Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Hum Mol Genet ; 29(22): 3631-3645, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33231680

ABSTRACT

OPA1 mutations are the major cause of dominant optic atrophy (DOA) and the syndromic form DOA plus, pathologies for which there is no established cure. We used a 'drug repurposing' approach to identify FDA-approved molecules able to rescue the mitochondrial dysfunctions induced by OPA1 mutations. We screened two different chemical libraries by using two yeast strains carrying the mgm1I322M and the chim3P646L mutations, identifying 26 drugs able to rescue their oxidative growth phenotype. Six of them, able to reduce the mitochondrial DNA instability in yeast, have been then tested in Opa1 deleted mouse embryonic fibroblasts expressing the human OPA1 isoform 1 bearing the R445H and D603H mutations. Some of these molecules were able to ameliorate the energetic functions and/or the mitochondrial network morphology, depending on the type of OPA1 mutation. The final validation has been performed in patients' fibroblasts, allowing to select the most effective molecules. Our current results are instrumental to rapidly translating the findings of this drug repurposing approach into clinical trial for DOA and other neurodegenerations caused by OPA1 mutations.


Subject(s)
Drug Repositioning , GTP Phosphohydrolases/genetics , Neurodegenerative Diseases/drug therapy , Optic Atrophy, Autosomal Dominant/drug therapy , Animals , DNA, Mitochondrial/drug effects , Fibroblasts/drug effects , GTP Phosphohydrolases/antagonists & inhibitors , Humans , Mice , Mitochondria/drug effects , Mitochondria/genetics , Mutation/drug effects , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Optic Atrophy, Autosomal Dominant/genetics , Optic Atrophy, Autosomal Dominant/pathology , Pedigree , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics
2.
Hum Mol Genet ; 29(8): 1319-1329, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32202296

ABSTRACT

Interpretation of variants of uncertain significance is an actual major challenge. We addressed this question on a set of OPA1 missense variants responsible for variable severity of neurological impairments. We used targeted metabolomics to explore the different signatures of OPA1 variants expressed in Opa1 deleted mouse embryonic fibroblasts (Opa1-/- MEFs), grown under selective conditions. Multivariate analyses of data discriminated Opa1+/+ from Opa1-/- MEFs metabolic signatures and classified OPA1 variants according to their in vitro severity. Indeed, the mild p.I382M hypomorphic variant was segregating close to the wild-type allele, while the most severe p.R445H variant was close to Opa1-/- MEFs, and the p.D603H and p.G439V alleles, responsible for isolated and syndromic presentations, respectively, were intermediary between the p.I382M and the p.R445H variants. The most discriminant metabolic features were hydroxyproline, the spermine/spermidine ratio, amino acid pool and several phospholipids, emphasizing proteostasis, endoplasmic reticulum (ER) stress and phospholipid remodeling as the main mechanisms ranking OPA1 allele impacts on metabolism. These results demonstrate the high resolving power of metabolomics in hierarchizing OPA1 missense mutations by their in vitro severity, fitting clinical expressivity. This suggests that our methodological approach can be used to discriminate the pathological significance of variants in genes responsible for other rare metabolic diseases and may be instrumental to select possible compounds eligible for supplementation treatment.


Subject(s)
Endoplasmic Reticulum Stress/genetics , GTP Phosphohydrolases/genetics , Metabolomics , Alleles , Animals , Fibroblasts/metabolism , Humans , Mice , Mutation, Missense/genetics , Phenotype , Proteostasis/genetics
3.
Biochim Biophys Acta Bioenerg ; 1859(4): 263-269, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29382469

ABSTRACT

OPA1 is a dynamin-related GTPase that controls mitochondrial dynamics, cristae integrity, energetics and mtDNA maintenance. The exceptional complexity of this protein is determined by the presence, in humans, of eight different isoforms that, in turn, are proteolytically cleaved into combinations of membrane-anchored long forms and soluble short forms. Recent advances highlight how each OPA1 isoform is able to fulfill "essential" mitochondrial functions, whereas only some variants carry out "specialized" features. Long forms determine fusion, long or short forms alone build cristae, whereas long and short forms together tune mitochondrial morphology. These findings offer novel challenging therapeutic potential to gene therapy.


Subject(s)
Alternative Splicing , GTP Phosphohydrolases/genetics , Mitochondria/enzymology , Mitochondrial Membranes/enzymology , Optic Atrophy, Autosomal Dominant/therapy , Animals , DNA, Mitochondrial/chemistry , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/therapeutic use , Gene Expression , Genetic Therapy/methods , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/therapeutic use , Mitochondria/ultrastructure , Mitochondrial Dynamics/genetics , Mitochondrial Membranes/ultrastructure , Optic Atrophy, Autosomal Dominant/enzymology , Optic Atrophy, Autosomal Dominant/genetics , Optic Atrophy, Autosomal Dominant/pathology , Oxidative Phosphorylation
4.
Biochim Biophys Acta Bioenerg ; 1859(3): 182-190, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29269267

ABSTRACT

A marked stimulation of complex II enzymatic activity was detected in cybrids bearing a homoplasmic MTCYB microdeletion causing disruption of both the activity and the assembly of complex III, but not in cybrids harbouring another MTCYB mutation affecting only the complex III activity. Moreover, complex II stimulation was associated with SDHA subunit tyrosine phosphorylation. Despite the lack of detectable hydrogen peroxide production, up-regulation of the levels of mitochondrial antioxidant defenses revealed a significant redox unbalance. This effect was also supported by the finding that treatment with N-acetylcysteine dampened the complex II stimulation, SDHA subunit tyrosine phosphorylation, and levels of antioxidant enzymes. In the absence of complex III, the cellular amount of succinate, but not fumarate, was markedly increased, indicating that enhanced activity of complex II is hampered due to the blockage of respiratory electron flow. Thus, we propose that complex II phosphorylation and stimulation of its activity represent a molecular mechanism triggered by perturbation of mitochondrial redox homeostasis due to severe dysfunction of respiratory complexes. Depending on the site and nature of the damage, complex II stimulation can either bypass the energetic deficit as an efficient compensatory mechanism, or be ineffectual, leaving cells to rely on glycolysis for survival.


Subject(s)
Electron Transport Complex III/metabolism , Electron Transport Complex II/metabolism , Homeostasis , Mitochondria/metabolism , Acetylcysteine/pharmacology , Cytochromes b/genetics , Cytochromes b/metabolism , Electron Transport/drug effects , Electron Transport Complex II/genetics , Electron Transport Complex III/genetics , Free Radical Scavengers/pharmacology , Humans , Hybrid Cells/metabolism , Hydrogen Peroxide/metabolism , Mitochondria/genetics , Mutation , Oxidation-Reduction , Phosphorylation/drug effects , Succinates/metabolism
5.
Pharmacol Res ; 131: 199-210, 2018 05.
Article in English | MEDLINE | ID: mdl-29454676

ABSTRACT

OPA1 is a GTPase that controls several functions, such as mitochondrial dynamics and energetics, mtDNA maintenance and cristae integrity. In the last years, there have been described other cellular pathways and mechanisms involving OPA1 directly or through its interaction. All this new information, by implementing our knowledge on OPA1 is instrumental to elucidating the pathogenic mechanisms of OPA1 mutations. Indeed, these are associated with dominant optic atrophy (DOA), one of the most common inherited optic neuropathies, and with an increasing number of heterogeneous neurodegenerative disorders. In this review, we overview all recent findings on OPA1 protein functions, on its dysfunction and related clinical phenotypes, focusing on the current therapeutic options and future perspectives to treat DOA and the other associated neurological disorders due to OPA1 mutations.


Subject(s)
GTP Phosphohydrolases/genetics , Mutation , Optic Atrophy, Autosomal Dominant/genetics , Animals , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , GTP Phosphohydrolases/metabolism , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Dynamics , Optic Atrophy, Autosomal Dominant/metabolism , Optic Atrophy, Autosomal Dominant/pathology , Optic Atrophy, Autosomal Dominant/therapy , Phenotype
6.
Hum Mol Genet ; 23(6): 1453-66, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24163135

ABSTRACT

Mitochondrial DNA mutations are currently investigated as modifying factors impinging on tumor growth and aggressiveness, having been found in virtually all cancer types and most commonly affecting genes encoding mitochondrial complex I (CI) subunits. However, it is still unclear whether they exert a pro- or anti-tumorigenic effect. We here analyzed the impact of three homoplasmic mtDNA mutations (m.3460G>A/MT-ND1, m.3571insC/MT-ND1 and m.3243A>G/MT-TL1) on osteosarcoma progression, chosen since they induce different degrees of oxidative phosphorylation impairment. In fact, the m.3460G>A/MT-ND1 mutation caused only a reduction in CI activity, whereas the m.3571insC/MT-ND1 and the m.3243A>G/MT-TL1 mutations induced a severe structural and functional CI alteration. As a consequence, this severe CI dysfunction determined an energetic defect associated with a compensatory increase in glycolytic metabolism and AMP-activated protein kinase activation. Osteosarcoma cells carrying such marked CI impairment displayed a reduced tumorigenic potential both in vitro and in vivo, when compared with cells with mild CI dysfunction, suggesting that mtDNA mutations may display diverse impact on tumorigenic potential depending on the type and severity of the resulting oxidative phosphorylation dysfunction. The modulation of tumor growth was independent from reactive oxygen species production but correlated with hypoxia-inducible factor 1α stabilization, indicating that structural and functional integrity of CI and oxidative phosphorylation are required for hypoxic adaptation and tumor progression.


Subject(s)
DNA, Mitochondrial/genetics , Electron Transport Complex I/genetics , Energy Metabolism , NADH Dehydrogenase/metabolism , Osteosarcoma/genetics , RNA, Transfer/genetics , AMP-Activated Protein Kinases/metabolism , Cell Line, Tumor , Disease Progression , Electron Transport Complex I/metabolism , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mutagenesis, Insertional , NADH Dehydrogenase/genetics , Osteosarcoma/pathology , Oxidative Phosphorylation , Point Mutation , Reactive Oxygen Species/metabolism
7.
Ann Neurol ; 78(1): 21-38, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25820230

ABSTRACT

OBJECTIVE: Mounting evidence links neurodegenerative disorders such as Parkinson disease and Alzheimer disease with mitochondrial dysfunction, and recent emphasis has focused on mitochondrial dynamics and quality control. Mitochondrial dynamics and mtDNA maintenance is another link recently emerged, implicating mutations in the mitochondrial fusion genes OPA1 and MFN2 in the pathogenesis of multisystem syndromes characterized by neurodegeneration and accumulation of mtDNA multiple deletions in postmitotic tissues. Here, we report 2 Italian families affected by dominant chronic progressive external ophthalmoplegia (CPEO) complicated by parkinsonism and dementia. METHODS: Patients were extensively studied by optical coherence tomography (OCT) to assess retinal nerve fibers, and underwent muscle and brain magnetic resonance spectroscopy (MRS), and muscle biopsy and fibroblasts were analyzed. Candidate genes were sequenced, and mtDNA was analyzed for rearrangements. RESULTS: Affected individuals displayed a slowly progressive syndrome characterized by CPEO, mitochondrial myopathy, sensorineural deafness, peripheral neuropathy, parkinsonism, and/or cognitive impairment, in most cases without visual complains, but with subclinical loss of retinal nerve fibers at OCT. Muscle biopsies showed cytochrome c oxidase-negative fibers and mtDNA multiple deletions, and MRS displayed defective oxidative metabolism in muscle and brain. We found 2 heterozygous OPA1 missense mutations affecting highly conserved amino acid positions (p.G488R, p.A495V) in the guanosine triphosphatase domain, each segregating with affected individuals. Fibroblast studies showed a reduced amount of OPA1 protein with normal mRNA expression, fragmented mitochondria, impaired bioenergetics, increased autophagy and mitophagy. INTERPRETATION: The association of CPEO and parkinsonism/dementia with subclinical optic neuropathy widens the phenotypic spectrum of OPA1 mutations, highlighting the association of defective mitochondrial dynamics, mtDNA multiple deletions, and altered mitophagy with parkinsonism.


Subject(s)
Dementia/genetics , GTP Phosphohydrolases/genetics , Mutation, Missense , Ophthalmoplegia, Chronic Progressive External/genetics , Parkinsonian Disorders/genetics , Aged , Dementia/complications , Female , Genetic Predisposition to Disease , Humans , Italy , Male , Ophthalmoplegia, Chronic Progressive External/complications , Parkinsonian Disorders/complications , Pedigree
8.
Hum Mol Genet ; 22(11): 2141-51, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23418307

ABSTRACT

Cytochrome b is the only mtDNA-encoded subunit of the mitochondrial complex III (CIII), the functional bottleneck of the respiratory chain. Previously, the human cytochrome b missense mutation m.15579A>G, which substitutes the Tyr 278 with Cys (p.278Y>C), was identified in a patient with severe exercise intolerance and multisystem manifestations. In this study, we characterized the biochemical properties of cybrids carrying this mutation and report that the homoplasmic p.278Y>C mutation caused a dramatic reduction in the CIII activity and in CIII-driven mitochondrial ATP synthesis. However, the CI, CI + CIII and CII + CIII activities and the rate of ATP synthesis driven by the CI or CII substrate were only partially reduced or unaffected. Consistent with these findings, mutated cybrids maintained the mitochondrial membrane potential in the presence of oligomycin, indicating that it originated from the respiratory electron transport chain. The p.278Y>C mutation enhanced superoxide production, as indicated by direct measurements in mitochondria and by the imbalance of glutathione homeostasis in intact cybrids. Remarkably, although the assembly of CI or CIII was not affected, the examination of respiratory supercomplexes revealed that the amounts of CIII dimer and III2IV1 were reduced, whereas those of I1III2IVn slightly increased. We therefore suggest that the deleterious effects of p.278Y>C mutation on cytochrome b are palliated when CIII is assembled into the supercomplexes I1III2IVn, in contrast to when it is found alone. These findings underline the importance of supramolecular interactions between complexes for maintaining a basal respiratory chain activity and shed light to the molecular basis of disease manifestations associated with this mutation.


Subject(s)
Cytochromes b/genetics , Electron Transport Complex IV/metabolism , Mutation , Superoxides/metabolism , Adenosine Triphosphate/biosynthesis , Cell Line , DNA, Mitochondrial/genetics , Electron Transport/genetics , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex IV/genetics , Energy Metabolism , Enzyme Activation , Glutathione/metabolism , Homeostasis/physiology , Humans , Mitochondria/genetics , Mitochondria/metabolism
9.
Hum Mutat ; 35(8): 954-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24863938

ABSTRACT

A novel heteroplasmic mitochondrial DNA (mtDNA) microdeletion affecting the cytochrome b gene (MT-CYB) was identified in an Italian female patient with a multisystem disease characterized by sensorineural deafness, cataracts, retinal pigmentary dystrophy, dysphagia, postural and gait instability, and myopathy with prominent exercise intolerance. The deletion is 18-base pair long and encompasses nucleotide positions 15,649-15,666, causing the loss of six amino acids (Ile-Leu-Ala-Met-Ile-Pro) in the protein, but leaving the remaining of the MT-CYB sequence in frame. The defective complex III function was cotransferred with mutant mtDNA in cybrids, thus unequivocally establishing its pathogenic role. Maternal relatives failed to show detectable levels of the deletion in blood and urinary epithelium, suggesting a de novo mutational event. This is the second report of an in-frame intragenic deletion in MT-CYB, which most likely occurred in early stages of embryonic development, associated with a severe multisystem disorder with prominent exercise intolerance.


Subject(s)
Base Sequence , Cytochromes b/genetics , Fatigue/genetics , Muscular Diseases/genetics , Sequence Deletion , Adult , Cataract/genetics , Cataract/pathology , DNA, Mitochondrial/genetics , Deglutition Disorders/genetics , Deglutition Disorders/pathology , Fatigue/pathology , Female , Gene Expression , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/pathology , Humans , Molecular Sequence Data , Muscular Diseases/pathology , Pigment Epithelium of Eye/pathology , Tooth Discoloration/genetics , Tooth Discoloration/pathology
10.
Biochim Biophys Acta ; 1827(11-12): 1332-9, 2013.
Article in English | MEDLINE | ID: mdl-23542447

ABSTRACT

In this mini review, we briefly survey the molecular processes that lead to reactive oxygen species (ROS) production by the respiratory complex III (CIII or cytochrome bc1). In particular, we discuss the "forward" and "reverse" electron transfer pathways that lead to superoxide generation at the quinol oxidation (Qo) site of CIII, and the components that affect these reactions. We then describe and compare the properties of a bacterial (Rhodobacter capsulatus) mutant enzyme producing ROS with its mitochondrial (human cybrids) counterpart associated with a disease. The mutation under study is located at a highly conserved tyrosine residue of cytochrome b (Y302C in R. capsulatus and Y278C in human mitochondria) that is at the heart of the quinol oxidation (Qo) site of CIII. Similarities of the major findings of bacterial and human mitochondrial cases, including decreased catalytic activity of CIII, enhanced ROS production and ensuing cellular responses and damages, are remarkable. This case illustrates the usefulness of undertaking parallel and complementary studies using biologically different yet evolutionarily related systems, such as α-proteobacteria and human mitochondria. It progresses our understanding of CIII mechanism of function and ROS production, and underlines the possible importance of supra-molecular organization of bacterial and mitochondrial respiratory chains (i.e., respirasomes) and their potential disease-associated protective roles. This article is part of a Special Issue entitled: Respiratory complex III and related bc complexes.


Subject(s)
Bacterial Proteins/metabolism , Electron Transport Complex III/metabolism , Mitochondrial Membranes/metabolism , Superoxides/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Electron Transport Complex III/chemistry , Electron Transport Complex III/genetics , Humans , Models, Molecular , Protein Conformation , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Rhodobacter capsulatus/genetics , Rhodobacter capsulatus/metabolism
11.
Genome Res ; 21(1): 12-20, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20974897

ABSTRACT

Eukaryotic cells harbor a small multiploid mitochondrial genome, organized in nucleoids spread within the mitochondrial network. Maintenance and distribution of mitochondrial DNA (mtDNA) are essential for energy metabolism, mitochondrial lineage in primordial germ cells, and to prevent mtDNA instability, which leads to many debilitating human diseases. Mounting evidence suggests that the actors of the mitochondrial network dynamics, among which is the intramitochondrial dynamin OPA1, might be involved in these processes. Here, using siRNAs specific to OPA1 alternate spliced exons, we evidenced that silencing of the OPA1 variants including exon 4b leads to mtDNA depletion, secondary to inhibition of mtDNA replication, and to marked alteration of mtDNA distribution in nucleoid and nucleoid distribution throughout the mitochondrial network. We demonstrate that a small hydrophobic 10-kDa peptide generated by cleavage of the OPA1-exon4b isoform is responsible for this process and show that this peptide is embedded in the inner membrane and colocalizes and coimmunoprecipitates with nucleoid components. We propose a novel synthetic model in which a peptide, including two trans-membrane domains derived from the N terminus of the OPA1-exon4b isoform in vertebrates or from its ortholog in lower eukaryotes, might contribute to nucleoid attachment to the inner mitochondrial membrane and promotes mtDNA replication and distribution. Thus, this study places OPA1 as a direct actor in the maintenance of mitochondrial genome integrity.


Subject(s)
DNA Replication/physiology , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , GTP Phosphohydrolases/metabolism , Genome, Mitochondrial , GTP Phosphohydrolases/genetics , Gene Silencing , Genome, Human , HeLa Cells , Hep G2 Cells , Humans , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
12.
Biochim Biophys Acta ; 1817(2): 363-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22086148

ABSTRACT

We have studied the effects of idebenone on mitochondrial function in cybrids derived from one normal donor (HQB17) and one patient harboring the G3460A/MT-ND1 mutation of Leber's Hereditary Optic Neuropathy (RJ206); and in XTC.UC1 cells bearing a premature stop codon at amino acid 101 of MT-ND1 that hampers complex I assembly. Addition of idebenone to HQB17 cells caused mitochondrial depolarization and NADH depletion, which were inhibited by cyclosporin (Cs) A and decylubiquinone, suggesting an involvement of the permeability transition pore (PTP). On the other hand, addition of dithiothreitol together with idebenone did not cause PTP opening and allowed maintenance of the mitochondrial membrane potential even in the presence of rotenone. Addition of dithiothreitol plus idebenone, or of idebenol, to HQB17, RJ206 and XTC.UC1 cells sustained membrane potential in intact cells and ATP synthesis in permeabilized cells even in the presence of rotenone and malonate, and restored a good level of coupled respiration in complex I-deficient XTC.UC1 cells. These findings demonstrate that idebenol can feed electrons at complex III. If the quinone is maintained in the reduced state, a task that in some cell types appears to be performed by dicoumarol-sensitive NAD(P)H:quinone oxidoreductase 1 [Haefeli et al. (2011) PLoS One 6, e17963], electron transfer to complex III may allow reoxidation of NADH in complex I deficiencies.


Subject(s)
Energy Metabolism/drug effects , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Ubiquinone/analogs & derivatives , Adenosine Triphosphate/metabolism , Animals , Antioxidants/pharmacology , Cell Respiration/drug effects , Cells, Cultured , Dithiothreitol/pharmacology , Drug Evaluation, Preclinical , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria, Liver/physiology , Oxygen Consumption/drug effects , Ubiquinone/pharmacology
13.
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
14.
Biochim Biophys Acta ; 1807(6): 633-42, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20732299

ABSTRACT

A prominent role for mitochondrial genes and metabolism has been recently characterized in oncocytic transformation of cancer cells. From mitochondrial ultrastructure alterations to respiratory complexes disruption and mutations within mitochondrial genes, oncocytic tumors present with a plethora of features that have helped understand the role that these organelles and their fundamental metabolic functions may play in cancer development. The history of this under-diagnosed subset of tumors and the bioenergetic implications of their mitochondrial derangement are discussed in this review along with the opportunities that oncocytic tumors offer to draw general conclusions on the involvement of mitochondria in cancer.


Subject(s)
Adenoma, Oxyphilic/etiology , Adenoma, Oxyphilic/pathology , Mitochondria/pathology , Neoplasms/etiology , Neoplasms/pathology , Adenoma, Oxyphilic/genetics , Adenoma, Oxyphilic/metabolism , Animals , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Efficiency/physiology , Gene Expression Profiling , Humans , Learning , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/physiology , Models, Biological , Neoplasms/genetics , Neoplasms/metabolism
15.
Hum Mol Genet ; 19(6): 1019-32, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20028790

ABSTRACT

We previously showed that disruptive complex I mutations in mitochondrial DNA are the main genetic hallmark of oncocytic tumors of the thyroid and kidney. We here report a high frequency of homoplasmic disruptive mutations in a large panel of oncocytic pituitary and head-and-neck tumors. The presence of such mutations implicates disassembly of respiratory complex I in vivo which in turn contributes to the inability of oncocytic tumors to stabilize HIF1alpha and to display pseudo-hypoxia. By utilizing transmitochondrial cytoplasmic hybrids (cybrids), we induced the shift to homoplasmy of a truncating mutation in the mitochondria-coded MTND1 gene. Such shift is associated with a profound metabolic impairment leading to the imbalance of alpha-ketoglutarate and succinate, the Krebs cycle metabolites which are the main responsible for HIF1alpha stabilization. We conclude that the main hallmarks of oncocytic transformation, namely the occurrence of homoplasmic disruptive mutations and complex I disassembly, may explain the benign nature of oncocytic neoplasms through lack of HIF1alpha stabilization.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Cell Hypoxia , Cell Respiration , DNA, Mitochondrial/genetics , Electron Transport Complex I/metabolism , Fumarate Hydratase/genetics , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Ketoglutaric Acids/metabolism , Mutation/genetics , NADH Dehydrogenase/genetics , Phenotype , Protein Biosynthesis , Protein Stability , RNA, Transfer/genetics , Reactive Oxygen Species/metabolism , Succinate Dehydrogenase/genetics , Succinic Acid/metabolism
17.
Life (Basel) ; 11(4)2021 Apr 17.
Article in English | MEDLINE | ID: mdl-33920624

ABSTRACT

The mitochondrial respiratory chain encompasses four oligomeric enzymatic complexes (complex I, II, III and IV) which, together with the redox carrier ubiquinone and cytochrome c, catalyze electron transport coupled to proton extrusion from the inner membrane. The protonmotive force is utilized by complex V for ATP synthesis in the process of oxidative phosphorylation. Respiratory complexes are known to coexist in the membrane as single functional entities and as supramolecular aggregates or supercomplexes (SCs). Understanding the assembly features of SCs has relevant biomedical implications because defects in a single protein can derange the overall SC organization and compromise the energetic function, causing severe mitochondrial disorders. Here we describe in detail the main types of SCs, all characterized by the presence of complex III. We show that the genetic alterations that hinder the assembly of Complex III, not just the activity, cause a rearrangement of the architecture of the SC that can help to preserve a minimal energetic function. Finally, the major metabolic disturbances associated with severe SCs perturbation due to defective complex III are discussed along with interventions that may circumvent these deficiencies.

18.
FEBS J ; 288(6): 1956-1974, 2021 03.
Article in English | MEDLINE | ID: mdl-32898935

ABSTRACT

Coenzyme Q10 (CoQ, ubiquinone) is a redox-active lipid endogenously synthesized by the cells. The final stage of CoQ biosynthesis is performed at the mitochondrial level by the 'complex Q', where coq2 is responsible for the prenylation of the benzoquinone ring of the molecule. We report that the competitive coq2 inhibitor 4-nitrobenzoate (4-NB) decreased the cellular CoQ content and caused severe impairment of mitochondrial function in the T67 human glioma cell line. In parallel with the reduction in CoQ biosynthesis, the cholesterol level increased, leading to significant perturbation of the plasma membrane physicochemical properties. We show that 4-NB treatment did not significantly affect the cell viability, because of an adaptive metabolic rewiring toward glycolysis. Hypoxia-inducible factor 1α (HIF-1α) stabilization was detected in 4-NB-treated cells, possibly due to the contribution of both reduction in intracellular oxygen tension and ROS overproduction. Exogenous CoQ supplementation partially recovered cholesterol content, HIF-1α degradation, and ROS production, whereas only weakly improved the bioenergetic impairment induced by the CoQ depletion. Our data provide new insights on the effect of CoQ depletion and contribute to shed light on the pathogenic mechanisms of ubiquinone deficiency syndrome.


Subject(s)
Energy Metabolism , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ubiquinone/analogs & derivatives , Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/metabolism , Ataxia/metabolism , Cell Line, Tumor , Cholesterol/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Muscle Weakness/metabolism , Nitrobenzoates/pharmacology , Protein Stability/drug effects , Ubiquinone/antagonists & inhibitors , Ubiquinone/biosynthesis , Ubiquinone/deficiency , Ubiquinone/metabolism
19.
Biochim Biophys Acta Bioenerg ; 1861(2): 148133, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31825807

ABSTRACT

The respiratory complexes are organized in supramolecular assemblies called supercomplexes thought to optimize cellular metabolism under physiological and pathological conditions. In this study, we used genetically and biochemically well characterized cells bearing the pathogenic microdeletion m.15,649-15,666 (ΔI300-P305) in MT-CYB gene, to investigate the effects of an assembly-hampered CIII on the re-organization of supercomplexes. First, we found that this mutation also affects the stability of both CI and CIV, and evidences the occurrence of a preferential structural interaction between CI and CIII2, yielding a small amount of active CI+CIII2 supercomplex. Indeed, a residual CI+CIII combined redox activity, and a low but detectable ATP synthesis driven by CI substrates are detectable, suggesting that the assembly of CIII into the CI+CIII2 supercomplex mitigates the detrimental effects of MT-CYB deletion. Second, measurements of oxygen consumption and ATP synthesis driven by NADH-linked and FADH2-linked substrates alone, or in combination, indicate a common ubiquinone pool for the two respiratory pathways. Finally, we report that prolonged incubation with rotenone enhances the amount of CI and CIII2, but reduces CIV assembly. Conversely, the antioxidant N-acetylcysteine increases CIII2 and CIV2 and partially restores respirasome formation. Accordingly, after NAC treatment, the rate of ATP synthesis increases by two-fold compared with untreated cell, while the succinate level, which is enhanced by the homoplasmic mutation, markedly decreases. Overall, our findings show that fine-tuning the supercomplexes stability improves the energetic efficiency of cells with the MT-CYB microdeletion.


Subject(s)
Adenosine Triphosphate/metabolism , Electron Transport Complex III/deficiency , Mitochondria/enzymology , Mitochondrial Membranes/enzymology , Oxygen Consumption , Animals , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Gene Deletion , Mitochondria/genetics , Oxidation-Reduction , Rotenone/pharmacology
20.
Hum Mutat ; 30(3): 391-6, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19086058

ABSTRACT

A disruptive frameshift mtDNA mutation affecting the ND5 subunit of complex I is present in homoplasmy in a nasopharyngeal oncocytic tumor and inherited as a heteroplasmic germline mutation recurring in two of the patient's siblings. Homoplasmic ND5 mutation in the tumor correlates with lack of the ND6 subunit, suggesting complex I disassembly. A few oncocytic areas, expressing ND6 and heteroplasmic for the ND5 mutation, harbor a de novo homoplasmic ND1 mutation. Since shift to homoplasmy of ND1 and ND5 mutations occurs exclusively in tumor cells, we conclude that complex I mutations may have a selective advantage and accompany oncocytic transformation.


Subject(s)
DNA, Mitochondrial/genetics , Electron Transport Complex I/genetics , Frameshift Mutation , Nasopharyngeal Neoplasms/genetics , Aged , Amino Acid Sequence , Base Sequence , Blotting, Western , DNA Mutational Analysis , DNA, Mitochondrial/chemistry , Electron Transport Complex I/metabolism , Germ-Line Mutation , Humans , Immunohistochemistry , Male , Microscopy, Electron , Molecular Sequence Data , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/ultrastructure , Pedigree , Sequence Deletion , Sequence Homology, Amino Acid , Siblings
SELECTION OF CITATIONS
SEARCH DETAIL