Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Science ; 381(6663): 1197-1205, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37708280

ABSTRACT

Inactivation of the ubiquitin ligase Ube3a causes the developmental disorder Angelman syndrome, whereas increased Ube3a dosage is associated with autism spectrum disorders. Despite the enriched localization of Ube3a in the axon terminals including presynapses, little is known about the presynaptic function of Ube3a and mechanisms underlying its presynaptic localization. We show that developmental synapse elimination requires presynaptic Ube3a activity in Drosophila neurons. We further identified the domain of Ube3a that is required for its interaction with the kinesin motor. Angelman syndrome-associated missense mutations in the interaction domain attenuate presynaptic targeting of Ube3a and prevent synapse elimination. Conversely, increased Ube3a activity in presynapses leads to precocious synapse elimination and impairs synaptic transmission. Our findings reveal the physiological role of Ube3a and suggest potential pathogenic mechanisms associated with Ube3a dysregulation.


Subject(s)
Angelman Syndrome , Autism Spectrum Disorder , Drosophila Proteins , Drosophila melanogaster , Synaptic Transmission , Ubiquitin-Protein Ligases , Animals , Angelman Syndrome/enzymology , Angelman Syndrome/genetics , Autism Spectrum Disorder/enzymology , Autism Spectrum Disorder/genetics , Down-Regulation , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Synapses/enzymology , Synapses/genetics
2.
J Biol Chem ; 293(47): 18400-18401, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30470726

ABSTRACT

Loss-of-function mutations in the UBE3A ubiquitin ligase are associated with Angelman syndrome (AS), a severe neurologic disorder. A new study defines the role of mutations in an N-terminal "AZUL" domain as mediating direct binding to a proteasomal subunit and shows that this interaction is correlated with the ability of UBE3A to promote Wnt/ß-catenin signaling. These results provide new insights into a central biomolecule in AS and suggest that defects in Wnt/ß-catenin signaling may underlie some AS phenotypes.


Subject(s)
Angelman Syndrome/enzymology , Point Mutation , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Angelman Syndrome/genetics , Humans , Proteasome Endopeptidase Complex/genetics , RNA-Binding Proteins , Ubiquitin-Protein Ligases/chemistry , Zinc/metabolism
3.
J Biol Chem ; 293(47): 18387-18399, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30257870

ABSTRACT

Deregulation of the HECT ubiquitin ligase UBE3A/E6AP has been implicated in Angelman syndrome as well as autism spectrum disorders. We and others have previously identified the 26S proteasome as one of the major UBE3A-interacting protein complexes. Here, we characterize the interaction of UBE3A and the proteasomal subunit PSMD4 (Rpn10/S5a). We map the interaction to the highly conserved Zn2+-binding N-terminal (AZUL) domain of UBE3A, the integrity of which is crucial for binding to PSMD4. Interestingly, two Angelman syndrome point mutations that affect the AZUL domain show an impaired ability to bind PSMD4. Although not affecting the ubiquitin ligase or the estrogen receptor α-mediated transcriptional regulation activities, these AZUL domain mutations prevent UBE3A from stimulating the Wnt/ß-catenin signaling pathway. Taken together, our data indicate that impaired binding to the 26S proteasome and consequential deregulation of Wnt/ß-catenin signaling might contribute to the functional defect of these mutants in Angelman syndrome.


Subject(s)
Angelman Syndrome/enzymology , Point Mutation , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Zinc/metabolism , Angelman Syndrome/genetics , Humans , Proteasome Endopeptidase Complex/genetics , RNA-Binding Proteins , Ubiquitin-Protein Ligases/metabolism , Wnt Signaling Pathway
4.
Neurobiol Dis ; 105: 99-108, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28576709

ABSTRACT

Angelman syndrome (AS) is a neurodevelopmental disorder characterized by severe intellectual and developmental disabilities. The disease is caused by the loss of function of maternally inherited UBE3A, a gene that exhibits paternal-specific imprinting in neuronal tissues. Ube3a-maternal deficient mice (AS mice) display many classical features of AS, although, the underlying mechanism of these behavioural deficits is poorly understood. Here we report that the absence of Ube3a in AS mice brain caused aberrant increase in HDAC1/2 along with decreased acetylation of histone H3/H4. Partial knockdown of Ube3a in cultured neuronal cells also lead to significant up-regulation of HDAC1/2 and consequent down-regulation of histones H3/H4 acetylation. Treatment of HDAC inhibitor, sodium valproate, to AS mice showed significant improvement in social, cognitive and motor impairment along with restoration of various proteins linked with synaptic function and plasticity. Interestingly, HDAC inhibitor also significantly increased the expression of Ube3a in cultured neuronal cells and in the brain of wild type mice but not in AS mice. These results indicate that anomalous HDAC1/2 activity might be linked with synaptic dysfunction and behavioural deficits in AS mice and suggests that HDAC inhibitors could be potential therapeutic molecule for the treatment of the disease.


Subject(s)
Angelman Syndrome/complications , Angelman Syndrome/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Histone Deacetylases/metabolism , Mental Disorders/etiology , Valproic Acid/pharmacology , Angelman Syndrome/drug therapy , Angelman Syndrome/genetics , Animals , Anxiety/etiology , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Line, Transformed , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Gene Expression Regulation, Enzymologic/genetics , Histone Deacetylases/therapeutic use , Male , Mice , Mice, Transgenic , Neurons/drug effects , Psychomotor Performance/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Valproic Acid/therapeutic use
5.
Cell Death Dis ; 6: e1625, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25633294

ABSTRACT

Angelman syndrome, a severe neurodevelopmental disease, occurs primarily due to genetic defects, which cause lack of expression or mutations in the wild-type E6AP/UBE3A protein. A proportion of the Angelman syndrome patients bear UBE3A point mutations, which do not interfere with the expression of the full-length protein, however, these individuals still develop physiological conditions of the disease. Interestingly, most of these mutations are catalytically defective, thereby indicating the importance of UBE3A enzymatic activity role in the Angelman syndrome pathology. In this study, we show that Angelman syndrome-associated mutants interact strongly with the proteasome via the S5a proteasomal subunit, resulting in an overall inhibitory effect on the proteolytic activity of the proteasome. Our results suggest that mutated catalytically inactive forms of UBE3A may cause defects in overall proteasome function, which could have an important role in the Angelman syndrome pathology.


Subject(s)
Angelman Syndrome/enzymology , Mutant Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Biocatalysis , Gene Knockdown Techniques , HEK293 Cells , Humans , Papillomavirus E7 Proteins/metabolism , Protein Binding , Protein Subunits/metabolism , Ubiquitination
6.
Learn Mem ; 21(2): 98-104, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24434871

ABSTRACT

Angelman Syndrome (AS) is a devastating neurological disorder caused by disruption of the maternal UBE3A gene. Ube3a protein is identified as an E3 ubiquitin ligase that shows neuron-specific imprinting. Despite extensive research evaluating the localization and basal expression profiles of Ube3a in mouse models, the molecular mechanisms whereby Ube3a deficiency results in AS are enigmatic. Using in vitro and in vivo systems we show dramatic changes in the expression of Ube3a following synaptic activation. In primary neuronal culture, neuronal depolarization was found to increase both nuclear and cytoplasmic Ube3a levels. Analogous up-regulation in maternal and paternal Ube3a expression was observed in Ube3a-YFP reporter mice following fear conditioning. Absence of Ube3a led to deficits in the activity-dependent increases in ERK1/2 phosphorylation, which may contribute to reported deficits in synaptic plasticity and cognitive function in AS mice. Taken together, our findings provide novel insight into the regulation of Ube3a by synaptic activity and its potential role in kinase regulation.


Subject(s)
Angelman Syndrome/physiopathology , Brain/physiopathology , Neurons/physiology , Ubiquitin-Protein Ligases/metabolism , Angelman Syndrome/enzymology , Animals , Cell Nucleus/metabolism , Cells, Cultured , Conditioning, Psychological , Cytoplasm/metabolism , Fear/physiology , Female , In Vitro Techniques , MAP Kinase Signaling System/physiology , Male , Membrane Potentials , Mice , Mice, Inbred C57BL , Mice, Transgenic , Parents , Synaptic Transmission , Ubiquitin-Protein Ligases/genetics
7.
Cell Rep ; 4(3): 405-12, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23911285

ABSTRACT

Angelman syndrome (AS) is associated with symptoms that include autism, intellectual disability, motor abnormalities, and epilepsy. We recently showed that AS model mice have increased expression of the alpha1 subunit of Na/K-ATPase (α1-NaKA) in the hippocampus, which was correlated with increased expression of axon initial segment (AIS) proteins. Our developmental analysis revealed that the increase in α1-NaKA expression preceded that of the AIS proteins. Therefore, we hypothesized that α1-NaKA overexpression drives AIS abnormalities and that by reducing its expression these and other phenotypes could be corrected in AS model mice. Herein, we report that the genetic normalization of α1-NaKA levels in AS model mice corrects multiple hippocampal phenotypes, including alterations in the AIS, aberrant intrinsic membrane properties, impaired synaptic plasticity, and memory deficits. These findings strongly suggest that increased expression of α1-NaKA plays an important role in a broad range of abnormalities in the hippocampus of AS model mice.


Subject(s)
Angelman Syndrome/genetics , Angelman Syndrome/pathology , Hippocampus/metabolism , Hippocampus/pathology , Sodium-Potassium-Exchanging ATPase/genetics , Angelman Syndrome/enzymology , Angelman Syndrome/metabolism , Animals , Ankyrins/biosynthesis , Disease Models, Animal , Female , Hippocampus/enzymology , Male , Mice , Mice, Inbred C57BL , NAV1.6 Voltage-Gated Sodium Channel/biosynthesis , Neurons/enzymology , Neurons/metabolism , Neurons/pathology , Protein Subunits , Sodium-Potassium-Exchanging ATPase/metabolism
8.
J Biol Chem ; 286(22): 19410-6, 2011 Jun 03.
Article in English | MEDLINE | ID: mdl-21493713

ABSTRACT

Deregulation of the ubiquitin-protein ligase E6AP contributes to the development of the Angelman syndrome and to cervical carcinogenesis suggesting that the activity of E6AP needs to be under tight control. However, how E6AP activity is regulated at the post-translational level under non-pathologic conditions is poorly understood. In this study, we report that the giant protein HERC2, which is like E6AP a member of the HECT family of ubiquitin-protein ligases, binds to E6AP. The interaction is mediated by the RCC1-like domain 2 of HERC2 and a region spanning amino acid residues 150-200 of E6AP. Furthermore, we provide evidence that HERC2 stimulates the ubiquitin-protein ligase activity of E6AP in vitro and within cells and that this stimulatory effect does not depend on the ubiquitin-protein ligase activity of HERC2. Thus, the data obtained indicate that HERC2 acts as a regulator of E6AP.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Angelman Syndrome/enzymology , Animals , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Mice , Mice, Knockout , Protein Binding , Ubiquitin-Protein Ligases/genetics
9.
Neurosci Lett ; 487(2): 129-33, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-19563863

ABSTRACT

Angelman syndrome (AS) is a severe neurological disorder caused by a deficiency of ubiquitin protein ligase E3A (UBE3A), but the pathophysiology of the disease remains unknown. We now report that in the brains of AS mice in which the maternal UBE3A allele is mutated (m-) and the paternal allele is potentially inactivated by imprinting (p+) (UBE3A m-\p+), the mitochondria are abnormal and exhibit a partial oxidative phosphorylation (OXPHOS) defect. Electron microscopy of the hippocampal region of the UBE3A m-\p+ mice (n=6) reveals small, dense mitochondria with altered cristae, relative to wild-type littermates (n=6) and reduced synaptic vesicle density. The specific activity of OXPHOS complex III is reduced in whole brain mitochondria in UBE3A m-\p+ (n=5) mice versus wild-type littermates (n=5). Therefore, mitochondrial dysfunction may contribute to the pathophysiology of Angelman syndrome.


Subject(s)
Angelman Syndrome/enzymology , CA1 Region, Hippocampal/enzymology , Disease Models, Animal , Mitochondria/enzymology , Neurons/enzymology , Ubiquitin-Protein Ligases/deficiency , Angelman Syndrome/genetics , Angelman Syndrome/pathology , Animals , CA1 Region, Hippocampal/pathology , Female , Genotype , Male , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/genetics , Mitochondria/pathology , Neurons/pathology , Neurons/physiology , Purkinje Cells/enzymology , Purkinje Cells/pathology , Synaptic Vesicles/genetics , Synaptic Vesicles/pathology , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
10.
Hum Mol Genet ; 18(3): 454-62, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-18996915

ABSTRACT

Angelman syndrome is a severe neurodevelopmental disorder mostly caused by loss-of-function mutations in the maternal allele of UBE3A, a gene that encodes an E3 ubiquitin ligase. Drosophila UBE3A (dUBE3A) is highly homologous to human UBE3A (hUBE3A) at the amino acid sequence level, suggesting their functional conservation. We generated dUBE3A-null mutant fly lines and found that dUBE3A is not essential for viability. However, loss of dUBE3A activity reduced dendritic branching of sensory neurons in the peripheral nervous system and slowed the growth of terminal dendritic fine processes. Several lines of evidence indicated that dUBE3A regulates dendritic morphogenesis in a cell autonomous manner. Moreover, overexpression of dUBE3A also decreased dendritic branching, suggesting that the proper level of dUBE3A is critically important for the normal dendritic patterning. These findings suggest that dendritic pathology may contribute to neurological deficits in patients with Angelman syndrome.


Subject(s)
Dendrites/enzymology , Drosophila Proteins/metabolism , Drosophila/enzymology , Ubiquitin-Protein Ligases/metabolism , Angelman Syndrome/enzymology , Angelman Syndrome/genetics , Animals , Dendrites/genetics , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/genetics , Humans , Morphogenesis , Peripheral Nervous System/cytology , Peripheral Nervous System/enzymology , Peripheral Nervous System/growth & development , Sensory Receptor Cells/cytology , Sensory Receptor Cells/enzymology , Ubiquitin-Protein Ligases/genetics
11.
Biochem Soc Trans ; 36(Pt 5): 797-801, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18793139

ABSTRACT

The ubiquitin ligase E6-AP (E6-associated protein) represents a prime example for the notion that deregulated modification of proteins with ubiquitin contributes to the development of human disease: loss of E6-AP function by mutation is responsible for the development of AS (Angelman syndrome), a neurological disorder, and unscheduled activation of E6-AP by complex formation with the E6 oncoprotein of HPVs (human papillomaviruses) contributes to cervical carcinogenesis. However, while there is a considerable amount of data concerning the oncogenic properties of the E6-E6-AP complex, only little is known about the function(s) of E6-AP in neurons. This is mainly due to the fact that although some E6-AP substrates have been identified, it is at present unclear whether deregulated modification/degradation of these proteins is involved in the pathogenesis of AS. Similarly, the cellular pathways involving E6-AP remain enigmatic. To obtain insights into the physiological functions of E6-AP, we are currently employing several strategies, including quantitative affinity proteomics and RNA interference approaches. The results obtained will eventually allow the introduction of E6-AP into functional protein networks and so reveal potential targets for molecular approaches in the treatment of E6-AP-associated diseases.


Subject(s)
Ubiquitin-Protein Ligases/metabolism , Angelman Syndrome/enzymology , Angelman Syndrome/genetics , Animals , DNA-Binding Proteins/metabolism , Enzyme Activation , Female , Humans , Mice , Mice, Knockout , Oncogene Proteins, Viral/metabolism , Ubiquitin-Protein Ligases/genetics , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/genetics
12.
Cell Mol Life Sci ; 65(4): 656-66, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18193166

ABSTRACT

E6-AP is a founding member of HECT (homologous to E6-AP C terminus) domain subfamily of E3 ubiquitin ligases. It degrades tumor suppressor p53 in association with the E6 oncoprotein of the human papilloma virus. However, there are conflicting reports on its role in the degradation of p53 in the absence of E6 oncoprotein. Here, we studied the role of E6-AP in regulation of p53 in mouse neuro 2a cells. Overexpression of E6-AP in neuro 2a cells increased the ubiquitylation and degradation of p53, which could be prevented upon deletion of HECT domain. E6-AP also directly ubiquitylated p53 in an in vitro ubiquitylation assay. Partial knockdown of E6-AP increased the levels of p53 and p53-dependent transcription. Partial knockdown also increased neuronal cell death, which may be mediated partly via p53. Our result suggests that E6-AP not only enhances the degradation of p53 but also regulates the neuronal cell growth.


Subject(s)
Angelman Syndrome/enzymology , Cell Division/drug effects , Intellectual Disability/enzymology , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Angelman Syndrome/genetics , Base Sequence , Cell Line, Tumor , DNA Primers , Gene Expression Regulation , Genes, Reporter , Humans , Intellectual Disability/genetics , Kinetics , Molecular Sequence Data , Neurons/cytology , Plasmids , Transfection , Tumor Suppressor Protein p53/genetics
13.
Hum Mol Genet ; 17(1): 111-8, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17940072

ABSTRACT

Loss of function of the maternally inherited allele for the UBE3A ubiquitin ligase gene causes Angelman syndrome (AS), which is characterized by severe neurological impairment and motor dysfunction. In addition, UBE3A lies within chromosome 15q11-q13 region, where maternal, but not paternal, duplications cause autism. The UBE3A gene product, E6-AP, has been shown to function both as an E3 ligase in the ubiquitin proteasome pathway and as a transcriptional coactivator. However, the specific role of E6-AP in the brain, or how loss of function of E6-AP results in AS, is unclear. Herein, we show, using a recombinant transgenic mouse expressing a Ube3a(YFP) fusion gene, that the maternal Ube3a(YFP) allele is upregulated and preferentially expressed in neurons, and that the fusion protein, E6-AP:YFP, is enriched in the nucleus and dendrites in vivo. We also show that E6-AP:YFP localizes to the nucleus and to presynaptic and postsynaptic compartments in cultured hippocampal neurons. Furthermore, we show that cerebellar Purkinje cell number and dendritic branching are not affected in Ube3a maternal-deficient mice, but that dendritic spine development, including spine morphology, number and length, is affected on cerebellar Purkinje cells and on pyramidal neurons in the hippocampus and cortex. Collectively, these data suggest that the neurological deficits observed in AS patients and in AS mice may result from specific abnormalities in synaptic development and/or plasticity.


Subject(s)
Angelman Syndrome/enzymology , Angelman Syndrome/genetics , Dendritic Spines/enzymology , Dendritic Spines/pathology , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/metabolism , Angelman Syndrome/pathology , Animals , Base Sequence , Cell Nucleus/enzymology , Cells, Cultured , DNA Probes/genetics , Female , Genomic Imprinting , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Purkinje Cells/metabolism , Purkinje Cells/pathology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Synapses/enzymology , Ubiquitin-Protein Ligases/genetics
15.
Curr Opin Pediatr ; 16(4): 419-26, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15273504

ABSTRACT

PURPOSE OF REVIEW: The goal of this review is to provide an overview of rapidly evolving information on a new group of genetic inborn errors affecting ubiquitination and proteasomal degradation of proteins and to suggest a classification scheme for these disorders. The relevant genes encode ubiquitin, ubiquitin enzymes (E1 and many E2s and E3s), deubiquitinating enzymes, proteasomal subunits, and substrates undergoing ubiquitination. RECENT FINDINGS: Since the initial recognition that Angelman syndrome is caused by maternal deficiency of the E6-AP ubiquitin E3 ligase (gene symbol UBE3A), several. other disorders of E3 ligases have been identified, including autosomal recessive juvenile Parkinson disease, the APECED form of autoimmune polyendocrinopathy syndrome, von Hippel-Lindau syndrome, and congenital polycythemia. Disorders that disturb ubiquitin regulatory signaling include at least two subtypes of Fanconi anemia, the BRCA1 and BRCA2 forms of breast and ovarian cancer susceptibility, incontinentia pigmenti, and cylindromatosis. Many disorders affect ubiquitin pathways secondarily. SUMMARY: The authors propose both a genetic and a functional classification for disorders of ubiquitination and proteasomal degradation, as follows. Genetic classes include mutations in (1) the UBB ubiquitin gene; (2) enzymes of ubiquitination including E1, E2, E3, and related proteins; (3) deubiquitinases; (4) proteasomal subunits; and (5) substrates of ubiquitination. Functional classes include defects in (1) proteolytic degradation, (2) ubiquitin signaling, and (3) subcellular localization of substrates. Additional functional classes are likely to be defined, and individual disorders may involve multiple functional defects.


Subject(s)
Genetic Diseases, Inborn/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitins/metabolism , Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Angelman Syndrome/enzymology , Angelman Syndrome/genetics , Angelman Syndrome/metabolism , Animals , Fanconi Anemia/enzymology , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Genetic Diseases, Inborn/enzymology , Genetic Diseases, Inborn/genetics , Genetic Predisposition to Disease , Humans , NF-kappa B/metabolism , Polyendocrinopathies, Autoimmune/enzymology , Polyendocrinopathies, Autoimmune/genetics , Polyendocrinopathies, Autoimmune/metabolism , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , von Hippel-Lindau Disease/enzymology , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/metabolism
16.
J Neurosci ; 23(7): 2634-44, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12684449

ABSTRACT

Angelman syndrome (AS) is a disorder of human cognition characterized by severe mental retardation and epilepsy. Recently, a mouse model for AS (Ube3a maternal null mutation) was developed that displays deficits in both context-dependent learning and hippocampal long-term potentiation (LTP). In the present studies, we examined the molecular basis for these LTP and learning deficits. Mutant animals exhibited a significant increase in hippocampal phospho-calcium/calmodulin-dependent protein kinase II (CaMKII), specifically at sites Thr(286) and Thr(305), with no corresponding change in the levels of total CaMKII. In addition, mutants show a reduction in CaMKII activity, autophosphorylation capability, and total CaMKII associated with postsynaptic density. These findings are the first to implicate misregulation of CaMKII as a molecular cause for the neurobehavioral deficits in a human learning disorder.


Subject(s)
Angelman Syndrome/enzymology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Hippocampus/enzymology , Intellectual Disability/enzymology , Angelman Syndrome/etiology , Angelman Syndrome/physiopathology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/immunology , Cells, Cultured , Hippocampus/physiology , Immunohistochemistry , Intellectual Disability/etiology , Intellectual Disability/physiopathology , Ligases/genetics , Long-Term Potentiation , Mice , Mutation , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Kinases/metabolism , Synapses/enzymology , Threonine/metabolism , Ubiquitin-Protein Ligases
18.
Hum Mutat ; 15(4): 387, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10737998

ABSTRACT

Angelman syndrome is a neurobehavioral disorder caused by defects of imprinted gene(s) on chromosome 15q11-13. AS-specific DNA methylation is found in patients carrying 3-4 Mb deletions ( approximately 70%), paternal uniparental disomy (3-5%) or imprinting center mutations (2-9%), while normal methylation pattern with biparental inheritance characterizes the remaining approximately 20-25% AS patients (Stalker et al.,1998; Tsai et al.,1998). Mutations in the Ubiquitin protein ligase 3A gene (UBE3A) have been found in the latter group, but only preliminary figures are available on their frequencies. We selected a sample of 25 AS patients with a clinical diagnosis of AS and a normal methylation pattern in order to search for mutations of the UBE3A gene. Automated sequencing of exons 8, 9, 10, 11 and 12 performed on our 25 patients allowed us to identify three novel mutations: an 897insA in two unrelated familial cases, a 2544insA and an E167X in two sporadic cases. Mutation R482X previously reported in a sporadic patient was identified in a third familial case. Hum Mutat 15:387, 2000.


Subject(s)
Angelman Syndrome/enzymology , Angelman Syndrome/genetics , Frameshift Mutation/genetics , Ligases/genetics , Mutation, Missense/genetics , Adolescent , Adult , Child , Child, Preschool , DNA Mutational Analysis , Female , Gene Frequency , Humans , Italy , Male , Microsatellite Repeats/genetics , Ubiquitin-Protein Ligases
19.
Hum Mol Genet ; 8(1): 129-35, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9887341

ABSTRACT

Angelman syndrome (AS) is characterized by mental retardation, absence of speech, seizures and motor dysfunction. AS is caused by maternal deletions for chromosome 15q11-q13, paternal uniparental disomy (UPD), imprinting defects or loss-of-function mutations in the UBE3A locus which encodes E6-AP ubiquitin-protein ligase. The UBE3A gene is imprinted with paternal silencing in human brain and similar silencing of the Ube3a locus in Purkinje cells and hippocampal neurons in the mouse. We have sequenced the major coding exons for UBE3A in 56 index patients with a clinical diagnosis of AS and a normal DNA methylation pattern. The analysis identified disease-causing mutations in 17 of 56 patients (30%) including 13 truncating mutations, two missense mutations, one single amino acid deletion and one stop codon mutation predicting an elongated protein. Mutations were identified in six of eight families (75%) with more than one affected case, and in 11 of 47 isolated cases (23%); no mutation was found in one family with two siblings, one with a typical and one with an atypical phenotype. Mutations were de novo in nine of the 11 isolated cases. An amino acid polymorphism of threonine substituted for alanine at codon 178 was identified, and a 3 bp length polymorphism was found in the intron upstream of exon 8. In all informative cases, phenotypic expression was consistent with imprinting with a normal phenotype when a mutation was on the paternal chromosome and an AS phenotype when a mutation was on the maternal chromosome. Laboratory diagnosis and genetic counseling for AS are complex, and mutation analysis is valuable in clinically typical AS patients with a normal methylation analysis.


Subject(s)
Angelman Syndrome/enzymology , Angelman Syndrome/genetics , Ligases/genetics , Mutation , Animals , Base Sequence , DNA Mutational Analysis , DNA Primers/genetics , Exons , Female , Genetic Counseling , Genetic Variation , Genomic Imprinting , Humans , Male , Mice , Pedigree , Phenotype , Ubiquitin-Protein Ligases
20.
Neurogenetics ; 1(4): 229-37, 1998 Aug.
Article in English | MEDLINE | ID: mdl-10732796

ABSTRACT

The clinical features of Angelman syndrome (AS) include microcephaly, severe mental retardation, "puppet-like" ataxic gait with jerky arm movements, hyperactivity, bouts of inappropriate laughter, EEG abnormalities, and seizures. The frequency of occurrence of AS is in the range of 1/10,000 to 1/20,000 births. The AS locus maps to the imprinted chromosome 15q11-q13 region and the disease is caused by the absence of a normal maternal contribution to this region. The genetic complexity of AS is revealed by the existence of at least four molecular classes. A candidate AS gene, ubiquitin protein ligase 3A (UBE3A/E6-AP), has been identified, and mutations of this gene have been detected in several cases of AS. Moreover, UBE3A is expressed predominantly from the maternal allele in brain, strongly supporting its causative role in AS. However, there is evidence to suggest that, in addition to UBE3A, another gene(s) may be involved either directly in AS and/or indirectly by regulating UBE3A expression.


Subject(s)
Angelman Syndrome/enzymology , Angelman Syndrome/genetics , Chromosomes, Human, Pair 15/genetics , Gene Silencing/physiology , Genomic Imprinting/genetics , Ligases/genetics , Alleles , Alternative Splicing , Angelman Syndrome/etiology , Animals , Humans , Isoenzymes/biosynthesis , Isoenzymes/genetics , Ligases/biosynthesis , Mice , Mutation , Organ Specificity , RNA, Messenger/biosynthesis , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL
...