1
|
Biagioni M, Baronchelli F, Fossati M. Multiscale spatio-temporal dynamics of UBE3A gene in brain physiology and neurodevelopmental disorders. Neurobiol Dis 2024; 201:106669. [PMID: 39293689 DOI: 10.1016/j.nbd.2024.106669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/13/2024] [Accepted: 09/15/2024] [Indexed: 09/20/2024] Open
Abstract
The UBE3A gene, located in the chromosomal region 15q11-13, is subject to neuron-specific genomic imprinting and it plays a critical role in brain development. Genetic defects of UBE3A cause severe neurodevelopmental disorders, namely the Angelman syndrome (AS) and the 15q11.2-q13.3 duplication syndrome (Dup15q). In the last two decades, the development of in vitro and in vivo models of AS and Dup15q were fundamental to improve the understanding of UBE3A function in the brain. However, the pathogenic mechanisms of these diseases remain elusive and effective treatments are lacking. Recent evidence suggests that UBE3A functions are both spatially and temporally specific, varying across subcellular compartments, brain regions, and neuronal circuits. In the present review, we summarize current knowledge on the role of UBE3A in neuronal pathophysiology under this spatio-temporal perspective. Additionally, we propose key research questions that will be instrumental to better understand the pathogenic mechanisms underpinning AS and Dup15q disorders and provide the rationale to develop novel therapies.
Collapse
Affiliation(s)
- Martina Biagioni
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano 20089, MI, Italy
| | - Federica Baronchelli
- CNR - Institute of Neuroscience, Section of Milano, via Manzoni 56, Rozzano 20089, MI, Italy; Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini, 20072 Pieve Emanuele, MI, Italy
| | - Matteo Fossati
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano 20089, MI, Italy; CNR - Institute of Neuroscience, Section of Milano, via Manzoni 56, Rozzano 20089, MI, Italy.
| |
Collapse
|
2
|
Ozarkar SS, Patel RKR, Vulli T, Smith AL, Shen MD, Burette AC, Philpot BD, Styner MA, Hazlett HC. Comparative profiling of white matter development in the human and mouse brain reveals volumetric deficits and delayed myelination in Angelman syndrome. RESEARCH SQUARE 2024:rs.3.rs-4681861. [PMID: 39149488 PMCID: PMC11326408 DOI: 10.21203/rs.3.rs-4681861/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Background Angelman syndrome (AS), a severe neurodevelopmental disorder resulting from the loss of the maternal UBE3A gene, is marked by changes in the brain's white matter (WM). The extent of WM abnormalities seems to correlate with the severity of clinical symptoms, but these deficits are still not well characterized or understood. This study provides the first large-scale measurement of WM volume reduction in children with AS. Furthermore, we probed the underlying neuropathology by examining the progression of myelination in an AS mouse model. Methods We conducted magnetic resonance imaging (MRI) on children with AS (n=32) and neurotypical controls (n=99) aged 0.5-12 years. In parallel, we examined myelination in postnatal Ube3a maternal-null mice (Ube3a m-/p+; AS model), Ube3a paternal-null mice (Ube3a m+/p-), and wildtype controls (Ube3a m+/p+) using immunohistochemistry, Western blotting, and electron microscopy. Results Our data revealed that AS individuals exhibit significant reductions in brain volume by ~1 year of age, with WM reduced by 26% and gray matter by 21% by 6-12 years of age-approximately twice the reductions observed in the adult AS mouse model. In our AS mouse model, we saw a global delay in the onset of myelination, which normalized within days (likely corresponding to months or years in human development). This myelination delay is caused by the loss of UBE3A in neurons rather than UBE3A haploinsufficiency in oligodendrocytes. Interestingly, ultrastructural analyses did not reveal any abnormalities in myelinated or unmyelinated axons. Limitations It is difficult to extrapolate the timing and duration of the myelination delay observed in AS model mice to individuals with AS. Conclusions This study reveals WM deficits as a hallmark in children with AS, demonstrating for the first time that these deficits are already apparent at 1 year of age. Parallel studies in a mouse model of AS show that these deficits may be associated with delayed onset of myelination due to the loss of neuronal (but not glial) UBE3A. These findings emphasize the potential of WM as both a therapeutic target for interventions and a valuable biomarker for tracking the progression of AS and the effectiveness of potential treatments.
Collapse
Affiliation(s)
- Siddhi S Ozarkar
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Ridthi K-R Patel
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Tasmai Vulli
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Audrey L Smith
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Mark D Shen
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Alain C Burette
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Benjamin D Philpot
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Martin A Styner
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel
| | - Heather C Hazlett
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
3
|
Gupta PK, Barak S, Feuermann Y, Goobes G, Kaphzan H. 1H-NMR-based metabolomics reveals metabolic alterations in early development of a mouse model of Angelman syndrome. Mol Autism 2024; 15:31. [PMID: 39049050 PMCID: PMC11267930 DOI: 10.1186/s13229-024-00608-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 07/07/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Angelman syndrome (AS) is a rare neurodevelopmental genetic disorder caused by the loss of function of the ubiquitin ligase E3A (UBE3A) gene, affecting approximately 1:15,000 live births. We have recently shown that mitochondrial function in AS is altered during mid to late embryonic brain development leading to increased oxidative stress and enhanced apoptosis of neural precursor cells. However, the overall alterations of metabolic processes are still unknown. Hence, as a follow-up, we aim to investigate the metabolic profiles of wild-type (WT) and AS littermates and to identify which metabolic processes are aberrant in the brain of AS model mice during embryonic development. METHODS We collected brain tissue samples from mice embryos at E16.5 and performed metabolomic analyses using proton nuclear magnetic resonance (1H-NMR) spectroscopy. Multivariate and Univariate analyses were performed to determine the significantly altered metabolites in AS mice. Pathways associated with the altered metabolites were identified using metabolite set enrichment analysis. RESULTS Our analysis showed that overall, the metabolomic fingerprint of AS embryonic brains differed from those of their WT littermates. Moreover, we revealed a significant elevation of distinct metabolites, such as acetate, lactate, and succinate in the AS samples compared to the WT samples. The elevated metabolites were significantly associated with the pyruvate metabolism and glycolytic pathways. LIMITATIONS Only 14 metabolites were successfully identified and investigated in the present study. The effect of unidentified metabolites and their unresolved peaks was not determined. Additionally, we conducted the metabolomic study on whole brain tissue samples. Employing high-resolution NMR studies on different brain regions could further expand our knowledge regarding metabolic alterations in the AS brain. Furthermore, increasing the sample size could reveal the involvement of more significantly altered metabolites in the pathophysiology of the AS brain. CONCLUSIONS Ube3a loss of function alters bioenergy-related metabolism in the AS brain during embryonic development. Furthermore, these neurochemical changes could be linked to the mitochondrial reactive oxygen species and oxidative stress that occurs during the AS embryonic development.
Collapse
Affiliation(s)
- Pooja Kri Gupta
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, 3103301, Israel
| | - Sharon Barak
- Department of Chemistry and The Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, 5290002, Israel
| | - Yonatan Feuermann
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, 3103301, Israel
| | - Gil Goobes
- Department of Chemistry and The Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, 5290002, Israel
| | - Hanoch Kaphzan
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, 3103301, Israel.
| |
Collapse
|
4
|
Gardner Z, Holbrook O, Tian Y, Odamah K, Man HY. The role of glia in the dysregulation of neuronal spinogenesis in Ube3a-dependent ASD. Exp Neurol 2024; 376:114756. [PMID: 38508482 PMCID: PMC11058030 DOI: 10.1016/j.expneurol.2024.114756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 02/14/2024] [Accepted: 03/14/2024] [Indexed: 03/22/2024]
Abstract
Overexpression of the Ube3a gene and the resulting increase in Ube3a protein are linked to autism spectrum disorder (ASD). However, the cellular and molecular processes underlying Ube3a-dependent ASD remain unclear. Using both male and female mice, we find that neurons in the somatosensory cortex of the Ube3a 2× Tg ASD mouse model display reduced dendritic spine density and increased immature filopodia density. Importantly, the increased gene dosage of Ube3a in astrocytes alone is sufficient to confer alterations in neurons as immature dendritic protrusions, as observed in primary hippocampal neuron cultures. We show that Ube3a overexpression in astrocytes leads to a loss of astrocyte-derived spinogenic protein, thrombospondin-2 (TSP2), due to a suppression of TSP2 gene transcription. By neonatal intraventricular injection of astrocyte-specific virus, we demonstrate that Ube3a overexpression in astrocytes in vivo results in a reduction in dendritic spine maturation in prelimbic cortical neurons, accompanied with autistic-like behaviors in mice. These findings reveal an astrocytic dominance in initiating ASD pathobiology at the neuronal and behavior levels. SIGNIFICANCE STATEMENT: Increased gene dosage of Ube3a is tied to autism spectrum disorders (ASDs), yet cellular and molecular alterations underlying autistic phenotypes remain unclear. We show that Ube3a overexpression leads to impaired dendritic spine maturation, resulting in reduced spine density and increased filopodia density. We find that dysregulation of spine development is not neuron autonomous, rather, it is mediated by an astrocytic mechanism. Increased gene dosage of Ube3a in astrocytes leads to reduced production of the spinogenic glycoprotein thrombospondin-2 (TSP2), leading to abnormalities in spines. Astrocyte-specific Ube3a overexpression in the brain in vivo confers dysregulated spine maturation concomitant with autistic-like behaviors in mice. These findings indicate the importance of astrocytes in aberrant neurodevelopment and brain function in Ube3a-depdendent ASD.
Collapse
Affiliation(s)
- Zachary Gardner
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, United States of America
| | - Otto Holbrook
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, United States of America
| | - Yuan Tian
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, United States of America
| | - KathrynAnn Odamah
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, United States of America
| | - Heng-Ye Man
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, United States of America; Department of Pharmacology, Physiology & Biophysics, Boston University School of Medicine, 72 East Concord St., L-603, Boston, MA 02118, United States of America; Center for Systems Neuroscience, Boston University, 610 Commonwealth Ave, Boston, MA 02215, United States of America.
| |
Collapse
|
5
|
Gonzalez Ramirez C, Salvador SG, Patel RKR, Clark S, Miller NW, James LM, Ringelberg NW, Simon JM, Bennett J, Amaral DG, Burette AC, Philpot BD. Regional and cellular organization of the autism-associated protein UBE3A/E6AP and its antisense transcript in the brain of the developing rhesus monkey. Front Neuroanat 2024; 18:1410791. [PMID: 38873093 PMCID: PMC11169893 DOI: 10.3389/fnana.2024.1410791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/17/2024] [Indexed: 06/15/2024] Open
Abstract
Angelman syndrome (AS) is a neurogenetic disorder caused by mutations or deletions in the maternally-inherited UBE3A allele, leading to a loss of UBE3A protein expression in neurons. The paternally-inherited UBE3A allele is epigenetically silenced in neurons during development by a noncoding transcript (UBE3A-ATS). The absence of neuronal UBE3A results in severe neurological symptoms, including speech and language impairments, intellectual disability, and seizures. While no cure exists, therapies aiming to restore UBE3A function-either by gene addition or by targeting UBE3A-ATS-are under development. Progress in developing these treatments relies heavily on inferences drawn from mouse studies about the function of UBE3A in the human brain. To aid translational efforts and to gain an understanding of UBE3A and UBE3A-ATS biology with greater relevance to human neurodevelopmental contexts, we investigated UBE3A and UBE3A-ATS expression in the developing brain of the rhesus macaque, a species that exhibits complex social behaviors, resembling aspects of human behavior to a greater degree than mice. Combining immunohistochemistry and in situ hybridization, we mapped UBE3A and UBE3A-ATS regional and cellular expression in normal prenatal, neonatal, and adolescent rhesus macaque brains. We show that key hallmarks of UBE3A biology, well-known in rodents, are also present in macaques, and suggest paternal UBE3A silencing in neurons-but not glial cells-in the macaque brain, with onset between gestational day 48 and 100. These findings support proposals that early-life, perhaps even prenatal, intervention is optimal for overcoming the maternal allele loss of UBE3A linked to AS.
Collapse
Affiliation(s)
- Chavely Gonzalez Ramirez
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Sarah G. Salvador
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ridthi Kartik Rekha Patel
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Sarah Clark
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Noah W. Miller
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lucas M. James
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Nicholas W. Ringelberg
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jeremy M. Simon
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, United States
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Jeffrey Bennett
- Department of Psychiatry and Behavioral Sciences, MIND Institute, Davis, CA, United States
- California National Primate Research Center, University of California, Davis, CA, United States
| | - David G. Amaral
- Department of Psychiatry and Behavioral Sciences, MIND Institute, Davis, CA, United States
- California National Primate Research Center, University of California, Davis, CA, United States
| | - Alain C. Burette
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Benjamin D. Philpot
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
6
|
Rohm D, Black JB, McCutcheon SR, Barrera A, Morone DJ, Nuttle X, de Esch CE, Tai DJ, Talkowski ME, Iglesias N, Gersbach CA. Activation of the imprinted Prader-Willi Syndrome locus by CRISPR-based epigenome editing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.03.583177. [PMID: 38496583 PMCID: PMC10942373 DOI: 10.1101/2024.03.03.583177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Epigenome editing with DNA-targeting technologies such as CRISPR-dCas9 can be used to dissect gene regulatory mechanisms and potentially treat associated disorders. For example, Prader-Willi Syndrome (PWS) is caused by loss of paternally expressed imprinted genes on chromosome 15q11.2-q13.3, although the maternal allele is intact but epigenetically silenced. Using CRISPR repression and activation screens in human induced pluripotent stem cells (iPSCs), we identified genomic elements that control expression of the PWS gene SNRPN from the paternal and maternal chromosomes. We showed that either targeted transcriptional activation or DNA demethylation can activate the silenced maternal SNRPN and downstream PWS transcripts. However, these two approaches function at unique regions, preferentially activating different transcript variants and involving distinct epigenetic reprogramming mechanisms. Remarkably, transient expression of the targeted demethylase leads to stable, long-term maternal SNRPN expression in PWS iPSCs. This work uncovers targeted epigenetic manipulations to reprogram a disease-associated imprinted locus and suggests possible therapeutic interventions.
Collapse
Affiliation(s)
- Dahlia Rohm
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Joshua B. Black
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Sean R. McCutcheon
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Alejandro Barrera
- Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27708, USA
| | - Daniel J. Morone
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Xander Nuttle
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Celine E. de Esch
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Derek J.C. Tai
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Michael E. Talkowski
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nahid Iglesias
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Charles A. Gersbach
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
7
|
Roy B, Amemasor E, Hussain S, Castro K. UBE3A: The Role in Autism Spectrum Disorders (ASDs) and a Potential Candidate for Biomarker Studies and Designing Therapeutic Strategies. Diseases 2023; 12:7. [PMID: 38248358 PMCID: PMC10814747 DOI: 10.3390/diseases12010007] [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: 11/13/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/23/2024] Open
Abstract
Published reports from the CDC's Autism and Development Disabilities Monitoring Networks have shown that an average of 1 in every 44 (2.3%) 8-year-old children were estimated to have ASD in 2018. Many of the ASDs exhibiting varying degrees of autism-like phenotypes have chromosomal anomalies in the Chr15q11-q13 region. Numerous potential candidate genes linked with ASD reside in this chromosomal segment. However, several clinical, in vivo, and in vitro studies selected one gene more frequently than others randomly and unbiasedly. This gene codes for UBE3A or Ubiquitin protein ligase E3A [also known as E6AP ubiquitin-protein ligase (E6AP)], an enzyme involved in the cellular degradation of proteins. This gene has been listed as one of the several genes with a high potential of causing ASD in the Autism Database. The gain of function mutations, triplication, or duplication in the UBE3A gene is also associated with ASDs like Angelman Syndrome (AS) and Dup15q Syndrome. The genetic imprinting of UBE3A in the brain and a preference for neuronal maternal-specific expression are the key features of various ASDs. Since the UBE3A gene is involved in two main important diseases associated with autism-like symptoms, there has been widespread research going on in understanding the link between this gene and autism. Additionally, since no universal methodology or mechanism exists for identifying UBE3A-mediated ASD, it continues to be challenging for neurobiologists, neuroscientists, and clinicians to design therapies or diagnostic tools. In this review, we focus on the structure and functional aspects of the UBE3A protein, discuss the primary relevance of the 15q11-q13 region in the cause of ASDs, and highlight the link between UBE3A and ASD. We try to broaden the knowledge of our readers by elaborating on the possible mechanisms underlying UBE3A-mediated ASDs, emphasizing the usage of UBE3A as a prospective biomarker in the preclinical diagnosis of ASDs and discuss the positive outcomes, advanced developments, and the hurdles in the field of therapeutic strategies against UBE3A-mediated ASDs. This review is novel as it lays a very detailed and comprehensive platform for one of the most important genes associated with diseases showing autistic-like symptoms. Additionally, this review also attempts to lay optimistic feedback on the possible steps for the diagnosis, prevention, and therapy of these UBE3A-mediated ASDs in the upcoming years.
Collapse
Affiliation(s)
- Bidisha Roy
- Life Science Centre, Department of Biological Sciences, Rutgers University-Newark, Newark, NJ 07102, USA; (E.A.); (S.H.); (K.C.)
| | | | | | | |
Collapse
|
8
|
Lin CW, Cheng YC, Yang CH, Huang HS. Light activates Ube3a, an Angelman syndrome-associated gene, by mediating the chromatin structures during postnatal development of mouse retina. J Neurochem 2023; 167:766-777. [PMID: 37994169 DOI: 10.1111/jnc.16018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 11/24/2023]
Abstract
Angelman syndrome, a severe neurodevelopmental disorder, is primarily caused by mutations or deletions of maternally inherited ubiquitin protein ligase E3A (UBE3A). Activation of the silenced paternal copy of UBE3A can occur with pharmacological perturbation; however, an environmental approach has not been examined. Here, we found Ube3a is highly expressed in embryonic and early neonatal mouse retina and is maternally-, but not paternally-, expressed in ganglion cells, amacrine cells, and horizontal cells. Moreover, we analyzed UBE3A expression in the retina and visual cortex of postnatal day 28 mice (P28) following exposure to light emissions from white compact-fluorescent bulbs or blue light-emitting diodes from postnatal day 0 (P0) to 28 (P28), encompassing a crucial phase of visual system development. We found higher levels of Ube3a RNA and protein in the retina, but not visual cortex compared with tissues from P28 mice exposure to typical lighting (controls). Levels of both paternal- and maternal-UBE3A protein in mouse retina were higher than controls in P28 mice exposed to white or blue light. Moreover, levels of open and repressive chromatin structures, indicated by histone H3 lysine 4 trimethylation (H3K4me3) and histone H3 lysine 27 trimethylation (H3K27me3), respectively, were increased in the Ube3a promoter from mouse retina exposed to white or blue light. Our findings strongly suggest that extended exposure to white or blue light constitutes a substantial environmental factor that can effectively promote UBE3A expression within the central nervous system.
Collapse
Affiliation(s)
- Chao-Wen Lin
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Ophthalmology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Chun Cheng
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Ophthalmology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chang-Hao Yang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Ophthalmology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsien-Sung Huang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
9
|
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
|
10
|
Chaudhary P, Proulx J, Park IW. Ubiquitin-protein ligase E3A (UBE3A) mediation of viral infection and human diseases. Virus Res 2023; 335:199191. [PMID: 37541588 PMCID: PMC10430597 DOI: 10.1016/j.virusres.2023.199191] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/06/2023]
Abstract
The Ubiquitin-protein ligase E3A, UBE3A, also known as E6-associated protein (E6-AP), is known to play an essential role in regulating the degradation of various proteins by transferring Ub from E2 Ub conjugating enzymes to the substrate proteins. Several studies indicate that UBE3A regulates the stabilities of key viral proteins in the virus-infected cells and, thereby, the infected virus-mediated diseases, even if it were reported that UBE3A participates in non-viral-related human diseases. Furthermore, mutations such as deletions and duplications in the maternally inherited gene in the brain cause human neurodevelopmental disorders such as Angelman syndrome (AS) and autism. It is also known that UBE3A functions as a transcriptional coactivator for the expression of steroid hormone receptors. These reports establish that UBE3A is distinguished by its multitudinous functions that are paramount to viral pathology and human diseases. This review is focused on molecular mechanisms for such intensive participation of UBE3A in disease formation and virus regulation.
Collapse
Affiliation(s)
- Pankaj Chaudhary
- Department of Microbiology, Immunology and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, United States.
| | - Jessica Proulx
- Department of Microbiology, Immunology and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, United States
| | - In-Woo Park
- Department of Microbiology, Immunology and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, United States.
| |
Collapse
|
11
|
Rabeling A, Goolam M. Cerebral organoids as an in vitro model to study autism spectrum disorders. Gene Ther 2023; 30:659-669. [PMID: 35790793 DOI: 10.1038/s41434-022-00356-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/01/2022] [Accepted: 06/23/2022] [Indexed: 11/09/2022]
Abstract
Autism spectrum disorders (ASDs) are a set of disorders characterised by social and communication deficits caused by numerous genetic lesions affecting brain development. Progress in ASD research has been hampered by the lack of appropriate models, as both 2D cell culture as well as animal models cannot fully recapitulate the developing human brain or the pathogenesis of ASD. Recently, cerebral organoids have been developed to provide a more accurate, 3D in vitro model of human brain development. Cerebral organoids have been shown to recapitulate the foetal brain gene expression profile, transcriptome, epigenome, as well as disease dynamics of both idiopathic and syndromic ASDs. They are thus an excellent tool to investigate development of foetal stage ASDs, as well as interventions that can reverse or rescue the altered phenotypes observed. In this review, we discuss the development of cerebral organoids, their recent applications in the study of both syndromic and idiopathic ASDs, their use as an ASD drug development platform, as well as limitations of their use in ASD research.
Collapse
Affiliation(s)
- Alexa Rabeling
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Mubeen Goolam
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa.
- UCT Neuroscience Institute, Cape Town, South Africa.
| |
Collapse
|
12
|
Xing L, Simon JM, Ptacek TS, Yi JJ, Loo L, Mao H, Wolter JM, McCoy ES, Paranjape SR, Taylor-Blake B, Zylka MJ. Autism-linked UBE3A gain-of-function mutation causes interneuron and behavioral phenotypes when inherited maternally or paternally in mice. Cell Rep 2023; 42:112706. [PMID: 37389991 PMCID: PMC10530456 DOI: 10.1016/j.celrep.2023.112706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 04/15/2023] [Accepted: 06/12/2023] [Indexed: 07/02/2023] Open
Abstract
The E3 ubiquitin ligase Ube3a is biallelically expressed in neural progenitors and glial cells, suggesting that UBE3A gain-of-function mutations might cause neurodevelopmental disorders irrespective of parent of origin. Here, we engineered a mouse line that harbors an autism-linked UBE3AT485A (T503A in mouse) gain-of-function mutation and evaluated phenotypes in animals that inherited the mutant allele paternally, maternally, or from both parents. We find that paternally and maternally expressed UBE3AT503A results in elevated UBE3A activity in neural progenitors and glial cells. Expression of UBE3AT503A from the maternal allele, but not the paternal one, leads to a persistent elevation of UBE3A activity in neurons. Mutant mice display behavioral phenotypes that differ by parent of origin. Expression of UBE3AT503A, irrespective of its parent of origin, promotes transient embryonic expansion of Zcchc12 lineage interneurons. Phenotypes of Ube3aT503A mice are distinct from Angelman syndrome model mice. Our study has clinical implications for a growing number of disease-linked UBE3A gain-of-function mutations.
Collapse
Affiliation(s)
- Lei Xing
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jeremy M Simon
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Campus Box #7255, Chapel Hill, NC 27599, USA; Department of Genetics, The University of North Carolina at Chapel Hill, Campus Box #7264, Chapel Hill, NC 27599, USA
| | - Travis S Ptacek
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Campus Box #7255, Chapel Hill, NC 27599, USA
| | - Jason J Yi
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Campus Box #7255, Chapel Hill, NC 27599, USA
| | - Lipin Loo
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hanqian Mao
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Campus Box #7255, Chapel Hill, NC 27599, USA
| | - Justin M Wolter
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Campus Box #7255, Chapel Hill, NC 27599, USA; Department of Genetics, The University of North Carolina at Chapel Hill, Campus Box #7264, Chapel Hill, NC 27599, USA
| | - Eric S McCoy
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Smita R Paranjape
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bonnie Taylor-Blake
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mark J Zylka
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Campus Box #7255, Chapel Hill, NC 27599, USA.
| |
Collapse
|
13
|
Simchi L, Gupta PK, Feuermann Y, Kaphzan H. Elevated ROS levels during the early development of Angelman syndrome alter the apoptotic capacity of the developing neural precursor cells. Mol Psychiatry 2023; 28:2382-2397. [PMID: 36991133 PMCID: PMC10611580 DOI: 10.1038/s41380-023-02038-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/01/2023] [Accepted: 03/14/2023] [Indexed: 03/31/2023]
Abstract
Angelman syndrome (AS) is a rare genetic neurodevelopmental disorder caused by the maternally inherited loss of function of the UBE3A gene. AS is characterized by a developmental delay, lack of speech, motor dysfunction, epilepsy, autistic features, happy demeanor, and intellectual disability. While the cellular roles of UBE3A are not fully understood, studies suggest that the lack of UBE3A function is associated with elevated levels of reactive oxygen species (ROS). Despite the accumulating evidence emphasizing the importance of ROS during early brain development and its involvement in different neurodevelopmental disorders, up to date, the levels of ROS in AS neural precursor cells (NPCs) and the consequences on AS embryonic neural development have not been elucidated. In this study we show multifaceted mitochondrial aberration in AS brain-derived embryonic NPCs, which exhibit elevated mitochondrial membrane potential (ΔΨm), lower levels of endogenous reduced glutathione, excessive mitochondrial ROS (mROS) levels, and increased apoptosis compared to wild-type (WT) littermates. In addition, we report that glutathione replenishment by glutathione-reduced ethyl ester (GSH-EE) corrects the excessive mROS levels and attenuates the enhanced apoptosis in AS NPCs. Studying the glutathione redox imbalance and mitochondrial abnormalities in embryonic AS NPCs provides an essential insight into the involvement of UBE3A in early neural development, information that can serve as a powerful avenue towards a broader view of AS pathogenesis. Moreover, since mitochondrial dysfunction and elevated ROS levels were associated with other neurodevelopmental disorders, the findings herein suggest some potential shared underlying mechanisms for these disorders as well.
Collapse
Affiliation(s)
- Lilach Simchi
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Pooja Kri Gupta
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Yonatan Feuermann
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Hanoch Kaphzan
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel.
| |
Collapse
|
14
|
Rigter PMF, de Konink C, van Woerden GM. Loss of CAMK2G affects intrinsic and motor behavior but has minimal impact on cognitive behavior. Front Neurosci 2023; 16:1086994. [PMID: 36685241 PMCID: PMC9853378 DOI: 10.3389/fnins.2022.1086994] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/07/2022] [Indexed: 01/08/2023] Open
Abstract
Introduction The gamma subunit of calcium/calmodulin-dependent protein kinase 2 (CAMK2G) is expressed throughout the brain and is associated with neurodevelopmental disorders. Research on the role of CAMK2G is limited and attributes different functions to specific cell types. Methods To further expand on the role of CAMK2G in brain functioning, we performed extensive phenotypic characterization of a Camk2g knockout mouse. Results We found different CAMK2G isoforms that show a distinct spatial expression pattern in the brain. Additionally, based on our behavioral characterization, we conclude that CAMK2G plays a minor role in hippocampus-dependent learning and synaptic plasticity. Rather, we show that CAMK2G is required for motor function and that the loss of CAMK2G results in impaired nest-building and marble burying behavior, which are innate behaviors that are associated with impaired neurodevelopment. Discussion Taken together, our results provide evidence for a unique function of this specific CAMK2 isozyme in the brain and further support the role of CAMK2G in neurodevelopment.
Collapse
Affiliation(s)
- Pomme M. F. Rigter
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, Netherlands
- Erfelijke Neuro-Cognitieve Ontwikkelingsstoornissen, Expertise Centre for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
| | - Charlotte de Konink
- Erfelijke Neuro-Cognitieve Ontwikkelingsstoornissen, Expertise Centre for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Geeske M. van Woerden
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, Netherlands
- Erfelijke Neuro-Cognitieve Ontwikkelingsstoornissen, Expertise Centre for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
15
|
Lee D, Chen W, Kaku HN, Zhuo X, Chao ES, Soriano A, Kuncheria A, Flores S, Kim JH, Rivera A, Rigo F, Jafar-nejad P, Beaudet AL, Caudill MS, Xue M. Antisense oligonucleotide therapy rescues disturbed brain rhythms and sleep in juvenile and adult mouse models of Angelman syndrome. eLife 2023; 12:e81892. [PMID: 36594817 PMCID: PMC9904759 DOI: 10.7554/elife.81892] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/30/2022] [Indexed: 01/04/2023] Open
Abstract
UBE3A encodes ubiquitin protein ligase E3A, and in neurons its expression from the paternal allele is repressed by the UBE3A antisense transcript (UBE3A-ATS). This leaves neurons susceptible to loss-of-function of maternal UBE3A. Indeed, Angelman syndrome, a severe neurodevelopmental disorder, is caused by maternal UBE3A deficiency. A promising therapeutic approach to treating Angelman syndrome is to reactivate the intact paternal UBE3A by suppressing UBE3A-ATS. Prior studies show that many neurological phenotypes of maternal Ube3a knockout mice can only be rescued by reinstating Ube3a expression in early development, indicating a restricted therapeutic window for Angelman syndrome. Here, we report that reducing Ube3a-ATS by antisense oligonucleotides in juvenile or adult maternal Ube3a knockout mice rescues the abnormal electroencephalogram (EEG) rhythms and sleep disturbance, two prominent clinical features of Angelman syndrome. Importantly, the degree of phenotypic improvement correlates with the increase of Ube3a protein levels. These results indicate that the therapeutic window of genetic therapies for Angelman syndrome is broader than previously thought, and EEG power spectrum and sleep architecture should be used to evaluate the clinical efficacy of therapies.
Collapse
Affiliation(s)
- Dongwon Lee
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Wu Chen
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Heet Naresh Kaku
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Xinming Zhuo
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Eugene S Chao
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | | | - Allen Kuncheria
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Stephanie Flores
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Joo Hyun Kim
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Armando Rivera
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Frank Rigo
- Ionis PharmaceuticalsCarlsbadUnited States
| | | | - Arthur L Beaudet
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Matthew S Caudill
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Mingshan Xue
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
16
|
Viho EMG, Punt AM, Distel B, Houtman R, Kroon J, Elgersma Y, Meijer OC. The Hippocampal Response to Acute Corticosterone Elevation Is Altered in a Mouse Model for Angelman Syndrome. Int J Mol Sci 2022; 24:ijms24010303. [PMID: 36613751 PMCID: PMC9820460 DOI: 10.3390/ijms24010303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Angelman Syndrome (AS) is a severe neurodevelopmental disorder, caused by the neuronal absence of the ubiquitin protein ligase E3A (UBE3A). UBE3A promotes ubiquitin-mediated protein degradation and functions as a transcriptional coregulator of nuclear hormone receptors, including the glucocorticoid receptor (GR). Previous studies showed anxiety-like behavior and hippocampal-dependent memory disturbances in AS mouse models. Hippocampal GR is an important regulator of the stress response and memory formation, and we therefore investigated whether the absence of UBE3A in AS mice disrupted GR signaling in the hippocampus. We first established a strong cortisol-dependent interaction between the GR ligand binding domain and a UBE3A nuclear receptor box in a high-throughput interaction screen. In vivo, we found that UBE3A-deficient AS mice displayed significantly more variation in circulating corticosterone levels throughout the day compared to wildtypes (WT), with low to undetectable levels of corticosterone at the trough of the circadian cycle. Additionally, we observed an enhanced transcriptomic response in the AS hippocampus following acute corticosterone treatment. Surprisingly, chronic corticosterone treatment showed less contrast between AS and WT mice in the hippocampus and liver transcriptomic responses. This suggests that UBE3A limits the acute stimulation of GR signaling, likely as a member of the GR transcriptional complex. Altogether, these data indicate that AS mice are more sensitive to acute glucocorticoid exposure in the brain compared to WT mice. This suggests that stress responsiveness is altered in AS which could lead to anxiety symptoms.
Collapse
Affiliation(s)
- Eva M. G. Viho
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Correspondence:
| | - A. Mattijs Punt
- Department of Clinical Genetics, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Ben Distel
- Department of Clinical Genetics, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - René Houtman
- Precision Medicine Lab, 5349 AB Oss, The Netherlands
| | - Jan Kroon
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Ype Elgersma
- Department of Clinical Genetics, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Onno C. Meijer
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
17
|
Tanas JK, Kerr DD, Wang L, Rai A, Wallaard I, Elgersma Y, Sidorov MS. Multidimensional analysis of behavior predicts genotype with high accuracy in a mouse model of Angelman syndrome. Transl Psychiatry 2022; 12:426. [PMID: 36192373 PMCID: PMC9529912 DOI: 10.1038/s41398-022-02206-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 12/03/2022] Open
Abstract
Angelman syndrome (AS) is a neurodevelopmental disorder caused by loss of expression of the maternal copy of the UBE3A gene. Individuals with AS have a multifaceted behavioral phenotype consisting of deficits in motor function, epilepsy, cognitive impairment, sleep abnormalities, as well as other comorbidities. Effectively modeling this behavioral profile and measuring behavioral improvement will be crucial for the success of ongoing and future clinical trials. Foundational studies have defined an array of behavioral phenotypes in the AS mouse model. However, no single behavioral test is able to fully capture the complex nature of AS-in mice, or in children. We performed multidimensional analysis (principal component analysis + k-means clustering) to quantify the performance of AS model mice (n = 148) and wild-type littermates (n = 138) across eight behavioral domains. This approach correctly predicted the genotype of mice based on their behavioral profile with ~95% accuracy, and remained effective with reasonable sample sizes (n = ~12-15). Multidimensional analysis was effective using different combinations of behavioral inputs and was able to detect behavioral improvement as a function of treatment in AS model mice. Overall, multidimensional behavioral analysis provides a tool for evaluating the effectiveness of preclinical treatments for AS. Multidimensional analysis of behavior may also be applied to rodent models of related neurodevelopmental disorders, and may be particularly valuable for disorders where individual behavioral tests are less reliable than in AS.
Collapse
Affiliation(s)
- Joseph K Tanas
- Center for Neuroscience Research, Children's National Hospital, Washington, DC, USA
| | - Devante D Kerr
- Center for Neuroscience Research, Children's National Hospital, Washington, DC, USA
- Howard University, Washington, DC, USA
| | - Li Wang
- Center for Neuroscience Research, Children's National Hospital, Washington, DC, USA
| | - Anika Rai
- Center for Neuroscience Research, Children's National Hospital, Washington, DC, USA
| | - Ilse Wallaard
- Department of Clinical Genetics and the ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, Netherlands
| | - Ype Elgersma
- Department of Clinical Genetics and the ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, Netherlands
| | - Michael S Sidorov
- Center for Neuroscience Research, Children's National Hospital, Washington, DC, USA.
- Departments of Pediatrics and Pharmacology & Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
18
|
Punt AM, Judson MC, Sidorov MS, Williams BN, Johnson NS, Belder S, den Hertog D, Davis CR, Feygin MS, Lang PF, Jolfaei MA, Curran PJ, van IJcken WF, Elgersma Y, Philpot BD. Molecular and behavioral consequences of Ube3a gene overdosage in mice. JCI Insight 2022; 7:e158953. [PMID: 36134658 PMCID: PMC9675564 DOI: 10.1172/jci.insight.158953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 08/17/2022] [Indexed: 12/01/2022] Open
Abstract
Chromosome 15q11.2-q13.1 duplication syndrome (Dup15q syndrome) is a severe neurodevelopmental disorder characterized by intellectual disability, impaired motor coordination, and autism spectrum disorder. Chromosomal multiplication of the UBE3A gene is presumed to be the primary driver of Dup15q pathophysiology, given that UBE3A exhibits maternal monoallelic expression in neurons and that maternal duplications typically yield far more severe neurodevelopmental outcomes than paternal duplications. However, studies into the pathogenic effects of UBE3A overexpression in mice have yielded conflicting results. Here, we investigated the neurodevelopmental impact of Ube3a gene overdosage using bacterial artificial chromosome-based transgenic mouse models (Ube3aOE) that recapitulate the increases in Ube3a copy number most often observed in Dup15q. In contrast to previously published Ube3a overexpression models, Ube3aOE mice were indistinguishable from wild-type controls on a number of molecular and behavioral measures, despite suffering increased mortality when challenged with seizures, a phenotype reminiscent of sudden unexpected death in epilepsy. Collectively, our data support a model wherein pathogenic synergy between UBE3A and other overexpressed 15q11.2-q13.1 genes is required for full penetrance of Dup15q syndrome phenotypes.
Collapse
Affiliation(s)
- A. Mattijs Punt
- Department of Clinical Genetics and Department of Neuroscience and
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, Netherlands
| | - Matthew C. Judson
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Michael S. Sidorov
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Brittany N. Williams
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Naomi S. Johnson
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Sabine Belder
- Department of Clinical Genetics and Department of Neuroscience and
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, Netherlands
| | - Dion den Hertog
- Department of Clinical Genetics and Department of Neuroscience and
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, Netherlands
| | - Courtney R. Davis
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Maximillian S. Feygin
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Patrick F. Lang
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Mehrnoush Aghadavoud Jolfaei
- Department of Clinical Genetics and Department of Neuroscience and
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, Netherlands
| | - Patrick J. Curran
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Ype Elgersma
- Department of Clinical Genetics and Department of Neuroscience and
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, Netherlands
| | - Benjamin D. Philpot
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| |
Collapse
|
19
|
Anderson DJ, Pauler FM, McKenna A, Shendure J, Hippenmeyer S, Horwitz MS. Simultaneous brain cell type and lineage determined by scRNA-seq reveals stereotyped cortical development. Cell Syst 2022; 13:438-453.e5. [PMID: 35452605 DOI: 10.1016/j.cels.2022.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 01/21/2022] [Accepted: 03/30/2022] [Indexed: 11/30/2022]
Abstract
Mutations are acquired frequently, such that each cell's genome inscribes its history of cell divisions. Common genomic alterations involve loss of heterozygosity (LOH). LOH accumulates throughout the genome, offering large encoding capacity for inferring cell lineage. Using only single-cell RNA sequencing (scRNA-seq) of mouse brain cells, we found that LOH events spanning multiple genes are revealed as tracts of monoallelically expressed, constitutionally heterozygous single-nucleotide variants (SNVs). We simultaneously inferred cell lineage and marked developmental time points based on X chromosome inactivation and the total number of LOH events while identifying cell types from gene expression patterns. Our results are consistent with progenitor cells giving rise to multiple cortical cell types through stereotyped expansion and distinct waves of neurogenesis. This type of retrospective analysis could be incorporated into scRNA-seq pipelines and, compared with experimental approaches for determining lineage in model organisms, is applicable where genetic engineering is prohibited, such as humans.
Collapse
Affiliation(s)
- Donovan J Anderson
- Allen Discovery Center for Lineage Tracing and Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA 98109, USA
| | - Florian M Pauler
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | | | - Jay Shendure
- Allen Discovery Center for Lineage Tracing, Department of Genome Sciences, and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98109, USA
| | - Simon Hippenmeyer
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Marshall S Horwitz
- Allen Discovery Center for Lineage Tracing and Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
20
|
Shi SQ, Mahoney CE, Houdek P, Zhao W, Anderson MP, Zhuo X, Beaudet A, Sumova A, Scammell TE, Johnson CH. Circadian Rhythms and Sleep Are Dependent Upon Expression Levels of Key Ubiquitin Ligase Ube3a. Front Behav Neurosci 2022; 16:837523. [PMID: 35401134 PMCID: PMC8989470 DOI: 10.3389/fnbeh.2022.837523] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Normal neurodevelopment requires precise expression of the key ubiquitin ligase gene Ube3a. Comparing newly generated mouse models for Ube3a downregulation (models of Angelman syndrome) vs. Ube3a upregulation (models for autism), we find reciprocal effects of Ube3a gene dosage on phenotypes associated with circadian rhythmicity, including the amount of locomotor activity. Consistent with results from neurons in general, we find that Ube3a is imprinted in neurons of the suprachiasmatic nuclei (SCN), the pacemaking circadian brain locus, despite other claims that SCN neurons were somehow exceptional to these imprinting rules. In addition, Ube3a-deficient mice lack the typical drop in wake late in the dark period and have blunted responses to sleep deprivation. Suppression of physical activity by light in Ube3a-deficient mice is not due to anxiety as measured by behavioral tests and stress hormones; quantification of stress hormones may provide a mechanistic link to sleep alteration and memory deficits caused by Ube3a deficiency, and serve as an easily measurable biomarker for evaluating potential therapeutic treatments for Angelman syndrome. We conclude that reduced Ube3a gene dosage affects not only neurodevelopment but also sleep patterns and circadian rhythms.
Collapse
Affiliation(s)
- Shu-qun Shi
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
| | - Carrie E. Mahoney
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Pavel Houdek
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Wenling Zhao
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Matthew P. Anderson
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Xinming Zhuo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | | | - Alena Sumova
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Thomas E. Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Carl Hirschie Johnson
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
- *Correspondence: Carl Hirschie Johnson,
| |
Collapse
|
21
|
Modeling and Targeting Neuroglial Interactions with Human Pluripotent Stem Cell Models. Int J Mol Sci 2022; 23:ijms23031684. [PMID: 35163606 PMCID: PMC8836094 DOI: 10.3390/ijms23031684] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/26/2022] [Accepted: 01/30/2022] [Indexed: 02/05/2023] Open
Abstract
Generation of relevant and robust models for neurological disorders is of main importance for both target identification and drug discovery. The non-cell autonomous effects of glial cells on neurons have been described in a broad range of neurodegenerative and neurodevelopmental disorders, pointing to neuroglial interactions as novel alternative targets for therapeutics development. Interestingly, the recent breakthrough discovery of human induced pluripotent stem cells (hiPSCs) has opened a new road for studying neurological and neurodevelopmental disorders “in a dish”. Here, we provide an overview of the generation and modeling of both neuronal and glial cells from human iPSCs and a brief synthesis of recent work investigating neuroglial interactions using hiPSCs in a pathophysiological context.
Collapse
|
22
|
Hyperexcitable Phenotypes in Induced Pluripotent Stem Cell-Derived Neurons From Patients With 15q11-q13 Duplication Syndrome, a Genetic Form of Autism. Biol Psychiatry 2021; 90:756-765. [PMID: 34538422 PMCID: PMC8571044 DOI: 10.1016/j.biopsych.2021.07.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 06/04/2021] [Accepted: 07/10/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Chromosome 15q11-q13 duplication syndrome (Dup15q) is a neurogenetic disorder caused by duplications of the maternal copy of this region. In addition to hypotonia, motor deficits, and language impairments, patients with Dup15q commonly meet the criteria for autism spectrum disorder and have a high prevalence of seizures. It is known from mouse models that synaptic impairments are a strong component of Dup15q pathophysiology; however, cellular phenotypes that relate to seizures are less clear. The development of patient-derived induced pluripotent stem cells provides a unique opportunity to study human neurons with the exact genetic disruptions that cause Dup15q. METHODS Here, we explored electrophysiological phenotypes in induced pluripotent stem cell-derived neurons from 4 patients with Dup15q compared with 6 unaffected control subjects, 1 patient with a 15q11-q13 paternal duplication, and 3 patients with Angelman syndrome. RESULTS We identified several properties of Dup15q neurons that could contribute to neuronal hyperexcitability and seizure susceptibility. Compared with control neurons, Dup15q neurons had increased excitatory synaptic event frequency and amplitude, increased density of dendritic protrusions, increased action potential firing, and decreased inhibitory synaptic transmission. Dup15q neurons also showed impairments in activity-dependent synaptic plasticity and homeostatic synaptic scaling. Finally, Dup15q neurons showed an increased frequency of spontaneous action potential firing compared with control neurons, in part due to disruption of KCNQ2 potassium channels. CONCLUSIONS Together, these data point to multiple electrophysiological mechanisms of hyperexcitability that may provide new targets for the treatment of seizures and other phenotypes associated with Dup15q.
Collapse
|
23
|
Judson MC, Shyng C, Simon JM, Davis CR, Punt AM, Salmon MT, Miller NW, Ritola KD, Elgersma Y, Amaral DG, Gray SJ, Philpot BD. Dual-isoform hUBE3A gene transfer improves behavioral and seizure outcomes in Angelman syndrome model mice. JCI Insight 2021; 6:144712. [PMID: 34676830 PMCID: PMC8564914 DOI: 10.1172/jci.insight.144712] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 09/02/2021] [Indexed: 12/23/2022] Open
Abstract
Loss of the maternal UBE3A allele causes Angelman syndrome (AS), a debilitating neurodevelopmental disorder. Here, we devised an AS treatment strategy based on reinstating dual-isoform expression of human UBE3A (hUBE3A) in the developing brain. Kozak sequence engineering of our codon-optimized vector (hUBE3Aopt) enabled translation of both short and long hUBE3A protein isoforms at a near-endogenous 3:1 (short/long) ratio, a feature that could help to support optimal therapeutic outcomes. To model widespread brain delivery and early postnatal onset of hUBE3A expression, we packaged the hUBE3Aopt vector into PHP.B capsids and performed intracerebroventricular injections in neonates. This treatment significantly improved motor learning and innate behaviors in AS mice, and it rendered them resilient to epileptogenesis and associated hippocampal neuropathologies induced by seizure kindling. hUBE3A overexpression occurred frequently in the hippocampus but was uncommon in the neocortex and other major brain structures; furthermore, it did not correlate with behavioral performance. Our results demonstrate the feasibility, tolerability, and therapeutic potential for dual-isoform hUBE3A gene transfer in the treatment of AS.
Collapse
Affiliation(s)
- Matthew C Judson
- Neuroscience Center.,Department of Cell Biology and Physiology.,Carolina Institute for Developmental Disabilities
| | - Charles Shyng
- Carolina Institute for Developmental Disabilities.,Gene Therapy Center, and
| | - Jeremy M Simon
- Neuroscience Center.,Carolina Institute for Developmental Disabilities.,Department of Genetics, University of North Carolina (UNC), Chapel Hill, North Carolina, USA
| | | | - A Mattijs Punt
- Department of Clinical Genetics and.,Department of Neuroscience, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | - Noah W Miller
- Neuroscience Center.,Department of Cell Biology and Physiology
| | - Kimberly D Ritola
- Neuroscience Center.,Department of Pharmacology, UNC, Chapel Hill, North Carolina, USA.,Scientific Operations Manager-Viral Tools, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
| | - Ype Elgersma
- Department of Clinical Genetics and.,Department of Neuroscience, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - David G Amaral
- Department of Psychiatry and Behavioral Sciences, MIND Institute, and.,California National Primate Research Center, University of California, Davis, California, USA
| | - Steven J Gray
- Gene Therapy Center, and.,Department of Pediatrics and.,Eugene McDermott Center for Human Growth and Development, University of Texas (UT) Southwestern Medical Center, Dallas, Texas, USA
| | - Benjamin D Philpot
- Neuroscience Center.,Department of Cell Biology and Physiology.,Carolina Institute for Developmental Disabilities
| |
Collapse
|
24
|
Rayi PR, Kaphzan H. Electrophysiological Characterization of Regular and Burst Firing Pyramidal Neurons of the Dorsal Subiculum in an Angelman Syndrome Mouse Model. Front Cell Neurosci 2021; 15:670998. [PMID: 34512263 PMCID: PMC8427506 DOI: 10.3389/fncel.2021.670998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 08/04/2021] [Indexed: 11/21/2022] Open
Abstract
Angelman syndrome (AS) is a debilitating neurogenetic disorder characterized by severe developmental delay, speech impairment, gait ataxia, sleep disturbances, epilepsy, and a unique behavioral phenotype. AS is caused by a microdeletion or mutation in the maternal 15q11-q13 chromosome region containing UBE3A gene. The hippocampus is one of the important brain regions affected in AS mice leading to substantial hippocampal-dependent cognitive and behavioral deficits. Recent studies have suggested an abnormal increase in the α1-Na/K-ATPase (α1-NaKA) in AS mice as the precipitating factor leading to the hippocampal deficits. A subsequent study showed that the hippocampal-dependent behavioral deficits occur as a result of altered calcium (Ca+2) dynamics in the CA1 pyramidal neurons (PNs) caused by the elevated α1-NaKA expression levels in the AS mice. Nonetheless, a causal link between hippocampal deficits and major behavioral phenotypes in AS is still obscure. Subiculum, a region adjacent to the hippocampal CA1 is the major output source of the hippocampus and plays an important role in the transfer of information from the CA1 region to the cortical areas. However, in spite of the robust hippocampal deficits and several known electrophysiological alterations in multiple brain regions in AS mice, the neuronal properties of the subicular neurons were never investigated in these mice. Additionally, subicular function is also implied in many neuropsychiatric disorders such as autism, schizophrenia, Alzheimer’s disease, and epilepsy that share some common features with AS. Therefore, given the importance of the subiculum in these neuropsychiatric disorders and the altered electrophysiological properties of the hippocampal CA1 PNs projecting to the subiculum, we sought to examine the subicular PNs. We performed whole-cell recordings from dorsal subiculum of both WT and AS mice and found three distinct populations of PNs based on their ability to fire bursts or single action potentials following somatic current injection: strong bursting, weak bursting, and regular firing neurons. We found no overall differences in the distribution of these different subicular PN populations among AS and WT controls. However, the different cell types showed distinct alterations in their intrinsic membrane properties. Further, none of these populations were altered in their excitatory synaptic properties. Altogether, our study characterized the different subtypes of PNs in the subicular region of an AS mouse model.
Collapse
Affiliation(s)
- Prudhvi Raj Rayi
- Sagol Department of Neurobiology, The Integrated Brain and Behavior Research Center, University of Haifa, Haifa, Israel
| | - Hanoch Kaphzan
- Sagol Department of Neurobiology, The Integrated Brain and Behavior Research Center, University of Haifa, Haifa, Israel
| |
Collapse
|
25
|
Sirois CL, Bloom JE, Fink JJ, Gorka D, Keller S, Germain ND, Levine ES, Chamberlain SJ. Abundance and localization of human UBE3A protein isoforms. Hum Mol Genet 2021; 29:3021-3031. [PMID: 32833011 DOI: 10.1093/hmg/ddaa191] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/13/2020] [Accepted: 08/17/2020] [Indexed: 01/01/2023] Open
Abstract
Loss of UBE3A expression, a gene regulated by genomic imprinting, causes Angelman syndrome (AS), a rare neurodevelopmental disorder. The UBE3A gene encodes an E3 ubiquitin ligase with three known protein isoforms in humans. Studies in mouse suggest that the human isoforms may have differences in localization and neuronal function. A recent case study reported mild AS phenotypes in individuals lacking one specific isoform. Here we have used CRISPR/Cas9 to generate isogenic human embryonic stem cells (hESCs) that lack the individual protein isoforms. We demonstrate that isoform 1 accounts for the majority of UBE3A protein in hESCs and neurons. We also show that UBE3A predominantly localizes to the cytoplasm in both wild type and isoform-null cells. Finally, we show that neurons lacking isoform 1 display a less severe electrophysiological AS phenotype.
Collapse
Affiliation(s)
- Carissa L Sirois
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Judy E Bloom
- Richard D. Berlin Center for Cell Analysis and Modeling, University of Connecticut Health Center, Farmington, CT 06030, USA.,Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - James J Fink
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Dea Gorka
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Steffen Keller
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Noelle D Germain
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Eric S Levine
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Stormy J Chamberlain
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA.,Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
26
|
Copping NA, McTighe SM, Fink KD, Silverman JL. Emerging Gene and Small Molecule Therapies for the Neurodevelopmental Disorder Angelman Syndrome. Neurotherapeutics 2021; 18:1535-1547. [PMID: 34528170 PMCID: PMC8608975 DOI: 10.1007/s13311-021-01082-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2021] [Indexed: 02/07/2023] Open
Abstract
Angelman syndrome (AS) is a rare (~1:15,000) neurodevelopmental disorder characterized by severe developmental delay and intellectual disability, impaired communication skills, and a high prevalence of seizures, sleep disturbances, ataxia, motor deficits, and microcephaly. AS is caused by loss-of-function of the maternally inherited UBE3A gene. UBE3A is located on chromosome 15q11-13 and is biallelically expressed throughout the body but only maternally expressed in the brain due to an RNA antisense transcript that silences the paternal copy. There is currently no cure for AS, but advancements in small molecule drugs and gene therapies offer a promising approach for the treatment of the disorder. Here, we review AS and how loss-of-function of the maternal UBE3A contributes to the disorder. We also discuss the strengths and limitations of current animal models of AS. Furthermore, we examine potential small molecule drug and gene therapies for the treatment of AS and associated challenges faced by the therapeutic design. Finally, gene therapy offers the opportunity for precision medicine in AS and advancements in the treatment of this disorder can serve as a foundation for other single-gene neurodevelopmental disorders.
Collapse
Affiliation(s)
- Nycole A Copping
- School of Medicine, Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California, Research II Building 96, 4625 2nd Avenue, Suite 1001B, Davis, Sacramento, CA, 95817, USA
- Stem Cell Program and Gene Therapy Center, Department of Neurology, MIND Institute, University of California, Davis, Sacramento, CA, USA
| | | | - Kyle D Fink
- Stem Cell Program and Gene Therapy Center, Department of Neurology, MIND Institute, University of California, Davis, Sacramento, CA, USA
| | - Jill L Silverman
- School of Medicine, Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California, Research II Building 96, 4625 2nd Avenue, Suite 1001B, Davis, Sacramento, CA, 95817, USA.
| |
Collapse
|
27
|
Yang L, Shu X, Mao S, Wang Y, Du X, Zou C. Genotype-Phenotype Correlations in Angelman Syndrome. Genes (Basel) 2021; 12:987. [PMID: 34203304 PMCID: PMC8304328 DOI: 10.3390/genes12070987] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/15/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Angelman syndrome (AS) is a rare neurodevelopmental disease that is caused by the loss of function of the maternal copy of ubiquitin-protein ligase E3A (UBE3A) on the chromosome 15q11-13 region. AS is characterized by global developmental delay, severe intellectual disability, lack of speech, happy disposition, ataxia, epilepsy, and distinct behavioral profile. There are four molecular mechanisms of etiology: maternal deletion of chromosome 15q11-q13, paternal uniparental disomy of chromosome 15q11-q13, imprinting defects, and maternally inherited UBE3A mutations. Different genetic types may show different phenotypes in performance, seizure, behavior, sleep, and other aspects. AS caused by maternal deletion of 15q11-13 appears to have worse development, cognitive skills, albinism, ataxia, and more autistic features than those of other genotypes. Children with a UBE3A mutation have less severe phenotypes and a nearly normal development quotient. In this review, we proposed to review genotype-phenotype correlations based on different genotypes. Understanding the pathophysiology of the different genotypes and the genotype-phenotype correlations will offer an opportunity for individualized treatment and genetic counseling. Genotype-phenotype correlations based on larger data should be carried out for identifying new treatment modalities.
Collapse
Affiliation(s)
- Lili Yang
- Department of Genetics and Metabolism, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China;
| | - Xiaoli Shu
- Department of Laboratory Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China;
| | - Shujiong Mao
- Division of Neonatology, Department of Pediatrics, Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China;
| | - Yi Wang
- Department of Neurology, Children’s Hospital of Fudan University, Shanghai 201102, China; (Y.W.); (X.D.)
| | - Xiaonan Du
- Department of Neurology, Children’s Hospital of Fudan University, Shanghai 201102, China; (Y.W.); (X.D.)
| | - Chaochun Zou
- Department of Endocrinology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| |
Collapse
|
28
|
Adhikari A, Copping NA, Beegle J, Cameron DL, Deng P, O'Geen H, Segal DJ, Fink KD, Silverman JL, Anderson JS. Functional rescue in an Angelman syndrome model following treatment with lentivector transduced hematopoietic stem cells. Hum Mol Genet 2021; 30:1067-1083. [PMID: 33856035 PMCID: PMC8188406 DOI: 10.1093/hmg/ddab104] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 12/14/2022] Open
Abstract
Angelman syndrome (AS) is a rare neurodevelopmental disorder characterized by impaired communication skills, ataxia, motor and balance deficits, intellectual disabilities, and seizures. The genetic cause of AS is the neuronal loss of UBE3A expression in the brain. A novel approach, described here, is a stem cell gene therapy which uses lentivector-transduced hematopoietic stem and progenitor cells to deliver functional UBE3A to affected cells. We have demonstrated both the prevention and reversal of AS phenotypes upon transplantation and engraftment of human CD34+ cells transduced with a Ube3a lentivector in a novel immunodeficient Ube3amat−/pat+ IL2rg−/y mouse model of AS. A significant improvement in motor and cognitive behavioral assays as well as normalized delta power measured by electroencephalogram was observed in neonates and adults transplanted with the gene modified cells. Human hematopoietic profiles observed in the lymphoid organs by detection of human immune cells were normal. Expression of UBE3A was detected in the brains of the adult treatment group following immunohistochemical staining illustrating engraftment of the gene-modified cells expressing UBE3A in the brain. As demonstrated with our data, this stem cell gene therapy approach offers a promising treatment strategy for AS, not requiring a critical treatment window.
Collapse
Affiliation(s)
- Anna Adhikari
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Nycole A Copping
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Julie Beegle
- Stem Cell Program, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - David L Cameron
- Stem Cell Program, Department of Neurology, Institute for Regenerative Cures, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Peter Deng
- Stem Cell Program, Department of Neurology, Institute for Regenerative Cures, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Henriette O'Geen
- Department of Biochemistry and Medical Microbiology, UC Davis Genome Center, University of California Davis School of Medicine, Davis, CA 95616, USA
| | - David J Segal
- Department of Biochemistry and Medical Microbiology, UC Davis Genome Center, University of California Davis School of Medicine, Davis, CA 95616, USA
| | - Kyle D Fink
- Stem Cell Program, Department of Neurology, Institute for Regenerative Cures, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Jill L Silverman
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Joseph S Anderson
- Stem Cell Program, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| |
Collapse
|
29
|
Sen D, Drobna Z, Keung AJ. Evaluation of UBE3A antibodies in mice and human cerebral organoids. Sci Rep 2021; 11:6323. [PMID: 33737669 PMCID: PMC7973473 DOI: 10.1038/s41598-021-85923-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/08/2021] [Indexed: 11/20/2022] Open
Abstract
UBE3A is an E3 ubiquitin ligase encoded by the neurally imprinted UBE3A gene. The abundance and subcellular distribution of UBE3A has been the topic of many previous studies as its dosage and localization has been linked to neurodevelopmental disorders including Autism, Dup15q syndrome, and Angelman syndrome. While commercially available antibodies have been widely employed to determine UBE3A localization, an extensive analysis and comparison of the performance of different UBE3A antibodies has not been conducted. Here we evaluated the specificities of seven commercial UBE3A antibodies in two of the major experimental models used in UBE3A research, mouse and human pluripotent stem cell-derived neural cells and tissues. We tested these antibodies in their two most common assays, immunofluorescence and western blot. In addition, we also assessed the ability of these antibodies to capture dynamic spatiotemporal changes of UBE3A by utilizing human cerebral organoid models. Our results reveal that among the seven antibodies tested, three antibodies demonstrated substantial nonspecific immunoreactivity. While four antibodies show specific localization patterns in both mouse brain sections and human cerebral organoids, these antibodies varied significantly in background signals and staining patterns in undifferentiated human pluripotent stem cells.
Collapse
Affiliation(s)
- Dilara Sen
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC, 27695-7905, USA
| | - Zuzana Drobna
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC, 27695-7905, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695-7614, USA
| | - Albert J Keung
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC, 27695-7905, USA.
| |
Collapse
|
30
|
Frohlich J, Toker D, Monti MM. Consciousness among delta waves: a paradox? Brain 2021; 144:2257-2277. [PMID: 33693596 DOI: 10.1093/brain/awab095] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 02/12/2021] [Accepted: 02/25/2021] [Indexed: 01/29/2023] Open
Abstract
A common observation in EEG research is that consciousness vanishes with the appearance of delta (1 - 4 Hz) waves, particularly when those waves are high amplitude. High amplitude delta oscillations are very frequently observed in states of diminished consciousness, including slow wave sleep, anaesthesia, generalised epileptic seizures, and disorders of consciousness such as coma and vegetative state. This strong correlation between loss of consciousness and high amplitude delta oscillations is thought to stem from the widespread cortical deactivation that occurs during the "down states" or troughs of these slow oscillations. Recently, however, many studies have reported the presence of prominent delta activity during conscious states, which casts doubt on the hypothesis that high amplitude delta oscillations are an indicator of unconsciousness. These studies include work in Angelman syndrome, epilepsy, behavioural responsiveness during propofol anaesthesia, postoperative delirium, and states of dissociation from the environment such as dreaming and powerful psychedelic states. The foregoing studies complement an older, yet largely unacknowledged, body of literature that has documented awake, conscious patients with high amplitude delta oscillations in clinical reports from Rett syndrome, Lennox-Gastaut syndrome, schizophrenia, mitochondrial diseases, hepatic encephalopathy, and nonconvulsive status epilepticus. At the same time, a largely parallel body of recent work has reported convincing evidence that the complexity or entropy of EEG and magnetoencephalogram or MEG signals strongly relates to an individual's level of consciousness. Having reviewed this literature, we discuss plausible mechanisms that would resolve the seeming contradiction between high amplitude delta oscillations and consciousness. We also consider implications concerning theories of consciousness, such as integrated information theory and the entropic brain hypothesis. Finally, we conclude that false inferences of unconscious states can be best avoided by examining measures of electrophysiological complexity in addition to spectral power.
Collapse
Affiliation(s)
- Joel Frohlich
- Department of Psychology, University of California Los Angeles, 3423 Franz Hall, Los Angeles, California 90095, USA
| | - Daniel Toker
- Department of Psychology, University of California Los Angeles, 3423 Franz Hall, Los Angeles, California 90095, USA
| | - Martin M Monti
- Department of Psychology, University of California Los Angeles, 3423 Franz Hall, Los Angeles, California 90095, USA.,Department of Neurosurgery, UCLA Brain Injury Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
31
|
Pauler FM, Hudson QJ, Laukoter S, Hippenmeyer S. Inducible uniparental chromosome disomy to probe genomic imprinting at single-cell level in brain and beyond. Neurochem Int 2021; 145:104986. [PMID: 33600873 DOI: 10.1016/j.neuint.2021.104986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 01/23/2021] [Accepted: 02/06/2021] [Indexed: 12/27/2022]
Abstract
Genomic imprinting is an epigenetic mechanism that results in parental allele-specific expression of ~1% of all genes in mouse and human. Imprinted genes are key developmental regulators and play pivotal roles in many biological processes such as nutrient transfer from the mother to offspring and neuronal development. Imprinted genes are also involved in human disease, including neurodevelopmental disorders, and often occur in clusters that are regulated by a common imprint control region (ICR). In extra-embryonic tissues ICRs can act over large distances, with the largest surrounding Igf2r spanning over 10 million base-pairs. Besides classical imprinted expression that shows near exclusive maternal or paternal expression, widespread biased imprinted expression has been identified mainly in brain. In this review we discuss recent developments mapping cell type specific imprinted expression in extra-embryonic tissues and neocortex in the mouse. We highlight the advantages of using an inducible uniparental chromosome disomy (UPD) system to generate cells carrying either two maternal or two paternal copies of a specific chromosome to analyze the functional consequences of genomic imprinting. Mosaic Analysis with Double Markers (MADM) allows fluorescent labeling and concomitant induction of UPD sparsely in specific cell types, and thus to over-express or suppress all imprinted genes on that chromosome. To illustrate the utility of this technique, we explain how MADM-induced UPD revealed new insights about the function of the well-studied Cdkn1c imprinted gene, and how MADM-induced UPDs led to identification of highly cell type specific phenotypes related to perturbed imprinted expression in the mouse neocortex. Finally, we give an outlook on how MADM could be used to probe cell type specific imprinted expression in other tissues in mouse, particularly in extra-embryonic tissues.
Collapse
Affiliation(s)
- Florian M Pauler
- Institute of Science and Technology Austria, Am Campus 1, 3400, Klosterneuburg, Austria
| | - Quanah J Hudson
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Susanne Laukoter
- Institute of Science and Technology Austria, Am Campus 1, 3400, Klosterneuburg, Austria
| | - Simon Hippenmeyer
- Institute of Science and Technology Austria, Am Campus 1, 3400, Klosterneuburg, Austria.
| |
Collapse
|
32
|
Ravi K, Paidas MJ, Saad A, Jayakumar AR. Astrocytes in rare neurological conditions: Morphological and functional considerations. J Comp Neurol 2021; 529:2676-2705. [PMID: 33496339 DOI: 10.1002/cne.25118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 01/16/2021] [Accepted: 01/19/2021] [Indexed: 01/06/2023]
Abstract
Astrocytes are a population of central nervous system (CNS) cells with distinctive morphological and functional characteristics that differ within specific areas of the brain and are widely distributed throughout the CNS. There are mainly two types of astrocytes, protoplasmic and fibrous, which differ in morphologic appearance and location. Astrocytes are important cells of the CNS that not only provide structural support, but also modulate synaptic activity, regulate neuroinflammatory responses, maintain the blood-brain barrier, and supply energy to neurons. As a result, astrocytic disruption can lead to widespread detrimental effects and can contribute to the pathophysiology of several neurological conditions. The characteristics of astrocytes in more common neuropathologies such as Alzheimer's and Parkinson's disease have significantly been described and continue to be widely studied. However, there still exist numerous rare neurological conditions in which astrocytic involvement is unknown and needs to be explored. Accordingly, this review will summarize functional and morphological changes of astrocytes in various rare neurological conditions based on current knowledge thus far and highlight remaining neuropathologies where astrocytic involvement has yet to be investigated.
Collapse
Affiliation(s)
- Karthik Ravi
- University of Michigan, Ann Arbor, Michigan, USA
| | - Michael J Paidas
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami School of Medicine, Miami, Florida, USA
| | - Ali Saad
- Pathology and Laboratory Medicine, University of Miami School of Medicine, Miami, Florida, USA
| | - Arumugam R Jayakumar
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami School of Medicine, Miami, Florida, USA.,South Florida VA Foundation for Research and Education Inc, Miami, Florida, USA.,General Medical Research Neuropathology Section, R&D Service, Veterans Affairs Medical Centre, Miami, Florida, USA
| |
Collapse
|
33
|
Keute M, Miller MT, Krishnan ML, Sadhwani A, Chamberlain S, Thibert RL, Tan WH, Bird LM, Hipp JF. Angelman syndrome genotypes manifest varying degrees of clinical severity and developmental impairment. Mol Psychiatry 2021; 26:3625-3633. [PMID: 32792659 PMCID: PMC8505254 DOI: 10.1038/s41380-020-0858-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 11/18/2022]
Abstract
Angelman Syndrome (AS) is a severe neurodevelopmental disorder due to impaired expression of UBE3A in neurons. There are several genetic mechanisms that impair UBE3A expression, but they differ in how neighboring genes on chromosome 15 at 15q11-q13 are affected. There is evidence that different genetic subtypes present with different clinical severity, but a systematic quantitative investigation is lacking. Here we analyze natural history data on a large sample of individuals with AS (n = 250, 848 assessments), including clinical scales that quantify development of motor, cognitive, and language skills (Bayley Scales of Infant Development, Third Edition; Preschool Language Scale, Fourth Edition), adaptive behavior (Vineland Adaptive Behavioral Scales, Second Edition), and AS-specific symptoms (AS Clinical Severity Scale). We found that clinical severity, as captured by these scales, differs between genetic subtypes: individuals with UBE3A pathogenic variants and imprinting defects (IPD) are less affected than individuals with uniparental paternal disomy (UPD); of those with UBE3A pathogenic variants, individuals with truncating mutations are more impaired than those with missense mutations. Individuals with a deletion that encompasses UBE3A and other genes are most impaired, but in contrast to previous work, we found little evidence for an influence of deletion length (class I vs. II) on severity of manifestations. The results of this systematic analysis highlight the relevance of genomic regions beyond UBE3A as contributing factors in the AS phenotype, and provide important information for the development of new therapies for AS. More generally, this work exemplifies how increasing genetic irregularities are reflected in clinical severity.
Collapse
Affiliation(s)
- Marius Keute
- grid.417570.00000 0004 0374 1269Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, Basel, Switzerland ,grid.5807.a0000 0001 1018 4307Department of Neurology, Otto-von-Guericke-University, Magdeburg, Germany
| | - Meghan T. Miller
- grid.417570.00000 0004 0374 1269Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, Basel, Switzerland
| | - Michelle L. Krishnan
- grid.417570.00000 0004 0374 1269Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, Basel, Switzerland
| | - Anjali Sadhwani
- grid.2515.30000 0004 0378 8438Department of Psychiatry, Boston Children’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XHarvard Medical School, Boston, MA USA
| | - Stormy Chamberlain
- grid.63054.340000 0001 0860 4915Department of Genetics and Genome Sciences, University of Connecticut, Farmington, CT USA
| | - Ronald L. Thibert
- grid.32224.350000 0004 0386 9924Department of Neurology, Massachusetts General Hospital, Boston, MA USA
| | - Wen-Hann Tan
- grid.38142.3c000000041936754XHarvard Medical School, Boston, MA USA ,grid.2515.30000 0004 0378 8438Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA USA
| | - Lynne M. Bird
- grid.266100.30000 0001 2107 4242Department of Pediatrics, University of California, San Diego, CA USA ,grid.286440.c0000 0004 0383 2910Department of Genetics/Dysmorphology, Rady Children’s Hospital, San Diego, CA USA
| | - Joerg F. Hipp
- grid.417570.00000 0004 0374 1269Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
34
|
Buonfiglio D, Hummer DL, Armstrong A, Christopher Ehlen J, DeBruyne JP. Angelman syndrome and melatonin: What can they teach us about sleep regulation. J Pineal Res 2020; 69:e12697. [PMID: 32976638 PMCID: PMC7577950 DOI: 10.1111/jpi.12697] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 01/20/2023]
Abstract
In 1965, Dr Harry Angelman reported a neurodevelopmental disorder affecting three unrelated children who had similar symptoms: brachycephaly, mental retardation, ataxia, seizures, protruding tongues, and remarkable paroxysms of laughter. Over the past 50 years, the disorder became Angelman's namesake and symptomology was expanded to include hyper-activity, stereotypies, and severe sleep disturbances. The sleep disorders in many Angelman syndrome (AS) patients are broadly characterized by difficulty falling and staying asleep at night. Some of these patients sleep less than 4 hours a night and, in most cases, do not make up this lost sleep during the day-leading to the speculation that AS patients may "need" less sleep. Most AS patients also have severely reduced levels of melatonin, a hormone produced by the pineal gland exclusively at night. This nightly pattern of melatonin production is thought to help synchronize internal circadian rhythms and promote nighttime sleep in humans and other diurnal species. It has been proposed that reduced melatonin levels contribute to the sleep problems in AS patients. Indeed, emerging evidence suggests melatonin replacement therapy can improve sleep in many AS patients. However, AS mice show sleep problems that are arguably similar to those in humans despite being on genetic backgrounds that do not make melatonin. This suggests the hypothesis that the change in nighttime melatonin may be a secondary factor rather than the root cause of the sleeping disorder. The goals of this review article are to revisit the sleep and melatonin findings in both AS patients and animal models of AS and discuss what AS may tell us about the underlying mechanisms of, and interplay between, melatonin and sleep.
Collapse
Affiliation(s)
- Daniella Buonfiglio
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Daniel L Hummer
- Department of Psychology, Morehouse College, Atlanta, GA, USA
| | - Ariel Armstrong
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, USA
| | | | - Jason P DeBruyne
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
35
|
An Unbiased Drug Screen for Seizure Suppressors in Duplication 15q Syndrome Reveals 5-HT 1A and Dopamine Pathway Activation as Potential Therapies. Biol Psychiatry 2020; 88:698-709. [PMID: 32507391 PMCID: PMC7554174 DOI: 10.1016/j.biopsych.2020.03.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/06/2020] [Accepted: 04/02/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Duplication 15q (Dup15q) syndrome is a rare neurogenetic disorder characterized by autism and pharmacoresistant epilepsy. Most individuals with isodicentric duplications have been on multiple medications to control seizures. We recently developed a model of Dup15q in Drosophila by elevating levels of fly Dube3a in glial cells using repo-GAL4, not neurons. In contrast to other Dup15q models, these flies develop seizures that worsen with age. METHODS We screened repo>Dube3a flies for approved compounds that can suppress seizures. Flies 3 to 5 days old were exposed to compounds in the fly food during development. Flies were tested using a bang sensitivity assay for seizure recovery time. At least 40 animals were tested per experiment, with separate testing for male and female flies. Studies of K+ content in glial cells of the fly brain were also performed using a fluorescent K+ indicator. RESULTS We identified 17 of 1280 compounds in the Prestwick Chemical Library that could suppress seizures. Eight compounds were validated in secondary screening. Four of these compounds regulated either serotonergic or dopaminergic signaling, and subsequent experiments confirmed that seizure suppression occurred primarily through stimulation of serotonin receptor 5-HT1A. Additional studies of K+ levels showed that Dube3a regulation of the Na+/K+ exchanger ATPα (adenosine triphosphatase α) in glia may be modulated by serotonin/dopamine signaling, causing seizure suppression. CONCLUSIONS Based on these pharmacological and genetic studies, we present an argument for the use of 5-HT1A agonists in the treatment of Dup15q epilepsy.
Collapse
|
36
|
Townsend LB, Jones KA, Dorsett CR, Philpot BD, Smith SL. Deficits in higher visual area representations in a mouse model of Angelman syndrome. J Neurodev Disord 2020; 12:28. [PMID: 33076843 PMCID: PMC7574469 DOI: 10.1186/s11689-020-09329-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 09/11/2020] [Indexed: 11/10/2022] Open
Abstract
Background Sensory processing deficits are common in individuals with neurodevelopmental disorders. One hypothesis is that deficits may be more detectable in downstream, “higher” sensory areas. A mouse model of Angelman syndrome (AS), which lacks expression of the maternally inherited Ube3a allele, has deficits in synaptic function and experience-dependent plasticity in the primary visual cortex. Thus, we hypothesized that AS model mice have deficits in visually driven neuronal responsiveness in downstream higher visual areas (HVAs). Methods Here, we used intrinsic signal optical imaging and two-photon calcium imaging to map visually evoked neuronal activity in the primary visual cortex and HVAs in response to an array of stimuli. Results We found a highly specific deficit in HVAs. Drifting gratings that changed speed caused a strong response in HVAs in wildtype mice, but this was not observed in littermate AS model mice. Further investigation with two-photon calcium imaging revealed the effect to be largely driven by aberrant responses of inhibitory interneurons, suggesting a cellular basis for higher level, stimulus-selective cortical dysfunction in AS. Conclusion Assaying downstream, or “higher” circuitry may provide a more sensitive measure for circuit dysfunction in mouse models of neurodevelopmental disorders. Trial registration Not applicable.
Collapse
Affiliation(s)
- Leah B Townsend
- Neuroscience Curriculum, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Kelly A Jones
- Neuroscience Curriculum, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA.,Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Christopher R Dorsett
- Neuroscience Curriculum, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Benjamin D Philpot
- Neuroscience Curriculum, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA.,Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA.,Carolina Institute for Developmental Disabilities, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA.,Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Spencer L Smith
- Department of Electrical & Computer Engineering, Neuroscience Research Institute, Center for BioEngineering, University of California Santa Barbara, 2002 BioEngineering Building; Mail code 5100, Santa Barbara, CA, 93106-5100, USA.
| |
Collapse
|
37
|
Mitake M, Hirano S, Kishino T. Imprinting analysis by droplet digital PCR coupled with locked nucleic acid TaqMan probes. Epigenetics 2020; 16:729-740. [PMID: 32970510 DOI: 10.1080/15592294.2020.1823160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Imprinted genes are differentially expressed in a parent-of-origin-specific manner. Parental origin of the alleles is discriminated by intragenic DNA polymorphisms. Comparisons of parental allelic expression have been analysed by semiquantitative RT-PCR. Here, we developed a novel quantitative method for allelic expression of the imprinted gene Ube3a, which inactivation and mutations cause Angelman syndrome and predominantly expressed by the maternal allele in neuronal tissues. In this method, cDNA was amplified by droplet digital PCR (ddPCR) coupled with allele-specific locked nucleic acid (LNA) TaqMan probes, which labelled by FAM and HEX were designed to detect the SNPs in the target regions. ddPCR assay demonstrated that the sense transcript of Ube3a was equally expressed from both parental alleles in adult tissues except neuronal tissues, where Ube3a expression from the paternal allele was about 10 to 14% of total Ube3a expression in adult brain, and 20% in spinal cord. The antisense transcript of Ube3a was expressed at 60% to 70% of the sense transcript of Ube3a in adult brain. Changes in the Ube3a transcripts during postnatal brain development were also evaluated by ddPCR. The ddPCR method is far more reliable and simpler to use than semiquantitative PCR to analyse skewed or faint allelic expression of imprinted genes.
Collapse
Affiliation(s)
- Maiko Mitake
- Division of Functional Genomics, Centre for Frontier Life Sciences, Nagasaki University, Nagasaki, Japan
| | - Shiori Hirano
- Division of Functional Genomics, Centre for Frontier Life Sciences, Nagasaki University, Nagasaki, Japan
| | - Tatsuya Kishino
- Division of Functional Genomics, Centre for Frontier Life Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
38
|
Laukoter S, Pauler FM, Beattie R, Amberg N, Hansen AH, Streicher C, Penz T, Bock C, Hippenmeyer S. Cell-Type Specificity of Genomic Imprinting in Cerebral Cortex. Neuron 2020; 107:1160-1179.e9. [PMID: 32707083 PMCID: PMC7523403 DOI: 10.1016/j.neuron.2020.06.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 05/20/2020] [Accepted: 06/24/2020] [Indexed: 12/20/2022]
Abstract
In mammalian genomes, a subset of genes is regulated by genomic imprinting, resulting in silencing of one parental allele. Imprinting is essential for cerebral cortex development, but prevalence and functional impact in individual cells is unclear. Here, we determined allelic expression in cortical cell types and established a quantitative platform to interrogate imprinting in single cells. We created cells with uniparental chromosome disomy (UPD) containing two copies of either the maternal or the paternal chromosome; hence, imprinted genes will be 2-fold overexpressed or not expressed. By genetic labeling of UPD, we determined cellular phenotypes and transcriptional responses to deregulated imprinted gene expression at unprecedented single-cell resolution. We discovered an unexpected degree of cell-type specificity and a novel function of imprinting in the regulation of cortical astrocyte survival. More generally, our results suggest functional relevance of imprinted gene expression in glial astrocyte lineage and thus for generating cortical cell-type diversity.
Collapse
Affiliation(s)
- Susanne Laukoter
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Florian M Pauler
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Robert Beattie
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Nicole Amberg
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Andi H Hansen
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Carmen Streicher
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Thomas Penz
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Simon Hippenmeyer
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria.
| |
Collapse
|
39
|
Sonzogni M, Zhai P, Mientjes EJ, van Woerden GM, Elgersma Y. Assessing the requirements of prenatal UBE3A expression for rescue of behavioral phenotypes in a mouse model for Angelman syndrome. Mol Autism 2020; 11:70. [PMID: 32948244 PMCID: PMC7501605 DOI: 10.1186/s13229-020-00376-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/04/2020] [Indexed: 11/26/2022] Open
Abstract
Background Angelman syndrome (AS) is a rare neurodevelopmental disorder caused by the loss of functional ubiquitin protein ligase E3A (UBE3A). In neurons, UBE3A expression is tightly regulated by a mechanism of imprinting which suppresses the expression of the paternal UBE3A allele. Promising treatment strategies for AS are directed at activating paternal UBE3A gene expression. However, for such strategies to be successful, it is important to know when such a treatment should start, and how much UBE3A expression is needed for normal embryonic brain development. Methods Using a conditional mouse model of AS, we further delineated the critical period for UBE3A expression during early brain development. Ube3a gene expression was induced around the second week of gestation and mouse phenotypes were assessed using a behavioral test battery. To investigate the requirements of embryonic UBE3A expression, we made use of mice in which the paternal Ube3a allele was deleted. Results We observed a full behavioral rescue of the AS mouse model phenotypes when Ube3a gene reactivation was induced around the start of the last week of mouse embryonic development. We found that full silencing of the paternal Ube3a allele was not completed till the first week after birth but that deletion of the paternal Ube3a allele had no significant effect on the assessed phenotypes. Limitations Direct translation to human is limited, as we do not precisely know how human and mouse brain development aligns over gestational time. Moreover, many of the assessed phenotypes have limited translational value, as the underlying brain regions involved in these tasks are largely unknown. Conclusions Our findings provide further important insights in the requirement of UBE3A expression during brain development. We found that loss of up to 50% of UBE3A protein during prenatal mouse brain development does not significantly impact the assessed mouse behavioral phenotypes. Together with previous findings, our results indicate that the most critical function for mouse UBE3A lies in the early postnatal period between birth and P21.
Collapse
Affiliation(s)
- Monica Sonzogni
- Department of Neuroscience and the ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Peipei Zhai
- Department of Neuroscience and the ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, 3015 CN, Rotterdam, The Netherlands.,Department of Neurology, The First Affiliated Hospital of Henan University, No.357, Ximendajie Street, Kaifeng City, Henan Province, China
| | - Edwin J Mientjes
- Department of Neuroscience and the ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Geeske M van Woerden
- Department of Neuroscience and the ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Ype Elgersma
- Department of Neuroscience and the ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, 3015 CN, Rotterdam, The Netherlands.
| |
Collapse
|
40
|
Human Cerebral Organoids Reveal Early Spatiotemporal Dynamics and Pharmacological Responses of UBE3A. Stem Cell Reports 2020; 15:845-854. [PMID: 32916124 PMCID: PMC7561513 DOI: 10.1016/j.stemcr.2020.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/22/2022] Open
Abstract
Angelman syndrome is a complex neurodevelopmental disorder characterized by delayed development, intellectual disability, speech impairment, and ataxia. It results from the loss of UBE3A protein, an E3 ubiquitin ligase, in neurons of the brain. Despite the dynamic spatiotemporal expression of UBE3A observed in rodents and the potential clinical importance of when and where it is expressed, its expression pattern in humans remains unknown. This reflects a common challenge of studying human neurodevelopment: prenatal periods are hard to access experimentally. In this work, human cerebral organoids reveal a change from weak to strong UBE3A in neuronal nuclei within 3 weeks of culture. Angelman syndrome human induced pluripotent stem cell-derived organoids also exhibit early silencing of paternal UBE3A, with topoisomerase inhibitors partially rescuing UBE3A levels and calcium transient phenotypes. This work establishes human cerebral organoids as an important model for studying UBE3A and motivates their broader use in understanding complex neurodevelopmental disorders. UBE3A signals in neuronal nuclei in hCOs correlate to early stages of development UBE3A exhibits a change from weakly to strongly nuclear in cortical layers UBE3A is imprinted and aberrantly localized in Angelman syndrome hCOs Topoisomerase inhibitors partially rescue UBE3A and neuronal function in AS hCOs
Collapse
|
41
|
Javed S, Selliah T, Lee YJ, Huang WH. Dosage-sensitive genes in autism spectrum disorders: From neurobiology to therapy. Neurosci Biobehav Rev 2020; 118:538-567. [PMID: 32858083 DOI: 10.1016/j.neubiorev.2020.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/26/2020] [Accepted: 08/17/2020] [Indexed: 12/24/2022]
Abstract
Autism spectrum disorders (ASDs) are a group of heterogenous neurodevelopmental disorders affecting 1 in 59 children. Syndromic ASDs are commonly associated with chromosomal rearrangements or dosage imbalance involving a single gene. Many of these genes are dosage-sensitive and regulate transcription, protein homeostasis, and synaptic function in the brain. Despite vastly different molecular perturbations, syndromic ASDs share core symptoms including social dysfunction and repetitive behavior. However, each ASD subtype has a unique pathogenic mechanism and combination of comorbidities that require individual attention. We have learned a great deal about how these dosage-sensitive genes control brain development and behaviors from genetically-engineered mice. Here we describe the clinical features of eight monogenic neurodevelopmental disorders caused by dosage imbalance of four genes, as well as recent advances in using genetic mouse models to understand their pathogenic mechanisms and develop intervention strategies. We propose that applying newly developed quantitative molecular and neuroscience technologies will advance our understanding of the unique neurobiology of each disorder and enable the development of personalized therapy.
Collapse
Affiliation(s)
- Sehrish Javed
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Tharushan Selliah
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Yu-Ju Lee
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Wei-Hsiang Huang
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.
| |
Collapse
|
42
|
Lopez SJ, Laufer BI, Beitnere U, Berg EL, Silverman JL, O'Geen H, Segal DJ, LaSalle JM. Imprinting effects of UBE3A loss on synaptic gene networks and Wnt signaling pathways. Hum Mol Genet 2020; 28:3842-3852. [PMID: 31625566 DOI: 10.1093/hmg/ddz221] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/21/2019] [Accepted: 09/04/2019] [Indexed: 12/13/2022] Open
Abstract
Ubiquitin E3 ligase 3A (UBE3A) encodes an E3 ubiquitin ligase whose loss from the maternal allele causes the neurodevelopmental disorder Angelman syndrome (AS). Previous studies of UBE3A function have not examined full Ube3a deletion in mouse, the complexity of imprinted gene networks in brain nor the molecular basis of systems-level cognitive dysfunctions in AS. We therefore utilized a systems biology approach to elucidate how UBE3A loss impacts the early postnatal brain in a novel CRISPR/Cas9-engineered rat Angelman model of a complete Ube3a deletion. Strand-specific transcriptome analysis of offspring from maternally or paternally inherited Ube3a deletions revealed the expected parental expression patterns of Ube3a sense and antisense transcripts by postnatal day 2 (P2) in hypothalamus and day 9 (P9) in cortex, compared to wild-type littermates. The dependency of genome-wide effects on parent-of-origin, Ube3a genotype and time (P2 and P9) was investigated through transcriptome (RNA sequencing of cortex and hypothalamus) and methylome (whole-genome bisulfite sequencing of hypothalamus). Weighted gene co-expression and co-methylation network analyses identified co-regulated networks in maternally inherited Ube3a deletion offspring enriched in postnatal developmental processes including Wnt signaling, synaptic regulation, neuronal and glial functions, epigenetic regulation, ubiquitin, circadian entrainment and splicing. Furthermore, we showed that loss of the paternal Ube3a antisense transcript resulted in both unique and overlapping dysregulated gene pathways with maternal loss, predominantly at the level of differential methylation. Together, these results provide a holistic examination of the molecular impacts of UBE3A loss in brain, supporting the existence of interactive epigenetic networks between maternal and paternal transcripts at the Ube3a locus.
Collapse
Affiliation(s)
- S Jesse Lopez
- Medical Immunology and Microbiology, University of California (UC) Davis School of Medicine, Davis, CA 95616, USA.,Genome Center, UC Davis, Davis, CA, USA.,Integrative Genetics and Genomics, UC Davis, Davis, CA 95616, USA.,Biochemistry and Molecular Medicine, UC Davis School of Medicine, Davis, CA 95616, USA.,Medical Investigation of Neurodevelopmental Disorders Institute, UC Davis Health, Sacramento, CA 95817, USA
| | - Benjamin I Laufer
- Medical Immunology and Microbiology, University of California (UC) Davis School of Medicine, Davis, CA 95616, USA.,Genome Center, UC Davis, Davis, CA, USA.,Medical Investigation of Neurodevelopmental Disorders Institute, UC Davis Health, Sacramento, CA 95817, USA
| | - Ulrika Beitnere
- Genome Center, UC Davis, Davis, CA, USA.,Biochemistry and Molecular Medicine, UC Davis School of Medicine, Davis, CA 95616, USA.,Medical Investigation of Neurodevelopmental Disorders Institute, UC Davis Health, Sacramento, CA 95817, USA
| | - Elizabeth L Berg
- Medical Investigation of Neurodevelopmental Disorders Institute, UC Davis Health, Sacramento, CA 95817, USA.,Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacromento, CA 95817, USA
| | - Jill L Silverman
- Medical Investigation of Neurodevelopmental Disorders Institute, UC Davis Health, Sacramento, CA 95817, USA.,Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacromento, CA 95817, USA
| | - Henriette O'Geen
- Genome Center, UC Davis, Davis, CA, USA.,Biochemistry and Molecular Medicine, UC Davis School of Medicine, Davis, CA 95616, USA.,Medical Investigation of Neurodevelopmental Disorders Institute, UC Davis Health, Sacramento, CA 95817, USA
| | - David J Segal
- Genome Center, UC Davis, Davis, CA, USA.,Integrative Genetics and Genomics, UC Davis, Davis, CA 95616, USA.,Biochemistry and Molecular Medicine, UC Davis School of Medicine, Davis, CA 95616, USA.,Medical Investigation of Neurodevelopmental Disorders Institute, UC Davis Health, Sacramento, CA 95817, USA
| | - Janine M LaSalle
- Medical Immunology and Microbiology, University of California (UC) Davis School of Medicine, Davis, CA 95616, USA.,Genome Center, UC Davis, Davis, CA, USA.,Integrative Genetics and Genomics, UC Davis, Davis, CA 95616, USA.,Medical Investigation of Neurodevelopmental Disorders Institute, UC Davis Health, Sacramento, CA 95817, USA
| |
Collapse
|
43
|
Transcriptomic and proteomic profiling of glial versus neuronal Dube3a overexpression reveals common molecular changes in gliopathic epilepsies. Neurobiol Dis 2020; 141:104879. [PMID: 32344153 DOI: 10.1016/j.nbd.2020.104879] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 04/04/2020] [Accepted: 04/23/2020] [Indexed: 01/05/2023] Open
Abstract
Epilepsy affects millions of individuals worldwide and many cases are pharmacoresistant. Duplication 15q syndrome (Dup15q) is a genetic disorder caused by duplications of the 15q11.2-q13.1 region. Phenotypes include a high rate of pharmacoresistant epilepsy. We developed a Dup15q model in Drosophila melanogaster that recapitulates seizures in Dup15q by over-expressing fly Dube3a or human UBE3A in glial cells, but not neurons, implicating glia in the Dup15q epilepsy phenotype. We compared Dube3a overexpression in glia (repo>Dube3a) versus neurons (elav>Dube3a) using transcriptomics and proteomics of whole fly head extracts. We identified 851 transcripts differentially regulated in repo>Dube3a, including an upregulation of glutathione S-transferase (GST) genes that occurred cell autonomously within glial cells. We reliably measured approximately 2,500 proteins by proteomics, most of which were also quantified at the transcript level. Combined transcriptomic and proteomic analysis revealed an enrichment of 21 synaptic transmission genes downregulated at the transcript and protein in repo>Dube3a indicating synaptic proteins change in a cell non-autonomous manner in repo>Dube3a flies. We identified 6 additional glia originating bang-sensitive seizure lines and found upregulation of GSTs in 4 out of these 6 lines. These data suggest GST upregulation is common among gliopathic seizures and may ultimately provide insight for treating epilepsy.
Collapse
|
44
|
Rotaru DC, Mientjes EJ, Elgersma Y. Angelman Syndrome: From Mouse Models to Therapy. Neuroscience 2020; 445:172-189. [PMID: 32088294 DOI: 10.1016/j.neuroscience.2020.02.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/19/2022]
Abstract
The UBE3A gene is part of the chromosome 15q11-q13 region that is frequently deleted or duplicated, leading to several neurodevelopmental disorders (NDD). Angelman syndrome (AS) is caused by the absence of functional maternally derived UBE3A protein, while the paternal UBE3A gene is present but silenced specifically in neurons. Patients with AS present with severe neurodevelopmental delay, with pronounced motor deficits, absence of speech, intellectual disability, epilepsy, and sleep problems. The pathophysiology of AS is still unclear and a treatment is lacking. Animal models of AS recapitulate the genotypic and phenotypic features observed in AS patients, and have been invaluable for understanding the disease process as well as identifying apropriate drug targets. Using these AS mouse models we have learned that loss of UBE3A probably affects many areas of the brain, leading to increased neuronal excitability and a loss of synaptic spines, along with changes in a number of distinct behaviours. Inducible AS mouse models have helped to identify the critical treatment windows for the behavioral and physiological phenotypes. Additionally, AS mouse models indicate an important role for the predominantly nuclear UBE3A isoform in generating the characteristic AS pathology. Last, but not least, the AS mice have been crucial in guiding Ube3a gene reactivation treatments, which present a very promising therapy to treat AS.
Collapse
Affiliation(s)
- Diana C Rotaru
- Department of Neuroscience, The ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Edwin J Mientjes
- Department of Neuroscience, The ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ype Elgersma
- Department of Neuroscience, The ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
45
|
Furumai R, Tamada K, Liu X, Takumi T. UBE3A regulates the transcription of IRF, an antiviral immunity. Hum Mol Genet 2020; 28:1947-1958. [PMID: 30690483 DOI: 10.1093/hmg/ddz019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/10/2019] [Accepted: 01/14/2019] [Indexed: 12/12/2022] Open
Abstract
UBE3A is a gene responsible for the pathogenesis of Angelman syndrome (AS), a neurodevelopmental disorder characterized by symptoms such as intellectual disability, delayed development and severe speech impairment. UBE3A encodes an E3 ubiquitin ligase, for which several targets have been identified, including synaptic molecules. Although proteolysis mainly occurs in the cytoplasm, UBE3A is localized to the cytoplasm and the nucleus. In fact, UBE3A is also known as a transcriptional regulator of the family of nuclear receptors. However, the function of UBE3A in the nucleus remains unclear. Therefore, we examined the involvement of UBE3A in transcription in the nuclei of neurons. Genome-wide transcriptome analysis revealed an enrichment of genes downstream of interferon regulatory factor (IRF) in a UBE3A-deficient AS mouse model. In vitro biochemical analyses further demonstrated that UBE3A interacted with IRF and, more importantly, that UBE3A enhanced IRF-dependent transcription. These results suggest a function for UBE3A as a transcriptional regulator of the immune system in the brain. These findings also provide informative molecular insights into the function of UBE3A in the brain and in AS pathogenesis.
Collapse
Affiliation(s)
- Ryohei Furumai
- RIKEN Brain Science Institute, Wako, Saitama, Japan.,Graduate School of Biomedical Sciences, Hiroshima University, Minami, Hiroshima, Japan
| | - Kota Tamada
- RIKEN Brain Science Institute, Wako, Saitama, Japan.,Graduate School of Biomedical Sciences, Hiroshima University, Minami, Hiroshima, Japan
| | - Xiaoxi Liu
- RIKEN Brain Science Institute, Wako, Saitama, Japan
| | - Toru Takumi
- RIKEN Brain Science Institute, Wako, Saitama, Japan.,Graduate School of Biomedical Sciences, Hiroshima University, Minami, Hiroshima, Japan
| |
Collapse
|
46
|
Yang X. Towards an understanding of Angelman syndrome in mice studies. J Neurosci Res 2019; 98:1162-1173. [PMID: 31867793 DOI: 10.1002/jnr.24576] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/13/2022]
Abstract
Angelman syndrome (AS) is a rare neurodevelopmental disorder characterized by severe mental retardation, absence of speech, abnormal motor coordination, abnormal EEG, and spontaneous seizure. AS is caused by a deficiency in the ubiquitin ligase E3A (Ube3a) gene product, known to play a dual role as both ubiquitin ligase and transcription coactivator. In AS animal models, multiple Ube3a substrates are accumulated in neurons. So far, studies in mouse models have either aimed at re-expressing Ube3a or manipulating downstream signaling pathways. Reintroducing Ube3a in AS mice showed promising results but may have two caveats. First, it may cause an overdosage in the Ube3a expression, which in turn is known to contribute to autism spectrum disorders. Second, in mutation cases, the exogenous Ube3a may have to compete with the mutated endogenous form. Such two caveats left spaces for developing therapies or interventions directed to targets downstream Ube3a. Notably, Ube3a expression is dynamically regulated by neuronal activity and plays a crucial role in synaptic plasticity. The abnormal synaptic plasticity uncovered in AS mice has been frequently rescued, but circuits symptoms like seizure are resistant to treatment. Future investigations are needed to further clarify the function (s) of Ube3a during development. Here I reviewed the recently identified major Ube3a substrates and signaling pathways involved in AS pathology, the Ube3a expression, imprinting and evolution, the AS mouse models that have been generated and inspired therapeutic potentials, and finally proposed some future explorations to better understand the AS pathology.
Collapse
Affiliation(s)
- Xin Yang
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
47
|
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
|
48
|
Frohlich J, Reiter LT, Saravanapandian V, DiStefano C, Huberty S, Hyde C, Chamberlain S, Bearden CE, Golshani P, Irimia A, Olsen RW, Hipp JF, Jeste SS. Mechanisms underlying the EEG biomarker in Dup15q syndrome. Mol Autism 2019; 10:29. [PMID: 31312421 PMCID: PMC6609401 DOI: 10.1186/s13229-019-0280-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 06/11/2019] [Indexed: 12/11/2022] Open
Abstract
Background Duplications of 15q11.2-q13.1 (Dup15q syndrome), including the paternally imprinted gene UBE3A and three nonimprinted gamma-aminobutyric acid type-A (GABAA) receptor genes, are highly penetrant for neurodevelopmental disorders such as autism spectrum disorder (ASD). To guide targeted treatments of Dup15q syndrome and other forms of ASD, biomarkers are needed that reflect molecular mechanisms of pathology. We recently described a beta EEG phenotype of Dup15q syndrome, but it remains unknown which specific genes drive this phenotype. Methods To test the hypothesis that UBE3A overexpression is not necessary for the beta EEG phenotype, we compared EEG from a reference cohort of children with Dup15q syndrome (n = 27) to (1) the pharmacological effects of the GABAA modulator midazolam (n = 12) on EEG from healthy adults, (2) EEG from typically developing (TD) children (n = 14), and (3) EEG from two children with duplications of paternal 15q (i.e., the UBE3A-silenced allele). Results Peak beta power was significantly increased in the reference cohort relative to TD controls. Midazolam administration recapitulated the beta EEG phenotype in healthy adults with a similar peak frequency in central channels (f = 23.0 Hz) as Dup15q syndrome (f = 23.1 Hz). Both paternal Dup15q syndrome cases displayed beta power comparable to the reference cohort. Conclusions Our results suggest a critical role for GABAergic transmission in the Dup15q syndrome beta EEG phenotype, which cannot be explained by UBE3A dysfunction alone. If this mechanism is confirmed, the phenotype may be used as a marker of GABAergic pathology in clinical trials for Dup15q syndrome.
Collapse
Affiliation(s)
- Joel Frohlich
- Roche Pharma Research and Early Development, Neuroscience, Ophthalmology and Rare Diseases, Roche Innovation Center Basel, Basel, Switzerland
- Center for Autism Research and Treatment, University of California Los Angeles, Semel Institute for Neuroscience, Los Angeles, CA 90024 USA
- Department of Psychology, University of California Los Angeles, 3423 Franz Hall, Los Angeles, CA 90095 USA
| | - Lawrence T. Reiter
- Departments of Neurology, Pediatrics and Anatomy & Neurobiology, The University of Tennessee Health Science Center, 855 Monroe Ave., Link, Memphis, TN 415 USA
| | - Vidya Saravanapandian
- Center for Autism Research and Treatment, University of California Los Angeles, Semel Institute for Neuroscience, Los Angeles, CA 90024 USA
| | - Charlotte DiStefano
- Center for Autism Research and Treatment, University of California Los Angeles, Semel Institute for Neuroscience, Los Angeles, CA 90024 USA
| | - Scott Huberty
- Center for Autism Research and Treatment, University of California Los Angeles, Semel Institute for Neuroscience, Los Angeles, CA 90024 USA
- McGill University, MUHC Research Institute, 5252, boul. de Maisonneuve Ouest, 3E.19, Montreal, QC H4A 3S5 Canada
| | - Carly Hyde
- Center for Autism Research and Treatment, University of California Los Angeles, Semel Institute for Neuroscience, Los Angeles, CA 90024 USA
| | - Stormy Chamberlain
- Genetics and Genome Sciences, UConn Health, 400 Farmington Avenue, Farmington, CT 06030-6403 USA
| | - Carrie E. Bearden
- Department of Psychiatry and Biobehavioral Sciences and Department of Psychology, University of California Los Angeles, Suite A7-460, 760 Westwood Plaza, Los Angeles, CA 90095 USA
| | - Peyman Golshani
- Department of Neurology and Psychiatry, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA 90095 USA
| | - Andrei Irimia
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave., Suite 228C, California, Los Angeles 90089 USA
| | - Richard W. Olsen
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, California, Los Angeles 90095 USA
| | - Joerg F. Hipp
- Roche Pharma Research and Early Development, Neuroscience, Ophthalmology and Rare Diseases, Roche Innovation Center Basel, Basel, Switzerland
| | - Shafali S. Jeste
- Center for Autism Research and Treatment, University of California Los Angeles, Semel Institute for Neuroscience, Los Angeles, CA 90024 USA
| |
Collapse
|
49
|
Khatri N, Man HY. The Autism and Angelman Syndrome Protein Ube3A/E6AP: The Gene, E3 Ligase Ubiquitination Targets and Neurobiological Functions. Front Mol Neurosci 2019; 12:109. [PMID: 31114479 PMCID: PMC6502993 DOI: 10.3389/fnmol.2019.00109] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 04/12/2019] [Indexed: 12/18/2022] Open
Abstract
UBE3A is a gene implicated in neurodevelopmental disorders. The protein product of UBE3A is the E3 ligase E6-associated protein (E6AP), and its expression in the brain is uniquely regulated via genetic imprinting. Loss of E6AP expression leads to the development of Angelman syndrome (AS), clinically characterized by lack of speech, abnormal motor development, and the presence of seizures. Conversely, copy number variations (CNVs) that result in the overexpression of E6AP are strongly associated with the development of autism spectrum disorders (ASDs), defined by decreased communication, impaired social interest, and increased repetitive behavior. In this review article, we focus on the neurobiological function of Ube3A/E6AP. As an E3 ligase, many functional target proteins of E6AP have been discovered, including p53, Arc, Ephexin5, and SK2. On a neuronal level, E6AP is widely expressed within the cell, including dendritic arbors, spines, and the nucleus. E6AP regulates neuronal morphological maturation and plays an important role in synaptic plasticity and cortical development. These molecular findings provide insight into our understanding of the molecular events underlying AS and ASDs.
Collapse
Affiliation(s)
- Natasha Khatri
- Department of Biology, Boston University, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
50
|
Lee HJ, Choi NY, Lee SW, Lee Y, Ko K, Kim GJ, Hwang HS, Ko K. Alteration of Genomic Imprinting Status of Human Parthenogenetic Induced Pluripotent Stem Cells during Neural Lineage Differentiation. Int J Stem Cells 2019; 12:31-42. [PMID: 30836722 PMCID: PMC6457707 DOI: 10.15283/ijsc18084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 11/19/2018] [Accepted: 12/29/2018] [Indexed: 12/16/2022] Open
Abstract
Background and Objectives Genomic imprinting modulates growth and development in mammals and is associated with genetic disorders. Although uniparental embryonic stem cells have been used to study genomic imprinting, there is an ethical issue associated with the destruction of human embryos. In this study, to investigate the genomic imprinting status in human neurodevelopment, we used human uniparental induced pluripotent stem cells (iPSCs) that possessed only maternal alleles and differentiated into neural cell lineages. Methods Human somatic iPSCs (hSiPSCs) and human parthenogenetic iPSCs (hPgiPSCs) were differentiated into neural stem cells (NSCs) and named hSi-NSCs and hPgi-NSCs respectively. DNA methylation and gene expression of imprinted genes related neurodevelopment was analyzed during reprogramming and neural lineage differentiation. Results The DNA methylation and expression of imprinted genes were altered or maintained after differentiation into NSCs. The imprinting status in NSCs were maintained after terminal differentiation into neurons and astrocytes. In contrast, gene expression was differentially presented in a cell type-specific manner. Conclusions This study suggests that genomic imprinting should be determined in each neural cell type because the genomic imprinting status can differ in a cell type-specific manner. In addition, the in vitro model established in this study would be useful for verifying the epigenetic alteration of imprinted genes which can be differentially changed during neurodevelopment in human and for screening novel imprinted genes related to neurodevelopment. Moreover, the confirmed genomic imprinting status could be used to find out an abnormal genomic imprinting status of imprinted genes related with neurogenetic disorders according to uniparental genotypes.
Collapse
Affiliation(s)
- Hye Jeong Lee
- Departement of Stem Cell Biology, Konkuk University School of Medicine, Seoul, Korea.,Center for Stem Cell Research, Institute of Advanced Biomedical Science, Konkuk University, Seoul, Korea
| | - Na Young Choi
- Departement of Stem Cell Biology, Konkuk University School of Medicine, Seoul, Korea.,Center for Stem Cell Research, Institute of Advanced Biomedical Science, Konkuk University, Seoul, Korea
| | - Seung-Wong Lee
- Departement of Stem Cell Biology, Konkuk University School of Medicine, Seoul, Korea.,Center for Stem Cell Research, Institute of Advanced Biomedical Science, Konkuk University, Seoul, Korea
| | - Yukyeong Lee
- Departement of Stem Cell Biology, Konkuk University School of Medicine, Seoul, Korea.,Center for Stem Cell Research, Institute of Advanced Biomedical Science, Konkuk University, Seoul, Korea
| | - Kisung Ko
- Departments of Medicine, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Gwang Jun Kim
- Departments of Obstetrics and Gynecology, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Han Sung Hwang
- Department of Obstetrics and Gynecology, Konkuk University School of Medicine, Seoul, Korea
| | - Kinarm Ko
- Departement of Stem Cell Biology, Konkuk University School of Medicine, Seoul, Korea.,Center for Stem Cell Research, Institute of Advanced Biomedical Science, Konkuk University, Seoul, Korea.,Research Institute of Medical Science, Konkuk University, Seoul, Korea
| |
Collapse
|