1
|
van Hoof S, Kreye J, Cordero-Gómez C, Hoffmann J, Momsen Reincke S, Sánchez-Sendin E, Duong SL, Upadhya M, Dhangar D, Michór P, Woodhall GL, Küpper M, Oder A, Kuchling J, Koch SP, Mueller S, Boehm-Sturm P, von Kries JP, Finke C, Kirschstein T, Wright SK, Prüss H. Human cerebrospinal fluid monoclonal CASPR2 autoantibodies induce changes in electrophysiology, functional MRI, and behavior in rodent models. Brain Behav Immun 2024; 122:266-278. [PMID: 39142424 DOI: 10.1016/j.bbi.2024.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/02/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024] Open
Abstract
Anti-contactin associated protein receptor 2 (CASPR2) encephalitis is a severe autoimmune encephalitis with a variable clinical phenotype including behavioral abnormalities, cognitive decline, epileptic seizures, peripheral nerve hyperexcitability and neuropathic pain. The detailed mechanisms of how CASPR2 autoantibodies lead to synaptic dysfunction and clinical symptoms are largely unknown. Aiming for analyses from the molecular to the clinical level, we isolated antibody-secreting cells from the cerebrospinal fluid of two patients with CASPR2 encephalitis. From these we cloned four anti-CASPR2 human monoclonal autoantibodies (mAbs) with strong binding to brain and peripheral nerves. All were highly hypermutated and mainly of the IgG4 subclass. Mutagenesis studies determined selective binding to the discoidin domain of CASPR2. Surface plasmon resonance revealed affinities with dissociation constants KD in the pico- to nanomolar range. CASPR2 mAbs interrupted the interaction of CASPR2 with its binding partner contactin 2 in vitro and were internalized after binding to CASPR2-expressing cells. Electrophysiological recordings of rat hippocampal slices after stereotactic injection of CASPR2 mAbs showed characteristic afterpotentials following electrical stimulation. In vivo experiments with intracerebroventricular administration of human CASPR2 mAbs into mice and rats showed EEG-recorded brain hyperexcitability but no spontaneous recurrent seizures. Behavioral assessment of infused mice showed a subtle clinical phenotype, mainly affecting sociability. Mouse brain MRI exhibited markedly reduced resting-state functional connectivity without short-term structural changes. Together, the experimental data support the direct pathogenicity of CASPR2 autoantibodies. The minimally invasive EEG and MRI techniques applied here may serve as novel objective, quantifiable tools for improved animal models, in particular for subtle neuropsychiatric phenotypes or repeated measurements.
Collapse
Affiliation(s)
- Scott van Hoof
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Helmholtz Innovation Lab BaoBab (Brain Antibody-omics and B-cell Lab), Berlin, Germany
| | - Jakob Kreye
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Helmholtz Innovation Lab BaoBab (Brain Antibody-omics and B-cell Lab), Berlin, Germany; Department of Pediatric Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - César Cordero-Gómez
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Julius Hoffmann
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - S Momsen Reincke
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Elisa Sánchez-Sendin
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Helmholtz Innovation Lab BaoBab (Brain Antibody-omics and B-cell Lab), Berlin, Germany
| | - Sophie L Duong
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Manoj Upadhya
- Institute of Health and Neurodevelopment, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Divya Dhangar
- Institute of Health and Neurodevelopment, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Paulina Michór
- Institute of Health and Neurodevelopment, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Gavin L Woodhall
- Institute of Health and Neurodevelopment, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Maraike Küpper
- Oscar Langendorff Institute of Physiology, University of Rostock, Germany, Center of Transdisciplinary Neurosciences Rostock (CTNR), Germany
| | - Andreas Oder
- Screening Unit, Leibniz Forschungsinstitut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Joseph Kuchling
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Neurocure Cluster of Excellence, NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
| | - Stefan Paul Koch
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Center for Stroke Research Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité - Universitätsmedizin Berlin, Germany; Charité 3R, Replace, Reduce, Refine, Charité - Universitätsmedizin Berlin, Germany
| | - Susanne Mueller
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Center for Stroke Research Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité - Universitätsmedizin Berlin, Germany; Charité 3R, Replace, Reduce, Refine, Charité - Universitätsmedizin Berlin, Germany
| | - Philipp Boehm-Sturm
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Center for Stroke Research Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité - Universitätsmedizin Berlin, Germany; Charité 3R, Replace, Reduce, Refine, Charité - Universitätsmedizin Berlin, Germany
| | - Jens Peter von Kries
- Screening Unit, Leibniz Forschungsinstitut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Carsten Finke
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Neurocure Cluster of Excellence, NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University of Rostock, Germany, Center of Transdisciplinary Neurosciences Rostock (CTNR), Germany
| | - Sukhvir K Wright
- Institute of Health and Neurodevelopment, College of Health and Life Sciences, Aston University, Birmingham, UK; Department of Paediatric Neurology, The Birmingham Women's and Children's Hospital National Health Service Foundation Trust, Birmingham, UK
| | - Harald Prüss
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany; Helmholtz Innovation Lab BaoBab (Brain Antibody-omics and B-cell Lab), Berlin, Germany.
| |
Collapse
|
2
|
Biagioni M, Baronchelli F, Fossati M. Multiscale spatio-temporal dynamics of UBE3A gene in brain physiology and neurodevelopmental disorders. Neurobiol Dis 2024; 201:106669. [PMID: 39293689 DOI: 10.1016/j.nbd.2024.106669] [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: 07/30/2024] [Revised: 09/13/2024] [Accepted: 09/15/2024] [Indexed: 09/20/2024] Open
Abstract
The UBE3A gene, located in the chromosomal region 15q11-13, is subject to neuron-specific genomic imprinting and it plays a critical role in brain development. Genetic defects of UBE3A cause severe neurodevelopmental disorders, namely the Angelman syndrome (AS) and the 15q11.2-q13.3 duplication syndrome (Dup15q). In the last two decades, the development of in vitro and in vivo models of AS and Dup15q were fundamental to improve the understanding of UBE3A function in the brain. However, the pathogenic mechanisms of these diseases remain elusive and effective treatments are lacking. Recent evidence suggests that UBE3A functions are both spatially and temporally specific, varying across subcellular compartments, brain regions, and neuronal circuits. In the present review, we summarize current knowledge on the role of UBE3A in neuronal pathophysiology under this spatio-temporal perspective. Additionally, we propose key research questions that will be instrumental to better understand the pathogenic mechanisms underpinning AS and Dup15q disorders and provide the rationale to develop novel therapies.
Collapse
Affiliation(s)
- Martina Biagioni
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano 20089, MI, Italy
| | - Federica Baronchelli
- CNR - Institute of Neuroscience, Section of Milano, via Manzoni 56, Rozzano 20089, MI, Italy; Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini, 20072 Pieve Emanuele, MI, Italy
| | - Matteo Fossati
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano 20089, MI, Italy; CNR - Institute of Neuroscience, Section of Milano, via Manzoni 56, Rozzano 20089, MI, Italy.
| |
Collapse
|
3
|
Huie EZ, Yang X, Rioult-Pedotti MS, Naik M, Huang YWA, Silverman JL, Marshall J. Peptidomimetic inhibitors targeting TrkB/PSD-95 signaling improves cognition and seizure outcomes in an Angelman Syndrome mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.597833. [PMID: 38895218 PMCID: PMC11185757 DOI: 10.1101/2024.06.07.597833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Angelman syndrome (AS) is a rare genetic neurodevelopmental disorder with profoundly debilitating symptoms with no FDA-approved cure or therapeutic. Brain-derived neurotrophic factor (BDNF), and its receptor TrkB, have a well-established role as regulators of synaptic plasticity, dendritic outgrowth, dendritic spine formation and maintenance. Previously, we reported that the association of PSD-95 with TrkB is critical for intact BDNF signaling in the AS mouse model, as illustrated by attenuated PLCγ and PI3K signaling and intact MAPK pathway signaling. These data suggest that drugs tailored to enhance the TrkB-PSD-95 interaction may provide a novel approach for the treatment of AS and a variety of NDDs. To evaluate this critical interaction, we synthesized a class of high-affinity PSD-95 ligands that bind specifically to the PDZ3 domain of PSD-95, denoted as Syn3 peptidomimetic ligands. We evaluated Syn3 and its analog D-Syn3 (engineered using dextrorotary (D)-amino acids) in vivo using the Ube3a exon 2 deletion mouse model of AS. Following systemic administration of Syn3 and D-Syn3, we demonstrated improvement in the seizure domain of AS. Learning and memory using the novel object recognition assay also illustrated improved cognition following Syn3 and D-Syn3, along with restored long-term potentiation. Finally, D-Syn3 treated mice showed a partial rescue in motor learning. Neither Syn3 nor D-Syn3 improved gross exploratory locomotion deficits, nor gait impairments that have been well documented in the AS rodent models. These findings highlight the need for further investigation of this compound class as a potential therapeutic for AS and other genetic NDDs.
Collapse
|
4
|
Qu S, Wang J, Guan X, Song C, Wang Y. Sleep disturbance in Angelman syndrome patients. Orphanet J Rare Dis 2024; 19:146. [PMID: 38580983 PMCID: PMC10996173 DOI: 10.1186/s13023-024-03154-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/28/2024] [Indexed: 04/07/2024] Open
Abstract
Angelman syndrome (AS) is a neurodevelopmental disorder caused by abnormal expression of the maternal ubiquitin protein ligase E3A gene (UBE3A). As one of the most challenging symptoms and important focuses of new treatment, sleep disturbance is reported to occur in 70-80% of patients with AS and has a serious impact on the lives of patients and their families. Although clinical studies and animal model studies have provided some clues, recent research into sleep disorders in the context of AS is still very limited. It is generally accepted that there is an interaction between neurodevelopment and sleep; however, there is no recognized mechanism for sleep disorders in AS patients. Accordingly, there are no aetiologically specific clinical treatments for AS-related sleep disorders. The most common approaches involve ameliorating symptoms through methods such as behavioural therapy and symptomatic pharmacotherapy. In recent years, preclinical and clinical studies on the targeted treatment of AS have emerged. Although precision therapy for restoring the UBE3A level and the function of its signalling pathways is inevitably hindered by many remaining obstacles, this approach has the potential to address AS-related sleep disturbance.
Collapse
Affiliation(s)
- Song Qu
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing, China
| | - Junyi Wang
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xingying Guan
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing, China
| | - Cui Song
- Department of Endocrinology and Genetic Metabolism Disease, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China.
| | - Yanyan Wang
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing, China.
| |
Collapse
|
5
|
Clarke MT, Remesal L, Lentz L, Tan DJ, Young D, Thapa S, Namuduri SR, Borges B, Kirn G, Valencia J, Lopez ME, Lui JH, Shiow LR, Dindot S, Villeda S, Sanders SJ, MacKenzie TC. Prenatal delivery of a therapeutic antisense oligonucleotide achieves broad biodistribution in the brain and ameliorates Angelman syndrome phenotype in mice. Mol Ther 2024; 32:935-951. [PMID: 38327047 PMCID: PMC11163203 DOI: 10.1016/j.ymthe.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 11/20/2023] [Accepted: 02/02/2024] [Indexed: 02/09/2024] Open
Abstract
Angelman syndrome (AS), an early-onset neurodevelopmental disorder characterized by abnormal gait, intellectual disabilities, and seizures, occurs when the maternal allele of the UBE3A gene is disrupted, since the paternal allele is silenced in neurons by the UBE3A antisense (UBE3A-AS) transcript. Given the importance of early treatment, we hypothesized that prenatal delivery of an antisense oligonucleotide (ASO) would downregulate the murine Ube3a-AS, resulting in increased UBE3A protein and functional rescue. Using a mouse model with a Ube3a-YFP allele that reports on-target ASO activity, we found that in utero, intracranial (IC) injection of the ASO resulted in dose-dependent activation of paternal Ube3a, with broad biodistribution. Accordingly, in utero injection of the ASO in a mouse model of AS also resulted in successful restoration of UBE3A and phenotypic improvements in treated mice on the accelerating rotarod and fear conditioning. Strikingly, even intra-amniotic (IA) injection resulted in systemic biodistribution and high levels of UBE3A reactivation throughout the brain. These findings offer a novel strategy for early treatment of AS using an ASO, with two potential routes of administration in the prenatal window. Beyond AS, successful delivery of a therapeutic ASO into neurons has implications for a clinically feasible prenatal treatment for numerous neurodevelopmental disorders.
Collapse
Affiliation(s)
- Maria T Clarke
- Department of Surgery, University of California San Francisco, San Francisco, California, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, California, USA; Center for Maternal-Fetal Precision Medicine, University of California San Francisco, San Francisco, California, USA
| | - Laura Remesal
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA
| | - Lea Lentz
- Department of Surgery, University of California San Francisco, San Francisco, California, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, California, USA; Center for Maternal-Fetal Precision Medicine, University of California San Francisco, San Francisco, California, USA
| | | | - David Young
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, California, USA; Institute for Molecular and Cell Biology, Agency for Science, Technology and Research, 138632, Singapore, Singapore
| | - Slesha Thapa
- BioMarin Pharmaceutical, San Rafael, California, USA
| | - Shalini R Namuduri
- Department of Surgery, University of California San Francisco, San Francisco, California, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, California, USA; Center for Maternal-Fetal Precision Medicine, University of California San Francisco, San Francisco, California, USA
| | - Beltran Borges
- Department of Surgery, University of California San Francisco, San Francisco, California, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, California, USA; Center for Maternal-Fetal Precision Medicine, University of California San Francisco, San Francisco, California, USA
| | - Georgia Kirn
- Department of Surgery, University of California San Francisco, San Francisco, California, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, California, USA; Center for Maternal-Fetal Precision Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jasmine Valencia
- Department of Surgery, University of California San Francisco, San Francisco, California, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, California, USA
| | | | - Jan H Lui
- BioMarin Pharmaceutical, San Rafael, California, USA
| | | | - Scott Dindot
- Department of Veterinary Pathobiology, School of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Saul Villeda
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, California, USA; Department of Anatomy, University of California San Francisco, San Francisco, California, USA
| | - Stephan J Sanders
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, California, USA; Institute of Developmental and Regenerative Medicine, Department of Paediatrics, University of Oxford, Oxford OX3 7TY, United Kingdom
| | - Tippi C MacKenzie
- Department of Surgery, University of California San Francisco, San Francisco, California, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, California, USA; Center for Maternal-Fetal Precision Medicine, University of California San Francisco, San Francisco, California, USA.
| |
Collapse
|
6
|
Fitzgerald PJ. Neural hyperexcitability in Angelman syndrome: Genetic factors and pharmacologic treatment approaches. Epilepsy Res 2024; 200:107286. [PMID: 38217951 DOI: 10.1016/j.eplepsyres.2024.107286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/18/2023] [Accepted: 01/02/2024] [Indexed: 01/15/2024]
Abstract
Angelman syndrome (AS) is a rare neurodevelopmental disorder that is typically caused by deletion or a loss-of-function mutation of the maternal copy of the ubiquitin ligase E3A (UBE3A) gene. The disorder is characterized by severe intellectual disability, deficits in speech, motor abnormalities, altered electroencephalography (EEG) activity, spontaneous epileptic seizures, sleep disturbances, and a happy demeanor with frequent laughter. Regarding electrophysiologic abnormalities in particular, enhanced delta oscillatory power and an elevated excitatory/inhibitory (E/I) ratio have been documented in AS, with E/I ratio especially studied in rodent models. These electrophysiologic characteristics appear to relate with the greatly elevated rates of epilepsy in individuals with AS, and associated hypersynchronous neural activity. Here we briefly review findings on EEG, E/I ratio, and epileptic seizures in AS, including data from rodent models of the disorder. We summarize pharmacologic approaches that have been used to treat behavioral aspects of AS, including neuropsychiatric phenomena and sleep disturbances, as well as seizures in the context of the disorder. Antidepressants such as SSRIs and atypical antipsychotics are among the medications that have been used behaviorally, whereas anticonvulsant drugs such as valproic acid and lamotrigine have frequently been used to control seizures in AS. We end by suggesting novel uses for some existing pharmacologic agents in AS, including noradrenergic transmission reducing drugs (alpha2 agonists, beta blockers, alpha1 antagonists) and cholinesterase inhibitors, where these various classes of drugs may have the ability to ameliorate both behavioral disturbances and seizures.
Collapse
Affiliation(s)
- Paul J Fitzgerald
- Department of Psychological Sciences, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
7
|
Dierdorff L, Garcia-Forn M, von Mueffling A, De Rubeis S. Assessing motor development and function in mouse models of neurodevelopmental disorders. Methods Cell Biol 2024; 188:171-181. [PMID: 38880523 DOI: 10.1016/bs.mcb.2024.04.005] [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] [Indexed: 06/18/2024]
Abstract
Alterations in motor development often accompany neurodevelopmental disorders (NDD) and can have an impact on social interaction and communication. Studying motor development and function in mouse models of NDDs can offer a window to identify underlying biological mechanisms and establish preclinical outcome measures for testing therapeutics. This chapter describes tests to measure motor developmental milestones early postnatally and adult motor functions in mouse models of NDDs.
Collapse
Affiliation(s)
- Lauren Dierdorff
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; The Alper Center for Neural Development and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Marta Garcia-Forn
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; The Alper Center for Neural Development and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Alexa von Mueffling
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; The Alper Center for Neural Development and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; The Alper Center for Neural Development and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
8
|
Cording KR, Bateup HS. Altered motor learning and coordination in mouse models of autism spectrum disorder. Front Cell Neurosci 2023; 17:1270489. [PMID: 38026686 PMCID: PMC10663323 DOI: 10.3389/fncel.2023.1270489] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder with increasing prevalence. Over 1,000 risk genes have now been implicated in ASD, suggesting diverse etiology. However, the diagnostic criteria for the disorder still comprise two major behavioral domains - deficits in social communication and interaction, and the presence of restricted and repetitive patterns of behavior (RRBs). The RRBs associated with ASD include both stereotyped repetitive movements and other motor manifestations including changes in gait, balance, coordination, and motor skill learning. In recent years, the striatum, the primary input center of the basal ganglia, has been implicated in these ASD-associated motor behaviors, due to the striatum's role in action selection, motor learning, and habit formation. Numerous mouse models with mutations in ASD risk genes have been developed and shown to have alterations in ASD-relevant behaviors. One commonly used assay, the accelerating rotarod, allows for assessment of both basic motor coordination and motor skill learning. In this corticostriatal-dependent task, mice walk on a rotating rod that gradually increases in speed. In the extended version of this task, mice engage striatal-dependent learning mechanisms to optimize their motor routine and stay on the rod for longer periods. This review summarizes the findings of studies examining rotarod performance across a range of ASD mouse models, and the resulting implications for the involvement of striatal circuits in ASD-related motor behaviors. While performance in this task is not uniform across mouse models, there is a cohort of models that show increased rotarod performance. A growing number of studies suggest that this increased propensity to learn a fixed motor routine may reflect a common enhancement of corticostriatal drive across a subset of mice with mutations in ASD-risk genes.
Collapse
Affiliation(s)
- Katherine R. Cording
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| | - Helen S. Bateup
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
- Molecular and Cell Biology Department, University of California, Berkeley, Berkeley, CA, United States
- Chan Zuckerberg Biohub, San Francisco, CA, United States
| |
Collapse
|
9
|
Xing L, Simon JM, Ptacek TS, Yi JJ, Loo L, Mao H, Wolter JM, McCoy ES, Paranjape SR, Taylor-Blake B, Zylka MJ. Autism-linked UBE3A gain-of-function mutation causes interneuron and behavioral phenotypes when inherited maternally or paternally in mice. Cell Rep 2023; 42:112706. [PMID: 37389991 PMCID: PMC10530456 DOI: 10.1016/j.celrep.2023.112706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 04/15/2023] [Accepted: 06/12/2023] [Indexed: 07/02/2023] Open
Abstract
The E3 ubiquitin ligase Ube3a is biallelically expressed in neural progenitors and glial cells, suggesting that UBE3A gain-of-function mutations might cause neurodevelopmental disorders irrespective of parent of origin. Here, we engineered a mouse line that harbors an autism-linked UBE3AT485A (T503A in mouse) gain-of-function mutation and evaluated phenotypes in animals that inherited the mutant allele paternally, maternally, or from both parents. We find that paternally and maternally expressed UBE3AT503A results in elevated UBE3A activity in neural progenitors and glial cells. Expression of UBE3AT503A from the maternal allele, but not the paternal one, leads to a persistent elevation of UBE3A activity in neurons. Mutant mice display behavioral phenotypes that differ by parent of origin. Expression of UBE3AT503A, irrespective of its parent of origin, promotes transient embryonic expansion of Zcchc12 lineage interneurons. Phenotypes of Ube3aT503A mice are distinct from Angelman syndrome model mice. Our study has clinical implications for a growing number of disease-linked UBE3A gain-of-function mutations.
Collapse
Affiliation(s)
- Lei Xing
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jeremy M Simon
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Campus Box #7255, Chapel Hill, NC 27599, USA; Department of Genetics, The University of North Carolina at Chapel Hill, Campus Box #7264, Chapel Hill, NC 27599, USA
| | - Travis S Ptacek
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Campus Box #7255, Chapel Hill, NC 27599, USA
| | - Jason J Yi
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Campus Box #7255, Chapel Hill, NC 27599, USA
| | - Lipin Loo
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hanqian Mao
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Campus Box #7255, Chapel Hill, NC 27599, USA
| | - Justin M Wolter
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Campus Box #7255, Chapel Hill, NC 27599, USA; Department of Genetics, The University of North Carolina at Chapel Hill, Campus Box #7264, Chapel Hill, NC 27599, USA
| | - Eric S McCoy
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Smita R Paranjape
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bonnie Taylor-Blake
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mark J Zylka
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Campus Box #7255, Chapel Hill, NC 27599, USA.
| |
Collapse
|
10
|
Alberini CM. IGF2 in memory, neurodevelopmental disorders, and neurodegenerative diseases. Trends Neurosci 2023; 46:488-502. [PMID: 37031050 PMCID: PMC10192130 DOI: 10.1016/j.tins.2023.03.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/27/2023] [Accepted: 03/12/2023] [Indexed: 04/08/2023]
Abstract
Insulin-like growth factor 2 (IGF2) emerged as a critical mechanism of synaptic plasticity and learning and memory. Deficits in IGF2 in the brain, serum, or cerebrospinal fluid (CSF) are associated with brain diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS). Increasing IGF2 levels enhances memory in healthy animals and reverses numerous symptoms in laboratory models of aging, neurodevelopmental disorders, and neurodegenerative diseases. These effects occur via the IGF2 receptor (IGF2R) - a receptor that is highly expressed in neurons and regulates protein trafficking, synthesis, and degradation. Here, I summarize the current knowledge regarding IGF2 expression and functions in the brain, particularly in memory, and propose a novel conceptual model for IGF2/IGF2R mechanisms of action in brain health and diseases.
Collapse
|
11
|
Egawa K, Watanabe M, Shiraishi H, Sato D, Takahashi Y, Nishio S, Fukuda A. Imbalanced expression of cation-chloride cotransporters as a potential therapeutic target in an Angelman syndrome mouse model. Sci Rep 2023; 13:5685. [PMID: 37069177 PMCID: PMC10110603 DOI: 10.1038/s41598-023-32376-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 03/27/2023] [Indexed: 04/19/2023] Open
Abstract
Angelman syndrome is a neurodevelopmental disorder caused by loss of function of the maternally expressed UBE3A gene. Treatments for the main manifestations, including cognitive dysfunction or epilepsy, are still under development. Recently, the Cl- importer Na+-K+-Cl- cotransporter 1 (NKCC1) and the Cl- exporter K+-Cl- cotransporter 2 (KCC2) have garnered attention as therapeutic targets for many neurological disorders. Dysregulation of neuronal intracellular Cl- concentration ([Cl-]i) is generally regarded as one of the mechanisms underlying neuronal dysfunction caused by imbalanced expression of these cation-chloride cotransporters (CCCs). Here, we analyzed the regulation of [Cl-]i and the effects of bumetanide, an NKCC1 inhibitor, in Angelman syndrome models (Ube3am-/p+ mice). We observed increased NKCC1 expression and decreased KCC2 expression in the hippocampi of Ube3am-/p+ mice. The average [Cl-]i of CA1 pyramidal neurons was not significantly different but demonstrated greater variance in Ube3am-/p+ mice. Tonic GABAA receptor-mediated Cl- conductance was reduced, which may have contributed to maintaining the normal average [Cl-]i. Bumetanide administration restores cognitive dysfunction in Ube3am-/p+ mice. Seizure susceptibility was also reduced regardless of the genotype. These results suggest that an imbalanced expression of CCCs is involved in the pathophysiological mechanism of Ube3am-/p+ mice, although the average [Cl-]i is not altered. The blockage of NKCC1 may be a potential therapeutic strategy for patients with Angelman syndrome.
Collapse
Affiliation(s)
- Kiyoshi Egawa
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 060-8638, Japan.
| | - Miho Watanabe
- Department of Neurophysiology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-Ku, Hamamatsu City, Shizuoka, 431-3192, Japan
| | - Hideaki Shiraishi
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Daisuke Sato
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Yukitoshi Takahashi
- Department of Clinical Research, National Epilepsy Center, NHO, Shizuoka Institute of Epilepsy and Neurological Disorders, Urushiyama 886, Aoi-Ku, Shizuoka, 420-8688, Japan
| | - Saori Nishio
- Department of Rheumatology, Endocrinology, and Nephrology, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-Ku, Hamamatsu City, Shizuoka, 431-3192, Japan
| |
Collapse
|
12
|
O'Geen H, Beitnere U, Garcia MS, Adhikari A, Cameron DL, Fenton TA, Copping NA, Deng P, Lock S, Halmai JANM, Villegas IJ, Liu J, Wang D, Fink KD, Silverman JL, Segal DJ. Transcriptional reprogramming restores UBE3A brain-wide and rescues behavioral phenotypes in an Angelman syndrome mouse model. Mol Ther 2023; 31:1088-1105. [PMID: 36641623 PMCID: PMC10124086 DOI: 10.1016/j.ymthe.2023.01.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/19/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Angelman syndrome (AS) is a neurogenetic disorder caused by the loss of ubiquitin ligase E3A (UBE3A) gene expression in the brain. The UBE3A gene is paternally imprinted in brain neurons. Clinical features of AS are primarily due to the loss of maternally expressed UBE3A in the brain. A healthy copy of paternal UBE3A is present in the brain but is silenced by a long non-coding antisense transcript (UBE3A-ATS). Here, we demonstrate that an artificial transcription factor (ATF-S1K) can silence Ube3a-ATS in an adult mouse model of Angelman syndrome (AS) and restore endogenous physiological expression of paternal Ube3a. A single injection of adeno-associated virus (AAV) expressing ATF-S1K (AAV-S1K) into the tail vein enabled whole-brain transduction and restored UBE3A protein in neurons to ∼25% of wild-type protein. The ATF-S1K treatment was highly specific to the target site with no detectable inflammatory response 5 weeks after AAV-S1K administration. AAV-S1K treatment of AS mice showed behavioral rescue in exploratory locomotion, a task involving gross and fine motor abilities, similar to low ambulation and velocity in AS patients. The specificity and tolerability of a single injection of AAV-S1K therapy for AS demonstrate the use of ATFs as a promising translational approach for AS.
Collapse
Affiliation(s)
| | | | | | - Anna Adhikari
- MIND Institute, UC Davis Health System, Sacramento, CA, USA; Department of Psychiatry and Behavioral Sciences, UC Davis Health System, Sacramento, CA, USA
| | - David L Cameron
- Neurology Department, Stem Cell Program and Gene Therapy Center, UC Davis Health System, Sacramento, CA, USA; MIND Institute, UC Davis Health System, Sacramento, CA, USA
| | - Timothy A Fenton
- MIND Institute, UC Davis Health System, Sacramento, CA, USA; Department of Psychiatry and Behavioral Sciences, UC Davis Health System, Sacramento, CA, USA
| | - Nycole A Copping
- MIND Institute, UC Davis Health System, Sacramento, CA, USA; Department of Psychiatry and Behavioral Sciences, UC Davis Health System, Sacramento, CA, USA
| | - Peter Deng
- Neurology Department, Stem Cell Program and Gene Therapy Center, UC Davis Health System, Sacramento, CA, USA; MIND Institute, UC Davis Health System, Sacramento, CA, USA
| | - Samantha Lock
- Neurology Department, Stem Cell Program and Gene Therapy Center, UC Davis Health System, Sacramento, CA, USA; MIND Institute, UC Davis Health System, Sacramento, CA, USA
| | - Julian A N M Halmai
- Neurology Department, Stem Cell Program and Gene Therapy Center, UC Davis Health System, Sacramento, CA, USA; MIND Institute, UC Davis Health System, Sacramento, CA, USA
| | - Isaac J Villegas
- Neurology Department, Stem Cell Program and Gene Therapy Center, UC Davis Health System, Sacramento, CA, USA; MIND Institute, UC Davis Health System, Sacramento, CA, USA
| | - Jiajian Liu
- Genome Editing and Novel Modalities (GENM), MilliporeSigma, St. Louis, MO, USA
| | - Danhui Wang
- Genome Editing and Novel Modalities (GENM), MilliporeSigma, St. Louis, MO, USA
| | - Kyle D Fink
- Neurology Department, Stem Cell Program and Gene Therapy Center, UC Davis Health System, Sacramento, CA, USA; MIND Institute, UC Davis Health System, Sacramento, CA, USA
| | - Jill L Silverman
- MIND Institute, UC Davis Health System, Sacramento, CA, USA; Department of Psychiatry and Behavioral Sciences, UC Davis Health System, Sacramento, CA, USA
| | - David J Segal
- Genome Center, UC Davis, Davis, CA, USA; Department of Biochemistry and Molecular Medicine, UC Davis, Davis, CA, USA; MIND Institute, UC Davis Health System, Sacramento, CA, USA.
| |
Collapse
|
13
|
Martinez LA, Born HA, Harris S, Regnier-Golanov A, Grieco JC, Weeber EJ, Anderson AE. Quantitative EEG Analysis in Angelman Syndrome: Candidate Method for Assessing Therapeutics. Clin EEG Neurosci 2023; 54:203-212. [PMID: 33203220 DOI: 10.1177/1550059420973095] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The goal of these studies was to use quantitative (q)EEG techniques on data from children with Angelman syndrome (AS) using spectral power analysis, and to evaluate this as a potential biomarker and quantitative method to evaluate therapeutics. Although characteristic patterns are evident in visual inspection, using qEEG techniques has the potential to provide quantitative evidence of treatment efficacy. We first assessed spectral power from baseline EEG recordings collected from children with AS compared to age-matched neurotypical controls, which corroborated the previously reported finding of increased total power driven by elevated delta power in children with AS. We then retrospectively analyzed data collected during a clinical trial evaluating the safety and tolerability of minocycline (3 mg/kg/d) to compare pretreatment recordings from children with AS (4-12 years of age) to EEG activity at the end of treatment and following washout for EEG spectral power and epileptiform events. At baseline and during minocycline treatment, the AS subjects demonstrated increased delta power; however, following washout from minocycline treatment the AS subjects had significantly reduced EEG spectral power and epileptiform activity. Our findings support the use of qEEG analysis in evaluating AS and suggest that this technique may be useful to evaluate therapeutic efficacy in AS. Normalizing EEG power in AS therefore may become an important metric in screening therapeutics to gauge overall efficacy. As therapeutics transition from preclinical to clinical studies, it is vital to establish outcome measures that can quantitatively evaluate putative treatments for AS and neurological disorders with distinctive EEG patterns.
Collapse
Affiliation(s)
- Luis A Martinez
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,The Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.,The Gordon and Mary Cain Pediatric Neurology Research Foundation Laboratories, Texas Children's Hospital, Houston, TX, USA
| | - Heather A Born
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,The Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.,The Gordon and Mary Cain Pediatric Neurology Research Foundation Laboratories, Texas Children's Hospital, Houston, TX, USA
| | - Sarah Harris
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,The Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.,The Gordon and Mary Cain Pediatric Neurology Research Foundation Laboratories, Texas Children's Hospital, Houston, TX, USA
| | - Angelique Regnier-Golanov
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,The Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.,The Gordon and Mary Cain Pediatric Neurology Research Foundation Laboratories, Texas Children's Hospital, Houston, TX, USA
| | - Joseph C Grieco
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - Edwin J Weeber
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - Anne E Anderson
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,The Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.,The Gordon and Mary Cain Pediatric Neurology Research Foundation Laboratories, Texas Children's Hospital, Houston, TX, USA.,Departments of Neuroscience and Neurology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
14
|
Romero LO, Caires R, Kaitlyn Victor A, Ramirez J, Sierra-Valdez FJ, Walsh P, Truong V, Lee J, Mayor U, Reiter LT, Vásquez V, Cordero-Morales JF. Linoleic acid improves PIEZO2 dysfunction in a mouse model of Angelman Syndrome. Nat Commun 2023; 14:1167. [PMID: 36859399 PMCID: PMC9977963 DOI: 10.1038/s41467-023-36818-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/17/2023] [Indexed: 03/03/2023] Open
Abstract
Angelman syndrome (AS) is a neurogenetic disorder characterized by intellectual disability and atypical behaviors. AS results from loss of expression of the E3 ubiquitin-protein ligase UBE3A from the maternal allele in neurons. Individuals with AS display impaired coordination, poor balance, and gait ataxia. PIEZO2 is a mechanosensitive ion channel essential for coordination and balance. Here, we report that PIEZO2 activity is reduced in Ube3a deficient male and female mouse sensory neurons, a human Merkel cell carcinoma cell line and female human iPSC-derived sensory neurons with UBE3A knock-down, and de-identified stem cell-derived neurons from individuals with AS. We find that loss of UBE3A decreases actin filaments and reduces PIEZO2 expression and function. A linoleic acid (LA)-enriched diet increases PIEZO2 activity, mechano-excitability, and improves gait in male AS mice. Finally, LA supplementation increases PIEZO2 function in stem cell-derived neurons from individuals with AS. We propose a mechanism whereby loss of UBE3A expression reduces PIEZO2 function and identified a fatty acid that enhances channel activity and ameliorates AS-associated mechano-sensory deficits.
Collapse
Affiliation(s)
- Luis O Romero
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38103, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, Memphis, TN, 38163, USA
| | - Rebeca Caires
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38103, USA
| | - A Kaitlyn Victor
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38103, USA
| | - Juanma Ramirez
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, UPV/EHU, Leioa, Bizkaia, Spain
| | - Francisco J Sierra-Valdez
- School of Engineering and Sciences, Tecnológico de Monterrey, Ave. Eugenio Garza Sada 2501 Sur, Monterrey, 64849, Mexico
| | | | | | - Jungsoo Lee
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38103, USA
| | - Ugo Mayor
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, UPV/EHU, Leioa, Bizkaia, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Bizkaia, Spain
| | - Lawrence T Reiter
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38103, USA
- Department of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38104, USA
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38104, USA
| | - Valeria Vásquez
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38103, USA.
| | - Julio F Cordero-Morales
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38103, USA.
| |
Collapse
|
15
|
Rigter PMF, de Konink C, van Woerden GM. Loss of CAMK2G affects intrinsic and motor behavior but has minimal impact on cognitive behavior. Front Neurosci 2023; 16:1086994. [PMID: 36685241 PMCID: PMC9853378 DOI: 10.3389/fnins.2022.1086994] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/07/2022] [Indexed: 01/08/2023] Open
Abstract
Introduction The gamma subunit of calcium/calmodulin-dependent protein kinase 2 (CAMK2G) is expressed throughout the brain and is associated with neurodevelopmental disorders. Research on the role of CAMK2G is limited and attributes different functions to specific cell types. Methods To further expand on the role of CAMK2G in brain functioning, we performed extensive phenotypic characterization of a Camk2g knockout mouse. Results We found different CAMK2G isoforms that show a distinct spatial expression pattern in the brain. Additionally, based on our behavioral characterization, we conclude that CAMK2G plays a minor role in hippocampus-dependent learning and synaptic plasticity. Rather, we show that CAMK2G is required for motor function and that the loss of CAMK2G results in impaired nest-building and marble burying behavior, which are innate behaviors that are associated with impaired neurodevelopment. Discussion Taken together, our results provide evidence for a unique function of this specific CAMK2 isozyme in the brain and further support the role of CAMK2G in neurodevelopment.
Collapse
Affiliation(s)
- Pomme M. F. Rigter
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, Netherlands
- Erfelijke Neuro-Cognitieve Ontwikkelingsstoornissen, Expertise Centre for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
| | - Charlotte de Konink
- Erfelijke Neuro-Cognitieve Ontwikkelingsstoornissen, Expertise Centre for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Geeske M. van Woerden
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, Netherlands
- Erfelijke Neuro-Cognitieve Ontwikkelingsstoornissen, Expertise Centre for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
16
|
Lee D, Chen W, Kaku HN, Zhuo X, Chao ES, Soriano A, Kuncheria A, Flores S, Kim JH, Rivera A, Rigo F, Jafar-nejad P, Beaudet AL, Caudill MS, Xue M. Antisense oligonucleotide therapy rescues disturbed brain rhythms and sleep in juvenile and adult mouse models of Angelman syndrome. eLife 2023; 12:e81892. [PMID: 36594817 PMCID: PMC9904759 DOI: 10.7554/elife.81892] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/30/2022] [Indexed: 01/04/2023] Open
Abstract
UBE3A encodes ubiquitin protein ligase E3A, and in neurons its expression from the paternal allele is repressed by the UBE3A antisense transcript (UBE3A-ATS). This leaves neurons susceptible to loss-of-function of maternal UBE3A. Indeed, Angelman syndrome, a severe neurodevelopmental disorder, is caused by maternal UBE3A deficiency. A promising therapeutic approach to treating Angelman syndrome is to reactivate the intact paternal UBE3A by suppressing UBE3A-ATS. Prior studies show that many neurological phenotypes of maternal Ube3a knockout mice can only be rescued by reinstating Ube3a expression in early development, indicating a restricted therapeutic window for Angelman syndrome. Here, we report that reducing Ube3a-ATS by antisense oligonucleotides in juvenile or adult maternal Ube3a knockout mice rescues the abnormal electroencephalogram (EEG) rhythms and sleep disturbance, two prominent clinical features of Angelman syndrome. Importantly, the degree of phenotypic improvement correlates with the increase of Ube3a protein levels. These results indicate that the therapeutic window of genetic therapies for Angelman syndrome is broader than previously thought, and EEG power spectrum and sleep architecture should be used to evaluate the clinical efficacy of therapies.
Collapse
Affiliation(s)
- Dongwon Lee
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Wu Chen
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Heet Naresh Kaku
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Xinming Zhuo
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Eugene S Chao
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | | | - Allen Kuncheria
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Stephanie Flores
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Joo Hyun Kim
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Armando Rivera
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Frank Rigo
- Ionis PharmaceuticalsCarlsbadUnited States
| | | | - Arthur L Beaudet
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Matthew S Caudill
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Mingshan Xue
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
17
|
Sun J, Liu Y, Hao X, Baudry M, Bi X. Lack of UBE3A-Mediated Regulation of Synaptic SK2 Channels Contributes to Learning and Memory Impairment in the Female Mouse Model of Angelman Syndrome. Neural Plast 2022; 2022:3923384. [PMID: 36237484 PMCID: PMC9553421 DOI: 10.1155/2022/3923384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/16/2022] [Indexed: 11/29/2022] Open
Abstract
Angelman syndrome (AS) is a rare neurodevelopmental disorder characterized by severe developmental delay, motor impairment, language and cognition deficits, and often with increased seizure activity. AS is caused by deficiency of UBE3A, which is both an E3 ligase and a cofactor for transcriptional regulation. We previously showed that the small conductance potassium channel protein SK2 is a UBE3A substrate, and that increased synaptic SK2 levels contribute to impairments in synaptic plasticity and fear-conditioning memory, as inhibition of SK2 channels significantly improved both synaptic plasticity and fear memory in male AS mice. In the present study, we investigated UBE3a-mediated regulation of synaptic plasticity and fear-conditioning in female AS mice. Results from both western blot and immunofluorescence analyses showed that synaptic SK2 levels were significantly increased in hippocampus of female AS mice, as compared to wild-type (WT) littermates. Like in male AS mice, long-term potentiation (LTP) was significantly reduced while long-term depression (LTD) was enhanced at hippocampal CA3-CA1 synapses of female AS mice, as compared to female WT mice. Both alterations were significantly reduced by treatment with the SK2 inhibitor, apamin. The shunting effect of SK2 channels on NMDA receptor was significantly larger in female AS mice as compared to female WT mice. Female AS mice also showed impairment in both contextual and tone memory recall, and this impairment was significantly reduced by apamin treatment. Our results indicate that like male AS mice, female AS mice showed significant impairment in both synaptic plasticity and fear-conditioning memory due to increased levels of synaptic SK2 channels. Any therapeutic strategy to reduce SK2-mediated inhibition of NMDAR should be beneficial to both male and female patients.
Collapse
Affiliation(s)
- Jiandong Sun
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766, USA
| | - Yan Liu
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766, USA
| | - Xiaoning Hao
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766, USA
| | - Michel Baudry
- College of Dental Medicine, Western University of Health Sciences, Pomona, California 91766, USA
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766, USA
| |
Collapse
|
18
|
Negrón-Moreno PN, Diep DT, Guoynes CD, Sidorov MS. Dissociating motor impairment from five-choice serial reaction time task performance in a mouse model of Angelman syndrome. Front Behav Neurosci 2022; 16:968159. [PMID: 36212189 PMCID: PMC9539753 DOI: 10.3389/fnbeh.2022.968159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 09/05/2022] [Indexed: 12/02/2022] Open
Abstract
Angelman syndrome (AS) is a single-gene neurodevelopmental disorder associated with cognitive and motor impairment, seizures, lack of speech, and disrupted sleep. AS is caused by loss-of-function mutations in the UBE3A gene, and approaches to reinstate functional UBE3A are currently in clinical trials in children. Behavioral testing in a mouse model of AS (Ube3a m-/p+ ) represents an important tool to assess the effectiveness of current and future treatments preclinically. Existing behavioral tests effectively model motor impairments, but not cognitive impairments, in Ube3a m-/p+ mice. Here we tested the hypothesis that the 5-choice serial reaction time task (5CSRTT) can be used to assess cognitive behaviors in Ube3a m-/p+ mice. Ube3a m-/p+ mice had more omissions during 5CSRTT training than wild-type littermate controls, but also showed impaired motor function including open field hypoactivity and delays in eating pellet rewards. Motor impairments thus presented an important confound for interpreting this group difference in omissions. We report that despite hypoactivity during habituation, Ube3a m-/p+ mice had normal response latencies to retrieve rewards during 5CSRTT training. We also accounted for delays in eating pellet rewards by assessing omissions solely on trials where eating delays would not impact results. Thus, the increase in omissions in Ube3a m-/p+ mice is likely not caused by concurrent motor impairments. This work underscores the importance of considering how known motor impairments in Ube3a m-/p+ mice may affect behavioral performance in other domains. Our results also provide guidance on how to design a 5CSRTT protocol that is best suited for future studies in Ube3a mutants.
Collapse
Affiliation(s)
- Paola N. Negrón-Moreno
- University of Puerto Rico-Cayey, Cayey, PR, United States
- Department of Cell Biology and Physiology, Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - David T. Diep
- University of Maryland, College Park, College Park, MD, United States
- Center for Neuroscience Research, Children’s National Medical Center, Washington, DC, United States
| | - Caleigh D. Guoynes
- Center for Neuroscience Research, Children’s National Medical Center, Washington, DC, United States
| | - Michael S. Sidorov
- Department of Cell Biology and Physiology, Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Center for Neuroscience Research, Children’s National Medical Center, Washington, DC, United States
- Departments of Pediatrics and Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| |
Collapse
|
19
|
Generation and Characterization of a Novel Angelman Syndrome Mouse Model with a Full Deletion of the Ube3a Gene. Cells 2022; 11:cells11182815. [PMID: 36139390 PMCID: PMC9496699 DOI: 10.3390/cells11182815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/28/2022] [Accepted: 08/31/2022] [Indexed: 11/29/2022] Open
Abstract
Angelman syndrome (AS) is a neurodevelopmental disorder caused by deficits in maternally inherited UBE3A. The disease is characterized by intellectual disability, impaired motor skills, and behavioral deficits, including increased anxiety and autism spectrum disorder features. The mouse models used so far in AS research recapitulate most of the cardinal AS characteristics. However, they do not mimic the situation found in the majority of AS patients who have a large deletion spanning 4–6 Mb. There is also a large variability in phenotypes reported in the available models, which altogether limits development of therapeutics. Therefore, we have generated a mouse model in which the Ube3a gene is deleted entirely from the 5′ UTR to the 3′ UTR of mouse Ube3a isoform 2, resulting in a deletion of 76 kb. To investigate its phenotypic suitability as a model for AS, we employed a battery of behavioral tests directed to reveal AS pathology and to find out whether this model better mirrors AS development compared to other available models. We found that the maternally inherited Ube3a-deficient line exhibits robust motor dysfunction, as seen in the rotarod and DigiGait tests, and displays abnormalities in additional behavioral paradigms, including reduced nest building and hypoactivity, although no apparent cognitive phenotype was observed in the Barnes maze and novel object recognition tests. The AS mice did, however, underperform in more complex cognition tasks, such as place reversal in the IntelliCage system, and exhibited a different circadian rhythm activity pattern. We show that the novel UBE3A-deficient model, based on a whole-gene deletion, is suitable for AS research, as it recapitulates important phenotypes characteristic of AS. This new mouse model provides complementary possibilities to study the Ube3a gene and its function in health and disease as well as possible therapeutic interventions to restore function.
Collapse
|
20
|
Antoine MW. Paradoxical Hyperexcitability in Disorders of Neurodevelopment. Front Mol Neurosci 2022; 15:826679. [PMID: 35571370 PMCID: PMC9102973 DOI: 10.3389/fnmol.2022.826679] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/14/2022] [Indexed: 01/29/2023] Open
Abstract
Autism Spectrum Disorder (ASD), Rett syndrome (RTT) and Angelman Syndrome (AS) are neurodevelopmental disorders (NDDs) that share several clinical characteristics, including displays of repetitive movements, developmental delays, language deficits, intellectual disability, and increased susceptibility to epilepsy. While several reviews address the biological basis of non-seizure-related ASD phenotypes, here, I highlight some shared biological mechanisms that may contribute to increased seizure susceptibility. I focus on genetic studies identifying the anatomical origin of the seizure phenotype in loss-of-function, monogenic, mouse models of these NDDs, combined with insights gained from complementary studies quantifying levels of synaptic excitation and inhibition. Epilepsy is characterized by a sudden, abnormal increase in synchronous activity within neuronal networks, that is posited to arise from excess excitation, largely driven by reduced synaptic inhibition. Primarily for this reason, elevated network excitability is proposed to underlie the causal basis for the ASD, RTT, and AS phenotypes. Although, mouse models of these disorders replicate aspects of the human condition, i.e., hyperexcitability discharges or seizures on cortical electroencephalograms, measures at the synaptic level often reveal deficits in excitatory synaptic transmission, rather than too much excitation. Resolving this apparent paradox has direct implications regarding expected outcomes of manipulating GABAergic tone. In particular, in NDDs associated with seizures, cortical circuits can display reduced, rather than normal or increased levels of synaptic excitation, and therefore suggested treatments aimed at increasing inhibition could further promote hypoactivity instead of normality. In this review, I highlight shared mechanisms across animal models for ASD, RTT, and AS with reduced synaptic excitation that nevertheless promote hyperexcitability in cortical circuits.
Collapse
Affiliation(s)
- Michelle W. Antoine
- Section on Neural Circuits, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
21
|
Adhikari A, Buchanan FKB, Fenton TA, Cameron DL, Halmai JANM, Copping NA, Fink KD, Silverman JL. Touchscreen Cognitive Deficits, Hyperexcitability, and Hyperactivity in Males and Females Using Two Models of Cdkl5 Deficiency. Hum Mol Genet 2022; 31:3032-3050. [PMID: 35445702 PMCID: PMC9476626 DOI: 10.1093/hmg/ddac091] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 11/17/2022] Open
Abstract
Many neurodevelopmental disorders (NDDs) are the result of mutations on the X chromosome. One severe NDD resulting from mutations on the X chromosome is CDKL5 deficiency disorder (CDD). CDD is an epigenetic, X-linked NDD characterized by intellectual disability (ID), pervasive seizures and severe sleep disruption, including recurring hospitalizations. CDD occurs at a 4:1 ratio, with a female bias. CDD is driven by the loss of cyclin-dependent kinase-like 5 (CDKL5), a serine/threonine kinase that is essential for typical brain development, synapse formation and signal transmission. Previous studies focused on male subjects from animal models, likely to avoid the complexity of X mosaicism. For the first time, we report translationally relevant behavioral phenotypes in young adult (8–20 weeks) females and males with robust signal size, including impairments in learning and memory, substantial hyperactivity and increased susceptibility to seizures/reduced seizure thresholds, in both sexes, and in two models of CDD preclinical mice, one with a general loss-of-function mutation and one that is a patient-derived mutation.
Collapse
Affiliation(s)
- Anna Adhikari
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA.,Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA
| | - Fiona K B Buchanan
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA.,Stem Cell Program and Gene Therapy Center, University of California Davis School of Medicine, Sacramento, CA
| | - Timothy A Fenton
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA.,Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA
| | - David L Cameron
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA.,Stem Cell Program and Gene Therapy Center, University of California Davis School of Medicine, Sacramento, CA
| | - Julian A N M Halmai
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA.,Stem Cell Program and Gene Therapy Center, University of California Davis School of Medicine, Sacramento, CA
| | - Nycole A Copping
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA.,Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA
| | - Kyle D Fink
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA.,Department of Neurology, University of California Davis School of Medicine, Sacramento, CA.,Stem Cell Program and Gene Therapy Center, University of California Davis School of Medicine, Sacramento, CA
| | - Jill L Silverman
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA.,Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA
| |
Collapse
|
22
|
Shi SQ, Mahoney CE, Houdek P, Zhao W, Anderson MP, Zhuo X, Beaudet A, Sumova A, Scammell TE, Johnson CH. Circadian Rhythms and Sleep Are Dependent Upon Expression Levels of Key Ubiquitin Ligase Ube3a. Front Behav Neurosci 2022; 16:837523. [PMID: 35401134 PMCID: PMC8989470 DOI: 10.3389/fnbeh.2022.837523] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Normal neurodevelopment requires precise expression of the key ubiquitin ligase gene Ube3a. Comparing newly generated mouse models for Ube3a downregulation (models of Angelman syndrome) vs. Ube3a upregulation (models for autism), we find reciprocal effects of Ube3a gene dosage on phenotypes associated with circadian rhythmicity, including the amount of locomotor activity. Consistent with results from neurons in general, we find that Ube3a is imprinted in neurons of the suprachiasmatic nuclei (SCN), the pacemaking circadian brain locus, despite other claims that SCN neurons were somehow exceptional to these imprinting rules. In addition, Ube3a-deficient mice lack the typical drop in wake late in the dark period and have blunted responses to sleep deprivation. Suppression of physical activity by light in Ube3a-deficient mice is not due to anxiety as measured by behavioral tests and stress hormones; quantification of stress hormones may provide a mechanistic link to sleep alteration and memory deficits caused by Ube3a deficiency, and serve as an easily measurable biomarker for evaluating potential therapeutic treatments for Angelman syndrome. We conclude that reduced Ube3a gene dosage affects not only neurodevelopment but also sleep patterns and circadian rhythms.
Collapse
Affiliation(s)
- Shu-qun Shi
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
| | - Carrie E. Mahoney
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Pavel Houdek
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Wenling Zhao
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Matthew P. Anderson
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Xinming Zhuo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | | | - Alena Sumova
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Thomas E. Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Carl Hirschie Johnson
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
- *Correspondence: Carl Hirschie Johnson,
| |
Collapse
|
23
|
Levin Y, Hosamane NS, McNair TE, Kunnam SS, Philpot BD, Fan Z, Sidorov MS. Evaluation of electroencephalography biomarkers for Angelman syndrome during overnight sleep. Autism Res 2022; 15:1031-1042. [PMID: 35304979 PMCID: PMC9227959 DOI: 10.1002/aur.2709] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 01/31/2022] [Accepted: 03/10/2022] [Indexed: 11/13/2022]
Abstract
Angelman syndrome (AS) is a neurodevelopmental disorder caused by loss‐of‐function mutations in the maternal copy of the UBE3A gene. AS is characterized by intellectual disability, impaired speech and motor skills, epilepsy, and sleep disruptions. Multiple treatment strategies to re‐express functional neuronal UBE3A from the dormant paternal allele were successful in rodent models of AS and have now moved to early phase clinical trials in children. Developing reliable and objective AS biomarkers is essential to guide the design and execution of current and future clinical trials. Our prior work quantified short daytime electroencephalograms (EEGs) to define promising biomarkers for AS. Here, we asked whether overnight sleep is better suited to detect AS EEG biomarkers. We retrospectively analyzed EEGs from 12 overnight sleep studies from individuals with AS with age and sex‐matched Down syndrome and neurotypical controls, focusing on low frequency (2–4 Hz) delta rhythms and sleep spindles. Delta EEG rhythms were increased in individuals with AS during all stages of overnight sleep, but overnight sleep did not provide additional benefit over wake in the ability to detect increased delta. Abnormal sleep spindles were not reliably detected in EEGs from individuals with AS during overnight sleep, suggesting that delta rhythms represent a more reliable biomarker. Overall, we conclude that periods of wakefulness are sufficient, and perhaps ideal, to quantify delta EEG rhythms for use as AS biomarkers.
Collapse
Affiliation(s)
- Yuval Levin
- Center for Neuroscience Research, Children's National Medical Center, Washington, District of Columbia, USA.,The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Nishitha S Hosamane
- Center for Neuroscience Research, Children's National Medical Center, Washington, District of Columbia, USA
| | - Taylor E McNair
- Center for Neuroscience Research, Children's National Medical Center, Washington, District of Columbia, USA
| | - Shrujana S Kunnam
- Center for Neuroscience Research, Children's National Medical Center, Washington, District of Columbia, USA
| | - Benjamin D Philpot
- Department of Cell Biology & Physiology, Carolina Institute for Developmental Disabilities, and UNC Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Zheng Fan
- Department of Neurology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Michael S Sidorov
- Center for Neuroscience Research, Children's National Medical Center, Washington, District of Columbia, USA.,Departments of Pediatrics and Pharmacology & Physiology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| |
Collapse
|
24
|
Petkova SP, Adhikari A, Berg EL, Fenton TA, Duis J, Silverman JL. Gait as a quantitative translational outcome measure in Angelman syndrome. Autism Res 2022; 15:821-833. [PMID: 35274462 PMCID: PMC9311146 DOI: 10.1002/aur.2697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 02/17/2022] [Accepted: 02/20/2022] [Indexed: 02/05/2023]
Abstract
Angelman syndrome (AS) is a genetic neurodevelopmental disorder characterized by developmental delay, lack of speech, seizures, intellectual disability, hypotonia, and motor coordination deficits. Motor abilities are an important outcome measure in AS as they comprise a broad repertoire of metrics including ataxia, hypotonia, delayed ambulation, crouched gait, and poor posture, and motor dysfunction affects nearly every individual with AS. Guided by collaborative work with AS clinicians studying gait, the goal of this study was to perform an in‐depth gait analysis using the automated treadmill assay, DigiGait. Our hypothesis is that gait presents a strong opportunity for a reliable, quantitative, and translational metric that can serve to evaluate novel pharmacological, dietary, and genetic therapies. In this study, we used an automated gait analysis system, in addition to standard motor behavioral assays, to evaluate components of motor, exploration, coordination, balance, and gait impairments across the lifespan in an AS mouse model. Our study demonstrated marked global motoric deficits in AS mice, corroborating previous reports. Uniquely, this is the first report of nuanced aberrations in quantitative spatial and temporal components of gait in AS mice compared to sex‐ and age‐matched wildtype littermates followed longitudinally using metrics that are analogous in AS individuals. Our findings contribute evidence toward the use of nuanced motor outcomes (i.e., gait) as valuable and translationally powerful metrics for therapeutic development for AS, as well as other genetic neurodevelopmental syndromes.
Collapse
Affiliation(s)
- Stela P Petkova
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, USA
| | - Anna Adhikari
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, USA
| | - Elizabeth L Berg
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, USA
| | - Timothy A Fenton
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, USA
| | - Jessica Duis
- Section of Genetics & Inherited Metabolic Disease, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anshutz Medical Campus, Aurora, Colorado, USA
| | - Jill L Silverman
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, USA
| |
Collapse
|
25
|
Dodge A, Morrill N, Weeber EJ, Nash KR. Recovery of Angelman syndrome rat deficits with UBE3A protein supplementation. Mol Cell Neurosci 2022; 120:103724. [DOI: 10.1016/j.mcn.2022.103724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/03/2022] [Accepted: 03/23/2022] [Indexed: 11/27/2022] Open
|
26
|
Cosgrove JA, Kelly LK, Kiffmeyer EA, Kloth AD. Sex-dependent influence of postweaning environmental enrichment in Angelman syndrome model mice. Brain Behav 2022; 12:e2468. [PMID: 34985196 PMCID: PMC8865162 DOI: 10.1002/brb3.2468] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/09/2021] [Accepted: 12/12/2021] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION Angelman syndrome (AS) is a rare neurodevelopmental disorder caused by mutation or loss of UBE3A and marked by intellectual disability, ataxia, autism-like symptoms, and other atypical behaviors. One route to treatment may lie in the role that environment plays early in postnatal life. Environmental enrichment (EE) is one manipulation that has shown therapeutic potential in preclinical models of many brain disorders, including neurodevelopmental disorders. Here, we examined whether postweaning EE can rescue behavioral phenotypes in Ube3a maternal deletion mice (AS mice), and whether any improvements are sex-dependent. METHODS Male and female mice (C57BL/6J Ube3atm1Alb mice and wild-type (WT) littermates; ≥10 mice/group) were randomly assigned to standard housing (SH) or EE at weaning. EE had a larger footprint, a running wheel, and a variety of toys that promoted foraging, burrowing, and climbing. Following 6 weeks of EE, animals were submitted to a battery of tests that reliably elicit behavioral deficits in AS mice, including rotarod, open field, marble burying, and forced swim; weights were also monitored. RESULTS In male AS-EE mice, we found complete restoration of motor coordination, marble burying, and forced swim behavior to the level of WT-SH mice. We also observed a complete normalization of exploratory distance traveled in the open field, but we found no rescue of vertical behavior or center time. AS-EE mice also had weights comparable to WT-SH mice. Intriguingly, in the female AS-EE mice, we found a failure of EE to rescue the same behavioral deficits relative to female WT-SH mice. CONCLUSIONS Environmental enrichment is an effective route to correcting the most penetrant phenotypes in male AS mice but not female AS mice. This finding has important implications for the translatability of early behavioral intervention for AS patients, most importantly the potential dependency of treatment response on sex.
Collapse
Affiliation(s)
- Jameson A. Cosgrove
- Department of BiologyAugustana University2001 S. Summit AvenueSioux FallsSouth DakotaUSA
| | - Lauren K. Kelly
- Department of BiologyAugustana University2001 S. Summit AvenueSioux FallsSouth DakotaUSA
| | - Elizabeth A. Kiffmeyer
- Department of BiologyAugustana University2001 S. Summit AvenueSioux FallsSouth DakotaUSA
| | - Alexander D. Kloth
- Department of BiologyAugustana University2001 S. Summit AvenueSioux FallsSouth DakotaUSA
| |
Collapse
|
27
|
Deng P, Halmai JANM, Beitnere U, Cameron D, Martinez ML, Lee CC, Waldo JJ, Thongphanh K, Adhikari A, Copping N, Petkova SP, Lee RD, Lock S, Palomares M, O’Geen H, Carter J, Gonzalez CE, Buchanan FKB, Anderson JD, Fierro FA, Nolta JA, Tarantal AF, Silverman JL, Segal DJ, Fink KD. An in vivo Cell-Based Delivery Platform for Zinc Finger Artificial Transcription Factors in Pre-clinical Animal Models. Front Mol Neurosci 2022; 14:789913. [PMID: 35153670 PMCID: PMC8829036 DOI: 10.3389/fnmol.2021.789913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/01/2021] [Indexed: 11/28/2022] Open
Abstract
Zinc finger (ZF), transcription activator-like effectors (TALE), and CRISPR/Cas9 therapies to regulate gene expression are becoming viable strategies to treat genetic disorders, although effective in vivo delivery systems for these proteins remain a major translational hurdle. We describe the use of a mesenchymal stem/stromal cell (MSC)-based delivery system for the secretion of a ZF protein (ZF-MSC) in transgenic mouse models and young rhesus monkeys. Secreted ZF protein from mouse ZF-MSC was detectable within the hippocampus 1 week following intracranial or cisterna magna (CM) injection. Secreted ZF activated the imprinted paternal Ube3a in a transgenic reporter mouse and ameliorated motor deficits in a Ube3a deletion Angelman Syndrome (AS) mouse. Intrathecally administered autologous rhesus MSCs were well-tolerated for 3 weeks following administration and secreted ZF protein was detectable within the cerebrospinal fluid (CSF), midbrain, and spinal cord. This approach is less invasive when compared to direct intracranial injection which requires a surgical procedure.
Collapse
Affiliation(s)
- Peter Deng
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Biochemistry and Molecular Medicine, Genome Center, University of California, Davis, Davis, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Julian A. N. M. Halmai
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Ulrika Beitnere
- Department of Biochemistry and Molecular Medicine, Genome Center, University of California, Davis, Davis, CA, United States
| | - David Cameron
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Michele L. Martinez
- Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, Gene Therapy Center, and California National Primate Research Center, University of California, Davis, Davis, CA, United States
| | - Charles C. Lee
- Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, Gene Therapy Center, and California National Primate Research Center, University of California, Davis, Davis, CA, United States
| | - Jennifer J. Waldo
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Krista Thongphanh
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States
| | - Anna Adhikari
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Nycole Copping
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Stela P. Petkova
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Ruth D. Lee
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Samantha Lock
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Miranda Palomares
- Department of Biochemistry and Molecular Medicine, Genome Center, University of California, Davis, Davis, CA, United States
| | - Henriette O’Geen
- Department of Biochemistry and Molecular Medicine, Genome Center, University of California, Davis, Davis, CA, United States
| | - Jasmine Carter
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Casiana E. Gonzalez
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Fiona K. B. Buchanan
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Johnathan D. Anderson
- Department of Otolaryngology, University of California, Davis, Davis, CA, United States
| | - Fernando A. Fierro
- Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States
| | - Jan A. Nolta
- Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States
| | - Alice F. Tarantal
- Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, Gene Therapy Center, and California National Primate Research Center, University of California, Davis, Davis, CA, United States
| | - Jill L. Silverman
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - David J. Segal
- Department of Biochemistry and Molecular Medicine, Genome Center, University of California, Davis, Davis, CA, United States
| | - Kyle D. Fink
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States,*Correspondence: Kyle D. Fink,
| |
Collapse
|
28
|
Lu YC, Chang TK, Yeh ST, Lin TC, Lin HS, Chen CH, Huang CH, Huang CH. Evaluation of graphene-derived bone scaffold exposure to the calvarial bone_ in-vitro and in-vivo studies. Nanotoxicology 2022; 16:1-15. [PMID: 35085045 DOI: 10.1080/17435390.2022.2027036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Graphene is a novel material which has recently been gaining great interest in the biomedical fields. Our previous study observed that graphene-derived particles help induce bone formation in a murine calvarial model. Here, we further developed a blended graphene-contained polycaprolactone (PCL/G) filament for application in a 3D-printed bone scaffold. Since implants are expected to be for long-term usage, in vitro cell culture and in vivo scaffold implants were evaluated in a critical-size bone defect calvarial model for over 60 weeks. Graphene greatly improved the mechanical strength by 30.2% compared to pure PCL. The fabricated PCL/G scaffolds also showed fine cell viability. In animal model, an abnormal electroencephalogram power spectrum and early signs of aging, such as hair graying and hair loss, were found in the group with a PCL/G scaffold compared to pure PCL scaffold. Neither of the abnormal symptoms caused death of all animals in both groups. The long-term use of graphene-derived biomaterials for in-vivo implants seems to be safe. But the comprehensive biosafety still needs further evaluation.
Collapse
Affiliation(s)
- Yung-Chang Lu
- Department of Medicine, MacKay Medical College, Taipei, Taiwan.,Department of Orthopaedic Surgery, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Ting-Kuo Chang
- Department of Medicine, MacKay Medical College, Taipei, Taiwan.,Department of Orthopaedic Surgery, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Shu-Ting Yeh
- Department of Orthopaedic Surgery, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Tzu-Chiao Lin
- Department of Orthopaedic Surgery, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Hung-Shih Lin
- Department of Neurosurgery, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chun-Hung Chen
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.,School of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Hsiung Huang
- Department of Orthopaedic Surgery, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Orthopaedic Surgery, Changhau Christian Hospital, Changhau, Taiwan
| | - Chang-Hung Huang
- Department of Orthopaedic Surgery, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.,School of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
29
|
Fontana BD, Alnassar N, Parker MO. The zebrafish (Danio rerio) anxiety test battery: comparison of behavioral responses in the novel tank diving and light-dark tasks following exposure to anxiogenic and anxiolytic compounds. Psychopharmacology (Berl) 2022; 239:287-296. [PMID: 34651212 PMCID: PMC8770442 DOI: 10.1007/s00213-021-05990-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/20/2021] [Indexed: 11/25/2022]
Abstract
RATIONALE Triangulation of approaches (i.e., using several tests of the same construct) can be extremely useful for increasing the robustness of the findings being widely used when working with behavioral testing, especially when using rodents as a translational model. Although zebrafish are widely used in neuropharmacology research due to their high-throughput screening potential for new therapeutic drugs, behavioral test battery effects following pharmacological manipulations are still unknown. METHODS Here, we tested the effects of an anxiety test battery and test time following pharmacological manipulations in zebrafish by using two behavioral tasks: the novel tank diving task (NTT) and the light-dark test (LDT). Fluoxetine and conspecific alarm substance (CAS) were chosen to induce anxiolytic and anxiogenic-like behavior, respectively. RESULTS For non-drug-treated animals, no differences were observed for testing order (NTT → LDT or LDT → NTT) and there was a strong correlation between performances on the two behavioral tasks. However, we found that during drug treatment, NTT/LDT responses are affected by the tested order depending on the test time being fluoxetine effects higher at the second behavioral task (6 min later) and CAS effects lower across time. CONCLUSIONS Overall, our data supports the use of baseline behavior assessment using this anxiety test battery. However, when working with drug exposure, data analysis must carefully consider time-drug-response and data variability across behavioral tasks.
Collapse
Affiliation(s)
- Barbara D Fontana
- Brain and Behaviour Laboratory, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Old St Michael's Building, White Swan Road, Portsmouth, PO1 2DT, UK.
| | - Nancy Alnassar
- Brain and Behaviour Laboratory, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Old St Michael's Building, White Swan Road, Portsmouth, PO1 2DT, UK
| | - Matthew O Parker
- Brain and Behaviour Laboratory, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Old St Michael's Building, White Swan Road, Portsmouth, PO1 2DT, UK.
| |
Collapse
|
30
|
Sell GL, Xin W, Cook EK, Zbinden MA, Schaffer TB, O'Meally RN, Cole RN, Margolis SS. Deleting a UBE3A substrate rescues impaired hippocampal physiology and learning in Angelman syndrome mice. Sci Rep 2021; 11:19414. [PMID: 34593829 PMCID: PMC8484563 DOI: 10.1038/s41598-021-97898-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/30/2021] [Indexed: 11/09/2022] Open
Abstract
In humans, loss-of-function mutations in the UBE3A gene lead to the neurodevelopmental disorder Angelman syndrome (AS). AS patients have severe impairments in speech, learning and memory, and motor coordination, for which there is currently no treatment. In addition, UBE3A is duplicated in > 1-2% of patients with autism spectrum disorders-a further indication of the significant role it plays in brain development. Altered expression of UBE3A, an E3 ubiquitin ligase, is hypothesized to lead to impaired levels of its target proteins, but identifying the contribution of individual UBE3A targets to UBE3A-dependent deficits remains of critical importance. Ephexin5 is a putative UBE3A substrate that has restricted expression early in development, regulates synapse formation during hippocampal development, and is abnormally elevated in AS mice, modeled by maternally-derived Ube3a gene deletion. Here, we report that Ephexin5 can be directly ubiquitylated by UBE3A. Furthermore, removing Ephexin5 from AS mice specifically rescued hippocampus-dependent behaviors, CA1 physiology, and deficits in dendritic spine number. Our findings identify Ephexin5 as a key driver of hippocampal dysfunction and related behavioral deficits in AS mouse models. These results demonstrate the exciting potential of targeting Ephexin5, and possibly other UBE3A substrates, to improve symptoms of AS and other UBE3A-related developmental disorders.
Collapse
Affiliation(s)
- Gabrielle L Sell
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Wood Basic Science Building Room 517, 725 N. Wolfe St., Baltimore, MD, 21205, USA.
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Center for Neuroscience, University of California-Davis, One Shields Avenue, Davis, CA, 95616, USA.
| | - Wendy Xin
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
- Department of Neurology and the Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Emily K Cook
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Wood Basic Science Building Room 517, 725 N. Wolfe St., Baltimore, MD, 21205, USA
| | - Mark A Zbinden
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Wood Basic Science Building Room 517, 725 N. Wolfe St., Baltimore, MD, 21205, USA
- Human Metabolome Technologies America, Inc., Boston, MA, 02134, USA
| | - Thomas B Schaffer
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Wood Basic Science Building Room 517, 725 N. Wolfe St., Baltimore, MD, 21205, USA
- NextCure Inc., Beltsville, MD, 20705, USA
| | - Robert N O'Meally
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Wood Basic Science Building Room 517, 725 N. Wolfe St., Baltimore, MD, 21205, USA
- Mass Spectrometry and Proteomics Facility, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Robert N Cole
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Wood Basic Science Building Room 517, 725 N. Wolfe St., Baltimore, MD, 21205, USA
- Mass Spectrometry and Proteomics Facility, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Seth S Margolis
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Wood Basic Science Building Room 517, 725 N. Wolfe St., Baltimore, MD, 21205, USA.
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
31
|
Berg EL, Petkova SP, Born HA, Adhikari A, Anderson AE, Silverman JL. Insulin-like growth factor-2 does not improve behavioral deficits in mouse and rat models of Angelman Syndrome. Mol Autism 2021; 12:59. [PMID: 34526125 PMCID: PMC8444390 DOI: 10.1186/s13229-021-00467-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 09/02/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Angelman Syndrome (AS) is a rare neurodevelopmental disorder for which there is currently no cure or effective therapeutic. Since the genetic cause of AS is known to be dysfunctional expression of the maternal allele of ubiquitin protein ligase E3A (UBE3A), several genetic animal models of AS have been developed. Both the Ube3a maternal deletion mouse and rat models of AS reliably demonstrate behavioral phenotypes of relevance to AS and therefore offer suitable in vivo systems in which to test potential therapeutics. One promising candidate treatment is insulin-like growth factor-2 (IGF-2), which has recently been shown to ameliorate behavioral deficits in the mouse model of AS and improve cognitive abilities across model systems. METHODS We used both the Ube3a maternal deletion mouse and rat models of AS to evaluate the ability of IGF-2 to improve electrophysiological and behavioral outcomes. RESULTS Acute systemic administration of IGF-2 had an effect on electrophysiological activity in the brain and on a metric of motor ability; however the effects were not enduring or extensive. Additional metrics of motor behavior, learning, ambulation, and coordination were unaffected and IGF-2 did not improve social communication, seizure threshold, or cognition. LIMITATIONS The generalizability of these results to humans is difficult to predict and it remains possible that dosing schemes (i.e., chronic or subchronic dosing), routes, and/or post-treatment intervals other than that used herein may show more efficacy. CONCLUSIONS Despite a few observed effects of IGF-2, our results taken together indicate that IGF-2 treatment does not profoundly improve behavioral deficits in mouse or rat models of AS. These findings shed cautionary light on the potential utility of acute systemic IGF-2 administration in the treatment of AS.
Collapse
Affiliation(s)
- Elizabeth L. Berg
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA USA
| | - Stela P. Petkova
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA USA
| | - Heather A. Born
- Department of Pediatrics and Neurology, Baylor College of Medicine, Houston, TX USA
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Anna Adhikari
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA USA
| | - Anne E. Anderson
- Department of Pediatrics and Neurology, Baylor College of Medicine, Houston, TX USA
| | - Jill L. Silverman
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA USA
| |
Collapse
|
32
|
Copping NA, McTighe SM, Fink KD, Silverman JL. Emerging Gene and Small Molecule Therapies for the Neurodevelopmental Disorder Angelman Syndrome. Neurotherapeutics 2021; 18:1535-1547. [PMID: 34528170 PMCID: PMC8608975 DOI: 10.1007/s13311-021-01082-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2021] [Indexed: 02/07/2023] Open
Abstract
Angelman syndrome (AS) is a rare (~1:15,000) neurodevelopmental disorder characterized by severe developmental delay and intellectual disability, impaired communication skills, and a high prevalence of seizures, sleep disturbances, ataxia, motor deficits, and microcephaly. AS is caused by loss-of-function of the maternally inherited UBE3A gene. UBE3A is located on chromosome 15q11-13 and is biallelically expressed throughout the body but only maternally expressed in the brain due to an RNA antisense transcript that silences the paternal copy. There is currently no cure for AS, but advancements in small molecule drugs and gene therapies offer a promising approach for the treatment of the disorder. Here, we review AS and how loss-of-function of the maternal UBE3A contributes to the disorder. We also discuss the strengths and limitations of current animal models of AS. Furthermore, we examine potential small molecule drug and gene therapies for the treatment of AS and associated challenges faced by the therapeutic design. Finally, gene therapy offers the opportunity for precision medicine in AS and advancements in the treatment of this disorder can serve as a foundation for other single-gene neurodevelopmental disorders.
Collapse
Affiliation(s)
- Nycole A Copping
- School of Medicine, Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California, Research II Building 96, 4625 2nd Avenue, Suite 1001B, Davis, Sacramento, CA, 95817, USA
- Stem Cell Program and Gene Therapy Center, Department of Neurology, MIND Institute, University of California, Davis, Sacramento, CA, USA
| | | | - Kyle D Fink
- Stem Cell Program and Gene Therapy Center, Department of Neurology, MIND Institute, University of California, Davis, Sacramento, CA, USA
| | - Jill L Silverman
- School of Medicine, Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California, Research II Building 96, 4625 2nd Avenue, Suite 1001B, Davis, Sacramento, CA, 95817, USA.
| |
Collapse
|
33
|
Moreira-de-Sá A, Gonçalves FQ, Lopes JP, Silva HB, Tomé ÂR, Cunha RA, Canas PM. Motor Deficits Coupled to Cerebellar and Striatal Alterations in Ube3a m-/p+ Mice Modelling Angelman Syndrome Are Attenuated by Adenosine A 2A Receptor Blockade. Mol Neurobiol 2021; 58:2543-2557. [PMID: 33464534 DOI: 10.1007/s12035-020-02275-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/28/2020] [Indexed: 01/22/2023]
Abstract
Angelman syndrome (AS) is a neurogenetic disorder involving ataxia and motor dysfunction, resulting from the absence of the maternally inherited functional Ube3a protein in neurons. Since adenosine A2A receptor (A2AR) blockade relieves synaptic and motor impairments in Parkinson's or Machado-Joseph's diseases, we now tested if A2AR blockade was also effective in attenuating motor deficits in an AS (Ube3am-/p+) mouse model and if this involved correction of synaptic alterations in striatum and cerebellum. Chronic administration of the A2AR antagonist SCH58261 (0.1 mg/kg/day, ip) promoted motor learning of AS mice in the accelerating-rotarod task and rescued the grip strength impairment of AS animals. These motor impairments were accompanied by synaptic alterations in cerebellum and striatum typified by upregulation of synaptophysin and vesicular GABA transporters (vGAT) in the cerebellum of AS mice along with a downregulation of vGAT, vesicular glutamate transporter 1 (vGLUT1) and the dopamine active transporter in AS striatum. Notably, A2AR blockade prevented the synaptic alterations found in AS mice cerebellum as well as the downregulation of striatal vGAT and vGLUT1. This provides the first indications that A2AR blockade may counteract the characteristic motor impairments and synaptic changes of AS, although more studies are needed to unravel the underlying mechanisms.
Collapse
Affiliation(s)
- Ana Moreira-de-Sá
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine Building-Polo 1, 3004-504, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Francisco Q Gonçalves
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine Building-Polo 1, 3004-504, Coimbra, Portugal
| | - João P Lopes
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine Building-Polo 1, 3004-504, Coimbra, Portugal
| | - Henrique B Silva
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine Building-Polo 1, 3004-504, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Ângelo R Tomé
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine Building-Polo 1, 3004-504, Coimbra, Portugal
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456, Coimbra, Portugal
| | - Rodrigo A Cunha
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine Building-Polo 1, 3004-504, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Paula M Canas
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine Building-Polo 1, 3004-504, Coimbra, Portugal.
| |
Collapse
|
34
|
Adhikari A, Copping NA, Beegle J, Cameron DL, Deng P, O'Geen H, Segal DJ, Fink KD, Silverman JL, Anderson JS. Functional rescue in an Angelman syndrome model following treatment with lentivector transduced hematopoietic stem cells. Hum Mol Genet 2021; 30:1067-1083. [PMID: 33856035 PMCID: PMC8188406 DOI: 10.1093/hmg/ddab104] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 12/14/2022] Open
Abstract
Angelman syndrome (AS) is a rare neurodevelopmental disorder characterized by impaired communication skills, ataxia, motor and balance deficits, intellectual disabilities, and seizures. The genetic cause of AS is the neuronal loss of UBE3A expression in the brain. A novel approach, described here, is a stem cell gene therapy which uses lentivector-transduced hematopoietic stem and progenitor cells to deliver functional UBE3A to affected cells. We have demonstrated both the prevention and reversal of AS phenotypes upon transplantation and engraftment of human CD34+ cells transduced with a Ube3a lentivector in a novel immunodeficient Ube3amat−/pat+ IL2rg−/y mouse model of AS. A significant improvement in motor and cognitive behavioral assays as well as normalized delta power measured by electroencephalogram was observed in neonates and adults transplanted with the gene modified cells. Human hematopoietic profiles observed in the lymphoid organs by detection of human immune cells were normal. Expression of UBE3A was detected in the brains of the adult treatment group following immunohistochemical staining illustrating engraftment of the gene-modified cells expressing UBE3A in the brain. As demonstrated with our data, this stem cell gene therapy approach offers a promising treatment strategy for AS, not requiring a critical treatment window.
Collapse
Affiliation(s)
- Anna Adhikari
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Nycole A Copping
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Julie Beegle
- Stem Cell Program, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - David L Cameron
- Stem Cell Program, Department of Neurology, Institute for Regenerative Cures, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Peter Deng
- Stem Cell Program, Department of Neurology, Institute for Regenerative Cures, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Henriette O'Geen
- Department of Biochemistry and Medical Microbiology, UC Davis Genome Center, University of California Davis School of Medicine, Davis, CA 95616, USA
| | - David J Segal
- Department of Biochemistry and Medical Microbiology, UC Davis Genome Center, University of California Davis School of Medicine, Davis, CA 95616, USA
| | - Kyle D Fink
- Stem Cell Program, Department of Neurology, Institute for Regenerative Cures, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Jill L Silverman
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Joseph S Anderson
- Stem Cell Program, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| |
Collapse
|
35
|
Egawa K, Nakakubo S, Kimura S, Goto T, Manabe A, Shiraishi H. Flurothyl-induced seizure paradigm revealed higher seizure susceptibility in middle-aged Angelman syndrome mouse model. Brain Dev 2021; 43:515-520. [PMID: 33408038 DOI: 10.1016/j.braindev.2020.12.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/24/2020] [Accepted: 12/17/2020] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Epilepsy is one of the main clinical problems in Angelman syndrome (AS). Seizures typically start in early childhood then decrease or are often alleviated by young adulthood. Several studies using AS model mice showed comparable seizure susceptibility during young adulthood. In contrast, the course of epilepsy post young adulthood differs from persistently relieved to rerising among reports. To elucidate this, we evaluated the seizure susceptibility of AS model mice of two different ages. METHODS Mice lacking maternal Ube3a gene (Ube3am-/p+) of C57BL/6 background or their littermate wild type (WT) were divided into two groups by age, 2 to 3 months (2-3 M) and 6 to 12 months (6-12 M), corresponding to adolescent to young adult aged and middle aged humans, respectively. Seizure susceptibility was evaluated by flurothyl inhalation or intraperitoneal injection of pentylenetetrazole (PTZ IP)-induced acute seizure protocol. RESULTS In the flurothyl-induced seizure paradigm, the latency to seizure occurrence had a significant interaction with genotype and age. Post-hoc analysis revealed that the latency was significantly shorter at 6-12 M than at 2-3 M in Ube3am-/p+ mice, and in Ube3am-/p+ mice than in WT mice at 6-12 M. No significant interaction or difference was observed by PTZ IP. CONCLUSION The flurothyl-induced seizure paradigm revealed that seizure susceptibility of Ube3am-/p+ mice increased with age, similar to clinical studies reporting the reappearance of epilepsy in older age. The flurothyl-induced seizure paradigm applied to middle-aged Ube3am-/p+ mice could be a suitable protocol for screening drugs against seizures in AS.
Collapse
Affiliation(s)
- Kiyoshi Egawa
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo 060-8638, Japan
| | - Sachiko Nakakubo
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo 060-8638, Japan
| | - Shuhei Kimura
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo 060-8638, Japan
| | - Takeru Goto
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo 060-8638, Japan
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo 060-8638, Japan
| | - Hideaki Shiraishi
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo 060-8638, Japan.
| |
Collapse
|
36
|
Early Developmental EEG and Seizure Phenotypes in a Full Gene Deletion of Ubiquitin Protein Ligase E3A Rat Model of Angelman Syndrome. eNeuro 2021; 8:ENEURO.0345-20.2020. [PMID: 33531368 PMCID: PMC8114899 DOI: 10.1523/eneuro.0345-20.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
Angelman syndrome (AS) is a neurodevelopmental disorder with unique behavioral phenotypes, seizures, and distinctive electroencephalographic (EEG) patterns. Recent studies identified motor, social communication, and learning and memory deficits in a CRISPR engineered rat model with a complete maternal deletion of the Ube3a gene. It is unknown whether this model recapitulates other aspects of the clinical disorder. We report here the effect of Ube3a maternal deletion in the rat on epileptiform activity, seizure threshold, and quantitative EEG. Using video-synchronized EEG (vEEG) monitoring, we assessed spectral power and epileptiform activity early postnatally through adulthood. While EEG power was similar to wild-type (WT) at 1.5 weeks postnatally, at all other ages analyzed, our findings were similar to the AS phenotype in mice and humans with significantly increased δ power. Analysis of epileptiform activity in juvenile and adult rats showed increased time spent in epileptiform activity in AS compared with WT rats. We evaluated seizure threshold using pentylenetetrazol (PTZ), audiogenic stimulus, and hyperthermia to provoke febrile seizures (FSs). Behavioral seizure scoring following PTZ induction revealed no difference in seizure threshold in AS rats, however behavioral recovery from the PTZ-induced seizure was longer in the adult group with significantly increased hippocampal epileptiform activity during this phase. When exposed to hyperthermia, AS rat pups showed a significantly lower temperature threshold to first seizure than WT. Our findings highlight an age-dependence for the EEG and epileptiform phenotypes in a preclinical model of AS, and support the use of quantitative EEG and increased δ power as a potential biomarker of AS.
Collapse
|
37
|
Copping NA, Silverman JL. Abnormal electrophysiological phenotypes and sleep deficits in a mouse model of Angelman Syndrome. Mol Autism 2021; 12:9. [PMID: 33549123 PMCID: PMC7866697 DOI: 10.1186/s13229-021-00416-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 01/18/2021] [Indexed: 01/17/2023] Open
Abstract
Background Angelman Syndrome (AS) is a rare genetic disorder characterized by impaired communication, motor and balance deficits, intellectual disabilities, recurring seizures and abnormal sleep patterns. The genetic cause of AS is neuronal-specific loss of expression of UBE3A (ubiquitin-protein ligase E6-AP), an imprinted gene. Seizure and sleep disorders are highly prevalent (> 80%) in the AS population. The present experiments were designed to identify translational, neurophysiological outcome measures in a model of AS. Methods We used the exon-2 deletion mouse (Ube3a-del) on a C57BL/6J background to assess seizure, sleep and electrophysiological phenotypes. Seizure susceptibility has been reported in Ube3a-del mice with a variety of seizure induction methods. Here, we provoked seizures by a single high-dose injection of 80 mg/kg pentylenetetrazole. Novel experiments included the utilization of wireless telemetry devices to acquire global electroencephalogram (EEG) and neurophysiological data on electrographic seizures, power spectra, light–dark cycles, sleep stages and sleep spindles in Ube3a-del and WT mice. Results Ube3a-del mice exhibited reduced seizure threshold compared to WT. EEG illustrated that Ube3a-del mice had increased epileptiform spiking activity and delta power, which corroborates findings from other laboratories and recapitulates clinical reports in AS. This is the first report to use a cortical surface-based recording by a wireless telemetry device over tethered/fixed head-mount depth recordings. Less time in both paradoxical and slow-wave sleep, longer latencies to paradoxical sleep stages and total less sleep time in Ube3a-del mice were observed compared to WT. For the first time, we detected fewer sleep spindles in the AS mouse model. Limitations This study was limited to the exon 2 deletion mouse model, and future work will investigate the rat model of AS, containing a complete Ube3a deletion and pair EEG with behavior. Conclusions Our data enhance rigor and translatability as our study provides important corroboration of previous reports on epileptiform and elevated delta power. For the first time we report neurophysiological phenotypes collected via translational methodology. Furthermore, this is the first report of reduced sleep spindles, a critical marker of memory consolidation during sleep, in an AS model. Our results are useful outcomes for therapeutic testing.
Collapse
Affiliation(s)
- N A Copping
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001B, Research II Building 96, 4625 2nd Avenue, Sacramento, CA, 95817, USA
| | - J L Silverman
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001B, Research II Building 96, 4625 2nd Avenue, Sacramento, CA, 95817, USA.
| |
Collapse
|
38
|
Perrino PA, Chamberlain SJ, Eigsti IM, Fitch RH. Communication-related assessments in an Angelman syndrome mouse model. Brain Behav 2021; 11:e01937. [PMID: 33151040 PMCID: PMC7821623 DOI: 10.1002/brb3.1937] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 10/06/2020] [Accepted: 10/18/2020] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Angelman syndrome (AS) is a neurodevelopmental disorder characterized by motor deficits, seizures, some autistic-like behaviors, and severe impairment of speech. A dysfunction of the maternally imprinted UBE3A gene, coupled with a functional yet silenced paternal copy, results in AS. Although studies of transgenic mouse models have revealed a great deal about neural populations and rescue timeframes for specific features of AS, these studies have largely failed to examine intermediate phenotypes that contribute to the profound communicative disabilities associated with AS. METHODS Here, we use a variety of tasks, including assessments of rapid auditory processing and social communication. Expressive vocalizations were directly assessed and correlated against other core behavioral measures (motor, social, acoustic perception) to model putative influences on communication. RESULTS AS mice displayed the characteristic phenotypes associated with Angelman syndrome (i.e., social and motor deficits), as well as marginal enhancements in rapid auditory processing ability. Our characterization of adult ultrasonic vocalizations further showed that AS mice produce fewer vocalizations and vocalized for a shorter amount of time when compared to controls. Additionally, a strong correlation between motor indices and ultrasonic vocalization output was shown, suggesting that the motor impairments in AS may contribute heavily to communication impairments. CONCLUSION In summary, the combination of motor deficits, social impairment, marginal rapid auditory enhancements, and altered ultrasonic vocalizations reported in a mouse model of AS clearly parallel the human symptoms of the disorder. This mouse model offers a novel route to interrogate the underlying genetic, physiologic, and behavioral influences on the under-studied topic of impaired communication in AS.
Collapse
Affiliation(s)
- Peter A Perrino
- Department of Psychological Science/Behavioral Neuroscience, University of Connecticut, Storrs, CT, USA
| | - Stormy J Chamberlain
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Inge-Marie Eigsti
- Department of Psychological Science/Clinical Psychology, University of Connecticut, Storrs, CT, USA
| | - Roslyn Holly Fitch
- Department of Psychological Science/Behavioral Neuroscience, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
39
|
Cruz E, Descalzi G, Steinmetz A, Scharfman HE, Katzman A, Alberini CM. CIM6P/IGF-2 Receptor Ligands Reverse Deficits in Angelman Syndrome Model Mice. Autism Res 2021; 14:29-45. [PMID: 33108069 PMCID: PMC8579913 DOI: 10.1002/aur.2418] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/16/2020] [Accepted: 10/09/2020] [Indexed: 11/12/2022]
Abstract
Angelman syndrome (AS), a genetic disorder that primarily affects the nervous system, is characterized by delayed development, intellectual disability, severe speech impairment, and problems with movement and balance (ataxia). Most affected children also have recurrent seizures (epilepsy). No existing therapies are capable of comprehensively treating the deficits in AS; hence, there is an urgent need to identify new treatments. Here we show that insulin-like growth factor 2 (IGF-2) and mannose-6-phosphate (M6P), ligands of two independent binding sites of the cation-independent M6P/IGF-2 receptor (CIM6P/IGF-2R), reverse most major deficits of AS modeled in mice. Subcutaneous injection of IGF-2 or M6P in mice modeling AS restored cognitive impairments as assessed by measurements of contextual and recognition memories, motor deficits assessed by rotarod and hindlimb clasping, and working memory/flexibility measured by Y-maze. IGF-2 also corrected deficits in marble burying and significantly attenuated acoustically induced seizures. An observational battery of tests confirmed that neither ligand changed basic functions including physical characteristics, general behavioral responses, and sensory reflexes, indicating that they are relatively safe. Our data provide strong preclinical evidence that targeting CIM6P/IGF-2R is a promising approach for developing novel therapeutics for AS. LAY SUMMARY: There is no effective treatment for the neurodevelopmental disorder Angelman syndrome (AS). Using a validated AS mouse model, the Ube3am-/p+ , in this study we show that systemic administration of ligands of the cation independent mannose-6-phosphate receptor, also known as insulin-like growth factor 2 receptor (CIM6P/IGF-2R) reverses cognitive impairment, motor deficits, as well as seizures associated with AS. Thus, ligands that activate the CIM6P/IGF-2R may represent novel, potential therapeutic targets for AS.
Collapse
Affiliation(s)
- Emmanuel Cruz
- Center for Neural Science, New York University, New York, New York, USA
| | - Giannina Descalzi
- Center for Neural Science, New York University, New York, New York, USA
| | - Adam Steinmetz
- Center for Neural Science, New York University, New York, New York, USA
| | - Helen E Scharfman
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, USA
- Department of Neuroscience and Physiology, New York University Langone Health, New York, New York, USA
- Department of Psychiatry, New York University Langone Health, New York, New York, USA
| | - Aaron Katzman
- Center for Neural Science, New York University, New York, New York, USA
| | | |
Collapse
|
40
|
Sonzogni M, Zhai P, Mientjes EJ, van Woerden GM, Elgersma Y. Assessing the requirements of prenatal UBE3A expression for rescue of behavioral phenotypes in a mouse model for Angelman syndrome. Mol Autism 2020; 11:70. [PMID: 32948244 PMCID: PMC7501605 DOI: 10.1186/s13229-020-00376-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/04/2020] [Indexed: 11/26/2022] Open
Abstract
Background Angelman syndrome (AS) is a rare neurodevelopmental disorder caused by the loss of functional ubiquitin protein ligase E3A (UBE3A). In neurons, UBE3A expression is tightly regulated by a mechanism of imprinting which suppresses the expression of the paternal UBE3A allele. Promising treatment strategies for AS are directed at activating paternal UBE3A gene expression. However, for such strategies to be successful, it is important to know when such a treatment should start, and how much UBE3A expression is needed for normal embryonic brain development. Methods Using a conditional mouse model of AS, we further delineated the critical period for UBE3A expression during early brain development. Ube3a gene expression was induced around the second week of gestation and mouse phenotypes were assessed using a behavioral test battery. To investigate the requirements of embryonic UBE3A expression, we made use of mice in which the paternal Ube3a allele was deleted. Results We observed a full behavioral rescue of the AS mouse model phenotypes when Ube3a gene reactivation was induced around the start of the last week of mouse embryonic development. We found that full silencing of the paternal Ube3a allele was not completed till the first week after birth but that deletion of the paternal Ube3a allele had no significant effect on the assessed phenotypes. Limitations Direct translation to human is limited, as we do not precisely know how human and mouse brain development aligns over gestational time. Moreover, many of the assessed phenotypes have limited translational value, as the underlying brain regions involved in these tasks are largely unknown. Conclusions Our findings provide further important insights in the requirement of UBE3A expression during brain development. We found that loss of up to 50% of UBE3A protein during prenatal mouse brain development does not significantly impact the assessed mouse behavioral phenotypes. Together with previous findings, our results indicate that the most critical function for mouse UBE3A lies in the early postnatal period between birth and P21.
Collapse
Affiliation(s)
- Monica Sonzogni
- Department of Neuroscience and the ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Peipei Zhai
- Department of Neuroscience and the ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, 3015 CN, Rotterdam, The Netherlands.,Department of Neurology, The First Affiliated Hospital of Henan University, No.357, Ximendajie Street, Kaifeng City, Henan Province, China
| | - Edwin J Mientjes
- Department of Neuroscience and the ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Geeske M van Woerden
- Department of Neuroscience and the ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Ype Elgersma
- Department of Neuroscience and the ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, 3015 CN, Rotterdam, The Netherlands.
| |
Collapse
|
41
|
Javed S, Selliah T, Lee YJ, Huang WH. Dosage-sensitive genes in autism spectrum disorders: From neurobiology to therapy. Neurosci Biobehav Rev 2020; 118:538-567. [PMID: 32858083 DOI: 10.1016/j.neubiorev.2020.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/26/2020] [Accepted: 08/17/2020] [Indexed: 12/24/2022]
Abstract
Autism spectrum disorders (ASDs) are a group of heterogenous neurodevelopmental disorders affecting 1 in 59 children. Syndromic ASDs are commonly associated with chromosomal rearrangements or dosage imbalance involving a single gene. Many of these genes are dosage-sensitive and regulate transcription, protein homeostasis, and synaptic function in the brain. Despite vastly different molecular perturbations, syndromic ASDs share core symptoms including social dysfunction and repetitive behavior. However, each ASD subtype has a unique pathogenic mechanism and combination of comorbidities that require individual attention. We have learned a great deal about how these dosage-sensitive genes control brain development and behaviors from genetically-engineered mice. Here we describe the clinical features of eight monogenic neurodevelopmental disorders caused by dosage imbalance of four genes, as well as recent advances in using genetic mouse models to understand their pathogenic mechanisms and develop intervention strategies. We propose that applying newly developed quantitative molecular and neuroscience technologies will advance our understanding of the unique neurobiology of each disorder and enable the development of personalized therapy.
Collapse
Affiliation(s)
- Sehrish Javed
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Tharushan Selliah
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Yu-Ju Lee
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Wei-Hsiang Huang
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.
| |
Collapse
|
42
|
Lopez SJ, Laufer BI, Beitnere U, Berg EL, Silverman JL, O'Geen H, Segal DJ, LaSalle JM. Imprinting effects of UBE3A loss on synaptic gene networks and Wnt signaling pathways. Hum Mol Genet 2020; 28:3842-3852. [PMID: 31625566 DOI: 10.1093/hmg/ddz221] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/21/2019] [Accepted: 09/04/2019] [Indexed: 12/13/2022] Open
Abstract
Ubiquitin E3 ligase 3A (UBE3A) encodes an E3 ubiquitin ligase whose loss from the maternal allele causes the neurodevelopmental disorder Angelman syndrome (AS). Previous studies of UBE3A function have not examined full Ube3a deletion in mouse, the complexity of imprinted gene networks in brain nor the molecular basis of systems-level cognitive dysfunctions in AS. We therefore utilized a systems biology approach to elucidate how UBE3A loss impacts the early postnatal brain in a novel CRISPR/Cas9-engineered rat Angelman model of a complete Ube3a deletion. Strand-specific transcriptome analysis of offspring from maternally or paternally inherited Ube3a deletions revealed the expected parental expression patterns of Ube3a sense and antisense transcripts by postnatal day 2 (P2) in hypothalamus and day 9 (P9) in cortex, compared to wild-type littermates. The dependency of genome-wide effects on parent-of-origin, Ube3a genotype and time (P2 and P9) was investigated through transcriptome (RNA sequencing of cortex and hypothalamus) and methylome (whole-genome bisulfite sequencing of hypothalamus). Weighted gene co-expression and co-methylation network analyses identified co-regulated networks in maternally inherited Ube3a deletion offspring enriched in postnatal developmental processes including Wnt signaling, synaptic regulation, neuronal and glial functions, epigenetic regulation, ubiquitin, circadian entrainment and splicing. Furthermore, we showed that loss of the paternal Ube3a antisense transcript resulted in both unique and overlapping dysregulated gene pathways with maternal loss, predominantly at the level of differential methylation. Together, these results provide a holistic examination of the molecular impacts of UBE3A loss in brain, supporting the existence of interactive epigenetic networks between maternal and paternal transcripts at the Ube3a locus.
Collapse
Affiliation(s)
- S Jesse Lopez
- Medical Immunology and Microbiology, University of California (UC) Davis School of Medicine, Davis, CA 95616, USA.,Genome Center, UC Davis, Davis, CA, USA.,Integrative Genetics and Genomics, UC Davis, Davis, CA 95616, USA.,Biochemistry and Molecular Medicine, UC Davis School of Medicine, Davis, CA 95616, USA.,Medical Investigation of Neurodevelopmental Disorders Institute, UC Davis Health, Sacramento, CA 95817, USA
| | - Benjamin I Laufer
- Medical Immunology and Microbiology, University of California (UC) Davis School of Medicine, Davis, CA 95616, USA.,Genome Center, UC Davis, Davis, CA, USA.,Medical Investigation of Neurodevelopmental Disorders Institute, UC Davis Health, Sacramento, CA 95817, USA
| | - Ulrika Beitnere
- Genome Center, UC Davis, Davis, CA, USA.,Biochemistry and Molecular Medicine, UC Davis School of Medicine, Davis, CA 95616, USA.,Medical Investigation of Neurodevelopmental Disorders Institute, UC Davis Health, Sacramento, CA 95817, USA
| | - Elizabeth L Berg
- Medical Investigation of Neurodevelopmental Disorders Institute, UC Davis Health, Sacramento, CA 95817, USA.,Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacromento, CA 95817, USA
| | - Jill L Silverman
- Medical Investigation of Neurodevelopmental Disorders Institute, UC Davis Health, Sacramento, CA 95817, USA.,Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacromento, CA 95817, USA
| | - Henriette O'Geen
- Genome Center, UC Davis, Davis, CA, USA.,Biochemistry and Molecular Medicine, UC Davis School of Medicine, Davis, CA 95616, USA.,Medical Investigation of Neurodevelopmental Disorders Institute, UC Davis Health, Sacramento, CA 95817, USA
| | - David J Segal
- Genome Center, UC Davis, Davis, CA, USA.,Integrative Genetics and Genomics, UC Davis, Davis, CA 95616, USA.,Biochemistry and Molecular Medicine, UC Davis School of Medicine, Davis, CA 95616, USA.,Medical Investigation of Neurodevelopmental Disorders Institute, UC Davis Health, Sacramento, CA 95817, USA
| | - Janine M LaSalle
- Medical Immunology and Microbiology, University of California (UC) Davis School of Medicine, Davis, CA 95616, USA.,Genome Center, UC Davis, Davis, CA, USA.,Integrative Genetics and Genomics, UC Davis, Davis, CA 95616, USA.,Medical Investigation of Neurodevelopmental Disorders Institute, UC Davis Health, Sacramento, CA 95817, USA
| |
Collapse
|
43
|
Gu B, Zhu M, Glass MR, Rougié M, Nikolova VD, Moy SS, Carney PR, Philpot BD. Cannabidiol attenuates seizures and EEG abnormalities in Angelman syndrome model mice. J Clin Invest 2020; 129:5462-5467. [PMID: 31503547 DOI: 10.1172/jci130419] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/03/2019] [Indexed: 01/08/2023] Open
Abstract
Angelman syndrome (AS) is a neurodevelopmental disorder characterized by intellectual disability, lack of speech, ataxia, EEG abnormalities, and epilepsy. Seizures in individuals with AS are common, debilitating, and often drug resistant. Thus, there is an unmet need for better treatment options. Cannabidiol (CBD), a major phytocannabinoid constituent of cannabis, has shown antiseizure activity and behavioral benefits in preclinical and clinical studies for some disorders associated with epilepsy, suggesting that the same could be true for AS. Here, we show that acute CBD (100 mg/kg) treatment attenuated hyperthermia- and acoustically induced seizures in a mouse model of AS. However, neither acute CBD nor a 2-week-long course of CBD administered immediately after a kindling protocol could halt the proepileptogenic plasticity observed in AS model mice. CBD had a dose-dependent sedative effect but did not have an impact on motor performance. CBD abrogated the enhanced intracortical local field potential power, including the delta and theta rhythms observed in AS model mice, indicating that CBD administration could also help normalize the EEG deficits observed in individuals with AS. We believe our results provide critical preclinical evidence supporting CBD treatment of seizures and alleviation of EEG abnormalities in AS and will thus help guide the rational development of CBD as a treatment for AS.
Collapse
Affiliation(s)
- Bin Gu
- Department of Cell Biology and Physiology.,Neuroscience Center
| | | | | | | | | | - Sheryl S Moy
- Department of Psychiatry.,Carolina Institute for Developmental Disabilities
| | - Paul R Carney
- Neuroscience Curriculum.,Carolina Institute for Developmental Disabilities.,Department of Neurology, and.,Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Benjamin D Philpot
- Department of Cell Biology and Physiology.,Neuroscience Center.,Carolina Institute for Developmental Disabilities
| |
Collapse
|
44
|
Pirbhoy PS, Rais M, Lovelace JW, Woodard W, Razak KA, Binder DK, Ethell IM. Acute pharmacological inhibition of matrix metalloproteinase-9 activity during development restores perineuronal net formation and normalizes auditory processing in Fmr1 KO mice. J Neurochem 2020; 155:538-558. [PMID: 32374912 DOI: 10.1111/jnc.15037] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/31/2020] [Accepted: 04/30/2020] [Indexed: 12/13/2022]
Abstract
Individuals with Fragile X Syndrome (FXS) and autism spectrum disorder (ASD) exhibit cognitive impairments, social deficits, increased anxiety, and sensory hyperexcitability. Previously, we showed that elevated levels of matrix metalloproteinase-9 (MMP-9) may contribute to abnormal development of parvalbumin (PV) interneurons and perineuronal nets (PNNs) in the developing auditory cortex (AC) of Fmr1 knock-out (KO) mice, which likely underlie auditory hypersensitivity. Thus, MMP-9 may serve as a potential target for treatment of auditory hypersensitivity in FXS. Here, we used the MMP-2/9 inhibitor, SB-3CT, to pharmacologically inhibit MMP-9 activity during a specific developmental period and to test whether inhibition of MMP-9 activity reverses neural oscillation deficits and behavioral impairments by enhancing PNN formation around PV cells in Fmr1 KO mice. Electroencephalography (EEG) was used to measure resting state and sound-evoked electrocortical activity in auditory and frontal cortices of postnatal day (P)22-23 male mice before and one-day after treatment with SB-3CT (25 mg/kg) or vehicle. At P27-28, animal behaviors were tested to measure the effects of the treatment on anxiety and hyperactivity. Results show that acute inhibition of MMP-9 activity improved evoked synchronization to auditory stimuli and ameliorated mouse behavioral deficits. MMP-9 inhibition enhanced PNN formation, increased PV levels and TrkB phosphorylation yet reduced Akt phosphorylation in the AC of Fmr1 KO mice. Our results show that MMP-9 inhibition during early postnatal development is beneficial in reducing some auditory processing deficits in the FXS mouse model and may serve as a candidate therapeutic for reversing sensory hypersensitivity in FXS and possibly other ASDs.
Collapse
Affiliation(s)
- Patricia S Pirbhoy
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Maham Rais
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Jonathan W Lovelace
- Department of Psychology, University of California Riverside, Riverside, CA, USA
| | - Walker Woodard
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Khaleel A Razak
- Department of Psychology, University of California Riverside, Riverside, CA, USA
| | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| |
Collapse
|
45
|
Tsagkaris C, Papakosta V, Miranda AV, Zacharopoulou L, Danilchenko V, Matiashova L, Dhar A. Gene Therapy for Angelman Syndrome: Contemporary Approaches and Future Endeavors. Curr Gene Ther 2020; 19:359-366. [DOI: 10.2174/1566523220666200107151025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/28/2019] [Accepted: 01/01/2020] [Indexed: 01/20/2023]
Abstract
Background:
Angelman Syndrome (AS) is a congenital non inherited neurodevelopmental
disorder. The contemporary AS management is symptomatic and it has been accepted that gene therapy
may play a key role in the treatment of AS.
Objective:
The purpose of this study is to summarize existing and suggested gene therapy approaches
to Angelman syndrome.
Methods:
This is a literature review. Pubmed and Scopus databases were researched with keywords
(gene therapy, Angelman’s syndrome, neurological disorders, neonates). Peer-reviewed studies that
were closely related to gene therapies in Angelman syndrome and available in English, Greek, Ukrainian
or Indonesian were included. Studies that were published before 2000 were excluded and did not
align with the aforementioned criteria.
Results:
UBE3A serves multiple roles in signaling and degradation procedures. Although the restoration
of UBE3A expression rather than targeting known activities of the molecule would be the optimal
therapeutic goal, it is not possible so far. Reinstatement of paternal UBE3A appears as an adequate alternative.
This can be achieved by administering topoisomerase-I inhibitors or reducing UBE3A antisense
transcript (UBE3A-ATS), a molecule which silences paternal UBE3A.
Conclusion:
Understanding UBE3A imprinting unravels the path to an etiologic treatment of AS.
Gene therapy models tested on mice appeared less effective than anticipated pointing out that activation
of paternal UBE3A cannot counteract the existing CNS defects. On the other hand, targeting abnormal
downstream cell signaling pathways has provided promising rescue effects. Perhaps, combined
reinstatement of paternal UBE3A expression with abnormal signaling pathways-oriented treatment is
expected to provide better therapeutic effects. However, AS gene therapy remains debatable in pharmacoeconomics
and ethics context.
Collapse
Affiliation(s)
| | | | | | | | - Valeriia Danilchenko
- Department of Pediatrics #1 with Propaedeutics and Neonatology, Ukrainian Medical Stomatological Academy, Poltava, Ukraine
| | | | - Amrit Dhar
- Government Medical College, Jammu and Kashmir, India
| |
Collapse
|
46
|
Maranga C, Fernandes TG, Bekman E, da Rocha ST. Angelman syndrome: a journey through the brain. FEBS J 2020; 287:2154-2175. [PMID: 32087041 DOI: 10.1111/febs.15258] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/02/2020] [Accepted: 02/21/2020] [Indexed: 12/31/2022]
Abstract
Angelman syndrome (AS) is an incurable neurodevelopmental disease caused by loss of function of the maternally inherited UBE3A gene. AS is characterized by a defined set of symptoms, namely severe developmental delay, speech impairment, uncontrolled laughter, and ataxia. Current understanding of the pathophysiology of AS relies mostly on studies using the murine model of the disease, although alternative models based on patient-derived stem cells are now emerging. Here, we summarize the literature of the last decade concerning the three major brain areas that have been the subject of study in the context of AS: hippocampus, cortex, and the cerebellum. Our comprehensive analysis highlights the major phenotypes ascribed to the different brain areas. Moreover, we also discuss the major drawbacks of current models and point out future directions for research in the context of AS, which will hopefully lead us to an effective treatment of this condition in humans.
Collapse
Affiliation(s)
- Carina Maranga
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tiago G Fernandes
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Evguenia Bekman
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,The Discoveries Centre for Regenerative and Precision Medicine, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Simão Teixeira da Rocha
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
47
|
Dodge A, Peters MM, Greene HE, Dietrick C, Botelho R, Chung D, Willman J, Nenninger AW, Ciarlone S, Kamath SG, Houdek P, Sumová A, Anderson AE, Dindot SV, Berg EL, O'Geen H, Segal DJ, Silverman JL, Weeber EJ, Nash KR. Generation of a Novel Rat Model of Angelman Syndrome with a Complete Ube3a Gene Deletion. Autism Res 2020; 13:397-409. [PMID: 31961493 PMCID: PMC7787396 DOI: 10.1002/aur.2267] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 01/02/2020] [Accepted: 01/06/2020] [Indexed: 11/08/2022]
Abstract
Angelman syndrome (AS) is a rare genetic disorder characterized by severe intellectual disability, seizures, lack of speech, and ataxia. The gene responsible for AS was identified as Ube3a and it encodes for E6AP, an E3 ubiquitin ligase. Currently, there is very little known about E6AP's mechanism of action in vivo or how the lack of this protein in neurons may contribute to the AS phenotype. Elucidating the mechanistic action of E6AP would enhance our understanding of AS and drive current research into new avenues that could lead to novel therapeutic approaches that target E6AP's various functions. To facilitate the study of AS, we have generated a novel rat model in which we deleted the rat Ube3a gene using CRISPR. The AS rat phenotypically mirrors human AS with loss of Ube3a expression in the brain and deficits in motor coordination as well as learning and memory. This model offers a new avenue for the study of AS. Autism Res 2020, 13: 397-409. © 2020 International Society for Autism Research,Wiley Periodicals, Inc. LAY SUMMARY: Angelman syndrome (AS) is a rare genetic disorder characterized by severe intellectual disability, seizures, difficulty speaking, and ataxia. The gene responsible for AS was identified as UBE3A, yet very little is known about its function in vivo or how the lack of this protein in neurons may contribute to the AS phenotype. To facilitate the study of AS, we have generated a novel rat model in which we deleted the rat Ube3a gene using CRISPR. The AS rat mirrors human AS with loss of Ube3a expression in the brain and deficits in motor coordination as well as learning and memory. This model offers a new avenue for the study of AS.
Collapse
Affiliation(s)
- Andie Dodge
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| | - Melinda M Peters
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| | - Hayden E Greene
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| | - Clifton Dietrick
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| | - Robert Botelho
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| | - Diana Chung
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| | - Jonathan Willman
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| | - Austin W Nenninger
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| | - Stephanie Ciarlone
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
- PTC Therapeutics Inc., Plainfield, 07080, New Jersey
| | - Siddharth G Kamath
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| | - Pavel Houdek
- Department of Neurohumoral Regulations, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Alena Sumová
- Department of Neurohumoral Regulations, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Anne E Anderson
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Scott V Dindot
- Department of Veterinary Pathobiology, Texas A&M, College Station, Texas
| | - Elizabeth L Berg
- School of Medicine, MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California - Davis, Sacramento, California
| | - Henriette O'Geen
- Genome Center and MIND Institute, University of California - Davis, Davis, California
| | - David J Segal
- Genome Center and MIND Institute, University of California - Davis, Davis, California
| | - Jill L Silverman
- School of Medicine, MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California - Davis, Sacramento, California
| | - Edwin J Weeber
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
- PTC Therapeutics Inc., Plainfield, 07080, New Jersey
| | - Kevin R Nash
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| |
Collapse
|
48
|
Rotaru DC, Mientjes EJ, Elgersma Y. Angelman Syndrome: From Mouse Models to Therapy. Neuroscience 2020; 445:172-189. [PMID: 32088294 DOI: 10.1016/j.neuroscience.2020.02.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/19/2022]
Abstract
The UBE3A gene is part of the chromosome 15q11-q13 region that is frequently deleted or duplicated, leading to several neurodevelopmental disorders (NDD). Angelman syndrome (AS) is caused by the absence of functional maternally derived UBE3A protein, while the paternal UBE3A gene is present but silenced specifically in neurons. Patients with AS present with severe neurodevelopmental delay, with pronounced motor deficits, absence of speech, intellectual disability, epilepsy, and sleep problems. The pathophysiology of AS is still unclear and a treatment is lacking. Animal models of AS recapitulate the genotypic and phenotypic features observed in AS patients, and have been invaluable for understanding the disease process as well as identifying apropriate drug targets. Using these AS mouse models we have learned that loss of UBE3A probably affects many areas of the brain, leading to increased neuronal excitability and a loss of synaptic spines, along with changes in a number of distinct behaviours. Inducible AS mouse models have helped to identify the critical treatment windows for the behavioral and physiological phenotypes. Additionally, AS mouse models indicate an important role for the predominantly nuclear UBE3A isoform in generating the characteristic AS pathology. Last, but not least, the AS mice have been crucial in guiding Ube3a gene reactivation treatments, which present a very promising therapy to treat AS.
Collapse
Affiliation(s)
- Diana C Rotaru
- Department of Neuroscience, The ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Edwin J Mientjes
- Department of Neuroscience, The ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ype Elgersma
- Department of Neuroscience, The ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
49
|
Berg EL, Pride MC, Petkova SP, Lee RD, Copping NA, Shen Y, Adhikari A, Fenton TA, Pedersen LR, Noakes LS, Nieman BJ, Lerch JP, Harris S, Born HA, Peters MM, Deng P, Cameron DL, Fink KD, Beitnere U, O'Geen H, Anderson AE, Dindot SV, Nash KR, Weeber EJ, Wöhr M, Ellegood J, Segal DJ, Silverman JL. Translational outcomes in a full gene deletion of ubiquitin protein ligase E3A rat model of Angelman syndrome. Transl Psychiatry 2020; 10:39. [PMID: 32066685 PMCID: PMC7026078 DOI: 10.1038/s41398-020-0720-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/17/2019] [Accepted: 01/02/2020] [Indexed: 12/17/2022] Open
Abstract
Angelman syndrome (AS) is a rare neurodevelopmental disorder characterized by developmental delay, impaired communication, motor deficits and ataxia, intellectual disabilities, microcephaly, and seizures. The genetic cause of AS is the loss of expression of UBE3A (ubiquitin protein ligase E6-AP) in the brain, typically due to a deletion of the maternal 15q11-q13 region. Previous studies have been performed using a mouse model with a deletion of a single exon of Ube3a. Since three splice variants of Ube3a exist, this has led to a lack of consistent reports and the theory that perhaps not all mouse studies were assessing the effects of an absence of all functional UBE3A. Herein, we report the generation and functional characterization of a novel model of Angelman syndrome by deleting the entire Ube3a gene in the rat. We validated that this resulted in the first comprehensive gene deletion rodent model. Ultrasonic vocalizations from newborn Ube3am-/p+ were reduced in the maternal inherited deletion group with no observable change in the Ube3am+/p- paternal transmission cohort. We also discovered Ube3am-/p+ exhibited delayed reflex development, motor deficits in rearing and fine motor skills, aberrant social communication, and impaired touchscreen learning and memory in young adults. These behavioral deficits were large in effect size and easily apparent in the larger rodent species. Low social communication was detected using a playback task that is unique to rats. Structural imaging illustrated decreased brain volume in Ube3am-/p+ and a variety of intriguing neuroanatomical phenotypes while Ube3am+/p- did not exhibit altered neuroanatomy. Our report identifies, for the first time, unique AS relevant functional phenotypes and anatomical markers as preclinical outcomes to test various strategies for gene and molecular therapies in AS.
Collapse
Affiliation(s)
- E L Berg
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - M C Pride
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - S P Petkova
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - R D Lee
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - N A Copping
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Y Shen
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - A Adhikari
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - T A Fenton
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - L R Pedersen
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - L S Noakes
- Mouse Imaging Centre, Toronto Centre for Phenogenomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - B J Nieman
- Mouse Imaging Centre, Toronto Centre for Phenogenomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - J P Lerch
- Wellcome Centre for Integrative Neuroimaging, The University of Oxford, Oxford, UK
| | - S Harris
- Department of Pediatrics and Neurology, Baylor College of Medicine, Houston, TX, USA
| | - H A Born
- Department of Pediatrics and Neurology, Baylor College of Medicine, Houston, TX, USA
| | - M M Peters
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - P Deng
- Stem Cell Program, Institute for Regenerative Cures, and Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - D L Cameron
- Stem Cell Program, Institute for Regenerative Cures, and Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - K D Fink
- Stem Cell Program, Institute for Regenerative Cures, and Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - U Beitnere
- MIND Institute, Genome Center, and Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, CA, USA
| | - H O'Geen
- MIND Institute, Genome Center, and Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, CA, USA
| | - A E Anderson
- Department of Pediatrics and Neurology, Baylor College of Medicine, Houston, TX, USA
| | - S V Dindot
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - K R Nash
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - E J Weeber
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - M Wöhr
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University of Marburg, Marburg, Germany
| | - J Ellegood
- Mouse Imaging Centre, Toronto Centre for Phenogenomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - D J Segal
- MIND Institute, Genome Center, and Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, CA, USA
| | - J L Silverman
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
50
|
Yang X. Towards an understanding of Angelman syndrome in mice studies. J Neurosci Res 2019; 98:1162-1173. [PMID: 31867793 DOI: 10.1002/jnr.24576] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/13/2022]
Abstract
Angelman syndrome (AS) is a rare neurodevelopmental disorder characterized by severe mental retardation, absence of speech, abnormal motor coordination, abnormal EEG, and spontaneous seizure. AS is caused by a deficiency in the ubiquitin ligase E3A (Ube3a) gene product, known to play a dual role as both ubiquitin ligase and transcription coactivator. In AS animal models, multiple Ube3a substrates are accumulated in neurons. So far, studies in mouse models have either aimed at re-expressing Ube3a or manipulating downstream signaling pathways. Reintroducing Ube3a in AS mice showed promising results but may have two caveats. First, it may cause an overdosage in the Ube3a expression, which in turn is known to contribute to autism spectrum disorders. Second, in mutation cases, the exogenous Ube3a may have to compete with the mutated endogenous form. Such two caveats left spaces for developing therapies or interventions directed to targets downstream Ube3a. Notably, Ube3a expression is dynamically regulated by neuronal activity and plays a crucial role in synaptic plasticity. The abnormal synaptic plasticity uncovered in AS mice has been frequently rescued, but circuits symptoms like seizure are resistant to treatment. Future investigations are needed to further clarify the function (s) of Ube3a during development. Here I reviewed the recently identified major Ube3a substrates and signaling pathways involved in AS pathology, the Ube3a expression, imprinting and evolution, the AS mouse models that have been generated and inspired therapeutic potentials, and finally proposed some future explorations to better understand the AS pathology.
Collapse
Affiliation(s)
- Xin Yang
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|