1
|
Powers S, Anderson KN, Tan WH, Gwaltney A, Potter SN, Tillmann J, Daniel M, Thum A, Farmer C, Clinch S, Squassante L, Tjeertes J, Vincenzi B, Buzasi K, Wheeler AC, Sadhwani A. Developing Meaningful Score Differences for the Bayley-4 and Vineland-3 in Angelman Syndrome using a Delphi Panel. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.05.25325305. [PMID: 40297434 PMCID: PMC12036406 DOI: 10.1101/2025.04.05.25325305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Objectives To develop within-patient meaningful score differences (MSDs) on the Bayley Scales of Infant Development, Fourth Edition (Bayley-4), and the Vineland Adaptive Behavior Scales, Third Edition (Vineland-3), for individuals with Angelman syndrome (AS). Methods A Delphi method, involving a panel of 19 caregivers of individuals with AS, was used to establish MSDs for Bayley-4 and Vineland-3 Growth Scale Values. MSD was defined as the smallest change that would noticeably impact the daily functioning of an individual with AS or family quality of life in a way that was important to the individual with AS or their family. For each subscale of the Bayley-4 and Vineland-3, the panel was presented with 2 to 4 vignettes describing varying levels of baseline functioning and asked to select a MSD from a range of potential values. An iterative process involving three rounds of ratings and two rounds of discussion was used to build consensus. The median caregiver rating from round 3 was retained as the final recommended MSD value for each vignette. Results Final MSD ratings for the five subscales of Bayley-4 and 10 subscales of the Vineland-3 had an agreement rate of 70% or higher. MSD thresholds for each subscale were not single cut-offs, but rather reflected a range of MSD values dependent on level of baseline functioning. Conclusions The Delphi Panel method incorporates the caregiver perspective to provide preliminary estimates of what constitutes meaningful within person change on the Bayley-4 and Vineland-3 in individuals with AS with various levels of baseline functioning. Highlights ⍰ To acquire regulatory approval in drug development, sponsors must demonstrate both statistical significance and clinical meaningfulness of a treatment effect.While several clinical trials are underway in AS, within person meaningful score difference thresholds are not yet established for the most commonly used outcome measures, namely the Bayley and Vineland.⍰ Aligning with FDA guidance, we have developed an innovative qualitative approach using a Delphi panel to incorporate caregiver perspectives in defining meaningful change and generated preliminary patient-informed meaningful score differences (MSDs) for individuals with Angelman Syndrome.⍰ What caregivers of individuals with AS consider to be a MSD on the measures depends primarily on the baseline severity of their child's presentation.
Collapse
|
2
|
O'Donohoe DS, Whelan S, Mannion A, Tones M, Heussler H, Bellgard M, Leader G. Association between sleep disturbances and challenging behavior in children and adolescents with Angelman syndrome. Sleep Med 2024; 123:1-6. [PMID: 39222563 DOI: 10.1016/j.sleep.2024.07.033] [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: 06/29/2023] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
Angelman Syndrome (AS) is a neurodevelopmental disorder with severe symptoms and associated comorbidities. It is caused by the inactivity or lack of the UBE3a gene. Symptoms of the syndrome include intellectual disability and developmental delay. The current study investigated sleep disturbances (SD) in children and adolescents with AS, associations between SD and possible predictors of SD. Variables examined included age, gender, newborn and infancy history, challenging behavior, type of therapy received, genetic type of AS, and seizures. The sample included data from 109 participants with a mean age of 8.21, accessed via the Global Angelman Syndrome Registry. Chi-square tests were carried out to assess the associations between the variables and a logistical regression was carried out to assess the possible predictors of SD. Associations were found between SD and certain repetitive behaviors: slapping walls, focal hand movements, and agitation at new situations. From these associations, a regression formed a predictive model for sleep disturbances. The findings of this research demonstrated the importance of investigating the relationship between sleep disturbances and challenging behavior in children and adolescents with AS and the need for further research in this area.
Collapse
Affiliation(s)
- Darragh S O'Donohoe
- Irish Centre for Autism and Neurodevelopmental Research, School of Psychology, University of Galway, Galway, Ireland
| | - Sally Whelan
- Irish Centre for Autism and Neurodevelopmental Research, School of Psychology, University of Galway, Galway, Ireland
| | - Arlene Mannion
- Irish Centre for Autism and Neurodevelopmental Research, School of Psychology, University of Galway, Galway, Ireland
| | - Megan Tones
- EResearch, Queensland University of Technology, Brisbane, Australia
| | - Helen Heussler
- Children's Health and Hospital Services, QLD, Brisbane, Australia
| | - Matthew Bellgard
- EResearch, Queensland University of Technology, Brisbane, Australia; University of East London, United Kingdom
| | - Geraldine Leader
- Irish Centre for Autism and Neurodevelopmental Research, School of Psychology, University of Galway, Galway, Ireland.
| |
Collapse
|
3
|
Pizzella A, Penna E, Liu Y, Abate N, Lacivita E, Leopoldo M, Perrone-Capano C, Crispino M, Baudry M, Bi X. Alterations of synaptic plasticity in Angelman syndrome model mice are rescued by 5-HT7R stimulation. Prog Neurobiol 2024; 242:102684. [PMID: 39481590 DOI: 10.1016/j.pneurobio.2024.102684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/10/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
Angelman syndrome (AS) is a severe neurodevelopmental disorder characterized by motor disfunction, seizures, intellectual disability, speech deficits, and autism-like behavior, showing high comorbidity with Autism Spectrum Disorders (ASD). It is known that stimulation of the serotonin receptor 7 (5-HT7R) can rescue some of the behavioral and neuroplasticity dysfunctions in animal models of Fragile X and Rett syndrome, two pathologies associated with ASD. In view of these observations, we hypothesised that alterations of 5-HT7R signalling might also be involved in AS. To test this hypothesis, we stimulated 5-HT7R with the selective agonist LP-211 to investigate its possible beneficial effects on synaptic dysfunctions and altered behavior in the AS mice model. In mutant mice, we observed impairment of the synaptic machinery of protein synthesis, which was reversed by 5-HT7R activation. Moreover, stimulation of 5-HT7R was able to: i) enhance dendritic spine density in hippocampal neurons, which was reduced in AS mice; ii) restore impaired long-term potentiation (LTP) in hippocampal slices of the AS mice; iii) improve cognitive performance of the mutant animals subjected to the fear conditioning paradigm. Altogether, our results, showing beneficial effects of 5-HT7R stimulation in restoring molecular and cognitive deficits associated with AS, suggest that targeting 5-HT7R could be a promising therapeutic approach for the pathology.
Collapse
Affiliation(s)
- Amelia Pizzella
- Department of Biology, University of Naples Federico II, Naples, Italy; College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, USA.
| | - Eduardo Penna
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, USA.
| | - Yan Liu
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, USA.
| | - Natalia Abate
- Department of Biology, University of Naples Federico II, Naples, Italy.
| | - Enza Lacivita
- Department of Pharmacy - Drug Sciences, University of Bari Aldo Moro, Bari, Italy.
| | - Marcello Leopoldo
- Department of Pharmacy - Drug Sciences, University of Bari Aldo Moro, Bari, Italy.
| | | | - Marianna Crispino
- Department of Biology, University of Naples Federico II, Naples, Italy.
| | - Michel Baudry
- College of Dental Medicine, Western University of Health Sciences, Pomona, USA.
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, USA.
| |
Collapse
|
4
|
Belghiti HD, Abbassi M, Sayel H, Ahakoud M, El Makhzen BE, Lee N, Russo S, Chaouki S, Bouguenouch L. Impact of Deletion on Angelman Syndrome Phenotype Variability: Phenotype-Genotype Correlation in 97 Patients with Motor Developmental Delay. J Pediatr Genet 2024; 13:15-21. [PMID: 38567176 PMCID: PMC10984711 DOI: 10.1055/s-0042-1751268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 05/23/2022] [Indexed: 10/15/2022]
Abstract
Angelman syndrome (AS) is a rare neurodevelopmental disorder due to genetic defects involving chromosome 15, known by intellectual disability, cognitive and behavioral disorders, ataxia, delayed motor development, and seizures. This study highlights the clinical spectrum and molecular research to establish the genotype-phenotype correlation in the pediatric Moroccan population. Methylation-specific-polymerase chain reaction (MS-PCR) is a primordial technique not only to identify the genetic mechanism of AS but also to characterize the different molecular classes induced in the appearance of the clinical symptoms. Patients with positive methylation profile were additionally studied by fluorescent in situ hybridization. Sequencing analysis of the UBE3A gene was performed for patients with negative MS-PCR. We used Fisher's test to assess differences in the distribution of features frequencies among the deletional and the nondeletional group. Statistical analysis was performed using R project. We identified from 97 patients diagnosed with AS, 14 (2.06%) had a classical AS phenotype, while 70 (84.5%) patients displayed a subset of consistent and frequent criteria. Development delay was shown severe in 63% and moderate in 37%. Nineteen out of 97 of them had MS-PCR positive in which 17 (89.47%) had 15q11-q13 deletion. Deletion patients presented a higher incidence of epileptic seizures ( p = 0.04), ataxia ( p = 0.0008), and abnormal electroencephalogram (EEG) profile ( p = 0.003). We further found out a frameshift deletion located at exon 9 of the UBE3A gene discovered in a 5 years old patient. We report in this study the genotype-phenotype correlation using different molecular testing. Correlation analysis did not reveal any statistical differences in phenotypic dissimilarity between deletion and nondeletion groups for most clinical features, except the correlation was highly significant in the abnormal EEG. According to our findings, we recommend offering MS-PCR analysis to all patients with severe intellectual disability, developmental delay, speech impairment, happy demeanor, and hypopigmentation.
Collapse
Affiliation(s)
- Hanae Daha Belghiti
- Medical Center of Biomedical and Translational Research, Hassan II University Hospital, Fez, Morocco
| | - Meriame Abbassi
- Unit of Medical Genetics and Oncogenetics, University Hospital Hassan II, Fez, Morocco
| | - Hanane Sayel
- Unit of Medical Genetics and Oncogenetics, University Hospital Hassan II, Fez, Morocco
| | - Mohamed Ahakoud
- Unit of Medical Genetics and Oncogenetics, University Hospital Hassan II, Fez, Morocco
| | | | - Norman Lee
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, United States
| | - Silvia Russo
- Istituto Auxologico Italiano Istituto di Ricovero e Cura a Carattere Scientifico, Cytogenetics and Molecular Genetics Laboratory, Milano, Lombardia, Italy
| | - Sana Chaouki
- Department of Neuropediatrics, Hospital University Hassan II, Fez, Morocco
| | - Laila Bouguenouch
- Unit of Medical Genetics and Oncogenetics, University Hospital Hassan II, Fez, Morocco
| |
Collapse
|
5
|
Ali A, Khatoon A, Shao C, Murtaza B, Tanveer Q, Su Z. Therapeutic potential of natural antisense transcripts and various mechanisms involved for clinical applications and disease prevention. RNA Biol 2024; 21:1-18. [PMID: 38090817 PMCID: PMC10761088 DOI: 10.1080/15476286.2023.2293335] [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] [Revised: 11/26/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023] Open
Abstract
Antisense transcription, a prevalent occurrence in mammalian genomes, gives rise to natural antisense transcripts (NATs) as RNA molecules. These NATs serve as agents of diverse transcriptional and post-transcriptional regulatory mechanisms, playing crucial roles in various biological processes vital for cell function and immune response. However, when their normal functions are disrupted, they can contribute to human diseases. This comprehensive review aims to establish the molecular foundation linking NATs to the development of disorders like cancer, neurodegenerative conditions, and cardiovascular ailments. Additionally, we evaluate the potential of oligonucleotide-based therapies targeting NATs, presenting both their advantages and limitations, while also highlighting the latest advancements in this promising realm of clinical investigation.Abbreviations: NATs- Natural antisense transcripts, PRC1- Polycomb Repressive Complex 1, PRC2- Polycomb Repressive Complex 2, ADARs- Adenosine deaminases acting on RNA, BDNF-AS- Brain-derived neurotrophic factor antisense transcript, ASOs- Antisense oligonucleotides, SINEUPs- Inverted SINEB2 sequence-mediated upregulating molecules, PTBP1- Polypyrimidine tract binding protein-1, HNRNPK- heterogeneous nuclear ribonucleoprotein K, MAPT-AS1- microtubule-associated protein tau antisense 1, KCNQ1OT- (KCNQ1 opposite strand/antisense transcript 1, ERK- extracellular signal-regulated kinase 1, USP14- ubiquitin-specific protease 14, EGF- Epidermal growth factor, LSD1- Lysine Specific Demethylase 1, ANRIL- Antisense Noncoding RNA in the INK4 Locus, BWS- Beckwith-Wiedemann syndrome, VEGFA- Vascular Endothelial Growth component A.
Collapse
Affiliation(s)
- Ashiq Ali
- Department of Histology and Embryology, Shantou University Medical College, Shantou, China
| | - Aisha Khatoon
- Department of Pathology, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Chenran Shao
- Department of Histology and Embryology, Shantou University Medical College, Shantou, China
| | - Bilal Murtaza
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Qaisar Tanveer
- The Roslin Institute, The Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, UK
| | - Zhongjing Su
- Department of Histology and Embryology, Shantou University Medical College, Shantou, China
| |
Collapse
|
6
|
Keary C, Bird LM, de Wit MC, Hatti S, Heimer G, Heussler H, Kolevzon A, Mathews A, Ochoa-Lubinoff C, Tan WH, Yan Y, Adams M. Gaboxadol in angelman syndrome: A double-blind, parallel-group, randomized placebo-controlled phase 3 study. Eur J Paediatr Neurol 2023; 47:6-12. [PMID: 37639777 DOI: 10.1016/j.ejpn.2023.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/21/2023] [Accepted: 07/31/2023] [Indexed: 08/31/2023]
Abstract
PURPOSE To evaluate efficacy and safety of gaboxadol for treatment of children with Angelman syndrome (AS). METHOD In this international, double-blind, phase 3 trial, we randomized children 4-12 years old with a molecular diagnosis of AS and a Clinical Global Impression (CGI)-severity score ≥3 to either daily administration of weight-based gaboxadol or matching placebo for 12 weeks. The primary endpoint was the CGI-Improvement-AS (CGI-I-AS) score at week 12. Secondary endpoints included the proportion of participants with CGI-I-AS response of ≤3 (i.e., at least "minimal improvement") and ≤2 (i.e., at least "much improvement") at week 12. Safety and tolerability were monitored throughout the study. Weight based dosing of study drug ranged from 0.125 mg/kg to 0.24 mg/kg depending on weight range. RESULTS Between August 2019 and November 2020, 104 participants were enrolled: participants 4-12 years old were randomly (1:1) assigned to gaboxadol (n = 47) or placebo (n = 50), and 7 other participants 2─3 years old who received gaboxadol and were assessed for safety only. All gaboxadol-treated participants and 48 of 50 placebo-treated participants completed treatment. There was no significant difference in CGI-I-AS between groups: at week 12, mean CGI-I-AS score was 3.3 (SD, 1.00) and 3.2 (SD, 1.05) in the gaboxadol and placebo groups, respectively, yielding a least squares mean difference of zero (p = 0.83). There were no between-group significant differences with respect to CGI-I-AS responses. Gaboxadol was well tolerated in all age groups of this study. CONCLUSIONS There was no significant difference in CGI-I-AS between gaboxadol and placebo after 12 weeks of study treatment in pediatric AS participants. CLINICALTRIALS GOV: NCT04106557.
Collapse
Affiliation(s)
| | - Lynne M Bird
- University of California, San Diego and Rady Children's Hospital, San Diego, California, USA
| | - Marie-Claire de Wit
- Department of Pediatric Neurology, ENCORE Expertise Center, ERN-Ithaca Erasmus MC Sophia Children's Hospital, Rotterdam, the Netherlands
| | | | - Gali Heimer
- Sheba Medical Center, The Tel Aviv University, Ramat Gan, Israel
| | - Helen Heussler
- Centre for Children's Health Research, University of Queensland, Brisbane, Australia; Centre for Clinical Trials in Rare Neurodevelopmental Disorders, Queensland Children's Hospital, Brisbane, Australia
| | | | | | | | - Wen-Hann Tan
- Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ying Yan
- Ovid Therapeutics, Cambridge, MA, USA
| | | |
Collapse
|
7
|
Camões dos Santos J, Appleton C, Cazaux Mateus F, Covas R, Bekman EP, da Rocha ST. Stem cell models of Angelman syndrome. Front Cell Dev Biol 2023; 11:1274040. [PMID: 37928900 PMCID: PMC10620611 DOI: 10.3389/fcell.2023.1274040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/02/2023] [Indexed: 11/07/2023] Open
Abstract
Angelman syndrome (AS) is an imprinted neurodevelopmental disorder that lacks a cure, characterized by developmental delay, intellectual impairment, seizures, ataxia, and paroxysmal laughter. The condition arises due to the loss of the maternally inherited copy of the UBE3A gene in neurons. The paternally inherited UBE3A allele is unable to compensate because it is silenced by the expression of an antisense transcript (UBE3A-ATS) on the paternal chromosome. UBE3A, encoding enigmatic E3 ubiquitin ligase variants, regulates target proteins by either modifying their properties/functions or leading them to degradation through the proteasome. Over time, animal models, particularly the Ube3a mat-/pat+ Knock-Out (KO) mice, have significantly contributed to our understanding of the molecular mechanisms underlying AS. However, a shift toward human pluripotent stem cell models (PSCs), such as human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), has gained momentum. These stem cell models accurately capture human genetic and cellular characteristics, offering an alternative or a complement to animal experimentation. Human stem cells possess the remarkable ability to recapitulate neurogenesis and generate "brain-in-a-dish" models, making them valuable tools for studying neurodevelopmental disorders like AS. In this review, we provide an overview of the current state-of-the-art human stem cell models of AS and explore their potential to become the preclinical models of choice for drug screening and development, thus propelling AS therapeutic advancements and improving the lives of affected individuals.
Collapse
Affiliation(s)
- João Camões dos Santos
- iBB—Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Carolina Appleton
- iBB—Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Department of Animal Biology, Faculdade de Ciências da Universidade de Lisboa, Lisbon, Portugal
| | - Francisca Cazaux Mateus
- iBB—Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Rita Covas
- iBB—Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Evguenia Pavlovna Bekman
- iBB—Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- The Egas Moniz Center for Interdisciplinary Research (CiiEM), Caparica, Portugal
| | - Simão Teixeira da Rocha
- iBB—Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
8
|
Keary CJ, McDougle CJ. Current and emerging treatment options for Angelman syndrome. Expert Rev Neurother 2023; 23:835-844. [PMID: 37599585 DOI: 10.1080/14737175.2023.2245568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 08/03/2023] [Indexed: 08/22/2023]
Abstract
INTRODUCTION Angelman syndrome (AS) is a neurodevelopmental disorder characterized by intellectual disability, limited expressive language, epilepsy, and motor impairment. Angelman syndrome is caused by haploinsufficiency of the UBE3A gene on the maternal copy of chromosome 15. There have been ongoing advances in the understanding of neurological, behavioral, and sleep-based problems and associated treatments for patients with AS. These results along with gene-based therapies entering into clinical development prompted this review. AREAS COVERED The authors summarize the research basis describing phenomenology of epilepsy and behavioral concerns such as hyperactivity behavior, aggression, self-injury, repetitive behavior, and sleep disorder. The evidence for recent treatment advances in these target symptom domains of concern is reviewed, and the potential for emerging gene therapy treatments is considered. EXPERT OPINION The prospect for emerging gene therapies means that increasing efforts should be directed toward the early identification of AS implemented equitably. Recent studies emphasize the important role of behavioral therapy in addressing mental health concerns such as aggression and disordered sleep.
Collapse
Affiliation(s)
- Christopher J Keary
- Department is department of psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Lurie Center for Autism, Lexington, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Angelman Syndrome Program, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Christopher J McDougle
- Department is department of psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Lurie Center for Autism, Lexington, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Aria F, Pandey K, Alberini CM. Excessive Protein Accumulation and Impaired Autophagy in the Hippocampus of Angelman Syndrome Modeled in Mice. Biol Psychiatry 2023; 94:68-83. [PMID: 36764852 PMCID: PMC10276539 DOI: 10.1016/j.biopsych.2022.11.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 11/03/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Angelman syndrome (AS), a neurodevelopmental disorder caused by abnormalities of the 15q11.2-q13.1 chromosome region, is characterized by impairment of cognitive and motor functions, sleep problems, and seizures. How the genetic defects of AS produce these neurological symptoms is unclear. Mice modeling AS (AS mice) accumulate activity-regulated cytoskeleton-associated protein (ARC/ARG3.1), a neuronal immediate early gene (IEG) critical for synaptic plasticity. This accumulation suggests an altered protein metabolism. METHODS Focusing on the dorsal hippocampus (dHC), a brain region critical for memory formation and cognitive functions, we assessed levels and tissue distribution of IEGs, de novo protein synthesis, and markers of protein synthesis, endosomes, autophagy, and synaptic functions in AS mice at baseline and following learning. We also tested autophagic flux and memory retention following autophagy-promoting treatment. RESULTS AS dHC exhibited accumulation of IEGs ARC, FOS, and EGR1; autophagy proteins MLP3B, SQSTM1, and LAMP1; and reduction of the endosomal protein RAB5A. AS dHC also had increased levels of de novo protein synthesis, impaired autophagic flux with accumulation of autophagosome, and altered synaptic protein levels. Contextual fear conditioning significantly increased levels of IEGs and autophagy proteins, de novo protein synthesis, and autophagic flux in the dHC of normal mice, but not in AS mice. Enhancing autophagy in the dHC alleviated AS-related memory and autophagic flux impairments. CONCLUSIONS A major biological deficit of AS brain is a defective protein metabolism, particularly that dynamically regulated by learning, resulting in stalled autophagy and accumulation of neuronal proteins. Activating autophagy ameliorates AS cognitive impairments and dHC protein accumulation.
Collapse
Affiliation(s)
- Francesca Aria
- Center for Neural Science, New York University, New York, New York
| | - Kiran Pandey
- Center for Neural Science, New York University, New York, New York
| | | |
Collapse
|
10
|
Khan N, Cabo R, Burdine RD, Tan WH, Keary CJ, Ochoa-Lubinoff C, Bird LM. Health-related quality of life and medication use among individuals with Angelman syndrome. Qual Life Res 2023:10.1007/s11136-023-03375-4. [PMID: 37039911 DOI: 10.1007/s11136-023-03375-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2022] [Indexed: 04/12/2023]
Abstract
PURPOSE The primary goal of this analysis is to describe the health-related quality of life (HRQoL), medical history, and medication use among adolescents and adults individuals with Angelman syndrome (AS). METHODS The analysis uses baseline data collected during the STARS study, a double-blind placebo controlled trial of gaboxadol (OV101) in adolescents and adults with AS. The HRQoL was estimated using EuroQoL 5-Dimension 5-Level (EQ-5D) health questionnaire proxy 1 version, which was completed by the caregivers. EQ-5D consists of two parts, a 5-dimension descriptive and a visual analogue scale (VAS) component. The utility score derived from EQ-5D ranges from 0 to 1 (perfect health) and VAS ranges from 0 to 100 (perfect health). RESULTS 87 individuals with AS were included in the present analysis. The mean utility score was 0.44 ± 0.20 and VAS score was 84 ± 1.5. The EQ-5D data indicated that the self-care, mobility and daily activities were most impacted. All adolescents (100%) and most adults (93%) had at least moderate problems with self-care activities, such as washing or dressing themselves. More than half (55%) of the adolescents and adults had at least moderate issues with mobility and usual activities. Approximately, 30% of adolescents and adults had moderate to extreme problems with anxiety/depression. High baseline concomitant use of medications was observed across both age groups with an average of 5 medications being used per person. CONCLUSION This study highlights the impact of AS on HRQoL and medication utilization among adolescents and adults individuals with AS.
Collapse
Affiliation(s)
- Nasreen Khan
- RWEC, LLC, 73 Walsingham Road, Mendham, NJ, USA.
| | - Raquel Cabo
- Ovid Therapeutics Inc, 441 Ninth Avenue, New York, NY, 10001, USA
| | - Rebecca D Burdine
- Department of Molecular Biology, Princeton University, 159 Moffett Laboratory, Washington Road, Princeton, NJ, 08544-1014, USA
| | - Wen-Hann Tan
- Division of Genetics and Genomics: Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Christopher J Keary
- Angelman Syndrome Program, Lurie Center for Autism, Massachusetts General Hospital for Children, Harvard Medical School, One Maguire Road, Lexington, MA, 02421, USA
| | - Cesar Ochoa-Lubinoff
- Division of Developmental-Behavioral Pediatrics, Rush University Medical Center, 1725 West Harrison Street, Suite 710, Chicago, IL, 60612, USA
| | - Lynne M Bird
- University of California, San Diego, Department of Pediatrics and Rady Children's Hospital San Diego, Division of Dysmorphology/Genetics, 3020 Children's Way #5031, San Diego, CA, 92123, USA
| |
Collapse
|
11
|
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: 16] [Impact Index Per Article: 5.3] [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
|
12
|
Li H, Sheng Z, Khan S, Zhang R, Liu Y, Zhang Y, Yong VW, Xue M. Matrix Metalloproteinase-9 as an Important Contributor to the Pathophysiology of Depression. Front Neurol 2022; 13:861843. [PMID: 35370878 PMCID: PMC8971905 DOI: 10.3389/fneur.2022.861843] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are physiologically expressed in the central nervous system in neurons, astrocytes and microglia, and their aberrant elevation contributes to a number of diseases. Amongst the MMP members, MMP−9 has generated considerable attention because of its possible involvement in inflammatory responses, blood-brain barrier permeability, the regulation of perineuronal nets, demyelination, and synaptic long-term potentiation. Emerging evidence indicate an association between MMP−9 and the syndrome of depression. This review provides an updated and comprehensive summary of the probable roles of MMP−9 in depression with an emphasis on the mechanisms and potential of MMP−9 as a biomarker of depression.
Collapse
Affiliation(s)
- Hongmin Li
- The Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Zhaofu Sheng
- The Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Suliman Khan
- The Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Ruiyi Zhang
- The Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Yang Liu
- The Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Yan Zhang
- The Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - V. Wee Yong
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- *Correspondence: V. Wee Yong
| | - Mengzhou Xue
- The Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
- Mengzhou Xue
| |
Collapse
|
13
|
Godler DE, Ling L, Gamage D, Baker EK, Bui M, Field MJ, Rogers C, Butler MG, Murgia A, Leonardi E, Polli R, Schwartz CE, Skinner CD, Alliende AM, Santa Maria L, Pitt J, Greaves R, Francis D, Oertel R, Wang M, Simons C, Amor DJ. Feasibility of Screening for Chromosome 15 Imprinting Disorders in 16 579 Newborns by Using a Novel Genomic Workflow. JAMA Netw Open 2022; 5:e2141911. [PMID: 34982160 PMCID: PMC8728620 DOI: 10.1001/jamanetworkopen.2021.41911] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
IMPORTANCE Newborn screening for Angelman syndrome (AS), Prader-Willi syndrome (PWS), and chromosome 15 duplication syndrome (Dup15q) may lead to benefit from early diagnosis and treatment. OBJECTIVE To examine the feasibility of newborn screening for these chromosome 15 imprinting disorders at population scale. DESIGN, SETTING, AND PARTICIPANTS In this diagnostic study, the validation data set for the first-tier SNRPN test, called methylation-specific quantitative melt analysis (MS-QMA), included 109 PWS, 48 AS, 9 Dup15q, and 1190 population control newborn blood spots (NBS) and peripheral tissue samples from participants recruited from January 2000 to December 2016. The test data set included NBS samples from 16 579 infants born in 2011. Infants with an NBS identified as positive for PWS, AS, or Dup15q by the first-tier test were referred for droplet digital polymerase chain reaction, real-time polymerase chain reaction, and low-coverage whole-genome sequencing for confirmatory testing. Data analyses were conducted between February 12, 2015, and August 15, 2020. RESULTS In the validation data set, the median age for the 77 patients with PWS was 3.00 years (IQR, 0.01-44.50 years); for the 46 patients with AS, 2.76 years (IQR, 0.028 to 49.00 years); and for the 9 patients with Dup15q, 4.00 years (IQR, 1.00 to 28.00 years). Thirty-eight patients (51.4%) in the PWS group, 20 patients (45.5%) in the AS group, and 6 patients (66.7%) in the Dup15q group who had sex reported were male. The validation data set showed MS-QMA sensitivity of 99.0% for PWS, 93.8% for AS, and 77.8% for Dup15q; specificity of 100% for PWS, AS, and Dup15q; positive predictive and negative predictive values of 100% for PWS and AS; and a positive predictive value of 87.5% and negative predictive value of 100% for Dup15q. In the test data set of NBS samples from 16 579 infants, 92 had a positive test result using a methylation ratio cut-off of 3 standard deviations from the mean. Of these patients, 2 were confirmed to have PWS; 2, AS; and 1, maternal Dup15q. With the use of more conservative PWS- and AS-specific thresholds for positive calls from the validation data set, 9 positive NBS results were identified by MS-QMA in this cohort. The 2 PWS and 2 AS calls were confirmed by second-tier testing, but the 1 Dup15q case was not confirmed. Together, these results provided prevalence estimates of 1 in 8290 for both AS and PWS and 1 in 16 579 for maternal Dup15q, with positive predictive values for first-tier testing at 67.0% for AS, 33.0% for PWS, and 44.0% for combined detection of chromosome 15 imprinting disorders for the validation data set. CONCLUSIONS AND RELEVANCE The findings of this diagnostic study suggest that it is feasible to screen for all chromosome 15 imprinting disorders using SNRPN methylation analysis, with 5 individuals identified with these disorders out of 16 579 infants screened.
Collapse
Affiliation(s)
- David E. Godler
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Parkville, Victoria, Australia
- Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
- E.D.G. Innovations and Consulting, St Kilda, Victoria, Australia
| | - Ling Ling
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Parkville, Victoria, Australia
| | - Dinusha Gamage
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Parkville, Victoria, Australia
| | - Emma K. Baker
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Parkville, Victoria, Australia
- Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Minh Bui
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Carlton, Victoria, Australia
| | - Michael J. Field
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, New South Wales, Australia
| | - Carolyn Rogers
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, New South Wales, Australia
| | - Merlin G. Butler
- Departments of Psychiatry, Behavioral Sciences and Pediatrics, University of Kansas Medical Centre, Kansas City, Kansas
| | - Alessandra Murgia
- Molecular Genetics of Neurodevelopment, Department of Women's and Children's Health, University of Padua, Padua, Italy
- Istituto di Ricerca Pediatrica (IRP), Città della Speranza, Padua, Italy
| | - Emanuela Leonardi
- Molecular Genetics of Neurodevelopment, Department of Women's and Children's Health, University of Padua, Padua, Italy
- Istituto di Ricerca Pediatrica (IRP), Città della Speranza, Padua, Italy
| | - Roberta Polli
- Molecular Genetics of Neurodevelopment, Department of Women's and Children's Health, University of Padua, Padua, Italy
- Istituto di Ricerca Pediatrica (IRP), Città della Speranza, Padua, Italy
| | - Charles E. Schwartz
- Center for Molecular Studies, J.C. Self Research Institute of Human Genetics, Greenwood Genetic Center, Greenwood, South Carolina
| | - Cindy D. Skinner
- Center for Molecular Studies, J.C. Self Research Institute of Human Genetics, Greenwood Genetic Center, Greenwood, South Carolina
| | - Angelica M. Alliende
- Cytogenetics and Molecular Laboratory, Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
- Centre for Diagnosis and Treatment of Fragile X Syndrome (CDTSXF), INTA University of Chile, Santiago, Chile
| | - Lorena Santa Maria
- Cytogenetics and Molecular Laboratory, Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
- Centre for Diagnosis and Treatment of Fragile X Syndrome (CDTSXF), INTA University of Chile, Santiago, Chile
| | - James Pitt
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Ronda Greaves
- Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - David Francis
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Ralph Oertel
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Min Wang
- Translational Bioinformatics, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Cas Simons
- Translational Bioinformatics, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - David J. Amor
- Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
- Neurodisability and Rehabilitation, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
14
|
Grebe SC, Limon DL, McNeel MM, Guzick A, Peters SU, Tan WH, Sadhwani A, Bacino CA, Bird LM, Samaco RC, Berry LN, Goodman WK, Schneider SC, Storch EA. Anxiety in Angelman Syndrome. AMERICAN JOURNAL ON INTELLECTUAL AND DEVELOPMENTAL DISABILITIES 2022; 127:1-10. [PMID: 34979033 PMCID: PMC8803540 DOI: 10.1352/1944-7558-127.1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/20/2021] [Indexed: 06/14/2023]
Abstract
Angelman Syndrome (AS) is a neurodevelopmental disorder most commonly caused by the impaired expression of the maternal UBE3A gene on chromosome 15. Though anxiety has been identified as a frequently present characteristic in AS, there are limited studies examining anxiety in this population. Studies of anxiety in other neurodevelopmental disorders have found disorder specific symptoms of anxiety and age specific displays of anxiety symptoms. However, there is a consistent challenge in identifying anxiety in people with neurodevelopmental disorders given the lack of measurement instruments specifically designed for this population. Given the limited information about AS and anxiety, the aims of the current project were to (a) examine symptoms of anxiety in children with AS and (b) determine the correlates of anxiety in children with AS. Participants included 42 adult caregivers of youth with AS in the AS Natural History study who completed the Developmental Behavior Checklist (DBC). The results found that 26% of the sample demonstrated elevated symptoms of anxiety and established a relationship between elevated anxiety in youth with AS and higher levels of irritability, hyperactivity, self-absorbed behaviors, and disruptive/antisocial behaviors. Findings from this research provide a foundation for tailoring evidence-based assessments and treatments for youth with AS and anxiety.
Collapse
Affiliation(s)
- Stacey C Grebe
- Stacey C. Grebe, Danica L. Limon, Morgan M. McNeel, and Andrew Guzick, Baylor College of Medicine
| | - Danica L Limon
- Stacey C. Grebe, Danica L. Limon, Morgan M. McNeel, and Andrew Guzick, Baylor College of Medicine
| | - Morgan M McNeel
- Stacey C. Grebe, Danica L. Limon, Morgan M. McNeel, and Andrew Guzick, Baylor College of Medicine
| | - Andrew Guzick
- Stacey C. Grebe, Danica L. Limon, Morgan M. McNeel, and Andrew Guzick, Baylor College of Medicine
| | | | - Wen-Hann Tan
- Wen-Hann Tan and Anjali Sadhwani, Boston Children's Hospital
| | - Anjali Sadhwani
- Wen-Hann Tan and Anjali Sadhwani, Boston Children's Hospital
| | - Carlos A Bacino
- Carlos A. Bacino, Baylor College of Medicine and Texas Children's Hospital
| | - Lynne M Bird
- Lynne M. Bird, University of California and Boston Children's Hospital
| | | | - Leandra N Berry
- Leandra N. Berry, Baylor College of Medicine and Texas Children's Hospital
| | | | | | - Eric A Storch
- Sophie C. Schneider and Eric A. Storch, Baylor College of Medicine
| |
Collapse
|
15
|
Li S, Ma Y, Wang T, Jin H, Du X, Wang Y. Epilepsy and Molecular Phenotype Affect the Neurodevelopment of Pediatric Angelman Syndrome Patients in China. Front Psychiatry 2022; 13:886028. [PMID: 35573374 PMCID: PMC9096167 DOI: 10.3389/fpsyt.2022.886028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/08/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE This study investigated the mental development of children with Angelman syndrome (AS) in China and evaluated the relationship between neurodevelopment and molecular subtype, age, epilepsy, and sex using the Chinese version of the Griffith Mental Development Scale (GMDS-C) to provide detailed baseline data regarding neurodevelopment with AS in China. METHODS Participants were recruited from the AS Natural History Study. The GMDS-C was used to evaluate all participants' mental age and developmental quotients. The general quotient (GQ) and quotients of five subscales (sports, personal-social, auditory language, eye-hand coordination, and comprehensive performance) were calculated. RESULTS A total of 119 children (average age: 42.12 months; range, 7.5-95.5 months) with a genetic diagnosis of AS were enrolled. The median GQ score of the GMDS was 29.6 points (95% confidence interval, 28.6-33.25). The children had relatively good locomotor and personal-social skills but poor language skills. Overall, 89% (106/119) had mental ages younger than 24 months for all five subscales. The non-deletion group (i.e., without deletion in chromosome 15q11-13) had higher GQs and locomotor, personal-social, and performance subscale quotients. The GQ was significantly different among the three age subgroups and significantly correlated with age. Compared with the non-epilepsy group, the epilepsy group had lower GQs and lower quotients for the locomotor, personal-social, speech, language, and eye-hand coordination subscales. CONCLUSION Children with AS in China experience severe neurodevelopmental deterioration. In addition to age, molecular subtypes and the onset of seizures may also correlate with these patients' intellectual development. The GMDS-C is an accurate tool that can assess the clinical characteristics of AS. The data of this study can be used as baseline data for clinical trials performed to evaluate drug development or other AS treatment development.
Collapse
Affiliation(s)
- Shuang Li
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, China
| | - Yu Ma
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, China
| | - Tianqi Wang
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, China
| | - Huimin Jin
- Shanghai YangZhi Rehabilitation Hospital, School of Medicine, Tongji University, Shanghia, China
| | - Xiaonan Du
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, China
| | - Yi Wang
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
16
|
Rare Neurological Diseases: an Overreview of Pathophysiology, Epidemiology, Clinical Features and Pharmacoeconomic Considerations in the Treating. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2021. [DOI: 10.2478/sjecr-2021-0049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Rare diseases (RD) are serious chronic diseases affecting small number of people compared to the general population. There are between 6000 and 8000 RDs, which affect about 400 million people worldwide. Drugs used for causal treatment of RDs are called orphan drugs. RDs bear great clinical and economic burden for patients, their families, healthcare systems and society overall. There are at least two reasons for the high cost of treatment of RDs. First, there is no causal therapy for majority of RDs, so exacerbations, complications, and hospitalizations in those patients are common. The second reason is high price of available orphan drugs, which are not cost-effective when traditional pharmacoeconomic evaluation is employed. The pharmacoeconomic aspect of the treatment of RDs is especially important in the field of neurology, since at least one fifth of all RDs is composed of neurological conditions. The aim of this paper was to provide a concise overview of the pathophysiological, epidemiological and clinical characteristics of some of the most important and common rare neurological diseases, with special reference to their impact on society and economy.
Collapse
|
17
|
PACS1-Neurodevelopmental disorder: clinical features and trial readiness. Orphanet J Rare Dis 2021; 16:386. [PMID: 34517877 PMCID: PMC8438988 DOI: 10.1186/s13023-021-02001-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 08/24/2021] [Indexed: 11/10/2022] Open
Abstract
Background PACS1-Neurodevelopmental Disorder (PACS1-NDD) is an ultra-rare condition due to a recurrent mutation in the PACS1 gene. Little systematically collected data exist about the functional abilities and neurodevelopmental morbidities in children with PACS1-NDD Methods Parents of individuals with PACS1-NDD completed an on-line survey designed collaboratively by researchers, parents, and clinicians. Analyses focused on those with a confirmed R203W variant. Results Of 35 individuals with confirmed variants, 18 (51%) were female. The median age was 8 years (interquartile range 4.5–15). Seventeen (49%) had a diagnosis of epilepsy. Twelve (40%, of 30 responding to the question) reported autism and (N = 11/30, 37%) reported features of autism. Most children walked independently (N = 29/32, 91%), had a pincer grasp (N = 23/32, 72%), could feed themselves independently (N = 15/32, 47%), and used speech (N = 23/32, 72%). Sixteen of twenty-nine (55%) had simple pre-academic skills. Neither epilepsy nor autism was associated with functional abilities or other clinical features (all P > 0.05). Conclusions PACS1-NDD is a moderately-severe intellectual disability syndrome in which seizures occur but are not a defining or primary feature. Successful precision medicine clinical trials for this ultra-rare disorder must target important core features of this disorder and utilize assessment tools commensurate with the level of function in this clinical population.
Collapse
|
18
|
Carvill GL, Matheny T, Hesselberth J, Demarest S. Haploinsufficiency, Dominant Negative, and Gain-of-Function Mechanisms in Epilepsy: Matching Therapeutic Approach to the Pathophysiology. Neurotherapeutics 2021; 18:1500-1514. [PMID: 34648141 PMCID: PMC8608973 DOI: 10.1007/s13311-021-01137-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2021] [Indexed: 02/04/2023] Open
Abstract
This review summarizes the pathogenic mechanisms that underpin the monogenic epilepsies and discusses the potential of novel precision therapeutics to treat these disorders. Pathogenic mechanisms of epilepsy include recessive (null alleles), haploinsufficiency, imprinting, gain-of-function, and dominant negative effects. Understanding which pathogenic mechanism(s) that underlie each genetic epilepsy is pivotal to design precision therapies that are most likely to be beneficial for the patient. Novel therapeutics discussed include gene therapy, gene editing, antisense oligonucleotides, and protein replacement. Discussions are illustrated and reinforced with examples from the literature.
Collapse
Affiliation(s)
- Gemma L Carvill
- Departments of Neurology, Pharmacology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tyler Matheny
- Department Biochemistry and Molecular Genetics, School of Medicine, RNA Bioscience Initiative, University of Colorado, PO Box 6511, Aurora, CO, USA
| | - Jay Hesselberth
- Department Biochemistry and Molecular Genetics, School of Medicine, RNA Bioscience Initiative, University of Colorado, PO Box 6511, Aurora, CO, USA
| | - Scott Demarest
- Departments of Pediatrics and Neurology, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA.
| |
Collapse
|
19
|
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: 24] [Impact Index Per Article: 6.0] [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
|
20
|
Elgersma Y, Sonzogni M. UBE3A reinstatement as a disease-modifying therapy for Angelman syndrome. Dev Med Child Neurol 2021; 63:802-807. [PMID: 33543479 PMCID: PMC8248324 DOI: 10.1111/dmcn.14831] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/19/2021] [Indexed: 01/08/2023]
Abstract
Half a century ago, Harry Angelman reported three patients with overlapping clinical features, now well known as Angelman syndrome. Angelman syndrome is caused by mutations affecting the maternally inherited UBE3A gene, which encodes an E3-ubiquitin ligase that is critical for typical postnatal brain development. Emerging evidence indicates that UBE3A plays a particularly important role in the nucleus. However, the critical substrates that are controlled by UBE3A remain elusive, which hinders the search for effective treatments. Moreover, given the multitude of signalling mechanisms that are derailed, it is unlikely that targeting a single pathway is going to be very effective. Therefore, expectations are very high for approaches that aim to restore UBE3A protein levels. A particular promising strategy is an antisense oligonucleotide approach, which activates the silenced paternal UBE3A gene. When successful, such treatments potentially offer a disease-modifying therapy for Angelman syndrome and several other neurodevelopmental disorders. What this paper adds Loss of UBE3A affects multiple signalling pathways in the brain. Emerging evidence suggests that UBE3A plays a critical role in the cell nucleus. Trials using antisense oligonucleotides to restore UBE3A levels are continuing.
Collapse
Affiliation(s)
- Ype Elgersma
- Department of Neuroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
- Deptartment of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
- The ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, the Netherlands
| | - Monica Sonzogni
- Department of Neuroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
- The ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
21
|
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: 30] [Impact Index Per Article: 7.5] [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
|
22
|
Willgoss T, Cassater D, Connor S, Krishnan ML, Miller MT, Dias-Barbosa C, Phillips D, McCormack J, Bird LM, Burdine RD, Claridge S, Bichell TJ. Measuring What Matters to Individuals with Angelman Syndrome and Their Families: Development of a Patient-Centered Disease Concept Model. Child Psychiatry Hum Dev 2021; 52:654-668. [PMID: 32880036 PMCID: PMC8238699 DOI: 10.1007/s10578-020-01051-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Angelman syndrome (AS) is a complex, heterogeneous, and life-long neurodevelopmental disorder. Despite the considerable impact on individuals and caregivers, no disease-modifying treatments are available. To support holistic clinical management and the development of AS-specific outcome measures for clinical studies, we conducted primary and secondary research identifying the impact of symptoms on individuals with AS and their unmet need. This qualitative research adopted a rigorous step-wise approach, aggregating information from published literature, then evaluating it via disease concept elicitation interviews with clinical experts and caregivers. We found that the AS-defining concepts most relevant for treatment included: impaired expressive communication, seizures, maladaptive behavior, cognitive impairment, motor function difficulties, sleep disturbance, and limited self-care abilities. We highlight the relevance of age in experiencing these key AS concepts, and the difference between the perceptions of clinicians and caregivers towards the syndrome. Finally, we outline the impact of AS on individuals, caregivers, and families.
Collapse
Affiliation(s)
- Tom Willgoss
- Roche Products Limited, Hexagon Place, 6 Falcon Way, Shire Park, Welwyn Garden City, AL7 1TW, UK.
| | - Daiana Cassater
- grid.417570.00000 0004 0374 1269Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Siobhan Connor
- grid.419227.bRoche Products Limited, Hexagon Place, 6 Falcon Way, Shire Park, Welwyn Garden City, AL7 1TW UK
| | - Michelle L. Krishnan
- grid.417570.00000 0004 0374 1269Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Meghan T. Miller
- grid.417570.00000 0004 0374 1269Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | - Dawn Phillips
- grid.410711.20000 0001 1034 1720Division of Physical Therapy, School of Medicine, University of North Carolina, Chapel Hill, NC USA
| | - Julie McCormack
- grid.423257.50000 0004 0510 2209Evidera, Patient-Centered Research, Bethesda, MD USA
| | - Lynne M. Bird
- grid.266100.30000 0001 2107 4242Department of Pediatrics, University of California San Diego, San Diego, CA USA
| | - Rebecca D. Burdine
- grid.16750.350000 0001 2097 5006Department of Molecular Biology, Princeton University, Princeton, NJ USA
| | - Sharon Claridge
- Foundation for Angelman Syndrome Research (FAST), Downers Grove, IL USA
| | - Terry Jo Bichell
- Consortium for Outcome Measures and Biomarkers for Neurodevelopmental Disorders, Nashville, TN USA
| |
Collapse
|
23
|
Dong J, Liu J, Wen Y, Tobin SW, Zhang C, Zheng H, Huang Z, Feng Y, Zhang D, Liu S, Zhang Z, Li J. Down-Regulation of Lnc-CYP7A1-1 Rejuvenates Aged Human Mesenchymal Stem Cells to Improve Their Efficacy for Heart Repair Through SYNE1. Front Cell Dev Biol 2020; 8:600304. [PMID: 33330489 PMCID: PMC7710953 DOI: 10.3389/fcell.2020.600304] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 10/21/2020] [Indexed: 01/08/2023] Open
Abstract
Background Several long non-coding RNAs (lncRNAs) have been associated with cell senescence, termed senescence-associated lncRNAs (SAL-RNAs). However, the mechanisms involved for SAL-RNAs in aging are not fully elucidated. In the present study, we investigated the effects of SAL-RNAs on aged human bone marrow-derived mesenchymal stem cells (hBM-MSCs), and the possible means to counteract such effects to improve the regenerative capacity of aged hBM-MSCs. Methods By comparing the lncRNAs expression of hBM-MSCs derived from young and old individuals, lnc-CYP7A1-1 was identified as being significantly increased with age. Using predictive software, the expression of Spectrin Repeat Containing Nuclear Envelope Protein 1 (SYNE1), was found to be decreased with age. Next, through lentiviral constructs, we downregulated the expression of lnc-CYP7A1-1 or SYNE1 in hBM-MSCs separately. Additionally, hBM-MSCs proliferation, survival, migration, and senescence were investigated in vitro. In vivo, lnc-CYP7A1-1 downregulated aged hBM-MSCs were implanted into infarcted mouse hearts after myocardial infarction (MI), and cardiac function was measured. Through lentivirus-mediated downregulation of lnc-CYP7A1-1 in aged hBM-MSCs, we revealed that cell senescence was decreased, whereas cell proliferation, migration, and survival were increased. On the other hand, downregulation of SYNE1, the target gene of lnc-CYP7A1-1, in young hBM-MSCs increased cell senescence, yet decreased cell proliferation, migration, and survival. Downregulation of lnc-CYP7A1-1 in aged hBM-MSCs induced cell rejuvenation, yet this effect was attenuated by repression of SYNE1. In vivo, transplantation of lnc-CYP7A1-1 downregulated old hBM-MSCs improved cardiac function after MI. Conclusion Down-regulation of lnc-CYP7A1-1 rejuvenated aged hBM-MSCs and improved cardiac function when implanted into the infarcted mouse hearts, possibly through its target gene SYNE1.
Collapse
Affiliation(s)
- Jun Dong
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Sunnybrook Research Institute, Toronto, ON, Canada
| | - Jianwei Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yueqiang Wen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Stephanie W Tobin
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Chongyu Zhang
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Huiling Zheng
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zehan Huang
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yongtao Feng
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Department of Cardiovascular Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Dongcheng Zhang
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Department of Cardiovascular Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shiming Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhenhui Zhang
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiao Li
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
24
|
Dutta R, Crawley JN. Behavioral Evaluation of Angelman Syndrome Mice at Older Ages. Neuroscience 2020; 445:163-171. [PMID: 31730795 PMCID: PMC7214203 DOI: 10.1016/j.neuroscience.2019.10.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 12/20/2022]
Abstract
Angelman syndrome is a neurodevelopmental disorder presenting with severe deficits in motor, speech, and cognitive abilities. The primary genetic cause of Angelman syndrome is a maternally transmitted mutation in the Ube3a gene, which has been successfully modeled in Ube3a mutant mice. Phenotypes have been extensively reported in young adult Ube3a mice. Because symptoms continue throughout life in Angelman syndrome, we tested multiple behavioral phenotypes of male Ube3a mice and WT littermate controls at older adult ages. Social behaviors on both the 3-chambered social approach and male-female social interaction tests showed impairments in Ube3a at 12 months of age. Anxiety-related scores on both the elevated plus-maze and the light ↔ dark transitions assays indicated anxiety-like phenotypes in 12 month old Ube3a mice. Open field locomotion parameters were consistently lower at 12 months. Reduced general exploratory locomotion at this age prevented the interpretation of an anxiety-like phenotype, and likely impacted social tasks. Robust phenotypes in middle-aged Ube3a mice appear to result from continued motor decline. Motor deficits may provide the best outcome measures for preclinical testing of pharmacological targets, towards reductions of symptoms in adults with Angelman syndrome.
Collapse
Affiliation(s)
- Rebecca Dutta
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Jacqueline N Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, USA.
| |
Collapse
|
25
|
Schultz MN, Crawley JN. Evaluation of a TrkB agonist on spatial and motor learning in the Ube3a mouse model of Angelman syndrome. Learn Mem 2020; 27:346-354. [PMID: 32817301 PMCID: PMC7433657 DOI: 10.1101/lm.051201.119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/11/2020] [Indexed: 12/21/2022]
Abstract
Angelman syndrome is a rare neurodevelopmental disorder caused by a mutation in the maternal allele of the gene Ube3a The primary symptoms of Angelman syndrome are severe cognitive deficits, impaired motor functions, and speech disabilities. Analogous phenotypes have been detected in young adult Ube3a mice. Here, we investigate cognitive phenotypes of Ube3a mice as compared to wild-type littermate controls at an older adult age. Water maze spatial learning, swim speed, and rotarod motor coordination and balance were impaired at 6 mo of age, as predicted. Based on previous findings of reduced brain-derived neurotrophic factor in Ube3a mice, a novel therapeutic target, the TrkB agonist 7,8-DHF, was interrogated. Semichronic daily treatment with 7,8-DHF, 5 mg/kg i.p., did not significantly improve the impairments in performance during the acquisition of the water maze hidden platform location in Ube3a mice, after training with either massed or spaced trials, and had no effect on the swim speed and rotarod deficits. Robust behavioral phenotypes in middle-aged Ube3a mice appear to result from continued motor decline. Our results suggest that motor deficits could offer useful outcome measures for preclinical testing of many pharmacological targets, with the goal of reducing symptoms in adults with Angelman syndrome.
Collapse
Affiliation(s)
- Maria N Schultz
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California 95821, USA
| | - Jacqueline N Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California 95821, USA
| |
Collapse
|
26
|
Budisteanu M, Jurca C, Papuc SM, Focsa I, Riga D, Riga S, Jurca A, Arghir A. Treatment of Epilepsy Associated with Common Chromosomal Developmental Diseases. Open Life Sci 2020; 15:21-29. [PMID: 33987468 PMCID: PMC8114617 DOI: 10.1515/biol-2020-0003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/22/2019] [Indexed: 11/15/2022] Open
Abstract
Chromosomal diseases are heterogeneous conditions with complex phenotypes, which include also epileptic seizures. Each chromosomal syndrome has a range of specific characteristics regarding the type of seizures, EEG findings and specific response to antiepileptic drugs, significant in the context of the respective genetic etiology. Therefore, it is very important to know these particularities, in order to avoid an exacerbation of seizures or some side effects. In this paper we will present a review of the epileptic seizures and antiepileptic treatment in some of the most common chromosomal syndromes.
Collapse
Affiliation(s)
- Magdalena Budisteanu
- Prof. Dr. Alexandru Obregia” Clinical Hospital of Psychiatry, BucharestRomania
- ”Victor Babes“ National Institute of Pathology, BucharestRomania
- ”Titu Maiorescu” University – Faculty of Medicine, BucharestRomania
| | - Claudia Jurca
- University of Oradea, Faculty of Medicine and Pharmacy, Preclinical Department, OradeaRomania
| | | | - Ina Focsa
- ”Carol Davila” University of Pharmacy and Medicine, BucharestRomania
| | - Dan Riga
- Prof. Dr. Alexandru Obregia” Clinical Hospital of Psychiatry, BucharestRomania
| | - Sorin Riga
- Prof. Dr. Alexandru Obregia” Clinical Hospital of Psychiatry, BucharestRomania
| | - Alexandru Jurca
- University of Oradea, Faculty of Medicine and Pharmacy, Preclinical Department, OradeaRomania
| | - Aurora Arghir
- ”Victor Babes“ National Institute of Pathology, BucharestRomania
- ”Carol Davila” University of Pharmacy and Medicine, BucharestRomania
| |
Collapse
|
27
|
Herber DL, Weeber EJ, D'Agostino DP, Duis J. Evaluation of the safety and tolerability of a nutritional Formulation in patients with ANgelman Syndrome (FANS): study protocol for a randomized controlled trial. Trials 2020; 21:60. [PMID: 31918761 PMCID: PMC6953273 DOI: 10.1186/s13063-019-3996-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 12/13/2019] [Indexed: 01/12/2023] Open
Abstract
Background Ketogenic and low-glycemic-index diets are effective in treating drug-resistant seizures in children with Angelman syndrome. Cognition, mobility, sleep, and gastrointestinal health are intrinsically linked to seizure activity and overall quality of life. Ketogenic and low-glycemic diets restrict carbohydrate consumption and stabilize blood glucose levels. The ketogenic diet induces ketosis, a metabolic state where ketone bodies are preferentially used for fuel. The use of exogenous ketones in promoting ketosis in Angelman syndrome has not been previously studied. The study formulation evaluated herein contains the exogenous ketone beta-hydroxybutyrate to rapidly shift the body towards ketosis, resulting in enhanced metabolic efficiency. Methods/design This is a 16-week, randomized, double-blind, placebo-controlled, crossover study to assess the safety and tolerability of a nutritional formula containing exogenous ketones. It also examines the potential for exogenous ketones to improve the patient’s nutritional status which can impact the physiologic, symptomatic, and health outcome liabilities of living with Angelman syndrome. Discussion This manuscript outlines the rationale for a study designed to be the first to provide data on nutritional approaches for patients with Angelman syndrome using exogenous ketones. Trial registration ClinicalTrials.gov, ID: NCT03644693. Registered on 23 August 2018. Last updated on 23 August 2018.
Collapse
Affiliation(s)
- Donna L Herber
- Disruptive Nutrition, LLC, 300 West Morgan Street, Suite 1510, Durham, NC, 27701, USA
| | - Edwin J Weeber
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ, 07080-2449, USA
| | - Dominic P D'Agostino
- Morsani College of Medicine, Department of Molecular Pharmacology and Physiology, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA.,Institute for Human and Machine Cognition, Ocala, FL, USA
| | - Jessica Duis
- Division of Medical Genetics & Genomic Medicine, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232-2578, USA.
| |
Collapse
|
28
|
Zhu J, Tsai NP. Ubiquitination and E3 Ubiquitin Ligases in Rare Neurological Diseases with Comorbid Epilepsy. Neuroscience 2020; 428:90-99. [DOI: 10.1016/j.neuroscience.2019.12.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 12/19/2022]
|
29
|
Fisher K, Keng J, Ziegler J. Nutrition Assessment and Intervention in a Pediatric Patient with Angelman Syndrome: A Case Presentation Highlighting Clinical Challenges and Evidence-Based Solutions. Lifestyle Genom 2019; 13:43-52. [PMID: 31786575 DOI: 10.1159/000504300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 10/21/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Angelman syndrome (AS) is a rare disorder of genetic imprinting which results in intellectual and developmental disability. It meets criteria of a disorder of neurologic impairment. A deletion in the long arm of chromosome 15 (del 15q11.2-q13) is responsible for about 70% of cases of AS (deletion genotype). SUMMARY There is a paucity of evidence to allow algorithmic nutrition assessment and intervention in pediatric patients with AS. Therefore, our objective is to use a case presentation to provide an example of nutrition assessment and intervention in a pediatric patient with the deletion genotype of AS and then highlight common challenges to providing evidenced-based nutrition care. For the highlighted challenges, we suggest evidence-based solutions to provide a resource for clinicians who may encounter similar challenges in clinical practice. Key Messages: There are genotype-phenotype correlations in AS that can help guide clinicians regarding nutritionally relevant clinical characteristics and corresponding interventions that are patient specific. The deletion genotype in AS is associated with multiple characteristics that are relevant to nutrition care and may also be different and/or more severe than characteristics seen in other AS genetic mechanisms. There is also overlap in certain nutritionally relevant clinical characteristics between AS and other conditions, including Prader-Willi syndrome, autism spectrum disorders, and disorders of neurological impairment like cerebral palsy. Clinicians can utilize nutrition resources related to these conditions to expand the scope of relevant resources available.
Collapse
Affiliation(s)
- Kelly Fisher
- Department of Clinical and Preventive Nutrition Sciences, Doctor of Clinical Nutrition Program, School of Health Professions, Rutgers University, Newark, New Jersey, USA, .,Department of Nutritional Sciences, Texas Christian University, Fort Worth, Texas, USA,
| | - Jane Keng
- Gastroenterology and Nutrition Clinic, Cook Children's Medical Center, Fort Worth, Texas, USA
| | - Jane Ziegler
- Department of Clinical and Preventive Nutrition Sciences, Doctor of Clinical Nutrition Program, School of Health Professions, Rutgers University, Newark, New Jersey, USA
| |
Collapse
|
30
|
Bindels-de Heus KGCB, Mous SE, Ten Hooven-Radstaake M, van Iperen-Kolk BM, Navis C, Rietman AB, Ten Hoopen LW, Brooks AS, Elgersma Y, Moll HA, de Wit MCY. An overview of health issues and development in a large clinical cohort of children with Angelman syndrome. Am J Med Genet A 2019; 182:53-63. [PMID: 31729827 PMCID: PMC6916553 DOI: 10.1002/ajmg.a.61382] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/17/2019] [Accepted: 09/23/2019] [Indexed: 01/22/2023]
Abstract
This study presents a broad overview of health issues and psychomotor development of 100 children with Angelman syndrome (AS), seen at the ENCORE Expertise Center for AS in Rotterdam, the Netherlands. We aimed to further delineate the phenotype of AS, to evaluate the association of the phenotype with genotype and other determinants such as epilepsy and to get insight in possible targets for intervention. We confirmed the presence of a more severe phenotype in the 15q11.2‐q13 deletion subtype. Novel findings were an association of (early onset of) epilepsy with a negative effect on development, a high occurrence of nonconvulsive status epilepticus, a high rate of crouch gait in the older children with risk of deterioration of mobility, a relatively low occurrence of microcephaly, a higher mean weight for height in all genetic subtypes with a significant higher mean in the nondeletion children, and a high occurrence of hyperphagia across all genetic subtypes. Natural history data are needed to design future trials. With this large clinical cohort with structured prospective and multidisciplinary follow‐up, we provide unbiased data on AS to support further intervention studies to optimize outcome and quality of life of children with AS and their family.
Collapse
Affiliation(s)
- Karen G C B Bindels-de Heus
- Department of Pediatrics, Erasmus MC, Rotterdam, The Netherlands.,ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands
| | - Sabine E Mous
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands.,Department of Child- and Adolescent Psychiatry and Psychology, Erasmus MC, Rotterdam, The Netherlands
| | - Maartje Ten Hooven-Radstaake
- Department of Pediatrics, Erasmus MC, Rotterdam, The Netherlands.,ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands
| | - Bianca M van Iperen-Kolk
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands.,Department of Physical Therapy, Erasmus MC, Rotterdam, The Netherlands
| | - Cindy Navis
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands.,Department of ENT (Speech & Language Pathology), Erasmus MC, Rotterdam, The Netherlands
| | - André B Rietman
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands.,Department of Child- and Adolescent Psychiatry and Psychology, Erasmus MC, Rotterdam, The Netherlands
| | - Leontine W Ten Hoopen
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands.,Department of Child- and Adolescent Psychiatry and Psychology, Erasmus MC, Rotterdam, The Netherlands
| | - Alice S Brooks
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands
| | | | - Ype Elgersma
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands.,Department of Neuroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Henriëtte A Moll
- Department of Pediatrics, Erasmus MC, Rotterdam, The Netherlands.,ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands
| | - Marie-Claire Y de Wit
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands.,Department of Neurology and Pediatric Neurology, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
31
|
Summers J. Using Behavioral Approaches to Assess Memory, Imitation and Motor Performance in Children with Angelman Syndrome: Results of a Pilot Study. Dev Neurorehabil 2019; 22:516-526. [PMID: 31116614 DOI: 10.1080/17518423.2019.1619857] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Purpose: This study was designed to assess memory, imitation of motor actions and motor performance by 12 children (age range 40-151 months) with Angelman syndrome (AS), a rare neurogenetic disorder associated with learning and memory impairments. Methods: Children's functioning was assessed at several time points over a 3-month period. Results: Memory and motor performance tests had acceptable test-retest and inter-rater reliability whereas the motor imitation test did not. Children were able to recall action sequences after a 24-h delay. Memory and motor performance scores were correlated with children's chronological age and raw scores on subdomains of the Vineland-II. Conclusions: These behavioral tests require further development and evaluation but may show promise to accompany standardized assessments that are currently in use with children with AS.
Collapse
Affiliation(s)
- Jane Summers
- Hospital for Sick Children , Toronto , Ontario , Canada.,Department of Psychiatry, University of Toronto , Toronto , ON , Canada
| |
Collapse
|
32
|
Han J, Bichell TJ, Golden S, Anselm I, Waisbren S, Bacino CA, Peters SU, Bird LM, Kimonis V. A placebo-controlled trial of folic acid and betaine in identical twins with Angelman syndrome. Orphanet J Rare Dis 2019; 14:232. [PMID: 31640736 PMCID: PMC6806546 DOI: 10.1186/s13023-019-1216-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/25/2019] [Indexed: 11/21/2022] Open
Abstract
Background Angelman syndrome (AS) is a neurodevelopmental disorder that is caused by maternal genetic deficiency of a gene that encodes E6-AP ubiquitin-protein ligase (gene symbol UBE3A) mapping to chromosome 15q11-q13. AS leads to stiff and jerky gait, excess laughter, seizures, and severe intellectual disability. In some parts of the brain, the paternally inherited UBE3A gene is subject to genomic imprinting by the action of the UBE3A-antisense transcript (UBE3A-ATS) on the paternally inherited allele. Consequently, only the maternally inherited UBE3A gene is expressed in mature neurons. AS occurs due to deletions of the maternal 15q11 − 13 region, paternal uniparental disomy (UPD), imprinting center defects, mutations in the maternal UBE3A gene, or other unknown genetic malfunctions that result in a silenced maternal UBE3A gene in the specific imprinted regions of the brain. Results A potential treatment strategy for AS is to increase methylation of UBE3A-ATS to promote expression of the paternal UBE3A gene and thus ameliorate the clinical phenotypes of AS. We treated two sets of male identical twins with class I deletions with a 1 year treatment trial of either betaine and folic acid versus placebo. We found no statistically significant changes in the clinical parameters tested at the end of the 1 year trial, nor did we find any significant adverse events. Conclusions This study tested the hypothesis that by increasing the methylation of the UBE3A-antisense transcript in Angelman syndrome to promote expression of the silenced paternal UBE3A gene we may ameliorate the clinical phenotypes of AS. We treated two sets of identical twins with placebo versus betaine and folic acid. Although this study represented a novel approach to treating Angelman syndrome, the differences in the developmental testing results was not significant. This paper also discusses the value of monozygotic twin studies in minimizing confounding variables and its utility in conducting small treatment studies. Trial registration NCT00348933. Registered 6 July 2006.
Collapse
Affiliation(s)
- Julia Han
- Division of Genetics and Genomic Medicine, Department of Pediatrics, University of California at Irvine, 101 The City Drive South, Orange, CA, 92868, USA
| | - Terry Jo Bichell
- Consortium for Outcome Measures and Biomarkers for Neurodevelopmental Disorders, Nashville, TN, USA
| | - Stephanie Golden
- Division of Genetics and Metabolism, Department of Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Irina Anselm
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Susan Waisbren
- Division of Genetics and Metabolism, Department of Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Carlos A Bacino
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Sarika U Peters
- Department of Pediatrics, Vanderbilt University, Vanderbilt Kennedy Center for Research on Human Development, Nashville, TN, USA
| | - Lynne M Bird
- Department of Pediatrics, University of California, San Diego, CA, USA.,Division of Genetics/Dysmorphology, Rady Children's Hospital San Diego, San Diego, CA, USA
| | - Virginia Kimonis
- Division of Genetics and Genomic Medicine, Department of Pediatrics, University of California at Irvine, 101 The City Drive South, Orange, CA, 92868, USA. .,Consortium for Outcome Measures and Biomarkers for Neurodevelopmental Disorders, Nashville, TN, USA.
| |
Collapse
|
33
|
Wesseler K, Kraft F, Eggermann T. Molecular and Clinical Opposite Findings in 11p15.5 Associated Imprinting Disorders: Characterization of Basic Mechanisms to Improve Clinical Management. Int J Mol Sci 2019; 20:ijms20174219. [PMID: 31466347 PMCID: PMC6747273 DOI: 10.3390/ijms20174219] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/26/2019] [Accepted: 08/26/2019] [Indexed: 12/14/2022] Open
Abstract
Silver-Russell and Beckwith-Wiedemann syndromes (SRS, BWS) are rare congenital human disorders characterized by opposite growth disturbances. With the increasing knowledge on the molecular basis of SRS and BWS, it has become obvious that the disorders mirror opposite alterations at the same genomic loci in 11p15.5. In fact, these changes directly or indirectly affect the expression of IGF2 and CDKN1C and their associated pathways, and thereby, cause growth disturbances as key features of both diseases. The increase of knowledge has become possible with the development and implementation of new and comprehensive assays. Whereas, in the beginning molecular testing was restricted to single chromosomal loci, many tests now address numerous loci in the same run, and the diagnostic implementation of (epi)genome wide assays is only a question of time. These high-throughput approaches will be complemented by the analysis of other omic datasets (e.g., transcriptome, metabolome, proteome), and it can be expected that the integration of these data will massively improve the understanding of the pathobiology of imprinting disorders and their diagnostics. Especially long-read sequencing methods, e.g., nanopore sequencing, allowing direct detection of native DNA modification, will strongly contribute to a better understanding of genomic imprinting in the near future. Thereby, new genomic loci and types of pathogenic variants will be identified, resulting in more precise discrimination into different molecular subgroups. These subgroups serve as the basis for (epi)genotype-phenotype correlations, allowing a more directed prognosis, counseling, and therapy. By deciphering the pathophysiological consequences of SRS and BWS and their molecular disturbances, future therapies will be available targeting the basic cause of the disease and respective pathomechanisms and will complement conventional therapeutic strategies.
Collapse
Affiliation(s)
- Katharina Wesseler
- Institute of Human Genetics, University Hospital, Technical University Aachen (RWTH), 52074 Aachen, Germany
| | - Florian Kraft
- Institute of Human Genetics, University Hospital, Technical University Aachen (RWTH), 52074 Aachen, Germany
| | - Thomas Eggermann
- Institute of Human Genetics, University Hospital, Technical University Aachen (RWTH), 52074 Aachen, Germany.
| |
Collapse
|
34
|
|
35
|
Barone I, Hawks-Mayer H, Lipton JO. Mechanisms of sleep and circadian ontogeny through the lens of neurodevelopmental disorders. Neurobiol Learn Mem 2019; 160:160-172. [DOI: 10.1016/j.nlm.2019.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 12/05/2018] [Accepted: 01/11/2019] [Indexed: 12/20/2022]
|
36
|
Sousa‐Franco A, Rebelo K, da Rocha ST, Bernardes de Jesus B. LncRNAs regulating stemness in aging. Aging Cell 2019; 18:e12870. [PMID: 30456884 PMCID: PMC6351848 DOI: 10.1111/acel.12870] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 09/18/2018] [Accepted: 09/28/2018] [Indexed: 12/21/2022] Open
Abstract
One of the most outstanding observations from next-generation sequencing approaches was that only 1.5% of our genes code for proteins. The biggest part is transcribed but give rise to different families of RNAs without coding potential. The functional relevance of these abundant transcripts remains far from elucidated. Among them are the long non-coding RNAs (lncRNAs), a relatively large and heterogeneous group of RNAs shown to be highly tissue-specific, indicating a prominent role in processes controlling cellular identity. In particular, lncRNAs have been linked to both stemness properties and detrimental pathways regulating the aging process, being novel players in the intricate network guiding tissue homeostasis. Here, we summarize the up-to-date information on the role of lncRNAs that affect stemness and hence impact upon aging, highlighting the likelihood that lncRNAs may represent an unexploited reservoir of potential therapeutic targets for reprogramming applications and aging-related diseases.
Collapse
Affiliation(s)
- António Sousa‐Franco
- Instituto de Medicina MolecularFaculdade de Medicina da Universidade de LisboaLisboaPortugal
| | - Kenny Rebelo
- Instituto de Medicina MolecularFaculdade de Medicina da Universidade de LisboaLisboaPortugal
| | - Simão Teixeira da Rocha
- Instituto de Medicina MolecularFaculdade de Medicina da Universidade de LisboaLisboaPortugal
| | - Bruno Bernardes de Jesus
- Instituto de Medicina MolecularFaculdade de Medicina da Universidade de LisboaLisboaPortugal
- Department of Medical Sciences and Institute of Biomedicine—iBiMEDUniversity of AveiroAveiroPortugal
| |
Collapse
|
37
|
Sonzogni M, Wallaard I, Santos SS, Kingma J, du Mee D, van Woerden GM, Elgersma Y. A behavioral test battery for mouse models of Angelman syndrome: a powerful tool for testing drugs and novel Ube3a mutants. Mol Autism 2018; 9:47. [PMID: 30220990 PMCID: PMC6137919 DOI: 10.1186/s13229-018-0231-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/03/2018] [Indexed: 12/26/2022] Open
Abstract
Background Angelman syndrome (AS) is a neurodevelopmental disorder caused by mutations affecting UBE3A function. AS is characterized by intellectual disability, impaired motor coordination, epilepsy, and behavioral abnormalities including autism spectrum disorder features. The development of treatments for AS heavily relies on the ability to test the efficacy of drugs in mouse models that show reliable, and preferably clinically relevant, phenotypes. We previously described a number of behavioral paradigms that assess phenotypes in the domains of motor performance, repetitive behavior, anxiety, and seizure susceptibility. Here, we set out to evaluate the robustness of these phenotypes when tested in a standardized test battery. We then used this behavioral test battery to assess the efficacy of minocycline and levodopa, which were recently tested in clinical trials of AS. Methods We combined data of eight independent experiments involving 111 Ube3a mice and 120 wild-type littermate control mice. Using a meta-analysis, we determined the statistical power of the subtests and the effect of putative confounding factors, such as the effect of sex and of animal weight on rotarod performance. We further assessed the robustness of these phenotypes by comparing Ube3a mutants in different genetic backgrounds and by comparing the behavioral phenotypes of independently derived Ube3a-mutant lines. In addition, we investigated if the test battery allowed re-testing the same animals, which would allow a within-subject testing design. Results We find that the test battery is robust across different Ube3a-mutant lines, but confirm and extend earlier studies that several phenotypes are very sensitive to genetic background. We further found that the audiogenic seizure susceptibility phenotype is fully reversible upon pharmacological treatment and highly suitable for dose-finding studies. In agreement with the clinical trial results, we found that minocycline and levodopa treatment of Ube3a mice did not show any sign of improved performance in our test battery. Conclusions Our study provides a useful tool for preclinical drug testing to identify treatments for Angelman syndrome. Since the phenotypes are observed in several independently derived Ube3a lines, the test battery can also be employed to investigate the effect of specific Ube3a mutations on these phenotypes.
Collapse
Affiliation(s)
- Monica Sonzogni
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
| | - Ilse Wallaard
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
| | - Sara Silva Santos
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
| | - Jenina Kingma
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
| | - Dorine du Mee
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
| | - Geeske M. van Woerden
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
| | - Ype Elgersma
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
38
|
A randomized placebo controlled clinical trial to evaluate the efficacy and safety of minocycline in patients with Angelman syndrome (A-MANECE study). Orphanet J Rare Dis 2018; 13:144. [PMID: 30126448 PMCID: PMC6102900 DOI: 10.1186/s13023-018-0891-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/13/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Minocycline is an old tetracycline antibiotic that has shown antiinflammatory and antiapoptotic properties in different neurological disease mouse models. Previous single arm study in humans demonstrated benefits in individuals with Angelman Syndrome (AS); however, its efficacy in patients with Angelman Syndrome has not been assessed in a controlled trial. This was a randomized, double-blind, placebo-controlled, crossover trial in individuals with AS, aged 6 years to 30 years (n = 32, mean age 12 [SD 6·29] years). Participants were randomized to minocycline or placebo for 8 weeks and then switched to the other treatment (a subset of 22 patients) or to receive minocycline for up to 16 weeks (10 patients). After week 16, all patients entered a wash-out 8-week follow-up period. RESULTS Thirty-six subjects were screened and 34 were randomized. Thirty two subjects (94·1%) completed at least the first period and all of them completed the full trial. Intention-to-treat analysis demonstrated the lack of significantly greater improvements in the primary outcome, mean changes in age equivalent of the development index of the Merrill-Palmer Revised Scale after minocycline compared with placebo (1·90 ± 3·16 and 2·00 ± 3·28, respectively, p = 0·937). Longer treatment duration up to 16 weeks did not result in better treatment outcomes (1·86 ± 3·35 for 8 weeks treatment vs 1·20 ± 5·53 for 16 weeks treatment, p = 0·667). Side effects were not significantly different during minocycline and placebo treatments. No serious adverse events occurred on minocycline. CONCLUSIONS Minocycline treatment for up to 16 weeks in children and young adults with AS resulted in lack of significant improvements in development indexes compared to placebo treatment. Treatment with minocycline appears safe and well tolerated; even if it cannot be completely ruled out that longer trials might be required for a potential minocycline effect to be expressed, available results and lack of knowledge on the actual mechanism of action do not support this hypothesis. TRIAL REGISTRATION European Clinical Trial database ( EudraCT 2013-002154-67 ), registered 16th September 2013; US Clinical trials database ( NCT02056665 ), registered 6th February 2014.
Collapse
|
39
|
Tones M, Cross M, Simons C, Napier KR, Hunter A, Bellgard MI, Heussler H. Research protocol: The initiation, design and establishment of the Global Angelman Syndrome Registry. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2018; 62:431-443. [PMID: 29633452 DOI: 10.1111/jir.12482] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 12/18/2017] [Accepted: 02/09/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Angelman syndrome (AS) is a rare neurodevelopmental disorder affecting between 1 in 15 000 and 1 in 24 000 individuals. The condition results in severe developmental and expressive language delays, motor impairments and a unique behavioural phenotype consisting of excessive laughter, smiling and sociability. While many studies have contributed knowledge about the causes and natural history of the syndrome, large scale longitudinal studies are required to advance research and therapeutics for this rare syndrome. METHOD This article describes the protocol for the Global Angelman Syndrome Registry, and some initial findings. Due to the rarity of AS and the variability in symptom presentation, the registry team will strive for complete case ascertainment. Parents and caregivers will submit data to the registry via a secure internet connection. The registry consists of 10 modules that cover patient demographics; developmental, diagnostic, medical and surgical history, behaviour and development, epilepsy, medications and interventions and sleep. RESULTS Since its launch at https://angelmanregistry.info in September 2016, almost 470 individuals with AS have been signed up to the registry worldwide: 59% are from North and South America, 23% are from Europe, 17% are from the Asia Pacific region and 1% are from the Middle East or Africa. The majority of registrants are children, with only 16% aged over 20 years. Most participants indicated a chromosome deletion (76%), with fewer participants indicating a mutation, uniparental disomy or imprinting defect (20%). CONCLUSION Findings indicate a need to consider recruitment strategies that target caregivers of older children and adults, and parents and caregivers from non-English speaking backgrounds.
Collapse
Affiliation(s)
- M Tones
- Developmental Paediatric Group, Mater Medical Research Institute, South Brisbane, Queensland, Australia
| | - M Cross
- Foundation for Angelman Syndrome Therapeutics Australia, Brisbane, Queensland, Australia
| | - C Simons
- Foundation for Angelman Syndrome Therapeutics Australia, Brisbane, Queensland, Australia
| | - K R Napier
- Murdoch University, Centre for Comparative Genomics, Murdoch, Western Australia, Australia
| | - A Hunter
- Murdoch University, Centre for Comparative Genomics, Murdoch, Western Australia, Australia
| | - M I Bellgard
- eResearch Directorate, Queensland University of Technology, Brisbane, Queensland, Australia
| | - H Heussler
- Centre for Children's Health Research University of Queensland, Australia
| |
Collapse
|
40
|
Kim A, Fujimoto M, Hwa V, Backeljauw P, Dauber A. Adrenal Insufficiency, Sex Reversal, and Angelman Syndrome due to Uniparental Disomy Unmasking a Mutation in CYP11A1. Horm Res Paediatr 2018; 89:205-210. [PMID: 29566378 PMCID: PMC5906135 DOI: 10.1159/000487638] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 02/12/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND/AIMS Cholesterol side-chain cleavage enzyme (P450scc) deficiency is a rare genetic disorder causing primary adrenal insufficiency with or without a 46,XY disorder of sexual development (DSD). Herein, we report a case of the combination of primary adrenal insufficiency, a DSD (testes with female external genitalia in a setting of a 47,XXY karyotype), and Angelman syndrome. METHODS Comprehensive genetic analyses were performed, including a single nucleotide polymorphism microarray and whole-exome sequencing. In vitro studies were performed to evaluate the pathogenicity of the novel mutation that was identified by whole-exome sequencing. RESULTS The patient was found to have segmental uniparental disomy (UPD) of chromosome 15 explaining her diagnosis of Angelman syndrome. Whole-exome sequencing further revealed a novel homozygous intronic variant in CYP11A1, the gene encoding P450scc, found within the region of UPD. In vitro studies confirmed that this variant led to decreased efficiency of CYP11A1 splicing. CONCLUSION We report the first case of the combination of 2 rare genetic disorders, Angelman syndrome, and P450scc deficiency. After 20 years of diagnostic efforts, significant advances in genetic diagnostic technology allowed us to determine that these 2 disorders originate from a unified genetic etiology, segmental UPD unmasking a novel recessive mutation in CYP11A1.
Collapse
Affiliation(s)
- Ahlee Kim
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Masanobu Fujimoto
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Vivian Hwa
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Philippe Backeljauw
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Andrew Dauber
- *Andrew Dauber, MD, Division of Endocrinology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., MLC 7012, Cincinnati, OH 45229 (USA), E-Mail
| |
Collapse
|
41
|
George AJ, Hoffiz YC, Charles AJ, Zhu Y, Mabb AM. A Comprehensive Atlas of E3 Ubiquitin Ligase Mutations in Neurological Disorders. Front Genet 2018; 9:29. [PMID: 29491882 PMCID: PMC5817383 DOI: 10.3389/fgene.2018.00029] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/22/2018] [Indexed: 01/11/2023] Open
Abstract
Protein ubiquitination is a posttranslational modification that plays an integral part in mediating diverse cellular functions. The process of protein ubiquitination requires an enzymatic cascade that consists of a ubiquitin activating enzyme (E1), ubiquitin conjugating enzyme (E2) and an E3 ubiquitin ligase (E3). There are an estimated 600-700 E3 ligase genes representing ~5% of the human genome. Not surprisingly, mutations in E3 ligase genes have been observed in multiple neurological conditions. We constructed a comprehensive atlas of disrupted E3 ligase genes in common (CND) and rare neurological diseases (RND). Of the predicted and known human E3 ligase genes, we found ~13% were mutated in a neurological disorder with 83 total genes representing 70 different types of neurological diseases. Of the E3 ligase genes identified, 51 were associated with an RND. Here, we provide an updated list of neurological disorders associated with E3 ligase gene disruption. We further highlight research in these neurological disorders and discuss the advanced technologies used to support these findings.
Collapse
Affiliation(s)
- Arlene J. George
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Yarely C. Hoffiz
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | | | - Ying Zhu
- Creative Media Industries Institute & Department of Computer Science, Georgia State University, Atlanta, GA, United States
| | - Angela M. Mabb
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
42
|
Lovastatin suppresses hyperexcitability and seizure in Angelman syndrome model. Neurobiol Dis 2017; 110:12-19. [PMID: 29097328 DOI: 10.1016/j.nbd.2017.10.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 10/11/2017] [Accepted: 10/27/2017] [Indexed: 11/22/2022] Open
Abstract
Epilepsy is prevalent and often medically intractable in Angelman syndrome (AS). AS mouse model (Ube3am-/p+) shows reduced excitatory neurotransmission but lower seizure threshold. The neural mechanism linking the synaptic dysfunction to the seizure remains elusive. We show that the local circuits of Ube3am-/p+in vitro are hyperexcitable and display a unique epileptiform activity, a phenomenon that is reminiscent of the finding in fragile X syndrome (FXS) mouse model. Similar to the FXS model, lovastatin suppressed the epileptiform activity and audiogenic seizures in Ube3am-/p+. The in vitro model of Ube3am-/p+ is valuable for dissection of neural mechanism and epilepsy drug screening in vivo.
Collapse
|
43
|
Tan WH, Bird LM, Sadhwani A, Barbieri-Welge RL, Skinner SA, Horowitz LT, Bacino CA, Noll LM, Fu C, Hundley RJ, Wink LK, Erickson CA, Barnes GN, Slavotinek A, Jeremy R, Rotenberg A, Kothare SV, Olson HE, Poduri A, Nespeca MP, Chu HC, Willen JM, Haas KF, Weeber EJ, Rufo PA. A randomized controlled trial of levodopa in patients with Angelman syndrome. Am J Med Genet A 2017; 176:1099-1107. [PMID: 28944563 DOI: 10.1002/ajmg.a.38457] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 07/31/2017] [Accepted: 08/01/2017] [Indexed: 11/06/2022]
Abstract
Treatment for Angelman syndrome (AS) is currently limited to symptomatic interventions. A mouse model of AS has reduced calcium/calmodulin-dependent kinase II activity due to excessive phosphorylation of specific threonine residues, leading to diminished long-term potentiation. In a rat model of Parkinson disease, levodopa reduced phosphorylation of various proteins, including calcium/calmodulin-dependent kinase II. Further studies demonstrated that AS mice treated with levodopa performed better on rotarod testing than untreated AS mice. We conducted a multi-center double-blind randomized placebo-controlled 1-year trial of levodopa / carbidopa with either 10 or 15 mg/kg/day of levodopa in children with AS. The outcome of this intervention was assessed using either the Bayley Scales of Infant Development or the Mullen Scales of Early Learning, as well as the Vineland Adaptive Behavior Scales, and the Aberrant Behavior Checklist. Of the 78 participants enrolled, 67 participants received study medication (33 on levodopa, 34 on placebo), and 55 participants (29 on levodopa, 26 on placebo) completed the 1-year study. There were no clinically or statistically significant changes in any of the outcome measures over a 1-year period comparing the levodopa and placebo groups. The number of adverse events reported, including the more serious adverse events, was similar in both groups, but none were related to treatment with levodopa. Our data demonstrate that levodopa is well-tolerated by children with AS. However, in the doses used in this study, it failed to improve their neurodevelopment or behavioral outcome.
Collapse
Affiliation(s)
- Wen-Hann Tan
- Division of Genetics and Genomics, Boston Children's Hospital; Harvard Medical School, Boston, Massachusetts
| | - Lynne M Bird
- Genetics / Dysmorphology, Rady Children's Hospital San Diego; Department of Pediatrics, University of California, San Diego, California
| | - Anjali Sadhwani
- Department of Psychiatry, Boston Children's Hospital; Harvard Medical School, Boston, Massachusetts
| | | | | | | | - Carlos A Bacino
- Genetics Service, Texas Children's Hospital; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Lisa M Noll
- Psychology Service, Texas Children's Hospital; Baylor College of Medicine, Houston, Texas
| | - Cary Fu
- Division of Pediatric Neurology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Rachel J Hundley
- Division of Developmental Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Logan K Wink
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Craig A Erickson
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Gregory N Barnes
- Division of Pediatric Neurology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Anne Slavotinek
- Department of Pediatrics, University of California, San Francisco, California
| | - Rita Jeremy
- Department of Pediatrics, University of California, San Francisco, California
| | - Alexander Rotenberg
- Department of Neurology, Boston Children's Hospital; Harvard Medical School, Boston, Massachusetts
| | - Sanjeev V Kothare
- Department of Neurology, Boston Children's Hospital; Harvard Medical School, Boston, Massachusetts
| | - Heather E Olson
- Department of Neurology, Boston Children's Hospital; Harvard Medical School, Boston, Massachusetts
| | - Annapurna Poduri
- Department of Neurology, Boston Children's Hospital; Harvard Medical School, Boston, Massachusetts
| | - Mark P Nespeca
- Neurology, Rady Children's Hospital San Diego; University of California, San Diego, California
| | - Hillary C Chu
- Division of Genetics and Genomics, Boston Children's Hospital; Harvard Medical School, Boston, Massachusetts
| | - Jennifer M Willen
- Division of Genetics and Genomics, Boston Children's Hospital; Harvard Medical School, Boston, Massachusetts
| | - Kevin F Haas
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Edwin J Weeber
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| | - Paul A Rufo
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital; Harvard Medical School, Boston, Massachusetts
| |
Collapse
|