Next Article in Journal
Bergamottin and PAP-1 Induced ACE2 Degradation to Alleviate Infection of SARS-CoV-2
Next Article in Special Issue
Prenatal SAMe Treatment Changes via Epigenetic Mechanism/s USVs in Young Mice and Hippocampal Monoamines Turnover at Adulthood in a Mouse Model of Social Hierarchy and Depression
Previous Article in Journal
Compositional and Structural Modifications by Ion Beam in Graphene Oxide for Radiation Detection Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Correlation between Phenotype and Genotype in CTNNB1 Syndrome: A Systematic Review of the Literature

1
Department of Family Medicine, Medical Faculty Ljubljana, University of Ljubljana, 1000 Ljubljana, Slovenia
2
Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur 342027, Rajasthan, India
3
Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia
4
Department of Infectious Diseases, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
5
Department of Pediatric Neurology, University Children’s Hospital, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
6
EN-FIST Centre of Excellence, 1000 Ljubljana, Slovenia
7
Center for Developmental Neuroscience, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
*
Author to whom correspondence should be addressed.
These authors have contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(20), 12564; https://doi.org/10.3390/ijms232012564
Submission received: 20 September 2022 / Revised: 10 October 2022 / Accepted: 15 October 2022 / Published: 19 October 2022
(This article belongs to the Special Issue Epigenetic Regulation in Neurodegeneration Disease 2.0)

Abstract

:
The CTNNB1 Syndrome is a rare neurodevelopmental disorder associated with developmental delay, intellectual disability, and delayed or absent speech. The aim of the present study is to systematically review the available data on the prevalence of clinical manifestations and to evaluate the correlation between phenotype and genotype in published cases of patients with CTNNB1 Syndrome. Studies were identified by systematic searches of four major databases. Information was collected on patients’ genetic mutations, prenatal and neonatal problems, head circumference, muscle tone, EEG and MRI results, dysmorphic features, eye abnormalities, early development, language and comprehension, behavioral characteristics, and additional clinical problems. In addition, the mutations were classified into five groups according to the severity of symptoms. The study showed wide genotypic and phenotypic variability in patients with CTNNB1 Syndrome. The most common moderate-severe phenotype manifested in facial dysmorphisms, microcephaly, various motor disabilities, language and cognitive impairments, and behavioral abnormalities (e.g., autistic-like or aggressive behavior). Nonsense and missense mutations occurring in exons 14 and 15 were classified in the normal clinical outcome category/group because they had presented an otherwise normal phenotype, except for eye abnormalities. A milder phenotype was also observed with missense and nonsense mutations in exon 13. The autosomal dominant CTNNB1 Syndrome encompasses a wide spectrum of clinical features, ranging from normal to severe. While mutations cannot be more generally categorized by location, it is generally observed that the C-terminal protein region (exons 13, 14, 15) correlates with a milder phenotype.

1. Introduction

CTNNB1 Syndrome is a severe autosomal dominant neurodevelopmental disorder usually caused by de novo loss-of-function mutations in the CTNNB1 (Cadherin-associated protein, beta 1) gene [1]. CTNNB1 Syndrome manifests itself in a variety of developmental disorders including Neurodevelopmental Disorder with Spastic Diplegia and Visual Defects (NEDSDV), and visual disorders including Familial Exudative Vitreoretinopathy (FEVR). NEDSDV is a neurodevelopmental disorder characterized by global developmental delay, impaired intellectual development with absent or very limited speech, craniofacial anomalies and microencephaly, axial hypotonia, and spasticity [1,2]. FEVR is an autosomal dominant disorder characterized by incomplete development of the retinal vasculature [3]. De novo loss-of-function mutations in the CTNNB1 gene were first discovered in 2012 after diagnostic exome sequencing of individuals with severe intellectual disability [4], and since then the term CTNNB1 Syndrome has become the generic term for all disorders associated with CTNNB1 haploinsufficiency. Currently, this disorder is diagnosed in approximately 300 patients worldwide, although this number is likely an underestimation due to misdiagnosis in cerebral palsy [5,6], leading to efforts to reevaluate the diagnoses of cerebral palsy patients to enable genomics-based changes in their clinical care.
The CTNNB1 gene is located on chromosome 3 (locus 3p22.1, 41240942–41281939). It consists of 16 exons, with exons 2–15 (2346 bp) providing the coding sequence for β-catenin protein. β-catenin protein consists of 781 amino acids and belongs to the armadillo family of structural proteins involved in both embryonic development and adult homeostasis where it plays two essential roles: (1) as a transcriptional co-factor in the Wnt-signaling pathway, and (2) as an anchor in intracellular contacts and cell adhesion [7]. When the Wnt pathway is not stimulated, most of the newly expressed β-catenin is depleted from the cytoplasm by the destruction complex, while the remaining undergraded β-catenin engages with E-cadherin and α-catenin in membrane complexes that serve as cellular anchors. Within the destruction complex, which consists of Axin, Adenomatous Polyposis Coli (APC), and CK140 proteins, β-catenin undergoes a series of consecutive phosphorylations by the glycogen synthase kinase 3β (GSK3β) and CK1 kinases, which ultimately leads to β-catenin ubiquitination by β-TrCP and its proteasomal degradation. In the course of the canonical Wnt signaling pathway, Wnt ligands bind the membrane Frizzled family receptor that stimulates Dishevelled protein to sequester destruction complex proteins [8]. In this way, the degradation of β-catenin is inhibited, allowing the accumulation of free β-catenin which, transported to the nucleus, assists the T-cell factor/lymphoid enhancer factor (TCF-LEF) family of transcription factors in the transcription of various developmental genes, such as axin 1 and cyclin D. Structural and signaling roles of β-catenin are mutually exclusive, which is reflected in its protein structure. β-catenin consists of three regions with a distinct charge distribution: (1) an unstructured N-terminal region (130 amino acids), bearing amino-acid residues important for β-catenin degradation (S33, S37, Y41, S45); (2) a highly conserved central core region (550 amino acids) consisting of 12 armadillo repeats (each is a 42 amino-acid triple helix) [9,10] that form a positively charged groove [9], where β-catenin interacts with more than 20 protein partners including E-cadherin, TCF and degradation complex proteins [10,11,12]; and (3), the unstructured C-terminal region (100 amino acids), which is believed to enhance β-catenin stability by shielding the N-terminus from the destruction complex [9,10,13]. The molecular mechanism of binding exclusivity for the various β-catenin partners remains elusive—it is thought that the occlusion of ligand binding may be achieved by back-folding of termini.
Given the low prevalence of CTNNB1 Syndrome and its relatively recent discovery, little is known about the effect of CTNNB1 mutation type and exonic localization on the severity of clinical phenotypes. It is also not clear whether CTNNB1 mutations are null (in which case the mutated transcripts undergo nonsense-mediated RNA decay—NMD [14], or are translated into non-functional proteins) or, on the other hand, cause a partial loss of protein function due to the presence of an incompletely functioning protein. Another type of mutation can lead to the expression of proteins that interfere with the normal function of the protein from the wild type allele. These so-called antimorphic or dominant-negative mutations (mutated transcripts escape NMD and translate into truncated variants with potentially deleterious effects on the function of the healthy allele) are rare; however, given the variability of CTNNB1 Syndrome-associated mutations in terms of type and location, a production of auto-inhibitory truncated variants cannot be ruled out.
De novo mutations of the CTNNB1 gene have been associated with neurodevelopmental disorders, with cases of intellectual disability and speech delay [4]. Addressing these open questions through phenotype–genotype correlation studies is essential in order to develop targeted interventions and focused clinical care, specific to the mutational context in the affected individuals [15]. The availability of data for such studies has been aided by genomic microarray technology, which has tremendously changed diagnostic approaches in children with neurodevelopmental disorders. Genetic testing can identify the genetic etiology in approximately 40% of cases of cerebral palsy (CP) cases, particularly those diagnosed with autism spectrum disorders (ASD) and intellectual disability (ID) with no apparent causative factor related to CP [5,16]. Access to a large number of patients who have been reliably and systematically assessed is fundamental for understanding CTNNB1 Syndrome. For the first time, this study provides a systematic review of previously reported cases in which we a) analyze the prevalence of clinical manifestations, and b) classify mutations according to their type (missense/nonsense/frameshift/splicing), exonic location, associated clinical features, and disease severity. Based on the analysis of the collected data, the genotype–phenotype correlations for CTNNB1 Syndrome are explored in detail. These may serve as a classification standard for new case studies and as a reference for researchers working to develop personalized therapeutic approaches.

2. Methods

This systematic review’s methodology and presentation follow the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. All the records were managed using the Endnote software program Endnote X4 (Thompson/ISI ResearchSoft Berkley).

2.1. Search Strategy and Inclusion Criteria

We included studies reporting mutations in the CTNNB1 gene associated with NEDSDV and FEVR. Studies were included if they reported germline CTNNB1 mutations, regardless of the amount of detailed phenotypic data on these patients. Language was not a restriction. Reviews, meta-analyses, abstracts, or conference papers, as well as studies on cell and animal models, were excluded from the data analysis.
Studies were identified by searching the following electronic databases from January 2012 to October 2021: PubMed, EMBASE, Web of Science, and CINAHL. The year 2012 was chosen because this was the first time that de novo CTNNB1 mutations were reported [4]. The following key terms were used: CTNNB1 AND (de novo OR loss-of-function OR germline mutations OR novel mutations; see Table 1 for an example of the search strategy in PubMed). To identify all studies that were not found in the literature search, we also screened the bibliographic references of the retrieved studies and reviews.

2.2. Data Extraction

Researchers SM and SK thoroughly reviewed the papers. The following key information was extracted: genetic mutation (exon number, variant, amino acid change, and mutation type), prenatal-neonatal issues (intrauterine growth retardation, IUGR), feeding difficulties, APGAR score), head circumference (presence of microencephaly), presence of muscle tone abnormalities, EEG and MRI results (presence of seizures, structural changes in the brain), dysmorphic features (broad nasal tip, small alae nasi, long and/or flat philtrum, thin upper lip vermillion), eye issues (e.g., strabismus, hyperopia, FEVR), early development (sitting, crawling, walking with support, walking independently), verbal speech and language comprehension (severity of speech delay and level of language comprehension), and behavioral characteristics (e.g., autistic-like or aggressive behavior), and additional clinical issues were extracted when available. During the extractions of genetic information from reviewed studies, we observed several mistakes in reporting exon numbers. Thus, each piece of genetic information was double-checked and estimated from the paper reporting on the genomic organization of the human β-catenin gene [17].

2.3. Quality Assessment

Each case report included in this review was evaluated against the adopted validated tools reported in case reports/case series [18,19,20]. This criterion was adapted to the present research topic (see Table 2 for quality assessment). We included six categories: ‘mutation analysis’, ‘demographic data’, ‘clinical assessment’, ‘cognitive assessment’ and ‘neuroimaging’, and ’neurophysiological investigation’. The highest possible score was 8. Case reports with a score of 7 or more were considered to be high quality, reports with a score of 5 or more were considered to be of moderate quality, and reports with a score of 4 or less were considered to be low quality. No data were excluded from this review, although case reports scoring as low quality were only included in the Supplementary Materials and excluded from the genotype–phenotype analysis.

2.4. Genotype-Phenotype Analysis

In analyzing the patients’ mutation data, we classified mutations into five groups, according to the severity of symptoms: ‘normal’, ‘mild’, ‘moderate’, ‘moderate-severe’, and ‘severe’. Because this is the first attempt to perform genotype–phenotype analysis, there is no previous literature available for this rare syndrome, so we could not refer to the existing literature. Thus, the classification was based mainly on the patient’s eye contact (present/absent), speech (Viking Speech Scale, Pennington, 2010) [21], and cognition (ID; present/absent). Motor development was assessed only for normal and mild phenotypes because motor development in the moderate and severe groups was less related to phenotype/symptom severity. The classification into five severity groups based on phenotypic and symptomatic characteristics formed the basis for the assessment of genotypic–phenotypic correlation (Table 3).

3. Results

The search strategy identified 1221 articles and one study was additionally included based on the searches in previous reviews. After removal of duplicates and articles that did not meet the inclusion criteria, 28 articles were included in the review (see Figure 1 for flow diagram) [1,2,4,5,7,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44]. Of the 28 articles, data from 84 patients with a CTNNB1 mutation were pooled and were available for analysis. Data from all 84 patients are described in Table S1 (Supplement A) and in the text (see Section 3.1); however, only 35 patients with sufficient data were included in the main genotype–phenotype analysis.

3.1. Prevalence of Clinical Features

Clinical features are presented based on primary (>50%) and secondary criteria (20–49%) (see Table 4 and Figure 2 and Figure 3), which were established to distinguish between more common and less common features. Facial dysmorphism was one of the most commonly reported clinical features (>86.8% of cases), including small alae nasi, long and/or flat philtrum, thin upper lip vermillion, and broad nasal tip. The presence of microencephaly (occipitofrontal circumference (OFC) less than 3 SD) was noted in 73.7% of cases, while the majority of the remaining cases had an OFC smaller than average. Reported cases exhibited eye abnormalities, including strabismus (52.6%), FEVR (22.8%), hyperopia (14%), astigmatism (8.8%), myopia (5.8%), esotropia (5.3%), retinal detachment (3.8%), and optic atrophy (1.9%). Muscle tone abnormalities were found in the majority of the reported cases, including axial hypotonia (91.5%), peripheral spasticity (84.7%), and dystonia, which was reported in 11 cases and not systematically assessed.
An electroencephalogram (EEG) was performed and reported in 30 patients, of whom 27 reported normal EEG (90%), whereas three patients had abnormal EEG (e.g., diffuse fast background activity, epileptiform activity with a tendency to spread). In addition, one patient’s report described focal epilepsy; however, it was not clear whether an EEG had been performed [5]. Magnetic resonance imaging (MRI) results were available for 24 cases of which 20 reports were normal (83.3%). Abnormal results were reported in four cases. These included arachnoid cysts, an enlarged Sylvian sulcus, hypoplasia of the corpus callosum, osteolytic lesions, enlarged lateral ventricles, abnormal gyration of the temporal lobe, absence of the right fornix and a hypoplastic brainstem, delayed myelination in the frontal lobes, mild dilatation of the ventricles, and mild thinning of the corpus callosum.
The gross motor milestone “sitting” was reported for 27 cases, of which 21 (77.7%) had reached this milestone at the mean age of 16 months. The remaining six had not reached it at that time, however, only three of them were older (age > 30 months) and the others still had time to reach this milestone (age ≤ 15 months). Of the 40 reported cases, 24 (60%) were able to walk independently, although most of them had difficulties (e.g., ataxic, unstable gait, use of an orthosis to stabilize the ankle). The average age for reaching this milestone was 3.8 years (range 12 months to 8 years). Of the 46 reported cases for speech development (Table S1, Supplement A), 14 cases were nonverbal (30.4%), 19 cases used few words (41.3%), eight cases were able to speak short sentences (17.4%), and three cases were able to speak complete sentences and were only mildly delayed (6.5%). Two cases had no language delays in speech and had been achieving age-appropriate speech language milestones. In the majority of reported cases, receptive language was significantly better than expressive language. Of 25 reported cases, 13 (52%) reported “good” language comprehension, 11 (44%) reported “basic” language comprehension, and one reported “poor” language comprehension.
In most cases, the behavior was problematic. This information was available for 42 cases. Seven cases (16.6%) were described as ‘friendly and sociable’ and ‘with a generally cheerful demeanor’. In other cases, behavioral difficulties such as aggression (47.6%) were noted, ten cases showed stereotypic behavior (23.8%) and nine cases were diagnosed with autism spectrum disorder (21.4%). Three cases were diagnosed with ADHD (7.1%) and two with schizophrenia (4.8%). Data from eight cases indicated sleep problems, either in infancy (difficulty falling asleep) or in toddlerhood (night-time laughing fits).
Additional clinical features were considered to be rare (two or fewer cases; see Table S1, Supplement A): Scoliosis, osteogenesis imperfecta, blue sclera, sacral dimple, left clubfoot, increased dermatoglyphic whorls, type 1 diabetes, dysplastic bicuspid pulmonary valve, delayed bone age, absent left testis, brachydactyly, Achilles tendon contracture, abnormal lung growth, pulmonary hypertension, mild thumb adduction, eczema, bicoronal craniosynostosis, single supernumerary maxillary incisor, bilateral orchidopexy, syringomyelia, hypermobile joints, and glue ear.

3.2. Genotype

The CTNNB1 gene is located on chromosome 3 and spans 40.94 kb wherein the coding region is 2345 nt in length encoding a protein of 781 amino acids. β-catenin belongs to the armadillo family of structural proteins and is composed of three regions: an unstructured N-terminal region, bearing amino-acid residues important for β-catenin degradation; a highly conserved central core region consisting of 12 armadillo repeats, where several important interaction regions of β-catenin with many different protein partners reside; and the unstructured C-terminal region (Figure 4 and Figure 5).
Our dataset of 84 patients diagnosed with CTNNB1 Syndrome shows that CTNNB1 genetic mutations are scattered throughout the gene with the majority of mutations located in the central armadillo repeat region (75.3%). The remaining mutations are roughly equally distributed between the N-terminal domain (10.6%) and the C-terminal domain (14.1%) (Figure 6).
Most of the mutations leading to CTNNB1 Syndrome were nonsense mutations (47.6%), followed by frameshift mutations (34.5%), missense mutations (8.3%), splice mutations (7.1%), and complete gene deletions (2.4%) (Figure 5). For all six missense mutations, American College of Medical Genetics and Genomics (ACMG) scores were extracted from both ClinVar and the Human Gene Mutation Database (HGMD). This information was available for three of six cases. Mutations c.1163T > C and c.2128C > T are classified as mutations of ‘Uncertain Significance’ and mutation c.1723G > A is classified as ‘Pathogenic/Likely Pathogenic’. The occurrence of mutations was found in all exons, except exon 1 and 2. The most frequently reported mutations include mutations in exon 10 (c.1603C > T, p.R535*), exon 9 (c.1420C > T, p.R474*), exon 7 (c.998dupA, c.999C > G, c.999del causing p.Y333*), intron 5 (c.734 + 1G > T, c.734 + 1G > A, causing splice mutation), and exon 13 (c.2038_2041dup, p.S681*) (Figure 6). Specific mutations in exon 9 (c.1272_1275del, p.Ser425Thrfs*11, and p.Glu479Argfs*18) and exon 4 (c.283C > T, p.R95*) occurred twice. Notably, there were two patients with reported gross deletion of the entire gene (Figure 7 and Figure 8). Other mutations are listed in the Table S1 (Supplement A). All but six cases found in three articles were reported as de novo mutations [24,29,35].

3.3. Phenotype–Genotype Correlation Analysis

There were 35 case reports that sufficiently met the study criteria and were further selected for detailed genotype–phenotype correlation analysis. As described previously, patients were divided into five groups according to the severity of the phenotype (Table 5 and Table 6).

3.3.1. Severe Phenotype

Five cases included in this category had presented a severe speech impairment (absent speech or being able to speak only two words) and poor or no eye contact. Mutations were observed in intron 5 (n = 3, splice site mutation) and exon 6 (n = 2, frameshift and nonsense). Compared with the other groups, this group had the lowest IQ, indicating severe ID (IQ < 70). These cases showed more ritualistic and autistic-like behavior along with auto-aggressive behavior. The ability to walk was reported in four cases, and three of them were able to walk independently. The majority of the ‘severe group’ had unremarkable brain MRI and EEG reports and presented with axial hypotonia, peripheral spasticity, facial dysmorphism, and eye abnormalities (FEVR = 2, strabismus = 2 and hyperopia = 1); see Table S1, Supplement A). Of the three patients reported, two had microencephaly, and one female patient (c.755delTinsAAC, p.Leu252*) had an OFC in the 13th percentile.

3.3.2. Moderate-Severe Phenotype

Thirteen patients with a moderate-severe phenotype had mutations in exon 4 (p.Tyr142Valfs*4), exon 8 (p.Leu388Pro), exon 9 (p.Ser425Thrfs*11, p.Arg449GlnfsTer24, p.Arg474Ter), exon 10 (p.Arg535*), exon 11 (p.Gln601*), and exon 12 (Glu642Argfs*6, p.Glu642Valfs*5). The main characteristics of this group were severe speech impairments (no speech/few words; 92.3%) and inability to walk independently (84.6%). One patient lost the ability to walk due to progressive spasticity. The majority of included cases had good language comprehension (six out of seven reported cases; 87.5%). Eleven reports included information on behavioral problems, including aggression (72.7%), tantrums (45.5%), and stereotypic behavior (36.4%). All reported cases showed facial dysmorphism, axial hypotonia, and peripheral spasticity. All but one case had normal brain MRI and EEG reports. One case with missense mutation reported epileptiform activity with a tendency to spread. Seven cases had microencephaly (53.8%), and the remaining six cases had head size smaller than the 20th percentile. Eight cases reported strabismus, and hyperopia, optic atrophy, and FEVR were each reported one time (Table S1, Supplement A).

3.3.3. Moderate Phenotype

Twelve patients were included in this category. Mutations were present throughout the gene in exon 3 (p.Gly34Asnfs*15, p.Gln78*), exon 4 (p.Arg95*, p.Tyr142Valfs*4), exon 5 (p.Gly236Argfs*35), exon 7 (p.Gln309*, p.Tyr333Ter), Intron 7, and exon 10 (p.Gln538*), and two patients had entire gene deletions. This group is characterized by higher walking ability: Ten cases were able to walk (83.3%), and two cases may still have had time to start walking (ages 3 and 4). Seven reported cases showed good speech development (ability to combine words in seven cases) and the remaining five cases had only recently started speaking and may improve over time. Comprehension was good in all reported cases. Five patients in this group were described as having a friendly and sociable personality (41.6%), five currently or in the past exhibited aggressive behavior (41.6%), two exhibited temper tantrums (16.6%), and two exhibited stereotypical behavior (16.6%). Axial hypotonia was present in all twelve cases, while peripheral spasticity was present in eight cases (75%). All cases presented with CTNNB1-related facial dysmorphism (broad nasal tip, small alae nasi, long and flat philtrum, thin upper lip vermillion), and all but one case with eye abnormalities (strabismus = 5, hyperopia = 3, hypermetropia = 1, and esotropia = 1). Five cases were found to have microencephaly, while the rest (n = 7) had OFC smaller than the 33th percentile. EEG was reported normal in all except one case (diffused fast background activity during episodes; p.Gln78*). MRI reports showed abnormal results in four cases (see Section 3.1 for details of abnormal results).

3.3.4. Mild Phenotype

Three patients were included in this group and all had a nonsense mutation in exon 13 (p.Ser681*, p.Arg661*). Two reported cases were siblings who both had a similar phenotype: mild ID with good language comprehension, and mild expressive speech impairment, such that they were able to speak in complete sentences. Both siblings were able to sit before their first birthday and walk independently before their second birthday. The report for the third patient was similar (sitting at 11 months, walking independently at 30 months, and speaking their first words before age four). All of these cases had behavioral problems. The siblings displayed symptoms of ADHD, autism, anxiety, aggression, and frustration, and would occasionally self-harm. The non-sibling case exhibited ‘obsessional behavior’ (no further data available). Both siblings, clearly with borderline intellectual abilities or mild intellectual disability, could speak in sentences, and reportedly enjoyed social and verbal interaction with others. Their expressive speech was limited, while their language comprehension was better. Available reports (for siblings only) showed normal EEG and MRI scans. All three cases presented with axial hypotonia, and only the siblings showed peripheral spasticity. One of the siblings had an average head circumference, while the other two cases showed OFC of −3SD and −6SD, indicating microencephaly. All three patients showed facial dysmorphism, including thin upper lip vermilion; however, only data from the siblings include a more detailed report on craniofacial features: broad nasal tip small alae nasi, and long and flat philtrum. Both siblings presented with strabismus and myopia, while the third patient had no data on the presence of eye abnormalities.

3.3.5. Normal Phenotype

Two reported cases, a missense mutation in exon 14 (c.2128C > T, p.Arg710Cys) and a nonsense mutation in exon 15 (c.2142_2157dup16, p.His720*) presented with the Retinal Vascular Condition FEVR, but were otherwise developing within the normal range.

4. Discussion

The present paper provides a comprehensive and up-to-date review of published cases of CTNNB1 Syndrome, an analysis of the prevalence of the most common symptoms, and a classification of CTNNB1-associated mutations according to the severity of their respective phenotypes (Table 5, Table 6 and Table S1, Supplement A). Based on the availability of sufficient data, 35 patients were included in the analysis. While we acknowledge that the size of the analyzed cohort was too small to perform a statistically significant genotype–phenotype correlation, we believe that the classification performed in our study may nevertheless be informative for future correlation studies and provide the classification basis for further data collection and analysis.
The main finding of this paper is that there is substantial variability within genotypes and phenotypes of patients with CTNNB1 Syndrome. Regarding genotype, we found that mutations associated with CTNNB1 Syndrome are scattered throughout the coding sequence of the gene, with the exception of the first coding exon (exon 2), although we cannot exclude the possibility that pathological mutations also occur in this exon. In terms of phenotype, we were able to classify patients into a spectrum of disease severity (severe, severe-moderate, moderate, mild, and normal). Our analysis of the mutations available in each phenotype category suggested certain relationship between phenotype severity and mutation location and type. The majority of the mutations analyzed were associated with moderate or severe disease phenotypes, manifested by facial dysmorphisms, microcephaly, various motor disabilities, speech and cognitive impairments, and behavioral difficulties. From a biochemical perspective, this was expected because the critical interaction surface of β-catenin is large, extending from armadillo repeat 3 to 9 encoded in exons 5 to 10, respectively (Figure 3, Figure 4, Figure 5 and Figure 6).
Consistent with these expectations, the analyzed nonsense and missense mutations occurring in exons 14 and 15, which are part of the C-terminal domain of β-catenin, were classified in the normal clinical outcome as they presented with eye abnormalities only, and otherwise had a normal phenotype. Both mutations were found in all family members, which suggests that they were inherited rather than occurred de novo [35]. Interestingly, a classic ophthalmological feature of CTNNB1 Syndrome—FEVR—was found in many of these patients (Table S1, Supplement A), suggesting that it may be caused by alterations at the uncharacterized C-terminal domain. Biochemically, nonsense mutations impose a premature stop codon that results in a truncated protein, while missense mutations lead to the substitution of a single amino-acid in the protein sequence, in which case the severity of the consequences depend on the structural integrity of the mutant protein and possible disruption of binding sites for interacting proteins. Lack of disease severity for mutations in exons 14–15 is most likely attributed to the fact that a large part of the protein, should the transcript circumvent nonsense-mediated decay, is transcribed and is likely to be, at least partially, functional. Such hypomorphic mutations have also been observed in other genes (e.g., DMD, APC) [45,46]. Furthermore, helix C, a critical structural motif of the C-terminal domain with a role in the co-transcriptional activity of β-catenin [47], is encoded by exon 13; therefore, it lies upstream of mutations in exons 14–15. Accordingly, patients with mutations in exon 13 exhibit an array of additional cognitive and motor impairments (Table 5). One of the reported nonsense mutations in exon 13 is a mutation of a tyrosine residue at position 654 (Y654; Table S1, Supplement A), which, in its phosphorylated state, is directly linked to stabilizing helix C, and thus allows accessibility to co-activators of β-catenin-mediated transcription leading to nuclear localization of β-catenin [48]. However, the mutations in exon 13 (c.2038_2041dup, p.Ser681* and c.1981C > T, p.Arg661*) were at worst classified into the mild-moderate phenotype category, which can be attributed to the fact that, while important for signaling, helix C appears to be completely dispensable for the structural role of β-catenin in cell-cell adhesion [9].
An additional explanation for the milder phenotype in mutations in exons 14–15 could be that helix C, which is the most important feature of the C-terminal domain, critical for the co-transcriptional role of β-catenin [47], is encoded upstream, in exon 13. Accordingly, patients with mutations in this region exhibit a range of additional cognitive and motor impairments (Table 5). One of the reported nonsense mutations in exon 13 occurs at the Y654 residue (Table S1, Supplement A), which, in its phosphorylated state, is directly linked to keeping helix C accessible to co-activators of β-catenin-mediated transcription, and is consequently responsible for the nuclear localization of β-catenin [48]. Still, the mutations in exon 13 (c.2038_2041dup, p.Ser681* and c.1981C > T, p.Arg661*) were at worst classified into the mild-moderate phenotype category, which can be attributed to the fact that, while important for signaling, helix C appears to be completely dispensable for the structural role of β-catenin in cell-cell adhesion [9].
In contrast to nonsense and missense mutations, we found that frameshift mutations in the C-terminal region can cause a severe disease phenotype. A patient with a frameshift mutation (c.2273delA, p.His758Leufs*30) in exon 15 (Table S1, Supplement A) presented with severe intellectual disability and symptoms of autism spectrum disorder [38]. Frameshift mutations can lead to a variety of changes, from the introduction of a premature stop codon to a protein extension beyond its normal stop codon. The consequences of these changes are difficult to predict without experimental analysis; however, in general, such mutations are considered deleterious [49] because they can undermine the structural integrity of the entire protein. Furthermore, splicing mutations have also been found to cause severe or moderate disease. In our analysis, splice mutations were located at the 3′ acceptor splice sites of intron 5 and 7, with patients presenting mainly with severe cognitive disability and lack of eye contact, while the most robust characteristics of the cases categorized as moderate-severe included their inability to walk (exons from 8–12). Splicing mutations likely behave like frameshift mutations in that they can cause retention of the intron during the process of splicing, which can be detrimental for the reading frame. Interestingly, a pair of patients with identical mutations (c.2092_2096dup, p.Ile700Leufs*37) exhibited varied symptoms [24]. Thus, the influence of a so-called modifier gene and/or additional environmental factors may play an important role in disease manifestation.
An additional interesting observation was that gross gene deletions resulted in a moderately pathological phenotype. Considering that these types of mutations likely result in null activity of β-catenin, we would expect them to cause the most severe phenotype possible. However, because many point mutations from our review appear to lead to a more severe phenotype, this is a possible indicator of dominant-negative effects. Dominant-negative effects are a consequence of mutations that lead to the expression of truncated proteins that obstruct the normal function of the protein from the wild type allele. It has been reported that β-catenin truncations without a C-terminal domain, which lack transcriptional activity, nevertheless bind co-transcription activators, and thus interfere with their binding to the wild type variants [50].
The present study showed that mutations associated with CTNNB1 Syndrome are also found in exon 3, although mutations in this region are usually associated with cancer, because exon 3 bears important amino-acid residues for β-catenin degradation, such as the CK1-α phosphorylation site (S45), GSK3-β phosphorylation sites (S33, S37, and T41), and Fbw1 (D32 and G34). These are essential components of the inhibitory destruction complex that controls the levels of the free cytosolic β-catenin [51,52]. Missense mutations in these residues have been associated with colorectal cancers and other types of malignancies (e.g., melanoma, brain tumors) as they lead to the accumulation of nuclear β-catenin and tumorigenesis [53]. In our data, four CTNNB1 Syndrome-causing nonsense and frameshift mutations in exon 3 were reported (G34, E54, Q72, and Q78), which were associated with a moderate phenotype. There is no evidence that these mutations cause tumor growth and progression.
Our review could not confirm the results reported by Rossetti et al. (2021) that missense mutations are associated with vitreoretinopathy [22]. This condition was found in 13 cases (see Table S1, Supplement A), with no correlation with mutation type; however, we observed a higher prevalence of this condition in the Chinese population (found in 69.2% of Chinese patients) [24,29,33,35,39,40]. The FEVR condition affects the retina by preventing blood vessels from forming at the edges of the retina. It is predicted that reduced β-catenin levels increase the probability of causing FEVR, although the exact pathophysiology is yet unclear [35]. This condition is progressive in nature. Thus, regular screening of the retina can prevent unnecessary vision loss in patients with CTNNB1 Syndrome.
Some methodological considerations should be taken into account. The proportion of incomplete clinical data is perhaps the most significant and troubling issue in the current study. Most of the included studies were case reports or small case series, so referral, selection, and publication biases could occur. The results of our systematic review of the genotype–phenotype correlation are based on the data reported in the previous studies. It is, therefore, necessary to validate these results in prospectively gathered data and cell models. Importantly, this systematic review makes a clear demarcation of the exon borders of β-catenin, based on the article of Nollet et al. 1996 [17], and can guide future studies. The organization of the clinical criteria based on the prevalence of the phenotypical findings may help neurologists to determine whether they will screen their patients for CTNNB1 Syndrome. Based on the phenotype, this gene can be added to the appropriate panels.

5. Conclusions

In conclusion, it was possible to observe at least some evidence of genotype-phenotype correlation between the type and position of pathogenic variants and clinical expression. Most importantly, we found that nonsense and missense mutations in exon 14 and 15 result in a functional protein with a neurotypical phenotype and ocular abnormalities only, whereas the nonsense and missense mutations on exon 13 result in a milder phenotype. Splice mutations in intron 6 and mutations at the exon can lead to a severe phenotype. Mutations between E8 and E12 could be associated with severe motor disabilities. Global developmental delays, speech impairments, craniofacial features, and eye problems are the commonly observed phenotypes in almost all reported mutations. Future implications for neurologists, researchers, and patient advocates include using the developed correlation to predict the patient’s potential phenotype.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms232012564/s1.

Author Contributions

Conceptualization, Š.M., S.K., P.S., R.J. and D.O.; methodology, Š.M., S.K., P.S., N.Ž., V.F., D.L. and D.G.; software, Š.M., S.K. and V.F.; validation, Š.M., S.K., R.J. and D.O.; formal analysis, Š.M. and S.K.; investigation, Š.M., S.K., P.S., R.J. and D.O.; data curation, Š.M., S.K., D.G., D.L.; writing—original draft preparation, Š.M., S.K.; writing—review and editing, all authors; visualization, Š.M, S.K., N.Ž., D.L. and V.F.; supervision, R.J. and D.O. All authors have read and agreed to the published version of the manuscript.

Funding

The authors acknowledge the funding from the Slovenian Research Agency ARRS (SM: Program MR-39262; PS: Z1-3193; RJ: P4-0176; DO: J7-4537).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Verhoeven, W.M.A.; Egger, J.I.M.; Jongbloed, R.E.; van Putten, M.M.; de Bruin-van Zandwijk, M.; Zwemer, A.S.; Pfundt, R.; Willemsen, M.H. A de novo CTNNB1 Novel Splice Variant in an Adult Female with Severe Intellectual Disability. Int. Med. Case Rep. J. 2020, 13, 487–492. [Google Scholar] [CrossRef] [PubMed]
  2. Kuechler, A.; Willemsen, M.H.; Albrecht, B.; Bacino, C.A.; Bartholomew, D.W.; van Bokhoven, H.; van den Boogaard, M.J.; Bramswig, N.; Buttner, C.; Cremer, K.; et al. De novo mutations in beta-catenin (CTNNB1) appear to be a frequent cause of intellectual disability: Expanding the mutational and clinical spectrum. Hum. Genet. 2015, 134, 97–109. [Google Scholar] [CrossRef] [PubMed]
  3. Gilmour, D.F. Familial exudative vitreoretinopathy and related retinopathies. Eye 2015, 29, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. de Ligt, J.; Willemsen, M.H.; van Bon, B.W.; Kleefstra, T.; Yntema, H.G.; Kroes, T.; Vulto-van Silfhout, A.T.; Koolen, D.A.; de Vries, P.; Gilissen, C.; et al. Diagnostic exome sequencing in persons with severe intellectual disability. N. Engl. J. Med. 2012, 367, 1921–1929. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Jin, S.C.; Lewis, S.A.; Bakhtiari, S.; Zeng, X.; Sierant, M.C.; Shetty, S.; Nordlie, S.M.; Elie, A.; Corbett, M.A.; Norton, B.Y.; et al. Mutations disrupting neuritogenesis genes confer risk for cerebral palsy. Nat. Genet. 2020, 52, 1046–1056. [Google Scholar] [CrossRef]
  6. Moreno-De-Luca, A.; Millan, F.; Pesacreta, D.R.; Elloumi, H.Z.; Oetjens, M.T.; Teigen, C.; Wain, K.E.; Scuffins, J.; Myers, S.M.; Torene, R.I.; et al. Molecular Diagnostic Yield of Exome Sequencing in Patients With Cerebral Palsy. JAMA 2021, 325, 467–475. [Google Scholar] [CrossRef]
  7. Li, N.; Xu, Y.; Li, G.; Yu, T.; Yao, R.E.; Wang, X.; Wang, J. Exome sequencing identifies a de novo mutation of CTNNB1 gene in a patient mainly presented with retinal detachment, lens and vitreous opacities, microcephaly, and developmental delay: Case report and literature review. Medicine 2017, 96, e6914. [Google Scholar] [CrossRef]
  8. Schwarz-Romond, T.; Metcalfe, C.; Bienz, M. Dynamic recruitment of axin by Dishevelled protein assemblies. J. Cell Sci. 2007, 120, 2402–2412. [Google Scholar] [CrossRef] [Green Version]
  9. Xing, Y.; Takemaru, K.; Liu, J.; Berndt, J.D.; Zheng, J.J.; Moon, R.T.; Xu, W. Crystal structure of a full-length beta-catenin. Structure 2008, 16, 478–487. [Google Scholar] [CrossRef] [Green Version]
  10. Huber, A.H.; Weis, W.I. The structure of the beta-catenin/E-cadherin complex and the molecular basis of diverse ligand recognition by beta-catenin. Cell 2001, 105, 391–402. [Google Scholar] [CrossRef]
  11. Huber, A.H.; Nelson, W.J.; Weis, W.I. Three-dimensional structure of the armadillo repeat region of beta-catenin. Cell 1997, 90, 871–882. [Google Scholar] [CrossRef] [Green Version]
  12. Tian, X.; Liu, Z.; Niu, B.; Zhang, J.; Tan, T.K.; Lee, S.R.; Zhao, Y.; Harris, D.C.; Zheng, G. E-cadherin/beta-catenin complex and the epithelial barrier. J. Biomed. Biotechnol. 2011, 2011, 567305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Mo, R.; Chew, T.L.; Maher, M.T.; Bellipanni, G.; Weinberg, E.S.; Gottardi, C.J. The terminal region of beta-catenin promotes stability by shielding the Armadillo repeats from the axin-scaffold destruction complex. J. Biol. Chem. 2009, 284, 28222–28231. [Google Scholar] [CrossRef] [Green Version]
  14. Neu-Yilik, G.; Amthor, B.; Gehring, N.H.; Bahri, S.; Paidassi, H.; Hentze, M.W.; Kulozik, A.E. Mechanism of escape from nonsense-mediated mRNA decay of human beta-globin transcripts with nonsense mutations in the first exon. RNA 2011, 17, 843–854. [Google Scholar] [CrossRef] [Green Version]
  15. Wright, C.F.; Fitzgerald, T.W.; Jones, W.D.; Clayton, S.; McRae, J.F.; van Kogelenberg, M.; King, D.A.; Ambridge, K.; Barrett, D.M.; Bayzetinova, T.; et al. Genetic diagnosis of developmental disorders in the DDD study: A scalable analysis of genome-wide research data. Lancet 2015, 385, 1305–1314. [Google Scholar] [CrossRef] [Green Version]
  16. Segel, R.; Ben-Pazi, H.; Zeligson, S.; Fatal-Valevski, A.; Aran, A.; Gross-Tsur, V.; Schneebaum-Sender, N.; Shmueli, D.; Lev, D.; Perlberg, S.; et al. Copy number variations in cryptogenic cerebral palsy. Neurology 2015, 84, 1660–1668. [Google Scholar] [CrossRef] [PubMed]
  17. Nollet, F.; Berx, G.; Molemans, F.; van Roy, F. Genomic organization of the human beta-catenin gene (CTNNB1). Genomics 1996, 32, 413–424. [Google Scholar] [CrossRef]
  18. Murad, M.H.; Sultan, S.; Haffar, S.; Bazerbachi, F. Methodological quality and synthesis of case series and case reports. BMJ Evid. Based Med. 2018, 23, 60–63. [Google Scholar] [CrossRef] [Green Version]
  19. Orrico, A.; Galli, L.; Cavaliere, M.L.; Garavelli, L.; Fryns, J.P.; Crushell, E.; Rinaldi, M.M.; Medeira, A.; Sorrentino, V. Phenotypic and molecular characterisation of the Aarskog-Scott syndrome: A survey of the clinical variability in light of FGD1 mutation analysis in 46 patients. Eur. J. Hum. Genet. 2004, 12, 16–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Zanetti Drumond, V.; Sousa Salgado, L.; Sousa Salgado, C.; Oliveira, V.A.L.; de Assis, E.M.; Campos Ribeiro, M.; Furtado Valadao, A.; Orrico, A. The Prevalence of Clinical Features in Patients with Aarskog-Scott Syndrome and Assessment of Genotype-Phenotype Correlation: A Systematic Review. Genet. Res. 2021, 2021, 6652957. [Google Scholar] [CrossRef]
  21. Pennington, L.; Virella, D.; Mjoen, T.; da Graca Andrada, M.; Murray, J.; Colver, A.; Himmelmann, K.; Rackauskaite, G.; Greitane, A.; Prasauskiene, A.; et al. Development of The Viking Speech Scale to classify the speech of children with cerebral palsy. Res. Dev. Disabil. 2013, 34, 3202–3210. [Google Scholar] [CrossRef] [PubMed]
  22. Rossetti, L.Z.; Bekheirnia, M.R.; Lewis, A.M.; Mefford, H.C.; Golden-Grant, K.; Tarczy-Hornoch, K.; Briere, L.C.; Sweetser, D.A.; Walker, M.A.; Kravets, E.; et al. Missense variants in CTNNB1 can be associated with vitreoretinopathy-Seven new cases of CTNNB1-associated neurodevelopmental disorder including a previously unreported retinal phenotype. Mol. Genet. Genom. Med. 2021, 9, e1542. [Google Scholar] [CrossRef]
  23. Winczewska-Wiktor, A.; Badura-Stronka, M.; Monies-Nowicka, A.; Nowicki, M.M.; Steinborn, B.; Latos-Bielenska, A.; Monies, D. A de novo CTNNB1 nonsense mutation associated with syndromic atypical hyperekplexia, microcephaly and intellectual disability: A case report. BMC Neurol. 2016, 16, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ho, S.; Tsang, M.H.; Fung, J.L.; Huang, H.; Chow, C.B.; Cheng, S.S.; Luk, H.M.; Chung, B.H.; Lo, I.F. CTNNB1-related neurodevelopmental disorder in a Chinese population: A case series. Am. J. Med. Genet. Part A 2021, 188, 130–137. [Google Scholar] [CrossRef] [PubMed]
  25. Retterer, K.; Juusola, J.; Cho, M.T.; Vitazka, P.; Millan, F.; Gibellini, F.; Vertino-Bell, A.; Smaoui, N.; Neidich, J.; Monaghan, K.G.; et al. Clinical application of whole-exome sequencing across clinical indications. Genet. Med. 2016, 18, 696–704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Posey, J.E.; Harel, T.; Liu, P.; Rosenfeld, J.A.; James, R.A.; Coban Akdemir, Z.H.; Walkiewicz, M.; Bi, W.; Xiao, R.; Ding, Y.; et al. Resolution of Disease Phenotypes Resulting from Multilocus Genomic Variation. N. Engl. J. Med. 2017, 376, 21–31. [Google Scholar] [CrossRef]
  27. Levchenko, A.; Davtian, S.; Freylichman, O.; Zagrivnaya, M.; Kostareva, A.; Malashichev, Y. Beta-catenin in schizophrenia: Possibly deleterious novel mutation. Psychiatry Res. 2015, 228, 843–848. [Google Scholar] [CrossRef]
  28. Tucci, V.; Kleefstra, T.; Hardy, A.; Heise, I.; Maggi, S.; Willemsen, M.H.; Hilton, H.; Esapa, C.; Simon, M.; Buenavista, M.T.; et al. Dominant beta-catenin mutations cause intellectual disability with recognizable syndromic features. J. Clin. Investig. 2014, 124, 1468–1482. [Google Scholar] [CrossRef] [Green Version]
  29. Wang, H.; Zhao, Y.; Yang, L.; Han, S.; Qi, M. Identification of a novel splice mutation in CTNNB1 gene in a Chinese family with both severe intellectual disability and serious visual defects. Neurol. Sci. 2019, 40, 1701–1704. [Google Scholar] [CrossRef] [Green Version]
  30. Kharbanda, M.; Pilz, D.T.; Tomkins, S.; Chandler, K.; Saggar, A.; Fryer, A.; McKay, V.; Louro, P.; Smith, J.C.; Burn, J.; et al. Clinical features associated with CTNNB1 de novo loss of function mutations in ten individuals. Eur. J. Med. Genet. 2017, 60, 130–135. [Google Scholar] [CrossRef]
  31. Grozeva, D.; Carss, K.; Spasic-Boskovic, O.; Tejada, M.I.; Gecz, J.; Shaw, M.; Corbett, M.; Haan, E.; Thompson, E.; Friend, K.; et al. Targeted Next-Generation Sequencing Analysis of 1,000 Individuals with Intellectual Disability. Hum. Mutat. 2015, 36, 1197–1204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Prasad, M.K.; Geoffroy, V.; Vicaire, S.; Jost, B.; Dumas, M.; Le Gras, S.; Switala, M.; Gasse, B.; Laugel-Haushalter, V.; Paschaki, M.; et al. A targeted next-generation sequencing assay for the molecular diagnosis of genetic disorders with orodental involvement. J. Med. Genet. 2016, 53, 98–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Sun, W.; Xiao, X.; Li, S.; Jia, X.; Wang, P.; Zhang, Q. Germline Mutations in CTNNB1 Associated With Syndromic FEVR or Norrie Disease. Investig. Ophthalmol. Vis. Sci. 2019, 60, 93–97. [Google Scholar] [CrossRef] [Green Version]
  34. Cordeiro, D.; Bullivant, G.; Siriwardena, K.; Evans, A.; Kobayashi, J.; Cohn, R.D.; Mercimek-Andrews, S. Genetic landscape of pediatric movement disorders and management implications. Neurol. Genet. 2018, 4, e265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Panagiotou, E.S.; Sanjurjo Soriano, C.; Poulter, J.A.; Lord, E.C.; Dzulova, D.; Kondo, H.; Hiyoshi, A.; Chung, B.H.; Chu, Y.W.; Lai, C.H.Y.; et al. Defects in the Cell Signaling Mediator beta-Catenin Cause the Retinal Vascular Condition FEVR. Am. J. Hum, Genet. 2017, 100, 960–968. [Google Scholar] [CrossRef] [Green Version]
  36. Coussa, R.G.; Zhao, Y.; DeBenedictis, M.J.; Babiuch, A.; Sears, J.; Traboulsi, E.I. Novel mutation in CTNNB1 causes familial exudative vitreoretinopathy (FEVR) and microcephaly: Case report and review of the literature. Ophthalmic Genet. 2020, 41, 63–68. [Google Scholar] [CrossRef]
  37. Dixon, M.W.; Stem, M.S.; Schuette, J.L.; Keegan, C.E.; Besirli, C.G. CTNNB1 mutation associated with familial exudative vitreoretinopathy (FEVR) phenotype. Ophthalmic Genet. 2016, 37, 468–470. [Google Scholar] [CrossRef]
  38. Thevenon, J.; Duffourd, Y.; Masurel-Paulet, A.; Lefebvre, M.; Feillet, F.; El Chehadeh-Djebbar, S.; St-Onge, J.; Steinmetz, A.; Huet, F.; Chouchane, M.; et al. Diagnostic odyssey in severe neurodevelopmental disorders: Toward clinical whole-exome sequencing as a first-line diagnostic test. Clin. Genet. 2016, 89, 700–707. [Google Scholar] [CrossRef]
  39. Ke, Z.; Chen, Y. Case Report: A de novo CTNNB1 Nonsense Mutation Associated With Neurodevelopmental Disorder, Retinal Detachment, Polydactyly. Front. Pediatr 2020, 8, 575673. [Google Scholar] [CrossRef]
  40. Yoo, Y.; Jung, J.; Lee, Y.N.; Lee, Y.; Cho, H.; Na, E.; Hong, J.; Kim, E.; Lee, J.S.; Lee, J.S.; et al. GABBR2 mutations determine phenotype in rett syndrome and epileptic encephalopathy. Ann. Neurol. 2017, 82, 466–478. [Google Scholar] [CrossRef]
  41. Dubruc, E.; Putoux, A.; Labalme, A.; Rougeot, C.; Sanlaville, D.; Edery, P. A new intellectual disability syndrome caused by CTNNB1 haploinsufficiency. Am. J. Med. Genet. A 2014, 164A, 1571–1575. [Google Scholar] [CrossRef] [PubMed]
  42. Karolak, J.A.; Szafranski, P.; Kilner, D.; Patel, C.; Scurry, B.; Kinning, E.; Chandler, K.; Jhangiani, S.N.; Coban Akdemir, Z.H.; Lupski, J.R.; et al. Heterozygous CTNNB1 and TBX4 variants in a patient with abnormal lung growth, pulmonary hypertension, microcephaly, and spasticity. Clin. Genet. 2019, 96, 366–370. [Google Scholar] [CrossRef] [PubMed]
  43. O’Roak, B.J.; Vives, L.; Girirajan, S.; Karakoc, E.; Krumm, N.; Coe, B.P.; Levy, R.; Ko, A.; Lee, C.; Smith, J.D.; et al. Sporadic autism exomes reveal a highly interconnected protein network of de novo mutations. Nature 2012, 485, 246–250. [Google Scholar] [CrossRef] [Green Version]
  44. Krupp, D.R.; Barnard, R.A.; Duffourd, Y.; Evans, S.A.; Mulqueen, R.M.; Bernier, R.; Riviere, J.B.; Fombonne, E.; O’Roak, B.J. Exonic Mosaic Mutations Contribute Risk for Autism Spectrum Disorder. Am. J. Hum. Genet. 2017, 101, 369–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Torella, A.; Zanobio, M.; Zeuli, R.; Del Vecchio Blanco, F.; Savarese, M.; Giugliano, T.; Garofalo, A.; Piluso, G.; Politano, L.; Nigro, V. The position of nonsense mutations can predict the phenotype severity: A survey on the DMD gene. PLoS ONE 2020, 15, e0237803. [Google Scholar] [CrossRef]
  46. van der Luijt, R.B.; Meera Khan, P.; Vasen, H.F.; Breukel, C.; Tops, C.M.; Scott, R.J.; Fodde, R. Germline mutations in the 3′ part of APC exon 15 do not result in truncated proteins and are associated with attenuated adenomatous polyposis coli. Hum. Genet. 1996, 98, 727–734. [Google Scholar] [CrossRef]
  47. Gottardi, C.J.; Peifer, M. Terminal regions of beta-catenin come into view. Structure 2008, 16, 336–338. [Google Scholar] [CrossRef] [Green Version]
  48. van Veelen, W.; Le, N.H.; Helvensteijn, W.; Blonden, L.; Theeuwes, M.; Bakker, E.R.; Franken, P.F.; van Gurp, L.; Meijlink, F.; van der Valk, M.A.; et al. beta-catenin tyrosine 654 phosphorylation increases Wnt signalling and intestinal tumorigenesis. Gut 2011, 60, 1204–1212. [Google Scholar] [CrossRef] [Green Version]
  49. Claverie, J.M. Detecting frame shifts by amino acid sequence comparison. J. Mol. Biol. 1993, 234, 1140–1157. [Google Scholar] [CrossRef]
  50. Vonbrull, M.; Riegel, E.; Halter, C.; Aigner, M.; Bock, H.; Werner, B.; Lindhorst, T.; Czerny, T. A Dominant Negative Antisense Approach Targeting beta-Catenin. Mol. Biotechnol. 2018, 60, 339–349. [Google Scholar] [CrossRef]
  51. Liu, C.; Li, Y.; Semenov, M.; Han, C.; Baeg, G.H.; Tan, Y.; Zhang, Z.; Lin, X.; He, X. Control of beta-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell 2002, 108, 837–847. [Google Scholar] [CrossRef] [Green Version]
  52. Yanagawa, S.; Matsuda, Y.; Lee, J.S.; Matsubayashi, H.; Sese, S.; Kadowaki, T.; Ishimoto, A. Casein kinase I phosphorylates the Armadillo protein and induces its degradation in Drosophila. EMBO J. 2002, 21, 1733–1742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Gao, C.; Wang, Y.; Broaddus, R.; Sun, L.; Xue, F.; Zhang, W. Exon 3 mutations of CTNNB1 drive tumorigenesis: A review. Oncotarget 2018, 9, 5492–5508. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flow diagram of the systematic review.
Figure 1. Flow diagram of the systematic review.
Ijms 23 12564 g001
Figure 2. A 2-D bar chart representing clinical manifestations categorized based on the primary and secondary criteria.
Figure 2. A 2-D bar chart representing clinical manifestations categorized based on the primary and secondary criteria.
Ijms 23 12564 g002
Figure 3. A column chart representing percentages of facial dysmorphism, eye abnormalities, speech development, and behavioral problems found in reported CTNNB1 patients.
Figure 3. A column chart representing percentages of facial dysmorphism, eye abnormalities, speech development, and behavioral problems found in reported CTNNB1 patients.
Ijms 23 12564 g003
Figure 4. Schematic representation of the β catenin coding region; exon structure in correspondence to encoded protein domains.
Figure 4. Schematic representation of the β catenin coding region; exon structure in correspondence to encoded protein domains.
Ijms 23 12564 g004
Figure 5. Three-dimensional model of human β-catenin protein generated by I-Tasser. Annotation was performed according to Huber et al., 1997. The model shows the N-terminus (red), armadillo repeat arms 1–12, and a helix and unstructured region of the C-terminal domain.
Figure 5. Three-dimensional model of human β-catenin protein generated by I-Tasser. Annotation was performed according to Huber et al., 1997. The model shows the N-terminus (red), armadillo repeat arms 1–12, and a helix and unstructured region of the C-terminal domain.
Ijms 23 12564 g005
Figure 6. Distribution of mutation causative of CTNNB1 Syndrome throughout the protein coding region in accordance with exon location and subsequent encoded protein domains. Most mutations reside within the armadillo repeat region of the protein. Asterisk indicates a nonsense mutation and number in parenthesis indicates the number of cases reported the mutation.
Figure 6. Distribution of mutation causative of CTNNB1 Syndrome throughout the protein coding region in accordance with exon location and subsequent encoded protein domains. Most mutations reside within the armadillo repeat region of the protein. Asterisk indicates a nonsense mutation and number in parenthesis indicates the number of cases reported the mutation.
Ijms 23 12564 g006
Figure 7. Distribution of CTNNB1 mutation types.
Figure 7. Distribution of CTNNB1 mutation types.
Ijms 23 12564 g007
Figure 8. Number of mutations detected in individual intron or exon regions or whole gene deletions of analyzed samples in this study (n = 84).
Figure 8. Number of mutations detected in individual intron or exon regions or whole gene deletions of analyzed samples in this study (n = 84).
Ijms 23 12564 g008
Table 1. Search strategy in PubMed.
Table 1. Search strategy in PubMed.
NumberSearch Strategy
#1CTNNB1 [Text Word]
#2CTNNB1 Protein, Xenopus [MeSH Terms]
#3#1 OR #2
#2de novo OR loss-of-function OR germline mutation OR novel mutation [Text Word]
#3de novo OR novel mutation [Text Word]
#4loss of function mutation [MeSH Terms]
#5germ-line mutation [MeSH Terms]
#6#3 OR #4 OR #5
#7#2 OR #6
#8#3 OR #7
Table 2. Dataset for assessing quality of genotype–phenotype correlational studies.
Table 2. Dataset for assessing quality of genotype–phenotype correlational studies.
CategoryPoints
Mutational analysisNone0
Prescreening methods1
Full sequencing2
Demographic dataNone0
Sex, age, (ethnicity)1
Clinical assessmentNo0
Neurological assessment (facial dysmorphism, achieving motor milestones)1
Neurological and cognitive assessment2
Cognitive assessmentNone0
Diagnostic test done (autism, IQ tests)1
Brain screening testsNone0
MR1
MR and EEG2
Table 3. Categorization of the severity for the genotype/phenotype analysis.
Table 3. Categorization of the severity for the genotype/phenotype analysis.
Variable/SeverityEye ContactSpeechCognition (ID)Motor Development
Normal+No delaysNormal IDNo delays
Mild+Delayed; speaking in full sentencesmild ID (55–70)sitting before 1 year, walking before 2 years
Mild-Moderate+Delayed; can speak in sentences, speaking can be unclearmild-moderate ID (40–70)Sitting and walking independently with difficulties (ataxic)
Moderate+Delayed, speaking in simple sentences, can be unclearmild-moderate ID (40–70)Could be sitting and walking but with difficulties
Moderate-Severe+Simple words/no words; uses sign languagemoderate-severe ID (20–55)Could be sitting and walking but with difficulties
Severe-No speechsevere ID (20–40) Could be sitting and walking but with difficulties
Notes. + present; - absent.
Table 4. Summarization of the clinical features categorized according to their prevalence (n= 84).
Table 4. Summarization of the clinical features categorized according to their prevalence (n= 84).
Clinical Featuren (%)Clinical Featuresn (%)
Primary Criteria (>50%) Secondary Criteria (20–49%)
Presence of microencephaly (valid cases: 57)42 (73.7)Walking inability (valid cases: 40)16 (40)
Eye abnormalities (valid cases: 57) 153 (93)Aggression20 (47.6)
  Strabismus 30 (52.6)Stereotypic movements10 (23.8)
  FEVR 13 (22.8)Autism9 (21.4)
  Hyperopia 8 (14)Sleep problems8 (19)
  Astigmatism 5 (8.8)ADHD3 (7.1)
  Esotropia3 (5.3)Temper tantrums3 (7.1)
  Myopia 3 (5.3)Schizophrenia2 (4.8)
Speech difficulties (valid cases: 46)41 (89.1)Abnormal MR (valid cases: 24) 24 (16.7)
  Non-verbal14 (30.4)IGR (valid cases: 37)8 (21.6)
  A few words19 (41.3)Additional criteria
  Short sentences8 (17.4)  Scoliosis (not systematically assessed)2
  Full sentences, but delayed3 (6.5)  Feeding problems (not systematically assessed)5
  No delays2 (4.4)
1 Patient can have several eye abnormalities at the same time; 2 dilated ventricles, underdevelopment of the corpus callosum and brainstem, delayed myelination.
Table 5. Categorized CTNNB1 cases according to the severity of the phenotype (n = 35).
Table 5. Categorized CTNNB1 cases according to the severity of the phenotype (n = 35).
Genetic MutationGenderAge (yrs)Facial DysmorphismEye ConditionsMicroencephalyAxial Hypotonia/SpasticityAchieving MilestonesBehavior and IQ
Exon no. and VariantAmino Acid ChangeMutation TypeSitting (mo)Crawling (mo)Walking IndependentlySpeaking
SEVERE PHENOTYPE
I5, c.734 + 1G > TSplice mutationSpliceF32 +Strabismus++/+2–5 years (40mo)NANoAbsent speechRitualistic behaviours with temper tantrums, autism, severe ID (18 months)
I5, c.734 + 1G > ASplice mutationSpliceF49 +FEVRNA+/+NANANAAbsent speechNA, IQ = 40
I5, c.734 + 1G > ASplice mutationSpliceF27+FEVRNANANANAWalking at 49 yrs (ataxic)Absent speechNA, IQ = 20
E6, c.755delTinsAACp.Leu252*NonsenseF15.3 +Strabismus, hyperopia-+/+NANA10 yrs2 wordsAuto-aggressive behavior, stereotypic movements, short eye contact; severe IQ
E6, c.799_809delGAAGGAGCTAAinsGAAp. Gly268TrpfsTer5FrameshiftF7+NA++/+18 NA3 (broad based gait)No speechAutism, ID
MODERATE-SEVERE PHENOTYPE
E4, c.423_424insGp. Tyr142Valfs*4FrameshiftF5.6+Strabismus++/-NA24not yetSevere, few words (30), sign languageRepetitive movements, ID
E8, c.1163T > Cp.Leu388ProMissenseF6.8+NA++/+13182.5First word at 2 ½, 20 words at 4 years but not intelligibleID
E9, c.1272_1275delp.Ser425Thrfs*11FrameshiftF29+NA++/+243 years8 (progressive spasticity now with support)Started speaking first words 9–10 years; now able to speak a few wordsAggression, auto-mutilation, and fecal smearing
E9, c.1272_1275delp.Ser425Thrfs*11FrameshiftF3.25+Strabismus-+/+NANAnot yetBabbles now, some words are understandableVery happy and friendly, low frustration tolerance
E9, c. 1344_1345 InsertionAp.Arg449GlnfsTer24FrameshiftM8 NAStrabismus+NA/+NANA8 First words at 3 years, at 8 years able to speak short sentences Aggression sometimes when frustrated
E9, c.1420C > Tp.Arg474*NonsenseF13NAStrabismus++/+N13 months42 monthsFirst words at 4.5 yearsADHD, aggressive, teeth grinding; mouths objects
E9, c.1420C > Tp.Arg474*NonsenseF5.25+Strabismus++/+1823not yetNo wordsStereotypic outbursts
E9, c.1543C > Tp.Arg515*NonsenseF51+Optic atrophy++/+NANANoNot able to speak, but uses sign languageNormal behavior, ID, cognitive abilities gradually deteriorated
E10, c.1603C > Tp.Arg535TerNonsenseM3.25+Strabismus++/+8NAUnable to walkLots of noises but no wordsNA
E10, c.1603C > Tp.Arg535TerNonsenseM14 +NA--/+15NAnot walkingModerate; Single words at 14 yearsAggressive outbursts, self-harm
E11, c.1801C > Tp.Gln601TerNonsenseM6.2+FEVR++/NANANAnot yetSays Mom and Dad with understanding, uses Makaton, points to body parts Occasional temper; can bite others and self; repetitive movements
E12, c.1923dupAp.Glu642Argfs*6FrameshiftM8.5 +Strabismus, hyperopia++/+NA14 8 yearsSevere; few single words, gesturesGood social interaction, outburst of temper tantrums or crying, self/biting
E12, c.1925_1926delp.Glu642Valfs*5FrameshiftF14.2 +Strabismus--/+not yetNANoModerate, first words at 6 years; not speaking in sentencesRages and tantrums, friendly personality, short attention span and poor eye contact; autism
MODERATE PHENOTYPE
E3, c.99_100delTGp.Gly34Asnfs*15FrameshiftM5.5+Strabismus++/+14; still head-leg256 yrs (cannot stand alone)Short sentencesSocial and friendly boy; no behavioral problems; concentration is limited; sensitive to noises
E3, c.232C > Tp.Gln78*NonsenseM11+Strabismus, hyperopia, astigmatism-+/+NADidn’t crawl3 yrs; at 11 yrs coordination problemUnclear speech; at 11 yrs regressionTemper tantrums, aggression, frustration, anxiety, friendly personality, stereotypic movements
E4, c.283C > Tp.Arg95*NonsenseF4+Normal++/+NA124 yrsSpeech apraxia, ~50 wordsWhen young, biting, banging the head in the wall, this has improved now
E5, c.705dupAp.Gly236Argfs*35FrameshiftF14 +Strabismus++/-12NA4.5 yrsBabbling at 3 yrs, 14 yrs speaking simple sentences, read simple wordsAutism, IQ = 65
E7, c.925C > Tp.Gln309*NonsenseM4.5 +Hyperopia++/+18 NAwalking at 4.6 (short distances)Started speaking at 4 years, articulation was poor and hard to understandHappy personality
E7, c.998dupAp.Tyr333TerNonsenseF9 NANA-+/+14NA4.2 (still had difficulties)First words age 4; more fluent speech age 6; said to be 3 years behind with verbal skillsViolent outbursts associated with difficulty expressing emotions
E7, c.999C > Gp.Tyr333TerNonsenseF27 +NA-+/+30 NA4.5 (ataxic)First words at 4.5 years; can speak in sentences but speech very unclearAggressive, temper tantrums, self-injurious (biting, picking)
E7, c.1038_1044delGCTATCTinsGCTp.Val349AlafsTer9FrameshiftF11 +Strabismus, hypermetropia++/+NA13.53.5 (ataxic)Single words at age 5 years, talks in sentences at age 11 yearsStereotypies (clapping repeatedly, temper tantrums, aggressive to family)
I7, c.1081 + 1G > CIVS6 Intron 7SpliceM3.8 +Normal++/+not yetnot yetnot yetUnclear speechGood social interaction, very happy personality
E10, c.1612C > Tp.Gln538TerNonsenseF4.5+Strabismus++/+23NA2.5–3 yearsFirst words at 3.4 yearsAutism
333 kb incl. entire gene and ex. 35–37 of ULK4Gross delNoneF5.2+Hyperopia++/-14NA4.5 years (ataxic)At 4.5 years could combine several words, count to 10Friendly, social, short focus
505 kb incl. entire geneGross delNoneM3 +Esotropia++/-NAnot yetnot yetBabbles and say “mama” and “dada”, before age 3 yearsHappy, good eye contact
MILD PHENOTYPE
E13, c.1981C > Tp.Arg661TerNonsenseF9.2+NA++/-11 NA2.5First words at 3,4 yearsObsessional behavior; dyspraxia
E13, c.2038_2041dupp.Ser681*NonsenseF13.2+Strabismus, myopia-+/+12NA1.5Mild, full sentences, but delayedSocial, autism, aggressive behavior, ADHD
E13, c.2038_2041dupp.Ser681*NonsenseF11 +Strabismus, myopia++/+12 17 2Mild, full sentences, but delayedCommunicative, social, aggression, ADHD
NORMAL PHENOTYPE
E14, c.2128C > Tp.Arg710CysMissenseMNAnormalFEVRNANormalNormal normalnormalnormalNormal
E15, c.2142_2157dup16p.His720*NonsenseMNAnormalFEVRNANormalNormal normalnormalnormalNormal
Table 6. Phenotype–genotype analysis (n = 35).
Table 6. Phenotype–genotype analysis (n = 35).
LocusMutation TypeNumberClinical OutcomeRemarkable PhenotypesReferences
Intron 5Splice3SevereFacial dysmorphisms, small head/microcephaly, axial hypotonia, peripheral spasticity, optical alterations, absent speech, severe ID, no eye contact, behavioral difficultiesVerhoeven et al. 2020 [1], Wang et al. 2019 [29]
Exon 6Nonsense, frameshift2SevereFacial dysmorphisms, small head/microcephaly, axial hypotonia, peripheral spasticity, severe speech impairment (no speech/2 words), severe ID, behavioral difficultiesKuechler et al. 2015 [2], Kharbanda et al. 2017 [30]
Exon 4Frameshift 1Moderate-SevereFacial dysmorphisms, microcephaly, axial hypotonia, peripheral spasticity, no walking, no speech (few words), autistic behaviorKuechler et al. 2015 [2]
Exon 8Missense1Moderate-SevereFacial dysmorphisms, epilepsy microcephaly, axial hypotonia, peripheral spasticity, delayed walking, impaired speech (words not intelligible), IDKuechler et al. 2015 [2]
Exon 9Nonsense3Moderate-SevereFacial dysmorphisms, small head/microcephaly, optical alterations, axial hypotonia, peripheral spasticity, severe speech impairments (no speech/single words), severe ID and can have behavioral alterationsKuechler et al. 2015 [2], Ligt et al. 2012 [4], Tucci et al. 2014 [28], Kharbanda et al. 2017 [30]
Exon 9Frameshift3Moderate-SevereFacial dysmorphisms, small head/microcephaly, optical alterations, axial hypotonia, peripheral spasticity, moderate speech (can have understandable words, can repeat short sentences), absent walking and speech, and can have behavioral difficultiesKuechler et al. 2015 [2], Ligt et al. 2012 [4], Tucci et al. 2014 [28], Jin et al. 2020 [5], Kharbanda et al. 2017 [30]
Exon 10 Nonsense2Moderate-SevereFacial dysmorphisms, small head/microcephaly, impaired speech (noises, few words), moderate/severe ID, behavioral difficultiesKharbanda et al. 2017 [30]
Exon 11Nonsense1Moderate-SevereFacial dysmorphisms, optical alterations, axial hypotonia, unable to walk, impaired speech (few words), severe ID, behavioral difficultiesKharbanda et al. 2017 [30]
Exon 12Frameshift2Moderate-SevereFacial dysmorphisms, optical alterations, peripheral spasticity, impaired speech (few words), IDKuechler et al. 2015 [2]
Exon 3Nonsense, frameshift2ModerateFacial dysmorphisms, optical alterations, axial hypotonia, peripheral spasticity, impaired speech (short sentences, unclear speech), IDWinczewska-Wiktor et al. 2016 [23], Kuechler et al. 2015 [2]
Exon 4Nonsense1ModerateFacial dysmorphisms, microcephaly, axial hypotonia, peripheral spasticity, delayed walking, impaired speech (at 4 and 5 years < 50 words), ID, autistic behaviorKuechler et al. 2015 [2]
Exon 5Frameshift1ModerateFacial dysmorphisms, strabismus, microcephaly, axial hypotonia, peripheral spasticity, delayed walking, impaired speech (simple sentences, read simple words), mild ID, and autistic behavior Tucci et al. 2014 [28]
Exon 7Nonsense, frameshift4ModerateFacial dysmorphisms, axial hypotonia, peripheral spasticity, delayed walking (average 4,2 years), impaired speech (started speaking in 4 years, speaks in sentences, articulation poor), and can have behavioral difficultiesLigt et al. 2012 [4], Tucci et al. 2014 [28], Kharbanda et al. 2017 [30]
Intron 7Splice1ModerateFacial dysmorphisms, microcephaly, axial hypotonia, peripheral spasticity, absent walking, impaired speech, IDKuechler et al. 2015 [2]
Exon 10Nonsense1ModerateFacial dysmorphisms, microcephaly, axial hypotonia, peripheral spasticity, impaired speech, autismKharbanda et al. 2017 [30]
Entire geneGross deletion2ModerateFacial dysmorphisms, optical alterations, microcephaly, axial hypotonia, impaired walking, impaired speech, IDDubruc et al. 2014 [41], Kuechler et al. 2015 [2]
Exon 13Nonsense 3MildFacial dysmorphisms, axial hypotonia, delayed walking (before 24 months), mild ID, behavioral alteration (autism and aggression)Kuechler et al. 2015 [2], Kharbanda et al. 2017 [30]
Exon 14 and 15Missense and Nonsense2NormalNormal phenotype with only optical alterations (FEVR)Panagiotou et al. 2017 [35]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Miroševič, Š.; Khandelwal, S.; Sušjan, P.; Žakelj, N.; Gosar, D.; Forstnerič, V.; Lainšček, D.; Jerala, R.; Osredkar, D. Correlation between Phenotype and Genotype in CTNNB1 Syndrome: A Systematic Review of the Literature. Int. J. Mol. Sci. 2022, 23, 12564. https://doi.org/10.3390/ijms232012564

AMA Style

Miroševič Š, Khandelwal S, Sušjan P, Žakelj N, Gosar D, Forstnerič V, Lainšček D, Jerala R, Osredkar D. Correlation between Phenotype and Genotype in CTNNB1 Syndrome: A Systematic Review of the Literature. International Journal of Molecular Sciences. 2022; 23(20):12564. https://doi.org/10.3390/ijms232012564

Chicago/Turabian Style

Miroševič, Špela, Shivang Khandelwal, Petra Sušjan, Nina Žakelj, David Gosar, Vida Forstnerič, Duško Lainšček, Roman Jerala, and Damjan Osredkar. 2022. "Correlation between Phenotype and Genotype in CTNNB1 Syndrome: A Systematic Review of the Literature" International Journal of Molecular Sciences 23, no. 20: 12564. https://doi.org/10.3390/ijms232012564

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop