Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hum Mol Genet ; 32(3): 473-488, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36018820

ABSTRACT

Kinesins are motor proteins involved in microtubule (MT)-mediated intracellular transport. They contribute to key cellular processes, including intracellular trafficking, organelle dynamics and cell division. Pathogenic variants in kinesin-encoding genes underlie several human diseases characterized by an extremely variable clinical phenotype, ranging from isolated neurodevelopmental/neurodegenerative disorders to syndromic phenotypes belonging to a family of conditions collectively termed as 'ciliopathies.' Among kinesins, kinesin-1 is the most abundant MT motor for transport of cargoes towards the plus end of MTs. Three kinesin-1 heavy chain isoforms exist in mammals. Different from KIF5A and KIF5C, which are specifically expressed in neurons and established to cause neurological diseases when mutated, KIF5B is an ubiquitous protein. Three de novo missense KIF5B variants were recently described in four subjects with a syndromic skeletal disorder characterized by kyphomelic dysplasia, hypotonia and DD/ID. Here, we report three dominantly acting KIF5B variants (p.Asn255del, p.Leu498Pro and p.Leu537Pro) resulting in a clinically wide phenotypic spectrum, ranging from dilated cardiomyopathy with adult-onset ophthalmoplegia and progressive skeletal myopathy to a neurodevelopmental condition characterized by severe hypotonia with or without seizures. In vitro and in vivo analyses provide evidence that the identified disease-associated KIF5B variants disrupt lysosomal, autophagosome and mitochondrial organization, and impact cilium biogenesis. All variants, and one of the previously reported missense changes, were shown to affect multiple developmental processes in zebrafish. These findings document pleiotropic consequences of aberrant KIF5B function on development and cell homeostasis, and expand the phenotypic spectrum resulting from altered kinesin-mediated processes.


Subject(s)
Kinesins , Animals , Humans , Kinesins/genetics , Kinesins/metabolism , Mammals/metabolism , Muscle Hypotonia , Neurons/metabolism , Phenotype , Zebrafish/genetics , Zebrafish/metabolism
2.
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
3.
Int J Mol Sci ; 25(13)2024 Jun 24.
Article in English | MEDLINE | ID: mdl-39000022

ABSTRACT

CDKL5 deficiency disorder (CDD) is an X-linked dominant epileptic encephalopathy, characterized by early-onset and drug-resistant seizures, psychomotor delay, and slight facial features. Genomic variants inactivating CDKL5 or impairing its protein product kinase activity have been reported, making next-generation sequencing (NGS) and chromosomal microarray analysis (CMA) the standard diagnostic tests. We report a suspicious case of CDD in a female child who tested negative upon NGS and CMA and harbored an X chromosome de novo pericentric inversion. The use of recently developed genomic techniques (optical genome mapping and whole-genome sequencing) allowed us to finely characterize the breakpoints, with one of them interrupting CDKL5 at intron 1. This is the fifth case of CDD reported in the scientific literature harboring a structural rearrangement on the X chromosome, providing evidence for the hypothesis that this type of anomaly can represent a recurrent pathogenic mechanism, whose frequency is likely underestimated, with it being overlooked by standard techniques. The identification of the molecular etiology of the disorder is extremely important in evaluating the pathological outcome and to better investigate the mechanisms associated with drug resistance, paving the way for the development of specific therapies. Karyotype and genomic techniques should be considered in all cases presenting with CDD without molecular confirmation.


Subject(s)
Chromosomes, Human, X , Protein Serine-Threonine Kinases , Humans , Female , Chromosomes, Human, X/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/deficiency , Chromosome Inversion , Epileptic Syndromes/genetics , Genetic Diseases, X-Linked/genetics , Spasms, Infantile
4.
Clin Genet ; 104(5): 528-541, 2023 11.
Article in English | MEDLINE | ID: mdl-37455656

ABSTRACT

CTNNB1 [OMIM *116806] encodes ß-catenin, an integral part of the cadherin/catenin complex, which functions as effector of Wnt signaling. CTNNB1 is highly expressed in brain as well as in other tissues, including heart. Heterozygous CTNNB1 pathogenic variations are associated with a neurodevelopmental disorder characterized by spastic diplegia and visual defects (NEDSDV) [OMIM #615075], featuring psychomotor delay, intellectual disability, behavioral disturbances, movement disorders, visual defects and subtle facial and somatic features. We report on a new series of 19 NEDSDV patients (mean age 10.3 years), nine of whom bearing novel CTNNB1 variants. Notably, five patients showed congenital heart anomalies including absent pulmonary valve with intact ventricular septum, atrioventricular canal with hypoplastic aortic arch, tetralogy of Fallot, and mitral valve prolapse. We focused on the cardiac phenotype characterizing such cases and reviewed the congenital heart defects in previously reported NEDSDV patients. While congenital heart defects had occasionally been reported so far, the present findings configure a higher rate of cardiac anomalies, suggesting dedicated heart examination to NEDSDV clinical management.


Subject(s)
Heart Defects, Congenital , Intellectual Disability , Neurodevelopmental Disorders , Humans , Child , beta Catenin/genetics , Heart Defects, Congenital/diagnosis , Syndrome , Intellectual Disability/genetics
5.
Brain ; 145(10): 3383-3390, 2022 10 21.
Article in English | MEDLINE | ID: mdl-35737950

ABSTRACT

The endocannabinoid system is a highly conserved and ubiquitous signalling pathway with broad-ranging effects. Despite critical pathway functions, gene variants have not previously been conclusively linked to human disease. We identified nine children from eight families with heterozygous, de novo truncating variants in the last exon of DAGLA with a neuro-ocular phenotype characterized by developmental delay, ataxia and complex oculomotor abnormality. All children displayed paroxysms of nystagmus or eye deviation accompanied by compensatory head posture and worsened incoordination most frequently after waking. RNA sequencing showed clear expression of the truncated transcript and no differences were found between mutant and wild-type DAGLA activity. Immunofluorescence staining of patient-derived fibroblasts and HEK cells expressing the mutant protein showed distinct perinuclear aggregation not detected in control samples. This report establishes truncating variants in the last DAGLA exon as the cause of a unique paediatric syndrome. Because enzymatic activity was preserved, the observed mislocalization of the truncated protein may account for the observed phenotype. Potential mechanisms include DAGLA haploinsufficiency at the plasma membrane or dominant negative effect. To our knowledge, this is the first report directly linking an endocannabinoid system component with human genetic disease and sets the stage for potential future therapeutic avenues.


Subject(s)
Endocannabinoids , Nervous System Diseases , Humans , Child , Phenotype , Nervous System Diseases/genetics , Heterozygote , Syndrome , Mutant Proteins
6.
Epilepsy Behav ; 147: 109436, 2023 10.
Article in English | MEDLINE | ID: mdl-37717460

ABSTRACT

CSNK2B encodes a regulatory subunit of casein kinase II, which is highly expressed in the brain. Heterozygous pathogenic variants in CSNK2B are associated with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS) (OMIM #618732), characterized by facial dysmorphisms, seizures, intellectual disability, and behavioral disturbances. We report ten new patients with CSNK2B-related Neurodevelopmental Syndrome associated with heterozygous variants of CSNK2B. In three patients, a pathogenic variant was inherited from an affected parent. We describe both molecular and clinical features, focusing on epileptic and neurodevelopmental phenotypes. The median age at follow-up was 8.5 years (range 21 months-42 years). All patients had epilepsy, with onset at a median age of 10.5 months range 6 days-10 years). Seizures were both focal and generalized and were resistant to anti-seizure medications in two out of ten patients. Six patients had mild to moderate cognitive delays, whereas four patients had no cognitive disability. Although all previously reported patients had a de novo CSNK2B pathogenic variant, here we report, for the first time, two familial cases of CSNK2B-related Neurodevelopmental Syndrome. We confirmed the highly variable expressivity of the disease among both interfamilial and intrafamilial cases. Furthermore, this study provides information about the long-term outcome in adult patients and underlines the importance of detailed family history collection before performing genetic testing in patients with epilepsy and neurodevelopmental disorders.


Subject(s)
Epilepsy , Intellectual Disability , Neurodevelopmental Disorders , Adult , Humans , Infant , Infant, Newborn , Epilepsy/genetics , Epilepsy/pathology , Neurodevelopmental Disorders/complications , Neurodevelopmental Disorders/genetics , Brain/diagnostic imaging , Brain/pathology , Intellectual Disability/genetics , Syndrome , Phenotype
7.
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
8.
Hum Mutat ; 43(1): 67-73, 2022 01.
Article in English | MEDLINE | ID: mdl-34747546

ABSTRACT

Biallelic mutations in the BRAT1 gene, encoding BRCA1-associated ATM activator 1, result in variable phenotypes, from rigidity and multifocal seizure syndrome, lethal neonatal to neurodevelopmental disorder, and cerebellar atrophy with or without seizures, without obvious genotype-phenotype associations. We describe two families at the mildest end of the spectrum, differing in clinical presentation despite a common genotype at the BRAT1 locus. Two siblings displayed nonprogressive congenital ataxia and shrunken cerebellum on magnetic resonance imaging. A third unrelated patient showed normal neurodevelopment, adolescence-onset seizures, and ataxia, shrunken cerebellum, and ultrastructural abnormalities on skin biopsy, representing the mildest form of NEDCAS hitherto described. Exome sequencing identified the c.638dup and the novel c.1395G>A BRAT1 variants, the latter causing exon 10 skippings. The p53-MCL test revealed normal ATM kinase activity. Our findings broaden the allelic and clinical spectrum of BRAT1-related disease, which should be suspected in presence of nonprogressive cerebellar signs, even without a neurodevelopmental disorder.


Subject(s)
Nuclear Proteins , Seizures , Genetic Association Studies , Genotype , Humans , Mutation , Nuclear Proteins/genetics , Phenotype , Seizures/genetics
9.
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
10.
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
11.
Mol Biol Rep ; 47(1): 711-714, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31583567

ABSTRACT

De novo mutations in the IRF2BPL gene have been identified to date in 18 patients presenting with neuromotor regression, epilepsy and variable neurological signs. Here, we report a female child carrying a novel heterozygous truncating variant in IRF2BPL. Following normal development for two and half years, she developed a progressive neurological condition with psychomotor regression, dystonic tetraparesis with hyperkinetic movements, but no overt epilepsy. Skin biopsy revealed enlarged lysosomes containing granular and tubular material, suggestive of a lysosomal storage disorder. This case expands the IRF2BPL phenotypic spectrum, for the first time providing evidence of endolysosomal storage.


Subject(s)
Carrier Proteins/genetics , Lysosomal Storage Diseases , Lysosomes/pathology , Nuclear Proteins/genetics , Child , DNA Mutational Analysis , Diagnosis, Differential , Female , Humans , Lysosomal Storage Diseases/diagnosis , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/pathology , Mutation/genetics , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Phenotype , Skin/cytology , Skin/pathology
12.
Brain ; 142(10): 2965-2978, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31412107

ABSTRACT

Basal ganglia are subcortical grey nuclei that play essential roles in controlling voluntary movements, cognition and emotion. While basal ganglia dysfunction is observed in many neurodegenerative or metabolic disorders, congenital malformations are rare. In particular, dysplastic basal ganglia are part of the malformative spectrum of tubulinopathies and X-linked lissencephaly with abnormal genitalia, but neurodevelopmental syndromes characterized by basal ganglia agenesis are not known to date. We ascertained two unrelated children (both female) presenting with spastic tetraparesis, severe generalized dystonia and intellectual impairment, sharing a unique brain malformation characterized by agenesis of putamina and globi pallidi, dysgenesis of the caudate nuclei, olfactory bulbs hypoplasia, and anomaly of the diencephalic-mesencephalic junction with abnormal corticospinal tract course. Whole-exome sequencing identified two novel homozygous variants, c.26C>A; p.(S9*) and c.752A>G; p.(Q251R) in the GSX2 gene, a member of the family of homeobox transcription factors, which are key regulators of embryonic development. GSX2 is highly expressed in neural progenitors of the lateral and median ganglionic eminences, two protrusions of the ventral telencephalon from which the basal ganglia and olfactory tubercles originate, where it promotes neurogenesis while negatively regulating oligodendrogenesis. The truncating variant resulted in complete loss of protein expression, while the missense variant affected a highly conserved residue of the homeobox domain, was consistently predicted as pathogenic by bioinformatic tools, resulted in reduced protein expression and caused impaired structural stability of the homeobox domain and weaker interaction with DNA according to molecular dynamic simulations. Moreover, the nuclear localization of the mutant protein in transfected cells was significantly reduced compared to the wild-type protein. Expression studies on both patients' fibroblasts demonstrated reduced expression of GSX2 itself, likely due to altered transcriptional self-regulation, as well as significant expression changes of related genes such as ASCL1 and PAX6. Whole transcriptome analysis revealed a global deregulation in genes implicated in apoptosis and immunity, two broad pathways known to be involved in brain development. This is the first report of the clinical phenotype and molecular basis associated to basal ganglia agenesis in humans.


Subject(s)
Globus Pallidus/growth & development , Homeodomain Proteins/genetics , Putamen/growth & development , Adolescent , Adult , Basal Ganglia/growth & development , Basal Ganglia/metabolism , Basal Ganglia/physiopathology , Cell Differentiation/genetics , Child, Preschool , Embryo, Mammalian/metabolism , Female , Globus Pallidus/metabolism , Globus Pallidus/physiopathology , Homeodomain Proteins/metabolism , Humans , Male , Mutation , Neural Stem Cells/metabolism , Neurogenesis/physiology , Neurons/metabolism , Putamen/metabolism , Putamen/physiopathology , Telencephalon , Transcription Factors/genetics , Exome Sequencing/methods
13.
Biochim Biophys Acta Mol Basis Dis ; 1863(12): 3303-3312, 2017 12.
Article in English | MEDLINE | ID: mdl-28807751

ABSTRACT

The neuron-restricted isoform 3 of the plasma membrane Ca2+ ATPase plays a major role in the regulation of Ca2+ homeostasis in the brain, where the precise control of Ca2+ signaling is a necessity. Several function-affecting genetic mutations in the PMCA3 pump associated to X-linked congenital cerebellar ataxias have indeed been described. Interestingly, the presence of co-occurring mutations in additional genes suggest their synergistic action in generating the neurological phenotype as digenic modulators of the role of PMCA3 in the pathologies. Here we report a novel PMCA3 mutation (G733R substitution) in the catalytic P-domain of the pump in a patient affected by non-progressive ataxia, muscular hypotonia, dysmetria and nystagmus. Biochemical studies of the pump have revealed impaired ability to control cellular Ca2+ handling both under basal and under stimulated conditions. A combined analysis by homology modeling and molecular dynamics have revealed a role for the mutated residue in maintaining the correct 3D configuration of the local structure of the pump. Mutation analysis in the patient has revealed two additional function-impairing compound heterozygous missense mutations (R123Q and G214S substitution) in phosphomannomutase 2 (PMM2), a protein that catalyzes the isomerization of mannose 6-phosphate to mannose 1-phosphate. These mutations are known to be associated with Type Ia congenital disorder of glycosylation (PMM2-CDG), the most common group of disorders of N-glycosylation. The findings highlight the association of PMCA3 mutations to cerebellar ataxia and strengthen the possibility that PMCAs act as digenic modulators in Ca2+-linked pathologies.


Subject(s)
Ataxia/genetics , Ataxia/metabolism , Congenital Disorders of Glycosylation/metabolism , Mutation, Missense , Phosphotransferases (Phosphomutases)/deficiency , Plasma Membrane Calcium-Transporting ATPases/genetics , Brain/diagnostic imaging , Brain/pathology , Calcium/metabolism , Child, Preschool , Congenital Disorders of Glycosylation/diagnostic imaging , Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/pathology , Glycosylation , HeLa Cells , Humans , Male , Phosphotransferases (Phosphomutases)/genetics , Phosphotransferases (Phosphomutases)/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism
14.
Eur Radiol ; 27(12): 5080-5092, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28677066

ABSTRACT

OBJECTIVE: To determine the neuroimaging pattern of cerebellar dysplasia (CD) and other posterior fossa morphological anomalies associated with mutations in tubulin genes and to perform clinical and genetic correlations. METHODS: Twenty-eight patients harbouring 23 heterozygous pathogenic variants (ten novel) in tubulin genes TUBA1A (n = 10), TUBB2B (n = 8) or TUBB3 (n = 5) were studied by a brain MRI scan performed either on a 1.5 T (n = 10) or 3 T (n = 18) MR scanner with focus on the posterior fossa. RESULTS: Cerebellar anomalies were detected in 24/28 patients (86%). CD was recognised in 19/28 (68%) including cortical cerebellar dysplasia (CCD) in 18/28, either involving only the cerebellar hemispheres (12/28) or associated with vermis dysplasia (6/28). CCD was located only in the right hemisphere in 13/18 (72%), including four TUBB2B-, four TUBB3- and five TUBA1A-mutated patients, while in the other five TUBA1A cases it was located only in the left hemisphere or in both hemispheres. The postero-superior region of the cerebellar hemispheres was most frequently affected. CONCLUSIONS: The cerebellar involvement in tubulinopathies shows specific features that may be labelled as 'tubulin-related CD'. This pattern is unique and differs from other genetic causes of cerebellar dysplasia. KEY POINTS: • Cortical cerebellar dysplasia without cysts is suggestive of tubulin-related disorder. • Cerebellar dysplasia in tubulinopathies shows specific features labelled as 'tubulin-related CD'. • Focal and unilateral involvement of cerebellar hemispheres has important implications for counselling.


Subject(s)
Cerebellum/abnormalities , Mutation , Nervous System Malformations/diagnostic imaging , Neuroimaging/methods , Tubulin/genetics , Adult , Brain Stem/diagnostic imaging , Cerebellum/diagnostic imaging , Cerebellum/pathology , Child , Child, Preschool , Developmental Disabilities/diagnostic imaging , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Female , Humans , Infant , Magnetic Resonance Imaging , Male , Middle Aged , Nervous System Malformations/genetics , Nervous System Malformations/pathology , Young Adult
15.
J Med Genet ; 53(9): 608-15, 2016 09.
Article in English | MEDLINE | ID: mdl-27208211

ABSTRACT

BACKGROUND: Ciliopathies are an extensive group of autosomal recessive or X-linked disorders with considerable genetic and clinical overlap, which collectively share multiple organ involvement and may result in lethal or viable phenotypes. In large numbers of cases the genetic defect remains yet to be determined. The aim of this study is to describe the mutational frequency and phenotypic spectrum of the CEP120 gene. METHODS: Exome sequencing was performed in 145 patients with Joubert syndrome (JS), including 15 children with oral-facial-digital syndrome type VI (OFDVI) and 21 Meckel syndrome (MKS) fetuses. Moreover, exome sequencing was performed in one fetus with tectocerebellar dysraphia with occipital encephalocele (TCDOE), molar tooth sign and additional skeletal abnormalities. As a parallel study, 346 probands with a phenotype consistent with JS or related ciliopathies underwent next-generation sequencing-based targeted sequencing of 120 previously described and candidate ciliopathy genes. RESULTS: We present six probands carrying nine distinct mutations (of which eight are novel) in the CEP120 gene, previously found mutated only in Jeune asphyxiating thoracic dystrophy (JATD). The CEP120-associated phenotype ranges from mild classical JS in four patients to more severe conditions in two fetuses, with overlapping features of distinct ciliopathies that include TCDOE, MKS, JATD and OFD syndromes. No obvious correlation is evident between the type or location of identified mutations and the ciliopathy phenotype. CONCLUSION: Our findings broaden the spectrum of phenotypes caused by CEP120 mutations that account for nearly 1% of patients with JS as well as for more complex ciliopathy phenotypes. The lack of clear genotype-phenotype correlation highlights the relevance of comprehensive genetic analyses in the diagnostics of ciliopathies.


Subject(s)
Abnormalities, Multiple/genetics , Cell Cycle Proteins/genetics , Cerebellum/abnormalities , Eye Abnormalities/genetics , Kidney Diseases, Cystic/genetics , Mutation/genetics , Retina/abnormalities , Amino Acid Sequence , Cerebellar Diseases/genetics , Child , Ciliopathies/genetics , Encephalocele/genetics , Female , Genetic Association Studies/methods , Genetic Predisposition to Disease/genetics , Genetic Testing/methods , Humans , Male , Mutation Rate , Orofaciodigital Syndromes/genetics , Pedigree , Phenotype , Sequence Alignment
16.
Neurogenetics ; 17(3): 191-5, 2016 07.
Article in English | MEDLINE | ID: mdl-27251579

ABSTRACT

Dysequilibrium syndrome (DES) is a non-progressive congenital ataxia characterized by severe intellectual deficit, truncal ataxia and markedly delayed, quadrupedal or absent ambulation. Recessive loss-of-function mutations in the very low density lipoprotein receptor (VLDLR) gene represent the most common cause of DES. Only two families have been reported harbouring homozygous missense mutations, both with a similarly severe phenotype. We report an Italian girl with very mild DES caused by the novel homozygous VLDLR missense mutation p.(C419Y). This unusually benign phenotype possibly relates to a less disruptive effect of the mutation, falling within a domain (EGF-B) not predicted as crucial for the protein function.


Subject(s)
Cerebellar Ataxia/genetics , Intellectual Disability/genetics , Mutation, Missense , Receptors, LDL/genetics , Brain/diagnostic imaging , Brain/pathology , Cerebellar Ataxia/diagnostic imaging , Cerebellar Ataxia/pathology , Child , Female , Genetic Association Studies , Humans , Intellectual Disability/diagnostic imaging , Intellectual Disability/pathology , Pedigree
17.
Hum Genet ; 134(1): 123-6, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25407461

ABSTRACT

Oral-facial-digital type VI syndrome (OFDVI) is a rare phenotype of Joubert syndrome (JS). Recently, C5orf42 was suggested as the major OFDVI gene, being mutated in 9 of 11 families (82 %). We sequenced C5orf42 in 313 JS probands and identified mutations in 28 (8.9 %), most with a phenotype of pure JS. Only 2 out of 17 OFDVI patients (11.7 %) were mutated. A comparison of mutated vs. non-mutated OFDVI patients showed that preaxial and mesoaxial polydactyly, hypothalamic hamartoma and other congenital defects may predict C5orf42 mutations, while tongue hamartomas are more common in negative patients.


Subject(s)
Cerebellar Diseases/genetics , Eye Abnormalities/genetics , Hamartoma/genetics , Hypothalamic Diseases/genetics , Kidney Diseases, Cystic/genetics , Membrane Proteins/genetics , Mutation/genetics , Orofaciodigital Syndromes/genetics , Retina/abnormalities , Abnormalities, Multiple , Cerebellar Diseases/pathology , Cerebellum/abnormalities , Cohort Studies , Eye Abnormalities/pathology , Family , Female , Follow-Up Studies , Hamartoma/pathology , Humans , Hypothalamic Diseases/pathology , Kidney Diseases, Cystic/pathology , Male , Orofaciodigital Syndromes/pathology , Phenotype , Retina/pathology
18.
Hum Mutat ; 35(1): 137-46, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24166846

ABSTRACT

Joubert syndrome (JS) is characterized by a distinctive cerebellar structural defect, namely the << molar tooth sign >>. JS is genetically heterogeneous, involving 20 genes identified to date, which are all required for cilia biogenesis and/or function. In a consanguineous family with JS associated with optic nerve coloboma, kidney hypoplasia, and polydactyly, combined exome sequencing and mapping identified a homozygous splice-site mutation in PDE6D, encoding a prenyl-binding protein. We found that pde6d depletion in zebrafish leads to renal and retinal developmental anomalies and wild-type but not mutant PDE6D is able to rescue this phenotype. Proteomic analysis identified INPP5E, whose mutations also lead to JS or mental retardation, obesity, congenital retinal dystrophy, and micropenis syndromes, as novel prenyl-dependent cargo of PDE6D. Mutant PDE6D shows reduced binding to INPP5E, which fails to localize to primary cilia in patient fibroblasts and tissues. Furthermore, mutant PDE6D is unable to bind to GTP-bound ARL3, which acts as a cargo-release factor for PDE6D-bound INPP5E. Altogether, these results indicate that PDE6D is required for INPP5E ciliary targeting and suggest a broader role for PDE6D in targeting other prenylated proteins to the cilia. This study identifies PDE6D as a novel JS disease gene and provides the first evidence of prenyl-binding-dependent trafficking in ciliopathies.


Subject(s)
Cerebellar Diseases/genetics , Cerebellar Diseases/metabolism , Cilia/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Eye Abnormalities/genetics , Eye Abnormalities/metabolism , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Retina/abnormalities , ADP-Ribosylation Factors/metabolism , Abnormalities, Multiple , Animals , Cerebellum/abnormalities , Exome , Female , Genetic Predisposition to Disease , Homozygote , Humans , Male , Models, Molecular , Pedigree , Protein Prenylation , Proteomics , Retina/metabolism , Sequence Analysis, DNA , Zebrafish/abnormalities , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
19.
Front Pediatr ; 12: 1417873, 2024.
Article in English | MEDLINE | ID: mdl-38978838

ABSTRACT

Intracranial hemorrhage may represent a complication of the perinatal period that affects neonatal morbidity and mortality. Very poor data exist about a possible association between mutations of the type IV collagen a1 chain (COL4A1) gene and the development of intracranial hemorrhage, and only sporadic reports focus on intracerebral bleedings already developing in utero or in the neonatal period in infants with such a mutation. This study presents a case series of term neonates affected by intracranial hemorrhage, with no apparent risk factors for the development of this condition, who were carriers of COL4A1 gene variants. This study also provides a review of the most recent scientific literature on this topic, specifically focusing on the available scientific data dealing with the perinatal period.

20.
Neurol Genet ; 10(4): e200169, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39139262

ABSTRACT

Objectives: To provide a comprehensive description of neuroradiologic findings in a patient with a probable pathogenic variant of HUWE1, particularly in relation to pontine and cerebellar hypoplasia. Methods: We first report prenatal and postnatal neuroradiologic phenotype of a female patient carrying a HUWE1 likely pathogenic variant and discuss its function. Results: An ultrasound shows borderline ventriculomegaly, rotated cerebellar vermis, and dysgenetic corpus callosum. An MR study identify a short, thin corpus callosum, falcine sinus persistence, reduced cerebellar vermis size, wide inferior IV ventricle, and reduced pontine bulging. Discussion: HUWE1 is a gene encoding an E3 ubitiquine ligase protein involved in nervous system development, function, and disease. The mechanisms by which HUWE1 gene affects nervous system are still largely unclear, but a growing body of literature described disease-causing variants in this gene. This report may help prenatal diagnostic experts in consider also this entity, especially when dealing with pontine and cerebellar hypoplasia findings.

SELECTION OF CITATIONS
SEARCH DETAIL