Skip to main content
Erschienen in: Molecular Autism 1/2018

Open Access 01.12.2018 | Research

Delineation of the genetic and clinical spectrum of Phelan-McDermid syndrome caused by SHANK3 point mutations

verfasst von: Silvia De Rubeis, Paige M. Siper, Allison Durkin, Jordana Weissman, François Muratet, Danielle Halpern, Maria del Pilar Trelles, Yitzchak Frank, Reymundo Lozano, A. Ting Wang, J. Lloyd Holder Jr, Catalina Betancur, Joseph D. Buxbaum, Alexander Kolevzon

Erschienen in: Molecular Autism | Ausgabe 1/2018

Abstract

Background

Phelan-McDermid syndrome (PMS) is a neurodevelopmental disorder characterized by psychiatric and neurological features. Most reported cases are caused by 22q13.3 deletions, leading to SHANK3 haploinsufficiency, but also usually encompassing many other genes. While the number of point mutations identified in SHANK3 has increased in recent years due to large-scale sequencing studies, systematic studies describing the phenotype of individuals harboring such mutations are lacking.

Methods

We provide detailed clinical and genetic data on 17 individuals carrying mutations in SHANK3. We also review 60 previously reported patients with pathogenic or likely pathogenic SHANK3 variants, often lacking detailed phenotypic information.

Results

SHANK3 mutations in our cohort and in previously reported cases were distributed throughout the protein; the majority were truncating and all were compatible with de novo inheritance. Despite substantial allelic heterogeneity, four variants were recurrent (p.Leu1142Valfs*153, p.Ala1227Glyfs*69, p.Arg1255Leufs*25, and c.2265+1G>A), suggesting that these are hotspots for de novo mutations. All individuals studied had intellectual disability, and autism spectrum disorder was prevalent (73%). Severe speech deficits were common, but in contrast to individuals with 22q13.3 deletions, the majority developed single words, including 41% with at least phrase speech. Other common findings were consistent with reports among individuals with 22q13.3 deletions, including hypotonia, motor skill deficits, regression, seizures, brain abnormalities, mild dysmorphic features, and feeding and gastrointestinal problems.

Conclusions

Haploinsufficiency of SHANK3 resulting from point mutations is sufficient to cause a broad range of features associated with PMS. Our findings expand the molecular and phenotypic spectrum of PMS caused by SHANK3 point mutations and suggest that, in general, speech impairment and motor deficits are more severe in the case of deletions. In contrast, renal abnormalities associated with 22q13.3 deletions do not appear to be related to the loss of SHANK3.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13229-018-0205-9) contains supplementary material, which is available to authorized users.
Catalina Betancur, Joseph D. Buxbaum, and Alexander Kolevzon are co-senior authors.
Abkürzungen
ACMG
American College of Medical Genetics and Genomics
ADI-R
Autism Diagnostic Interview-Revised
ADOS-2
Autism Diagnostic Observation Schedule: Second Edition
ASD
Autism spectrum disorder
DAS-II
Differential Ability Scales, Second Edition
DSM-5
Diagnostic and Statistical Manual for Mental Disorders: Fifth Edition
EEG
Electroencephalography
EVS
Exome Variant Server
gnomAD
Genome Aggregation Database
ID
Intellectual disability
IQ
Intellectual quotient
MRI
Magnetic resonance imaging
OMIM
Online Mendelian Inheritance in Man
PMS
Phelan-McDermid syndrome

Background

Phelan-McDermid syndrome (PMS, OMIM 606232) is a rare neurodevelopmental disorder characterized by neonatal hypotonia, global developmental delay, intellectual disability (ID), severely delayed or absent speech, and frequent autism spectrum disorder (ASD) [1]. The neurobehavioral phenotype of PMS is usually severe. In a prospective study of 32 PMS individuals, 77% manifested severe-to-profound ID and 84% met criteria for ASD using gold standard diagnostic tools [2]. Dysmorphic features are usually mild and include long eyelashes, large or prominent ears, bulbous nose, pointed chin, fleshy hands, and dysplastic toenails [1]. Additional features include gastrointestinal problems, seizures, motor deficits, structural brain abnormalities, renal malformations, lymphedema, and recurrent infections [1].
The major neurodevelopmental features of PMS are caused by deletions or mutations of the SHANK3 gene, which encodes a scaffolding protein of the postsynaptic density of glutamatergic synapses. Most reported cases of PMS are caused by 22q13.3 deletions, which usually encompass many genes and can extend up to 9.2 Mb [24]. Genotype-phenotype analyses indicate that the size of the deletion and the number and/or severity of clinical manifestations are positively correlated [2, 47]. Specifically, correlations have been reported between deletion size and hypotonia [57], developmental delay [57], dysmorphic features [2, 7], speech abilities [4], social communication deficits related to ASD [2], and other medical conditions [2]. Furthermore, individuals with small terminal deletions may have more favorable developmental trajectories than those with larger deletions [8].
De novo truncating and missense mutations in SHANK3 have been identified in cohorts ascertained for ASD [916] or ID [1721]. In addition, there is a single report of two families ascertained for schizophrenia with mutations in SHANK3; affected individuals also had ID [22]. Despite the increasing number of mutations in SHANK3, their prevalence in PMS and more broadly in ASD is underestimated because clinical sequencing is still uncommon compared to chromosomal microarray. In addition, SHANK3 has been poorly covered by whole exome sequencing due to high GC content [13, 23], and there is little in the PMS phenotype that would prompt a clinician to specifically target SHANK3 for optimized Sanger sequencing. We and others estimate that SHANK3 haploinsufficiency might account for up to 1% of more severely affected ASD cases [13, 23].
Given the dearth of identified cases with SHANK3 mutations, analyses of PMS cohorts have largely focused on individuals with 22q13.3 deletions [28, 24]. Only two studies on PMS have included a few individuals carrying SHANK3 mutations [2, 24]. These observations have been complemented by the description of a small number of individuals identified through SHANK3 targeted sequencing in ASD cohorts [913]. Large-scale sequencing studies have been instrumental in revealing additional SHANK3 mutations but have not provided detailed phenotypic information [1416, 1921].
The limited number of subjects with SHANK3 mutations examined thus far, and the lack of systematic clinical evaluation have hindered the characterization of the phenotypic spectrum associated with SHANK3 mutations. Here, we aimed to delineate the genetic spectrum of SHANK3 mutations and their associated phenotype in relationship to PMS features.

Methods

Participants

The study includes 14 participants (S1–S14) enrolled at the Seaver Autism Center for Research and Treatment at the Icahn School of Medicine at Mount Sinai, and three individuals (B1–B3) evaluated at Baylor College of Medicine. Individuals were referred through the Phelan-McDermid Syndrome Foundation, ongoing research studies, and communication between families. The study was approved by the Program for the Protection of Human Subjects at the Icahn School of Medicine at Mount Sinai and the Baylor College of Medicine Institutional Review Board. Parents or legal guardians provided informed consent for participation and publication. Consent was also obtained to publish the photos shown in Fig. 1.

Genetic testing

All mutations were identified and/or validated by Clinical Laboratory Improvement Amendments (CLIA)-certified laboratories. The mutation in individual S1 was identified by whole exome sequencing (WES) and then validated by Sanger sequencing at the Seaver Autism Center [2] and by GeneDx. The mutation in S2 was identified by panel sequencing at the Michigan Medical Genetics Laboratories. The mutation in S3 was identified and validated at Seaver as previously reported [2] and further confirmed by Athena Diagnostics. The mutation in S4 was identified through clinical WES by the Columbia University Laboratory of Personalized Medicine. The mutations in S5, S11, and B1 were identified through clinical WES by the Medical Genetics Laboratory at the Baylor College of Medicine. The mutations in S6, S7, S9, S10, S12, and S14 were identified through clinical WES by GeneDx. The mutation in S8 was identified through clinical WES by AmbryGenetics. The variants in S13 were identified at the Seaver Autism Center and confirmed by GeneDx. The mutation in B2 and B3 was identified through clinical WES by Transgenomic.
Variants were described according to the Human Genome Variation Society guidelines. As reported previously [2], the human genome reference assembly (GRCh37/hg19 and GRCh38/hg38) is missing the beginning of exon 11 (NM_033517.1:c.1305_1346, 5′-cccgagcgggcccggcggccccggccccgcgcccggccccgg-3′, coding for 436-PSGPGGPGPAPGPG-449). We numbered nucleotide and amino acid positions according to the SHANK3 RefSeq mRNA (NM_033517.1) and protein (NP_277052.1) sequence, in which this mistake has been corrected. Variants were interpreted according to the American College of Medical Genetics and Genomics (ACMG) guidelines [25].

Review of previously reported SHANK3 mutations

We searched the literature for pathogenic or likely pathogenic mutations in SHANK3 and retrieved the molecular and clinical information (Additional file 1: Tables S1–S3). We also included mutations reported in ClinVar (http://​www.​ncbi.​nlm.​nih.​gov/​clinvar/​). To avoid duplicate counting of affected individuals, we reviewed all available information (including gender, country of origin, and phenotype) and contacted the authors when doubts persisted. Individuals reported more than once are indicated in Additional file 1: Table S1.

Clinical evaluation

Prospective clinical and psychological characterization was completed for 12 individuals seen at the Seaver Autism Center (S1–S4, S6–S8, S10–S14), including three previously reported (S1 and S3 [2] and S13 [26]). A battery of standardized assessments was used to examine ASD, intellectual functioning, adaptive behavior, language, motor skills, and sensory processing (see below). The medical evaluation included psychiatric, neurological, and clinical genetics examinations and medical record review. The evaluation of the individuals seen at Baylor College of Medicine (B1–B3) included parent interview, neurological examination, and medical record review. Their seizure phenotype and brain magnetic resonance imaging (MRI) findings were reported previously [24]. Two additional individuals (S5 and S9) received genetic testing through the Seaver Autism Center but were not evaluated clinically. Their caregivers completed surveys to capture developmental, medical, and behavioral health issues and were interviewed by phone.

ASD phenotype

Gold-standard ASD diagnostic testing included the Autism Diagnostic Observation Schedule, Second Edition (ADOS-2) [27], the Autism Diagnostic Interview-Revised (ADI-R) [28], and a clinical evaluation to assess Diagnostic and Statistical Manual for Mental Disorders, Fifth Edition (DSM-5) criteria for ASD [29]. The ADOS-2 and ADI-R were administered and scored by research reliable raters, and the psychiatric evaluation was completed by a board-certified child and adolescent psychiatrist. The ADOS-2 is a semi-structured observational assessment that provides scores in the domains of social affect, restricted and repetitive behavior, and a total score. A comparison score ranging from 1 to 10, with higher scores reflecting a greater number of symptoms, was calculated to examine symptom severity within each ADOS-2 domain and in total [30]. Nine individuals (S1–S4, S6, S8, S11, S13, S14) received module 1 of the ADOS, for children who are nonverbal or communicate using single words. Two individuals (S7, S10) received module 3, for children who are verbally fluent. The ADI-R is a structured caregiver interview that assesses ASD symptomatology within the domains of socialization, communication, and repetitive and restricted interests and behavior. A consensus diagnosis was determined for each participant based on results from the ADOS-2, ADI-R, and clinical evaluation using DSM-5.

Intellectual functioning

Global cognitive ability was measured using the Mullen Scales of Early Learning [31] (n = 10), the Stanford Binet Intelligence Scales, Fifth Edition [32] (n = 1), and the Differential Ability Scales, Second Edition (DAS-II) [33] (n = 1), depending on age and verbal ability. The Mullen is validated for children from birth to 68 months but is commonly used for older individuals with ID [34]. Developmental quotients were calculated using age equivalents divided by chronological age as has been done in previous studies [35]. For example, a nonverbal developmental quotient was computed by dividing the mean age equivalents on the visual reception and fine motor scales by the child’s chronological age and then multiplying by 100. The DAS-II is a measure of cognitive functioning that assesses a child’s verbal reasoning, nonverbal reasoning, and spatial abilities. A general conceptual ability can be calculated to assess overall intellectual functioning. The Stanford-Binet Intelligence Scales, Fifth Edition is an intelligence test that produces a nonverbal intellectual quotient (IQ), verbal IQ, and full scale IQ based on performance across five scales: fluid reasoning, knowledge, quantitative reasoning, visual-spatial, and working memory.

Adaptive behavior

The Vineland Adaptive Behavior Scales, Second Edition, Survey Interview Form (Vineland-II) [36] is a clinician-administered interview that assesses adaptive behavior in the domains of communication, daily living, socialization, and motor skills. The Vineland-II was completed for 11 individuals. The motor domain is intended for children ages 6 years and under but was assessed in all individuals given significant motor delays in this population. The Vineland-II was also used in conjunction with cognitive testing to identify the presence and severity of ID.

Language skills

Language milestones were assessed during the ADI-R (n = 11) and the psychiatric evaluation. Current expressive and receptive language abilities were assessed using the Mullen (n = 10), Vineland-II (n = 11), MacArthur-Bates Communicative Development Inventories [37] (n = 10), Peabody Picture Vocabulary Test, Fourth Edition [38] (n = 3), and Expressive Vocabulary Test [39] (n = 2).

Motor skills

Motor milestones were assessed during the ADI-R (n = 11) and the psychiatric evaluation (n = 12). Current motor skills were assessed using the Vineland-II (n = 11) and Mullen (n = 10) fine and gross motor skills domains. The Beery Visual-Motor Integration Test, 6th Edition [40] was completed when appropriate (n = 2).

Sensory processing

Sensory processing was assessed using the Short Sensory Profile [41] and the Sensory Assessment for Neurodevelopmental Disorders (SAND) [42]. The Short Sensory Profile is a 38-item caregiver report form that investigates daily life sensory experiences. The SAND is a standardized assessment that includes a clinician-administered observation and a 36-item corresponding caregiver interview. The scoring algorithm measures sensory hyperreactivity, hyporeactivity, and seeking behavior across visual, tactile, and auditory domains.

Results

SHANK3 mutations

We report 17 individuals (including two monozygotic twins) with SHANK3 mutations identified through WES or panel sequencing. The variants were distributed throughout the protein and included 13 frameshift, two nonsense, and one missense mutation (Table 1, Fig. 1a). Notably, we observed an identical frameshift mutation, c.3679dupG (p.Ala1227Glyfs*69), in three unrelated individuals. Mutations were confirmed to be de novo in 15 individuals and non-paternal or non-maternal in two (no DNA was available from the other two parents). In addition to a nonsense mutation, individual S13 carries a missense variant (p.Ser1291Leu) absent in the mother but present in the unaffected sister and in four individuals in the Genome Aggregation Database (gnomAD), suggesting it is likely benign, despite being predicted as damaging by several in silico tools (Additional file 1: Table S3). All other mutations are absent from the Exome Variant Server (EVS) and gnomAD. The missense mutation in S14 (p.Asp1672Tyr) affects a highly conserved residue and is predicted to be damaging by all algorithms used, including Polyphen-2, SIFT, PANTHER, MutPred2, Condel2, CADD, and M-CAP (Additional file 1: Table S3).
Table 1
SHANK3 point mutations in 17 individuals described in this study
ID
Coding DNA changea
Protein changeb
Genomic change (hg19)
Location
Effect
Inheritance
Variant classification [25]
S1c
c.1527G>A
p.Trp509*
chr22:g.51137146G>A
Exon 12
Nonsense
De novo
Pathogenic
S2
c.2471delC
p.Pro824Argfs*69
chr22:g.51158732delC
Exon 21
Frameshift
De novo
Pathogenic
S3d
c.2499delG
p.Pro834Argfs*59
chr22:g.51158760delG
Exon 21
Frameshift
De novo
Pathogenic
S4
c.2946_2949delCCGC
p.Arg983Serfs*94
chr22:g.51159207_51159210delCCGC
Exon 21
Frameshift
De novo
Pathogenic
S5
c.3095_3107delTGGGGGCCATCGA
p.Val1032Glyfs*42
chr22:g.51159356_51159368delTGGGGGCCATCGA
Exon 21
Frameshift
De novo
Pathogenic
S6
c.3424_3425delCT
p.Leu1142Valfs*153
chr22:g.51159685_51159686delCT
Exon 21
Frameshift
De novo
Pathogenic
S7
c.3679dupG
p.Ala1227Glyfs*69
chr22:g.51159940dupG
Exon 21
Frameshift
Non-paternal
Pathogenic
S8
c.3679dupG
p.Ala1227Glyfs*69
chr22:g.51159940dupG
Exon 21
Frameshift
De novo
Pathogenic
B1e
c.3679dupG
p.Ala1227Glyfs*69
chr22:g.51159940dupG
Exon 21
Frameshift
De novo
Pathogenic
S9
c.3764_3776delGGGCCCAGCCCCC
p.Arg1255Leufs*25
chr22:g.51160025_51160037delGGGCCCAGCCCCC
Exon 21
Frameshift
De novo
Pathogenic
B2, B3e,f
c.4065_4066delTG
p.Val1357Glyfs*4
chr22:g.51160326_51160327delTG
Exon 21
Frameshift
De novo
Pathogenic
S10
c.4229delC
p.Pro1410Hisfs*18
chr22:g.51160490delC
Exon 21
Frameshift
De novo
Pathogenic
S11
c.4577_4578delCC
p.Ala1526Glufs*16
chr22:g.51160838_51160839delCC
Exon 22
Frameshift
De novo
Pathogenic
S12
c.4906_4921dupTCCCCCTCGCCGTCGC
p.Pro1641Leufs*58
chr22:g.51169450_51169465dupTCCCCCTCGCCGTCGC
Exon 22
Frameshift
De novo
Pathogenic
S13g
c.5008A>T
p.Lys1670*
chr22:g.51169552A>T
Exon 22
Nonsense
Non-maternal
Likely pathogenic
c.3872C>T
p.Ser1291Leu
chr22:g.51160133C>T
Exon 21
Missense
Non-maternal
Likely benign
S14
c.5014G>T
p.Asp1672Tyr
chr22:g.51169558G>T
Exon 22
Missense
De novo
Likely pathogenic
aNM_033517.1
bNP_277052.1 (Q9BYB0-1)
cS1 also has a de novo pathogenic 17q12 microduplication [62]. Reported previously [2, 14, 26]
dReported previously [2, 26]
eReported previously [24]
fMonozygotic twins
gReported previously [26]. This individual has two variants in SHANK3; the missense variant is likely benign and is not shown in Fig. 1a
We also searched the literature and ClinVar for SHANK3 mutations and assessed their pathogenicity. Variants listed in Additional file 1: Table S1 meet the following criteria: (1) loss-of-function variants (frameshift, nonsense, and splice site), or de novo missense variants predicted to be deleterious by several bioinformatics predictors, and (2) absent from control databases (EVS and gnomAD). After removing cases ascertained or reported multiple times, we identified 60 additional individuals from 55 families with SHANK3 mutations classified as pathogenic or likely pathogenic according to ACMG [25]. All the mutations with parental samples available were de novo. Three families had multiple affected siblings, consistent with germline mosaicism [9, 22, 43]. Four de novo missense variants reported in children with ASD, ID, or infantile spasms (p.Thr337Ser, p.Ser1197Gly, p.Ala1214Pro, and p.Arg1255Gly) [15, 4446] were classified as variants of uncertain significance because, although not present in controls, in silico predictions did not provide consistent evidence for pathogenicity (Additional file 1: Tables S1, S3). Given that SHANK3 is highly constrained against missense variation (Exome Aggregation Consortium Z score 4.92) [47], further studies are needed to determine the pathogenicity of these and other missense variants.
Three of the mutations in our cohort are recurrent, having been previously observed in unrelated individuals (Fig. 1a, Additional file 1: Table S1). The mutation in S6, p.Leu1142Valfs*153, was reported in a boy with ASD [13]. The mutation c.3679dupG (p.Ala1227Glyfs*69), shared by three of our patients (S7, S8, B1), is within a stretch of eight guanines and has been identified in three independent cases [9, 15, 20]. p.Arg1255Leufs*25, present in S9, has been reported in three unrelated patients [13, 21]. The donor splice site at position c.2265+1 is another hotspot: there are three individuals with a G>A substitution [16, 24, 48], and one with a deletion of the same G (c.2265+1delG), shown to result in a frameshift (p.Ser755Serfs*1) [11]. Overall, there were four recurrent and 56 private pathogenic/likely pathogenic mutations in SHANK3 (Fig. 1a, Additional file 1: Table S1).
We also searched for potentially deleterious variants inherited from unaffected parents or present in population controls (Additional file 1: Table S4). An inherited frameshift variant reported as pathogenic in two unrelated children with ASD [12, 49], and classified as damaging in the Human Gene Mutation Database, is in fact intronic when annotated in the correct reference sequence, NM_033517.1 [49], and is present 173 times in gnomAD (chr22:g.51135705dupG, hg19). An inherited substitution in a splice region (c.1772-4G>A) reported in ASD [12] is present seven times in gnomAD and is thus unlikely to be deleterious. gnomAD contains 21 variants predicted to be loss-of-function when annotated in the Ensembl canonical transcript ENST00000262795 (which is missing the beginning of exon 11 and contains three extra, unvalidated exons). When annotated in NM_033517.1, many of these variants are in fact intronic. The remaining 10 loss-of-function variants are all singletons; seven are flagged because they were found in sites covered in a limited number of individuals, which may indicate low-quality sites, one is located at the extreme 3′ end, and one has an abnormal allele balance. These findings confirm that truncating variants in SHANK3 are highly penetrant and unlikely to be present in unaffected individuals.
Four in-frame deletions [10, 13, 19, 50] and one in-frame insertion [50] in SHANK3 have been reported in ASD/ID (Additional file 1: Table S4). Three of these variants were inherited [10, 13, 50], and one was found in two controls [50], suggesting that some short in-frame deletions or insertions may be tolerated. An in-frame deletion of five amino acids (p.Gly1453_Ala1457del) reported in an ASD proband and his unaffected mother [10] was detected in six individuals in the gnomAD database. gnomAD lists 15 in-frame deletions or insertions (after annotation in NM_033517.1); six are on multiallelic sites, and four others are flagged because of low coverage. Among the remaining in-frame variants, p.Glu1230del was observed in five individuals and p.Gly1518del in four (Additional file 1: Table S4). These findings indicate that at least some in-frame variants in SHANK3 can be present in seemingly unaffected individuals.

Clinical phenotype of SHANK3 haploinsufficiency

Phenotypic spectrum in the individuals from our cohort

Detailed clinical information of the 17 individuals (9 males and 8 females, 3–42 years old at evaluation) is summarized in Tables 2 and 3 and Additional file 2: Table S5.
Table 2
Main clinical features of individuals with SHANK3 mutations
 
S1
S2
S3
S4
S5a
S6
S7
S8
S9a
S10
S11
S12
S13
S14
B1
B2b
B3b
Total (%)
Gender
M
M
M
F
F
M
F
M
M
M
F
F
M
M
F
F
F
9 M, 8 F
Gestational age (wks)
36
39
40
Term
38
Term
33
40
39
Term
36.5
Term
40
41
40
36
36
 
Birth weight (g)
2863
3400
3657
4000
3175
2948
1940
4111
4630
3230
3000
3728
2700
3090
2438
2551
2523
 
Birth length (cm)
47
55
NK
53
NK
50
46
58
55
50
53
NK
48
51
48
NK
NK
 
Postnatal growth
         
 Age at examination (y)
12
5
7
3
9
5
7
9
12
9
6
42
15
4
14
14
14
3–42
 Height (cm, percentile)
131 (< 1)
110 (27)
108 (< 1)
103 (93)
NK
109 (50)
125 (45)
148 (95)
142 (17)
135 (56)
111 (4)
170 (85)
149 (< 1)
97 (4)
151 (25–50)
145 (< 1)
145 (< 1)
5/16 (31%) short
 Weight (kg, percentile)
32.6 (11)
20 (53)
18.8 (5)
17.2 (88)
NK
17.3 (50)
35 (96)
36 (78)
29.5 (3)
29.5 (55)
18.2 (7)
69.4 (82)
48.1 (18)
15.4 (19)
55.1 (75)
59.1 (75)
40.5 (12)
 
 OFC (cm, percentile)
56 (95)
50.8 (36)
52.7 (70)
50 (75)
NK
NK
51.2 (40)
54 (82)
NK
53 (64)
50.5 (27)
57 (99)
54.5 (40)
46 (< 1)
52.7 (30)
57 (98)
57 (98)
3/14 (21%) macrocephaly; 1 (7%) microcephaly
Psychomotor development
 Sat independently (mo)
12
5–6
9–10
6
12
9
8–11
6
8–9
6
Normal
8
NK
5
8
6
6
 
 Walked independently (mo)
24
16
14
15
16
14
18
14
14–15
12
24
20
14
13
14
19
19
 
 First words and current language ability
3 y; now non-verbal
No speech; vowel sounds and sounds of pleasure present
No speech; previously had 10 signs but regressed to 3–4, babbling present, apraxic
No speech; uses about 5 signs with word approximations
Non-verbal
36 mo; 2-word phrases at 3.5 y; primarily single words with some phrase speech, stereotyped speech and echolalia
14 mo; first phrases at 24 mo, verbally fluent, complex speech
15 mo; never developed 2-word phrases, eventually lost all words
Non-verbal
8 mo; 2-word phrases by 24 mo, now verbally fluent
4 y; had approximately 10 words; currently uses no words
15 mo; phrase speech at 3 y; was verbally fluent until 12–13 y but currently uses no words
10–15 words by 18 mo; understands roughly 40 signs; comprehension and expressive language is limited
10 mo; uses a few word approximations, some signs, apraxic
At 3 y had approximately 200 words but only used 50 routinely; can speak in 2–3 word sentences but mostly echolalia
19 mo; combined words at 3.5 y; currently speaks in full sentences but developed word finding difficulties
19 mo; combined words at 3.5 y; spoke in full sentences but regressed at 9 y to only say 2–3 words, regained some vocabulary but fluctuating language
Currently non-verbal 9/17 (53%), fluent speech 3/17 (18%)
 Intellectual disability (IQ or DQ)
Profound ID (Mullen: DQ 6.7, NVDQ 10.3, VDQ 3.1)
Profound ID (Mullen: DQ 21.3, NVDQ 30.1, VDQ 12.5)
Profound ID (Mullen: DQ 14.7, NVDQ 18.8, VDQ 10.6)
Profound ID (Mullen: DQ 16.5, NVDQ 19.5, VDQ 13.4)
ID (no testing available)
Severe ID (Mullen: CSS < 49, DQ 30.4, NVDQ 35, VDQ 25.8)
Mild ID (DAS-II: GCA 50, Verbal 52, NV reasoning 74, spatial 32, special NV 49)
Profound ID (Mullen: DQ 11.5, NVDQ 14.5, VDQ 8.5)
Severe ID (no testing available)
Mild ID (Stanford Binet: FSIQ 56, NVIQ 60, VIQ 56)
Profound ID (Mullen: DQ 10.5, NVDQ 13.2, VDQ 7.9)
Profound ID (Mullen: DQ 0.63, NVDQ 0.97, VDQ 0.29)
Profound ID (Mullen: DQ 10.4, NVDQ 15.5, VDQ 5.2)
Severe ID (Mullen: CSS < 49, DQ 26.4, NVDQ 33, VDQ 19.8)
Mild ID (BDI at 4 y: adaptive SS 65, cognitive 65, communication 65, fine motor 72, gross motor 69, social 65)
Mild ID (no testing available)
Mild ID (no testing available)
17/17 (100%)
 Feeding difficulties
+ (chewing problems)
+ (regurgitation, oral motor dysfunction, difficulty consuming solid foods, PEG tube)
+ (oral motor dysfunction since birth, dysphagia, drooling, overeating, chewing problems)
+ (failure to thrive, g-tube)
− (drooling)
+ (dysphagia, drooling, may induce vomiting when over-eats)
+ (difficulty chewing, dysphagia)
+ (history of oral motor dysfunction, drooling)
+ (history of dysphagia)
+ (drooling, dysphagia)
+ (difficulty latching, currently gagging and choking behaviors, dysphagia, drooling)
+ (oral motor dysfunction)
+ (difficulty latching)
+ (difficulty latching)
13/17 (76%)
 Hypotonia
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
16/17 (94%)
 Gait abnormalities
+ (apraxic, hypotonic, toe-walking)
+ (toe-walking, unsteady, needs assistance)
+
+
+ (slow pace)
+ (toe walking)
+ (mildly hypotonic)
+
+
+
+ (apraxia)
+ (slow, hesitant and apraxic; previously reported as wide-based gait)
+ (mild but went through 6-month period in early childhood when he was unable to ambulate due to muscle weakness)
+
14/17 (82%)
Behavioral abnormalities
 ASD
+
+
+
+
+
+
+
+
NK
+
+
+
11/16 (69%)
 Hyperactivity
+
+
+
+
+
+
+
+
+
+
+
11/17 (65%)
 Aggression
+
+
+
+
+
+
+
+
8/17 (47%)
 Self-injury
+
+
+
NK
NK
3/15 (20%)
 Sleep disturbance
+
+
+
+
+
+
+
+
+
+
10/17 (59%)
 Pica
+
+
+
+
+
+
+
+
8/17 (47%)
 Repetitive behaviors (type)
+ (stereotypic motor movements in upper and lower extremities, forced exhalations)
+ (spinning, hand-flapping, teeth grinding)
+ (repetitive motor mannerisms, stereotypic vocalizations)
+ (bouncing, tapping, upper extremity motor stereotypies)
+ (chewing, teeth grinding, breath holding)
+ (chewing, teeth grinding, hand flapping, stereotypic vocalizations)
+ (self-stimulation, insistence on routines)
+ (hand-flapping, chewing)
+ (hand-flapping)
+ (restricted interests, perseveration)
+ (teeth grinding, repeatedly taps objects, walks in circles)
+ (pacing, upper extremity motor stereotypies)
+ (hand flapping, chewing, stereotypic vocalizations, teeth grinding)
+ (chewing, hand flapping, repetitive jumping, stereotypic vocalizations)
+ (finger and toe tapping)
+ (chewing, tapping teeth with finger)
16/17 (94%)
 Psychosis
+ (12–13 y)
1/17 (6%)
 Regression (age and details)
+ (5 y; some language loss)
+ (6 y; motor regression and lost some sign language, at one point stopped walking for 6 weeks, slowly regained ambulatory skills; at 5 y, some language loss)
+ (15 mo; stopped babbling, loses motor skills when sick)
+ (3.5 y; language and motor skills, stopped walking, socially withdrawn and less responsive)
+ (3–4 y; loss of fine motor skills)
+ (4.5 y; loss of language and motor skills, lost ability to ambulate and eye contact, lethargic, developed unusual motor stereotypies, regression coincided with diagnosis of parasitic infection)
+ (12–13 y; intermittent periods of behavioral, motor, and language regressions sometimes preceded by viral infection, included psychiatric symptoms. Currently non-verbal and unable to walk unsupported)
+ (2 y; lost all words, 7 y; regression in handwriting [can no longer hold a pen], motor regression began roughly around when seizures started)
+ (12–18 mo; loss of babbling, loss of few words, eye contact, and gesturing to request)
+ (13 y; “manic-like” behavior)
+ (9–10 y; “manic-like” behavior)
11/17 (65%)
Neurological findings
 Brain MRI (age)
Diffuse ventricular enlargement, colpocephaly, communicating hydrocephalus, thinning of parieto-occipital white matter and corpus callosum (8 y)
No MRI
Leukodystrophy (5 y)
Grossly normal but scattered areas of subtle FLAIR hyperintensity (2 y)
NK
Grossly normal but hyper-intensity in the left inferior parietal subcortical white matter possibly related to gliosis (4 y)
Normal (5 y)
Normal (3 y)
Normal (5, 9, and 11 y)
Normal (7 y)
Normal (4.5 y)
Normal (14 and 18 y)
Venous angioma (6 y); normal (16 y)
Normal (3 y)
Bilateral T2 hyper-intensities of posterior centrum semiovale (12 y)
Mild cerebellar tonsillar ectopia (14 y)
Normal (14 y)
Abnormal in 5/15 (33%)
 Seizures (age of onset, type)
+ (5 y, Landau-Kleffner variant; 6 y epileptic encephalopathy)
− (10 y, suspected complex partial seizures)
+ (3 y febrile; 6 y focal; 15 y began 1–10 absence or partial seizures daily)
+ (4 y, generalized myoclonic seizures)
+ (14 y, atypical absence)
+ (7 y, atypical absence and tonic)
5/17 (29%)
 Abnormal EEG
+ (localized sleep potentiated epileptiform discharges mainly in the midline and central regions during slow wave sleep)
+ (increased theta wave activity; bilateral K-complexes, and spindles and vertex waves during sleep. Left frontal spike and wave activity)
+ (spikes)
+ (spike and wave activity in frontotemporal lobes; no seizures)
+ (right frontal lobe spikes, slowing)
+ (left frontal spikes/polyspikes, intermittent polymorphic slowing [L>R] in the temporal region, and background slowing during sleep)
+ (high-voltage spike and sharp activity in frontal regions)
+ (occasional generalized polyspikes or polyspike-wave with shifting hemispheric predominance during sleep)
+ (no occipital dominant rhythm)
9/17 (53%)
Gastrointestinal problems
 Gastroesophageal reflux
+
+
+
+
+
5/17 (29%)
 Constipation
+
+
+
+
+
+
+
+
+
9/17 (53%)
 Diarrhea
+
+
+
+
+
5/17 (29%)
Additional features
 Increased pain tolerance
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
16/17 (94%)
 Decreased perspiration/heat intolerance
+
NK
NK
NK
+
NK
NK
2/12 (17%)
 Recurrent infections
+ (otitis, MT)
+ (otitis)
+ (otitis, upper respiratory tract)
+ (otitis, MT)
+ (otitis, tonsillitis)
+ (otitis, MT; yeast)
+ (otitis, bronchitis)
+ (otitis, sinusitis)
+ (otitis, MT)
9/17 (53%)
 Visual problems
+ (strabismus, corrective surgery)
+ (mild hyperopic astigmatism)
+ (strabismus)
+ (myopia)
+ (myopia)
5/17 (29%)
 Congenital heart defect
+ (coronary artery fistula)
1/17 (6%)
 Renal abnormalities
c
c
c
c
c
c
0/17
 Allergies
+ (penicillin)
+ (food, seasonal)
+ (food)
+ (penicillin, seasonal)
+ (seasonal)
+ (food)
+ (seasonal)
+ (penicillin)
+ (seasonal)
+ (food, dust, pets)
+ (food)
+ (food)
12/17 (71%)
 Asthma
+
+ (allergy induced)
+
3/17 (18%)
 Eczema
+
+
+
+
+
+
6/17 (35%)
 Other
      
Birth by in vitro fertilization
 
Sleep apnea
  
Sleep apnea, atrial fibrillation, intermittent hypoglycemia
  
Left preauricular skin tag, scoliosis
Episode of idiopathic intracranial hypertension at 12 y
  
+ present, − absent, ASD autism spectrum disorder, BDI Battelle Developmental Inventory, CSS Composite Standard Score, DAS-II Differential Ability Scales, Second Edition, DQ developmental quotient, EEG electroencephalography, F female, FSIQ full scale intelligence quotient, GCA general conceptual ability, ID intellectual disability, M male, MRI magnetic resonance imaging, MT myringotomy tubes, NA not applicable, NK not known, NV non-verbal, NVDQ non-verbal developmental quotient, NVIQ non-verbal intelligence quotient, PEG percutaneous endoscopic gastrostomy, SS standard score, VDQ verbal developmental quotient, VIQ verbal intelligence quotient
aIndividuals not directly evaluated
bMonozygotic twins
cNormal renal evaluation (ultrasound or computed tomography)
Table 3
Dysmorphic features in individuals with SHANK3 mutations
 
S1a
S2
S3
S4
S7
S8
S10
S11
S12
S13
S14
Total (%)
Gender
M
M
M
F
F
M
M
F
F
M
M
7 M, 4 F
Age at examination (years)
12
5
7
3
7
9
9
6
42
15
4
3–42
Craniofacial features
 Microcephalyb
+
1/11 (9%)
 Macrocephalyc
+
1/11 (9%)
 Dolichocephaly
+
1/11 (9%)
 Synophrys
0
 Sparse eyebrows
+
+
2/11 (18%)
 Long eyelashes
+
+
+
+
+
+
+
+
8/11 (73%)
 Periorbital fullness
+
+
+
+
4/11 (36%)
 Deep set eyes
+
+
+
+
+
5/11 (45%)
 Ptosis
0
 Epicanthal folds
+
+
+
+
+
5/11 (45%)
 Hypertelorism
0
 Wide nasal bridge
+
+
+
+
+
+
6/11 (55%)
 Bulbous nose
+
+
+
+
+
+
6/11 (55%)
 Anteverted nares
+
1/11 (9%)
 Full cheeks
+
+
2/11 (18%)
 Malar hypoplasia
+
+
+
+
+
5/11 (45%)
 Thin upper vermillion
+
+
+
3/11 (27%)
 Thick lower vermillion
+
+
+
3/11 (27%)
 Short philtrum
0
 Long philtrum
+
+
+
3/11 (27%)
 Malocclusion
+
+
+
+
+
+
+
7/11 (64%)
 High arched palate
+
+
+
+
NE
+
+
6/10 (60%)
 Ear anomalies
+ (low set ears)
+ (overfolded helix)
+ (fleshy ears)
+ (prominent ears)
4/11 (36%)
 Micrognathia
0
 Macrognathia
+
+
+
3/11 (27%)
 Pointed chin
+
+
+
+
+
+
+
7/11 (64%)
Hand and feet anomalies
 Large fleshy hands
+
+
+
3/11 (27%)
 5th finger clinodactyly
+
+
+
+
+
+
+
+
+
9/11 (82%)
 Partial syndactyly of toes 2–3
+
+
+
+
+
5/11 (45%)
 Sandal gap
+
+
+
NE
+
+
NE
+
6/9 (67%)
 Hypoplasia of distal phalanges of 5th finger
+
+
2/11 (18%)
Ectodermal anomalies
 Hypertrichosis
0
 Abnormal hair whorl
+
+
2/11 (18%)
 Hypoplastic/dysplastic toenails
+
+
+
+
+
+
6/11 (55%)
 Hypoplastic/dysplastic fingernails
+
+
2/11 (18%)
Other features
 Short stature/delayed growthd
+
+
+
3/11 (27%)
 Tall stature/accelerated growthe
0
 Hyperextensibility
+
+
+
+
NE
+
+
6/10 (60%)
 Sacral dimple
NE
0
 Scoliosis
+
1/11 (9%)
Total dysmorphic features
15
16
5
18
10
9
10
6
11
17
11
11/11 (100%)
Only individuals that underwent a detailed evaluation by a clinical geneticist are shown
+ present, − absent, F female, M male, NE not evaluated
aS1 also has a de novo pathogenic 17q12 microduplication [62]
bHead circumference < 3rd percentile
cHead circumference > 98th percentile
dHeight < 3rd percentile
eHeight > 98th percentile
ASD
Findings of ASD were widespread, with 69% (11/16) receiving a diagnosis of ASD. Among the 11 individuals of the Seaver cohort who received ASD diagnostic testing and a psychiatric evaluation, 82% (9/11) met criteria for ASD on the ADOS and 73% (8/11) met criteria for ASD on the ADI-R. A consensus diagnosis of ASD, accounting for both standardized assessments and clinical impression based on DSM-5 criteria, was reached in 73% (8/11) (Additional file 2: Tables S5, S6). All three children who did not receive an ASD diagnosis (S3, S7, S10) showed relevant features, including two with scores above the ASD cutoff on the ADOS-2 or the ADI-R but not both. It is notable that two of these three individuals (S7 and S10) were verbally fluent with cognitive functioning on the cusp of mild ID/borderline cognitive functioning.
Additional behavioral findings
All participants from the Seaver cohort had significant repetitive behaviors (n = 14), including hand flapping and stereotypic motor movements (11/14, 79%), chewing and teeth grinding (7/14, 50%), pica and mouthing of objects (8/14, 57%), and stereotypic vocalizations (5/14, 36%). The majority of participants were described as hyperactive (11/17, 65%), although the extent and severity of hyperactivity varied widely as did the extent of impulsivity and inattention. Participants were also prone to aggression (8/17, 47%) and self-injury (3/15, 20%), particularly when frustrated. Sleep disturbance was common (10/17, 59%).
Intellectual functioning
ID was observed in all cases that received standardized testing (n = 13), with 10 cases falling in the range of a severe-to-profound ID and three cases in the mild range. Two individuals (B2, B3) who did not receive standardized testing were characterized as mildly intellectually disabled based on the extent of language and developmental delay. All individuals within the normed range of up to 68 months (S2, S4, S6, S14) achieved the lowest possible standard score on the Mullen Early Learning Composite (< 49, < 1st percentile), indicating that the instrument reached its lower limit for reliable data collection (“floor” effect). Developmental quotients (DQ) were calculated for all individuals (excluding the 42-year-old individual) and ranged from 6.7 to 30 (mean ± SD, 15.6 ± 8.0). Verbal DQ ranged from 9.2 to 35 (19.9 ± 9.2), and nonverbal DQ ranged from 3.1 to 25.8 (11.39 ± 7.16) (Additional file 2: Table S6). Results from three additional cases who received other cognitive measures (S7, S10, and B1) indicated the presence of mild-to-moderate ID (Table 2).
Adaptive behavior
Results from the Vineland-II indicated that adaptive functioning was consistent with cognitive functioning (Additional file 2: Table S5). Overall, motor skills and socialization skills were better developed than communication and daily living skills. Two children (S7, S10) fell within the borderline range; all others fell below the first percentile.
Language skills
Language impairment was prominent (17/17, 100%); results are summarized in Table 2. All subjects were delayed in achieving language milestones. With regard to current language abilities, the ADOS-2 (n = 11) indicated that five individuals used no words, three used < 5 recognizable words or word approximations, one used mostly single words, and two used complex speech with frequent grammatical errors. Receptive and expressive language were equally delayed (Additional file 2: Table S7). Three individuals (S7, S10, S14) were administered the Peabody Picture Vocabulary Test and achieved scores between < 1st and 7th percentiles. Two of these individuals (S7, S10) achieved scores of 70 (2nd percentile) on the Expressive Vocabulary Test, indicating that despite fluent speech, expressive language abilities were significantly delayed relative to same-aged peers. Two of the Baylor participants were also reported to speak in sentences, but one was mostly echolalic.
Motor skills
Most individuals achieved motor milestones on time, despite significant fine and gross motor delays in all participants at the time of evaluation. Hypotonia (16/17, 94%) and gait abnormalities (14/17, 82%) were present in the majority of individuals. Gross motor skills were significantly better developed than fine motor skills (n = 9, p = 0.02 for both the Mullen and the Vineland-II, Wilcoxon signed-rank test; Additional file 2: Table S7). Two individuals (S7, S10) were administered the Beery Visual-Motor Integration Test and received standard scores of 45 and 65, respectively, which is indicative of visual-motor deficits.
Sensory processing
According to parent report, 16 of 17 participants had increased pain tolerance (94%). Results from the Sensory Assessment for Neurodevelopmental Disorders (n = 10) and clinical observation indicated that sensory hyporeactivity (i.e., underresponsiveness to stimuli) was prominent. These findings are consistent with the results from the Short Sensory Profile (n = 11), indicating high scores in the underresponsive/seeks sensation domain (10/11) and the low energy/weak domain (9/11).
Neurological findings
Seizures were reported in five individuals (5/17, 29%), including febrile (n = 1), absence (n = 3), focal (n = 1), and generalized seizures (n = 2) (one individual had febrile, absence, and focal seizures) (Table 2). Age of onset ranged from 4 to 14 years (7.2 ± 4). Nine individuals had an abnormal electroencephalography (EEG) (9/17, 53%), including five without clinical seizures. MRI in 15 individuals revealed abnormal findings in five (33%), including white matter abnormalities (n = 3), venous angioma (n = 1), and mild cerebellar ectopia (n = 1).
Regression
For the purpose of this manuscript, we only document regression in patients who clearly and consistently acquired skills for a prolonged period of time and then lost these skills, either permanently or for an extended period. Regression, occurring at various stages of development from early childhood to early adolescence, and affecting language, motor, and behavioral domains, was reported in 11 of 17 cases (65%). At least two caregivers noted regression that was triggered by infection and one reported seizures preceding the onset of regression.
Other medical conditions
Gastrointestinal problems were common, including gastroesophageal reflux (5/17, 29%), constipation (9/17, 53%), and diarrhea (5/17, 29%). Feeding problems were also common (13/17, 76%), including dysphagia and chewing difficulties; two individuals required placement of a gastrostomy tube. Recurrent infections were reported in 53% (9/17) of individuals, most often affecting the ears. Visual problems, and strabismus in particular, have been described in carriers of 22q13.3 deletions [2, 4, 51] and were present in 29% (5/17) of patients, including strabismus (n = 2), myopia (n = 2), and astigmatism (n = 1). Renal or urinary tract abnormalities, reported in 26–40% of cases with 22q13 deletions [2, 4], were absent in our cohort. Similarly, congenital heart defects, reported in 3–13% of patients with 22q13 deletions [2, 52], were uncommon; one individual had a coronary artery fistula that did not require surgical intervention. Lymphedema, cellulitis, precocious or delayed puberty, hearing problems, and hypothyroidism have been reported in cases with 22q13 deletions [2, 4] but were not present in individuals with SHANK3 mutations (Table 4).
Table 4
Clinical features in individuals with SHANK3 mutations as compared to 22q13 deletions including SHANK3
Clinical features
Individuals with SHANK3 mutations (current study)
Individuals with 22q13 deletions [2]
Intellectual disability
17/17 (100%)
29/30 (97%)
ASD
11/16 (69%)
26/30 (87%)
Verbally fluent
3/17 (18%)
0/30
Repetitive behaviors
16/17 (94%)
30/30 (100%)
Hyperactivity
11/17 (65%)
14/30 (47%)
Aggression
8/17 (47%)
13/30 (43%)
Sleep disturbance
10/17 (59%)
12/30 (40%)
Hypotonia
16/17 (94%)
23/30 (77%)
Gait abnormalities
14/17 (82%)
13/14 (93%)
Seizures
5/17 (29%)
12/30 (40%)
Abnormal brain MRI
5/15 (33%)
18/26 (69%)
Short staturea
5/16 (31%)
3/30 (10%)
Tall statureb
0/16
1/30 (3%)
Microcephalyc
1/14 (7%)
2/30 (7%)
Macrocephalyd
3/14 (21%)
9/30 (30%)
Dolichocephaly
1/11 (9%)
7/30 (23%)
Sparse hair/abnormal whorl
2/11 (18%)
5/30 (17%)
Long eyelashes
8/11 (73%)
13/30 (43%)
Periorbital fullness
4/11 (36%)
8/30 (27%)
Hypertelorism
0/11
3/30 (10%)
Deep set eyes
5/11 (45%)
2/30 (7%)
Ptosis
0/11
2/30 (7%)
Epicanthal folds
5/11 (45%)
9/30 (30%)
Strabismus
2/17 (12%)
3/30 (10%)
Wide nasal bridge
6/11 (55%)
4/30 (13%)
Bulbous nose
6/11 (55%)
15/30 (50%)
Full cheeks
2/11 (18%)
8/30 (27%)
Malar hypoplasia
5/11 (45%)
3/30 (10%)
Long philtrum
3/11 (27%)
5/30 (17%)
Malocclusion
7/11 (64%)
5/30 (17%)
Widely spaced teeth
0/11
1/30 (3%)
High arched palate
6/10 (60%)
8/30 (27%)
Ear anomalies
4/11 (36%)
13/30 (43%)
Pointed chin
7/11 (64%)
7/30 (23%)
Large fleshy hands
3/11 (27%)
17/30 (57%)
5th finger clinodactyly
9/11 (82%)
3/30 (10%)
Syndactyly of toes 2–3
5/11 (45%)
3/30 (10%)
Hypoplastic/dysplastic nails
7/11 (64%)
11/30 (37%)
Hyperextensibility
6/10 (60%)
8/30 (27%)
Scoliosis
1/11 (9%)
7/30 (23%)
Sacral dimple
0/10
4/30 (13%)
Gastroesophageal reflux
5/17 (29%)
13/30 (43%)
Constipation/diarrhea
11/17 (65%)
11/30 (37%)
Increased pain tolerance
16/17 (94%)
26/30 (87%)
Recurrent infections
9/17 (53%)
16/30 (53%)
Renal abnormalities
0/17
12/30 (40%)
Congenital heart defect
1/17 (6%)
1/30 (3%)
Hypothyroidism
0/17
1/30 (3%)
Lymphedema
0/17
7/30 (23%)
ASD autism spectrum disorder
aHeight < 3rd percentile
bHeight > 98th percentile
cHead circumference < 3rd percentile
dHead circumference > 98th percentile
Dysmorphic features
Dysmorphology examinations were performed on 11 individuals from the Seaver cohort, using a PMS-specific checklist (Table 3, Fig. 1b). All had at least five usually mild dysmorphic features (range 5–18), without a distinctive facial gestalt. In general, findings were consistent with those reported in patients with 22q13 deletions [2, 46]. However, some features were more common than previously reported, including fifth finger clinodactyly (9/11, 82%), malocclusion (7/11, 64%), and wide nasal bridge (6/11, 55%) (Table 4). The use of a PMS-specific checklist could account in part for the higher frequency with which certain features were noted. Other features present in over 50% of the individuals were long eyelashes, bulbous nose, high arched palate, pointed chin, hyperextensibility, dysplastic toenails, and sandal gap.

Phenotype of individuals with SHANK3 mutations in the literature

The clinical features of 45 previously published individuals with pathogenic or likely pathogenic SHANK3 variants are summarized in Additional file 1: Table S2. (Fifteen individuals reported only in ClinVar are included in Additional file 1: Table S1 where we summarize the allelic spectrum but are not included here because no phenotype information was available for them.) Although only limited information was available for most cases, the phenotype was consistent with that observed in our cohort, including ID (33/33, 100%), severe language impairment (22/23, 96%), ASD (26/34, 76%), hypotonia (8/12, 67%), seizures (17/30, 57%), and dysmorphic features (13/21, 62%). Regression was reported in 11 individuals.

Discussion

This is the first study to comprehensively describe the phenotype in patients with PMS due to SHANK3 point mutations. Our findings demonstrate that loss of SHANK3 alone is sufficient to produce the characteristic features of PMS, including ID, ASD, severe speech impairment, hypotonia, epilepsy, motor skills deficits, feeding difficulties, mild dysmorphic features, increased pain tolerance, gastrointestinal problems, and neuroimaging abnormalities. In addition, we advance the understanding of the genetic architecture of PMS and, in so doing, provide information to aid in the interpretation of SHANK3 variants.

Genetic findings

Findings in our cohort and in previously reported patients indicate that SHANK3 mutations are fully penetrant. The identification of three families with SHANK3 mutations in multiple siblings due to germline mosaicism (5%, 3/57) [9, 22, 43] has important implications for genetic counseling. Of note, we identified four recurrent mutations in SHANK3, including p.Leu1142Valfs*153, p.Ala1227Glyfs*69, p.Arg1255Leufs*25, and c.2265+1G>A. The most common mutation, c.3679dupG (p.Ala1227Glyfs*69), identified in six individuals, is due to the duplication of a guanine in a stretch of eight guanines, indicating that this segment is prone to replication errors. Functional studies on several of the truncating mutations described here (p.Trp509* in S1, p.Pro834Argfs*59 in S3, p.Lys1670* in S13, and p.A1227Gfs*69 in S7, S8, and B1) provide further support for their deleterious effects [9, 26, 53, 54].
Although the majority of pathogenic/likely pathogenic SHANK3 variants identified to date are truncating, the interpretation of missense variants remains difficult. Missense variant assessment relies on inheritance, segregation within families, frequency in population databases, functional studies, and computational predictions of pathogenicity (see ACMG guidelines [25]). In the case of SHANK3, in silico prediction programs often provide contradictory results (Additional file 1: Table S3). Functional studies could help determine the pathogenicity of missense substitutions; however, previous in vitro analyses have identified synaptic defects associated with missense variants in ASD inherited from healthy parents and found in control databases [9, 53, 54]; hence, more discriminatory functional approaches will need to be developed.

ASD, ID, language, and motor skills

Our results demonstrate the high prevalence of ASD in individuals with PMS resulting from SHANK3 mutations, similar to our previous findings in individuals with 22q13 deletions [2]. The ADOS and ADI-R provided important information regarding ASD features, even in individuals with low mental ages; however, clinical evaluation and consensus discussion proved necessary to determine which individuals did not meet criteria for ASD. Negative ASD findings in the two verbally fluent individuals raise questions about the relationship between ASD diagnosis and severe global developmental delay. Interestingly, in spite of severe-to-profound ID, and significant expressive and receptive language delays in the majority of participants, language appears to be more preserved in individuals with SHANK3 mutations compared to those with 22q13 deletions seen at the same centers [2, 24]. Motor skills deficits were also pronounced, although early motor milestones were achieved on time for the majority of individuals. Gross motor skills were better developed than fine motor skills and, in most cases, appear to be less severely affected than in individuals with 22q13 deletions, particularly regarding gait. These results indicate that SHANK3 haploinsufficiency affects cognition, language, and motor functioning.

Regression and psychotic symptoms

Significant cognitive and behavioral regression has been reported in individuals with PMS [2, 3, 9, 51, 5559]. Over half of our sample reportedly experienced a regression in motor and language skills that occurred during different periods of development (early childhood or adolescence). These results indicate that SHANK3 haploinsufficiency alone is sufficient to increase risk for regression. However, reports of regression must be interpreted with caution based on a lack of well-defined criteria or standardized assessment instruments and potential recall biases in reporting. Further careful study is needed to characterize the regression phenotype in PMS using longitudinal designs and to begin to elucidate the underlying mechanisms.
Possibly related to regression, psychotic symptoms have emerged as an important area of study in PMS as several reports have suggested that as individuals with PMS age, they may be at increased risk for significant psychiatric disturbance, including bipolar disorder [51, 5557, 59]. Four of the reported patients had truncating mutations in SHANK3 [9, 56], indicating that SHANK3 is responsible for this phenotype. Mutations in SHANK3 have also been found in four individuals from two families with atypical schizophrenia associated with early onset and ID [22]. The monozygotic twins reported here (B2, B3) showed “manic-like” behavior beginning at 13 years of age in one and at 9–10 years of age in the other. Also, one individual (S12) experienced psychotic symptoms characterized by auditory and visual hallucinations beginning around 12–13 years of age. She had episodic periods of mania and depression, insomnia, decreased appetite and weight loss, unsteady gait, and catatonic posturing, similar to previous reports [51, 55, 56, 59]. Importantly, she also had significant regression in language and motor skills with documented cognitive decline from borderline intellectual functioning before puberty to profound ID based on the current assessment at age 42 (see Table 2). The patient was verbally fluent but became non-verbal. She was also walking independently at 20 months and currently is unable to walk more than several steps without support. Pubertal onset appears to be a potential trigger for shifts in the psychiatric phenotype in PMS; hence, it is important to note that only two of the 14 Seaver participants were post-pubertal.

Other medical findings

Common medical features in individuals with SHANK3 mutations were consistent with published literature in subjects with 22q13.3 deletions [1, 2, 46]. Epilepsy has been reported in PMS with a mean prevalence of 32% and a wide range of seizure types, frequencies, and severity [24]. The lower frequency of seizures in our study compared to that for previously reported individuals with SHANK3 point mutations (29% versus 57%) might be due to the young age of many of our patients (seizure onset occurred at ≥ 10 years in 41% [7/17] of new and previously reported individuals). In agreement with our findings, no specific EEG abnormalities have been reported in PMS, and EEG abnormalities (61%) are seen in children with and without a history of clinical seizures [24]. Structural brain abnormalities are observed in about a third of cases with 22q13 deletions (including corpus callosum and cerebellar abnormalities, dysmyelination, ventricular dilatation, and arachnoid cysts) [1, 2, 24]; results from patients with mutations are consistent with those with deletions. Overall, loss of SHANK3 is sufficient to cause seizures and structural brain changes, although findings remain non-specific to PMS.
Gastrointestinal problems, recurrent infections, and increased pain tolerance were common among individuals with SHANK3 mutations, consistent with previous estimates in 22q13 deletions [2, 4]. In agreement with these findings, studies in mice showed that SHANK3 is expressed in the spinal cord and primary sensory neurons, where it regulates pain sensitivity [60]. SHANK3 has also been shown to be expressed in intestinal epithelial cells, where it regulates barrier function [61]. In contrast, despite reports of renal and urinary tract abnormalities in 26–40% of cases with 22q13 deletions (including vesicoureteral reflux, hydronephrosis, renal agenesis, and dysplastic or polycystic kidneys) [2, 4], no such anomalies were observed in our cohort. While data from ongoing genotype-phenotype studies are still emerging, it is likely that the genetic risk for renal anomalies is not directly associated to SHANK3 haploinsufficiency and involves other gene(s) in 22q13.
Despite high variability, mild dysmorphic features were prevalent among patients with SHANK3 mutations and were consistent with the phenotype in patients with 22q13 deletions [1, 2, 4]. It has been previously reported that the number of dysmorphic features is correlated with deletion size [2] and that several dysmorphic features are associated with larger deletion sizes [7]. Our results suggest that some of the more common dysmorphic features associated with PMS are caused by SHANK3 mutations, but further studies are needed to determine the contribution of other genes involved in 22q13 deletions.

Conclusions

This represents a first detailed report of the genetic and phenotypic spectrum associated with SHANK3 mutations, which are being identified with greater frequency as clinical sequencing becomes more widespread. Our findings show that SHANK3 haploinsufficiency due to point mutations alone is sufficient to cause a broad range of phenotypic features associated with PMS. These include hypotonia, global developmental delay, ID, ASD, language deficits, sleep disturbance, increased pain tolerance, regression, motor skills deficits, seizures, abnormal EEG, brain imaging abnormalities, feeding difficulties, and gastrointestinal problems. We also describe frequent dysmorphic features in individuals with SHANK3 mutations, including fifth finger clinodactyly, long eyelashes, bulbous nose, wide nasal bridge, malocclusion, high arched palate, pointed chin, sandal gap, and dysplastic toenails. Importantly, we show that language and motor phenotypes appear to be less severe in individuals with point mutations, as compared to 22q13 deletions. These findings extend the role of SHANK3 dysfunction in human disease beyond its well-known role at the synapse in the central nervous system.

Acknowledgements

We thank the individuals with PMS and their families for their participation.

Funding

This work was supported by grants from the Beatrice and Samuel A. Seaver Foundation, the Phelan-McDermid Syndrome Foundation, NIMH (R34 MH100276-01 to AK and R21 MH107839 to JDB), and NINDS (U54 NS092090-01 to AK and K08 NS091381 to JLH). SDR is a Seaver fellow. JLH is funded by the Robbins Foundation. CB is a Research Director at INSERM. The funding bodies had no role in the design of the study, in the collection, analysis, and interpretation of data, or in the writing of the manuscript.

Availability of data and materials

All data generated or analyzed during this study are included in this article and its supplementary information files.
The institutional review boards of the Icahn School of Medicine at Mount Sinai and Baylor College of Medicine approved this study. Participants were enrolled after written informed consent was obtained from parents or legal guardians.
Written informed consent for publication was obtained from the parents or legal guardians.

Competing interests

JDB and Mount Sinai hold a shared parent for the use of insulin-like growth factor-1 (IGF-1) in Phelan-McDermid syndrome. JDB is on the scientific advisory board for Coronis Neuroscience and has consulted for the Gerson Lehrman Group. AK is on the advisory board of Vencerx Therapeutics and consults for Ovid Therapeutics. The remaining authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Kolevzon A, Angarita B, Bush L, Wang AT, Frank Y, Yang A, et al. Phelan-McDermid syndrome: a review of the literature and practice parameters for medical assessment and monitoring. J Neurodev Disord. 2014;6:39.CrossRefPubMedPubMedCentral Kolevzon A, Angarita B, Bush L, Wang AT, Frank Y, Yang A, et al. Phelan-McDermid syndrome: a review of the literature and practice parameters for medical assessment and monitoring. J Neurodev Disord. 2014;6:39.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Soorya L, Kolevzon A, Zweifach J, Lim T, Dobry Y, Schwartz L, et al. Prospective investigation of autism and genotype-phenotype correlations in 22q13 deletion syndrome and SHANK3 deficiency. Mol Autism. 2013;4:18.CrossRefPubMedPubMedCentral Soorya L, Kolevzon A, Zweifach J, Lim T, Dobry Y, Schwartz L, et al. Prospective investigation of autism and genotype-phenotype correlations in 22q13 deletion syndrome and SHANK3 deficiency. Mol Autism. 2013;4:18.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Bonaglia MC, Giorda R, Beri S, De Agostini C, Novara F, Fichera M, et al. Molecular mechanisms generating and stabilizing terminal 22q13 deletions in 44 subjects with Phelan/McDermid syndrome. PLoS Genet. 2011;7:e1002173.CrossRefPubMedPubMedCentral Bonaglia MC, Giorda R, Beri S, De Agostini C, Novara F, Fichera M, et al. Molecular mechanisms generating and stabilizing terminal 22q13 deletions in 44 subjects with Phelan/McDermid syndrome. PLoS Genet. 2011;7:e1002173.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Sarasua SM, Boccuto L, Sharp JL, Dwivedi A, Chen CF, Rollins JD, et al. Clinical and genomic evaluation of 201 patients with Phelan-McDermid syndrome. Hum Genet. 2014;133:847–59.CrossRefPubMed Sarasua SM, Boccuto L, Sharp JL, Dwivedi A, Chen CF, Rollins JD, et al. Clinical and genomic evaluation of 201 patients with Phelan-McDermid syndrome. Hum Genet. 2014;133:847–59.CrossRefPubMed
5.
Zurück zum Zitat Luciani JJ, de Mas P, Depetris D, Mignon-Ravix C, Bottani A, Prieur M, et al. Telomeric 22q13 deletions resulting from rings, simple deletions, and translocations: cytogenetic, molecular, and clinical analyses of 32 new observations. J Med Genet. 2003;40:690–6.CrossRefPubMedPubMedCentral Luciani JJ, de Mas P, Depetris D, Mignon-Ravix C, Bottani A, Prieur M, et al. Telomeric 22q13 deletions resulting from rings, simple deletions, and translocations: cytogenetic, molecular, and clinical analyses of 32 new observations. J Med Genet. 2003;40:690–6.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Wilson HL, Wong AC, Shaw SR, Tse WY, Stapleton GA, Phelan MC, et al. Molecular characterisation of the 22q13 deletion syndrome supports the role of haploinsufficiency of SHANK3/PROSAP2 in the major neurological symptoms. J Med Genet. 2003;40:575–84.CrossRefPubMedPubMedCentral Wilson HL, Wong AC, Shaw SR, Tse WY, Stapleton GA, Phelan MC, et al. Molecular characterisation of the 22q13 deletion syndrome supports the role of haploinsufficiency of SHANK3/PROSAP2 in the major neurological symptoms. J Med Genet. 2003;40:575–84.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Sarasua SM, Dwivedi A, Boccuto L, Rollins JD, Chen CF, Rogers RC, et al. Association between deletion size and important phenotypes expands the genomic region of interest in Phelan-McDermid syndrome (22q13 deletion syndrome). J Med Genet. 2011;48:761–6.CrossRefPubMed Sarasua SM, Dwivedi A, Boccuto L, Rollins JD, Chen CF, Rogers RC, et al. Association between deletion size and important phenotypes expands the genomic region of interest in Phelan-McDermid syndrome (22q13 deletion syndrome). J Med Genet. 2011;48:761–6.CrossRefPubMed
8.
Zurück zum Zitat Zwanenburg RJ, Ruiter SA, van den Heuvel ER, Flapper BC, Van Ravenswaaij-Arts CM. Developmental phenotype in Phelan-McDermid (22q13.3 deletion) syndrome: a systematic and prospective study in 34 children. J Neurodev Disord. 2016;8:16.CrossRefPubMedPubMedCentral Zwanenburg RJ, Ruiter SA, van den Heuvel ER, Flapper BC, Van Ravenswaaij-Arts CM. Developmental phenotype in Phelan-McDermid (22q13.3 deletion) syndrome: a systematic and prospective study in 34 children. J Neurodev Disord. 2016;8:16.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Durand CM, Betancur C, Boeckers TM, Bockmann J, Chaste P, Fauchereau F, et al. Mutations in the gene encoding the synaptic scaffolding protein SHANK3 are associated with autism spectrum disorders. Nat Genet. 2007;39:25–7.CrossRefPubMed Durand CM, Betancur C, Boeckers TM, Bockmann J, Chaste P, Fauchereau F, et al. Mutations in the gene encoding the synaptic scaffolding protein SHANK3 are associated with autism spectrum disorders. Nat Genet. 2007;39:25–7.CrossRefPubMed
10.
Zurück zum Zitat Moessner R, Marshall CR, Sutcliffe JS, Skaug J, Pinto D, Vincent J, et al. Contribution of SHANK3 mutations to autism spectrum disorder. Am J Hum Genet. 2007;81:1289–97.CrossRefPubMedPubMedCentral Moessner R, Marshall CR, Sutcliffe JS, Skaug J, Pinto D, Vincent J, et al. Contribution of SHANK3 mutations to autism spectrum disorder. Am J Hum Genet. 2007;81:1289–97.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Gauthier J, Spiegelman D, Piton A, Lafreniere RG, Laurent S, St-Onge J, et al. Novel de novo SHANK3 mutation in autistic patients. Am J Med Genet B Neuropsychiatr Genet. 2009;150B:421–4.CrossRefPubMed Gauthier J, Spiegelman D, Piton A, Lafreniere RG, Laurent S, St-Onge J, et al. Novel de novo SHANK3 mutation in autistic patients. Am J Med Genet B Neuropsychiatr Genet. 2009;150B:421–4.CrossRefPubMed
12.
Zurück zum Zitat Boccuto L, Lauri M, Sarasua SM, Skinner CD, Buccella D, Dwivedi A, et al. Prevalence of SHANK3 variants in patients with different subtypes of autism spectrum disorders. Eur J Hum Genet. 2013;21:310–6.CrossRefPubMed Boccuto L, Lauri M, Sarasua SM, Skinner CD, Buccella D, Dwivedi A, et al. Prevalence of SHANK3 variants in patients with different subtypes of autism spectrum disorders. Eur J Hum Genet. 2013;21:310–6.CrossRefPubMed
13.
Zurück zum Zitat Leblond CS, Nava C, Polge A, Gauthier J, Huguet G, Lumbroso S, et al. Meta-analysis of SHANK mutations in autism spectrum disorders: a gradient of severity in cognitive impairments. PLoS Genet. 2014;10:e1004580.CrossRefPubMedPubMedCentral Leblond CS, Nava C, Polge A, Gauthier J, Huguet G, Lumbroso S, et al. Meta-analysis of SHANK mutations in autism spectrum disorders: a gradient of severity in cognitive impairments. PLoS Genet. 2014;10:e1004580.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat De Rubeis S, He X, Goldberg AP, Poultney CS, Samocha K, Cicek AE, et al. Synaptic, transcriptional and chromatin genes disrupted in autism. Nature. 2014;515:209–15.CrossRefPubMedPubMedCentral De Rubeis S, He X, Goldberg AP, Poultney CS, Samocha K, Cicek AE, et al. Synaptic, transcriptional and chromatin genes disrupted in autism. Nature. 2014;515:209–15.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat O'Roak BJ, Stessman HA, Boyle EA, Witherspoon KT, Martin B, Lee C, et al. Recurrent de novo mutations implicate novel genes underlying simplex autism risk. Nat Commun. 2014;5:5595.CrossRefPubMedPubMedCentral O'Roak BJ, Stessman HA, Boyle EA, Witherspoon KT, Martin B, Lee C, et al. Recurrent de novo mutations implicate novel genes underlying simplex autism risk. Nat Commun. 2014;5:5595.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Yuen RK, Merico D, Bookman M, Howe JL. Thiruvahindrapuram B, Patel RV, et al. Whole genome sequencing resource identifies 18 new candidate genes for autism spectrum disorder. Nat Neurosci. 2017;20:602–11. Yuen RK, Merico D, Bookman M, Howe JL. Thiruvahindrapuram B, Patel RV, et al. Whole genome sequencing resource identifies 18 new candidate genes for autism spectrum disorder. Nat Neurosci. 2017;20:602–11.
17.
Zurück zum Zitat Hamdan FF, Gauthier J, Araki Y, Lin DT, Yoshizawa Y, Higashi K, et al. Excess of de novo deleterious mutations in genes associated with glutamatergic systems in nonsyndromic intellectual disability. Am J Hum Genet. 2011;88:306–16.CrossRefPubMedPubMedCentral Hamdan FF, Gauthier J, Araki Y, Lin DT, Yoshizawa Y, Higashi K, et al. Excess of de novo deleterious mutations in genes associated with glutamatergic systems in nonsyndromic intellectual disability. Am J Hum Genet. 2011;88:306–16.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Gong X, Jiang YW, Zhang X, An Y, Zhang J, Wu Y, et al. High proportion of 22q13 deletions and SHANK3 mutations in Chinese patients with intellectual disability. PLoS One. 2012;7:e34739.CrossRefPubMedPubMedCentral Gong X, Jiang YW, Zhang X, An Y, Zhang J, Wu Y, et al. High proportion of 22q13 deletions and SHANK3 mutations in Chinese patients with intellectual disability. PLoS One. 2012;7:e34739.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Redin C, Gerard B, Lauer J, Herenger Y, Muller J, Quartier A, et al. Efficient strategy for the molecular diagnosis of intellectual disability using targeted high-throughput sequencing. J Med Genet. 2014;51:724–36.CrossRefPubMedPubMedCentral Redin C, Gerard B, Lauer J, Herenger Y, Muller J, Quartier A, et al. Efficient strategy for the molecular diagnosis of intellectual disability using targeted high-throughput sequencing. J Med Genet. 2014;51:724–36.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Lelieveld SH, Reijnders MR, Pfundt R, Yntema HG, Kamsteeg EJ, de Vries P, et al. Meta-analysis of 2,104 trios provides support for 10 new genes for intellectual disability. Nat Neurosci. 2016;19:1194–6.CrossRefPubMed Lelieveld SH, Reijnders MR, Pfundt R, Yntema HG, Kamsteeg EJ, de Vries P, et al. Meta-analysis of 2,104 trios provides support for 10 new genes for intellectual disability. Nat Neurosci. 2016;19:1194–6.CrossRefPubMed
21.
Zurück zum Zitat Deciphering Developmental Disorders Study. Prevalence and architecture of de novo mutations in developmental disorders. Nature. 2017;542:433–8.CrossRef Deciphering Developmental Disorders Study. Prevalence and architecture of de novo mutations in developmental disorders. Nature. 2017;542:433–8.CrossRef
22.
Zurück zum Zitat Gauthier J, Champagne N, Lafreniere RG, Xiong L, Spiegelman D, Brustein E, et al. De novo mutations in the gene encoding the synaptic scaffolding protein SHANK3 in patients ascertained for schizophrenia. Proc Natl Acad Sci U S A. 2010;107:7863–8.CrossRefPubMedPubMedCentral Gauthier J, Champagne N, Lafreniere RG, Xiong L, Spiegelman D, Brustein E, et al. De novo mutations in the gene encoding the synaptic scaffolding protein SHANK3 in patients ascertained for schizophrenia. Proc Natl Acad Sci U S A. 2010;107:7863–8.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Betancur C, Buxbaum JD. SHANK3 haploinsufficiency: a “common” but underdiagnosed highly penetrant monogenic cause of autism spectrum disorders. Mol Autism. 2013;4:17.CrossRefPubMedPubMedCentral Betancur C, Buxbaum JD. SHANK3 haploinsufficiency: a “common” but underdiagnosed highly penetrant monogenic cause of autism spectrum disorders. Mol Autism. 2013;4:17.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Holder JL Jr, Quach MM. The spectrum of epilepsy and electroencephalographic abnormalities due to SHANK3 loss-of-function mutations. Epilepsia. 2016;57:1651–9.CrossRefPubMedPubMedCentral Holder JL Jr, Quach MM. The spectrum of epilepsy and electroencephalographic abnormalities due to SHANK3 loss-of-function mutations. Epilepsia. 2016;57:1651–9.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17:405–24.CrossRefPubMedPubMedCentral Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17:405–24.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Cochoy DM, Kolevzon A, Kajiwara Y, Schoen M, Pascual-Lucas M, Lurie S, et al. Phenotypic and functional analysis of SHANK3 stop mutations identified in individuals with ASD and/or ID. Mol Autism. 2015;6:23.CrossRefPubMedPubMedCentral Cochoy DM, Kolevzon A, Kajiwara Y, Schoen M, Pascual-Lucas M, Lurie S, et al. Phenotypic and functional analysis of SHANK3 stop mutations identified in individuals with ASD and/or ID. Mol Autism. 2015;6:23.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Lord C, Rutter M, DiLavore PS, Risi S, Gotham K, Bishop D. Autism Diagnostic Observation Schedule, 2nd edition (ADOS-2) manual (part I): modules 1–4. Torrance: Western Psychological Services; 2012. Lord C, Rutter M, DiLavore PS, Risi S, Gotham K, Bishop D. Autism Diagnostic Observation Schedule, 2nd edition (ADOS-2) manual (part I): modules 1–4. Torrance: Western Psychological Services; 2012.
28.
Zurück zum Zitat Lord C, Rutter M, Le Couteur A. Autism Diagnostic Interview-Revised: a revised version of a diagnostic interview for caregivers of individuals with possible pervasive developmental disorders. J Autism Dev Disord. 1994;24:659–85.CrossRefPubMed Lord C, Rutter M, Le Couteur A. Autism Diagnostic Interview-Revised: a revised version of a diagnostic interview for caregivers of individuals with possible pervasive developmental disorders. J Autism Dev Disord. 1994;24:659–85.CrossRefPubMed
29.
Zurück zum Zitat American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 5th edition, text revision. Washington, DC: American Psychiatric Association; 2013.CrossRef American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 5th edition, text revision. Washington, DC: American Psychiatric Association; 2013.CrossRef
30.
Zurück zum Zitat Hus V, Gotham K, Lord C. Standardizing ADOS domain scores: separating severity of social affect and restricted and repetitive behaviors. J Autism Dev Disord. 2014;44:2400–12.CrossRefPubMedPubMedCentral Hus V, Gotham K, Lord C. Standardizing ADOS domain scores: separating severity of social affect and restricted and repetitive behaviors. J Autism Dev Disord. 2014;44:2400–12.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Mullen EM. Mullen Scales of Early Learning. Circle Pines: American Guidance Services; 1995. Mullen EM. Mullen Scales of Early Learning. Circle Pines: American Guidance Services; 1995.
32.
Zurück zum Zitat Roid GH. Stanford Binet Intelligence Scales. 5th ed. Itasca: Riverside Publishing; 2003. Roid GH. Stanford Binet Intelligence Scales. 5th ed. Itasca: Riverside Publishing; 2003.
33.
Zurück zum Zitat Elliot CD. Differential Ability Scales—second edition: introductory and technical manual. San Antonio: Harcourt Assessment; 2007. Elliot CD. Differential Ability Scales—second edition: introductory and technical manual. San Antonio: Harcourt Assessment; 2007.
34.
Zurück zum Zitat Bishop SL, Guthrie W, Coffing M, Lord C. Convergent validity of the Mullen Scales of Early Learning and the Differential Ability Scales in children with autism spectrum disorders. Am J Intellect Dev Disabil. 2011;116:331–43.CrossRefPubMed Bishop SL, Guthrie W, Coffing M, Lord C. Convergent validity of the Mullen Scales of Early Learning and the Differential Ability Scales in children with autism spectrum disorders. Am J Intellect Dev Disabil. 2011;116:331–43.CrossRefPubMed
35.
Zurück zum Zitat Akshoomoff N. Use of the Mullen Scales of Early Learning for the assessment of young children with autism spectrum disorders. Child Neuropsychol. 2006;12:269–77.CrossRefPubMedPubMedCentral Akshoomoff N. Use of the Mullen Scales of Early Learning for the assessment of young children with autism spectrum disorders. Child Neuropsychol. 2006;12:269–77.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Sparrow SS, Cicchetti DV, Balla DA. Vineland Adaptive Behavior Scales: second edition (Vineland II), survey interview form/caregiver rating form. Livonia: Pearson Assessments; 2005. Sparrow SS, Cicchetti DV, Balla DA. Vineland Adaptive Behavior Scales: second edition (Vineland II), survey interview form/caregiver rating form. Livonia: Pearson Assessments; 2005.
37.
Zurück zum Zitat Fenson L, Marchman VA, Thal DJ, Dale PS, Reznick JS, Bates E. MacArthur-Bates communicative development inventories: user’s guide and technical manual. 2nd ed. Baltimore: Brookes Publishing Co.; 2007. Fenson L, Marchman VA, Thal DJ, Dale PS, Reznick JS, Bates E. MacArthur-Bates communicative development inventories: user’s guide and technical manual. 2nd ed. Baltimore: Brookes Publishing Co.; 2007.
38.
Zurück zum Zitat Dunn LM, Dunn DM. PPVT-4: Peabody Picture Vocabulary Test. Bloomington: Pearson Assessments; 2007. Dunn LM, Dunn DM. PPVT-4: Peabody Picture Vocabulary Test. Bloomington: Pearson Assessments; 2007.
39.
Zurück zum Zitat Williams KT. The Expressive Vocabulary Test. 2nd ed. Circle Pines: AGS Publishing; 2007. Williams KT. The Expressive Vocabulary Test. 2nd ed. Circle Pines: AGS Publishing; 2007.
40.
Zurück zum Zitat Beery KE, Buktenica NA, Beery NA. Beery-Buktenica Developmental Test of Visual-Motor Integration. 6th ed. Minneapolis: Pearson; 2010. Beery KE, Buktenica NA, Beery NA. Beery-Buktenica Developmental Test of Visual-Motor Integration. 6th ed. Minneapolis: Pearson; 2010.
41.
Zurück zum Zitat Dunn W, Westman K. The sensory profile: the performance of a national sample of children without disabilities. Am J Occup Ther. 1997;51:25–34.CrossRefPubMed Dunn W, Westman K. The sensory profile: the performance of a national sample of children without disabilities. Am J Occup Ther. 1997;51:25–34.CrossRefPubMed
42.
Zurück zum Zitat Siper PM, Kolevzon A, Wang AT, Buxbaum JD, Tavassoli T. A clinician-administered observation and corresponding caregiver interview capturing DSM-5 sensory reactivity symptoms in children with ASD. Autism Res. 2017;10:1133–40.CrossRefPubMed Siper PM, Kolevzon A, Wang AT, Buxbaum JD, Tavassoli T. A clinician-administered observation and corresponding caregiver interview capturing DSM-5 sensory reactivity symptoms in children with ASD. Autism Res. 2017;10:1133–40.CrossRefPubMed
43.
Zurück zum Zitat Nemirovsky SI, Cordoba M, Zaiat JJ, Completa SP, Vega PA, Gonzalez-Moron D, et al. Whole genome sequencing reveals a de novo SHANK3 mutation in familial autism spectrum disorder. PLoS One. 2015;10:e0116358.CrossRefPubMedPubMedCentral Nemirovsky SI, Cordoba M, Zaiat JJ, Completa SP, Vega PA, Gonzalez-Moron D, et al. Whole genome sequencing reveals a de novo SHANK3 mutation in familial autism spectrum disorder. PLoS One. 2015;10:e0116358.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Lim ET, Uddin M, De Rubeis S, Chan Y, Kamumbu AS, Zhang X, et al. Rates, distribution and implications of postzygotic mosaic mutations in autism spectrum disorder. Nat Neurosci. 2017;20:1217–24.CrossRefPubMedPubMedCentral Lim ET, Uddin M, De Rubeis S, Chan Y, Kamumbu AS, Zhang X, et al. Rates, distribution and implications of postzygotic mosaic mutations in autism spectrum disorder. Nat Neurosci. 2017;20:1217–24.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Zhang Y, Kong W, Gao Y, Liu X, Gao K, Xie H, et al. Gene mutation analysis in 253 Chinese children with unexplained epilepsy and intellectual/developmental disabilities. PLoS One. 2015;10:e0141782.CrossRefPubMedPubMedCentral Zhang Y, Kong W, Gao Y, Liu X, Gao K, Xie H, et al. Gene mutation analysis in 253 Chinese children with unexplained epilepsy and intellectual/developmental disabilities. PLoS One. 2015;10:e0141782.CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Bowling KM, Thompson ML, Amaral MD, Finnila CR, Hiatt SM, Engel KL, et al. Genomic diagnosis for children with intellectual disability and/or developmental delay. Genome Med. 2017;9:43.CrossRefPubMedPubMedCentral Bowling KM, Thompson ML, Amaral MD, Finnila CR, Hiatt SM, Engel KL, et al. Genomic diagnosis for children with intellectual disability and/or developmental delay. Genome Med. 2017;9:43.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Samocha KE, Robinson EB, Sanders SJ, Stevens C, Sabo A, McGrath LM, et al. A framework for the interpretation of de novo mutation in human disease. Nat Genet. 2014;46:944–50.CrossRefPubMedPubMedCentral Samocha KE, Robinson EB, Sanders SJ, Stevens C, Sabo A, McGrath LM, et al. A framework for the interpretation of de novo mutation in human disease. Nat Genet. 2014;46:944–50.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Bramswig NC, Ludecke HJ, Alanay Y, Albrecht B, Barthelmie A, Boduroglu K, et al. Exome sequencing unravels unexpected differential diagnoses in individuals with the tentative diagnosis of Coffin-Siris and Nicolaides-Baraitser syndromes. Hum Genet. 2015;134:553–68.CrossRefPubMed Bramswig NC, Ludecke HJ, Alanay Y, Albrecht B, Barthelmie A, Boduroglu K, et al. Exome sequencing unravels unexpected differential diagnoses in individuals with the tentative diagnosis of Coffin-Siris and Nicolaides-Baraitser syndromes. Hum Genet. 2015;134:553–68.CrossRefPubMed
49.
Zurück zum Zitat Kolevzon A, Cai G, Soorya L, Takahashi N, Grodberg D, Kajiwara Y, et al. Analysis of a purported SHANK3 mutation in a boy with autism: clinical impact of rare variant research in neurodevelopmental disabilities. Brain Res. 2011;1380:98–105.CrossRefPubMed Kolevzon A, Cai G, Soorya L, Takahashi N, Grodberg D, Kajiwara Y, et al. Analysis of a purported SHANK3 mutation in a boy with autism: clinical impact of rare variant research in neurodevelopmental disabilities. Brain Res. 2011;1380:98–105.CrossRefPubMed
50.
Zurück zum Zitat Waga C, Okamoto N, Ondo Y, Fukumura-Kato R, Goto Y, Kohsaka S, et al. Novel variants of the SHANK3 gene in Japanese autistic patients with severe delayed speech development. Psychiatr Genet. 2011;21:208–11.CrossRefPubMed Waga C, Okamoto N, Ondo Y, Fukumura-Kato R, Goto Y, Kohsaka S, et al. Novel variants of the SHANK3 gene in Japanese autistic patients with severe delayed speech development. Psychiatr Genet. 2011;21:208–11.CrossRefPubMed
51.
Zurück zum Zitat Denayer A, Van Esch H, de Ravel T, Frijns JP, Van Buggenhout G, Vogels A, et al. Neuropsychopathology in 7 patients with the 22q13 deletion syndrome: presence of bipolar disorder and progressive loss of skills. Mol Syndromol. 2012;3:14–20.PubMedPubMedCentral Denayer A, Van Esch H, de Ravel T, Frijns JP, Van Buggenhout G, Vogels A, et al. Neuropsychopathology in 7 patients with the 22q13 deletion syndrome: presence of bipolar disorder and progressive loss of skills. Mol Syndromol. 2012;3:14–20.PubMedPubMedCentral
52.
Zurück zum Zitat Jeffries AR, Curran S, Elmslie F, Sharma A, Wenger S, Hummel M, et al. Molecular and phenotypic characterization of ring chromosome 22. Am J Med Genet A. 2005;137:139–47.CrossRefPubMed Jeffries AR, Curran S, Elmslie F, Sharma A, Wenger S, Hummel M, et al. Molecular and phenotypic characterization of ring chromosome 22. Am J Med Genet A. 2005;137:139–47.CrossRefPubMed
53.
Zurück zum Zitat Durand CM, Perroy J, Loll F, Perrais D, Fagni L, Bourgeron T, et al. SHANK3 mutations identified in autism lead to modification of dendritic spine morphology via an actin-dependent mechanism. Mol Psychiatry. 2012;17:71–84.CrossRefPubMed Durand CM, Perroy J, Loll F, Perrais D, Fagni L, Bourgeron T, et al. SHANK3 mutations identified in autism lead to modification of dendritic spine morphology via an actin-dependent mechanism. Mol Psychiatry. 2012;17:71–84.CrossRefPubMed
54.
Zurück zum Zitat Arons MH, Thynne CJ, Grabrucker AM, Li D, Schoen M, Cheyne JE, et al. Autism-associated mutations in ProSAP2/Shank3 impair synaptic transmission and neurexin-neuroligin-mediated transsynaptic signaling. J Neurosci. 2012;32:14966–78.CrossRefPubMedPubMedCentral Arons MH, Thynne CJ, Grabrucker AM, Li D, Schoen M, Cheyne JE, et al. Autism-associated mutations in ProSAP2/Shank3 impair synaptic transmission and neurexin-neuroligin-mediated transsynaptic signaling. J Neurosci. 2012;32:14966–78.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Vucurovic K, Landais E, Delahaigue C, Eutrope J, Schneider A, Leroy C, et al. Bipolar affective disorder and early dementia onset in a male patient with SHANK3 deletion. Eur J Med Genet. 2012;55:625–9.CrossRefPubMed Vucurovic K, Landais E, Delahaigue C, Eutrope J, Schneider A, Leroy C, et al. Bipolar affective disorder and early dementia onset in a male patient with SHANK3 deletion. Eur J Med Genet. 2012;55:625–9.CrossRefPubMed
56.
Zurück zum Zitat Serret S, Thummler S, Dor E, Vesperini S, Santos A, Askenazy F. Lithium as a rescue therapy for regression and catatonia features in two SHANK3 patients with autism spectrum disorder: case reports. BMC Psychiatry. 2015;15:107.CrossRefPubMedPubMedCentral Serret S, Thummler S, Dor E, Vesperini S, Santos A, Askenazy F. Lithium as a rescue therapy for regression and catatonia features in two SHANK3 patients with autism spectrum disorder: case reports. BMC Psychiatry. 2015;15:107.CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat Egger JI, Zwanenburg RJ, van Ravenswaaij-Arts CM, Kleefstra T, Verhoeven WM. Neuropsychological phenotype and psychopathology in seven adult patients with Phelan-McDermid syndrome: implications for treatment strategy. Genes Brain Behav. 2016;15:395–404.CrossRefPubMed Egger JI, Zwanenburg RJ, van Ravenswaaij-Arts CM, Kleefstra T, Verhoeven WM. Neuropsychological phenotype and psychopathology in seven adult patients with Phelan-McDermid syndrome: implications for treatment strategy. Genes Brain Behav. 2016;15:395–404.CrossRefPubMed
58.
Zurück zum Zitat Reierson G, Bernstein J, Froehlich-Santino W, Urban A, Purmann C, Berquist S, et al. Characterizing regression in Phelan McDermid syndrome (22q13 deletion syndrome). J Psychiatr Res. 2017;91:139–44.CrossRefPubMed Reierson G, Bernstein J, Froehlich-Santino W, Urban A, Purmann C, Berquist S, et al. Characterizing regression in Phelan McDermid syndrome (22q13 deletion syndrome). J Psychiatr Res. 2017;91:139–44.CrossRefPubMed
59.
Zurück zum Zitat Verhoeven WM, Egger JI, Willemsen MH, de Leijer GJ, Kleefstra T. Phelan-McDermid syndrome in two adult brothers: atypical bipolar disorder as its psychopathological phenotype? Neuropsychiatr Dis Treat. 2012;8:175–9.CrossRefPubMedPubMedCentral Verhoeven WM, Egger JI, Willemsen MH, de Leijer GJ, Kleefstra T. Phelan-McDermid syndrome in two adult brothers: atypical bipolar disorder as its psychopathological phenotype? Neuropsychiatr Dis Treat. 2012;8:175–9.CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat Han Q, Kim YH, Wang X, Liu D, Zhang ZJ, Bey AL, et al. SHANK3 deficiency impairs heat hyperalgesia and TRPV1 signaling in primary sensory neurons. Neuron. 2016;92:1279–93.CrossRefPubMedPubMedCentral Han Q, Kim YH, Wang X, Liu D, Zhang ZJ, Bey AL, et al. SHANK3 deficiency impairs heat hyperalgesia and TRPV1 signaling in primary sensory neurons. Neuron. 2016;92:1279–93.CrossRefPubMedPubMedCentral
61.
Zurück zum Zitat Wei SC, Yang-Yen HF, Tsao PN, Weng MT, Tung CC, Yu LCH, et al. SHANK3 regulates intestinal barrier function through modulating ZO-1 expression through the PKCε-dependent pathway. Inflamm Bowel Dis. 2017;23:1730–40.CrossRefPubMed Wei SC, Yang-Yen HF, Tsao PN, Weng MT, Tung CC, Yu LCH, et al. SHANK3 regulates intestinal barrier function through modulating ZO-1 expression through the PKCε-dependent pathway. Inflamm Bowel Dis. 2017;23:1730–40.CrossRefPubMed
62.
Zurück zum Zitat Brandt T, Desai K, Grodberg D, Mehta L, Cohen N, Tryfon A, et al. Complex autism spectrum disorder in a patient with a 17q12 microduplication. Am J Med Genet A. 2012;158A:1170–7.CrossRefPubMed Brandt T, Desai K, Grodberg D, Mehta L, Cohen N, Tryfon A, et al. Complex autism spectrum disorder in a patient with a 17q12 microduplication. Am J Med Genet A. 2012;158A:1170–7.CrossRefPubMed
Metadaten
Titel
Delineation of the genetic and clinical spectrum of Phelan-McDermid syndrome caused by SHANK3 point mutations
verfasst von
Silvia De Rubeis
Paige M. Siper
Allison Durkin
Jordana Weissman
François Muratet
Danielle Halpern
Maria del Pilar Trelles
Yitzchak Frank
Reymundo Lozano
A. Ting Wang
J. Lloyd Holder Jr
Catalina Betancur
Joseph D. Buxbaum
Alexander Kolevzon
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Molecular Autism / Ausgabe 1/2018
Elektronische ISSN: 2040-2392
DOI
https://doi.org/10.1186/s13229-018-0205-9

Weitere Artikel der Ausgabe 1/2018

Molecular Autism 1/2018 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie