Next Article in Journal
Genome-Wide Identification, Characterization, and Expression Analysis of the HD-Zip Gene Family in Lagerstroemia for Regulating Plant Height
Next Article in Special Issue
Exploring the Role of the MUTYH Gene in Breast, Ovarian and Endometrial Cancer
Previous Article in Journal
Investigating the Influence of ANTXR2 Gene Mutations on Protective Antigen Binding for Heightened Anthrax Resistance
Previous Article in Special Issue
Novel Pathogenic Variants Leading to Sporadic Amyotrophic Lateral Sclerosis in Greek Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genetic Alterations in a Large Population of Italian Patients Affected by Neurodevelopmental Disorders

by
Annaluisa Ranieri
1,†,
Ilaria La Monica
1,2,†,
Maria Rosaria Di Iorio
1,2,
Barbara Lombardo
1,2,* and
Lucio Pastore
1,2
1
CEINGE-Biotecnologie Avanzate Franco Salvatore, via G. Salvatore 486, 80145 Naples, Italy
2
Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, via Sergio Pansini 5, 80131 Naples, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Genes 2024, 15(4), 427; https://doi.org/10.3390/genes15040427
Submission received: 21 February 2024 / Revised: 26 March 2024 / Accepted: 27 March 2024 / Published: 28 March 2024
(This article belongs to the Special Issue Genetics of Multifactorial Diseases)

Abstract

:
Neurodevelopmental disorders are a group of complex multifactorial disorders characterized by cognitive impairment, communication deficits, abnormal behaviour, and/or motor skills resulting from abnormal neural development. Copy number variants (CNVs) are genetic alterations often associated with neurodevelopmental disorders. We evaluated the diagnostic efficacy of the array-comparative genomic hybridization (a-CGH) method and its relevance as a routine diagnostic test in patients with neurodevelopmental disorders for the identification of the molecular alterations underlying or contributing to the clinical manifestations. In the present study, we analysed 1800 subjects with neurodevelopmental disorders using a CGH microarray. We identified 208 (7%) pathogenetic CNVs, 2202 (78%) variants of uncertain significance (VOUS), and 504 (18%) benign CNVs in the 1800 patients analysed. Some alterations contain genes potentially related to neurodevelopmental disorders including CHRNA7, ANKS1B, ANKRD11, RBFOX1, ASTN2, GABRG3, SHANK2, KIF1A SETBP1, SNTG2, CTNNA2, TOP3B, CNTN4, CNTN5, and CNTN6. The identification of interesting significant genes related to neurological disorders with a-CGH is therefore an essential step in the diagnostic procedure, allowing a better understanding of both the pathophysiology of these disorders and the mechanisms underlying their clinical manifestations.

1. Introduction

Molecular cytogenetic techniques, such as the array-comparative genomic hybridization (a-CGH) method, have increased our ability to identify clinically relevant submicroscopic copy number variants (CNVs) in patients with neurodevelopmental disorders (NDDs), such as autism spectrum disorder (ASD), intellectual disability (ID), attention deficit hyperactivity disorder (ADHD), psychomotor developmental delay, specific learning disorder, communication disorders, speech delay, epilepsy, and schizophrenia [1,2,3]. NDDs are a group of heterogeneous conditions whose onset and clinical expression occur during childhood or adolescence and are characterized by impairment in cognition, communication, adaptive behaviour, and psychomotor skills resulting from abnormal brain development [4,5]. Such disorders represent a serious health problem in our society, affecting 3% of children worldwide [6]. NDDs have a strong genetic basis; however, additional causal environmental factors have also been identified [7]. These factors may interact during brain development causing disorders that to date have not been fully elucidated. The influence of environmental insults and the presence of genetic alterations during any of these stages can alter brain development and cause these disorders [8,9]. Many studies suggest the presence of shared molecular pathways underlying multiple clinical signs typical of NDDs; in fact, the comorbidity of two or more of these disorders is often found in the same individual [10]. Identifying the pathogenetic mechanisms common to different NDDs will help explain the aforementioned comorbidity and will eventually lead to the identification of effective therapies [6,11]. CNVs represent the genetic aberrations most related to NDDs [12,13,14]. The identification of CNVs and mutations in specific genes as significant causative factors in NDDs, via a-CGH and exome sequencing or genome-wide next-generation sequencing (NGS) analyses, is critical for accurate genetic counselling and represents an essential first step toward a better understanding of the molecular pathways underlying these disorders [15,16,17,18,19]. In particular, a-CGH is currently considered a first-level diagnostic tool for identifying neurodevelopmental disorders with a significantly higher diagnostic performance (5–10%) than conventional karyotype analysis [12,20].
In the present study, we analysed subjects with ASD, ID, speech delay, psychomotor developmental delay, ADHD, epilepsy, schizophrenia, and other neurological disorders using a-CGH. We evaluated diagnostic efficacy of a-CGH and its importance as a routine genetic test in patients with NDDs to identify molecular alterations underlying or contributing to clinical manifestation [21]. Specifically, we have identified in our patients and, subsequently, studied through database and literature support the major pathogenic CNVs and variants of uncertain significance (VOUS) containing in some cases a single gene potentially related to their disorders. We also believe that the early diagnosis of NDDs, through careful observation of early signs that may occur during childhood and appropriate molecular diagnostics, can potentially allow significant changes in the clinical evolution of these disorders [22]. Early specific therapies can possibly improve their clinical progress and positively influence their evolution, preventing or reducing their evolution into more severe psychiatric disorders later in life [6].

2. Materials and Methods

2.1. Patients

In the period between January 2019 and March 2023, we evaluated by a-CGH a cohort of 1800 paediatric and adolescent patients aged between a few days and 18 years old; patients were affected by different neurodevelopmental disorders with a male-to-female ratio of 2.5:1. The patients were referred from different departments, in particular, 2% from the Dipartimento di Neuropsichiatria Infantile, Università della Campania “Luigi Vanvitelli” (A.O.U. Luigi Vanvitelli), Napoli, 39% from Dipartimento di Neuropsichiatria Infantile, Università degli Studi di Napoli “Federico II” (A.O.U. Federico II), Napoli, 25% from Dipartimento di Pediatria, Università degli Studi di Napoli “Federico II”, (A.O.U. Federico II), Napoli, 4% from Dipartimento di Genetica Medica, Azienda Ospedaliera di Rilievo Nazionale Santobono Pausilipon (A.O.R.N. Santobono Pausilipon), Napoli, 3% from Dipartimento di Genetica Medica, Azienda Ospedaliera di Rilievo Nazionale Antonio Cardarelli, (A.O.R.N. Antonio Cardarelli), Napoli, 11% from Dipartimento di Neuropsichiatria Infantile, Azienda Ospedaliera San Giovanni di Dio e Ruggi d’Aragona (A.O.U. San Giovanni di Dio e Ruggi d’Aragona), Salerno, and 16% from DAI Medicina di Laboratorio e Trasfusionale, Azienda Ospedaliera Federico II, Napoli (Ambulatorio DAIMELAB A.O.U. Federico II). When available, diagnostic investigations were conducted on the parents of the patients analysed, in order to determine the transmission or de novo origin of the identified CNVs.
Developmental delay and ID were evaluated by using different methods depending on the patients’ age. In early childhood, before the age of three, the assessment of motor and cognitive developmental was evaluated using the Bayley Scales of Infant Development and Toddler Development, the Griffith Mental Development Scales, and the Autism Diagnostic Observation Schedule, Second Edition (ADOS-2). After three years of age, the Autism Diagnostic Interview-Revised (ADI-R) was performed; for children six years old and older, Wechsler Intelligence Scale for Children (WISC) for ID, the Raven’s Matrices, and the Vineland Adaptive Behavior Scales for the adaptive quotient were used. All patients gave their written informed consent to the study, which was performed according to the Helsinki II Declaration and was approved by the Ethics Committee of our Faculty of Medicine (authorization no. 193/06, 25 October 2006; amendment no. 193/06/ESES1, 1 October 2014).

2.2. Genetic Testing

Patients’ DNA and their parents’ DNA, when available, was isolated from peripheral blood samples using the RSC Blood DNA Kit (Promega, Madison, WI, USA), using manufacturer’s protocol. DNA quality and concentration were determined with a NanoDrop 2000 spectrophotometer (ThermoFisher Scientific Inc., Bartlesville, OK, USA). Following extraction, parents’ DNA and their parents’ DNA were processed according to the Agilent protocol (Version 7.5, June 2016) using Human Reference DNA (Promega, Madison, WI, USA). In particular, chromosomal microarray analysis (CMA) was performed using SurePrint G3 Human CGH Array 4 × 180 K (Agilent Technologies, Santa Clara, CA, USA), according to the manufacturers’ recommendation. The Agilent SurePrint G3 Human CGH Array 4 × 180 K has an average spacing of 13 kb, allowing an average resolution of 25 kb. The array images were acquired using the Agilent laser scanner G2600D. Image files were quantified, and data were visualized by using Agilent’s Cytogenomics software version 4.0.3.12 (Agilent Technologies, Santa Clara, CA, USA). Finally, the detected pathogenic and likely pathogenic CNVs were validated using quantitative real-time polymerase chain reaction (RT-qPCR) technique. RT-qPCR was performed using the SYBR Green method and implemented in the Applied Biosystems 7900HT Fast Real-time PCR system, following the manufacturer’s protocol (Applied Biosystems, Life of technologies). Primers for each pathogenic/probably pathogenic CNV were designed using Primer Express 2.0 software (Applied Biosystems, Carlsbad, CA, USA); primers selected from the ZNF80 gene were used to generate a normalizing amplicon. Assays were performed in a total volume of 10 μL in duplicate in 96-well optical reaction plates (Applied Biosystems). For each sample, a relative quantification was carried out using 9 genomic DNA samples (ctrls). A comparative 2−∆∆Ct method was used to validate the rearrangements identified by a-CGH in this study [23].

2.3. Data Analysis

The analysis algorithm for CNVs was set in Agilent’s Cytogenomics version 4.0.3.12 ADM2 analysis software with a threshold of 6, with the minimum number of probes considered for the inclusion of a CNV equal to 3, and the Minimum Absolute Average Log Ratio of Region equal to 0.25. All CNVs of the same size as those detected with a minimum of 3 probes were analysed. The discovered CNVs were analysed and compared with aberrations present in a number of databases including: Database of Genomic Variants (DGV; http://dgv.tcag.ca/dgv/app/home, 15 February 2024), Database of Chromosomal Imbalance and Phenotype in Humans (DECIPHER; https://decipher.sanger.ac.uk/, 15 February 2024), ClinVar (National Center for Biotechnology Information, U.S. National Library of Medicine; http://www.ncbi.nlm.nih.gov/clinvar/, 15 February 2024), ClinGen (https://clinicalgenome.org, 15 February 2024), The Human Gene Database, GeneCards (https://www.genecards.org/, 15 February 2024), and Online Mendelian Inheritance in Man (OMIM; https://www.omim.org/, 15 February 2024). CNVs were classified into three groups (pathogenic, VOUS, and benign) according to the American College of Medical Genetics and Genomics (ACMG) standards and guidelines [24]. Benign CNVs have typically been reported in multiple peer-reviewed publications or annotated in curated databases as benign and are usually present in >1% of the population. Pathogenic CNVs included alterations with consistent clinical phenotypes across multiple peer-reviewed publications, well-documented penetrance, and expressivity and with almost one gene known to be dosage sensitive. VOUS represents a broad category and may include findings that are later demonstrated with additional evidence to be either pathogenic or benign. Moreover, we analysed our results by using the Simons Foundation Autism Research Initiative database gene autism (SFARI), in which 2290 different CNV regions and 1416 genes are reported and associated with ASD in different types of studies (update October 2023; https://gene.sfari.org/). The analysis was carried out by evaluating the correlation of the alterations with the main diagnostic suspicion present on the parents’ request, in order to avoid confusing interpretations of the results.

3. Results

Among the 1800 patients who came to our attention, 649 (36%) had ASD, 341 (19%) psychomotor developmental delay, 187 (10%) ID, 194 (11%) speech delay, 38 (2%) ADHD, 15 (1%) schizophrenia, 109 (6%) epilepsy, and 267 (15%) other neurological disorders (Figure 1). Since it was not possible to carry out a complete clinical evaluation and phenotyping on the patients, they were divided and classified on the basis of the main diagnostic suspicion reported on request. For this reason, it must be considered that comorbidities are common in neurodevelopmental disorders and the final diagnosis may include multiple phenotypic traits. We identified 208 (7%) pathogenic CNVs, 2202 (76%) VOUS, and 504 (17%) benign CNVs in our patients (all informations are available in the Supplementary Materials). In total, the number of deletions (1750, 60%) identified in patients is slightly higher than the number of duplications (1164, 40%). Microdeletions were more frequently classified as pathogenic or likely pathogenic compared to microduplications (60% vs. 40%, Figure 2); however, their sizes did not differ significantly. Among the 194 patients with 208 pathogenic/likely pathogenic CNVs, 33% of CNVs (68/208) were of de novo origin, 14% of CNVs (29/208) were inherited from a parent. In the remaining cases, parental DNA was not available for analysis.
In addition, 76% of all a-CGH findings (2202/2914) were classified as VOUS. Among the VOUS for which it was possible perform analysis on parental DNA, 21.9% of CNVs (482/2202) were of de novo origin, and 28% of CNVs (626/2202) were inherited. CNVs in 26.4% of the cases analysed were classified as likely benign with 504 CNVs in 476 patients (476/1800). In general, the likely benign CNVs found are poor in genes and they are most likely not correlated with the phenotype of the patients. A majority of benign CNVs were found to be inherited from unaffected parents when they were tested: in fact, 58% of the likely benign CNVs (292/504) were inherited, while 36 CNVs were of de novo origin.
Among the pathogenic and likely pathogenic alterations observed in our patients, we identified microdeletion and microduplication associated with specific syndromes: microdeletion 1q21.1-q21.2 (1q21.1 Deletion syndrome OMIM#612474, Thrombocytopenia-absent radius syndrome OMIM#274000), microduplication 1q21.1-q21.2 (1q21.1 Duplication syndrome OMIM# 612475), microduplication 15q11.2-q13.1 (Duplication 15q11-q13 syndrome OMIM#608636, Angelman syndrome OMIM#105830, Prader–Willi syndrome OMIM#176270), microdeletion 16p11.2 (16p11.2 deletion syndrome, OMIM#611913), microduplication 16p11.2 (16p11.2 duplication syndrome OMIM#614671), microdeletion 22q11.21 (DiGeorge syndrome OMIM#188400, Velocardiofacial syndrome OMIM#192430), microduplication 22q11.21 (22q11.2 microduplication syndrome OMIM#608363, DiGeorge syndrome OMIM#188400, Velocardiofacial syndrome OMIM#192430), microdeletion Xp21.1 (Duchenne muscular dystrophy OMIM#310200, Becker muscular dystrophy OMIM#300376), microduplication 17p12 (Charcot–Marie-Tooth disease, type 1A OMIM#118220), and microdeletion 17p11.2 (Smith–Magenis syndrome OMIM #182290) (Table 1). Instead, among the pathogenic/likely pathogenic CNVs, being unique non-recurrent and not associated with syndromes, we have identified interesting CNVs such as microduplication 11q13.1-q13.2, microduplication 5q14.3, microdeletion Xq28, microdeletion 13q21.32-q31.1, microdeletion 2p16.3, microduplication 20p12.3-p12.1, and microdeletion 15q26.3 (Table 2).
Among the VOUS observed in patients (Table 3), we selected by detailed gene content analysis CNVs of uncertain clinical significance structural containing genes significantly related to neurodevelopmental disorders, such as Ankyrin Repeat And Sterile Alpha Motif Domain-Containing 1B (ANKS1B, OMIM*607815), located in 12q23.1; Ankyrin Repeat Domain 11 (ANKRD11, OMIM*611192), located in 16q24.3; RNA Binding Fox-1 Homolog 1 (RBFOX1, OMIM*605104), located in 16p13.3; Cholinergic Receptor Nicotinic Alpha 7 Subunit (CHRNA7, OMIM*118511), located in 15q13.3; Gamma-Aminobutyric Acid Type A Receptor Subunit Gamma3 (GABRG3, OMIM*600233), located in 15q12; Astrotactin 2 (ASTN2, OMIM*612856), located in 9q33.1; SH3 And Multiple Ankyrin Repeat Domains 2 (SHANK2, OMIM*603290), located in 11q13.3-q13.4; Kinesin Family Member 1A (KIF1A, OMIM*601255) located in 2q37.3; Set-binding Protein 1 (SETBP1, OMIM*611060), located in 18q12.3; Syntrophin-gamma-2 (SNTG2, OMIM*608715), located in 2p25.3; Catenin, alpha-2 (CTNNA2, OMIM*114025), located in 2p12; Topoisomerase, DNA, III, beta (TOP3B, OMIM*603582), located in 22q11.22; Contactin 4 (CNTN4, OMIM*607280), located in 3p26.3-p26.2; Contactin 6 (CNTN6, OMIM*607220), located in 3p26.3; and Contactin 5 (CNTN5, OMIM*607219), located in 11q22.1.

4. Discussion

A-CGH is an extremely relevant diagnostic step for the discovery of new submicroscopic rearrangements involved in the pathogenesis of NDDs; its diagnostic yield has rapidly increased so that it was adopted and recommended as a first-level clinical diagnostic test for such disorders [25,26]. In order to adequately determine the role of the CNVs detected in patients, we considered their type (deletion or duplication), size, gene content, inheritance, and information present in public databases and in the literature. The identification of new microdeletions and microduplications in the human genome represents an important challenge to determine the role and clinical impact of these chromosomal rearrangements [27]. For this reason, during our 4-year study, we analysed patients with NDDs using a-CGH, identifying CNVs containing interesting genes, potentially related to the phenotypic characteristics found in our patients and which could contribute to clarifying the pathogenesis of these disorders. Many pathogenic CNVs result in similar clinical manifestations such as ID and ASD, but may differ in the occurrence of specific and different phenotypic traits for each subject. In fact, the impact of different variants in allelic loci present in each individual genome may act as modifiers; in addition, different expression of the alleles could also explain this phenomenon [28]. In particular, these phenomena could clarify cases in which patients inherit pathogenetic mutations from healthy parents causing incomplete penetrance or variable expressivity of the genes included in the detected CNVs [28]. For example, in patients with CNVs classified as pathogenic/likely pathogenic, we identified two cases of 1q21.1-q21.2 microdeletion of maternal origin and one case of paternal origin, one case of 1q21.1-q21.2 microduplication of paternal origin, one case of 22q11.21 microdeletion inherited from the father, one case of 22q11.21 microduplication inherited from the mother, one case of 16p11.2 microdeletion of maternal origin, one case of 17p12 microduplication of paternal origin, one case of 17p11.2 microdeletion inherited from the father, one case of microdeletion 15q26.3 of paternal origin, one case of 2p16.3 microdeletion of maternal origin, and one case of paternal origin. In these cases, the modifying impact of multiple loci in the human genome could contribute to explain the more severe phenotype found in children compared to their parents or the variability of phenotypes among children who share the same CNVs [29]. Paternal or maternal inheritance of a CNV should also consider a possible source of variability due to the possible presence of imprinted genes.
Among the genes contained in the VOUS, we focused our attention on a small number of genes, not well characterized on databases and in the literature, but which were present in the SFARI databases and correlated with the development of phenotypic features present in NDDs; this approach can lead to the identification of new causative or potentially causative genes that contribute to elucidate the etiopathogenesis of these disorders.
The first interesting case that we have selected is an 8-year-old male child with ASD, a communicative-relational disorder in the presence of nonverbal intellectual functioning in the normal range, slow language evolution, discontinuous and fleeting gaze attachment, and epilepsy with continuous spike waves in sleep. We identified a deletion in the 12q23.1 region of 180.0 kb extent, partially containing the Ankyrin Repeat and Sterile Alpha Motif Domain-Containing 1B (ANKS1B) gene. This deletion involves intron 9, exon 9, and intron 8 of the ANKS1B gene; this alteration appeared to be de novo after an in-depth diagnostic investigation conducted on the parents. In addition, smaller deletions involving only intronic regions were identified in three patients with ASD and one patient with ID, ADHD, and dysmorphic features. Only in one of the four cases could the maternal origin of the deletion containing ANKS1B be observed. The ANKS1B gene encodes for the protein AIDA-1, highly expressed in the brain, and enriched in hippocampal and cerebellar regions; AIDA-1 is one of the most abundant proteins at neuronal synapses [30], located as a central postsynaptic density protein and interacting with PSD95 in a complex that contains other factors associated with NDDs, including GRIN2B, SYNGAP1, and NLGN [31]. Mouse models with heterozygous deletions in the ANKS1B gene have shown a number of distinctive behavioural signs typical of autism [32,33]. Monogenic CNVs including the ANKS1B gene have been identified in individuals showing a spectrum of neurodevelopmental phenotypes, including ASD, ADHD, and motor and language deficits [30,34,35]. The ANKS1B gene is present in the SFARI database with a 2.1S, Strong Candidate, Syndromic score with evidence that mutations in this gene have an important functional effect and are correlated with an increased risk of developing ASD. Therefore, ANKS1B could represent a crucial gene in which rare variants with loss of function cause NDDs, including ASD, ADHD, and deficits in speech and motor function.
Another CNV that we have evaluated consists of an approximately 43 kb duplication on chromosome 18 in the q12.3 region encompassing the SET binding protein 1 (SETBP1) gene, identified in a 4-year-old male patient with a NDD characterized by growth and psychomotor developmental delay, behavioural disturbances, and language regression from 18 months of age. The duplication partially includes intron 5 and exon 6 of the gene SETBP1; analysis of parental DNA was not possible, so we could not establish whether it was a de novo alteration. SETBP1 has an important role in controlling forebrain progenitor expansion and neurogenic differentiation [36]. The SETBP1 gene is associated with several NDDs; in fact, haploinsufficiency due to deletions in the heterozygosity of the gene or mutations with the production of a nonfunctioning protein cause a dysfunction-related disorder of the SETBP1 gene, characterized by hypotonia and mild motor developmental delay; intellectual abilities ranging from normal to severely impaired; speech and language disorders; behavioural problems; and mild dysmorphic [37,38,39]. Its strong association with developmental delay along with language impairment makes SETBP1 a new candidate gene for NDDs [40,41]. This gene is present in SFARI with a score of 1.1, high confidence, indicating a clear implication in the development of ASD. Specifically, duplications or mutations of the SETBP1 gene likely result in a gain of function with subsequent degradation or production of a dominant negative suggesting a possible role of SETBP1 gene duplication in the development of the phenotype found in the patient [36].
In addition, we identified and studied an alteration in the Astrotactin 2 (ASTN2) gene, found in a 6-year-old male patient diagnosed with ASD, characterized by significant difficulties in social interaction and communication with a tendency toward isolation, and also the presence of severe psychomotor instability and some motor stereotypies. The CNV present in our patient is a de novo deletion of extent 167.9 kb present in the 9q33.1 region involving intron 1, exon 2, and partially intron 2 of the ASTN2 gene. ASTN2 encodes for an integral membrane protein, expressed in the brain, that plays a central role in neural-glial adhesion during neuronal migration and synaptic strength through the trafficking and degradation of surface proteins [42]. ASTN2 deletions has been identified in patients with ASD, ADHD, and schizophrenia, highlighting how ASTN2 alterations are a risk factors for NDDs [43,44]. ASTN2 knockout mice showed an increase in exploratory activity in a new environment, social behaviour and impulsivity, or a release of anxious and depressed behaviours [45]. Such behaviour is also seen in patients with ADHD and ASD, who are often unable to adapt to new situations [46,47]. According to the SFARI databases, ASTN2 is reported as a strong candidate with a score of 2.1, which demonstrates that the variants present in this gene could have a functional effect and correlate with an increased risk of developing disorders such as ASD, especially due to the role of ASTN2 in the modulation of synaptic activity during neural development.
Genetic variants involving a gamma-aminobutyric acid (GABA) receptor have been identified in patients with NDDs. In fact, we found a deletion in the 15q12 region encompassing the Gamma-Aminobutyric Acid Type A Receptor Subunit Gamma3 (GABRG3) gene in three ASD patients. GABA is the major inhibitory neurotransmitter in the mammalian brain where it acts on GABA-A receptors, which are ligand-gated chloride channels. Deletions or duplications of the 15q11-13 region containing the GABR genes are related to multiple neurological disorders including Prader–Willi syndrome (PWS), Angelman syndrome (AS), and autism [48]. In particular, we found in a 4 years old male patient, with ASD characterized by relational difficulties, absent eye contact, and severely impaired communication with indicative gesture and absent verbal language, a deletion of 47.0 kb involving intron 3 of the GABRG3 gene. The unavailability of parents’ DNA prevented us from clarifying whether it was a de novo alteration. Studies in rat models have pointed out that the neurotransmitter GABA appears early in neurogenesis; during embryogenesis, GABAergic neurons become widespread in all regions of the developing brain and spinal cord [49,50,51]. A similar pattern emerges in the developing human CNS, with a transient and widespread distribution of GABAergic neurons [52]. This suggests that GABA plays a role in neurogenesis before becoming the main fast-acting inhibitory neurotransmitter in the brain. The subunits that make up GABAA receptors have also been shown to change during development [53]. In a pilot study, the role of some genetic variants of the GABA type A receptor subunits GABRB3, GABRG3, and GABRA5 in the aetiology of ASD was investigated [54], supporting the hypothesis that ASD may be also caused by GABAergic dysfunctions [55]. In fact, GABRG3 gene is reported in SFARI as a strong candidate with a score of 2.1 that correlates with an increased risk of developing NDDs such as ASD.
From the analysis carried out in a 15-year-old female autistic patient, with impaired pragmatic communication, difficulty interacting with her peers due to a severe deficit in social skills, and a strong state of anxious activation with isolation and obsessive behaviours, we identified a paternal duplication in the 2q37.3 region of 52.7 kb that partially containing the Kinesin Family Member 1A (KIF1A) gene, which aroused our particular interest. This duplication of paternal origin involves from intron 27 to intron 45 and from exon 28 to exon 46 of the KIF1A gene. The KIF1A gene encodes for a microtubule-dependent motor protein responsible for fast anterograde transport of synaptic vesicle precursors in neurons [56]. KIF1A is a motor protein expressed exclusively in the brain; in fact, several studies have shown that de novo pathogenic variants determine an increased risk of ASD [57,58]. There is therefore a link between KIF1A mutations and ASD. Notably, de novo dominant mutations in the motor domain have been reported in association with complex phenotypes characterized by ID and the variable presence of cerebellar atrophy, spastic paraparesis, hyperactivity, and epilepsy [57,58,59]. KIF1A is listed in the SFARI database as a strong syndromic candidate with score 2.1S, associated with an increased risk of developing ASD.
Furthermore, among the alterations evaluated we observed a paternally derived duplication in the 2p25.3 region of size 457.1 kb in a 9-year-old patient with poor social interaction, severe communication delay, hyperresponsiveness to external stimuli, poor orientation, hyperactivity, and episodes of delirium consistent with a diagnosis of ASD made at age 4 years. This duplication included exons 1–6 of the Syntrophin-γ2 (SNTG2) gene. CNVs of different sizes containing the SNTG2 gene were also found in four patients with ASD, two patients with ID, and one patient with psychomotor developmental delay and speech delay. It was only possible in three cases to perform an in-depth diagnostic investigation in the parents, which defined in one patient the paternal origin of the duplication, in one patient the de novo origin, and in one patient the maternal origin. The SNTG2 gene encodes for a scaffolding protein that interacts with neuroligins (NL4X and NL4Y) as a crucial binding factor at inhibitory synapses. Mutations in the neuroligins (NL3 and NL4X) have been implicated in the development of autism and ID [60]. In particular, it was observed that the binding of neuroligins to SNTG2, at inhibitory synapses can be influenced by mutations in neuroligins, showing how interactions between neuroligins and SNTG2 could be implicated in the aetiology of autism [61]. Indeed, the SFARI database reports the SNTG2 gene with a score of 2.1, Strong Candidate, indicating an unambiguous implication of the gene in ASD. Further screening studies in autistic individuals for the SNTG2 gene could elucidate the interaction between SNTG2 and neuroligins and ultimately the association with neurodevelopmental disorders.
Finally, another interesting gene that we have analysed and explored is the cholinergic receptor nicotinic alpha 7 (CHRNA7) gene, which is altered in six patients with ASD, ID and psychomotor developmental delay, two of whom have a parental origin. Our index case is an 11-year-old child with ASD, a speech delay with the inability to formulate complete sentences and express himself clearly, and a behavioural disorder characterized by immaturity in social interactions. We were able to detect a 459.63 kb duplication of maternal origin in region 15q13.3, which affects intron 3 to intron 9 and exon 4 to exon 10 of the CHRNA7 gene. The CHRNA7 gene encodes for the α7 subunit of nicotinic acetylcholine receptors (nAChRs). The nAChRs are highly expressed in the human brain and play a role in synaptic plasticity, learning and memory through calcium signalling at the neuronal level [62]. In particular, the CHRNA7 gene is listed as a strong candidate in the SFARI database with a score of 2.1, making it an important candidate gene for neurodevelopmental disorders. Indeed, deletions and duplications of the CHRNA7 gene have been reported in patients with neurological phenotypes such as cognitive deficits, epilepsy, language disorders, abnormal behaviour, ASD, ADHD or attention deficit disorder, mood disorders, and schizophrenia, with varying degrees of penetrance and severity, suggesting that the human brain is sensitive to CHRNA7 dosage [63]. It could be hypothesized that altered CHRNA7 dosage leads to changes in α7 protein abundance, which in turn alters calcium signalling in the corresponding areas of the central nervous system [64]. Studies conducted on iPSCs and human neural progenitor cells (NPCs) have shown that heterozygous deletion of the 15q13.3 region results in lower levels of CHRNA7 reaching the cell membrane, leading to a decrease in channel activity, while duplication leads to an increased expression of CHRNA7 with the accumulation of the misfolded protein and decreased trafficking of CHRNA7 to the membrane with impaired calcium flux [65].

5. Conclusions

In recent years, great progress has been made in elucidating the aetiology of NDDs. The considerable percentage of familial cases associated with a high concordance rate in monozygotic twins sustains a genetic basis of these disorders [66]. On the other hand, the functional significance of CNVs affecting individual genes and/or genomic regions is still notably incomplete, making diagnostic processes and genetic counselling difficult [67]. Consequently, the discovery of significant genes that can be considered causative or associated with neurodevelopmental disorders represents a necessary step in the diagnostic procedure, for a better understanding the pathophysiology of NDDs [68,69]. A deeper knowledge will indeed help to better classify the clinical features of the patients and the mechanisms that underlie their clinical manifestations.
In this study, a-CGH allowed the detection of VOUS containing interesting genes potentially related to the clinical characteristics found in the patients analysed at our facility. In particular, we have been able to observe how the genes CHRNA7, ANKS1B, ANKRD11, RBFOX1, ASTN2, GABRG3, SHANK2, KIF1A SETBP1, SNTG2, CTNNA2, TOP3B, CNTN4, CNTN5, and CNTN6 are involved in important phases of brain development and the correct functioning of the central nervous system. In particular, the presence of alterations in these genes in other patients with NDDs, availability of studies on animal models, and data from the literature make them interesting candidate genes for NDDs, essential to clarify and contribute to the knowledge of the molecular bases of these complex disorders.
For this reason, the results of our study, according to the literature, underline the significance and the usefulness of a-CGH in the genetic diagnosis. In fact, a-CGH offers significant sensitivity and resolution for the identification of submicroscopic chromosomal rearrangement and represents one of the most used techniques in daily practice [70]. However, since a-CGH still leaves some patients with NDDs undiagnosed, the interplay between a-CGH and NGS panels of known disease genes or exome sequencing (ES) can be considered an added value in aetiological diagnosis of these disorders [71,72,73,74]. Furthermore, in recent years, ES has been indicated in many studies as a first-line diagnostic test for NDDs, and for this reason, in the future, the integration of CNV calling algorithms into ES analysis could improve the characterization of complex phenotypes [75].
Considering the wide heterogeneity of NDDs, both in terms of behavioural and genetic features, further efforts are also needed to explore the possible role of environmental and epigenetic elements in phenotype modulation, and moreover, to better categorize each patient, future studies should necessarily be conducted in tight collaboration between neuropsychiatry divisions and genetics laboratories.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/genes15040427/s1, Table S1: All CNVs observed in our study.

Author Contributions

Conceptualization, B.L. and L.P.; methodology, A.R., I.L.M. and M.R.D.I.; software, A.R., I.L.M. and M.R.D.I.; validation, B.L. and L.P.; writing—original draft preparation, A.R., I.L.M. and M.R.D.I.; writing—review and editing, L.P. and B.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted according to Helsinki Declaration guidelines. Biological samples were obtained from patients as part of their diagnostic process. Thus, no approval by Ethics Committee was required. Patients gave their informed consent to the use of their samples anonymously at the time of samples withdrawal, as specified below.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study. This consent informs the patients about the study and the patients declare to have understood the purposes and expresses their informed consent, as well as authorization to use their biological material for diagnostic purposes, also for the purposes of scientific research, as well as to process the clinical and laboratory data derived, anonymously, exclusively for statistical-case studies and scientific purposes. Patients are described in an absolutely anonymous way without any picture or personal references, and thus, it is not possible to associate the data reported herein to the subjects they belong.

Data Availability Statement

Data are contained within the text. Further details can be required to corresponding authors.

Acknowledgments

The authors thank Iris Scala (Pediatria, A.O:U. Federico II), Gianfranca Auricchio (Neuropsichiatria Infantile, A.O.U. Federico II), Giancarlo Parenti (Pediatria, A.O.U. Federico II), Daniela Melis (Pediatria, A.O.U. Federico II), Claudia Santoro (Neuropsichiatria Infantile, A.O.U. Luigi Vanvitelli), Daniele De Brasi (Genetica Medica, A.O.R.N. Santobono Pausilipon), Francesca Felicia Operto (Neuropsichiatria Infantile, A.O.U. San Giovanni di Dio e Ruggi d’Aragona), and Carmelo Piscopo (Genetica Medica, A.O.R.N. Cardarelli) for providing the patients’ samples for analysis.

Conflicts of Interest

Authors Annaluisa Ranieri, Ilaria La Monica, Maria Rosaria Di Iorio, Barbara Lombardo and Lucio Pastore were employed by the company CEINGE-Biotecnologie Avanzate Franco Salvatore. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Bartnik, M.; Szczepanik, E.; Derwińska, K.; Wiśniowiecka-Kowalnik, B.; Gambin, T.; Sykulski, M.; Ziemkiewicz, K.; Keogonekdzior, M.; Gos, M.; Hoffman-Zacharska, D.; et al. Application of array comparative genomic hybridization in 102 patients with epilepsy and additional neurodevelopmental disorders. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 2012, 159, 760–771. [Google Scholar] [CrossRef] [PubMed]
  2. Mullin, A.P.; Gokhale, A.; Moreno-De-Luca, A.; Sanyal, S.; Waddington, J.L.; Faundez, V. Neurodevelopmental disorders: Mechanisms and boundary definitions from genomes, interactomes and proteomes. Transl. Psychiatry 2013, 3, e329-6. [Google Scholar] [CrossRef] [PubMed]
  3. Morris-Rosendahl, D.J.; Crocq, M.A. Neurodevelopmental disorders-the history and future of a diagnostic concept. Dialogues Clin. Neurosci. 2020, 22, 65–72. [Google Scholar] [CrossRef] [PubMed]
  4. Jeste Spurling, S. Neurodevelopmental behavioral and cognitive disorders. Contin. Lifelong Learn. Neurol. 2015, 21, 690. [Google Scholar] [CrossRef]
  5. De Felice, A.; Ricceri, L.; Venerosi, A.; Chiarotti, F.; Calamandrei, G. Multifactorial origin of neurodevelopmental disorders: Approaches to understanding complex etiologies. Toxics 2015, 3, 89–129. [Google Scholar] [CrossRef] [PubMed]
  6. Parenti, I.; Rabaneda, L.G.; Schoen, H.; Novarino, G. Neurodevelopmental Disorders: From Genetics to Functional Pathways. Trends Neurosci. 2020, 43, 608–621. [Google Scholar] [CrossRef] [PubMed]
  7. Wiśniowiecka-Kowalnik, B.; Nowakowska, B.A. Genetics and epigenetics of autism spectrum disorder—Current evidence in the field. J. Appl. Genet. 2019, 60, 37–47. [Google Scholar] [CrossRef] [PubMed]
  8. Ghiani, A.; Faundez, V. Cellular and Molecular Mechanisms of Neurodevelopmental Disorders. Neurosci. Res. 2017, 95, 1093–1096. [Google Scholar] [CrossRef]
  9. Cheroni, C.; Caporale, N.; Testa, G. Autism spectrum disorder at the crossroad between genes and environment: Contributions, convergences, and interactions in ASD developmental pathophysiology. Mol. Autism 2020, 11, 69. [Google Scholar] [CrossRef] [PubMed]
  10. Fakhro, K.A. Genomics of Autism. Adv. Neurobiol. 2020, 24, 83–96. [Google Scholar] [CrossRef] [PubMed]
  11. Ní Ghrálaigh, F.; Gallagher, L.; Lopez, L.M. Autism spectrum disorder genomics: The progress and potential of genomic technologies. Genomics 2020, 112, 5136–5142. [Google Scholar] [CrossRef] [PubMed]
  12. Lovrečić, L.; Rajar, P.; Volk, M.; Bertok, S.; Gnidovec Stražišar, B.; Osredkar, D.; Jekovec Vrhovšek, M.; Peterlin, B. Diagnostic efficacy and new variants in isolated and complex autism spectrum disorder using molecular karyotyping. J. Appl. Genet. 2018, 59, 179–185. [Google Scholar] [CrossRef] [PubMed]
  13. Yamamoto, T. Genomic aberrations associated with the pathophysiological mechanisms of neurodevelopmental disorders. Cells 2021, 10, 2317. [Google Scholar] [CrossRef] [PubMed]
  14. Molloy, C.J.; Quigley, C.; McNicholas, Á.; Lisanti, L.; Gallagher, L. A review of the cognitive impact of neurodevelopmental and neuropsychiatric associated copy number variants. Transl. Psychiatry 2023, 13, 116. [Google Scholar] [CrossRef] [PubMed]
  15. Lombardo, B.; D’Argenio, V.; Monda, E.; Vitale, A.; Caiazza, M.; Sacchetti, L.; Pastore, L.; Limongelli, G.; Frisso, G.; Mazzaccara, C. Genetic analysis resolves differential diagnosis of a familial syndromic dilated cardiomyopathy: A new case of Alström syndrome. Mol. Genet. Genomic Med. 2020, 8, e1260. [Google Scholar] [CrossRef] [PubMed]
  16. Iossa, S.; Costa, V.; Corvino, V.; Auletta, G.; Barruffo, L.; Cappellani, S.; Ceglia, C.; Cennamo, G.; D’Adamo, A.P.; D’Amico, A.; et al. Phenotypic and genetic characterization of a family carrying two Xq21.1-21.3 interstitial deletions associated with syndromic hearing loss. Mol. Cytogenet. 2015, 8, 18. [Google Scholar] [CrossRef] [PubMed]
  17. Veneruso, I.; Di Resta, C.; Tomaiuolo, R.; D’argenio, V. Current Updates on Expanded Carrier Screening: New Insights in the Omics Era. Medicina 2022, 58, 455. [Google Scholar] [CrossRef] [PubMed]
  18. De Angelis, C.; Nardelli, C.; Concolino, P.; Pagliuca, M.; Setaro, M.; De Paolis, E.; De Placido, P.; Forestieri, V.; Scaglione, G.L.; Ranieri, A.; et al. Case Report: Detection of a Novel Germline PALB2 Deletion in a Young Woman With Hereditary Breast Cancer: When the Patient’s Phenotype History Doesn’t Lie. Front. Oncol. 2021, 11, 602523. [Google Scholar] [CrossRef] [PubMed]
  19. Arnett, A.B.; Wang, T.; Eichler, E.E.; Bernier, R.A. Reflections on the genetics-first approach to advancements in molecular genetic and neurobiological research on neurodevelopmental disorders. J. Neurodev. Disord. 2021, 13, 24. [Google Scholar] [CrossRef] [PubMed]
  20. Carter, M.T.; Srour, M.; Au, P.Y.B.; Buhas, D.; Dyack, S.; Eaton, A.; Inbar-Feigenberg, M.; Howley, H.; Kawamura, A.; Lewis, S.M.E.; et al. Genetic and metabolic investigations for neurodevelopmental disorders: Position statement of the Canadian College of Medical Geneticists (CCMG). J. Med. Genet. 2023, 60, 523–532. [Google Scholar] [CrossRef] [PubMed]
  21. Lombardo, B.; Pagani, M.; De Rosa, A.; Nunziato, M.; Migliarini, S.; Garofalo, M.; Terrile, M.; D’Argenio, V.; Galbusera, A.; Nuzzo, T.; et al. D-aspartate oxidase gene duplication induces social recognition memory deficit in mice and intellectual disabilities in humans. Transl. Psychiatry 2022, 12, 305. [Google Scholar] [CrossRef] [PubMed]
  22. Carlsson, T.; Molander, F.; Taylor, M.J.; Jonsson, U.; Bölte, S. Early environmental risk factors for neurodevelopmental disorders—A systematic review of twin and sibling studies. Dev. Psychopathol. 2021, 33, 1448–1495. [Google Scholar] [CrossRef] [PubMed]
  23. Di Stefano, C.; Lombardo, B.; Fabbricatore, C.; Munno, C.; Caliendo, I.; Gallo, F.; Pastore, L. Oculo-facio-cardio-dental (OFCD) syndrome: The first Italian case of BCOR and co-occurring OTC gene deletion. Gene 2015, 559, 203–206. [Google Scholar] [CrossRef] [PubMed]
  24. Riggs, E.; Andersen, E.; Cherry, A.; Kantarci, S.; Kearney, H.; Patel, A.; Raca, G.; Ritter, D.; South, S.; Thorland, E.; et al. Technical standards for the interpretation and reporting of constitutional copy-number variants: A joint consensus recommendation of the American College of Medical Genetics and Genomics (ACMG) and the Clinical Genome Resource (ClinGen). Genet Med. 2020, 22, 245–257. [Google Scholar] [CrossRef] [PubMed]
  25. Miller, D.T.; Adam, M.P.; Aradhya, S.; Biesecker, L.G.; Brothman, A.R.; Carter, N.P.; Church, D.M.; Crolla, J.A.; Eichler, E.E.; Epstein, C.J.; et al. Consensus Statement: Chromosomal Microarray Is a First-Tier Clinical Diagnostic Test for Individuals with Developmental Disabilities or Congenital Anomalies. Am. J. Hum. Genet. 2010, 86, 749–764. [Google Scholar] [CrossRef] [PubMed]
  26. Heil, K.M.; Schaaf, C.P. The genetics of autism spectrum disorders—A guide for clinicians. Curr. Psychiatry Rep. 2013, 15, 334. [Google Scholar] [CrossRef] [PubMed]
  27. Watson, C.T.; Marques-Bonet, T.; Sharp, A.J.; Mefford, H.C. The Genetics of Microdeletion and Microduplication Syndromes: An Update. Annu. Rev. Genom. Hum. Genet. 2014, 15, 215–244. [Google Scholar] [CrossRef] [PubMed]
  28. Rylaarsdam, L.; Guemez-Gamboa, A. Genetic Causes and Modifiers of Autism Spectrum Disorder. Front. Cell Neurosci. 2019, 13, 385. [Google Scholar] [CrossRef] [PubMed]
  29. Wayhelova, M.; Smetana, J.; Vallova, V.; Hladilkova, E.; Filkova, H.; Hanakova, M.; Vilemova, M.; Nikolova, P.; Gromesova, B.; Gaillyova, R.; et al. The clinical benefit of array-based comparative genomic hybridization for detection of copy number variants in Czech children with intellectual disability and developmental delay. BMC Med. Genom. 2019, 12, 111. [Google Scholar] [CrossRef]
  30. Carbonell, A.U.; Cho, C.H.; Tindi, J.O.; Counts, P.A.; Bates, J.C.; Erdjument-Bromage, H.; Cvejic, S.; Iaboni, A.; Kvint, I.; Rosensaft, J.; et al. Haploinsufficiency in the ANKS1B gene encoding AIDA-1 leads to a neurodevelopmental syndrome. Nat. Commun. 2019, 10, 3529. [Google Scholar] [CrossRef]
  31. Jordan, B.A.; Fernholz, B.D.; Khatri, L.; Ziff, E.B. Activity-dependent AIDA-1 nuclear signaling regulates nucleolar numbers and protein synthesis in neurons. Nat. Neurosci. 2007, 10, 427–435. [Google Scholar] [CrossRef]
  32. Carbonell, A.U.; Freire-Cobo, C.; Deyneko, I.V.; Dobariya, S.; Erdjument-Bromage, H.; Clipperton-Allen, A.E.; Page, D.T.; Neubert, T.A.; Jordan, B.A. Comparing synaptic proteomes across five mouse models for autism reveals converging molecular similarities including deficits in oxidative phosphorylation and Rho GTPase signaling. Front. Aging Neurosci. 2023, 15, 1152562. [Google Scholar] [CrossRef] [PubMed]
  33. Enga, R.M.; Rice, A.C.; Weller, P.; Subler, M.A.; Lee, D.; Hall, C.P.; Windle, J.J.; Beardsley, P.M.; van den Oord, E.J.; McClay, J.L. Initial characterization of behavior and ketamine response in a mouse knockout of the post-synaptic effector gene Anks1b. Neurosci. Lett. 2017, 641, 26–32. [Google Scholar] [CrossRef] [PubMed]
  34. Uddin, M.; Tammimies, K.; Pellecchia, G.; Alipanahi, B.; Hu, P.; Wang, Z.; Pinto, D.; Lau, L.; Nalpathamkalam, T.; Marshall, C.R.; et al. Brain-expressed exons under purifying selection are enriched for de novo mutations in autism spectrum disorder. Nat. Genet. 2014, 46, 742–747. [Google Scholar] [CrossRef] [PubMed]
  35. Pinto, D.; Delaby, E.; Merico, D.; Barbosa, M.; Merikangas, A.; Klei, L.; Thiruvahindrapuram, B.; Xu, X.; Ziman, R.; Wang, Z.; et al. Convergence of genes and cellular pathways dysregulated in autism spectrum disorders. Am. J. Hum. Genet. 2014, 94, 677–694. [Google Scholar] [CrossRef] [PubMed]
  36. Cardo, L.F.; de la Fuente, D.C.; Li, M. Impaired neurogenesis and neural progenitor fate choice in a human stem cell model of SETBP1 disorder. Mol. Autism 2023, 14, 8. [Google Scholar] [CrossRef] [PubMed]
  37. Filges, I.; Shimojima, K.; Okamoto, N.; Röthlisberger, B.; Weber, P.; Huber, A.R.; Nishizawa, T.; Datta, A.N.; Miny, P.; Yamamoto, T. Reduced expression by SETBP1 haploinsufficiency causes developmental and expressive language delay indicating a phenotype distinct from Schinzel-Giedion syndrome. J. Med. Genet. 2011, 48, 117–122. [Google Scholar] [CrossRef] [PubMed]
  38. Schinzel, A.; Binkert, F.; Lillington, D.M.; Sands, M.; Stocks, R.J.; Lindenbaum, R.H.; Matthews, H.; Sheridan, H. Interstitial deletion of the long arm of chromosome 18, del(18)(q12.2q21.1): A report of three cases of an autosomal deletion with a mild phenotype. J. Med. Genet. 1991, 28, 352–355. [Google Scholar] [CrossRef] [PubMed]
  39. Bouquillon, S.; Andrieux, J.; Landais, E.; Duban-Bedu, B.; Boidein, F.; Lenne, B.; Vallée, L.; Leal, T.; Doco-Fenzy, M.; Delobel, B. A 5.3Mb deletion in chromosome 18q12.3 as the smallest region of overlap in two patients with expressive speech delay. Eur. J. Med. Genet. 2011, 54, 194–197. [Google Scholar] [CrossRef] [PubMed]
  40. Marseglia, G.; Scordo, M.R.; Pescucci, C.; Nannetti, G.; Biagini, E.; Scandurra, V.; Gerundino, F.; Magi, A.; Benelli, M.; Torricelli, F. 372 kb microdeletion in 18q12.3 causing SETBP1 haploinsufficiency associated with mild mental retardation and expressive speech impairment. Eur. J. Med. Genet. 2012, 55, 216–221. [Google Scholar] [CrossRef] [PubMed]
  41. Jansen, N.A.; Braden, R.O.; Srivastava, S.; Otness, E.F.; Lesca, G.; Rossi, M.; Nizon, M.; Bernier, R.A.; Quelin, C.; van Haeringen, A.; et al. Clinical delineation of SETBP1 haploinsufficiency disorder. Eur. J. Hum. Genet. 2021, 29, 1198–1205. [Google Scholar] [CrossRef] [PubMed]
  42. Behesti, H.; Fore, T.R.; Wu, P.; Horn, Z.; Leppert, M.; Hull, C.; Hattena, M.E. ASTN2 modultes synaptic strength by trafficking and degradation of surface proteins. Proc. Natl. Acad. Sci. USA 2018, 115, E9717–E9726. [Google Scholar] [CrossRef] [PubMed]
  43. Glessner, J.T.; Wang, K.; Cai, G.; Korvatska, O.; Kim, C.E.; Wood, S.; Zhang, H.; Estes, A.; Brune, C.W.; Bradfield, J.P.; et al. Autism genome-wide copy number variation reveals ubiquitin and neuronal genes. Nature 2009, 459, 569–572. [Google Scholar] [CrossRef] [PubMed]
  44. Lionel, A.C.; Tammimies, K.; Vaags, A.K.; Rosenfeld, J.A.; Ahn, J.W.; Merico, D.; Noor, A.; Runke, C.K.; Pillalamarri, V.K.; Carter, M.T.; et al. Disruption of the ASTN2/TRIM32 locus at 9q33.1 is a risk factor in males for autism spectrum disorders, ADHD and other neurodevelopmental phenotypes. Hum. Mol. Genet. 2014, 23, 2752–2768. [Google Scholar] [CrossRef] [PubMed]
  45. Ito, T.; Yoshida, M.; Aida, T.; Kushima, I.; Hiramatsu, Y.; Ono, M.; Yoshimi, A.; Tanaka, K.; Ozaki, N.; Noda, Y. Astrotactin 2 (ASTN2) regulates emotional and cognitive functions by affecting neuronal morphogenesis and monoaminergic systems. J. Neurochem. 2023, 165, 211–229. [Google Scholar] [CrossRef] [PubMed]
  46. Hartman, C.A.; Geurts, H.M.; Franke, B.; Buitelaar, J.K.; Rommelse, N.N.J. Changing ASD-ADHD symptom co-occurrence across the lifespan with adolescence as crucial time window: Illustrating the need to go beyond childhood. Neurosci. Biobehav. Rev. 2016, 71, 529–541. [Google Scholar] [CrossRef] [PubMed]
  47. Mikami, A.Y.; Miller, M.; Lerner, M.D. Social functioning in youth with attention-deficit/hyperactivity disorder and autism spectrum disorder: Transdiagnostic commonalities and differences. Clin. Psychol. Rev. 2019, 68, 54–70. [Google Scholar] [CrossRef] [PubMed]
  48. Hogart, A.; Nagarajan, R.; Patzel, K.; Yasui, D.; Lasalle, J. GABAA receptor genes are normally biallelically expressed in brain yet are subject to epigenetic dysregulation in autism-spectrum disorders. Hum. Mol. Genet. 2007, 16, 691–703. [Google Scholar] [CrossRef] [PubMed]
  49. Wang, L.; Li, J.; Shuang, M.; Lu, T.; Wang, Z.; Zhang, T.; Yue, W.; Jia, M.; Ruan, Y.; Liu, J.; et al. Association study and mutation sequencing of genes on chromosome 15q11-q13 identified GABRG3 as a susceptibility gene for autism in Chinese Han population. Transl. Psychiatry 2018, 8, 152. [Google Scholar] [CrossRef] [PubMed]
  50. DeLorey, T.M.; Sahbaie, P.; Hashemi, E.; Homanics, G.E.; Clark, J.D. Gabrb3 gene deficient mice exhibit impaired social and exploratory behaviors, deficits in non-selective attention and hypoplasia of cerebellar vermal lobules: A potential model of autism spectrum disorder. Behav. Brain Res. 2008, 187, 207–220. [Google Scholar] [CrossRef]
  51. Smith, S.E.P.; Zhou, Y.-D.; Zhang, G.; Jin, Z.; Stoppel, D.C.; Anderson, M.P. Increased gene dosage of Ube3a results in autism traits and decreased glutamate synaptic transmission in mice. Sci. Transl. Med. 2011, 3, 103ra97. [Google Scholar] [CrossRef] [PubMed]
  52. DeLorey, T.M.; Sahbaie, P.; Hashemi, E.; Li, W.W.; Salehi, A.; Clark, D.J. Somatosensory and sensorimotor consequences associated with the heterozygous disruption of the autism candidate gene, Gabrb3. Behav. Brain Res. 2011, 216, 36–45. [Google Scholar] [CrossRef] [PubMed]
  53. Menold, M.M.; Shao, Y.; Wolpert, C.M.; Donnelly, S.L.; Raiford, K.L.; Martin, E.R.; Ravan, S.A.; Abramson, R.K.; Wright, H.H.; Delong, G.R.; et al. Association Analysis of Chromosome 15 GABAA Receptor Subunit Genes in Autistic Disorder. J. Neurogenet. 2001, 15, 245–259. [Google Scholar] [CrossRef] [PubMed]
  54. Adak, P.; Sinha, S.; Banerjee, N. An Association Study of Gamma-Aminobutyric Acid Type A Receptor Variants and Susceptibility to Autism Spectrum Disorders. J. Autism Dev. Disord. 2021, 51, 4043–4053. [Google Scholar] [CrossRef] [PubMed]
  55. Coskunpinar, E.M.; Tur, S.; Cevher Binici, N.; Yazan Songür, C. Association of GABRG3, GABRB3, HTR2A gene variants with autism spectrum disorder. Gene 2023, 870, 147399. [Google Scholar] [CrossRef] [PubMed]
  56. Aguilera, C.; Hümmer, S.; Masanas, M.; Gabau, E.; Guitart, M.; Jeyaprakash, A.A.; Segura, M.F.; Santamaria, A.; Ruiz, A. The Novel KIF1A Missense Variant (R169T) Strongly Reduces Microtubule Stimulated ATPase Activity and Is Associated With NESCAV Syndrome. Front. Neurosci. 2021, 15, 618098. [Google Scholar] [CrossRef] [PubMed]
  57. Nair, A.; Greeny, A.; Rajendran, R.; Abdelgawad, M.A.; Ghoneim, M.M.; Raghavan, R.P.; Sudevan, S.T.; Mathew, B.; Kim, H. KIF1A-Associated Neurological Disorder: An Overview of a Rare Mutational Disease. Pharmaceuticals 2023, 16, 147. [Google Scholar] [CrossRef] [PubMed]
  58. Tomaselli, P.J.; Rossor, A.M.; Horga, A.; Laura, M.; Blake, J.C.; Houlden, H.; Reilly, M.M. A de novo dominant mutation in KIF1A associated with axonal neuropathy, spasticity and autism spectrum disorder. J. Peripher. Nerv. Syst. 2017, 22, 460–463. [Google Scholar] [CrossRef] [PubMed]
  59. Demily, C.; Lesca, G.; Poisson, A.; Till, M.; Barcia, G.; Chatron, N.; Sanlaville, D.; Munnich, A. Additive Effect of Variably Penetrant 22q11.2 Duplication and Pathogenic Mutations in Autism Spectrum Disorder: To Which Extent Does the Tree Hide the Forest? J. Autism Dev. Disord. 2018, 48, 2886–2889. [Google Scholar] [CrossRef]
  60. Rio, M.; Royer, G.; Gobin, S.; de Blois, M.; Ozilou, C.; Bernheim, A.; Nizon, M.; Munnich, A.; Bonnefont, J.P.; Romana, S.; et al. Monozygotic twins discordant for submicroscopic chromosomal anomalies in 2p25.3 region detected by array CGH. Clin. Genet. 2013, 84, 31–36. [Google Scholar] [CrossRef]
  61. Yamakawa, H.; Oyama, S.; Mitsuhashi, H.; Sasagawa, N.; Uchino, S.; Kohsaka, S.; Ishiura, S. Neuroligins 3 and 4X interact with syntrophin-γ2, and the interactions are affected by autism-related mutations. Biochem. Biophys. Res. Commun. 2007, 355, 41–46. [Google Scholar] [CrossRef] [PubMed]
  62. Felix, R.A.; Chavez, V.A.; Novicio, D.M.; Morley, B.J.; Portfors, C.V. Nicotinic acetylcholine receptor subunit α7-knockout mice exhibit degraded auditory temporal processing. J. Neurophysiol. 2019, 122, 451–465. [Google Scholar] [CrossRef] [PubMed]
  63. Budisteanu, M.; Papuc, S.M.; Streata, I.; Cucu, M.; Pirvu, A.; Serban-Sosoi, S.; Erbescu, A.; Andrei, E.; Iliescu, C.; Ioana, D.; et al. The phenotypic spectrum of 15q13.3 region duplications: Report of 5 patients. Genes 2021, 12, 1025. [Google Scholar] [CrossRef] [PubMed]
  64. Gillentine, M.A.; Schaaf, C.P. The Human Clinical Phenotypes of Altered CHRNA7 Copy Number. Biochem. Pharmacol. 2015, 97, 352–362. [Google Scholar] [CrossRef] [PubMed]
  65. Antony, I.; Narasimhan, M.; Shen, R.; Prakasam, R.; Kaushik, K.; Chapman, G.; Kroll, K.L. Duplication Versus Deletion through the Lens of 15q13.3: Clinical and Research Implications of Studying Copy Number Variants Associated with Neuropsychiatric Disorders in Induced Pluripotent Stem Cell-Derived Neurons. Stem. Cell Rev. Rep. 2023, 19, 639–650. [Google Scholar] [CrossRef] [PubMed]
  66. Lichtenstein, P.; Carlström, E.; Råstam, M.; Gillberg, C.; Anckarsäter, H. The genetics of autism spectrum disorders and related neuropsychiatric disorders in childhood. Am. J. Psychiatry 2010, 167, 1357–1363. [Google Scholar] [CrossRef] [PubMed]
  67. Ravel, J.M.; Renaud, M.; Muller, J.; Becker, A.; Renard, É.; Remen, T.; Lefort, G.; Dexheimer, M.; Jonveaux, P.; Leheup, B.; et al. Clinical utility of periodic reinterpretation of CNVs of uncertain significance: An 8-year retrospective study. Genome Med. 2023, 15, 39. [Google Scholar] [CrossRef]
  68. Zamora-Moratalla, A.; Martínez de Lagrán, M.; Dierssen, M. Neurodevelopmental disorders: 2021 update. Free Neuropathol. 2021, 2, 2–6. [Google Scholar] [CrossRef] [PubMed]
  69. Firdaus, Z.; Li, X. Unraveling the Genetic Landscape of Neurological Disorders: Insights into Pathogenesis, Techniques for Variant Identification, and Therapeutic Approaches. Int. J. Mol. Sci. 2024, 25, 2320. [Google Scholar] [CrossRef]
  70. Lombardo, B.; Ceglia, C.; Tarsitano, M.; Pierucci, I.; Salvatore, F.; Pastore, L. Identification of a deletion in the NDUFS4 gene using array-comparative genomic hybridization in a patient with suspected mitochondrial respiratory disease. Gene 2014, 535, 376–379. [Google Scholar] [CrossRef]
  71. Alotibi, R.S.; Sannan, N.S.; AlEissa, M.; Aldriwesh, M.G.; Al Tuwaijri, A.; Akiel, M.A.; Almutairi, M.; Alsamer, A.; Altharawi, N.; Aljawfan, G.; et al. The diagnostic yield of CGH and WES in neurodevelopmental disorders. Front. Pediatr. 2023, 11, 1133789. [Google Scholar] [CrossRef] [PubMed]
  72. Ranieri, A.; Veneruso, I.; La Monica, I.; Pascale, M.G.; Pastore, L.; D’argenio, V.; Lombardo, B. Combined aCGH and Exome Sequencing Analysis Improves Autism Spectrum Disorders Diagnosis: A Case Report. Medicina 2022, 58, 522. [Google Scholar] [CrossRef] [PubMed]
  73. Au, P.Y.B.; Eaton, A.; Dyment, D.A. Genetic Mechanisms of Neurodevelopmental Disorders, 1st ed.; Elsevier: Amsterdam, The Netherlands, 2020; Volume 173, ISBN 9780444641502. [Google Scholar]
  74. Wayhelova, M.; Vallova, V.; Broz, P.; Mikulasova, A.; Smetana, J.; Dynkova Filkova, H.; Machackova, D.; Handzusova, K.; Gaillyova, R.; Kuglik, P. Exome sequencing improves the molecular diagnostics of paediatric unexplained neurodevelopmental disorders. Orphanet J. Rare Dis. 2024, 19, 41. [Google Scholar] [CrossRef] [PubMed]
  75. Srivastava, S.; Love-Nichols, J.A.; Dies, K.A.; Ledbetter, D.H.; Martin, C.L.; Chung, W.K.; Firth, H.V.; Frazier, T.; Hansen, R.L.; Prock, L.; et al. Meta-analysis and multidisciplinary consensus statement: Exome sequencing is a first-tier clinical diagnostic test for individuals with neurodevelopmental disorders. Genet. Med. 2019, 21, 2413–2421. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Frequency of main diagnostic suspects reported on the request of our patients.
Figure 1. Frequency of main diagnostic suspects reported on the request of our patients.
Genes 15 00427 g001
Figure 2. Comparison of the number and significance of loss and gain identified in patients with neurodevelopmental disorders.
Figure 2. Comparison of the number and significance of loss and gain identified in patients with neurodevelopmental disorders.
Genes 15 00427 g002
Table 1. Pathogenic/Likely pathogenic CNVs associated with specific syndromes identified in patients with neurodevelopmental disorder. The number of patients analysed column indicates the number of patients in which the single alteration reported in the a-CGH results. The inheritance column reports the segregation/de novo nature of the single CNV reported in the result. ASD: autism spectrum disorder; NDD: neurodevelopmental disorder; ID: intellectual disability; ADHD: attention deficit hyperactivity disorder; ND: not determined.
Table 1. Pathogenic/Likely pathogenic CNVs associated with specific syndromes identified in patients with neurodevelopmental disorder. The number of patients analysed column indicates the number of patients in which the single alteration reported in the a-CGH results. The inheritance column reports the segregation/de novo nature of the single CNV reported in the result. ASD: autism spectrum disorder; NDD: neurodevelopmental disorder; ID: intellectual disability; ADHD: attention deficit hyperactivity disorder; ND: not determined.
Number of
Patients Analysed
Reason for ReferralArray-CGH ResultsCNV ClassificationInheritanceRelated
Syndrome/OMIM Number
7ID, dysmorphic features, psychomotor development delay, ASDarr 16p11.2 deletionPathogenic/Likely pathogenic1 maternal origin
2 De novo
4 ND
16p11.2 Deletion syndrome (#611913)
9 ADHD, ID, ASD, speech delayarr 16p11.2 duplicationPathogenic/Likely pathogenic3 De novo
6 ND
16p11.2 Duplication syndrome (#614671)
22 ID, psychomotor development delay, dysmorphic features, DiGeorge syndromearr 22q11.21 deletionPathogenic1 paternal origin
6 De novo
15 ND
DiGeorge syndrome (#188400)
Velocardiofacial syndrome (#192430)
15 Psychomotor development delay, learning deficit, growth delayarr 22q11.21 duplicationPathogenic1 maternal origin
4 De novo
10 ND
22q11.2 microduplication syndrome (#608363)
DiGeorge syndrome (#188400)
Velocardiofacial syndrome (#192430)
8 ASD, psychomotor development delayarr 15q11.2-q13.1 duplicationPathogenic2 De novo
6 ND
Duplication 15q11-q13 syndrome (#608636)
Angelman syndrome (#105830)
Prader–Willi syndrome (#176270)
10 Psychomotor development delay, ASD, ADHD, ID, psychomotor and growth delay, dysmorphic features, microcephalyarr 1q21.1-q21.2 deletionPathogenic/Likely pathogenic2 maternal origin
1 paternal origin
3 De novo
4 ND
1q21.1 Deletion syndrome (#612474)
Thrombocytopenia-absent
radius syndrome (#274000)
11 ASD, psychomotor development delay and epilepsyarr 1q21.1-q21.2 duplicationPathogenic/Likely pathogenic1 paternal origin
4 De novo
6 ND
1q21.1 Duplication syndrome (#612475)
6 ASD, psychomotor development delay, Becker muscular dystrophyarr Xp21.1 deletionPathogenic6 NDDuchenne muscular dystrophy (#310200)
Becker muscular dystrophy (#300376)
5 ADHD, speech delay, Charcot–Marie-Tooth disease type 1arr 17p12 duplicationPathogenic1 paternal origin
1 De novo
3 ND
Charcot–Marie-Tooth disease, type 1A (#118220)
4 NDD, psychomotor development delay, speech delayarr 17p11.2 deletionPathogenic1 paternal origin
3 ND
Smith–Magenis syndrome (#182290)
Table 2. Pathogenic/Likely pathogenic CNVs non-recurrent and not associated with syndromes identified in patients with neurodevelopmental disorder. The Number of Patients Analysed column indicates the number of patients in which the single alteration was reported in the a-CGH results. The Inheritance column reports the segregation/de novo nature of the single CNV reported in the result. ASD: autism spectrum disorder; ADHD: attention deficit hyperactivity disorder; ND: not determined.
Table 2. Pathogenic/Likely pathogenic CNVs non-recurrent and not associated with syndromes identified in patients with neurodevelopmental disorder. The Number of Patients Analysed column indicates the number of patients in which the single alteration was reported in the a-CGH results. The Inheritance column reports the segregation/de novo nature of the single CNV reported in the result. ASD: autism spectrum disorder; ADHD: attention deficit hyperactivity disorder; ND: not determined.
Number of Patients AnalysedReason for ReferralArray-CGH ResultsCNV ClassificationInheritance
1 ASDarr 11q13.1-q13.2 duplicationPathogenic/Likely pathogenicDe novo
1 ASDarr 5q14.3 duplicationPathogenic/Likely pathogenicND
3 ASD, psychomotor development delayarr Xq28 deletionPathogenic/Likely pathogenic3 ND
1 ADHDarr 13q21.32-q31.1 deletionPathogenic/Likely pathogenicND
9 ASD, psychomotor development delay, speech delayarr 2p16.3 deletionPathogenic/Likely pathogenic1 maternal origin
1 paternal origin
2 De novo
5 ND
1 Speech delayarr 20p12.3-p12.1 duplicationPathogenic/Likely pathogenicND
2 Psychomotor development delay, growth delay, ASDarr 15q26.3 deletionPathogenic/Likely pathogenic1 paternal origin
1 De novo
Table 3. The table show lists the most interesting variants of uncertain significance (VOUS) identified in our patients with neurodevelopmental disorders. The Number of Patients Analysed column indicates the number of patients in which the single alteration reported in the a-CGH results. The inheritance column reports the segregation/de novo nature of the single CNV reported in the result. These VOUS were selected on the basis of the score assigned to each gene in SFARI and the information present in the databases and literature. ASD: autism spectrum disorder; NDD: neurodevelopmental disorder; ID: intellectual disability; ADHD: attention deficit hyperactivity disorder; ND: not determined.
Table 3. The table show lists the most interesting variants of uncertain significance (VOUS) identified in our patients with neurodevelopmental disorders. The Number of Patients Analysed column indicates the number of patients in which the single alteration reported in the a-CGH results. The inheritance column reports the segregation/de novo nature of the single CNV reported in the result. These VOUS were selected on the basis of the score assigned to each gene in SFARI and the information present in the databases and literature. ASD: autism spectrum disorder; NDD: neurodevelopmental disorder; ID: intellectual disability; ADHD: attention deficit hyperactivity disorder; ND: not determined.
Number of
Patients
Analysed
Clinical DiagnosisArray-CGH ResultsCNV
Classification
InheritanceGene OMIM Number and Score SFARIBiological Function
12 ASD, psychomotor
developmental delay, speech delay
arr 3p26.3-p26.2 deletionVOUS1 paternal origin
2 De novo
9 ND
CNTN4 (*607280) Strong candidateEssential for promoting dendrite growth and dendritic spine formation
1 ASDarr 9q33.1 deletionVOUSDe novoASTN2 (*612856) Strong candidateA central role in neural-glial adhesion during neuronal migration and synaptic strength through the trafficking and degradation of surface proteins
1 NDDarr 18q12.3 duplicationVOUSNDSETBP1 (*611060) High confidenceRole in controlling forebrain progenitor expansion and neurogenic
differentiation
6 ASD, epilepsy,
psychomotor developmental delay
arr 15q13.3 duplicationVOUS1 maternal origin
1 paternal origin
4 ND
CHRNA7 (*118511) Strong candidateInvolved in signal transmission at
synapses
1 ASDarr 11q13.3-q13.4 deletionVOUSNDSHANK2 (*603290) High confidenceMolecular scaffolds in the postsynaptic density of excitatory synapses
1 ASDarr 2q37.3 duplicationVOUSPaternal originKIF1A (*601255) Strong syndromic candidateMicrotubule-dependent motor protein responsible for fast anterograde transport of synaptic vesicle precursors in neurons
3 ASDarr 15q12 deletionVOUS3 NDGABRG3 (*600233) Strong candidateMajor inhibitory neurotransmitter in the mammalian brain
3 ASDarr 3p26.3 duplicationVOUS1 De novo
2 ND
CNTN6 (*607220) Strong candidateRole in the formation of axon connections in the developing nervous system
1 ASD, epilepsyarr 16q24.3 deletionVOUSNDANKRD11 (*611192) High confidence
syndromic
Regulates pyramidal neuron migration and dendritic differentiation in the developing mouse cerebral cortex
3 ASD, IDarr 11q22.1 deletionVOUS1 maternal origin
2 ND
CNTN5 (*607219) Strong candidateRole in the formation of axon connections in the developing nervous system
5 ASD, ID, ADHD
dysmorphic features
arr 12q23.1 deletionVOUS1 maternal origin
1 De novo
3 ND
ANKS1B (*607815)
Strong syndromic candidate
Abundant in neuronal synapses and important role in normal brain development
8 ASD, ID, psychomotor developmental delay,
speech delay
arr 2p25.3 duplicationVOUS1 maternal origin
2 paternal origin
1 De novo
4 ND
SNTG2 (*608715) Strong candidateA scaffolding protein that interacts with neuroligins as crucial binding factor at inhibitory synapses
7 ASD, ID, psychomotor developmental delay,
speech delay,
dysmorphic features
arr 22q11.2 duplicationVOUS1 paternal origin
6 ND
TOP3B (*603582) Strong candidateControls the DNA state during transcription by catalysing transient breakage and junction of a single strand of DNA
3 ASD, psychomotor
developmental delay
arr 2p12 deletionVOUS1 maternal origin
1 paternal origin
1 ND
CTNNA2 (*114025) SyndromicInvolved in the negative regulation of actin nucleation and in the regulation of neuron migration and the development of neuron projection
12 ASD, ID, ADHDarr 16p13.3 deletionVOUS1 paternal origin
2 De novo
9 ND
RBFOX1 (*605104) Strong candidateRole in the regulation of the alternative splicing of large neuronal gene networks
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

Ranieri, A.; La Monica, I.; Di Iorio, M.R.; Lombardo, B.; Pastore, L. Genetic Alterations in a Large Population of Italian Patients Affected by Neurodevelopmental Disorders. Genes 2024, 15, 427. https://doi.org/10.3390/genes15040427

AMA Style

Ranieri A, La Monica I, Di Iorio MR, Lombardo B, Pastore L. Genetic Alterations in a Large Population of Italian Patients Affected by Neurodevelopmental Disorders. Genes. 2024; 15(4):427. https://doi.org/10.3390/genes15040427

Chicago/Turabian Style

Ranieri, Annaluisa, Ilaria La Monica, Maria Rosaria Di Iorio, Barbara Lombardo, and Lucio Pastore. 2024. "Genetic Alterations in a Large Population of Italian Patients Affected by Neurodevelopmental Disorders" Genes 15, no. 4: 427. https://doi.org/10.3390/genes15040427

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