Next Article in Journal
Mechanisms Limiting Renal Tissue Protection and Repair in Glomerulonephritis
Next Article in Special Issue
Epigenetic Analysis of the Dopamine Transporter Gene DAT1 with a Focus on Personality Traits in Athletes
Previous Article in Journal
Impact of Novel Active Layer-by-Layer Edible Coating on the Qualitative and Biochemical Traits of Minimally Processed ‘Annurca Rossa del Sud’ Apple Fruit
Previous Article in Special Issue
Staphylococcal Resistance Patterns, blaZ and SCCmec Cassette Genes in the Nasopharyngeal Microbiota of Pregnant Women
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Chromosome 22q11.2 Deletion Syndrome: A Comprehensive Review of Molecular Genetics in the Context of Multidisciplinary Clinical Approach

by
Aleksandra Szczawińska-Popłonyk
1,*,
Eyal Schwartzmann
2,
Zuzanna Chmara
3,
Antonina Głukowska
3,
Tomasz Krysa
3,
Maksymilian Majchrzycki
3,
Maurycy Olejnicki
3,
Paulina Ostrowska
3 and
Joanna Babik
4
1
Department of Pediatric Pneumonology, Allergy and Clinical Immunology, Institute of Pediatrics, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
2
Medical Student Scientific Society, English Division, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
3
Medical Student Scientific Society, Karol Marcinkowski University of Medical Sciences, 60-572 Poznań, Poland
4
Gynecology and Obstetrics with Pregnancy Pathology Unit, Franciszek Raszeja Municipal Hospital, 60-834 Poznań, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(9), 8317; https://doi.org/10.3390/ijms24098317
Submission received: 14 April 2023 / Revised: 30 April 2023 / Accepted: 3 May 2023 / Published: 5 May 2023
(This article belongs to the Special Issue State-of-the-Art Molecular Genetics and Genomics in Poland 2.0)

Abstract

:
The 22q11.2 deletion syndrome is a multisystemic disorder characterized by a marked variability of phenotypic features, making the diagnosis challenging for clinicians. The wide spectrum of clinical manifestations includes congenital heart defects—most frequently conotruncal cardiac anomalies—thymic hypoplasia and predominating cellular immune deficiency, laryngeal developmental defects, midline anomalies with cleft palate and velar insufficiency, structural airway defects, facial dysmorphism, parathyroid and thyroid gland hormonal dysfunctions, speech delay, developmental delay, and neurocognitive and psychiatric disorders. Significant progress has been made in understanding the complex molecular genetic etiology of 22q11.2 deletion syndrome underpinning the heterogeneity of clinical manifestations. The deletion is caused by chromosomal rearrangements in meiosis and is mediated by non-allelic homologous recombination events between low copy repeats or segmental duplications in the 22q11.2 region. A range of genetic modifiers and environmental factors, as well as the impact of hemizygosity on the remaining allele, contribute to the intricate genotype-phenotype relationships. This comprehensive review has been aimed at highlighting the molecular genetic background of 22q11.2 deletion syndrome in correlation with a clinical multidisciplinary approach.

1. Introduction

The chromosome 22q11.2 deletion syndrome (22q11.2 DS), also known as DiGeorge syndrome (DGS) or velocardiofacial syndrome (VCFS), is a genetic condition resulting from the impaired development of structures originating from the third and fourth pharyngeal pouches in the germinal stage. The clinical features of the syndrome include hypoparathyroidism and hypocalcemia, thymic hypoplasia, conotruncal heart defects, facial dysmorphism, and palatoschisis. The complex phenotype of children affected with 22q11.2 DS may show considerable intersubject variability, and the expanded clinical manifestations comprise craniofacial, neurological, cognitive, behavioral, ocular, speech and hearing, musculoskeletal, and internal organs, as well as airway, gastrointestinal or renal abnormalities [1]. The phenotypes may vary considerably among patients with immunodeficiency and immune dysregulation, including autoimmunity, allergy, and lymphoproliferative sequelae.
The incidence of 22q11.2 DS has been estimated to range from 1:3000 [2] to 1:4000 live births [1], placing it among frequent syndromic diseases. However, the rate of its clinical suspicion is still challenging, and the process of establishing the definitive diagnosis is an odyssey [3] due to the remarkable heterogeneity of clinical phenotypic expressions and overlapping manifestations with other categories of syndromic disorders [4,5,6]. The disease entities, such as, but not limited to, coloboma-heart defects-choanal atresia-retardation of growth and development-ear anomalies (CHARGE) syndrome [7,8], cardio-facio-cutaneous (CFC) syndrome [9,10], and Takenouchi-Kosaki syndrome (TKS) [11,12], require special pediatrician awareness and multidisciplinary care, as their distinctive phenotypes are associated with immunodeficiencies. Furthermore, a common denominator of 22q11.2 DS and other syndromic disorders is an increased susceptibility to recurrent infections, which may not result only from inborn errors of immunity; a multiplicity of developmental anatomical malformations and organ dysfunctions are also important contributing factors. These extra-immune phenotypes in 22q11.2 DS, with neurological, psychomotor, hormonal, circulatory, and respiratory pathophysiological mechanisms, are substantially underpinning the definitive clinical manifestations.
However, there is a paucity of reports on the clinical course of 22q11.2 DS in pediatric patients, and the data are scattered, thereby making them difficult to apply in the all-embracing pediatric practice. In this review, we aimed to establish a comprehensive molecular genetic and clinical approach to 22q11.2 DS addressed to pediatricians and specialists in other fields of medicine who encounter children affected with this syndrome and provide in-depth clinical care. We also sought to determine what is known and what is new in the multidisciplinary genetic and clinical diagnostic process and prophylactic measures, as well as therapeutic modalities in 22q11.2 DS.

2. Pathophysiology

Several clinical disorders make up 22q11.2 DS, such as velocardiofacial (VCF, pharyngeal dysfunction, cardiac anomaly, facial dysmorphism) and DiGeorge (DGS, cardiac anomaly, hypoparathyroidism, thymic hypoplasia) syndromes, a group of disorders described by the acronym CATCH22 (cardiac defect, abnormal facial features, thymic hypoplasia, cleft palate, hypocalcemia). The nomenclature may be confusing due to the remarkable heterogeneity of clinical phenotype and marked intersubject variability of the presenting symptomatology in 22q11.2 DS [13]. A variety of anatomical and functional developmental abnormalities occur in the fetus and multiple conditions appear later in childhood. The syndrome is characterized by haploinsufficiency resulting from a hemizygous deletion in the region 11.2 on the long arm of chromosome 22, meaning that the gene alleles have no homologic counterparts. Interestingly, a deletion on 22q11.2, in combination with a single gene variant on the other allele, can reveal autosomal recessive conditions, such as Bernard-Soulier syndrome with thrombocytopenia and increased megakaryocytes (platelet glycoprotein 1B beta-polypeptide, GP1BB variant) and cerebral dysgenesis (polymicrogyria), neuropathy, ichthyosis, and palmoplantar keratoderma (CEDNIK) syndrome (synaptosomal-associated protein 29 kDa, SNAP29 variant) [14].
The deletion types and sizes in 22q11.2 DS show a high degree of variability due to several low-copy-number repeat sequences (LCR22A, LCR22B, LCR22C, LCR22D, LCR22E, and LCR22F) flanking the deleted region. Most patients (about 90%) show a 2.54 Mb heterozygous deletion comprising four repeats extending from LCR22A to LCR22D and involving approximately 40 genes [15]. The disease-causing genes mapping to the region spanned by LCR22A-LCR22D are, among others, TBX1, proline dehydrogenase PRODH, catechol-O-methyltransferase COMT, cell division cycle 45 CDC45, GP1BB, SNAP29, and DiGeorge critical regions DGCR2, DGCR6, DGCR6L, and DGCR8. A proximal 1.25 Mb deletion comprising LCR22A to LCR22B affects about 5% of individuals with 22q11.2 DS, whereas 2% of them show a deletion spanning the LCR22A to LCR22C region, and the next 5% have a smaller, atypical, nested deletion extending from LCR22B or LCR22C to LCR22D. Distal deletions, flanked by LCR22D-E and LCR22D-F, have been reported less frequently [15,16]. The complexity of 22q11.2 genetics is even more prominent due to the regulatory effect of deleted DGCR6, DGCR6L, and DGCR8 gene alleles and mi-RNAs, such as miR-185, miR-4716, miR-3618, miR-1286, miR-1306, and miR-6816. Several miRs—miR-96-5, miR-451a, and miR-17-92—target TBX1, thereby influencing its expression and, consequently, the clinical phenotype [17,18,19]. The 22q11.2 region encompassing protein-coding genes, long non-coding RNAs, microRNAs, and genes expressed in functional haploinsufficiency in 22q11.2 DS are displayed in Figure 1. Additionally, genome-wide DNA methylation analysis in a group of patients with 22q11.2 DS has demonstrated differentially methylated regions located apart from the chromosome 22, which may involve clinically relevant genes or regulatory elements, thereby determining the variability of phenotypic features. This phenomenon may also shed light on the impact of 22q11.2 haploinsufficiency in altering the genomic methylation level [20].
The de novo occurrence of the syndrome is observable in about 90–95% of affected individuals in all populations, yet autosomal dominant inheritance has also been ascertained in 6–28% of subjects [17]. With its prevalence ranging broadly from 1:2000 to 1:6000 live births [14], 22q11.2 DS is put among frequent syndromic disorders. In seemingly anatomically normal fetuses, 22q11.2 deletion occurs with an approximate frequency of 1:1000, and it reaches 1:100 in fetuses with major structural defects, such as congenital heart disorders [14]. Furthermore, the current estimation of 22q11.2 DS frequency needs to take into consideration the reproductive fitness and increase in survival of children who can pass on the deletion to their offspring, thereby increasing the frequency of its occurrence [21,22,23]. Although 22q11.2 DS is the most common microdeletion in humans, it may be underdiagnosed in diverse populations due to the heterogeneity of clinical phenotypic features, including less recognizable facial dysmorphism, largely depending on the patient’s ethnicity [24]. Thus, the recognition rate of 22q11.2 in affected children of non-European—in particular, African or Asian descent—may be insufficient and the actual prevalence underestimated.

3. The Burden of 22q11.2 in Pediatrics

With its average frequency estimated to be 1:4000 live births and 1:1000 in unselected fetuses [14], 22q.11.2 DS has a global burden and remarkable impact on affected patients, their families, society, and health providers, as well. Whereas the number of patients reported in the medical literature is disproportionately lower than anticipated due to the estimated frequency, important concerns are raised regarding all the newborns awaiting the genetic molecular diagnosis or children who escaped from the timely recognition of this syndrome [25]. The latter group of undiagnosed individuals may embrace all those children who experience a protracted period of the diagnostic odyssey [2]; they are consequently burdened by unfavorable outcomes. Of note, 22q11.2 DS is an important cause of morbidity and mortality across the lifespan, and both major birth defects and complications that develop later in childhood or even adulthood make an early definitive diagnosis of paramount importance. In addition to being the most common cause of syndromic palatal anomalies and velopharyngeal dysfunction [26,27], 22q11.2 DS is the also most common cause of schizophrenia [28,29] and the second most common cause of developmental delay and congenital heart disease after Down syndrome, as well as a more common cause of conotruncal heart disease [30,31]. The occurrence of 22q11.2 deletion is found in about 2% of all subjects with congenital heart disease [32,33], and, thus, the syndrome should always be considered in prenatal settings in fetuses affected with congenital heart developmental abnormalities [34]. Notably, after the surgery for congenital heart disease, the rate of postoperative non-cardiac complications, such as infections or the need for dialysis [35], length of stay in the intensive care unit, and hypocalcemia [36], is higher in children with 22q11.2 DS than in those with no chromosomal defects. Furthermore, severe forms of congenital heart disease are a primary factor contributing to a lower life expectancy in adults with 22q11.2 DS because the probability to survive to the age of 45 is approximately 72% in those individuals [22].

4. Clinical Symptomatology

Craniofacial dysmorphism in 22q11.2 DS is characterized by a multiplicity of facial phenotypic features. Some of these facial characteristics are not easily recognized clinically in children as they are mild in nature, and age is an important determinant of facial morphology. These features include an elongated face, hypertelorism, wide nasal bridge, hooded eyelids, upslanted palpebral fissures, epicanthus, long nose with a bulbous tip, narrow alar base, short philtrum, small mouth, micrognathia, and low-set, posteriorly rotated, small ears. The retrognathic posture of the mandible, inward displacement of the lower part of the face and the ocular region, and an enlarged cranial base angle have been identified as the uppermost dysmorphology features in the cephalometric and spatially dense 3-dimensional analysis [37]. High prevalence of dental features, including tooth abnormalities of their eruption, shape, and number—such as agenesis of permanent dentition or supernumerary teeth, as well as enamel hypomineralization and hypoplasia—are attributable to 22q11 DS [37,38,39].
Among oropharyngeal developmental disorders, palatoschisis is a frequently observable phenotypic feature, occurring in the form of open cleft, submucosal cleft, palatopharyngeal disproportion, and cleft lingula [40,41]. Velopharyngeal insufficiency and hypotonia accompanied by hypernasal speech have been estimated to occur in as many as 30–80% of children with 22q11.2 DS, rendering the surgical intervention particularly challenging due to the complexity of coexisting serious medical conditions [41,42]. Over the past decade, the number of clinical centers of excellence dedicated to the care of 22q11.2 DS patients, with surgeons specialized in velopharyngeal operations, has grown across different regions of the world.
Developmental disorders of the upper airways in 22q11.2 DS include congenital laryngeal anomalies. Laryngeal atresia due to a glottic web or partial laryngeal stenosis, laryngomalacia, laryngeal cleft, and vocal fold abnormalities are characteristic parts of the syndromic features [43,44]. The laryngeal web has been estimated to be the most frequent upper airway anomaly and 40% sensitive for suggesting a diagnosis of 22q11.2 DS [45]. Velopharyngeal and laryngeal anomalies may be accompanied by lower respiratory tract disorders, such as tracheal stenosis, tracheo- and bronchomalacia, short trachea with reduced numbers of tracheal rings, and tracheoesophageal fistula, as well as aberrant tracheal bronchus [46,47]. These structural airway abnormalities may be related to obstruction due to anatomically and functionally reduced patency, recurrent and persistent respiratory tract infections, atelectasis of the lung, and pulmonary edema resulting from congenital heart disease. All these overlapping clinical encumbrances may be significant causes of morbidity and mortality in children with 22q11.2 DS and result in respiratory distress and the need for ventilation support and tracheostomy. Impaired coordination of swallowing and breathing, the risk of aspiration, and nasopharyngeal reflux, as well as the need to provide caloric and nutritional support, necessitate the placement of an enteral percutaneous feeding tube [48].
Congenital heart disease has been recognized in approximately 60–80% of children affected with 22q11.2 DS. The most commonly occurring subset of cardiac anomaly is a conotruncal defect, such as tetralogy of Fallot, pulmonary atresia with ventricular septal defect, truncus arteriosus, interrupted aortic arch type B, conoventricular and/or atrial septal defects, and aortic arch anomalies [32]. The anatomical complexity of congenital heart disease in 22q11.2 DS requires special perioperative care, particularly in children with pulmonary atresia-ventricular septal defect (PA-VSD) and major portopulmonary collateral arteries (MAPCAs), which pose the risk of increased mortality due to vasomotor instability, airway hyperresponsiveness, coexisting airway abnormalities, increased frequency of airway bleeding, and infectious—mainly fungal—complications [49]. The syndrome is therefore associated with unfavorable early perioperative results which, in turn, may be associated with worse cognitive and neuropsychiatric outcomes [50]. Cardiovascular complications are the most common cause of premature death in adults with 22q11.2 DS. Major associated conditions, such as hypocalcemia, thyroid disorders, autoimmune diseases, behavioral problems, and neurodevelopmental disability contribute to the worse fallout [51]. Interestingly, it has been suggested that variance in congenital heart disease, conotruncal-type penetration, in the population of individuals with 22q11.2 DS may be associated with variants in GH22J020947 affecting the expression of CRKL encoding for a cytoplasmic adaptor protein (CRK-like proto-oncogene) involved in growth factor signaling [52]. Moreover, it has been hypothesized that rare copy number variants outside the deleted 22q11.2 region may act as modifiers of the risk of congenital heart disease in 22q11.2 microdeletion-affected children [53].
The complex and variable phenotypes related to 22q11.2 DS also include mild to moderate intellectual disability, which occurs in approximately one-third of affected pediatric patients [54]. In these patients, global reduction in brain volume, with widespread decay in frontal, temporal, parietal, and occipital lobes, atrophy of the cerebellum and hippocampus, as well as polymicrogyria and altered cortical thickness are observable in neuroradiological imaging [55,56]. The syndrome is associated with neurological disorders, such as epilepsy and increased incidence of seizures, which may be provoked by other concomitant conditions and complications, including hypoxia due to congenital heart disease and cardiac surgery, hypocalcemia, thyroid hormonal dysfunction, infections, and fever [57]. Movement disorders in 22q11.2 DS include catatonia and the increased risk of Parkinson’s disease [58].
Noteworthy, 22q11.2 DS is one of the rare examples of a cytogenetic abnormality occurring in conjunction with a psychiatric disease, schizophrenia, autism spectrum disorders (ASD) with some level of social-communication impairment, and attention deficit/hyperreactivity disorder (ADHD), which occur in as many as 20% of affected children and adolescents [54,59]. The spectrum of psychopathology in 22q11.2 DS is complex and comprises anxiety disorders, disruptive, and mood disorders. Importantly, symptoms of psychotic conditions may be misinterpreted and wrongly attributed to cognitive impairment, with low social and communicative skills. Moreover, both low intellectual ability early in life and its subsequent decline, impaired verbal and perceptual reasoning abilities, as well as altered auditory and visual processing from preschool to adolescence, are associated with increased risk of schizophrenia [60,61,62,63]. Ocular abnormalities are frequently encountered in children with 22q11.2 DS in the form of refractive errors, strabismus, amblyopia, and other structural ophthalmological disorders. such as posterior embryotoxon aka Axenfeld-Rieger anomaly, which may contribute to learning difficulties and cognitive impairment [64,65].
Endocrine manifestations are hallmarks of 22q11.2 DS and primarily comprise hypoparathyroidism, thyroid dysfunction, and growth retardation [66,67]. Hypocalcemia is considered a classical feature of the syndrome, resulting from low parathormone serum levels, accompanied by 25(OH)D3 deficiency. In addition to the major effect of parathyroid gland endocrinopathy, hypothyroidism and hypomagnesemia may play a role in hypocalcemia by suppressing parathyroid hormone and, thereby, are further contributing factors to hypocalcemia [68]. The presentation of hypocalcemia is more likely to occur at a young age and may vary considerably, ranging from transient neonatal silent hypocalcemia to hypocalcemic tetany and overt hypoparathyroidism throughout the lifespan. Affected patients may also experience fatigue and paresthesia, as well as more severe manifestations including low seizure threshold and prolongation of the QT interval, which significantly affect both early life neurodevelopment [69] and clinical outcome in cardiac and non-cardiac surgical procedures [70,71,72]. Thyroid dysfunction often presents later, in older childhood or adulthood, and is commonly due to autoimmune thyroiditis. Thyroid autoantibodies, particularly anti-thyroperoxidase antibodies, have been found in up to 5% of children and in approximately 30% of adults affected with 22q11.2 DS [73]. Two clinical disease entities of autoimmune thyroiditis have been reported in children with the syndrome: Hashimoto thyroiditis occurs in 20% of them and is characterized by inhomogeneous thyroid echostructure and progression from normal function to hypothyroidism and, less frequently, Grave’s disease, which presents with overt hyperthyroidism and is observable in 1.4% of affected pediatric patients [67,73,74,75,76,77]. Congenital thyroid gland abnormalities have also been recognized in children with 22q11.2 DS and include an absent thyroid isthmus, retrocarotid and retroesophageal extension, and absence/hypoplasia of the left thyroid lobe. It is noteworthy that developmental thyroid disorders coexist in as many as 71% of children with congenital heart disease, compared to 31% of those with normal thyroid volume, highlighting the role of the TBX1 gene in the formation of cardiac outflow structure and positioning of the thyroid gland [78]. Growth restriction due to a growth hormone deficiency that is consistent from infancy to final height has been reported, and the association between patients’ height and congenital heart disease has been shown in pediatric and young adult patients with 22q11.2 DS [66]. Whereas immune dysregulation is associated with 22q11.2 DS, autoimmune endocrinopathies, such as diabetes mellitus with serum insulin antibodies [79] or adrenal antibodies with normal adrenal function [80], are concomitant clinical features. Metabolic disorders, such as type 2 diabetes [81], hypertriglyceridemia [82], and obesity [83], have been noted in patients with the syndrome, with the high frequency of the latter condition reaching approximately 43% of adult individuals affected with 22q11.2 DS [83].
The summary of multisystemic clinical phenotypic features and their estimated frequency in patients with 22q11.2 deletion syndrome, based on literature reviews, is shown in Table 1 [13,14,32,37,38,39,43,44,46,47,55,56,64,65,66,67,84].

5. Immune Deficiency and Immune Dysregulation

Immunodeficiency is a key feature of 22q11.2 DS and is secondary to thymic aplasia or hypoplasia with subsequent impaired thymocyte development. The third and fourth pharyngeal pouches are a common embryonic precursor for the thymus, parathyroid glands, and conotruncal regions of the heart. In 22q11.2 DS, maldevelopment of these organs is due to impaired migration of the neural crest cell into pouch ectoderm [85]. In the setting of abnormal thymic migration, but with the preservation of residual microscopic nests of thymic epithelial cells, mild to moderate reductions in T cell numbers accompanied by only a mild deficit in T cell function occur in most affected children. However, even in these patients, the T-cell thymic output of recent thymic emigrants, assessed by T-cell receptor excision circles (TRECs) analysis, is very low and decreases with age [86,87]. Full thymic aplasia appears occasionally, in approximately 1% of cases with 22q11.2 DS [86,88]. Interestingly, beyond the TBX1 hemizygosity, other genes, such as CRKL in the affected 22q11.2 region, may also have a gene dosing, modifying effect on the phenotypic expression of the syndrome. CRKL is expressed in neural crest-derived tissues and involves thymic development. The effect of compound heterozygosity for TBX1 and CRKL deletion on clinical features and thymus development is additive [89].
Consequently, a wide spectrum of T-cell alterations is seen in 22q11.2 DS, ranging from near normal to near completely immunodeficient. Mild T cell immunodeficiency may be found in children with apparently hypoplastic thymus because ectopic retropharyngeal thymic tissue may be preserved [90,91]. Furthermore, dynamic changes in immunodeficiency are observable over time, and the direct effect of thymic hypoplasia on T cell counts is most apparent in early infancy [86,87]. They tend to normalize by adulthood in most patients, due to the increased secretion of interleukin (IL)-7 that stimulates the thymic output and peripheral proliferation of T cells [92]. The most common deficits include low total CD3+ T cell percentage and absolute count, as well as low numbers of naive CD4+ T helper and CD8+ T cytotoxic/suppressor cells. The naive T cell compartment shows a progressive decline with patients’ age, leading to a predominantly memory phenotype of T cells at the periphery [93].
Referring to anatomical maldevelopment and dysfunction in generating T cells, the humoral immunodeficiency and B cell abnormalities in children with 22q11.2 DS are secondary to T cell deficits [94]. Low immunoglobulin production, most frequently affecting IgM and occasionally IgG and rendering the need for immunoglobulin replacement therapy, as well as the defective immune response to polysaccharide antigens, have been reported in children with the syndrome [95]. Among the B cell subsets, low switched memory B cells expanding by adulthood, accompanied by decreased somatic hypermutation despite increased follicular T helper cells, have been shown, reflecting a dysregulated B cell compartment and compromised T cell help [96,97,98]. Recurrent respiratory infections, such as adenotonsillitis, otitis media, bronchitis, pneumonia, as well as sepsis [99,100], also occur.
Immune dysregulation in 22q11.2 DS has been foremostly ascribed to T-cell lymphopenia and deficiency in CD3+CD4+CD25++, FOXP3+ regulatory T cells, which play a crucial role in maintaining immune homeostasis and self-tolerance. Reduced thymic output related to an increased naive T cell subset, and subsequent T regulatory cell activation, control the expansion of the T cell compartment. A reduced number of regulatory T cells, with their activated phenotype and loss of suppressive capacity in children with 22q11.2 DS, are key features of deregulated T cell homeostasis [101]. Beyond the T cell compartment, immunophenotype anomalies also encompass peculiar B cell developmental disorders with increased naive B cells and deficit in switched memory B cells [102] which are biomarkers of immune dysregulation in 22q11.2 DS. It has been postulated that the individual patient’s immunophenotype may be influenced by genetic modifiers outside the microdeletion locus which regulatethe expression of TBX1. Rare DNA variants in transcriptional regulators involved in retinoic acid signaling, NCOR2 and EP300, were found to be associated with parameters of the immune functions, such as immunoglobulin levels, lymphocyte response to antigens and mitogens, and flow cytometric lymphocyte compartment. Retinoic acid plays an important role in maintaining immune homeostasis by enhancing the differentiation of regulatory T cells, modulating epithelial and mucosal immune responses, and regulating proinflammatory cytokine activity. Hence, genetic modifiers contributing to the individual’s genetic background and modulating variable penetrance may influence the immune response in 22q11.2 DS [103,104].
Autoimmune disorders have been described in as many as 23% of pediatric patients with the syndrome [105], manifesting as autoimmune thyroid disease, juvenile idiopathic arthritis, autoimmune cytopenia (thrombocytopenia, hemolytic anemia, neutropenia), celiac disease, psoriasis, vitiligo, autoimmune hepatitis, and inflammatory bowel disease [100,105,106,107]. It has also been hypothesized that psychotic disorders, developmental regression, and cognitive impairment in children with 22q11.2 DS may have a causal relationship with autoimmune encephalitis [108].
The peripheral homeostatic expansion of T cells driven by low thymic output and T cell lymphopenia may contribute to Th2-skewed lymphocyte phenotype and atopic manifestations, such as eczema and asthma [109]. In these children with 22q.11.2 DS, the overall frequency of atopic diseases has been estimated to reach 70% and the frequency of asthma to as many as 50% [110]. Coexisting gastroesophageal reflux and sinopulmonary infections may have an impact on its clinical course [111].
The thymus dysfunction and immunophenotypic abnormalities within the T cell compartments in 22q11.2 DS make affected children susceptible to cancerogenic viruses, such as Epstein-Barr virus (EBV) and human papilloma virus (HPV), that might be linked to an increased risk of malignant transformation. It has also been postulated that chronic immune activation of peripherally expanded T cell population in dysfunctional cellular immunity may underpin the predisposition to developing lymphoproliferative disorders and lymphomagenesis [112], as T cell and B cell lymphomas [113,114,115], as well as acute lymphoblastic leukemia [116], have been reported in children with 22q11.2 DS. The spectrum of malignancies in affected pediatric patients also includes solid tumors—namely Wilms tumor—hepatoblastoma, neuroblastoma, thyroid carcinoma [116], pineoblastoma [112], and xanthoastrocytoma [117].

6. Diagnosis

The chromosomal 22q11.2 DS is a clinically highly variable microdeletion syndrome with differently expressed phenotypes, with wide interfamilial and intrafamilial variability in patients sharing the same genetic underpinnings [118]. This is due to both the remarkable complexity of the 22q11.2 region with LCR blocks and the high susceptibility of this region to meiotic errors, as well as the epigenomic and environmental factors influencing the phenotypic variability [119]. Based on functional genomic assessments, it has also been hypothesized that theories on single-gene haploinsufficiency in 22q11.2 DS cannot be supported. In the setting of diminished 22q11.2 gene dosage, shared molecular functions, convergence on cellular processes, and related consequences on the genetic level point to the matrix or multigenic interactions that translate into the multiplicity of phenotypes [120]. All these genetic factors, together with age-related developing symptomatology, contribute to diagnostic challenges in 22q11.2 DS pediatric patients on the diverse individual, family, social, and population levels. The summary of modifying variants influencing the TBX1 penetrance and related phenotypic expression is shown in Table 2 [17,18,19,52,53,103,104,118,120,121,122,123,124].
The diagnosis of 22q11.2 DS has been traditionally based on the recognition of clinical features and cytogenetic testing using the fluorescence in situ hybridization (FISH) technique. FISH is perceived as the golden standard genetic testing method to confirm the diagnosis of microdeletion syndromes. However, poor clinical accuracy, the low confirmatory rate in the screening of suspected microdeletion syndromes, and failure to detect other than the targeted microdeletion are the major drawbacks of this method. An important limitation of this method is a failure to identify atypical and nested deletions because it can recognize deletions in the proximal part of the critical region, including the typical LCR22A-D deletion. FISH is admittedly inexpensive, yet still a highly labor-intensive and time-consuming procedure [125]. Importantly, due to a marked clinical variability from minimal to full manifestation in patients with 22q11.2 DS, precision in defining clinical criteria is important for referring patients to undergo FISH analysis. Since FISH alone cannot provide a reliable diagnosis of 22q11.2 DS, other diagnostic methods have been developed, such as comparative genomic hybridization (CGH), multiplex ligation-dependent probe amplification (MLPA), multiplex quantitative real-time polymerase chain reaction (qPCR), and high-resolution single-nucleotide polymorphism (SNP) microarray analysis [126]. Although the FISH method is still routinely used in laboratories, the MLPA assay has been perceived as an alternative that is superior to the FISH technique as it is less costly, less time-consuming, and laborious, and does not require cell cultures. It has been proposed that a locus-specific approach, using FISH or MLPA assays, could be offered to children strongly meeting clinical and dysmorphology criteria for 22q11.2 DS [126,127]. Even though FISH is a state-of-the-art procedure for patients with the clinical suspicion of 22q11.2 DS, at present, patients are usually diagnosed by indirect whole genome studies [128]. Referring to the regulatory role of the TBX1 gene during development of the heart, thymus, and parathyroid glands, as well as during formation of the palate, teeth, and craniofacial features, there has been growing evidence that TBX1 is a candidate gene for 22q11.2 DS [129,130]. Therefore, in patients with clinically evident disease in whom a deletion of 22q11.2 has not been identified by FISH or microarray tests, TBX1 gene testing is recommended. Genotype first approach has therefore been proposed, and whole genome sequencing as the first-line method in the not-too-distant future, with FISH to be used as a confirmation of patient and family screening results [125].
Newborn screening (NBS) for severe combined immunodeficiency (SCID) is identifying a subset of infants with 22q11.2 DS due to T cell lymphopenia and low TREC numbers, and, hence, it is not a universally reliable detecting method. Therefore, direct NBS for 22q11.2 DS to recognize affected neonates using the genomic approach with multiplex qPCR assay, targeted to the TBX1 gene within the LCR22A-B region and CRKL within the LCR22C-D region, has been elaborated [131].
Whereas postnatal phenotypes have been widely characterized and categorized, prenatal diagnosis of 22q11.2 DS remains challenging due to a low rate of inheritance (10% of cases) and mild unrecognized parental features. Fetal ultrasound imaging may provide information on findings characteristic for 22q11.2 DS, such as polyhydramnios, hypoplasia or aplasia of the fetal thymus, central nervous system anomalies, such as asymmetric ventriculomegaly and dilated cavum septum pellucidum, and, rarely, skeletal anomalies, among others, such as bilateral talipes and anomalous vertebrae [132].
The golden standard method for detecting 22q11.2 microdeletions remains first trimester screening by chromosomal microarray (CMA), which is performed in invasively obtained prenatal samples, such as chorionic villi and amniotic fluid. An important technical advance in prenatal noninvasive screening for 22q11.2 is cell-free DNA testing [133]. Cell-free DNA fragments present in maternal plasma, which derive from both the mother and the embryo as a result of apoptosis of the cytotrophoblast. An external layer of the placenta is used for qualitative and quantitative assays. The fetal fraction is screened for common trisomy and 22q11.2 deletion, as well as for other rare trisomies and microdeletions. Targeted technologies, such as single-nucleotide polymorphism (SNP)-based, digital analysis of selected regions (DANSR), and targeted capture enrichment assay (TCEA) technologies, as well as the genome-wide methodology massively parallel shotgun sequencing (MPSS), are advanced techniques used in cell-free DNA analysis [133,134,135,136,137].

7. Therapeutic Approach

The marked phenotypic variability of 22q11.2 DS is accompanied by a wide scope of immune deficits, ranging from mild to moderate T cell lymphopenia in the partial form of DiGeorge syndrome (pDGS) to profound combined T and B cell immunodeficiency in the complete form (cDGS). In the first case, hypoplastic ectopic thymus or microscopic thymic rests are found and successful spontaneous immunocorrection has been reported, whereas the latter case is characterized by complete athymia [138]. In those most severely immunocompromised children, immune reconstitution may be achieved by thymus transplantation, providing the ability to produce naive T cells showing a broad T cell receptor repertoire. To facilitate the optimal establishment of thymic allograft, stability of comorbidities, such as attaining cardiopulmonary function, upper airway stabilization, appropriate weight gain, and metabolic compensation, are essential to avoid graft failure. The management of concurrent disorders plays a fundamental role in the timing of thymus transplantation, which requires optimal planning and sequencing [139]. Another approach to cDGS is T cell-replete hematopoietic stem cell transplantation; however, due to the absence of the thymus, engraftment of post-thymic T cells may result in poor quality of immune reconstitution [94,140,141]. Although 22q11.2 DS has been perceived as a T cell deficiency, disorders of B cell maturation and reduced numbers and functions in naive, unswitched, and switched memory B cells have also been reported. Humoral immunodeficiency in children with 22q11.2 DS may considerably vary from hypogammaglobulinemia with low all immunoglobulin isotypes and the need to receive immunoglobulin replacement therapy (approximately 6% and 3% of them, respectively) [95] to low serum IgA or IgM and impaired antigen-specific vaccine response in sporadic cases [142,143]. Despite the vast majority of children with 22q11.2 DS having normal serum immunoglobulin levels, due to the variable degree of cellular immunity impairment, they are susceptible to acute and recurrent infections, among others such as sinusitis, otitis media, mastoiditis, pneumonia, urinary tract infections, and viral infections [144,145]. Important questions are then raised about indications for preventive measures against infections in those children who do not qualify for immunoglobulin replacement therapy but present with T-cell lymphopenia. Antibiotic prophylaxis is indicated for children with 22q11.2 DS, first of all in those with low IgA serum levels and panhypogammaglobulinemia presenting with recurrent respiratory tract infections during epidemic season. The prophylactic regimens include daily or alternate-daily use of amoxicillin or azithromycin and co-trimoxazole in children with advanced T-cell lymphopenia posing the risk of Pneumocystis jiroveci infection [136,146,147].
Active immunization in children with 22q11.2 DS requires optimizing to provide vaccination coverage against vaccine-preventable infections. Live vaccine practices with Bacille Calmette-Guerin (BCG), and vaccines against measles-mumps-rubella (MMR), varicella (VAR), and an intranasal live attenuated influenza vaccine (LAIV), are contraindicated in children with this syndrome as T cell lymphopenia makes them susceptible to adverse effects following live immunization (AEFLI). Furthermore, in those children who, due to hypogammaglobulinemia, receive immunoglobulin replacement therapy either intravenously or subcutaneously, live vaccines are inactivated by administered antibodies and thereby are contraindicated. Inactivated vaccines can be safely administered to immunodeficient patients as they do not pose the risk of an uncontrolled spreading of vaccine microorganisms in the patient’s body, and they are not inactivated by supplemented immunoglobulins. However, the immune response to vaccines with antigen-specific antibodies and memory B cell generation may be significantly reduced [148].
However, many individuals with 22q11.2 DS with mild to moderate immunosuppression receive live viral MMR and varicella vaccines despite the known diagnosis and tolerate them well, without serious adverse effects [149,150]. Given the risk of natural infection, the benefits of protection following immunizations with live vaccines outweigh the risks of potential AEFLI. To assess the safety of live attenuated vaccines and evaluate the ability to generate an effective immune response in children with 22q11.2 DS, immunological investigations prior to the administration of live vaccines have been proposed [151]. The recommended immunology workup practices include lymphocyte immunophenotyping with the evaluation of total CD3+ T cells, CD4+ T helper cells, CD8+ T cytotoxic cells, and CD3+CD4+CD45RA+CD31+ recent thymic emigrants, as well as response to mitogen phytohemagglutinin (PHA). Live vaccines can be safely administered in children showing a total T cell count above 0.5 × 109/L, a cytotoxic T cell count above 0.2 × 109/L, and a normal response to mitogen [151].

8. Conclusions

Reports on nationwide studies capturing patients with 22q11.2 DS [89,145,152] stand in contrast to the frequency of the syndrome, which has been estimated to occur in approximately from 1:3000 to 1:4000 births and show a remarkable discrepancy between the estimated frequency and the diagnostic rate in 22q11.2 DS. Whereas clinical symptomatology of the syndrome may be heterogeneous and finding the genetic etiology may be arduous due to complex molecular genetics, the role of genetic modifiers, and epigenetic and environmental factors, as well as the influence of mutations on the remaining genes uncovering rare recessive conditions as shown in Table 3 [14,153,154], the clinical diagnosis may be challenging for clinicians. Increased awareness of pediatricians and specialists in different fields of medicine about the broad spectrum of phenotypic features of 22q11.2 DS they may encounter is therefore indispensable. Comprehensive multidisciplinary care should be provided to patients with 22q11.2 DS by cardiologists, cardiosurgeons, endocrinologists, laryngologists, neurologists, surgeons, and geneticists, under the clinical immunologist’s supervision [155,156,157]. Clinical centers of excellence with multidisciplinary expertise in comprehensive care for patients with 22q11.2 DS provide careful monitoring and timely interventions. Multidisciplinary care is associated with significantly higher guidelines adherence and optimal outcomes in affected individuals [158,159,160].

Author Contributions

A.S.-P.: Conceptualization, methodology, data curation, writing—original draft preparation, review and editing, supervision; E.S.: data curation, writing—original draft preparation; Z.C., A.G., T.K., M.M., M.O. and P.O.: writing—original draft preparation (These six authors equally contributed to this work); J.B.: writing—revision. All authors have read and agreed to the published version of the manuscript.

Funding

No fundings have been secured for this study.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Research data available by authors.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cortés-Martín, J.; Peñuela, N.L.; Sánchez-García, J.C.; Montiel-Troya, M.; Díaz-Rodríguez, L.; Rodríguez-Blanque, R. Deletion Syndrome 22q11.2: A Systematic Review. Children 2022, 9, 1168. [Google Scholar] [CrossRef] [PubMed]
  2. Fomin, A.B.; Pastorino, A.C.; Kim, C.A.; Pereira, A.C.; Carneiro-Sampaio, M.; Jacob, C.M.A. DiGeorge Syndrome: A not so rare disease. Clinics 2010, 65, 865–869. [Google Scholar] [CrossRef] [PubMed]
  3. Palmer, L.D.; Butcher, N.J.; Boot, E.; Hodgkinson, K.A.; Heung, T.; Chow, E.W.C.; Guna, A.; Crowley, T.B.; Zackai, E.; McDonald-McGinn, D.M.; et al. Elucidating the diagnostic odyssey of 22q11.2 deletion syndrome. Am. J. Med. Genet. Part A 2018, 176, 936–944. [Google Scholar] [CrossRef] [PubMed]
  4. Kersseboom, R.; Brooks, A.; Weemaes, C. Educational paper: Syndromic forms of primary immunodeficiency. Eur. J. Pediatr. 2011, 170, 295–308. [Google Scholar] [CrossRef]
  5. Bardou, M.L.D.; Henriques, M.T.; Grumach, A.S. Inborn errors of immunity associated with characteristic phenotypes. J. Pediatr. 2021, 97, S75–S83. [Google Scholar] [CrossRef]
  6. Szczawinska-Poplonyk, A.; Begier, K.; Dorota, A.; Dabrowska, M.; Galecka, D.; Wawrzeniak, K.; Wroblewski, K. Syndromic immunodeficiencies: A pediatrician’s perspective on selected diseases. Allergol. Immunopathol. 2020, 49, 117–136. [Google Scholar] [CrossRef]
  7. Hsu, P.; Ma, A.; Wilson, M.; Williams, G.; Curotta, J.; Munns, C.F.; Mehr, S. CHARGE syndrome: A review. J. Paediatr. Child Health 2014, 50, 504–511. [Google Scholar] [CrossRef]
  8. Trider, C.-L.; Arra-Robar, A.; van Ravenswaaij-Arts, C.; Blake, K. Developing a CHARGE syndrome checklist: Health supervision across the lifespan (from head to toe). Am. J. Med. Genet. Part A 2017, 173, 684–691. [Google Scholar] [CrossRef]
  9. Szczawińska-Popłonyk, A.; Popłonyk, N.; Niedziela, M.; Sowińska-Seidler, A.; Sztromwasser, P.; Jamsheer, A.; Obara-Moszyńska, M. Case report: The cardio-facio-cutaneous syndrome due to a novel germline mutation in MAP2K1: A multifaceted disease with immunodeficiency and short stature. Front. Pediatr. 2022, 10, 990111. [Google Scholar] [CrossRef]
  10. Pierpont, M.E.M.; Magoulas, P.L.; Adi, S.; Kavamura, M.I.; Neri, G.; Noonan, J.; Pierpont, E.I.; Reinker, K.; Roberts, A.E.; Shankar, S.; et al. Cardio-Facio-Cutaneous Syndrome: Clinical Features, Diagnosis, and Management Guidelines. Pediatrics 2014, 134, e1149–e1162. [Google Scholar] [CrossRef]
  11. Bucciol, G.; Pillay, B.; Casas-Martin, J.; Delafontaine, S.; Proesmans, M.; Lorent, N.; Coolen, J.; Tousseyn, T.; Bossuyt, X.; Ma, C.S.; et al. Systemic Inflammation and Myelofibrosis in a Patient with Takenouchi-Kosaki Syndrome due to CDC42 Tyr64Cys Mutation. J. Clin. Immunol. 2020, 40, 567–570. [Google Scholar] [CrossRef]
  12. Martinelli, S.; Krumbach, O.H.; Pantaleoni, F.; Coppola, S.; Amin, E.; Pannone, L.; Nouri, K.; Farina, L.; Dvorsky, R.; Lepri, F.; et al. Functional Dysregulation of CDC42 Causes Diverse Developmental Phenotypes. Am. J. Hum. Genet. 2018, 102, 309–320. [Google Scholar] [CrossRef]
  13. Kobrynski, L.J.; Sullivan, K.E. Velocardiofacial syndrome, DiGeorge syndrome: The chromosome 22q11.2 deletion syndromes. Lancet 2007, 370, 1443–1452. [Google Scholar] [CrossRef]
  14. McDonald-McGinn, D.M.; Sullivan, K.E.; Marino, B.; Philip, N.; Swillen, A.; Vorstman, J.A.S.; Zackai, E.H.; Emanuel, B.S.; Vermeesch, J.R.; Morrow, B.E.; et al. 22q11.2 deletion syndrome. Nat. Rev. Dis. Prim. 2016, 1, 15071. [Google Scholar] [CrossRef]
  15. Gavril, E.-C.; Popescu, R.; Nucă, I.; Ciobanu, C.-G.; Butnariu, L.I.; Rusu, C.; Pânzaru, M.-C. Different Types of Deletions Created by Low-Copy Repeats Sequences Location in 22q11.2 Deletion Syndrome: Genotype–Phenotype Correlation. Genes 2022, 13, 2083. [Google Scholar] [CrossRef]
  16. Burnside, R.D. 22q11.21 Deletion Syndromes: A Review of Proximal, Central, and Distal Deletions and Their Associated Features. Cytogenet. Genome Res. 2015, 146, 89–99. [Google Scholar] [CrossRef]
  17. Funato, N. Craniofacial Phenotypes and Genetics of DiGeorge Syndrome. J. Dev. Biol. 2022, 10, 18. [Google Scholar] [CrossRef]
  18. Gao, S.; Moreno, M.; Eliason, S.; Cao, H.; Li, X.; Yu, W.; Bidlack, F.B.; Margolis, H.C.; Baldini, A.; Amendt, B.A. TBX1 protein interactions and microRNA-96-5p regulation controls cell proliferation during craniofacial and dental development: Implications for 22q11.2 deletion syndrome. Hum. Mol. Genet. 2015, 24, 2330–2348. [Google Scholar] [CrossRef]
  19. Du, Q.; De La Morena, M.T.; Van Oers, N.S.C. The Genetics and Epigenetics of 22q11.2 Deletion Syndrome. Front. Genet. 2020, 10, 1365. [Google Scholar] [CrossRef] [PubMed]
  20. Rooney, K.; Levy, M.A.; Haghshenas, S.; Kerkhof, J.; Rogaia, D.; Tedesco, M.G.; Imperatore, V.; Mencarelli, A.; Squeo, G.M.; Di Venere, E.; et al. Identification of a DNA Methylation Episignature in the 22q11.2 Deletion Syndrome. Int. J. Mol. Sci. 2021, 22, 8611. [Google Scholar] [CrossRef] [PubMed]
  21. Costain, G.; Chow, E.W.C.; Silversides, C.K.; Bassett, A.S. Sex differences in reproductive fitness contribute to preferential maternal transmission of 22q11.2 deletions. J. Med. Genet. 2011, 48, 819–824. [Google Scholar] [CrossRef] [PubMed]
  22. Van, L.; Heung, T.; Graffi, J.; Ng, E.; Malecki, S.; Van Mil, S.; Boot, E.; Corral, M.; Chow, E.W.C.; Hodgkinson, K.A.; et al. All-cause mortality and survival in adults with 22q11.2 deletion syndrome. Anesth. Analg. 2019, 21, 2328–2335. [Google Scholar] [CrossRef] [PubMed]
  23. Palmer, L.D.; McManus, Z.; Heung, T.; McAlpine, G.; Blagojevic, C.; Corral, M.; Bassett, A.S. Reproductive Outcomes in Adults with 22q11.2 Deletion Syndrome. Genes 2022, 13, 2126. [Google Scholar] [CrossRef] [PubMed]
  24. Kruszka, P.; Addissie, Y.A.; McGinn, D.E.; Porras, A.R.; Biggs, E.; Share, M.; Crowley, T.B.; Chung, B.H.Y.; Mok, G.T.K.; Mak, C.C.Y.; et al. 22q11.2 deletion syndrome in diverse populations. Am. J. Med. Genet. Part A 2017, 173, 879–888. [Google Scholar] [CrossRef]
  25. Mc Donald-McGinn, D. 22q11.2 deletion syndrome: A tiny piece leading to a big picture. Nat. Rev. Dis. Prim. 2020, 6, 33. [Google Scholar] [CrossRef]
  26. Jackson, O.; Crowley, T.B.; Sharkus, R.; Smith, R.; Jeong, S.; Solot, C.; McDonald-Mcginn, D. Palatal evaluation and treatment in 22q11.2 deletion syndrome. Am. J. Med. Genet. Part A 2019, 179, 1184–1195. [Google Scholar] [CrossRef]
  27. Seselgyte, R.; Swan, M.C.; Birch, M.J.; Kangesu, L. Velopharyngeal Incompetence in Children With 22q11.2 Deletion Syndrome: Velar and Pharyngeal Dimensions. J. Craniofacial Surg. 2021, 32, 578–580. [Google Scholar] [CrossRef]
  28. Cleynen, I.; Engchuan, W.; Hestand, M.S.; Heung, T.; Holleman, A.M.; Johnston, H.R.; Monfeuga, T.; McDonald-McGinn, D.M.; Gur, R.E.; Morrow, B.E.; et al. Genetic contributors to risk of schizophrenia in the presence of a 22q11.2 deletion. Mol. Psychiatry 2021, 26, 4496–4510. [Google Scholar] [CrossRef]
  29. Van, L.; Boot, E.; Bassett, A.S. Update on the 22q11.2 deletion syndrome and its relevance to schizophrenia. Curr. Opin. Psychiatry 2017, 30, 191–196. [Google Scholar] [CrossRef]
  30. Putotto, C.; Pugnaloni, F.; Unolt, M.; Maiolo, S.; Trezzi, M.; Digilio, M.C.; Cirillo, A.; Limongelli, G.; Marino, B.; Calcagni, G.; et al. 22q11.2 Deletion Syndrome: Impact of Genetics in the Treatment of Conotruncal Heart Defects. Children 2022, 9, 772. [Google Scholar] [CrossRef]
  31. Calcagni, G.; Pugnaloni, F.; Digilio, M.C.; Unolt, M.; Putotto, C.; Niceta, M.; Baban, A.; Piceci Sparascio, F.; Drago, F.; De Luca, A.; et al. Cardiac defects and genetic syndromes: Old incertainities and new insights. Genes 2021, 12, 1047. [Google Scholar] [CrossRef]
  32. Goldmuntz, E. 22q11.2 deletion syndrome and congenital heart disease. Am. J. Med. Genet. Part C Semin. Med. Genet. 2020, 184, 64–72. [Google Scholar] [CrossRef]
  33. Tan, M.; Wang, X.; Liu, H.; Peng, X.; Yang, Y.; Yu, H.; Xu, L.; Li, J.; Cao, H. Genetic Diagnostic Yield and Novel Causal Genes of Congenital Heart Disease. Front. Genet. 2022, 13, 941364. [Google Scholar] [CrossRef]
  34. Petrescu, A.M.; Ruican, D.; Tudorache, S.; Cernea, N.; Dobrescu, M.A.; Iliescu, D.G. Associated chromosomal abnormalities in fetuses diagnosed prenatally with right aortic arch. Curr. Health Sci. J. 2021, 47, 170–176. [Google Scholar]
  35. McDonald, R.; Dodgen, A.; Goyal, S.; Gossett, J.M.; Shinkawa, T.; Uppu, S.C.; Blanco, C.; Garcia, X.; Bhutta, A.T.; Imamura, M.; et al. Impact of 22q11.2 Deletion on the Postoperative Course of Children After Cardiac Surgery. Pediatr. Cardiol. 2013, 34, 341–347. [Google Scholar] [CrossRef]
  36. Cuturilo, G.; Drakulic, D.; Jovanovic, I.; Ilic, S.; Kalanj, J.; Vulicevic, I.; Raus, M.; Skoric, D.; Mijovic, M.; Medjo, B.; et al. The Impact of 22q11.2 Microdeletion on Cardiac Surgery Postoperative Outcome. Pediatr. Cardiol. 2017, 38, 1680–1685. [Google Scholar] [CrossRef]
  37. Lewyllie, A.; Roosenboom, J.; Indencleef, K.; Claes, P.; Swillen, A.; Devriendt, K.; Carels, C.; Pérula, M.C.D.L.; Willems, G.; Hens, G.; et al. A Comprehensive Craniofacial Study of 22q11.2 Deletion Syndrome. J. Dent. Res. 2017, 96, 1386–1391. [Google Scholar] [CrossRef]
  38. AlQarni, M.A.; Alharbi, A.; Merdad, L. Dental management of a patient with 22q11.2 deletion syndrome (22q11.2DS). BMJ Case Rep. 2018, 2018, bcr2018225765. [Google Scholar] [CrossRef] [PubMed]
  39. Wong, D.H.; Rajan, S.; Hallett, K.B.; Manton, D.J. Medical and dental characteristics of children with 22q11.2 deletion syndrome at the Royal Children’s Hospital, Melbourne. Int. J. Paediatr. Dent. 2021, 31, 682–690. [Google Scholar] [CrossRef]
  40. Cardenas-Nieto, D.; Forero-Castro, M.; Esteban-Perez, C.; Martinez-Lozano, J.; Briceno-Balcazar, I. The 22q11.2 microdeletion in pediatric patients with cleft lip, palate, or both and congenital heart disease: A systematic review. J. Pediatr. Genet. 2020, 9, 1–8. [Google Scholar] [CrossRef]
  41. Kirschner, R.E.; Baylis, A.L. Surgical Considerations in 22Q11.2 Deletion Syndrome. Clin. Plast. Surg. 2014, 41, 271–282. [Google Scholar] [CrossRef] [PubMed]
  42. Failla, S.; You, P.; Rajakumar, C.; Dworschak-Stokan, A.; Doyle, P.C.; Husein, M. Characteristics of velopharyngeal dysfunction in 22q11.2 deletion syndrome: A retrospective case-control study. J. Otolaryngol. Head Neck Surg. 2020, 49, 54. [Google Scholar] [CrossRef] [PubMed]
  43. Abe, Y.; Hirade, T.; Koike, D.; Matama, C.; Kato, F. Laryngeal web with 22q11.2 deletion syndrome. Int. J. Pediatr. Adolesc. Med. 2022, 9, 182–184. [Google Scholar] [PubMed]
  44. Komasińska, P.; Szczawińska-Popłonyk, A.; Jończyk-Potoczna, K.; Bręborowicz, A. Congenital atresia of the larynx and esophagus in a girl with 22q11.2 deletion—A case report. Polish J. Pediatr. 2017, 92, 335–341. [Google Scholar]
  45. Hankey, P.B.; Ghulmiyyah, J.; Yeh, H.-W.; Tracy, M.; Arganbright, J. Airway anomalies in patients with 22q11.2 deletion syndrome: A scoping review. Int. J. Pediatr. Otorhinolaryngol. 2022, 163, 111373. [Google Scholar] [CrossRef]
  46. Verheij, E.; Speleman, L.; van der Molen, A.B.M.; Thomeer, H.G. Congenital respiratory tract disorders in 22q11.2 deletion syndrome. Int. J. Pediatr. Otorhinolaryngol. 2018, 104, 1–4. [Google Scholar] [CrossRef]
  47. Huang, R.Y.; Shapiro, N.L. Structural airway anomalies in patients with DiGeorge syndrome: A current review. Am. J. Otolaryngol. 2000, 21, 326–330. [Google Scholar] [CrossRef]
  48. Ebert, B.; Morrell, N.; Zavala, H.; Chinnadurai, S.; Tibesar, R.; Roby, B.B. Percutaneous Enteral Feeding in Patients with 22q11.2 Deletion Syndrome. Cleft Palate-Craniofacial J. 2022, 59, 121–125. [Google Scholar] [CrossRef]
  49. Unolt, M.; Versacci, P.; Anaclerio, S.; Lambiase, C.; Calcagni, G.; Trezzi, M.; Carotti, A.; Crowley, T.B.; Zackai, E.H.; Goldmuntz, E.; et al. Congenital heart diseases and cardiovascular abnormalities in 22q11.2 deletion syndrome: From well-established knowledge to new frontiers. Am. J. Med. Genet. Part A 2018, 176, 2087–2098. [Google Scholar] [CrossRef]
  50. Yi, J.J.; Tang, S.X.; McDonald-McGinn, D.M.; Calkins, M.E.; Whinna, D.A.; Souders, M.C.; Zackai, E.H.; Goldmuntz, E.; Gaynor, J.W.; Gur, R.C.; et al. Contribution of congenital heart disease to neuropsychiatric outcome in school-age children with 22q11.2 deletion syndrome. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 2013, 165, 137–147. [Google Scholar] [CrossRef]
  51. Rayannavar, A.; Katz, L.E.L.; Crowley, T.B.; Lessig, M.; Grand, K.; Goldmuntz, E.; Zackai, E.H.; McDonald-McGinn, N.M. Association of hypocalcemia with congenital heart disease in 22q11.2 deletion syndrome. Am. J. Med. Genet. Part A 2018, 176, 2099–2103. [Google Scholar] [CrossRef]
  52. 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. 2020, 106, 26–40. [Google Scholar] [CrossRef]
  53. 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]
  54. Quach, T.T.; Stratton, H.J.; Khanna, R.; Kolattukudy, P.E.; Honnorat, J.; Meyer, K.; Duchemin, A.-M. Intellectual disability: Dendritic anomalies and emerging genetic perspectives. Acta Neuropathol. 2020, 141, 139–158. [Google Scholar] [CrossRef]
  55. Hopkins, S.E.; Chadehumbe, M.; Crowley, T.B.; Zackai, E.H.; Bilaniuk, L.T.; McDonald-McGinn, D.M. Neurologic challenges in 22q11.2 deletion syndrome. Am. J. Med. Genet. Part A 2018, 176, 2140–2145. [Google Scholar] [CrossRef]
  56. Bagautdinova, J.; Zöller, D.; Schaer, M.; Padula, M.C.; Mancini, V.; Schneider, M.; Eliez, S. Altered cortical thickness development in 22q11.2 deletion syndrome and association with psychotic symptoms. Mol. Psychiatry 2021, 26, 7671–7678. [Google Scholar] [CrossRef]
  57. Mudigoudar, B.; Nune, S.; Fulton, S.; Dayyat, E.; Wheless, J.W. Epilepsy in 22q11.2 Deletion Syndrome: A Case Series and Literature Review. Pediatr. Neurol. 2017, 76, 86–90. [Google Scholar] [CrossRef]
  58. Bayat, M.; Bayat, A. Neurologic manifestations of 22q11.2 deletion syndrome. Neurol Sci. 2022, 43, 1695–1700. [Google Scholar] [CrossRef]
  59. Osley, O.; Evans, N.A.; Fernandes-Carriba, S.; Smearman, E.L.; Rockers, K.; Morrier, M.J.; Evans, D.W.; Coleman, K.; Cubells, J. Examining the overlap between autism spectrum disorders and 22q11.2 deletion syndrome. Int. J. Mol. Sci. 2017, 18, 1071. [Google Scholar] [CrossRef]
  60. Fiksinski, A.M.; Schneider, M.; Zinstok, J.; Baribeau, D.; Chawner, S.J.R.A.; Vorstman, J.A.S. Neurodevelomental trajectories and psychiatric morbidity: Lessons learned from the 22q11.2 deletion syndrome. Curr. Psychiatry Rep. 2021, 23, 13. [Google Scholar] [CrossRef]
  61. Swillen, A. The importance of understanding cognitive trajectories: The case of 22q11.2 deletion syndrome. Curr. Opin. Psychiatry. 2016, 29, 133–137. [Google Scholar] [CrossRef] [PubMed]
  62. Biria, M.; Tomescu, M.I.; Custo, A.; Cantonas, L.M.; Song, K.-W.; Schneider, M.; Murray, M.M.; Eliez, S.; Michel, C.M.; Rihs, T.A. Visual processing deficits in 22q11.2 Deletion Syndrome. NeuroImage Clin. 2017, 17, 976–986. [Google Scholar] [CrossRef] [PubMed]
  63. Francisco, A.A.; Foxe, J.J.; Horsthuis, D.J.; DeMaio, D.; Molholm, S. Assessing auditory processing endophenotypes associated with Schizophrenia in individuals with 22q11.2 deletion syndrome. Transl. Psychiatry 2020, 10, 85. [Google Scholar] [CrossRef] [PubMed]
  64. Casteels, I.; Casaer, P.; Gewillig, M.; Swillen, A.; Devriendt, K. Ocular findings in children with a microdeletion in chromosome 22q11.2. Eur. J. Pediatr. 2008, 167, 751–755. [Google Scholar] [CrossRef]
  65. Gokturk, B.; Topcu-Yilmaz, P.; Bozkurt, B.; Yildirim, M.S.; Guner, S.N.; Sayar, E.H.; Reisli, I. Ocular Findings in Children with 22q11.2 Deletion Syndrome. J. Pediatr. Ophthalmol. Strabismus 2016, 53, 218–222. [Google Scholar] [CrossRef]
  66. Levy-Shraga, Y.; Gothelf, D.; Goichberg, Z.; Katz, U.; Somech, R.; Pinhas-Hamiel, O.; Modan-Moses, D. Growth characteristics and endocrine abnormalities in 22q11.2 deletion syndrome. Am. J. Med. Genet. Part A 2017, 173, 1301–1308. [Google Scholar] [CrossRef]
  67. Choi, J.-H.; Shin, Y.-L.; Kim, G.-H.; Seo, E.-J.; Kim, Y.; Park, I.-S.; Yoo, H.-W. Endocrine Manifestations of Chromosome 22q11.2 Microdeletion Syndrome. Horm. Res. Paediatr. 2005, 63, 294–299. [Google Scholar] [CrossRef]
  68. Cheung, E.N.M.; George, S.R.; Costain, G.A.; Andrade, D.M.; Chow, E.W.C.; Silversides, C.K.; Bassett, A.S. Prevalence of hypocalcemia and its associated features in 22q11.2 deletion syndrome. Clin. Endocrinol. 2014, 81, 190–196. [Google Scholar] [CrossRef]
  69. Cheung, E.N.; George, S.R.; Andrade, D.M.; Chow, E.W.; Silversides, C.K.; Bassett, A.S. Neonatal hypocalcemia, neonatal seizures and intellectual disability in 22q11.2 deletion syndrome. Genet. Med. 2014, 16, 40–44. [Google Scholar] [CrossRef]
  70. Arganbright, J.M.; Tracy, M.; Feldt, M.; Narayanan, S.; Mahadev, A.; Noel-MacDonnell, J. Postoperative Hypocalcemia following Non-Cardiac Surgical Procedures in Children with 22q11.2 Deletion Syndrome. Genes 2022, 13, 1905. [Google Scholar] [CrossRef]
  71. Yang, C.; Ge, J.; Zhang, R.; Chen, C.; Yi, L.; Shen, L. The Correlation between Severity of Postoperative Hypocalcemia and Perioperative Mortality in Chromosome 22q11.2 Microdeletion (22q11DS) Patient After Cardiac-Correction Surgery: A Retrospective Analysis. Heart Surg. Forum 2022, 23, E549–E554. [Google Scholar] [CrossRef]
  72. Fujii, S.; Nakanishi, T. Clinical manifestations and frequency of hypocalcemia in 22q11.2 deletion syndrome. Pediatr. Int. 2015, 57, 1086–1089. [Google Scholar] [CrossRef]
  73. Kyritsi, E.M.; Kanaka-Gantenbaum, C. Autoimmune thyroide disease in specific genetic syndromes in childhood and adolescence. Front. Endocrinol. 2020, 11, 543. [Google Scholar] [CrossRef]
  74. Ricci, S.; Sarli, W.A.; Lodi, L.; Canessa, C.; Lippi, F.; Azzari, C.; Stagi, S. Characterization of autoimmune thyroid disease in cohort of 73 pediatric patients affected by 22q11.2 deletion syndrome: Longitudinal single-centre study. Genes 2022, 13, 1552. [Google Scholar] [CrossRef]
  75. Shugar, A.L.; Shapiro, J.M.; Cytrynbaum, C.; Hedges, S.; Weksberg, R.; Fishman, L. An increased prevalence of thyroid disease in children with 22q11.2 deletion syndrome. Am. J. Med. Genet. Part A 2015, 167, 1560–1564. [Google Scholar] [CrossRef]
  76. Ueda, Y.; Uraki, S.; Inaba, H.; Nakashima, S.; Ariyasu, H.; Iwakura, H.; Ota, T.; Furuta, H.; Nishi, M.; Akamizu, T. Grave’s disease in pediatric and elederly patients with 22q11.2 deletion syndrome. Intern. Med. 2017, 56, 1169–1173. [Google Scholar] [CrossRef]
  77. Brown, J.; Datta, V.; Browning, M.; Swift, P. Graves’ Disease in DiGeorge Syndrome: Patient Report with a Review of Endocrine Autoimmunity Associated with 22q11.2 Deletion. J. Pediatr. Endocrinol. Metab. 2004, 17, 1575–1580. [Google Scholar] [CrossRef]
  78. Stagi, S.; Lapi, E.; Gambineri, E.; Salti, R.; Genuardi, M.; Colarusso, G.; Conti, C.; Jenuso, R.; Chiarelli, F.; Azzari, C.; et al. Thyroid function and morphology in subjects with microdeletion of chromosome 22q11 (del(22)(q11)). Clin. Endocrinol. 2009, 72, 839–844. [Google Scholar] [CrossRef]
  79. Elder, D.A.; Kaiser-Rogers, K.; Aylsworth, A.S.; Calikoglu, A.S. Type I diabetes mellitus in a patient with 22q11.2 deletion syndrome. Am. J. Med. Genet. 2001, 101, 17–19. [Google Scholar] [CrossRef]
  80. Lima, K.; Abrahamsen, T.G.; Wolff, A.B.; Husebye, E.; Alimohammadi, M.; Kämpe, O.; Følling, I. Hypoparathyroidism and autoimmunity in the 22q11.2 deletion syndrome. Eur. J. Endocrinol. 2011, 165, 345–352. [Google Scholar] [CrossRef]
  81. Van, L.; Heung, T.; Malecki, S.L.; Fenn, C.; Tyrer, A.; Sanches, R.; Chow, E.W.; Boot, E.; Corral, M.; Dash, S.; et al. 22q11.2 microdeletion and risk for type 2 diabetes. EClinicalMedicine 2020, 26, 100528. [Google Scholar] [CrossRef] [PubMed]
  82. Blagojevic, C.; Heung, T.; Malecki, S.; Ying, S.; Cancelliere, S.; Hegele, R.A.; Bassett, A.S. Hypertriglyceridemia in young adults with a 22q11.2 microdeletion. Eur. J. Endocrinol. 2022, 187, 91–99. [Google Scholar] [CrossRef] [PubMed]
  83. Voll, S.L.; Boot, E.; Butcher, N.J.; Cooper, S.; Heung, T.; Chow, E.W.; Silversides, C.K.; Bassett, A.S. Obesity in adults with 22q11.2 deletion syndrome. Anesth. Analg. 2017, 19, 204–208. [Google Scholar] [CrossRef] [PubMed]
  84. Sullivan, K.E. Chromosome 22q11.2 deletion syndrome and DiGeorge syndrome. Immunol. Rev. 2019, 287, 186–201. [Google Scholar] [CrossRef]
  85. Gennery, A.R. Immunological aspects of 22q11.2 deletion syndrome. Cell. Mol. Life Sci. 2012, 69, 17–27. [Google Scholar] [CrossRef]
  86. Dar, N.; Gothelf, D.; Korn, D.; Frisch, A.; Weizman, A.; Michaelovsky, E.; Carmel, M.; Yeshayahu, Y.; Dubnov-Raz, G.; Pessach, I.M.; et al. Thymic and bone marrow output in individuals with 22q11.2 deletion syndrome. Pediatr. Res. 2015, 77, 579–585. [Google Scholar] [CrossRef]
  87. Gul, K.A.; Øverland, T.; Osnes, L.; Baumbusch, L.O.; Pettersen, R.D.; Lima, K.; Abrahamsen, T.G. Neonatal Levels of T-cell Receptor Excision Circles (TREC) in Patients with 22q11.2 Deletion Syndrome and Later Disease Features. J. Clin. Immunol. 2015, 35, 408–415. [Google Scholar] [CrossRef]
  88. Collins, C.; Sharpe, E.; Silber, A.; Kulke, S.; Hsieh, E.W.Y. Congenital Athymia: Genetic Etiologies, Clinical Manifestations, Diagnosis, and Treatment. J. Clin. Immunol. 2021, 41, 881–895. [Google Scholar] [CrossRef]
  89. Guris, D.L.; Duester, G.; Papaioannou, V.; Imamoto, A. Dose-Dependent Interaction of Tbx1 and Crkl and Locally Aberrant RA Signaling in a Model of del22q11 Syndrome. Dev. Cell 2006, 10, 81–92. [Google Scholar] [CrossRef]
  90. Shah, S.S.; Lai, S.Y.; Ruchelli, E.; Kazahaya, K.; Mahboubi, S. Retropharyngeal Aberrant Thymus. Pediatrics 2001, 108, e94. [Google Scholar] [CrossRef]
  91. Grudzień, K.; Kuzaj, J.; Dębicka, M.; Kwiatkowski, S.; Milczarek, O. Retropharyngeal Ectopic Thymus in a Pediatric Patient With 22q11.2 Deletion Syndrome. Cureus 2023, 15, e33350. [Google Scholar] [CrossRef]
  92. Crowley, B.; Ruffner, M.; McGinn, D.M.M.; Sullivan, K.E. Variable immune deficiency related to deletion size in chromosome 22q11.2 deletion syndrome. Am. J. Med. Genet. Part A 2018, 176, 2082–2086. [Google Scholar] [CrossRef]
  93. Giardino, G.; Borzacchiello, C.; De Luca, M.; Romano, R.; Prencipe, R.; Cirillo, E.; Pignata, C. T-Cell Immunodeficiencies with Congenital Alterations of Thymic Development: Genes Implicated and Differential Immunological and Clinical Features. Front. Immunol. 2020, 11, 1837. [Google Scholar] [CrossRef]
  94. Davies, E.G. Immunodeficiency in DiGeorge Syndrome and Options for Treating Cases with Complete Athymia. Front. Immunol. 2013, 4, 322. [Google Scholar] [CrossRef]
  95. Patel, K.; Akhter, J.; Kobrynski, L.; Gathman, B.; Davis, O.; Sullivan, K.E. Immunoglobulin Deficiencies: The B-Lymphocyte Side of DiGeorge Syndrome. J. Pediatr. 2012, 161, 950–953.e1. [Google Scholar] [CrossRef]
  96. Zemble, R.; Prak, E.L.; McDonald, K.; McDonald-McGinn, D.; Zackai, E.; Sullivan, K. Secondary immunologic consequences in chromosome 22q11.2 deletion syndrome (DiGeorge syndrome/velocardiofacial syndrome). Clin. Immunol. 2010, 136, 409–418. [Google Scholar] [CrossRef]
  97. Derfalvi, B.; Maurer, K.; McGinn, D.M.M.; Zackai, E.; Meng, W.; Prak, E.T.L.; Sullivan, K.E. B cell development in chromosome 22q11.2 deletion syndrome. Clin. Immunol. 2016, 163, 1–9. [Google Scholar] [CrossRef]
  98. Montin, D.; Marolda, A.; Licciardi, F.; Robasto, F.; DI Cesare, S.; Ricotti, E.; Ferro, F.; Scaioli, G.; Giancotta, C.; Amodio, D.; et al. Immunophenotype Anomalies Predict the Development of Autoimmune Cytopenia in 22q11.2 Deletion Syndrome. J. Allergy Clin. Immunol. Pract. 2019, 7, 2369–2376. [Google Scholar] [CrossRef]
  99. Wahrmann, S.; Kainulainen, L.; Kytö, V.; Lempainen, J. Childhood manifestations of 22q11.2 deletion syndrome: A Finnish nationwide register-based cohort study. Acta Paediatr. 2023; online. [Google Scholar] [CrossRef]
  100. Di Cesare, S.; Puliafito, P.; Ariganello, P.; Marcovecchio, G.E.; Mandolesi, M.; Capolino, S.; Digilio, M.C.; Aiuti, A.; Rossi, P.; Cancrini, C. Autoimmunity and regulatory T cells in 22q11.2 deletion syndrome patients. Pediatr. Allergy Immunol. 2015, 26, 578–594. [Google Scholar] [CrossRef]
  101. Ferrando-Martínez, S.; Lorente, R.; Gurbindo, D.; De José, M.I.; Leal, M.; Muñoz-Fernández, M.A.; Correa-Rocha, R. Low Thymic Output, Peripheral Homeostasis Deregulation, and Hastened Regulatory T Cells Differentiation in Children with 22q11.2 Deletion Syndrome. J. Pediatr. 2014, 164, 882–889. [Google Scholar] [CrossRef] [PubMed]
  102. Framme, J.L.; Lundqvist, C.; Lundell, A.C.; van Schouwenburg, P.A.; Lemarquis, A.L.; Thörn, K.; Lindgren, S.; Gudmundsdottir, J.; Lundberg, V.; Degerman, S.; et al. Long-term follow-up of newborns with 22q11 deletion syndrome and low TRECs. J. Clin. Immunol. 2022, 42, 618–633. [Google Scholar] [CrossRef] [PubMed]
  103. Pinnaro, C.T.; Henry, T.; Major, H.J.; Parida, M.; DesJardin, L.E.; Manak, J.R.; Darbro, B.W. Candidate modifier genes for immune function in 22q11.2 deletion syndrome. Mol. Genet. Genom. Med. 2019, 8, e1057. [Google Scholar] [CrossRef] [PubMed]
  104. Oliveira, L.D.M.; Teixeira, F.M.E.; Sato, M.N. Impact of Retinoic Acid on Immune Cells and Inflammatory Diseases. Mediat. Inflamm. 2018, 2018, 3067126. [Google Scholar] [CrossRef]
  105. Cancrini, C.; Puliafito, P.; Digilio, M.C.; Soresina, A.; Martino, S.; Rondelli, R.; Consolini, R.; Ruga, E.M.; Cardinale, F.; Finocchi, A.; et al. Clinical Features and Follow-Up in Patients with 22q11.2 Deletion Syndrome. J. Pediatr. 2014, 164, 1475–1480.e2. [Google Scholar] [CrossRef]
  106. Deshpande, D.R.; Demirdag, Y.Y.; Marsh, R.A.; Sullivan, K.E.; Orange, J.S.; The USIDNET Consortium. Relationship between Severity of T Cell Lymphopenia and Immune Dysregulation in Patients with DiGeorge Syndrome (22q11.2 Deletions and/or Related TBX1 Mutations): A USIDNET Study. J. Clin. Immunol. 2021, 41, 29–37. [Google Scholar] [CrossRef]
  107. Mahé, P.; Nagot, N.; Portales, P.; Lozano, C.; Vincent, T.; Sarda, P.; Perez, M.; Amedro, P.; Marin, G.; Jeziorski, E. Risk factors of clinical dysimmune manifestations in a cohort of 86 children with 22q11.2 deletion syndrome: A retrospective study in France. Am. J. Med. Genet. Part A 2019, 179, 2207–2213. [Google Scholar] [CrossRef]
  108. Ciano-Petersen, N.L.; Hamad-Cueto, O.; Drissi-Reyes, H.; Dona-Diaz, A.; Garcia-Martin, G. Case report: Autoimmune psychosis in 22q11.2 deletion syndrome. Front. Immunol. 2021, 12, 708625. [Google Scholar] [CrossRef]
  109. Jesenak, M.; Zelieskova, M.; Repko, M.; Banovcin, P. Successful treatment of severe allergic asthma with omalizumab in a girl with DiGeorge syndrome. Central Eur. J. Immunol. 2020, 45, 361–363. [Google Scholar] [CrossRef]
  110. Morsheimer, M.; Brown Whitehorn, T.F.; Heimall, J.; Sullivan, K.E. The immune deficiency of chromosome 22q11.2 deletion syndrome. Am. J. Med. Genet. Part A 2017, 173, 2366–2372. [Google Scholar] [CrossRef]
  111. Maggadottir, S.M.; Sullivan, K.E. The Diverse Clinical Features of Chromosome 22q11.2 Deletion Syndrome (DiGeorge Syndrome). J. Allergy Clin. Immunol. Pract. 2013, 1, 589–594. [Google Scholar] [CrossRef]
  112. Stevens, T.; Van Der Werff Ten Bosch, J.; De Rademaeker, M.; Van Den Bogaert, A.; Akker, M.V.D. Risk of malignancy in 22q11.2 deletion syndrome. Clin. Case Rep. 2017, 5, 486–490. [Google Scholar] [CrossRef]
  113. Itoh, S.; Ohno, T.; Kakizaki, S.; Ichinohasama, R. Epstein-Barr virus-positive T-cell lymphoma cells having chromosome 22q11.2 deletion: An autopsy report of DiGeorge syndrome. Hum. Patol. 2011, 42, 2037–2041. [Google Scholar] [CrossRef]
  114. Hong, R.; Shen, V.; Rooney, C.; Hughes, D.P.; Smith, C.; Comoli, P.; Zhang, L. Correction od DiGeorge anomaly with EBV-induced lymphoma by transplantation of organ-cultured thymus and Epstein Barr-specific cytotixic T lymphocytes. Clin. Immunol. 2001, 91, 54–61. [Google Scholar] [CrossRef]
  115. Pongpruttipan, T.; Cook, J.R.; Reyes-Mugica, M.; Spahr, E.; Swerdlow, S.H. Pulmonary extranodal marginal zone lymphoma of mucosa-associated lymphoid tissue associated with granulomatous inflammation in a child with chromosome 22q11.2 deletion syndrome (DiGeorge syndrome). J. Pediatr. 2012, 161, 954–958. [Google Scholar] [CrossRef]
  116. McDonald-McGinn, D.M.; Reilly, A.; Wallgren-Pettersson, C.; Hoyme, H.E.; Yang, S.P.; Adam, M.P.; Zackai, E.H.; Sullivan, K.E. Malignancy in chromosome 22q11.2 deletion syndrome (DiGeorge syndrome/velocardiofacial syndrome). Am. J. Med. Genet. Part A 2006, 140, 906–909. [Google Scholar] [CrossRef]
  117. Murray, J.C.; Donahue, D.J.; Malik, S.I.; Dzurik, Y.B.; Braly, E.Z.; Dougherty, M.J.; Eaton, K.W.; Biegel, J.A. Temporal lobe pleomorphic xanthoastrocytoma and acquired BRAF mutation in an adolescent with the constitutional 22q11.2 deletion syndrome. J. Neuro-Oncol. 2011, 102, 509–514. [Google Scholar] [CrossRef]
  118. Digilio, M.C.; Angioni, A.; De Santis, M.; Lombardo, A.; Giannotti, A.; Dallapiccola, B.; Marino, B. Spectrum of clinical variability in familial deletion 22q11.2: From full manifestation to extremely mild clinical anomalies. Clin. Genet. 2003, 63, 308–313. [Google Scholar] [CrossRef]
  119. Karbarz, M. Consequences of 22q11.2 microdeletion on the genome, individual and population level. Genes 2020, 11, 977. [Google Scholar] [CrossRef]
  120. Motahari, Z.; Moody, S.A.; Maynard, T.M.; LaMantia, A.S. In the line-up: Deleted genes associated with DiGeorge/22q11.2 deletion syndrome: Are they all suspects? J. Neurodev. Disord. 2019, 11, 7. [Google Scholar] [CrossRef]
  121. Giacomelli, M.; Kumar, R.; Soresina, A.; Tamassia, N.; Lorenzini, T.; Moratto, D.; Gasperini, S.; Cassatella, M.; Plebani, A.; Lougaris, V.; et al. Reduction of CRKL expression in patients with partial DiGeorege syndrome is associated with impairment of T cell functions. J. Allergy Clin. Immunol. 2016, 138, 229–240. [Google Scholar] [CrossRef] [PubMed]
  122. Guo, T.; Chung, J.H.; Wang, T.; McDonald-McGinn, D.M.; Kates, W.R.; Hawuła, W.; Coleman, K.; Zackai, E.; Emanuel, B.S.; Morrow, B.E. Histone Modifier Genes Alter Conotruncal Heart Phenotypes in 22q11.2 Deletion Syndrome. Am. J. Hum. Genet. 2015, 97, 869–877. [Google Scholar] [CrossRef] [PubMed]
  123. León, L.E.; Benavides, F.; Espinoza, K.; Vial, C.; Alvarez, P.; Palomares, M.; Lay-Son, G.; Miranda, M.; Repetto, G.M. Partial microduplication in the histone acetyltransferase complex member KANSL1 is associated with congenital heart defects in 22q11.2 microdeletion syndrome patients. Sci. Rep. 2017, 7, 1795. [Google Scholar] [CrossRef] [PubMed]
  124. 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]
  125. Halder, A.; Jain, M.; Chaudhary, I.; Gupta, N.; Kabra, M. Fluorescence in situ hybridization (FISH) using non-commercial probes in the diagnosis of clically suspected microdeletion syndromes. Indian J. Med. Res. 2013, 138, 135–142. [Google Scholar]
  126. Maran, S.; Faten, S.A.; Lim, S.H.E.; Lai, K.S.; Ibrahim, W.P.W.; Ankathil, R.; Gan, S.H.; Tan, H.L. Screening of 22q11.2 DS using multiplex ligation as alternative diagnostic method. Biomed. Res. Int. 2020, 2020, 6945730. [Google Scholar] [CrossRef]
  127. 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]
  128. Koczkowska, M.; Wierzba, J.; Śmigiel, R.; Sąsiadek, M.; Cabała, M.; Ślężak, R.; Iliszko, M.; Kardaś, I.; Limon, J.; Lipska-Ziętkiewicz, B.S. Genomic findings in patients with clinical suspicion of 22q11.2 deletion syndrome. J. Appl. Genet. 2017, 58, 93–98. [Google Scholar] [CrossRef]
  129. Gao, S.; Li, X.; Amendt, B.A. Understanding the Role of Tbx1 as a Candidate Gene for 22q11.2 Deletion Syndrome. Curr. Allergy Asthma Rep. 2013, 13, 613–621. [Google Scholar] [CrossRef]
  130. Yagi, H.; Furutani, Y.; Hamada, H.; Sasaki, T.; Asakawa, S.; Minoshima, S.; Ichida, F.; Joo, K.; Kimura, M.; Imamura, S.-I.; et al. Role of TBX1 in human del22q11.2 syndrome. Lancet 2003, 362, 1366–1373. [Google Scholar] [CrossRef]
  131. Barry, J.C.; Crowley, T.B.; Jyonouchi, S.; Heimall, J.; Zackai, E.H.; Sullivan, K.E.; McDonald-McGinn, D.M. Identification of 22q11.2 Deletion Syndrome via Newborn Screening for Severe Combined Immunodeficiency. J. Clin. Immunol. 2017, 37, 476–485. [Google Scholar] [CrossRef]
  132. Schindewolf, E.; Khalek, N.; Johnson, M.P.; Gebb, J.; Coleman, B.; Crowley, T.B.; Zackai, E.H.; McDonald-McGinn, N.M.; Moldenhauer, J.S. Expanding the fetal phenotype: Prenatal sonographic findings and perinatal outcomes in a cohort of patients with a confirmed 22q11.2 deletion syndrome. Am. J. Med. Genet. Part A 2018, 176, 1735–1741. [Google Scholar] [CrossRef]
  133. Grati, F.R.; Gross, S.J. Noninvasive screening by cell-free DNA for 22q11.2 deletion: Benefits, limitations, and challenges. Prenat. Diagn. 2019, 39, 70–80. [Google Scholar] [CrossRef]
  134. Bevilacqua, E.; Jani, C.C.; Chaoui, R.; Suk, E.K.A.; Palma-Dias, R.; Ko, T.M.; Warsof, S.; Stokowski, R.; Jones, K.J.; Grati, F.R.; et al. Performance of targeted cell-free DNA prenatal test for 22q11.2 deletion in a large clinical cohort. Ultrasound Obstet. Gynecol. 2021, 58, 597–602. [Google Scholar] [CrossRef]
  135. Dar, P.; Jacobsson, B.; Clifton, R.; Egbert, M.; Malone, F.; Wapner, R.J.; Roman, A.S.; Khalil, A.; Faro, R.; Madankumar, R.; et al. Cell-free DNA screening for prenatal detection of 22q11.2 deletion syndrome. Am. J. Obstet. Gynecol. 2022, 227, 79.e1–79.e11. [Google Scholar] [CrossRef]
  136. Kagan, K.O.; Hoopmann, M.; Pfaff, T.; Prodan, N.; Wagner, P.; Schmid, M.; Dufke, A.; Mau-Holzmann, U.; Brucker, S.; Marcato, L.; et al. First Trimester Screening for Common Trisomies and Microdeletion 22q11.2 Syndrome Using Cell-Free DNA: A Prospective Clinical Study. Fetal Diagn. Ther. 2020, 47, 841–852. [Google Scholar] [CrossRef]
  137. Blagowidow, N.; Nowakowska, B.; Schindewolf, E.; Grati, F.R.; Putotto, C.; Breckpot, J.; Swillen, A.; Crowley, T.B.; Loo, J.C.Y.; Lairson, L.A.; et al. Prenatal Screening and Diagnostic Considerations for 22q11.2 Microdeletions. Genes 2023, 14, 160. [Google Scholar] [CrossRef]
  138. Biggs, S.E.; Gilchrist, B.; May, K.R. Chromosome 22q11.2 Deletion (DiGeorge Syndrome): Immunologic Features, Diagnosis, and Management. Curr. Allergy Asthma Rep. 2023, 23, 213–222. [Google Scholar] [CrossRef]
  139. Howley, E.; Davies, E.G.; Kreins, A.Y. Congenital athtmia: Unmet needs and practical guidance. Ther. Clin. Risk Manag. 2023, 19, 239–254. [Google Scholar] [CrossRef]
  140. Kreins, A.Y.; Bonfanti, P.; Davies, E.G. Current and future therapeutic approaches for thymic stomal cell defects. Front. Immunol. 2021, 12, 655354. [Google Scholar] [CrossRef]
  141. Davies, E.G.; Cheung, M.; Gilmour, K.; Maimaris, J.; Curry, J.; Furmanski, A.; Sebire, N.; Halliday, N.; Mengrelis, K.; Adams, S.; et al. Thymus transplantation for complete DiGeorge syndrome: European experience. J. Allergy Clin. Immunol. 2017, 140, 1660–1670.e16. [Google Scholar] [CrossRef] [PubMed]
  142. Caka, C.; Cimen, O.; Kahyaoglu Tezcan, I.; Cagdas, D. Selective IgM deficiency: Follow-up and outcome. Pediatr. Allergy Immunol. 2021, 32, 1327–1334. [Google Scholar] [CrossRef]
  143. Kung, S.-J.; Gripp, K.W.; Stephan, M.J.; Fairchok, M.P.; McGeady, S.J. Selective IgM deficiency and 22q11.2 deletion syndrome. Ann. Allergy Asthma Immunol. 2007, 99, 87–92. [Google Scholar] [CrossRef] [PubMed]
  144. Ozen, S.; Akcal, O.; Taskirdi, I.; Haci, I.A.; Karaca, N.E.; Gulez, N.; Aksu, G.; Genel, F.; Kutukculer, N. 22q11.2 deletion syndrome: 20 years of experience from two pediatric immunology units and review of clues for diagnosis and disease management. Allergol. Immunopathol. 2021, 49, 95–100. [Google Scholar] [CrossRef] [PubMed]
  145. Nissan, E.; Katz, U.; Levy-Shraga, Y.; Frizinsky, S.; Carmel, E.; Gothelf, D.; Somech, R. Clinical Features in a Large Cohort of Patients With 22q11.2 Deletion Syndrome. J. Pediatr. 2021, 238, 215–220.e5. [Google Scholar] [CrossRef]
  146. Habel, A.; Herriot, R.; Kumararatne, D.; Allgrove, J.; Baker, K.; Baxendale, H.; Bu’lock, F.; Firth, H.; Gennery, A.; Holland, A.; et al. Towards a safety net for management of 22q11.2 deletion syndrome: Guidelines for our times. Eur. J. Pediatr. 2014, 173, 757–765. [Google Scholar] [CrossRef]
  147. Kuruvilla, M.; de la Morena, M.T. Antibiotic prophylaxis in primary immune deficiency disorders. J. Allergy Clin. Immunol. Pract. 2013, 1, 573–582. [Google Scholar] [CrossRef]
  148. Szczawinska-Poplonyk, A.; Breborowicz, A.; Samara, H.; Ossowska, L.; Dworacki, G. Impaired Antigen-Specific Immune Response to Vaccines in Children with Antibody Production Defects. Clin. Vaccine Immunol. 2015, 22, 875–882. [Google Scholar] [CrossRef]
  149. Hofstetter, A.M.; Jakob, K.; Klein, N.P.; Dekker, C.L.; Edwards, K.M.; Halsey, N.A.; Baxter, R.; Williams, S.E.; Graham, P.L.; LaRussa, P. Live Vaccine Use and Safety in DiGeorge Syndrome. Pediatrics 2014, 133, e946–e954. [Google Scholar] [CrossRef] [PubMed]
  150. Lim, S.M.; Shin, J.H.; Baek, J.Y.; Lee, J.Y.; Kang, J.-M.; Ahn, J.G. Safety of Live Immunization in DiGeorge Syndrome: A Retrospective Single-Center Study in Korea, 2005–2021. Vaccines 2022, 10, 2165. [Google Scholar] [CrossRef]
  151. Berkhout, A.; Preece, K.; Varghese, V.; Prasad, V.; Heussler, H.; Clark, J.; Wen, S.C.H. Optimising immunisation in children with 22q11 microdeletion. Ther. Adv. Vaccines Immunother. 2020, 8, 1–7. [Google Scholar] [CrossRef] [PubMed]
  152. Morrow, B.; McDonald-McGinn, D.M.; Emanuel, B.S.; Vermeesch, J.R.; Scambler, P.J. Molecular Genetics of 22.q11.2 deletion syndrome. Am. J. Med. Genet. 2018, 176, 2070–2081. [Google Scholar] [CrossRef] [PubMed]
  153. Mustillo, P.J.; Sullivan, K.E.; Chinn, I.K.; Notarangelo, L.D.; Haddad, E.; Davies, E.G.; de la Morena, M.T.; Hartog, N.; Yu, J.E.; Hernandez-Trujillo, V.P.; et al. Clinical Practice Guidelines for the Immunological Management of Chromosome 22q11.2 Deletion Syndrome and Other Defects in Thymic Development. J. Clin. Immunol. 2023, 43, 247–270. [Google Scholar] [CrossRef]
  154. 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]
  155. McGovern, P.E.; Crowley, T.B.; Zackai, E.H.; Burrows, E.; McDonald-McGinn, D.M.; Nance, M.L. Surgical insights and management in patients with 22q11.2 deletion syndrome. Pediatr. Surg. Int. 2022, 38, 899–905. [Google Scholar] [CrossRef]
  156. Abu-Ghname, A.; Perdanasari, A.T.; Raj, S.; Seema, J.; Wilson, K.T.; Maricevich, R.S. Access to multidisciplinary care for patients with 22q11.2 deletion syndrome: Identifying breakdowns in the screening process. J. Craniofac. Surg. 2020, 31, 428–431. [Google Scholar] [CrossRef]
  157. Boot, E.; Óskarsdóttir, S.; Loo, J.C.; Crowley, T.B.; Orchanian-Cheff, A.; Andrade, D.M.; Arganbright, J.M.; Castelein, R.M.; Cserti-Gazdewich, C.; de Reuver, S.; et al. Updated clinical practice recommendations for managing adults with 22q11.2 deletion syndrome. Anesth. Analg. 2023, 25, 100344. [Google Scholar] [CrossRef]
  158. Hickey, S.E.; Kellogg, B.; O’brien, M.; Hall, C.; Kirschner, R.E.; Santoro, S.L.; Leonard, H.; Baylis, A.L. Impact of Interdisciplinary Team Care for Children with 22q11.2 Deletion Syndrome. Cleft Palate-Craniofacial J. 2020, 57, 1362–1369. [Google Scholar] [CrossRef]
  159. Meneses, Z.; Durant, J.; Ale, H. The Unique Experience of a New Multidisciplinary Program for 22q Deletion and Duplication Syndromes in a Community Hospital in Florida: A Reaffirmation That Multidisciplinary Care Is Essential for Best Outcomes in These Patients. Genes 2022, 13, 1949. [Google Scholar] [CrossRef]
  160. Bassett, A.S.; McDonald-McGinn, D.M.; Devriendt, K.; Digilio, M.C.; Goldenberg, P.; Habel, A.; Marino, B.; Oskarsdottir, S.; Philip, N.; Sullivan, K.; et al. Practical Guidelines for Managing Patients with 22q11.2 Deletion Syndrome. J. Pediatr. 2011, 159, 332–339.e1. [Google Scholar] [CrossRef]
Figure 1. The schematic representation of the 22q11.2 region comprising LCR22 A, B, C, and D sequences and encompassing protein-coding genes, long non-coding RNAs, microRNAs, and genes expressed in functional haploinsufficiency in 22q11.2 DS.
Figure 1. The schematic representation of the 22q11.2 region comprising LCR22 A, B, C, and D sequences and encompassing protein-coding genes, long non-coding RNAs, microRNAs, and genes expressed in functional haploinsufficiency in 22q11.2 DS.
Ijms 24 08317 g001
Table 1. Multisystemic anatomical anomalies and dysfunctions in patients with 22q11.2 deletion syndrome.
Table 1. Multisystemic anatomical anomalies and dysfunctions in patients with 22q11.2 deletion syndrome.
Systemic InvolvementPhenotypic FeaturesFrequency in 22q11.2 DS
Facial dysmorphismElongated face
Hooded eyelids
Upslanted palpebral fissures
Epicanthus
Wide nasal bridge
Long nose with a bulbous tip
Narrow alar base
Short philtrum
Small mouth
Micrognathia and retrognathia
Low-set small ears
80–99%
Ocular findingsPosterior embryotoxon
Tortous retinal vessels
Refractive errors
Strabismus
Amblyopia
7–70%
DentitionDelayed teeth eruption
Agenesis of permanent dentition
Supernumerary teeth
Enamel hypoplasia
Impaired enamel calcification
2.5%
Palatal anomaliesVelopharyngeal insufficiency and hypotonia
Cleft palate
Submucous cleft palate
Bifid uvula
69–100%
Laryngeal anomaliesGlottic web
Laryngeal stenosis
Laryngeal cleft
Laryngomalacia
Vocal fold anomalies
25–43%
Lower airway anomaliesTracheo and bronchomalacia
Tracheal stenosis
Short trachea with reduced tracheal rings
Aberrant tracheal bronchus
Tracheoesophageal fistula
21%
Cardiovascular anomaliesInterrupted aortic arch type B
Truncus arteriosus
Tetrealogy of Fallot
Conoventricular septal defect
Isolated aortic arch anomaly
Double outlet right ventricle
Transposision of the great arteries
Hypoplastic left heart syndrome
Valvar pulmonary stenosis
49–83%
Genitourinary anomaliesRenal agenesis
Multicystic dysplastic kidney
Hydronephrosis
Duplicated collecting system
Absent uterus
Hypospadias, cryptorchidism
33%
Gastrointestinal anomaliesGastroesophageal reflux
Esophageal atresia
Impaired swallowing
Hirschprung disease
Imperforate anus
30%
Central nervous system anomaliesCerebral atrophy
Polymicrogyria
Atrophy of the hippocampus
Cerebellar atrophy
8%
Endocrine anomaliesThyroid gland aplasia/hypoplasia
Retrocarotid and retroesphageal thyroid extension
Inhomogeneous thyroid structure
Parathyroid gland dysfunction
Growth hormone deficiency
65%
Skeletal and muscular anomaliesCervical spine anomalies
Thoracic vertebral anomalies
Arachnodactyly, Camptodactyly, Syndactyly
Hammer toes
Skull malformations
Diaphragmatic hernia
17–19%
Immune disordersAthymia
Thymic hypoplasia
Ectopic thymus
75%
Table 2. Genetic modifiers influencing the TBX1 penetrance and affecting the phenotypic expression in 22q11.2 DS.
Table 2. Genetic modifiers influencing the TBX1 penetrance and affecting the phenotypic expression in 22q11.2 DS.
Genetic ModifierRolePhenotypic Expression
CRKL
(CRK like proto-oncogene adaptor protein)
Activates the RAS and JUN kinase signaling pathways, mediates transduction of intracellular signalsDevelopment of organs originating from the neural crest, thymus, parathyroid glands, craniofacial structures, T lymphocytes, cardiac outflow region
SLC2A3 aka GLUT3
(Solute carrier family 2 member 3)
Facilitated glucose transporterConotruncal heart region, aortic arch
KANSL1
(KAT8 regulatory NSL complex subunit 1)
Histone acetyltransferase complex memberDevelopmment of aortic arch, semilunar valve, cardiac septa, pulmonary artery
JMJD1C
(jumonji domain containing 1C)
Chromatin expression modification, histone demethylationPharyngeal apparatus, cardiac outflow region
RREB1
(Ras responsive element binding protein 1)
Chromatin expression modification, histone demethylationConotruncal heart region
SEC24C
(SEC24 family member C)
Role in transporting proteins from the endoplasmic reticulum to the Golgi apparatusEmbryonic development, cardiac outflow region
MINA
(MYC induced nuclear antigen)
Chromatin expression modification, histone demethylationCardiac development
KDM7A
(Lysine-specific demethylase 7A)
Chromatin expression modification, histone demethylationCardiac development
DGCR8
(DiGeorge syndrome critical region 8)
miRNAs and lnRNAs regulationEmbryo development, Immune, naurological, and cardiac functions
NCOR2
(Nuclear receptor corepressor 2)
Transcriptional regulator of the retinoic acid signalingB and T lymphocytes, regulation of the immune response
EP300
(E1A binding protein P300)
Transcriptional regulator of the retinoic acid signalingB and T lymphocytes, regulation of the immune response
Table 3. Autosomal recessive conditions resulting from mutations associated with 22q11.2 hemizygosity.
Table 3. Autosomal recessive conditions resulting from mutations associated with 22q11.2 hemizygosity.
GeneDiseaseOMIM#Phenotype
PRODHHyperprolinemia type 1239500General: neurological deficits
Specific: psychomotor delay, hypotonia, seizures
SLC25A1D2A2AD syndrome615182General: severe muscular weakness, respiratory distress, failed psychomotor development, early death
Specific: encephalopathy, seizures
GP1BBBernard-Soulier syndrome231200Specific: hematologic disease, thrombocytopenia, increased megakaryocytes
SCARF2Van den Ende-Gupta syndrome600920General: joint dislocations
Specific: contractual arachnodactyly, hooked clavicles, blepharophimosis
SNAP29CEDNIK syndrome609528General: neuropathy
Specific: cerebral dysgenesis, ichtyosis, keratoderma
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

Szczawińska-Popłonyk, A.; Schwartzmann, E.; Chmara, Z.; Głukowska, A.; Krysa, T.; Majchrzycki, M.; Olejnicki, M.; Ostrowska, P.; Babik, J. Chromosome 22q11.2 Deletion Syndrome: A Comprehensive Review of Molecular Genetics in the Context of Multidisciplinary Clinical Approach. Int. J. Mol. Sci. 2023, 24, 8317. https://doi.org/10.3390/ijms24098317

AMA Style

Szczawińska-Popłonyk A, Schwartzmann E, Chmara Z, Głukowska A, Krysa T, Majchrzycki M, Olejnicki M, Ostrowska P, Babik J. Chromosome 22q11.2 Deletion Syndrome: A Comprehensive Review of Molecular Genetics in the Context of Multidisciplinary Clinical Approach. International Journal of Molecular Sciences. 2023; 24(9):8317. https://doi.org/10.3390/ijms24098317

Chicago/Turabian Style

Szczawińska-Popłonyk, Aleksandra, Eyal Schwartzmann, Zuzanna Chmara, Antonina Głukowska, Tomasz Krysa, Maksymilian Majchrzycki, Maurycy Olejnicki, Paulina Ostrowska, and Joanna Babik. 2023. "Chromosome 22q11.2 Deletion Syndrome: A Comprehensive Review of Molecular Genetics in the Context of Multidisciplinary Clinical Approach" International Journal of Molecular Sciences 24, no. 9: 8317. https://doi.org/10.3390/ijms24098317

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