Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Kidney Dis (Basel) ; 8(2): 148-159, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35527992

ABSTRACT

Aims: This study aimed to investigate associations between renal and extrarenal manifestations of mitochondrial diseases and their natural history as well as predictors of renal disease severity and overall disease outcome. The secondary aim was to generate a protocol of presymptomatic assessment and monitoring of renal function in patients with a defined mitochondrial disease. Methods: A multicenter, retrospective cohort study was performed by the Mitochondrial Clinical and Research Network (MCRN). Patients of any age with renal manifestations associated with a genetically verified mitochondrial disease were included from 8 expert European centers specializing in mitochondrial diseases: Gothenburg, Oulu, Copenhagen, Bergen, Helsinki, Stockholm, Rotterdam, and Barcelona. Results: Of the 36 patients included, two-thirds had mitochondrial DNA-associated disease. Renal manifestations were the first sign of mitochondrial disease in 19%, and renal involvement was first identified by laboratory tests in 57% of patients. Acute kidney injury occurred in 19% of patients and was the first sign of renal disease in the majority of these. The most common renal manifestation was chronic kidney disease (75% with stage 2 or greater), followed by tubulopathy (44.4%), the latter seen mostly among patients with single large-scale mitochondrial DNA deletions. Acute kidney injury and tubulopathy correlated with worse survival outcome. The most common findings on renal imaging were increased echogenicity and renal dysplasia/hypoplasia. Renal histology revealed focal segmental glomerulosclerosis, nephrocalcinosis, and nephronophthisis. Conclusion: Acute kidney injury is a distinct renal phenotype in patients with mitochondrial disease. Our results highlight the importance to recognize renal disease as a sign of an underlying mitochondrial disease. Acute kidney injury and tubulopathy are 2 distinct indicators of poor survival in patients with mitochondrial diseases.

2.
Mitochondrion ; 62: 187-204, 2022 01.
Article in English | MEDLINE | ID: mdl-34740866

ABSTRACT

Although mitochondrial dysfunction is the known cause of primary mitochondrial disease, mitochondrial dysfunction is often difficult to measure and prove, especially when biopsies of affected tissue are not available. In order to identify blood biomarkers of mitochondrial dysfunction, we reviewed studies that measured blood biomarkers in genetically, clinically or biochemically confirmed primary mitochondrial disease patients. In this way, we were certain that there was an underlying mitochondrial dysfunction which could validate the biomarker. We found biomarkers of three classes: 1) functional markers measured in blood cells, 2) biochemical markers of serum/plasma and 3) DNA markers. While none of the reviewed single biomarkers may perfectly reveal all underlying mitochondrial dysfunction, combining biomarkers that cover different aspects of mitochondrial impairment probably is a good strategy. This biomarker panel may assist in the diagnosis of primary mitochondrial disease patients. As mitochondrial dysfunction may also play a significant role in the pathophysiology of multifactorial disorders such as Alzheimer's disease and glaucoma, the panel may serve to assess mitochondrial dysfunction in complex multifactorial diseases as well and enable selection of patients who could benefit from therapies targeting mitochondria.


Subject(s)
Biomarkers/blood , Mitochondrial Diseases/blood , Mitochondrial Diseases/diagnosis , Humans , Mitochondrial Diseases/metabolism
3.
J Inherit Metab Dis ; 43(4): 726-736, 2020 07.
Article in English | MEDLINE | ID: mdl-32391929

ABSTRACT

BACKGROUND: Variants in POLG are one of the most common causes of inherited mitochondrial disease. Phenotypic classification of POLG disease has evolved haphazardly making it complicated and difficult to implement in everyday clinical practise. The aim of our study was to simplify the classification and facilitate better clinical recognition. METHODS: A multinational, retrospective study using data from 155 patients with POLG variants recruited from seven European countries. RESULTS: We describe the spectrum of clinical features associated with POLG variants in the largest known cohort of patients. While clinical features clearly form a continuum, stratifying patients simply according to age of onset-onset prior to age 12 years; onset between 12 and 40 years and onset after the age of 40 years, permitted us to identify clear phenotypic and prognostic differences. Prior to 12 years of age, liver involvement (87%), seizures (84%), and feeding difficulties (84%) were the major features. For those with onset between 12 and 40 years, ataxia (90%), peripheral neuropathy (84%), and seizures (71%) predominated, while for those with onset over 40 years, ptosis (95%), progressive external ophthalmoplegia (89%), and ataxia (58%) were the major clinical features. The earlier the onset the worse the prognosis. Patients with epilepsy and those with compound heterozygous variants carried significantly worse prognosis. CONCLUSION: Based on our data, we propose a simplified POLG disease classification, which can be used to guide diagnostic investigations and predict disease course.


Subject(s)
DNA Polymerase gamma/genetics , Genetic Predisposition to Disease/genetics , Mitochondrial Diseases/classification , Mitochondrial Diseases/genetics , Adolescent , Adult , Age of Onset , Aged , Aged, 80 and over , Child , Child, Preschool , Europe , Female , Humans , Infant , Male , Middle Aged , Mitochondrial Diseases/mortality , Mutation , Retrospective Studies , Survival Analysis , Young Adult
4.
Eur J Paediatr Neurol ; 23(5): 692-706, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31481303

ABSTRACT

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


Subject(s)
Algorithms , Decision Support Systems, Clinical , Spinocerebellar Degenerations/diagnosis , Adolescent , Child , Diagnosis, Differential , Female , Humans , Male
5.
Clin Genet ; 93(1): 111-118, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28671271

ABSTRACT

Mitochondrial respiratory chain complex I consists of 44 different subunits and contains 3 functional modules: the Q-, the N- and the P-module. NDUFA9 is a Q-module subunit required for complex I assembly or stability. However, its role in complex I biogenesis has not been studied in patient fibroblasts. So far, a single patient carrying an NDUFA9 variant with a severe neonatally fatal phenotype has been reported. Via exome sequencing, we identified a novel homozygous NDUFA9 missense variant in another patient with a milder phenotype including childhood-onset progressive generalized dystonia and axonal peripheral neuropathy. We performed complex I assembly analysis using primary skin fibroblasts of both patients. Reduced complex I abundance and an accumulation of Q-module subassemblies were present in both patients but more pronounced in the severe clinical phenotype patient. The latter displayed additional accumulation of P-module subassemblies, which was not present in the milder-phenotype patient. Lentiviral complementation of both patient fibroblast cell lines with wild-type NDUFA9 rescued complex I deficiency and the assembly defects. Our report further characterizes the phenotypic spectrum of NDUFA9 deficiency and demonstrates that the severity of the clinical phenotype correlates with the severity of the effects of the different NDUFA9 variants on complex I assembly.


Subject(s)
Electron Transport Complex I/genetics , Mitochondrial Proteins/genetics , Point Mutation , Cells, Cultured , Electron Transport Complex I/metabolism , Fatal Outcome , HEK293 Cells , Humans , Infant, Newborn , Male , Mitochondrial Proteins/metabolism , Exome Sequencing/methods
6.
Clin Genet ; 91(1): 121-125, 2017 01.
Article in English | MEDLINE | ID: mdl-26951855

ABSTRACT

The inherited optic neuropathies comprise a group of genetically heterogeneous disorders causing optic nerve dysfunction. In some cases, optic neuropathies are associated with cerebellar atrophy which mainly affects the vermis. Here, we describe a Moroccan girl of consanguineous parents with optic atrophy and cerebellar atrophy. Exome sequencing revealed a novel homozygous mutation (c.283+3G>T) in the donor splice site for exon 1 of SLC25A46. RNA analysis revealed that an alternative splice site within exon 1 was used leading to a premature termination codon within exon 2. SLC25A46 mRNA expression showed there is no wild-type transcript present in the patient and the mutant transcript does not undergo nonsense-mediated mRNA decay. Futhermore, we observed c.283+3G>T SLC25A46 mutation induces mitochondrial fragmentation. An additional 10 patients with optic atrophy and cerebellar atrophy, which were negative for mtDNA and OPA1 variants, were tested for pathogenic mutations in the SLC25A46 gene. However, no additional variants were identified. Our findings confirm the recent report of pathogenic SLC25A46 mutations as a novel cause for optic atrophy spectrum disorder.


Subject(s)
Genetic Predisposition to Disease/genetics , Mitochondrial Proteins/genetics , Mutation , Optic Atrophies, Hereditary/genetics , Phosphate Transport Proteins/genetics , RNA Splice Sites/genetics , Base Sequence , Consanguinity , Exome/genetics , Exons/genetics , Family Health , Female , Humans , Male , Parents , Pedigree , Sequence Analysis, DNA/methods
7.
Mutat Res Rev Mutat Res ; 764: 16-30, 2015.
Article in English | MEDLINE | ID: mdl-26041263

ABSTRACT

Several mutations in nuclear genes encoding for mitochondrial components have been associated with an increased cancer risk or are even causative, e.g. succinate dehydrogenase (SDHB, SDHC and SDHD genes) and iso-citrate dehydrogenase (IDH1 and IDH2 genes). Recently, studies have suggested an eminent role for mitochondrial DNA (mtDNA) mutations in the development of a wide variety of cancers. Various studies associated mtDNA abnormalities, including mutations, deletions, inversions and copy number alterations, with mitochondrial dysfunction. This might, explain the hampered cellular bioenergetics in many cancer cell types. Germline (e.g. m.10398A>G; m.6253T>C) and somatic mtDNA mutations as well as differences in mtDNA copy number seem to be associated with cancer risk. It seems that mtDNA can contribute as driver or as complementary gene mutation according to the multiple-hit model. This can enhance the mutagenic/clonogenic potential of the cell as observed for m.8993T>G or influences the metastatic potential in later stages of cancer progression. Alternatively, other mtDNA variations will be innocent passenger mutations in a tumor and therefore do not contribute to the tumorigenic or metastatic potential. In this review, we discuss how reported mtDNA variations interfere with cancer treatment and what implications this has on current successful pharmaceutical interventions. Mutations in MT-ND4 and mtDNA depletion have been reported to be involved in cisplatin resistance. Pharmaceutical impairment of OXPHOS by metformin can increase the efficiency of radiotherapy. To study mitochondrial dysfunction in cancer, different cellular models (like ρ(0) cells or cybrids), in vivo murine models (xenografts and specific mtDNA mouse models in combination with a spontaneous cancer mouse model) and small animal models (e.g. Danio rerio) could be potentially interesting to use. For future research, we foresee that unraveling mtDNA variations can contribute to personalized therapy for specific cancer types and improve the outcome of the disease.


Subject(s)
DNA, Mitochondrial/genetics , Neoplasms/genetics , Neoplasms/therapy , Animals , Drug Resistance, Neoplasm , Humans , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mutation , Precision Medicine , Radiation Tolerance
8.
J Neuromuscul Dis ; 1(1): 99-109, 2014.
Article in English | MEDLINE | ID: mdl-27858664

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) is a progressive muscle disease. No curative therapy is currently available, but in recent decades standards of care have improved. These improvements include the use of corticosteroids and mechanical ventilation. OBJECTIVE: To present a detailed population based report of the DMD disease course in The Netherlands (1980-2006) and evaluate the effect of changes in care by comparing it with an historical Dutch DMD cohort (1961-1974). METHODS: Information about DMD patients was gathered through the Dutch Dystrophinopathy Database using a standardized questionnaire and information from treating physicians. RESULTS: The study population involved 336 DMD patients (70% of the estimated prevalence), of whom 285 were still alive. Mean age at disease milestones was: diagnosis 4.3 years, wheelchair dependence 9.7 years, scoliosis surgery 14 years, cardiomyopathy (fractional shortening <27%) 15 years, mechanical ventilation 17 years and death 19 years. Within our cohort, corticosteroid use was associated with an increased age of wheelchair dependence from 9.8 to 11.6 years (p < 0.001). When comparing the recent cohort to the historical cohort, mean survival improved from 17 to 27 years (p < 0.001). CONCLUSION: The current study gives detailed information about the disease course of DMD patients, provides evidence for the positive effect of steroid treatment and mechanical ventilation and supports the use of patient registries as a valuable resource for evaluating improvements in care.

9.
Pediatr Diabetes ; 14(7): 519-25, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23710907

ABSTRACT

OBJECTIVE: The compound muscle action potential (CMAP) scan is a novel neurophysiological technique that appears more sensitive in detecting peripheral motor neuropathy than conventional methods. This study explores the value of the CMAP scan for the detection of subclinical diabetic peripheral motor neuropathy. METHODS: In this cross-sectional pilot study, CMAP scanning of the peroneal nerve was performed in (i) 13 well-controlled patients (8-25 yr old) with type 1 diabetes mellitus (T1DM) duration between 2.5 and 5 yr; (ii) 17 patients (10-25 yr old) with a duration of T1DM of at least 10 yr, poorly controlled and/or with microvascular complications and (iii) 13 adults with T1DM and established clinical diabetic peripheral neuropathy (DPN). Various CMAP scan variables, including measures of axonal excitability and axonal loss and reinnervation, were compared between patients and healthy controls. RESULTS: Axonal excitability was significantly decreased in the young patient groups as compared to their controls. The CMAP scan measures of axonal loss and reinnervation differed only between patients with clinical DPN and their controls. CONCLUSIONS: Motor nerve axonal excitability seems to be reduced early in T1DM, even in well-controlled young patients, and probably before (irreversible) axonal damage occurs. These changes can be measured by the CMAP scan, which makes this a promising tool for detecting nerve dysfunction in T1DM.


Subject(s)
Action Potentials , Diabetes Mellitus, Type 1/physiopathology , Neural Conduction/physiology , Peroneal Nerve/physiology , Adolescent , Adult , Axons/physiology , Child , Cross-Sectional Studies , Diabetic Angiopathies/physiopathology , Diabetic Neuropathies/physiopathology , Female , Humans , Male , Pilot Projects
10.
Neuropediatrics ; 43(6): 332-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23065766

ABSTRACT

AIM: Leukoencephalopathy with brainstem and spinal cord involvement and lactate elevation (LBSL) is known as a relatively mild leukoencephalopathy. We investigated the occurrence of severe variants of LBSL with extensive brain magnetic resonance imaging (MRI) abnormalities. METHOD: MRIs of approximately 3,000 patients with an unknown leukoencephalopathy were retrospectively reviewed for extensive signal abnormalities of the cerebral and cerebellar white matter, posterior limb of the internal capsule, cerebellar peduncles, pyramids, and medial lemniscus. Clinical data were retrospectively collected. RESULTS: Eleven patients fulfilled the MRI criteria (six males); six had DARS2 mutations. Clinical and laboratory findings did not distinguish between patients with and without DARS2 mutations, but MRI did. Patients with DARS2 mutations more often had involvement of structures typically affected in LBSL, including decussatio of the medial lemniscus, anterior spinocerebellar tracts, and superior and inferior cerebellar peduncles. Also, involvement of the globus pallidus was associated with DARS2 mutations. Earliest disease onset was neonatal; earliest death at 20 months. INTERPRETATION: This study confirms the occurrence of early infantile, severe LBSL, extending the known phenotypic range of LBSL. Abnormality of specific brainstem tracts and cerebellar peduncles are MRI findings that point to the correct diagnosis.


Subject(s)
Brain/pathology , Leukoencephalopathies/pathology , Mitochondrial Diseases/pathology , Nerve Fibers, Myelinated/pathology , Aspartate-tRNA Ligase/deficiency , Aspartate-tRNA Ligase/genetics , Child , Child, Preschool , Female , Humans , Infant , Leukoencephalopathies/genetics , Magnetic Resonance Imaging , Male , Mitochondrial Diseases/genetics , Mutation , Retrospective Studies , Severity of Illness Index , Spinal Cord/pathology
11.
J Clin Endocrinol Metab ; 97(9): 3261-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22802091

ABSTRACT

CONTEXT: Conflicting data exist on mitochondrial function and physical activity in type 2 diabetes mellitus (T2DM) development. OBJECTIVE: The aim was to assess mitochondrial function at different stages during T2DM development in combination with physical exercise in longstanding T2DM patients. DESIGN AND METHODS: We performed cross-sectional analysis of skeletal muscle from 12 prediabetic 11 longstanding T2DM male subjects and 12 male controls matched by age and body mass index. INTERVENTION: One-year intrasubject controlled supervised exercise training intervention was done in longstanding T2DM patients. MAIN OUTCOME MEASUREMENTS: Extensive ex vivo analyses of mitochondrial quality, quantity, and function were collected and combined with global gene expression analysis and in vivo ATP production capacity after 1 yr of training. RESULTS: Mitochondrial density, complex I activity, and the expression of Krebs cycle and oxidative phosphorylation system-related genes were lower in longstanding T2DM subjects but not in prediabetic subjects compared with controls. This indicated a reduced capacity to generate ATP in longstanding T2DM patients only. Gene expression analysis in prediabetic subjects suggested a switch from carbohydrate toward lipid as an energy source. One year of exercise training raised in vivo skeletal muscle ATP production capacity by 21 ± 2% with an increased trend in mitochondrial density and complex I activity. In addition, expression levels of ß-oxidation, Krebs cycle, and oxidative phosphorylation system-related genes were higher after exercise training. CONCLUSIONS: Mitochondrial dysfunction is apparent only in inactive longstanding T2DM patients, which suggests that mitochondrial function and insulin resistance do not depend on each other. Prolonged exercise training can, at least partly, reverse the mitochondrial impairments associated with the longstanding diabetic state.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Mitochondria, Muscle/physiology , Mitochondrial Myopathies/metabolism , Mitochondrial Myopathies/therapy , Motor Activity/physiology , Muscle, Skeletal/metabolism , Adenosine Triphosphate/biosynthesis , Aged , Blood Pressure/physiology , Body Composition/physiology , Body Mass Index , Citric Acid Cycle/genetics , Citric Acid Cycle/physiology , Diabetes Mellitus, Type 2/therapy , Disease Progression , Female , Gene Expression/physiology , Humans , Magnetic Resonance Spectroscopy , Male , Middle Aged , Mitochondria, Muscle/metabolism , Oxidative Phosphorylation , Physical Fitness/physiology , Prediabetic State/metabolism , Real-Time Polymerase Chain Reaction
12.
Biochim Biophys Acta ; 1817(11): 1971-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22796146

ABSTRACT

Oxidative phosphorylation disorders are often associated with increased oxidative stress and antioxidant therapy is frequently given as treatment. However, the role of oxidative stress in oxidative phosphorylation disorders or patients is far from clear and consequently the preventive or therapeutic effect of antioxidants is highly anecdotic. Therefore, we performed a systematic study of a panel of oxidative stress parameters (reactive oxygen species levels, damage and defense) in fibroblasts of twelve well-characterized oxidative phosphorylation patients with a defect in the POLG1 gene, in the mitochondrial DNA-encoded tRNA-Leu gene (m.3243A>G or m.3302A>G) and in one of the mitochondrial DNA-encoded NADH dehydrogenase complex I (CI) subunits. All except two cell lines (one POLG1 and one tRNA-Leu) showed increased reactive oxygen species levels compared with controls, but only four (two CI and two tRNA-Leu) cell lines provided evidence for increased oxidative protein damage. The absence of a correlation between reactive oxygen species levels and oxidative protein damage implies differences in damage prevention or correction. This was investigated by gene expression studies, which showed adaptive and compensating changes involving antioxidants and the unfolded protein response, especially in the POLG1 group. This study indicated that patients display individual responses and that detailed analysis of fibroblasts enables the identification of patients that potentially benefit from antioxidant therapy. Furthermore, the fibroblast model can also be used to search for and test novel, more specific antioxidants or explore ways to stimulate compensatory mechanisms.


Subject(s)
Antioxidants/therapeutic use , Fibroblasts/metabolism , Mitochondrial Diseases/drug therapy , Oxidative Phosphorylation , Oxidative Stress , Adolescent , Adult , Cell Line , Child , Child, Preschool , DNA Polymerase gamma , DNA, Mitochondrial/genetics , DNA-Directed DNA Polymerase/genetics , Female , Humans , Infant , Male , Mitochondrial Diseases/metabolism , Mutation , RNA, Transfer, Leu/genetics , Reactive Oxygen Species/metabolism
13.
Hum Reprod Update ; 18(4): 341-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22456975

ABSTRACT

BACKGROUND: Mitochondrial disorders are often fatal multisystem disorders, partially caused by heteroplasmic mitochondrial DNA (mtDNA) point mutations. Prenatal diagnosis is generally not possible for these maternally inherited mutations because of extensive variation in mutation load among embryos and the inability to accurately predict the clinical expression. The aim of this study is to investigate if PGD could be a better alternative, by investigating the existence of a minimal mutation level below which the chance of an embryo being affected is acceptably low, irrespective of the mtDNA mutation. METHODS: We performed a systematic review of muscle mutation levels, evaluating 159 different heteroplasmic mtDNA point mutations derived from 327 unrelated patients or pedigrees, and reviewed three overrepresented mtDNA mutations (m.3243A>G, m.8344A>G and m.8993T>C/G) separately. RESULTS: Mutation levels were included for familial mtDNA point mutations only, covering all affected (n = 195) and unaffected maternal relatives (n = 19) from 137 pedigrees. Mean muscle mutation levels were comparable between probands and affected maternal relatives, and between affected individuals with tRNA- versus protein-coding mutations. Using an estimated a priori prevalence of being affected in pedigrees of 0.477, we calculated that a 95% or higher chance of being unaffected was associated with a muscle mutation level of 18% or less. At a mutation level of 18%, the predicted probability of being affected is 0.00744. The chance of being unaffected was lower only for the m.3243A>G mutation (P < 0.001). Most carriers of mtDNA mutations will have oocytes with mutation levels below this threshold. CONCLUSIONS: Our data show, for the first time, that carriers of heteroplasmic mtDNA mutations will have a fair chance of having healthy offspring, by applying PGD. Nevertheless, our conclusions are partly based on estimations and, as indicated, do not provide absolute certainty. Carriers of mtDNA should be informed about these constraints.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/genetics , Preimplantation Diagnosis/statistics & numerical data , Heterozygote , Humans , Muscle, Skeletal , Pedigree , Point Mutation , RNA, Transfer/genetics
14.
J Inherit Metab Dis ; 35(3): 505-11, 2012 May.
Article in English | MEDLINE | ID: mdl-22008944

ABSTRACT

Classic infantile Pompe disease is an inherited generalized glycogen storage disorder caused by deficiency of lysosomal acid α-glucosidase. If left untreated, patients die before one year of age. Although enzyme-replacement therapy (ERT) has significantly prolonged lifespan, it has also revealed new aspects of the disease. For up to 11 years, we investigated the frequency and consequences of facial-muscle weakness, speech disorders and dysphagia in long-term survivors. Sequential photographs were used to determine the timing and severity of facial-muscle weakness. Using standardized articulation tests and fibreoptic endoscopic evaluation of swallowing, we investigated speech and swallowing function in a subset of patients. This study included 11 patients with classic infantile Pompe disease. Median age at the start of ERT was 2.4 months (range 0.1-8.3 months), and median age at the end of the study was 4.3 years (range 7.7 months -12.2 years). All patients developed facial-muscle weakness before the age of 15 months. Speech was studied in four patients. Articulation was disordered, with hypernasal resonance and reduced speech intelligibility in all four. Swallowing function was studied in six patients, the most important findings being ineffective swallowing with residues of food (5/6), penetration or aspiration (3/6), and reduced pharyngeal and/or laryngeal sensibility (2/6). We conclude that facial-muscle weakness, speech disorders and dysphagia are common in long-term survivors receiving ERT for classic infantile Pompe disease. To improve speech and reduce the risk for aspiration, early treatment by a speech therapist and regular swallowing assessments are recommended.


Subject(s)
Deglutition Disorders/pathology , Enzyme Therapy/methods , Facial Muscles/pathology , Glycogen Storage Disease Type II/pathology , Muscle Weakness/pathology , Speech Disorders/pathology , Child , Child, Preschool , Deglutition , Deglutition Disorders/diagnosis , Female , Glycogen Storage Disease Type II/diagnosis , Humans , Infant , Larynx/pathology , Male , Muscle Weakness/diagnosis , Pharynx/pathology , Speech , Speech Therapy/methods
15.
J Med Genet ; 49(1): 10-5, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22114105

ABSTRACT

BACKGROUND: Mitochondrial disorders are associated with abnormalities of the oxidative phosphorylation (OXPHOS) system and cause significant morbidity and mortality in the population. The extensive clinical and genetic heterogeneity of these disorders due to a broad variety of mutations in several hundreds of candidate genes, encoded by either the mitochondrial DNA (mtDNA) or nuclear DNA (nDNA), impedes a straightforward genetic diagnosis. A new disease gene is presented here, identified in a single Kurdish patient born from consanguineous parents with neonatally fatal Leigh syndrome and complex I deficiency. METHODS AND RESULTS: Using homozygosity mapping and subsequent positional candidate gene analysis, a total region of 255.8 Mb containing 136 possible mitochondrial genes was identified. A pathogenic mutation was found in the complex I subunit encoding the NDUFA9 gene, changing a highly conserved arginine at position 321 to proline. This is the first disease-causing mutation ever reported for NDUFA9. Complex I activity was restored in fibroblasts of the patient by lentiviral transduction with wild type but not mutant NDUFA9, confirming that the mutation causes the complex I deficiency and related disease. CONCLUSIONS: The data show that homozygosity mapping and candidate gene analysis remain an efficient way to detect mutations even in small consanguineous pedigrees with OXPHOS deficiency, especially when the enzyme deficiency in fibroblasts allows appropriate candidate gene selection and functional complementation.


Subject(s)
Electron Transport Complex I/genetics , Leigh Disease/diagnosis , Leigh Disease/genetics , Mutation, Missense , Amino Acid Sequence , Cells, Cultured , Consanguinity , DNA Mutational Analysis , Electron Transport Complex I/metabolism , Fatal Outcome , Genetic Association Studies , Homozygote , Humans , Infant, Newborn , Magnetic Resonance Imaging , Male , Molecular Sequence Data , Neuroimaging
16.
Eur J Med Genet ; 54(3): 299-300, 2011.
Article in English | MEDLINE | ID: mdl-21194575

ABSTRACT

Recently in this journal, Masurel-Paulet et al. reported the association between pulmonary disease and a mutation in X-linked FLNA in a male patient. We confirm this association in a female patient, showing that this complication is not sex-specific. Our patient has a FLNA missense mutation (c.220G > A) and presented with cerebral periventricular nodular heterotopia, cardiovascular abnormalities, and pulmonary disease consisting of lobar emphysema of the right middle pulmonary lobe with severe malacia of the right sided bronchus intermedius. Surgical resection of the right middle lobe was necessary and she had long-term oxygen dependency. Symptoms improved with age.


Subject(s)
Abnormalities, Multiple/genetics , Contractile Proteins/genetics , Lung Diseases/pathology , Microfilament Proteins/genetics , Mutation, Missense , Abnormalities, Multiple/pathology , Cardiovascular Abnormalities/pathology , Child, Preschool , Female , Filamins , Humans , Infant , Periventricular Nodular Heterotopia/pathology
17.
Mol Syndromol ; 1(3): 113-120, 2010 Sep.
Article in English | MEDLINE | ID: mdl-21031080

ABSTRACT

Partial monosomy 21 has been reported, but the phenotypes described are variable with location and size of the deletion. We present 2 patients with a partially overlapping microdeletion of 21q22 and a striking phenotypic resemblance. They both presented with severe psychomotor delay, behavioral problems, no speech, microcephaly, feeding problems with frequent regurgitation, idiopathic thrombocytopenia, obesity, deep set eyes, down turned corners of the mouth, dysplastic ears, and small chin. Brain MRI showed cerebral atrophy mostly evident in frontal and temporal lobes, widened ventricles and thin corpus callosum in both cases, and in one patient evidence of a migration disorder. The first patient also presented with epilepsy and a ventricular septum defect. The second patient had a unilateral Peters anomaly. Microarray analysis showed a partially overlapping microdeletion spanning about 2.5 Mb in the 21q22.1-q22.2 region including the DYRK1A gene and excluding RUNX1. These patients present with a recognizable phenotype specific for this 21q22.1-q22.2 locus. We searched the literature for patients with overlapping deletions including the DYRK1A gene, in order to define other genes responsible for this presentation.

18.
Seizure ; 19(7): 450-2, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20637656

ABSTRACT

We report a case of a girl who presented with typical absence seizures at age of 4.5 years. EEG showed absence seizures of sudden onset with 3 Hz spike-and-waves that also correlated with the clinical absences. The seizure semiology included subtle deviation of the eyes which prompted MRI investigation of the brain. This showed a periventricular nodular heterotopia in the mid to anterior horn of the right lateral ventricle. Although possibly coincidental, periventricular heterotopia are considered to be epileptogenic and this association has been reported once before. Migration disorders, such as in the periventricular heterotopia of our patient, may influence the formation and excitability of the striato-thalamo-cortical network involved in the generation of 3 Hz spike-waves.


Subject(s)
Epilepsy, Absence/etiology , Epilepsy, Absence/pathology , Periventricular Nodular Heterotopia/complications , Periventricular Nodular Heterotopia/pathology , Anticonvulsants/therapeutic use , Child, Preschool , Drug Therapy, Combination , Electroencephalography , Epilepsy, Absence/drug therapy , Female , Humans , Lamotrigine , Magnetic Resonance Imaging , Triazines/administration & dosage , Valproic Acid/administration & dosage
19.
J Med Genet ; 47(8): 507-12, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19542079

ABSTRACT

BACKGROUND: Leigh syndrome is an early onset, progressive, neurodegenerative disorder with developmental and motor skills regression. Characteristic magnetic resonance imaging abnormalities consist of focal bilateral lesions in the basal ganglia and/or the brainstem. The main cause is a deficiency in oxidative phosphorylation due to mutations in an mtDNA or nuclear oxidative phosphorylation gene. METHODS AND RESULTS: A consanguineous Moroccan family with Leigh syndrome comprise 11 children, three of which are affected. Marker analysis revealed a homozygous region of 11.5 Mb on chromosome 20, containing 111 genes. Eight possible mitochondrial candidate genes were sequenced. Patients were homozygous for an unclassified variant (p.P193L) in the cardiolipin synthase gene (CRLS1). As this variant was present in 20% of a Moroccan control population and enzyme activity was only reduced to 50%, this could not explain the rare clinical phenotype in our family. Patients were also homozygous for an amino acid substitution (p.L159F) in C20orf7, a new complex I assembly factor. Parents were heterozygous and unaffected sibs heterozygous or homozygous wild type. The mutation affects the predicted S-adenosylmethionine (SAM) dependent methyltransferase domain of C20orf7, possibly involved in methylation of NDUFB3 during the assembly process. Blue native gel electrophoresis showed an altered complex I assembly with only 30-40% of mature complex I present in patients and 70-90% in carriers. CONCLUSIONS: A new cause of Leigh syndrome can be a defect in early complex I assembly due to C20orf7 mutations.


Subject(s)
Electron Transport Complex I/metabolism , Leigh Disease/enzymology , Leigh Disease/genetics , Methyltransferases/genetics , Mitochondrial Proteins/genetics , Mutation/genetics , Adolescent , Adult , Amino Acid Sequence , Amino Acid Substitution/genetics , Base Sequence , Child, Preschool , DNA Mutational Analysis , Electron Transport Complex I/genetics , Family , Female , Homozygote , Humans , Leigh Disease/diagnostic imaging , Leigh Disease/metabolism , Leukocytes, Mononuclear/enzymology , Magnetic Resonance Imaging , Male , Methyltransferases/chemistry , Mitochondrial Proteins/chemistry , Molecular Sequence Data , Morocco , Pedigree , Tomography, X-Ray Computed , Young Adult
20.
Neurogenetics ; 10(4): 333-6, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19384555

ABSTRACT

We report a child with a severe choreadystonic movement disorder, bilateral periventricular nodular heterotopia (BPNH), and secondary microcephaly based on compound heterozygosity for two new ARFGEF2 mutations (c.2031_2038dup and c.3798_3802del), changing the limited knowledge about the phenotype. The brain MRI shows bilateral hyperintensity of the putamen, BPNH, and generalized atrophy. Loss of ARFGEF2 function affects vesicle trafficking, proliferation/apoptosis, and neurotransmitter receptor function. This can explain BPNH and microcephaly. We hypothesize that the movement disorder and the preferential damage to the basal ganglia, specifically to the putamen, may be caused by an increased sensitivity to degeneration, a dynamic dysfunction due to neurotransmitter receptor mislocalization or a combination of both.


Subject(s)
Cell Movement/physiology , Guanine Nucleotide Exchange Factors/genetics , Movement Disorders/genetics , Mutation , Neurons/physiology , Periventricular Nodular Heterotopia/genetics , Abnormalities, Multiple/genetics , Base Sequence , Brain/cytology , Brain/pathology , Child , DNA Mutational Analysis , Female , Humans , Male , Microcephaly/genetics , Microcephaly/pathology , Molecular Sequence Data , Movement Disorders/pathology , Movement Disorders/physiopathology , Periventricular Nodular Heterotopia/pathology , Periventricular Nodular Heterotopia/physiopathology , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...