Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Synchrotron Radiat ; 27(Pt 1): 185-198, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31868751

ABSTRACT

Friedreich's ataxia (FRDA) is a neurodegenerative disease characterized by an increase in intracytoplasmic iron concentration. Here the nanoscale iron distribution within single fibroblasts from FRDA patients was investigated using synchrotron-radiation-based nanoscopic X-ray fluorescence and X-ray in-line holography at the ID16A nano-imaging beamline of the ESRF. This unique probe was deployed to uncover the iron cellular two-dimensional architecture of freeze-dried FRDA fibroblasts. An unsurpassed absolute detection capability of 180 iron atoms within a 30 nm × 50 nm nanoscopic X-ray beam footprint was obtained using state-of-the-art X-ray focusing optics and a large-solid-angle detection system. Various micrometre-sized iron-rich organelles could be revealed for the first time, tentatively identified as endoplasmic reticulum, mitochondria and lysosomes. Also a multitude of nanoscopic iron hot-spots were observed in the cytosol, interpreted as chaperoned iron within the fibroblast's labile iron pool. These observations enable new hypotheses on the storage and trafficking of iron in the cell and ultimately to a better understanding of iron-storage diseases such as Friedreich's ataxia.


Subject(s)
Fibroblasts/chemistry , Friedreich Ataxia/pathology , Holography/methods , Iron/analysis , Single-Cell Analysis/methods , Spectrometry, X-Ray Emission/methods , Carbon , Cytoplasm/chemistry , Fibroblasts/ultrastructure , Freeze Drying , Humans , Nanostructures , Organelles/chemistry , Organelles/ultrastructure , Single-Cell Analysis/instrumentation , Synchrotrons , Tissue Fixation/methods
2.
Am J Hum Genet ; 99(1): 217-27, 2016 Jul 07.
Article in English | MEDLINE | ID: mdl-27374774

ABSTRACT

Complex I deficiency is the most common biochemical phenotype observed in individuals with mitochondrial disease. With 44 structural subunits and over 10 assembly factors, it is unsurprising that complex I deficiency is associated with clinical and genetic heterogeneity. Massively parallel sequencing (MPS) technologies including custom, targeted gene panels or unbiased whole-exome sequencing (WES) are hugely powerful in identifying the underlying genetic defect in a clinical diagnostic setting, yet many individuals remain without a genetic diagnosis. These individuals might harbor mutations in poorly understood or uncharacterized genes, and their diagnosis relies upon characterization of these orphan genes. Complexome profiling recently identified TMEM126B as a component of the mitochondrial complex I assembly complex alongside proteins ACAD9, ECSIT, NDUFAF1, and TIMMDC1. Here, we describe the clinical, biochemical, and molecular findings in six cases of mitochondrial disease from four unrelated families affected by biallelic (c.635G>T [p.Gly212Val] and/or c.401delA [p.Asn134Ilefs(∗)2]) TMEM126B variants. We provide functional evidence to support the pathogenicity of these TMEM126B variants, including evidence of founder effects for both variants, and establish defects within this gene as a cause of complex I deficiency in association with either pure myopathy in adulthood or, in one individual, a severe multisystem presentation (chronic renal failure and cardiomyopathy) in infancy. Functional experimentation including viral rescue and complexome profiling of subject cell lines has confirmed TMEM126B as the tenth complex I assembly factor associated with human disease and validates the importance of both genome-wide sequencing and proteomic approaches in characterizing disease-associated genes whose physiological roles have been previously undetermined.


Subject(s)
Alleles , Electron Transport Complex I/deficiency , Membrane Proteins/genetics , Mitochondrial Diseases/genetics , Mutation/genetics , Phenotype , Adolescent , Adult , Age of Onset , Amino Acid Sequence , Child , Electron Transport Complex I/genetics , Female , Humans , Infant , Male , Membrane Proteins/chemistry , Middle Aged , Pedigree , Young Adult
4.
Am J Hum Genet ; 95(6): 708-20, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25434004

ABSTRACT

Respiratory chain deficiencies exhibit a wide variety of clinical phenotypes resulting from defective mitochondrial energy production through oxidative phosphorylation. These defects can be caused by either mutations in the mtDNA or mutations in nuclear genes coding for mitochondrial proteins. The underlying pathomechanisms can affect numerous pathways involved in mitochondrial physiology. By whole-exome and candidate gene sequencing, we identified 11 individuals from 9 families carrying compound heterozygous or homozygous mutations in GTPBP3, encoding the mitochondrial GTP-binding protein 3. Affected individuals from eight out of nine families presented with combined respiratory chain complex deficiencies in skeletal muscle. Mutations in GTPBP3 are associated with a severe mitochondrial translation defect, consistent with the predicted function of the protein in catalyzing the formation of 5-taurinomethyluridine (τm(5)U) in the anticodon wobble position of five mitochondrial tRNAs. All case subjects presented with lactic acidosis and nine developed hypertrophic cardiomyopathy. In contrast to individuals with mutations in MTO1, the protein product of which is predicted to participate in the generation of the same modification, most individuals with GTPBP3 mutations developed neurological symptoms and MRI involvement of thalamus, putamen, and brainstem resembling Leigh syndrome. Our study of a mitochondrial translation disorder points toward the importance of posttranscriptional modification of mitochondrial tRNAs for proper mitochondrial function.


Subject(s)
Acidosis, Lactic/genetics , Brain Diseases/genetics , Cardiomyopathy, Hypertrophic/genetics , GTP-Binding Proteins/genetics , Protein Processing, Post-Translational , Acidosis, Lactic/physiopathology , Amino Acid Sequence , Brain/pathology , Brain Diseases/physiopathology , Cardiomyopathy, Hypertrophic/physiopathology , Cell Line , Child , Child, Preschool , Consanguinity , Female , Fibroblasts , GTP-Binding Proteins/metabolism , Humans , Infant , Infant, Newborn , Male , Molecular Sequence Data , Mutation , Pedigree , Protein Biosynthesis , RNA Interference , RNA, Transfer/genetics , RNA, Transfer/metabolism , Sequence Alignment
5.
Mol Genet Metab ; 122(4): 172-181, 2017 12.
Article in English | MEDLINE | ID: mdl-29126765

ABSTRACT

Mutations in FARS2 are known to cause dysfunction of mitochondrial translation due to deficient aminoacylation of the mitochondrial phenylalanine tRNA. Here, we report three novel mutations in FARS2 found in two patients in a compound heterozygous state. The missense mutation c.1082C>T (p.Pro361Leu) was detected in both patients. The mutations c.461C>T (p.Ala154Val) and c.521_523delTGG (p.Val174del) were each detected in one patient. We report abnormal in vitro aminoacylation assays as a functional validation of the molecular genetic findings. Based on the phenotypic data of previously reported subjects and the two subjects reported here, we conclude that FARS2 deficiency can be associated with two phenotypes: (i) an epileptic phenotype, and (ii) a spastic paraplegia phenotype.


Subject(s)
Epilepsy/genetics , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Phenotype , Phenylalanine-tRNA Ligase/deficiency , Phenylalanine-tRNA Ligase/genetics , Spastic Paraplegia, Hereditary/genetics , Adolescent , Amino Acyl-tRNA Synthetases/metabolism , Aminoacylation , Brain/diagnostic imaging , Cells, Cultured , Exome , Female , Fibroblasts/metabolism , Heterozygote , Humans , Infant , Magnetic Resonance Imaging , Male , Mitochondria/enzymology , Mitochondria/metabolism , Muscle, Skeletal/pathology , Mutation, Missense/genetics , Oxygen Consumption , RNA, Transfer/metabolism , Sequence Analysis, DNA
6.
Biochim Biophys Acta ; 1853(2): 285-98, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25450972

ABSTRACT

The tight interrelationship between peroxisomes and mitochondria is illustrated by their cooperation in lipid metabolism, antiviral innate immunity and shared use of proteins executing organellar fission. In addition, we previously reported that disruption of peroxisome biogenesis in hepatocytes severely impacts on mitochondrial integrity, primarily damaging the inner membrane. Here we investigated the molecular impairments of the dysfunctional mitochondria in hepatocyte selective Pex5 knockout mice. First, by using blue native electrophoresis and in-gel activity stainings we showed that the respiratory complexes were differentially affected with reduction of complexes I and III and incomplete assembly of complex V, whereas complexes II and IV were normally active. This resulted in impaired oxygen consumption in cultured Pex5(-/-) hepatocytes. Second, mitochondrial DNA was depleted causing an imbalance in the expression of mitochondrial- and nuclear-encoded subunits of the respiratory chain complexes. Third, mitochondrial membranes showed increased permeability and fluidity despite reduced content of the polyunsaturated fatty acid docosahexaenoic acid. Fourth, the affected mitochondria in peroxisome deficient hepatocytes displayed increased oxidative stress. Acute deletion of PEX5 in vivo using adeno-Cre virus phenocopied these effects, indicating that mitochondrial perturbations closely follow the loss of functional peroxisomes in time. Likely to compensate for the functional impairments, the volume of the mitochondrial compartment was increased several folds. This was not driven by PGC-1α but mediated by activation of PPARα, possibly through c-myc overexpression. In conclusion, loss of peroxisomal metabolism in hepatocytes perturbs the mitochondrial inner membrane, depletes mitochondrial DNA and causes mitochondrial biogenesis independent of PGC-1α.


Subject(s)
DNA, Mitochondrial/metabolism , Hepatocytes/metabolism , Mitochondria/metabolism , Peroxisomes/metabolism , Transcription Factors/metabolism , Animals , Cell Compartmentation , Cell Proliferation , Cell Respiration , Electron Transport , Gene Deletion , Hepatocytes/ultrastructure , Lipids/chemistry , Membrane Fluidity , Mice, Knockout , Mitochondria/ultrastructure , Oxidation-Reduction , Oxidative Phosphorylation , Oxidative Stress , PPAR alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Peroxisome-Targeting Signal 1 Receptor , Protein Subunits/metabolism , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/metabolism
7.
Nat Genet ; 39(4): 534-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17384640

ABSTRACT

Leukoencephalopathy with brain stem and spinal cord involvement and lactate elevation (LBSL) has recently been defined based on a highly characteristic constellation of abnormalities observed by magnetic resonance imaging and spectroscopy. LBSL is an autosomal recessive disease, most often manifesting in early childhood. Affected individuals develop slowly progressive cerebellar ataxia, spasticity and dorsal column dysfunction, sometimes with a mild cognitive deficit or decline. We performed linkage mapping with microsatellite markers in LBSL families and found a candidate region on chromosome 1, which we narrowed by means of shared haplotypes. Sequencing of genes in this candidate region uncovered mutations in DARS2, which encodes mitochondrial aspartyl-tRNA synthetase, in affected individuals from all 30 families. Enzyme activities of mutant proteins were decreased. We were surprised to find that activities of mitochondrial complexes from fibroblasts and lymphoblasts derived from affected individuals were normal, as determined by different assays.


Subject(s)
Aspartate-tRNA Ligase/genetics , Genetic Linkage , Lactic Acid/metabolism , Mitochondria/genetics , Spinocerebellar Degenerations/genetics , Aspartate-tRNA Ligase/metabolism , Genetic Markers , Haplotypes , Humans , Mitochondria/enzymology , Mitochondrial Diseases/genetics , Polymorphism, Genetic , Spinocerebellar Degenerations/metabolism
8.
Hum Mutat ; 36(2): 222-31, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25385316

ABSTRACT

A homozygous missense mutation (c.822G>C) was found in the gene encoding the mitochondrial asparaginyl-tRNA synthetase (NARS2) in two siblings born to consanguineous parents. These siblings presented with different phenotypes: one had mild intellectual disability and epilepsy in childhood, whereas the other had severe myopathy. Biochemical analysis of the oxidative phosphorylation (OXPHOS) complexes in both siblings revealed a combined complex I and IV deficiency in skeletal muscle. In-gel activity staining after blue native-polyacrylamide gel electrophoresis confirmed the decreased activity of complex I and IV, and, in addition, showed the presence of complex V subcomplexes. Considering the consanguineous descent, homozygosity mapping and whole-exome sequencing were combined revealing the presence of one single missense mutation in the shared homozygous region. The c.822G>C variant affects the 3' splice site of exon 7, leading to skipping of the whole exon 7 and a part of exon 8 in the NARS2 mRNA. In EBV-transformed lymphoblasts, a specific decrease in the amount of charged mt-tRNA(Asn) was demonstrated as compared with controls. This confirmed the pathogenic nature of the variant. To conclude, the reported variant in NARS2 results in a combined OXPHOS complex deficiency involving complex I and IV, making NARS2 a new member of disease-associated aaRS2.


Subject(s)
Aspartate-tRNA Ligase/genetics , Mutation, Missense , Adult , Aspartate-tRNA Ligase/metabolism , Base Sequence , Cells, Cultured , Consanguinity , DNA Mutational Analysis , Female , Genetic Association Studies , Homozygote , Humans , Male , Muscular Diseases/genetics , Protein Biosynthesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Splice Sites
9.
Hum Mol Genet ; 22(13): 2590-602, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23462291

ABSTRACT

Two siblings from consanguineous parents died perinatally with a condition characterized by generalized hypotonia, respiratory insufficiency, arthrogryposis, microcephaly, congenital brain malformations and hyperglycinemia. Catalytic activities of the mitochondrial respiratory complexes I and II were deficient in skeletal muscle, a finding suggestive of an inborn error in mitochondrial biogenesis. Homozygosity mapping identified IBA57 located in the largest homozygous region on chromosome 1 as a culprit candidate gene. IBA57 is known to be involved in the biosynthesis of mitochondrial [4Fe-4S] proteins. Sequence analysis of IBA57 revealed the homozygous mutation c.941A > C, p.Gln314Pro. Severely decreased amounts of IBA57 protein were observed in skeletal muscle and cultured skin fibroblasts from the affected subjects. HeLa cells depleted of IBA57 showed biochemical defects resembling the ones found in patient-derived cells, including a decrease in various mitochondrial [4Fe-4S] proteins and in proteins covalently linked to lipoic acid (LA), a cofactor produced by the [4Fe-4S] protein LA synthase. The defects could be complemented by wild-type IBA57 and partially by mutant IBA57. As a result of the mutation, IBA57 protein was excessively degraded, an effect ameliorated by protease inhibitors. Hence, we propose that the mutation leads to partial functional impairment of IBA57, yet the major pathogenic impact is due to its proteolytic degradation below physiologically critical levels. In conclusion, the ensuing lethal complex biochemical phenotype of a novel metabolic syndrome results from multiple Fe/S protein defects caused by a deficiency in the Fe/S cluster assembly protein IBA57.


Subject(s)
Brain Diseases/genetics , Carrier Proteins/genetics , Muscular Diseases/genetics , Mutation , Brain/pathology , Brain Diseases/diagnosis , Carrier Proteins/metabolism , Consanguinity , DNA Mutational Analysis , Electron Transport/genetics , Female , Fibroblasts/metabolism , Humans , Infant, Newborn , Magnetic Resonance Imaging , Male , Muscle, Skeletal/metabolism , Muscular Diseases/diagnosis , Pedigree , Phenotype , Siblings , Skin/metabolism
10.
Anesthesiology ; 122(2): 343-52, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25296107

ABSTRACT

BACKGROUND: Propofol is a short-acting intravenous anesthetic agent. In rare conditions, a life-threatening complication known as propofol infusion syndrome can occur. The pathophysiologic mechanism is still unknown. Some studies suggested that propofol acts as uncoupling agent, others suggested that it inhibits complex I or complex IV, or causes increased oxidation of cytochrome c and cytochrome aa3, or inhibits mitochondrial fatty acid metabolism. Although the exact site of interaction is not known, most hypotheses point to the direction of the mitochondria. METHODS: Eight rats were ventilated and sedated with propofol up to 20 h. Sequential biopsy specimens were taken from liver and skeletal muscle and used for determination of respiratory chain activities and propofol concentration. Activities were also measured in skeletal muscle from a patient who died of propofol infusion syndrome. RESULTS: In rats, authors detected a decrease in complex II+III activity starting at low tissue concentration of propofol (20 to 25 µM), further declining at higher concentrations. Before starting anesthesia, the complex II+III/citrate synthase activity ratio in liver was 0.46 (0.25) and in skeletal muscle 0.23 (0.05) (mean [SD]). After 20 h of anesthesia, the ratios declined to 0.17 (0.03) and 0.12 (0.02), respectively. When measured individually, the activities of complexes II and III remained normal. Skeletal muscle from one patient taken in the acute phase of propofol infusion syndrome also shows a selective decrease in complex II+III activity (z-score: -2.96). CONCLUSION: Propofol impedes the electron flow through the respiratory chain and coenzyme Q is the main site of interaction with propofol.


Subject(s)
Anesthetics, Intravenous/toxicity , Propofol/toxicity , Ubiquinone/metabolism , Animals , Citric Acid Cycle/drug effects , Electron Transport/drug effects , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Rats , Rats, Wistar , Respiration, Artificial , Syndrome
11.
J Inherit Metab Dis ; 38(6): 1147-53, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25971455

ABSTRACT

Leukodystrophies are a heterogeneous group of severe genetic neurodegenerative disorders. A multiple mitochondrial dysfunctions syndrome was found in an infant presenting with a progressive leukoencephalopathy. Homozygosity mapping, whole exome sequencing, and functional studies were used to define the underlying molecular defect. Respiratory chain studies in skeletal muscle isolated from the proband revealed a combined deficiency of complexes I and II. In addition, western blotting indicated lack of protein lipoylation. The combination of these findings was suggestive for a defect in the iron-sulfur (Fe/S) protein assembly pathway. SNP array identified loss of heterozygosity in large chromosomal regions, covering the NFU1 and BOLA3, and the IBA57 and ABCB10 candidate genes, in 2p15-p11.2 and 1q31.1-q42.13, respectively. A homozygous c.436C > T (p.Arg146Trp) variant was detected in IBA57 using whole exome sequencing. Complementation studies in a HeLa cell line depleted for IBA57 showed that the mutant protein with the semi-conservative amino acid exchange was unable to restore the biochemical phenotype indicating a loss-of-function mutation of IBA57. In conclusion, defects in the Fe/S protein assembly gene IBA57 can cause autosomal recessive neurodegeneration associated with progressive leukodystrophy and fatal outcome at young age. In the affected patient, the biochemical phenotype was characterized by a defect in the respiratory chain complexes I and II and a decrease in mitochondrial protein lipoylation, both resulting from impaired assembly of Fe/S clusters.


Subject(s)
Carrier Proteins/genetics , Iron-Sulfur Proteins/genetics , Leukoencephalopathies/diagnosis , Leukoencephalopathies/genetics , Mitochondrial Diseases/diagnosis , Electron Transport Complex I/genetics , Fatal Outcome , Heterozygote , Homozygote , Humans , Infant , Magnetic Resonance Imaging , Male , Mitochondria/genetics , Mutation , Phenotype
12.
J Inherit Metab Dis ; 38(3): 417-26, 2015 May.
Article in English | MEDLINE | ID: mdl-25326274

ABSTRACT

OBJECTIVES: TMEM70 deficiency is the most common nuclear-encoded defect affecting the ATP synthase. In this multicentre retrospective study we characterise the natural history of the disease, treatment and outcome in 48 patients with mutations in TMEM70. Eleven centers from eight European countries, Turkey and Israel participated. RESULTS: All 27 Roma and eight non-Roma patients were homozygous for the common mutation c.317-2A > G. Five patients were compound heterozygotes for the common mutation and mutations c.470 T > A, c.628A > C, c.118_119insGT or c.251delC. Six Arab Muslims and two Turkish patients were homozygous for mutations c.238C > T, c.316 + 1G > T, c.336 T > A, c.578_579delCA, c.535C > T, c.359delC. Age of onset was neonatal in 41 patients, infantile in six cases and two years in one child. The most frequent symptoms at onset were poor feeding, hypotonia, lethargy, respiratory and heart failure, accompanied by lactic acidosis, 3-methylglutaconic aciduria and hyperammonaemia. Symptoms further included: developmental delay (98%), hypotonia (95%), faltering growth (94%), short stature (89%), non-progressive cardiomyopathy (89%), microcephaly (71%), facial dysmorphism (66%), hypospadias (50% of the males), persistent pulmonary hypertension of the newborn (22%) and Wolff-Parkinson-White syndrome (13%). One or more acute metabolic crises occurred in 24 surviving children, frequently followed by developmental regression. Hyperammonaemic episodes responded well to infusion with glucose and lipid emulsion, and ammonia scavengers or haemodiafiltration. Ten-year survival was 63%, importantly for prognostication, no child died after the age of five years. CONCLUSION: TMEM70 deficiency is a panethnic, multisystemic disease with variable outcome depending mainly on adequate management of hyperammonaemic crises in the neonatal period and early childhood.


Subject(s)
Hyperammonemia/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Muscle, Skeletal/pathology , Acidosis, Lactic/genetics , Adolescent , Adult , Cardiomyopathies/genetics , Child , Child, Preschool , Disease Management , Europe , Female , Heterozygote , Homozygote , Humans , Infant , Infant, Newborn , Israel , Kaplan-Meier Estimate , Male , Metabolism, Inborn Errors/genetics , Mutation , Retrospective Studies , Turkey , Young Adult
13.
Eur J Pediatr ; 174(2): 267-70, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25009317

ABSTRACT

UNLABELLED: The propositus presented with hypotonia, respiratory failure, and seizures in the newborn period and was found to have severe hyperlactacidemia and a hypertrophic heart. He carried a de novo pathogenic mutation (m.8993 T>G) in the gene encoding subunit 6 of the mitochondrial ATP synthase (MTATP6). Although the lactate concentration in serum normalized and the proband recovered after a short period at the neonatal intensive care unit, his ultimate motor and cognitive development was poor. Brain MRI at the age of 6 months showed bilaterally signal abnormalities in the caudate nucleus, putamen, thalamus, and mesencephalon. He died at the age of 9 months. The difficulty in genetic counseling in families with a maternal mitochondrial mutation disorder is emphasized. CONCLUSION: Here, we report on a neonate with the m.8993 T>G mutation and emphasize implications of mtDNA disorders on family planning decisions.


Subject(s)
Acidosis, Lactic/genetics , DNA, Mitochondrial/genetics , Leigh Disease/genetics , Mitochondria/genetics , Mitochondrial Proton-Translocating ATPases/genetics , Point Mutation/genetics , Fatal Outcome , Genetic Counseling , Humans , Infant, Newborn , Magnetic Resonance Imaging , Male
14.
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
15.
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
16.
Pediatr Res ; 72(3): 232-40, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22728747

ABSTRACT

BACKGROUND: Protons are pumped from the mitochondrial matrix via oxidative phosphorylation (OXPHOS) into the intermembrane space, creating an electric membrane potential (ΔΨ) that is used for adenosine triphosphate (ATP) production. Defects in one or more of the OXPHOS complexes are associated with a variety of clinical symptoms, often making it difficult to pinpoint the causal mutation. METHODS: In this article, a microscopic method for the quantitative evaluation of ΔΨ in cultured skin fibroblasts is described. The method using 5,5',6,6'-tetraethylbenzimidazolyl-carbocyanine iodide (JC-1) fluorescence staining was tested in a selection of OXPHOS-deficient cell lines. RESULTS: A significant reduction of ΔΨ was found in the cell lines of patients with either an isolated defect in complex I, II, or IV or a combined defect (complex I + complex IV). ΔΨ was not reduced in the fibroblasts of two patients with severe complex V deficiency. Addition of the complex I inhibitor rotenone induced a significant reduction of ΔΨ and perinuclear relocalization of the mitochondria. In cells with a heteroplasmic mitochondrial DNA (mtDNA) defect, a more heterogeneous reduction of ΔΨ was detected. CONCLUSION: Our data show that imaging of ΔΨ in cultured skin fibroblasts is a useful method for the evaluation of OXPHOS functioning in cultured cell lines.


Subject(s)
Mitochondria/metabolism , Mitochondrial Diseases/diagnosis , Skin/metabolism , Cell Line , Fibroblasts/metabolism , Humans , Immunohistochemistry , Microscopy, Fluorescence , Skin/cytology
17.
Cells ; 11(3)2022 01 30.
Article in English | MEDLINE | ID: mdl-35159298

ABSTRACT

Human mitochondrial disease exhibits large variation of clinical phenotypes, even in patients with the same causative gene defect. We illustrate this heterogeneity by confronting clinical and biochemical data of two patients with the uncommon pathogenic homoplasmic NC_012920.1(MT-ATP6):m.9035T>C variant in MT-ATP6. Patient 1 presented as a toddler with severe motor and speech delay and spastic ataxia without extra-neurologic involvement. Patient 2 presented in adolescence with ataxia and ophthalmoplegia without cognitive or motor impairment. Respiratory chain complex activities were normal in cultured skin fibroblasts from both patients when calculated as ratios over citrate synthase activity. Native gels found presence of subcomplexes of complex V in fibroblast and/or skeletal muscle. Bioenergetic measurements in fibroblasts from both patients detected reduced spare respiratory capacities and altered extracellular acidification rates, revealing a switch from mitochondrial respiration to glycolysis to uphold ATP production. Thus, in contrast to the differing disease presentation, biochemical evidence of mitochondrial deficiency turned out quite similar. We conclude that biochemical analysis remains a valuable tool to confirm the genetic diagnosis of mitochondrial disease, especially in patients with new gene variants or atypical clinical presentation.


Subject(s)
Mitochondrial Diseases , Mitochondrial Proton-Translocating ATPases , Adolescent , Ataxia/genetics , Genotype , Humans , Infant , Mitochondrial Diseases/genetics , Mitochondrial Proton-Translocating ATPases/genetics , Mutation/genetics , Phenotype
18.
Oncogene ; 41(1): 15-25, 2022 01.
Article in English | MEDLINE | ID: mdl-34508176

ABSTRACT

Long non-coding RNAs (lncRNAs) can exhibit cell-type and cancer-type specific expression profiles, making them highly attractive as therapeutic targets. Pan-cancer RNA sequencing data revealed broad expression of the SAMMSON lncRNA in uveal melanoma (UM), the most common primary intraocular malignancy in adults. Currently, there are no effective treatments for UM patients with metastatic disease, resulting in a median survival time of 6-12 months. We aimed to investigate the therapeutic potential of SAMMSON inhibition in UM. Antisense oligonucleotide (ASO)-mediated SAMMSON inhibition impaired the growth and viability of a genetically diverse panel of uveal melanoma cell lines. These effects were accompanied by an induction of apoptosis and were recapitulated in two uveal melanoma patient derived xenograft (PDX) models through subcutaneous ASO delivery. SAMMSON pulldown revealed several candidate interaction partners, including various proteins involved in mitochondrial translation. Consequently, inhibition of SAMMSON impaired global, mitochondrial and cytosolic protein translation levels and mitochondrial function in uveal melanoma cells. The present study demonstrates that SAMMSON expression is essential for uveal melanoma cell survival. ASO-mediated silencing of SAMMSON may provide an effective treatment strategy to treat primary and metastatic uveal melanoma patients.


Subject(s)
Cell Survival/genetics , Melanoma/mortality , RNA, Long Noncoding/metabolism , Uveal Neoplasms/mortality , Animals , Cell Line, Tumor , Humans , Mice
19.
Genome Med ; 14(1): 38, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35379322

ABSTRACT

BACKGROUND: Lack of functional evidence hampers variant interpretation, leaving a large proportion of individuals with a suspected Mendelian disorder without genetic diagnosis after whole genome or whole exome sequencing (WES). Research studies advocate to further sequence transcriptomes to directly and systematically probe gene expression defects. However, collection of additional biopsies and establishment of lab workflows, analytical pipelines, and defined concepts in clinical interpretation of aberrant gene expression are still needed for adopting RNA sequencing (RNA-seq) in routine diagnostics. METHODS: We implemented an automated RNA-seq protocol and a computational workflow with which we analyzed skin fibroblasts of 303 individuals with a suspected mitochondrial disease that previously underwent WES. We also assessed through simulations how aberrant expression and mono-allelic expression tests depend on RNA-seq coverage. RESULTS: We detected on average 12,500 genes per sample including around 60% of all disease genes-a coverage substantially higher than with whole blood, supporting the use of skin biopsies. We prioritized genes demonstrating aberrant expression, aberrant splicing, or mono-allelic expression. The pipeline required less than 1 week from sample preparation to result reporting and provided a median of eight disease-associated genes per patient for inspection. A genetic diagnosis was established for 16% of the 205 WES-inconclusive cases. Detection of aberrant expression was a major contributor to diagnosis including instances of 50% reduction, which, together with mono-allelic expression, allowed for the diagnosis of dominant disorders caused by haploinsufficiency. Moreover, calling aberrant splicing and variants from RNA-seq data enabled detecting and validating splice-disrupting variants, of which the majority fell outside WES-covered regions. CONCLUSION: Together, these results show that streamlined experimental and computational processes can accelerate the implementation of RNA-seq in routine diagnostics.


Subject(s)
RNA , Transcriptome , Alleles , Humans , Sequence Analysis, RNA/methods , Exome Sequencing
20.
J Biol Chem ; 285(6): 4099-4109, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-19933271

ABSTRACT

Studies in yeast have shown that a deficiency in Atp12p prevents assembly of the extrinsic domain (F(1)) of complex V and renders cells unable to make ATP through oxidative phosphorylation. De Meirleir et al. (De Meirleir, L., Seneca, S., Lissens, W., De Clercq, I., Eyskens, F., Gerlo, E., Smet, J., and Van Coster, R. (2004) J. Med. Genet. 41, 120-124) have reported that a homozygous missense mutation in the gene for human Atp12p (HuAtp12p), which replaces Trp-94 with Arg, was linked to the death of a 14-month-old patient. We have investigated the impact of the pathogenic W94R mutation on Atp12p structure/function. Plasmid-borne wild type human Atp12p rescues the respiratory defect of a yeast ATP12 deletion mutant (Deltaatp12). The W94R mutation alters the protein at the most highly conserved position in the Pfam sequence and renders HuAtp12p insoluble in the background of Deltaatp12. In contrast, the yeast protein harboring the corresponding mutation, ScAtp12p(W103R), is soluble in the background of Deltaatp12 but not in the background of Deltaatp12Deltafmc1, a strain that also lacks Fmc1p. Fmc1p is a yeast mitochondrial protein not found in higher eukaryotes. Tryptophan 94 (human) or 103 (yeast) is located in a positively charged region of Atp12p, and hence its mutation to arginine does not alter significantly the electrostatic properties of the protein. Instead, we provide evidence that the primary effect of the substitution is on the dynamic properties of Atp12p.


Subject(s)
Chaperonins/genetics , Molecular Chaperones/genetics , Mutation , Proton-Translocating ATPases/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Amino Acid Substitution , Arginine/genetics , Arginine/metabolism , Blotting, Western , Cells, Cultured , Chaperonins/chemistry , Chaperonins/metabolism , Electron Transport/genetics , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Genetic Complementation Test , Humans , Microscopy, Electron , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proton-Translocating ATPases , Models, Molecular , Molecular Chaperones/metabolism , Protein Conformation , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/metabolism , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Solubility , Static Electricity , Tryptophan/genetics , Tryptophan/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL