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

Country/Region as subject
Publication year range
1.
Am J Hum Genet ; 106(3): 412-421, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32142645

ABSTRACT

Primary familial brain calcification (PFBC) is a rare neurodegenerative disorder characterized by a combination of neurological, psychiatric, and cognitive decline associated with calcium deposition on brain imaging. To date, mutations in five genes have been linked to PFBC. However, more than 50% of individuals affected by PFBC have no molecular diagnosis. We report four unrelated families presenting with initial learning difficulties and seizures and later psychiatric symptoms, cerebellar ataxia, extrapyramidal signs, and extensive calcifications on brain imaging. Through a combination of homozygosity mapping and exome sequencing, we mapped this phenotype to chromosome 21q21.3 and identified bi-allelic variants in JAM2. JAM2 encodes for the junctional-adhesion-molecule-2, a key tight-junction protein in blood-brain-barrier permeability. We show that JAM2 variants lead to reduction of JAM2 mRNA expression and absence of JAM2 protein in patient's fibroblasts, consistent with a loss-of-function mechanism. We show that the human phenotype is replicated in the jam2 complete knockout mouse (jam2 KO). Furthermore, neuropathology of jam2 KO mouse showed prominent vacuolation in the cerebral cortex, thalamus, and cerebellum and particularly widespread vacuolation in the midbrain with reactive astrogliosis and neuronal density reduction. The regions of the human brain affected on neuroimaging are similar to the affected brain areas in the myorg PFBC null mouse. Along with JAM3 and OCLN, JAM2 is the third tight-junction gene in which bi-allelic variants are associated with brain calcification, suggesting that defective cell-to-cell adhesion and dysfunction of the movement of solutes through the paracellular spaces in the neurovascular unit is a key mechanism in CNS calcification.


Subject(s)
Age of Onset , Alleles , Brain Diseases/genetics , Calcinosis/genetics , Cell Adhesion Molecules/genetics , Genes, Recessive , Adolescent , Adult , Animals , Brain Diseases/diagnostic imaging , Calcinosis/diagnostic imaging , Child , Female , Humans , Male , Mice , Middle Aged , Pedigree
2.
Hum Mutat ; 41(4): 837-849, 2020 04.
Article in English | MEDLINE | ID: mdl-31898846

ABSTRACT

IFIH1 gain-of-function has been reported as a cause of a type I interferonopathy encompassing a spectrum of autoinflammatory phenotypes including Aicardi-Goutières syndrome and Singleton Merten syndrome. Ascertaining patients through a European and North American collaboration, we set out to describe the molecular, clinical and interferon status of a cohort of individuals with pathogenic heterozygous mutations in IFIH1. We identified 74 individuals from 51 families segregating a total of 27 likely pathogenic mutations in IFIH1. Ten adult individuals, 13.5% of all mutation carriers, were clinically asymptomatic (with seven of these aged over 50 years). All mutations were associated with enhanced type I interferon signaling, including six variants (22%) which were predicted as benign according to multiple in silico pathogenicity programs. The identified mutations cluster close to the ATP binding region of the protein. These data confirm variable expression and nonpenetrance as important characteristics of the IFIH1 genotype, a consistent association with enhanced type I interferon signaling, and a common mutational mechanism involving increased RNA binding affinity or decreased efficiency of ATP hydrolysis and filament disassembly rate.


Subject(s)
Gain of Function Mutation , Genetic Association Studies , Genotype , Interferon-Induced Helicase, IFIH1/genetics , Phenotype , Alleles , Autoimmune Diseases of the Nervous System/diagnosis , Autoimmune Diseases of the Nervous System/genetics , DNA Mutational Analysis , Female , Genetic Association Studies/methods , High-Throughput Nucleotide Sequencing , Humans , Interferon-Induced Helicase, IFIH1/chemistry , Male , Models, Molecular , Nervous System Malformations/diagnosis , Nervous System Malformations/genetics , Protein Conformation , Structure-Activity Relationship
3.
Clin Genet ; 98(2): 147-154, 2020 08.
Article in English | MEDLINE | ID: mdl-32385905

ABSTRACT

Variants in the FIG4 gene, which encodes a phosphatidylinositol-3,5-bisphosphatase lead to obstruction of endocytic trafficking, causing accumulation of enlarged vesicles in murine peripheral neurons and fibroblasts. Bi-allelic pathogenic variants in FIG4 are associated with neurological disorders including Charcot-Marie-Tooth disease type-4J (CMT4J) and Yunis-Varón syndrome (YVS). We present four probands from three unrelated families, all homozygous for a recurrent FIG4 missense variant c.506A>C p.(Tyr169Ser), with a novel phenotype involving features of both CMT4J and YVS. Three presented with infant-onset dystonia and one with hypotonia. All have depressed lower limb reflexes and distal muscle weakness, two have nerve conduction studies (NCS) consistent with severe sensorimotor demyelinating peripheral neuropathy and one had NCS showing patchy intermediate/mildly reduced motor conduction velocities. All have cognitive impairment and three have swallowing difficulties. MRI showed cerebellar atrophy and bilateral T2 hyperintense medullary swellings in all patients. These children represent a novel clinicoradiological phenotype and suggest that phenotypes associated with FIG4 missense variants do not neatly fall into previously described diagnoses but can present with variable features. Analysis of this gene should be considered in patients with central and peripheral neurological signs and medullary radiological changes, providing earlier diagnosis and informing reproductive choices.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Cleidocranial Dysplasia/genetics , Ectodermal Dysplasia/genetics , Flavoproteins/genetics , Genetic Predisposition to Disease , Limb Deformities, Congenital/genetics , Micrognathism/genetics , Phosphoric Monoester Hydrolases/genetics , Age of Onset , Charcot-Marie-Tooth Disease/complications , Charcot-Marie-Tooth Disease/pathology , Child , Child, Preschool , Cleidocranial Dysplasia/complications , Cleidocranial Dysplasia/pathology , Dystonia/complications , Dystonia/genetics , Dystonia/pathology , Ectodermal Dysplasia/complications , Ectodermal Dysplasia/pathology , Female , Genotype , Humans , Limb Deformities, Congenital/complications , Limb Deformities, Congenital/pathology , Male , Micrognathism/complications , Micrognathism/pathology , Muscle Hypotonia/complications , Muscle Hypotonia/genetics , Muscle Hypotonia/pathology , Mutation/genetics , Pedigree , Phenotype
4.
Neuropediatrics ; 51(4): 302-306, 2020 08.
Article in English | MEDLINE | ID: mdl-32464672

ABSTRACT

We report the clinical and molecular characterization of a novel biallelic mutation in the CSF1R gene leading to an autosomal recessive form of childhood onset leukoencephalopathy in a consanguineous family. The female child experienced acute encephalopathy at the age of 2 years, followed by spasticity and loss of all achieved milestones over 6 months. Her elder brother presented with encephalopathy at 4 years of age, with a subsequent loss of all achieved milestones over 8 months. Brain imaging in both children revealed multiple well-defined areas of calcification in the parietal and frontal regions and the occipital horns of both lateral ventricles. Clinical exome trio analysis showed homozygosity for a p.T833M mutation in CSF1R in the girl. Heterozygous family members, including both parents, were asymptomatic, with the eldest being 68 years of age. Total CSF1R protein expression levels were normal as compared with wild-type allele, but CSF1 ligand dependent autophosphorylation was consistent with a hypomorphic allele.


Subject(s)
Leukoencephalopathies , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Child, Preschool , Consanguinity , Fatal Outcome , Female , Humans , Leukoencephalopathies/genetics , Leukoencephalopathies/pathology , Leukoencephalopathies/physiopathology , Male , Pedigree
5.
Neuropediatrics ; 51(3): 178-184, 2020 06.
Article in English | MEDLINE | ID: mdl-31779033

ABSTRACT

BACKGROUND: A homozygous founder mutation in MTPAP/TENT6, encoding mitochondrial poly(A) polymerase (MTPAP), was first reported in six individuals of Old Order Amish descent demonstrating an early-onset, progressive spastic ataxia with optic atrophy and learning difficulties. MTPAP contributes to the regulation of mitochondrial gene expression through the polyadenylation of mitochondrially encoded mRNAs. Mitochondrial mRNAs with severely truncated poly(A) tails were observed in affected individuals, and mitochondrial protein expression was altered. OBJECTIVE: To determine the genetic basis of a perinatal encephalopathy associated with stereotyped neuroimaging and infantile death in three patients from two unrelated families. METHODS: Whole-exome sequencing was performed in two unrelated patients and the unaffected parents of one of these individuals. Variants and familial segregation were confirmed by Sanger sequencing. Polyadenylation of mitochondrial transcripts and de novo synthesis of mitochondrial proteins were assessed in patient's fibroblasts. RESULTS: Compound heterozygous p.Ile428Thr and p.Arg523Trp substitutions in MTPAP were recorded in two affected siblings from one family, and a homozygous p.Ile385Phe missense variant identified in a further affected child from a second sibship. Mitochondrial poly(A) tail analysis demonstrated shorter posttranscriptional additions to the mitochondrial transcripts, as well as an altered expression of mitochondrial proteins in the fibroblasts of the two siblings compared with healthy controls. CONCLUSION: Mutations in MTPAP likely cause an autosomal recessive perinatal encephalopathy with lethality in the first year of life.


Subject(s)
Brain Diseases/genetics , Brain Diseases/metabolism , DNA-Directed RNA Polymerases/genetics , Fibroblasts/metabolism , Mitochondrial Proteins/metabolism , Female , Humans , Infant , Infant Death , Male , Mitochondrial Proteins/genetics , Pedigree , Exome Sequencing
6.
Dev Med Child Neurol ; 62(1): 42-47, 2020 01.
Article in English | MEDLINE | ID: mdl-31175662

ABSTRACT

Comprehensive reviews of the clinical characteristics and pathogenesis of Aicardi-Goutières syndrome (AGS), particularly its contextualization within a putative type I interferonopathy framework, already exist. However, recent reports of attempts at treatment suggest that an assessment of the field from a therapeutic perspective is warranted at this time. Here, we briefly summarize the neurological phenotypes associated with mutations in the seven genes so far associated with AGS, rehearse current knowledge of the pathology as it relates to possible treatment approaches, critically appraise the potential utility of therapies, and discuss the challenges in assessing clinical efficacy. WHAT THIS PAPER ADDS: Progress in understanding AGS disease pathogenesis has led to the first attempts at targeted treatment. Further rational therapies are expected to become available in the short- to medium-term.


Subject(s)
Autoimmune Diseases of the Nervous System/therapy , Nervous System Malformations/therapy , Autoimmune Diseases of the Nervous System/etiology , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/immunology , Humans , Nervous System Malformations/etiology , Nervous System Malformations/genetics , Nervous System Malformations/immunology
7.
Hum Mutat ; 40(5): 619-630, 2019 05.
Article in English | MEDLINE | ID: mdl-30740813

ABSTRACT

The lipid phosphatase gene FIG4 is responsible for Yunis-Varón syndrome and Charcot-Marie-Tooth disease Type 4J, a peripheral neuropathy. We now describe four families with FIG4 variants and prominent abnormalities of central nervous system (CNS) white matter (leukoencephalopathy), with onset in early childhood, ranging from severe hypomyelination to mild undermyelination, in addition to peripheral neuropathy. Affected individuals inherited biallelic FIG4 variants from heterozygous parents. Cultured fibroblasts exhibit enlarged vacuoles characteristic of FIG4 dysfunction. Two unrelated families segregate the same G > A variant in the +1 position of intron 21 in the homozygous state in one family and compound heterozygous in the other. This mutation in the splice donor site of exon 21 results in read-through from exon 20 into intron 20 and truncation of the final 115 C-terminal amino acids of FIG4, with retention of partial function. The observed CNS white matter disorder in these families is consistent with the myelination defects in the FIG4 null mouse and the known role of FIG4 in oligodendrocyte maturation. The families described here the expanded clinical spectrum of FIG4 deficiency to include leukoencephalopathy.


Subject(s)
Alleles , Demyelinating Diseases/diagnosis , Demyelinating Diseases/genetics , Flavoproteins/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Mutation , Phosphoric Monoester Hydrolases/genetics , Child , Child, Preschool , DNA Mutational Analysis , Demyelinating Diseases/metabolism , Fibroblasts/metabolism , Genotype , Humans , Inheritance Patterns , Magnetic Resonance Imaging , Male , Neuroimaging , Pedigree , Phenotype
8.
Hum Mutat ; 39(8): 1076-1080, 2018 08.
Article in English | MEDLINE | ID: mdl-29782060

ABSTRACT

We describe progressive spastic paraparesis in two male siblings and the daughter of one of these individuals. Onset of disease occurred within the first decade, with stiffness and gait difficulties. Brisk deep tendon reflexes and extensor plantar responses were present, in the absence of intellectual disability or dermatological manifestations. Cerebral imaging identified intracranial calcification in all symptomatic family members. A marked upregulation of interferon-stimulated gene transcripts was recorded in all three affected individuals and in two clinically unaffected relatives. A heterozygous IFIH1 c.2544T>G missense variant (p.Asp848Glu) segregated with interferon status. Although not highly conserved (CADD score 10.08 vs. MSC-CADD score of 19.33) and predicted as benign by in silico algorithms, this variant is not present on publically available databases of control alleles, and expression of the D848E construct in HEK293T cells indicated that it confers a gain-of-function. This report illustrates, for the first time, the occurrence of autosomal-dominant spastic paraplegia with intracranial calcifications due to an IFIH1-related type 1 interferonopathy.


Subject(s)
Interferon-Induced Helicase, IFIH1/genetics , Paraparesis, Spastic/genetics , Algorithms , Brain Diseases/genetics , Calcinosis/genetics , Female , Gain of Function Mutation/genetics , HEK293 Cells , Heterozygote , Humans , Male , Mutation, Missense/genetics , Pedigree
9.
Neuropediatrics ; 49(2): 118-122, 2018 04.
Article in English | MEDLINE | ID: mdl-29253910

ABSTRACT

Alexander disease (AD) is a leukodystrophy caused by heterozygous mutations in the gene encoding the glial fibrillary acidic protein (GFAP). Currently, de novo heterozygous missense mutations in the GFAP gene are identified in over 95% of patients with AD. However, patients with biopsy-proven AD have been reported in whom no GFAP mutation has been identified. We report identical twin boys presenting in infancy with seizures and developmental delay in whom MR appearances were suggestive of AD with the exception of an unusual, bilateral, arc of calcification at the frontal white-gray junction. Initial mutation screening of the GFAP gene did not identify a mutation. Whole exome sequencing in both brothers revealed a de novo heterozygous in-frame deletion of the whole of exon 5 of the GFAP gene. Mutations in the GFAP gene are thought to result in a toxic effect of mutant GFAP disrupting the formation of the normal intermediate filament network and resulting in Rosenthal fiber formation, which has hitherto not been linked to exonic scale copy number variants in GFAP. Further studies on mutation negative AD patients are warranted to determine whether a similar mechanism underlies their disease.


Subject(s)
Alexander Disease/genetics , Exons/genetics , Gene Deletion , Glial Fibrillary Acidic Protein/genetics , Alexander Disease/diagnostic imaging , Brain/diagnostic imaging , Child , Child, Preschool , DNA Mutational Analysis , Follow-Up Studies , Humans , Magnetic Resonance Imaging , Male , Tomography Scanners, X-Ray Computed
10.
J Clin Immunol ; 36(3): 220-34, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26951490

ABSTRACT

PURPOSE: Spondyloenchondrodysplasia is a rare immuno-osseous dysplasia caused by biallelic mutations in ACP5. We aimed to provide a survey of the skeletal, neurological and immune manifestations of this disease in a cohort of molecularly confirmed cases. METHODS: We compiled clinical, genetic and serological data from a total of 26 patients from 18 pedigrees, all with biallelic ACP5 mutations. RESULTS: We observed a variability in skeletal, neurological and immune phenotypes, which was sometimes marked even between affected siblings. In total, 22 of 26 patients manifested autoimmune disease, most frequently autoimmune thrombocytopenia and systemic lupus erythematosus. Four patients were considered to demonstrate no clinical autoimmune disease, although two were positive for autoantibodies. In the majority of patients tested we detected upregulated expression of interferon-stimulated genes (ISGs), in keeping with the autoimmune phenotype and the likely immune-regulatory function of the deficient protein tartrate resistant acid phosphatase (TRAP). Two mutation positive patients did not demonstrate an upregulation of ISGs, including one patient with significant autoimmune disease controlled by immunosuppressive therapy. CONCLUSIONS: Our data expand the known phenotype of SPENCD. We propose that the OMIM differentiation between spondyloenchondrodysplasia and spondyloenchondrodysplasia with immune dysregulation is no longer appropriate, since the molecular evidence that we provide suggests that these phenotypes represent a continuum of the same disorder. In addition, the absence of an interferon signature following immunomodulatory treatments in a patient with significant autoimmune disease may indicate a therapeutic response important for the immune manifestations of spondyloenchondrodysplasia.


Subject(s)
Autoimmune Diseases/genetics , Intellectual Disability/genetics , Lupus Erythematosus, Systemic/genetics , Mutation , Osteochondrodysplasias/genetics , Purpura, Thrombocytopenic, Idiopathic/genetics , Tartrate-Resistant Acid Phosphatase/genetics , Adolescent , Adult , Alleles , Autoantibodies/biosynthesis , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Bone and Bones/immunology , Bone and Bones/pathology , Brain/immunology , Brain/pathology , Child , Child, Preschool , Female , Gene Expression , Genotype , Humans , Intellectual Disability/immunology , Intellectual Disability/pathology , Interferon Type I/genetics , Interferon Type I/immunology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Male , Osteochondrodysplasias/immunology , Osteochondrodysplasias/pathology , Pedigree , Phenotype , Purpura, Thrombocytopenic, Idiopathic/immunology , Purpura, Thrombocytopenic, Idiopathic/pathology , Tartrate-Resistant Acid Phosphatase/deficiency , Tartrate-Resistant Acid Phosphatase/immunology
11.
Neuropediatrics ; 47(6): 355-360, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27643693

ABSTRACT

The Aicardi-Goutières syndrome (AGS) was first described in 1984, and over the following years was defined by the clinical and radiological features of an early onset, severe, neurologic disorder with intracranial calcification, leukoencephalopathy, and cerebral atrophy, usually associated with a cerebrospinal fluid (CSF) pleocytosis and elevated CSF interferon α activity. It is now recognized that mutations in any of the following seven genes may result in the classical AGS phenotype: TREX1 (AGS1), RNASEH2A (AGS2), RNASEH2B (AGS3), RNASEH2C (AGS4), SAMHD1 (AGS5), ADAR1 (AGS6), and IFIH1 (AGS7). All of these genes encode proteins involved in nucleotide metabolism and/or sensing. Mutations in these genes result in the induction of type 1 interferon production and an upregulation of interferon stimulated genes. As more patients harboring mutations in these genes have been described, in particular facilitated by the advent of whole exome sequencing, a remarkably broad spectrum of associated neurologic phenotypes has been revealed, which we summarize here. We propose that the term AGS has continued clinical utility in the designation of a characteristic phenotype, which suggests relevant diagnostic investigations and can inform outcome predictions. However, we also suggest that the use of the term "type 1 interferonopathy" is appropriate for the wider spectrum of disease consequent upon dysfunction of these genes and proteins since it implies the possibility of a common "anti-interferon" approach to therapy as such treatments become available.


Subject(s)
Autoimmune Diseases of the Nervous System/genetics , Mutation/genetics , Nervous System Malformations/genetics , Adenosine Deaminase/genetics , Autoimmune Diseases of the Nervous System/cerebrospinal fluid , Autoimmune Diseases of the Nervous System/diagnostic imaging , Exodeoxyribonucleases/genetics , Genetic Association Studies , Humans , Interferon-Induced Helicase, IFIH1/genetics , Interferons/cerebrospinal fluid , Magnetic Resonance Imaging , Monomeric GTP-Binding Proteins/genetics , Nervous System Malformations/cerebrospinal fluid , Nervous System Malformations/diagnostic imaging , Phosphoproteins/genetics , RNA-Binding Proteins/genetics , Ribonuclease H/genetics , SAM Domain and HD Domain-Containing Protein 1
12.
Hum Mutat ; 36(5): 489-95, 2015 May.
Article in English | MEDLINE | ID: mdl-25726928

ABSTRACT

Primary familial brain calcification (PFBC) is a heterogeneous neuropsychiatric disorder, with affected individuals presenting a wide variety of motor and cognitive impairments, such as migraine, parkinsonism, psychosis, dementia, and mood swings. Calcifications are usually symmetrical, bilateral, and found predominantly in the basal ganglia, thalamus, and cerebellum. So far, variants in three genes have been linked to PFBC: SLC20A2, PDGFRB, and PDGFB. Variants in SLC20A2 are responsible for most cases identified so far and, therefore, the present review is a comprehensive worldwide summary of all reported variants to date. SLC20A2 encodes an inorganic phosphate transporter, PiT-2, widely expressed in various tissues, including brain, and is part of a major family of solute carrier membrane transporters. Fifty variants reported in 55 unrelated patients so far have been identified in families of diverse ethnicities and only few are recurrent. Various types of variants were detected (missense, nonsense, frameshift) including full or partial SLC20A2 deletions. The recently reported SLC20A2 knockout mouse will enhance our understanding of disease mechanism and allow for screening of therapeutic compounds. In the present review, we also discuss the implications of these recent exciting findings and consider the possibility of treatments based on manipulation of inorganic phosphate homeostasis.


Subject(s)
Brain Diseases/genetics , Brain Diseases/pathology , Calcinosis/genetics , Mutation , Sodium-Phosphate Cotransporter Proteins, Type III/genetics , Alleles , Amino Acid Substitution , Brain Diseases/diagnosis , DNA Mutational Analysis , Exons , Genetic Association Studies , Genetic Variation , Humans , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism
14.
Brain ; 137(Pt 5): 1350-60, 2014 May.
Article in English | MEDLINE | ID: mdl-24645144

ABSTRACT

The first described patients with pyridox(am)ine 5'-phosphate oxidase deficiency all had neonatal onset seizures that did not respond to treatment with pyridoxine but responded to treatment with pyridoxal 5'-phosphate. Our data suggest, however, that the clinical spectrum of pyridox(am)ine 5'-phosphate oxidase deficiency is much broader than has been reported in the literature. Sequencing of the PNPO gene was undertaken for a cohort of 82 individuals who had shown a reduction in frequency and severity of seizures in response to pyridoxine or pyridoxal 5'-phosphate. Novel sequence changes were studied using a new cell-free expression system and a mass spectrometry-based assay for pyridoxamine phosphate oxidase. Three groups of patients with PNPO mutations that had reduced enzyme activity were identified: (i) patients with neonatal onset seizures responding to pyridoxal 5'-phosphate (n = 6); (ii) a patient with infantile spasms (onset 5 months) responsive to pyridoxal 5'-phosphate (n = 1); and (iii) patients with seizures starting under 3 months of age responding to pyridoxine (n = 8). Data suggest that certain genotypes (R225H/C and D33V) are more likely to result in seizures that to respond to treatment with pyridoxine. Other mutations seem to be associated with infertility, miscarriage and prematurity. However, the situation is clearly complex with the same combination of mutations being seen in patients who responded and did not respond to pyridoxine. It is possible that pyridoxine responsiveness in PNPO deficiency is affected by prematurity and age at the time of the therapeutic trial. Other additional factors that are likely to influence treatment response and outcome include riboflavin status and how well the foetus has been supplied with vitamin B6 by the mother. For some patients there was a worsening of symptoms on changing from pyridoxine to pyridoxal 5'-phosphate. Many of the mutations in PNPO affected residues involved in binding flavin mononucleotide or pyridoxal 5'-phosphate and many of them showed residual enzyme activity. One sequence change (R116Q), predicted to affect flavin mononucleotide binding and binding of the two PNPO dimers, and with high residual activity was found in Groups (ii) and (iii). This sequence change has been reported in the 1000 Genomes project suggesting it could be a polymorphism but alternatively it could be a common mutation, perhaps responsible for the susceptibility locus for genetic generalized epilepsy on 17q21.32 (close to rs72823592). We believe the reduction in PNPO activity and B6-responsive epilepsy in the patients reported here indicates that it contributes to the pathogenesis of epilepsy.


Subject(s)
Environment , Epilepsy/genetics , Mutation/genetics , Pyridoxaminephosphate Oxidase/genetics , Anticonvulsants/therapeutic use , Child , Child, Preschool , Electroencephalography , Epilepsy/therapy , Female , HeLa Cells , Humans , Infant , Male , Mutagenesis, Site-Directed/methods , Pyridoxal Phosphate/therapeutic use , Pyridoxaminephosphate Oxidase/metabolism , Transfection , Young Adult
15.
J Med Genet ; 51(2): 76-82, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24262145

ABSTRACT

BACKGROUND: We recently observed mutations in ADAR1 to cause a phenotype of bilateral striatal necrosis (BSN) in a child with the type I interferonopathy Aicardi-Goutières syndrome (AGS). We therefore decided to screen patients with apparently non-syndromic BSN for ADAR1 mutations, and for an upregulation of interferon-stimulated genes (ISGs). METHODS: We performed Sanger sequencing of ADAR1 in a series of patients with BSN presenting to us during our routine clinical practice. We then undertook detailed clinical and neuroradiological phenotyping in nine mutation-positive children. We also measured the expression of ISGs in peripheral blood from these patients, and in children with BSN who did not have ADAR1 mutations. RESULTS: Nine ADAR1 mutation-positive patients from seven families demonstrated an acute (five cases) or subacute (four cases) onset of refractory, four-limb dystonia starting between 8 months and 5 years of age. Eight patients were developmentally normal at initial presentation. In seven cases, the disease was inherited as an autosomal recessive trait, while two related patients were found to have a heterozygous (dominant) ADAR1 mutation. All seven mutation-positive patients assayed showed an upregulation of ISGs (median: 12.50, IQR: 6.43-36.36) compared to controls (median: 0.93, IQR: 0.57-1.30), a so-called interferon signature, present many years after disease onset. No interferon signature was present in four children with BSN negative for mutations in ADAR1 (median: 0.63, IQR: 0.47-1.10). CONCLUSIONS: ADAR1-related disease should be considered in the differential diagnosis of apparently non-syndromic BSN with severe dystonia of varying evolution. The finding of an interferon signature provides a useful screening test for the presence of ADAR1 mutations in this context, and may suggest novel treatment approaches.


Subject(s)
Adenosine Deaminase/genetics , Interferon Type I/physiology , Striatonigral Degeneration/congenital , Case-Control Studies , Child, Preschool , DNA Mutational Analysis , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Infant , Male , Molecular Diagnostic Techniques , Mutation, Missense , RNA-Binding Proteins , Striatonigral Degeneration/enzymology , Striatonigral Degeneration/genetics
16.
Nat Genet ; 38(8): 917-20, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16845398

ABSTRACT

Aicardi-Goutières syndrome (AGS) presents as a severe neurological brain disease and is a genetic mimic of the sequelae of transplacentally acquired viral infection. Evidence exists for a perturbation of innate immunity as a primary pathogenic event in the disease phenotype. Here, we show that TREX1, encoding the major mammalian 3' --> 5' DNA exonuclease, is the AGS1 gene, and AGS-causing mutations result in abrogation of TREX1 enzyme activity. Similar loss of function in the Trex1(-/-) mouse leads to an inflammatory phenotype. Our findings suggest an unanticipated role for TREX1 in processing or clearing anomalous DNA structures, failure of which results in the triggering of an abnormal innate immune response.


Subject(s)
Exodeoxyribonucleases/genetics , Heredodegenerative Disorders, Nervous System/enzymology , Heredodegenerative Disorders, Nervous System/genetics , Mutation , Phosphoproteins/genetics , Proteins/genetics , Animals , Base Sequence , DNA/genetics , Exodeoxyribonucleases/deficiency , Heredodegenerative Disorders, Nervous System/immunology , Humans , Immunity, Innate , Mice , Mice, Knockout , Molecular Sequence Data , Phosphoproteins/deficiency , Syndrome
17.
Neuropediatrics ; 45(6): 386-93, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25243380

ABSTRACT

BACKGROUND: Hereditary spastic paraplegia is a neurodegenerative phenotype characterized by a progressive loss of corticospinal motor tract function. In a majority of affected individuals the pathogenesis remains undetermined. METHODS: We identified a series of patients with a phenotype of nonsyndromic spastic paraplegia in whom no diagnosis had been reached before exome sequencing. We measured the expression of interferon stimulated genes (ISGs) in peripheral blood from these patients. RESULTS: Five patients from four families with previously unexplained spastic paraplegia were identified with mutations in either ADAR1 (one patient), IFIH1 (one patient), or RNASEH2B (three patients from two families). All patients were developmentally normal before the onset of features beginning in the second year of life. All patients remain of normal intellect. Four patients demonstrated normal neuroimaging, while a single patient had features of nonspecific dysmyelination. The patients with ADAR1 and IFIH1-related disease showed a robust interferon signature. The patients with mutations in RNASEH2B demonstrated no (two patients) or a minimal (one patient) upregulation of ISGs compared with controls. CONCLUSIONS: Mutations in ADAR1, IFIH1, and RNASEH2B can cause a phenotype of spastic paraplegia with normal neuroimaging, or in association with nonspecific dysmyelination. Although the presence of an interferon signature can be helpful in interpreting the significance of gene variants in this context, patients with pathogenic mutations in RNASEH2B may demonstrate no upregulation of ISGs in peripheral blood. However, it remains possible that type I interferons act as a neurotoxin in the context of all genotypes.


Subject(s)
Adenosine Deaminase/genetics , DEAD-box RNA Helicases/genetics , Mutation , RNA-Binding Proteins/genetics , Ribonuclease H/genetics , Spastic Paraplegia, Hereditary/genetics , Adult , Child , Child, Preschool , Female , Humans , Interferon Type I/metabolism , Interferon-Induced Helicase, IFIH1 , Male , Siblings , Spastic Paraplegia, Hereditary/blood
18.
Neuropediatrics ; 45(3): 175-82, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24407470

ABSTRACT

OBJECTIVE: With the identification of mutations in the conserved telomere maintenance component 1 (CTC1) gene as the cause of Coats plus (CP) disease, it has become evident that leukoencephalopathy with calcifications and cysts (LCC) is a distinct genetic entity. PATIENTS AND METHODS: A total of 15 patients with LCC were identified from our database of patients with intracranial calcification. The clinical and radiological features are described. RESULTS: The median age (range) at presentation was 10 months (range, 2 days-54 years). Of the 15 patients, 9 presented with epileptic seizures, 5 with motor abnormalities, and 1 with developmental delay. Motor abnormalities developed in 14 patients and cognitive problems in 13 patients. Dense calcification occurred in the basal ganglia, thalami, dentate nucleus, brain stem, deep gyri, deep white matter, and in a pericystic distribution. Diffuse leukoencephalopathy was present in all patients, and it was usually symmetrical involving periventricular, deep, and sometimes subcortical, regions. Cysts developed in the basal ganglia, thalamus, deep white matter, cerebellum, or brain stem. In unaffected areas, normal myelination was present. No patient demonstrated cerebral atrophy. CONCLUSION: LCC shares the neuroradiological features of CP. However, LCC is a purely neurological disorder distinguished genetically by the absence of mutations in CTC1. The molecular cause(s) of LCC has (have) not yet been determined.


Subject(s)
Brain Diseases/diagnosis , Calcinosis/diagnosis , Cysts/diagnosis , Leukoencephalopathies/diagnosis , Nervous System Diseases/diagnosis , Adolescent , Adult , Brain Diseases/complications , Calcinosis/complications , Child , Child, Preschool , Cysts/complications , Humans , Infant , Infant, Newborn , Leukoencephalopathies/complications , Magnetic Resonance Imaging , Middle Aged , Tomography Scanners, X-Ray Computed , Young Adult
19.
Dev Med Child Neurol ; 56(7): 612-26, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24372060

ABSTRACT

Intracranial calcification (ICC) is a common finding on neuroimaging in paediatric neurology practice. In approximately half of all cases the calcification occurs in damaged, neoplastic, or malformed brain. For the large number of other disorders in which ICC occurs, no common pathogenetic mechanism can be suggested. Congenital infection, particularly with cytomegalovirus, accounts for a significant proportion of all cases. However, some genetic diseases, in particular Aicardi-Goutières syndrome, Band-like calcification, and RNASET2-related disease, may mimic congenital infection; therefore, a full consideration of the radiological and clinical features is necessary before concluding that congenital infection is the cause. In some disorders calcification is a universal finding, in others it is a frequent occurrence, and in some it is only an occasional finding. Characteristic patterns of calcification are seen in a number of conditions, and a systematic approach to the identification and description of radiological findings, taken together in the context of the clinical scenario, allows a diagnosis to be made in many cases. Nonetheless, there remain a number of presumed genetic disorders associated with ICC for which the underlying molecular cause has not yet been identified.


Subject(s)
Brain Diseases/etiology , Brain/abnormalities , Calcinosis/etiology , Phenotype , Brain Diseases/diagnostic imaging , Brain Diseases/genetics , Calcinosis/diagnostic imaging , Calcinosis/genetics , Child , Humans , Neuroimaging , Radiography
20.
Childs Nerv Syst ; 30(3): 375-85, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24452481

ABSTRACT

PURPOSE: Cerebellar mutism is a serious neurosurgical complication after posterior fossa surgery, but the cause, incidence and outcome remain incompletely defined. The aim of this paper was to identify and review all reports of this phenomenon to better delineate and improve the evidence base. METHODS: A systematic search and retrieval of databases was conducted using advanced search techniques. Review/outcomes criteria were developed, and study quality was determined. RESULTS: The retrieval identified 2,281 papers of which 96 were relevant, identifying 650 children with cerebellar mutism. Causative factors, clinical features and outcomes were reported variably; papers focussed on multiple areas, the majority reporting incidence in single or series of case studies with little or no analysis further than description. CONCLUSIONS: The complexity and variability of data reporting, likely contributing factors and outcomes make cerebellar mutism difficult to predict in incidence and the degree of impact that may ensue. A clear and accepted universal definition would help improve reporting, as would the application of agreed outcome measures. Clear and consistent reporting of surgical technique remains absent. Recommendations for practice are provided.


Subject(s)
Cerebellar Diseases/complications , Cerebellar Diseases/therapy , Mutism/etiology , Mutism/therapy , Adolescent , Cerebellar Diseases/psychology , Cerebellar Diseases/surgery , Child , Child, Preschool , Cognition/physiology , Data Interpretation, Statistical , Female , Humans , Infant , Infant, Newborn , Male , Mutism/psychology , Mutism/surgery , Neurosurgical Procedures , Publication Bias , Risk Factors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL