1
|
Necpál J, Schneider SA, Zech M, Jech R. Paradoxical caffeine-responsive paroxysmal nonkinesigenic dyskinesias. Acta Neurol Belg 2025; 125:253-255. [PMID: 39436557 DOI: 10.1007/s13760-024-02666-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024]
Affiliation(s)
- Ján Necpál
- Department of Neurology, Zvolen Hospital, Kuzmányho nábrežie 28, Zvolen, 960 01, Slovakia.
- Parkinsonism and Movement Disorders Treatment Center, Zvolen Hospital, Zvolen, Slovakia.
| | | | - Michael Zech
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Robert Jech
- Department of Neurology, Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| |
Collapse
|
2
|
Chen Y, Jiang Y, Sarvanantharajah N, Apirakkan O, Yang M, Milcova A, Topinka J, Abbate V, Arlt VM, Stürzenbaum SR. Genome-modified Caenorhabditis elegans expressing the human cytochrome P450 (CYP1A1 and CYP1A2) pathway: An experimental model for environmental carcinogenesis and pharmacological research. ENVIRONMENT INTERNATIONAL 2024; 194:109187. [PMID: 39671827 DOI: 10.1016/j.envint.2024.109187] [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: 09/28/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/15/2024]
Abstract
Polycyclic aromatic hydrocarbons (PAHs), including the Group 1 human carcinogen benzo[a]pyrene (BaP), are produced by the incomplete combustion of organic matter and thus are present in tobacco smoke, charbroiled food and diesel exhaust. The nematode Caenorhabditis elegans is an established model organism, however it lacks the genetic components of the classical mammalian cytochrome P450 (CYP)-mediated BaP-diol-epoxide metabolism pathway. We therefore introduced human CYP1A1 or CYP1A2 together with human epoxide hydrolase (EPHX) into the worm genome by Mos1-mediated Single Copy Insertion (MosSCI) and evaluated their response to BaP exposure via toxicological endpoints. Compared to wild-type control, CYP-humanised worms were characterised by an increase in pharyngeal pumping rate and a decrease in volumetric surface area. Furthermore, BaP exposure reduced reproductive performance, as reflected in smaller brood size, which coincided with the downregulation of the nematode-specific major sperm protein as determined by transcriptomics (RNAseq). BaP-mediated reproductive toxicity was exacerbated in CYP-humanised worms at higher exposure levels. Collagen-related genes were downregulated in BaP-exposed animals, which correlate with the reduction in volumetric size. Whole genome DNA sequencing revealed a higher frequency of T > G (A > C) base substitution mutations in worms expressing human CYP1A1;EPHX which aligned with an increase in DNA adducts identified via an ELISA method (but not classical 32P-postlabelling). Overall, the CYP-humanised worms provided new insights into the value of genome-optimised invertebrate models by identifying the benefits and limitations within the context of the (3Rs) concept which aims to replace, reduce and refine the use of animals in research.
Collapse
Affiliation(s)
- Yuzhi Chen
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Yang Jiang
- Hubrecht Institute, Developmental Biology and Stem Cell Research, Utrecht, Netherlands
| | - Nirujah Sarvanantharajah
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Orapan Apirakkan
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Mengqi Yang
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Alena Milcova
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic
| | - Jan Topinka
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic
| | - Vincenzo Abbate
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Volker M Arlt
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK; Toxicology Department, GAB Consulting GmbH, 69126 Heidelberg, Germany
| | - Stephen R Stürzenbaum
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK.
| |
Collapse
|
3
|
Solis GP, Koval A, Valnohova J, Kazemzadeh A, Savitsky M, Katanaev VL. Neomorphic Gαo mutations gain interaction with Ric8 proteins in GNAO1 encephalopathies. J Clin Invest 2024; 134:e172057. [PMID: 38874642 PMCID: PMC11291268 DOI: 10.1172/jci172057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 06/04/2024] [Indexed: 06/15/2024] Open
Abstract
GNAO1 mutated in pediatric encephalopathies encodes the major neuronal G protein Gαo. Of the more than 80 pathogenic mutations, most are single amino acid substitutions spreading across the Gαo sequence. We performed extensive characterization of Gαo mutants, showing abnormal GTP uptake and hydrolysis and deficiencies in binding Gβγ and RGS19. Plasma membrane localization of Gαo was decreased for a subset of mutations that leads to epilepsy; dominant interactions with GPCRs also emerged for the more severe mutants. Pathogenic mutants massively gained interaction with Ric8A and, surprisingly, Ric8B proteins, relocalizing them from cytoplasm to Golgi. Of these 2 mandatory Gα-subunit chaperones, Ric8A is normally responsible for the Gαi/Gαo, Gαq, and Gα12/Gα13 subfamilies, and Ric8B solely responsible for Gαs/Gαolf. Ric8 mediates the disease dominance when engaging in neomorphic interactions with pathogenic Gαo through imbalance of the neuronal G protein signaling networks. As the strength of Gαo-Ric8B interactions correlates with disease severity, our study further identifies an efficient biomarker and predictor for clinical manifestations in GNAO1 encephalopathies. Our work uncovers the neomorphic molecular mechanism of mutations underlying pediatric encephalopathies and offers insights into other maladies caused by G protein malfunctioning and further genetic diseases.
Collapse
Affiliation(s)
- Gonzalo P. Solis
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Alexey Koval
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jana Valnohova
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Arghavan Kazemzadeh
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Mikhail Savitsky
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Vladimir L. Katanaev
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- School of Medicine and Life Sciences, Department of Pharmacy and Pharmacology, Far Eastern Federal University, Vladivostok, Russia
| |
Collapse
|
4
|
Lanza E, Lucente V, Nicoletti M, Schwartz S, Cavallo IF, Caprini D, Connor CW, Saifuddin MFA, Miller JM, L’Etoile ND, Folli V. See Elegans: Simple-to-use, accurate, and automatic 3D detection of neural activity from densely packed neurons. PLoS One 2024; 19:e0300628. [PMID: 38517838 PMCID: PMC10959381 DOI: 10.1371/journal.pone.0300628] [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: 10/13/2023] [Accepted: 02/29/2024] [Indexed: 03/24/2024] Open
Abstract
In the emerging field of whole-brain imaging at single-cell resolution, which represents one of the new frontiers to investigate the link between brain activity and behavior, the nematode Caenorhabditis elegans offers one of the most characterized models for systems neuroscience. Whole-brain recordings consist of 3D time series of volumes that need to be processed to obtain neuronal traces. Current solutions for this task are either computationally demanding or limited to specific acquisition setups. Here, we propose See Elegans, a direct programming algorithm that combines different techniques for automatic neuron segmentation and tracking without the need for the RFP channel, and we compare it with other available algorithms. While outperforming them in most cases, our solution offers a novel method to guide the identification of a subset of head neurons based on position and activity. The built-in interface allows the user to follow and manually curate each of the processing steps. See Elegans is thus a simple-to-use interface aimed at speeding up the post-processing of volumetric calcium imaging recordings while maintaining a high level of accuracy and low computational demands. (Contact: enrico.lanza@iit.it).
Collapse
Affiliation(s)
- Enrico Lanza
- Center for Life Nano- and Neuro-Science@Sapienza, Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Valeria Lucente
- Center for Life Nano- and Neuro-Science@Sapienza, Istituto Italiano di Tecnologia (IIT), Rome, Italy
- D-tails s.r.l., Rome, Italy
| | - Martina Nicoletti
- Center for Life Nano- and Neuro-Science@Sapienza, Istituto Italiano di Tecnologia (IIT), Rome, Italy
- Department of Engineering, Campus Bio-Medico University, Rome, Italy
| | - Silvia Schwartz
- Center for Life Nano- and Neuro-Science@Sapienza, Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Ilaria F. Cavallo
- Center for Life Nano- and Neuro-Science@Sapienza, Istituto Italiano di Tecnologia (IIT), Rome, Italy
- D-tails s.r.l., Rome, Italy
| | - Davide Caprini
- Center for Life Nano- and Neuro-Science@Sapienza, Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Christopher W. Connor
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Mashel Fatema A. Saifuddin
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States of America
| | - Julia M. Miller
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States of America
| | - Noelle D. L’Etoile
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States of America
| | - Viola Folli
- Center for Life Nano- and Neuro-Science@Sapienza, Istituto Italiano di Tecnologia (IIT), Rome, Italy
- D-tails s.r.l., Rome, Italy
| |
Collapse
|
5
|
Benedetti MC, D'andrea T, Colantoni A, Silachev D, de Turris V, Boussadia Z, Babenko VA, Volovikov EA, Belikova L, Bogomazova AN, Pepponi R, Whye D, Buttermore ED, Tartaglia GG, Lagarkova MA, Katanaev VL, Musayev I, Martinelli S, Fucile S, Rosa A. Cortical neurons obtained from patient-derived iPSCs with GNAO1 p.G203R variant show altered differentiation and functional properties. Heliyon 2024; 10:e26656. [PMID: 38434323 PMCID: PMC10907651 DOI: 10.1016/j.heliyon.2024.e26656] [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: 08/03/2023] [Revised: 01/24/2024] [Accepted: 02/16/2024] [Indexed: 03/05/2024] Open
Abstract
Pathogenic variants in the GNAO1 gene, encoding the alpha subunit of an inhibitory heterotrimeric guanine nucleotide-binding protein (Go) highly expressed in the mammalian brain, have been linked to encephalopathy characterized by different combinations of neurological symptoms, including developmental delay, hypotonia, epilepsy and hyperkinetic movement disorder with life-threatening paroxysmal exacerbations. Currently, there are only symptomatic treatments, and little is known about the pathophysiology of GNAO1-related disorders. Here, we report the characterization of a new in vitro model system based on patient-derived induced pluripotent stem cells (hiPSCs) carrying the recurrent p.G203R amino acid substitution in Gαo, and a CRISPR-Cas9-genetically corrected isogenic control line. RNA-Seq analysis highlighted aberrant cell fate commitment in neuronal progenitor cells carrying the p.G203R pathogenic variant. Upon differentiation into cortical neurons, patients' cells showed reduced expression of early neural genes and increased expression of astrocyte markers, as well as premature and defective differentiation processes leading to aberrant formation of neuronal rosettes. Of note, comparable defects in gene expression and in the morphology of neural rosettes were observed in hiPSCs from an unrelated individual harboring the same GNAO1 variant. Functional characterization showed lower basal intracellular free calcium concentration ([Ca2+]i), reduced frequency of spontaneous activity, and a smaller response to several neurotransmitters in 40- and 50-days differentiated p.G203R neurons compared to control cells. These findings suggest that the GNAO1 pathogenic variant causes a neurodevelopmental phenotype characterized by aberrant differentiation of both neuronal and glial populations leading to a significant alteration of neuronal communication and signal transduction.
Collapse
Affiliation(s)
- Maria Cristina Benedetti
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Tiziano D'andrea
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Alessio Colantoni
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Denis Silachev
- School of Medicine and Life Sciences, Far Eastern Federal University, 690090, Vladivostok, Russia
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Moscow State University, 119992, Moscow, Russia
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Translational Research Center in Oncohaematology, University of Geneva, 1211, Geneva, Switzerland
| | - Valeria de Turris
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Zaira Boussadia
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Valentina A. Babenko
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Moscow State University, 119992, Moscow, Russia
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Translational Research Center in Oncohaematology, University of Geneva, 1211, Geneva, Switzerland
| | - Egor A. Volovikov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
| | - Lilia Belikova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
| | - Alexandra N. Bogomazova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
| | - Rita Pepponi
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Dosh Whye
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center and F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, MA, USA
| | - Elizabeth D. Buttermore
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center and F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, MA, USA
| | - Gian Gaetano Tartaglia
- Center for Human Technologies (CHT), Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Maria A. Lagarkova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
| | - Vladimir L. Katanaev
- School of Medicine and Life Sciences, Far Eastern Federal University, 690090, Vladivostok, Russia
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Translational Research Center in Oncohaematology, University of Geneva, 1211, Geneva, Switzerland
| | | | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Sergio Fucile
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Alessandro Rosa
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| |
Collapse
|
6
|
Solis GP, Larasati YA, Thiel M, Koval A, Koy A, Katanaev VL. GNAO1 Mutations Affecting the N-Terminal α-Helix of Gαo Lead to Parkinsonism. Mov Disord 2024; 39:601-606. [PMID: 38358016 DOI: 10.1002/mds.29720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/07/2023] [Accepted: 01/02/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Patients carrying pathogenic variants in GNAO1 present a phenotypic spectrum ranging from severe early-onset epileptic encephalopathy and developmental delay to mild adolescent/adult-onset dystonia. Genotype-phenotype correlation and molecular mechanisms underlying the disease remain understudied. METHODS We analyzed the clinical course of a child carrying the novel GNAO1 mutation c.38T>C;p.Leu13Pro, and structural, biochemical, and cellular properties of the corresponding mutant Gαo-GNAO1-encoded protein-alongside the related mutation c.68T>C;p.Leu23Pro. RESULTS The main clinical feature was parkinsonism with bradykinesia and rigidity, unlike the hyperkinetic movement disorder commonly associated with GNAO1 mutations. The Leu ➔ Pro substitutions have no impact on enzymatic activity or overall folding of Gαo but uniquely destabilize the N-terminal α-helix, blocking formation of the heterotrimeric G-protein and disabling activation by G-protein-coupled receptors. CONCLUSIONS Our study defines a parkinsonism phenotype within the spectrum of GNAO1 disorders and suggests a genotype-phenotype correlation by GNAO1 mutations targeting the N-terminal α-helix of Gαo. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Gonzalo P Solis
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Yonika A Larasati
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Moritz Thiel
- Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Alexey Koval
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Anne Koy
- Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Rare Diseases, Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Vladimir L Katanaev
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- School of Medicine and Life Sciences, Department of Pharmacy and Pharmacology, Far Eastern Federal University, Vladivostok, Russia
| |
Collapse
|
7
|
Rahimpouresfahani F, Tabatabaei N, Rezai P. High-throughput light sheet imaging of adult and larval C. elegans Parkinson's disease model using a low-cost optofluidic device and a fluorescent microscope. RSC Adv 2024; 14:626-639. [PMID: 38173569 PMCID: PMC10759043 DOI: 10.1039/d3ra06323b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
Recent advancements at the interface of microfluidics technology and light sheet fluorescence microscopy have opened the door for high-throughput and high-content investigation of C. elegans disease models. In this paper, we report on the development of a simple, miniaturized, and low-cost optofluidic platform that can be added to a conventional inverted fluorescent microscope for continuous light sheet imaging of transgenic worm populations with high lateral and axial resolutions of 1.1 µm and 2.4 µm, respectively. The optofluidic device is made entirely of PDMS with integrated optics for light sheet generation. Laser excitation is delivered to the device via a low-cost free space laser, and cross-sections of worm populations are imaged as they pass continuously through a channel. Results show the platform can image NW1229 whole worms with pan-neural fluorescent expression at a throughput of >20 worms per minute at L3 and young adult (YA) stages. As a benchmark test, we show that the low-cost device can quantify the reduced neuronal expressions of L3 and YA NW1229 worms when exposed to 500 µM 6-OHDA neurodegenerative agent. Following the benchmark validation, we utilized the platform in a novel application for imaging human alpha-synuclein reporter in populations of Parkinson's transgenic model (ERS100). Results show the ability of the low-cost platform to reliably detect and quantify the anomalous neural phenotypic changes in ERS100 populations at L3 and YA stages with high spatial resolution. The findings of this study show the potential of our low-cost optofluidic add-on platform to equip conventional fluorescent microscopes with light sheet capability for quantitative phenotypic studies of transgenic C. elegans at high resolution and throughput.
Collapse
Affiliation(s)
- Faraz Rahimpouresfahani
- Department of Mechanical Engineering, York University 4700 Keele St Toronto M3J 1P3 Ontario Canada +1-416-7362100
| | - Nima Tabatabaei
- Department of Mechanical Engineering, York University 4700 Keele St Toronto M3J 1P3 Ontario Canada +1-416-7362100
| | - Pouya Rezai
- Department of Mechanical Engineering, York University 4700 Keele St Toronto M3J 1P3 Ontario Canada +1-416-7362100
| |
Collapse
|
8
|
Catalano F, O’Brien TJ, Mekhova AA, Sepe LV, Elia M, De Cegli R, Gallotta I, Santonicola P, Zampi G, Ilyechova EY, Romanov AA, Samuseva PD, Salzano J, Petruzzelli R, Polishchuk EV, Indrieri A, Kim BE, Brown AEX, Puchkova LV, Di Schiavi E, Polishchuk RS. A new Caenorhabditis elegans model to study copper toxicity in Wilson disease. Traffic 2024; 25:e12920. [PMID: 37886910 PMCID: PMC10841361 DOI: 10.1111/tra.12920] [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: 07/27/2023] [Revised: 09/28/2023] [Accepted: 10/03/2023] [Indexed: 10/28/2023]
Abstract
Wilson disease (WD) is caused by mutations in the ATP7B gene that encodes a copper (Cu) transporting ATPase whose trafficking from the Golgi to endo-lysosomal compartments drives sequestration of excess Cu and its further excretion from hepatocytes into the bile. Loss of ATP7B function leads to toxic Cu overload in the liver and subsequently in the brain, causing fatal hepatic and neurological abnormalities. The limitations of existing WD therapies call for the development of new therapeutic approaches, which require an amenable animal model system for screening and validation of drugs and molecular targets. To achieve this objective, we generated a mutant Caenorhabditis elegans strain with a substitution of a conserved histidine (H828Q) in the ATP7B ortholog cua-1 corresponding to the most common ATP7B variant (H1069Q) that causes WD. cua-1 mutant animals exhibited very poor resistance to Cu compared to the wild-type strain. This manifested in a strong delay in larval development, a shorter lifespan, impaired motility, oxidative stress pathway activation, and mitochondrial damage. In addition, morphological analysis revealed several neuronal abnormalities in cua-1 mutant animals exposed to Cu. Further investigation suggested that mutant CUA-1 is retained and degraded in the endoplasmic reticulum, similarly to human ATP7B-H1069Q. As a consequence, the mutant protein does not allow animals to counteract Cu toxicity. Notably, pharmacological correctors of ATP7B-H1069Q reduced Cu toxicity in cua-1 mutants indicating that similar pathogenic molecular pathways might be activated by the H/Q substitution and, therefore, targeted for rescue of ATP7B/CUA-1 function. Taken together, our findings suggest that the newly generated cua-1 mutant strain represents an excellent model for Cu toxicity studies in WD.
Collapse
Affiliation(s)
- Federico Catalano
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Institute of Biosciences and BioResources (IBBR), National Research Council (CNR), Napoli, Italy
| | - Thomas J O’Brien
- MRC London Institute of Medical Sciences, London, United Kingdom
- Institute of Clinical Sciences, Imperial College London, London, United Kingdom
| | - Aleksandra A Mekhova
- Research center of advanced functional materials and laser communication systems, ADTS Institute, ITMO University, St. Petersburg, Russia
| | | | | | - Rossella De Cegli
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Ivan Gallotta
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso (IGB-ABT), National Research Council (CNR), Napoli, Italy
| | - Pamela Santonicola
- Institute of Biosciences and BioResources (IBBR), National Research Council (CNR), Napoli, Italy
| | - Giuseppina Zampi
- Institute of Biosciences and BioResources (IBBR), National Research Council (CNR), Napoli, Italy
| | - Ekaterina Y Ilyechova
- Research center of advanced functional materials and laser communication systems, ADTS Institute, ITMO University, St. Petersburg, Russia
- Department of Molecular Genetics, Research Institute of Experimental Medicine, St. Petersburg, Russia
| | - Aleksei A Romanov
- Department of applied mathematics, Institute of applied mathematics and mechanics, Peter the Great Polytechnic University, St. Petersburg, Russia
| | - Polina D Samuseva
- Research center of advanced functional materials and laser communication systems, ADTS Institute, ITMO University, St. Petersburg, Russia
| | - Josephine Salzano
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Raffaella Petruzzelli
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Scuola Superiore Meridionale (SSM, School of Advanced Studies), Genomics and Experimental Medicine program, University of Naples Federico II, Naples, Italy
| | - Elena V. Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Institute of Biosciences and BioResources (IBBR), National Research Council (CNR), Napoli, Italy
| | - Alessia Indrieri
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Institute for Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan, Italy
| | - Byung-Eun Kim
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, USA
| | - André EX Brown
- MRC London Institute of Medical Sciences, London, United Kingdom
- Institute of Clinical Sciences, Imperial College London, London, United Kingdom
| | - Ludmila V Puchkova
- Research center of advanced functional materials and laser communication systems, ADTS Institute, ITMO University, St. Petersburg, Russia
| | - Elia Di Schiavi
- Institute of Biosciences and BioResources (IBBR), National Research Council (CNR), Napoli, Italy
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso (IGB-ABT), National Research Council (CNR), Napoli, Italy
| | - Roman S. Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Institute for Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan, Italy
| |
Collapse
|
9
|
Cevik S, Zhao P, Zorluer A, Pir MS, Bian W, Kaplan OI. Matching variants for functional characterization of genetic variants. G3 (BETHESDA, MD.) 2023; 13:jkad227. [PMID: 37933433 PMCID: PMC10700107 DOI: 10.1093/g3journal/jkad227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 09/06/2023] [Indexed: 11/08/2023]
Abstract
Rapid and low-cost sequencing, as well as computer analysis, have facilitated the diagnosis of many genetic diseases, resulting in a substantial rise in the number of disease-associated genes. However, genetic diagnosis of many disorders remains problematic due to the lack of interpretation for many genetic variants, especially missenses, the infeasibility of high-throughput experiments on mammals, and the shortcomings of computational prediction technologies. Additionally, the available mutant databases are not well-utilized. Toward this end, we used Caenorhabditis elegans mutant resources to delineate the functions of eight missense variants (V444I, V517D, E610K, L732F, E817K, H873P, R1105K, and G1205E) and two stop codons (W937stop and Q1434stop), including several matching variants (MatchVar) with human in ciliopathy associated IFT-140 (also called CHE-11)//IFT140 (intraflagellar transport protein 140). Moreover, MatchVars carrying C. elegans mutants, including IFT-140(G680S) and IFT-140(P702A) for the human (G704S) (dbSNP: rs150745099) and P726A (dbSNP: rs1057518064 and a conflicting variation) were created using CRISPR/Cas9. IFT140 is a key component of IFT complex A (IFT-A), which is involved in the retrograde transport of IFT along cilia and the entrance of G protein-coupled receptors into cilia. Functional analysis of all 10 variants revealed that P702A and W937stop, but not others phenocopied the ciliary phenotypes (short cilia, IFT accumulations, mislocalization of membrane proteins, and cilia entry of nonciliary proteins) of the IFT-140 null mutant, indicating that both P702A and W937stop are phenotypic in C. elegans. Our functional data offered experimental support for interpreting human variants, by using ready-to-use mutants carrying MatchVars and generating MatchVars with CRISPR/Cas9.
Collapse
Affiliation(s)
- Sebiha Cevik
- Rare Disease Laboratory, School of Life and Natural Sciences, Abdullah Gul University, Kayseri 38080, Turkey
| | - Pei Zhao
- School of Applied Science and Engineering, Fuzhou Institute of Technology, Fuzhou 350014, China
- SunyBiotech Co., Ltd., Fuzhou 35000, China
| | - Atiyye Zorluer
- Rare Disease Laboratory, School of Life and Natural Sciences, Abdullah Gul University, Kayseri 38080, Turkey
| | - Mustafa S Pir
- Rare Disease Laboratory, School of Life and Natural Sciences, Abdullah Gul University, Kayseri 38080, Turkey
| | | | - Oktay I Kaplan
- Rare Disease Laboratory, School of Life and Natural Sciences, Abdullah Gul University, Kayseri 38080, Turkey
| |
Collapse
|
10
|
Galosi S, Novelli M, Di Rocco M, Flex E, Messina E, Pollini L, Parrini E, Pisani F, Guerrini R, Leuzzi V, Martinelli S. GNAO1 Haploinsufficiency: The Milder End of the GNAO1 Phenotypic Spectrum. Mov Disord 2023; 38:2313-2314. [PMID: 37632268 DOI: 10.1002/mds.29585] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Affiliation(s)
- Serena Galosi
- Department of Human Neuroscience, "Sapienza" University of Rome, Rome, Italy
| | - Maria Novelli
- Department of Human Neuroscience, "Sapienza" University of Rome, Rome, Italy
| | - Martina Di Rocco
- Department of Human Neuroscience, "Sapienza" University of Rome, Rome, Italy
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Elisabetta Flex
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Elena Messina
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Luca Pollini
- Department of Human Neuroscience, "Sapienza" University of Rome, Rome, Italy
| | - Elena Parrini
- Neuroscience Department, Children's Hospital Meyer IRCCS, Florence, Italy
| | - Francesco Pisani
- Department of Human Neuroscience, "Sapienza" University of Rome, Rome, Italy
| | - Renzo Guerrini
- Neuroscience Department, Children's Hospital Meyer IRCCS, Florence, Italy
| | - Vincenzo Leuzzi
- Department of Human Neuroscience, "Sapienza" University of Rome, Rome, Italy
| | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
11
|
Zea Vera A, DiSabella M, Tochen L, Meltzer M, Gropman A. Awakening-Related Bouts of Severe Opisthotonos in GNAO1. Mov Disord Clin Pract 2023; 10:1698-1699. [PMID: 37982118 PMCID: PMC10654818 DOI: 10.1002/mdc3.13873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/25/2023] [Accepted: 08/24/2023] [Indexed: 11/21/2023] Open
Affiliation(s)
- Alonso Zea Vera
- Department of NeurologyChildren's National HospitalWashingtonDistrict of ColumbiaUSA
- Department of NeurologyGeorge Washington University School of Medicine & Health SciencesWashingtonDistrict of ColumbiaUSA
| | - Marc DiSabella
- Department of NeurologyChildren's National HospitalWashingtonDistrict of ColumbiaUSA
- Department of NeurologyGeorge Washington University School of Medicine & Health SciencesWashingtonDistrict of ColumbiaUSA
| | - Laura Tochen
- Department of NeurologyChildren's National HospitalWashingtonDistrict of ColumbiaUSA
- Department of NeurologyGeorge Washington University School of Medicine & Health SciencesWashingtonDistrict of ColumbiaUSA
| | - Meira Meltzer
- Department of Neurogenetics and NeurodevelopmentalChildren's National HospitalWashingtonDistrict of ColumbiaUSA
| | - Andrea Gropman
- Department of Neurogenetics and NeurodevelopmentalChildren's National HospitalWashingtonDistrict of ColumbiaUSA
| |
Collapse
|
12
|
Domínguez-Carral J, Ludlam WG, Segarra MJ, Marti MF, Balsells S, Muchart J, Petrović DČ, Espinoza I, Ortigoza-Escobar JD, Martemyanov KA. Severity of GNAO1-Related Disorder Correlates with Changes in G-Protein Function. Ann Neurol 2023; 94:987-1004. [PMID: 37548038 PMCID: PMC10681096 DOI: 10.1002/ana.26758] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023]
Abstract
OBJECTIVE GNAO1-related disorders (OMIM #615473 and #617493), caused by variants in the GNAO1 gene, are characterized by developmental delay or intellectual disability, hypotonia, movement disorders, and epilepsy. Neither a genotype-phenotype correlation nor a clear severity score have been established for this disorder. The objective of this prospective and retrospective observational study was to develop a severity score for GNAO1-related disorders, and to delineate the correlation between the underlying molecular mechanisms and clinical severity. METHODS A total of 16 individuals with GNAO1-related disorders harboring 12 distinct missense variants, including four novel variants (p.K46R, p.T48I, p.R209P, and p.L235P), were examined with repeated clinical assessments, video-electroencephalogram monitoring, and brain magnetic resonance imaging. The molecular pathology of each variant was delineated using a molecular deconvoluting platform. RESULTS The patients displayed a wide variability in the severity of their symptoms. This heterogeneity was well represented in the GNAO1-related disorders severity score, with a broad range of results. Patients with the same variant had comparable severity scores, indicating that differences in disease profiles are not due to interpatient variability, but rather, to unique disease mechanisms. Moreover, we found a significant correlation between clinical severity scores and molecular mechanisms. INTERPRETATION The clinical score proposed here provides further insight into the correlation between pathophysiology and phenotypic severity in GNAO1-related disorders. We found that each variant has a unique profile of clinical phenotypes and pathological molecular mechanisms. These findings will contribute to better understanding GNAO1-related disorders. Additionally, the severity score will facilitate standardization of patients categorization and assessment of response to therapies in development. ANN NEUROL 2023;94:987-1004.
Collapse
Affiliation(s)
- Jana Domínguez-Carral
- Epilepsy Unit, Department of Child Neurology, Institut de
Recerca Sant Joan de Déu, Barcelona, Spain
| | - William Grant Ludlam
- Department of Neuroscience, The Herbert Wertheim UF
Scripps Institute for Biomedical Innovation & Technology, University of Florida,
Jupiter, FL 33458, USA
| | | | | | - Sol Balsells
- Department of Statistics Institut de Recerca Sant Joan de
Déu Barcelona Spain
| | - Jordi Muchart
- Department of Pediatric Radiology, Hospital Sant Joan de
Déu, Barcelona, Spain
| | | | - Iván Espinoza
- Pediatric Neurology Department, Hospital Nacional Cayetano
Heredia, Lima, Perú
| | | | - Juan Dario Ortigoza-Escobar
- Movement Disorders Unit, Department of Child Neurology,
Institut de Recerca Sant Joan de Déu
- U-703 Centre for Biomedical Research on Rare Diseases
(CIBER-ER), Instituto de Salud Carlos III, 08002 Barcelona, Spain
- European Reference Network for Rare Neurological
Diseases (ERN-RND), Barcelona, Spain
| | - Kirill A. Martemyanov
- Department of Neuroscience, The Herbert Wertheim UF
Scripps Institute for Biomedical Innovation & Technology, University of Florida,
Jupiter, FL 33458, USA
| |
Collapse
|
13
|
Larasati YA, Solis GP, Koval A, Griffiths ST, Berentsen R, Aukrust I, Lesca G, Chatron N, Ville D, Korff CM, Katanaev VL. Clinical Cases and the Molecular Profiling of a Novel Childhood Encephalopathy-Causing GNAO1 Mutation P170R. Cells 2023; 12:2469. [PMID: 37887313 PMCID: PMC10605901 DOI: 10.3390/cells12202469] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/29/2023] [Accepted: 10/12/2023] [Indexed: 10/28/2023] Open
Abstract
De novo mutations in GNAO1, the gene encoding the major neuronal G protein Gαo, cause a spectrum of pediatric encephalopathies with seizures, motor dysfunction, and developmental delay. Of the >80 distinct missense pathogenic variants, many appear to uniformly destabilize the guanine nucleotide handling of the mutant protein, speeding up GTP uptake and deactivating GTP hydrolysis. Zinc supplementation emerges as a promising treatment option for this disease, as Zn2+ ions reactivate the GTP hydrolysis on the mutant Gαo and restore cellular interactions for some of the mutants studied earlier. The molecular etiology of GNAO1 encephalopathies needs further elucidation as a prerequisite for the development of efficient therapeutic approaches. In this work, we combine clinical and medical genetics analysis of a novel GNAO1 mutation with an in-depth molecular dissection of the resultant protein variant. We identify two unrelated patients from Norway and France with a previously unknown mutation in GNAO1, c.509C>G that results in the production of the Pro170Arg mutant Gαo, leading to severe developmental and epileptic encephalopathy. Molecular investigations of Pro170Arg identify this mutant as a unique representative of the pathogenic variants. Its 100-fold-accelerated GTP uptake is not accompanied by a loss in GTP hydrolysis; Zn2+ ions induce a previously unseen effect on the mutant, forcing it to lose the bound GTP. Our work combining clinical and molecular analyses discovers a novel, biochemically distinct pathogenic missense variant of GNAO1 laying the ground for personalized treatment development.
Collapse
Affiliation(s)
- Yonika A. Larasati
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland (G.P.S.); (A.K.)
| | - Gonzalo P. Solis
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland (G.P.S.); (A.K.)
| | - Alexey Koval
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland (G.P.S.); (A.K.)
| | - Silja T. Griffiths
- Department of Pediatrics, Haukeland University Hospital, 5009 Bergen, Norway
| | - Ragnhild Berentsen
- Department of Medical Genetics, Haukeland University Hospital, 5009 Bergen, Norway; (R.B.)
| | - Ingvild Aukrust
- Department of Medical Genetics, Haukeland University Hospital, 5009 Bergen, Norway; (R.B.)
- Department of Clinical Science, University of Bergen, 5008 Bergen, Norway
| | - Gaetan Lesca
- Department of Medical Genetics, University Hospital of Lyon, 69002 Lyon, France; (G.L.); (N.C.)
| | - Nicolas Chatron
- Department of Medical Genetics, University Hospital of Lyon, 69002 Lyon, France; (G.L.); (N.C.)
| | - Dorothée Ville
- Pediatric Neurology Department, University Hospital of Lyon, 69002 Lyon, France;
| | - Christian M. Korff
- Pediatric Neurology Unit, University Hospitals of Geneva, CH-1211 Geneva, Switzerland;
| | - Vladimir L. Katanaev
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland (G.P.S.); (A.K.)
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok 690090, Russia
| |
Collapse
|
14
|
Leuzzi V, Galosi S. Experimental pharmacology: Targeting metabolic pathways. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 169:259-315. [PMID: 37482395 DOI: 10.1016/bs.irn.2023.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Since the discovery of the treatment for Wilson disease a growing number of treatable inherited dystonias have been identified and their search and treatment have progressively been implemented in the clinics of patients with dystonia. While waiting for gene therapy to be more widely and adequately translated into the clinical setting, the efforts to divert the natural course of dystonia reside in unveiling its pathogenesis. Specific metabolic treatments can rewrite the natural history of the disease by preventing neurotoxic metabolite accumulation or interfering with the cell accumulation of damaging metabolites, restoring energetic cell fuel, supplementing defective metabolites, and supplementing the defective enzyme. A metabolic derangement of cell homeostasis is part of the progression of many non-metabolic genetic lesions and could be the target for possible metabolic approaches. In this chapter, we provided an update on treatment strategies for treatable inherited dystonias and an overview of genetic dystonias with new experimental therapeutic approaches available or close to clinical translation.
Collapse
Affiliation(s)
- Vincenzo Leuzzi
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | - Serena Galosi
- Department of Human Neuroscience, Sapienza University, Rome, Italy.
| |
Collapse
|
15
|
Pannone L, Muto V, Nardecchia F, Di Rocco M, Marchei E, Tosato F, Petrini S, Onorato G, Lanza E, Bertuccini L, Manti F, Folli V, Galosi S, Di Schiavi E, Leuzzi V, Tartaglia M, Martinelli S. The recurrent pathogenic Pro890Leu substitution in CLTC causes a generalized defect in synaptic transmission in Caenorhabditis elegans. Front Mol Neurosci 2023; 16:1170061. [PMID: 37324589 PMCID: PMC10264582 DOI: 10.3389/fnmol.2023.1170061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/09/2023] [Indexed: 06/17/2023] Open
Abstract
De novo CLTC mutations underlie a spectrum of early-onset neurodevelopmental phenotypes having developmental delay/intellectual disability (ID), epilepsy, and movement disorders (MD) as major clinical features. CLTC encodes the widely expressed heavy polypeptide of clathrin, a major component of the coated vesicles mediating endocytosis, intracellular trafficking, and synaptic vesicle recycling. The underlying pathogenic mechanism is largely unknown. Here, we assessed the functional impact of the recurrent c.2669C > T (p.P890L) substitution, which is associated with a relatively mild ID/MD phenotype. Primary fibroblasts endogenously expressing the mutated protein show reduced transferrin uptake compared to fibroblast lines obtained from three unrelated healthy donors, suggesting defective clathrin-mediated endocytosis. In vitro studies also reveal a block in cell cycle transition from G0/G1 to the S phase in patient's cells compared to control cells. To demonstrate the causative role of the p.P890L substitution, the pathogenic missense change was introduced at the orthologous position of the Caenorhabditis elegans gene, chc-1 (p.P892L), via CRISPR/Cas9. The resulting homozygous gene-edited strain displays resistance to aldicarb and hypersensitivity to PTZ, indicating defective release of acetylcholine and GABA by ventral cord motor neurons. Consistently, mutant animals show synaptic vesicle depletion at the sublateral nerve cords, and slightly defective dopamine signaling, highlighting a generalized deficit in synaptic transmission. This defective release of neurotransmitters is associated with their secondary accumulation at the presynaptic membrane. Automated analysis of C. elegans locomotion indicates that chc-1 mutants move slower than their isogenic controls and display defective synaptic plasticity. Phenotypic profiling of chc-1 (+/P892L) heterozygous animals and transgenic overexpression experiments document a mild dominant-negative behavior for the mutant allele. Finally, a more severe phenotype resembling that of chc-1 null mutants is observed in animals harboring the c.3146 T > C substitution (p.L1049P), homologs of the pathogenic c.3140 T > C (p.L1047P) change associated with a severe epileptic phenotype. Overall, our findings provide novel insights into disease mechanisms and genotype-phenotype correlations of CLTC-related disorders.
Collapse
Affiliation(s)
- Luca Pannone
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Valentina Muto
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | | | - Martina Di Rocco
- Department of Human Neuroscience, “Sapienza” University of Rome, Rome, Italy
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Emilia Marchei
- National Centre on Addiction and Doping, Istituto Superiore di Sanità, Rome, Italy
| | - Federica Tosato
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Stefania Petrini
- Confocal Microscopy Core Facility, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Giada Onorato
- Institute of Biosciences and Bioresources, National Research Council, Naples, Italy
- Department of Environmental, Biological and Pharmaceutical Science and Technologies, Università degli Studi della Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Enrico Lanza
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Rome, Italy
- D-Tails s.r.l., Rome, Italy
| | | | - Filippo Manti
- Department of Human Neuroscience, “Sapienza” University of Rome, Rome, Italy
| | - Viola Folli
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Rome, Italy
- D-Tails s.r.l., Rome, Italy
| | - Serena Galosi
- Department of Human Neuroscience, “Sapienza” University of Rome, Rome, Italy
| | - Elia Di Schiavi
- Institute of Biosciences and Bioresources, National Research Council, Naples, Italy
| | - Vincenzo Leuzzi
- Department of Human Neuroscience, “Sapienza” University of Rome, Rome, Italy
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
16
|
Novelli M, Galosi S, Zorzi G, Martinelli S, Capuano A, Nardecchia F, Granata T, Pollini L, Di Rocco M, Marras CE, Nardocci N, Leuzzi V. GNAO1-related movement disorder: An update on phenomenology, clinical course, and response to treatments. Parkinsonism Relat Disord 2023:105405. [PMID: 37142469 DOI: 10.1016/j.parkreldis.2023.105405] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/09/2023] [Accepted: 04/16/2023] [Indexed: 05/06/2023]
Abstract
AIM To evaluate clinical phenotype and molecular findings of 157 cases with GNAO1 pathogenic or likely pathogenic variants delineating the clinical spectrum, course, and response to treatments. METHOD Clinical phenotype, genetic data, and pharmacological and surgical treatment history of 11 novel cases and 146 previously published patients were analyzed. RESULTS Complex hyperkinetic movement disorder (MD) characterizes 88% of GNAO1 patients. Severe hypotonia and prominent disturbance of postural control seem to be hallmarks in the early stages preceding the hyperkinetic MD. In a subgroup of patients, paroxysmal exacerbations became so severe as to require admission to intensive care units (ICU). Almost all patients had a good response to deep brain stimulation (DBS). Milder phenotypes with late-onset focal/segmental dystonia, mild to moderate intellectual disability, and other minor neurological signs (i.e., parkinsonism and myoclonus) are emerging. MRI, previously considered noncontributory to a diagnosis, can show recurrent findings (i.e., cerebral atrophy, myelination and/or basal ganglia abnormalities). Fifty-eight GNAO1 pathogenic variants, including missense changes and a few recurrent splice site defects, have been reported. Substitutions at residues Gly203, Arg209 and Glu246, together with the intronic c.724-8G > A change, account for more than 50% of cases. INTERPRETATION Infantile or childhood-onset complex hyperkinetic MD (chorea and/or dystonia) with or without paroxysmal exacerbations, associated hypotonia, and developmental disorders should prompt research for GNAO1 mutations. DBS effectively controls and prevents severe exacerbations and should be considered early in patients with specific GNAO1 variants and refractory MD. Prospective and natural history studies are necessary to define genotype-phenotype correlations further and clarify neurological outcomes.
Collapse
Affiliation(s)
- Maria Novelli
- Department of Human Neuroscience, Sapienza University of Rome, Italy
| | - Serena Galosi
- Department of Human Neuroscience, Sapienza University of Rome, Italy.
| | - Giovanna Zorzi
- Department of Pediatric Neuroscience, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy
| | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Tiziana Granata
- Department of Pediatric Neuroscience, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy
| | - Luca Pollini
- Department of Human Neuroscience, Sapienza University of Rome, Italy
| | - Martina Di Rocco
- Department of Human Neuroscience, Sapienza University of Rome, Italy; Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Nardo Nardocci
- Department of Pediatric Neuroscience, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy
| | - Vincenzo Leuzzi
- Department of Human Neuroscience, Sapienza University of Rome, Italy
| |
Collapse
|
17
|
AlAbdi L, Desbois M, Rusnac DV, Sulaiman RA, Rosenfeld JA, Lalani S, Murdock DR, Burrage LC, Billie Au PY, Towner S, Wilson WG, Wong L, Brunet T, Strobl-Wildemann G, Burton JE, Hoganson G, McWalter K, Begtrup A, Zarate YA, Christensen EL, Opperman KJ, Giles AC, Helaby R, Kania A, Zheng N, Grill B, Alkuraya FS. Loss-of-function variants in MYCBP2 cause neurobehavioural phenotypes and corpus callosum defects. Brain 2023; 146:1373-1387. [PMID: 36200388 PMCID: PMC10319777 DOI: 10.1093/brain/awac364] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 08/11/2022] [Accepted: 08/22/2022] [Indexed: 11/14/2022] Open
Abstract
The corpus callosum is a bundle of axon fibres that connects the two hemispheres of the brain. Neurodevelopmental disorders that feature dysgenesis of the corpus callosum as a core phenotype offer a valuable window into pathology derived from abnormal axon development. Here, we describe a cohort of eight patients with a neurodevelopmental disorder characterized by a range of deficits including corpus callosum abnormalities, developmental delay, intellectual disability, epilepsy and autistic features. Each patient harboured a distinct de novo variant in MYCBP2, a gene encoding an atypical really interesting new gene (RING) ubiquitin ligase and signalling hub with evolutionarily conserved functions in axon development. We used CRISPR/Cas9 gene editing to introduce disease-associated variants into conserved residues in the Caenorhabditis elegans MYCBP2 orthologue, RPM-1, and evaluated functional outcomes in vivo. Consistent with variable phenotypes in patients with MYCBP2 variants, C. elegans carrying the corresponding human mutations in rpm-1 displayed axonal and behavioural abnormalities including altered habituation. Furthermore, abnormal axonal accumulation of the autophagy marker LGG-1/LC3 occurred in variants that affect RPM-1 ubiquitin ligase activity. Functional genetic outcomes from anatomical, cell biological and behavioural readouts indicate that MYCBP2 variants are likely to result in loss of function. Collectively, our results from multiple human patients and CRISPR gene editing with an in vivo animal model support a direct link between MYCBP2 and a human neurodevelopmental spectrum disorder that we term, MYCBP2-related developmental delay with corpus callosum defects (MDCD).
Collapse
Affiliation(s)
- Lama AlAbdi
- Department of Zoology, College of Science, King Saud University, Riyadh 11362, Saudi Arabia
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | - Muriel Desbois
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Domniţa-Valeria Rusnac
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Raashda A Sulaiman
- Department of Medical Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Seema Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - David R Murdock
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lindsay C Burrage
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Ping Yee Billie Au
- Department of Medical Genetics, Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Shelley Towner
- Pediatric Genetics, University of Virginia, Charlottesville, VA 22903, USA
| | - William G Wilson
- Pediatric Genetics, University of Virginia, Charlottesville, VA 22903, USA
| | - Lawrence Wong
- Department of Genetics, Northern California Kaiser Permanente, Oakland, CA 94611, USA
| | - Theresa Brunet
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- Institute of Neurogenomics (ING), Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | | | - Jennifer E Burton
- Department of Genetics, University of Illinois College of Medicine at Peoria, Peoria, IL 61605, USA
| | - George Hoganson
- Department of Genetics, University of Illinois College of Medicine at Peoria, Peoria, IL 61605, USA
| | - Kirsty McWalter
- Genedx, Inc., 207 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Amber Begtrup
- Genedx, Inc., 207 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Yuri A Zarate
- Section of Genetics and Metabolism, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Elyse L Christensen
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Karla J Opperman
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Andrew C Giles
- Division of Medical Sciences, University of Northern British Columbia, Prince George, BC V2N 4Z9, Canada
| | - Rana Helaby
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | - Artur Kania
- Institut de recherches cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, QC H3A 2B4, Canada
- Division of Experimental Medicine, McGill University, Montréal, QC H3A 2B2, Canada
- Department of Anatomy and Cell Biology, McGill University, Montréal, QC H3A 0C7, Canada
| | - Ning Zheng
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Brock Grill
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98101, USA
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| |
Collapse
|
18
|
Katanaev VL. Gln52 mutations in GNAO1-related disorders and personalized drug discovery. Epilepsy Behav Rep 2023; 24:100598. [PMID: 38106673 PMCID: PMC10724470 DOI: 10.1016/j.ebr.2023.100598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
•Gln52 mutations have been found in patients with GNAO1-related disorders.•Gln52 can be mutated to Pro and Arg, leading to different clinical manifestations.•Personalized drug discovery is tailored to specific GNAO1 mutations.
Collapse
Affiliation(s)
- Vladimir L. Katanaev
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690090 Vladivostok, Russia
| |
Collapse
|
19
|
Di Rocco M, Galosi S, Follo FC, Lanza E, Folli V, Martire A, Leuzzi V, Martinelli S. Phenotypic Assessment of Pathogenic Variants in GNAO1 and Response to Caffeine in C. elegans Models of the Disease. Genes (Basel) 2023; 14:319. [PMID: 36833246 PMCID: PMC9957173 DOI: 10.3390/genes14020319] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/13/2023] [Accepted: 01/22/2023] [Indexed: 01/28/2023] Open
Abstract
De novo mutations affecting the G protein α o subunit (Gαo)-encoding gene (GNAO1) cause childhood-onset developmental delay, hyperkinetic movement disorders, and epilepsy. Recently, we established Caenorhabditis elegans as an informative experimental model for deciphering pathogenic mechanisms associated with GNAO1 defects and identifying new therapies. In this study, we generated two additional gene-edited strains that harbor pathogenic variants which affect residues Glu246 and Arg209-two mutational hotspots in Gαo. In line with previous findings, biallelic changes displayed a variable hypomorphic effect on Gαo-mediated signaling that led to the excessive release of neurotransmitters by different classes of neurons, which, in turn, caused hyperactive egg laying and locomotion. Of note, heterozygous variants showed a cell-specific dominant-negative behavior, which was strictly dependent on the affected residue. As with previously generated mutants (S47G and A221D), caffeine was effective in attenuating the hyperkinetic behavior of R209H and E246K animals, indicating that its efficacy is mutation-independent. Conversely, istradefylline, a selective adenosine A2A receptor antagonist, was effective in R209H animals but not in E246K worms, suggesting that caffeine acts through both adenosine receptor-dependent and receptor-independent mechanisms. Overall, our findings provide new insights into disease mechanisms and further support the potential efficacy of caffeine in controlling dyskinesia associated with pathogenic GNAO1 mutations.
Collapse
Affiliation(s)
- Martina Di Rocco
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
- Department of Human Neuroscience, ‘Sapienza’ University of Rome, 00185 Rome, Italy
| | - Serena Galosi
- Department of Human Neuroscience, ‘Sapienza’ University of Rome, 00185 Rome, Italy
| | - Francesca C. Follo
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Enrico Lanza
- Center for Life Nano Science, Istituto Italiano di Tecnologia, 00161 Rome, Italy
| | - Viola Folli
- Center for Life Nano Science, Istituto Italiano di Tecnologia, 00161 Rome, Italy
- D-tails s.r.l., 00165 Rome, Italy
| | - Alberto Martire
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Vincenzo Leuzzi
- Department of Human Neuroscience, ‘Sapienza’ University of Rome, 00185 Rome, Italy
| | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| |
Collapse
|
20
|
JoJo Yang QZ, Porter BE, Axeen ET. GNAO1-related neurodevelopmental disorder: Literature review and caregiver survey. Epilepsy Behav Rep 2022; 21:100582. [PMID: 36654732 PMCID: PMC9841045 DOI: 10.1016/j.ebr.2022.100582] [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: 11/01/2021] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/01/2023] Open
Abstract
Background GNAO1-related neurodevelopmental disorder is a heterogeneous condition characterized by hypotonia, developmental delay, epilepsy, and movement disorder. This study aims to better understand the spectrum of epilepsy associated with GNAO1 variants and experience with anti-seizure medications, and to review published epilepsy phenotypes in GNAO1. Methods An online survey was distributed to caregivers of individuals diagnosed with GNAO1 pathogenic variants, and a literature review was conducted. Results Fifteen respondents completed the survey with the median age of 39 months, including a novel variant p.Q52P. Nine had epilepsy - six had onset in the first week of life, three in the first year of life - but two reported no ongoing seizures. Seizure types varied. Individuals were taking a median of 3 seizure medications without a single best treatment. Our cohort was compared to a literature review of epilepsy in GNAO1. In 86 cases, 38 discrete variants were described; epilepsy is reported in 53 % cases, and a developmental and epileptic encephalopathy in 36 %. Conclusions While GNAO1-related epilepsy is most often early-onset and severe, seizures may not always be drug resistant or lifelong. Experience with anti-seizure medications is varied. Certain variant "hotspots" may correlate with epilepsy phenotype though genotype-phenotype correlation is poorly understood.
Collapse
Affiliation(s)
- Qian-Zhou JoJo Yang
- Division of Child Neurology, Department of Neurology, University of North Carolina, Chapel Hill, NC, United States,Corresponding author at: 170 Manning Dr, Campus Box 7025, Chapel Hill, NC 27599, United States
| | - Brenda E Porter
- Division of Child Neurology, Department of Neurology, Stanford University, Palo Alto, CA, United States
| | - Erika T Axeen
- Division of Pediatric Neurology, Department of Neurology, University of Virginia, United States
| |
Collapse
|
21
|
Huang P, Wang Y, Liu SS, Wang ZJ, Xu YQ. SAHmap: Synergistic-antagonistic heatmap to evaluate the combined synergistic effect of mixtures of three pesticides on multiple endpoints of Caenorhabditis elegans. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 315:120378. [PMID: 36220575 DOI: 10.1016/j.envpol.2022.120378] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2023]
Abstract
The environmental pollution caused by toxic chemicals such as pesticides has become a global problem. The mixture of dichlorvos (DIC), dimethoate (DIM), aldicarb (ALD) poses potential risks to the environment and human health. To fully explore the interaction of complex mixtures on Caenorhabditis elegans behavioral toxicity endpoint. This study created a synergistic-antagonistic heatmap (SAHmap) based on the combination index to systematically describe the toxicological interaction prospect of the mixture system. It was shown that the three pesticides and their binary as well as ternary mixture rays have significant concentration-response relationship on three behavioral endpoints of nematodes, From the perspective of synergistic-antagonistic heatmaps, all the mixture rays in the DIC-DIM mixture system showed strong synergism on the three behavioral and lethal endpoints. In the ternary mixture system, the five mixture rays showed different interaction between the behavioral endpoint and the lethal endpoint, and showed slight synergism to two behavioral endpoints as a whole. The emergence of synergism should arouse our attention to these hazardous chemicals. In addition, the use of SAHmap and the significant linear correlation among three behavioral endpoints further improved the efficiency of the study on the behavioral toxicity of pesticide mixtures to Caenorhabditis elegans.
Collapse
Affiliation(s)
- Peng Huang
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China; State Key Laboratory of Pollution Control and Resource Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China
| | - Yu Wang
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China; State Key Laboratory of Pollution Control and Resource Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China
| | - Shu-Shen Liu
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China; State Key Laboratory of Pollution Control and Resource Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai, 200092, PR China.
| | - Ze-Jun Wang
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China; State Key Laboratory of Pollution Control and Resource Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China
| | - Ya-Qian Xu
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China; State Key Laboratory of Pollution Control and Resource Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China
| |
Collapse
|
22
|
Hopkins CE, Brock T, Caulfield TR, Bainbridge M. Phenotypic screening models for rapid diagnosis of genetic variants and discovery of personalized therapeutics. Mol Aspects Med 2022; 91:101153. [PMID: 36411139 PMCID: PMC10073243 DOI: 10.1016/j.mam.2022.101153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/22/2022] [Accepted: 10/23/2022] [Indexed: 11/19/2022]
Abstract
Precision medicine strives for highly individualized treatments for disease under the notion that each individual's unique genetic makeup and environmental exposures imprints upon them not only a disposition to illness, but also an optimal therapeutic approach. In the realm of rare disorders, genetic predisposition is often the predominant mechanism driving disease presentation. For such, mostly, monogenic disorders, a causal gene to phenotype association is likely. As a result, it becomes important to query the patient's genome for the presence of pathogenic variations that are likely to cause the disease. Determining whether a variant is pathogenic or not is critical to these analyses and can be challenging, as many disease-causing variants are novel and, ergo, have no available functional data to help categorize them. This problem is exacerbated by the need for rapid evaluation of pathogenicity, since many genetic diseases present in young children who will experience increased morbidity and mortality without rapid diagnosis and therapeutics. Here, we discuss the utility of animal models, with a focus mainly on C. elegans, as a contrast to tissue culture and in silico approaches, with emphasis on how these systems are used in determining pathogenicity of variants with uncertain significance and then used to screen for novel therapeutics.
Collapse
Affiliation(s)
| | | | - Thomas R Caulfield
- Mayo Clinic, Department of Neuroscience, Department of Computational Biology, Department of Clinical Genomics, Jacksonville, FL, 32224, Rochester, MN, 55905, USA
| | | |
Collapse
|
23
|
Mew M, Caldwell KA, Caldwell GA. From bugs to bedside: functional annotation of human genetic variation for neurological disorders using invertebrate models. Hum Mol Genet 2022; 31:R37-R46. [PMID: 35994032 PMCID: PMC9585664 DOI: 10.1093/hmg/ddac203] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 02/02/2023] Open
Abstract
The exponential accumulation of DNA sequencing data has opened new avenues for discovering the causative roles of single-nucleotide polymorphisms (SNPs) in neurological diseases. The opportunities emerging from this are staggering, yet only as good as our abilities to glean insights from this surplus of information. Whereas computational biology continues to improve with respect to predictions and molecular modeling, the differences between in silico and in vivo analysis remain substantial. Invertebrate in vivo model systems represent technically advanced, experimentally mature, high-throughput, efficient and cost-effective resources for investigating a disease. With a decades-long track record of enabling investigators to discern function from DNA, fly (Drosophila) and worm (Caenorhabditis elegans) models have never been better poised to serve as living engines of discovery. Both of these animals have already proven useful in the classification of genetic variants as either pathogenic or benign across a range of neurodevelopmental and neurodegenerative disorders-including autism spectrum disorders, ciliopathies, amyotrophic lateral sclerosis, Alzheimer's and Parkinson's disease. Pathogenic SNPs typically display distinctive phenotypes in functional assays when compared with null alleles and frequently lead to protein products with gain-of-function or partial loss-of-function properties that contribute to neurological disease pathogenesis. The utility of invertebrates is logically limited by overt differences in anatomical and physiological characteristics, and also the evolutionary distance in genome structure. Nevertheless, functional annotation of disease-SNPs using invertebrate models can expedite the process of assigning cellular and organismal consequences to mutations, ascertain insights into mechanisms of action, and accelerate therapeutic target discovery and drug development for neurological conditions.
Collapse
Affiliation(s)
- Melanie Mew
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Kim A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
- Alabama Research Institute on Aging, The University of Alabama, Tuscaloosa, AL 35487, USA
- Center for Convergent Bioscience and Medicine, The University of Alabama, Tuscaloosa, AL 35487, USA
- Departments of Neurobiology and Neurology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center of Excellence for Research in the Basic Biology of Aging, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Guy A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
- Center for Convergent Bioscience and Medicine, The University of Alabama, Tuscaloosa, AL 35487, USA
- Departments of Neurobiology and Neurology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center of Excellence for Research in the Basic Biology of Aging, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
24
|
Larasati YA, Savitsky M, Koval A, Solis GP, Valnohova J, Katanaev VL. Restoration of the GTPase activity and cellular interactions of Gα o mutants by Zn 2+ in GNAO1 encephalopathy models. SCIENCE ADVANCES 2022; 8:eabn9350. [PMID: 36206333 PMCID: PMC9544338 DOI: 10.1126/sciadv.abn9350] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 08/22/2022] [Indexed: 06/16/2023]
Abstract
De novo point mutations in GNAO1, gene encoding the major neuronal G protein Gαo, have recently emerged in patients with pediatric encephalopathy having motor, developmental, and epileptic dysfunctions. Half of clinical cases affect codons Gly203, Arg209, or Glu246; we show that these mutations accelerate GTP uptake and inactivate GTP hydrolysis through displacement Gln205 critical for GTP hydrolysis, resulting in constitutive GTP binding by Gαo. However, the mutants fail to adopt the activated conformation and display aberrant interactions with signaling partners. Through high-throughput screening of approved drugs, we identify zinc pyrithione and Zn2+ as agents restoring active conformation, GTPase activity, and cellular interactions of the encephalopathy mutants, with negligible effects on wild-type Gαo. We describe a Drosophila model of GNAO1 encephalopathy where dietary zinc restores the motor function and longevity of the mutant flies. Zinc supplements are approved for diverse human neurological conditions. Our work provides insights into the molecular etiology of GNAO1 encephalopathy and defines a potential therapy for the patients.
Collapse
Affiliation(s)
- Yonika A. Larasati
- Translational Research Centre in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Mikhail Savitsky
- Translational Research Centre in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Alexey Koval
- Translational Research Centre in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Gonzalo P. Solis
- Translational Research Centre in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Jana Valnohova
- Translational Research Centre in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Vladimir L. Katanaev
- Translational Research Centre in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690090 Vladivostok, Russia
| |
Collapse
|
25
|
Lasa-Aranzasti A, Cazurro-Gutiérrez A, Bescós A, González V, Ispierto L, Tardáguila M, Valenzuela I, Plaja A, Moreno-Galdó A, Macaya-Ruiz A, Pérez-Dueñas B. 16q12.2q21 deletion: A newly recognized cause of dystonia related to GNAO1 haploinsufficiency. Parkinsonism Relat Disord 2022; 103:112-114. [PMID: 36096018 DOI: 10.1016/j.parkreldis.2022.08.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 11/19/2022]
Affiliation(s)
- Amaia Lasa-Aranzasti
- Department of Clinical and Molecular Genetics, Vall d'Hebron University Hospital, Barcelona, Spain; Pediatric Neurology Research Group, Vall d'Hebron Research Institute (VHIR), Autonomous University of Barcelona, Barcelona, Spain; Medicine Department, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ana Cazurro-Gutiérrez
- Pediatric Neurology Research Group, Vall d'Hebron Research Institute (VHIR), Autonomous University of Barcelona, Barcelona, Spain; Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Agustín Bescós
- Pediatric Neuromodulation Unit, Hospital Vall d'Hebrón and Hospital Germans Trias I Pujol, Barcelona, Spain; Department of Neurosurgery, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Victoria González
- Pediatric Neuromodulation Unit, Hospital Vall d'Hebrón and Hospital Germans Trias I Pujol, Barcelona, Spain; Department of Neurology, Department of Neurology, Vall Hebron University Hospital Barcelona, Spain
| | - Lourdes Ispierto
- Pediatric Neuromodulation Unit, Hospital Vall d'Hebrón and Hospital Germans Trias I Pujol, Barcelona, Spain; Neurodegenerative Diseases Unit, Neurology Service and Neurosciences Department, University Hospital Germans Trias i Pujol, Barcelona, Spain
| | - Manel Tardáguila
- Pediatric Neuromodulation Unit, Hospital Vall d'Hebrón and Hospital Germans Trias I Pujol, Barcelona, Spain; Department of Neurological Surgery, University Hospital Germans Trias i Pujol, Barcelona, Spain
| | - Irene Valenzuela
- Department of Clinical and Molecular Genetics, Vall d'Hebron University Hospital, Barcelona, Spain; Medicine Genetics Group, Vall d'Hebron Research Institute (VHIR), Autonomous University of Barcelona, Barcelona, Spain
| | - Alberto Plaja
- Department of Clinical and Molecular Genetics, Vall d'Hebron University Hospital, Barcelona, Spain; Medicine Genetics Group, Vall d'Hebron Research Institute (VHIR), Autonomous University of Barcelona, Barcelona, Spain
| | - Antonio Moreno-Galdó
- Department of Pediatrics, Universitat Autónoma de Barcelona, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain; CIBER of Rare diseases (CIBERER), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Alfons Macaya-Ruiz
- Department of Pediatrics, Universitat Autónoma de Barcelona, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain; Pediatric Neurology Research Group, Vall d'Hebron Research Institute (VHIR), Autonomous University of Barcelona, Barcelona, Spain
| | - Belen Pérez-Dueñas
- Pediatric Neurology Research Group, Vall d'Hebron Research Institute (VHIR), Autonomous University of Barcelona, Barcelona, Spain; Pediatric Neuromodulation Unit, Hospital Vall d'Hebrón and Hospital Germans Trias I Pujol, Barcelona, Spain; CIBER of Rare diseases (CIBERER), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain.
| |
Collapse
|
26
|
Ling W, Huang D, Yang F, Yang Z, Liu M, Zhu Q, Huang J, Zhou R, Chen X. Treating GNAO1 mutation-related severe movement disorders with oxcarbazepine: a case report. Transl Pediatr 2022; 11:1577-1587. [PMID: 36247896 PMCID: PMC9561508 DOI: 10.21037/tp-22-297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/16/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND GNAO1 variants have been found to be associated with epileptic encephalopathies, developmental delays (DDs), and movement disorders (MDs). Therapies for patients with GNAO1 variants vary. However, treatments for GNAO1-related diseases are still in their infancy. Previous reports suggest that few pharmacological treatments are effective for patients with GNAO1 variant-related MDs. Deep brain stimulation (DBS) treatment appears to be effective, however surgical procedures and equipment failures pose risks to the patients. Effectiveness for oxcarbazepine (OXC) in GNAO1 variant-related MDs is first reported in our study, and it expand the effective drugs for MD treatment. CASE DESCRIPTION We report the case of a 5-year-old male patient with a MD, who suffered from hypotonia and refractory choreoathetosis. The patient was found to have a DD and an intellectual disability. A de-novo variant of the GNAO1 gene (NM_138736: exom6: c.709G>A [p. Glu237Lys]) was identified by whole exome sequencing (WES) when he was 8 months old. The patient visited our hospital at the age of 4 years and 3 months because of fever and recurrent convulsions. Electroencephalogram (EEG) results show abnormal spikes, and magnetic resonance imaging (MRI) showed the enlargement of the lateral ventricles. The administration of tiapride hydrochloride, phenobarbital, midazolam, and hormones had no effect. OXC treatment was then initiated. No MD behaviors, such as rigidity and twisting of the limbs and trunk, or chorea, were observed after 10 days OXC treatment. Eventually, incremental doses of OXC were effective, and our patient achieved good control of his MD. CONCLUSIONS We are the first to demonstrate the role of OXC in alleviating MDs associated with GNAO1 mutations. This report provides a novel possibility for the clinical treatment of this rare disease. To manage MDs associated with GNAO1 mutations, we recommend that OXC treatment be attempted before invasive surgical therapy.
Collapse
Affiliation(s)
- Weihao Ling
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, China
| | - Danping Huang
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, China
| | | | | | - Min Liu
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, China
| | - Qiujiao Zhu
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, China
| | - Jing Huang
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, China
| | - Rui Zhou
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, China
| | - Xuqin Chen
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
27
|
Galosi S, Pollini L, Novelli M, Bernardi K, Di Rocco M, Martinelli S, Leuzzi V. Motor, epileptic, and developmental phenotypes in genetic disorders affecting G protein coupled receptors-cAMP signaling. Front Neurol 2022; 13:886751. [PMID: 36003298 PMCID: PMC9393484 DOI: 10.3389/fneur.2022.886751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Over the last years, a constantly increasing number of genetic diseases associated with epilepsy and movement disorders have been recognized. An emerging group of conditions in this field is represented by genetic disorders affecting G-protein-coupled receptors (GPCRs)–cAMP signaling. This group of postsynaptic disorders includes genes encoding for proteins highly expressed in the central nervous system and involved in GPCR signal transduction and cAMP production (e.g., GNAO1, GNB1, ADCY5, GNAL, PDE2A, PDE10A, and HPCA genes). While the clinical phenotype associated with ADCY5 and GNAL is characterized by movement disorder in the absence of epilepsy, GNAO1, GNB1, PDE2A, PDE10A, and HPCA have a broader clinical phenotype, encompassing movement disorder, epilepsy, and neurodevelopmental disorders. We aimed to provide a comprehensive phenotypical characterization of genetic disorders affecting the cAMP signaling pathway, presenting with both movement disorders and epilepsy. Thus, we reviewed clinical features and genetic data of 203 patients from the literature with GNAO1, GNB1, PDE2A, PDE10A, and HPCA deficiencies. Furthermore, we delineated genotype–phenotype correlation in GNAO1 and GNB1 deficiency. This group of disorders presents with a highly recognizable clinical phenotype combining distinctive motor, epileptic, and neurodevelopmental features. A severe hyperkinetic movement disorder with potential life-threatening exacerbations and high susceptibility to a wide range of triggers is the clinical signature of the whole group of disorders. The existence of a distinctive clinical phenotype prompting diagnostic suspicion and early detection has relevant implications for clinical and therapeutic management. Studies are ongoing to clarify the pathophysiology of these rare postsynaptic disorders and start to design disease-specific treatments.
Collapse
Affiliation(s)
- Serena Galosi
- Department Human Neuroscience, Sapienza University, Rome, Italy
- *Correspondence: Serena Galosi
| | - Luca Pollini
- Department Human Neuroscience, Sapienza University, Rome, Italy
| | - Maria Novelli
- Department Human Neuroscience, Sapienza University, Rome, Italy
| | | | - Martina Di Rocco
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Vincenzo Leuzzi
- Department Human Neuroscience, Sapienza University, Rome, Italy
| |
Collapse
|
28
|
Lange KI, Best S, Tsiropoulou S, Berry I, Johnson CA, Blacque OE. Interpreting ciliopathy-associated missense variants of uncertain significance (VUS) in Caenorhabditis elegans. Hum Mol Genet 2022; 31:1574-1587. [PMID: 34964473 PMCID: PMC9122650 DOI: 10.1093/hmg/ddab344] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 12/26/2022] Open
Abstract
Better methods are required to interpret the pathogenicity of disease-associated variants of uncertain significance (VUS), which cannot be actioned clinically. In this study, we explore the use of an animal model (Caenorhabditis elegans) for in vivo interpretation of missense VUS alleles of TMEM67, a cilia gene associated with ciliopathies. CRISPR/Cas9 gene editing was used to generate homozygous knock-in C. elegans worm strains carrying TMEM67 patient variants engineered into the orthologous gene (mks-3). Quantitative phenotypic assays of sensory cilia structure and function (neuronal dye filling, roaming and chemotaxis assays) measured how the variants impacted mks-3 gene function. Effects of the variants on mks-3 function were further investigated by looking at MKS-3::GFP localization and cilia ultrastructure. The quantitative assays in C. elegans accurately distinguished between known benign (Asp359Glu, Thr360Ala) and known pathogenic (Glu361Ter, Gln376Pro) variants. Analysis of eight missense VUS generated evidence that three are benign (Cys173Arg, Thr176Ile and Gly979Arg) and five are pathogenic (Cys170Tyr, His782Arg, Gly786Glu, His790Arg and Ser961Tyr). Results from worms were validated by a genetic complementation assay in a human TMEM67 knock-out hTERT-RPE1 cell line that tests a TMEM67 signalling function. We conclude that efficient genome editing and quantitative functional assays in C. elegans make it a tractable in vivo animal model for rapid, cost-effective interpretation of ciliopathy-associated missense VUS alleles.
Collapse
Affiliation(s)
- Karen I Lange
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sunayna Best
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, West Yorkshire, UK
| | - Sofia Tsiropoulou
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Ian Berry
- Bristol Genetics Laboratory, Pathology Sciences, Southmead Hospital, Bristol BS10 5NB, UK
| | - Colin A Johnson
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, West Yorkshire, UK
| | - Oliver E Blacque
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
29
|
Méneret A, Mohammad SS, Cif L, Doummar D, DeGusmao C, Anheim M, Barth M, Damier P, Demonceau N, Friedman J, Gallea C, Gras D, Gurgel-Giannetti J, Innes EA, Necpál J, Riant F, Sagnes S, Sarret C, Seliverstov Y, Paramanandam V, Shetty K, Tranchant C, Doulazmi M, Vidailhet M, Pringsheim T, Roze E. Efficacy of Caffeine in ADCY5-Related Dyskinesia: A Retrospective Study. Mov Disord 2022; 37:1294-1298. [PMID: 35384065 DOI: 10.1002/mds.29006] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/08/2022] [Accepted: 03/15/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND ADCY5-related dyskinesia is characterized by early-onset movement disorders. There is currently no validated treatment, but anecdotal clinical reports and biological hypotheses suggest efficacy of caffeine. OBJECTIVE The aim is to obtain further insight into the efficacy and safety of caffeine in patients with ADCY5-related dyskinesia. METHODS A retrospective study was conducted worldwide in 30 patients with a proven ADCY5 mutation who had tried or were taking caffeine for dyskinesia. Disease characteristics and treatment responses were assessed through a questionnaire. RESULTS Caffeine was overall well tolerated, even in children, and 87% of patients reported a clear improvement. Caffeine reduced the frequency and duration of paroxysmal movement disorders but also improved baseline movement disorders and some other motor and nonmotor features, with consistent quality-of-life improvement. Three patients reported worsening. CONCLUSION Our findings suggest that caffeine should be considered as a first-line therapeutic option in ADCY5-related dyskinesia. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Aurélie Méneret
- Inserm U1127, CNRS UMR7225, UM75, Paris Brain Institute, Assistance Publique-Hôpitaux de Paris, DMU Neurosciences, Sorbonne University, Paris, France
| | - Shekeeb S Mohammad
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Laura Cif
- Département de Neurochirurgie, Hôpital Gui de Chauliac, Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - Diane Doummar
- Service de Neuropédiatrie-Pathologie du développement, centre de référence mouvements anormaux enfant, Hôpital Trousseau AP-HP.SU, FHU I2D2, Sorbonne Université, Paris, France
| | | | - Mathieu Anheim
- Service de Neurologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch-Graffenstaden, France.,Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | | | - Philippe Damier
- CHU de Nantes, INSERM, CIC 1314, Hôpital Laennec, Nantes, France
| | | | - Jennifer Friedman
- Departments of Neurosciences and Pediatrics, University of California San Diego, La Jolla, California, USA.,Division of Neurology, Rady Children's Hospital, San Diego, California, USA.,Rady Children's Institute for Genomic Medicine, San Diego, California, USA
| | - Cécile Gallea
- Sorbonne University, INSERM, CNRS, Paris Brain Institute, Paris, France
| | - Domitille Gras
- U1141 Neurodiderot, équipe 5 inDev, Inserm, CEA, UP, UNIACTNeurospin, Joliot, DRF, CEA, Saclay, France
| | | | - Emily A Innes
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia.,University of Notre Dame Australia, School of Medicine, Sydney, NSW, Australia
| | - Ján Necpál
- Department of Neurology, Zvolen Hospital, Zvolen, Slovakia
| | - Florence Riant
- Service de Génétique Moléculaire, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Sandrine Sagnes
- Délégation à la Recherche Clinique et à l'Innovation-DRCI (Clinical Research and Innovation Department) and URC (Clinical Research Unit) GH Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Catherine Sarret
- Service de pédiatrie, hôpital Estaing, Centre hospitalier universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Yury Seliverstov
- Research Center of Neurology, Moscow, Russia.,Kazaryan Clinic of Epileptology and Neurology, Moscow, Russia
| | | | - Kuldeep Shetty
- Department of Neurology, Mazumdar Shaw Medical Center, Bangalore, India
| | - Christine Tranchant
- Service de Neurologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch-Graffenstaden, France.,Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Mohamed Doulazmi
- Adaptation Biologique et Vieillissement, Institut de Biologie Paris Seine, Sorbonne University, CNRS, Paris, France
| | - Marie Vidailhet
- Inserm U1127, CNRS UMR7225, UM75, Paris Brain Institute, Assistance Publique-Hôpitaux de Paris, DMU Neurosciences, Sorbonne University, Paris, France
| | - Tamara Pringsheim
- Department of Clinical Neurosciences, Psychiatry, Pediatrics and Community Health Sciences, University of Calgary, Calgary, Canada
| | - Emmanuel Roze
- Inserm U1127, CNRS UMR7225, UM75, Paris Brain Institute, Assistance Publique-Hôpitaux de Paris, DMU Neurosciences, Sorbonne University, Paris, France
| |
Collapse
|
30
|
An intronic GNAO1 variant leading to in-frame insertion cause movement disorder controlled by deep brain stimulation. Neurogenetics 2022; 23:129-135. [PMID: 35147852 DOI: 10.1007/s10048-022-00686-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/06/2022] [Indexed: 12/27/2022]
Abstract
GNAO1 variants are associated with a wide range of neurodevelopmental disorders including epileptic encephalopathies and movement disorders. It has been reported that some GNAO1 variants are associated with movement disorders, and the 207-246 amino acid region was proposed as a mutational hotspot. Here, we report an intronic variant (NM_020988.3:c.724-8G>A) in GNAO1 in a Japanese girl who showed mild developmental delay and movement disorders including dystonia and myoclonus. Her movement disorders were improved by deep brain stimulation treatment as previously reported. This variant has been recurrently reported in four patients and was transmitted from her mother who possessed the variant as low-prevalent mosaicism. Using RNA extracted from lymphoblastoid cells derived from the patient, we demonstrated that the variant caused abnormal splicing of in-frame 6-bp intronic retention, leading to 2 amino acid insertion (p.Thr241_Asn242insProGln). Immunoblotting and immunostaining using WT and mutant GNAO1 vectors showed no significant differences in protein expression levels, but the cellular localization pattern of this mutant was partially shifted to the cytoplasm whereas WT was exclusively localized in the cellular membrane. Our report first clarified abnormal splicing and resulting mutant protein caused by the c.724-8G>A variant.
Collapse
|
31
|
Silachev D, Koval A, Savitsky M, Padmasola G, Quairiaux C, Thorel F, Katanaev VL. Mouse models characterize GNAO1 encephalopathy as a neurodevelopmental disorder leading to motor anomalies: from a severe G203R to a milder C215Y mutation. Acta Neuropathol Commun 2022; 10:9. [PMID: 35090564 PMCID: PMC8796625 DOI: 10.1186/s40478-022-01312-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/08/2022] [Indexed: 02/07/2023] Open
Abstract
GNAO1 encephalopathy characterized by a wide spectrum of neurological deficiencies in pediatric patients originates from de novo heterozygous mutations in the gene encoding Gαo, the major neuronal G protein. Efficient treatments and even the proper understanding of the underlying etiology are currently lacking for this dominant disease. Adequate animal models of GNAO1 encephalopathy are urgently needed. Here we describe establishment and characterization of mouse models of the disease based on two point mutations in GNAO1 with different clinical manifestations. One of them is G203R leading to the early-onset epileptic seizures, motor dysfunction, developmental delay and intellectual disability. The other is C215Y producing much milder clinical outcomes, mostly-late-onset hyperkinetic movement disorder. The resultant mouse models show distinct phenotypes: severe neonatal lethality in GNAO1[G203R]/ + mice vs. normal vitality in GNAO1[C215Y]/ + . The latter model further revealed strong hyperactivity and hyperlocomotion in a panel of behavioral assays, without signs of epilepsy, recapitulating the patients' manifestations. Importantly, despite these differences the two models similarly revealed prenatal brain developmental anomalies, such as enlarged lateral ventricles and decreased numbers of neuronal precursor cells in the cortex. Thus, our work unveils GNAO1 encephalopathy as to a large extent neurodevelopmental malady. We expect that this understanding and the tools we established will be instrumental for future therapeutic developments.
Collapse
Affiliation(s)
- Denis Silachev
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Moscow State University, 119992, Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow, 117997, Russia
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Translational Research Center in Oncohaematology, University of Geneva, 1211, Geneva, Switzerland
- School of Biomedicine, Far Eastern Federal University, 690090, Vladivostok, Russia
| | - Alexey Koval
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Translational Research Center in Oncohaematology, University of Geneva, 1211, Geneva, Switzerland
| | - Mikhail Savitsky
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Translational Research Center in Oncohaematology, University of Geneva, 1211, Geneva, Switzerland
| | - Guru Padmasola
- Department of Basic Neuroscience, Faculty of Medicine, University of Geneva, 1211, Geneva, Switzerland
| | - Charles Quairiaux
- Department of Basic Neuroscience, Faculty of Medicine, University of Geneva, 1211, Geneva, Switzerland
| | - Fabrizio Thorel
- Transgenesis Core Facility, Faculty of Medicine, University of Geneva, 1211, Geneva, Switzerland
| | - Vladimir L Katanaev
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Translational Research Center in Oncohaematology, University of Geneva, 1211, Geneva, Switzerland.
- School of Biomedicine, Far Eastern Federal University, 690090, Vladivostok, Russia.
| |
Collapse
|