Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 36
1.
Acta Neuropathol Commun ; 12(1): 62, 2024 Apr 18.
Article En | MEDLINE | ID: mdl-38637827

BACKGROUND: Helsmoortel-Van der Aa syndrome is a neurodevelopmental disorder in which patients present with autism, intellectual disability, and frequent extra-neurological features such as feeding and gastrointestinal problems, visual impairments, and cardiac abnormalities. All patients exhibit heterozygous de novo nonsense or frameshift stop mutations in the Activity-Dependent Neuroprotective Protein (ADNP) gene, accounting for a prevalence of 0.2% of all autism cases worldwide. ADNP fulfills an essential chromatin remodeling function during brain development. In this study, we investigated the cerebellum of a died 6-year-old male patient with the c.1676dupA/p.His559Glnfs*3 ADNP mutation. RESULTS: The clinical presentation of the patient was representative of the Helsmoortel-Van der Aa syndrome. During his lifespan, he underwent two liver transplantations after which the child died because of multiple organ failure. An autopsy was performed, and various tissue samples were taken for further analysis. We performed a molecular characterization of the cerebellum, a brain region involved in motor coordination, known for its highest ADNP expression and compared it to an age-matched control subject. Importantly, epigenome-wide analysis of the ADNP cerebellum identified CpG methylation differences and expression of multiple pathways causing neurodevelopmental delay. Interestingly, transcription factor motif enrichment analysis of differentially methylated genes showed that the ADNP binding motif was the most significantly enriched. RNA sequencing of the autopsy brain further identified downregulation of the WNT signaling pathway and autophagy defects as possible causes of neurodevelopmental delay. Ultimately, label-free quantification mass spectrometry identified differentially expressed proteins involved in mitochondrial stress and sirtuin signaling pathways amongst others. Protein-protein interaction analysis further revealed a network including chromatin remodelers (ADNP, SMARCC2, HDAC2 and YY1), autophagy-related proteins (LAMP1, BECN1 and LC3) as well as a key histone deacetylating enzyme SIRT1, involved in mitochondrial energy metabolism. The protein interaction of ADNP with SIRT1 was further biochemically validated through the microtubule-end binding proteins EB1/EB3 by direct co-immunoprecipitation in mouse cerebellum, suggesting important mito-epigenetic crosstalk between chromatin remodeling and mitochondrial energy metabolism linked to autophagy stress responses. This is further supported by mitochondrial activity assays and stainings in patient-derived fibroblasts which suggest mitochondrial dysfunctions in the ADNP deficient human brain. CONCLUSION: This study forms the baseline clinical and molecular characterization of an ADNP autopsy cerebellum, providing novel insights in the disease mechanisms of the Helsmoortel-Van der Aa syndrome. By combining multi-omic and biochemical approaches, we identified a novel SIRT1-EB1/EB3-ADNP protein complex which may contribute to autophagic flux alterations and impaired mitochondrial metabolism in the Helsmoortel-Van der Aa syndrome and holds promise as a new therapeutic target.


Autistic Disorder , Intellectual Disability , Male , Child , Animals , Mice , Humans , Intellectual Disability/genetics , Autistic Disorder/genetics , Sirtuin 1/genetics , Sirtuin 1/metabolism , Genes, Mitochondrial , Homeodomain Proteins/genetics , Cerebellum/metabolism , Autopsy , Methylation , Nerve Tissue Proteins/metabolism , DNA-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Mol Genet Genomic Med ; 9(9): e1768, 2021 09.
Article En | MEDLINE | ID: mdl-34402213

BACKGROUND: Primary microcephaly (PM) is defined as a significant reduction in occipitofrontal circumference (OFC) of prenatal onset. Clinical and genetic heterogeneity of PM represents a diagnostic challenge. METHODS: We performed detailed phenotypic and genomic analyses in a large cohort (n = 169) of patients referred for PM and could establish a molecular diagnosis in 38 patients. RESULTS: Pathogenic variants in ASPM and WDR62 were the most frequent causes in non-consanguineous patients in our cohort. In consanguineous patients, microarray and targeted gene panel analyses reached a diagnostic yield of 67%, which contrasts with a much lower rate in non-consanguineous patients (9%). Our series includes 11 novel pathogenic variants and we identify novel candidate genes including IGF2BP3 and DNAH2. We confirm the progression of microcephaly over time in affected children. Epilepsy was an important associated feature in our PM cohort, affecting 34% of patients with a molecular confirmation of the PM diagnosis, with various degrees of severity and seizure types. CONCLUSION: Our findings will help to prioritize genomic investigations, accelerate molecular diagnoses, and improve the management of PM patients.


Consanguinity , Epilepsy/genetics , Genotype , Microcephaly/genetics , Phenotype , Cell Cycle Proteins/genetics , Child , Epilepsy/epidemiology , Epilepsy/pathology , Female , Gene Frequency , Genetic Heterogeneity , Humans , Incidence , Male , Microcephaly/complications , Microcephaly/pathology , Nerve Tissue Proteins/genetics
3.
Nat Commun ; 11(1): 4932, 2020 10 01.
Article En | MEDLINE | ID: mdl-33004838

Most genes associated with neurodevelopmental disorders (NDDs) were identified with an excess of de novo mutations (DNMs) but the significance in case-control mutation burden analysis is unestablished. Here, we sequence 63 genes in 16,294 NDD cases and an additional 62 genes in 6,211 NDD cases. By combining these with published data, we assess a total of 125 genes in over 16,000 NDD cases and compare the mutation burden to nonpsychiatric controls from ExAC. We identify 48 genes (25 newly reported) showing significant burden of ultra-rare (MAF < 0.01%) gene-disruptive mutations (FDR 5%), six of which reach family-wise error rate (FWER) significance (p < 1.25E-06). Among these 125 targeted genes, we also reevaluate DNM excess in 17,426 NDD trios with 6,499 new autism trios. We identify 90 genes enriched for DNMs (FDR 5%; e.g., GABRG2 and UIMC1); of which, 61 reach FWER significance (p < 3.64E-07; e.g., CASZ1). In addition to doubling the number of patients for many NDD risk genes, we present phenotype-genotype correlations for seven risk genes (CTCF, HNRNPU, KCNQ3, ZBTB18, TCF12, SPEN, and LEO1) based on this large-scale targeted sequencing effort.


Genetic Predisposition to Disease , Neurodevelopmental Disorders/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , CCCTC-Binding Factor/genetics , Case-Control Studies , Cohort Studies , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Female , Genetic Association Studies , Heterogeneous-Nuclear Ribonucleoprotein U/genetics , High-Throughput Nucleotide Sequencing , Humans , KCNQ3 Potassium Channel/genetics , Male , Mutation , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/genetics
5.
Eur J Paediatr Neurol ; 24: 100-104, 2020 Jan.
Article En | MEDLINE | ID: mdl-31926845

Many pathways have been involved in pathophysiology of the fragile X syndrome, one of the more frequent genetic causes of intellectual disability and autism. This review highlights the recent insights in the role the abnormalities in the GABAergic system play in the disorder. Since the initial observations showed that the expression of specific subunits of the GABA(A) receptor were underexpressed in the fragile X knockout mouse model more than a decade ago, evidence has accumulated that the expression of approximately half of the GABAergic system is compromised in multiple species, including in fragile X patients. Functional consequences of the GABAergic deficiencies could be measured using whole-cell voltage clamp recordings. Pharmalogical treatment with agonist of the receptor was been able to restore several behavioral deficits in the fragile X mouse model, including seizures, marble burying and, in part, prepulse inhibition. Trials in patients with the same agonist have demonstrated encouraging post-hoc results in the most severely affected patients, although no effect could be demonstrated in the patient group as a whole. In conclusion, there can be little doubt that the GABAergic system is compromised in the fragile X syndrome and that these abnormalities contribute to the clinical abnormalities observed. However, at the moment the difference in treatment effectiveness of agonist of the receptor in animal models as opposed to in patients remains unexplained.


Fragile X Syndrome/genetics , Fragile X Syndrome/physiopathology , Receptors, GABA/genetics , Animals , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , Humans , Mice
6.
Am J Hum Genet ; 105(3): 493-508, 2019 09 05.
Article En | MEDLINE | ID: mdl-31447100

Histones mediate dynamic packaging of nuclear DNA in chromatin, a process that is precisely controlled to guarantee efficient compaction of the genome and proper chromosomal segregation during cell division and to accomplish DNA replication, transcription, and repair. Due to the important structural and regulatory roles played by histones, it is not surprising that histone functional dysregulation or aberrant levels of histones can have severe consequences for multiple cellular processes and ultimately might affect development or contribute to cell transformation. Recently, germline frameshift mutations involving the C-terminal tail of HIST1H1E, which is a widely expressed member of the linker histone family and facilitates higher-order chromatin folding, have been causally linked to an as-yet poorly defined syndrome that includes intellectual disability. We report that these mutations result in stable proteins that reside in the nucleus, bind to chromatin, disrupt proper compaction of DNA, and are associated with a specific methylation pattern. Cells expressing these mutant proteins have a dramatically reduced proliferation rate and competence, hardly enter into the S phase, and undergo accelerated senescence. Remarkably, clinical assessment of a relatively large cohort of subjects sharing these mutations revealed a premature aging phenotype as a previously unrecognized feature of the disorder. Our findings identify a direct link between aberrant chromatin remodeling, cellular senescence, and accelerated aging.


Cellular Senescence/physiology , Histones/physiology , Aneuploidy , Cell Nucleolus/metabolism , Child , Chromatin/metabolism , DNA Methylation , Female , Histones/chemistry , Humans , Infant , Male , Middle Aged
9.
Am J Med Genet A ; 170(3): 670-5, 2016 Mar.
Article En | MEDLINE | ID: mdl-26842493

We report on 19 individuals with a recurrent de novo c.607C>T mutation in PACS1. This specific mutation gives rise to a recognizable intellectual disability syndrome. There is a distinctive facial appearance (19/19), characterized by full and arched eyebrows, hypertelorism with downslanting palpebral fissures, long eye lashes, ptosis, low set and simple ears, bulbous nasal tip, wide mouth with downturned corners and a thin upper lip with an unusual "wavy" profile, flat philtrum, and diastema of the teeth. Intellectual disability, ranging from mild to moderate, was present in all. Hypotonia is common in infancy (8/19). Seizures are frequent (12/19) and respond well to anticonvulsive medication. Structural malformations are common, including heart (10/19), brain (12/16), eye (10/19), kidney (3/19), and cryptorchidism (6/12 males). Feeding dysfunction is presenting in infancy with failure to thrive (5/19), gastroesophageal reflux (6/19), and gastrostomy tube placement (4/19). There is persistence of oral motor dysfunction. We provide suggestions for clinical work-up and management and hope that the present study will facilitate clinical recognition of further cases.


Abnormalities, Multiple/genetics , Intellectual Disability/genetics , Point Mutation , Seizures/genetics , Vesicular Transport Proteins/genetics , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/drug therapy , Abnormalities, Multiple/pathology , Adolescent , Anticonvulsants/therapeutic use , Child , Child, Preschool , Facies , Failure to Thrive/diagnosis , Failure to Thrive/drug therapy , Failure to Thrive/genetics , Failure to Thrive/pathology , Female , Gene Expression , Humans , Intellectual Disability/diagnosis , Intellectual Disability/drug therapy , Intellectual Disability/pathology , Male , Muscle Hypotonia/diagnosis , Muscle Hypotonia/drug therapy , Muscle Hypotonia/genetics , Muscle Hypotonia/pathology , Seizures/diagnosis , Seizures/drug therapy , Seizures/pathology , Severity of Illness Index , Syndrome , Young Adult
10.
Eur J Hum Genet ; 24(5): 652-9, 2016 May.
Article En | MEDLINE | ID: mdl-26306646

The Koolen-de Vries syndrome (KdVS; OMIM #610443), also known as the 17q21.31 microdeletion syndrome, is a clinically heterogeneous disorder characterised by (neonatal) hypotonia, developmental delay, moderate intellectual disability, and characteristic facial dysmorphism. Expressive language development is particularly impaired compared with receptive language or motor skills. Other frequently reported features include social and friendly behaviour, epilepsy, musculoskeletal anomalies, congenital heart defects, urogenital malformations, and ectodermal anomalies. The syndrome is caused by a truncating variant in the KAT8 regulatory NSL complex unit 1 (KANSL1) gene or by a 17q21.31 microdeletion encompassing KANSL1. Herein we describe a novel cohort of 45 individuals with KdVS of whom 33 have a 17q21.31 microdeletion and 12 a single-nucleotide variant (SNV) in KANSL1 (19 males, 26 females; age range 7 months to 50 years). We provide guidance about the potential pitfalls in the laboratory testing and emphasise the challenges of KANSL1 variant calling and DNA copy number analysis in the complex 17q21.31 region. Moreover, we present detailed phenotypic information, including neuropsychological features, that contribute to the broad phenotypic spectrum of the syndrome. Comparison of the phenotype of both the microdeletion and SNV patients does not show differences of clinical importance, stressing that haploinsufficiency of KANSL1 is sufficient to cause the full KdVS phenotype.


Abnormalities, Multiple/diagnosis , Intellectual Disability/diagnosis , Nuclear Proteins/genetics , Phenotype , Polymorphism, Single Nucleotide , Abnormalities, Multiple/genetics , Adolescent , Adult , Child , Chromosome Deletion , Chromosomes, Human, Pair 17/genetics , Female , Humans , Intellectual Disability/genetics , Male , Middle Aged
11.
Hum Mutat ; 36(11): 1112, 2015 Nov.
Article En | MEDLINE | ID: mdl-26457590

The original article to which this Erratum refers was published in Human Mutation 36(6):593­598(DOI:10.1002/humu22795).The authors realized that a co-author, Nuria C. Bramswig, was left off of the title page of this article at the time of submission. This erratum serves to correct this error by including Dr. Bramswig and Dr. Bramswig's institution in the title page information.The authors regret the error.

12.
Eur J Med Genet ; 58(10): 503-8, 2015 Oct.
Article En | MEDLINE | ID: mdl-26327614

Recurrent rearrangements of chromosome 1q21.1 that occur as a consequence of non-allelic homologous recombination (NAHR) show considerable variability in phenotypic expression and penetrance. Chromosome 1q21.1 deletions (OMIM 612474) have been associated with microcephaly, intellectual disability, autism, schizophrenia, cardiac abnormalities and cataracts. Phenotypic features in individuals with 1q21.1 duplications (OMIM 612475) include macrocephaly, learning difficulties, developmental delay, intellectual disability and mild dysmorphic features. Half of these patients show autistic behavior. For the first time, we describe five patients, including monozygotic twins, with a triplication of the 1q21.1 chromosomal segment. Facial features common to all patients include a high, broad forehead; a flat and broad nasal bridge; long, downslanted palpebral fissures and dysplastic, low-set ears. Likely associated features include macrocephaly and increased weight. We observed that the triplications arose through different mechanisms in the patients: it was de novo in one patient, inherited from a triplication carrier in two cases, while the father of the twins is a 1q21.1 duplication carrier. The de novo triplication contained copies of both maternal alleles, suggesting it was generated by a combination of inter- and intrachromosomal recombination.


Chromosomes, Human, Pair 1/genetics , Craniofacial Abnormalities/genetics , Megalencephaly/genetics , Overweight/genetics , Trisomy , Child , Child, Preschool , Craniofacial Abnormalities/diagnosis , Female , Humans , Infant , Male , Megalencephaly/diagnosis , Overweight/diagnosis , Syndrome , Twins, Monozygotic/genetics
13.
PLoS One ; 10(7): e0131486, 2015.
Article En | MEDLINE | ID: mdl-26222316

Over the last several years, evidence has accumulated that the GABAA receptor is compromised in animal models for fragile X syndrome (FXS), a common hereditary form of intellectual disability. In mouse and fly models, agonists of the GABAA receptor were able to rescue specific consequences of the fragile X mutation. Here, we imaged and quantified GABAA receptors in vivo in brain of fragile X patients using Positron Emission Topography (PET) and [11C]flumazenil, a known high-affinity and specific ligand for the benzodiazepine site of GABAA receptors. We measured regional GABAA receptor availability in 10 fragile X patients and 10 control subjects. We found a significant reduction of on average 10% in GABAA receptor binding potential throughout the brain in fragile X patients. In the thalamus, the brain region showing the largest difference, the GABAA receptor availability was even reduced with 17%. This is one of the first reports of a PET study of human fragile X brain and directly demonstrates that the GABAA receptor availability is reduced in fragile X patients. The study reinforces previous hypotheses that the GABAA receptor is a potential target for rational pharmacological treatment of fragile X syndrome.


Brain , Flumazenil/administration & dosage , Fragile X Syndrome , Positron-Emission Tomography , Receptors, GABA-A/metabolism , Adolescent , Adult , Animals , Brain/diagnostic imaging , Brain/metabolism , Female , Fragile X Syndrome/diagnostic imaging , Fragile X Syndrome/metabolism , Humans , Male , Mice , Middle Aged , Radiography
14.
BMC Med Genet ; 16: 51, 2015 Jul 21.
Article En | MEDLINE | ID: mdl-26189493

BACKGROUND: Identification of the first de novo mutation in potassium voltage-gated channel, shal-related subfamily, member 3 (KCND3) in a patient with complex early onset cerebellar ataxia in order to expand the genetic and phenotypic spectrum. METHODS: Whole exome sequencing in a cerebellar ataxia patient and subsequent immunocytochemistry, immunoblotting and patch clamp assays of the channel were performed. RESULTS: A de novo KCND3 mutation (c.877_885dupCGCGTCTTC; p.Arg293_Phe295dup) was found duplicating the RVF motif and thereby adding an extra positive charge to voltage-gated potassium 4.3 (Kv4.3) in the voltage-sensor domain causing a severe shift of the voltage-dependence gating to more depolarized voltages. The patient displayed a severe phenotype with early onset cerebellar ataxia complicated by intellectual disability, epilepsy, attention deficit hyperactivity disorder, strabismus, oral apraxia and joint hyperlaxity. CONCLUSIONS: We identified a de novo KCND3 mutation causing the most marked change in Kv4.3's channel properties reported so far, which correlated with a severe and unique spinocerebellar ataxia (SCA) type 19/22 disease phenotype.


Apraxias/genetics , Intellectual Disability/genetics , Shal Potassium Channels/genetics , Spinocerebellar Degenerations/genetics , Base Sequence , Cell Line, Tumor , Child , Epilepsy/genetics , Genetic Markers , HeLa Cells , Humans , Male , Patch-Clamp Techniques , Sequence Analysis, DNA
16.
Hum Mutat ; 36(6): 593-8, 2015 Jun.
Article En | MEDLINE | ID: mdl-25824905

Adams-Oliver syndrome (AOS) is characterized by the association of aplasia cutis congenita with terminal transverse limb defects, often accompanied by additional cardiovascular or neurological features. Both autosomal-dominant and autosomal-recessive disease transmission have been observed, with recent gene discoveries indicating extensive genetic heterogeneity. Mutations of the DOCK6 gene were first described in autosomal-recessive cases of AOS and only five DOCK6-related families have been reported to date. Recently, a second type of autosomal-recessive AOS has been attributed to EOGT mutations in three consanguineous families. Here, we describe the identification of 13 DOCK6 mutations, the majority of which are novel, across 10 unrelated individuals from a large cohort comprising 47 sporadic cases and 31 AOS pedigrees suggestive of autosomal-recessive inheritance. DOCK6 mutations were strongly associated with structural brain abnormalities, ocular anomalies, and intellectual disability, thus suggesting that DOCK6-linked disease represents a variant of AOS with a particularly poor prognosis.


Brain/abnormalities , Ectodermal Dysplasia/diagnosis , Ectodermal Dysplasia/genetics , Eye Abnormalities/genetics , Genes, Recessive , Genetic Association Studies , Guanine Nucleotide Exchange Factors/genetics , Limb Deformities, Congenital/diagnosis , Limb Deformities, Congenital/genetics , Mutation , Scalp Dermatoses/congenital , Adolescent , Brain/pathology , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Magnetic Resonance Imaging , Male , Scalp Dermatoses/diagnosis , Scalp Dermatoses/genetics , Tomography, X-Ray Computed , Young Adult
17.
Eur J Hum Genet ; 23(6): 803-7, 2015 Jun.
Article En | MEDLINE | ID: mdl-25205402

AUTS2 syndrome is characterized by low birth weight, feeding difficulties, intellectual disability, microcephaly and mild dysmorphic features. All affected individuals thus far were caused by chromosomal rearrangements, variants at the base pair level disrupting AUTS2 have not yet been described. Here we present the full clinical description of two affected men with intragenic AUTS2 variants (one two-base pair deletion in exon 7 and one deletion of exon 6). Both variants are de novo and are predicted to cause a frameshift of the full-length transcript but are unlikely to affect the shorter 3' transcript starting in exon 9. The similarities between the phenotypes of both men are striking and further support that AUTS2 syndrome is a single gene disorder.


Gene Deletion , Intellectual Disability/genetics , Microcephaly/genetics , Polymorphism, Genetic , Proteins/genetics , Cytoskeletal Proteins , Exons , Frameshift Mutation , Humans , Intellectual Disability/diagnosis , Male , Phenotype , Syndrome , Transcription Factors , Young Adult
18.
Am J Med Genet A ; 164A(12): 3061-8, 2014 Dec.
Article En | MEDLINE | ID: mdl-25256099

Small interstitial deletions affecting chromosome region 3p25.3 have been reported in only five patients so far, four of them with overlapping telomeric microdeletions 3p25.3 and variable features of 3p- syndrome, and one patient with a small proximal microdeletion and a distinct phenotype with intellectual disability (ID) and multiple congenital anomalies. Here we report on three novel patients with overlapping proximal microdeletions 3p25.3 of 1.1-1.5 Mb in size showing a consistent non-3p- phenotype with ID, epilepsy/EEG abnormalities, poor speech, ataxia and stereotypic hand movements. The smallest region of overlap contains two genes encoding sodium- and chloride-dependent GABA transporters which have not been associated with this disease phenotype in humans so far. The protein function, the phenotype in transporter deficient animal models and the effects of specific pharmacological transporter inhibition in mice and humans provide evidence that these GABA transporters are plausible candidates for seizures/EEG abnormalities, ataxia and ID in this novel group of patients. A fourth novel patient deleted for a 3.16 Mb region, both telomeric and centromeric to 3p25.3, confirms that the telomeric segment is critical for the 3p- syndrome phenotype. Finally, a region of 643 kb is suggested to harbor one or more genes causative for polydactyly which is part of the 3p- syndrome.


Abnormalities, Multiple/genetics , Chromosome Aberrations , Chromosomes, Human, Pair 3/genetics , Epilepsy/genetics , GABA Plasma Membrane Transport Proteins/deficiency , Intellectual Disability/genetics , Abnormalities, Multiple/pathology , Female , GABA Plasma Membrane Transport Proteins/genetics , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Male , Stereotyped Behavior
19.
Am J Med Genet C Semin Med Genet ; 166C(3): 315-26, 2014 Sep.
Article En | MEDLINE | ID: mdl-25169753

Mutations in ADNP were recently identified as a frequent cause of syndromic autism, characterized by deficits in social communication and interaction and restricted, repetitive behavioral patterns. Based on its functional domains, ADNP is a presumed transcription factor. The gene interacts closely with the SWI/SNF complex by direct and experimentally verified binding of its C-terminus to three of its core components. A detailed and systematic clinical assessment of the symptoms observed in our patients allows a detailed comparison with the symptoms observed in other SWI/SNF disorders. While the mutational mechanism of the first 10 patients identified suggested a gain of function mechanism, an 11th patient reported here is predicted haploinsufficient. The latter observation may raise hope for therapy, as addition of NAP, a neuroprotective octapeptide named after the first three amino acids of the sequence NAPVSPIQ, has been reported by others to ameliorate some of the cognitive abnormalities observed in a knockout mouse model. It is concluded that detailed clinical and molecular studies on larger cohorts of patients are necessary to establish a better insight in the genotype phenotype correlation and in the mutational mechanism.


Autistic Disorder/genetics , Homeodomain Proteins/genetics , Mutation , Nerve Tissue Proteins/genetics , Abnormalities, Multiple/genetics , Animals , Autistic Disorder/etiology , Child, Preschool , DNA Helicases/genetics , DNA Helicases/metabolism , Face/abnormalities , Hand Deformities, Congenital/genetics , Haploinsufficiency/genetics , Humans , Infant , Intellectual Disability/genetics , Mice, Knockout , Micrognathism/genetics , Neck/abnormalities , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligopeptides/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism
...