Anda di halaman 1dari 18

American Journal of Medical Genetics Part C (Seminars in Medical Genetics)

R E S E A R C H R E V I E W

Angelman Syndrome: Current and Emerging


Therapies in 2016
WEN-HANN TAN AND LYNNE M. BIRD*

Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by a loss of the maternally-inherited
UBE3A; the paternal UBE3A is silenced in neurons by a mechanism involving an antisense transcript (UBE3A-AS) at
the unmethylated paternal locus. We reviewed all published information on the clinical trials that have been
completed as well as the publicly available information on ongoing trials of therapies in AS. To date, all clinical trials
that strove to improve neurodevelopment in AS have been unsuccessful. Attempts at hypermethylating the
maternal locus through dietary compounds were ineffective. The results of an 8-week open-label trial using
minocycline as a matrix metalloproteinase-9 inhibitor were inconclusive, while a subsequent randomized placebo-
controlled trial suggested that treatment with minocycline for 8 weeks did not result in any neurodevelopmental
gains. A 1-year randomized placebo-controlled trial using levodopa to alter the phosphorylation of calcium/
calmodulin-dependent kinase II did not lead to any improvement in neurodevelopment. Topoisomerase inhibitors
and antisense oligonucleotides are being developed to directly inhibit UBE3A-AS. Artificial transcription factors are
being developed to “super activate” UBE3A or inhibit UBE3A-AS. Other strategies targeting specific pathways are
briefly discussed. We also reviewed the medications that are currently used to treat seizures and sleep disturbances,
which are two of the more common complications of AS. © 2016 Wiley Periodicals, Inc.

KEY WORDS: Angelman syndrome; clinical trial; mouse model; therapy; therapeutic

How to cite this article: Tan W-H, Bird LM. 2016. Angelman syndrome: Current and emerging therapies in
2016. Am J Med Genet Part C Semin Med Genet 9999C:1–18.

INTRODUCTION AND skills being more delayed than their motor younger individuals with the development
BACKGROUND skills, and their expressive language being of peripheral hypertonia in older individ-
far more delayed than their receptive uals, ataxia, epilepsy, as well as resting and
Angelman syndrome (AS) is a rare neuro- language, usually having only minimal intention tremorsin adolescents and adults.
developmental disorder with recent esti- speech. Their behavioral profile is charac- Additional clinical features have been
mates of its prevalence being between terized by a happy demeanor with easily- described in recent reviews, and clinical
1:22,000 and 1:52,000 [Oiglane-Shlik provoked laughter, fascination with water, diagnostic criteria for AS have been
et al., 2006; Mertz et al., 2013; Luk and mouthing behaviors, hyperkinetic behav- proposed [Williams et al., 2006; Thibert
Lo, 2016], although previous studies had iors, and maladaptive behaviors such as et al., 2013; Bird, 2014; Larson et al., 2015].
suggested that the prevalence was about 1 hair-pulling. In addition, most individuals
in 10,000 to 1 in 20,000 [Kyllerman, 1995; with AS go through prolonged periods of
Petersen et al., 1995; Buckley et al., 1998]. sleep disturbance in the form of increased Lack of expression of the
Individuals with AS have global develop- sleep latency and multiple awakenings
mental delay that evolves to severe throughout the night. Other neurologic
maternally-inherited UBE3A
intellectual disability, with their language features include generalized hypotonia in gene in the brain results in

Conflict of interest: Wen-Hann Tan and Lynne M. Bird declare that they have no conflict of interest.
Wen-Hann Tan is a clinical geneticist at Boston Children's Hospital and an assistant professor of pediatrics at Harvard Medical School. He has been
actively involved in a nationwide natural history study and clinical trials in Angelman syndrome since 2006. He also serves on the Angelman Syndrome
Foundation Scientific Advisory Committee.
Lynne M. Bird is a professor of clinical pediatrics at University of California, San Diego, and a clinical geneticist/dysmorphologist at Rady Children's
Hospital San Diego with more than 15 years of experience in the clinical care and research of patients with Angelman syndrome. She served as the
Principal Investigator for the Angelman Syndrome Natural History Study sponsored by the Rare Diseases Clinical Research Network.
This is an updated version of a manuscript entitled “Pharmacological therapies for Angelman syndrome” that was previously published by the same
authors in Wiener Medizinische Wochenschrift 2016 Jan 12 [Epub ahead of print]. It is reproduced in part with permission from Springer.
*Correspondence to: Lynne M. Bird, M.D., Rady Children's Hospital San Diego, Division of Genetics/Dysmorphology, 3020 Children's Way, #5031
San Diego, CA 92123. E-mail: lbird@rchsd.org
DOI 10.1002/ajmg.c.31536
Article first published online in Wiley Online Library (wileyonlinelibrary.com).

ß 2016 Wiley Periodicals, Inc.


2 AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) RESEARCH REVIEW

AS. Unlike most genes, the mice, seven genes were up-regulated forms multiple long non-coding RNAs
paternally-inherited copy of and 57 were down-regulated. Most of (lncRNAs) such as IPW, small nucleolar
these genes have been implicated in RNAs (snoRNAs) such as SNORD116/
UBE3A is silenced (i.e., cellular signaling, regulation of cell HBII-85, and small nucleolar long non-
imprinted) in neurons, so death, neurogenesis, and neurotransmis- coding RNAs (snolncRNAs). The ex-
sion. Among the proteins encoded by pression of these non-coding RNAs may
neurons are entirely dependent these genes are transcription factors for be regulated by the multiple alternative
on UBE3A expressed from the the differentiation of dopaminergic promoters and exons 50 (i.e., “upstream”)
neurons, dopamine receptors, cytoskel- of SNURF-SNRPN exon 1. These
maternal allele. etal proteins, heat shock proteins, and a upstream exons, some of which are
sodium-potassium ATPase transporter differentially methylated between the
[Low and Chen, 2010; Jensen et al., paternal and maternal allele, are used
Lack of expression of the mater- 2013]. However, as noted in a recent primarily in neurons [Dittrich et al.,
nally-inherited UBE3A gene in the review on the putative substrates of 1996; Farber et al., 1999]. The mecha-
brain results in AS. Unlike most genes, UBE3A, there are inter-species differ- nism of action of the AS-IC remains
the paternally-inherited copy of ences in the substrates that interact with unclear. Recent data suggest that in
UBE3A is silenced (i.e., imprinted) in UBE3A, and different substrates may oocytes, a SNURF-SNRPN upstream
neurons, so neurons are entirely depen- interact with UBE3A at different stages exon within the AS-IC initiates (i.e.,
dent on UBE3A expressed from the in development [Sell and Margolis, promotes) transcription of the DNA
maternal allele. Located on chromo- 2015]. Nonetheless, these data suggest sequence into SNRPN exon 2 by
some 15q11.2, UBE3A encodes ubiq- that although AS is due to the loss of a splicing out exon 1. This process results
uitin-protein ligase E3A, which is also single gene, its pathogenesis involves the in DNA methylation at the PWS-IC by
known as E6AP ubiquitin-protein ligase dysregulation of many genes involved means that are still poorly understood,
or human papillomavirus E6-associated in multiple cellular and physiological perhaps through chromatin modification
protein. UBE3A is a member of the pathways. [Lewis et al., 2015]. In most human cell
HECT (Homologous to the E6AP The imprinting of UBE3A is regu- types, the SNURF-SNRPN non-coding
Carboxyl Terminus) E3 ligase family. It lated by a bipartite imprinting center on transcript extends to the IPW gene,
catalyzes the last step in the ubiquitina- chromosome 15q11.2 about one mega- which itself encodes a lncRNA. How-
tion pathway in which activated ubiq- base (Mb) centromeric to UBE3A ever, in neurons, the transcript extends
uitin is transferred from the E2 comprising a centromeric AS imprinting beyond IPW, resulting in the generation
conjugating enzyme and covalently center (AS-IC) and a telomeric Prader- of other lncRNAs, including UBE3A-
ligated to lysine residues of proteins Willi syndrome imprinting center AS (UBE3A anti-sense transcript),
designated for degradation in the pro- (PWS-IC) about 35 kilobases (kb) apart which is also known as SNHG14
teasome. A given protein can be either (Fig. 1) [Buiting et al., 1999]. This (snoRNA host gene 14). UBE3A-AS
monoubiquitinated (i.e., only one ubiq- imprinting center is flanked by NPAP1 represses the expression of paternal
uitin molecule attached to a single lysine (previously known as C15ORF2) on the UBE3A in cis, but the exact mechanism
residue) or polyubiquitinated (i.e., mul- centromeric side and by SNURF- remains unclear [Chamberlain, 2013].
tiple ubiquitin molecules attached to SNRPN on the telomeric side. The Since only the paternally-inherited
multiple lysine residues). In general, PWS-IC extends from the SNURF- SNURF-SNRPN and UBE3A-AS tran-
monoubiquitination regulates intracel- SNRPN promoter through the first scripts are expressed due to the methyl-
lular transport and transcription, exon of SNRPN and includes a differen- ation of the PWS-IC on the maternal
whereas polyubiquitination regulates tially methylated region (DMR) that is allele, only the paternally-inherited
the concentrations of specific intracel- methylated on the maternal allele and UBE3A allele is silenced. In fact, deletion
lular proteins temporally and spatially unmethylated on the paternal allele of the PWS-IC on the paternal allele in a
through proteasome-mediated protein [Dubose et al., 2011]. SNURF-SNRPN mouse model resulted in the re-activa-
degradation [Dagli et al., 2012; Bird, is bicistronic and encodes two over- tion of the paternal Ube3a in neurons
2014; Buiting et al., 2016]. Notwith- lapping transcripts, SNURF (SNRPN [Chamberlain and Brannan, 2001].
standing its role in the ubiquitination of upstream reading frame) and SNRPN There are four mechanisms that can
specific proteins, studies of gene expres- (Small nuclear ribonucleoprotein poly- result in the loss of UBE3A expression
sion in the cerebellum of maternal peptide N); the first three exons of from the maternal allele, viz. deletion on
Ube3a knockout mice and in the heads SNRPN encode SNURF, while the start the maternal copy of chromosome
of Drosophila with Dube3a null muta- codon for the SNRPN protein resides in 15q11.2q13.1 (about 70% of AS indi-
tions have suggested that loss of this the fourth exon [Dubose et al., 2011]. In viduals), paternal uniparental disomy
protein results in either the up-regula- addition, SNURF-SNRPN encodes a (about 9%), imprinting defect due to
tion or down-regulation of specific long non-coding transcript covering either a deletion in the maternal AS-IC
genes; in the maternal Ube3a knockout about 600 kb of genomic DNA that or an epimutation that results in the
RESEARCH REVIEW AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) 3

Figure 1. Genomic region of human chromosome 15q11q13 in neurons: Blue rectangles represent imprinted transcripts that are
paternally expressed, red rectangles represent imprinted transcripts that are maternally expressed, black rectangles represent the repressed
alleles of imprinted genes, and gray rectangles represent transcripts that are not imprinted. Reproduced with permission from:
Chamberlain SJ. RNAs of the human chromosome 15q11-q13 imprinted region. Wiley Interdiscip Rev RNA. 2013;4:155-66.

maternal allele having the “imprint” of of neocortical synapses [Jiang et al., RESULTS
the paternal allele (about 8%), and a 1998; Yashiro et al., 2009].
pathogenic variant in the maternal In addition to treating clinical Activation of Paternal UBE3A
UBE3A allele (about 11%) [Bird, symptoms empirically, attempts at treat-
In theory, the imprinted paternal UBE3A
2014]. Unless the paternal UBE3A allele ing AS have focused primarily on one of
allele may be expressed through one of
harbors a pathogenic variant, the pater- three strategies, viz. (i) activation of the
three mechanisms: (i) Hypermethylation
nal UBE3A allele should remain intact, paternal UBE3A allele that is normally
of the SNURF-SNRPN DMR in the
albeit silenced, regardless of the mecha- silenced, (ii) direct introduction of a
paternally-inherited copy of PWS-IC,
nism by which expression of the mater- functional copy of UBE3A, or (iii)
thereby repressing the expression of the
nal UBE3A allele is lost. correcting specific cellular pathways
SNURF-SNRPN transcript that includes
Although the genetic etiology of that are disrupted by the lack of
UBE3A-AS on the paternal chromosome
AS has been elucidated, the pathogene- UBE3A. Although a few clinical trials
15; (ii) Direct repression of UBE3A-AS by
sis remains poorly understood at the have been conducted as described
interfering with the binding of UBE3A-
cellular level. UBE3A localizes to the below, a cure for AS remains elusive.
AS to the UBE3A DNA bases; or (iii)
nucleus, and to a lesser extent the
Either “super activation” of UBE3A
dendrites, of neurons as well as to the
METHODS promoter or repression of UBE3A-AS
pre- and post-synaptic compartments.
using an artificial transcription factor.
Mice that lacked maternal expression of For this review of therapies in Angelman
Ube3a have reduced dendritic spine syndrome, we searched the PubMed
length and density, suggesting that database (http://www.ncbi.nlm.nih. Hypermethylation of SNURF-SNRPN
UBE3A may be responsible for synaptic gov/pubmed/) using the search terms: DMR
formation at a structural level [Dindot “Angelman AND (therapy OR therapies Studies in rodent models of carcinogen-
et al., 2008]. Moreover, these mice have OR therapeutic OR therapeutics OR esis have suggested that a severely
deficits in long-term potentiation treatment OR treatments).” We also “methyl-deficient diet” resulted in hy-
(LTP), which is a cellular model for searched the ClinicalTrials.gov registry pomethylation of genomic DNA, lead-
memory formation, in hippocampal- (https://www.clinicaltrials.gov/) using ing to the overexpression of specific
dependent associative learning, and in the term “Angelman” to identify un- genes and repression of other genes
the experience-dependent maturation published and ongoing clinical trials. [Dizik et al., 1991]. The major methyl
4 AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) RESEARCH REVIEW

donor for DNA methylation is S- paternal UBE3A by Although the levels of betaine, dime-
adenosylmethionine (SAM), which is hypermethylating the paternal thylglycine, and red blood cell (RBC)
synthesized directly from methionine. folate were higher in the treatment
The methionine-homocysteine cycle SNURF-SNRPN locus group than in the control group at the
that generates SAM is shown in Figure 2. through the administration of end of the 1-year study, there were no
Methionine synthase transfers a methyl statistically significant differences in the
group from 5-methyltetrahydrofolate these “pro-methylation” oral levels of methionine and homocysteine
(5-MTHF) to homocysteine, thereby supplements, have been between the two groups. The develop-
converting homocysteine to methio- mental outcomes as assessed by the
nine, and 5-MTHF to tetrahydrofolate
published. Bayley Scales of Infant Development
(THF). Methylcobalamin, a form of (2nd edition) and the Preschool Lan-
vitamin B12, is the cofactor for methi- guage Scale (3rd edition) were not
onine synthase. In the liver and probably It has been hypothesized that “pro- significantly different between the two
in the kidneys, methionine can also be methylation” dietary supplements such groups either [Peters et al., 2010].
synthesized by the hydrolysis of betaine as folic acid derivatives and betaine In a follow-up clinical trial (Clin-
by betaine-homocysteine methyltrans- would lead to an increase in the icalTrials.gov identifier: NCT00348933),
ferase to form dimethylglycine. The concentration of SAM, which in turn, 65 children with AS aged between
main source of THF is dietary folic acid, would result in a global increase in DNA 5 months and 5 years old completed a
which is converted to THF by dihy- methylation and hence activation of the 1-year open-label trial in which all
drofolate reductase. After SAM has paternal UBE3A allele [Peters et al., subjects received a combination treatment
donated its methyl group, it is converted 2010]. Based on this hypothesis, two comprising L-5-methyltetrahydrofolate,
to S-adenosylhomocysteine (SAH). clinical trials, both with the overall goal vitamin B12, betaine, and creatine. The
SAH inhibits DNA methyltransferases, of activating the paternal UBE3A by rationale for this combination was to try to
which are the enzymes that methylate hypermethylating the paternal SNURF- enhance the availability of SAM by
genomic DNA, and the “SAM/SAH SNRPN locus through the administra- providing the cofactor for methionine
ratio” is termed the “methylation tion of these “pro-methylation” oral synthase (i.e., vitamin B12) and reducing
index” [Van den Veyver, 2002]. supplements, have been published. In the utilization of SAM in the synthesis of
the first study, 48 children with AS aged creatine by supplying exogenous creatine
between 5 months and 14 years old (although SAM is also used for the
Based on this hypothesis, two completed a 1-year double-blind pla- synthesis of many other methylated
cebo-controlled trial of folic acid and compounds). L-5-methyltetrahydrofolate
clinical trials, both with the betaine (n ¼ 28 in placebo group; was used instead of folic acid due to
overall goal of activating the n ¼ 20 in the treatment group). concerns that excess “unmetabolized”

Figure 2. The methionine-homocysteine cycle. The enzymes are in italics and the cofactor is in square brackets. The compounds that
were administered in the two clinical trials involving “pro-methylation” compounds, as described in the text, are shown in the boxes. SAM,
S-adenosylmethionine; SAH, S-adenosylhomocysteine; THF, tetrahydrofolate; 5-MTHF, 5-methyltetrahydrofolate; MTHFR,
methylene tetrahydrofolate reductase.
RESEARCH REVIEW AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) 5

folic acid might be harmful in the long one molecule, irinotecan, that produced Inhibition of topoisomerase results in
term [Bird et al., 2011]. For the analysis of a positive signal. Irinotecan is an the accumulation of R-loops, increased
this open-label trial, the control group was inhibitor of the topoisomerase type I chromatin decondensation (through an
composed of the 22 subjects in the enzyme, a complex that relaxes the unknown mechanism), and inhibition
placebo arm of the previous folic acid- supercoiling of DNA. On further of transcription elongation at the loci
betaine trial described above [Peters et al., screening of topoisomerase inhibitors, where the R-loops have accumulated; it
2010] who were between 15 and the authors identified 15 other topo- remains unknown whether chromatin
59 months old. The only biochemical isomerase type I and II inhibitors that decondensation is a cause or effect of R-
changes in the treatment group that were were also capable of activating paternal loop formation. The consequence of R-
statistically significant were increases in Ube3a in vitro in their cultured neurons. loop stabilization at the paternal
betaine, dimethylglycine, and creatine; In particular, topotecan, a topoisomer- Snord116 locus is the cessation of
there were no statistically significant ase type I inhibitor, which is approved transcription beyond this locus, resulting
changes in the levels of RBC folate, by the Food and Drug Administration in the lack of Ube3a-As and expression of
methionine, and homocysteine. There (FDA) as a chemotherapeutic agent in paternal Ube3a [Powell et al., 2013;
were no detectable changes in the specific cancers, was found to be far Skourti-Stathaki and Proudfoot, 2014].
methylation patterns at 27,578 CpG more potent than irinotecan, which was In cultured human and mouse
dinucleotide sites as measured by the the only other FDA-approved topo- neurons, topoisomerases promote the
Infinium HumanMethylation27 Bead- isomerase inhibitor identified through expression of many genes longer than
Chip array in the whole blood samples this assay. The extent to which top- 67 kb and almost all genes longer than
of 16 subjects who were treated for 1 year otecan was able to promote the expres- 200 kb (i.e., “long genes”), and the
compared to their baseline level of sion of paternal Ube3a in mouse models repression of these genes by topotecan
methylation. Although the daily living when administered through intracere- and a topoisomerase II inhibitor was
skills and motor skills as measured by the bral infusion directly into the lateral both dose-dependent and gene-length-
Vineland Adaptive Behavior Scales (2nd ventricles was dependent on the dose, dependent (longer genes being more
edition) (VABS-II) were better in the with the lower doses resulting in the susceptible to repression than shorter
treatment group compared to the control activation of paternal Ube3a only in the genes). Interestingly, the expression of
group, there were no statistically signifi- cerebral cortex, hippocampus, and stri- most genes shorter than 67 kb was
cant differences in the developmental atum, and the higher doses resulting in increased, albeit to a “very small” extent,
domains measured by the Bayley Scales the activation of paternal Ube3a in the by the application of topotecan [King
of Infant Development (3rd edition) cerebellum as well. Moreover, paternal et al., 2013]. More recently, the gene
(BSID-III); of note, the VABS-II is based Ube3a expression, at least in the spinal expression profile of cultured mouse
on parental reports through a semi- cord neurons, was sustained for up to neurons treated with topotecan was
structured interview while the BSID-III 12 weeks after intrathecal administration compared to that of mouse neurons
is based on objective assessments by a of topotecan. The in vitro experiments with a homozygous deletion of Top1,
psychologist. The authors felt that overall, also demonstrated that topotecan re- the gene that encodes topoisomerase I.
this treatment cocktail did not have any pressed the expression of Snurf-Snrpn Interestingly, far more genes were both
appreciable impact on the developmental and Ube3a-As in cultured neurons upregulated and downregulated in the
outcome of children with AS [Bird et al., [Huang et al., 2012]. Subsequent ex- topotecan treated wild-type neurons
2011]. The findings from these two periments have suggested that topotecan than in those neurons with the Top1
clinical trials suggest that the paternal represses the expression of paternal deletion; in particular, many immediate
UBE3A allele cannot be activated to any Ube3a-As by stabilizing R-loops at the early genes were downregulated in the
clinically significant degree, if at all, paternal Snord116 locus [Powell et al., topotecan treated neurons but not in the
through the use of oral “pro-methylation” 2013]. The mechanism of R-loop neurons with the Top1 deletion, al-
compounds, although the authors did not formation remains unclear, but R-loops though the authors suggested that this
assess the degree of UBE3A activation in may be formed during transcription and might be due to “reduced spontaneous
the participants in each of these trials. comprise a triple-stranded nascent neuronal activity” following treatment
mRNA-template DNA hybrid with with topotecan. Moreover, Ube3a ex-
Inhibiting transcription of Snurf-Snrpn and the displaced single-stranded non-tem- pression was restored in the topotecan
Ube3a-As by topoisomerase inhibitors plate DNA. R-loops can result in treated but not in the Top1-deleted
In an attempt to discover compounds “genomic instability” due to various neurons. The data from these authors
that might activate paternal UBE3A, putative mechanisms, and they can also also suggest that topotecan inhibits
Huang et al. screened 2,306 small prevent the elongation of transcription. topoisomerase type I by binding to
molecules in cultured mouse cortical One of the means through which R- both topoisomerase type I and DNA,
neurons containing an engineered loop formation is suppressed is through forming a topoisomerase type I cleavage
Ube3a-YFP (yellow fluorescent protein) the activity of topoisomerases [Skourti- complex [Mabb et al., 2016]. These data
gene on the paternal allele and identified Stathaki and Proudfoot, 2014]. raise the concern that the systemic use of
6 AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) RESEARCH REVIEW

topoisomerase inhibitors such as top- other, resulting in the premature termi- administered intravenously [Agarwala
otecan for the treatment of AS may nation of both transcripts, and the et al., 2015; Marafini et al., 2015; Santos
result in unintended off-target effects incomplete Ube3a transcript is then et al., 2015], as well as some monogenic
due to the repression of many different degraded [Meng et al., 2013]. These neuromuscular and neurodegenerative
long genes and possibly over-expression data suggest that direct repression of conditions.
of the shorter genes [Plasschaert and Ube3a-As could be a potential thera- In September 2016, the FDA
Bartolomei, 2013]. peutic strategy for AS. approved the use of an intravenous
ASO, eteplirsen (EXONDYS 51), for
Inhibiting transcription of Ube3a-As by the treatment of Duchenne muscular
antisense oligonucleotides dystrophy (DMD), although the evi-
Since the expression of paternal UBE3A Since the expression of dence for efficacy was weak [Food and
is normally repressed by UBE3A-AS, it paternal UBE3A is normally Drug Administration, 2016]. Eteplirsen
has been hypothesized that loss of binds to exon 51 of dystrophin pre-
UBE3A-AS would result in transcrip- repressed by UBE3A-AS, it mRNA, resulting in the deletion of this
tion of the paternal UBE3A allele. To has been hypothesized that exon from the mature mRNA. In
evaluate this hypothesis in vivo, Meng patients with specific frameshift muta-
loss of UBE3A-AS would
et al. engineered a mouse model that had tions, this results in the restoration of the
a poly(A) “stop cassette” inserted be- result in transcription of the reading frame, thereby converting the
tween Snord115 and Ube3a that should paternal UBE3A allele. To clinical phenotype of DMD to that of
theoretically prevent the expression of the less severe Becker muscular dystro-
Ube3a-As downstream of the cassette evaluate this hypothesis in phy [Jacobson and Feldman, 2016;
without affecting transcription of the vivo, Meng et al. engineered a Shimizu-Motohashi et al., 2016]. Nu-
other non-coding RNAs that are all sinersen is an ASO that binds to exon 7
upstream of Snord115. As expected, this
mouse model that had a of the paralogous SMN2 pre-mRNA
reduction in Ube3a-As expression was poly(A) “stop cassette” and promotes the formation of a full-
observed only when the stop cassette inserted between Snord115 length mature mRNA by preventing the
was on the paternally-inherited chro- normal skipping of exon 7 in SMN2,
mosome, but not when it was on the and Ube3a that should hence allowing the presence of SMN2
maternally-inherited chromosome since theoretically prevent the to compensate for the loss of SMN1
Ube3a-As is not normally expressed on [Hua et al., 2008]. Based on the interim
the maternal allele. In mice with the expression of Ube3a-As analysis of a phase 3 trial of nusinersen in
poly(A) cassette on the paternal allele, downstream of the cassette infantile-onset SMA (ClinicalTrials.gov
the amount of Ube3a protein was identifier: NCT02193074), a New
without affecting transcription
increased to between 50% and 70% of Drug Application was filed with the
wild type (WT) levels in different of the other non-coding RNAs FDA in September 2016 for the use of
regions of the brain. Mice engineered that are all upstream of nusinersen in SMA. ASOs are also being
with maternal Ube3a knockout and developed to treat Huntington disease
paternal poly(A) stop cassette demon- Snord115. by reducing expression of the mutant
strated improved performance in motor HTT (huntingtin) gene [Godinho et al.,
coordination (as assessed by the acceler- 2015] (ClinicalTrials.gov identifier:
ating rotarod test) and long-term mem- The use of antisense oligonucleo- NCT02519036), familial amyotrophic
ory (as assessed by the fear conditioning tides (ASOs) to modulate gene expres- lateral sclerosis (ClinicalTrials.gov iden-
test). Obesity, which is typically seen in sion in vivo has been attempted for a tifier: NCT02623699), and some forms
AS mice, was not observed in these variety of systemic diseases and malig- of frontotemporal dementia [Mis et al.,
engineered mice. Deficits in LTP were nancies. ASOs are synthetic single- 2016; Picher-Martel et al., 2016]. All of
also restored in these mice. However, stranded oligonucleotides that bind to these clinical trials will help provide
some other phenotypic features typically specific target RNAs in the nucleus and evidence for the potential safety and
seen in AS mice such as hypoactivity the ASO-RNA duplexes are then efficacy of ASO therapy administered
were not significantly different between cleaved by ribonuclease H (RNase H), intrathecally for the treatment of a wide
the engineered and WT mice. The with the resulting RNA strands being variety of neurologic disorders due to
authors hypothesized that paternal degraded by exonucleases. Thus, ASOs mutations in a single gene, thereby
Ube3a-As mediates the imprinting of can be used to target and degrade RNAs raising the hope that this may also be a
Ube3a through “transcriptional colli- with specific sequences. This approach potentially effective therapeutic strategy
sion” whereby the RNA polymerases has been found to be effective in treating for AS. It should be noted, however, that
that transcribe Ube3a and Ube3a-As in some types of hypercholesterolemia and the cognitive impairment in AS is
opposite directions collide with each inflammatory bowel disease when congenital in onset, so it remains
RESEARCH REVIEW AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) 7

unknown whether postnatal treatment domain that either activates or represses the Morris water maze was not con-
would be effective. the target gene, and a nuclear localiza- vincing, and there was no improvement
In order to target and repress tion signal that guides the ATF into the in rotarod performance. Moreover,
Ube3a-As directly, Meng et al., in nucleus. The DNA-binding protein deficits in LTP were only partially
collaboration with Ionis Pharmaceuti- may be a zinc finger protein (ZFP), restored in the treated mice [Daily
cals, developed a set of ASOs designed to transcription activator-like effectors et al., 2011]. As such, viral gene therapy
bind to a 113 kb sequence in mouse (TALEs), or a clustered regularly inter- is promising, but it is not a feasible
Ube3a-As. One ASO injected directly spaced short palindromic repeat therapeutic strategy yet. Recently, Agilis
into the lateral cerebral ventricle of adult (CRISPR)-derived protein. ZFP- Biotherapeutics, Inc. (Cambridge, MA)
WT mice resulted in a reduction in ATFs are being developed for the licensed this technology from these
paternal Ube3a-As by 60–70%, and an treatment of Huntington disease, retini- authors and it is now developing DNA
upregulation of Ube3a protein in the tis pigmentosa [Agustin-Pavon and constructs and novel vectors in an
cortex, hippocampus, and spinal cord by Isalan, 2014], amyotrophic lateral scle- attempt to develop a gene transfer
33–82% 4 weeks after injection; expres- rosis (ClinicalTrials.gov identifier: product for AS [Agilis Biotherapeutics
sion of Ube3a-As remained suppressed NCT00748501) and diabetic neuropa- Inc., 2016].
for 16 weeks after injection of a single thy (ClinicalTrials.gov identifier:
dose of ASO. These data suggested that NCT01079325). Bailus et al. (2016)
Treating Disruptions in Specific
this ASO was able to partially unsilence created 11 different ZFP-ATFs targeting
Cellular Pathways Caused by Lack
paternal Ube3a. Importantly, the expres- Ube3a and its regulatory regions, with a
of UBE3A
sion of Snrpn, Snord115, Snord116, and mixture of activation and repressor
five of the long genes that they domains. The ATF that had the stron- As discussed in the Introduction, loss of
specifically examined was not down- gest activity and could be produced in UBE3A leads to both the up-regulation
regulated by treatment with this ASO. sufficient amount was TAT-S1, a repres- and down-regulation of multiple genes,
When this ASO was subsequently sor ATF that binds between the AS-IC/ which results in disruption of various
injected into the lateral ventricle of PWS-IC and SNURF/SNRPN locus. signaling pathways and synaptic trans-
adult AS mice, Ube3a was expressed in The authors found that both subcutane- missions, some of which may be
the cortex, hippocampus, and cerebel- ous and intraperitoneal administration modulated by specific therapeutic
lum to about 35–47% of that seen in WT of TAT-S1 into adult AS mice resulted in compounds.
mice. Behavioral assays on these AS an increase in Ube3a throughout the
mice 4 weeks after treatment suggested a brain, especially in the hippocampus and Dopaminergic pathways and
partial restoration of the WT phenotype cerebellum [Bailus et al., 2016]. phosphorylation of calcium/calmodulin-
with normalized “contextual freezing” dependent kinase II (CaMKII)
behavior (indicative of long-term mem- CaMKII, an enzyme in the post-synap-
Direct Injection of UBE3A Into the
ory), but no improvement in other tic density of neurons, is activated when
Brain Using Recombinant Adeno-
neurologic assays such as the “marble it binds to the calcium-calmodulin
Associated Virus (rAAV)
burying” and accelerating rotarod tests. complex and it is essential for LTP in
The authors postulated that a complete Another therapeutic approach that has the cerebral cortex and hippocampus.
reversal of the AS phenotype may not be been attempted in a mouse model is the Activated CaMKII phosphorylates the
possible unless the mice were treated direct injection of Ube3A into the GluR1 subunit of the AMPA (a-amino-
“before a critical developmental win- hippocampi bilaterally using a rAAV 3-hydroxy-5-methylisoxazole-4-pro-
dow” or a greater amount of Ube3a is vector (serotype 9). When AS mice were prionic acid) glutamate receptor, in-
produced [Meng et al., 2015]. Never- so treated, it was found that Ube3a creasing the conductance of the sodium
theless, these data suggest that the use of expression in the hippocampus was up- ion channel. CaMKII also autophos-
ASOs might be a viable form of therapy regulated to a level comparable to that in phorylates its own threonine residues at
for AS if a sufficient amount of paternal the WT mice, but Ube3a was not over- amino acid positions 286, 305, and 306
UBE3A can be expressed throughout expressed; however, Ube3a expression in (Thr286, Thr305/306). Phosphoryla-
the brain in early infancy. the cerebellum was not increased, at least tion at Thr286 results in prolonged
not after a single injection of the rAAV- activation of CaMKII, the inhibition of
Repression of Ube3a-As by zinc finger Ube3a vector. The rAAV-Ube3a treated which impairs LTP in the hippocampus.
artificial transcription factor mice demonstrated normalization in In contrast, phosphorylation at Thr305/
Artificial transcription factors (ATFs) are fear conditioning as measured by appro- 306 prevents the binding of substrates to
synthetic compounds that regulate tran- priate contextual freezing 24 hr after CaMKII and regulates the cellular
scription of genes by binding to specific training, which suggested an improve- localization of CaMKII in the post-
target DNA sequences. At a minimum, ment in associative learning. However, synaptic density. The regulation of
each ATF comprises a specific DNA- the apparent improvement in spatial CaMKII activity and translocation by
binding protein (domain), an effector memory assessed through the use of phosphorylation of these specific
8 AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) RESEARCH REVIEW

threonine residues is critical for “cogni- initiated in children with AS immature dendrites. In AS mice, the
tive flexibility” and fine-tuning of spatial aged between 4 and 12 years. density of the dendritic spines was found
learning [Giese et al., 1998; Elgersma to be lower than that in wild-type mice,
et al., 2002; Lisman et al., 2002; Blitzer Among the 55 children who and the spines that were present were
et al., 2005]. completed this 1-year study, shorter with more variable head sizes
In a study on LTP in AS mice, it was [Dindot et al., 2008].
found that these mice had increased no clinically or statistically Matrix metalloproteinases (MMPs)
phosphorylation at the Thr286 and significant differences were are a family of zinc-binding endopepti-
Thr305/306 residues in CaMKII in dases that catalyze the proteolysis of
the hippocampus and hence reduced
found in any of the outcome specific proteins in the extracellular
CaMKII kinase activity, resulting in measures, which included the matrix, cell adhesion molecules, and
impaired LTP induction and memory BSID-III or Mullen Scales of cell surface receptors. MMPs are also
formation [Weeber et al., 2003]. Impor- synthesized and secreted by neurons,
tantly, when AS mice were crossed with Early Learning, Vineland and to a lesser extent, glia, in the central
mice that had a specific mutation in the Adaptive Behavior Scales, and nervous system. Some MMPs are ex-
CaMKII-Thr305/306 residues that pre- pressed more highly in specific brain
vented these residues from becoming Aberrant Behavior Checklist. regions. In particular, MMP9 localizes
phosphorylated, the double mutants to the hippocampal dendrites and
(i.e., AS mice with these mutated dendritic spines, as well as the cerebral
To evaluate the potential efficacy of
Thr305/306 residues) had normal cortex and cerebellum [Dziembowska
levodopa for the treatment of AS, a
CaMKII activity in the hippocampus, and Wlodarczyk, 2012; Huntley, 2012].
double-blind placebo-controlled trial of
normal hippocampal-dependent learn- MMP3 and MMP9 are believed to be
levodopa/carbidopa was initiated in
ing, normal spatial learning, improve- important in both associative and non-
children with AS aged between 4 and
ment in gait ataxia, and a decrease in associative learning and in the formation
12 years. Among the 55 children who
seizure tendency [van Woerden et al., of memory [Huntley, 2012]. Although
completed this 1-year study, no clini-
2007]. These data suggest that thera- genetically-engineered mice that over-
cally or statistically significant differ-
peutic interventions that prevent the express Mmp9 in neurons had increased
ences were found in any of the outcome
excessive phosphorylation of Thr305/ dendritic spine density, prolonged main-
measures, which included the BSID-III
306 residues in CaMKII may be tenance and greater potentiation during
or Mullen Scales of Early Learning,
beneficial in individuals with AS. LTP, and improvement in the “non-
Vineland Adaptive Behavior Scales, and
It has previously been found in a rat spatial novel object recognition” task,
Aberrant Behavior Checklist. These
model of Parkinson disease that levo- which suggests greater neuronal plastic-
findings suggest that L-dopa is not an
dopa (L-dopa) can reverse the excessive ity [Fragkouli et al., 2012], hippocampal
effective therapy for AS, at least at the
phosphorylation of threonine residues neurons that were chronically exposed
dose used in this study (i.e., 15 mg/kg/
in specific synaptic and neuronal en- to MMP9 in a “bath” (i.e., affecting
day); as noted by the authors, it is
zymes including CaMKII [Brown et al., entire neuron rather than dendritic
unknown whether use of a much lower
2005]. Preliminary data suggest that spines only) in vitro formed long and
dose could have been effective since in
treatment of AS mice with L-dopa leads thin immature dendritic spines [Dziem-
theory, L-dopa inhibits the phosphory-
to reduced phosphorylation at both bowska and Wlodarczyk, 2012; Hunt-
lation of the threonine residues in a non-
Thr286 and Thr305/306 residues in ley, 2012]. Moreover, in vivo and in
specific manner, and inhibition of
CaMKII, and an improvement in motor vitro data suggest that pharmacologic
Thr286 in CaMKII could impair LTP
skills [Tan et al., 2016]. It was therefore inhibition of MMP9 results in arrest of
[Tan et al., 2016].
hypothesized that the use of L-dopa in dendritic spine growth and maturation
AS individuals may result in an improve- [Bilousova et al., 2009; Dziembowska
ment in their motor and cognitive Dendritic spine maturation and matrix and Wlodarczyk, 2012]. There are no
development. metalloproteinases published reports on the activity of any
UBE3A localizes to the nuclei, growth MMP in AS. However, increased
cones, and dendrites, as well as the pre- MMP9 activity has been found in the
and post-synaptic compartments of cerebral cortex of post-mortem fetuses
To evaluate the potential neurons. Dendrites are usually lined and adults with Down syndrome [Iulita
with short protrusions (i.e., dendritic et al., 2014; Vafadari et al., 2015] and in
efficacy of levodopa for the spines) that form synapses with axons. the cerebral cortex and hippocampus in
treatment of AS, a double- Mature dendrites usually have long Fragile X mice and in humans with
dendritic spines with relatively large Fragile X syndrome. There are data from
blind placebo-controlled trial “mushroom-shaped” heads instead of Fragile X mice suggesting that normali-
of levodopa/carbidopa was the “filopodia-like” spines seen in zation of MMP9 activity using a double
RESEARCH REVIEW AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) 9

knockout model was beneficial [Sidhu identifier: NCT02056665) was con- development is impaired [Margolis
et al., 2014]; however, attempts to alter ducted by a different group of inves- et al., 2010]. Reducing E5 levels did
MMP9 activity using minocycline in tigators, and no differences in cognitive not alter the audiogenic stimulus-in-
humans to treat Fragile X syndrome did and motor skills were detected between duced seizure like freezing behavior in
not produce convincing results [Dziem- the minocycline and placebo cohorts. AS mice [Mandel-Brehm et al., 2015].
bowska et al., 2013]. These results were presented at the 12th Because the common deletion
Minocycline is a tetracycline anti- Congress of the European Association causing AS includes three g-amino-
biotic that has been used to treat acne for for Clinical Pharmacology and Thera- butyric acid type A (GABAA) receptor
almost 50 years [Cullen and Cohan, peutics in 2015 [Ruiz-Antora n et al., subunits: b3, a5, and g3, GABAergic
1976]. Tetracyclines bind to bacterial 2015], but the full manuscript has not dysfunction is thought to account for
ribosomes and divalent cations, and the been published yet. the observation that the deletion sub-
chelation of zinc by tetracyclines may type of AS has a more severe phenotype
inhibit the activity of MMPs. Tetracy- Synaptic signaling pathways than AS caused by other molecular
clines may also modulate pro-inflamma- There is evidence from animal studies mechanisms. GABA, the main inhibi-
tory cytokines, resulting in secondary that several synaptic signaling pathways tory neurotransmitter in the mammalian
inhibition of MMP transcription. Min- are altered in AS. Presumably, defective central nervous system, acts to regulate
ocycline is one of the most lipophilic Ube3a activity impairs ubiquitination muscle tone and reduce neuronal excit-
tetracyclines and it crosses the blood- and subsequent degradation of its sub- ability. In frozen post-mortem cortical
brain barrier well [Jonas and Cunha, strates, resulting in increased abundance samples of four human AS brains,
1982; Griffin et al., 2010]. Despite the of its target proteins. Several Ube3a Roden et al. [2010] found alterations
lack of studies showing altered MMP9 targets have been identified, including in GABAA receptor subunit expression
activity, a trial of minocycline in AS was Arc [Greer et al., 2010] and Ephexin5 that were consistent with impaired
undertaken based on an in vitro small [Margolis et al., 2010]. Ube3a regulates cortical extra-synaptic GABAergic ac-
molecule screen and preliminary mouse Arc levels either by regulating estradiol- tivity. GABA transporter 1 (GAT1)
data. Twenty five children with AS induced transcription of Arc [Kuhnle activity is increased in AS mice, pre-
between the age of 4 and 12 years et al., 2013] or by direct ubiquitination sumably because Ube3a deficiency de-
were treated with minocycline for of Arc [Greer et al., 2010]. Arc in turn creases GAT1 ubiquitination. Since
8 weeks in an open-label trial. Devel- regulates surface expression of a-amino- GAT1 takes up GABA from the extra-
opmental assessments were performed at 3-hydroxy-5-methyl-4-isoxazole-pro- synaptic space, tonic inhibition, a
the end of the 8-week trial and at pionic acid receptors (AMPARs). When GABAergic form of continuous inhibi-
16 weeks after initiation of therapy (i.e., Ube3a is deficient, Arc levels are tory transmission mediated through
8 weeks after discontinuation of mino- excessive, and the excitatory postsynap- specialized GABAA receptors, is de-
cycline). The authors found slight tic AMPARs are internalized, which creased in the cerebellar granule cells of
overall improvement, as measured by impairs synaptic transmission [Greer AS mice due to reduced availability of
the Clinical Global Impressions—Se- et al., 2010]. Using a Ube3a/Arc GABA in the extra-synaptic space
verity scale. There was also an improve- double knockout mouse model, Man- [Egawa et al., 2012]. Tonic inhibition
ment in receptive communication on del-Brehm et al. showed that reducing regulates neuronal excitability in the
the VABS-II, which is based on parental the levels of Arc decreases abnormal entire brain, including the neocortex,
report, at the end of the 8-week trial, but brain spiking in AS mice and rescues the hippocampus, thalamus, hypothalamus,
this improvement was not sustained increased duration of freezing (seizure- basal ganglia, and cerebellum, as well as
8 weeks after discontinuation of mino- like) behavior in response to an audio- the spinal cord, and in retinal ganglion
cycline [Grieco et al., 2014]. However, genic stimulus. However, other behav- cells. In various mouse models, loss of
multiple flaws in the study design, ioral measures (ultrasonic vocalizations, tonic inhibition results in increased
including the lack of a control group, hind limb-clasp phenotype, hypoactiv- seizure susceptibility and in some in-
have made it impossible to draw any ity) in the double knockout mice stances, sleep disturbances, depending
conclusions regarding the efficacy of remained unchanged compared with on the GABAA receptor subtype that is
minocycline in AS from this trial. AS mice [Mandel-Brehm et al., 2015]. knocked out [Lee and Maguire, 2014].
Moreover, MMP9 activity was not Ephexins are guanine nucleotide Egawa et al. also found that cerebellar
measured in this study, so it is unknown exchange factors (GEFs), which modu- ataxia, as demonstrated by gait analysis
whether minocycline therapy had any late the development of excitatory and the clasping reflex, improved when
impact on MMP9 activity in AS. synapses. Ephexin5 (E5) is one such the AS mice were treated with 4,5,6,7-
Subsequently, an 8-week cross-over GEF, which functions to suppress excit- tetrahydroisothiazolo-[5,4-c]pyridin-3-
double-blind placebo-controlled trial atory synapse development. Ube3a ol (THIP), an extra-synaptic GABAA
of minocycline in 32 children and adults mediates the degradation of E5, so receptor agonist selective for those
with AS, randomized 2:1 to minocy- when Ube3a is absent, E5 levels are receptors that contain the d subunit.
cline or placebo, (ClinicalTrials.gov elevated and excitatory synapse These results suggested that the
10 AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) RESEARCH REVIEW

cerebellar ataxia in AS is mediated implications of this finding are unclear, compared to the wild type mice, so it is
through reduced tonic inhibition the authors postulated that this may be unlikely that the increased Nrg1-ErbB4
[Egawa et al., 2012]. In light of these related to the unusually happy personal- signaling is mediated through these
findings, Ovid Therapeutics Inc. (New ity in AS since individuals who are ionotropic glutamate receptors [Kaph-
York, NY) will be conducting a phase 2 depressed have reduced BDNF concen- zan et al., 2012].
randomized double-blind placebo-con- trations [Wink et al., 2015]. The mechanistic target of rapamy-
trolled trial of THIP, also known as In a pilot study of 11 children and cin, mTOR, plays critical roles in brain
gaboxadol, in adults with AS to assess its adults with AS, it was found that the development and synaptic plasticity.
safety and potential efficacy in this plasma concentrations of amyloid pre- mTOR is the catalytic subunit of two
population [Ovid Therapeutics Inc., cursor protein (APP) and its break- complexes, mTORC1 and mTORC2
2016]. However, a recent study sug- down products were higher in those (mechanistic target of rapamycin com-
gested that the “locomotor deficits” in with AS compared to age- and gender- plexes 1 and 2, respectively). Sun et al.
AS mice, as determined by the acceler- matched controls, and the levels of total found increased activity of mTORC1
ating rotarod test, are not due to a APP and APP alpha were positively with increased phosphorylation of some
cerebellar defect but may be due to correlated with plasma BDNF concen- of its substrates including 40S ribosomal
loss of Ube3a in the “extracerebellar trations, but the levels of total APP and protein S6 and ribosomal protein S6
motor circuits” instead [Bruinsma et al., its breakdown products did not correlate kinase beta-1 (S6K1), but decreased
2015]. with the two neurodevelopmental out- phosphorylation of mTORC2 sub-
As previously indicated, dendritic come measures that were used in the strates such as AKT in the cerebellum
spine abnormalities are seen in mouse study [Erickson et al., 2016]. The of AS mice. These changes in
models of AS [Dindot et al., 2008]. clinical implications of these findings mTORC1 and mTORC2 activity
Shortly after LTP induction, dendritic remain unclear, but the authors sug- were mediated by an increase in the
spine actin polymerization occurs, and gested that APP (or its breakdown concentration of TSC2 (tuberous scle-
this is tightly correlated with LTP products) might be a therapeutic target rosis complex 2), which is usually
stabilization, consolidation and memory in the future. degraded by UBE3A. Treatment with
[Lynch et al., 2008; Simmons et al., Because of perceived clinical and intraperitoneal rapamycin (also known
2009; Baudry et al., 2011]. The induc- neuropathological similarities between as sirolimus), an mTOR inhibitor,
tion of LTP in the hippocampal slices of AS and schizophrenia, Kaphzan et al. corrected the activity of both complexes
AS mice is apparently normal, but LTP [2012] examined AS mice for dysregu- and improved motor function and
stabilization, which requires dendritic lation of the neuregulin 1 (Nrg1)- dendritic spine morphology in the AS
spine actin polymerization in response ErbB4 pathway, which is implicated in mice [Sun et al., 2015a]. These authors
to the inductive stimulus, is severely the pathophysiology of schizophrenia. subsequently reported that treatment of
impaired in AS mice. Deficits in LTP NRG1 signals through its receptor AS mice with intraperitoneal rapamycin
stabilization and spine actin polymeri- tyrosine kinase ErbB4 (Erb-B2 Recep- also led to an improvement in LTP and
zation are reversed in the hippocampal tor Tyrosine Kinase 4). Nrg1 concen- in their learning and memory as deter-
slices by short-term treatment with an trations are significantly increased in the mined by the fear conditioning assay
ampakine, which also reversed deficits in hippocampi of AS mice compared to [Sun et al., 2016].
contextual fear conditioning in AS mice wild-type littermates; high levels of Reelin is a ligand of the apo-
[Baudry et al., 2012]. Ampakines work Nrg1 result in constitutive activation lipoprotein receptor family, which is
by binding to the glutamatergic AMPA (phosphorylation) of ErbB4. Constitu- primarily secreted by GABAergic in-
receptor, enhancing fast excitatory tive activation of ErbB4 results in terneurons. Reelin regulates AMPA
transmission, facilitating LTP and upre- impaired LTP [Kwon et al., 2005; receptor function and NMDA receptor
gulating brain-derived neurotrophic Pitcher et al., 2008], and ErbB inhibitors insertion, and it is necessary for proper
factor (BDNF) (reviewed in [Chang can rescue LTP impairments in AS mice neuronal migration as well as adult
et al., 2012; Panja and Bramham, 2014]). hippocampal slices [Kaphzan et al., neuronal synaptic plasticity. Purified
AS mice have defective BDNF signaling 2012]. Deficits in contextual fear con- recombinant Reelin injected into the
(NOT necessarily concentration) due to ditioning, a hippocampus-dependent cerebral ventricles of WT [Rogers et al.,
elevated Arc levels [Cao et al., 2013], form of associative memory, were 2011] and AS [Hethorn et al.,
and BDNF is required for proper rescued in AS mice by post-training 2015] adult mice enhances LTP, in-
postsynaptic signaling [Yoshii and Con- injection of an ErbB inhibitor into both creases dendritic spine density, and
stantine-Paton, 2007; Yoshii et al., hippocampi. Neither Nrg1 nor ErbB4 improves associative learning and
2011]. Interestingly, in 12 children and interacts directly with Ube3a. More- memory. Reelin did not rescue motor
adults with AS, it was found that the over, there were no differences in the deficits in the AS mice [Hethorn et al.,
plasma BDNF concentrations were expression or phosphorylation status of 2015].
elevated compared to typically develop- AMPA receptors or N-methyl-D-aspar- Another substrate that is ubiquiti-
ing controls. Although the clinical tate (NMDA) receptors in the AS mice nated by UBE3A for endocytosis is the
RESEARCH REVIEW AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) 11

small conductance calcium-acti- hippocampal neurons of AS mice, with an unknown prevalence, but in
vated potassium channel protein 2 normalized the electroencephalogram probably no less than 10%. Those with
(SK2). SK2 is a postsynaptic potassium (EEG) pattern, and improved motor deletions had the highest rates of
channel that regulates LTP and long- coordination and anxiety [Llewellyn epilepsy, and those with imprinting
term depression (LTD), and hence et al., 2015]. More recently, it was defects the lowest. Pelc et al.
synaptic plasticity. As expected, there found that mitochondrial superoxide [2008a] reported that approximately
was an increase in the density of SK2 production was increased in AS mice half of patients with AS have multiple
channels in the postsynaptic membrane compared to wild type mice, but seizures types. Those with the larger
of hippocampal neurons derived from treatment with intraperitoneal MitoQ deletions tend to have more frequent
AS mice compared to wild type mice 10-methanesuflonate, an antioxidant and disabling seizures and are more
[Lizarraga and Morrow, 2015; Sun et al., that contains coenzyme Q10, normal- likely to need polytherapy compared to
2015b]. Interestingly, treatment of wild ized the superoxide concentration as those with the smaller deletions [Valente
type mice and rats with apamin (a SK well as hippocampal LTP and contex- et al., 2013]. There are no obvious
antagonist) resulted in an improvement tual fear conditioning. These data candidate genes in the non-overlapping
in their learning and memory [Moreno suggest that oxidative stress may con- deletion region (NIPA1, NIPA2,
and Giralt, 2015]. When hippocampal tribute to the learning and memory CYF1P1, GCP5) that readily explain
slices from AS mice were incubated with deficits seen in AS [Santini et al., 2015]. this observation.
apamin, there was an improvement in
LTP and LTD; when AS mice were
Strategies Addressing Specific
treated with intraperitoneal apamin,
Symptoms Although valproic acid, and to
there was an improvement in their
learning and memory, as determined Seizures a lesser extent, clonazepam,
by the fear-conditioning assay [Sun Seizures occur in about 80–95% of
et al., 2015b]. These data suggest that individuals with AS with the age of were the most commonly-
an excessive concentration of SK2 onset being less than 3 years old in about prescribed anti-epileptic
channels on the postsynaptic membrane 75% of them (reviewed in [Thibert et al.,
medications in AS,
in neurons contributes to the patho- 2013]). The epilepsy of AS is considered
physiology of AS, including the LTP and a generalized epilepsy, but partial seiz- levetiracetam, lamotrigine,
LTD deficits. Fluoxetine, a serotonin ures are seen commonly as well. The and clobazam were perceived
selective reuptake inhibitor (SSRI), has mechanism of epileptogenesis in AS is
been found to block all three SK not well understood (reviewed in [Pelc to be more efficacious,
channels in vitro [Terstappen et al., et al., 2008a]), but a recent mouse study especially when compared
2003]. Treatment of adult AS mice suggested that susceptibility to seizures is
with subcutaneous fluoxetine partially caused by loss of Ube3a in cortical
with clonazepam.
restored hippocampal neurogenesis, but GABAergic, but not glutamatergic,
learning and behavioral assays were not neurons resulting in neuronal hyperex-
reported for these fluoxetine-treated citability [Santini and Klann, 2016]. Although valproic acid, and to a
mice [Godavarthi et al., 2015], and Based on a questionnaire survey of lesser extent, clonazepam, were the most
there are no reports on the outcome of 461 respondents (of which 86% indi- commonly-prescribed anti-epileptic
any individual with AS who has received cated a family member with AS had medications in AS, levetiracetam, lamo-
fluoxetine. experienced seizures), Thibert et al. trigine, and clobazam were perceived to
[2009] reported that the most frequently be more efficacious, especially when
reported types of seizure were atonic, compared with clonazepam. More im-
Mitochondrial Dysfunction
generalized tonic-clonic, atypical ab- portantly, compared to the newer anti-
AS mice exhibit deficits in hippocam- sence, and partial complex seizures; focal epileptic medications, valproic acid was
pal LTP, and Su et al. [2011] observed motor, tonic, secondarily generalized associated with more side effects, in-
that the hippocampal mitochondria of and myoclonic seizures as well as cluding thrombocytopenia, pancreatitis,
AS mice are small with disorganized infantile spasms and Lennox–Gastaut tremor, balance, and gait difficulties
cristae and reduced complex III activ- syndrome were also reported. Some [Thibert et al., 2013; Shaaya et al.,
ity. Coenzyme Q10, a cofactor in the authors report myoclonic seizures as one 2016]. Carbamazepine and phenobarbi-
electron transport chain, is frequently of the most common seizure types tal had the lowest rates of efficacy, the
prescribed as a supplement in patients [Valente et al., 2013]. The average age highest rates of seizure exacerbation, and
with mitochondrial disease. Adminis- of onset of seizures was 2.9 years (earlier the highest rates of side effects [Franz
tration of a coenzyme Q10 analogue in those with deletions) [Thibert et al., et al., 2000; Dion et al., 2007; Thibert
increased mitochondrial complex III 2009]. Both convulsive and non-con- et al., 2009]. Vigabatrin and oxcarbaze-
and complex IV expression in vulsive status epilepticus are reported pine are also associated with seizure
12 AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) RESEARCH REVIEW

exacerbation [Ostergaard and Balslev, ketosis for the treatment of epilepsy in Sleep
2001; Valente et al., 2006]. AS, Ciarlone et al. [2016] fed 4–6 Sleep disturbance is one of the most
Non-pharmacological therapies for week-old AS mice a ketone ester, R,S- vexing problems in AS. Sleep disruption
epilepsy in AS include the ketogenic diet 1,3-butanediol acetoacetate diester, in in children with AS correlates positively
(KD), low glycemic index treatment addition to their regular rodent chow ad with sleep disturbance in adult care-
(LGIT) and vagal nerve stimulator. KD lib for 8 weeks, after they had initially takers and with some measures of
is a highly limited carbohydrate diet, fasted for 8 hr. The authors found that parental stress [Didden and Sigafoos,
with approximately 90% of calories the mice whose feeds were supple- 2001; Goldman et al., 2012]. There
coming from fat, intending to mimic mented with ketone ester had lower appears to be a reduced need for sleep in
the fasting state. There are few random- blood glucose and higher beta-hydrox- individuals with AS, and problems with
ized controlled trials of KD in children ybutyrate levels compared to the con- both the initiation and maintenance of
[Neal et al., 2008; Neal et al., 2009], and trol mice. Although the treated mice sleep have been reported [Goldman
none in AS specifically [Levy et al., did not show any improvement in their et al., 2012]. Severe sleep problems
2012]. KD is associated with a significant anxiety and general locomotion, their affect a significant portion of individuals
reduction in seizure frequency in a performance on the accelerating ro- with AS. A questionnaire study of sleep
pharmaco-resistant population (38% tarod and “wire hang” was better than problems in 49 Italian individuals with
vs. 6% in controls) [Neal et al., 2008], the untreated AS mice, suggesting an AS showed that 70% of those children
and a recent systematic review by the improvement in motor coordination slept for less than 8 hr per night, and 32%
Cochrane Epilepsy Group agreed that and motor learning, although not as of them took longer than 30 min to fall
KD could be a viable therapeutic option well as the wild type mice. The treated asleep. Sixty-two percent had more than
for children, but its efficacy has not been AS mice were also less likely to develop two awakenings per night, and 57%
demonstrated in adults with epilepsy seizures in response to audiogenic and reported difficulty in resuming sleep
[Martin et al., 2016]. KD has been used chemical (kainic acid) stimulation after awakening at night. Poor sleep
anecdotally in AS with great benefit in [Ciarlone et al., 2016]. These findings quality was reported by 70% of re-
some cases and remains a valuable suggest that ketone ester supplementa- spondents. Only 24% reported daytime
treatment modality for patients with tion may be an alternative to treatment somnolence and only 8% reported
refractory epilepsy [Valente et al., 2006; with KD or LGIT, although some falling asleep at school, indicating that
Evangeliou et al., 2010; Stein et al., clinical experts opined that this strategy despite the sleep disruption, most indi-
2010]. The main side effects of KD are might not work unless the “regular viduals with AS do not exhibit daytime
gastrointestinal disturbances such as diet” is at least somewhat ketogenic somnolence [Bruni et al., 2004]. An-
constipation, metabolic acidosis, hyper- [Ronald L. Thibert, Personal Commu- other questionnaire study of 109 Dutch-
lipidemia, and occasionally renal calculi nication, July 2016]. speaking individuals found 40% had
[Kossoff et al., 2009; Bergqvist, 2012]. Corticosteroids have been used to severe sleep problems, mostly consisting
There may be substantial difficulties treat various forms of epilepsy, most of night-time wakening rather than
adhering to the diet, and in one series, notably infantile spasms. Forrest et al. prolonged sleep latency [Didden et al.,
only about 10% of individuals remained reported four children with AS (all with 2004]. Walz et al. examined the sleep
on it for an extended period of time a deletion) who responded to predniso- patterns of 339 individuals registered
[Hemingway et al., 2001]. lone with cessation of seizures and with the Angelman Syndrome Founda-
In order to address issues with improvement in sleep disturbance. tion using a validated sleep question-
palatability leading to reduced adher- When prednisolone was tapered, seiz- naire. Difficulty falling asleep and
ence, modifications to the KD have ures frequently recurred, but were less sleeping less than peers were reported
been made. LGIT limits the amount of severe [Forrest et al., 2009]. in 48% and 42% of respondents respec-
carbohydrates to 10% of the diet and the Cannabinoids are being used clini- tively [Walz et al., 2005]. Sleep problems
type of carbohydrates to those with a cally to treat refractory epilepsy [dos did not appear to abate with age,
glycemic index less than 50, while Santos et al., 2015; Fasinu et al., 2016]. although the sleep issues may be most
permitting up to 30% protein. In a small Anecdotal evidence and preclinical severe between the ages of 2 and 6 years
prospective study of LGIT in AS, five of studies support this as a potentially [Bruni et al., 2004; Didden et al., 2004;
six patients experienced a more than useful modality in some situations, but Walz et al., 2005; Robinson-Shelton
80% reduction in seizures (including no specific reports on the use of and Malow, 2016].
one patient who did not achieve the cannabinoids in AS are available. Cur- In the only study to examine sleep
target carbohydrate reduction), and 50% rently, there are three ongoing open- objectively in AS, Miano et al.
became seizure free [Thibert et al., label trials of cannabinoids in children [2004] performed polygraphy on 15
2012]. with epilepsy, but none are specific for children with AS and found decreased
Given the challenges of adhering to AS (ClinicalTrials.gov identifiers: sleep efficiency and REM sleep, as well
a low glycemic index diet and to NCT02447198, NCT02397863, as an increased percentage of time spent
explore alternative means of inducing NCT02523183). in slow wave sleep in those with AS
RESEARCH REVIEW AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) 13

under the age of 8 years compared to a Behavior modification can be ef- features that are present in most of these
control group of children, and decreased fective in some patients with AS [Allen mouse models include impaired associa-
REM sleep in those more than 8 years of et al., 2013], as it can be in other tive and contextual learning, seizures/
age compared with controls. Sleep- conditions with developmental disabil- abnormal EEG, and gait ataxia [Jana,
disordered breathing and periodic leg ity [Grigg-Damberger and Ralls, 2013]. 2012]. The only other animal model
movements did not occur more fre- The importance of sleep hygiene, that has been widely used in studies of
quently in AS than in other disorders including the imposition of fixed bed- AS has been Drosophila whose UBE3A
with intellectual disability with or times, and a consistent bedtime routine homolog, Dube3a, is about 62% identi-
without epilepsy [Miano et al., 2005]. and sleep schedule in the management cal to human UBE3A; these flies had
The etiology of the sleep problems in of sleep disturbance is crucial. Allen et al. deficits in motor activities and “associa-
AS is unknown, but it is speculated to be [2013] showed that a behavioral “pack- tive olfactory memory” [Gatto and
related to thalamocortical dysfunction age” could result in independent sleep Broadie, 2011]. However, the substrates
(reviewed in [Pelc et al., 2008b]). In initiation, modest increase in total sleep of mouse Ube3a and Drosophila Dube3a
addition, pathophysiological processes time, and reduction of disruptive be- are not identical, so the findings from
unrelated to the neurologic regulation haviors (both sleep- and non-sleep- Drosophila are not always translatable to
of sleep such as epilepsy, gastrointestinal related). Severity of epilepsy correlates mice, much less to humans.
discomfort, and the use of certain with sleep problems, but whether more While animal models have been
medications, may adversely impact the severe seizures create sleep disturbance useful in advancing our understanding of
quality or duration of sleep in any or whether poor sleep patterns exacer- AS, they have limitations. Some of these
individual. In a small study, melatonin bate epilepsy remains unclear [Conant mouse models have been incompletely
levels were found to be low in those with et al., 2009]. characterized [Margolis et al., 2015]. It
AS [Takaesu et al., 2012], corroborating has been found that the genetic back-
the prior results of small open-label and ground and the age of the AS mouse
ANIMAL MODELS OF
randomized placebo-controlled trials. model greatly impacts the phenotype.
ANGELMAN SYNDROME
Use of melatonin shortened the time to Huang et al. found that the AS 129S7/
AND FUTURE CHALLENGES
sleep onset, decreased sleep latency, SvEvBrd-Hprtb-m2 mice exhibited no
increased total sleep time, and reduced Much of what we know about the significant differences from their WT
night awakenings [Zhdanova et al., 1999; pathogenesis of AS has come from counterparts in phenotypic assays that
Braam et al., 2008]. The effects of studies in animal models of AS that assessed motor skills, but they were
melatonin are not unique to AS; melato- could not possibly be conducted in deficient in their “reversal learning” skills
nin has been shown to be effective in humans, and it is only through a better compared to the WT mice. In contrast,
diverse conditions associated with devel- understanding of the basic biology of AS the AS C57BL/6J mice demonstrated
opmental disability [Braam et al., 2009; that investigators have been able to marked deficiencies in various motor and
Schwichtenberg and Malow, 2015]. Clo- develop potential therapies for AS that cognitive skills compared to their WT
nidine was effective in a small cohort of are safe, specific, and potentially effec- counterparts. However, even among the
patients with severe intellectual disability tive for AS such as some of those C57BL/6J mice, some of the differences
[Ingrassia and Turk, 2005] and it seems to described above. Animal models have between the AS and WT mice were only
be effective in some patients with AS also been helpful in enabling investi- observed in mice in certain age groups
(Bird & Tan, personal observations), but it gators to assess the efficacy and safety of [Huang et al., 2013]. Similarly, “cued fear
has not been studied specifically in the AS potential therapeutic compounds before conditioning” is only seen in the mutant
population. Other medications that are they are tested in human clinical trials. exon 15/16 deletion mouse on a C57
used less often in the AS population but The mouse Ube3a protein shares about background strain, but not in the exon 2
could be considered in specific circum- 99% similarity with human UBE3A, deletion mouse on a 129 background
stances include: (i) oral iron supplemen- and murine Ube3a is imprinted just as strain [Margolis et al., 2015]. As such, the
tation—if there is periodic limb human UBE3A is. A few different AS genetic background, the age of the mice,
movement disorder or serum ferritin mouse models have been developed, all and possibly the sex, should be taken into
levels are low, (ii) ramelteon (melatonin of which recapitulate some, but not all, consideration when interpreting the
agonist)—clinical benefit over melatonin of the human AS phenotypic findings. results of studies using mouse models of
remains to be determined, (iii) gabapen- Among the more commonly used AS. Another major limitation of both
tin—this is a precursor of GABA and mouse models are one that harbors a mouse and Drosophila models of AS is the
increases synaptic GABA concentration, mutation in exon 2 of the maternally- inherent inability to capture one of the
(iv) clonazepam—as a benzodiazepine, inherited Ube3a only resulting in a most striking phenotypes in AS—the
there is an increased risk for daytime complete loss of Ube3a expression [Jiang lack of expressive language. There is
somnolence and habituation, (v) zolpi- et al., 1998], and one with a deletion therefore an urgent need to develop new
dem, and (vi) trazodone [Blackmer and including part of exon 15 and all of exon animal models of AS that recapitulate
Feinstein, 2016]. 16 [Miura et al., 2002]. The phenotypic more fully the human phenotype.
14 AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) RESEARCH REVIEW

Since AS is a congenital disorder, it reactivated by Cre REFERENCES


has long been suspected that there are recombinase, it was found that
“windows of opportunity” for the ame- Agarwala A, Jones P, Nambi V. 2015. The role of
antisense oligonucleotide therapy in patients
lioration of the phenotype beyond which reactivation of Ube3a during with familial hypercholesterolemia: Risks,
some phenotypic manifestations will not embryonic development benefits, and management recommenda-
be amenable to treatment. This suspicion tions. Curr Atheroscler Rep 17:467.
was further reinforced by findings from resulted in phenotypically Agilis Biotherapeutics Inc. 2016. Agilis DNA
therapeutic programs: DNA therapeutics for
the studies in which AS mice treated with normal offspring. Angelman syndrome (AS). Updated: 2016.
topotecan, an ASO, or viral gene therapy, Accessed: 2016 Oct 22. Available from:
http://www.agilisbio.com/pipeline/dna-
failed to normalize the phenotype therapeutic-programs/
completely even though there were The development of new thera- Agustin-Pavon C, Isalan M. 2014. Synthetic
improvements in some aspects of their peutic compounds for AS is currently biology and therapeutic strategies for the
degenerating brain: Synthetic biology ap-
deficits [Daily et al., 2011; Huang et al., limited by the lack of valid and reliable proaches can transform classical cell and gene
2012; Meng et al., 2015]. In a recent biomarkers that would allow for the therapies, to provide new cures for neuro-
study using an AS mouse model in which rapid detection of potential clinical degenerative diseases. Bioessays 36:979–990.
Allen KD, Kuhn BR, DeHaai KA, Wallace DP.
the maternally-inherited copy of the efficacy, rather than having to await 2013. Evaluation of a behavioral treatment
Ube3a gene is inactivated by a floxed the outcomes of neurodevelopmental package to reduce sleep problems in children
cassette that can be reactivated by Cre assessments, which are generally not with Angelman Syndrome. Res Dev Disabil
34:676–686.
recombinase, it was found that reactiva- valid if conducted at less than 6-monthly Bailus BJ, Pyles B, McAlister MM, O’Geen H,
tion of Ube3a during embryonic devel- intervals. Given the slow developmental Lockwood SH, Adams AN, Nguyen JT, Yu
opment resulted in phenotypically trajectories typical of most individuals A, Berman RF, Segal DJ. 2016. Protein
delivery of an artificial transcription factor
normal offspring. In contrast, when with AS, we do not believe that restores widespread ube3a expression in an
Ube3a was reactivated at different postna- meaningful changes can be detected by Angelman syndrome mouse brain. Mol Ther
tal ages, viz. 3 weeks, 6 weeks, and assessments that are performed less than 24:548–555.
Baudry M, Bi X, Gall C, Lynch G. 2011. The
14 weeks of age, corresponding very 1 year apart. Our knowledge of the biochemistry of memory: The 26 year
approximately to human childhood, clinical presentation and phenotypes of journey of a ’new and specific hypothesis’.
adolescence, and adulthood, the pheno- adults with AS is limited because most Neurobiol Learn Mem 95:125–133.
Baudry M, Kramar E, Xu X, Zadran H, Moreno S,
typic deficits that can be rescued were clinical studies on AS have traditionally Lynch G, Gall C, Bi X. 2012. Ampakines
different. For example, reactivation of focused on children. Hence, the natural promote spine actin polymerization, long-
Ube3a at 3 weeks old led to the complete history of AS in adults needs to be term potentiation, and learning in a mouse
model of Angelman syndrome. Neurobiol
resolution of the “motor coordination” further explored in order to identify the Dis 47:210–215.
deficit, whereas reactivation at 6 weeks medical needs in that population. Bergqvist AG. 2012. Long-term monitoring of the
old resulted in only a partial rescue of the Nevertheless, with the progress that ketogenic diet: Do’s and Don’ts. Epilepsy
Res 100:261–266.
same deficit, and reactivation at 14 weeks we have made in our understanding of Bilousova TV, Dansie L, Ngo M, Aye J, Charles JR,
of age failed to rescue this deficit. AS and the development of new genetic Ethell DW, Ethell IM. 2009. Minocycline
Similarly, other phenotypic features engineering tools over the last 10 years, promotes dendritic spine maturation and
improves behavioural performance in the fragile
such as autism and anxiety could only an effective therapy for AS is an exciting X mouse model. J Med Genet 46:94–102.
be rescued if Ube3a were reactivated prior prospect for the near future. It is possible Bird LM. 2014. Angelman syndrome: Review of
to a certain age. Interestingly, the epilepsy that successful treatment of AS may clinical and molecular aspects. Appl Clin
Genet 7:93–104.
phenotype could not be rescued postna- require a therapeutic “cocktail” com- Bird LM, Tan WH, Bacino CA, Peters SU,
tally [Silva-Santos et al., 2015]. These prising compounds that activate the Skinner SA, Anselm I, Barbieri-Welge R,
findings underscore the critical impor- normally imprinted paternal UBE3A, Bauer-Carlin A, Gentile JK, Glaze DG,
Horowitz LT, Mohan KN, Nespeca MP,
tance of identifying and treating children compounds that target some of the Sahoo T, Sarco D, Waisbren SE, Beaudet AL.
with AS as early as possible since a cure pathways that underlie the pathophysi- 2011. A therapeutic trial of pro-methylation
may only be possible up to a certain (as yet ology of AS, and compounds that dietary supplements in Angelman syndrome.
Am J Med Genet A 155A:2956–2963.
undetermined) age. address some of the specific symptoms Blackmer AB, Feinstein JA. 2016. Management of
as discussed above. sleep disorders in children with neuro-
developmental disorders: A review. Phar-
macotherapy 36:84–98.
ACKNOWLEDGMENTS Blitzer RD, Iyengar R, Landau EM. 2005.
In a recent study using an AS Postsynaptic signaling networks: Cellular
The authors would like to thank Stormy cogwheels underlying long-term plasticity.
mouse model in which the J. Chamberlain, Ph.D., (University of
Biol Psychiatry 57:113–119.
Braam W, Didden R, Smits MG, Curfs LM. 2008.
maternally-inherited copy of Connecticut Health Center) for review- Melatonin for chronic insomnia in Angel-
ing and revising the sections in this man syndrome: A randomized placebo-
the Ube3a gene is inactivated manuscript that describe imprinting in
controlled trial. J Child Neurol 23:649–654.
Braam W, Smits MG, Didden R, Korzilius H, Van
by a floxed cassette that can be Angelman syndrome. Geijlswijk IM, Curfs LM. 2009. Exogenous
RESEARCH REVIEW AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) 15

melatonin for sleep problems in individuals Didden R, Sigafoos J. 2001. A review of the nature cannabidiol preparations as new therapeutic
with intellectual disability: A meta-analysis. and treatment of sleep disorders in individu- agents. Pharmacotherapy 36:781–796.
Dev Med Child Neurol 51:340–349. als with developmental disabilities. Res Dev Food and Drug Administration. 2016. FDA grants
Brown AM, Deutch AY, Colbran RJ. 2005. Disabil 22:255–272. accelerated approval to first drug for Duchenne
Dopamine depletion alters phosphorylation Dindot SV, Antalffy BA, Bhattacharjee MB, muscular dystrophy. Updated: 2016 Sep 19.
of striatal proteins in a model of Parkinson- Beaudet AL. 2008. The Angelman syn- Accessed: 2016 Oct 22. Available from:
ism. Eur J Neurosci 22:247–256. drome ubiquitin ligase localizes to the http://www.fda.gov/NewsEvents/Newsroom/
Bruinsma CF, Schonewille M, Gao Z, Aronica synapse and nucleus, and maternal defi- PressAnnouncements/ucm521263.htm
EM, Judson MC, Philpot BD, Hoebeek FE, ciency results in abnormal dendritic spine Forrest KM, Young H, Dale RC, Gill DS. 2009.
van Woerden GM, De Zeeuw CI, Elgersma morphology. Hum Mol Genet 17:111–118. Benefit of corticosteroid therapy in Angel-
Y. 2015. Dissociation of locomotor and Dion MH, Novotny EJ Jr., Carmant L, Cossette P, man syndrome. J Child Neurol 24:952–958.
cerebellar deficits in a murine Angelman Nguyen DK. 2007. Lamotrigine therapy of Fragkouli A, Papatheodoropoulos C, Georgopou-
syndrome model. J Clin Invest epilepsy with Angelman’s syndrome. Epi- los S, Stamatakis A, Stylianopoulou F,
125:4305–4315. lepsia 48:593–596. Tsilibary EC, Tzinia AK. 2012. Enhanced
Bruni O, Ferri R, D’Agostino G, Miano S, Dittrich B, Buiting K, Korn B, Rickard S, Buxton neuronal plasticity and elevated endogenous
Roccella M, Elia M. 2004. Sleep disturban- J, Saitoh S, Nicholls RD, Poustka A, sAPPalpha levels in mice over-expressing
ces in Angelman syndrome: A questionnaire Winterpacht A, Zabel B, Horsthemke B. MMP9. J Neurochem 121:239–251.
study. Brain Dev 26:233–240. 1996. Imprint switching on human chro- Franz DN, Glauser TA, Tudor C, Williams S.
Buckley RH, Dinno N, Weber P. 1998. Angelman mosome 15 may involve alternative tran- 2000. Topiramate therapy of epilepsy associ-
syndrome: Are the estimates too low? Am J scripts of the SNRPN gene. Nat Genet ated with Angelman’s syndrome. Neurology
Med Genet 80:385–390. 14:163–170. 54:1185–1188.
Buiting K, Lich C, Cottrell S, Barnicoat A, Dizik M, Christman JK, Wainfan E. 1991. Gatto CL, Broadie K. 2011. Drosophila modeling
Horsthemke B. 1999. A 5-kb imprinting Alterations in expression and methylation of heritable neurodevelopmental disorders.
center deletion in a family with Angelman of specific genes in livers of rats fed a cancer Curr Opin Neurobiol 21:834–841.
syndrome reduces the shortest region of promoting methyl-deficient diet. Carcino- Giese KP, Fedorov NB, Filipkowski RK, Silva AJ.
deletion overlap to 880 bp. Hum Genet genesis 12:1307–1312. 1998. Autophosphorylation at Thr286 of
105:665–666. dos Santos RG, Hallak JE, Leite JP, Zuardi AW, the alpha calcium-calmodulin kinase II in
Buiting K, Williams C, Horsthemke B. 2016. Crippa JA. 2015. Phytocannabinoids and LTP and learning. Science 279:870–873.
Angelman syndrome—insights into a rare epilepsy. J Clin Pharm Ther 40:135–143. Godavarthi SK, Dey P, Sharma A, Jana NR. 2015.
neurogenetic disorder. Nat Rev Neurol Dubose AJ, Smith EY, Yang TP, Johnstone KA, Impaired adult hippocampal neurogenesis
12:584–593. Resnick JL. 2011. A new deletion refines the and its partial reversal by chronic treatment
Cao C, Rioult-Pedotti MS, Migani P, Yu CJ, boundaries of the murine Prader-Willi of fluoxetine in a mouse model of Angelman
Tiwari R, Parang K, Spaller MR, Goebel syndrome imprinting center. Hum Mol syndrome. Biochem Biophys Res Commun
DJ, Marshall J. 2013. Impairment of TrkB- Genet 20:3461–3466. 464:1196–1201.
PSD-95 signaling in Angelman syndrome. Dziembowska M, Pretto DI, Janusz A, Kaczmarek Godinho BM, Malhotra M, O’Driscoll CM,
PLoS Biol 11:e1001478. L, Leigh MJ, Gabriel N, Durbin-Johnson B, Cryan JF. 2015. Delivering a disease-modi-
Chamberlain SJ. 2013. RNAs of the human Hagerman RJ, Tassone F. 2013. High fying treatment for Huntington’s disease.
chromosome 15q11-q13 imprinted region. MMP-9 activity levels in fragile X syndrome Drug Discov Today 20:50–64.
Wiley Interdiscip Rev RNA 4:155–166. are lowered by minocycline. Am J Med Goldman SE, Bichell TJ, Surdyka K, Malow BA.
Chamberlain SJ, Brannan CI. 2001. The Prader- Genet Part A 161A:1897–1903. 2012. Sleep in children and adolescents with
Willi syndrome imprinting center activates Dziembowska M, Wlodarczyk J. 2012. MMP9: A Angelman syndrome: Association with par-
the paternally expressed murine Ube3a novel function in synaptic plasticity. Int J ent sleep and stress. J Intellect Disabil Res
antisense transcript but represses paternal Biochem Cell Biol 44:709–713. 56:600–608.
Ube3a. Genomics 73:316–322. Egawa K, Kitagawa K, Inoue K, Takayama M, Greer PL, Hanayama R, Bloodgood BL, Mardinly
Chang PK, Verbich D, McKinney RA. 2012. Takayama C, Saitoh S, Kishino T, Kitagawa AR, Lipton DM, Flavell SW, Kim TK,
AMPA receptors as drug targets in neuro- M, Fukuda A. 2012. Decreased tonic Griffith EC, Waldon Z, Maehr R, Ploegh
logical disease-advantages, caveats, and fu- inhibition in cerebellar granule cells causes HL, Chowdhury S, Worley PF, Steen J,
ture outlook. Eur J Neurosci 35:1908–1916. motor dysfunction in a mouse model of Greenberg ME. 2010. The Angelman
Ciarlone SL, Grieco JC, D’Agostino DP, Weeber Angelman syndrome. Sci Transl Med Syndrome protein Ube3A regulates synapse
EJ. 2016. Ketone ester supplementation 4:163ra157. development by ubiquitinating arc. Cell
attenuates seizure activity, and improves Elgersma Y, Fedorov NB, Ikonen S, Choi ES, 140:704–716.
behavior and hippocampal synaptic plasticity Elgersma M, Carvalho OM, Giese KP, Silva Grieco JC, Ciarlone SL, Gieron-Korthals M,
in an Angelman syndrome mouse model. AJ. 2002. Inhibitory autophosphorylation of Schoenberg MR, Smith AG, Philpot RM,
Neurobiol Dis 96:38–46. CaMKII controls PSD association, plasticity, Heussler HS, Banko JL, Weeber EJ. 2014.
Conant KD, Thibert RL, Thiele EA. 2009. and learning. Neuron 36:493–505. An open-label pilot trial of minocycline in
Epilepsy and the sleep-wake patterns found Erickson CA, Wink LK, Baindu B, Ray B, children as a treatment for Angelman
in Angelman syndrome. Epilepsia Schaefer TL, Pedapati EV, Lahiri DK. 2016. syndrome. BMC Neurol 14:232.
50:2497–2500. Analysis of peripheral amyloid precursor Griffin MO, Fricovsky E, Ceballos G, Villarreal
Cullen SI, Cohan RH. 1976. Mino- protein in Angelman Syndrome. Am J Med F. 2010. Tetracyclines: A pleitropic family
cycline therapy in acne vulgaris. Cutis Genet Part A 170:2334–2337. of compounds with promising thera-
17:1208–1210,1214. Evangeliou A, Doulioglou V, Haidopoulou K, peutic properties. Review of the litera-
Dagli A, Buiting K, Williams CA. 2012. Molecu- Aptouramani M, Spilioti M, Varlamis G. ture. Am J Physiol Cell Physiol 299:
lar and clinical aspects of Angelman syn- 2010. Ketogenic diet in a patient with C539–C548.
drome. Mol Syndromol 2:100–112. Angelman syndrome. Pediatr Int Grigg-Damberger M, Ralls F. 2013. Treatment
Daily JL, Nash K, Jinwal U, Golde T, Rogers J, 52:831–834. strategies for complex behavioral insomnia
Peters MM, Burdine RD, Dickey C, Banko Farber C, Dittrich B, Buiting K, Horsthemke B. in children with neurodevelopmental dis-
JL, Weeber EJ. 2011. Adeno-associated 1999. The chromosome 15 imprinting orders. Curr Opin Pulm Med 19:616–625.
virus-mediated rescue of the cognitive centre (IC) region has undergone multiple Hemingway C, Freeman JM, Pillas DJ, Pyzik PL.
defects in a mouse model for Angelman duplication events and contains an upstream 2001. The ketogenic diet: A 3- to 6-year
syndrome. PLoS ONE 6:e27221. exon of SNRPN that is deleted in all follow-up of 150 children enrolled prospec-
Didden R, Korzilius H, Smits MG, Curfs LM. Angelman syndrome patients with an IC tively. Pediatrics 108:898–905.
2004. Sleep problems in individuals with microdeletion. Hum Mol Genet 8:337–343. Hethorn WR, Ciarlone SL, Filonova I, Rogers JT,
Angelman syndrome. Am J Ment Retard Fasinu PS, Phillips S, ElSohly MA, Walker LA. Aguirre D, Ramirez RA, Grieco JC, Peters
109:275–284. 2016. Current status and prospects for MM, Gulick D, Anderson AEJLB, Lussier
16 AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) RESEARCH REVIEW

AL, Weeber EJ. 2015. Reelin supplementa- YM, Nation J, Nordli DR Jr., Pfeifer HH, Marafini I, Di Fusco D, Calabrese E, Sedda S,
tion recovers synaptic plasticity and cogni- Rho JM, Stafstrom CE, Thiele EA, Turner Pallone F, Monteleone G. 2015. Antisense
tive deficits in a mouse model for Angelman Z, Wirrell EC, Wheless JW, Veggiotti P, approach to inflammatory bowel disease:
syndrome. Eur J Neurosci 41:1372–1380. Vining EP, Charlie Foundation PCotCNS. Prospects and challenges. Drugs
Hua Y, Vickers TA, Okunola HL, Bennett CF, 2009. Optimal clinical management of 75:723–730.
Krainer AR. 2008. Antisense masking of an children receiving the ketogenic diet: Rec- Margolis SS, Salogiannis J, Lipton DM, Mandel-
hnRNP A1/A2 intronic splicing silencer ommendations of the International Keto- Brehm C, Wills ZP, Mardinly AR, Hu L,
corrects SMN2 splicing in transgenic mice. genic Diet Study Group. Epilepsia Greer PL, Bikoff JB, Ho HY, Soskis MJ,
Am J Hum Genet 82:834–848. 50:304–317. Sahin M, Greenberg ME. 2010. EphB-
Huang HS, Allen JA, Mabb AM, King IF, Miriyala Kuhnle S, Mothes B, Matentzoglu K, Scheffner mediated degradation of the RhoA GEF
J, Taylor-Blake B, Sciaky N, Dutton JW Jr., M. 2013. Role of the ubiquitin ligase E6AP/ Ephexin5 relieves a developmental brake on
Lee HM, Chen X, Jin J, Bridges AS, Zylka UBE3A in controlling levels of the synaptic excitatory synapse formation. Cell
MJ, Roth BL, Philpot BD. 2012. Topoisom- protein Arc. Proc Natl Acad Sci USA 143:442–455.
erase inhibitors unsilence the dormant allele 110:8888–8893. Margolis SS, Sell GL, Zbinden MA, Bird LM.
of Ube3a in neurons. Nature 481:185–189. Kwon OB, Longart M, Vullhorst D, Hoffman DA, 2015. Angelman syndrome. Neurothera-
Huang HS, Burns AJ, Nonneman RJ, Baker LK, Buonanno A. 2005. Neuregulin-1 reverses peutics 12:641–650.
Riddick NV, Nikolova VD, Riday TT, long-term potentiation at CA1 hippocampal Martin K, Jackson CF, Levy RG, Cooper PN.
Yashiro K, Philpot BD, Moy SS. 2013. synapses. J Neurosci 25:9378–9383. 2016. Ketogenic diet and other dietary
Behavioral deficits in an Angelman syn- Kyllerman M. 1995. On the prevalence of treatments for epilepsy. Cochrane Database
drome model: Effects of genetic background Angelman syndrome. Am J Med Genet Syst Rev 2:CD001903.
and age. Behav Brain Res 243:79–90. 59:405. Meng L, Person RE, Huang W, Zhu PJ, Costa-
Huntley GW. 2012. Synaptic circuit remodelling Larson AM, Shinnick JE, Shaaya EA, Thiele EA, Mattioli M, Beaudet AL. 2013. Truncation
by matrix metalloproteinases in health and Thibert RL. 2015. Angelman syndrome in of Ube3a-ATS unsilences paternal Ube3a
disease. Nat Rev Neurosci 13:743–757. adulthood. Am J Med Genet A and ameliorates behavioral defects in the
Ingrassia A, Turk J. 2005. The use of clonidine for 167A:331–344. Angelman syndrome mouse model. PLoS
severe and intractable sleep problems in Lee V, Maguire J. 2014. The impact of tonic Genet 9:e1004039.
children with neurodevelopmental disor- GABAA receptor-mediated inhibition on Meng L, Ward AJ, Chun S, Bennett CF, Beaudet
ders-a case series. Eur Child Adolesc neuronal excitability varies across brain AL, Rigo F. 2015. Towards a therapy for
Psychiatry 14:34–40. region and cell type. Front Neural Circuits Angelman syndrome by targeting a long
Iulita MF, Do Carmo S, Ower AK, Fortress AM, 8:3. non-coding RNA. Nature 518:409–412.
Aguilar LF, Hanna M, Wisniewski T, Levy RG, Cooper PN, Giri P. 2012. Ketogenic Mertz LG, Christensen R, Vogel I, Hertz JM,
Granholm AC, Buhusi M, Busciglio J, diet and other dietary treatments for epilepsy. Nielsen KB, Gronskov K, Ostergaard JR.
Cuello AC. 2014. Nerve growth factor Cochrane Database Syst Rev 3:CD001903. 2013. Angelman syndrome in Denmark.
metabolic dysfunction in Down’s syndrome Lewis MW, Brant JO, Kramer JM, Moss JI, Yang birth incidence, genetic findings, and age at
brains. Brain 137:860–872. TP, Hansen PJ, Williams RS, Resnick JL. diagnosis. Am J Med Genet Part A
Jacobson RD, Feldman EL. 2016. Antisense 2015. Angelman syndrome imprinting cen- 161A:2197–2203.
oligonucleotides for duchenne muscular ter encodes a transcriptional promoter. Proc Miano S, Bruni O, Elia M, Musumeci SA, Verrillo
dystrophy: Why No neurologist should Natl Acad Sci USA 112:6871–6875. E, Ferri R. 2005. Sleep breathing and
skip this. JAMA Neurol 73:259–260. Lisman J, Schulman H, Cline H. 2002. The periodic leg movement pattern in Angelman
Jana NR. 2012. Understanding the pathogenesis molecular basis of CaMKII function in Syndrome: A polysomnographic study. Clin
of Angelman syndrome through animal synaptic and behavioural memory. Nat Neurophysiol 116:2685–2692.
models. Neural Plast 2012:710943. Rev Neurosci 3:175–190. Miano S, Bruni O, Leuzzi V, Elia M, Verrillo E,
Jensen L, Farook MF, Reiter LT. 2013. Proteomic Lizarraga SB, Morrow EM. 2015. Uncovering a Ferri R. 2004. Sleep polygraphy in Angel-
profiling in Drosophila reveals potential role for SK2 in Angelman syndrome. Cell man syndrome. Clin Neurophysiol
Dube3a regulation of the actin cytoskeleton Rep 12:359–360. 115:938–945.
and neuronal homeostasis. PLoS ONE 8: Llewellyn KJ, Nalbandian A, Gomez A, Wei D, Mis MS, Brajkovic S, Tafuri F, Bresolin N, Comi
e61952. Walker N, Kimonis VE. 2015. Administra- GP, Corti S. 2016. Development of thera-
Jiang YH, Armstrong D, Albrecht U, Atkins CM, tion of CoQ10 analogue ameliorates dys- peutics for C9ORF72 ALS/FTD-Related
Noebels JL, Eichele G, Sweatt JD, Beaudet function of the mitochondrial respiratory disorders. Mol Neurobiol DOI: 10.1007/
AL. 1998. Mutation of the Angelman chain in a mouse model of Angelman s12035-016-9993-0 [Epub ahead of print].
ubiquitin ligase in mice causes increased syndrome. Neurobiol Dis 76:77–86. Miura K, Kishino T, Li E, Webber H, Dikkes P,
cytoplasmic p53 and deficits of contextual Low D, Chen KS. 2010. Genome-wide gene Holmes GL, Wagstaff J. 2002. Neurobeha-
learning and long-term potentiation. Neu- expression profiling of the Angelman syn- vioral and electroencephalographic abnor-
ron 21:799–811. drome mice with Ube3a mutation. Eur J malities in Ube3a maternal-deficient mice.
Jonas M, Cunha BA. 1982. Minocycline. Ther Hum Genet 18:1228–1235. Neurobiol Dis 9:149–159.
Drug Monit 4:137–145. Luk HM, Lo IF. 2016. Angelman syndrome in Moreno M, Giralt E. 2015. Three valuable
Kaphzan H, Hernandez P, Jung JI, Cowansage Hong Kong chinese: A 20 years’ experience. peptides from bee and wasp venoms for
KK, Deinhardt K, Chao MV, Abel T, Klann Eur J Med Genet 59:315–319. therapeutic and biotechnological use: Me-
E. 2012. Reversal of impaired hippocampal Lynch G, Rex CS, Chen LY, Gall CM. 2008. The littin, apamin and mastoparan. Toxins
long-term potentiation and contextual fear substrates of memory: Defects, treatments, 7:1126–1150.
memory deficits in Angelman syndrome and enhancement. Eur J Pharmacol Neal EG, Chaffe H, Schwartz RH, Lawson MS,
model mice by ErbB inhibitors. Biol 585:2–13. Edwards N, Fitzsimmons G, Whitney A,
Psychiatry 72:182–190. Mabb AM, Simon JM, King IF, Lee HM, An LK, Cross JH. 2008. The ketogenic diet for the
King IF, Yandava CN, Mabb AM, Hsiao JS, Huang Philpot BD, Zylka MJ. 2016. Topoisomerase treatment of childhood epilepsy: A random-
HS, Pearson BL, Calabrese JM, Starmer J, 1 regulates gene expression in neurons ised controlled trial. Lancet Neurol
Parker JS, Magnuson T, Chamberlain SJ, through cleavage complex-dependent and 7:500–506.
Philpot BD, Zylka MJ. 2013. Topoisomer- Independent mechanisms. PloS ONE 11: Neal EG, Chaffe H, Schwartz RH, Lawson MS,
ases facilitate transcription of long genes e0156439. Edwards N, Fitzsimmons G, Whitney A,
linked to autism. Nature 501:58–62. Mandel-Brehm C, Salogiannis J, Dhamne SC, Cross JH. 2009. A randomized trial of
Kossoff EH, Zupec-Kania BA, Amark PE, Rotenberg A, Greenberg ME. 2015. Sei- classical and medium-chain triglyceride
Ballaban-Gil KR, Christina Bergqvist AG, zure-like activity in a juvenile Angelman ketogenic diets in the treatment of child-
Blackford R, Buchhalter JR, Caraballo RH, syndrome mouse model is attenuated by hood epilepsy. Epilepsia 50:1109–1117.
Helen Cross J, Dahlin MG, Donner EJ, reducing Arc expression. Proc Natl Acad Sci Oiglane-Shlik E, Talvik T, Zordania R, Poder H,
Klepper J, Jehle RS, Kim HD, Christiana Liu USA 112:5129–5134. Kahre T, Raukas E, Ilus T, Tasa G, Bartsch O,
RESEARCH REVIEW AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) 17

Vaisanen ML, Ounap K. 2006. Prevalence of supplementation enhances cognitive ability, mouse model for Angelman syndrome.
Angelman syndrome and Prader-Willi syn- synaptic plasticity, and dendritic spine Neurosci Lett 487:129–133.
drome in Estonian children: Sister syn- density. Learn Mem 18:558–564. Sun J, Liu Y, Moreno S, Baudry M, Bi X. 2015a.
dromes not equally represented. Am J Med Ruiz-Antora n B, Sancho L opez A, Cazorla R, Imbalanced mechanistic target of rapamycin
Genet Part A 140:1936–1943. Iglesias G, Lara J, Lopez Pajaro L, Marın E, C1 and C2 activity in the cerebellum of
Ostergaard JR, Balslev T. 2001. Efficacy of Avenda~ no-Sola C. 2015. Randomized clin- Angelman syndrome mice impairs motor
different antiepileptic drugs in children ical trial, placebo compared to evaluate the function. J Neurosci 35:4706–4718.
with Angelman syndrome associated with efficacy and safety of minocycline In Angel- Sun J, Zhu G, Liu Y, Standley S, Ji A, Tunuguntla
15q11-13 deletion: The Danish experience. man syndrome (A-Manece study). Clin R, Wang Y, Claus C, Luo Y, Baudry M, Bi
Dev Med Child Neurol 43:718–719. Ther 37:e154–e155. X. 2015b. UBE3A regulates synaptic plas-
Ovid Therapeutics Inc. 2016. Ovid therapeutics Santini E, Klann E. 2016. Genetically dissecting ticity and learning and memory by control-
receives orphan drug designation from cortical neurons involved in epilepsy in ling SK2 channel endocytosis. Cell Rep
the U.S. FDA for OV101 for the treatment Angelman syndrome. Neuron 90:1–3. 12:449–461.
of patients with Angelman syndrome. Up- Santini E, Turner KL, Ramaraj AB, Murphy MP, Sun J, Liu Y, Tran J, O’Neal P, Baudry M, Bi X.
dated: 2016 Sep 8. Accessed: 2016 Oct 22. Klann E, Kaphzan H. 2015. Mitochondrial 2016. MTORC1-S6K1 inhibition or
Available from: http://www.ovidrx.com/ superoxide contributes to hippocampal mTORC2 activation improves hippocampal
press-release/ovid-therapeutics-receives- synaptic dysfunction and memory deficits synaptic plasticity and learning in Angelman
orphan-drug-designation-from-the-u-s-fda- in Angelman syndrome model mice. syndrome mice. Cell Mol Life Sci
for-ov101-for-the-treatment-of-patients-with- J Neurosci 35:16213–16220. 73:4303–4314.
angelman-syndrome Santos RD, Raal FJ, Catapano AL, Witztum JL, Takaesu Y, Komada Y, Inoue Y. 2012. Melatonin
Panja D, Bramham CR. 2014. BDNF mechanisms Steinhagen-Thiessen E, Tsimikas S. 2015. profile and its relation to circadian rhythm
in late LTP formation: A synthesis and Mipomersen, an antisense oligonucleotide sleep disorders in Angelman syndrome
breakdown. Neuropharmacology 76 Pt to apolipoprotein B-100, reduces patients. Sleep Med 13:1164–1170.
C:664–676. lipoprotein(a) in various populations with Tan WH, Bird LM, Sadhwani A, Barbieri-Welge
Pelc K, Boyd SG, Cheron G, Dan B. 2008a. hypercholesterolemia: Results of 4 phase III R, Skinner SA, Horowitz LT, Bacino CA,
Epilepsy in Angelman syndrome. Seizure trials. Arterioscler Thromb Vasc Biol Noll LM, Fu C, Hundley RJ, Wink LK,
17:211–217. 35:689–699. Erickson CA, Barnes GN, Slavotinek A,
Pelc K, Cheron G, Boyd SG, Dan B. 2008b. Are Schwichtenberg AJ, Malow BA. 2015. Melatonin Jeremy R, Rotenberg A, Kothare SV, Olson
there distinctive sleep problems in Angelman treatment in children with developmental HE, Poduri A, Nespeca MP, Chu HC,
syndrome? Sleep Med 9:434–441. disabilities. Sleep Med Clin 10:181–187. Willen JM, Haas KF, Weeber EJ, Rufo PA.
Peters SU, Bird LM, Kimonis V, Glaze DG, Sell GL, Margolis SS. 2015. From UBE3A to 2016. Levodopa in Angelman syndrome—
Shinawi LM, Bichell TJ, Barbieri-Welge R, Angelman syndrome: A substrate perspec- outcome of a randomized controlled trial.
Nespeca M, Anselm I, Waisbren S, Sanborn tive. Front Neurosci 9:322. Am J Med Genet A Submitted.
E, Sun Q, O’Brien WE, Beaudet AL, Shaaya EA, Grocott OR, Laing O, Thibert RL. Terstappen GC, Pellacani A, Aldegheri L, Graziani
Bacino CA. 2010. Double-blind therapeutic 2016. Seizure treatment in Angelman syn- F, Carignani C, Pula G, Virginio C. 2003.
trial in Angelman syndrome using betaine drome: A case series from the Angelman The antidepressant fluoxetine blocks the
and folic acid. Am J Med Genet Part A syndrome clinic at massachusetts general human small conductance calcium-activated
152A:1994–2001. hospital. Epilepsy Behav 60:138–141. potassium channels SK1, SK2 and SK3.
Petersen MB, Brondum-Nielsen K, Hansen LK, Shimizu-Motohashi Y, Miyatake S, Komaki H, Neurosci Lett 346:85–88.
Wulff K. 1995. Clinical, cytogenetic, and Takeda S, Aoki Y. 2016. Recent advances in Thibert RL, Conant KD, Braun EK, Bruno P, Said
molecular diagnosis of Angelman syndrome: innovative therapeutic approaches for Duch- RR, Nespeca MP, Thiele EA. 2009.
Estimated prevalence rate in a Danish enne muscular dystrophy: From discovery to Epilepsy in Angelman syndrome: A ques-
county. Am J Med Genet 60:261–262. clinical trials. Am J Transl Res 8:2471–2489. tionnaire-based assessment of the natural
Picher-Martel V, Valdmanis PN, Gould PV, Julien Sidhu H, Dansie LE, Hickmott PW, Ethell DW, history and current treatment options.
JP, Dupre N. 2016. From animal models to Ethell IM. 2014. Genetic removal of matrix Epilepsia 50:2369–2376.
human disease: A genetic approach for metalloproteinase 9 rescues the symptoms of Thibert RL, Larson AM, Hsieh DT, Raby AR,
personalized medicine in ALS. Acta Neuro- fragile X syndrome in a mouse model. Thiele EA. 2013. Neurologic manifestations
pathol Commun 4:70. J Neurosci 34:9867–9879. of Angelman syndrome. Pediatric Neurol
Pitcher GM, Beggs S, Woo RS, Mei L, Salter MW. Silva-Santos S, van Woerden GM, Bruinsma CF, 48:271–279.
2008. ErbB4 is a suppressor of long-term Mientjes E, Jolfaei MA, Distel B, Kushner Thibert RL, Pfeifer HH, Larson AM, Raby AR,
potentiation in the adult hippocampus. SA, Elgersma Y. 2015. Ube3a reinstatement Reynolds AA, Morgan AK, Thiele EA.
Neuroreport 19:139–143. identifies distinct developmental windows in 2012. Low glycemic index treatment for
Plasschaert RN, Bartolomei MS. 2013. Autism: A a murine Angelman syndrome model. J Clin seizures in Angelman syndrome. Epilepsia
long genetic explanation. Nature Invest 125:2069–2076. 53:1498–1502.
501:36–37. Simmons DA, Rex CS, Palmer L, Pandyarajan V, Vafadari B, Salamian A, Kaczmarek L. 2015.
Powell WT, Coulson RL, Gonzales ML, Crary Fedulov V, Gall CM, Lynch G. 2009. Up- MMP-9 in translation: From molecule to
FK, Wong SS, Adams S, Ach RA, Tsang P, regulating BDNF with an ampakine rescues brain physiology, pathology and therapy.
Yamada NA, Yasui DH, Chedin F, LaSalle synaptic plasticity and memory in Hunting- J Neurochem DOI: 10.1111/jnc.13415
JM. 2013. R-loop formation at Snord116 ton’s disease knockin mice. Proc Natl Acad [Epub ahead of print].
mediates topotecan inhibition of Ube3a- Sci USA 106:4906–4911. Valente KD, Koiffmann CP, Fridman C, Varella M,
antisense and allele-specific chromatin de- Skourti-Stathaki K, Proudfoot NJ. 2014. Kok F, Andrade JQ, Grossmann RM,
condensation. Proc Natl Acad Sci USA A double-edged sword: R loops as threats Marques-Dias MJ. 2006. Epilepsy in patients
110:13938–13943. to genome integrity and powerful regulators with Angelman syndrome caused by dele-
Robinson-Shelton A, Malow BA. 2016. Sleep of gene expression. Genes Dev tion of the chromosome 15q11-13. Arch
disturbances in neurodevelopmental disor- 28:1384–1396. Neurol 63:122–128.
ders. Curr Psychiatry Rep 18:6. Stein D, Chetty M, Rho JM. 2010. A “happy” Valente KD, Varela MC, Koiffmann CP, Andrade
Roden WH, Peugh LD, Jansen LA. 2010. Altered toddler presenting with sudden, life-threat- JQ, Grossmann R, Kok F, Marques-Dias MJ.
GABA(A) receptor subunit expression and ening seizures. Semin Pediatr Neurol 2013. Angelman syndrome caused by dele-
pharmacology in human Angelman syn- 17:35–38. tion: A genotype-phenotype correlation
drome cortex. Neurosci Lett 483:167–172. Su H, Fan W, Coskun PE, Vesa J, Gold JA, Jiang determined by breakpoint. Epilepsy Res
Rogers JT, Rusiana I, Trotter J, Zhao L, YH, Potluri P, Procaccio V, Acab A, Weiss 105:234–239.
Donaldson E, Pak DT, Babus LW, Peters JH, Wallace DC, Kimonis VE. 2011. Van den Veyver IB. 2002. Genetic effects of
M, Banko JL, Chavis P, Rebeck GW, Hoe Mitochondrial dysfunction in CA1 hippo- methylation diets. Ann Rev Nutr
HS, Weeber EJ. 2011. Reelin campal neurons of the UBE3A deficient 22:255–282.
18 AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS) RESEARCH REVIEW

van Woerden GM, Harris KD, Hojjati MR, mental retardation syndrome. J Neurosci required for experience-dependent matura-
Gustin RM, Qiu S, de Avila Freire R, Jiang 23:2634–2644. tion of the neocortex. Nat Neurosci
YH, Elgersma Y, Weeber EJ. 2007. Rescue Williams CA, Beaudet AL, Clayton-Smith J, 12:777–783.
of neurological deficits in a mouse model for Knoll JH, Kyllerman M, Laan LA, Magenis Yoshii A, Constantine-Paton M. 2007. BDNF
Angelman syndrome by reduction of alpha- RE, Moncla A, Schinzel AA, Summers JA, induces transport of PSD-95 to dendrites
CaMKII inhibitory phosphorylation. Nat Wagstaff J. 2006. Angelman syndrome 2005: through PI3K-AKT signaling after NMDA
Neurosci 10:280–282. Updated consensus for diagnostic criteria. receptor activation. Nat Neurosci
Walz NC, Beebe D, Byars K. 2005. Sleep in Am J Med Genet A 140:413–418. 10:702–711.
individuals with Angelman syndrome: Par- Wink LK, Fitzpatrick S, Shaffer R, Melnyk S, Yoshii A, Murata Y, Kim J, Zhang C, Shokat KM,
ent perceptions of patterns and problems. Begtrup AH, Fox E, Schaefer TL, Mathieu- Constantine-Paton M. 2011. TrkB and
Am J Ment Retard 110:243–252. Frasier L, Ray B, Lahiri D, Horn PA, Erickson protein kinase Mzeta regulate synaptic
Weeber EJ, Jiang YH, Elgersma Y, Varga AW, CA. 2015. The neurobehavioral and molec- localization of PSD-95 in developing cortex.
Carrasquillo Y, Brown SE, Christian JM, ular phenotype of Angelman Syndrome. Am J Neurosci 31:11894–11904.
Mirnikjoo B, Silva A, Beaudet AL, Sweatt J Med Genet Part A 167A:2623–2628. Zhdanova IV, Wurtman RJ, Wagstaff J. 1999.
JD. 2003. Derangements of hippocampal Yashiro K, Riday TT, Condon KH, Roberts AC, Effects of a low dose of melatonin on sleep in
calcium/calmodulin-dependent protein ki- Bernardo DR, Prakash R, Weinberg RJ, children with Angelman syndrome.
nase II in a mouse model for Angelman Ehlers MD, Philpot BD. 2009. Ube3a is J Pediatric Endocrinol Metab 12:57–67.

Anda mungkin juga menyukai