Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Stem Cell Res ; 76: 103332, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38354648

ABSTRACT

We established two iPSC lines starting from skin fibroblasts of two healthy individuals using Sendai-virus-based technique. The obtained iPSCs were characterized showing same STR profile as starting fibroblasts, normal karyotype, loss of stemness vectors, expression of stemness markers, both through real-time PCR and immunofluorescence, (OCT4, SOX2, TRA-1-60, NANOG and SSEA4) and in vitro differentiation into three germ layers.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Sendai virus/genetics , Fibroblasts/metabolism , Cell Differentiation
2.
Stem Cell Res ; 66: 103002, 2023 02.
Article in English | MEDLINE | ID: mdl-36521382

ABSTRACT

We produced an iPSC line from a patient with Joubert syndrome carrying the homozygous c.787dupC variant in the AHI1 gene. The iPSC line was obtained by reprogramming skin fibroblasts, mycoplasma-free, using Sendai-virus-based technique. Characterization of iPSCs showed the same Short Tandem Repeats profile than fibroblasts, normal karyotype, expression of staminal markers (OCT4, SOX2, SSEA4 and NANOG) and ability to differentiate into three germ layers in vitro.


Subject(s)
Abnormalities, Multiple , Eye Abnormalities , Induced Pluripotent Stem Cells , Kidney Diseases, Cystic , Humans , Induced Pluripotent Stem Cells/metabolism , Abnormalities, Multiple/metabolism , Eye Abnormalities/genetics , Eye Abnormalities/metabolism , Kidney Diseases, Cystic/metabolism , Retina , Cerebellum , Fibroblasts/metabolism , Cell Differentiation
3.
Cerebellum ; 21(4): 525-530, 2022 Aug.
Article in English | MEDLINE | ID: mdl-34089469

ABSTRACT

The glycophosphatidylinositol (GPI) anchor pathway plays an essential role in posttranslational modification of proteins to facilitate proper membrane anchoring and trafficking to lipid rafts, which is critical for many cell functions, including embryogenesis and neurogenesis. GPI biosynthesis is a multi-step process requiring the activity of over 25 distinct genes, most of them belonging to the phosphatidylinositol glycan (PIG) family and associated with rare neurodevelopmental disorders. PIGQ encodes the phosphatidylinositol glycan class Q protein and is part of the GPI-N-acetylglucosaminyltransferase complex that initiates GPI biosynthesis from phosphatidylinositol (PI) and N-acetylglucosamine (GlcNAc) on the cytoplasmic side of the endoplasmic reticulum (ER). Pathogenic variants in the PIGQ gene have been previously reported in 10 patients with congenital hypotonia, early-infantile epileptic encephalopathy, and premature death occurring in more than half cases. We detected a novel homozygous variant in PIGQ (NM_004204.5: c.1631dupA; p.Tyr544fs*79) by WES trio-analysis of a male patient with a neurodevelopmental disorder characterized by nonprogressive congenital ataxia, intellectual disability, generalized epilepsy, and cerebellar atrophy. Flow cytometry confirmed deficiency of several GPI-anchored proteins on leukocytes (CD14, FLAER). Clinical features of this case broaden the phenotypic spectrum of PIGQ-related GPI deficiency, outlining the importance of glycophosphatidylinositol (GPI) anchor pathway in the pathogenesis of cerebellar ataxia.


Subject(s)
Cerebellar Ataxia , Glycosylphosphatidylinositols , Cerebellar Ataxia/genetics , Glycosylphosphatidylinositols/genetics , Glycosylphosphatidylinositols/metabolism , Humans , Male , Membrane Proteins/genetics , Muscle Hypotonia/genetics , Muscle Hypotonia/pathology , Mutation , Pedigree , Seizures
4.
Eur J Paediatr Neurol ; 23(5): 692-706, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31481303

ABSTRACT

Early onset cerebellar Ataxia (EOAc) comprises a large group of rare heterogeneous disorders. Determination of the underlying etiology can be difficult given the broad differential diagnosis and the complexity of the genotype-phenotype relationships. This may change the diagnostic work-up into a time-consuming, costly and not always rewarding task. In this overview, the Childhood Ataxia and Cerebellar Group of the European Pediatric Neurology Society (CACG-EPNS) presents a diagnostic algorithm for EOAc patients. In seven consecutive steps, the algorithm leads the clinician through the diagnostic process, including EOA identification, application of the Inventory of Non-Ataxic Signs (INAS), consideration of the family history, neuro-imaging, laboratory investigations, genetic testing by array CGH and Next Generation Sequencing (NGS). In children with EOAc, this algorithm is intended to contribute to the diagnostic process and to allow uniform data entry in EOAc databases.


Subject(s)
Algorithms , Decision Support Systems, Clinical , Spinocerebellar Degenerations/diagnosis , Adolescent , Child , Diagnosis, Differential , Female , Humans , Male
5.
AJNR Am J Neuroradiol ; 38(12): 2385-2390, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28838911

ABSTRACT

In Joubert syndrome, the "molar tooth" sign can be associated with several additional supra- and infratentorial malformations. Here we report on 3 subjects (2 siblings, 8-14 years of age) with Joubert syndrome, showing an abnormal thick bulging of the anterior profile of the mesencephalon causing a complete obliteration of the interpeduncular fossa. DTI revealed that the abnormal tissue consisted of an ectopic white matter tract with a laterolateral transverse orientation. Tractographic reconstructions support the hypothesis of impaired axonal guidance mechanisms responsible for the malformation. The 2 siblings were compound heterozygous for 2 missense variants in the TMEM67 gene, while no mutations in a panel of 120 ciliary genes were detected in the third patient. The name "anterior mesencephalic cap dysplasia," referring to the peculiar aspect of the mesencephalon on sagittal MR imaging, is proposed for this new malformative feature.


Subject(s)
Abnormalities, Multiple/pathology , Cerebellum/abnormalities , Eye Abnormalities/pathology , Kidney Diseases, Cystic/pathology , Mesencephalon/abnormalities , Retina/abnormalities , Abnormalities, Multiple/diagnostic imaging , Adolescent , Cerebellum/diagnostic imaging , Cerebellum/pathology , Child , Eye Abnormalities/diagnostic imaging , Female , Humans , Kidney Diseases, Cystic/diagnostic imaging , Magnetic Resonance Imaging , Male , Mesencephalon/diagnostic imaging , Retina/diagnostic imaging , Retina/pathology
6.
Clin Genet ; 91(3): 441-447, 2017 03.
Article in English | MEDLINE | ID: mdl-27102574

ABSTRACT

NDUFB11, a component of mitochondrial complex I, is a relatively small integral membrane protein, belonging to the "supernumerary" group of subunits, but proved to be absolutely essential for the assembly of an active complex I. Mutations in the X-linked nuclear-encoded NDUFB11 gene have recently been discovered in association with two distinct phenotypes, i.e. microphthalmia with linear skin defects and histiocytoid cardiomyopathy. We report on a male with complex I deficiency, caused by a de novo mutation in NDUFB11 and displaying early-onset sideroblastic anemia as the unique feature. This is the third report that describes a mutation in NDUFB11, but all are associated with a different phenotype. Our results further expand the molecular spectrum and associated clinical phenotype of NDUFB11 defects.


Subject(s)
Acidosis, Lactic/genetics , Anemia, Sideroblastic/genetics , Electron Transport Complex I/genetics , Microphthalmos/genetics , Acidosis, Lactic/complications , Acidosis, Lactic/physiopathology , Anemia, Sideroblastic/complications , Anemia, Sideroblastic/physiopathology , Child , DNA, Mitochondrial/genetics , Electron Transport Complex I/deficiency , Genetic Predisposition to Disease , Humans , Male , Microphthalmos/physiopathology , Mutation , Pedigree , Phenotype , Tyrosine-tRNA Ligase
8.
Neurobiol Dis ; 91: 21-36, 2016 07.
Article in English | MEDLINE | ID: mdl-26916954

ABSTRACT

Heterozygous mutations in the PINK1 gene are considered a susceptibility factor to develop early-onset Parkinson's disease (PD), as supported by dopamine hypometabolism in asymptomatic mutation carriers and subtle alterations of dopamine-dependent striatal synaptic plasticity in heterozygous PINK1 knockout (PINK1(+/-)) mice. The aim of the present study was to investigate whether exposure to low-dose rotenone of heterozygous PINK1(+/-) mice, compared to their wild-type PINK1(+/+) littermates, could impact on dopamine-dependent striatal synaptic plasticity, in the absence of apparent structural alterations. Mice were exposed to a range of concentrations of rotenone (0.01-1mg/kg). Chronic treatment with concentrations of rotenone up to 0.8mg/kg did not cause manifest neuronal loss or changes in ATP levels both in the striatum or substantia nigra of PINK1(+/-) and PINK1(+/+) mice. Moreover, rotenone (up to 0.8mg/kg) treatment did not induce mislocalization of the mitochondrial membrane protein Tom20 and release of cytochrome c in PINK1(+/-) striata. Accordingly, basic electrophysiological properties of nigral dopaminergic and striatal medium spiny neurons (MSNs) were normal. Despite the lack of gross alterations in neuronal viability in chronically-treated PINK1(+/-), a complete loss of both long-term depression (LTD) and long-term potentiation (LTP) was recorded in MSNs from PINK1(+/-) mice treated with a low rotenone (0.1mg/kg) concentration. Even lower concentrations (0.01mg/kg) blocked LTP induction in heterozygous PINK1(+/-) MSNs compared to PINK1(+/+) mice. Of interest, chronic pretreatment with the antioxidants alpha-tocopherol and Trolox, a water-soluble analog of vitamin E and powerful antioxidant, rescued synaptic plasticity impairment, confirming that, at the doses we utilized, rotenone did not induce irreversible alterations. In this model, chronic exposure to low-doses of rotenone was not sufficient to alter mitochondrial integrity and ATP production, but profoundly impaired the expression of long-term plasticity at corticostriatal synapses in PINK1 heterozygous knockout mice, suggesting that disruption of synaptic plasticity may represent an early feature of a pre-manifesting state of the disease, and a potential tool to test novel neuroprotective agents.


Subject(s)
Corpus Striatum/drug effects , Neuronal Plasticity/drug effects , Protein Kinases/genetics , Rotenone/pharmacology , Substantia Nigra/drug effects , Synapses/drug effects , Animals , Corpus Striatum/metabolism , Dopamine/metabolism , Heterozygote , Long-Term Potentiation/drug effects , Mice, Knockout , Neuronal Plasticity/genetics , Neurons/drug effects , Neurons/metabolism , Protein Kinases/drug effects , Substantia Nigra/metabolism , Synapses/metabolism
9.
Eur J Neurol ; 22(1): 178-86, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25164370

ABSTRACT

BACKGROUND AND PURPOSE: Mutations in the PLA2G6 gene are causative of PLA2G6-associated neurodegeneration (PLAN), a spectrum of neurodegenerative conditions including infantile, childhood and adult onset forms. METHODS: Seventeen North African patients with a clinical suspicion of infantile-onset PLAN underwent clinical, neurophysiological and neuroimaging examinations, and PLA2G6 sequencing. Haplotype analysis was performed to date the identified founder mutation. RESULTS: All patients carried biallelic mutations in PLA2G6. Sixteen children had the commonest form of infantile-onset PLAN, with early onset of psychomotor regression, hypotonia, pyramidal and cerebellar signs, and abnormal ocular movements. The phenotype was highly homogeneous, with rapid development of severe spastic tetraparesis, cognitive impairment and optic atrophy. Neuroimaging showed cerebellar atrophy and claval hypertrophy to be the commonest and earliest signs, whilst cerebellar cortex hyperintensity and pallidal iron deposition were later findings. Motor or sensory-motor neuropathy and electroencephalogram fast rhythms were also frequent. Nine patients from six families shared the same founder mutation (p.V691del) which probably arose by the late seventeenth century. Only one patient fitted the diagnosis of the much rarer childhood-onset PLAN. Despite the early onset (18 months), clinical progression was slower, with behavioral disturbances and dystonia. Typical features of infantile-onset PLAN such as hypotonia, nystagmus/strabismus, optic atrophy, electroencephalogram fast rhythms and motor neuropathy were absent. Cerebellar atrophy, claval hypertrophy and pallidal hypointensity were evident at brain magnetic resonance imaging. This patient carried a missense variant predicted to be less deleterious. CONCLUSIONS: The PLAN-associated phenotypes and the challenges of diagnosing the childhood-onset form are delineated, and a common North African founder mutation is identifed.


Subject(s)
Age of Onset , Group VI Phospholipases A2/genetics , Mutation/genetics , Neuroaxonal Dystrophies/classification , Atrophy/pathology , Child , Child, Preschool , Electroencephalography , Electromyography , Female , Founder Effect , Humans , Infant , Libya , Magnetic Resonance Imaging , Male , Neuroaxonal Dystrophies/genetics , Neuroaxonal Dystrophies/pathology , Neuroaxonal Dystrophies/physiopathology , Pedigree , Phenotype , Tunisia
10.
Curr Mol Med ; 14(8): 1079-1088, 2014.
Article in English | MEDLINE | ID: mdl-25323865

ABSTRACT

Parkinson Disease (PD) is a common neurodegenerative disorder of intricate etiology, caused by progressive loss of aminergic neurons and accumulation of Lewy bodies. The predominant role of genetics in the etiology of the disease has emerged since the identification of the first pathogenetic mutation in SNCA (alpha-synuclein) gene, back in 1997. Mendelian parkinsonisms, a minority among all PD forms, have been deeply investigated, with 19 loci identified. More recently, genome wide association studies have provided convincing evidence that variants in some of these genes, as well as in other genes, may confer an increased risk for late onset, sporadic PD. Moreover, the finding that heterozygous mutations in the GBA gene (mutated in Gaucher disease) are among the strongest genetic susceptibility factors for PD, has widened the scenario of PD genetic background to enclose a number of genes previously associated to distinct disorders, such as genes causative of spinocerebellar ataxias, mitochondrial disorders and fragile X syndrome. At present, the genetic basis of PD defines a continuum from purely mendelian forms (such as those caused by autosomal recessive genes) to multifactorial inheritance, resulting from the variable interplay of many distinct genetic variants and environmental factors.

11.
Eur J Neurol ; 21(9): 1242-6, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24471704

ABSTRACT

BACKGROUND AND PURPOSE: Akinetic crisis (AC) is the most severe and possibly lethal complication of parkinsonism. It occurs with an incidence of 3‰ Parkinson's disease patients per year, but it is not known whether genetically determined parkinsonism is more or less susceptible to this complication. METHODS: In a cohort of 756 parkinsonian patients the incidence and outcome of AC was prospectively assessed. A total of 142 of the parkinsonian patients were tested for genetic mutations because of familial parkinsonism, and 20 patients resulted positive: in four the mutation definitely involved mitochondrial functions (POLG1, PINK1), two presented with LRRK2 mutation, nine presented with GBA mutation and five presented with Park 4 different mutations. RESULTS: Akinetic crisis occurred in 30 patients for an incidence of 2.8‰ persons/year and was lethal in seven (23%), not dissimilarly from known incidences of this complication. Yet six of 30 patients were carriers of genetic mutations, one GBA, one LRRK2, one POLG1 and three PINK1. In POLG1 and PINK1 carriers, the syndrome was recurrent and was fatal in three. Incidence of AC was 3.0‰ in familiar parkinsonism, 21.2‰ in genetic parkinsonisms. CONCLUSIONS: Our preliminary findings suggest that the incidence of AC is remarkably increased in carriers of these genetic mutations.


Subject(s)
Mitochondria/genetics , Mutation/genetics , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology , Aged , Cohort Studies , DNA Polymerase gamma , DNA-Directed DNA Polymerase/genetics , Female , Glucosylceramidase/genetics , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Male , Middle Aged , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics
12.
Cell Death Differ ; 20(7): 920-30, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23519076

ABSTRACT

Mutations in the PINK1 gene are a frequent cause of autosomal recessive Parkinson's disease (PD). PINK1 encodes a mitochondrial kinase with neuroprotective activity, implicated in maintaining mitochondrial homeostasis and function. In concurrence with Parkin, PINK1 regulates mitochondrial trafficking and degradation of damaged mitochondria through mitophagy. Moreover, PINK1 can activate autophagy by interacting with the pro-autophagic protein Beclin-1. Here, we report that, upon mitochondrial depolarization, PINK1 interacts with and phosphorylates Bcl-xL, an anti-apoptotic protein also known to inhibit autophagy through its binding to Beclin-1. PINK1-Bcl-xL interaction does not interfere either with Beclin-1 release from Bcl-xL or the mitophagy pathway; rather it protects against cell death by hindering the pro-apoptotic cleavage of Bcl-xL. Our data provide a functional link between PINK1, Bcl-xL and apoptosis, suggesting a novel mechanism through which PINK1 regulates cell survival. This pathway could be relevant for the pathogenesis of PD as well as other diseases including cancer.


Subject(s)
Apoptosis/physiology , Mitochondria/physiology , Protein Kinases/physiology , bcl-X Protein/metabolism , Apoptosis Regulatory Proteins/physiology , Autophagy/physiology , Beclin-1 , Cell Line, Tumor , Cell Survival/physiology , HEK293 Cells , Humans , Membrane Proteins/physiology , Phosphorylation/physiology , Signal Transduction/physiology
13.
Arch Ital Biol ; 150(2-3): 194-217, 2012.
Article in English | MEDLINE | ID: mdl-23165879

ABSTRACT

Mutations in the PTEN-induced putative kinase1 (PINK1) represent the second most frequent cause of autosomal recessive Parkinson's disease. The PINK1 protein mainly localizes to mitochondria and interacts with a variety of proteins, including the pro-autophagy protein beclin1 and the ubiquitin-ligase parkin. Upon stress conditions, PINK1 is known to recruit parkin at the surface of dysfunctional mitochondria and to activate the mitophagy cascade. Aim of this study was to use a simple and highly reproducible catecholamine cell model and transmission electron microscopy to characterize whether PINK1 could affect mitochondrial homeostasis, the recruitment of specific proteins at mitochondria, mitophagy and apoptosis. Samples were analyzed both in baseline conditions and following treatment with methamphetamine (METH), a neurotoxic compound which strongly activates autophagy and produces mitochondrial damage. Our data provide robust sub-cellular evidence that the modulation of PINK1 levels dramatically affects the morphology and number of mitochondria and the amount of cell death. In particular, especially upon METH exposure, PINK1 is able to increase the total number of mitochondria, concurrently recruit beclin1, parkin and ubiquitin and enhance the clearance of damaged mitochondria. In the absence of functional PINK1 and upon autophagy stress, we observe a failure of the autophagy system at large, with marked accumulation of dysfunctional mitochondria and dramatic increase of apoptotic cell death. These findings highlight the strong neuroprotective role of PINK1 as a key protein in the surveillance and regulation of mitochondrial homeostasis.


Subject(s)
Autophagy/genetics , Mitochondria/genetics , Mutation/genetics , Protein Kinases/genetics , Animals , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Cell Death/genetics , Central Nervous System Stimulants/pharmacology , Humans , Membrane Proteins/metabolism , Methamphetamine/pharmacology , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/ultrastructure , PC12 Cells/drug effects , PC12 Cells/ultrastructure , RNA, Small Interfering/genetics , Rats , Subcellular Fractions/metabolism , Subcellular Fractions/ultrastructure , Transfection , Ubiquitin-Protein Ligases/metabolism
14.
Neurobiol Dis ; 38(2): 192-200, 2010 May.
Article in English | MEDLINE | ID: mdl-20053375

ABSTRACT

DYT1 dystonia is an autosomal-dominantly inherited movement disorder, which is usually caused by a GAG deletion in the TOR1A gene. Due to the reduced penetrance of approximately 30-40%, the determination of the mutation in a subject is of limited use with regard to actual manifestation of symptoms. In the present study, we used Affymetrix oligonucleotide microarrays to analyze global gene expression in blood samples of 15 manifesting and 15 non-manifesting mutation carriers in order to identify a susceptibility profile beyond the GAG deletion which is associated with the manifestation of symptoms in DYT1 dystonia. We identified a genetic signature which distinguished between asymptomatic mutation carriers and symptomatic DYT1 patients with 86.7% sensitivity and 100% specificity. This genetic signature could correctly predict the disease state in an independent test set with a sensitivity of 87.5% and a specificity of 85.7%. Conclusively, this genetic signature might provide a possibility to distinguish DYT1 patients from asymptomatic mutation carriers.


Subject(s)
Dystonia Musculorum Deformans/genetics , Gene Expression Profiling , Molecular Chaperones/genetics , Adult , Female , Genetic Predisposition to Disease , Genetic Testing , Humans , Male , Middle Aged , Mutation , Oligonucleotide Array Sequence Analysis , Penetrance , Trinucleotide Repeats
15.
Cell Death Differ ; 17(6): 962-74, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20057503

ABSTRACT

Mutations in the PINK1 gene cause autosomal recessive Parkinson's disease. The PINK1 gene encodes a protein kinase that is mitochondrially cleaved to generate two mature isoforms. In addition to its protective role against mitochondrial dysfunction and apoptosis, PINK1 is also known to regulate mitochondrial dynamics acting upstream of the PD-related protein Parkin. Recent data showed that mitochondrial Parkin promotes the autophagic degradation of dysfunctional mitochondria, and that stable PINK1 silencing may have an indirect role in mitophagy activation. Here we report a new interaction between PINK1 and Beclin1, a key pro-autophagic protein already implicated in the pathogenesis of Alzheimer's and Huntington's diseases. Both PINK1 N- and C-terminal are required for the interaction, suggesting that full-length PINK1, and not its cleaved isoforms, interacts with Beclin1. We also demonstrate that PINK1 significantly enhances basal and starvation-induced autophagy, which is reduced by knocking down Beclin1 expression or by inhibiting the Beclin1 partner Vps34. A mutant, PINK1(W437X), interaction of which with Beclin1 is largely impaired, lacks the ability to enhance autophagy, whereas this is not observed for PINK1(G309D), a mutant with defective kinase activity but unaltered ability to bind Beclin1. These findings identify a new function of PINK1 and further strengthen the link between autophagy and proteins implicated in the neurodegenerative process.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy , Membrane Proteins/metabolism , Protein Kinases/metabolism , Apoptosis Regulatory Proteins/analysis , Beclin-1 , Cell Line, Tumor , HeLa Cells , Humans , Membrane Proteins/analysis , Mitochondria/chemistry , Mitochondria/ultrastructure , Mutation , Protein Kinases/analysis , Protein Kinases/genetics , Sequence Deletion , Two-Hybrid System Techniques
16.
Eye (Lond) ; 24(2): 222-5, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19461662

ABSTRACT

PURPOSE: Joubert syndrome (JS) is an autosomal-recessive inherited complex malformation of the midbrain-hindbrain. It has been associated with ocular and oculomotor abnormalities. The aim of our study was to extend the ophthalmic knowledge in JS and to add new findings. METHODS: In a retrospective study, 10 consecutive patients, who met the revised diagnostic criteria of JS were included. Mutation analysis was carried out in all the cases. Each patient underwent a comprehensive neuro-ophthalmological examination. RESULTS: Bilateral drusen of the optic disc were found in two patients. Four patients showed bilateral morphological and functional signs of retinal dystrophy (CEP290 mutation in two cases and AHI1 mutation in one case). In nine patients performance during smooth pursuit, saccades, and vestibulo-ocular reflex (VOR) cancellation was poor. CONCLUSIONS: To the best of our knowledge, the association of optic disc drusen with JS has not yet been described. In support of the earlier findings, decreased smooth pursuit and VOR cancellation, as well as partial-to-complete oculomotor apraxia seem to be the key oculomotor features of JS. Genotype-phenotype correlations showed the predictive value of CEP290 and AHI1 mutations for retinal involvement.


Subject(s)
Retina/pathology , Abnormalities, Multiple , Adolescent , Adult , Cerebellar Diseases/genetics , Cerebellar Diseases/physiopathology , Cerebellum/abnormalities , Child , DNA Mutational Analysis , Eye Abnormalities/genetics , Eye Abnormalities/physiopathology , Female , Genotype , Humans , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/physiopathology , Male , Ocular Motility Disorders/physiopathology , Optic Disk Drusen/pathology , Retina/abnormalities , Retina/physiopathology , Retinal Pigment Epithelium/pathology , Retrospective Studies , Saccades/physiology , Visual Acuity/physiology , Young Adult
17.
Am J Med Genet A ; 149A(7): 1511-5, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19533793

ABSTRACT

Joubert syndrome (JS) is characterized by hypotonia, ataxia, developmental delay, and a typical neuroimaging finding, the so-called "molar tooth sign" (MTS). The association of MTS and polymicrogyria (PMG) has been reported as a distinct JS-related disorder (JSRD). So far, five patients have been reported with this phenotype, only two of them being siblings. We report on one additional family, describing a living child with JS and PMG, and the corresponding neuropathological picture in the aborted brother. No mutations were detected in the AHI1 gene, the only so far associated with the JS + PMG phenotype. Moreover, linkage analysis allowed excluding all known gene loci, suggesting further genetic heterogeneity.


Subject(s)
Abnormalities, Multiple/diagnosis , Malformations of Cortical Development/diagnosis , Malformations of Cortical Development/pathology , Siblings , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Abortion, Eugenic , Child , DNA Mutational Analysis , Female , Fetal Diseases/diagnosis , Fetal Diseases/genetics , Fetal Diseases/pathology , Humans , Male , Malformations of Cortical Development/complications , Malformations of Cortical Development/genetics , Pregnancy , Syndrome
18.
Eur J Neurol ; 16(8): 949-52, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19473353

ABSTRACT

BACKGROUND AND PURPOSE: Adult-onset dystonia may be related, amongst other factors, to abnormal neuronal plasticity in cortical and subcortical structures. Brain-derived neurotrophic factor is a major modulator of synaptic efficiency and neuronal plasticity. Recent works documented that a single nucleotide polymorphism (SNP) of the BDNF gene, the Val66Met SNP, modulates short-term plastic changes within motor cortical circuits. In this study we aimed at exploring the effect of this SNP upon the risk of developing common forms of primary adult-onset dystonia. METHODS: We explored the influence of the Val66Met SNP of the BDNF gene on the risk of cranial and cervical dystonia in a cohort of 156 Italian patients and 170 age- and gender-matched healthy control subjects drawn from the same population. RESULTS: The presence of the rare Met allele was not significantly associated with the diagnosis of dystonia (age- and gender-adjusted odds ratios of 1.22, P = 0.38). The study had a >90% power to detect a 50% change in the risk of developing cranial-cervical dystonia associated with the presence of the Met allele. Moreover, there was no relationship between Val66Met SNP and age at dystonia onset or type of dystonia. CONCLUSION: Our data do not support the common variant Val66Met of the BDNF gene as an etiologic factor shared by the various forms of primary adult-onset dystonia.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Dystonic Disorders/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Age of Onset , Case-Control Studies , Cohort Studies , Female , Humans , Italy/epidemiology , Male , Middle Aged , Odds Ratio , Sequence Analysis, DNA
19.
Neuropediatrics ; 40(6): 287-90, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20446224

ABSTRACT

Developmental delay and subsequent impaired cognitive functions are present in almost all patients with Joubert syndrome (JS). We report on a 20-year-old woman with mild clinical signs of JS (minimal truncal ataxia and oculomotor apraxia) but typical molar tooth sign on neuroimaging, normal full scale (IQ=93), verbal (IQ=93), and performance intelligence quotient (IQ=94). Only minor difficulties in visual-spatial organization and in some executive functions could be detected. This pattern of deficits is partly reminiscent of the cerebellar cognitive affective syndrome. Her diagnosis was only reached following the diagnosis of JS in two brothers with severe cognitive impairment. Molecular investigations demonstrated a homozygous mutation in the INPP5E gene. This exceptional observation confirms that normal cognitive functions are possible in JS and corroborates the well known intrafamilial variability.


Subject(s)
Cerebellar Diseases/complications , Cognition Disorders/etiology , Developmental Disabilities/complications , Movement Disorders/complications , Adolescent , Humans , Magnetic Resonance Imaging/methods , Neuropsychological Tests
20.
Clin Neurophysiol ; 119(8): 1864-1869, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18571468

ABSTRACT

OBJECTIVE: The only known genetic cause of early-onset primary torsion dystonia is the GAG deletion in the DYT1 gene. Due to the reduced penetrance, many mutation carriers remain clinically asymptomatic, despite the presence of subclinical abnormalities, mainly in the motor control circuitry. Our aim was to investigate whether the DYT1 mutation impairs the inner simulation of movements, a fundamental function for motor planning and execution, which relies upon cortical and subcortical systems, dysfunctional in dystonia. METHODS: DYT1 manifesting patients, DYT1 non-manifesting carriers and control subjects were asked to fixate body (hand, foot, face) or non-body (car) stimuli on a computer screen. Stimuli were presented at different degrees of orientations and subjects had to mentally rotate them, in order to give a laterality judgement. Reaction times and accuracy were collected. RESULTS: DYT1 carriers, manifesting and non-manifesting dystonic symptoms, were slower in mentally rotating body parts (but not cars) than control subjects. CONCLUSIONS: The DYT1 gene mutation is associated with a slowness in mental simulation of movements, independently from the presence of motor symptoms. SIGNIFICANCE: These findings suggest that the cognitive representation of body movements may be altered subclinically in dystonia, thus contributing to the endophenotypic trait of disease.


Subject(s)
Dystonia/genetics , Dystonia/physiopathology , Molecular Chaperones/genetics , Movement/physiology , Mutation/genetics , Trinucleotide Repeats/genetics , Adolescent , Adult , Aged , Analysis of Variance , Child , DNA Mutational Analysis , Female , Functional Laterality/genetics , Humans , Male , Middle Aged , Orientation/physiology , Photic Stimulation/methods , Posture , Psychomotor Performance , Reaction Time/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...