the formation of a ring in the middle of the dividing cell. The ring is composed of myosin motor proteins and filaments made of a protein called actin. The movements of the motor proteins along the filaments cause the ring to contract and tighten. This pulls the cell membrane inward and physically pinches the cell into two. For a long time, the mechanism of cytokinesis was assumed to be same across different types of animal cell, but later evidence suggested otherwise. For example, in liver, heat and bone cells, cytokinesis naturally fails during development to create cells with two or more nuclei. If a similar ‘failure’ happened in other cell types, it could lead to diseases such as cancers or blood disorders. This raised the question: what are the molecular mechanisms that allow cytokinesis to happen differently in different cell types? Davies et al. investigated this question using embryos of the worm Caenorhabditis elegans at a stage in their development when they consist of just four cells. The proteins forming the contractile ring in this worm are the same as those in humans. However, in the worm, the contractile ring can easily be damaged using chemical inhibitors or by mutating the genes that encode its proteins. Davies et al. show that when the contractile ring was damaged, two of the four cells in the worm embryo still divided successfully. This result indicates the existence of new mechanisms to divide the cytoplasm that allow division even with a weak contractile ring. In a further experiment, the embryos were dissected to isolate each of the four cells. Davies et al. saw that one of the two dividing cells could still divide on its own, while the other cell could not. This shows that this new method of cytokinesis is regulated both by factors inherent to the dividing cell and by external signals from other cells. Moreover, one of these extrinsic signals was found to be a signaling protein that had previously been implicated in human cancers. Future work will determine if these variations in cytokinesis between the different cell types found in the worm apply to humans too; and, more importantly from a therapeutic standpoint, if these new mechanisms exist in human cancers.
Collapse
Affiliation(s)
- Tim Davies
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States
| | - Han X Kim
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States.,Department of Genetics and Development, Columbia University Medical Center, New York, United States
| | - Natalia Romano Spica
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States
| | - Benjamin J Lesea-Pringle
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States
| | - Julien Dumont
- Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Paris, France
| | - Mimi Shirasu-Hiza
- Department of Genetics and Development, Columbia University Medical Center, New York, United States
| | - Julie C Canman
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States
| |
Collapse
|
48
|
Pallavicini G, Sgrò F, Garello F, Falcone M, Bitonto V, Berto GE, Bianchi FT, Gai M, Chiotto AM, Filippi M, Cutrin JC, Ala U, Terreno E, Turco E, Cunto FD. Inactivation of Citron Kinase Inhibits Medulloblastoma Progression by Inducing Apoptosis and Cell Senescence. Cancer Res 2018; 78:4599-4612. [DOI: 10.1158/0008-5472.can-17-4060] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 05/01/2018] [Accepted: 06/07/2018] [Indexed: 11/16/2022]
|
49
|
Abstract
Primary microcephaly (MCPH, for "microcephaly primary hereditary") is a disorder of brain development that results in a head circumference more than 3 standard deviations below the mean for age and gender. It has a wide variety of causes, including toxic exposures, in utero infections, and metabolic conditions. While the genetic microcephaly syndromes are relatively rare, studying these syndromes can reveal molecular mechanisms that are critical in the regulation of neural progenitor cells, brain size, and human brain evolution. Many of the causative genes for MCPH encode centrosomal proteins involved in centriole biogenesis. However, other MCPH genes fall under different mechanistic categories, notably DNA replication and repair. Recent gene discoveries and functional studies have implicated novel cellular processes, such as cytokinesis, centromere and kinetochore function, transmembrane or intracellular transport, Wnt signaling, and autophagy, as well as the apical polarity complex. Thus, MCPH genes implicate a wide variety of molecular and cellular mechanisms in the regulation of cerebral cortical size during development.
Collapse
Affiliation(s)
- Divya Jayaraman
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Harvard-MIT MD-PhD Program, Harvard Medical School, Boston, Massachusetts 02115, USA.,Current affiliation: Boston Combined Residency Program (Child Neurology), Boston Children's Hospital, Boston, Massachusetts 02115, USA;
| | - Byoung-Il Bae
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06510, USA;
| | - Christopher A Walsh
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA.,Departments of Pediatrics and Neurology, Harvard Medical School, Boston, Massachusetts 02115, USA;
| |
Collapse
|
50
|
Makrythanasis P, Maroofian R, Stray-Pedersen A, Musaev D, Zaki MS, Mahmoud IG, Selim L, Elbadawy A, Jhangiani SN, Coban Akdemir ZH, Gambin T, Sorte HS, Heiberg A, McEvoy-Venneri J, James KN, Stanley V, Belandres D, Guipponi M, Santoni FA, Ahangari N, Tara F, Doosti M, Iwaszkiewicz J, Zoete V, Backe PH, Hamamy H, Gleeson JG, Lupski JR, Karimiani EG, Antonarakis SE. Biallelic variants in KIF14 cause intellectual disability with microcephaly. Eur J Hum Genet 2018; 26:330-339. [PMID: 29343805 PMCID: PMC5839044 DOI: 10.1038/s41431-017-0088-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/31/2017] [Accepted: 11/29/2017] [Indexed: 02/07/2023] Open
Abstract
Kinesin proteins are critical for various cellular functions such as intracellular transport and cell division, and many members of the family have been linked to monogenic disorders and cancer. We report eight individuals with intellectual disability and microcephaly from four unrelated families with parental consanguinity. In the affected individuals of each family, homozygosity for likely pathogenic variants in KIF14 were detected; two loss-of-function (p.Asn83Ilefs*3 and p.Ser1478fs), and two missense substitutions (p.Ser841Phe and p.Gly459Arg). KIF14 is a mitotic motor protein that is required for spindle localization of the mitotic citron rho-interacting kinase, CIT, also mutated in microcephaly. Our results demonstrate the involvement of KIF14 in development and reveal a wide phenotypic variability ranging from fetal lethality to moderate developmental delay and microcephaly.
Collapse
Affiliation(s)
- Periklis Makrythanasis
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Reza Maroofian
- Medical Research, RILD Welcome Wolfson Centre, Exeter Medical School, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
- Genetics and Molecular Cell Sciences Research Centre, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Asbjørg Stray-Pedersen
- Norwegian National Unit for Newborn Screening, Oslo University Hospital, Oslo, Norway
- Department of Pediatrics, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Damir Musaev
- Laboratory for Pediatric Brain Disease, Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, CA, 92093, USA
| | - Maha S Zaki
- Human Genetics and Genome Research Division, Clinical Genetics Department, National Research Centre, Cairo, Egypt
| | - Iman G Mahmoud
- Pediatric Neurology and Neurometabolic Unit, Pediatric Department, Cairo University Children Hospital, Cairo, Egypt
| | - Laila Selim
- Pediatric Neurology and Neurometabolic Unit, Pediatric Department, Cairo University Children Hospital, Cairo, Egypt
| | - Amera Elbadawy
- Pediatric Neurology and Neurometabolic Unit, Pediatric Department, Cairo University Children Hospital, Cairo, Egypt
| | - Shalini N Jhangiani
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Human Genome Sequencing Center of Baylor College of Medicine, Houston, TX, 77030, USA
| | - Zeynep H Coban Akdemir
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Tomasz Gambin
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Institute of Computer Science, Warsaw University of Technology, Warsaw, Poland
| | - Hanne S Sorte
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Arvid Heiberg
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Jennifer McEvoy-Venneri
- Laboratory for Pediatric Brain Disease, Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, CA, 92093, USA
| | - Kiely N James
- Laboratory for Pediatric Brain Disease, Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, CA, 92093, USA
| | - Valentina Stanley
- Laboratory for Pediatric Brain Disease, Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, CA, 92093, USA
| | - Denice Belandres
- Laboratory for Pediatric Brain Disease, Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, CA, 92093, USA
| | - Michel Guipponi
- Service of Genetic Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Federico A Santoni
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
- Service of Genetic Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Najmeh Ahangari
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical/Molecular Genetics, Hope Generation Genetic Polyclinic, Mashhad, Iran
| | - Fatemeh Tara
- Women's Health Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Doosti
- Department of Medical/Molecular Genetics, Hope Generation Genetic Polyclinic, Mashhad, Iran
| | - Justyna Iwaszkiewicz
- Molecular Modeling Group, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Vincent Zoete
- Molecular Modeling Group, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Paul Hoff Backe
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
- Department of Medical Biochemistry, University of Oslo, Oslo, Norway
| | - Hanan Hamamy
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Joseph G Gleeson
- Laboratory for Pediatric Brain Disease, Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, CA, 92093, USA
| | - James R Lupski
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Human Genome Sequencing Center of Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ehsan Ghayoor Karimiani
- Razavi Cancer Research Center, Razavi Hospital, Imam Reza International University, Mashhad, Iran
- Innovative Medical Research Center, Faculty of Medicine, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Stylianos E Antonarakis
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland.
- Service of Genetic Medicine, University Hospitals of Geneva, Geneva, Switzerland.
- Institute of Genetics and Genomics of Geneva, Geneva, Switzerland.
| |
Collapse
|