Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Mol Genet Genomic Med ; 7(2): e00527, 2019 02.
Article in English | MEDLINE | ID: mdl-30548430

ABSTRACT

BACKGROUND: The diagnostic workup in patients with a clinical suspicion of lysosomal storage diseases (LSD) is often difficult due to the variability in the clinical phenotype. The gold standard for diagnosis of LSDs consists of enzymatic testing. However, due to the sequential nature of this methodology and inconsistent genotype-phenotype correlations of certain LSDs, finding a diagnosis can be challenging. METHOD: We developed and clinically implemented a gene panel covering 50 genes known to cause LSDs when mutated. Over a period of 18 months, we analyzed 150 patients who were referred for LSD testing and compared these results with the data of patients who were previously enrolled in a scheme of classical biochemical testing. RESULTS: Our panel was able to determine the molecular cause of the disease in 22 cases (15%), representing an increase in diagnostic yield compared to biochemical tests developed for 21 LSDs (4.6%). We were furthermore able to redirect the diagnosis of a mucolipidosis patient who was initially suspected to be affected with galactosialidosis. Several patients were identified as being affected with neuronal ceroid lipofuscinosis, which cannot readily be detected by enzyme testing. Finally, several carriers of pathogenic mutations in LSD genes related to the disease phenotype were identified as well, thus potentially increasing the diagnostic yield of the panel as heterozygous deletions cannot be detected. CONCLUSION: We show that the implementation of a gene panel for LSD diagnostics results in an increased yield in comparison to classical biochemical testing. As the panel is able to cover a wider range of diseases, we propose to implement this methodology as a first-tier test in cases of an aspecific LSD presentation, while enzymatic testing remains the first choice in patients with a more distinctive clinical presentation. Positive panel results should however still be enzymatically confirmed whenever possible.


Subject(s)
Genetic Testing/methods , Lysosomal Storage Diseases/genetics , Sequence Analysis, DNA/methods , Cells, Cultured , Fibroblasts/metabolism , Genetic Testing/standards , Humans , Immunoassay/methods , Lysosomal Storage Diseases/diagnosis , Molecular Diagnostic Techniques/methods , Sequence Analysis, DNA/standards
2.
J Med Genet ; 54(6): 432-440, 2017 06.
Article in English | MEDLINE | ID: mdl-28258187

ABSTRACT

BACKGROUND: Collagens are one of the major constituents of the pial membrane, which plays a crucial role in neuronal migration and cortical lamination during brain development. Type III procollagen, the chains of which are encoded by COL3A1, is the ligand of the G protein-coupled receptor 56 (GPR56), also known as adhesion G protein-coupled receptor G1. Bi-allelic mutations in GPR56 give rise to cobblestone-like malformation, white matter changes and cerebellar dysplasia. This report shows that bi-allelic mutations in COL3A1 are associated with a similar phenotype. METHODS: Exome analysis was performed in a family consisting of two affected and two non-affected siblings. Brain imaging studies of this family and of two previously reported individuals with bi-allelic mutations in COL3A1 were reviewed. Functional assays were performed on dermal fibroblasts. RESULTS: Exome analysis revealed a novel homozygous variant c.145C>G (p.Pro49Ala) in exon 2 of COL3A1. Brain MRI in the affected siblings as well as in the two previously reported individuals with bi-allelic COL3A1 mutations showed a brain phenotype similar to that associated with mutations in GPR56. CONCLUSION: Homozygous or compound heterozygous mutations in COL3A1 are associated with cobblestone-like malformation in all three families reported to date. The variability of the phenotype across patients suggests that genetic alterations in distinct domains of type III procollagen can lead to different outcomes. The presence of cobblestone-like malformation in patients with bi-allelic COL3A1 mutations emphasises the critical role of the type III collagen-GPR56 axis and the pial membrane in the regulation of brain development and cortical lamination.


Subject(s)
Collagen Type III/genetics , Cysts/genetics , Malformations of Cortical Development/genetics , Receptors, G-Protein-Coupled/genetics , White Matter/pathology , Adult , Alleles , Cells, Cultured , Cerebellar Diseases/genetics , Cerebellar Diseases/pathology , Child , Child, Preschool , Cysts/pathology , Exome/genetics , Exons/genetics , Female , Fibroblasts/pathology , Humans , Ligands , Magnetic Resonance Imaging/methods , Male , Malformations of Cortical Development/pathology , Mutation/genetics , Phenotype , Young Adult
3.
BMC Bioinformatics ; 17(1): 425, 2016 Oct 19.
Article in English | MEDLINE | ID: mdl-27760515

ABSTRACT

BACKGROUND: Predict whether a mutation is deleterious based on the custom 3D model of a protein. RESULTS: We have developed MODICT, a mutation prediction tool which is based on per residue RMSD (root mean square deviation) values of superimposed 3D protein models. Our mathematical algorithm was tested for 42 described mutations in multiple genes including renin (REN), beta-tubulin (TUBB2B), biotinidase (BTD), sphingomyelin phosphodiesterase-1 (SMPD1), phenylalanine hydroxylase (PAH) and medium chain Acyl-Coa dehydrogenase (ACADM). Moreover, MODICT scores corresponded to experimentally verified residual enzyme activities in mutated biotinidase, phenylalanine hydroxylase and medium chain Acyl-CoA dehydrogenase. Several commercially available prediction algorithms were tested and results were compared. The MODICT PERL package and the manual can be downloaded from https://github.com/IbrahimTanyalcin/MODICT . CONCLUSIONS: We show here that MODICT is capable tool for mutation effect prediction at the protein level, using superimposed 3D protein models instead of sequence based algorithms used by POLYPHEN and SIFT.


Subject(s)
Computational Biology/methods , Models, Molecular , Mutation/genetics , Proteins/chemistry , Proteins/genetics , Software , Acyl-CoA Dehydrogenase/genetics , Humans , Protein Conformation , Renin/genetics , Tubulin/genetics
4.
Biomed Res Int ; 2016: 6191307, 2016.
Article in English | MEDLINE | ID: mdl-26925412

ABSTRACT

Sertoli cell-only syndrome is defined by the complete absence of germ cells in testicular tissues and always results in male infertility. The aetiology often remains unknown. In this paper, we have investigated possible causes of Sertoli cell-only syndrome with a special focus on genetic causes. Our results show that, for a large part of the patients (>23% in an unselected group), the sex chromosomes are involved. The majority of patients had a Klinefelter syndrome, followed by patients with Yq microdeletions. Array comparative genomic hybridization in a selected group of "idiopathic patients" showed no known infertility related copy number variations.


Subject(s)
DNA Copy Number Variations/genetics , Infertility, Male/genetics , Klinefelter Syndrome/genetics , Sertoli Cell-Only Syndrome/genetics , Chromosome Deletion , Chromosomes, Human, Y/genetics , Comparative Genomic Hybridization/methods , Gene Expression Regulation , Humans , Infertility, Male/pathology , Klinefelter Syndrome/complications , Klinefelter Syndrome/pathology , Male , Sertoli Cell-Only Syndrome/etiology , Sertoli Cell-Only Syndrome/pathology , Sertoli Cells/pathology , Spermatogenesis/genetics , Testis/pathology
5.
Bioinformatics ; 32(3): 447-9, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26454277

ABSTRACT

SUMMARY: Today's genome browsers and protein databanks supply vast amounts of information about proteins. The challenge is to concisely bring together this information in an interactive and easy to generate format. AVAILABILITY AND IMPLEMENTATION: We have developed an interactive CIRCOS module called i-PV to visualize user supplied protein sequence, conservation and SNV data in a live presentable format. I-PV can be downloaded from http://www.i-pv.org. CONTACT: ibrahim.tanyalcin@i-pv.org, itanyalc@vub.ac.be or support@i-pv.org SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Amino Acids/chemistry , Computer Graphics , Proteins/metabolism , Sequence Analysis, Protein/methods , Software , Animals , Databases, Protein , Humans , Internet , Mice , Polymorphism, Genetic/genetics , Proteins/chemistry , Proteins/genetics , User-Computer Interface
6.
Eur J Hum Genet ; 24(3): 400-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26036855

ABSTRACT

SCN5A mutations involving the α-subunit of the cardiac voltage-gated muscle sodium channel (NaV1.5) result in different cardiac channelopathies with an autosomal-dominant inheritance such as Brugada syndrome. On the other hand, mutations in SCN4A encoding the α-subunit of the skeletal voltage-gated sodium channel (NaV1.4) cause non-dystrophic myotonia and/or periodic paralysis. In this study, we investigated whether cardiac arrhythmias or channelopathies such as Brugada syndrome can be part of the clinical phenotype associated with SCN4A variants and whether patients with Brugada syndrome present with non-dystrophic myotonia or periodic paralysis and related gene mutations. We therefore screened seven families with different SCN4A variants and non-dystrophic myotonia phenotypes for Brugada syndrome and performed a neurological, neurophysiological and genetic work-up in 107 Brugada families. In the families with an SCN4A-associated non-dystrophic myotonia, three patients had a clinical diagnosis of Brugada syndrome, whereas we found a remarkably high prevalence of myotonic features involving different genes in the families with Brugada syndrome. One Brugada family carried an SCN4A variant that is predicted to probably affect function, one family suffered from a not genetically confirmed non-dystrophic myotonia, one family was diagnosed with myotonic dystrophy (DMPK gene) and one family had a Thomsen disease myotonia congenita (CLCN1 variant that affects function). Our findings and data suggest a possible involvement of SCN4A variants in the pathophysiological mechanism underlying the development of a spontaneous or drug-induced type 1 electrocardiographic pattern and the occurrence of malignant arrhythmias in some patients with Brugada syndrome.


Subject(s)
Brugada Syndrome/genetics , Channelopathies/genetics , Genetic Predisposition to Disease , Muscle, Skeletal/pathology , Mutation/genetics , Myocardium/pathology , NAV1.4 Voltage-Gated Sodium Channel/genetics , Adult , Aged , Brugada Syndrome/diagnostic imaging , Electrocardiography , Electromyography , Female , Genetic Testing , Humans , Male , Middle Aged , Phenotype , Ultrasonography
7.
Thromb Res ; 135(6): 1179-85, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25837307

ABSTRACT

BACKGROUND: Hereditary antithrombin (AT) deficiency is a rare autosomal dominant disorder characterised by decreased AT activity in plasma and predisposition to recurrent venous thromboembolism (VTE). Thrombotic risk is thought to vary according to the subtype of deficiency, with Heparin Binding Site (HBS) deficiencies being the less thrombogenic. OBJECTIVES AND METHODS: The study population consisted of 82 genetically confirmed HBS deficient patients sharing six different mutations. Plasma samples of 35 of them, including one homozygous patient, were used for the evaluation of 4 commercial activity assays in their ability to diagnose HBS deficiency. We assessed mutation-specific prevalence of venous and arterial thrombosis and the contribution of additional thrombophilic risk factors. RESULTS AND CONCLUSIONS: Only one assay showed 100% sensitivity for all HBS mutations. The other ones failed mainly in the cases with p.Pro73Leu and p.Arg79His mutations. Shortening of incubation time resulted in an increase in sensitivity. In one patient, a novel HBS mutation, p.Asn77His, was identified, a quite exceptional and important finding given the restricted number of causal mutations reported so far in AT HBS deficiency. The overall prevalence of VTE in our study population (35%) was higher than previously reported (6-8%) in these patients. The presence of additional thrombophilic risk factors such as Factor V Leiden or prothrombin gene mutation G20210A contributed to a higher risk of VTE. Interestingly, the p.Pro73Leu and p.Arg79His mutations were associated with a high prevalence of arterial thrombosis. Our data suggest that AT HBS deficiencies are probably more prevalent and less benign than previously assumed.


Subject(s)
Antithrombin III Deficiency/genetics , Antithrombin III/genetics , Antithrombins/chemistry , Heparin/chemistry , Thrombophilia/genetics , Adolescent , Adult , Antithrombin III Deficiency/blood , Antithrombin III Deficiency/diagnosis , Antithrombins/blood , Arteries/physiopathology , Binding Sites , Child , Child, Preschool , Factor V/genetics , Female , Heterozygote , Homozygote , Humans , Infant , Infant, Newborn , Male , Middle Aged , Mutation , Phenotype , Prevalence , Reproducibility of Results , Risk , Risk Factors , Sensitivity and Specificity , Thrombophilia/blood , Thrombophilia/diagnosis , Thrombosis/physiopathology , Young Adult
8.
Eur J Hum Genet ; 23(1): 41-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24667782

ABSTRACT

Next-generation sequencing (NGS), an innovative sequencing technology that enables the successful analysis of numerous gene sequences in a massive parallel sequencing approach, has revolutionized the field of molecular biology. Although NGS was introduced in a rather recent past, the technology has already demonstrated its potential and effectiveness in many research projects, and is now on the verge of being introduced into the diagnostic setting of routine laboratories to delineate the molecular basis of genetic disease in undiagnosed patient samples. We tested a benchtop device on retrospective genomic DNA (gDNA) samples of controls and patients with a clinical suspicion of a mitochondrial DNA disorder. This Ion Torrent Personal Genome Machine platform is a high-throughput sequencer with a fast turnaround time and reasonable running costs. We challenged the chemistry and technology with the analysis and processing of a mutational spectrum composed of samples with single-nucleotide substitutions, indels (insertions and deletions) and large single or multiple deletions, occasionally in heteroplasmy. The output data were compared with previously obtained conventional dideoxy sequencing results and the mitochondrial revised Cambridge Reference Sequence (rCRS). We were able to identify the majority of all nucleotide alterations, but three false-negative results were also encountered in the data set. At the same time, the poor performance of the PGM instrument in regions associated with homopolymeric stretches generated many false-positive miscalls demanding additional manual curation of the data.


Subject(s)
Genome, Mitochondrial , Genomics , Genetic Testing/methods , Genetic Testing/standards , Genetic Variation , Genomics/methods , Genomics/standards , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Humans , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/genetics , Reproducibility of Results , Sensitivity and Specificity , Sequence Deletion
9.
PLoS One ; 9(11): e112950, 2014.
Article in English | MEDLINE | ID: mdl-25383547

ABSTRACT

The advent of massive parallel sequencing (MPS) has revolutionized the field of human molecular genetics, including the diagnostic study of mitochondrial (mt) DNA dysfunction. The analysis of the complete mitochondrial genome using MPS platforms is now common and will soon outrun conventional sequencing. However, the development of a robust and reliable protocol is rather challenging. A previous pilot study for the re-sequencing of human mtDNA revealed an uneven coverage, affecting predominantly part of the plus strand. In an attempt to address this problem, we undertook a comparative study of standard and modified protocols for the Ion Torrent PGM system. We could not improve strand representation by altering the recommended shearing methodology of the standard workflow or omitting the DNA polymerase amplification step from the library construction process. However, we were able to associate coverage bias of the plus strand with a specific sequence motif. Additionally, we compared coverage and variant calling across technologies. The same samples were also sequenced on a MiSeq device which showed that coverage and heteroplasmic variant calling were much improved.


Subject(s)
Genome, Mitochondrial , High-Throughput Nucleotide Sequencing/methods , Molecular Diagnostic Techniques/methods , Sequence Analysis, DNA/methods , DNA, Mitochondrial/genetics , Gene Library , Humans , Pilot Projects , Sensitivity and Specificity
10.
Ann Endocrinol (Paris) ; 75(2): 109-11, 2014 May.
Article in English | MEDLINE | ID: mdl-24768008

ABSTRACT

Male infertility, affecting around half of the couples with a problem to get pregnant, is a very heterogeneous condition. Part of patients are having a defect in spermatogenesis of which the underlying causes (including genetic ones) remain largely unknown. The only genetic tests routinely used in the diagnosis of male infertility are the analyses for the presence of Yq microdeletions and/or chromosomal abnormalities. Various other single gene or polygenic defects have been proposed to be involved in male fertility. Yet, their causative effect often remains to be proven. The recent evolution in the development of whole genome-based techniques may help in clarifying the role of genes and other genetic factors involved in spermatogenesis and spermatogenesis defects.


Subject(s)
Infertility, Male/genetics , Azoospermia/diagnosis , Azoospermia/genetics , Causality , Chromosome Aberrations , Chromosome Deletion , Chromosomes, Human, Y/genetics , Chromosomes, Human, Y/ultrastructure , Comparative Genomic Hybridization/methods , DNA Mutational Analysis/methods , Genetic Predisposition to Disease , Genetic Testing , Humans , Infertility, Male/diagnosis , Klinefelter Syndrome/diagnosis , Klinefelter Syndrome/genetics , Male , Polymorphism, Single Nucleotide , Sequence Deletion , Sex Chromosome Aberrations , Sex Chromosome Disorders of Sex Development/genetics , Spermatogenesis/genetics
11.
Phytother Res ; 28(2): 312-6, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23620374

ABSTRACT

Few therapeutic options are available to patients with oxidative phosphorylation disorders. Administering pharmacological agents that are able to stimulate mitochondrial biogenesis have been put forward as a possible treatment, yet the approach remains in need of thorough testing. We investigated the effect of resveratrol in an in vitro setting. Mitochondrial enzymatic activities were tested in cultured skin fibroblasts from patients harboring a nuclear defect in either complex II or complex IV (n = 11), and in fibroblasts from healthy controls (n = 11). In the latter, preincubation with resveratrol resulted in a significant increase of citrate synthase, complex II and complex IV enzyme activity. In patients with complex II or complex IV deficiency, however, activity of the deficient complex could not be substantially augmented, and response was dependent upon the residual activity. We conclude that resveratrol is not capable of normalizing oxidative phosphorylation activities in deficient cell lines.


Subject(s)
Cytochrome-c Oxidase Deficiency/enzymology , Electron Transport Complex II/deficiency , Fibroblasts/drug effects , Oxidative Phosphorylation/drug effects , Stilbenes/pharmacology , Cells, Cultured , Citrate (si)-Synthase/metabolism , Cytochrome-c Oxidase Deficiency/physiopathology , Electron Transport Complex II/metabolism , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Fibroblasts/enzymology , Humans , Mitochondria/drug effects , Mitochondria/enzymology , Resveratrol
12.
J Inherit Metab Dis ; 37(1): 53-62, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23774949

ABSTRACT

Primary coenzyme Q10 (CoQ10) deficiencies are associated with mutations in genes encoding enzymes important for its biosynthesis and patients are responsive to CoQ10 supplementation. Early treatment allows better prognosis of the disease and therefore, early diagnosis is desirable. The complex phenotype and genotype and the frequent secondary CoQ10 deficiencies make it difficult to achieve a definitive diagnosis by direct quantification of CoQ10. We developed a non-radioactive methodology for the quantification of CoQ10 biosynthesis in fibroblasts that allows the identification of primary deficiencies. Fibroblasts were incubated 72 h with 28 µmol/L (2)H3-mevalonate or 1.65 mmol/L (13)C6-p-hydroxybenzoate. The newly synthesized (2)H3- and (13)C6- labelled CoQ10 were analysed by high performance liquid chromatography-tandem mass spectrometry. The mean and the reference range for (13)C6-CoQ10 and (2)H3-CoQ10 biosynthesis were 0.97 (0.83-1.1) and 0.13 (0.09-0.17) nmol/Unit of citrate synthase, respectively. We validated the methodology through the study of one patient with COQ2 mutations and six patients with CoQ10 deficiency secondary to other inborn errors of metabolism. Afterwards we investigated 16 patients' fibroblasts and nine showed decreased CoQ10 biosynthesis. Therefore, the next step is to study the COQ genes in order to reach a definitive diagnosis in these nine patients. In the patients with normal rates the deficiency is probably secondary. In conclusion, we have developed a non-invasive non-radioactive method suitable for the detection of defects in CoQ10 biosynthesis, which offers a good tool for the stratification of patients with these treatable mitochondrial diseases.


Subject(s)
Ataxia/diagnosis , Ataxia/metabolism , Fibroblasts/metabolism , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/metabolism , Muscle Weakness/diagnosis , Muscle Weakness/metabolism , Mutation , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Cell Line , Chromatography, High Pressure Liquid , Citrate (si)-Synthase/metabolism , Genotype , Humans , Molecular Diagnostic Techniques , Phenotype , Reference Values , Reproducibility of Results , Skin/metabolism , Tandem Mass Spectrometry , Time Factors , Ubiquinone/biosynthesis , Ubiquinone/metabolism
13.
Mol Genet Metab Rep ; 1: 223-231, 2014.
Article in English | MEDLINE | ID: mdl-27896091

ABSTRACT

A 1-year-old girl born to consanguineous parents presented with unexplained liver failure, leading to transplantation at 19 months. Subsequent partial splenectomy for persistent cytopenia showed the presence of foamy cells, and Gaucher disease was confirmed by homozygosity for the p.Leu483Pro mutation in the GBA gene. She was treated by enzyme replacement therapy (ERT). Clinical follow-up showed mild developmental delay, strabismus, nystagmus and oculomotor apraxia. Biochemical studies revealed multiple respiratory chain deficiencies and a mosaic pattern of deficient complex IV immunostaining in liver and fibroblast. Molecular analysis identified a mtDNA depletion syndrome due to the homozygous p.Pro98Leu mutation in MPV17. A younger sister unaffected by mtDNA depletion, presented with pancytopenia and hepatosplenomegaly. ERT for Gaucher disease resulted in visceral normalization without any neurological symptom. A third sister, affected by both conditions, had marked developmental delay, strabismus and ophthalmoplegia but no liver cirrhosis. In conclusion, intrafamilal variability occurs in MPV17-related disease. The combined pathological effect of Gaucher and mitochondrial diseases can negatively impact neurological and liver functions and influence the outcome in consanguineous families. The immunocytochemical staining of OXPHOS protein in tissues and cultured cells is a powerful tool revealing mosaic pattern of deficiency pointing to mtDNA-related mitochondrial disorders.

14.
Eur J Paediatr Neurol ; 17(6): 625-30, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23816342

ABSTRACT

BACKGROUND: Primary coenzyme Q10 (CoQ10) deficiencies are heterogeneous autosomal recessive disorders. CoQ2 mutations have been identified only rarely in patients. All affected individuals presented with nephrotic syndrome in the first year of life. METHODS: An infant is studied with myoclonic seizures and hypertrophic cardiomyopathy in the first months of life and developed a nephrotic syndrome in a later stage. RESULTS: At three weeks of age, the index patient developed myoclonic seizures. In addition, he had hypertrophic cardiomyopathy and increased CSF lactate. A skeletal muscle biopsy performed at two months of age disclosed normal activities of the oxidative phosphorylation complexes. The child was supplemented with CoQ10 (5 mg/kg/day). At the age of four months, brain MR images showed bilateral increased signal intensities in putamen and cerebral cortex. After that age, he developed massive proteinuria. The daily dose of CoQ10 was increased to 30 mg/kg. Renal biopsy showed focal segmental glomerulosclerosis. Biochemical analyses of a kidney biopsy sample revealed a severely decreased activity of succinate cytochrome c reductase [complex II + III] suggesting ubiquinone depletion. Incorporation of labelled precursors necessary for CoQ10 synthesis was significantly decreased in cultured skin fibroblasts. His condition deteriorated and he died at the age of five months. A novel homozygous mutation c.326G > A (p.Ser109Asn) was found in COQ2. CONCLUSIONS: In contrast to previously reported patients with CoQ2 the proband presented with early myoclonic epilepsy, hypertrophic cardiomyopathy and only in a later stage developed a nephrotic syndrome. The phenotype of this patient enlarges the phenotypical spectrum of the multisystem infantile variant.


Subject(s)
Alkyl and Aryl Transferases/genetics , Ataxia/genetics , Cardiomyopathy, Hypertrophic/genetics , Epilepsies, Myoclonic/genetics , Mitochondrial Diseases/genetics , Muscle Weakness/genetics , Mutation/genetics , Nephrotic Syndrome/genetics , Ubiquinone/deficiency , Ataxia/complications , Ataxia/pathology , Cardiomyopathy, Hypertrophic/complications , Cardiomyopathy, Hypertrophic/pathology , Diffusion Magnetic Resonance Imaging , Electroencephalography , Epilepsies, Myoclonic/complications , Epilepsies, Myoclonic/pathology , Genetic Testing , Humans , Infant , Kidney/pathology , Kidney/ultrastructure , Magnetic Resonance Spectroscopy , Male , Microscopy, Electron, Transmission , Mitochondrial Diseases/complications , Mitochondrial Diseases/pathology , Muscle Weakness/complications , Muscle Weakness/pathology , Muscle, Skeletal/pathology , Nephrotic Syndrome/etiology , Nephrotic Syndrome/pathology , Ubiquinone/genetics
15.
Eur J Paediatr Neurol ; 17(6): 666-70, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23755938

ABSTRACT

BACKGROUND: The BIG2 protein, coded by ARFGEF2 indirectly assists neuronal proliferation and migration during cortical development. Mutations in ARFGEF2 have been reported as a rare cause of periventricular heterotopia. METHODS: The presence of periventricular heterotopia, acquired microcephaly and suspected recessive inheritance led to mutation analysis of ARFGEF2 in two affected siblings and their healthy consanguineous parents, after mutations in FLNA had been ruled out. RESULTS: A homozygous c.242_249delins7 (p.Pro81fs) mutation in exon 3 of ARFGEF2 was identified in the siblings. The alteration is a combination of 2 missense mutations (c.242C > A and c.247G > T) and a frameshift mutation (c.249delA) resulting in a premature stop codon. The clinical phenotype was characterized by dystonic quadriplegia, marked developmental delay, obstructive cardiomyopathy, recurrent infections and feeding difficulties. Degenerative features included early regression, acquired microcephaly and cerebral atrophy. Brain MRI revealed bilateral periventricular heterotopia, small corpus callosum, cerebral and hippocampal atrophy and hyperintensity in the putamen. CONCLUSION: Mutations in ARFGEF2 can be anticipated based on characteristic clinical and imaging features.


Subject(s)
Guanine Nucleotide Exchange Factors/genetics , Mutation, Missense/genetics , Periventricular Nodular Heterotopia/genetics , Animals , Brain/pathology , DNA Mutational Analysis , Family Health , Humans , Infant , Magnetic Resonance Imaging , Male , Phenotype
16.
JIMD Rep ; 10: 107-11, 2013.
Article in English | MEDLINE | ID: mdl-23430811

ABSTRACT

The nucleus-encoded mitochondrial pyruvate dehydrogenase enzyme complex plays key roles in cellular energy metabolism and acid-base equilibrium. Pyruvate dehydrogenase complex deficiency is due to loss-of-function mutation in one of the five component enzymes, most commonly E1α-subunit. The common clinical presentation ranges from fatal infantile lactic acidosis in newborns to chronic neurological dysfunction. We describe here an unusual presentation of E1α-subunit deficiency presenting as recurrent demyelination, Guillain-Barré syndrome-like demyelinating polyneuropathy at the onset, and ophthalmoplegia in a young infant. The clinical phenotype of the mutation in the patient was unique as compared to the previous reported cases of pyruvate dehydrogenase deficiency. The mother was found to be a mosaic carrier of the mutation. This phenotypic variability of pyruvate dehydrogenase complex deficiency and early suspicion of its unusual neurological manifestations is highlighted. Thiamine and ketogenic diet can be helpful.

17.
Hum Mutat ; 34(5): 785-91, 2013 May.
Article in English | MEDLINE | ID: mdl-23420552

ABSTRACT

Pathogenic Alu element insertions are rarely reported, whereas their occurrence is expected to be much higher. Alu containing alleles are usually out-competed during the PCR process and consequently undetectable with the classical screening methods. However, with the introduction of the next generation sequencing (NGS) technology in the diagnostic field, new opportunities are emerging. NGS data for a particular genomic region can be seen as the summation of all the individual sequences (reads) obtained for that region and no longer as the mean of this sum as it is the case for traditional Sanger sequencing. Because each single read covering that region is expected to be generated from a different template molecule, the presence of one single mutant read must theoretically be sufficient to identify the mutation. However, generation and identification of mutant reads bearing Alu insertions remains challenging and several wet/dry bench parameters need to be optimized. Hereby we present the proof of principle of a NGS-based mutation screening procedure allowing the detection of inherited Alu insertions within any predefined sequence by investigating 2 cases: c.1739_1740insAlu in BRCA1 and c.156_157insAlu in BRCA2.


Subject(s)
Genes, BRCA1 , Genes, BRCA2 , Sequence Analysis, DNA/methods , Base Sequence , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Homology, Amino Acid , Software
18.
Eur J Hum Genet ; 21(6): 695-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23188049

ABSTRACT

Multiple endocrine neoplasia type 1 (MEN1) is an autosomal-dominant cancer syndrome that is caused by a germline mutation in the MEN1 gene encoding a tumour-suppressor protein, menin. MEN1 causes a combination of endocrine tumours such as parathyroid adenomas, pituitary adenomas, glucagonomas, gastrinomas, insulinomas, adrenocortical adenomas and non-endocrine tumours. We here present a large MEN1 family where the carriers developed mild hyperparathyroidism, multiple well-differentiated functionally active neuroendocrine tumours of the pancreas and no pituitary tumour. The causal mutation is a new double substitution in the coding region of exon 2 in the MEN1 gene c.[428T>A; 429C>T], p.Leu143His. This new mutation in the MEN1 gene is clinically relevant leading to a limited penetrance and specific phenotype.


Subject(s)
Amino Acid Substitution/genetics , Multiple Endocrine Neoplasia Type 1/genetics , Mutation/genetics , Penetrance , Proto-Oncogene Proteins/genetics , Adult , Family , Female , Heterozygote , Humans , Male , Pedigree , Phenotype
19.
J Mol Diagn ; 14(6): 560-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22921311

ABSTRACT

This study evaluated a large set of blinded, previously analyzed prenatal DNA samples with a novel, CGG triplet-repeat primed (TP)-PCR assay (Amplidex FMR1 PCR Kit; Asuragen, Austin, TX). This cohort of 67 fetal DNAs contained 18 full mutations (270 to 1100 repeats, including 1 mosaic), 12 premutations (59 to 150 repeats), 9 intermediate mutations (54 to 58 repeats), and 28 normal samples (17 to 50 repeats, including 3 homozygous female samples). TP-PCR accurately identified FMR1 genotypes, ranging from normal to full- mutation alleles, with a 100% specificity (95% CI, 85.0% to 100%) and a 97.4% sensitivity (95% CI, 84.9% to 99.9%) in comparison with Southern blot analysis results. Exact sizing was possible for a spectrum of normal, intermediate, and premutation (up to 150 repeats) alleles, but CGG repeat numbers >200 are only identified as full mutations. All homozygous alleles were correctly resolved. The assay is also able to reproducibly detect a 2.5% premutation and a 3% full-mutation mosaicism in a normal male background, but a large premutation in a full male mutation background was masked when the amount of the latter was >5%. Implementation of this TP-PCR will significantly reduce reflex testing using Southern blot analyses. Additional testing with methylation-informative techniques might still be needed for a few cases with (large) premutations or full mutations.


Subject(s)
DNA , Fragile X Mental Retardation Protein/genetics , Genetic Testing/methods , Mutation , Polymerase Chain Reaction/methods , Prenatal Diagnosis/methods , Alleles , DNA/genetics , Female , Genetic Testing/economics , Genotype , Humans , Male , Mosaicism , Polymerase Chain Reaction/economics , Pregnancy , Prenatal Diagnosis/economics , Reproducibility of Results , Retrospective Studies , Sensitivity and Specificity , Time Factors , Trinucleotide Repeats
20.
Muscle Nerve ; 46(2): 246-56, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22806374

ABSTRACT

INTRODUCTION: Giant axonal neuropathy (GAN) is a progressive hereditary disease that affects the peripheral and central nervous systems. It is characterized morphologically by aggregates of intermediate filaments in different tissues. Mutations have been reported in the gene that codes for gigaxonin. Nevertheless, the underlying molecular mechanism remains obscure. METHODS: Cell lines from 4 GAN patients and 4 controls were analyzed by iTRAQ. RESULTS: Among the dysregulated proteins were ribosomal protein L29, ribosomal protein L37, galectin-1, glia-derived nexin, and aminopeptidase N. Also, nuclear proteins linked to formin-binding proteins were found to be dysregulated. Although the major role of gigaxonin is reported to be degradation of cytoskeleton-associated proteins, the amount of 76 structural cytoskeletal proteins was unaltered. CONCLUSIONS: Several of the dysregulated proteins play a role in cytoskeletal reorganization. Based on these findings, we speculate that disturbed cytoskeletal regulation is responsible for formation of aggregates of intermediate filaments.


Subject(s)
Axons/metabolism , Giant Axonal Neuropathy/metabolism , CD13 Antigens/metabolism , Female , Fibroblasts/metabolism , Galectin 1/metabolism , Humans , Male , Proteomics , Ribosomal Proteins/metabolism , Serpin E2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...