Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 59
1.
Parkinsonism Relat Disord ; 84: 98-104, 2021 03.
Article En | MEDLINE | ID: mdl-33607528

INTRODUCTION: Mitochondrial membrane protein-associated neurodegeneration (MPAN) is a rare neurological syndrome caused by pathogenic variants in the C19orf12 and is characterized by iron deposition in the basal ganglia and substantia nigra. Only a limited number of cohort studies were published to date and the prevalence of MPAN remains uncertain. METHODS: Recruited subjects with MPAN in Russia were diagnosed by whole-exome sequencing or Sanger sequencing of the C19orf12 gene. Data of over 14000 whole exome sequencing analyses was used to calculate the estimated disease frequency. RNA analysis was performed by RT-PCR. QSVanalyzer software was used to quantify the allelic disbalance. RESULTS: We describe the clinical and molecular characterizations of 17 patients with MPAN. DNA analysis detected three previously undescribed pathogenic/likely pathogenic variants in the C19orf12 gene. The estimated disease frequency was calculated to be 1:619150. We describe unusual clinical observations in several cases. One patient showed severe neurogenic muscle weakness along with a lack of marked spasticity or optic nerve atrophy. In another mild clinical case with the NM_001031726.3:c.204_214del (p.(Gly69Argfs*10)) variant in a heterozygous state, a marked allelic disbalance was observed on the RNA level with reduced expression level of the wild-type allele. Thus, this case became the first one of a possible regulatory variant causing MPAN. CONCLUSION: We reported a detailed clinical and molecular characterization of the third-largest MPAN cohort. We expanded the mutational and clinical spectrum of MPAN. Moreover, we calculated the estimated MPAN frequency in the Russian population for the first time.


Globus Pallidus/pathology , Iron Metabolism Disorders , Membrane Proteins , Mitochondrial Membranes , Mitochondrial Proteins , Neuroaxonal Dystrophies , Substantia Nigra/pathology , Adolescent , Adult , Child , Female , Globus Pallidus/diagnostic imaging , Humans , Iron Metabolism Disorders/epidemiology , Iron Metabolism Disorders/genetics , Iron Metabolism Disorders/pathology , Iron Metabolism Disorders/physiopathology , Magnetic Resonance Imaging , Neuroaxonal Dystrophies/epidemiology , Neuroaxonal Dystrophies/genetics , Neuroaxonal Dystrophies/pathology , Neuroaxonal Dystrophies/physiopathology , Retrospective Studies , Russia/epidemiology , Substantia Nigra/diagnostic imaging , Exome Sequencing
4.
Ann Clin Transl Neurol ; 7(8): 1436-1442, 2020 08.
Article En | MEDLINE | ID: mdl-32767480

FBXO7 is implicated in the ubiquitin-proteasome system and parkin-mediated mitophagy. FBXO7defects cause a levodopa-responsive parkinsonian-pyramidal syndrome(PPS). METHODS: We investigated the disease molecular bases in a child with PPS and brain iron accumulation. RESULTS: A novel homozygous c.368C>G (p.S123*) FBXO7 mutation was identified in a child with spastic paraplegia, epilepsy, cerebellar degeneration, levodopa nonresponsive parkinsonism, and brain iron deposition. Patient's fibroblasts assays demonstrated an absence of FBXO7 RNA expression leading to impaired proteasome degradation and accumulation of poly-ubiquitinated proteins. CONCLUSION: This novel FBXO7 phenotype associated with impaired proteasome activity overlaps with neurodegeneration with brain iron accumulation disorders.


F-Box Proteins/genetics , Iron Metabolism Disorders , Neuroaxonal Dystrophies , Parkinsonian Disorders , Proteasome Endopeptidase Complex/metabolism , Adult , Consanguinity , Epilepsy/enzymology , Epilepsy/genetics , Epilepsy/pathology , Epilepsy/physiopathology , Female , Humans , Iron Metabolism Disorders/enzymology , Iron Metabolism Disorders/genetics , Iron Metabolism Disorders/pathology , Iron Metabolism Disorders/physiopathology , Neuroaxonal Dystrophies/enzymology , Neuroaxonal Dystrophies/genetics , Neuroaxonal Dystrophies/pathology , Neuroaxonal Dystrophies/physiopathology , Paraplegia/enzymology , Paraplegia/genetics , Paraplegia/pathology , Paraplegia/physiopathology , Parkinsonian Disorders/enzymology , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology , Parkinsonian Disorders/physiopathology , Spinocerebellar Degenerations/enzymology , Spinocerebellar Degenerations/genetics , Spinocerebellar Degenerations/pathology , Spinocerebellar Degenerations/physiopathology , Syndrome , Young Adult
5.
Pediatr Neurol ; 109: 56-62, 2020 08.
Article En | MEDLINE | ID: mdl-32387008

BACKGROUND: Mutations in the X-linked gene WDR45 cause neurodegeneration with brain iron accumulation type 5. Global developmental delay occurs at an early age with slow progression to dystonia, parkinsonism, and dementia due to progressive iron accumulation in the brain. METHODS: We present 17 new cases and reviewed 106 reported cases of neurodegeneration with brain iron accumulation type 5. Detailed information related to developmental history and key time to event measures was collected. RESULTS: Within this cohort, there were 19 males. Most individuals were molecularly diagnosed by whole-exome testing. Overall 10 novel variants were identified across 11 subjects. All individuals were affected by developmental delay, most prominently in verbal skills. Most individuals experienced a decline in motor and cognitive skills. Although most individuals were affected by seizures, the spectrum ranged from provoked seizures to intractable epilepsy. The imaging findings varied as well, often evolving over time. The classic iron accumulation in the globus pallidus and substantia nigra was noted in half of our cohort and was associated with older age of image acquisition, whereas myelination abnormalities were associated with younger age. CONCLUSIONS: WDR45 mutations lead to a progressive and evolving disorder whose diagnosis is often delayed. Developmental delay and seizures predominate in early childhood, followed by a progressive decline of neurological function. There is variable expressivity in the clinical phenotypes of individuals with WDR45 mutations, suggesting that this gene should be considered in the diagnostic evaluation of children with myelination abnormalities, iron deposition, developmental delay, and epilepsy depending on the age at evaluation.


Carrier Proteins/genetics , Demyelinating Diseases , Developmental Disabilities , Epilepsy , Iron Metabolism Disorders , Neuroaxonal Dystrophies , Adolescent , Adult , Child , Child, Preschool , Cohort Studies , Demyelinating Diseases/diagnosis , Demyelinating Diseases/etiology , Demyelinating Diseases/genetics , Demyelinating Diseases/physiopathology , Developmental Disabilities/diagnosis , Developmental Disabilities/etiology , Developmental Disabilities/genetics , Developmental Disabilities/physiopathology , Epilepsy/diagnosis , Epilepsy/etiology , Epilepsy/genetics , Epilepsy/physiopathology , Female , Humans , Infant , Iron Metabolism Disorders/complications , Iron Metabolism Disorders/diagnosis , Iron Metabolism Disorders/genetics , Iron Metabolism Disorders/physiopathology , Male , Middle Aged , Neuroaxonal Dystrophies/complications , Neuroaxonal Dystrophies/diagnosis , Neuroaxonal Dystrophies/genetics , Neuroaxonal Dystrophies/physiopathology , Phenotype , Exome Sequencing , Young Adult
6.
Eur J Med Genet ; 63(3): 103765, 2020 Mar.
Article En | MEDLINE | ID: mdl-31536831

Neurodegeneration with brain iron accumulation (NBIA) comprises a group of rare genetic disorders characterized by progressive extrapyramidal and other neurological symptoms due to focal iron accumulation in the basal ganglia (Adidi et al., 2016). ß-Propeller protein-associated neurodegeneration (BPAN) is the most recently identified subtype of NBIA caused by heterozygous variants in WDR45 (OMIM: *300526) at Xp11.23. We report the clinical neurophysiological and neuro-imaging findings of a new subtype of BPAN in a 6 year-old female patient, who was identified to have a large de novo WDR45 deletion who presented in the first year of life with early onset global developmental delay, severe cognitive impairment, generalized hypotonia and a corticosteroid responsive epileptic encephalopathy.


Carrier Proteins/genetics , Iron Metabolism Disorders/diagnosis , Neuroaxonal Dystrophies/diagnosis , Basal Ganglia/diagnostic imaging , Basal Ganglia/pathology , Brain/diagnostic imaging , Brain/pathology , Child , Epileptic Syndromes/genetics , Female , Heterozygote , Humans , Iron Metabolism Disorders/diagnostic imaging , Iron Metabolism Disorders/genetics , Iron Metabolism Disorders/physiopathology , Magnetic Resonance Imaging , Neuroaxonal Dystrophies/diagnostic imaging , Neuroaxonal Dystrophies/genetics , Neuroaxonal Dystrophies/physiopathology , Sequence Deletion
7.
Neurol Sci ; 41(3): 679-685, 2020 Mar.
Article En | MEDLINE | ID: mdl-31773358

BACKGROUND AND AIM: Neurodegeneration with brain iron accumulation (NBIA) and Wilson's disease (WD) is considered the prototype of neurodegenerative disorders characterised by the overloading of iron and copper in the central nervous system. Growing evidence has unveiled the involvement of these metals in brain cortical neurotransmission. Aim of this study was to assess cortical excitability profile due to copper and iron overload. METHODS: Three patients affected by NBIA, namely two patients with a recessive hereditary parkinsonism (PARK9) and one patient with aceruloplasminemia and 7 patients with neurological WD underwent transcranial magnetic stimulation (TMS) protocols to assess cortical excitability. Specifically, we evaluated the motor thresholds that reflect membrane excitability related to the voltage-gated sodium channels in the neurons of the motor system and the ease of activation of motor cortex via glutamatergic networks, and ad hoc TMS protocols to probe inhibitory-GABAergic (short interval intracortical inhibition, SICI; short-latency afferent inhibition, SAI; cortical silent period, CSP) and excitatory intracortical circuitry (intracortical facilitation, ICF). RESULTS: Patients with NBIA exhibited an abnormal prolongation of CSP respect to HC and WD patients. On the contrary, neurological WD displayed higher motor thresholds and reduced CSP and SICI. CONCLUSION: Hereditary conditions due to overload of copper and iron exhibited peculiar cortical excitability profiles that can help during differential diagnosis between these conditions. Moreover, such results can give us more clues about the role of metals in acquired neurodegenerative disorders, such as Parkinson disease, Alzheimer disease, and multiple sclerosis.


Ceruloplasmin/deficiency , Cortical Excitability/physiology , Hepatolenticular Degeneration/physiopathology , Iron Metabolism Disorders/physiopathology , Neuroaxonal Dystrophies/physiopathology , Neurodegenerative Diseases/physiopathology , Parkinsonian Disorders/physiopathology , Adult , Female , Humans , Male , Middle Aged , Transcranial Magnetic Stimulation , Young Adult
11.
Parkinsonism Relat Disord ; 61: 179-186, 2019 04.
Article En | MEDLINE | ID: mdl-30340910

INTRODUCTION: PLA2G6-associated neurodegeneration (PLAN) comprises a continuum of three phenotypes with overlapping clinical and radiologic features. METHODS: Observational clinical study in a cohort of infantile and childhood onset PLAN patients and genetic analysis of the PLA2G6 gene. We analysed chronological evolution in terms of age at onset and disease course through a 66-item questionnaire. We performed qualitative and quantitative assessment of MRI abnormalities and searched for clinical and radiological phenotype and genotype correlations. RESULTS: Sixteen PLAN patients (mean age: 10.2 years, range 3-33) were evaluated, with a median onset (years) of signs/symptoms as follows: neurological regression (1.5), oculomotor abnormalities (1.5), hypotonia (1.8), gait loss (2.2), pyramidal signs (3.0), axonal neuropathy (3.0), dysphagia (4.0), optic atrophy (4.0), psychiatric symptoms (4.0), seizures (5.9), joint contractures (6.0), dystonia (8.0), bladder dysfunction (13.0) and parkinsonism (15.0). MRI assessment identified cerebellar atrophy (19/19), brain iron deposition (10/19), clava hypertrophy (8/19) and T2/FLAIR hyperintensity of the cerebellar cortex (6/19). The mid-sagittal vermis relative diameter (MVRD) correlated with age at onset of clinical variants, meaning that the earlier the onset, the more severe the cerebellar atrophy. All patients harboured missense, nonsense and frameshift mutations in PLA2G6, including four novel variants. CONCLUSIONS: Cerebellar atrophy was a universal radiological sign in infantile and childhood onset PLAN, and correlated with the severity of the phenotype. Iron accumulation within the globus pallidum and substantia nigra was also a common and strikingly uniform feature regardless of the phenotype.


Cerebellum/pathology , Globus Pallidus/metabolism , Neuroaxonal Dystrophies/pathology , Neuroaxonal Dystrophies/physiopathology , Substantia Nigra/metabolism , Adolescent , Adult , Age of Onset , Atrophy/pathology , Cerebellum/diagnostic imaging , Child , Child, Preschool , Cross-Sectional Studies , Globus Pallidus/diagnostic imaging , Group VI Phospholipases A2/genetics , Humans , Magnetic Resonance Imaging , Neuroaxonal Dystrophies/diagnostic imaging , Phenotype , Severity of Illness Index , Substantia Nigra/diagnostic imaging , Young Adult
13.
Neurocase ; 24(3): 161-165, 2018 06.
Article En | MEDLINE | ID: mdl-30088953

Mitochondrial membrane protein-associated neurodegeneration (MPAN) is an autosomal recessive disorder caused by mutation in the C19orf12 gene. We report a compound heterozygous c.[32C>T];[205G>A;424A>G] (p.[Thr11Met];[Gly69Arg;Lys142Glu]) Czech patient who manifested with right foot dystonia, impaired handwriting, attention deficit, and signs of iron accumulation on brain MRI. Gradually, he developed dysarthria, spastic-dystonic gait, pedes cavi, and atrophy of leg muscles. Additionally, we report demographic parameters, clinical signs, and allelic frequencies of C19orf12 mutations of all published MPAN cases. We compared the most frequent mutations, p.Thr11Met and p.Gly69ArgfsX10; the latter was associated with younger age at onset and more frequent optic atrophy in homozygotes.


Interleukins/genetics , Iron Metabolism Disorders , Mitochondrial Membrane Transport Proteins , Neuroaxonal Dystrophies , Adult , Humans , Iron Metabolism Disorders/diagnostic imaging , Iron Metabolism Disorders/genetics , Iron Metabolism Disorders/physiopathology , Male , Neuroaxonal Dystrophies/diagnostic imaging , Neuroaxonal Dystrophies/genetics , Neuroaxonal Dystrophies/physiopathology , Young Adult
14.
Eur J Paediatr Neurol ; 22(5): 854-861, 2018 Sep.
Article En | MEDLINE | ID: mdl-29859652

BACKGROUND AND AIMS: Phospholipase A2 associated neurodegeneration (PLAN) is a heterogeneous autosomal recessive disorder caused by mutations in the ubiquitously expressed PLA2G6 gene. It is responsible for delayed brain iron accumulation and induces progressive psychomotor regression. We report the concomitant clinical, radiological and neurophysiological findings in PLAN patients in an attempt to determine the contribution of each test to guide diagnosis. METHODS: Concomitant clinical, radiological, electroencephalographic (EEG) and electrodiagnostic testing (EDX) findings in a series of 8 consecutive genetically confirmed PLAN patients were collected. RESULTS: All patients presented marked motor axonal loss, with decreased or absent distal compound muscle action potentials, acute and chronic denervation at needle electromyography, in contrast with preservation of sensory conduction. EEG showed high-amplitude fast activity in all patients aged above 15 months. Two patients showing severe neonatal hypotonia displayed atypical hypsarhythmia and epileptic spasms. Iron deposition in globus pallidus was observed in only two patients aged above 6 years. CONCLUSIONS: Peripheral involvement is an early feature in PLAN recognizable by EDX at an earlier stage than typical iron accumulation in the brain. Furthermore, the association of West syndrome and axonal motor neuropathy may represent positive clues in favor of PLAN. This results emphasize the interest of early and repeated EDX.


Neuroaxonal Dystrophies/physiopathology , Child , Electroencephalography , Electromyography , Female , Group VI Phospholipases A2/genetics , Humans , Infant , Male , Mutation , Neuroaxonal Dystrophies/genetics
16.
Parkinsonism Relat Disord ; 49: 88-94, 2018 04.
Article En | MEDLINE | ID: mdl-29454663

INTRODUCTION: Phospholipase A2-associated neurodegeneration (PLAN) is an autosomal recessive movement disorder with abnormal iron deposition in basal ganglia, substantial nigra and adjacent areas, and cerebellar atrophy. It is caused by PLA2G6 mutations and comprises three phenotypes. We aimed to investigate genetic mutations in patients with predominantly extrapyramidal symptoms. METHODS: Eighteen Chinese patients with early onset of extrapyramidal symptoms were identified and underwent targeted next-generation sequencing, followed by Sanger sequencing. Detailed clinical and radiological features are presented. Prediction software was used to evaluate the pathogenicity of the identified variants. RESULTS: We identified 7 PLA2G6 variants including five known variants (c.668C > T, c.991G > T, c.1117G > A, c.1982C > T, and c.2218G > A) and two novel variants (c.1511C > T, and c.1915G > A) in four index cases. Among them, three cases had initial symptoms of difficulty walking or gait disturbance around the age of 30, and one case and his sibling developed mental handicap at age 7. Two cases exhibited a phenotype of "early parkinsonism" and the other two cases mimicked a phenotype of "hereditary spastic paraplegia (HSP)". Iron deposition in globus pallidus and substantia nigra was seen in three cases. Cerebellar atrophy was present in all four cases. CONCLUSIONS: Our study expands the mutation spectrum of the PLA2G6 gene and further supports the hypothesis that PLA2G6 mutations are associated with a continuous clinical spectrum from PLAN to HSP.


Group VI Phospholipases A2/genetics , Neuroaxonal Dystrophies/genetics , Neuroaxonal Dystrophies/physiopathology , Adult , China , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Parkinsonian Disorders/physiopathology , Phenotype , Sequence Analysis, DNA , Spastic Paraplegia, Hereditary/physiopathology
17.
Br J Ophthalmol ; 102(1): 102-108, 2018 01.
Article En | MEDLINE | ID: mdl-28487376

BACKGROUND/AIMS: Neurodegeneration with brain iron accumulation (NBIA) type I is a rare disease that can be divided into a classical or atypical variant, according to age of onset and clinical pattern. Neuro-ophthalmological involvement has been documented in the classical variant but only anecdotically in the atypical variant. We sought to describe the visual and ocular motor function in patients with atypical form of NBIA type I. METHODS: Cross-sectional study, including patients with genetically confirmed NBIA type I and classified as atypical variant, who underwent ophthalmological examination with best corrected visual acuity (BCVA), optical coherence tomography (OCT), fundus autofluorescence (FAF), electroretinography (ERG), visual evoked potentials (VEP) and video-oculography. RESULTS: Seven patients with a mean BCVA of 0.12±0.14 logMAR were included. Only two patients showed structural evidence of advanced retinopathy in OCT and FAF, and there were no cases of optic atrophy. ERG data, however, showed abnormal scotopic and/or photopic responses in all patients. VEP were normal in all three patients. Ocular fixation was markedly unstable (eg, increased rate of saccadic pulses) in the majority of patients (5). Additional mild ocular motor disturbances included low gain pursuit (2), hypermetric saccades (1), low gain optokinetic (2) and caloric and rotatory responses (3). CONCLUSION: Functional retinal changes associated with marked instability of ocular fixation should be included in the clinical spectrum of NBIA, particularly in the atypical form.


Evoked Potentials, Visual/physiology , Eye Movements/physiology , Iron Metabolism Disorders/complications , Neuroaxonal Dystrophies/complications , Ocular Motility Disorders/physiopathology , Retina/physiopathology , Retinal Diseases/physiopathology , Visual Acuity , Adult , Cross-Sectional Studies , Electroretinography , Female , Fluorescein Angiography , Fundus Oculi , Humans , Iron Metabolism Disorders/diagnosis , Iron Metabolism Disorders/physiopathology , Male , Middle Aged , Neuroaxonal Dystrophies/diagnosis , Neuroaxonal Dystrophies/physiopathology , Ocular Motility Disorders/diagnosis , Ocular Motility Disorders/etiology , Retina/diagnostic imaging , Retinal Diseases/diagnosis , Retinal Diseases/etiology , Tomography, Optical Coherence , Young Adult
18.
J Neurol Sci ; 381: 209-212, 2017 Oct 15.
Article En | MEDLINE | ID: mdl-28991683

Phospholipase A2-associated neurodegeneration (PLAN), a syndrome of Neurodegeneration with Brain Iron Accumulation (NBIA), is an autosomal recessive disorder caused by mutations in PLA2G6 gene. This gene encodes a calcium-independent group VI phospholipase A2 (iPLA-VI) critical in cell membrane homeostasis. PLAN syndrome encompasses a group of phenotypes with a different age of onset: classic infantile neuroaxonal dystrophy (INAD), atypical neuroaxonal dystrophy of childhood-onset (atypical NAD) and adult-onset PLA2G6-related dystonia-parkinsonism (PARK14). INAD is a severe progressive psychomotor disorder characterized by the presence of axonal spheroids throughout the central and peripheral nervous system. Here we report clinical, genetic and histopathological findings of an INAD consanguineous-family from Senegal. Sanger sequencing analysis revealed a new homozygous PLA2G6-mutation in the proband (c.1483C>T) and the co-segregation of the mutation in this family. Electron microscopy on skin biopsy showed degenerated axons confirming the phenotype. This study contributes to enrich the landscape of PLA2G6-associated INAD mutations and enforce the genotype-phenotype correlation.


Group VI Phospholipases A2/genetics , Mutation , Neuroaxonal Dystrophies/genetics , Child, Preschool , Consanguinity , Diagnosis, Differential , Family , Humans , Male , Neuroaxonal Dystrophies/pathology , Neuroaxonal Dystrophies/physiopathology , Phenotype , Skin/pathology
20.
Brain Dev ; 39(2): 93-100, 2017 Feb.
Article En | MEDLINE | ID: mdl-27884548

Infantile neuroaxonal dystrophy is a rare neurodegenerative disorder characterized by infantile onset of rapid motor and cognitive regression and hypotonia evolving into spasticity. Recessively inherited mutations of the PLA2G6 gene are causative of infantile neuroaxonal dystrophy and other PLA2G6-associated neurodegeneration, which includes conditions known as atypical neuroaxonal dystrophy, Karak syndrome and early-onset dystonia-parkinsonism with cognitive impairment. Phenotypic spectrum continues to evolve and genotype-phenotype correlations are currently limited. Due to the overlapping phenotypes and heterogeneity of clinical findings characterization of the syndrome is not always achievable. We reviewed the most recent clinical and neuroradiological information in the way to make easier differential diagnosis with other degenerative disorders in the paediatric age. Recognizing subtle signs and symptoms is a fascinating challenge to drive towards better diagnostic and genetic investigations.


Group VI Phospholipases A2/genetics , Neuroaxonal Dystrophies/diagnosis , Neuroaxonal Dystrophies/genetics , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/genetics , Adolescent , Brain/diagnostic imaging , Brain/physiopathology , Child , Child, Preschool , Early Diagnosis , Humans , Infant , Neuroaxonal Dystrophies/physiopathology , Neurodegenerative Diseases/physiopathology
...