Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Am J Physiol Cell Physiol ; 321(1): C176-C186, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34106788

ABSTRACT

Maintaining mitochondrial function and dynamics is crucial for cellular health. In muscle, defects in mitochondria result in severe myopathies where accumulation of damaged mitochondria causes deterioration and dysfunction. Importantly, understanding the role of mitochondria in disease is a necessity to determine future therapeutics. One of the most common myopathies is mitochondrial encephalopathy lactic acidosis stroke-like episodes (MELAS), which has no current treatment. Recently, patients with MELAS treated with rapamycin exhibited improved clinical outcomes. However, the cellular mechanisms of rapamycin effects in patients with MELAS are currently unknown. In this study, we used cultured skin fibroblasts as a window into the mitochondrial dysfunction evident in MELAS cells, as well as to study the mechanisms of rapamycin action, compared with control, healthy individuals. We observed that mitochondria from patients were fragmented, had a threefold decline in the average speed of motility, a twofold reduced mitochondrial membrane potential, and a 1.5- to 2-fold decline in basal respiration. Despite the reduction in mitochondrial function, mitochondrial import protein Tim23 was elevated in patient cell lines. MELAS fibroblasts exhibited increased MnSOD levels and lysosomal function when compared with healthy controls. Treatment of MELAS fibroblasts with rapamycin for 24 h resulted in increased mitochondrial respiration compared with control cells, a higher lysosome content, and a greater localization of mitochondria to lysosomes. Our studies suggest that rapamycin has the potential to improve cellular health even in the presence of mtDNA defects, primarily via an increase in lysosomal content.


Subject(s)
Fibroblasts/drug effects , Lysosomes/drug effects , MELAS Syndrome/genetics , Mitochondria/drug effects , Sirolimus/pharmacology , Case-Control Studies , Child, Preschool , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Humans , Infant , Lysosomes/metabolism , MELAS Syndrome/drug therapy , MELAS Syndrome/metabolism , MELAS Syndrome/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Mutation , Oxidative Phosphorylation/drug effects , Primary Cell Culture , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism , Young Adult
2.
J Neurosci ; 40(23): 4576-4585, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32341096

ABSTRACT

An impediment to the development of effective therapies for neurodegenerative disease is that available animal models do not reproduce important clinical features such as adult-onset and stereotypical patterns of progression. Using in vivo magnetic resonance imaging and behavioral testing to study male and female decrepit mice, we found a stereotypical neuroanatomical pattern of progression of the lesion along the limbic system network and an associated memory impairment. Using structural variant analysis, we identified an intronic mutation in a mitochondrial-associated gene (Mrpl3) that is responsible for the decrepit phenotype. While the function of this gene is unknown, embryonic lethality in Mrpl3 knock-out mice suggests it is critical for early development. The observation that a mutation linked to energy metabolism precipitates a pattern of neurodegeneration via cell death across disparate but linked brain regions may explain how stereotyped patterns of neurodegeneration arise in humans or define a not yet identified human disease.SIGNIFICANCE STATEMENT The development of novel therapies for adult-onset neurodegenerative disease has been impeded by the limitations of available animal models in reproducing many of the clinical features. Here, we present a novel spontaneous mutation in a mitochondrial-associated gene in a mouse (termed decrepit) that results in adult-onset neurodegeneration with a stereotypical neuroanatomical pattern of progression and an associated memory impairment. The decrepit mouse model may represent a heretofore undiagnosed human disease and could serve as a new animal model to study neurodegenerative disease.


Subject(s)
Genetic Variation/genetics , Memory Disorders/diagnostic imaging , Memory Disorders/genetics , Mitochondrial Proteins/genetics , Neurodegenerative Diseases/diagnostic imaging , Neurodegenerative Diseases/genetics , Ribosomal Proteins/genetics , Age Factors , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
3.
Int J Mol Sci ; 18(8)2017 Jul 31.
Article in English | MEDLINE | ID: mdl-28758966

ABSTRACT

Creatine deficiency syndrome (CDS) comprises three separate enzyme deficiencies with overlapping clinical presentations: arginine:glycine amidinotransferase (GATM gene, glycine amidinotransferase), guanidinoacetate methyltransferase (GAMT gene), and creatine transporter deficiency (SLC6A8 gene, solute carrier family 6 member 8). CDS presents with developmental delays/regression, intellectual disability, speech and language impairment, autistic behaviour, epileptic seizures, treatment-refractory epilepsy, and extrapyramidal movement disorders; symptoms that are also evident in children with autism. The objective of the study was to test the hypothesis that genetic variability in creatine metabolism genes is associated with autism. We sequenced GATM, GAMT and SLC6A8 genes in 166 patients with autism (coding sequence, introns and adjacent untranslated regions). A total of 29, 16 and 25 variants were identified in each gene, respectively. Four variants were novel in GATM, and 5 in SLC6A8 (not present in the 1000 Genomes, Exome Sequencing Project (ESP) or Exome Aggregation Consortium (ExAC) databases). A single variant in each gene was identified as non-synonymous, and computationally predicted to be potentially damaging. Nine variants in GATM were shown to have a lower minor allele frequency (MAF) in the autism population than in the 1000 Genomes database, specifically in the East Asian population (Fisher's exact test). Two variants also had lower MAFs in the European population. In summary, there were no apparent associations of variants in GAMT and SLC6A8 genes with autism. The data implying there could be a lower association of some specific GATM gene variants with autism is an observation that would need to be corroborated in a larger group of autism patients, and with sub-populations of Asian ethnicities. Overall, our findings suggest that the genetic variability of creatine synthesis/transport is unlikely to play a part in the pathogenesis of autism spectrum disorder (ASD) in children.


Subject(s)
Amidinotransferases/genetics , Autism Spectrum Disorder/genetics , Creatinine/metabolism , Genetic Variation , Guanidinoacetate N-Methyltransferase/genetics , Nerve Tissue Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Amidinotransferases/metabolism , Autism Spectrum Disorder/metabolism , Child , Child, Preschool , Female , Guanidinoacetate N-Methyltransferase/metabolism , Humans , Male , Nerve Tissue Proteins/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/metabolism , Prospective Studies
4.
Mol Genet Metab Rep ; 12: 23-27, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28540186

ABSTRACT

Coenzyme Q10 (CoQ10) or ubiquinone is one of the two electron carriers in the mitochondrial respiratory chain which has an essential role in the process of oxidative phosphorylation. Defects in CoQ10 synthesis are usually associated with the impaired function of CoQ10-dependent complexes I, II and III. The recessively transmitted CoQ10 deficiency has been associated with a number of phenotypically and genetically heterogeneous groups of disorders manifesting at variable age of onset. The infantile, multisystemic presentation is usually caused by mutations in genes directly involved in CoQ10 biosynthesis. To date, mutations in COQ1 (PDSS1 and PDSS2), COQ2, COQ4, COQ6, COQ7, COQ8A/ADCK3, COQ8B/ADCK4, and COQ9 genes have been identified in patients with primary form of CoQ10 deficiency. Here we report novel mutations in the COQ4 gene, which were identified in an infant with profound mitochondrial disease presenting with perinatal seizures, hypertrophic cardiomyopathy and severe muscle CoQ10 deficiency.

5.
Mol Genet Metab ; 118(1): 21-7, 2016 May.
Article in English | MEDLINE | ID: mdl-26971250

ABSTRACT

We report a patient from a consanguineous family who presented with transient acute liver failure and biochemical patterns suggestive of disturbed urea cycle and mitochondrial function, for whom conventional genetic and metabolic investigations for acute liver failure failed to yield a diagnosis. Whole exome sequencing revealed a homozygous 12-bp deletion in PCK1 (MIM 614168) encoding cytosolic phosphoenolpyruvate carboxykinase (PEPCK); enzymatic studies subsequently confirmed its pathogenic nature. We propose that PEPCK deficiency should be considered in the young child with unexplained liver failure, especially where there are marked, accumulations of TCA cycle metabolites on urine organic acid analysis and/or an amino acid profile with hyperammonaemia suggestive of a proximal urea cycle defect during the acute episode. If suspected, intravenous administration of dextrose should be initiated. Long-term management comprising avoidance of fasting with the provision of a glucose polymer emergency regimen for illness management may be sufficient to prevent future episodes of liver failure. This case report provides further insights into the (patho-)physiology of energy metabolism, confirming the power of genomic analysis of unexplained biochemical phenotypes.


Subject(s)
Base Sequence , Carbohydrate Metabolism, Inborn Errors/diagnosis , Gastroenteritis/etiology , Intracellular Signaling Peptides and Proteins/genetics , Liver Diseases/diagnosis , Liver Failure, Acute/etiology , Phosphoenolpyruvate Carboxykinase (GTP)/deficiency , Sequence Deletion , Carbohydrate Metabolism, Inborn Errors/drug therapy , Carbohydrate Metabolism, Inborn Errors/genetics , Consanguinity , Exome , Gastroenteritis/genetics , Glucose/administration & dosage , Glucose/therapeutic use , High-Throughput Nucleotide Sequencing , Humans , Infant , Liver Diseases/drug therapy , Liver Diseases/genetics , Liver Failure, Acute/genetics , Male , Pedigree , Phosphoenolpyruvate Carboxykinase (GTP)/genetics
6.
Eur J Paediatr Neurol ; 19(5): 525-32, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26008862

ABSTRACT

BACKGROUND: Two siblings with hypertrophic cardiomyopathy and brain atrophy were diagnosed with Complex I deficiency based on low enzyme activity in muscle and high lactate/pyruvate ratio in fibroblasts. METHODS: Whole exome sequencing results of fibroblast gDNA from one sibling was narrowed down to 190 SNPs or In/Dels in 185 candidate genes by selecting non-synonymous coding sequence base pair changes that were not present in the SNP database. RESULTS: Two compound heterozygous mutations were identified in both siblings in NDUFV2, encoding the 24 kDa subunit of Complex I. The intronic mutation (c.IVS2 + 1delGTAA) is disease causing and has been reported before. The other mutation is novel (c.669_670insG, p.Ser224Valfs*3) and predicted to cause a pathogenic frameshift in the protein. Subsequent investigation of 10 probands with complex I deficiency from different families revealed homozygosity for the intronic c.IVS2 + 1delGTAA mutation in a second, consanguineous family. In this family three of five siblings were affected. Interestingly, they presented with Leigh syndrome but no cardiac involvement. The same genotype had been reported previously in a two families but presenting with hypertrophic cardiomyopathy, trunk hypotonia and encephalopathy. CONCLUSION: We have identified NDUFV2 mutations in two families with Complex I deficiency, including a novel mutation. The diagnosis of Leigh syndrome expands the clinical phenotypes associated with the c.IVS2 + 1delGTAA mutation in this gene.


Subject(s)
Exome , Leigh Disease/genetics , Mutation , NADH Dehydrogenase/genetics , Exome/genetics , Female , Humans , Pedigree , Phenotype , Siblings
7.
Brain ; 137(Pt 2): 366-79, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24334290

ABSTRACT

Patients with nonketotic hyperglycinemia and deficient glycine cleavage enzyme activity, but without mutations in AMT, GLDC or GCSH, the genes encoding its constituent proteins, constitute a clinical group which we call 'variant nonketotic hyperglycinemia'. We hypothesize that in some patients the aetiology involves genetic mutations that result in a deficiency of the cofactor lipoate, and sequenced genes involved in lipoate synthesis and iron-sulphur cluster biogenesis. Of 11 individuals identified with variant nonketotic hyperglycinemia, we were able to determine the genetic aetiology in eight patients and delineate the clinical and biochemical phenotypes. Mutations were identified in the genes for lipoate synthase (LIAS), BolA type 3 (BOLA3), and a novel gene glutaredoxin 5 (GLRX5). Patients with GLRX5-associated variant nonketotic hyperglycinemia had normal development with childhood-onset spastic paraplegia, spinal lesion, and optic atrophy. Clinical features of BOLA3-associated variant nonketotic hyperglycinemia include severe neurodegeneration after a period of normal development. Additional features include leukodystrophy, cardiomyopathy and optic atrophy. Patients with lipoate synthase-deficient variant nonketotic hyperglycinemia varied in severity from mild static encephalopathy to Leigh disease and cortical involvement. All patients had high serum and borderline elevated cerebrospinal fluid glycine and cerebrospinal fluid:plasma glycine ratio, and deficient glycine cleavage enzyme activity. They had low pyruvate dehydrogenase enzyme activity but most did not have lactic acidosis. Patients were deficient in lipoylation of mitochondrial proteins. There were minimal and inconsistent changes in cellular iron handling, and respiratory chain activity was unaffected. Identified mutations were phylogenetically conserved, and transfection with native genes corrected the biochemical deficiency proving pathogenicity. Treatments of cells with lipoate and with mitochondrially-targeted lipoate were unsuccessful at correcting the deficiency. The recognition of variant nonketotic hyperglycinemia is important for physicians evaluating patients with abnormalities in glycine as this will affect the genetic causation and genetic counselling, and provide prognostic information on the expected phenotypic course.


Subject(s)
Genetic Variation/genetics , Glutaredoxins/genetics , Hyperglycinemia, Nonketotic/genetics , Mutation/genetics , Proteins/genetics , Sulfurtransferases/genetics , Atrophy , Child , Child, Preschool , Fatal Outcome , Female , Glutaredoxins/chemistry , Humans , Hyperglycinemia, Nonketotic/diagnosis , Hyperglycinemia, Nonketotic/pathology , Infant , Male , Mitochondrial Proteins , Proteins/chemistry , Severity of Illness Index , Sulfurtransferases/chemistry
8.
PLoS One ; 8(3): e57195, 2013.
Article in English | MEDLINE | ID: mdl-23469184

ABSTRACT

Alaskan Husky Encephalopathy (AHE) has been previously proposed as a mitochondrial encephalopathy based on neuropathological similarities with human Leigh Syndrome (LS). We studied 11 Alaskan Husky dogs with AHE, but found no abnormalities in respiratory chain enzyme activities in muscle and liver, or mutations in mitochondrial or nuclear genes that cause LS in people. A genome wide association study was performed using eight of the affected dogs and 20 related but unaffected control AHs using the Illumina canine HD array. SLC19A3 was identified as a positional candidate gene. This gene controls the uptake of thiamine in the CNS via expression of the thiamine transporter protein THTR2. Dogs have two copies of this gene located within the candidate interval (SLC19A3.2 - 43.36-43.38 Mb and SLC19A3.1 - 43.411-43.419 Mb) on chromosome 25. Expression analysis in a normal dog revealed that one of the paralogs, SLC19A3.1, was expressed in the brain and spinal cord while the other was not. Subsequent exon sequencing of SLC19A3.1 revealed a 4bp insertion and SNP in the second exon that is predicted to result in a functional protein truncation of 279 amino acids (c.624 insTTGC, c.625 C>A). All dogs with AHE were homozygous for this mutation, 15/41 healthy AH control dogs were heterozygous carriers while 26/41 normal healthy AH dogs were wild type. Furthermore, this mutation was not detected in another 187 dogs of different breeds. These results suggest that this mutation in SLC19A3.1, encoding a thiamine transporter protein, plays a critical role in the pathogenesis of AHE.


Subject(s)
Brain Diseases, Metabolic/genetics , Brain Diseases, Metabolic/veterinary , Dog Diseases/genetics , Membrane Transport Proteins/genetics , Mutation , Animals , Base Sequence , Biological Transport/genetics , Brain Diseases, Metabolic/metabolism , Brain Diseases, Metabolic/pathology , Dog Diseases/metabolism , Dog Diseases/pathology , Dogs , Exons , Female , Genetic Loci , Genome-Wide Association Study , Heterozygote , Homozygote , Humans , Leigh Disease/genetics , Leigh Disease/metabolism , Leigh Disease/pathology , Male , Membrane Transport Proteins/metabolism , Molecular Sequence Data , Polymorphism, Single Nucleotide , Thiamine/metabolism
9.
Am J Med Genet A ; 161A(2): 285-94, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23322652

ABSTRACT

Complex II deficiency is a rare cause of mitochondrial respiratory chain defects with a prevalence of 2-23%. It is exclusively nuclear encoded and functions in the citric acid cycle by oxidizing succinate to fumarate and in the mitochondrial electron transport chain (ETC) by transferring electrons to ubiquinone. Of the four subunits, SDHA and SDHB are catalytic and SDHC and SDHD are anchoring. Mutations in SDHA and SDHAF1 (assembly factor) have been found in patients with CII deficiency and a mitochondrial phenotype. We present a patient with CII deficiency with a previously undescribed phenotype of dilated cardiomyopathy, left ventricular noncompaction, failure to thrive, hypotonia, and developmental delay. Also, a comprehensive review of 36 cases published in the literature was undertaken. The results show that CII deficiency has a variable phenotype with no correlation with residual complex activity in muscle although the phenotype and enzyme activities are comparable within a family. For some, the condition was fatal in infancy, others had multisystem involvement and some had onset in adulthood with mild symptoms and normal cognition. Neurological involvement is most commonly observed and brain imaging commonly shows leukoencephalopathy, Leigh syndrome, or cerebellar atrophy. Mutations in SDHAF1 are associated with leukoencephalopathy. Other organ systems like heart, muscle, and eyes are only involved in about 50% of the cases but cardiomyopathy is associated with high mortality and morbidity. In some patients, riboflavin has provided clinical improvement.


Subject(s)
Brain Diseases, Metabolic, Inborn/diagnosis , Succinate Dehydrogenase/deficiency , Brain Diseases, Metabolic, Inborn/blood , Brain Diseases, Metabolic, Inborn/enzymology , Electron Transport Complex II/deficiency , Electron Transport Complex II/genetics , Fatal Outcome , Female , Humans , Infant , Lactic Acid/blood , Lactic Acid/cerebrospinal fluid , Succinate Dehydrogenase/genetics
10.
Mol Genet Metab ; 108(1): 40-50, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23266196

ABSTRACT

We report on two families with Sengers syndrome and mutations in the acylglycerol kinase gene (AGK). In the first family, two brothers presented with vascular strokes, lactic acidosis, cardiomyopathy and cataracts, abnormal muscle cell histopathology and mitochondrial function. One proband had very abnormal mitochondria with citrate synthase crystals visible in electron micrographs, associated with markedly high citrate synthase activity. Exome sequencing was used to identify mutations in the AGK gene in the index patient. Targeted sequencing confirmed the same homozygous mutation (c.3G>A, p.M1I) in the brother. The second family had four affected members, of which we examined two. They also presented with similar clinical symptoms, but no strokes. Postmortem heart and skeletal muscle tissues showed low complex I, III and IV activities in the heart, but normal in the muscle. Skin fibroblasts showed elevated lactate/pyruvate ratios and low complex I+III activity. Targeted sequencing led to identification of a homozygous c.979A>T, p.K327* mutation. AGK is located in the mitochondria and phosphorylates monoacylglycerol and diacylglycerol to lysophosphatidic acid and phosphatidic acid. Disruption of these signaling molecules affects the mitochondria's response to superoxide radicals, resulting in oxidative damage to mitochondrial DNA, lipids and proteins, and stimulation of cellular detoxification pathways. High levels of manganese superoxide dismutase protein were detected in all four affected individuals, consistent with increased free radical damage. Phosphatidic acid is also involved in the synthesis of phospholipids and its loss will result in changes to the lipid composition of the inner mitochondrial membrane. These effects manifest as cataract formation in the eye, respiratory chain dysfunction and cardiac hypertrophy in heart tissue. These two pedigrees confirm that mutation of AGK is responsible for the severe neonatal presentation of Sengers syndrome. The identification of citrate synthase precipitates by electron microscopy and the presence of vascular strokes in two siblings may expand the cellular and clinical phenotype of this disease.


Subject(s)
Cardiomyopathies/enzymology , Cataract/enzymology , Citrate (si)-Synthase/chemistry , Mitochondria/enzymology , Mutation , Phosphotransferases (Alcohol Group Acceptor)/genetics , Base Sequence , Child, Preschool , Crystallization , DNA Primers , Female , Humans , Infant , Male , Pedigree
11.
Biochem J ; 441(1): 275-83, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21880015

ABSTRACT

LRPPRC (leucine-rich pentatricopeptide repeat-containing) has been shown to be essential for the maturation of COX (cytochrome c oxidase), possibly by stabilizing RNA transcripts of COXI, COXII and COXIII genes encoded in mtDNA (mitochondrial DNA). We established a mouse 'gene-trap' model using ES cells (embryonic stem cells) in which the C-terminus of LRPPRC has been replaced with a ß-geo construct. Mice homozygous for this modification were found to be subject to embryonic lethality, with death before 12.5 dpc (days post-coitum). Biochemical analysis of MEFs (mouse embryonic fibroblasts) isolated from homozygous mutants showed a major decrease in COX activity, with slight reductions in other respiratory chain complexes with mtDNA encoded components. Constructs of LRPPRC containing different numbers of PPRs (pentatricopeptide repeats) were expressed as recombinant proteins and tested for their ability to bind to the COXI mRNA transcript. Full binding required the first 19 PPR motifs. A specific segment of COXI mRNA was identified as the binding target for LRPPRC, encoded by mouse mtDNA nucleotides 5961-6020. These data strongly suggest that LRPPRC is involved in the maturation of COX, and is involved in stabilizing of mitochondrial mRNAs encoding COX transcripts.


Subject(s)
Electron Transport Complex IV/metabolism , Gene Expression Regulation, Developmental/physiology , Mitochondria/metabolism , Neoplasm Proteins/metabolism , RNA/metabolism , Animals , Cell Line , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Electron Transport Complex IV/classification , Electron Transport Complex IV/genetics , Embryo, Mammalian , Embryonic Stem Cells , Fibroblasts/cytology , Fibroblasts/metabolism , Genotype , Mice , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mutation , Neoplasm Proteins/genetics , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , Protein Binding , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , beta-Galactosidase/metabolism
12.
Am J Hum Genet ; 89(4): 486-95, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21944046

ABSTRACT

Severe combined deficiency of the 2-oxoacid dehydrogenases, associated with a defect in lipoate synthesis and accompanied by defects in complexes I, II, and III of the mitochondrial respiratory chain, is a rare autosomal recessive syndrome with no obvious causative gene defect. A candidate locus for this syndrome was mapped to chromosomal region 2p14 by microcell-mediated chromosome transfer in two unrelated families. Unexpectedly, analysis of genes in this area identified mutations in two different genes, both of which are involved in [Fe-S] cluster biogenesis. A homozygous missense mutation, c.545G>A, near the splice donor of exon 6 in NFU1 predicting a p.Arg182Gln substitution was found in one of the families. The mutation results in abnormal mRNA splicing of exon 6, and no mature protein could be detected in fibroblast mitochondria. A single base-pair duplication c.123dupA was identified in BOLA3 in the second family, causing a frame shift that produces a premature stop codon (p.Glu42Argfs(∗)13). Transduction of fibroblast lines with retroviral vectors expressing the mitochondrial, but not the cytosolic isoform of NFU1 and with isoform 1, but not isoform 2 of BOLA3 restored both respiratory chain function and oxoacid dehydrogenase complexes. NFU1 was previously proposed to be an alternative scaffold to ISCU for the biogenesis of [Fe-S] centers in mitochondria, and the function of BOLA3 was previously unknown. Our results demonstrate that both play essential roles in the production of [Fe-S] centers for the normal maturation of lipoate-containing 2-oxoacid dehydrogenases, and for the assembly of the respiratory chain complexes.


Subject(s)
Carrier Proteins/genetics , Mutation , Oxidoreductases/metabolism , Proteins/genetics , Cytosol/metabolism , Electron Transport , Exons , Family Health , Female , Fibroblasts/metabolism , Homozygote , Humans , Iron-Sulfur Proteins/metabolism , Male , Mitochondria/metabolism , Mitochondrial Proteins , Mutation, Missense
13.
Mitochondrion ; 11(1): 191-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20920610

ABSTRACT

Mutations in the TMEM70 gene are responsible for a familial form of complex V deficiency presenting with 3-methylglutaconic aciduria, lactic acidosis, cardiomyopathy and mitochondrial myopathy. Here we present a case of TMEM70 deficiency due to compound heterozygous mutations, who displayed abnormal mitochondria with whorled cristae in muscle. Immunogold electron microscopy and tomography shows for the first time that nucleoid clusters of mtDNA are disrupted in the abnormal mitochondria, with both nucleoids and mitochondrial respiratory chain complexes confined to the outer rings of the whorls. This could explain the differential effects on the expression and assembly of complex V in different tissues.


Subject(s)
DNA, Mitochondrial/genetics , Heterozygote , Membrane Proteins/deficiency , Mitochondria, Muscle/ultrastructure , Mitochondria/ultrastructure , Mitochondrial Diseases/genetics , Mitochondrial Proteins/deficiency , Mutation , Submitochondrial Particles/ultrastructure , Acidosis, Lactic/genetics , Acidosis, Lactic/metabolism , Acidosis, Lactic/pathology , Adult , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Female , Fibroblasts/metabolism , Humans , Infant, Newborn , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria, Muscle/genetics , Mitochondria, Muscle/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Submitochondrial Particles/genetics , Tomography
14.
Mitochondrion ; 11(2): 264-72, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21050896

ABSTRACT

Cytochrome c oxidase (COX) activity reflects the expressed level of respiratory chain complexes, mtDNA levels, titer and mass of mitochondria. Activity is also indicative of the overall fitness of mt-transcription factors and the import, transcription and translation of mt-proteins. We have developed a high-throughput assay to measure COX activity using live cells to screen chemical libraries for compounds capable of increasing COX activity. These libraries have revealed four examples which elevated the activities of COX in NIH-3T3 fibroblasts and in fibroblasts from patients with COX defects independent of the peroxisome proliferator activated receptor family.


Subject(s)
Electron Transport Complex IV/drug effects , Animals , Blotting, Western , Cell Line, Transformed , Coloring Agents , Electron Transport Complex IV/metabolism , Enzyme Activation , Fibroblasts/enzymology , Humans , Mice , NIH 3T3 Cells
15.
Biochem Biophys Res Commun ; 399(3): 331-5, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20637729

ABSTRACT

Mitochondrial dysfunction is involved in the underlying pathology of Parkinson's Disease (PD). PINK1 deficiency, which gives rise to familial early-onset PD, is associated with this dysfunction as well as increased oxidative stress. We have established primary fibroblast cell lines from two patients with PD who carry mutations in the PINK1 gene. The phosphorylation of Akt is abrogated in the presence of oxidative stressors in the complete absence of PINK1 suggesting enhanced apoptotic signalling. We have found an imbalance between the production of reactive oxygen species where the capacity of the cell to remove these toxins by anti-oxidative enzymes is greatly reduced. The expression levels of the anti-oxidant enzymes glutathione peroxidase-1, MnSOD, peroxiredoxin-3 and thioredoxin-2 were diminished. The p66(Shc) adaptor protein has recently been identified to become activated by oxidative stress by phosphorylation at residue Ser36 which then translocates to the mitochondrial inner membrane space. The phosphorylation of p66(Shc) at Ser36 is significantly increased in PINK1 deficient cell lines under normal tissue culture conditions, further still in the presence of compounds which elicit oxidative stress. The stable transfection of PINK1 in the fibroblasts which display the null phenotype ameliorates the hyper-phosphorylation of p66(Shc).


Subject(s)
Oxidative Stress , Parkinson Disease/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Shc Signaling Adaptor Proteins/metabolism , Cell Line , Fibroblasts/metabolism , Glutathione Peroxidase/metabolism , Humans , Peroxiredoxins/metabolism , Phosphorylation , Protein Kinases/genetics , Serine/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1 , Superoxide Dismutase/metabolism , Thioredoxins/metabolism , Glutathione Peroxidase GPX1
16.
Mol Genet Metab ; 98(4): 378-82, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19699667

ABSTRACT

We report here the identification of a patient with muscle-specific glycogen synthase deficiency. The 8-year-old patient showed no prior signs of distress before collapsing during a bout of exercise, resulting in death. Initial post-mortem analysis of tissues suggested death was due to metabolic complications of mitochondrial myopathy, but upon further examination it was found that the anomalies were indicative of mitochondrial proliferation and oxidative compensation. A homozygous two base pair deletion was identified in exon 2 of GYS1, and the parents and sibling were confirmed as heterozygous carriers of the deletion. This case highlights the importance of differentiating between mitochondrial compensatory phenomena and true mitochondrial disease, and suggests that GYS1 deficiency could be a common cause of sudden cardiac death in children. Children with abnormal cardiac responses to increased workloads as well as those with defined myocardial disease should therefore be tested for GYS1 deficiency.


Subject(s)
Death, Sudden, Cardiac/etiology , Fibroblasts/enzymology , Fibroblasts/pathology , Glycogen Phosphorylase, Muscle Form/genetics , Mutation/genetics , Skin/pathology , Base Sequence , Cell Extracts , Child , DNA Mutational Analysis , Fatal Outcome , Female , Humans , Lactates/metabolism , Male , Mitochondria/enzymology , Mitochondria/pathology , Mitochondria/ultrastructure , Molecular Sequence Data , Pedigree , Sonication , Staining and Labeling
17.
Mol Genet Metab ; 98(4): 349-55, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19640754

ABSTRACT

Fibroblast cell lines are frequently used to diagnose genetic mitochondrial defects in children. The effect of enzyme deficiency on overall flux rate through metabolic pathways is, however, not generally considered. We have transposed an experimental paradigm that was developed for isolated perfused organs using (13)C-labeled substrates and (13)C-isotopomer analysis to probe pyruvate mitochondrial metabolism in cultured human fibroblast cell lines with normal or genetically mutant pyruvate decarboxylation (PDC) or carboxylation (PC) activity. Cells were incubated with 1mM [U-(13)C]pyruvate, and the (13)C-molar percent enrichment (MPE) of intracellular pyruvate, citrate, malate (as a surrogate of oxaloacetate) and aspartate was assessed by mass spectrometry. We estimated various flux ratios relevant to metabolic pathways involved in energy production, namely pyruvate formation, PDC, PC, and citrate recycling in the citric acid cycle (CAC). In all cell lines, exogenous pyruvate was predominately decarboxylated (PC/PDC ratios 0.01-0.3). PC-deficient cell lines displayed an expected negligible contribution of PC flux to oxaloacetate formation for citrate synthesis (PC/CS), which was associated with a greater contribution of PDC to acetyl-CoA formation (PDC/CS), and greater recycling of (13)C-labeled citrate into the CAC. In PDH-deficient cell lines, metabolic flux alterations were most apparent in cells with more than 50% reduction in enzyme activity. This led to an unexpected lower PC/CS flux ratio, while the PDC/CS flux ratio was unchanged. These data illustrate the usefulness of this approach in identifying unexpected metabolic consequences of genetic defects related to pyruvate metabolism.


Subject(s)
Fibroblasts/metabolism , Mass Spectrometry/methods , Mutation/genetics , Pyruvic Acid/metabolism , Aspartic Acid/metabolism , Carbon Isotopes , Cell Line , Chromatography, Liquid , Citric Acid/metabolism , Citric Acid Cycle/drug effects , Fibroblasts/drug effects , Gas Chromatography-Mass Spectrometry , Humans , Isotope Labeling , Malates/metabolism , Male , Pyruvic Acid/pharmacology
18.
Biochem J ; 416(1): 15-26, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18729827

ABSTRACT

Mice homozygous for a defect in the PTCD2 (pentatricopeptide repeat domain protein 2) gene were generated in order to study the role of this protein in mitochondrial RNA metabolism. These mice displayed specific but variable reduction of ubiquinol-cytochrome c reductase complex activity in mitochondria of heart, liver and skeletal muscle due to a decrease in the expression of mitochondrial DNA-encoded cytochrome b, the catalytic core of the complex. This reduction in mitochondrial function has a profound effect on the myocardium, with replacement of ventricular cardiomyocytes by fibro-fatty tissue. Northern blotting showed a reduction in the mRNA for the mitochondrial DNA encoded proteins cytochrome b (cytb) and ND5 (NADH dehydrogenase subunit 5) and an elevation in a combined pre-processed ND5-CYTB transcript. This suggests that the PTCD2 protein is involved in processing RNA transcripts involving cytochrome b derived from mitochondrial DNA. This defines the site for PTCD2 action in mammalian mitochondria and suggests a possible role for dysfunction of this protein in the aetiology of heart failure.


Subject(s)
Cytochromes b/biosynthesis , Electron Transport Complex III/biosynthesis , Genes, Mitochondrial/physiology , Mitochondria, Heart/enzymology , Mitochondrial Proteins/genetics , RNA-Binding Proteins/genetics , Animals , Gene Expression Regulation, Enzymologic , HeLa Cells , Humans , Mice , Microscopy, Electron , Mitochondria, Heart/ultrastructure , Mitochondria, Liver/enzymology , Mitochondria, Muscle/enzymology , Mitochondrial Proteins/physiology , RNA/metabolism , RNA, Mitochondrial , RNA-Binding Proteins/physiology
19.
Mol Genet Metab ; 90(1): 15-23, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17095275

ABSTRACT

Exercise intolerance syndromes are well known to be associated with inborn errors of metabolism affecting glycolysis (phosphorylase and phosphofructokinase deficiency) and fatty acid oxidation (palmitoyl carnitine transferase deficiency). We have identified a canine model for profound exercise intolerance caused by a deficit in PDP1 (EC 3.1.3.43), the phosphatase enzyme that activates the pyruvate dehydrogenase complex (PDHc). The Clumber spaniel breed was originated in 1760 by the Duc de Noailles, as a hunting dog with a gentle temperament suitable for the 'elderly gentleman'. Here we report that 20% of the current Clumber and Sussex spaniel population are carriers for a null mutation in PDP1, and that homozygosity produces severe exercise intolerance. Human pyruvate dehydrogenase phosphatase deficiency was recently characterized at the molecular level. However, the nature of the human mutation (loss of a single amino acid altering PDP1 activity) made it impossible to discern the role of the second phosphatase isoform, PDP2, in the deficient phenotype. Here we show that the null mutation in dogs provides a valuable animal model with which to study the effects of dysregulation of the PDHc. Knowledge of the molecular defect has allowed for the institution of a rapid restriction enzyme test for the canine mutation that will allow for selective breeding and has led to a suggested dietary therapy for affected dogs that has proven to be beneficial. Pharmacological and genetic therapies for PDP1 deficiency can now be investigated and the role of PDP2 can be fully characterized.


Subject(s)
Dogs , Isoenzymes/deficiency , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/deficiency , Animals , Cells, Cultured , Disease Models, Animal , Female , Humans , Isoenzymes/genetics , Male , Pedigree , Physical Conditioning, Animal/physiology , Point Mutation , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/genetics
20.
Am J Med Genet A ; 140(14): 1542-52, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16770810

ABSTRACT

We have diagnosed dihydrolipoamide dehydrogenase (DLD) deficiency in two male second cousins, who presented with markedly different clinical phenotypes. Patient 1 had a recurrent encephalopathy, and patient 2 had microcephaly and lactic acidosis. Their presentation is unusual, in that the DLD subunit deficiency had little effect on pyruvate dehydrogenase complex activity, but caused a severe reduction in the activities of other enzymes that utilize this subunit. We have identified two mutations in the DLD gene in each patient. The second cousins have one novel mutation in common resulting in a substitution of isoleucine for threonine (I47T), which has not been previously reported in the literature. Patient 1 has a second mutation that has been reported to be common in the Ashkenazi Jewish population, G229C. Patient 2 has a second mutation, E375K, which has also been previously reported in the literature. Enzyme kinetic measurements on patient fibroblasts show that under certain conditions, one heteroallelic mutation may have a higher K(m). This may account for the differing clinical phenotypes. These findings have important repercussions for other patients with similar clinical phenotypes, as DLD activity is not normally measured in cases with normal PDHc activity.


Subject(s)
Dihydrolipoamide Dehydrogenase/deficiency , Dihydrolipoamide Dehydrogenase/genetics , Point Mutation , Pyruvate Dehydrogenase Complex/metabolism , Acidosis, Lactic/enzymology , Acidosis, Lactic/genetics , Amino Acid Substitution , Base Sequence , Brain Diseases, Metabolic/enzymology , Brain Diseases, Metabolic/genetics , Child , Child, Preschool , DNA/genetics , Dihydrolipoamide Dehydrogenase/chemistry , Female , Fibroblasts/enzymology , Heterozygote , Humans , In Vitro Techniques , Kinetics , Male , Microcephaly/enzymology , Microcephaly/genetics , Models, Molecular , Phenotype , Protein Subunits , Pyruvate Dehydrogenase Complex/chemistry , Pyruvate Dehydrogenase Complex/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...