1
|
Höltje M, Wolkowicz A, Brunk I, Baron J, Ahnert‐Hilger G. Gα o1 and Gα o1/Gα o2 deletion differentially affect hippocampal mossy fiber tract anatomy and neuronal morphogenesis. J Neurochem 2025; 169:e16248. [PMID: 39466989 PMCID: PMC11808459 DOI: 10.1111/jnc.16248] [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: 07/24/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/30/2024]
Abstract
The heterotrimeric G-protein αo subunit is ubiquitously expressed in the CNS as two splice variants Gαo1 and Gαo2, regulating various brain functions. Here, we investigated the effect of single Gαo1, Gαo2, and double Gαo1/2 knockout on the postnatal development of the murine mossy fiber tract, a central pathway of the hippocampal connectivity circuit. The size of the hippocampal synaptic termination fields covered by mossy fiber boutons together with various fiber length parameters of the tract was analyzed by immunohistochemical staining of the vesicular Zinc transporter 3 (ZnT3) or Synaptoporin at postnatal days 2, 4, 8, 12, 16, and in the adult. Ultimately, Gαo1 knockout resulted in a reduced developmental growth of synaptic mossy fiber terminal fields by 37% in the adult Stratum lucidum and by 30% in the total mossy fiber tract size. Other morphological parameters such as projection length of the infrapyramidal bundle of the tract were increased (+52% in Gαo1 -/- mice). In contrast, Gαo2 knockout had no effects on the mossy fiber tract. Moreover, by using primary heterozygous and homozygous Gαo1 knockout hippocampal cultures, we detected a strongly pronounced reduction in axon and dendrite length (-50% and -38%, respectively) as well as axon and dendrite arborization complexity (-75% and -72% branch nodes, respectively) in the homozygous knockout. Deletion of both splice variants Gαo1 and Gαo2 partially rescued the in vivo and completely reconstituted the in vitro effects, indicating an opposing functional relevance of the two Gαo splice variants for neuronal development and synaptic connectivity.
Collapse
Affiliation(s)
- Markus Höltje
- Institut für Integrative Neuroanatomie, Charité–Universitätsmedizin BerlinCorporate Member of Freie Universität Berlin and Humboldt‐Universität Zu BerlinBerlinGermany
| | - Anton Wolkowicz
- Institut für Integrative Neuroanatomie, Charité–Universitätsmedizin BerlinCorporate Member of Freie Universität Berlin and Humboldt‐Universität Zu BerlinBerlinGermany
| | - Irene Brunk
- Institut für Integrative Neuroanatomie, Charité–Universitätsmedizin BerlinCorporate Member of Freie Universität Berlin and Humboldt‐Universität Zu BerlinBerlinGermany
| | - Jens Baron
- Institut für Integrative Neuroanatomie, Charité–Universitätsmedizin BerlinCorporate Member of Freie Universität Berlin and Humboldt‐Universität Zu BerlinBerlinGermany
| | - Gudrun Ahnert‐Hilger
- Laboratory of NeurobiologyMax‐Planck‐Institute for Biophysical Chemistry and University of GöttingenGöttingeGermany
| |
Collapse
|
2
|
Choi JM, Acharya R, Cha HL, Lee KW, Seo J, Yang E, Kim H, Yoon JH, Chang DY, Kim SS, Kim SJ, Birnbaumer L, Suh-Kim H. CB1R activates the epilepsy-associated protein Go to regulate neurotransmitter release and synaptic plasticity in the cerebellum. Proc Natl Acad Sci U S A 2024; 121:e2409773121. [PMID: 39602265 PMCID: PMC11626142 DOI: 10.1073/pnas.2409773121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
GNAO1 encodes the alpha subunit of the heterotrimeric Go protein. Despite being the most abundant G protein at synapses, the role of Go in the brain remains unclear, primarily because of the high mortality associated with developmental and epileptic encephalopathy (DEE) 17 in Gnao1 mutated animals. Here, we conducted proteomic analyses with a brain synaptosomal fraction to investigate the Go-interactome and then generated a non-DEE model using Gli1CreERT2 mice to selectively knockout (KO) the presynaptic Gαo within cerebellum. Our findings revealed that Gαo interacts with multiple proteins involved in neurotransmitter release, as well as cannabinoid receptor type 1 (CB1R), a key Gi/o-coupled receptor in presynaptic terminals. In Gnao1 KO mice, synapse formation was reduced in the cerebellum with a concomitant reduction in depolarization-induced suppression of excitation, a manifestation of CB1R-mediated synaptic plasticity found in the cerebellum. These mice displayed motor deficits in rotarod, grip strength, gait, and beam balance tests. Our results suggest that Go plays a critical role in regulating neurotransmitter releases at the presynaptic terminals and its absence in the entire brain may contribute to DEE pathogenesis. This study also provides valuable insights into the signaling pathways in the brain from a Go-dependent perspective.
Collapse
Affiliation(s)
- Jung-Mi Choi
- Department of Anatomy, Ajou University School of Medicine, Suwon16499, South Korea
| | - Rakshya Acharya
- Department of Anatomy, Ajou University School of Medicine, Suwon16499, South Korea
| | - Hye Lim Cha
- Department of Anatomy, Ajou University School of Medicine, Suwon16499, South Korea
| | - Kwang-Wook Lee
- Department of Anatomy, Ajou University School of Medicine, Suwon16499, South Korea
| | - Jewoo Seo
- Department Physiology, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Esther Yang
- Department of Anatomy, College of Medicine, Korea University, Seoul02841, Korea
| | - Hyun Kim
- Department of Anatomy, College of Medicine, Korea University, Seoul02841, Korea
| | - Jong Hyuk Yoon
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu41062, South Korea
| | - Da-Young Chang
- Research Center, CELLeBRAIN Co., Ltd., Suwon16681, South Korea
| | - Sung-Soo Kim
- Department of Anatomy, Ajou University School of Medicine, Suwon16499, South Korea
| | - Sang Jeong Kim
- Department Physiology, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Lutz Birnbaumer
- Institute of Biomedical Research, School of Biomedical Sciences, Catholic University of Argentina, Buenos AiresC1107AFF, Argentina
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC27709
| | - Haeyoung Suh-Kim
- Department of Anatomy, Ajou University School of Medicine, Suwon16499, South Korea
- Research Center, CELLeBRAIN Co., Ltd., Suwon16681, South Korea
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon16499, Korea
| |
Collapse
|
3
|
Kanai SM, Garcia CR, Augustus MR, Sharafeldeen SA, Brooks EP, Sucharov J, Lencer ES, Nichols JT, Clouthier DE. The Gq/11 family of Gα subunits is necessary and sufficient for lower jaw development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.611698. [PMID: 39345358 PMCID: PMC11430119 DOI: 10.1101/2024.09.17.611698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Vertebrate jaw development is coordinated by highly conserved ligand-receptor systems such as the peptide ligand Endothelin 1 (Edn1) and Endothelin receptor type A (Ednra), which are required for patterning of lower jaw structures. The Edn1/Ednra signaling pathway establishes the identity of lower jaw progenitor cells by regulating expression of numerous patterning genes, but the intracellular signaling mechanisms linking receptor activation to gene regulation remain poorly understood. As a first step towards elucidating this mechanism, we examined the function of the Gq/11 family of Gα subunits in zebrafish using pharmacological inhibition and genetic ablation of Gq/11 activity and transgenic induction of a constitutively active Gq protein in edn1 -/- embryos. Genetic loss of Gq/11 activity fully recapitulated the edn1 -/- phenotype, with genes encoding G11 being most essential. Furthermore, inducing Gq activity in edn1 -/- embryos not only restored Edn1/Ednra-dependent jaw structures and gene expression signatures but also caused homeosis of the upper jaw structure into a lower jaw-like structure. These results indicate that Gq/11 is necessary and sufficient to mediate the lower jaw patterning mechanism for Ednra in zebrafish.
Collapse
Affiliation(s)
- Stanley M. Kanai
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Chloe R. Garcia
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - MaCalia R. Augustus
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Shujan A. Sharafeldeen
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Elliott P. Brooks
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Juliana Sucharov
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Ezra S. Lencer
- Department of Biology, Lafayette College, Easton, PA USA
| | - James T. Nichols
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - David E. Clouthier
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| |
Collapse
|
4
|
Bernhard SM, Han J, Che T. GPCR-G protein selectivity revealed by structural pharmacology. FEBS J 2024; 291:2784-2791. [PMID: 38151714 PMCID: PMC11209754 DOI: 10.1111/febs.17049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/28/2023] [Accepted: 12/27/2023] [Indexed: 12/29/2023]
Abstract
Receptor-G protein promiscuity is frequently observed in class A G protein-coupled receptors (GPCRs). In particular, GPCRs can couple with G proteins from different families (Gαs, Gαq/11, Gαi/o, and Gα12/13) or the same family subtypes. The molecular basis underlying the selectivity/promiscuity is not fully revealed. We recently reported the structures of kappa opioid receptor (KOR) in complex with the Gi/o family subtypes [Gαi1, GαoA, Gαz, and Gustducin (Gαg)] determined by cryo-electron microscopy (cryo-EM). The structural analysis, in combination with pharmacological studies, provides insights into Gi/o subtype selectivity. Given the conserved sequence identity and activation mechanism between different G protein families, the findings within Gi/o subtypes could be likely extended to other families. Understanding the KOR-Gi/o or GPCR-G protein selectivity will facilitate the development of more precise therapeutics targeting a specific G protein subtype.
Collapse
MESH Headings
- Humans
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/chemistry
- Cryoelectron Microscopy
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, kappa/chemistry
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/genetics
- GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
- GTP-Binding Protein alpha Subunits, Gi-Go/chemistry
- GTP-Binding Protein alpha Subunits, Gi-Go/genetics
- GTP-Binding Proteins/metabolism
- GTP-Binding Proteins/chemistry
- GTP-Binding Proteins/genetics
- Protein Binding
- Animals
- Protein Conformation
- Models, Molecular
Collapse
Affiliation(s)
- Sarah M. Bernhard
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Center for Clinical Pharmacology, University of Health Sciences & Pharmacy and Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Jianming Han
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Center for Clinical Pharmacology, University of Health Sciences & Pharmacy and Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Tao Che
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Center for Clinical Pharmacology, University of Health Sciences & Pharmacy and Washington University School of Medicine; St. Louis, MO 63110, USA
| |
Collapse
|
5
|
Abu Obaid A, Ivandic I, Korsching SI. Deciphering the function of the fifth class of Gα proteins: regulation of ionic homeostasis as unifying hypothesis. Cell Mol Life Sci 2024; 81:213. [PMID: 38727814 PMCID: PMC11087313 DOI: 10.1007/s00018-024-05228-w] [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: 02/26/2024] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 05/13/2024]
Abstract
Trimeric G proteins transduce signals from a superfamily of receptors and each G protein controls a wide range of cellular and systemic functions. Their highly conserved alpha subunits fall in five classes, four of which have been well investigated (Gs, Gi, G12, Gq). In contrast, the function of the fifth class, Gv is completely unknown, despite its broad occurrence and evolutionary ancient origin (older than metazoans). Here we show a dynamic presence of Gv mRNA in several organs during early development of zebrafish, including the hatching gland, the pronephros and several cartilage anlagen, employing in situ hybridisation. Next, we generated a Gv frameshift mutation in zebrafish and observed distinct phenotypes such as reduced oviposition, premature hatching and craniofacial abnormalities in bone and cartilage of larval zebrafish. These phenotypes could suggest a disturbance in ionic homeostasis as a common denominator. Indeed, we find reduced levels of calcium, magnesium and potassium in the larvae and changes in expression levels of the sodium potassium pump atp1a1a.5 and the sodium/calcium exchanger ncx1b in larvae and in the adult kidney, a major osmoregulatory organ. Additionally, expression of sodium chloride cotransporter slc12a3 and the anion exchanger slc26a4 is altered in complementary ways in adult kidney. It appears that Gv may modulate ionic homeostasis in zebrafish during development and in adults. Our results constitute the first insight into the function of the fifth class of G alpha proteins.
Collapse
Affiliation(s)
- Asmaa Abu Obaid
- Institute of Genetics, Faculty of Mathematics and Natural Sciences of the University at Cologne, Zülpicher Str. 47A, 50674, Cologne, Germany
- Department of Optometry, Faculty of Modern Sciences, The Arab American University, Yousef Asfour Street, Ramallah, Palestine
| | - Ivan Ivandic
- Institute of Genetics, Faculty of Mathematics and Natural Sciences of the University at Cologne, Zülpicher Str. 47A, 50674, Cologne, Germany
| | - Sigrun I Korsching
- Institute of Genetics, Faculty of Mathematics and Natural Sciences of the University at Cologne, Zülpicher Str. 47A, 50674, Cologne, Germany.
| |
Collapse
|
6
|
Falsaperla R, Sortino V, Marino SD, Collotta AD, Gammeri C, Sipala FM, Volti GL, Ruggieri M, Ronsisvalle S. Molecular Dynamic Simulations to Determine Individualized Therapy: Tetrabenazine for the GNAO1 Encephalopathy E246K Variant. Mol Diagn Ther 2024; 28:329-337. [PMID: 38581611 DOI: 10.1007/s40291-024-00706-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2024] [Indexed: 04/08/2024]
Abstract
INTRODUCTION GNAO1 encephalopathy is characterized by severe hypotonia, psychomotor retardation, epilepsy, and movement disorders. Genetic variations in GNAO1 have been linked to neurological symptoms including movement disorders like dystonia. The correlation between the E246K mutation in the Gα subunit and aberrant signal transduction of G proteins has been established but no data are reported regarding the efficacy of medical treatment with tetrabenazine. METHODS Molecular modeling studies were performed to elucidate the molecular mechanisms underlying this mutation. We developed drug efficacy models using molecular dynamic simulations that replicated the behavior of wild-type and mutated proteins in the presence or absence of ligands. RESULTS AND DISCUSSION We demonstrated that the absence of the mutation leads to normal signal transduction upon receptor activation by the endogenous ligand, but not in the presence of tetrabenazine. In contrast, the presence of the mutation resulted in abnormal signal transduction in the presence of the endogenous ligand, which was corrected by the drug tetrabenazine. Tetrabenazine was identified as a promising therapeutic option for pediatric patients suffering from encephalopathy due to an E246K mutation in the GNAO1 gene validated through molecular dynamics. This is a potential first example of the use of this technique in a rare neurological pediatric disease.
Collapse
Affiliation(s)
- Raffaele Falsaperla
- Neonatal Intensive Care Unit and Neonatal Accompaniment Unit, Azienda Ospedaliero-Universitaria Policlinico "Rodolico-San Marco", San Marco Hospital, University of Catania, Catania, Italy.
- Unit of Clinical Paediatrics, Azienda Ospedaliero-Universitaria Policlinico, "Rodolico-San Marco", San Marco Hospital, Catania, Italy.
- Pediatric Clinic, University of Ferrara, Ferrara, Italy.
| | - Vincenzo Sortino
- Unit of Clinical Paediatrics, Azienda Ospedaliero-Universitaria Policlinico, "Rodolico-San Marco", San Marco Hospital, Catania, Italy
| | - Simona Domenica Marino
- Unit of Clinical Paediatrics, Azienda Ospedaliero-Universitaria Policlinico, "Rodolico-San Marco", San Marco Hospital, Catania, Italy
| | - Ausilia Desiree Collotta
- Unit of Clinical Paediatrics, Azienda Ospedaliero-Universitaria Policlinico, "Rodolico-San Marco", San Marco Hospital, Catania, Italy
- Postgraduate Training Program in Pediatrics, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Carmela Gammeri
- Postgraduate Training Program in Pediatrics, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Federica Maria Sipala
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 87, 95125, Catania, Italy
| | - Martino Ruggieri
- Unit of Clinical Pediatrics, Unit of Rare Diseases, AOU "Policlinico", PO "G. Rodolico", University of Catania, Catania, Italy
| | - Simone Ronsisvalle
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| |
Collapse
|
7
|
Nelic D, Chetverikov N, Hochmalová M, Diaz C, Doležal V, Boulos J, Jakubík J, Martemyanov K, Janoušková-Randáková A. Agonist-selective activation of individual G-proteins by muscarinic receptors. Sci Rep 2024; 14:9652. [PMID: 38671143 PMCID: PMC11053168 DOI: 10.1038/s41598-024-60259-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/20/2024] [Indexed: 04/28/2024] Open
Abstract
Selective activation of individual subtypes of muscarinic receptors is a promising way to safely alleviate a wide range of pathological conditions in the central nervous system and the periphery as well. The flexible G-protein interface of muscarinic receptors allows them to interact with several G-proteins with various efficacy, potency, and kinetics. Agonists biased to the particular G-protein mediated pathway may result in selectivity among muscarinic subtypes and, due to the non-uniform expression of individual G-protein alpha subunits, possibly achieve tissue specificity. Here, we demonstrate that novel tetrahydropyridine-based agonists exert specific signalling profiles in coupling with individual G-protein α subunits. These signalling profiles profoundly differ from the reference agonist carbachol. Moreover, coupling with individual Gα induced by these novel agonists varies among subtypes of muscarinic receptors which may lead to subtype selectivity. Thus, the novel tetrahydropyridine-based agonist can contribute to the elucidation of the mechanism of pathway-specific activation of muscarinic receptors and serve as a starting point for the development of desired selective muscarinic agonists.
Collapse
Affiliation(s)
- Dominik Nelic
- Department of Neurochemistry, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Nikolai Chetverikov
- Department of Neurochemistry, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Martina Hochmalová
- Department of Neurochemistry, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Christina Diaz
- Department of Physical Sciences, Barry University, Miami Shores, Miami, FL, USA
| | - Vladimír Doležal
- Department of Neurochemistry, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - John Boulos
- Department of Physical Sciences, Barry University, Miami Shores, Miami, FL, USA
| | - Jan Jakubík
- Department of Neurochemistry, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Kirill Martemyanov
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, Jupiter, FL, 33458, USA.
| | - Alena Janoušková-Randáková
- Department of Neurochemistry, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic.
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, Jupiter, FL, 33458, USA.
| |
Collapse
|
8
|
Jarysta A, Tadenev ALD, Day M, Krawchuk B, Low BE, Wiles MV, Tarchini B. Inhibitory G proteins play multiple roles to polarize sensory hair cell morphogenesis. eLife 2024; 12:RP88186. [PMID: 38651641 PMCID: PMC11037916 DOI: 10.7554/elife.88186] [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] [Indexed: 04/25/2024] Open
Abstract
Inhibitory G alpha (GNAI or Gαi) proteins are critical for the polarized morphogenesis of sensory hair cells and for hearing. The extent and nature of their actual contributions remains unclear, however, as previous studies did not investigate all GNAI proteins and included non-physiological approaches. Pertussis toxin can downregulate functionally redundant GNAI1, GNAI2, GNAI3, and GNAO proteins, but may also induce unrelated defects. Here, we directly and systematically determine the role(s) of each individual GNAI protein in mouse auditory hair cells. GNAI2 and GNAI3 are similarly polarized at the hair cell apex with their binding partner G protein signaling modulator 2 (GPSM2), whereas GNAI1 and GNAO are not detected. In Gnai3 mutants, GNAI2 progressively fails to fully occupy the sub-cellular compartments where GNAI3 is missing. In contrast, GNAI3 can fully compensate for the loss of GNAI2 and is essential for hair bundle morphogenesis and auditory function. Simultaneous inactivation of Gnai2 and Gnai3 recapitulates for the first time two distinct types of defects only observed so far with pertussis toxin: (1) a delay or failure of the basal body to migrate off-center in prospective hair cells, and (2) a reversal in the orientation of some hair cell types. We conclude that GNAI proteins are critical for hair cells to break planar symmetry and to orient properly before GNAI2/3 regulate hair bundle morphogenesis with GPSM2.
Collapse
Affiliation(s)
| | | | - Matthew Day
- The Jackson LaboratoryBar HarborUnited States
| | | | | | | | - Basile Tarchini
- The Jackson LaboratoryBar HarborUnited States
- Tufts University School of MedicineBostonUnited States
| |
Collapse
|
9
|
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
|
10
|
Jarysta A, Tadenev ALD, Day M, Krawchuk B, Low BE, Wiles MV, Tarchini B. Inhibitory G proteins play multiple roles to polarize sensory hair cell morphogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.25.542257. [PMID: 37292807 PMCID: PMC10245865 DOI: 10.1101/2023.05.25.542257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Inhibitory G alpha (GNAI or Gαi) proteins are critical for the polarized morphogenesis of sensory hair cells and for hearing. The extent and nature of their actual contributions remains unclear, however, as previous studies did not investigate all GNAI proteins and included non-physiological approaches. Pertussis toxin can downregulate functionally redundant GNAI1, GNAI2, GNAI3 and GNAO proteins, but may also induce unrelated defects. Here we directly and systematically determine the role(s) of each individual GNAI protein in mouse auditory hair cells. GNAI2 and GNAI3 are similarly polarized at the hair cell apex with their binding partner GPSM2, whereas GNAI1 and GNAO are not detected. In Gnai3 mutants, GNAI2 progressively fails to fully occupy the subcellular compartments where GNAI3 is missing. In contrast, GNAI3 can fully compensate for the loss of GNAI2 and is essential for hair bundle morphogenesis and auditory function. Simultaneous inactivation of Gnai2 and Gnai3 recapitulates for the first time two distinct types of defects only observed so far with pertussis toxin: 1) a delay or failure of the basal body to migrate off-center in prospective hair cells, and 2) a reversal in the orientation of some hair cell types. We conclude that GNAI proteins are critical for hair cells to break planar symmetry and to orient properly before GNAI2/3 regulate hair bundle morphogenesis with GPSM2.
Collapse
|
11
|
Baris RO, Sahin N, Bilgic AD, Ozdemir C, Edgunlu TG. Molecular and in silico analyses of SYN III gene variants in autism spectrum disorder. Ir J Med Sci 2023; 192:2887-2895. [PMID: 37166614 DOI: 10.1007/s11845-023-03402-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/04/2023] [Indexed: 05/12/2023]
Abstract
BACKGROUND Defects in neurotransmission and synaptogenesis are noteworthy in the pathogenesis of ASD. Synapsin III (SYN III) is defined as a synaptic vesicle protein that plays an important role in synaptogenesis and regulation of neurotransmitter release and neurite outgrowth. Therefore, SYN III may associate with many neurodevelopmental diseases, including ASD. AIM The aim of this study was to investigate whether the SYN III gene -631 C > G (rs133946) and -196 G > A (rs133945) polymorphisms are associated with susceptibility to ASD. METHODS SYN III variants and the risk of ASD were investigated in 26 healthy children and 24 ASD children. SYN III gene variants were genotyped by PCR-RFLP methods. The differences in genotype and allele frequencies between the ASD and control groups were calculated using the chi-square (χ2). We analysed the SYN III gene using web-based tools. RESULTS Our results suggest that the presence of the AA genotype of the SYN III -196 G > A (rs133945) polymorphism affects the characteristics and development of ASD in children (p = 0.012). SYN III -631 C > G (rs133946) polymorphism was not associated with ASD (p = 0.524). We have shown the prediction of gene-gene interaction that SYN III is co-expressed with 17 genes, physical interaction with 3 genes, and co-localization with 12 genes. The importance of different genes (SYN I, II, III, GABRD, NOS1AP, GNAO1) for ASD pathogenesis was revealed by GO analysis. CONCLUSION Considering the role of SYN III and related genes, especially in the synaptic vesicle pathway and neurotransmission, its effect on ASD can be further investigated.
Collapse
Affiliation(s)
- Remzi Oguz Baris
- Faculty of Medicine, Mugla Sitki Kocman University, Mugla, Turkey
| | - Nilfer Sahin
- Department of Child and Adolescent Mental Health Diseases School of Medicine, Muğla Sıtkı Koçman University, Mugla, Turkey
| | - Ayşegül Demirtas Bilgic
- Department of Medical Biology, Health Sciences Institution, Muğla Sıtkı Koçman University, Mugla, Turkey
| | - Cilem Ozdemir
- Department of Medical Biology, Health Sciences Institution, Muğla Sıtkı Koçman University, Mugla, Turkey.
| | - Tuba Gokdogan Edgunlu
- Department of Medical Biology, School of Medicine, Muğla Sıtkı Koçman University, Mugla, 48000, Turkey
| |
Collapse
|
12
|
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
|
13
|
Li Y, Chen H, Li L, Cao X, Ding X, Chen L, Cao D. Phenotypes in children with GNAO1 encephalopathy in China. Front Pediatr 2023; 11:1086970. [PMID: 37705601 PMCID: PMC10495587 DOI: 10.3389/fped.2023.1086970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 08/14/2023] [Indexed: 09/15/2023] Open
Abstract
Background The GNAO1 gene encodes the α-subunit (Gαo) of the heterotrimeric guanine nucleotide-binding protein (G protein). The aim of this study was to explore the clinical characteristics of patients with GNAO1 pathogenic variations. Methods Ten patients with pathogenic variations in GNAO1 were enrolled from the Shenzhen Children's Hospital. Clinical data from several cases previously reported from China were also included and analyzed. Results Twenty-seven patients with variations in GNAO1 were analyzed (10 patients from Shenzhen Children's Hospital, 17 patients from previously published studies) including 12 boys and 15 girls. The median age of onset was 3 months with moderate to severe global developmental delay. Nineteen different GNAO1 heterozygous variants were identified. Epilepsy was observed in 18 patients (67%, 18/27), movement disorder (MD) was observed in 22 patients (81%, 22/27), and both were seen in 13 patients (48%, 13/27). Seizures typically presented as focal seizures in all patients with epilepsy. MD typically presented as dystonia and chorea. Loss-of-function (LOF) or partial loss-of-function (PLOF) mutations were more frequent in patients with developmental and epileptic encephalopathy (p = 0.029). Interictal electroencephalograms showed multifocal or diffuse epileptiform discharges. The most common magnetic resonance imaging finding was widened extracerebral space. In contrast to MD, in which improvements were not common, seizures were easily controlled by anti-seizure medications. Severe dystonia in three patients was effectively treated by deep brain stimulation. Seven (26%, 7/27) patients died of respiratory complications, status dystonicus, choreoathetosis, or sudden unexpected death in epilepsy. Conclusion We analyzed clinical data of 27 cases of GNAO1-related encephalopathy in China. MD seemed to be the central feature and was most difficult to control. LOF or PLOF variants were significantly associated with developmental and epileptic encephalopathy. The active intervention of severe dystonia may prevent death due to status dystonicus. However, future studies with larger samples are needed to confirm these results.
Collapse
Affiliation(s)
- Yanmei Li
- Shenzhen Children’s Hospital, Shantou University, Shenzhen, China
- Department of Neurology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Hong Chen
- Department of Neurology, Shenzhen Children’s Hospital, Shenzhen, China
- Surgery Division, Epilepsy Center, Shenzhen Children’s Hospital, Shenzhen, China
| | - Lin Li
- Surgery Division, Epilepsy Center, Shenzhen Children’s Hospital, Shenzhen, China
| | - Xueyan Cao
- Department of Neurology, Shenzhen Children’s Hospital, Shenzhen, China
- Surgery Division, Epilepsy Center, Shenzhen Children’s Hospital, Shenzhen, China
| | - Xin Ding
- Department of Neurology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Li Chen
- Department of Neurology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Dezhi Cao
- Department of Neurology, Shenzhen Children’s Hospital, Shenzhen, China
- Surgery Division, Epilepsy Center, Shenzhen Children’s Hospital, Shenzhen, China
| |
Collapse
|
14
|
Polikarpova AV, Egorova TV, Lunev EA, Tsitrina AA, Vassilieva SG, Savchenko IM, Silaeva YY, Deykin AV, Bardina MV. CRISPR/Cas9-generated mouse model with humanizing single-base substitution in the Gnao1 for safety studies of RNA therapeutics. Front Genome Ed 2023; 5:1034720. [PMID: 37077890 PMCID: PMC10106585 DOI: 10.3389/fgeed.2023.1034720] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 03/20/2023] [Indexed: 04/05/2023] Open
Abstract
The development of personalized medicine for genetic diseases requires preclinical testing in the appropriate animal models. GNAO1 encephalopathy is a severe neurodevelopmental disorder caused by heterozygous de novo mutations in the GNAO1 gene. GNAO1 c.607 G>A is one of the most common pathogenic variants, and the mutant protein Gαo-G203R likely adversely affects neuronal signaling. As an innovative approach, sequence-specific RNA-based therapeutics such as antisense oligonucleotides or effectors of RNA interference are potentially applicable for selective suppression of the mutant GNAO1 transcript. While in vitro validation can be performed in patient-derived cells, a humanized mouse model to rule out the safety of RNA therapeutics is currently lacking. In the present work, we employed CRISPR/Cas9 technology to introduce a single-base substitution into exon 6 of the Gnao1 to replace the murine Gly203-coding triplet (GGG) with the codon used in the human gene (GGA). We verified that genome-editing did not interfere with the Gnao1 mRNA or Gαo protein synthesis and did not alter localization of the protein in the brain structures. The analysis of blastocysts revealed the off-target activity of the CRISPR/Cas9 complexes; however, no modifications of the predicted off-target sites were detected in the founder mouse. Histological staining confirmed the absence of abnormal changes in the brain of genome-edited mice. The created mouse model with the “humanized” fragment of the endogenous Gnao1 is suitable to rule out unintended targeting of the wild-type allele by RNA therapeutics directed at lowering GNAO1 c.607 G>A transcripts.
Collapse
Affiliation(s)
- Anna V. Polikarpova
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
- Marlin Biotech, Sochi, Russia
| | - Tatiana V. Egorova
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
- Marlin Biotech, Sochi, Russia
| | - Evgenii A. Lunev
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
- Marlin Biotech, Sochi, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alexandra A. Tsitrina
- Koltzov Institute of Developmental Biology Russian Academy of Sciences, Moscow, Russia
| | - Svetlana G. Vassilieva
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
- Marlin Biotech, Sochi, Russia
| | - Irina M. Savchenko
- Marlin Biotech, Sochi, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Yuliya Y. Silaeva
- Core Facility Center, Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
| | - Alexey V. Deykin
- Marlin Biotech, Sochi, Russia
- Core Facility Center, Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
- Laboratory of Genetic Technologies and Genome Editing for Biomedicine and Animal Health, Joint Center for Genetic Technologies, Belgorod National Research University, Belgorod, Russia
| | - Maryana V. Bardina
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
- Marlin Biotech, Sochi, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
- *Correspondence: Maryana V. Bardina,
| |
Collapse
|
15
|
Koval A, Larasati YA, Savitsky M, Solis GP, Good JM, Quinodoz M, Rivolta C, Superti-Furga A, Katanaev VL. In-depth molecular profiling of an intronic GNAO1 mutant as the basis for personalized high-throughput drug screening. MED 2023; 4:311-325.e7. [PMID: 37001522 DOI: 10.1016/j.medj.2023.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 03/01/2023] [Accepted: 03/07/2023] [Indexed: 04/03/2023]
Abstract
BACKGROUND The GNAO1 gene, encoding the major neuronal G protein Gαo, is mutated in a subset of pediatric encephalopathies. Most such mutations consist of missense variants. METHODS In this study, we present a precision medicine workflow combining next-generation sequencing (NGS) diagnostics, molecular etiology analysis, and personalized drug discovery. FINDINGS We describe a patient carrying a de novo intronic mutation (NM_020988.3:c.724-8G>A), leading to epilepsy-negative encephalopathy with motor dysfunction from the second decade. Our data show that this mutation creates a novel splice acceptor site that in turn causes an in-frame insertion of two amino acid residues, Pro-Gln, within the regulatory switch III region of Gαo. This insertion misconfigures the switch III loop and creates novel interactions with the catalytic switch II region, resulting in increased GTP uptake, defective GTP hydrolysis, and aberrant interactions with effector proteins. In contrast, intracellular localization, Gβγ interactions, and G protein-coupled receptor (GPCR) coupling of the Gαo[insPQ] mutant protein remain unchanged. CONCLUSIONS This in-depth analysis characterizes the heterozygous c.724-8G>A mutation as partially dominant negative, providing clues to the molecular etiology of this specific pathology. Further, this analysis allows us to establish and validate a high-throughput screening platform aiming at identifying molecules that could correct the aberrant biochemical functions of the mutant Gαo. FUNDING This work was supported by the Joint Seed Money Funding scheme between the University of Geneva and the Hebrew University of Jerusalem.
Collapse
Affiliation(s)
- Alexey Koval
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Yonika A Larasati
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Mikhail Savitsky
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Gonzalo P Solis
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Jean-Marc Good
- Division of Genetic Medicine, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Mathieu Quinodoz
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), 4031 Basel, Switzerland; Department of Ophthalmology, University of Basel, 4031 Basel, Switzerland; Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Carlo Rivolta
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), 4031 Basel, Switzerland; Department of Ophthalmology, University of Basel, 4031 Basel, Switzerland; Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Andrea Superti-Furga
- Division of Genetic Medicine, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Vladimir L Katanaev
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690090 Vladivostok, Russia.
| |
Collapse
|
16
|
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
|
17
|
Xu S, Yu W, Zhang X, Wang W, Wang X. The regulatory role of Gnao1 protein in diabetic encephalopathy in KK-Ay mice and streptozotocin-induced diabetic rats. Brain Res 2022; 1792:148012. [PMID: 35839930 DOI: 10.1016/j.brainres.2022.148012] [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: 05/04/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 11/02/2022]
Abstract
AIMS To investigate the regulation and functional role of Gnao1 in the brain of diabetic encephalopathy (DE) in various animal models. RESULTS Data from the biochemical and behavioral studies showed that DE models were successful induced in streptozotocin treatment animals and KK-Ay mice. Gnao1 was down regulated in the brain tissues of these two diabetes animal models with significant cognition deficiency. It suggested that the changes in DE are also related to dementia such as Alzheimer's disease (AD). Our study also showed that the expression of adrenergic α2 receptor (Adr-α2R), the upstream protein of Gnao1, was decreased in DE animal models. Furthermore, many downstream proteins of Gnao1 also altered, among which cAMP and PKA proteins were increased, CREB and BDNF proteins were decreased both in animal models and in the cell levels. In addition, Gnao1 silencing leads to the increase of reactive oxygen species (ROS) and the decreased proliferation in cultured primary astrocytes, which means that the deficiency of Gnao1 might not be benefit for DE. CONCLUSION Our findings demonstrated the importance of Gnao1 in DE and suggested Gnao1 as a novel marker and a promising therapeutic target for DE and dementia in animal models.
Collapse
Affiliation(s)
- Shuhong Xu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenwen Yu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiang Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weiping Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoliang Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
Inazumi H, Kuwahara K, Nakagawa Y, Kuwabara Y, Numaga-Tomita T, Kashihara T, Nakada T, Kurebayashi N, Oya M, Nonaka M, Sugihara M, Kinoshita H, Moriuchi K, Yanagisawa H, Nishikimi T, Motoki H, Yamada M, Morimoto S, Otsu K, Mortensen RM, Nakao K, Kimura T. NRSF- GNAO1 Pathway Contributes to the Regulation of Cardiac Ca 2+ Homeostasis. Circ Res 2022; 130:234-248. [PMID: 34875852 DOI: 10.1161/circresaha.121.318898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 12/06/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND During the development of heart failure, a fetal cardiac gene program is reactivated and accelerates pathological cardiac remodeling. We previously reported that a transcriptional repressor, NRSF (neuron restrictive silencer factor), suppresses the fetal cardiac gene program, thereby maintaining cardiac integrity. The underlying molecular mechanisms remain to be determined, however. METHODS We aim to elucidate molecular mechanisms by which NRSF maintains normal cardiac function. We generated cardiac-specific NRSF knockout mice and analyzed cardiac gene expression profiles in those mice and mice cardiac-specifically expressing a dominant-negative NRSF mutant. RESULTS We found that cardiac expression of Gαo, an inhibitory G protein encoded in humans by GNAO1, is transcriptionally regulated by NRSF and is increased in the ventricles of several mouse models of heart failure. Genetic knockdown of Gnao1 ameliorated the cardiac dysfunction and prolonged survival rates in these mouse heart failure models. Conversely, cardiac-specific overexpression of GNAO1 in mice was sufficient to induce cardiac dysfunction. Mechanistically, we observed that increasing Gαo expression increased surface sarcolemmal L-type Ca2+ channel activity, activated CaMKII (calcium/calmodulin-dependent kinase-II) signaling, and impaired Ca2+ handling in ventricular myocytes, which led to cardiac dysfunction. CONCLUSIONS These findings shed light on a novel function of Gαo in the regulation of cardiac Ca2+ homeostasis and systolic function and suggest Gαo may be an effective therapeutic target for the treatment of heart failure.
Collapse
Affiliation(s)
- Hideaki Inazumi
- Cardiovascular Medicine (H.I., Y.N., H.K., K.M., H.Y., T. Nishikimi, T. Kimura), Graduate School of Medicine, Kyoto University
| | - Koichiro Kuwahara
- Cardiovascular Medicine (K.K., M.O., H.M.), School of Medicine, Shinshu University, Matsumoto
| | - Yasuaki Nakagawa
- Cardiovascular Medicine (H.I., Y.N., H.K., K.M., H.Y., T. Nishikimi, T. Kimura), Graduate School of Medicine, Kyoto University
| | - Yoshihiro Kuwabara
- Center for Accessing Early Promising Treatment, Kyoto University Hospital (Y.K.)
| | - Takuro Numaga-Tomita
- Molecular Pharmacology (T.N.-T., M.Y.), School of Medicine, Shinshu University, Matsumoto
| | - Toshihide Kashihara
- Molecular Pharmacology, School of Pharmaceutical Sciences, Kitasato University, Tokyo (T. Kashihara)
| | - Tsutomu Nakada
- Research Center for Supports to Advanced Science (T. Nakada), School of Medicine, Shinshu University, Matsumoto
| | - Nagomi Kurebayashi
- Cellular and Molecular Pharmacology, School of Medicine, Juntendo University, Tokyo (N.K.)
| | - Miku Oya
- Cardiovascular Medicine (K.K., M.O., H.M.), School of Medicine, Shinshu University, Matsumoto
| | - Miki Nonaka
- Pain Control Research, The Jikei University School of Medicine (M.N.)
| | - Masami Sugihara
- Clinical Laboratory Medicine, School of Medicine, Juntendo University, Tokyo (M.S.)
| | - Hideyuki Kinoshita
- Cardiovascular Medicine (H.I., Y.N., H.K., K.M., H.Y., T. Nishikimi, T. Kimura), Graduate School of Medicine, Kyoto University
| | - Kenji Moriuchi
- Cardiovascular Medicine (H.I., Y.N., H.K., K.M., H.Y., T. Nishikimi, T. Kimura), Graduate School of Medicine, Kyoto University
| | | | - Toshio Nishikimi
- Cardiovascular Medicine (H.I., Y.N., H.K., K.M., H.Y., T. Nishikimi, T. Kimura), Graduate School of Medicine, Kyoto University
- Wakakusa Tatsuma Rehabilitation Hospital, Osaka (T. Nishikimi)
| | - Hirohiko Motoki
- Cardiovascular Medicine (K.K., M.O., H.M.), School of Medicine, Shinshu University, Matsumoto
| | - Mitsuhiko Yamada
- Molecular Pharmacology (T.N.-T., M.Y.), School of Medicine, Shinshu University, Matsumoto
| | - Sachio Morimoto
- School of Health Sciences Fukuoka, International University of Health and Welfare, Okawa (S.M.)
| | - Kinya Otsu
- The School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, United Kingdom (K.O.)
| | | | - Kazuwa Nakao
- Medical Innovation Center (K.N.), Graduate School of Medicine, Kyoto University
| | - Takeshi Kimura
- Cardiovascular Medicine (H.I., Y.N., H.K., K.M., H.Y., T. Nishikimi, T. Kimura), Graduate School of Medicine, Kyoto University
| |
Collapse
|
21
|
Solis GP, Kozhanova TV, Koval A, Zhilina SS, Mescheryakova TI, Abramov AA, Ishmuratov EV, Bolshakova ES, Osipova KV, Ayvazyan SO, Lebon S, Kanivets IV, Pyankov DV, Troccaz S, Silachev DN, Zavadenko NN, Prityko AG, Katanaev VL. Pediatric Encephalopathy: Clinical, Biochemical and Cellular Insights into the Role of Gln52 of GNAO1 and GNAI1 for the Dominant Disease. Cells 2021; 10:2749. [PMID: 34685729 PMCID: PMC8535069 DOI: 10.3390/cells10102749] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/29/2021] [Accepted: 10/12/2021] [Indexed: 11/19/2022] Open
Abstract
Heterotrimeric G proteins are immediate transducers of G protein-coupled receptors-the biggest receptor family in metazoans-and play innumerate functions in health and disease. A set of de novo point mutations in GNAO1 and GNAI1, the genes encoding the α-subunits (Gαo and Gαi1, respectively) of the heterotrimeric G proteins, have been described to cause pediatric encephalopathies represented by epileptic seizures, movement disorders, developmental delay, intellectual disability, and signs of neurodegeneration. Among such mutations, the Gln52Pro substitutions have been previously identified in GNAO1 and GNAI1. Here, we describe the case of an infant with another mutation in the same site, Gln52Arg. The patient manifested epileptic and movement disorders and a developmental delay, at the onset of 1.5 weeks after birth. We have analyzed biochemical and cellular properties of the three types of dominant pathogenic mutants in the Gln52 position described so far: Gαo[Gln52Pro], Gαi1[Gln52Pro], and the novel Gαo[Gln52Arg]. At the biochemical level, the three mutant proteins are deficient in binding and hydrolyzing GTP, which is the fundamental function of the healthy G proteins. At the cellular level, the mutants are defective in the interaction with partner proteins recognizing either the GDP-loaded or the GTP-loaded forms of Gαo. Further, of the two intracellular sites of Gαo localization, plasma membrane and Golgi, the former is strongly reduced for the mutant proteins. We conclude that the point mutations at Gln52 inactivate the Gαo and Gαi1 proteins leading to aberrant intracellular localization and partner protein interactions. These features likely lie at the core of the molecular etiology of pediatric encephalopathies associated with the codon 52 mutations in GNAO1/GNAI1.
Collapse
Affiliation(s)
- 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.); (S.T.); (D.N.S.)
| | - Tatyana V. Kozhanova
- St. Luka’s Clinical Research Center for Children, 119620 Moscow, Russia; (T.V.K.); (S.S.Z.); (T.I.M.); (A.A.A.); (E.V.I.); (E.S.B.); (K.V.O.); (S.O.A.); (A.G.P.)
- Department of Neurology, Neurosurgery and Medical Genetics, Faculty of Pediatrics, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - 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.); (S.T.); (D.N.S.)
| | - Svetlana S. Zhilina
- St. Luka’s Clinical Research Center for Children, 119620 Moscow, Russia; (T.V.K.); (S.S.Z.); (T.I.M.); (A.A.A.); (E.V.I.); (E.S.B.); (K.V.O.); (S.O.A.); (A.G.P.)
- Department of Neurology, Neurosurgery and Medical Genetics, Faculty of Pediatrics, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Tatyana I. Mescheryakova
- St. Luka’s Clinical Research Center for Children, 119620 Moscow, Russia; (T.V.K.); (S.S.Z.); (T.I.M.); (A.A.A.); (E.V.I.); (E.S.B.); (K.V.O.); (S.O.A.); (A.G.P.)
| | - Aleksandr A. Abramov
- St. Luka’s Clinical Research Center for Children, 119620 Moscow, Russia; (T.V.K.); (S.S.Z.); (T.I.M.); (A.A.A.); (E.V.I.); (E.S.B.); (K.V.O.); (S.O.A.); (A.G.P.)
| | - Evgeny V. Ishmuratov
- St. Luka’s Clinical Research Center for Children, 119620 Moscow, Russia; (T.V.K.); (S.S.Z.); (T.I.M.); (A.A.A.); (E.V.I.); (E.S.B.); (K.V.O.); (S.O.A.); (A.G.P.)
| | - Ekaterina S. Bolshakova
- St. Luka’s Clinical Research Center for Children, 119620 Moscow, Russia; (T.V.K.); (S.S.Z.); (T.I.M.); (A.A.A.); (E.V.I.); (E.S.B.); (K.V.O.); (S.O.A.); (A.G.P.)
| | - Karina V. Osipova
- St. Luka’s Clinical Research Center for Children, 119620 Moscow, Russia; (T.V.K.); (S.S.Z.); (T.I.M.); (A.A.A.); (E.V.I.); (E.S.B.); (K.V.O.); (S.O.A.); (A.G.P.)
| | - Sergey O. Ayvazyan
- St. Luka’s Clinical Research Center for Children, 119620 Moscow, Russia; (T.V.K.); (S.S.Z.); (T.I.M.); (A.A.A.); (E.V.I.); (E.S.B.); (K.V.O.); (S.O.A.); (A.G.P.)
| | - Sébastien Lebon
- Unit of Pediatric Neurology and Neurorehabilitation, Division of Pediatrics, Woman-Mother-Child Department, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland;
| | - Ilya V. Kanivets
- Center of Medical Genetics, Genomed Ltd., 115093 Moscow, Russia; (I.V.K.); (D.V.P.)
| | - Denis V. Pyankov
- Center of Medical Genetics, Genomed Ltd., 115093 Moscow, Russia; (I.V.K.); (D.V.P.)
| | - Sabina Troccaz
- 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.); (S.T.); (D.N.S.)
| | - Denis N. Silachev
- 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.); (S.T.); (D.N.S.)
- 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, 117997 Moscow, Russia
- School of Biomedicine, Far Eastern Federal University, 690090 Vladivostok, Russia
| | - Nikolay N. Zavadenko
- Department of Neurology, Neurosurgery and Medical Genetics, Faculty of Pediatrics, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Andrey G. Prityko
- St. Luka’s Clinical Research Center for Children, 119620 Moscow, Russia; (T.V.K.); (S.S.Z.); (T.I.M.); (A.A.A.); (E.V.I.); (E.S.B.); (K.V.O.); (S.O.A.); (A.G.P.)
- Department of Neurology, Neurosurgery and Medical Genetics, Faculty of Pediatrics, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - 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.); (S.T.); (D.N.S.)
- School of Biomedicine, Far Eastern Federal University, 690090 Vladivostok, Russia
| |
Collapse
|
22
|
Di Rocco M, Galosi S, Lanza E, Tosato F, Caprini D, Folli V, Friedman J, Bocchinfuso G, Martire A, Di Schiavi E, Leuzzi V, Martinelli S. Caenorhabditis elegans provides an efficient drug screening platform for GNAO1-related disorders and highlights the potential role of caffeine in controlling dyskinesia. Hum Mol Genet 2021; 31:929-941. [PMID: 34622282 PMCID: PMC8947233 DOI: 10.1093/hmg/ddab296] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022] Open
Abstract
Dominant GNAO1 mutations cause an emerging group of childhood-onset neurological disorders characterized by developmental delay, intellectual disability, movement disorders, drug-resistant seizures and neurological deterioration. GNAO1 encodes the α-subunit of an inhibitory GTP/GDP-binding protein regulating ion channel activity and neurotransmitter release. The pathogenic mechanisms underlying GNAO1-related disorders remain largely elusive and there are no effective therapies. Here, we assessed the functional impact of two disease-causing variants associated with distinct clinical features, c.139A > G (p.S47G) and c.662C > A (p.A221D), using Caenorhabditis elegans as a model organism. The c.139A > G change was introduced into the orthologous position of the C. elegans gene via CRISPR/Cas9, whereas a knock-in strain carrying the p.A221D variant was already available. Like null mutants, homozygous knock-in animals showed increased egg laying and were hypersensitive to aldicarb, an inhibitor of acetylcholinesterase, suggesting excessive neurotransmitter release by different classes of motor neurons. Automated analysis of C. elegans locomotion indicated that goa-1 mutants move faster than control animals, with more frequent body bends and a higher reversal rate and display uncoordinated locomotion. Phenotypic profiling of heterozygous animals revealed a strong hypomorphic effect of both variants, with a partial dominant-negative activity for the p.A221D allele. Finally, caffeine was shown to rescue aberrant motor function in C. elegans harboring the goa-1 variants; this effect is mainly exerted through adenosine receptor antagonism. Overall, our findings establish a suitable platform for drug discovery, which may assist in accelerating the development of new therapies for this devastating condition, and highlight the potential role of caffeine in controlling GNAO1-related dyskinesia.
Collapse
Affiliation(s)
- Martina Di Rocco
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome 00161, Italy.,Department of Human Neuroscience, "Sapienza" University of Rome, Rome 00185, Italy
| | - Serena Galosi
- Department of Human Neuroscience, "Sapienza" University of Rome, Rome 00185, Italy
| | - Enrico Lanza
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Rome 00161, Italy
| | - Federica Tosato
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Davide Caprini
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Rome 00161, Italy
| | - Viola Folli
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Rome 00161, Italy
| | - Jennifer Friedman
- UCSD Department of Neuroscience and Pediatrics, Rady Children's Hospital Division of Neurology; Rady Children's Institute for Genomic Medicine, San Diego, USA
| | - Gianfranco Bocchinfuso
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Alberto Martire
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Elia Di Schiavi
- Institute of Biosciences and BioResources, National Research Council, Naples 80131, Italy
| | - Vincenzo Leuzzi
- Department of Human Neuroscience, "Sapienza" University of Rome, Rome 00185, Italy
| | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome 00161, Italy
| |
Collapse
|
23
|
Wang D, Dao M, Muntean BS, Giles AC, Martemyanov KA, Grill B. Genetic modeling of GNAO1 disorder delineates mechanisms of Gαo dysfunction. Hum Mol Genet 2021; 31:510-522. [PMID: 34508586 PMCID: PMC8863422 DOI: 10.1093/hmg/ddab235] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/30/2021] [Accepted: 08/09/2021] [Indexed: 12/26/2022] Open
Abstract
GNAO1 encephalopathy is a neurodevelopmental disorder with a spectrum of symptoms that include dystonic movements, seizures and developmental delay. While numerous GNAO1 mutations are associated with this disorder, the functional consequences of pathological variants are not completely understood. Here, we deployed the invertebrate C. elegans as a whole-animal behavioral model to study the functional effects of GNAO1 disorder-associated mutations. We tested several pathological GNAO1 mutations for effects on locomotor behaviors using a combination of CRISPR/Cas9 gene editing and transgenic overexpression in vivo. We report that all three mutations tested (G42R, G203R and R209C) result in strong loss of function defects when evaluated as homozygous CRISPR alleles. In addition, mutations produced dominant negative effects assessed using both heterozygous CRISPR alleles and transgenic overexpression. Experiments in mice confirmed dominant negative effects of GNAO1 G42R, which impaired numerous motor behaviors. Thus, GNAO1 pathological mutations result in conserved functional outcomes across animal models. Our study further establishes the molecular genetic basis of GNAO1 encephalopathy, and develops a CRISPR-based pipeline for functionally evaluating mutations associated with neurodevelopmental disorders.
Collapse
Affiliation(s)
- Dandan Wang
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Maria Dao
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Brian S Muntean
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Andrew C Giles
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Brock Grill
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA.,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA.,Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
24
|
Roldán-Sastre A, Aguado C, Martín-Belmonte A, Alfaro-Ruiz R, Moreno-Martínez AE, Luján R. Cellular Diversity and Differential Subcellular Localization of the G-Protein G αo Subunit in the Mouse Cerebellum. Front Neuroanat 2021; 15:686279. [PMID: 34248508 PMCID: PMC8267243 DOI: 10.3389/fnana.2021.686279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/02/2021] [Indexed: 11/27/2022] Open
Abstract
Heterotrimeric guanine nucleotide-binding proteins (G proteins) transduce signals from G protein-coupled receptors (GPCRs) to effector ion channels and enzymes Gαo, a member of the pertussis toxin-sensitive Gi/o family, is widely expressed in the brain, although its role within a neuronal context remains largely unknown. Using immunohistochemical and quantitative immunoelectron microscopy techniques, we have investigated the expression, cellular and subcellular localization of Gαo in the cerebellar cortex. Histoblot revealed that Gαo is expressed in many brain regions, including the cerebellum. At the cellular level, Gαo protein was distributed in Purkinje cells, basket cells, stellate cells, granule cells and Golgi cells. At the subcellular level, pre-embedding immunoelectron microscopy revealed mainly a postsynaptic localization of Gαo along the extrasynaptic plasma membrane of Purkinje cell dendritic shafts and spines, and dendrites of basket, stellate and granule cells. To a lesser extent, immunolabeling for Gαo was localized in different types of axon terminals establishing excitatory synapses. Moreover, post-embedding immunoelectron microscopy revealed the synaptic localization of Gαo on PSDs of glutamatergic synapses between Purkinje cell spines and parallel fiber terminals and its co-localization with GABAB1 in the same spines. Quantitative analysis of Gαo immunoparticles revealed they preferentially localized on the cytoplasmic face of the plasma membrane. Furthermore, the analysis revealed a high concentration of Gαo around excitatory synapses on Purkinje cell dendritic spines, but a uniform distribution in granule cell dendrites. These molecular-anatomical findings suggest that Gαo is a major signal transducer of specific GPCRs in different neuronal populations in the cerebellum.
Collapse
Affiliation(s)
- Alberto Roldán-Sastre
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Department of Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Carolina Aguado
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Department of Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Alejandro Martín-Belmonte
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Department of Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Rocío Alfaro-Ruiz
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Department of Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Ana Esther Moreno-Martínez
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Department of Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Rafael Luján
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Department of Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| |
Collapse
|
25
|
Yang X, Niu X, Yang Y, Cheng M, Zhang J, Chen J, Yang Z, Zhang Y. Phenotypes of GNAO1 Variants in a Chinese Cohort. Front Neurol 2021; 12:662162. [PMID: 34122306 PMCID: PMC8193119 DOI: 10.3389/fneur.2021.662162] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/06/2021] [Indexed: 11/30/2022] Open
Abstract
This study aimed to analyze the genotypes and phenotypes of GNAO1 variants in a Chinese cohort. Seven male and four female patients with GNAO1 variants were enrolled, including siblings of brothers. Ten different GNAO1 variants (nine missense and one splicing site) were identified, among which six were novel. All the variants were confirmed to be de novo in peripheral blood DNA. Eight (73%, 8/11) patients had epilepsy; the seizure onset age ranged from 6 h after birth to 4 months (median age, 2.5 months). Focal seizures were observed in all eight patients, epileptic spasms occurred in six (75%, 6/8), tonic spasm in four (50%, 4/8), tonic seizures in two, atypical absence in one, and generalized tonic–clonic seizures in one. Seven patients had multiple seizure types. Eight (73%, 8/11) patients had movement disorders, seven of them having only dystonia, and one having dystonia with choreoathetosis. Varying degrees of developmental delay (DD) were present in all 11 patients. The phenotypes were diagnosed as early infantile epileptic encephalopathy (EIEE) in two (18%) patients, which were further diagnosed as West syndrome. Movement disorders (MD) with developmental delay were diagnosed in two (18%) brothers. EIEE and MD were overlapped in six (55%) patients, among which two were diagnosed with West syndrome, one with Ohtahara syndrome, and the other three with non-specific EIEE. One (9%) patient was diagnosed as DD alone. The onset age of GNAO1-related disorders was early infancy. The phenotypic spectrum of GNAO1 included EIEE, MD with DD, and DD alone.
Collapse
Affiliation(s)
- Xiaoling Yang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Xueyang Niu
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Ying Yang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Miaomiao Cheng
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Jing Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Jiaoyang Chen
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Zhixian Yang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yuehua Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| |
Collapse
|
26
|
Randáková A, Jakubík J. Functionally selective and biased agonists of muscarinic receptors. Pharmacol Res 2021; 169:105641. [PMID: 33951507 DOI: 10.1016/j.phrs.2021.105641] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 12/24/2022]
Abstract
Disruption of cholinergic signalling via muscarinic receptors is associated with various pathologies, like Alzheimer's disease or schizophrenia. Selective muscarinic agonists possess therapeutic potential in the treatment of diabetes, pain or Sjögren's syndrome. The orthosteric binding site of all subtypes of the muscarinic receptor is structurally identical, making the development of affinity-based selective agonists virtually impossible. Some agonists, however, are functionally selective; they activate only a subset of receptors or signalling pathways. Others may stabilise specific conformations of the receptor leading to non-uniform modulation of individual signalling pathways (biased agonists). Functionally selective and biased agonists represent a promising approach for selective activation of individual subtypes of muscarinic receptors. In this work we review chemical structures, receptor binding and agonist-specific conformations of currently known functionally selective and biased muscarinic agonists in the context of their intricate intracellular signalling. Further, we take a perspective on the possible use of biased agonists for tissue and organ-specific activation of muscarinic receptors.
Collapse
Affiliation(s)
- Alena Randáková
- Institute of Physiology Czech Academy of Sciences, Prague, Czech Republic.
| | - Jan Jakubík
- Institute of Physiology Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
27
|
Akasaka M, Kamei A, Tanifuji S, Asami M, Ito J, Mizuma K, Oyama K, Tokutomi T, Yamamoto K, Fukushima A, Takenouchi T, Uehara T, Suzuki H, Kosaki K. GNAO1 mutation-related severe involuntary movements treated with gabapentin. Brain Dev 2021; 43:576-579. [PMID: 33358199 DOI: 10.1016/j.braindev.2020.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/28/2020] [Accepted: 12/02/2020] [Indexed: 01/24/2023]
Abstract
BACKGROUND Mutations in GNAO1 typically result in neurodevelopmental disorders, including involuntary movements. They may be improved using calcium-channel modulators. CASE The patient visited our hospital at age 2 years because of moderate global developmental delay. Her intermittent, generalized involuntary movements started at age 8 years. A de novo GNAO1 mutation, NM_020988.2:c.626G > A, (p.Arg209Cys), was identified by whole exome sequencing. At age 9 years, she experienced severe, intermittent involuntary movements, which led to rhabdomyolysis. She needed intensive care with administration of midazolam, dantrolene sodium hydrate, and plasma exchange. We started treating her with gabapentin (GBP), after which she recovered completely. At age 11 years, she developed continuous, generalized involuntary movements. This prompted us to increase the GBP dose, which again resolved the involuntary movements completely. CONCLUSION In the case of movement disorders associated with GNAO1 mutations, GBP treatment may be attempted before more invasive procedures are performed.
Collapse
Affiliation(s)
- Manami Akasaka
- Department of Pediatrics, School of Medicine, Iwate Medical University, Japan.
| | - Atsushi Kamei
- Department of Pediatrics, School of Medicine, Iwate Medical University, Japan
| | - Sachiko Tanifuji
- Department of Pediatrics, School of Medicine, Iwate Medical University, Japan
| | - Maya Asami
- Department of Pediatrics, School of Medicine, Iwate Medical University, Japan
| | - Jun Ito
- Department of Pediatrics, School of Medicine, Iwate Medical University, Japan
| | - Kanako Mizuma
- Department of Pediatrics, School of Medicine, Iwate Medical University, Japan
| | - Kotaro Oyama
- Department of Pediatrics, School of Medicine, Iwate Medical University, Japan
| | - Tomoharu Tokutomi
- Department of Clinical Genetics, School of Medicine, Iwate Medical University, Japan
| | - Kayono Yamamoto
- Department of Clinical Genetics, School of Medicine, Iwate Medical University, Japan
| | - Akimune Fukushima
- Department of Clinical Genetics, School of Medicine, Iwate Medical University, Japan
| | - Toshiki Takenouchi
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Tomoko Uehara
- Department of Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Hisato Suzuki
- Department of Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Kenjiro Kosaki
- Department of Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
28
|
Gαo is a major determinant of cAMP signaling in the pathophysiology of movement disorders. Cell Rep 2021; 34:108718. [PMID: 33535037 PMCID: PMC7903328 DOI: 10.1016/j.celrep.2021.108718] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/07/2020] [Accepted: 01/11/2021] [Indexed: 01/20/2023] Open
Abstract
The G protein alpha subunit o (Gαo) is one of the most abundant proteins in the nervous system, and pathogenic mutations in its gene (GNAO1) cause movement disorder. However, the function of Gαo is ill defined mechanistically. Here, we show that Gαo dictates neuromodulatory responsiveness of striatal neurons and is required for movement control. Using in vivo optical sensors and enzymatic assays, we determine that Gαo provides a separate transduction channel that modulates coupling of both inhibitory and stimulatory dopamine receptors to the cyclic AMP (cAMP)-generating enzyme adenylyl cyclase. Through a combination of cell-based assays and rodent models, we demonstrate that GNAO1-associated mutations alter Gαo function in a neuron-type-specific fashion via a combination of a dominant-negative and loss-of-function mechanisms. Overall, our findings suggest that Gαo and its pathological variants function in specific circuits to regulate neuromodulatory signals essential for executing motor programs. Muntean et al. describe biochemical, cellular, and physiological mechanisms by which the heterotrimeric G protein subunit Gαo controls neuromodulatory signaling in the striatum and elucidate mechanisms by which Gαo mutations compromise movements in GNAO1 disorder.
Collapse
|
29
|
Wang W, Frankel WN. Overlaps, gaps, and complexities of mouse models of Developmental and Epileptic Encephalopathy. Neurobiol Dis 2021; 148:105220. [PMID: 33301879 PMCID: PMC8547712 DOI: 10.1016/j.nbd.2020.105220] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/17/2020] [Accepted: 12/04/2020] [Indexed: 11/28/2022] Open
Abstract
Mouse models have made innumerable contributions to understanding the genetic basis of neurological disease and pathogenic mechanisms and to therapy development. Here we consider the current state of mouse genetic models of Developmental and Epileptic Encephalopathy (DEE), representing a set of rare but devastating and largely intractable childhood epilepsies. By examining the range of mouse lines available in this rapidly moving field and by detailing both expected and unusual features in representative examples, we highlight lessons learned in an effort to maximize the full potential of this powerful resource for preclinical studies.
Collapse
Affiliation(s)
- Wanqi Wang
- Department of Genetics & Development, Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, United States of America.
| | - Wayne N Frankel
- Department of Genetics & Development, Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, United States of America.
| |
Collapse
|
30
|
Corey EA, Ukhanov K, Bobkov YV, McIntyre JC, Martens JR, Ache BW. Inhibitory signaling in mammalian olfactory transduction potentially mediated by Gα o. Mol Cell Neurosci 2020; 110:103585. [PMID: 33358996 DOI: 10.1016/j.mcn.2020.103585] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/27/2020] [Accepted: 12/09/2020] [Indexed: 01/12/2023] Open
Abstract
Olfactory GPCRs (ORs) in mammalian olfactory receptor neurons (ORNs) mediate excitation through the Gαs family member Gαolf. Here we tentatively associate a second G protein, Gαo, with inhibitory signaling in mammalian olfactory transduction by first showing that odor evoked phosphoinositide 3-kinase (PI3K)-dependent inhibition of signal transduction is absent in the native ORNs of mice carrying a conditional OMP-Cre based knockout of Gαo. We then identify an OR from native rat ORNs that are activated by octanol through cyclic nucleotide signaling and inhibited by citral in a PI3K-dependent manner. We show that the OR activates cyclic nucleotide signaling and PI3K signaling in a manner that reflects its functionality in native ORNs. Our findings lay the groundwork to explore the interesting possibility that ORs can interact with two different G proteins in a functionally identified, ligand-dependent manner to mediate opponent signaling in mature mammalian ORNs.
Collapse
Affiliation(s)
- Elizabeth A Corey
- Whitney Laboratory, Center for Smell and Taste, University of Florida, Gainesville, FL 32610, United States of America
| | - Kirill Ukhanov
- Dept. of Pharmacology and Therapeutics, Center for Smell and Taste, University of Florida, Gainesville, FL 32610, United States of America
| | - Yuriy V Bobkov
- Whitney Laboratory, Center for Smell and Taste, University of Florida, Gainesville, FL 32610, United States of America
| | - Jeremy C McIntyre
- Dept. of Neuroscience, Center for Smell and Taste, University of Florida, Gainesville, FL 32610, United States of America
| | - Jeffrey R Martens
- Dept. of Pharmacology and Therapeutics, Center for Smell and Taste, University of Florida, Gainesville, FL 32610, United States of America
| | - Barry W Ache
- Whitney Laboratory, Dept. of Biology, Center for Smell and Taste, University of Florida, Gainesville, FL 32610, United States of America; Whitney Laboratory, Dept. of Neuroscience, Center for Smell and Taste, University of Florida, Gainesville, FL 32610, United States of America.
| |
Collapse
|
31
|
Akamine S, Okuzono S, Yamamoto H, Setoyama D, Sagata N, Ohgidani M, Kato TA, Ishitani T, Kato H, Masuda K, Matsushita Y, Ono H, Ishizaki Y, Sanefuji M, Saitsu H, Matsumoto N, Kang D, Kanba S, Nakabeppu Y, Sakai Y, Ohga S. GNAO1 organizes the cytoskeletal remodeling and firing of developing neurons. FASEB J 2020; 34:16601-16621. [PMID: 33107105 DOI: 10.1096/fj.202001113r] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/03/2020] [Accepted: 10/13/2020] [Indexed: 01/03/2023]
Abstract
Developmental and epileptic encephalopathy (DEE) represents a group of neurodevelopmental disorders characterized by infantile-onset intractable seizures and unfavorable prognosis of psychomotor development. To date, hundreds of genes have been linked to the onset of DEE. GNAO1 is a DEE-associated gene encoding the alpha-O1 subunit of guanine nucleotide-binding protein (GαO ). Despite the increasing number of reported children with GNAO1 encephalopathy, the molecular mechanisms underlying their neurodevelopmental phenotypes remain elusive. We herein present that co-immunoprecipitation and mass spectrometry analyses identified another DEE-associated protein, SPTAN1, as an interacting partner of GαO . Silencing of endogenous Gnao1 attenuated the neurite outgrowth and calcium-dependent signaling. Inactivation of GNAO1 in human-induced pluripotent stem cells gave rise to anomalous brain organoids that only weakly expressed SPTAN1 and Ankyrin-G. Furthermore, GNAO1-deficient organoids failed to conduct synchronized firing to adjacent neurons. These data indicate that GαO and other DEE-associated proteins organize the cytoskeletal remodeling and functional polarity of neurons in the developing brain.
Collapse
Affiliation(s)
- Satoshi Akamine
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sayaka Okuzono
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Yamamoto
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daiki Setoyama
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Noriaki Sagata
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masahiro Ohgidani
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takahiro A Kato
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tohru Ishitani
- Division of Integrated Signaling Systems, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan.,Department of Homeostatic Regulation, Division of Cellular and Molecular Biology. Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Hiroki Kato
- Division of Oral Biological Sciences, Department of Molecular Cell Biology and Oral Anatomy, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Keiji Masuda
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yuki Matsushita
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroaki Ono
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshito Ishizaki
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masafumi Sanefuji
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirotomo Saitsu
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Dongchon Kang
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shigenobu Kanba
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yusaku Nakabeppu
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yasunari Sakai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
32
|
Savitsky M, Solis GP, Kryuchkov M, Katanaev VL. Humanization of Drosophila Gαo to Model GNAO1 Paediatric Encephalopathies. Biomedicines 2020; 8:E395. [PMID: 33036271 PMCID: PMC7599900 DOI: 10.3390/biomedicines8100395] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 12/15/2022] Open
Abstract
Several hundred genes have been identified to contribute to epilepsy-the disease affecting 65 million people worldwide. One of these genes is GNAO1 encoding Gαo, the major neuronal α-subunit of heterotrimeric G proteins. An avalanche of dominant de novo mutations in GNAO1 have been recently described in paediatric epileptic patients, suffering, in addition to epilepsy, from motor dysfunction and developmental delay. Although occurring in amino acids conserved from humans to Drosophila, these mutations and their functional consequences have only been poorly analysed at the biochemical or neuronal levels. Adequate animal models to study the molecular aetiology of GNAO1 encephalopathies have also so far been lacking. As the first step towards modeling the disease in Drosophila, we here describe the humanization of the Gαo locus in the fruit fly. A two-step CRISPR/Cas9-mediated replacement was conducted, first substituting the coding exons 2-3 of Gαo with respective human GNAO1 sequences. At the next step, the remaining exons 4-7 were similarly replaced, keeping intact the gene Cyp49a1 embedded in between, as well as the non-coding exons, exon 1 and the surrounding regulatory sequences. The resulting flies, homozygous for the humanized GNAO1 loci, are viable and fertile without any visible phenotypes; their body weight, locomotion, and longevity are also normal. Human Gαo-specific antibodies confirm the endogenous-level expression of the humanized Gαo, which fully replaces the Drosophila functions. The genetic model we established will make it easy to incorporate encephalopathic GNAO1 mutations and will permit intensive investigations into the molecular aetiology of the human disease through the powerful toolkit of Drosophila genetics.
Collapse
Affiliation(s)
- Mikhail Savitsky
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; (M.S.); (G.P.S.); (M.K.)
| | - Gonzalo P. Solis
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; (M.S.); (G.P.S.); (M.K.)
| | - Mikhail Kryuchkov
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; (M.S.); (G.P.S.); (M.K.)
| | - Vladimir L. Katanaev
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; (M.S.); (G.P.S.); (M.K.)
- School of Biomedicine, Far Eastern Federal University, 690690 Vladivostok, Russia
| |
Collapse
|
33
|
Gold MS, Baron D, Bowirrat A, Blum K. Neurological correlates of brain reward circuitry linked to opioid use disorder (OUD): Do homo sapiens acquire or have a reward deficiency syndrome? J Neurol Sci 2020; 418:117137. [PMID: 32957037 PMCID: PMC7490287 DOI: 10.1016/j.jns.2020.117137] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/19/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022]
Abstract
The extant literature confirms that an array of polymorphic genes related to- neurotransmitters and second messengers govern the net release of dopamine in the Nucleus Accumbens (NAc) in the mesolimbic region of the brain. They are linked predominantly to motivation, anti-stress, incentive salience (wanting), and wellbeing. Notably, in 2000 the Nobel Prize was awarded to Carlsson, Greengard, and Kandel for their work on the molecular and cellular function of dopaminergic activity at neurons. This historical psychopharmacological work involved neurotransmission of serotonin, endorphins, glutamate, and dopamine, and the seminal work of Blum, Gold, Volkow, Nestler, and others related to neurotransmitter function and related behaviors. Currently, Americans are facing their second and worst opioid epidemic, prescribed opioids, and easy access drive this epidemic of overdoses, and opioid use disorders (OUDs). Presently the clinical consensus is to treat OUD, as if it were an opioid deficiency syndrome, with long-term to life-long opioid substitution therapy. Opioid agonist administration is seen as necessary to replace missing opioids, treat OUD, and prevent overdoses, like insulin is used to treat diabetes. Treatment of OUD and addiction, in general, is similar to the endocrinopathy conceptualization in that it views opioid agonist MATs as an essential core to therapy. Is this approach logical? Other than as harm reduction, is using opioids to treat OUD therapeutic or harmful in the long term? This historical Trieste provides a molecular framework to understand the current underpinnings of endorphinergic/dopaminergic mechanisms related to opioid deficiency syndrome and generalized reward processing depletion. WC 249.
Collapse
Affiliation(s)
- Mark S Gold
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States.
| | - David Baron
- Graduate School of Biomedical Sciences, Western University Health Sciences, Pomona, CA, United States
| | - Abdalla Bowirrat
- Department of Neuroscience and Genetics, Interdisciplinary Center Herzliya, Israel
| | - Kenneth Blum
- Graduate School of Biomedical Sciences, Western University Health Sciences, Pomona, CA, United States
| |
Collapse
|
34
|
Cha HL, Choi JM, Oh HH, Bashyal N, Kim SS, Birnbaumer L, Suh-Kim H. Deletion of the α subunit of the heterotrimeric Go protein impairs cerebellar cortical development in mice. Mol Brain 2019; 12:57. [PMID: 31221179 PMCID: PMC6585000 DOI: 10.1186/s13041-019-0477-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/29/2019] [Indexed: 02/02/2023] Open
Abstract
Go is a member of the pertussis toxin-sensitive Gi/o family. Despite its abundance in the central nervous system, the precise role of Go remains largely unknown compared to other G proteins. In the present study, we explored the functions of Go in the developing cerebellar cortex by deleting its gene, Gnao. We performed a histological analysis with cerebellar sections of adult mice by cresyl violet- and immunostaining. Global deletion of Gnao induced cerebellar hypoplasia, reduced arborization of Purkinje cell dendrites, and atrophied Purkinje cell dendritic spines and the terminal boutons of climbing fibers from the inferior olivary nucleus. These results indicate that Go-mediated signaling pathway regulates maturation of presynaptic parallel fibers from granule cells and climbing fibers during the cerebellar cortical development.
Collapse
Affiliation(s)
- Hye Lim Cha
- Departments of Anatomy, Ajou University School of Medicine, Woldcup-ro 164, Yeongtong-gu, Suwon, 16499 South Korea
| | - Jung-Mi Choi
- Departments of Anatomy, Ajou University School of Medicine, Woldcup-ro 164, Yeongtong-gu, Suwon, 16499 South Korea
| | - Huy-Hyen Oh
- Departments of Anatomy, Ajou University School of Medicine, Woldcup-ro 164, Yeongtong-gu, Suwon, 16499 South Korea
| | - Narayan Bashyal
- Departments of Anatomy, Ajou University School of Medicine, Woldcup-ro 164, Yeongtong-gu, Suwon, 16499 South Korea
- Departments of Biomedical Sciences, The Graduate School, Ajou University School of Medicine, World cup-ro 164, Yeongtong-gu, Suwon, 16499 South Korea
| | - Sung-Soo Kim
- Departments of Anatomy, Ajou University School of Medicine, Woldcup-ro 164, Yeongtong-gu, Suwon, 16499 South Korea
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, 27709 NC USA
- Institute of Biomedical Research (BIOMED), School of Medical Sciences, Catholic University of Argentina, Av. Alicia Moreau de Justo 1300, Edificio San Jose Piso 3, C1107AAZ Buenos Aires, Argentina
| | - Haeyoung Suh-Kim
- Departments of Anatomy, Ajou University School of Medicine, Woldcup-ro 164, Yeongtong-gu, Suwon, 16499 South Korea
- Departments of Biomedical Sciences, The Graduate School, Ajou University School of Medicine, World cup-ro 164, Yeongtong-gu, Suwon, 16499 South Korea
| |
Collapse
|
35
|
Cholecalciferol (Vitamin D 3) Reduces Rat Neuropathic Pain by Modulating Opioid Signaling. Mol Neurobiol 2019; 56:7208-7221. [PMID: 31001801 DOI: 10.1007/s12035-019-1582-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/21/2019] [Indexed: 01/13/2023]
Abstract
The impact of vitamin D on sensory function, including pain processing, has been receiving increasing attention. Indeed, vitamin D deficiency is associated with various chronic pain conditions, and several lines of evidence indicate that vitamin D supplementation may trigger pain relief. However, the underlying mechanisms of action remain poorly understood. We used inflammatory and non-inflammatory rat models of chronic pain to evaluate the benefits of vitamin D3 (cholecalciferol) on pain symptoms. We found that cholecalciferol supplementation improved mechanical nociceptive thresholds in monoarthritic animals and reduced mechanical hyperalgesia and cold allodynia in a model of mononeuropathy. Transcriptomic analysis of cerebrum, dorsal root ganglia, and spinal cord tissues indicate that cholecalciferol supplementation induces a massive gene dysregulation which, in the cerebrum, is associated with opioid signaling (23 genes), nociception (14), and allodynia (8), and, in the dorsal root ganglia, with axonal guidance (37 genes) and nociception (17). Among the identified cerebral dysregulated nociception-, allodynia-, and opioid-associated genes, 21 can be associated with vitamin D metabolism. However, it appears that their expression is modulated by intermediate regulators such as diverse protein kinases and not, as expected, by the vitamin D receptor. Overall, several genes-Oxt, Pdyn, Penk, Pomc, Pth, Tac1, and Tgfb1-encoding for peptides/hormones stand out as top candidates to explain the therapeutic benefit of vitamin D3 supplementation. Further studies are now warranted to detail the precise mechanisms of action but also the most favorable doses and time windows for pain relief.
Collapse
|
36
|
Abstract
Although the majority of seizures in neonates are related to acute brain injury, a substantial minority are the first symptom of a neonatal-onset epilepsy often linked to a pathogenic genetic variant. Historically, studies on neonatal seizures including treatment response and long-term consequences have lumped all etiologies together. However, etiology has been consistently shown to be the most important determinant of outcome. In the past few years, an increasing number of monogenic disorders have been described and might explain up to a third of neonatal-onset epilepsy syndromes previously included under the umbrella of Ohtahara syndrome and early myoclonic encephalopathy. In this chapter, we define the concept of genetic epilepsy and review the classification. Then, we review the most relevant monogenic neonatal-onset epilepsies, detail their underlying pathophysiologic mechanisms, and present their electroclinical phenotypes. We highlight that, in some cases, such as neonates with KCNQ2 or KCNT1 gene mutations, the early recognition of the electroclinical phenotype can lead to targeted diagnostic testing and precision medicine treatment, enabling the possibility of improved outcome.
Collapse
|
37
|
Abela L, Kurian MA. Postsynaptic movement disorders: clinical phenotypes, genotypes, and disease mechanisms. J Inherit Metab Dis 2018; 41:1077-1091. [PMID: 29948482 PMCID: PMC6326993 DOI: 10.1007/s10545-018-0205-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/13/2018] [Accepted: 05/18/2018] [Indexed: 12/30/2022]
Abstract
Movement disorders comprise a group of heterogeneous diseases with often complex clinical phenotypes. Overlapping symptoms and a lack of diagnostic biomarkers may hamper making a definitive diagnosis. Next-generation sequencing techniques have substantially contributed to unraveling genetic etiologies underlying movement disorders and thereby improved diagnoses. Defects in dopaminergic signaling in postsynaptic striatal medium spiny neurons are emerging as a pathogenic mechanism in a number of newly identified hyperkinetic movement disorders. Several of the causative genes encode components of the cAMP pathway, a critical postsynaptic signaling pathway in medium spiny neurons. Here, we review the clinical presentation, genetic findings, and disease mechanisms that characterize these genetic postsynaptic movement disorders.
Collapse
Affiliation(s)
- Lucia Abela
- Molecular Neurosciences, Developmental Neuroscience, UCL Institute of Child Health, London, UK
| | - Manju A Kurian
- Molecular Neurosciences, Developmental Neuroscience, UCL Institute of Child Health, London, UK.
- Developmental Neurosciences Programme, UCL GOS - Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK.
| |
Collapse
|
38
|
Koy A, Cirak S, Gonzalez V, Becker K, Roujeau T, Milesi C, Baleine J, Cambonie G, Boularan A, Greco F, Perrigault PF, Cances C, Dorison N, Doummar D, Roubertie A, Beroud C, Körber F, Stüve B, Waltz S, Mignot C, Nava C, Maarouf M, Coubes P, Cif L. Deep brain stimulation is effective in pediatric patients with GNAO1 associated severe hyperkinesia. J Neurol Sci 2018; 391:31-39. [DOI: 10.1016/j.jns.2018.05.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/21/2018] [Indexed: 12/27/2022]
|
39
|
Gerald B, Ramsey K, Belnap N, Szelinger S, Siniard AL, Balak C, Russell M, Richholt R, De Both M, Claasen AM, Schrauwen I, Huentelman MJ, Craig DW, Rangasamy S, Narayanan V. Neonatal epileptic encephalopathy caused by de novo GNAO1 mutation misdiagnosed as atypical Rett syndrome: Cautions in interpretation of genomic test results. Semin Pediatr Neurol 2018; 26:28-32. [PMID: 29961512 DOI: 10.1016/j.spen.2017.08.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Epileptic encephalopathies are childhood brain disorders characterized by a variety of severe epilepsy syndromes that differ by the age of onset and seizure type. Until recently, the cause of many epileptic encephalopathies was unknown. Whole exome or whole genome sequencing has led to the identification of several causal genes in individuals with epileptic encephalopathy, and the list of genes has now expanded greatly. Genetic testing with epilepsy gene panels is now done quite early in the evaluation of children with epilepsy, following brain imaging, electroencephalogram, and metabolic profile. Early infantile epileptic encephalopathy (EIEE1; OMIM #308350) is the earliest of these age-dependent encephalopathies, manifesting as tonic spasms, myoclonic seizures, or partial seizures, with severely abnormal electroencephalogram, often showing a suppression-burst pattern. In this case study, we describe a 33-month-old female child with severe, neonatal onset epileptic encephalopathy. An infantile epilepsy gene panel test revealed 2 novel heterozygous variants in the MECP2 gene; a 70-bp deletion resulting in a frameshift and truncation (p.Lys377ProfsX9) thought to be pathogenic, and a 6-bp in-frame deletion (p.His371_372del), designated as a variant of unknown significance. Based on this test result, the diagnosis of atypical Rett syndrome (RTT) was made. Family-based targeted testing and segregation analysis, however, raised questions about the pathogenicity of these specific MECP2 variants. Whole exome sequencing was performed in this family trio, leading to the discovery of a rare, de novo, missense mutation in GNAO1 (p. Leu284Ser). De novo, heterozygous mutations in GNAO1 have been reported to cause early infantile epileptic encephalopathy-17 (EIEE17; OMIM 615473). The child's severe phenotype, the family history and segregation analysis of variants and prior reports of GNAO1-linked disease allowed us to conclude that the GNAO1 mutation, and not the MECP2 variants, was the cause of this child's neurological disease. With the increased use of genetic panels and whole exome sequencing, we will be confronted with lists of gene variants suspected to be pathogenic or of unknown significance. It is important to integrate clinical information, genetic testing that includes family members and correlates this with the published clinical and scientific literature, to help one arrive at the correct genetic diagnosis.
Collapse
Affiliation(s)
- Brittany Gerald
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ; School of Life Sciences, Arizona State University, Tempe, AZ
| | - Keri Ramsey
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Newell Belnap
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Szabolcs Szelinger
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Ashley L Siniard
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Chris Balak
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Megan Russell
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Ryan Richholt
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Matt De Both
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Ana M Claasen
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Isabelle Schrauwen
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Matthew J Huentelman
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ
| | - David W Craig
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Sampathkumar Rangasamy
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ.
| | - Vinodh Narayanan
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ; School of Life Sciences, Arizona State University, Tempe, AZ.
| |
Collapse
|
40
|
Genome-wide association study identifies genes associated with neuropathy in patients with head and neck cancer. Sci Rep 2018; 8:8789. [PMID: 29884837 PMCID: PMC5993794 DOI: 10.1038/s41598-018-27070-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/21/2018] [Indexed: 12/18/2022] Open
Abstract
Neuropathic pain (NP), defined as pain initiated or caused by a primary lesion or dysfunction in the nervous system, is a debilitating chronic pain condition often resulting from cancer treatment. Among cancer patients, neuropathy during cancer treatment is a predisposing event for NP. To identify genetic variants influencing the development of NP, we conducted a genome-wide association study in 1,043 patients with squamous cell carcinoma of the head and neck, based on 714,494 tagging single-nucleotide polymorphisms (SNPs) (130 cases, 913 controls). About 12.5% of the patients, who previously had cancer treatment, had neuropathy-associated diagnoses, as defined using the ICD-9/ICD-10 codes. We identified four common SNPs representing four genomic regions: 7q22.3 (rs10950641; SNX8; P = 3.39 × 10−14), 19p13.2 (rs4804217; PCP2; P = 2.95 × 10−9), 3q27.3 (rs6796803; KNG1; P = 6.42 × 10−9) and 15q22.2 (rs4775319; RORA; P = 1.02 × 10−8), suggesting SNX8, PCP2, KNG1 and RORA might be novel target genes for NP in patients with head and neck cancer. Future experimental validation to explore physiological effects of the identified SNPs will provide a better understanding of the biological mechanisms underlying NP and may provide insights into novel therapeutic targets for treatment and management of NP.
Collapse
|
41
|
Feng H, Khalil S, Neubig RR, Sidiropoulos C. A mechanistic review on GNAO1-associated movement disorder. Neurobiol Dis 2018; 116:131-141. [PMID: 29758257 DOI: 10.1016/j.nbd.2018.05.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/28/2018] [Accepted: 05/10/2018] [Indexed: 02/07/2023] Open
Abstract
Mutations in the GNAO1 gene cause a complex constellation of neurological disorders including epilepsy, developmental delay, and movement disorders. GNAO1 encodes Gαo, the α subunit of Go, a member of the Gi/o family of heterotrimeric G protein signal transducers. Go is the most abundant membrane protein in the mammalian central nervous system and plays major roles in synaptic neurotransmission and neurodevelopment. GNAO1 mutations were first reported in early infantile epileptic encephalopathy 17 (EIEE17) but are also associated with a more common syndrome termed neurodevelopmental disorder with involuntary movements (NEDIM). Here we review a mechanistic model in which loss-of-function (LOF) GNAO1 alleles cause epilepsy and gain-of-function (GOF) alleles are primarily associated with movement disorders. We also develop a signaling framework related to cyclic AMP (cAMP), synaptic vesicle release, and neural development and discuss gene mutations perturbing those mechanisms in a range of genetic movement disorders. Finally, we analyze clinical reports of patients carrying GNAO1 mutations with respect to their symptom onset and discuss pharmacological/surgical treatments in the context of our mechanistic model.
Collapse
Affiliation(s)
- Huijie Feng
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Suad Khalil
- Department of Neurology & Ophthalmology, Michigan State University, East Lansing, MI 48824, USA
| | - Richard R Neubig
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA.
| | - Christos Sidiropoulos
- Department of Neurology & Ophthalmology, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
42
|
Baron J, Bilbao A, Hörtnagl H, Birnbaumer L, Leixner S, Spanagel R, Ahnert-Hilger G, Brunk I. Balance of Go1α and Go2α expression regulates motor function via the striatal dopaminergic system. J Neurochem 2018; 146:374-389. [PMID: 29747224 DOI: 10.1111/jnc.14460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/13/2018] [Accepted: 04/19/2018] [Indexed: 11/27/2022]
Abstract
The heterotrimeric G-protein Go with its splice variants, Go1α and Go2α, seems to be involved in the regulation of motor function but isoform-specific effects are still unclear. We found that Go1α-/- knockouts performed worse on the rota-rod than Go2α-/- and wild-type (WT) mice. In Go1+2α-/- mice motor function was partially recovered. Furthermore, Go1+2α-/- mice showed an increased spontaneous motor activity. Compared to wild types or Go2α-/- mice, Go1+2α-/- mice developed increased behavioural sensitization following repetitive cocaine treatment, but failed to develop conditioned place preference. Analysis of dopamine concentration and expression of D1 and D2 receptors unravelled splice-variant-specific imbalances in the striatal dopaminergic system: In Go1α-/- mice dopamine concentration and vesicular monoamine uptake were increased compared to wild types. The expression of the D2 receptor was higher in Go1α-/- compared to wild type littermates, but unchanged in Go2α-/- mice. Deletion of both Go1α and Go2α re-established both dopamine and D2 receptor levels comparable to those in the wild-type. Cocaine treatment had no effect on the ratio of D1 receptor to D2 receptor in Go1+2α-/- mutants, but decreased this ratio in Go2α-/- mice. Finally, we observed that the deletion of Go1α led to a threefold higher striatal expression of Go2α. Taken together our data suggest that a balance in the expression of Go1α and Go2α sustains normal motor function. Deletion of either splice variant results in divergent behavioural and molecular alterations in the striatal dopaminergic system. Deletion of both splice variants partially restores the behavioural and molecular changes. Open Data: Materials are available on https://cos.io/our-services/open-science-badges/ https://osf.io/93n6m/.
Collapse
Affiliation(s)
- Jens Baron
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin, Berlin, Germany
| | - Ainhoa Bilbao
- Medical Faculty, Behavioural Genetics Research Group, Institute of Psychopharmacology, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Heide Hörtnagl
- Department of Pharmacology, Innsbruck Medical University, Innsbruck, Austria
| | - Lutz Birnbaumer
- School of Medical Sciences, Catholic University of Argentina, Institute of Biomedical Research (BIOMED UCA-CONICET), Buenos Aires, Argentina
| | - Sarah Leixner
- Medical Faculty, Behavioural Genetics Research Group, Institute of Psychopharmacology, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Rainer Spanagel
- Medical Faculty, Institute of Psychopharmacology, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Gudrun Ahnert-Hilger
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin, Berlin, Germany
| | - Irene Brunk
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
43
|
Xiong J, Peng J, Duan HL, Chen C, Wang XL, Chen SM, Yin F. [Recurrent convulsion and pulmonary infection complicated by psychomotor retardation in an infant]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20:154-157. [PMID: 29429466 PMCID: PMC7389246 DOI: 10.7499/j.issn.1008-8830.2018.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/11/2018] [Indexed: 06/08/2023]
Abstract
A 4-month-old girl developed convulsion in the neonatal period, which was focal motor seizures in the initial stage and later became spasm and tonic spasm. And the girl also had psychomotor retardation and recurrent pulmonary infection. Electroencephalography showed hypsarrhythmia, normal results were obtained from cranial magnetic resonance imaging, cerebrospinal fluid examination, and urine organic acid analysis, as well as the spectral analyses of blood ammonia, blood lactic acid, blood amino acids, and acylcarnitines. Gene detection revealed a de novo heterozygous mutation, c.607G>A (p.G203R) , in GNAO1. The girl was then diagnosed with GNAO1-associated early infantile epileptic encephalopathy (EIEE type 17). The seizures were well controlled by topiramate and vigabatrin, but there was no improvement in psychomotor development. She also suffered from recurrent pulmonary infection and died at the age of 12 months due to severe pneumonia. For children with unexplained early infantile epileptic encephalopathy, GNAO1 gene mutations should be considered and genetic tests should be performed as early as possible. Recurrent pulmonary infection should also be taken seriously.
Collapse
Affiliation(s)
- Juan Xiong
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China.
| | | | | | | | | | | | | |
Collapse
|
44
|
Presence of Androgen Receptor Variant in Neuronal Lipid Rafts. eNeuro 2017; 4:eN-NWR-0109-17. [PMID: 28856243 PMCID: PMC5575139 DOI: 10.1523/eneuro.0109-17.2017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 08/03/2017] [Accepted: 08/14/2017] [Indexed: 11/21/2022] Open
Abstract
Fast, nongenomic androgen actions have been described in various cell types, including neurons. However, the receptor mediating this cell membrane–initiated rapid signaling remains unknown. This study found a putative androgen receptor splice variant in a dopaminergic N27 cell line and in several brain regions (substantia nigra pars compacta, entorhinal cortex, and hippocampus) from gonadally intact and gonadectomized (young and middle-aged) male rats. This putative splice variant protein has a molecular weight of 45 kDa and lacks an N-terminal domain, indicating it is homologous to the human AR45 splice variant. Interestingly, AR45 was highly expressed in all brain regions examined. In dopaminergic neurons, AR45 is localized to plasma membrane lipid rafts, a microdomain involved in cellular signaling. Further, AR45 protein interacts with membrane-associated G proteins Gαq and Gαo. Neither age nor hormone levels altered AR45 expression in dopaminergic neurons. These results provide the first evidence of AR45 protein expression in the brain, specifically plasma membrane lipid rafts. AR45 presence in lipid rafts indicates that it may function as a membrane androgen receptor to mediate fast, nongenomic androgen actions.
Collapse
|
45
|
Feng H, Sjögren B, Karaj B, Shaw V, Gezer A, Neubig RR. Movement disorder in GNAO1 encephalopathy associated with gain-of-function mutations. Neurology 2017; 89:762-770. [PMID: 28747448 DOI: 10.1212/wnl.0000000000004262] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 04/17/2017] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To define molecular mechanisms underlying the clinical spectrum of epilepsy and movement disorder in individuals with de novo mutations in the GNAO1 gene. METHODS We identified all GNAO1 mutations reported in individuals with epilepsy (early infantile epileptiform encephalopathy 17) or movement disorders through April 2016; 15 de novo mutant alleles from 25 individuals were introduced into the Gαo subunit by site-directed mutagenesis in a mammalian expression plasmid. We assessed protein expression and function in vitro in HEK-293T cells by Western blot and determined functional Gαo-dependent cyclic adenosine monophosphate (cAMP) inhibition with a coexpressed α2A adrenergic receptor. RESULTS Of the 15 clinical GNAO1 mutations studied, 9 show reduced expression and loss of function (LOF; <90% maximal inhibition). Six other mutations show variable levels of expression but exhibit normal or even gain-of-function (GOF) behavior, as demonstrated by significantly lower EC50 values for α2A adrenergic receptor-mediated inhibition of cAMP. The GNAO1 LOF mutations are associated with epileptic encephalopathy while GOF mutants (such as G42R, G203R, and E246K) or normally functioning mutants (R209) were found in patients with movement disorders with or without seizures. CONCLUSIONS Both LOF and GOF mutations in Gαo (encoded by GNAO1) are associated with neurologic pathophysiology. There appears to be a strong predictive correlation between the in vitro biochemical phenotype and the clinical pattern of epilepsy vs movement disorder.
Collapse
Affiliation(s)
- Huijie Feng
- From the Department of Pharmacology & Toxicology, Michigan State University, East Lansing
| | - Benita Sjögren
- From the Department of Pharmacology & Toxicology, Michigan State University, East Lansing
| | - Behirda Karaj
- From the Department of Pharmacology & Toxicology, Michigan State University, East Lansing
| | - Vincent Shaw
- From the Department of Pharmacology & Toxicology, Michigan State University, East Lansing
| | - Aysegul Gezer
- From the Department of Pharmacology & Toxicology, Michigan State University, East Lansing
| | - Richard R Neubig
- From the Department of Pharmacology & Toxicology, Michigan State University, East Lansing.
| |
Collapse
|
46
|
Sakamoto S, Monden Y, Fukai R, Miyake N, Saito H, Miyauchi A, Matsumoto A, Nagashima M, Osaka H, Matsumoto N, Yamagata T. A case of severe movement disorder with GNAO1 mutation responsive to topiramate. Brain Dev 2017; 39:439-443. [PMID: 27916449 DOI: 10.1016/j.braindev.2016.11.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 11/15/2016] [Accepted: 11/16/2016] [Indexed: 11/15/2022]
Abstract
We report the case of a 19-year-old female patient who had progressive chorea associated with a GNAO1 mutation. Chorea was refractory to multiple anticonvulsants, and the patient suffered from tiapride-induced neuroleptic malignant syndrome. After identification of a GNAO1 missense mutation at the age of 18years, topiramate treatment was initiated and the frequency of chorea decreased dramatically. The efficacy of topiramate may have been related to the inhibitory modulation of voltage-activated Ca2+ channels. Given the side effects and complications associated with neuroleptics and deep brain stimulation, respectively, topiramate is recommended for the first-line management of severe chorea associated with a GNAO1 mutation.
Collapse
Affiliation(s)
- Saori Sakamoto
- Department of Pediatrics, Jichi Medical University, Tochigi, Japan
| | - Yukifumi Monden
- Department of Pediatrics, Jichi Medical University, Tochigi, Japan; Department of Pediatrics, International University of Health and Welfare, 537-3 Iguchi, Shiobara, Tochigi 329-2763, Japan.
| | - Ryoko Fukai
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan; Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroshi Saito
- Department of Pediatrics, Jichi Medical University, Tochigi, Japan
| | - Akihiko Miyauchi
- Department of Pediatrics, Jichi Medical University, Tochigi, Japan
| | - Ayumi Matsumoto
- Department of Pediatrics, Jichi Medical University, Tochigi, Japan
| | - Masako Nagashima
- Department of Pediatrics, Jichi Medical University, Tochigi, Japan
| | - Hitoshi Osaka
- Department of Pediatrics, Jichi Medical University, Tochigi, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | | |
Collapse
|
47
|
Danti FR, Galosi S, Romani M, Montomoli M, Carss KJ, Raymond FL, Parrini E, Bianchini C, McShane T, Dale RC, Mohammad SS, Shah U, Mahant N, Ng J, McTague A, Samanta R, Vadlamani G, Valente EM, Leuzzi V, Kurian MA, Guerrini R. GNAO1 encephalopathy: Broadening the phenotype and evaluating treatment and outcome. Neurol Genet 2017; 3:e143. [PMID: 28357411 PMCID: PMC5362187 DOI: 10.1212/nxg.0000000000000143] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 02/13/2017] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To describe better the motor phenotype, molecular genetic features, and clinical course of GNAO1-related disease. METHODS We reviewed clinical information, video recordings, and neuroimaging of a newly identified cohort of 7 patients with de novo missense and splice site GNAO1 mutations, detected by next-generation sequencing techniques. RESULTS Patients first presented in early childhood (median age of presentation 10 months, range 0-48 months), with a wide range of clinical symptoms ranging from severe motor and cognitive impairment with marked choreoathetosis, self-injurious behavior, and epileptic encephalopathy to a milder phenotype, featuring moderate developmental delay associated with complex stereotypies, mainly facial dyskinesia and mild epilepsy. Hyperkinetic movements were often exacerbated by specific triggers, such as voluntary movement, intercurrent illnesses, emotion, and high ambient temperature, leading to hospital admissions. Most patients were resistant to drug intervention, although tetrabenazine was effective in partially controlling dyskinesia for 2/7 patients. Emergency deep brain stimulation (DBS) was life saving in 1 patient, resulting in immediate clinical benefit with complete cessation of violent hyperkinetic movements. Five patients had well-controlled epilepsy and 1 had drug-resistant seizures. Structural brain abnormalities, including mild cerebral atrophy and corpus callosum dysgenesis, were evident in 5 patients. One patient had a diffuse astrocytoma (WHO grade II), surgically removed at age 16. CONCLUSIONS Our findings support the causative role of GNAO1 mutations in an expanded spectrum of early-onset epilepsy and movement disorders, frequently exacerbated by specific triggers and at times associated with self-injurious behavior. Tetrabenazine and DBS were the most useful treatments for dyskinesia.
Collapse
Affiliation(s)
- Federica Rachele Danti
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Serena Galosi
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Marta Romani
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Martino Montomoli
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Keren J Carss
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - F Lucy Raymond
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Elena Parrini
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Claudia Bianchini
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Tony McShane
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Russell C Dale
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Shekeeb S Mohammad
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Ubaid Shah
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Neil Mahant
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Joanne Ng
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Amy McTague
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Rajib Samanta
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Gayatri Vadlamani
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Enza Maria Valente
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Vincenzo Leuzzi
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Manju A Kurian
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Renzo Guerrini
- Department of Paediatrics, Child Neurology and Psychiatry (F.R.D., S.G., V.L.), Sapienza University of Rome, Italy; Molecular Neurosciences, Developmental Neurosciences Programme (F.R.D., J.N., A.M., M.A.K.), University College London Institute of Child Health, UK; Department of Neurology (F.R.D., J.N., A.M., M.A.K.), Great Ormond Street Hospital for Children, London, UK; GENOMA Group (M.R.), Molecular Genetics Laboratory, Rome, Italy; Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (M.M., E.P., C.B., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence, Italy; Department of Haematology (K.J.C.), University of Cambridge, NHS Blood and Transplant Centre, UK; NIHR Bioresource Rare Diseases (K.J.C., F.L.R.), University of Cambridge, UK; Department of Neurology (N.M.), Westmead Hospital, Sydney, Australia; Childrens Hospital Oxford (T.M.), John Radcliffe Hospital, UK; Institute for Neuroscience and Muscle Research (R.C.D., S.S.M., U.S.), the Children's Hospital at Westmead, University of Sydney, Australia; Department of Medical Genetics (F.L.R.), Cambridge Institute for Medical Research, University of Cambridge, UK; Department of Neurology (R.S.), University Hospitals Leicester NHS Trust, UK; Department of Paediatric Neurology (G.V.), Leeds Teaching Hospitals NHS Trust, UK; Section of Neurosciences (E.M.V.), Department of Medicine and Surgery, University of Salerno, Italy; and Neurogenetics Unit (E.M.V.), IRCCS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
48
|
Sanna MD, Ghelardini C, Galeotti N. HuD-mediated distinct BDNF regulatory pathways promote regeneration after nerve injury. Brain Res 2017; 1659:55-63. [DOI: 10.1016/j.brainres.2017.01.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/13/2017] [Accepted: 01/14/2017] [Indexed: 11/30/2022]
|
49
|
Development of the main olfactory system and main olfactory epithelium-dependent male mating behavior are altered in Go-deficient mice. Proc Natl Acad Sci U S A 2016; 113:10974-9. [PMID: 27625425 DOI: 10.1073/pnas.1613026113] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In mammals, initial detection of olfactory stimuli is mediated by sensory neurons in the main olfactory epithelium (MOE) and the vomeronasal organ (VNO). The heterotrimeric GTP-binding protein Go is widely expressed in the MOE and VNO of mice. Early studies indicated that Go expression in VNO sensory neurons is critical for directing social and sexual behaviors in female mice [Oboti L, et al. (2014) BMC Biol 12:31]. However, the physiological functions of Go in the MOE have remained poorly defined. Here, we examined the role of Go in the MOE using mice lacking the α subunit of Go Development of the olfactory bulb (OB) was perturbed in mutant mice as a result of reduced neurogenesis and increased cell death. The balance between cell types of OB interneurons was altered in mutant mice, with an increase in the number of tyrosine hydroxylase-positive interneurons at the expense of calbindin-positive interneurons. Sexual behavior toward female mice and preference for female urine odors by olfactory sensory neurons in the MOE were abolished in mutant male mice. Our data suggest that Go signaling is essential for the structural and functional integrity of the MOE and for specification of OB interneurons, which in turn are required for the transmission of pheromone signals and the initiation of mating behavior with the opposite sex.
Collapse
|
50
|
Ramírez VT, Ramos-Fernández E, Henríquez JP, Lorenzo A, Inestrosa NC. Wnt-5a/Frizzled9 Receptor Signaling through the Gαo-Gβγ Complex Regulates Dendritic Spine Formation. J Biol Chem 2016; 291:19092-107. [PMID: 27402827 DOI: 10.1074/jbc.m116.722132] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Indexed: 02/04/2023] Open
Abstract
Wnt ligands play crucial roles in the development and regulation of synapse structure and function. Specifically, Wnt-5a acts as a secreted growth factor that regulates dendritic spine formation in rodent hippocampal neurons, resulting in postsynaptic development that promotes the clustering of the PSD-95 (postsynaptic density protein 95). Here, we focused on the early events occurring after the interaction between Wnt-5a and its Frizzled receptor at the neuronal cell surface. Additionally, we studied the role of heterotrimeric G proteins in Wnt-5a-dependent synaptic development. We report that FZD9 (Frizzled9), a Wnt receptor related to Williams syndrome, is localized in the postsynaptic region, where it interacts with Wnt-5a. Functionally, FZD9 is required for the Wnt-5a-mediated increase in dendritic spine density. FZD9 forms a precoupled complex with Gαo under basal conditions that dissociates after Wnt-5a stimulation. Accordingly, we found that G protein inhibition abrogates the Wnt-5a-dependent pathway in hippocampal neurons. In particular, the activation of Gαo appears to be a key factor controlling the Wnt-5a-induced dendritic spine density. In addition, we found that Gβγ is required for the Wnt-5a-mediated increase in cytosolic calcium levels and spinogenesis. Our findings reveal that FZD9 and heterotrimeric G proteins regulate Wnt-5a signaling and dendritic spines in cultured hippocampal neurons.
Collapse
Affiliation(s)
- Valerie T Ramírez
- From the Centro de Envejecimiento y Regeneración, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - Eva Ramos-Fernández
- From the Centro de Envejecimiento y Regeneración, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - Juan Pablo Henríquez
- the Laboratorio de Neurobiología del Desarrollo, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Núcleo Milenio de Biología Regenerativa, Centro de Microscopía Avanzada, Universidad de Concepción, 4089100 Concepción, Chile
| | - Alfredo Lorenzo
- the Laboratorio de Neuropatología Experimental, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, 5016 Córdoba, Argentina
| | - Nibaldo C Inestrosa
- From the Centro de Envejecimiento y Regeneración, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile, the Center for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, 2031 New South Wales, Australia, and the Centro de Excelencia en Biomedicina de Magallanes, Universidad de Magallanes, 6200000 Punta Arenas, Chile
| |
Collapse
|