Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Hum Genet ; 110(7): 1098-1109, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37301203

ABSTRACT

Although the best-known spinocerebellar ataxias (SCAs) are triplet repeat diseases, many SCAs are not caused by repeat expansions. The rarity of individual non-expansion SCAs, however, has made it difficult to discern genotype-phenotype correlations. We therefore screened individuals who had been found to bear variants in a non-expansion SCA-associated gene through genetic testing, and after we eliminated genetic groups that had fewer than 30 subjects, there were 756 subjects bearing single-nucleotide variants or deletions in one of seven genes: CACNA1A (239 subjects), PRKCG (175), AFG3L2 (101), ITPR1 (91), STUB1 (77), SPTBN2 (39), or KCNC3 (34). We compared age at onset, disease features, and progression by gene and variant. There were no features that reliably distinguished one of these SCAs from another, and several genes-CACNA1A, ITPR1, SPTBN2, and KCNC3-were associated with both adult-onset and infantile-onset forms of disease, which also differed in presentation. Nevertheless, progression was overall very slow, and STUB1-associated disease was the fastest. Several variants in CACNA1A showed particularly wide ranges in age at onset: one variant produced anything from infantile developmental delay to ataxia onset at 64 years of age within the same family. For CACNA1A, ITPR1, and SPTBN2, the type of variant and charge change on the protein greatly affected the phenotype, defying pathogenicity prediction algorithms. Even with next-generation sequencing, accurate diagnosis requires dialogue between the clinician and the geneticist.


Subject(s)
Cerebellar Ataxia , Spinocerebellar Ataxias , Humans , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/diagnosis , Cerebellar Ataxia/genetics , Phenotype , Ataxia/genetics , Genetic Testing , ATPases Associated with Diverse Cellular Activities/genetics , ATP-Dependent Proteases/genetics , Ubiquitin-Protein Ligases/genetics
2.
Am J Med Genet C Semin Med Genet ; : e32089, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38884529

ABSTRACT

Blepharophimosis with intellectual disability (BIS) is a recently recognized disorder distinct from Nicolaides-Baraister syndrome that presents with distinct facial features of blepharophimosis, developmental delay, and intellectual disability. BIS is caused by pathogenic variants in SMARCA2, that encodes the catalytic subunit of the superfamily II helicase group of the BRG1 and BRM-associated factors (BAF) forming the BAF complex, a chromatin remodeling complex involved in transcriptional regulation. Individuals bearing variants within the bipartite nuclear localization (BNL) signal domain of ADNP present with the neurodevelopmental disorder known as Helsmoortel-Van Der Aa Syndrome (HVDAS). Distinct DNA methylation profiles referred to as episignatures have been reported in HVDAS and BAF complex disorders. Due to molecular interactions between ADNP and BAF complex, and an overlapping craniofacial phenotype with narrowing of the palpebral fissures in a subset of patients with HVDAS and BIS, we hypothesized the possibility of a common phenotype-specific episignature. A distinct episignature was shared by 15 individuals with BIS-causing SMARCA2 pathogenic variants and 12 individuals with class II HVDAS caused by truncating pathogenic ADNP variants. This represents first evidence of a sensitive phenotype-specific episignature biomarker shared across distinct genetic conditions that also exhibit unique gene-specific episignatures.

3.
J Med Genet ; 60(9): 885-893, 2023 09.
Article in English | MEDLINE | ID: mdl-36788019

ABSTRACT

BACKGROUND: Joubert syndrome (JS) is a neurodevelopmental ciliopathy characterised by a distinctive mid-hindbrain malformation, the 'molar tooth sign'. Over 40 JS-associated genes are known, accounting for two-thirds of cases. METHODS: While most variants are novel or extremely rare, we report on 11 recurring variants in seven genes, including three known 'founder variants' in the Ashkenazi Jewish, Hutterite and Finnish populations. We evaluated variant frequencies in ~550 European patients with JS and compared them with controls (>15 000 Italian plus gnomAD), and with an independent cohort of ~600 JS probands from the USA. RESULTS: All variants were markedly enriched in the European JS cohort compared with controls. When comparing allele frequencies in the two JS cohorts, the Ashkenazim founder variant (TMEM216 c.218G>T) was significantly enriched in American compared with European patients with JS, while MKS1 c.1476T>G was about 10 times more frequent among European JS. Frequencies of other variants were comparable in the two cohorts. Genotyping of several markers identified four novel European founder haplotypes.Two recurrent variants (MKS1 c.1476T>G and KIAA0586 c.428delG), have been detected in homozygosity in unaffected individuals, suggesting they could act as hypomorphic variants. However, while fibroblasts from a MKS1 c.1476T>G healthy homozygote showed impaired ability to form primary cilia and mildly reduced ciliary length, ciliary parameters were normal in cells from a KIAA0586 c.428delG healthy homozygote. CONCLUSION: This study contributes to understand the complex genetic landscape of JS, explain its variable prevalence in distinct geographical areas and characterise two recurrent hypomorphic variants.


Subject(s)
Abnormalities, Multiple , Eye Abnormalities , Kidney Diseases, Cystic , Humans , Cerebellum/abnormalities , Abnormalities, Multiple/genetics , Eye Abnormalities/genetics , Kidney Diseases, Cystic/genetics , Retina/abnormalities
4.
Am J Med Genet A ; 191(5): 1350-1354, 2023 05.
Article in English | MEDLINE | ID: mdl-36680497

ABSTRACT

The ubiquitin-specific protease USP9X has been found to play a role in multiple aspects of neural development including processes of neuronal migrations. In males, hemizygous partial loss of function variants in USP9X lead to a clinical phenotype primarily characterized by intellectual disability, hypotonia, speech and language impairment, behavioral disturbances accompanied by additional clinical features with variable expressivity. Structural brain abnormalities are reported in all cases where neuro-imaging was performed. The most common radiological features described include hypoplasia/agenesis of the corpus callosum, widened ventricles, white matter disturbances, and cerebellar hypoplasia. Here we report a child harboring a missense variant in USP9X presenting with the classical neurodevelopmental phenotype and a previously unreported radiological picture of periventricular heterotopia. This case expands the phenotypic landscape of this emergent condition and supports the critical role of USP9X in neuronal migration processes.


Subject(s)
Intellectual Disability , Periventricular Nodular Heterotopia , Humans , Child , Male , Periventricular Nodular Heterotopia/diagnostic imaging , Periventricular Nodular Heterotopia/genetics , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Intellectual Disability/complications , Mutation, Missense , Developmental Disabilities/genetics , Radiography , Ubiquitin Thiolesterase/genetics
5.
Dev Med Child Neurol ; 65(4): 544-550, 2023 04.
Article in English | MEDLINE | ID: mdl-36175354

ABSTRACT

AIM: To assess whether microcephaly with pontine and cerebellar hypoplasia (MICPCH) could manifest in the prenatal period in patients with calcium/calmodulin-dependent serine protein kinase (CASK) gene disorders. METHOD: In this international multicentre retrospective study, we contacted a CASK parents' social media group and colleagues with expertise in cerebellar malformations and asked them to supply clinical and imaging information. Centiles and standard deviations (SD) were calculated according to age by nomograms. RESULTS: The study consisted of 49 patients (44 females and 5 males). Information regarding prenatal head circumference was available in 19 patients; 11 out of 19 had a fetal head circumference below -2SD (range -4.1SD to -2.02SD, mean gestational age at diagnosis 20 weeks). Progressive prenatal deceleration of head circumference growth rate was observed in 15 out of 19. At birth, 20 out of 42 had a head circumference below -2SD. A total of 6 out of 15 fetuses had a TCD z-score below -2 (range -5.88 to -2.02). INTERPRETATION: This study expands the natural history of CASK-related disorders to the prenatal period, showing evidence of progressive deceleration of head circumference growth rate, head circumference below -2SD, or small TCD. Most cases will not be diagnosed according to current recommendations for fetal central nervous system routine assessment. Consecutive measurements and genetic studies are advised in the presence of progressive deceleration of head circumference growth rates or small TCD. WHAT THIS PAPER ADDS: Progressive deceleration of fetal head circumference growth rate can be observed. A small transcerebellar diameter is an additional important manifestation. Most cases will not be diagnosed according to current recommendations for fetal central nervous system routine assessment. Consecutive measurements are advised when measurements are within the low range of norm.


Subject(s)
Microcephaly , Nervous System Malformations , Female , Humans , Infant , Infant, Newborn , Male , Pregnancy , Fetus , Gestational Age , Microcephaly/diagnosis , Nervous System Malformations/genetics , Retrospective Studies
6.
Neurol Sci ; 44(1): 263-271, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36114979

ABSTRACT

PURPOSE: Fibrocartilaginous nucleus pulposus components herniation and embolism rarely causes acute ischaemic events involving the spinal cord. Few reports have suggested this as a mechanism leading to anterior spinal artery syndrome. The purpose of this study was to evaluate the topography and pattern of this rare myelopathy by MRI. METHODS: A retrospective observational case series of patients, admitted to our Institute between 2008 and 2021, with a diagnosis of fibrocartilaginous embolism based on typical clinical and radiological features. RESULTS: Five patients were identified (2 men and 3 women; range 13-38 years). No one had pre-existing vascular risk factors. All referred potential precipitating event in the 24 h prior to symptom onset. MRI findings showed increased signal intensity of the spinal cord on T2-weighted images in all cases and degenerative disc changes opposite to it in four of them. The outcome was poor: three showed only partial sensitivity and motor improvement (mRs 4, 3, and 2, respectively); one completely recovered except for isolated hand paresis (mRs 1); and one remained severely neurologically affected (mRs 5). CONCLUSIONS: Fibrocartilaginous embolism must be a differential diagnosis in case of otherwise unexplained spinal cord infarction in adult and paediatric low risk population. Neuroradiological findings such as abnormal spinal cord signal intensity and degenerative disc changes can aid in early diagnosis of this rare myelopathy. The prevalent myelopathy location was thoracic. All signal alterations were detected in the anterior region of the spinal cord in the territories of the anterior spinal artery.


Subject(s)
Embolism , Spinal Cord Diseases , Male , Adult , Child , Humans , Female , Retrospective Studies , Spinal Cord/diagnostic imaging , Spinal Cord/blood supply , Spinal Cord Diseases/complications , Spinal Cord Diseases/diagnostic imaging , Embolism/complications , Embolism/diagnostic imaging , Magnetic Resonance Imaging , Infarction/diagnostic imaging , Infarction/etiology
7.
J Med Genet ; 59(9): 888-894, 2022 Sep.
Article in English | MEDLINE | ID: mdl-34675124

ABSTRACT

BACKGROUND: Joubert syndrome (JS) is a recessively inherited ciliopathy characterised by congenital ocular motor apraxia (COMA), developmental delay (DD), intellectual disability, ataxia, multiorgan involvement, and a unique cerebellar and brainstem malformation. Over 40 JS-associated genes are known with a diagnostic yield of 60%-75%.In 2018, we reported homozygous hypomorphic missense variants of the SUFU gene in two families with mild JS. Recently, heterozygous truncating SUFU variants were identified in families with dominantly inherited COMA, occasionally associated with mild DD and subtle cerebellar anomalies. METHODS: We reanalysed next generation sequencing (NGS) data in two cohorts comprising 1097 probands referred for genetic testing of JS genes. RESULTS: Heterozygous truncating and splice-site SUFU variants were detected in 22 patients from 17 families (1.5%) with strong male prevalence (86%), and in 8 asymptomatic parents. Patients presented with COMA, hypotonia, ataxia and mild DD, and only a third manifested intellectual disability of variable severity. Brain MRI showed consistent findings characterised by vermis hypoplasia, superior cerebellar dysplasia and subtle-to-mild abnormalities of the superior cerebellar peduncles. The same pattern was observed in two out of three tested asymptomatic parents. CONCLUSION: Heterozygous truncating or splice-site SUFU variants cause a novel neurodevelopmental syndrome encompassing COMA and mild JS, which likely represent overlapping entities. Variants can arise de novo or be inherited from a healthy parent, representing the first cause of JS with dominant inheritance and reduced penetrance. Awareness of this condition will increase the diagnostic yield of JS genetic testing, and allow appropriate counselling about prognosis, medical monitoring and recurrence risk.


Subject(s)
Abnormalities, Multiple , Cerebellar Ataxia , Eye Abnormalities , Intellectual Disability , Kidney Diseases, Cystic , Abnormalities, Multiple/genetics , Cerebellar Ataxia/genetics , Cerebellum/abnormalities , Cerebellum/diagnostic imaging , Eye Abnormalities/genetics , Haploinsufficiency/genetics , Humans , Intellectual Disability/genetics , Kidney Diseases, Cystic/diagnosis , Kidney Diseases, Cystic/genetics , Male , Phenotype , Repressor Proteins/genetics , Retina/abnormalities
8.
J Med Genet ; 59(4): 399-409, 2022 04.
Article in English | MEDLINE | ID: mdl-34085948

ABSTRACT

BACKGROUND: Pontocerebellar hypoplasias (PCH) comprise a group of genetically heterogeneous disorders characterised by concurrent hypoplasia of the pons and the cerebellum and variable clinical and imaging features. The current classification includes 13 subtypes, with ~20 known causative genes. Attempts have been made to delineate the phenotypic spectrum associated to specific PCH genes, yet clinical and neuroradiological features are not consistent across studies, making it difficult to define gene-specific outcomes. METHODS: We performed deep clinical and imaging phenotyping in 56 probands with a neuroradiological diagnosis of PCH, who underwent NGS-based panel sequencing of PCH genes and MLPA for CASK rearrangements. Next, we conducted a phenotype-based unsupervised hierarchical cluster analysis to investigate associations between genes and specific phenotypic clusters. RESULTS: A genetic diagnosis was obtained in 43 probands (77%). The most common causative gene was CASK, which accounted for nearly half cases (45%) and was mutated in females and occasionally in males. The European founder mutation p.Ala307Ser in TSEN54 and pathogenic variants in EXOSC3 accounted for 18% and 9% of cases, respectively. VLDLR, TOE1 and RARS2 were mutated in single patients. We were able to confirm only few previously reported associations, including jitteriness and clonus with TSEN54 and lower motor neuron signs with EXOSC3. When considering multiple features simultaneously, a clear association with a phenotypic cluster only emerged for EXOSC3. CONCLUSION: CASK represents the major PCH causative gene in Italy. Phenotypic variability associated with the most common genetic causes of PCH is wider than previously thought, with marked overlap between CASK and TSEN54-associated disorders.


Subject(s)
Cerebellar Diseases , Olivopontocerebellar Atrophies , Cerebellar Diseases/genetics , Cerebellum/diagnostic imaging , Cerebellum/pathology , Female , Humans , Male , Mutation/genetics , Nuclear Proteins/genetics , Olivopontocerebellar Atrophies/diagnosis , Olivopontocerebellar Atrophies/genetics , Olivopontocerebellar Atrophies/pathology , Phenotype
9.
Hum Mutat ; 43(12): 2222-2233, 2022 12.
Article in English | MEDLINE | ID: mdl-36259739

ABSTRACT

Trichothiodystrophy (TTD) is a rare hereditary disease whose prominent feature is brittle hair. Additional clinical signs are physical and neurodevelopmental abnormalities and in about half of the cases hypersensitivity to UV radiation. The photosensitive form of TTD (PS-TTD) is most commonly caused by mutations in the ERCC2/XPD gene encoding a subunit of the transcription/DNA repair complex TFIIH. Here we report novel ERCC2/XPD mutations affecting proper protein folding, which generate thermo-labile forms of XPD associated with thermo-sensitive phenotypes characterized by reversible aggravation of TTD clinical signs during episodes of fever. In patient cells, the newly identified XPD variants result in thermo-instability of the whole TFIIH complex and consequent temperature-dependent defects in DNA repair and transcription. Improving the protein folding process by exposing patient cells to low temperature or to the chemical chaperone glycerol allowed rescue of TFIIH thermo-instability and a concomitant recovery of the complex activities. Besides providing a rationale for the peculiar thermo-sensitive clinical features of these new cases, the present findings demonstrate how variations in the cellular concentration of mutated TFIIH impact the cellular functions of the complex and underlie how both quantitative and qualitative TFIIH alterations contribute to TTD clinical features.


Subject(s)
Hair Diseases , Skin Diseases , Trichothiodystrophy Syndromes , Xeroderma Pigmentosum , Humans , Transcription Factor TFIIH/genetics , Transcription Factor TFIIH/metabolism , Trichothiodystrophy Syndromes/genetics , Trichothiodystrophy Syndromes/complications , DNA Repair , Hair Diseases/genetics , Transcription, Genetic , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum Group D Protein/genetics , Xeroderma Pigmentosum Group D Protein/metabolism
10.
Am J Med Genet C Semin Med Genet ; 190(1): 102-108, 2022 03.
Article in English | MEDLINE | ID: mdl-35488810

ABSTRACT

Biallelic loss-of-function (LoF) variants in CENPF gene are responsible for Strømme syndrome, a condition presenting with intestinal atresia, anterior ocular chamber anomalies, and microcephaly. Through an international collaboration, four individuals (three males and one female) carrying CENPF biallelic variants, including two missense variants in homozygous state and four LoF variants, were identified by exome sequencing. All individuals had variable degree of developmental delay/intellectual disability and microcephaly (ranging from -2.9 SDS to -5.6 SDS) and a recognizable pattern of dysmorphic facial features including inverted-V shaped interrupted eyebrows, epicanthal fold, depressed nasal bridge, and pointed chin. Although one of the cases had duodenal atresia, all four individuals did not have the combination of internal organ malformations of Strømme syndrome (intestinal atresia and anterior eye segment abnormalities). Immunofluorescence analysis on skin fibroblasts on one of the four cases with the antibody for ARL13B that decorates primary cilia revealed shorter primary cilia that are consistent with a ciliary defect. This case-series of individuals with biallelic CENPF variants suggests the spectrum of clinical manifestations of the disorder that may be related to CENPF variants is broad and can include phenotypes lacking the cardinal features of Strømme syndrome.


Subject(s)
Chromosomal Proteins, Non-Histone , Intellectual Disability , Intestinal Atresia , Microcephaly , Microfilament Proteins , Chromosomal Proteins, Non-Histone/genetics , Eye Abnormalities , Female , Humans , Intestinal Atresia/genetics , Male , Microcephaly/genetics , Microfilament Proteins/genetics , Mutation/genetics , Phenotype
11.
Am J Med Genet A ; 188(6): 1667-1675, 2022 06.
Article in English | MEDLINE | ID: mdl-35146895

ABSTRACT

TRPM3 encodes a transient receptor potential cation channel of the melastatin family, expressed in the central nervous system and in peripheral sensory neurons of the dorsal root ganglia. The recurrent substitution in TRPM3: c.2509G>A, p.(Val837Met) has been associated with syndromic intellectual disability and seizures. In this report, we present the clinical and molecular features of seven previously unreported individuals, identified by exome sequencing, with the recurrent p.(Val837Met) variant and global developmental delay. Other shared clinical features included congenital hypotonia, dysmorphic facial features (broad forehead, deep-set eyes, and down turned mouth), exotropia, and musculoskeletal issues (hip dysplasia, hip dislocation, scoliosis). Seizures were observed in two of seven individuals (febrile seizure in one and generalized tonic-clonic seizures with atonic drops in another), and epileptiform activity was observed in an additional two individuals. This report extends the number of affected individuals to 16 who are heterozygous for the de novo recurrent substitution p.(Val837Met). In contrast with the initial report, epilepsy was not a mandatory feature observed in this series. TRPM3 pathogenic variation should be considered in individuals with global developmental delays, moderate-severe intellectual disability with, or without, childhood-onset epilepsy.


Subject(s)
Epilepsy , Infant, Newborn, Diseases , Intellectual Disability , TRPM Cation Channels , Child , Developmental Disabilities/genetics , Humans , Infant, Newborn , Intellectual Disability/genetics , Muscle Hypotonia/genetics , Mutation, Missense , TRPM Cation Channels/genetics , Exome Sequencing
12.
J Med Genet ; 58(7): 475-483, 2021 07.
Article in English | MEDLINE | ID: mdl-32737135

ABSTRACT

BACKGROUND: Dominant and recessive variants in the KIF1A gene on chromosome 2q37.3 are associated with several phenotypes, although only three syndromes are currently listed in the OMIM classification: hereditary sensory and autonomic neuropathy type 2 and spastic paraplegia type 30, both recessively inherited, and mental retardation type 9 with dominant inheritance. METHODS: In this retrospective multicentre study, we describe the clinical, neuroradiological and genetic features of 19 Caucasian patients (aged 3-65 years) harbouring heterozygous KIF1A variants, and extensively review the available literature to improve current classification of KIF1A-related disorders. RESULTS: Patients were divided into two groups. Group 1 comprised patients with a complex phenotype with prominent pyramidal signs, variably associated in all but one case with additional features (ie, epilepsy, ataxia, peripheral neuropathy, optic nerve atrophy); conversely, patients in group 2 presented an early onset or congenital ataxic phenotype. Fourteen different heterozygous missense variants were detected by next-generation sequencing screening, including three novel variants, most falling within the kinesin motor domain. CONCLUSION: The present study further enlarges the clinical and mutational spectrum of KIF1A-related disorders by describing a large series of patients with dominantly inherited KIF1A pathogenic variants ranging from pure to complex forms of hereditary spastic paraparesis/paraplegias (HSP) and ataxic phenotypes in a lower proportion of cases. A comprehensive review of the literature indicates that KIF1A screening should be implemented in HSP regardless of its mode of inheritance or presentations as well as in other complex neurodegenerative or neurodevelopmental disorders showing congenital or early onset ataxia.


Subject(s)
Kinesins/genetics , Neurodegenerative Diseases/genetics , Neurodevelopmental Disorders/genetics , Adolescent , Adult , Aged , Ataxia/congenital , Ataxia/genetics , Child , Child, Preschool , Female , Heterozygote , Humans , Male , Middle Aged , Mutation , Retrospective Studies , Young Adult
13.
Neuropediatrics ; 52(6): 484-488, 2021 12.
Article in English | MEDLINE | ID: mdl-33853164

ABSTRACT

KIRREL3 is a gene important for the central nervous system development-in particular for the process of neuronal migration, axonal fasciculation, and synaptogenesis-and colocalizes and cooperates in neurons with CASK gene. Alterations of KIRREL3 have been linked to neurodevelopmental disorders, ranging from developmental delay, to autism spectrum disorder, to attention deficit/hyperactivity disorder. The underlying mechanism is not yet fully understood, as it has been hypothesized a fully dominant effect, a risk factor role of KIRREL3 partially penetrating variants, and a recessive inheritance pattern. We report a novel and de novo KIRREL3 mutation in a child affected by severe neurodevelopmental disorder and with brain magnetic resonance imaging evidence of mega cisterna magna and mild cerebellar hypoplasia. This case strengthens the hypothesis that dominant KIRREL3 variants may lead to neurodevelopmental disruption; furthermore, given the strong interaction between KIRREL3 and CASK, we discuss as posterior fossa anomalies may also be part of the phenotype of KIRREL3-related syndrome.


Subject(s)
Autism Spectrum Disorder , Nervous System Malformations , Neurodevelopmental Disorders , Autism Spectrum Disorder/diagnostic imaging , Autism Spectrum Disorder/genetics , Cerebellum/abnormalities , Child , Developmental Disabilities/diagnostic imaging , Developmental Disabilities/genetics , Humans , Nervous System Malformations/diagnostic imaging , Nervous System Malformations/genetics , Neurodevelopmental Disorders/diagnostic imaging , Neurodevelopmental Disorders/genetics
14.
Am J Hum Genet ; 101(4): 552-563, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28965847

ABSTRACT

The Sonic Hedgehog (SHH) pathway is a key signaling pathway orchestrating embryonic development, mainly of the CNS and limbs. In vertebrates, SHH signaling is mediated by the primary cilium, and genetic defects affecting either SHH pathway members or ciliary proteins cause a spectrum of developmental disorders. SUFU is the main negative regulator of the SHH pathway and is essential during development. Indeed, Sufu knock-out is lethal in mice, and recessive pathogenic variants of this gene have never been reported in humans. Through whole-exome sequencing in subjects with Joubert syndrome, we identified four children from two unrelated families carrying homozygous missense variants in SUFU. The children presented congenital ataxia and cerebellar vermis hypoplasia with elongated superior cerebellar peduncles (mild "molar tooth sign"), typical cranio-facial dysmorphisms (hypertelorism, depressed nasal bridge, frontal bossing), and postaxial polydactyly. Two siblings also showed polymicrogyria. Molecular dynamics simulation predicted random movements of the mutated residues, with loss of the native enveloping movement of the binding site around its ligand GLI3. Functional studies on cellular models and fibroblasts showed that both variants significantly reduced SUFU stability and its capacity to bind GLI3 and promote its cleavage into the repressor form GLI3R. In turn, this impaired SUFU-mediated repression of the SHH pathway, as shown by altered expression levels of several target genes. We demonstrate that germline hypomorphic variants of SUFU cause deregulation of SHH signaling, resulting in recessive developmental defects of the CNS and limbs which share features with both SHH-related disorders and ciliopathies.


Subject(s)
Abnormalities, Multiple/genetics , Bone Diseases, Developmental/genetics , Cerebellum/abnormalities , Craniofacial Abnormalities/genetics , Eye Abnormalities/genetics , Genes, Recessive , Hedgehog Proteins/metabolism , Kidney Diseases, Cystic/genetics , Mutation, Missense , Repressor Proteins/genetics , Retina/abnormalities , Abnormalities, Multiple/pathology , Bone Diseases, Developmental/pathology , Cells, Cultured , Cerebellum/pathology , Child , Cohort Studies , Craniofacial Abnormalities/pathology , Eye Abnormalities/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation, Developmental , Humans , Kidney Diseases, Cystic/pathology , Kruppel-Like Transcription Factors/metabolism , Male , Nerve Tissue Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Retina/pathology , Sequence Analysis, DNA , Signal Transduction , Skin/metabolism , Skin/pathology , Zinc Finger Protein Gli3
15.
Cerebellum ; 19(5): 629-635, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32472476

ABSTRACT

Chromosomal microarray analysis is commonly used as screening test for children with neurodevelopmental issues, also in case of complex neurological phenotypes. Developmental delay/intellectual disability is a common presentation sign in pediatric ataxias, diseases with high clinical and genetic heterogeneity. In order to determine the diagnostic yield of Array-CGH in such conditions, all the tests performed in the last 10-year activity of a single referral center in children who present, besides the neurodevelopmental impairment, cerebellar abnormalities have been systematically gathered. The study demonstrates that, except for Dandy-Walker malformation or poly-malformative phenotypes, chromosomal microarray analysis should be discouraged as first-line diagnostic test in pediatric ataxias with neurodevelopmental disability.


Subject(s)
Cerebellar Cortex/abnormalities , Developmental Disabilities/genetics , Intellectual Disability/genetics , Nervous System Malformations/genetics , Child , Child, Preschool , Developmental Disabilities/diagnosis , Diagnostic Tests, Routine/methods , Female , Humans , Infant , Intellectual Disability/diagnosis , Male , Microarray Analysis/methods , Nervous System Malformations/diagnosis
16.
Am J Med Genet A ; 182(10): 2317-2324, 2020 10.
Article in English | MEDLINE | ID: mdl-33043631

ABSTRACT

Potocki-Lupski syndrome is a condition mainly characterized by infantile hypotonia, developmental delay/intellectual disability (DD/ID), and congenital anomalies, caused by duplications of the 17p11.2 region, encompassing RAI1 gene. Its clinical presentation is extremely variable, especially for what concerns the cognitive level and the behavioral phenotype. Such aspects, as well as the dysmorphic/malformative ones, have been covered by previous studies; otherwise neurological features have never been systematically described. In order to delineate the neurological phenotype of Potocki-Lupski Syndrome, we collect an 8-patients cohort. Developmental milestones are delayed and a mild to moderate cognitive impairment is present in all patients, variably associated with features of autism spectrum disorder, behavioral disturb, and sleep disturb. Hypotonia appears a less frequent finding than what previously reported, while motor clumsiness/coordination impairment is frequent. EGG registration demonstrated a common pattern with excess of diffuse rhythmic activity in sleep phases or while the patient is falling asleep. Brain MRI did not reveal common anomalies, although unspecific white matter changes may be present. We discuss such findings and compare them to literature data, offering an overview on the neurological and cognitive-behavioral presentation of the syndrome.


Subject(s)
Abnormalities, Multiple/diagnostic imaging , Chromosome Disorders/diagnostic imaging , Developmental Disabilities/diagnostic imaging , Intellectual Disability/diagnostic imaging , Nervous System Diseases/diagnostic imaging , Sleep/physiology , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Adolescent , Child , Child, Preschool , Chromosome Disorders/genetics , Chromosome Disorders/pathology , Chromosome Duplication/genetics , Cognitive Dysfunction/diagnostic imaging , Comparative Genomic Hybridization , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Electroencephalography , Female , Humans , Infant , Intellectual Disability/genetics , Intellectual Disability/pathology , Magnetic Resonance Imaging , Male , Muscle Hypotonia/diagnostic imaging , Muscle Hypotonia/genetics , Muscle Hypotonia/pathology , Nervous System Diseases/genetics , Nervous System Diseases/pathology , Phenotype
17.
Am J Med Genet A ; 182(1): 229-249, 2020 01.
Article in English | MEDLINE | ID: mdl-31710777

ABSTRACT

Joubert syndrome (JS) is a recessive neurodevelopmental disorder defined by a characteristic cerebellar and brainstem malformation recognizable on axial brain magnetic resonance imaging as the "Molar Tooth Sign". Although defined by the neurological features, JS is associated with clinical features affecting many other organ systems, particularly progressive involvement of the retina, kidney, and liver. JS is a rare condition; therefore, many affected individuals may not have easy access to subspecialty providers familiar with JS (e.g., geneticists, neurologists, developmental pediatricians, ophthalmologists, nephrologists, hepatologists, psychiatrists, therapists, and educators). Expert recommendations can enable practitioners of all types to provide quality care to individuals with JS and know when to refer for subspecialty care. This need will only increase as precision treatments targeting specific genetic causes of JS emerge. The goal of these recommendations is to provide a resource for general practitioners, subspecialists, and families to maximize the health of individuals with JS throughout the lifespan.


Subject(s)
Abnormalities, Multiple/epidemiology , Cerebellum/abnormalities , Eye Abnormalities/epidemiology , Health Personnel , Kidney Diseases, Cystic/epidemiology , Neurodevelopmental Disorders/epidemiology , Retina/abnormalities , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Abnormalities, Multiple/therapy , Brain Stem/pathology , Cerebellum/pathology , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Eye Abnormalities/therapy , Health Planning Guidelines , Humans , Kidney/pathology , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/pathology , Kidney Diseases, Cystic/therapy , Liver/pathology , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Neurodevelopmental Disorders/therapy , Retina/pathology
18.
Nephrol Dial Transplant ; 35(7): 1195-1202, 2020 07 01.
Article in English | MEDLINE | ID: mdl-30403813

ABSTRACT

BACKGROUND: Joubert syndrome (JS) is an inherited ciliopathy characterized by a complex midbrain-hindbrain malformation and multiorgan involvement. Renal disease, mainly juvenile nephronophthisis (NPH), was reported in 25-30% patients although only ∼18% had a confirmed diagnosis of chronic kidney disease (CKD). NPH often remains asymptomatic for many years, resulting in delayed diagnosis. The aim of the study was to identify a biomarker able to quantify the risk of progressive CKD in young children with JS. METHODS: Renal features were investigated in 93 Italian patients, including biochemical tests, ultrasound and 1-deamino-8D-arginine vasopressin test in children with reduced basal urine osmolality. A subset of patients was followed-up over time. RESULTS: At last examination, 27 of 93 subjects (29%) presented with CKD, ranging from isolated urinary concentration defect (UCD) to end-stage renal disease. Both normal and pathological urine osmolality levels remained stable over time, even when obtained at very early ages. Follow-up data showed that the probability of developing CKD can be modelled as a function of the urine osmolality value, exceeding 75% for levels <600 mOsm/kg H2O, and significantly increased in patients with an early diagnosis of isolated UCD. CONCLUSIONS: We conclude that the frequency of CKD in JS increases with age and is higher than previously reported. Urine osmolality represents an early sensitive quantitative biomarker of the risk of CKD progression.


Subject(s)
Biomarkers/urine , Cerebellum/abnormalities , Eye Abnormalities/complications , Kidney Diseases, Cystic/complications , Renal Insufficiency, Chronic/diagnosis , Retina/abnormalities , Abnormalities, Multiple , Adolescent , Adult , Child , Child, Preschool , Disease Progression , Female , Humans , Infant , Infant, Newborn , Male , Osmolar Concentration , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/urine , Risk Factors , Survival Rate , Young Adult
19.
Neuropediatrics ; 51(3): 173-177, 2020 06.
Article in English | MEDLINE | ID: mdl-31784971

ABSTRACT

Heterozygous deletions of the gene PMP22 are associated to hereditary neuropathy with liability to pressure palsies (HNPP), a demyelinating neuromuscular disease causing variable transitory focal muscles weakness. Deletions involving both copies of PMP22 cause more severe phenotypes, with early-onset neuropathy and impairment in motor development. We report a patient with a severe early-onset demyelinating neuropathy, caused by two different inherited deletions of PMP22, whose parents had an HNPP. The patient showed neurological signs and delay in motor development but normal intellective abilities. A motor and sensitive conduction study showed severe signs of demyelination, suggestive for Dejerine Sottas Syndrome (DSS). The patient's father had a typical HNPP caused by a heterozygous 17p11.2 deletion, encompassing PMP22. The patient's mother reported no neuropathic symptoms, but in a nerve conduction studies, parents and several relatives showed signs of sensory-motor deficit with focal slowing of conduction at common sites of entrapment. Quantitative analysis of PMP22, performed in our patient by multiplex ligation-dependent probe amplification, revealed a compound heterozygous status with the same deletion of the father and a deletion of PMP22 exon 5, after proved to be inherited from the mother. Therefore, when we face an early-onset, severe form of neuropathy, we have to consider rare forms of hereditary neuropathy caused by homozygous or compound heterozygous mutations in PMP22, even if parents are asymptomatic; an exhaustive family history and an electrodiagnostic study are essential to guide genetic tests and to make a diagnosis.


Subject(s)
Arthrogryposis , Hereditary Sensory and Motor Neuropathy , Myelin Proteins/genetics , Arthrogryposis/diagnosis , Arthrogryposis/genetics , Arthrogryposis/physiopathology , Child, Preschool , Female , Gene Deletion , Hereditary Sensory and Motor Neuropathy/diagnosis , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Neural Conduction/physiology , Pedigree
20.
Mol Genet Metab ; 126(4): 489-494, 2019 04.
Article in English | MEDLINE | ID: mdl-30826161

ABSTRACT

BACKGROUND: Aicardi-Goutières syndrome (AGS) is a rare genetic leukoencephalopathy related to inappropriate activation of type I interferon. Neuroradiological findings are typically characterized by white matter abnormalities, cerebral atrophy and cerebral calcification. The disease usually manifests itself during the first year of life in the form of an initial "encephalitic-like" phase followed by a chronic phase of stabilization of the neurological signs. Recently new therapeutic strategies have been proposed aimed at blocking the abnormal activation of the interferon cascade. MATERIALS AND METHODS: We reviewed clinical and MRI findings in three young RNASEH2B-mutated patients studied with serial CT and MRI studies. RESULTS: All three patients presented clinical and MRI features consistent with AGS but, very unexpectedly, an improving neuroradiological course. In patient 1, the MRI improvement was noted some months after treatment with high-dose steroid and IVIg treatment; in patients 2 and 3 it occurred spontaneously. Patient 2 did not show cerebral calcification on CT images. CONCLUSIONS: Our series highlights the possibility of spontaneous neuroradiological improvement in AGS2 patients, as well as the possibility of absence of cerebral calcification in AGS. The study underlines the need for extreme caution when using MRI as an outcome measure in therapeutic trials specific for this disease. MRI follow-up studies in larger series are necessary to describe the natural course of AGS.


Subject(s)
Autoimmune Diseases of the Nervous System/diagnostic imaging , Brain/diagnostic imaging , Calcinosis , Nervous System Malformations/diagnostic imaging , Autoimmune Diseases of the Nervous System/genetics , Brain/pathology , Child, Preschool , Female , Follow-Up Studies , Humans , Magnetic Resonance Imaging , Male , Mutation , Nervous System Malformations/genetics , Neuroimaging , Ribonuclease H/genetics , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL