Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters











Publication year range
1.
Nature ; 599(7886): 650-656, 2021 11.
Article in English | MEDLINE | ID: mdl-34732887

ABSTRACT

Loss of functional mitochondrial complex I (MCI) in the dopaminergic neurons of the substantia nigra is a hallmark of Parkinson's disease1. Yet, whether this change contributes to Parkinson's disease pathogenesis is unclear2. Here we used intersectional genetics to disrupt the function of MCI in mouse dopaminergic neurons. Disruption of MCI induced a Warburg-like shift in metabolism that enabled neuronal survival, but triggered a progressive loss of the dopaminergic phenotype that was first evident in nigrostriatal axons. This axonal deficit was accompanied by motor learning and fine motor deficits, but not by clear levodopa-responsive parkinsonism-which emerged only after the later loss of dopamine release in the substantia nigra. Thus, MCI dysfunction alone is sufficient to cause progressive, human-like parkinsonism in which the loss of nigral dopamine release makes a critical contribution to motor dysfunction, contrary to the current Parkinson's disease paradigm3,4.


Subject(s)
Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Animals , Axons/drug effects , Axons/metabolism , Axons/pathology , Cell Death , Dendrites/metabolism , Dendrites/pathology , Disease Models, Animal , Disease Progression , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Female , Levodopa/pharmacology , Levodopa/therapeutic use , Male , Mice , Motor Skills/drug effects , NADH Dehydrogenase/deficiency , NADH Dehydrogenase/genetics , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/physiopathology , Phenotype , Substantia Nigra/cytology , Substantia Nigra/drug effects , Substantia Nigra/metabolism
2.
Cell Rep ; 35(3): 109002, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33882309

ABSTRACT

Complex I (CI) is the largest enzyme of the mitochondrial respiratory chain, and its defects are the main cause of mitochondrial disease. To understand the mechanisms regulating the extremely intricate biogenesis of this fundamental bioenergetic machine, we analyze the structural and functional consequences of the ablation of NDUFS3, a non-catalytic core subunit. We show that, in diverse mammalian cell types, a small amount of functional CI can still be detected in the complete absence of NDUFS3. In addition, we determine the dynamics of CI disassembly when the amount of NDUFS3 is gradually decreased. The process of degradation of the complex occurs in a hierarchical and modular fashion in which the ND4 module remains stable and bound to TMEM126A. We, thus, uncover the function of TMEM126A, the product of a disease gene causing recessive optic atrophy as a factor necessary for the correct assembly and function of CI.


Subject(s)
Electron Transport Complex I/genetics , Membrane Proteins/genetics , Mitochondria/genetics , NADH Dehydrogenase/genetics , Optic Atrophy/genetics , Animals , Binding Sites , CRISPR-Cas Systems , Cell Line, Tumor , Electron Transport Complex I/deficiency , Gene Editing , Gene Expression Regulation , Gene Knockout Techniques , HCT116 Cells , Humans , Melanocytes/metabolism , Melanocytes/pathology , Membrane Proteins/metabolism , Mice , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Membranes/chemistry , Mitochondrial Membranes/metabolism , Models, Molecular , NADH Dehydrogenase/deficiency , Optic Atrophy/metabolism , Optic Atrophy/pathology , Osteoblasts/metabolism , Osteoblasts/pathology , Protein Binding , Protein Conformation , Proteomics
3.
Oxid Med Cell Longev ; 2021: 5545261, 2021.
Article in English | MEDLINE | ID: mdl-33763166

ABSTRACT

Mitochondrial dysfunction has been suggested to be the key factor in the development and progression of cardiac hypertrophy. The onset of mitochondrial dysfunction and the mechanisms underlying the development of cardiac hypertrophy (CH) are incompletely understood. The present study is based on the use of multiple bioinformatics analyses for the organization and analysis of scRNA-seq and microarray datasets from a transverse aortic constriction (TAC) model to examine the potential role of mitochondrial dysfunction in the pathophysiology of CH. The results showed that NADH:ubiquinone oxidoreductase core subunit S1- (Ndufs1-) dependent mitochondrial dysfunction plays a key role in pressure overload-induced CH. Furthermore, in vivo animal studies using a TAC mouse model of CH showed that Ndufs1 expression was significantly downregulated in hypertrophic heart tissue compared to that in normal controls. In an in vitro model of angiotensin II- (Ang II-) induced cardiomyocyte hypertrophy, Ang II treatment significantly downregulated the expression of Ndufs1 in cardiomyocytes. In vitro mechanistic studies showed that Ndufs1 knockdown induced CH; decreased the mitochondrial DNA content, mitochondrial membrane potential (MMP), and mitochondrial mass; and increased the production of mitochondrial reactive oxygen species (ROS) in cardiomyocytes. On the other hand, Ang II treatment upregulated the expression levels of atrial natriuretic peptide, brain natriuretic peptide, and myosin heavy chain beta; decreased the mitochondrial DNA content, MMP, and mitochondrial mass; and increased mitochondrial ROS production in cardiomyocytes. The Ang II-mediated effects were significantly attenuated by overexpression of Ndufs1 in rat cardiomyocytes. In conclusion, our results demonstrate downregulation of Ndufs1 in hypertrophic heart tissue, and the results of mechanistic studies suggest that Ndufs1 deficiency may cause mitochondrial dysfunction in cardiomyocytes, which may be associated with the development and progression of CH.


Subject(s)
Cardiomegaly/metabolism , Membrane Potential, Mitochondrial , Myocardium/metabolism , Myocardium/pathology , NADH Dehydrogenase/deficiency , Pressure , Angiotensin II , Animals , Atrial Natriuretic Factor/metabolism , Biomarkers/metabolism , Cardiomegaly/pathology , Constriction, Pathologic , Down-Regulation , Male , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/metabolism , NADH Dehydrogenase/metabolism , Natriuretic Peptide, Brain/metabolism , RNA-Seq , Rats , Single-Cell Analysis
4.
Clin Chem Lab Med ; 58(11): 1809-1817, 2020 10 25.
Article in English | MEDLINE | ID: mdl-32432562

ABSTRACT

Objectives Leigh syndrome (LS) is one of the most common mitochondrial diseases and has variable clinical symptoms. However, the genetic variant spectrum of this disease is incomplete. Methods Next-generation sequencing (NGS) was used to identify the m.14430A > G (p.W82R) variant in a patient with LS. The pathogenesis of this novel complex I (CI) variant was verified by determining the mitochondrial respiration, assembly of CI, ATP, MMP and lactate production, and cell growth rate in cybrids with and without this variant. Results A novel m.14430A > G (p.W82R) variant in the NADH dehydrogenase 6 (ND6) gene was identified in the patient; the mutant loads of m.14430A > G (p.W82R) in the patient were much higher than those in his mother. Although the transmitochondrial cybrid-based study showed that mitochondrial CI assembly remains unaffected in cells with the m.14430G variant, control cells had significantly higher endogenous and CI-dependent mitochondrial respiration than mutant cells. Accordingly, mutant cells had a lower ATP, MMP and higher extracellular lactate production than control cells. Notably, mutant cells had impaired growth in a galactose-containing medium when compared to wild-type cells. Conclusions A novel m.14430A > G (p.W82R) variant in the ND6 gene was identified from a patient suspected to have LS, and this variant impaired mitochondrial respiration by decreasing the activity of mitochondrial CI.


Subject(s)
Leigh Disease/genetics , NADH Dehydrogenase/deficiency , NADH Dehydrogenase/genetics , Cell Line, Tumor , Child, Preschool , High-Throughput Nucleotide Sequencing , Humans , Leigh Disease/enzymology , Male , Mutation, Missense
5.
Sci Rep ; 8(1): 1165, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29348607

ABSTRACT

Mitochondrial complex I (CI) deficiency is the most frequent cause of oxidative phosphorylation (OXPHOS) disorders in humans. In order to benchmark the effects of CI deficiency on mitochondrial bioenergetics and dynamics, respiratory chain (RC) and endoplasmic reticulum (ER)-mitochondria communication, and superoxide production, fibroblasts from patients with mutations in the ND6, NDUFV1 or ACAD9 genes were analyzed. Fatty acid metabolism, basal and maximal respiration, mitochondrial membrane potential, and ATP levels were decreased. Changes in proteins involved in mitochondrial dynamics were detected in various combinations in each cell line, while variable changes in RC components were observed. ACAD9 deficient cells exhibited an increase in RC complex subunits and DDIT3, an ER stress marker. The level of proteins involved in ER-mitochondria communication was decreased in ND6 and ACAD9 deficient cells. |ΔΨ| and cell viability were further decreased in all cell lines. These findings suggest that disruption of mitochondrial bioenergetics and dynamics, ER-mitochondria crosstalk, and increased superoxide contribute to the pathophysiology in patients with ACAD9 deficiency. Furthermore, treatment of ACAD9 deficient cells with JP4-039, a novel mitochondria-targeted reactive oxygen species, electron and radical scavenger, decreased superoxide level and increased basal and maximal respiratory rate, identifying a potential therapeutic intervention opportunity in CI deficiency.


Subject(s)
Acyl-CoA Dehydrogenases/genetics , Electron Transport Complex I/deficiency , Fibroblasts/enzymology , Mitochondrial Diseases/genetics , NADH Dehydrogenase/genetics , Reactive Oxygen Species/metabolism , Acyl-CoA Dehydrogenases/deficiency , Adenosine Triphosphate/agonists , Adenosine Triphosphate/biosynthesis , Electron Transport/drug effects , Electron Transport/genetics , Electron Transport Complex I/genetics , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Fibroblasts/drug effects , Fibroblasts/pathology , Free Radical Scavengers/pharmacology , Gene Expression , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/pathology , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/pathology , NADH Dehydrogenase/deficiency , Nitrogen Oxides/pharmacology , Oxidative Phosphorylation/drug effects , Primary Cell Culture , Reactive Oxygen Species/antagonists & inhibitors
6.
Glia ; 66(4): 874-888, 2018 04.
Article in English | MEDLINE | ID: mdl-29285794

ABSTRACT

Mitochondrial defects associated with respiratory chain complex I deficiency lead to heterogeneous fatal syndromes. While the role of NDUFS8, an essential subunit of the core assembly of the complex I, is established in mitochondrial diseases, the mechanisms underlying neuropathology are poorly understood. We developed a Drosophila model of NDUFS8 deficiency by knocking down the expression of its fly homologue in neurons or in glial cells. Downregulating ND23 in neurons resulted in shortened lifespan, and decreased locomotion. Although total brain ATP levels were decreased, histological analysis did not reveal any signs of neurodegeneration except for photoreceptors of the retina. Interestingly, ND23 deficiency-associated phenotypes were rescued by overexpressing the glucose transporter hGluT3 demonstrating that boosting glucose metabolism in neurons was sufficient to bypass altered mitochondrial functions and to confer neuroprotection. We then analyzed the consequences of ND23 knockdown in glial cells. In contrast to neuronal knockdown, loss of ND23 in glia did not lead to significant behavioral defects nor to reduced lifespan, but induced brain degeneration, as visualized by numerous vacuoles found all over the nervous tissue. This phenotype was accompanied by the massive accumulation of lipid droplets at the cortex-neuropile boundaries, suggesting an alteration of lipid metabolism in glia. These results demonstrate that complex I deficiency triggers metabolic alterations both in neurons and glial cells which may contribute to the neuropathology.


Subject(s)
Drosophila Proteins/deficiency , Lipid Metabolism/physiology , Mitochondrial Diseases/pathology , NADH Dehydrogenase/deficiency , Neurodegenerative Diseases/pathology , Neuroglia/pathology , Adenosine Triphosphate/metabolism , Animals , Animals, Genetically Modified , Brain/metabolism , Brain/pathology , Disease Models, Animal , Drosophila , Drosophila Proteins/genetics , Female , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Homeostasis/physiology , Humans , Mitochondrial Diseases/metabolism , Motor Activity/physiology , NADH Dehydrogenase/genetics , Neurodegenerative Diseases/metabolism , Neuroglia/metabolism , Neurons/metabolism , Neurons/pathology , Photoreceptor Cells, Invertebrate/metabolism , Photoreceptor Cells, Invertebrate/pathology , RNA Interference , RNA, Messenger/metabolism
7.
Molecules ; 22(9)2017 Sep 11.
Article in English | MEDLINE | ID: mdl-28891993

ABSTRACT

The chloroplast genome (CPG) of Pinus massoniana belonging to the genus Pinus (Pinaceae), which is a primary source of turpentine, was sequenced and analyzed in terms of gene rearrangements, ndh genes loss, and the contraction and expansion of short inverted repeats (IRs). P. massoniana CPG has a typical quadripartite structure that includes large single copy (LSC) (65,563 bp), small single copy (SSC) (53,230 bp) and two IRs (IRa and IRb, 485 bp). The 108 unique genes were identified, including 73 protein-coding genes, 31 tRNAs, and 4 rRNAs. Most of the 81 simple sequence repeats (SSRs) identified in CPG were mononucleotides motifs of A/T types and located in non-coding regions. Comparisons with related species revealed an inversion (21,556 bp) in the LSC region; P. massoniana CPG lacks all 11 intact ndh genes (four ndh genes lost completely; the five remained truncated as pseudogenes; and the other two ndh genes remain as pseudogenes because of short insertions or deletions). A pair of short IRs was found instead of large IRs, and size variations among pine species were observed, which resulted from short insertions or deletions and non-synchronized variations between "IRa" and "IRb". The results of phylogenetic analyses based on whole CPG sequences of 16 conifers indicated that the whole CPG sequences could be used as a powerful tool in phylogenetic analyses.


Subject(s)
Gene Deletion , Gene Rearrangement , Genes, Plant , Genome, Chloroplast , NADH Dehydrogenase/genetics , Pinus/genetics , Chromosome Mapping , Gene Ontology , Inverted Repeat Sequences , Isoenzymes/deficiency , Isoenzymes/genetics , Molecular Sequence Annotation , NADH Dehydrogenase/deficiency , Phylogeny , Pinus/classification , Whole Genome Sequencing
8.
Stem Cell Res Ther ; 8(1): 150, 2017 06 24.
Article in English | MEDLINE | ID: mdl-28646906

ABSTRACT

BACKGROUND: Disorders of the oxidative phosphorylation (OXPHOS) system represent a large group among the inborn errors of metabolism. The most frequently observed biochemical defect is isolated deficiency of mitochondrial complex I (CI). No effective treatment strategies for CI deficiency are so far available. The purpose of this study was to investigate whether and how mesenchymal stem cells (MSCs) are able to modulate metabolic function in fibroblast cell models of CI deficiency. METHODS: We used human and murine fibroblasts with a defect in the nuclear DNA encoded NDUFS4 subunit of CI. Fibroblasts were co-cultured with MSCs under different stress conditions and intercellular mitochondrial transfer was assessed by flow cytometry and fluorescence microscopy. Reactive oxygen species (ROS) levels were measured using MitoSOX-Red. Protein levels of CI were analysed by blue native polyacrylamide gel electrophoresis (BN-PAGE). RESULTS: Direct cellular interactions and mitochondrial transfer between MSCs and human as well as mouse fibroblast cell lines were demonstrated. Mitochondrial transfer was visible in 13.2% and 6% of fibroblasts (e.g. fibroblasts containing MSC mitochondria) for human and mouse cell lines, respectively. The transfer rate could be further stimulated via treatment of cells with TNF-α. MSCs effectively lowered cellular ROS production in NDUFS4-deficient fibroblast cell lines (either directly via co-culture or indirectly via incubation of cell lines with cell-free MSC supernatant). However, CI protein expression and activity were not rescued by MSC treatment. CONCLUSION: This study demonstrates the interplay between MSCs and fibroblast cell models of isolated CI deficiency including transfer of mitochondria as well as modulation of cellular ROS levels. Further exploration of these cellular interactions might help to develop MSC-based treatment strategies for human CI deficiency.


Subject(s)
Electron Transport Complex I , Fibroblasts/enzymology , Mesenchymal Stem Cells/enzymology , Mitochondria/enzymology , NADH Dehydrogenase , Oxidative Phosphorylation , Animals , Coculture Techniques , Electron Transport Complex I/deficiency , Electron Transport Complex I/metabolism , Fibroblasts/cytology , Humans , Mesenchymal Stem Cells/cytology , Mice , Mitochondria/genetics , NADH Dehydrogenase/deficiency , NADH Dehydrogenase/metabolism
9.
Mitochondrion ; 28: 73-8, 2016 May.
Article in English | MEDLINE | ID: mdl-27079373

ABSTRACT

The genetic causes of Leigh syndrome are heterogeneous, with a poor correlation between the phenotype and genotype. Here, we present a patient with an NDUFS4 mutation to expand the clinical and biochemical spectrum of the disease. A combined defect in the CoQ, PDH and RCC activities in our patient was due to an inappropriate assembly of the RCC complex I (CI), which was confirmed using Blue-Native polyacrylamide gel electrophoresis (BN-PAGE) analysis. Targeted exome sequencing analysis allowed for the genetic diagnosis of this patient. We reviewed 198 patients with 24 different genetic defects causing RCC I deficiency and compared them to 22 NDUFS4 patients. We concluded that NDUFS4-related Leigh syndrome is invariably linked to an early onset severe phenotype that results in early death. Some data, including the clinical phenotype, neuroimaging and biochemical findings, can guide the genetic study in patients with RCC I deficiency.


Subject(s)
Leigh Disease/diagnosis , NADH Dehydrogenase/deficiency , Electron Transport Complex I , Electrophoresis, Polyacrylamide Gel , Female , Humans , Infant, Newborn , Ubiquinone/deficiency
10.
J Exp Bot ; 67(10): 3079-93, 2016 05.
Article in English | MEDLINE | ID: mdl-27122571

ABSTRACT

The mitochondrial NADH dehydrogenase complex (complex I) is of particular importance for the respiratory chain in mitochondria. It is the major electron entry site for the mitochondrial electron transport chain (mETC) and therefore of great significance for mitochondrial ATP generation. We recently described an Arabidopsis thaliana double-mutant lacking the genes encoding the carbonic anhydrases CA1 and CA2, which both form part of a plant-specific 'carbonic anhydrase domain' of mitochondrial complex I. The mutant lacks complex I completely. Here we report extended analyses for systematically characterizing the proteome of the ca1ca2 mutant. Using various proteomic tools, we show that lack of complex I causes reorganization of the cellular respiration system. Reduced electron entry into the respiratory chain at the first segment of the mETC leads to induction of complexes II and IV as well as alternative oxidase. Increased electron entry at later segments of the mETC requires an increase in oxidation of organic substrates. This is reflected by higher abundance of proteins involved in glycolysis, the tricarboxylic acid cycle and branched-chain amino acid catabolism. Proteins involved in the light reaction of photosynthesis, the Calvin cycle, tetrapyrrole biosynthesis, and photorespiration are clearly reduced, contributing to the significant delay in growth and development of the double-mutant. Finally, enzymes involved in defense against reactive oxygen species and stress symptoms are much induced. These together with previously reported insights into the function of plant complex I, which were obtained by analysing other complex I mutants, are integrated in order to comprehensively describe 'life without complex I'.


Subject(s)
Arabidopsis/metabolism , Mitochondria/enzymology , NADH Dehydrogenase/deficiency , Arabidopsis/cytology , Arabidopsis/genetics , Arabidopsis Proteins/metabolism , Cell Respiration/physiology , Electrophoresis, Polyacrylamide Gel , Mass Spectrometry , Mitochondria/metabolism , Mutation , NADH Dehydrogenase/metabolism , Oxygen Consumption , Peroxisomes/metabolism , Plastids/metabolism , Proteome
11.
Article in English | MEDLINE | ID: mdl-24963768

ABSTRACT

Mitochondrial energy metabolism disorder is one of the important reasons of leukodystrophy. Mutations of mitochondrial complex I genes have been implicated in more common neurological disorders such as Leigh syndrome. We describe a case of a child manifested as regression of mental and motor development, aggravated obviously after suffering infection. Physical and auxiliary examinations demonstrated that a series of changes including white matter lesions of magnetic resonance imaging, peripheral neuropathy with high muscle tension and hyperreflexia of limbs pointed to the diagnosis of leukodystrophy, with what can't explain the high levels of lactate and creatine kinase. Spontaneously, genetic analysis covered known leukodystrophy and mitochondrial genes were adapted for this child and his parents. Results showed the child was compound heterozygous mutation (c.278A > G; c.247G > A) within exon 2 in the NDUFAF1 gene, his parents carried a heterozygous mutation each. The authors report a case of leukodystrophy associated with mitochondrial complex I deficiency due to a novel mutation in the NDUFAF1 gene. This is the first report that NDUFAF1 mutations cause leukodystrophy.


Subject(s)
Brain Diseases, Metabolic, Inborn/genetics , Exons , Heterozygote , Mitochondrial Proteins/deficiency , NADH Dehydrogenase/deficiency , Point Mutation , Brain Diseases, Metabolic, Inborn/diagnosis , Brain Diseases, Metabolic, Inborn/enzymology , Brain Diseases, Metabolic, Inborn/pathology , Humans , Infant , Male
12.
Microbiology (Reading) ; 161(11): 2220-31, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26377309

ABSTRACT

YajL is the closest prokaryotic homologue of Parkinson's disease-associated DJ-1, a protein of undefined function involved in the oxidative stress response. We reported recently that YajL and DJ-1 protect cells against oxidative stress-induced protein aggregation by acting as covalent chaperones for the thiol proteome, including the NuoG subunit of NADH dehydrogenase 1, and that NADH dehydrogenase 1 activity is negligible in the yajL mutant. We report here that this mutant compensates for low NADH dehydrogenase activity by utilizing NADH-independent alternative dehydrogenases, including pyruvate oxidase PoxB and d-amino acid dehydrogenase DadA, and mixed acid aerobic fermentations characterized by acetate, lactate, succinate and ethanol excretion. The yajL mutant has a low adenylate energy charge favouring glycolytic flux, and a high NADH/NAD ratio favouring fermentations over pyruvate dehydrogenase and the Krebs cycle. DNA array analysis showed upregulation of genes involved in glycolytic and pentose phosphate pathways and alternative respiratory pathways. Moreover, the yajL mutant preferentially catabolized pyruvate-forming amino acids over Krebs cycle-related amino acids, and thus the yajL mutant utilizes pyruvate-centred respiro-fermentative metabolism to compensate for the NADH dehydrogenase 1 defect and constitutes an interesting model for studying eukaryotic respiratory complex I deficiencies, especially those associated with Alzheimer's and Parkinson's diseases.


Subject(s)
Escherichia coli/enzymology , Escherichia coli/metabolism , NADH Dehydrogenase/deficiency , Ribosomal Proteins/deficiency , Aerobiosis , Escherichia coli Proteins , Fermentation , Gene Expression Profiling , Humans , Metabolic Flux Analysis , Microarray Analysis , Models, Biological , Molecular Sequence Data , Parkinsonian Disorders/pathology , Sequence Analysis, DNA
13.
FEBS Lett ; 588(23): 4520-5, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25447539

ABSTRACT

We investigated H(+) and Na(+) transport by complex I from Escherichia coli devoid of the NuoL subunit, which is probably part of the ion translocating machinery. We observed that complex I devoid of the NuoL subunit still translocates H(+), although to a smaller extension than the complete version of complex I, but does not transport Na(+). Our results unequivocally reinforce the observation that E. coli complex I transports Na(+) in the opposite direction to that of the H+ and show that NuoL subunit is involved in the translocation of both ions by complex I.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , NADH Dehydrogenase/deficiency , NADH Dehydrogenase/metabolism , Protein Subunits/deficiency , Protein Subunits/metabolism , Sodium/metabolism , Biological Transport , Cell Membrane/metabolism , Escherichia coli/cytology , Escherichia coli Proteins/chemistry , Models, Molecular , NADH Dehydrogenase/chemistry , Protein Conformation , Protein Subunits/chemistry , Protons
14.
PLoS One ; 8(7): e68591, 2013.
Article in English | MEDLINE | ID: mdl-23861923

ABSTRACT

The re-colonization of aquatic habitats by angiosperms has presented a difficult challenge to plants whose long evolutionary history primarily reflects adaptations to terrestrial conditions. Many aquatics must complete vital stages of their life cycle on the water surface by means of floating or emergent leaves and flowers. Only a few species, mainly within the order Alismatales, are able to complete all aspects of their life cycle including pollination, entirely underwater. Water-pollinated Alismatales include seagrasses and water nymphs (Najas), the latter being the only freshwater genus in the family Hydrocharitaceae with subsurface water-pollination. We have determined the complete nucleotide sequence of the plastid genome of Najas flexilis. The plastid genome of N. flexilis is a circular AT-rich DNA molecule of 156 kb, which displays a quadripartite structure with two inverted repeats (IR) separating the large single copy (LSC) from the small single copy (SSC) regions. In N. flexilis, as in other Alismatales, the rps19 and trnH genes are localized in the LSC region instead of within the IR regions as in other monocots. However, the N. flexilis plastid genome presents some anomalous modifications. The size of the SSC region is only one third of that reported for closely related species. The number of genes in the plastid is considerably less. Both features are due to loss of the eleven ndh genes in the Najas flexilis plastid. In angiosperms, the absence of ndh genes has been related mainly to the loss of photosynthetic function in parasitic plants. The ndh genes encode the NAD(P)H dehydrogenase complex, believed essential in terrestrial environments, where it increases photosynthetic efficiency in variable light intensities. The modified structure of the N. flexilis plastid genome suggests that adaptation to submersed environments, where light is scarce, has involved the loss of the NDH complex in at least some photosynthetic angiosperms.


Subject(s)
Adaptation, Biological/genetics , Alismatales/genetics , Genes, Plant , Genome, Plastid , NADH Dehydrogenase/genetics , Plant Proteins/genetics , Plastids/genetics , Aquatic Organisms , Biological Evolution , Genome Size , NADH Dehydrogenase/deficiency , Photosynthesis/physiology , Phylogeny , Plant Proteins/metabolism , Plastids/classification , Pollination/physiology , Repetitive Sequences, Nucleic Acid
15.
PLoS One ; 7(11): e50644, 2012.
Article in English | MEDLINE | ID: mdl-23226344

ABSTRACT

Defects in mitochondrial electron transport chain (ETC) function have been implicated in a number of neurodegenerative disorders, cancer, and aging. Mitochondrial complex I (NADH dehydrogenase) is the largest and most complicated enzyme of the ETC with 45 subunits originating from two separate genomes. The biogenesis of complex I is an intricate process that requires multiple steps, subassemblies, and assembly factors. Here, we report the generation and characterization of a Drosophila model of complex I assembly factor deficiency. We show that CG7598 (dCIA30), the Drosophila homolog of human complex I assembly factor Ndufaf1, is necessary for proper complex I assembly. Reduced expression of dCIA30 results in the loss of the complex I holoenzyme band in blue-native polyacrylamide gel electrophoresis and loss of NADH:ubiquinone oxidoreductase activity in isolated mitochondria. The complex I assembly defect, caused by mutation or RNAi of dCIA30, has repercussions both during development and adulthood in Drosophila, including developmental arrest at the pupal stage and reduced stress resistance during adulthood. Expression of the single-subunit yeast alternative NADH dehydrogenase, Ndi1, can partially or wholly rescue phenotypes associated with the complex I assembly defect. Our work shows that CG7598/dCIA30 is a functional homolog of Ndufaf1 and adds to the accumulating evidence that transgenic NDI1 expression is a viable therapy for disorders arising from complex I deficiency.


Subject(s)
Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Electron Transport Complex I/genetics , NADH Dehydrogenase/chemistry , NADH Dehydrogenase/metabolism , Saccharomyces cerevisiae Proteins/genetics , Animals , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Electron Transport Complex I/deficiency , Gene Expression , Holoenzymes/chemistry , Holoenzymes/deficiency , Holoenzymes/genetics , Holoenzymes/metabolism , Humans , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Diseases/genetics , Mitochondrial Diseases/therapy , NADH Dehydrogenase/deficiency , NADH Dehydrogenase/genetics , Phenotype , RNA Interference , Sequence Homology, Amino Acid
16.
Biometals ; 24(5): 827-35, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21390523

ABSTRACT

Copper is both an essential nutrient and a toxic element able to catalyze free radicals formation which damage lipids and proteins. Although the available copper redox species in aerobic environment is Cu(II), proteins that participate in metal homeostasis use Cu(I). With isolated Escherichia coli membranes, we have previously shown that electron flow through the respiratory chain promotes cupric ions reduction by NADH dehydrogenase-2 and quinones. Here, we determined Cu(II)-reductase activity by whole cells using strains deficient in these respiratory chain components. Measurements were done by the appearance of Cu(I) in the supernatants of cells exposed to sub-lethal Cu(II) concentrations. In the absence of quinones, the Cu(II)-reduction rate decreased ~70% in respect to the wild-type strain, while this diminution was about 85% in a strain lacking both NDH-2 and quinones. The decrease was ~10% in the absence of only NDH-2. In addition, we observed that quinone deficient strains failed to grow in media containing either excess or deficiency of copper, as we have described for NDH-2 deficient mutants. Thus, the Cu(II)-reduction by E. coli intact cells is mainly due to quinones and to a lesser extent to NDH-2, in a quinone-independent way. To our knowledge, this is the first in vivo demonstration of the involvement of E. coli respiratory components in the Cu(II)-reductase activity which contributes to the metal homeostasis.


Subject(s)
Copper/metabolism , Electron Transport , Escherichia coli/cytology , Escherichia coli/metabolism , NADH Dehydrogenase/metabolism , Cell Membrane/metabolism , Escherichia coli/chemistry , Iron/metabolism , NADH Dehydrogenase/deficiency , NADH Dehydrogenase/genetics , Oxidation-Reduction , Oxidoreductases/metabolism , Quinones/metabolism
17.
Arch Biochem Biophys ; 505(2): 155-9, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20933494

ABSTRACT

Respiratory NADH dehydrogenase-2 (NDH-2) of Escherichia coli is a membrane-bound flavoprotein. Bioinformatics approaches suggested the involvement of NDH-2 C-terminal region in membrane anchorage. Here, we demonstrated that NDH-2 is a peripheral membrane protein and that its predicted C-terminal amphipathic Arg390-Ala406 helix is sufficient to bind the protein to lipid membranes. Additionally, a cytosolic NDH-2 protein (Trun-3), lacking the last 43 aminoacids, was purified and characterized. FAD cofactor was absent in purified Trun-3. Upon the addition of FAD, Trun-3 maximum velocity was similar to native NDH-2 rate with ferricyanide and MTT acceptors. However, Trun-3 activity was around 5-fold lower with quinones. No significant difference in K(m) values was observed for both enzymes. For the first time, an active and water soluble NDH-2 was obtained, representing a major improvement for structural/functional characterizations.


Subject(s)
Cell Membrane/metabolism , Escherichia coli/cytology , Escherichia coli/enzymology , Hydrophobic and Hydrophilic Interactions , NADH Dehydrogenase/chemistry , NADH Dehydrogenase/metabolism , Amino Acid Sequence , Kinetics , Molecular Sequence Data , NADH Dehydrogenase/deficiency , NADH Dehydrogenase/genetics , Protein Structure, Secondary , Protein Transport , Sequence Deletion , Solubility , Water/chemistry
18.
Intern Med ; 49(19): 2039-42, 2010.
Article in English | MEDLINE | ID: mdl-20930427

ABSTRACT

BACKGROUND: It is known that patients with myocarditis of unknown etiology and with dilated cardiomyopathy show a high incidence of serum autoantibodies (M7) directed against mitochondrial flavoproteins. The tissue concentration of mitochondrial flavoproteins in the myocardium obtained from patients with dilated cardiomyopathy (DCM) was examined to further investigate the immunopathological mechanism of cardiomyopathy. METHODS AND RESULTS: Myocardial tissue specimens were obtained from patients who underwent cardiac catheterization and a subendomyocardial biopsy for the diagnosis of cardiomyopathy. All samples were analyzed by Western blotting. NADH dehydrogenase ubiquinone flavoprotein (NDUFV1) production in the myocardium decreased significantly with DCM, in comparison to fumarate hydratase and flavoprotein SDHA. There was a significant negative-correlation between the left ventricular end-diastolic dimension and NDUFV1 production (R(2)=0.291, p value<0.05). CONCLUSION: NDUFV1 may be involved in the pathogenesis of DCM. A defect of mitochondrial NDUFV1 may reduce complex I, which produces most of the superoxide, which is then scavenged by the mitochondrial enzyme Mn-superoxide dismutase to produce H(2)O(2). Exploring the nature of the candidate protein found in the myocardium in this study will provide further insight into the immunological mechanism of DCM.


Subject(s)
Cardiomyopathy, Dilated/enzymology , Myocardium/enzymology , NADH Dehydrogenase/deficiency , Adult , Aged , Aged, 80 and over , Animals , Autoantibodies/metabolism , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/pathology , Cells, Cultured , Electron Transport Complex I , Female , Humans , Male , Mice , Middle Aged , Mitochondria, Heart/enzymology , Myocardium/immunology , Myocardium/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/immunology , NADH Dehydrogenase/immunology
19.
Mitochondrion ; 10(2): 125-36, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19900588

ABSTRACT

Mitochondrial dysfunction and associated oxidant stress have been linked with numerous complex diseases and aging largely by in vitro determination of mitochondria oxidant production and scavenging. We applied targeted in vivo fluorescence analyses of mitochondria-dense pharyngeal tissue in Caenorhabditis elegans to better understand relative mitochondrial effects, particularly on matrix oxidant burden, of respiratory chain complex, MnSOD, and insulin receptor mutants displaying variable longevity. The data demonstrate significantly elevated in vivo matrix oxidant burden in the short-lived complex I mutant, gas-1(fc21), which was associated with limited superoxide scavenging capacity despite robust MnSOD induction, as well as decreased mitochondria content and membrane potential. Significantly increased MnSOD activity was associated with in vivo matrix oxidant levels similar to wild-type in the long-lived respiratory chain complex III mutant, isp-1(qm150). Yet, despite greater superoxide scavenging capacity in the complex III mutant than in the significantly longer-lived insulin receptor mutant, daf-2(e1368), only the former showed modest oxidative stress sensitivity. Furthermore, increased longevity was seen in MnSOD knockout mutants (sod-2(ok1030) and sod-2(gk257)) that had decreased MnSOD scavenging capacity and increased in vivo matrix oxidant burden. Thus, factors beside oxidant stress must underlie RC mutant longevity in C. elegans. This work highlights the utility of the C. elegans model as a tractable means to non-invasively monitor multi-dimensional in vivo consequences of primary mitochondrial dysfunction.


Subject(s)
Caenorhabditis elegans/physiology , Electron Transport , Mitochondria/physiology , Oxidants/toxicity , Oxidative Stress , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins , Electron Transport Complex III/deficiency , Longevity , Membrane Potential, Mitochondrial , NADH Dehydrogenase/deficiency , Receptor, Insulin/deficiency , Superoxide Dismutase/deficiency
20.
Eur J Heart Fail ; 12(1): 4-12, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20023039

ABSTRACT

AIMS: The transcription factor MEF2 is a downstream target for several hypertrophic signalling pathways in the heart, suggesting that MEF2 may act as a valuable therapeutic target in the treatment of heart failure. METHODS AND RESULTS: In this study, we investigated the potential benefits of overall MEF2 inhibition in a mouse model of chronic pressure overloading, by subjecting transgenic mice expressing a dominant negative form of MEF2 (DN-MEF2 Tg) in the heart, to transverse aortic constriction (TAC). Histological analysis revealed no major differences in cardiac remodelling between DN-MEF2 Tg and control mice after TAC. Surprisingly, echocardiographic analysis revealed that DN-MEF2 Tg mice had a decrease in cardiac function compared with control animals. Analysis of the mitochondrial respiratory chain showed that DN-MEF2 Tg mice displayed lower expression of NADH dehydrogenase subunit 6 (ND6), part of mitochondrial Complex I. The reduced expression of ND6 in DN-MEF2 Tg mice after pressure overload correlated with an increase in cell death secondary to overproduction of reactive oxygen species (ROS). CONCLUSION: Our data suggest that MEF2 transcriptional activity is required for mitochondrial function and its inhibition predisposes the heart to impaired mitochondrial function, overproduction of ROS, enhanced cell death, and cardiac dysfunction, following pressure overload.


Subject(s)
Cardiomegaly/etiology , Heart Failure/etiology , Mitochondria, Heart/physiology , Myogenic Regulatory Factors/antagonists & inhibitors , 8-Hydroxy-2'-Deoxyguanosine , Animals , Apoptosis/genetics , Cardiomegaly/metabolism , Cardiomegaly/pathology , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/genetics , Disease Models, Animal , Echocardiography , Gene Expression , Heart Failure/metabolism , Heart Failure/pathology , Integrases/genetics , MEF2 Transcription Factors , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myogenic Regulatory Factors/genetics , NADH Dehydrogenase/deficiency , Transcription, Genetic , Treatment Outcome , Ventricular Remodeling/genetics
SELECTION OF CITATIONS
SEARCH DETAIL