Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Hum Mol Genet ; 20(10): 1893-905, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21349918

ABSTRACT

Dominant optic atrophy (DOA) is genetically heterogeneous and pathogenic mutations have been identified in the OPA1 and OPA3 genes, both encoding for mitochondrial proteins. We characterized clinical and laboratory features in a large OPA1-negative family with complicated DOA. Search for mitochondrial dysfunction was performed by studying muscle biopsies, fibroblasts, platelets and magnetic resonance (MR) spectroscopy. Genetic investigations included mitochondrial DNA (mtDNA) analysis, linkage analysis, copy number variation (CNV) analysis and candidate gene screening. Optic neuropathy was undistinguishable from that in OPA1-DOA and frequently associated with late-onset sensorineural hearing loss, increases of central conduction times at somato-sensory evoked potentials and various cardiac abnormalities. Serum lactic acid after exercise, platelet respiratory complex activities, adenosine triphosphate (ATP) content in fibroblasts and muscle phosphorus MR spectroscopy all failed to reveal a mitochondrial dysfunction. However, muscle biopsies and their mtDNA analysis showed increased mitochondrial biogenesis. Furthermore, patient's fibroblasts grown in the galactose medium were unable to increase ATP content compared with controls, and exhibited abnormally high rate of fusion activity. Genome-wide linkage revealed a locus on chromosome 16q21-q22 with a maximum two-point LOD score of 8.84 for the marker D16S752 and a non-recombinant interval of ∼ 6.96 cM. Genomic screening of 45 genes in this interval including several likely candidate genes (CALB2, CYB5B, TK2, DHODH, PLEKHG4) revealed no mutation. Moreover, we excluded the presence of CNVs using array-based comparative genome hybridization. The identification of a new OPA locus (OPA8) in this pedigree demonstrates further genetic heterogeneity in DOA, and our results indicate that the pathogenesis may still involve mitochondria.


Subject(s)
Chromosomes, Human, Pair 16/genetics , Optic Atrophy, Autosomal Dominant/genetics , Adolescent , Adult , Child , Comparative Genomic Hybridization , DNA Copy Number Variations , DNA, Mitochondrial/genetics , Female , Genome-Wide Association Study , Haplotypes , Humans , Male , Microsatellite Repeats/genetics , Middle Aged , Mitochondria/metabolism , Mitochondria/pathology , Mitochondria/ultrastructure , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Pedigree , Polymorphism, Genetic/genetics , Retinal Vessels/pathology , Young Adult
3.
Ophthalmology ; 117(8): 1547-53, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20417568

ABSTRACT

PURPOSE: To analyze the influence of OPA1 gene mutations on the optic nerve head (ONH) morphology in patients with dominant optic atrophy (DOA). DESIGN: Cross-sectional study. PARTICIPANTS: Twenty-eight patients with DOA from 11 pedigrees, which were positive for the presence of OPA1 gene mutations, and 56 age-matched control subjects, were enrolled. METHODS: The ONH of DOA patients was studied by optical coherence tomography and compared with an age-matched control group of 56 individuals. MAIN OUTCOME MEASURES: ONH area, and vertical and horizontal diameters. RESULTS: The ONH analysis of DOA patients showed a significantly smaller optic disc area (P<0.0001), vertical (P = 0.018), and horizontal (P<0.0001) disc diameters, compared with controls. Stratification of the results for the single OPA1 mutation revealed normal ONH area with 2 mutations, whereas the only missense mutation linked to a "DOA plus" phenotype had the smallest ONH measurements. CONCLUSIONS: The DOA patients carrying OPA1 gene mutations present, as a group, a significantly smaller ONH compared with the range of size observed in a control population; this feature may be mutation specific. This observation suggests that OPA1 is involved in shaping the anatomic conformation of the ONH in patients with DOA. The relevance of OPA1 in regulating apoptosis and modeling the eye development has been recently shown by others. Thus, mutations in the OPA1 gene may determine the previously unrecognized feature of a smaller optic disc size and this in turn may have relevance for DOA pathogenesis. Furthermore, OPA1 gene polymorphic variants may contribute to the normal variability of ONH size in the general population.


Subject(s)
GTP Phosphohydrolases/genetics , Mutation , Optic Atrophy, Autosomal Dominant/genetics , Optic Disk/pathology , Adolescent , Adult , Aged , Child , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Optic Atrophy, Autosomal Dominant/diagnosis , Pedigree , Polymerase Chain Reaction , Tomography, Optical Coherence , Young Adult
4.
Brain ; 131(Pt 2): 352-67, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18222991

ABSTRACT

Dominant optic atrophy (DOA) is characterized by retinal ganglion cell degeneration leading to optic neuropathy. A subset of DOA is caused by mutations in the OPA1 gene, encoding for a dynamin-related GTPase required for mitochondrial fusion. The functional consequences of OPA1 mutations in DOA patients are still poorly understood. This study investigated the effect of five different OPA1 pathogenic mutations on the energetic efficiency and mitochondrial network dynamics of skin fibroblasts from patients. Although DOA fibroblasts maintained their ATP levels and grew in galactose medium, i.e. under forced oxidative metabolism, a significant impairment in mitochondrial ATP synthesis driven by complex I substrates was found. Furthermore, balloon-like structures in the mitochondrial reticulum were observed in galactose medium and mitochondrial fusion was completely inhibited in about 50% of DOA fibroblasts, but not in control cells. Respiratory complex assembly and the expression level of complex I subunits were similar in control and DOA fibroblasts. Co-immunoprecipitation experiments revealed that OPA1 directly interacts with subunits of complexes I, II and III, but not IV and with apoptosis inducing factor. The results disclose a novel link between OPA1, apoptosis inducing factor and the respiratory complexes that may shed some light on the pathogenic mechanism of DOA.


Subject(s)
GTP Phosphohydrolases/genetics , Mitochondria/metabolism , Optic Atrophy, Autosomal Dominant/genetics , Oxidative Phosphorylation , Adenosine Triphosphate/biosynthesis , Apoptosis/drug effects , Cells, Cultured , DNA Mutational Analysis/methods , DNA, Mitochondrial/analysis , Electron Transport , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/ultrastructure , GTP Phosphohydrolases/metabolism , Gene Deletion , Humans , Mitochondria/ultrastructure , Optic Atrophy, Autosomal Dominant/metabolism , Optic Atrophy, Autosomal Dominant/pathology , Oxidants/pharmacology , Skin/metabolism , Skin/ultrastructure
5.
Hum Mutat ; 29(1): 106-12, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17722006

ABSTRACT

Autosomal dominant optic atrophy (adOA) is most commonly caused by mutations in the OPA1 gene. There is a considerable allelic heterogeneity among adOA-associated OPA1 mutations, however these mutations have mostly been identified and studied only at the genomic DNA level. Here we report the identification of 22 novel OPA1 mutations and their analysis at the cDNA level along with 15 already known OPA1 mutations. We found that 18 of these mutations cause splice defects that involve either skipping of the adjacent exon or the activation of a cryptic splice site. We also observed a reduced level of the mutant transcript in several adOA subjects. Allele-specific quantification of the transcript steady-state level was performed for 13 different OPA1 mutations applying pyrosequencing to a RT-PCR amplified cSNP (c.2109C>T) in OPA1. Using this new assay we could demonstrate that the majority of OPA1 mutations that lead to a premature termination codon (PTC) undergo nonsense-mediated mRNA decay (NMD). Mutant transcript levels were reduced between 1.25- and 2.5-fold and varied between PTC containing mutations, and between subjects. Our results emphasize the value of cDNA analysis in the characterization of OPA1 mutations and further strengthen the model of haploinsufficiency as a major pathomechanism in OPA1-associated adOA.


Subject(s)
Codon, Nonsense/genetics , DNA, Complementary/metabolism , GTP Phosphohydrolases/genetics , Mutation , Optic Atrophy, Autosomal Dominant/genetics , Alleles , DNA Mutational Analysis/methods , DNA, Complementary/blood , DNA, Complementary/genetics , Genetic Variation , Humans , Optic Atrophy, Autosomal Dominant/blood
6.
Brain ; 130(Pt 4): 1029-42, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17314202

ABSTRACT

Autosomal dominant optic atrophy (adOA) is a juvenile onset, progressive ocular disorder characterized by bilateral loss of vision, central visual field defects, colour vision disturbances, and optic disc pallor. adOA is most frequently associated with mutations in OPA1 encoding a dynamin-related large GTPase that localizes to mitochondria. Histopathological studies in adOA patients have shown a degeneration of retinal ganglion cells (RGCs) and a loss of axons in the optic nerve. However little is known about the molecular mechanism and pathophysiology of adOA due to the lack of appropriate in vivo models. Here we report a first mouse model carrying a splice site mutation (c.1065 + 5G --> A) in the Opa1 gene. The mutation induces a skipping of exon 10 during transcript processing and leads to an in-frame deletion of 27 amino acid residues in the GTPase domain. Western blot analysis showed no evidence of a shortened mutant protein but a approximately 50% reduced OPA1 protein level supporting haploinsufficiency as a major disease mechanism in adOA. Homozygous mutant mice die in utero during embryogenesis with first notable developmental delay at E8.5 as detected by magnetic resonance imaging (MRI). Heterozygous mutants are viable and of normal habitus but exhibit an age-dependent loss of RGCs that eventually progresses to a severe degeneration of the ganglion cell and nerve fibre layer. In addition optic nerves of mutant mice showed a reduced number of axons, and a swelling and abnormal shape of the remaining axons. Mitochondria in these axons showed disorganized cristae structures. All these defects recapitulate crucial features of adOA in humans and therefore document the validity and importance of this model for future research.


Subject(s)
GTP Phosphohydrolases/genetics , Optic Atrophy, Autosomal Dominant/genetics , Amino Acids/genetics , Animals , Cells, Cultured , DNA, Circular/genetics , DNA, Mitochondrial/genetics , Disease Models, Animal , Electroretinography/methods , Exons/genetics , Hearing/genetics , Magnetic Resonance Imaging/methods , Mice , Mice, Inbred C3H , Microscopy, Electron, Transmission/methods , Mitochondria/genetics , Mutation/genetics , Optic Atrophy, Autosomal Dominant/pathology , Optic Nerve/pathology , RNA Splice Sites/genetics , Retina/pathology , Retinal Ganglion Cells/pathology , Sensory Thresholds/physiology , Transcription, Genetic/genetics
7.
Arch Neurol ; 68(1): 67-73, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20837821

ABSTRACT

OBJECTIVE: To assess whether impaired energy metabolism in skeletal muscle is a hallmark feature of patients with dominant optic atrophy due to several different mutations in the OPA1 gene. DESIGN: We used phosphorus 31 magnetic resonance spectroscopy to assess calf muscle oxidative metabolism in subjects with molecularly defined dominant optic atrophy carrying different mutations in the OPA1 gene. In a subset of patients, we also evaluated serum lactate levels after exercise and muscle biopsy results for histology and mitochondrial DNA analysis. SETTING: University neuromuscular and neurogenetics and magnetic resonance imaging units. PATIENTS: Eighteen patients with dominant optic atrophy were enrolled from 8 unrelated families, 7 of which carried an OPA1 mutation predicted to induce haploinsufficiency and 1 with a missense mutation in exon 27. Fifteen patients had documented optic atrophy. MAIN OUTCOME MEASURES: Presence of skeletal muscle mitochondrial oxidative phosphorylation dysfunction as assessed by phosphorus 31 magnetic resonance spectroscopy, serum lactate levels, and histological and mitochondrial DNA analysis. RESULTS: Phosphorus 31 magnetic resonance spectroscopy showed reduced phosphorylation potential in the calf muscle at rest in patients with an OPA1 mutation (-24% from normal mean; P = .003) as well as a reduced maximum rate of mitochondrial adenosine triphosphate synthesis (-36%; P < .001; ranging from -28% to -49% in association with different mutations). In 4 of 10 patients (40%), the serum lactate level after exercise was elevated. Only 2 of 5 muscle biopsies, from the 2 patients with a missense mutation, showed slight myopathic changes. Low levels of mitochondrial DNA multiple deletions were found in all muscle biopsies. CONCLUSIONS: Defective oxidative phosphorylation in skeletal muscle is a subclinical feature of patients with OPA1-related dominant optic atrophy, indicating a systemic expression of the OPA1 defect, similar to that previously reported for Leber hereditary optic neuropathy due to complex I dysfunction. This defect of oxidative phosphorylation does not appear to depend on the low amounts of mitochondrial DNA multiple deletions detected in muscle biopsies.


Subject(s)
Adenosine Triphosphate/genetics , DNA, Mitochondrial/genetics , GTP Phosphohydrolases/genetics , Muscle, Skeletal/pathology , Mutation/genetics , Optic Atrophy, Autosomal Dominant/genetics , Adenosine Triphosphate/biosynthesis , Adult , Aged , DNA, Mitochondrial/metabolism , Female , Humans , Male , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Mutation, Missense/genetics , Optic Atrophy, Autosomal Dominant/metabolism , Oxidative Phosphorylation , Young Adult
8.
Mol Neurodegener ; 5: 25, 2010 Jun 14.
Article in English | MEDLINE | ID: mdl-20546606

ABSTRACT

BACKGROUND: Up to the 1950s, there was an ongoing debate about the diversity of hereditary optic neuropathies, in particular as to whether all inherited optic atrophies can be ascribed to Leber's hereditary optic neuropathy (LHON) or represent different disease entities. In 1954 W. Jaeger published a detailed clinical and genealogical investigation of a large family with explicit autosomal dominant segregation of optic atrophy thus proving the existence of a discrete disease different from LHON, which is nowadays known as autosomal dominant optic atrophy (ADOA). Since the year 2000 ADOA is associated with genomic mutations in the OPA1 gene, which codes for a protein that is imported into mitochondria where it is required for mitochondrial fusion. Interestingly enough, the underlying mutation in this family has not been identified since then. RESULTS: We have reinvestigated this family with the aim to identify the mutation and to further clarify the underlying pathomechanism. Patients showed a classical non-syndromic ADOA. The long term deterioration in vision in the two teenagers examined 50 years later is of particular note 5/20 to 6/120. Multiplex ligation probe amplification revealed a duplication of the OPA1 exons 7-9 which was confirmed by long distance PCR and cDNA analysis, resulting in an in-frame duplication of 102 amino acids. Segregation was verified in 53 available members of the updated pedigree and a penetrance of 88% was calculated. Fibroblast cultures from skin biopsies were established to assess the mitochondrial network integrity and to qualitatively and quantitatively study the consequences of the mutation on transcript and protein level. Fibroblast cultures demonstrated a fragmented mitochondrial network. Processing of the OPA1 protein was altered. There was no correlation of the OPA1 transcript levels and the OPA1 protein levels in the fibroblasts. Intriguingly an overall decrease of mitochondrial proteins was observed in patients' fibroblasts, while the OPA1 transcript levels were elevated. CONCLUSIONS: The thorough study of this family provides a detailed clinical picture accompanied by a molecular investigation of patients' fibroblasts. Our data show a classic OPA1-associated non-syndromic ADOA segregating in this family. Cell biological findings suggest that OPA1 is regulated by post-translational mechanisms and we would like to hypothesize that loss of OPA1 function might lead to impaired mitochondrial quality control. With the clinical, genetic and cell biological characterisation of a family described already more than 50 years ago, we span more than half a century of research in optic neuropathies.

9.
Hum Genet ; 118(6): 767-71, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16323009

ABSTRACT

Mutations in OPA1 are the most frequent cause underlying autosomal dominant optic atrophy (adOA). Until now only few putative splicing mutations in the OPA1 gene have been investigated at the mRNA level and all these result in exon skipping. Here, we report the identification and cDNA analysis of four intronic and three exonic OPA1 gene mutations that cause a variety of splicing defects including activation of cryptic splice sites in either flanking exon or intron sequences, and a leaky splicing mutation. Our results show that cDNA analysis is of prime importance for the full evaluation of the effect of putative splicing mutations in the OPA1 gene.


Subject(s)
GTP Phosphohydrolases/genetics , RNA Splice Sites/genetics , Alternative Splicing , Animals , COS Cells , Chlorocebus aethiops , DNA Mutational Analysis , Exons , Humans , Introns , Mutation , Optic Atrophy, Autosomal Dominant/genetics , Transfection
10.
Exp Eye Res ; 83(3): 702-6, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16698014

ABSTRACT

Autosomal dominant optic atrophy (ADOA) is the most frequent hereditary optic neuropathy. Three loci have been reported for ADOA: a major locus, harboring all identified mutations to date, maps to 3q28 (OPA1), a second locus is linked to 18q12.2-q12.3 (OPA4) and a third locus on 22q12.1-q13.1 (OPA5) has been reported recently. We describe a six-generation Iranian family in which optic atrophy runs as an autosomal dominant trait with an age of onset at 14-15years. We performed linkage analysis with markers mapping to 3q28 and 18q12.2-q12.3 and found linkage to 3q28. Subsequent sequencing of OPA1 identified a novel heterozygous missense mutation (c.1313A>G) replacing aspartic acid by glycine (p.D438G) in the GTPase domain of OPA1. Interestingly, another missense mutation at the same position (c.1313A>T, D438V) has been reported before in two unrelated German families, indicating a possible mutation hot spot. Further evidence supporting the importance of D438 is its conservation from human to acoelomata. OPA1 is believed to be the human orthologue of yeast MGM1, a dynamin-related protein required for the integrity of mitochondrial DNA. Homology modeling of the OPA1 GTPase domain revealed extensive structural similarity to the Dictyostelium dynamin A GTPase domain and showed that D438 may interact with residues of the G1 and the G4 motifs, which are crucial in coordinating GTP. Based on this analysis, we propose a mechanism which explains the gradual decline of vision in ADOA patients with OPA1 mutations at position 438.


Subject(s)
GTP Phosphohydrolases/genetics , Mutation, Missense , Optic Atrophy, Autosomal Dominant/genetics , Adolescent , Animals , Humans , Models, Molecular , Protein Structure, Tertiary , Sequence Homology
11.
Ann Neurol ; 56(5): 719-23, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15505825

ABSTRACT

Dominant optic atrophy has been associated with mutations in the OPA1 gene, which encodes for a dynamin-related GTPase, a mitochondrial protein implicated in the formation and maintenance of mitochondrial network and morphology. We used phosphorus magnetic resonance spectroscopy to assess calf muscle oxidative metabolism in six patients from two unrelated families carrying the c.2708-2711delTTAG deletion in exon 27 of the OPA1 gene. The rate of postexercise phosphocreatine resynthesis, a measure of mitochondrial adenosine triphosphate production rate, was significantly delayed in the patients. Our in vivo results show for the first time to our knowledge a deficit of oxidative phosphorylation in OPA1-related DOA.


Subject(s)
Adenosine Triphosphate/deficiency , GTP Phosphohydrolases/genetics , Mitochondria, Muscle/metabolism , Optic Atrophy, Autosomal Dominant/genetics , Adolescent , Adult , Aged , DNA Mutational Analysis/methods , Exons/genetics , Family Health , Female , Humans , Magnetic Resonance Spectroscopy , Male , Middle Aged , Optic Atrophy, Autosomal Dominant/metabolism , Phosphocreatine/metabolism , Sequence Deletion , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL