Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
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.
Molecules ; 27(4)2022 Feb 16.
Article in English | MEDLINE | ID: mdl-35209128

ABSTRACT

The finding that the most common mitochondrial DNA mutation m.11778G>A/MT-ND4 (p.R340H) associated with Leber's hereditary optic neuropathy (LHON) induces rotenone resistance has produced a long-standing debate, because it contrasts structural evidence showing that the ND4 subunit is far away from the quinone-reaction site in complex I, where rotenone acts. However, recent cryo-electron microscopy data revealed that rotenone also binds to the ND4 subunit. We investigated the possible structural modifications induced by the LHON mutation and found that its amino acid replacement would disrupt a possible hydrogen bond between native R340 and Q139 in ND4, thereby destabilizing rotenone binding. Our analysis thus explains rotenone resistance in LHON patients as a biochemical signature of its pathogenic effect on complex I.


Subject(s)
Alleles , Amino Acid Substitution , Drug Resistance/genetics , Electron Transport Complex I/genetics , Mutation , Optic Atrophy, Hereditary, Leber/genetics , Rotenone/pharmacology , Amino Acid Sequence , Binding Sites , Conserved Sequence , Electron Transport Complex I/chemistry , Electron Transport Complex I/metabolism , Models, Molecular , Optic Atrophy, Hereditary, Leber/metabolism , Protein Binding , Protein Conformation , Rotenone/chemistry , Structure-Activity Relationship , Uncoupling Agents/pharmacology
4.
FASEB J ; 34(6): 7675-7686, 2020 06.
Article in English | MEDLINE | ID: mdl-32304340

ABSTRACT

Mutations in mitochondrial transfer RNA (mt-tRNA) genes are responsible for a wide range of syndromes, for which no effective treatment is available. We previously reported that transfection of the nucleotide sequence encoding for the 16-residue ß32_33 peptide from mitochondrial leucyl-tRNA synthetase ameliorates the cell phenotype caused by the mitochondrial tRNA mutations. In this work, we demonstrated that both the ß32_33 peptide linked with the known (L)-Phe-(D)-Arg-(L)-Phe-(L)-Lys (FrFK) mitochondrial penetrating sequence and, strikingly, the ß32_33 peptide per se, are able to penetrate both the plasma and mitochondrial membranes and exert the rescuing activity when exogenously administered to cells bearing the mutations m.3243A > G and m.8344A > G. These mutations are responsible for the most common and severe mt-tRNA-related diseases. In addition, we dissected the molecular determinants of constructs activity by showing that both the order of amino acids along the sequence and presence of positive charges are essential determinants of the peptide activity in cells and mt-tRNA structures stabilization in vitro. In view of future in vivo studies, this information may be required to design of ß32_33 peptide-mimetic derivatives. The ß32_33 and FrFK-ß32_33 peptides are, therefore, promising molecules for the development of therapeutic agents against diseases caused by the mt-tRNA point mutations.


Subject(s)
Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Membranes/metabolism , Peptides/metabolism , RNA, Transfer/metabolism , Amino Acids/metabolism , Cell Line , Humans , Point Mutation/physiology
5.
Hum Mutat ; 39(1): 92-102, 2018 01.
Article in English | MEDLINE | ID: mdl-28967163

ABSTRACT

Respiratory complex III (CIII) is the first enzymatic bottleneck of the mitochondrial respiratory chain both in its native dimeric form and in supercomplexes. The mammalian CIII comprises 11 subunits among which cytochrome b is central in the catalytic core, where oxidation of ubiquinol occurs at the Qo site. The Qo- or PEWY-motif of cytochrome b is the most conserved through species. Importantly, the highly conserved glutamate at position 271 (Glu271) has never been studied in higher eukaryotes so far and its role in the Q-cycle remains debated. Here, we showed that the homoplasmic m.15557G > A/MT-CYB, which causes the p.Glu271Lys amino acid substitution predicted to dramatically affect CIII, induces a mild mitochondrial dysfunction in human transmitochondrial cybrids. Indeed, we found that the severity of such mutation is mitigated by the proper assembly of CIII into supercomplexes, which may favor an optimal substrate channeling and buffer superoxide production in vitro.


Subject(s)
Alleles , Cytochromes b/genetics , Genetic Association Studies , Mutation , Phenotype , Adenosine Triphosphate , Amino Acid Sequence , Amino Acid Substitution , Cell Line , Cell Survival/genetics , Conserved Sequence , Electron Transport Complex III/genetics , Electron Transport Complex III/metabolism , Energy Metabolism , Humans , Membrane Potential, Mitochondrial , Reactive Oxygen Species/metabolism
6.
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
7.
Neurobiol Dis ; 114: 129-139, 2018 06.
Article in English | MEDLINE | ID: mdl-29486301

ABSTRACT

There is growing evidence that the sequence variation of mitochondrial DNA (mtDNA), which clusters in population- and/or geographic-specific haplogroups, may result in functional effects that, in turn, become relevant in disease predisposition or protection, interaction with environmental factors and ultimately in modulating longevity. To unravel functional differences between mtDNA haplogroups we here employed transmitochondrial cytoplasmic hybrid cells (cybrids) grown in galactose medium, a culture condition that forces oxidative phosphorylation, and in the presence of rotenone, the classic inhibitor of respiratory Complex I. Under this experimental paradigm we assessed functional parameters such as cell viability and respiration, ATP synthesis, reactive oxygen species production and mtDNA copy number. Our analyses show that haplogroup J1, which is common in western Eurasian populations, is the most sensitive to rotenone, whereas K1 mitogenomes orchestrate the best compensation, possibly because of the haplogroup-specific missense variants impinging on Complex I function. Remarkably, haplogroups J1 and K1 fit the genetic associations previously established with Leber's hereditary optic neuropathy (LHON) for J1, as a penetrance enhancer, and with Parkinson's disease (PD) for K1, as a protective background. Our findings provide functional evidences supporting previous well-established genetic associations of specific haplogroups with two neurodegenerative pathologies, LHON and PD. Our experimental paradigm is instrumental to highlighting the subtle functional differences characterizing mtDNA haplogroups, which will be increasingly needed to dissect the role of mtDNA genetic variation in health, disease and longevity.


Subject(s)
DNA, Mitochondrial/genetics , Genome, Mitochondrial/genetics , Haplotypes/genetics , Parkinson Disease, Secondary/genetics , Pesticides/toxicity , Rotenone/toxicity , Cell Survival/drug effects , Cell Survival/genetics , DNA, Mitochondrial/chemistry , Fibroblasts/drug effects , Fibroblasts/physiology , Genome, Mitochondrial/drug effects , Haplotypes/drug effects , Humans , Oxygen Consumption/drug effects , Oxygen Consumption/physiology , Parkinson Disease, Secondary/chemically induced , Phylogeny , Protein Structure, Secondary
8.
Int J Mol Sci ; 19(3)2018 Mar 07.
Article in English | MEDLINE | ID: mdl-29518970

ABSTRACT

Mammalian respiratory complex I (CI) biogenesis requires both nuclear and mitochondria-encoded proteins and is mostly organized in respiratory supercomplexes. Among the CI proteins encoded by the mitochondrial DNA, NADH-ubiquinone oxidoreductase chain 1 (ND1) is a core subunit, evolutionary conserved from bacteria to mammals. Recently, ND1 has been recognized as a pivotal subunit in maintaining the structural and functional interaction among the hydrophilic and hydrophobic CI arms. A critical role of human ND1 both in CI biogenesis and in the dynamic organization of supercomplexes has been depicted, although the proof of concept is still missing and the critical amount of ND1 protein necessary for a proper assembly of both CI and supercomplexes is not defined. By exploiting a unique model in which human ND1 is allotopically re-expressed in cells lacking the endogenous protein, we demonstrated that the lack of this protein induces a stall in the multi-step process of CI biogenesis, as well as the alteration of supramolecular organization of respiratory complexes. We also defined a mutation threshold for the m.3571insC truncative mutation in mitochondrially encoded NADH:ubiquinone oxidoreductase core subunit 1 (MT-ND1), below which CI and its supramolecular organization is recovered, strengthening the notion that a certain amount of human ND1 is required for CI and supercomplexes biogenesis.


Subject(s)
Alleles , Electron Transport Complex I/chemistry , Electron Transport Complex I/genetics , Mutation , NADH Dehydrogenase/chemistry , NADH Dehydrogenase/genetics , Cell Respiration , DNA, Mitochondrial/genetics , Electron Transport Complex I/metabolism , Mitochondria/genetics , Mitochondria/metabolism , NADH Dehydrogenase/metabolism , Oxygen Consumption , Protein Binding , Structure-Activity Relationship
9.
Biochim Biophys Acta Bioenerg ; 1858(8): 582-590, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28213331

ABSTRACT

Energy homeostasis is pivotal for cell fate since metabolic regulation, cell proliferation and death are strongly dependent on the balance between catabolic and anabolic pathways. In particular, metabolic and energetic changes have been observed in cancer cells even before the discovery of oncogenes and tumor suppressors, but have been neglected for a long time. Instead, during the past 20years a renaissance of the study of tumor metabolism has led to a revised and more accurate sight of the metabolic landscape of cancer cells. In this scenario, genetic, biochemical and clinical evidences place mitochondria as key actors in cancer metabolic restructuring, not only because there are energy and biosynthetic intermediates manufacturers, but also because occurrence of mutations in metabolic enzymes encoded by both nuclear and mitochondrial DNA has been associated to different types of cancer. Here we provide an overview of the possible mechanisms modulating mitochondrial energy production and homeostasis in the intriguing scenario of neoplastic cells, focusing on the double-edged role of 5'-AMP activated protein kinase in cancer metabolism. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.


Subject(s)
Energy Metabolism , Mitochondria/metabolism , Neoplasms/metabolism , AMP-Activated Protein Kinases/physiology , Adenosine Triphosphate/metabolism , Animals , Disease Progression , Genes, Mitochondrial , Homeostasis , Humans , Mice , Mitochondrial Proteins/genetics , Mitochondrial Proteins/physiology , Models, Biological , Neoplasm Proteins/physiology , Neoplasms/pathology , Neoplasms, Experimental/metabolism , Tumor Suppressor Proteins/physiology
10.
Hum Mol Genet ; 24(24): 6921-31, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26410888

ABSTRACT

Leber's hereditary optic neuropathy (LHON) is a maternally inherited blinding disease characterized by degeneration of retinal ganglion cells (RGCs) and consequent optic nerve atrophy. Peculiar features of LHON are incomplete penetrance and gender bias, with a marked male prevalence. Based on the different hormonal metabolism between genders, we proposed that estrogens play a protective role in females and showed that these hormones ameliorate mitochondrial dysfunction in LHON through the estrogen receptors (ERs). We also showed that ERß localize to the mitochondria of RGCs. Thus, targeting ERß may become a therapeutic strategy for LHON specifically aimed at avoiding or delaying the onset of disease in mutation carriers. Here, we tested the effects of ERß targeting on LHON mitochondrial defective metabolism by treating LHON cybrid cells carrying the m.11778G>A mutation with a combination of natural estrogen-like compounds that bind ERß with high selectivity. We demonstrated that these molecules improve cell viability by reducing apoptosis, inducing mitochondrial biogenesis and strongly reducing the levels of reactive oxygen species in LHON cells. These effects were abolished in cells with ERß knockdown by silencing receptor expression or by using specific receptor antagonists. Our observations support the hypothesis that estrogen-like molecules may be useful in LHON prophylactic therapy. This is particularly important for lifelong disease prevention in unaffected LHON mutation carriers. Current strategies attempting to combat degeneration of RGCs during the acute phase of LHON have not been very effective. Implementing a different and preemptive approach with a low risk profile may be very helpful.


Subject(s)
Estrogen Receptor beta/antagonists & inhibitors , Optic Atrophy, Hereditary, Leber/prevention & control , Phytoestrogens/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Respiration , Cell Survival/drug effects , Cells, Cultured , Estrogen Receptor beta/genetics , Female , Gene Knockdown Techniques , Humans , Male , Mitochondria/drug effects , Mitochondria/metabolism , Mutation , Optic Atrophy, Hereditary, Leber/metabolism , Organelle Biogenesis , Oxidative Stress/drug effects , Oxygen Consumption , Retinal Ganglion Cells/metabolism
11.
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
12.
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
13.
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
14.
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
15.
Biochim Biophys Acta ; 1832(3): 445-52, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23246842

ABSTRACT

Complex I (CI) deficiency is a frequent cause of mitochondrial disorders and, in most cases, is due to mutations in CI subunit genes encoded by mitochondrial DNA (mtDNA). In this study, we establish the pathogenic role of the heteroplasmic mtDNA m.3890G>A/MT-ND1 (p.R195Q) mutation, which affects an extremely conserved amino acid position in ND1 subunit of CI. This mutation was found in a young-adult male with optic atrophy resembling Leber's hereditary optic neuropathy (LHON) and bilateral brainstem lesions. The only previously reported case with this mutation was a girl with fatal infantile Leigh syndrome with bilateral brainstem lesions. Transfer of the mutant mtDNA in the cybrid cell system resulted in a marked reduction of CI activity and CI-dependent ATP synthesis in the presence of a normally assembled enzyme. These findings establish the pathogenicity of the m.3890G>A/MT-ND1 mutation and remark the link between CI mutations affecting the mtDNA-encoded ND subunits and LHON-like optic atrophy, which may be complicated by bilateral and symmetric lesions affecting the central nervous system. Peculiar to this mutation is the distribution of the brainstem lesions, with sparing of the striatum in both patients.


Subject(s)
Brain Stem/metabolism , DNA, Mitochondrial/genetics , Mutation, Missense , NADH Dehydrogenase/genetics , Optic Atrophies, Hereditary/genetics , Adult , Amino Acid Sequence , Brain Stem/pathology , Cell Line, Tumor , Female , Humans , Hybrid Cells , Lactates/blood , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Molecular Sequence Data , Muscle, Skeletal/metabolism , NADH Dehydrogenase/metabolism , Optic Atrophies, Hereditary/blood , Optic Atrophies, Hereditary/metabolism , Pedigree , Sequence Homology, Amino Acid
16.
Biochim Biophys Acta Biomembr ; 1866(3): 184291, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38296218

ABSTRACT

Ionic liquids (ILs) are salts composed of a combination of organic or inorganic cations and anions characterized by a low melting point, often below 100 °C. This property, together with an extremely low vapor pressure, low flammability and high thermal stability, makes them suitable for replacing canonical organic solvents, with a reduction of industrial activities impact on the environment. Although in the last decades the eco-compatibility of ILs has been extensively verified through toxicological tests performed on model organisms, a detailed understanding of the interaction of these compounds with biological membranes is far from being exhaustive. In this context, we have chosen to evaluate the effect of some ILs on native membranes by using chromatophores, photosynthetic vesicles that can be isolated from Rhodobacter capsulatus, a member of the purple non­sulfur bacteria. Here, carotenoids associated with the light-harvesting complex II, act as endogenous spectral probes of the transmembrane electrical potential (ΔΨ). By measuring through time-resolved absorption spectroscopy the evolution of the carotenoid band shift induced by a single excitation of the photosynthetic reaction center, information on the ΔΨ dissipation due to ionic currents across the membrane can be obtained. We found that some ILs cause a rather fast dissipation of the transmembrane ΔΨ even at low concentrations, and that this behavior is dose-dependent. By using two different models to analyze the decay of the carotenoid signals, we attempted to interpret at a mechanistic level the marked increase of ionic permeability caused by specific ILs.


Subject(s)
Ionic Liquids , Ionic Liquids/pharmacology , Ionic Liquids/chemistry , Solvents/chemistry , Spectrum Analysis , Permeability , Carotenoids
17.
Int J Biol Macromol ; 273(Pt 2): 133086, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38871105

ABSTRACT

Variants found in the respiratory complex I (CI) subunit genes encoded by mitochondrial DNA can cause severe genetic diseases. However, it is difficult to establish a priori whether a single or a combination of CI variants may impact oxidative phosphorylation. Here we propose a computational approach based on coarse-grained molecular dynamics simulations aimed at investigating new CI variants. One of the primary CI variants associated with the Leber hereditary optic neuropathy (m.14484T>C/MT-ND6) was used as a test case and was investigated alone or in combination with two additional rare CI variants whose role remains uncertain. We found that the primary variant positioned in the E-channel region, which is fundamental for CI function, stiffens the enzyme dynamics. Moreover, a new mechanism for the transition between π- and α-conformation in the helix carrying the primary variant is proposed. This may have implications for the E-channel opening/closing mechanism. Finally, our findings show that one of the rare variants, located next to the primary one, further worsens the stiffening, while the other rare variant does not affect CI function. This approach may be extended to other variants candidate to exert a pathogenic impact on CI dynamics, or to investigate the interaction of multiple variants.


Subject(s)
Electron Transport Complex I , Molecular Dynamics Simulation , Mutation, Missense , Electron Transport Complex I/genetics , Electron Transport Complex I/chemistry , Electron Transport Complex I/metabolism , Humans , Optic Atrophy, Hereditary, Leber/genetics , Computational Biology/methods , NADH Dehydrogenase
18.
Cell Rep Med ; 5(2): 101383, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38272025

ABSTRACT

Idebenone, the only approved treatment for Leber hereditary optic neuropathy (LHON), promotes recovery of visual function in up to 50% of patients, but we can neither predict nor understand the non-responders. Idebenone is reduced by the cytosolic NAD(P)H oxidoreductase I (NQO1) and directly shuttles electrons to respiratory complex III, bypassing complex I affected in LHON. We show here that two polymorphic variants drastically reduce NQO1 protein levels when homozygous or compound heterozygous. This hampers idebenone reduction. In its oxidized form, idebenone inhibits complex I, decreasing respiratory function in cells. By retrospectively analyzing a large cohort of idebenone-treated LHON patients, classified by their response to therapy, we show that patients with homozygous or compound heterozygous NQO1 variants have the poorest therapy response, particularly if carrying the m.3460G>A/MT-ND1 LHON mutation. These results suggest consideration of patient NQO1 genotype and mitochondrial DNA mutation in the context of idebenone therapy.


Subject(s)
Optic Atrophy, Hereditary, Leber , Ubiquinone/analogs & derivatives , Humans , Optic Atrophy, Hereditary, Leber/drug therapy , Optic Atrophy, Hereditary, Leber/genetics , Optic Atrophy, Hereditary, Leber/metabolism , Antioxidants/therapeutic use , Antioxidants/pharmacology , Retrospective Studies , Ubiquinone/pharmacology , Ubiquinone/therapeutic use , Ubiquinone/metabolism , Electron Transport Complex I/genetics , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL