Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Haematologica ; 103(12): 2008-2015, 2018 12.
Article in English | MEDLINE | ID: mdl-30026338

ABSTRACT

YARS2 variants have previously been described in patients with myopathy, lactic acidosis and sideroblastic anemia 2 (MLASA2). YARS2 encodes the mitochondrial tyrosyl-tRNA synthetase, which is responsible for conjugating tyrosine to its cognate mt-tRNA for mitochondrial protein synthesis. Here we describe 14 individuals from 11 families presenting with sideroblastic anemia and YARS2 variants that we identified using a sideroblastic anemia gene panel or exome sequencing. The phenotype of these patients ranged from MLASA to isolated congenital sideroblastic anemia. As in previous cases, inter- and intra-familial phenotypic variability was observed, however, this report includes the first cases with isolated sideroblastic anemia and patients with biallelic YARS2 variants that have no clinically ascertainable phenotype. We identified ten novel YARS2 variants and three previously reported variants. In vitro amino-acylation assays of five novel missense variants showed that three had less effect on the catalytic activity of YARS2 than the most commonly reported variant, p.(Phe52Leu), associated with MLASA2, which may explain the milder phenotypes in patients with these variants. However, the other two missense variants had a more severe effect on YARS2 catalytic efficiency. Several patients carried the common YARS2 c.572 G>T, p.(Gly191Val) variant (minor allele frequency =0.1259) in trans with a rare deleterious YARS2 variant. We have previously shown that the p.(Gly191Val) variant reduces YARS2 catalytic activity. Consequently, we suggest that biallelic YARS2 variants, including severe loss-of-function alleles in trans of the common p.(Gly191Val) variant, should be considered as a cause of isolated congenital sideroblastic anemia, as well as the MLASA syndromic phenotype.


Subject(s)
Acidosis, Lactic/genetics , Anemia, Sideroblastic/genetics , Genetic Diseases, X-Linked/genetics , Germ-Line Mutation , MELAS Syndrome/genetics , Mitochondrial Proteins/genetics , Tyrosine-tRNA Ligase/genetics , Acidosis, Lactic/enzymology , Adolescent , Anemia, Sideroblastic/enzymology , Female , Genetic Association Studies , Genetic Diseases, X-Linked/enzymology , Humans , Infant , MELAS Syndrome/enzymology , Male , Middle Aged , Mutation, Missense , Young Adult
2.
Biochim Biophys Acta Mol Basis Dis ; 1864(5 Pt A): 1596-1608, 2018 May.
Article in English | MEDLINE | ID: mdl-29454073

ABSTRACT

Ketogenic diet (KD) which combined carbohydrate restriction and the addition of ketone bodies has emerged as an alternative metabolic intervention used as an anticonvulsant therapy or to treat different types of neurological or mitochondrial disorders including MELAS syndrome. MELAS syndrome is a severe mitochondrial disease mainly due to the m.3243A > G mitochondrial DNA mutation. The broad success of KD is due to multiple beneficial mechanisms with distinct effects of very low carbohydrates and ketones. To evaluate the metabolic part of carbohydrate restriction, transmitochondrial neuronal-like cybrid cells carrying the m.3243A > G mutation, shown to be associated with a severe complex I deficiency was exposed during 3 weeks to glucose restriction. Mitochondrial enzyme defects were combined with an accumulation of complex I (CI) matrix intermediates in the untreated mutant cells, leading to a drastic reduction in CI driven respiration. The severe reduction of CI was also paralleled in post-mortem brain tissue of a MELAS patient carrying high mutant load. Importantly, lowering significantly glucose concentration in cell culture improved CI assembly with a significant reduction of matrix assembly intermediates and respiration capacities were restored in a sequential manner. In addition, OXPHOS protein expression and mitochondrial DNA copy number were significantly increased in mutant cells exposed to glucose restriction. The accumulation of CI matrix intermediates appeared as a hallmark of MELAS pathophysiology highlighting a critical pathophysiological mechanism involving CI disassembly, which can be alleviated by lowering glucose fuelling and the induction of mitochondrial biogenesis, emphasizing the usefulness of metabolic interventions in MELAS syndrome.


Subject(s)
Electron Transport Complex I/metabolism , Glucose/metabolism , MELAS Syndrome/enzymology , Mitochondria/enzymology , Neurons/enzymology , Point Mutation , Cell Line, Tumor , Electron Transport Complex I/genetics , Female , Humans , MELAS Syndrome/genetics , MELAS Syndrome/pathology , Male , Mitochondria/genetics , Mitochondria/pathology , Neurons/pathology , Oxidative Phosphorylation
3.
Zhonghua Yi Xue Za Zhi ; 93(16): 1247-50, 2013 Apr 23.
Article in Chinese | MEDLINE | ID: mdl-23902617

ABSTRACT

OBJECTIVE: To explore the expression and significance of respiratory chain enzyme of cells in urine sediment in mitochondrial encephalopathy myopathy, lactic acidosis and stroke-like episodes (MELAS) syndrome. METHODS: Through enzyme histochemistry, the authors analyzed the changes of respiratory chain enzyme in urine sediment in 20 MELAS patients due to mitochondrial A3243G mutation (MELAS group) and 20 health peoples (control group). And the impact on the expression of protein encoded by nuclear DNA (A21347) and mitochondrial DNA (A6404) was detected by immunochemistry. Image pro Plus 6.0 software was used for analysis of absorbance (A) of staining images as staining intensity. The data were expressed as M (Q1, Q3) and analyzed through statistical software. RESULTS: The staining intensity of complexes Iin the MELAS group was lower than that in the control group (0.06(0.01, 0.12) vs 0.12(0.01, 0.62), P = 0.010). The intergroup staining intensity of complex II showed no marked difference. Increased density of blue particle and cytoplasmic gathering was found in 13 cased (65%) of the MELAS group under light microscope. The staining intensity of complexes IV was expressed at a low level in the MELAS group (0.14(0.03, 0.32) vs 0.23(0.06, 0.43), P = 0.038). The expression of protein encoded by nuclear DNA (A21347) was lower than that in the control group (0.05(0.02, 0.45) vs 0.17(0.03, 0.70), P = 0.000). The expression of protein encoded by mitochondrial DNA (A6404) was also lower than that in the control group (0.03(0.01, 0.07) vs 0.15 (0.09, 0.23), P = 0.000). CONCLUSION: Abnormal change of respiratory chain enzyme in urine sediment in MELAS due to mitochondrial A3243G mutation and a low expression of proteins encoded by two kinds of DNA in complexes IV can help to confirm the genetic diagnosis of mitochondrial encephalomyopathies so that different subtypes may be classified and its pathogenesis elucidated.


Subject(s)
Electron Transport Complex IV/urine , MELAS Syndrome/enzymology , Adolescent , Adult , Aged , Case-Control Studies , Cell Nucleus/genetics , Child , Child, Preschool , DNA, Mitochondrial/genetics , Electron Transport , Electron Transport Complex IV/genetics , Female , Humans , MELAS Syndrome/metabolism , MELAS Syndrome/urine , Male , Middle Aged , Mitochondrial Membranes/metabolism , Mutation , Young Adult
4.
Stem Cells ; 31(7): 1298-308, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23553816

ABSTRACT

Mitochondrial diseases display pathological phenotypes according to the mixture of mutant versus wild-type mitochondrial DNA (mtDNA), known as heteroplasmy. We herein examined the impact of nuclear reprogramming and clonal isolation of induced pluripotent stem cells (iPSC) on mitochondrial heteroplasmy. Patient-derived dermal fibroblasts with a prototypical mitochondrial deficiency diagnosed as mitochondrial encephalomyopathy with lactic acidosis and stroke-like episodes (MELAS) demonstrated mitochondrial dysfunction with reduced oxidative reserve due to heteroplasmy at position G13513A in the ND5 subunit of complex I. Bioengineered iPSC clones acquired pluripotency with multilineage differentiation capacity and demonstrated reduction in mitochondrial density and oxygen consumption distinguishing them from the somatic source. Consistent with the cellular mosaicism of the original patient-derived fibroblasts, the MELAS-iPSC clones contained a similar range of mtDNA heteroplasmy of the disease-causing mutation with identical profiles in the remaining mtDNA. High-heteroplasmy iPSC clones were used to demonstrate that extended stem cell passaging was sufficient to purge mutant mtDNA, resulting in isogenic iPSC subclones with various degrees of disease-causing genotypes. On comparative differentiation of iPSC clones, improved cardiogenic yield was associated with iPSC clones containing lower heteroplasmy compared with isogenic clones with high heteroplasmy. Thus, mtDNA heteroplasmic segregation within patient-derived stem cell lines enables direct comparison of genotype/phenotype relationships in progenitor cells and lineage-restricted progeny, and indicates that cell fate decisions are regulated as a function of mtDNA mutation load. The novel nuclear reprogramming-based model system introduces a disease-in-a-dish tool to examine the impact of mutant genotypes for MELAS patients in bioengineered tissues and a cellular probe for molecular features of individual mitochondrial diseases.


Subject(s)
DNA, Mitochondrial/genetics , Induced Pluripotent Stem Cells/metabolism , MELAS Syndrome/genetics , MELAS Syndrome/pathology , Mitochondria/genetics , Cellular Reprogramming/genetics , Cellular Reprogramming/physiology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/pathology , MELAS Syndrome/enzymology , MELAS Syndrome/metabolism , Mitochondria/pathology
5.
Biol Direct ; 7: 26, 2012 Aug 09.
Article in English | MEDLINE | ID: mdl-22873568

ABSTRACT

BACKGROUND: In previous work, we introduced a concept, a mathematical model and its computer realization that describe the interaction between bacterial and phage type RNA polymerases, protein factors, DNA and RNA secondary structures during transcription, including transcription initiation and termination. The model accurately reproduces changes of gene transcription level observed in polymerase sigma-subunit knockout and heat shock experiments in plant plastids. The corresponding computer program and a user guide are available at http://lab6.iitp.ru/en/rivals. Here we apply the model to the analysis of transcription and (partially) translation processes in the mitochondria of frog, rat and human. Notably, mitochondria possess only phage-type polymerases. We consider the entire mitochondrial genome so that our model allows RNA polymerases to complete more than one circle on the DNA strand. RESULTS: Our model of RNA polymerase interaction during transcription initiation and elongation accurately reproduces experimental data obtained for plastids. Moreover, it also reproduces evidence on bulk RNA concentrations and RNA half-lives in the mitochondria of frog, human with or without the MELAS mutation, and rat with normal (euthyroid) or hyposecretion of thyroid hormone (hypothyroid). The transcription characteristics predicted by the model include: (i) the fraction of polymerases terminating at a protein-dependent terminator in both directions (the terminator polarization), (ii) the binding intensities of the regulatory protein factor (mTERF) with the termination site and, (iii) the transcription initiation intensities (initiation frequencies) of all promoters in all five conditions (frog, healthy human, human with MELAS syndrome, healthy rat, and hypothyroid rat with aberrant mtDNA methylation). Using the model, absolute levels of all gene transcription can be inferred from an arbitrary array of the three transcription characteristics, whereas, for selected genes only relative RNA concentrations have been experimentally determined. Conversely, these characteristics and absolute transcription levels can be obtained using relative RNA concentrations and RNA half-lives known from various experimental studies. In this case, the "inverse problem" is solved with multi-objective optimization. CONCLUSIONS: In this study, we demonstrate that our model accurately reproduces all relevant experimental data available for plant plastids, as well as the mitochondria of chordates. Using experimental data, the model is applied to estimate binding intensities of phage-type RNA polymerases to their promoters as well as predicting terminator characteristics, including polarization. In addition, one can predict characteristics of phage-type RNA polymerases and the transcription process that are difficult to measure directly, e.g., the association between the promoter's nucleotide composition and the intensity of polymerase binding. To illustrate the application of our model in functional predictions, we propose a possible mechanism for MELAS syndrome development in human involving a decrease of Phe-tRNA, Val-tRNA and rRNA concentrations in the cell. In addition, we describe how changes in methylation patterns of the mTERF binding site and three promoters in hypothyroid rat correlate with changes in intensities of the mTERF binding and transcription initiations. Finally, we introduce an auxiliary model to describe the interaction between polysomal mRNA and ribonucleases.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Mitochondria/enzymology , Models, Molecular , Promoter Regions, Genetic , Animals , Anura/genetics , Anura/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Binding Sites , Chaperonin 60/genetics , Chaperonin 60/metabolism , DNA Methylation , DNA-Directed RNA Polymerases/genetics , Genome, Mitochondrial , Half-Life , Humans , Hypothyroidism/enzymology , Hypothyroidism/genetics , MELAS Syndrome/enzymology , MELAS Syndrome/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Polyribosomes/enzymology , Polyribosomes/genetics , Protein Interaction Mapping/methods , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , Rats , Reproducibility of Results , Transcription, Genetic
6.
Hum Mol Genet ; 20(23): 4634-43, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21890497

ABSTRACT

MTU1 (TRMU) is a mitochondrial enzyme responsible for the 2-thiolation of the wobble U in tRNA(Lys), tRNA(Glu) and tRNA(Gln), a post-transcriptional modification believed to be important for accurate and efficient synthesis of the 13 respiratory chain subunits encoded by mtDNA. Mutations in MTU1 are associated with acute infantile liver failure, and this has been ascribed to a transient lack of cysteine, the sulfur donor for the thiouridylation reaction, resulting in a mitochondrial translation defect during early development. A mutation in tRNA(Lys) that causes myoclonic epilepsy with ragged-red fibers (MERRF) is also reported to prevent modification of the wobble U. Here we show that mitochondrial translation is unaffected in fibroblasts from an MTU1 patient, in which MTU1 is undetectable by immunoblotting, despite the severe reduction in the 2-thiolation of mitochondrial tRNA(Lys), tRNA(Glu) and tRNA(Gln). The only respiratory chain abnormality that we could observe in these cells was an accumulation of a Complex II assembly intermediate, which, however, did not affect the level of the fully assembled enzyme. The identical phenotype was observed by siRNA-mediated knockdown of MTU1 in HEK 293 cells. Further, the mitochondrial translation deficiencies present in myoblasts from mitochondrial encephalomyopathy, lactic acidosis and stroke-like episode and MERRF patients, which are associated with defects in post-transcriptional modification of mitochondrial tRNAs, did not worsen following knockdown of MTU1 in these cells. This study demonstrates that MTU1 is not required for mitochondrial translation at normal steady-state levels of tRNAs, and that it may possess an as yet uncharacterized function in another sulfur-trafficking pathway.


Subject(s)
Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Protein Biosynthesis , tRNA Methyltransferases/metabolism , Electrophoresis, Polyacrylamide Gel , Fibroblasts/enzymology , Fibroblasts/pathology , Gene Knockdown Techniques , HEK293 Cells , Humans , MELAS Syndrome/enzymology , MELAS Syndrome/pathology , MERRF Syndrome/enzymology , MERRF Syndrome/pathology , Mitochondrial Proteins/deficiency , Mutation/genetics , Myoblasts/enzymology , Myoblasts/pathology , Oxidative Phosphorylation , RNA, Transfer/metabolism , Thiouridine/metabolism , tRNA Methyltransferases/deficiency
7.
Mol Cell Biol ; 30(9): 2147-54, 2010 May.
Article in English | MEDLINE | ID: mdl-20194621

ABSTRACT

Mutations in mitochondrial tRNA genes are associated with a wide spectrum of human diseases. In particular, the tRNA(Leu(UUR)) A3243G mutation causes mitochondrial encephalomyopathy, lactic acidosis, and stroke-like symptoms (MELAS) and 2% of cases of type 2 diabetes. The primary defect in this mutation was an inefficient aminoacylation of the tRNA(Leu(UUR)). In the present study, we have investigated the molecular mechanism of the A3243G mutation and whether the overexpression of human mitochondrial leucyl-tRNA synthetase (LARS2) in the cytoplasmic hybrid (cybrid) cells carrying the A3243G mutation corrects the mitochondrial dysfunctions. Human LARS2 localizes exclusively to mitochondria, and LARS2 is expressed ubiquitously but most abundantly in tissues with high metabolic rates. We showed that the alteration of aminoacylation tRNA(Leu(UUR)) caused by the A3243G mutation led to mitochondrial translational defects and thereby reduced the aminoacylated efficiencies of tRNA(Leu(UUR)) as well as tRNA(Ala) and tRNA(Met). We demonstrated that the transfer of human mitochondrial leucyl-tRNA synthetase into the cybrid cells carrying the A3243G mutation improved the efficiency of aminoacylation and stability of mitochondrial tRNAs and then increased the rates of mitochondrial translation and respiration, consequently correcting the mitochondrial dysfunction. These findings provide new insights into the molecular mechanism of maternally inherited diseases and a step toward therapeutic interventions for these disorders.


Subject(s)
Diabetes Mellitus, Type 2/enzymology , Genetic Predisposition to Disease , Leucine-tRNA Ligase/genetics , MELAS Syndrome/enzymology , Mitochondria/enzymology , Mutation/genetics , RNA, Transfer, Leu/genetics , Cell Line, Tumor , Cell Respiration , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Gene Expression Regulation, Enzymologic , Humans , Kinetics , Leucine-tRNA Ligase/metabolism , MELAS Syndrome/complications , MELAS Syndrome/genetics , MELAS Syndrome/pathology , Protein Biosynthesis , Protein Transport , RNA Processing, Post-Transcriptional , Subcellular Fractions/enzymology , Transfection , Transfer RNA Aminoacylation
8.
J Med Genet ; 45(2): 117-21, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18245391

ABSTRACT

BACKGROUND: MELAS (mitochondrial myopathy, encephalopathy, lactic acidosis and stroke-like episodes) is one of the more common mitochondrial encephalomyopathies. About 80% of MELAS cases are caused by transition 3243A-->G in the mitochondrial tRNA(Leu(UUR)) gene (MT-TL1). Other mutations in MT-TL1, other mitochondrial tRNA genes and mitochondrial-encoded subunits of respiratory complex I account for the remainder of cases. OBJECTIVE: To characterise the molecular basis of a MELAS case without a mutation in any recognised MELAS target gene. RESULTS AND METHODS: Deletion of a single nucleotide (7630delT) within MT-CO2, the gene of subunit II of cytochrome c oxidase (COX), was identified by mitochondrial DNA (mtDNA) sequencing. The deletion-induced frameshift results in a stop codon close to the 5' end of the reading frame. The lack of subunit II (COII) precludes the assembly of COX and leads to the degradation of unassembled subunits, even those not directly affected by the mutation. Despite mitochondrial proliferation and transcriptional upregulation of nuclear and mtDNA-encoded COX genes (including MT-CO2), a severe COX deficiency was found with all investigations of the muscle biopsy (histochemistry, biochemistry, immunoblotting). CONCLUSIONS: The 7630delT mutation in MT-CO2 leads to a lack of COII with subsequent misassembly and degradation of respiratory complex IV despite transcriptional upregulation of its subunits. The causal association of the resulting isolated COX deficiency with MELAS is at odds with current concepts of the biochemical deficits underlying this common mitochondrial disease, and indicates that the genetic and pathobiochemical heterogeneity of MELAS is greater than previously appreciated.


Subject(s)
Cytochrome-c Oxidase Deficiency/enzymology , Cytochrome-c Oxidase Deficiency/genetics , Electron Transport Complex IV/genetics , MELAS Syndrome/enzymology , MELAS Syndrome/genetics , Adult , Amino Acid Sequence , Base Sequence , Cytochrome-c Oxidase Deficiency/complications , DNA, Mitochondrial/genetics , Humans , MELAS Syndrome/etiology , MELAS Syndrome/pathology , Male , Molecular Sequence Data , Muscles/enzymology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Deletion
9.
Arch Neurol ; 62(3): 473-6, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15767514

ABSTRACT

BACKGROUND: The mitochondrial DNA gene encoding subunit 5 of complex I (ND5) has turned out to be a hot spot for mutations associated with mitochondrial encephalomyopathy with lactic acidosis and strokelike episodes (MELAS) and various overlap syndromes. OBJECTIVE: To describe a novel mutation in the ND5 gene in a young man man with an overlap syndrome of MELAS and myoclonus epilepsy with ragged-red fibers. DESIGN: Case report. PATIENT: A 25-year-old man had recurrent strokes, seizures, and myoclonus. His mother also had multiple strokes. A muscle biopsy specimen showed no ragged-red fibers but several strongly succinate dehydrogenase-reactive blood vessels. RESULTS: Biochemical analysis showed isolated complex I deficiency and molecular analysis revealed a novel heteroplasmic mutation (G13042A) in the ND5 gene. CONCLUSIONS: These data confirm that ND5 is a genetic hot spot for overlap syndromes, including MELAS and strokelike and myoclonus epilepsy with ragged-red fibers.


Subject(s)
DNA, Mitochondrial/genetics , Electron Transport Complex I/genetics , MELAS Syndrome/genetics , MERRF Syndrome/genetics , Mutation , Protein Subunits/genetics , Adult , Amino Acid Sequence , Humans , MELAS Syndrome/enzymology , MELAS Syndrome/pathology , MERRF Syndrome/enzymology , MERRF Syndrome/pathology , Male , Mitochondrial Proteins , Molecular Sequence Data , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism
10.
J Med Genet ; 40(3): 188-91, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12624137

ABSTRACT

Leigh syndrome is a subacute necrotising encephalomyopathy frequently ascribed to mitochondrial respiratory chain deficiency. This condition is genetically heterogeneous, as mutations in both mitochondrial (mt) and nuclear genes have been reported. Here, we report the G13513A transition in the ND5 mtDNA gene in three unrelated children with complex I deficiency and a peculiar MRI aspect distinct from typical Leigh syndrome. Brain MRI consistently showed a specific involvement of the substantia nigra and medulla oblongata sparing the basal ganglia. Variable degrees of heteroplasmy were found in all tissues tested and a high percentage of mutant mtDNA was observed in muscle. The asymptomatic mothers presented low levels of mutant mtDNA in blood leucocytes. This mutation, which affects an evolutionary conserved amino acid (D393N), has been previously reported in adult patients with MELAS or LHON/MELAS syndromes, emphasising the clinical heterogeneity of mitochondrial DNA mutations. Since the G13513A mutation was found in 21% of our patients with Leigh syndrome and complex I deficiency (3/14), it appears that this mutation represents a frequent cause of Leigh-like syndrome, which should be systematically tested for molecular diagnosis in affected children and for genetic counselling in their maternal relatives.


Subject(s)
DNA, Mitochondrial/genetics , Leigh Disease/genetics , MELAS Syndrome/genetics , NADH Dehydrogenase/genetics , NADH, NADPH Oxidoreductases/deficiency , Brain/pathology , Child, Preschool , DNA, Mitochondrial/metabolism , Deoxyribonucleases, Type II Site-Specific/metabolism , Electron Transport Complex I , Humans , Infant , Leigh Disease/enzymology , Leigh Disease/pathology , MELAS Syndrome/enzymology , Magnetic Resonance Imaging , Male , NADH, NADPH Oxidoreductases/genetics , Point Mutation
11.
J Neuropathol Exp Neurol ; 61(10): 885-95, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12387454

ABSTRACT

To assess the detailed expression pattern of mitochondrial-encoded proteins in skeletal muscle of patients with mitochondrial diseases we performed determinations of cytochrome content and enzyme activities of respiratory chain complexes of 12 patients harboring large-scale deletions and of 10 patients harboring the A3243G mutation. For large-scale deletions we observed a mutation gene dose-dependent linear decline of cytochrome aa3 content, cytochrome c oxidase (COX) activity, and complex I activity. The content of cytochromes b and the complex III activity was either not affected or only weakly affected by the deletion mutation and did not correlate to the degree of heteroplasmy. In contrast, in skeletal muscle harboring the A3243G mutation all investigated enzymes containing mitochondrial-encoded subunits were equally affected by the mutation, but we observed milder enzyme deficiencies at a comparable mutation gene dose. The results of single fiber analysis of selected biopsies supported these findings but revealed differences in the distribution of COX deficiency. Whereas predominantly type I fibers were affected in A3243G and deletion CPEO biopsies, we observed in MELAS and KSS biopsies higher quantities of COX-deficient type 2 fibers. Our findings indicate different pathomechanisms of deletion and A3243G mutations.


Subject(s)
Citrate (si)-Synthase/genetics , Cytochromes/genetics , DNA, Mitochondrial/genetics , Mitochondria, Muscle/enzymology , Muscle, Skeletal/enzymology , Point Mutation , Sequence Deletion , Base Sequence , Female , Humans , MELAS Syndrome/enzymology , MELAS Syndrome/genetics , MELAS Syndrome/pathology , Male , Mitochondria, Muscle/pathology , Muscle, Skeletal/pathology
12.
Eur J Hum Genet ; 9(10): 805-9, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11781695

ABSTRACT

We report a novel point mutation in the gene for the mitochondrially encoded ND6 subunit of the NADH:ubiquinone oxidoreductase (complex I of the respiratory chain) in a patient with MELAS syndrome. The mutation causes a change from alanine to valine in the most conserved region of the ND6 subunit. The patient was heteroplasmic for the mutation in both muscle and blood, but the mutation was not detected in the patient's mother. A marked reduction of complex I activity was found in the patient's muscular tissue. This is the first report of a mutation in the ND6 subunit causing MELAS. Our data confirm the genetic heterogeneity in mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes syndrome, and confirms that MELAS can be caused by mutation in polypeptide-coding mtDNA genes.


Subject(s)
DNA, Mitochondrial/genetics , MELAS Syndrome/enzymology , MELAS Syndrome/genetics , Mutation/genetics , NADH Dehydrogenase/chemistry , NADH Dehydrogenase/genetics , Base Sequence , Child, Preschool , DNA Mutational Analysis , DNA, Mitochondrial/blood , Female , Humans , MELAS Syndrome/blood , Mitochondria, Muscle/enzymology , NADH Dehydrogenase/metabolism , Protein Subunits , Restriction Mapping
13.
J Child Neurol ; 15(11): 755-8, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11108510

ABSTRACT

A variety of endocrine and metabolic defects, including hypothalamopituitary hypofunction and diabetes mellitus, has been reported in association with mitochondrial disorders. We describe two sisters affected by mitochondrial encephalomyopathy, lactic acidosis, and strokelike episodes (MELAS) syndrome in whom DNA analysis showed an A-->G transition at the 3243rd nucleotide position on the transfer RNALeu(UUR) gene with 65% and 45% of mutant-type mitochondrial DNA present in the blood cells of the younger and the older sister, respectively. The younger sister had severe involvement of the central nervous system with mental retardation, epilepsia partialis continua, and strokelike episodes. Endocrine investigations showed an extensive neuroendocrine dysfunction with growth hormone deficiency, hypothalamopituitary hypothyroidism, prepubertal gonadotropin levels, and absence of any secondary sexual characteristics at the age of 12 6/12 years. The neurologically normal older sister was affected by diabetes mellitus and had normal hypothalamopituitary function. Our report confirms that the endocrine system can be affected differently by the same mitochondrial DNA mutation, depending on the heteroplasmia phenomenon. A complete endocrine evaluation must be performed in patients affected by mitochondrial disease and the existence of a mitochondrial disorder should be taken into account in patients with endocrine abnormalities, even if neuromuscular signs are lacking.


Subject(s)
DNA, Mitochondrial/genetics , Endocrine System Diseases/diagnosis , Intellectual Disability/genetics , MELAS Syndrome/genetics , MELAS Syndrome/physiopathology , Mutation , Adolescent , Child , DNA, Mitochondrial/metabolism , Diagnosis, Differential , Endocrine System Diseases/genetics , Fatal Outcome , Female , Gene Expression Regulation , Genotype , Humans , MELAS Syndrome/enzymology , Phenotype
14.
Neuropediatrics ; 31(3): 114-21, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10963097

ABSTRACT

The aim of this study was to assess an optimal screening for paediatric patients suspected of mitochondriocytopathy to justify a muscle biopsy. Forty-five patients were included. Medical history, physical examination, cardiac and ophthalmologic evaluation, clinical chemical investigations, in vivo function tests, neuroimaging and a skeletal muscle biopsy were performed in all patients. The results of the biochemical muscle studies were compared with the results of the other investigations. First, parameters with a statistical relationship with the result in muscle, normal or deficient, were selected. Secondly, a prognostic index was constructed using these parameters. Five parameters were selected: age <4 years, elevated fasting lactate to pyruvate ratio, elevated thrombocyte count, elevated lactate, and elevated alanine. Each parameter was scored 0 (not present) or 1 (present). The chance of a normal biopsy with a given value of this index (sum of the scores) was calculated: logit (Pr) = alpha + beta x index; alpha: -0.8167 and beta: 0.8331. (Pr: probability of normal biopsy.) The chance of a normal biopsy with an index value of 5 is 0.03, 4 is 0.07, 3 is 0.16, 2 is 0.30, 1 is 0.50 and 0 is 0.69. This prognostic index is a valuable instrument in deciding whether the suspicion of mitochondriocytopathy is strong enough to merit a muscle biopsy.


Subject(s)
MELAS Syndrome/diagnosis , Muscle, Skeletal/pathology , Adolescent , Adult , Biopsy , Child , Child, Preschool , DNA, Mitochondrial/genetics , Electron Transport/physiology , Female , Humans , Infant , Infant, Newborn , Lactic Acid/blood , MELAS Syndrome/enzymology , MELAS Syndrome/genetics , Male , Mitochondrial ADP, ATP Translocases/metabolism , Muscle, Skeletal/enzymology , Oxidative Stress , Phosphorylation , Prognosis , Pyruvate Dehydrogenase Complex/metabolism , Reference Values , Severity of Illness Index
15.
Arch Dis Child ; 82(5): 407-11, 2000 May.
Article in English | MEDLINE | ID: mdl-10799437

ABSTRACT

AIMS: To clarify the phenotype-genotype relation associated with the A3243G mitochondrial DNA mutation. METHODS: Five unrelated probands harbouring the A3243G mutation but presenting different clinical phenotype were analysed. Probands include Leigh syndrome (LS(3243)), mitochondrial myopathy, encephalopathy, lactic acidosis and stroke like episodes (MELAS(3243)), progressive external ophthalmoplegia (PEO(3243)), and mitochondrial diabetes mellitus (MDM(3243)). Extensive clinical, histological, biochemical, and molecular genetic studies were performed on five families. RESULTS: All patients showed ragged red fibres (RRF), and focal cytochrome c oxidase (COX) deficiency except for the patient with MDM(3243). The mutation load was highest in the proband with LS(3243) (>90%), who also presented the highest proportion of RRF (68%) and COX negative fibres (10%), and severe complex I plus IV deficiency. These proportions were lower in the probands with PEO(3243) and with MDM(3243). CONCLUSION: The most severe clinical phenotype, LS(3243), was associated with the highest proportion of the A3243G mutation as well as the most prominent histological and biochemical abnormalities.


Subject(s)
DNA, Mitochondrial/genetics , Mutation/genetics , RNA, Transfer/genetics , RNA/genetics , Adolescent , Adult , Child , Child, Preschool , Diabetes Mellitus/genetics , Female , Genotype , Humans , Infant , Leigh Disease/enzymology , Leigh Disease/genetics , MELAS Syndrome/enzymology , MELAS Syndrome/genetics , Male , Middle Aged , Mitochondrial Encephalomyopathies/enzymology , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Myopathies/enzymology , Mitochondrial Myopathies/genetics , Molecular Biology , Ophthalmoplegia, Chronic Progressive External/enzymology , Ophthalmoplegia, Chronic Progressive External/genetics , Pedigree , Phenotype , RNA, Mitochondrial
16.
Brain ; 123 Pt 3: 591-600, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10686181

ABSTRACT

Cytochrome c oxidase (COX) is encoded by three mitochondrial and nine nuclear genes. COX deficiency is genetically heterogeneous but current diagnostic methods cannot easily distinguish between mitochondrial and nuclear defects. We hypothesized that there may be differential expression of COX subunits depending on the underlying mutation. COX subunit expression was investigated in five patients with known mtDNA mutations. Severe and selective reduction of mtDNA-encoded COX subunits I and II was consistently observed in all these patients and was restricted to COX-deficient fibres. Immunostaining of nuclear-encoded subunits COX IV and Va was normal, whilst subunit VIc, also nuclear-encoded, was decreased. Twelve of 36 additional patients with histochemically defined COX deficiency also had this pattern of staining, suggesting that they had mtDNA defects. Clinical features in this group were heterogeneous, including infantile encephalopathy, multisystem disease, cardiomyopathy and childhood-onset isolated myopathy. The remaining patients did not have the same pattern of immunostaining. Fourteen had reduced staining of all subunits, whilst 10 had normal staining of all subunits despite reduced enzyme activity. Patients with COX deficiency secondary to mtDNA mutations have a specific pattern of subunit loss, but the majority of children with COX deficiency do not have this pattern of subunit loss and are likely to have nuclear gene defects.


Subject(s)
DNA, Mitochondrial/analysis , Electron Transport Complex IV/analysis , Electron Transport Complex IV/genetics , MELAS Syndrome/genetics , MERRF Syndrome/genetics , Adolescent , Child , Child, Preschool , Cytochrome-c Oxidase Deficiency , Electron Transport , Female , Gene Expression Regulation, Enzymologic , Humans , Immunoenzyme Techniques , Infant , Infant, Newborn , MELAS Syndrome/enzymology , MELAS Syndrome/pathology , MERRF Syndrome/enzymology , MERRF Syndrome/pathology , Male , Middle Aged , Muscle, Skeletal/enzymology , Mutation
17.
Biochim Biophys Acta ; 1500(1): 10-6, 2000 Jan 03.
Article in English | MEDLINE | ID: mdl-10564713

ABSTRACT

The MELAS syndrome (mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes) is most commonly caused by the 3243A-->G mutation in mitochondrial DNA, resulting in impaired mitochondrial protein synthesis and decreased activities of the respiratory chain complexes. These defects may cause a reduced capacity for ATP synthesis and an increased rate of production of reactive oxygen species. Myoblasts cultured from controls and patients carrying the 3243A-->G mutation were used to measure ATP, ADP, catalase and superoxide dismutase, which was also measured from blood samples. ATP and ADP concentrations were decreased in myoblasts with the 3243A-->G mutation, but the ATP/ADP ratio remained constant, suggesting a decrease in the adenylate pool. The superoxide dismutase and catalase activities were higher than in control cells, and superoxide dismutase activity was slightly, but not significantly higher in the blood of patients with the mutation than in controls. We conclude that impairment of mitochondrial ATP production in myoblasts carrying the 3243A-->G mutation results in adenylate catabolism, causing a decrease in the total adenylate pool. The increase in superoxide dismutase and catalase activities could be an adaptive response to increased production of reactive oxygen species due to dysfunction of the mitochondrial respiratory chain.


Subject(s)
Adenosine Triphosphate/metabolism , Antioxidants/metabolism , DNA, Mitochondrial/genetics , MELAS Syndrome/enzymology , Muscle, Skeletal/enzymology , Adenosine Diphosphate/metabolism , Adult , Catalase/metabolism , Cells, Cultured , Female , Humans , MELAS Syndrome/blood , MELAS Syndrome/genetics , Male , Middle Aged , Oxidative Phosphorylation , Point Mutation , Superoxide Dismutase/blood , Superoxide Dismutase/metabolism
18.
J Child Neurol ; 14(8): 518-23, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10456762

ABSTRACT

Cultured human skin fibroblasts from 12 patients with a variety of mitochondrial respiratory chain defects were examined for their capacity to oxidize dihydrorhodamine-123 to the fluorescent molecule rhodamine-123 using a flow cytometer. We found that cells from patients with functional defects in respiratory chain enzymes were less able to oxidize dihydrorhodamine-123 than those of healthy controls. Ten of the cell strains had reduced activity in at least one of the respiratory chain complexes and also showed significantly reduced fluorescence when compared to the mean of eight normal control cell strains. One patient had mitochondrial encephalomyopathy, lactic acidosis, and strokelike episodes (with the A3243G mutation) and reduced respiratory chain activities in muscle and liver. Molecular analysis did not show the mutation in cultured skin fibroblasts, and had correspondingly normal fluorescence. The 12th cell strain showed reduced fluorescence but did not reach statistical significance. This strategy could be of use in helping direct further investigations in patients, and in studying the biochemical pathogenesis of mitochondrial DNA mutations in cybrid studies.


Subject(s)
Electron Transport/physiology , Flow Cytometry , MELAS Syndrome/diagnosis , Mitochondria/enzymology , Mitochondrial Encephalomyopathies/diagnosis , Adenosine Triphosphate/metabolism , Child, Preschool , Consanguinity , DNA Mutational Analysis , DNA, Mitochondrial/genetics , Electron Transport/genetics , Female , Humans , Infant , Infant, Newborn , MELAS Syndrome/enzymology , MELAS Syndrome/genetics , Male , Mitochondria/genetics , Mitochondria, Liver/genetics , Mitochondria, Liver/physiology , Mitochondrial Encephalomyopathies/enzymology , Mitochondrial Encephalomyopathies/genetics , NAD(P)H Dehydrogenase (Quinone)/deficiency , NAD(P)H Dehydrogenase (Quinone)/genetics , Polymerase Chain Reaction , Reactive Oxygen Species/metabolism
19.
J Neurol Sci ; 163(2): 168-74, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10371079

ABSTRACT

Out of 90 Portuguese patients with mitochondrial cytopathy, six harbored the A3243G mutation in the mtDNA tRNA(Leu(UUR)) gene ('MELAS mutation'). They had heterogeneous clinical features, including myopathy with stroke-like episodes, progressive external ophthalmoparesis, diabetes mellitus, and subacute encephalopathy. Histochemical and biochemical analyses of muscle biopsies showed abundant ragged-red fibers reacting positively with the cytochrome oxidase stain, and decreased respiratory chain enzyme activities. On average, the proportion of mutated mtDNA was 67% (20-88%) in tissues from patients and 21% (0-49%) in blood from 20 maternal relatives. The proportion of mutated mitochondrial genomes in muscle did not correlate with clinical presentation or duration of disease. This study, the first in Portuguese patients, confirms the frequent occurrence of the A3243G mutation in patients with mitochondrial diseases, and emphasises the usefulness of genetic testing in reaching a correct diagnosis.


Subject(s)
DNA, Mitochondrial/genetics , MELAS Syndrome/genetics , Point Mutation , RNA, Transfer, Leu/genetics , Adolescent , Adult , Child , Electron Transport Complex IV/metabolism , Female , Humans , MELAS Syndrome/enzymology , MELAS Syndrome/pathology , Male , Middle Aged , Mitochondria, Muscle/genetics , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/pathology , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Oxygen Consumption , Pedigree , Portugal
20.
FEBS Lett ; 433(1-2): 93-7, 1998 Aug 14.
Article in English | MEDLINE | ID: mdl-9738940

ABSTRACT

Mutations in the human mtDNA gene encoding subunit III of cytochrome c oxidase (CO) have been reported to cause MELAS and LHON. Poracoccus denitrificans cells expressing substitutions homologous to these MELAS- and LHON-causing mutations had lower growth yield than wild type cells and lower efficiency of proton pumping by CO (e.g. lower H+/e ratio and lower deltapsi), but had similar CO activity. These results indicate that both substitutions (F263L > A212T) cause intrinsic uncoupling, which may be the direct cause of the diseases. These results also suggest that subunit III is involved in proton pumping.


Subject(s)
DNA, Mitochondrial/genetics , Electron Transport Complex IV/genetics , MELAS Syndrome/genetics , Optic Atrophies, Hereditary/genetics , Ascorbic Acid/pharmacology , Gene Deletion , Gene Expression , Humans , Kinetics , MELAS Syndrome/enzymology , Onium Compounds/metabolism , Optic Atrophies, Hereditary/enzymology , Organophosphorus Compounds/metabolism , Paracoccus denitrificans/enzymology , Paracoccus denitrificans/genetics , Paracoccus denitrificans/growth & development , Proton Pumps/metabolism , Tetramethylphenylenediamine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...