Next Article in Journal
High Throughput SARS-CoV-2 Genome Sequencing from 384 Respiratory Samples Using the Illumina COVIDSeq Protocol
Previous Article in Journal / Special Issue
Language Profiles of School-Aged Children with 22q11.2 Copy Number Variants
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Coexisting Conditions Modifying Phenotypes of Patients with 22q11.2 Deletion Syndrome

by
Marta Smyk
1,
Maciej Geremek
1,
Kamila Ziemkiewicz
1,
Tomasz Gambin
1,2,
Anna Kutkowska-Kaźmierczak
1,
Katarzyna Kowalczyk
1,
Izabela Plaskota
1,
Barbara Wiśniowiecka-Kowalnik
1,
Magdalena Bartnik-Głaska
1,
Magdalena Niemiec
1,
Dominika Grad
1,
Małgorzata Piotrowicz
3,
Dorota Gieruszczak-Białek
4,
Aleksandra Pietrzyk
5,
T. Blaine Crowley
6,
Victoria Giunta
6,
Daniel E. McGinn
6,7,
Elaine H. Zackai
6,7,
Oanh Tran
6,
Beverly S. Emanuel
6,7,
Donna M. McDonald-McGinn
6,7 and
Beata A. Nowakowska
1,*
add Show full author list remove Hide full author list
1
Department of Medical Genetics, Institute of Mother and Child, 01-211 Warsaw, Poland
2
Institute of Computer Science, Warsaw University of Technology, 75, 00-662 Warsaw, Poland
3
Department of Genetics, Polish Mother’s Memorial Hospital Research Institute, 70-445 Łódź, Poland
4
Department of Medical Genetics, Children’s Memorial Health Institute, 04730 Warsaw, Poland
5
Department of Genetics and Pathology, Pomeranian Medical University, 70-204 Szczecin, Poland
6
Division of Human Genetics and 22q and You Center, the Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
7
Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA 19104, USA
*
Author to whom correspondence should be addressed.
Submission received: 19 November 2022 / Revised: 15 February 2023 / Accepted: 22 February 2023 / Published: 9 March 2023
(This article belongs to the Special Issue 22q11.2 Deletion Syndrome)

Abstract

:
22q11.2 deletion syndrome (22q11.2DS) is the most common genomic disorder with an extremely broad phenotypic spectrum. The aim of our study was to investigate how often the additional variants in the genome can affect clinical variation among patients with the recurrent deletion. To examine the presence of additional variants affecting the phenotype, we performed microarray in 82 prenatal and 77 postnatal cases and performed exome sequencing in 86 postnatal patients with 22q11.2DS. Within those 159 patients where array was performed, 5 pathogenic and 5 likely pathogenic CNVs were identified outside of the 22q11.2 region. This indicates that in 6.3% cases, additional CNVs most likely contribute to the clinical presentation. Additionally, exome sequencing in 86 patients revealed 3 pathogenic (3.49%) and 5 likely pathogenic (5.81%) SNVs and small CNV. These results show that the extension of diagnostics with genome-wide methods can reveal other clinically relevant changes in patients with 22q11 deletion syndrome.

Graphical Abstract

1. Introduction

22q11.2DS is the most common chromosomal microdeletion syndrome in humans, with a prevalence of 1:2148 live births [1,2]. The morbidity in utero is higher with the frequency of 1:1000 fetuses [3,4] and 1:1500 miscarriage samples [5]. 22q11 deletion is a result of non-allelic homologous recombination between high identity low copy repeats (LCRs). Most of the affected individuals (~90%) carry a typical hemizygous 3 Mb deletion on chromosome 22q11.2, mediated by LCRs 22-A and 22-D, but smaller deletions mediated by other combinations of LCRs are also described [6]. This leads to a diminished dosage of nearly 60 genes, including coding and noncoding RNAs.
The characteristics of this contiguous gene deletion syndrome is highly variable phenotypic severity with many common significant physical and behavioral clinical findings described, as well as numerous rare significant features impacting clinical care, but none appear to be fully penetrant and each exhibits variable expressivity [2,7,8]. Variability is observed across unrelated patients and between affected family members who have inherited identical deletions, including amongst identical twins [9,10,11,12]. Approximately 10% of chromosome 22q11.2 deletions are parentally transmitted, and in those cases, high penetrance and wide expressivity are observed [11,13,14]. Frequently associated features of 22q11.2DS are congenital heart defects, palatal differences, immunodeficiency, endocrine abnormalities, developmental delay, cognitive deficits, and behavioral phenotypes [7,8,14,15]. Less commonly observed are structural ophthalmologic abnormalities, choanal atresia, hearing loss, laryngo-tracheo-esophageal abnormalities, gastrointestinal differences, congenital diaphragmatic hernia, vertebral anomalies, polydactyly, IUGR, idiopathic seizures, microcephaly, neural tube defects, ADHD, and autism [14]. Patients with 22q11.2DS have been reported to have a diminished adult life expectancy, with an increased risk of sudden death and developing psychiatric disorders [16,17].
It remains unknown why individuals with deletion of the same size present such a wide range of phenotypes. Some of the genes located within the standard deletion have major clinical impact, particularly T-box 1 (TBX1). The TBX1 gene is part of the larger family of T-box genes, which help to regulate tissue and organ formation during development. However, a different minority of patients harbor nested distal deletions but retain two copies of the TBX1 gene [2]. Therefore, the single dose of deleted genes alone cannot explain the tremendous variation in the severity and penetrance of the associated clinical features among affected individuals. Differences in deletion extents also cannot account for the huge variability in phenotypic severity among patients with 22q11.2DS [18]. Currently, there is no association between deletion size and the presence of the most common clinical symptoms, congenital heart disease or palate anomalies [19], although the common clinical features are less penetrant in smaller atypical nested deletions [14]. This points out that genetic variants, beyond the deleted genes themselves, may play a role in determining the 22q11.2DS clinical complications. Therefore, sequencing and microarray methods can help uncover, at least partially, genetic differences among patients with 22q11.2DS that can influence disease severity.
Several potential genetic mechanisms can underlie these highly variable phenotypic manifestations, including that they may be attributed to the hemizygous variants on the remaining allele, due to additional rare pathogenic variants elsewhere in the genome, as well as nongenetic environmental factors. Unmasking autosomal recessive disorders by having a deletion in one 22q11.2 allele and a mutation on the other non-deleted allele has been described in GP1BB (Bernard–Soulier syndrome type B) [20], SNAP29 (CEDNIK syndrome—Cerebral dysgenesis, neuropathy, ichthyosis, and keratoderma) [21], LZTR1 (Noonan syndrome) [22,23], SCARF2 (van den Ende-Gupta syndrome (VDEGS) [24], and CDC45 (CGS syndrome—craniosynostosis, gastrointestinal differences, short stature, and skeletal differences/Meier–Gorlin syndrome/Baller–Gerold syndrome) genes [25,26]. Mapping the variation in the remaining alleles in patients with 22q11.2DS also identified potential variants in PI4KA and PRODH [27]. Recently, known pathogenic genetic factors beyond the 22q11.2 region, including mutations and chromosomal structural aberrations, were described in ~0.9% of patients with 22q11.2DS with atypical phenotypic features [28].
Our goal was to determine the presence of additional variants affecting the phenotype of patients with 22q11.2DS.

2. Materials and Methods

2.1. Patients and Deep Phenotyping

225 patients were enrolled in the study after receiving genetic counselling and signing informed consent. The study group included 82 prenatal cases and 143 postnatal patients. During routine aCGH diagnostics at the Institute of Mother and Child, we identified 159 patients with 22q11.2 deletion (82 prenatal and 77 postnatal). For exome sequencing, 86 postnatal patients (40 females and 46 males) were recruited/evaluated at the Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland (76 probands), and within the 22q and You Center, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA (10 probands). 20 patients were analyzed by both methods: array CGH and exome sequencing. In the remaining 66 patients, the presence of a chromosome 22q11.2 deletion had been identified by FISH or MLPA.
To obtain the most detailed clinical description of the patients in each case, medical consultation was carried out by experienced clinical geneticists. A standard form (Supplementary Table S1) for deep phenotyping was prepared at the Institute of Mother and Child on the basis of review of the literature and included clinical features that may occur in individuals with 22q11.2DS. The clinical symptoms were grouped by category: cardiovascular system, immunodeficiency, autoimmune disease, palatal abnormalities/velopharyngeal insufficiency/dysfunction, laryngo-tracheo-esophageal anomalies, endocrinologic problems including hypocalcemia, short stature, intrauterine growth deficiency/failure to thrive (FTT), macrocephaly, microcephaly, structural eye defects and ocular disorders, skeletal abnormalities, dental problems, structural ear defects and hearing problems, structural CNS anomalies, unprovoked seizures, genitourinary tract anomalies, inguinal hernia, gastrointestinal anomalies and functional problems, umbilical hernia, craniofacial dysmorphic features, psychomotor and intellectual development, learning disabilities/cognitive deficits, behavioural abnormalities, and psychiatric illness. A detailed form with over 230 clinical features was completed where possible (Supplementary Table S2). The research was approved by the Bioethics Committee of the Institute of Mother and Child in Warsaw and the Institutional Review Board of the Children’s Hospital of Philadelphia.

2.2. aCGH

We performed array comparative genomic hybridization (arrayCGH), with commercially available arrays such as the 60K CytoSure Constitutional v3 microarray (Oxford Gene Technology, Oxford, UK) according to manufacturer’s instructions. CNVs that showed partial or complete overlap with known segmental duplications were excluded from further analysis, due to the high variability of copy number variations in those regions. CNVs were classified as pathogenic, likely pathogenic, likely benign, benign and of uncertain significance (VUS) based on clinical data in known CNV databases: Database of Genomic Variants (http://dgv.tcag.ca/dgv/app/home accessed on 18 November 2022), ClinVar (https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/clinvar/ accessed on 18 November 2022), DECIPHER (https://www.deciphergenomics.org/ accessed on 18 November 2022), ClinGen (https://www.clinicalgenome.org/ accessed on 18 November 2022). Recurrent CNVs or CNVs associated with known microdeletion or microduplication syndromes were classified as pathogenic or likely pathogenic depending on the penetrance and clinical features present in probands. In 159 patients (82 prenatal and 77 postnatal), array CGH was performed as a first diagnostic test. Additionally, we conducted aCGH where exome sequencing revealed CNV, which could be confirmed (due to size and array coverage).

2.3. Exome Sequencing

Exome sequencing (ES) was applied to identify rare variants that could affect the phenotype in 86 patients with 22q11.2DS. Detailed phenotyping of all patients was performed (Supplementary Table S2) to estimate the correlation between ES results and specific phenotypes.
Sequencing was performed using Illumina HiSeq (San Diego, CA, USA) instruments after exome capture with the Sure Select All Human V6 design. Raw sequence data were post-processed using the bc-bio pipeline (https://github.com/chapmanb/bcbio-nextgen accessed on 18 November 2022). The bc-bio pipeline performs mapping of the short reads against a human genome reference sequence (GRCh38) using the Burrows-Wheeler Alignment (BWA), BAM post-processing using GATK, and variant calling using the GATKHaplotype Caller. Finally, ANNOVAR [29] was used to annotate relevant information about gene names, predicted variant pathogenicity, reference allele frequencies and metadata from external resources, and then to add these to the Variant Call Format (VCF) file.
Next, we used HMZDelFinder algorithm [30] to search for small, hemizygous deletions within the 22q11.2 region and small, heterozygous deletions beyond 22q11.2 region. As a control data set, we used WES data from 164 samples sequenced at the same platform and processed using the same pipeline, as our patients with 22q11.2DS. To prepare input data for HMZDelFinder algorithm, for each sample for every exonic target from the capture design we computed RPKM values, i.e., number of reads per kilobase of transcript per million mapped reads. To detect heterozyous CNVs outside of the 22q11.2 region we used CoNIFER tool with its default parameters [30]. As an input we reused the same rpkm data previously precomputed to run HMZDelFinder. Each putative CNV identified by CoNIFER has been visualized and undergone manual inspection to remove common polymorphisms and likely false positive calls.

3. Results

3.1. aCGH Analysis

In 21 patients, in addition to the chromosome 22q11.2 loss, we identified 31 copy number variants. Among those 31 CNVs, 5 were pathogenic, 5 were likely pathogenic, and 21 CNVs were classified as variants of uncertain significance (Table 1). In 8 patients, more than one additional CNV was observed. Ten out of thirty-one additional CNVs also occurred on chromosome 22q, but in different regions, however in 7 of those cases the additional deleted regions most likely were the extension of the typical or nested 22q11. Importantly, these variants would not be possible to identify using targeted methods, like FISH or MLPA.

3.2. Exome Sequencing Analysis

3.2.1. Variants in Genes from Remaining 22q11.2 Region

Hemizygotic variants in the 22q11.2 deletion region were analyzed with a frequency of 0.01 in gnomAD database. Next, only exonic, stopgain, nonsynonymous, and frameshift variants were taken into analysis. Across all 86 patients, a total of 29 variants in 21 patients were identified in the remaining 22q11.2 region (Supplementary Table S3). None of the variants were previously described as pathogenic or likely pathogenic, therefore all variants were classified as variants of uncertain clinical significance. However, investigation of the remaining allele in our tested cohort revealed a rare nonsynonymous-damaging variant in CDC45 in one patient out of 8 with craniosynostosis. In addition to bilateral coronal craniosynostosis, this patient (GC028958) also presented with additional clinical features overlapping with the previously described MGORS7/CGS syndrome, such as an anteriorly placed anus.
HMZDelFinder identified two putative hemizygous deletions in the remaining 22q11.2 region. The first deletion encompasses two genes; DGCR6, PRODH (Chr22:18906447-18931248). This deletion is classified in ClinVar as likely benign and had no effect on the patient’s phenotype, because the region is commonly deleted in control populations. However, this deletion was never described in patients with 22q11.2DS. Therefore, we cannot exclude the possibility that nullisomy of PRODH has an impact on the patient’s long-term phenotype. This deletion was classified as uncertain clinical significance. The second deletion (hemizygous), also identified in the 22q11.2 region, encompasses 8 exons of TANGO2 (chr22:20043265-20064650). Bi-allelic truncating mutations in this gene have been associated with TANGO2-related metabolic encephalopathy and arrhythmias presenting with recurrent muscle weakness with rhabdomyolysis, metabolic crises, and cardiac arrhythmia [31]. Therefore, this variation was confirmed by a qPCR method, classified as pathogenic, and was consistent with the patient’s phenotype (Figure 1).

3.2.2. Filtering for SNVs Elsewhere in the Genome

Filtering for known pathogenic variants, defined as a frequency less than or equal to 0.001 in gnomAD genome database, autosomal dominant inheritance, and classified as pathogenic in ClinVar database, revealed 1 rare stopgain, 1 rare frameshift deletion, 1 rare UTR3 variant, and 4 rare non-synonymous variants. In total, 6 genes across 86 patients carry a rare protein-altering variant. The clinical outcome of these variants was discussed with clinicians for all individuals (Table 2). For two cases, the clinical features were consistent with the predicted consequences of genomic variant and were classified as pathogenic. In five cases, the patients did not present expected phenotype features, and therefore the variants have been classified as potentially pathogenic.
Rare SNVs were selected using filtering with an allele frequency less than 0.001 in gnomAD and were then filtered against an in-house control dataset. Variants in genes associated with OMIM diseases, of autosomal and X-linked dominant inheritance or X-linked recessive inheritance in male patients, were chosen. Effects of missense variants were predicted using four in silico analysis tools (SIFT; Polyphen2_HDIV; Polyphen2_HVAR; MutationTaster). SNVs with deleterious predictions in at least 2 out of 4 prediction tools or SNV automatically deleterious in Mutation Taster were selected. Next, phenotypic features of patients with those SNVs were compared with clinical features related to the implicated genes. Only those variants, where the clinical features overlapped with predicted phenotypes, were included in the table (Table 3). The pathogenicity of all selected SNVs was estimated using Alamut-2.11-0 software.
HMZDelFinder revealed three heterozygous deletions in two genes: GABRA5 and KARS (Table 4).
Additionally known cancer susceptibility SNVs in genes associated with cancer risk have been identified in 5 patients (Supplementary Table S3). The variants have been revealed in 3 genes: ATM, BRCA1, and BRCA2. All patients were informed regarding these risk genes and were referred to the oncology clinic.
Overall, 885 variants of uncertain clinical significance were selected based on the frequency (less than 0.0001 in gnomAD database), function (exonic), and type of change (frameshift deletion, frameshift insertion, stopgain, or nonsynonymous (Supplementary Table S4).
In summary, we have identified 8 pathogenic variants, and 10 likely pathogenic variants in 225 patients, which is much more than previously expected (Table 5).

4. Discussion

In our study, we have analyzed genomes of 225 patients with 22q11.2 deletion syndrome, using array CGH and/or exome sequencing methodology. The aim of our research was to identify variants beyond the 22q11.2 deletion, which may contribute to the clinical complexity in our patients.
Hemizygous variants are one of the potential genetic mechanisms that could underlie this clinical variability. Allelic variations of genes within a critical region of the non-deleted chromosome can have an impact on phenotypes by unmasking recessive diseases, which has already been proven for several genes within the 22q11.2 region, like SNAP29 and GP1BB [21,32]. Recent studies have mainly focused on mapping mutations on the remaining allele [27,33]. This strategy has demonstrated high efficiency in cases with atypical and less common clinical features. For example, biallelic mutations in CDC45 are thought to be the cause of craniosynostosis in Meier–Gorlin syndrome (MGORS7, OMIM 617063) [25], a rare autosomal recessive primordial dwarfism disorder, characterized by microtia, short stature, and absent or hypoplastic patellae. Recently, in patients with 22q11.2DS, the alteration of CDC45 was associated with the pathogenesis of craniosynostosis, as well as phenotypic features more frequently reported in association with Baller–Gerold (OMIM 218600) and RAPADILINO syndrome (OMIM 266280), as well as unique previously-unreported features [26]. In fact, rare hemizygous variants in the CDC45 gene were found in 5/15 (33%) of the patients reported by Unolt et al., with both a 22q11.2 deletion and atypical clinical features in 3/7 of the patients (43%) including craniosynostosis (2 bicoronal, 1 metopic) and none of the 133 22q11.2DS patients from the control group (Fisher’s test p value < 0.01). These patients also had important structural anomalies not typically associated with 22q11.2DS, most significantly some combination of anal/intestinal anomalies, limb differences, short stature, and craniofacial anomalies. Investigation of the remaining allele in our tested cohort revealed a rare nonsynonymous damaging variant in CDC45 in one patient out of eight with craniosynostosis. In addition to bilateral coronal craniosynostosis, this patient also presented other clinical features overlapping with MGORS7/CGS, such as an anteriorly placed anus.
In two patients, we found additional deletions on the remaining allele. In one patient, an intragenic deletion of 8 exons in TANGO2 was identified following a postmortem study. Mutations in this gene are responsible for TANGO2-related metabolic encephalopathy and arrhythmias, an autosomal recessive recurrent metabolic encephalomyopathic crises, rhabdomyolysis, cardiac arrhythmia, and intellectual disability syndrome (OMIM 616878). A large proportion of patients described in the literature have succumbed in childhood due to cardiac arrest related to arrhythmias or following seizures related to a hypoglycemic crisis. Some patients, with variants in the TANGO2 gene, presented primarily with neurologic features [34]. In our second patient, we identified a deletion encompassing the whole PRODH gene. Mutations or small deletions in PRODH cause recessive hyperprolinemia (OMIM 239500) with neurologic manifestations, including seizures. It is well known that individuals with 22q11.2DS are at high risk for developing schizophrenia and schizoaffective disorder. PRODH was suggested to play a role in the later stages of neural development [35]. Although the latest research does not support the theory that high proline level is responsible for psychosis, it cannot be excluded that nullisomy of the PRODH gene has no clinical effect. Despite the association between variants in PRODH and an increased risk of schizophrenia shown in some studies, the potential role of this gene in psychiatric diseases remains unclear [36,37].
Hemizygous variants on a second allele may explain only part of the clinical variability associated with 22q11.2DS. The phenotypic variability may also be associated with additional copy number variations, as was shown in our study.
Analysis of other microdeletions with variable expressivity suggests that a two-hit model may be more generally applicable to neuropsychiatric disease [38]. Additional CNVs were already suggested to be genomic modifiers in patients with a 22q11.2 deletion or duplication [39,40]. In our study we have identified 31 additional CNVs in 21 patients, among 159 tested by array CGH. It is worth noting that more than one CNV was observed in 8 patients in our cohort, worsening the overall clinical picture. In 10 cases the additional CNV was located on chromosome 22q, however in 7 of those cases the additional deleted regions most likely were the extension of the typical or nested 22q11 deletions. It is particularly interesting that additional CNVs occurred in 7 patients with nested 22q11 deletions and 14 patients with A-D typical deletion. Taking into account the ratio of nested to the typical deletions (28:131 in our study), the result can suggest that if the smaller deletions have a milder phenotype than the classical ones, they may more likely have additional CNVs affecting phenotype. However, to draw final conclusions, more cases with different sizes of the deletion need to be studied. Moreover, in 3 cases, recurrent deletions with variable expressivity on chromosome 15 and 16 were revealed. Recurrent proximal 16p11.2 microdeletion found in case GC034796 can also influence some phenotypic features like global developmental delay, hypotonia, or cardiac anomalies. Rare CNVs outside of the 22q11.2 region may modify risk for congenital cardiac defects in some patients, with 22q11.2DS probably being due to the content of genes influencing heart development [41,42]. In patient GC034823, who demonstrated cognitive regression and dementia, both of which are unusual for 22q11.2DS, the deletion region is expanded and encompasses the TUBA8 gene. Mutations in TUBA8 were associated with recessive polymicrogyria [43]. Recently, Dantas et al. reported downregulation of this gene in patients with 22q11.2 deletion syndrome [44]. Another CNV encompassing gene associated with an intellectual disability is partial duplication of the MID2 gene, which was revealed in male patient GC034808. Previously, a duplication overlapping MID2 was reported in a boy with FG syndrome, an X-linked multiple congenital anomalies syndrome (OMIM 300581), who had hypotonia and developmental delay [45]. A missense mutation in MID2 was described in a large Indian family with global developmental delay and minor facial features [46]. Recently, an Xq22 duplication including MID2 was described in a patient whose phenotypic features were not consistent with FG syndrome [47]. Another mechanism modifying patients’ clinical features is the influence of single nucleotide variations (SNVs) in genes that reside outside of the deleted region, e.g., SNVs in genes that lie on the same pathway with genes from the deleted region can modify the severity of symptoms. A good example of such a mechanism demonstrates patient GC034900, with a variant in the TLL1 gene. Heterozygous mutations in the TLL1 gene are responsible for atrial septal defect (ASD6). In about 1% of patients with 22q11.2DS. another known pathogenic genetic factor, such as mutations, chromosomal structural aberrations. or mosaic aneuploidy, was observed, resulting in dual diagnoses [28]. An example of a dual diagnosis in our cohort is patient GC030951, with both 22q11.2DS and a pathogenic variant in the NF1 gene resulting in Neurofibromatosis as well as 22q11.2DS.
Historically, medical evaluation of genetic conditions relies upon pattern recognition, which recognizes a phenotype, signs, or symptoms to guide targeted genetic testing. The clinical features of many genetic conditions and 22q11.2 deletion syndrome may overlap. Therefore, most often, when a 22q11.2 deletion is identified, further diagnosis is frequently not considered in most patients, and additional or rare clinical features are attributed to the presence of the deletion.
22q11.2DS is a multi-system disorder caused by a single deletion that is genetically complex and can be seen as a model for gene–gene interactions and phenotype–genotype correlations. Recent advances in genetics and systems biology provide excellent opportunities to gain novel insights into pathways underlying the morbidities associated with 22q11.2DS across the lifespan.
Our study demonstrates that secondary diagnoses should be taken into account in all patients with 22q11.2DS. Only a whole-exome/whole-genome study of large cohorts of patients with 22q11.2DS will provide a robust opportunity to determine genotype–phenotype correlations for individual genes with the 22q11.2 deletion and beyond. Knowing how often additional variants can modify the patient’s phenotype, it is recommended to use the aCGH/SNP microarray methodology in cases of suspected 22q11.2 deletion at minimum.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/genes14030680/s1, Supplementary Table S1: On the basis of physical examination for every patient, a detailed form with over 230 clinical features was completed. Supplementary Table S2: Summary of phenotypic characteristics of 86 patients with 22q11DS recruited for exome sequencing. Supplementary Table S3: Hemizygotic variants in the 22q11.2 deletion region. Supplementary Table S4: ClinVar pathogenic cancer predisposition. Supplementary Table S5: Variants of uncertain clinical significance. 885 variants of uncertain clinical significance were selected based on the frequency (less than 0.0001 in gnomAD database), function (exonic) and type of change (frameshift deletion, frameshift insertion, stopgain, or nonsynonymous.

Author Contributions

Conceptualization: B.A.N., D.M.M.-M., M.G. and M.S.; Data curation: T.B.C., V.G., D.E.M., O.T. and B.S.E.; exome data analysis: M.G., T.G., M.S. and B.A.N.; array data analysis: M.S., K.K., I.P., B.W.-K., M.B.-G., M.N. and D.G.; Data collection/investigation: M.G., A.K.-K., M.P., D.G.-B., A.P., T.B.C., V.G., D.E.M., E.H.Z., O.T. and B.S.E. Methodology: T.G. and M.G. Writing, revising and reviewing: B.A.N., D.M.M.-M., M.S. and K.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported with a grant from the National Science Centre (OPUS NCN 2015/17/B/NZ5/01357 to BN).

Institutional Review Board Statement

The study was conducted in accordance with the approved by the Ethics Committee of Institute of Mother and Child (protocol code; 39/2021, date of approval; 22 April 2021).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Any data requests can be directed to the corresponding author.

Acknowledgments

The authors thank the patients and their families for their participation.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Blagojevic, C.; Heung, T.; Theriault, M.; Tomita-Mitchell, A.; Chakraborty, P.; Kernohan, K.; Bulman, D.E.; Bassett, A.S. Estimate of the contemporary live-birth prevalence of recurrent 22q11.2 deletions: A cross-sectional analysis from population-based newborn screening. CMAJ Open 2021, 9, E802–E809. [Google Scholar] [CrossRef] [PubMed]
  2. Campbell, I.M.; Sheppard, S.E.; Crowley, T.B.; McGinn, D.E.; Bailey, A.; McGInn, M.J.; Unolt, M.; Homans, J.F.; Chen, E.Y.; Salmons, H.I.; et al. What is new with 22q? An update from the 22q and You Center at the Children’s Hospital of Philadelphia. Am. J. Med. Genet. A 2018, 176, 2058–2069. [Google Scholar] [CrossRef] [PubMed]
  3. Grati, F.R.; Molina Gomes, D.; Ferreira, J.C.P.B.; Dupont, C.; Alesi, V.; Gouas, L.; Horelli-Kuitunen, N.; Choy, K.W.; García-Herrero, S.; De La Vega, A.G.; et al. Prevalence of recurrent pathogenic microdeletions and microduplications in over 9500 pregnancies. Prenat. Diagn. 2015, 35, 801–809. [Google Scholar] [CrossRef] [PubMed]
  4. Martin, K.; Iyengar, S.; Kalyan, A.; Lan, C.; Simon, A.; Stosic, M.; Kobara, K.; Ravi, H.; Truong, T.; Ryan, A.; et al. Clinical experience with a single-nucleotide polymorphism-based non-invasive prenatal test for five clinically significant microdeletions. Clin. Genet. 2017, 93, 293–300. [Google Scholar] [CrossRef] [Green Version]
  5. Maisenbacher, M.K.; Merrion, K.; Pettersen, B.; Young, M.; Paik, K.; Iyengar, S.; Kareht, S.; Sigurjonsson, S.; Demko, Z.P.; Martin, K.A. Incidence of the 22q11.2 deletion in a large cohort of miscarriage samples. Mol. Cytogenet. 2017, 10, 6. [Google Scholar] [CrossRef] [Green Version]
  6. Shaikh, T.H.; Kurahashi, H.; Saitta, S.C.; O’Hare, A.M.; Hu, P.; Roe, B.A.; Driscoll, D.A.; McDonald-McGinn, D.M.; Zackai, E.H.; Budarf, M.L.; et al. Chromosome 22-specific low copy repeats and the 22q11.2 deletion syndrome: Genomic organization and deletion endpoint analysis. Hum. Mol. Genet. 2000, 9, 489–501. [Google Scholar] [CrossRef] [Green Version]
  7. Shprintzen, R.J. Velo-cardio-facial syndrome: 30 Years of study. Dev. Disabil. Res. Rev. 2008, 14, 3–10. [Google Scholar] [CrossRef]
  8. Monteiro, F.; Vieira, T.P.; Sgardioli, I.C.; Molck, M.C.; Damiano, A.P.; Souza, J.; Monlleo, I.; Fontes, M.I.B.; Fett-Conte, A.C.; Félix, T.M.; et al. Defining new guidelines for screening the 22q11.2 deletion based on a clinical and dysmorphologic evaluation of 194 individuals and review of the literature. Eur. J. Pediatr. 2013, 172, 927–945. [Google Scholar] [CrossRef]
  9. Desmaze, C.; Prieur, M.; Amblard, F.; Aikem, M.; LeDeist, F.; Demczuk, S.; Zucman, J.; Plougastel, B.; Delattre, O.; Croquette, M.F. Physical mapping by FISH of the DiGeorge critical region (DGCR): Involvement of the region in familial cases. Am. J. Hum. Genet. 1993, 53, 1239–1249. [Google Scholar]
  10. Scambler, P.J.; Carey, A.H.; Wyse, R.K.; Roach, S.; Dumanski, J.P.; Nordenskjold, M.; Williamson, R. Microdeletions within 22q11 associated with sporadic and familial DiGeorge syndrome. Genomics 1991, 10, 201–206. [Google Scholar] [CrossRef]
  11. McDonald-McGinn, D.M.; Tonnesen, M.K.; Laufer-Cahana, A.; Finucane, B.; Driscoll, D.A.; Emanuel, B.S.; Zackai, E.H. Phenotype of the 22q11.2 deletion in individuals identified through an affected relative: Cast a wide FISHing net! Genet. Med. 2001, 3, 23–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Van Hemel, J.O.; Schaap, C.; Van Opstal, D.; Mulder, M.P.; Niermeijer, M.F.; Meijers, J.H. Recurrence of DiGeorge syndrome: Prenatal detection by FISH of a molecular 22q11 deletion. J. Med. Genet. 1995, 32, 657–658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. McDonald-McGinn, D.M.; Kirschner, R.; Goldmuntz, E.; Sullivan, K.; Eicher, P.; Gerdes, M.; Moss, E.; Solot, C.; Wang, P.; Jacobs, I.; et al. The Philadelphia story: The 22q11.2 deletion: Report on 250 patients. Genet. Couns. 1999, 10, 11–24. [Google Scholar] [PubMed]
  14. McDonald-McGinn, D.M.; Sullivan, K.; Marino, B.; Philip, N.; Swillen, A.; Vortsman, J.; Zackai, E.H.; Emanuel, B.; Vermeesch, J.; Morrow, B.; et al. 22q11.2 deletion syndrome. Nat. Rev. Dis. Primers. 2015, 19, 15071. [Google Scholar] [CrossRef] [Green Version]
  15. Swillen, A.; Vogels, A.; Devriendt, K.; Fryns, J.P. Chromosome 22q11 deletion syndrome: Update and review of the clinical features, cognitive-behavioral spectrum, and psychiatric complications. Am. J. Med. Genet. 2000, 97, 128–135. [Google Scholar] [CrossRef]
  16. Bassett, A.S.; Chow, E.W.C. Schizophrenia and 22q11.2 deletion syndrome. Curr. Psychiatry Rep. 2008, 10, 148–157. [Google Scholar] [CrossRef] [Green Version]
  17. Bassett, A.S.; Chow, E.W.C.; Husted, J.; Hodgkinson, K.A.; Oechslin, E.; Harris, L.; Silversides, C. Premature death in adults with 22q11.2 deletion syndrome. J. Med. Genet. 2009, 46, 324–330. [Google Scholar] [CrossRef] [Green Version]
  18. Bertini, V.; Azzarà, A.; Legitimo, A.; Milone, R.; Battini, R.; Consolini, R.; Valetto, A. Deletion Extents Are Not the Cause of Clinical Variability in 22q11.2 Deletion Syndrome: Does the Interaction between DGCR8 and miRNA-CNVs Play a Major Role? Front Genet. 2017, 8, 47. [Google Scholar] [CrossRef] [Green Version]
  19. Rozas, M.F.; Benavides, F.; León, L.; Repetto, G.M. Association between phenotype and deletion size in 22q11.2 microdeletion syndrome: Systematic review and meta-analysis. Orphanet J. Rare Dis. 2019, 14, 195. [Google Scholar] [CrossRef]
  20. Budarf, M.L.; Konkle, B.A.; Ludlow, L.B.; Michaud, D.; Li, M.; Yamashiro, D.J.; McDonald-McGinn, D.; Zackai, E.H.; Driscoll, D.A. Identification of a patient with Bernard-Soulier syndrome and a deletion in the DiGeorge/velo-cardio-facial chromosomal region in 22q11.2. Hum. Mol. Genet. 1995, 4, 763–766. [Google Scholar] [CrossRef]
  21. McDonald-McGinn, D.M.; Fahiminiya, S.; Revil, T.; Nowakowska, B.A.; Suhl, J.; Bailey, A.; Mlynarski, E.; Lynch, D.R.; Yan, A.C.; Bilaniuk, L.T.; et al. Hemizygous mutations in SNAP29 unmask autosomal recessive conditions and contribute to atypical findings in patients with 22q11.2DS. J. Med. Genet. 2012, 50, 80–90. [Google Scholar] [CrossRef] [Green Version]
  22. Johnston, J.J.; van der Smagt, J.J.; Rosenfeld, J.A.; Pagnamenta, A.T.; Alswaid, A.; Baker, E.H.; Blair, E.; Borck, G.; Brinkmann, J.; Craigen, W.; et al. Autosomal recessive Noonan syndrome associated with biallelic LZTR1 variants. Genet. Med. 2018, 20, 1175–1185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Chinton, J.; Huckstadt, V.; Foncuberta, M.E.; Perez, M.M.; Bonetto, M.C.; Gravina, L.P.; Obregon, M.G. Challenges in genetic diagnosis, co-occurrence of 22q11.2 deletion syndrome and Noonan syndrome. Am. J. Med. Genet. Part A 2022, 188, 2505–2508. [Google Scholar] [CrossRef] [PubMed]
  24. Bedeschi, M.; Colombo, L.; Mari, F.; Hofmann, K.; Rauch, A.; Gentilin, B.; Renieri, A.; Clerici, D. Unmasking of a Recessive SCARF2 Mutation by a 22q11.12 de novo Deletion in a Patient with Van den Ende-Gupta Syndrome. Mol. Syndr. 2010, 1, 239–245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Fenwick, A.L.; Kliszczak, M.; Cooper, F.; Murray, J.; Pulido, L.S.; Twigg, S.; Goriely, A.; McGowan, S.; Miller, K.A.; Taylor, I.B.; et al. Mutations in CDC45, Encoding an Essential Component of the Pre-initiation Complex, Cause Meier-Gorlin Syndrome and Craniosynostosis. Am. J. Hum. Genet. 2016, 99, 125–138. [Google Scholar] [CrossRef] [Green Version]
  26. Unolt, M.; Kammoun, M.; Nowakowska, B.; Graham, G.E.; Crowley, T.B.; Hestand, M.S.; Demaerel, W.; Geremek, M.; Emanuel, B.S.; Zackai, E.H.; et al. Pathogenic variants in CDC45 on the remaining allele in patients with a chromosome 22q11.2 deletion result in a novel autosomal recessive condition. Genet. Med. 2019, 22, 326–335. [Google Scholar] [CrossRef]
  27. Hestand, M.S.; Nowakowska, B.A.; Vergaelen, E.; Van Houdt, J.; Dehaspe, L.; Suhl, J.A.; Del-Favero, J.; Mortier, G.; Zackai, E.; Swillen, A.; et al. A catalog of hemizygous variation in 127 22q11 deletion patients. Hum. Genome Var. 2016, 3, 15065. [Google Scholar] [CrossRef] [Green Version]
  28. Cohen, J.L.; Crowley, T.B.; McGinn, D.E.; McDougall, C.; Unolt, M.; Lambert, M.P.; Emanuel, B.S.; Zackai, E.H.; McDonald-McGinn, n.M. 22q and two: 22q11.2 deletion syndrome and coexisting conditions. Am. J. Med. Genet. Part A 2018, 176, 2203–2214. [Google Scholar] [CrossRef]
  29. Wang, K.; Li, M.; Hakonarson, H. ANNOVAR: Functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010, 38, e164. [Google Scholar] [CrossRef]
  30. Gambin, T.; Akdemir, Z.C.; Yuan, B.; Gu, S.; Chiang, T.; Carvalho, C.M.; Shaw, C.; Jhangiani, S.; Boone, P.M.; Eldomery, M.K.; et al. Homozygous and hemizygous CNV detection from exome sequencing data in a Mendelian disease cohort. Nucleic Acids Res. 2016, 45, 1633–1648. [Google Scholar] [CrossRef]
  31. Lalani, S.R.; Liu, P.; Rosenfeld, J.A.; Watkin, L.B.; Chiang, T.; Leduc, M.S.; Zhu, W.; Ding, Y.; Pan, S.; Vetrini, F.; et al. Recurrent Muscle Weakness with Rhabdomyolysis, Metabolic Crises, and Cardiac Arrhythmia Due to Bi-allelic TANGO2 Mutations. Am. J. Hum. Genet. 2016, 98, 347–357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kunishima, S.; Imai, T.; Kobayashi, R.; Kato, M.; Ogawa, S.; Saito, H. Bernard-Soulier syndrome caused by a hemizygous GPIbβ mutation and 22q11.2 deletion. Pediatr Int. 2013, 55, 434–437. [Google Scholar] [CrossRef] [PubMed]
  33. Zhao, Y.; Diacou, A.; Johnston, H.R.; Musfee, F.I.; McDonald-McGinn, D.M.; McGinn, D.; Crowley, T.B.; Repetto, G.M.; Swillen, A.; Breckpot, J.; et al. Complete Sequence of the 22q11.2 Allele in 1053 Subjects with 22q11.2 Deletion Syndrome Reveals Modifiers of Conotruncal Heart Defects. Am. J. Hum. Genet. 2019, 106, 26–40. [Google Scholar] [CrossRef] [PubMed]
  34. Dines, J.N.; Golden-Grant, K.; LaCroix, A.; Muir, A.M.; Cintrón, D.L.; McWalter, K.; Cho, M.T.; Sun, A.; Merritt, J.L.; Thies, J.; et al. TANGO2: Expanding the clinical phenotype and spectrum of pathogenic variants. Genet. Med. 2018, 21, 601–607. [Google Scholar] [CrossRef] [Green Version]
  35. Lin, M.; Pedrosa, E.; Hrabovsky, A.; Chen, J.; Puliafito, B.R.; Gilbert, S.R.; Zheng, D.; Lachman, H.M. Integrative transcriptome network analysis of iPSC-derived neurons from schizophrenia and schizoaffective disorder patients with 22q11.2 deletion. BMC Syst. Biol. 2016, 10, 105. [Google Scholar] [CrossRef] [Green Version]
  36. Ota, V.K.; Bellucco, F.T.; Gadelha, A.; Santoro, M.L.; Noto, C.; Christofolini, D.M.; Assunção, I.B.; Yamada, K.M.; Ribeiro-dos-Santos, A.K.; Santos, S.; et al. PRODH polymorphisms, cortical volumes and thickness in schizophrenia. PLoS One 2014, 9, e87686. [Google Scholar] [CrossRef] [Green Version]
  37. Guo, X.; Tang, P.; Yang, C.; Li, R. Proline dehydrogenase gene (PRODH) polymorphisms and schizophrenia susceptibility: A meta-analysis. Metab. Brain Dis. 2017, 33, 89–97. [Google Scholar] [CrossRef]
  38. Girirajan, S.; Rosenfeld, J.A.; Cooper, G.M.; Antonacci, F.; Siswara, P.; Itsara, A.; Vives, L.; Walsh, T.; McCarthy, S.E.; Baker, C.; et al. A recurrent 16p12.1 microdeletion supports a two-hit model for severe developmental delay. Nat. Genet. 2010, 42, 203–209. [Google Scholar] [CrossRef]
  39. Li, D.; Tekin, M.; Buch, M.; Fan, Y.S. Co-existence of other copy number variations with 22q11.2 deletion or duplication: A modifier for variable phenotypes of the syndrome? Mol. Cytogenet. 2012, 5, 18. [Google Scholar] [CrossRef] [Green Version]
  40. Bartnik, M.; Nowakowska, B.; Derwińska, K.; Wiśniowiecka-Kowalnik, B.; Kędzior, M.; Bernaciak, J.; Ziemkiewicz, K.; Gambin, T.; Sykulski, M.; Bezniakow, N.; et al. Application of array comparative genomic hybridization in 256 patients with developmental delay or intellectual disability. J. Appl. Genet. 2013, 55, 125–144. [Google Scholar] [CrossRef] [Green Version]
  41. Mlynarski, E.E.; Sheridan, M.B.; Xie, M.; Guo, T.; Racedo, S.E.; McDonald-McGinn, D.M.; Gai, X.; Chow, E.W.; Vorstman, J.; Swillen, A.; et al. Copy-Number Variation of the Glucose Transporter Gene SLC2A3 and Congenital Heart Defects in the 22q11.2 Deletion Syndrome. Am. J. Hum. Genet. 2015, 96, 753–764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Mlynarski, E.E.; Xie, M.; Taylor, D.; Sheridan, M.B.; Guo, T.; Racedo, S.E.; McDonald-McGinn, D.M.; Chow, E.W.C.; Vorstman, J.; Swillen, A.; et al. Rare copy number variants and congenital heart defects in the 22q11.2 deletion syndrome. Hum. Genet. 2016, 135, 273–285. [Google Scholar] [CrossRef] [Green Version]
  43. Abdollahi, M.R.; Morrison, E.; Sirey, T.; Molnar, Z.; Hayward, B.E.; Carr, I.M.; Springell, K.; Woods, C.G.; Ahmed, M.; Hattingh, L.; et al. Mutation of the Variant α-Tubulin TUBA8 Results in Polymicrogyria with Optic Nerve Hypoplasia. Am. J. Hum. Genet. 2009, 85, 737–744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Dantas, A.G.; Santoro, M.L.; Nunes, N.; de Mello, C.B.; Pimenta, L.S.E.; Meloni, V.A.; Soares, D.C.Q.; Belangero, S.N.; Carvalheira, G.; Kim, C.A.; et al. Downregulation of genes outside the deleted region in individuals with 22q11.2 deletion syndrome. Hum. Genet. 2019, 138, 93–103. [Google Scholar] [CrossRef] [PubMed]
  45. Jehee, F.S.; Rosenberg, C.; Krepischi, A.; Kok, F.; Knijnenburg, J.; Froyen, G.; Vianna-Morgante, A.M.; Opitz, J.M.; Passos-Bueno, M.R. An Xq22.3 duplication detected by comparative genomic hybridization microarray (Array-CGH) defines a new locus (FGS5) for FG syndrome. Am. J. Med. Genet. Part A 2005, 139A, 221–226. [Google Scholar] [CrossRef] [Green Version]
  46. Geetha, T.S.; Michealraj, K.A.; Kabra, M.; Kaur, G.; Juyal, R.C.; Thelma, B. Targeted Deep Resequencing Identifies MID2 Mutation for X-Linked Intellectual Disability with Varied Disease Severity in a Large Kindred from India. Hum. Mutat. 2013, 35, 41–44. [Google Scholar] [CrossRef]
  47. Chanchani, S.R.; Xie, H.; Sekhon, G.; Melikishvili, A.M.; Moyer Harasink, S.; Pall, H.; Giampietro, P.F. A male infant with Xq22.2q22.3 duplication containing PLP1 and MID2. Mol. Genet. Genom. Med. 2020, 8, e1078. [Google Scholar]
Figure 1. HMZDelFinder identified; (A) clinically uncertain, hemizygous deletion, in the remaining 22q11.2 region. The variant encompasses two genes: DGCR6, PRODH (Chr22:18906447-18931248). (B) Pathogenic, hemizygous deletion, in the remaining 22q11.2 region. The variant encompasses 8 exons of TANGO2 (chr22:20043265-20064650).
Figure 1. HMZDelFinder identified; (A) clinically uncertain, hemizygous deletion, in the remaining 22q11.2 region. The variant encompasses two genes: DGCR6, PRODH (Chr22:18906447-18931248). (B) Pathogenic, hemizygous deletion, in the remaining 22q11.2 region. The variant encompasses 8 exons of TANGO2 (chr22:20043265-20064650).
Genes 14 00680 g001
Table 1. Copy number variants identified in 159 (82 prenatal cases and 77 postnatal patients) patients, in addition to the 22q11 deletion. CNVs were classified as pathogenic, likely pathogenic, and of uncertain significance (VOUS) based on clinical data in known, free-access CNV databases. OMIM morbid genes are bolded.
Table 1. Copy number variants identified in 159 (82 prenatal cases and 77 postnatal patients) patients, in addition to the 22q11 deletion. CNVs were classified as pathogenic, likely pathogenic, and of uncertain significance (VOUS) based on clinical data in known, free-access CNV databases. OMIM morbid genes are bolded.
Patient IDVariantSexAgeSizePathogenicity/
CNV Classification
Protein Coding Genes or Known CNV RegionPatients’ Phenotype22q11.2 Recurrent Region
PD5864arr[GRCh37] 11q24.2q25(125857822_134868420)x1M17–18 Hbd9.01 Mbpathogenic38 protein coding genes, 11q (Jacobsen syndrome) regionFetal death, lip and cleft palate, edema.proximal, A-B
arr[hg19] 22q11.1q11.21(16940617_18706059)x1 1.77 Mblikely pathogenicCCT8L2, XKR3, GAB4, IL17RA, TMEM121B, HDHD5, ADA2, CECR2, SLC25A18, ATP6V1E1, BCL2L13, BID, MICAL3, PEX26, TUBA8, USP18
PD8038arr[GRCh37] 22q11.21q11.23(21759521_23822984)x1M17–18 Hbd2.06 Mbpathogenic22q11.2 recurrent region (distal type I, D-E/F)Symmetrical hypotrophy of the fetus, cardiac defectproximal, B-D
GC034796arr[GRCh37] 13q21.32(67204211_67215612)x1M1 y 3 m11 kbuncertain clinical significanceexon 4 of PCDH9Global developmental delay, ventricular septal defect, interrupted aortic arch, bicuspid aortic valve, atrial septal defect, aberrant subclavian, thymic aplasia with absent T cells, hypocalcemia, nasal regurgitation, thyroid hypoplasia, hypothyroidism, growth hormone deficiency, growth delay, dysmorphic features (micrognathia), scoliosis, butterfly vertebrae, additional ribs, hypotonia, ligamentous laxity, delayed dental eruption, chronic constipationproximal, A-C
arr[GRCh37] 16p11.2(29673967_30190593)x1 517 kbpathogenic16p11.2 recurrent region (proximal, BP4-BP5) (includes TBX6)
arr[GRCh37] 22q11.21(18375151_18661758)x1 287 kbuncertain clinical significanceexons 1–11 of MICAL3, PEX26, TUBA8, USP18
113787arr[GRCh37] 20p12.3(8256689_8558204)x1F5 y301 kbuncertain clinical significanceexon 3 of PLCB1Global developmental delay, delay of speech development, dysmorphic featuresproximal, A-B
arr[GRCh37] 22q11.1q11.21(16940617_18848020)x3 1.91 Mbpathogenic22q11.21 recurrent (Cat eye syndrome) region (includes CECR2)
GC034823arr[GRCh37] 22q11.1q11.21(17397633_18661758)x1F25 y1.26 Mblikely pathogenicGAB4, IL17RA, TMEM121B, HDHD5, ADA2, CECR2, SLC25A18, ATP6V1E1, BCL2L13, BID, MICAL3, PEX26, TUBA8, USP18Ventricular septal defect, double outlet right ventricle, pulmonary (valve) stenosis, unilateral cleft palate and lip, bifid uvula, velopharyngeal insufficiency, growth delay, dysmorphic features, myopia, scoliosis, dental problems, conductive hearing loss, nasopharyngeal reflux, chronic constipation, global developmental delay, delayed speech and language development, moderate intellectual disability, mood changesproximal, A-B
arr[GRCh37] 22q13.33(51162483_51178213)x3 16 kbuncertain clinical significanceACR, exons 1–21 of SHANK3
111183arr[GRCh37] 16p12.2(21959950_22407951)x1F1 m448 kblikely pathogenic16p12.2 recurrent region (proximal) (includes EEF2K, CDR2)Facial dysmorphic features, cardiac defect, urinary system defectproximal, A-D
105620arr[GRCh37] 12q24.33(132239944_133773393)x1,M6 y1.53 Mblikely pathogenicSFSWAP, MMP17, ULK1, PUS1, EP400, DDX51, NOC4L, GALNT9, FBRSL1, LRCOL1, P2RX2, POLE, PXMP2, PGAM5, ANKLE2, GOLGA3, CHFR, ZNF605, ZNF26, ZNF84, ZNF140, ZNF891, ZNF10, ZNF268Hypotonia (low muscle tone), dysmorphic features, lack of speech development, cardiac defectproximal, A-B
arr[GRCh37] 22q11.1q11.21(17397633_18661758)x1 1.26 Mblikely pathogenicGAB4, IL17RA, TMEM121B, HDHD5, ADA2, CECR2, SLC25A18, ATP6V1E1, BCL2L13, BID, MICAL3, PEX26, TUBA8, USP18
GC034824 /106502arr[GRCh37] Xp11.23(47616035_48204099)x3F2 y 3 m588 kbuncertain clinical significanceZNF81, ZNF182, ZNF630, SPACA5, SPACA5B, SSX5, SSX1Aberrant subclavian artery, recurrent infections, thymic hypoplasia, hypernasal speech, nasal regurgitation, pronunciation defects, short stature, dysmorphic features (hypotelorism, bitemporal narrowing, micrognathia, retrognathia) long fingers, hypotonia, ligamentous laxity, dental problems, enuresis, gastrointestinal problems, global developmental delayproximal, A-D
108894arr[GRCh37] 1p36.23(8736229_9105539)x3 matM2 y369 kbuncertain clinical significanceexons 1–2 of RERE, ENO1, CA6, SLC2A7, exons 5–12 of SLC2A5Global developmental delay, facial dysmorphic features, hypotonia (low muscle tone)proximal, A-D
110226arr[GRCh37] 4q13.1(65794809_66356976)x1 matM11 y562 kbuncertain clinical significanceexons 1–2 of EPHA5Hypotonia (low muscle tone), delay of speech developmentproximal, A-D
111711arr[GRCh37] 12q12(44202928_44445117)x1M1 m242 kbuncertain clinical significanceexons 1–3 of TMEM117Facial dysmorphic features, cardiac defectproximal, A-D
112367arr[GRCh37] 22q11.22q11.23(23012069_23648159)x1 patM9 m637 kbuncertain clinical significance22q11.2 recurrent region (distal type II, E-F)Cardiac defectproximal, A-D
112587arr[GRCh37] 15q13.2q13.3(30419801_32861612)x3M6 y2.44 Mbuncertain clinical significance15q13.3 recurrent region (BP4-BP5) (includes CHRNA7)No dataproximal, A-D
113324arr[GRCh37] 7q21.3q22.1(97939915_98557740)x3 matF1 y 4 m618 kbuncertain clinical significanceexons 1–8 of BAIAP2L1, NPTX2, TMEM130, exons 1–44 of TRRAPDefect of the larynx, thymic hypoplasia, dysmorphic featuresproximal, A-D
arr[GRCh37] 22q11.23(23720181_25066484)x3 mat 1.35 Mbuncertain clinical significance22q11.2 recurrent region (distal type III, F-H)
116123arr[GRCh37] 18p11.31p11.23(7094706_8359012)x3F1 m1.26 Mbuncertain clinical significanceexon 1 of LAMA1, LRRC30, exons 1–23 of PTPRMFacial dysmorphic features, defect of the urinary system, cardiac defect (vascular ring), long fingers.proximal, A-D
PD2305arr[GRCh37] 7p12.3p12.2(46094932_49190408)x3M22 Hbd3.1 Mbuncertain clinical significanceTNS3, PKD1L1, HUS1, SUN3, C7orf57, UPP1, ABCA13, CDC14CScreening test result showing increased risk of chromosomal aberration, fetal cardiac defectproximal, A-D
PD3311arr[GRCh37] 22q11.22q11.23(23012069_23648159)x1 patM21 Hbd636 kbuncertain clinical significance22q11.2 recurrent region (distal type II, E-F)Fetal cardiac defectproximal, A-D
GC034808arr[GRCh37] Xq22.3(106791395_107156386)x2M6y465 kbuncertain clinical significanceFRMPD3, NCBP2L, PRPS1, TSC22D3, exons 1–5 of MID2Recurrent infections, acute otitis media, submucosal cleft palate, bifid uvula, secondary hearing loss, pronunciation defects, speech articulation difficulties, short stature, dysmorphic features, hypotonia, ligamentous laxity, feeding difficulties, umbilical hernia, global developmental delay, delayed speech and language development, attention difficulties proximal, A-D
108366arr[GRCh37] 8p12(30498811_30732891)x3M9 y234 kbuncertain clinical significanceexons 1–2 of GTF2E2, SMIM18, GSR, UBXN8, PPP2CB, exons 2–7 of TEX15Global developmental delay, facial dysmorphic features, epilepsyproximal, A-B
GC034815arr[GRCh37] 3p26.3(1539221_2206719)x1F17 y667 kbuncertain clinical significanceexons 1–2 of CNTN4Patent foramen ovale, defects in humoral immunity, IgA deficiency, recurrent infections, acute otitis media, chronic sinusitis, T-cell lymphopenia, vitiligo, thrombocytopenia, Hashimoto thyroiditis, velopharyngeal insufficiency, laryngotracheoesophageal anomalies, hypothyroidism, hypoparathyroidism, dysmorphic features, myopia, amblyopia, hypotonia, ligamentous laxity, delayed dental eruption, feeding and swallowing problems, delayed speech and language development, supernumerary spleensproximal, A-D
arr[GRCh37] 7q21.3q22.1(97939915_98557740)x3 618 kbuncertain clinical significanceexons 1–8 of BAIAP2L1, NPTX2, TMEM130, exons 1–44 of TRRAP
GC028958arr[GRCh37] 8q22.1(96815252_97229772)x1F2 weeks414 kbuncertain clinical significanceGDF6Child succumbed at 14 days of life following her heart repair on day of life 13.
Ventricular septal defect, persistent truncus arteriosus, atrial septal defect, bilateral coronal craniosynostosis, diffuse white matter gliosis, reduced cortical thickness, thymic aplasia with absent T cells, hypocalcemia, hypoparathyroidism, intestinal malrotation, anteriorly placed anus, diaphragmatic hernia
proximal, A-D
arr[GRCh37] Xq28(151896983_152351582)x3455 kbuncertain clinical significanceCETN2, CSAG1, MAGEA12, MAGEA2, MAGEA3, NSDHL, PNMA3, PNMA5, PNMA6A, PNMA6C, ZNF185
(8)x2~3146.36 Mpzpathogenicmosaic trisomy
Table 2. Pathogenic and likely pathogenic SNVs, of autosomal dominant inheritance, were selected using filtering with an allele frequency less than 0.001 in gnomAD, then filtered against an in-house control dataset. EBD—Epidermolysis bullosa dystrophica.
Table 2. Pathogenic and likely pathogenic SNVs, of autosomal dominant inheritance, were selected using filtering with an allele frequency less than 0.001 in gnomAD, then filtered against an in-house control dataset. EBD—Epidermolysis bullosa dystrophica.
IDSexAgeChrChangeZygGeneFunctionOMIM_Diseases Linked to GeneExonic FunctionGnomAD Exome ALLPatient’s Clinical Features Associated with Variant
GC034931M7 ychr2NM_000384:c.9115_9119del:p.F3039fshetAPOBexonicHypercholesterolemia, familial, 2; Hypobetalipoproteinemiaframeshift deletion4.071 × 10−6NA
GC034813M12 ychrXNM_000495:c.G1871A:p.G624DhomCOL4A5exonicAlport syndrome 1, X-linkednonsynonymous SNV8.97 × 10−5NA
GC034793M7 ychr3NM_000094:c.A425G:p.K142RhetCOL7A1exonicEBD inversa; EBD, Bart type;
EBD, localisata variant;
Epidermolysis bullosa dystrophica, AD; Epidermolysis bullosa dystrophica AR; Epidermolysis bullosa pruriginosa; Epidermolysis bullosa, pretibial;
Toenail dystrophy, isolated;
Transient bullous of the newborn
nonsynonymous SNV4.468 × 10−5NA
GC034931M7 ychr11 hetF2UTR3Dysprothrombinemia; Hypoprothrombinemia;
Thrombophilia due to thrombin defect
0NA
GC030951M6 ychr17NM_000267:c.C5839T:p.R1947XhetNF1exonicLeukemia, juvenile myelomonocytic; Neurofibromatosis, familial spinal; Neurofibromatosis, type 1; Neurofibromatosis-Noonan syndrome; Watson syndromestopgain0Neurofibromatosis
GC034900F1 ychr4NM_012464.5:c.713T>C:p.p.Val238AlahetTLL1exonicAtrial septal defect 6nonsynonymous SNV2 × 10−4Atrial septal defect
GC034800F7 ychr12NM_000552:c.G2561A:p.R854QhetVWFexonicvon Willebrand disease, type 1;
von Willebrand disease, type 3;
von Willebrand disease, types 2A, 2B, 2M, and 2N
nonsynonymous SNV0.0034NA
Table 3. Rare SNVs of uncertain clinical significance. CNS—central nervous system; VSD—Ventricular septal defect; IAA—interrupted aortic arch; BAV—bicuspid aortic valve; ASD—atrial septal defect; PTA—patent ductus arteriosus; DORV—double outlet right ventricle.
Table 3. Rare SNVs of uncertain clinical significance. CNS—central nervous system; VSD—Ventricular septal defect; IAA—interrupted aortic arch; BAV—bicuspid aortic valve; ASD—atrial septal defect; PTA—patent ductus arteriosus; DORV—double outlet right ventricle.
IdentifierSexAgeInheritanceChrStartEndRefAltZygGeneOMIM DiseasesPatient’s Phenotype Features Correlating with OMIM Phenotype
GC028956F13 yADchr9128607934128607934AChetSPTAN1Developmental and epileptic encephalopathy 5Structural CNS anomalies (spina bifida, polyhydramnios), unprovoked seizures, developmental delay
GC028956 F13 yADchr183479422334794223TGhetDTNALeft ventricular noncompaction 1, with or without congenital heart defectsCongenital heart defects (VSD)
GC030952 F24 yADchr2120982720120982720AChetGLI2Culler-Jones syndrome; Holoprosencephaly 9Palatal abnormalities (bifid uvula), growth delay, delayed psychomotor development
GC034768M6 yADchr191098624610986246GAhetSMARCA4Coffin-Siris syndrome 4Congenital heart defects (VSD, IAA, BAV, ASD), dental problems, hypotonia, global developmental delay, delayed speech and language development, learning disability
GC034778F10 yADchr184495209344952093GAhetSETBP1Intellectual developmental disorder, autosomal dominant 29; Schinzel–Giedion midface retraction syndromeGlobal developmental delay, delayed speech and language development, learning disability
GC034779F7 yADchr21594572315945723AThetMYCNFeingold syndrome 1Asymmetric face, narrow palpebral fissures, epicantic folds, micrognathia, palatal abnormalities (bifid uvula), hearing loss, global developmental delay, delayed speech and language development, learning disability
GC034783F8 yADchr1211082791211082791GAhetKCNH1Temple-Baraitser syndrome;
Zimmermann-Laband syndrome 1
Cardiovascular system defects (ASD, conotruncal cardiac anomaly, aberrant subclavian), hypertelorism, skeletal abnormalities (proximal implantation of thumbs), learning disability
GC034785M19 yADchr86080042260800422GAhetCHD7CHARGE syndrome; Hypogonadotropic hypogonadism 5 with or without anosmiaSubmucosal cleft palate, dysmorphic face (facial asymmetry, hypertelorism, malar flattening, micrognathia, cup ear), hearing loss, skeletal anomalies of limbs, feeding and swallowing problems, intellectual disability, learning disability, thymic hypoplasia, T-cell lymphopenia
GC034786M2 y 6 mADchr177477767747776AGhetCAMTA1Cerebellar dysfunction with variable cognitive and behavioral abnormalitiesBulbous, wide nose, low-set ears, delayed speech and language development, hypotonia, dental problems, gastrointestinal problems
GC034787M1 yADchr3119402046119402046CGhetARHGAP31Adams-Oliver syndrome 1Microcephaly, palatal anomalies (bifid uvula), cardiovascular system defects (VSD, PTA, Pulmonary artery stenosis), hypotonia, global developmental delay
GC034789F6 mXLDchrX4113476341134763CThetUSP9XIntellectual developmental disorder, X-linked 99, syndromic, female-restrictedCongenital heart defects (VSD, ASD), dysmorphic face (prominent forehead, bitemporal narrowing, posteriorly rotated ears, broad nasal bridge, bulbous nose), hypotonia, sensory processing problems
GC034796M1 y 3 mADchr1151440971151440971GThetPOGZWhite-Sutton syndromeCongenital heart defects (VSD, ASD, IAA, BAV, aberrant subclavian), growth delay, dysmorphic face (low-set ears, posteriorly rotated ears, short philtrum), hypotonia, global developmental delay
GC034797F2 y 2 mADchr2227309265227309265CThetCOL4A3Alport syndrome 3, autosomal dominant; Hematuria, benign familialSensorineural hearing loss
GC034800F7 yADchr12755205527552055-GhetAHDC1Xia-Gibbs syndromeLow-set ears, global developmental delay, delayed speech and language development, intellectual disability, learning disability
GC034800F7 yADchr64549205445492054CThetRUNX2Cleidocranial dysplasia; Cleidocranial dysplasia, forme fruste, dental anomalies only; Cleidocranial dysplasia, forme fruste, with brachydactyly; Metaphyseal dysplasia with maxillary hypoplasia with or without brachydactylyProminent forehead, dental problems, high-arched palate, skeletal abnormalities
GC034813M12 yADchr1637693453769345CAhetCREBBPMenke-Hennekam syndrome 1;
Rubinstein-Taybi syndrome 1
Congenital heart defects (Tetralogy of Fallot, VSD, ASD, conotruncal heart defects) submucosal cleft palate, dysmorphic face (prominent forehead, broad nasal bridge, micrognathia, retrognathia, low-set ears), scoliosis, syndactyly, dental problems, cryptorchidism, recurrent infections, hypotonia, developmental delay, delayed speech and language development, intellectual disability
GC034813M12 yXLDchrX4506139645061396TChomKDM6AKabuki syndrome 2Congenital heart defects (conotruncal heart defects, Tetralogy of Fallot, VSD, ASD), submucosal cleft palate, cupped ears, dental problems, hypotonia, ligamentous laxity, developmental delay, behavioral difficulties
GC034822M10 yADchr191330055913300559GChetCACNA1ADevelopmental and epileptic encephalopathy 42; Episodic ataxia, type 2; Migraine, familial hemiplegic, 1; Migraine, familial hemiplegic, 1, with progressive cerebellar ataxia; Spinocerebellar ataxia 6Global developmental delay, hypotonia, intellectual disability (borderline), learning difficulties
GC034822M 10 yXLRchrX7045427770454277GAhomDLG3Intellectual developmental disorder, X-linked 90Global developmental delay, delayed speech development, intellectual disability (borderline), learning difficulties
GC034824F2 y 3 mADchr8143728049143728049CThetFAM83HAmelogenesis imperfecta, type IIIADental problems
GC034930F4 mADchr3111649747111649747AChetCD96C syndromeMicrognathia, posteriorly rotated ears, epicantic folds, broad nasal bridge, short nose, congenital heart defect (VSD)
GC034930F4 mADchr1119916604119916604CThetNOTCH2Alagille syndrome 2; Hajdu-Cheney syndromeCongenital heart defects (VSD, ASD, PTA), Bulbous nasal tip
GC034932F 1 yADchr12115996599115996599CThetMED13LImpaired intellectual development and distinctive facial features with or without cardiac defectsGlobal developmental delay
GC034823F25 yADchr8105801714105801714GAhetZFPM2Tetralogy of FallotVSD, DORV with pulmonary stenosis
GC034789F6 mADchr5173233189173233189CAhetNKX2-5Atrial septal defect 7, with or without AV conduction defects; Hypoplastic left heart syndrome 2; Hypothyroidism, congenital nongoitrous, 5; Tetralogy of Fallot; Ventricular septal defect 3Congenital heart defects (ASD, VSD)
GC034808M6 yADchr616102021610202CThomFOXC1Axenfeld-Rieger syndrome, type 3Secondary hearing loss, redundant periumbilical skin
Table 4. Pathogenic and of uncertain clinical significance deletions identified by HMZDelFinder.
Table 4. Pathogenic and of uncertain clinical significance deletions identified by HMZDelFinder.
IDSexAgeReults of HMZdelfinder AnalysisGene ContentGene Function
pathogenic
GC034806M 5 ychr22_20043265_20064650exons 2–9 of TANGO2encodes transport and golgi organization 2 homologs; bi-allelic mutations in TANGO2 cause recurrent muscle weakness with rhabdomyolysis, metabolic crises, and cardiac arrhythmia
uncertain pathogenicity
GC028957F27 ychr15_26914781_26914903exon 7 of GABRA5encodes γ-aminobutyric acid type A receptor alpha5 subunit; restricted expression toward brain; GABA is the major inhibitory neurotransmitter in the mammalian brain where it acts at GABA-A receptors, which are ligand-gated chloride channels
GC028958F2 weekschr15_26937134_26937376exon 8 of GABRA5encodes γ-aminobutyric acid type A receptor alpha5 subunit; restricted expression toward brain; GABA is the major inhibitory neurotransmitter in the mammalian brain where it acts at GABA-A receptors, which are ligand-gated chloride channels
GC030952F24 ychr16_75635620_75635862exon 7 of KARSencodes lysyl-tRNA synthetase; defects in KARS are associated with the recessive form of Charcot-Mary-Tooth polyneuropathy, the autosomal recessive non-syndromic hearing loss, congenital visual impairment and progressive microcephaly, hypertrophic cardiomyopathy and combined mitochondrial respiratory chain defect
chr22_26601827_26608074exons 4–5 of CRYBB1encodes crystallin β B1; mutations in CRYBB1 are associated with congenital cataract with nystagmus
GC035414M9 yChr22:18906447-18931248DGCR6, PRODHProline dehydrogenase is involved in the degradation of the amino acid proline. It catalyzes the conversion of proline to pyrroline-5-carboxylate, or P5C. It is associated with Hyperprolinemia, type I, and susceptibility to schizophrenia
GC030953M3 ychr12_21568814_21580552exons 9–12 of GYS2encodes glycogen synthase 2; mutations in this gene cause autosomal recessive glycogen storage disease type 0 (GSD-0)—a rare type of early childhood fasting hypoglycemia with decreased liver glycogen content
chr23_55751086_55751208exon 7 of RRAGBencodes Ras related GTP binding protein B
GC034767F3 ychr1_202135375_202154374exons 1–5 of ARL8A, exons 6–10 of PTPN7ARL8A—encodes ADP ribosylation factor like GTPase 8A; PTPN7—encodes protein tyrosine phosphatase non-receptor type 7
GC034773M6 ychr2_3702340_4228633exon 12 of ALLC, DCDC2C, LINC01304, ENSG00000215960ALLC encodes allantoicase, expressed in testis; DCDC2C encodes doublecortin domain containing 2C, low expression in testis; LINC01304 expressed in testis
GC034774F35 ychr12_6452218_6452340exon 1 of TAPBPLencodes TAP binding protein like, involved in controlling peptide presentation to the immune system
GC034780M10 ychr8_3029293_3230255exons 28–52 of CSMD1encodes CUB and Sushi multiple domains 1, expressed at intermediate level in brain, including cerebellum, substantia nigra, hippocampus and fetal brain; potential suppressor of squamous cell carcinomas; altered expression of the CSMD1 gene in the peripheral blood of schizophrenia patients; association with cognitive function
GC034789F6 mchr1_145850735_145854625exons 8–12 of PIAS3encodes protein inhibitor of activated STAT 3, plays a crucial role as a transcriptional coregulation in various cellular pathways, including the STAT pathway and the steroid hormone signaling pathway
chr1_145868270_145875066exons 8–11 of ANKRD35encodes ankyrin repeat domain 35, expression in skin, esophagus, association with chronic lymphocytic leukemia
GC034850M3 mchr1_196902403_196914600exons 2–8 of CFHR4encodes complement factor H related 4, expressed in liver, can associate with lipoproteins and may play a role in lipid metabolism
GC034825F34 ychr14_23988867_23990346exon 2 of DHRS4L2encodes a member of the short chain dehydrogenase reductase family. The encoded protein may be a NADPH-dependent retinol oxidoreductase
Table 5. Pathogenic, likely pathogenic, and variants of uncertain significance identified by array and exome sequencing. For the likely pathogenic variants, only CNVs and SNVs which correlated with the patient phenotypes have been included.
Table 5. Pathogenic, likely pathogenic, and variants of uncertain significance identified by array and exome sequencing. For the likely pathogenic variants, only CNVs and SNVs which correlated with the patient phenotypes have been included.
PathogenicLikely PathogenicVariants of Uncertain Significance
aCGH
159 cases
CNV + aneuploidy
5 (3.14%)5 (3.14%)21 (13.21%)
ES
86 cases
SNV + small deletions
1 deletion + 2 SNVs
3 (3.49%)
5 SNVs
5 (5.81%)
15 deletions +
911 SNVs
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Smyk, M.; Geremek, M.; Ziemkiewicz, K.; Gambin, T.; Kutkowska-Kaźmierczak, A.; Kowalczyk, K.; Plaskota, I.; Wiśniowiecka-Kowalnik, B.; Bartnik-Głaska, M.; Niemiec, M.; et al. Coexisting Conditions Modifying Phenotypes of Patients with 22q11.2 Deletion Syndrome. Genes 2023, 14, 680. https://0-doi-org.brum.beds.ac.uk/10.3390/genes14030680

AMA Style

Smyk M, Geremek M, Ziemkiewicz K, Gambin T, Kutkowska-Kaźmierczak A, Kowalczyk K, Plaskota I, Wiśniowiecka-Kowalnik B, Bartnik-Głaska M, Niemiec M, et al. Coexisting Conditions Modifying Phenotypes of Patients with 22q11.2 Deletion Syndrome. Genes. 2023; 14(3):680. https://0-doi-org.brum.beds.ac.uk/10.3390/genes14030680

Chicago/Turabian Style

Smyk, Marta, Maciej Geremek, Kamila Ziemkiewicz, Tomasz Gambin, Anna Kutkowska-Kaźmierczak, Katarzyna Kowalczyk, Izabela Plaskota, Barbara Wiśniowiecka-Kowalnik, Magdalena Bartnik-Głaska, Magdalena Niemiec, and et al. 2023. "Coexisting Conditions Modifying Phenotypes of Patients with 22q11.2 Deletion Syndrome" Genes 14, no. 3: 680. https://0-doi-org.brum.beds.ac.uk/10.3390/genes14030680

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