Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
iScience ; 23(8): 101362, 2020 Aug 21.
Article in English | MEDLINE | ID: mdl-32738610

ABSTRACT

Dopaminergic (DA) neurons have been implicated as key targets in neurological disorders, notably those involving locomotor impairment, and are considered to be highly vulnerable to mitochondrial dysfunction, a common feature of such diseases. Here we investigated a Drosophila model of locomotor disorders in which functional impairment is brought about by pan-neuronal RNAi knockdown of subunit COX7A of cytochrome oxidase (COX). Despite minimal neuronal loss by apoptosis, the expression and activity of tyrosine hydroxylase was decreased by half. Surprisingly, COX7A knockdown specifically targeted to DA neurons did not produce locomotor defect. Instead, using various drivers, we found that COX7A knockdown in specific groups of cholinergic and glutamatergic neurons underlay the phenotype. Based on our main finding, the vulnerability of DA neurons to mitochondrial dysfunction as a cause of impaired locomotion in other organisms, including mammals, warrants detailed investigation.

2.
Sci Adv ; 6(16): eaba0694, 2020 04.
Article in English | MEDLINE | ID: mdl-32426457

ABSTRACT

Mitochondria play an important role in sensing both acute and chronic hypoxia in the pulmonary vasculature, but their primary oxygen-sensing mechanism and contribution to stabilization of the hypoxia-inducible factor (HIF) remains elusive. Alteration of the mitochondrial electron flux and increased superoxide release from complex III has been proposed as an essential trigger for hypoxic pulmonary vasoconstriction (HPV). We used mice expressing a tunicate alternative oxidase, AOX, which maintains electron flux when respiratory complexes III and/or IV are inhibited. Respiratory restoration by AOX prevented acute HPV and hypoxic responses of pulmonary arterial smooth muscle cells (PASMC), acute hypoxia-induced redox changes of NADH and cytochrome c, and superoxide production. In contrast, AOX did not affect the development of chronic hypoxia-induced pulmonary hypertension and HIF-1α stabilization. These results indicate that distal inhibition of the mitochondrial electron transport chain in PASMC is an essential initial step for acute but not chronic oxygen sensing.

3.
PLoS Genet ; 15(10): e1008410, 2019 10.
Article in English | MEDLINE | ID: mdl-31584940

ABSTRACT

Mitochondria have been increasingly recognized as a central regulatory nexus for multiple metabolic pathways, in addition to ATP production via oxidative phosphorylation (OXPHOS). Here we show that inducing mitochondrial DNA (mtDNA) stress in Drosophila using a mitochondrially-targeted Type I restriction endonuclease (mtEcoBI) results in unexpected metabolic reprogramming in adult flies, distinct from effects on OXPHOS. Carbohydrate utilization was repressed, with catabolism shifted towards lipid oxidation, accompanied by elevated serine synthesis. Cleavage and translocation, the two modes of mtEcoBI action, repressed carbohydrate rmetabolism via two different mechanisms. DNA cleavage activity induced a type II diabetes-like phenotype involving deactivation of Akt kinase and inhibition of pyruvate dehydrogenase, whilst translocation decreased post-translational protein acetylation by cytonuclear depletion of acetyl-CoA (AcCoA). The associated decrease in the concentrations of ketogenic amino acids also produced downstream effects on physiology and behavior, attributable to decreased neurotransmitter levels. We thus provide evidence for novel signaling pathways connecting mtDNA to metabolism, distinct from its role in supporting OXPHOS.


Subject(s)
Cellular Reprogramming/genetics , DNA, Mitochondrial/genetics , Diabetes Mellitus, Type 2/genetics , Mitochondria/genetics , Adenosine Triphosphate/genetics , Animals , Carbohydrate Metabolism/genetics , Carbohydrates/genetics , DNA Restriction Enzymes/genetics , Diabetes Mellitus, Type 2/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Humans , Metabolic Networks and Pathways/genetics , Mitochondria/metabolism , Oxidative Phosphorylation , Oxidative Stress/genetics
4.
Biochim Biophys Acta Mol Basis Dis ; 1865(4): 854-866, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30342157

ABSTRACT

The alternative respiratory chain (aRC), comprising the alternative NADH dehydrogenases (NDX) and quinone oxidases (AOX), is found in microbes, fungi and plants, where it buffers stresses arising from restrictions on electron flow in the oxidative phosphorylation system. The aRC enzymes are also found in species belonging to most metazoan phyla, including some chordates and arthropods species, although not in vertebrates or in Drosophila. We postulated that the aRC enzymes might be deployed to alleviate pathological stresses arising from mitochondrial dysfunction in a wide variety of disease states. However, before such therapies can be contemplated, it is essential to understand the effects of aRC enzymes on cell metabolism and organismal physiology. Here we report and discuss new findings that shed light on the functions of the aRC enzymes in animals, and the unexpected benefits and detriments that they confer on model organisms. In Ciona intestinalis, the aRC is induced by hypoxia and by sulfide, but is unresponsive to other environmental stressors. When expressed in Drosophila, AOX results in impaired survival under restricted nutrition, in addition to the previously reported male reproductive anomalies. In contrast, it confers cold resistance to developing and adult flies, and counteracts cell signaling defects that underlie developmental dysmorphologies. The aRC enzymes may also influence lifespan and stress resistance more generally, by eliciting or interfering with hormetic mechanisms. In sum, their judicious use may lead to major benefits in medicine, but this will require a thorough characterization of their properties and physiological effects.


Subject(s)
Drosophila Proteins/metabolism , Mitochondria/metabolism , NADH Dehydrogenase/metabolism , Quinone Reductases/metabolism , Animals , Cell Respiration , Ciona intestinalis , Drosophila Proteins/genetics , Drosophila melanogaster , Electron Transport , Mitochondria/enzymology , NADH Dehydrogenase/genetics , Quinone Reductases/genetics
5.
Mol Cell Biol ; 38(24)2018 12 15.
Article in English | MEDLINE | ID: mdl-30224521

ABSTRACT

Downregulation of Jun N-terminal kinase (JNK) signaling inhibits cell migration in diverse model systems. In Drosophila pupal development, attenuated JNK signaling in the thoracic dorsal epithelium leads to defective midline closure, resulting in cleft thorax. Here we report that concomitant expression of the Ciona intestinalis alternative oxidase (AOX) was able to compensate for JNK pathway downregulation, substantially correcting the cleft thorax phenotype. AOX expression also promoted wound-healing behavior and single-cell migration in immortalized mouse embryonic fibroblasts (iMEFs), counteracting the effect of JNK pathway inhibition. However, AOX was not able to rescue developmental phenotypes resulting from knockdown of the AP-1 transcription factor, the canonical target of JNK, nor its targets and had no effect on AP-1-dependent transcription. The migration of AOX-expressing iMEFs in the wound-healing assay was differentially stimulated by antimycin A, which redirects respiratory electron flow through AOX, altering the balance between mitochondrial ATP and heat production. Since other treatments affecting mitochondrial ATP did not stimulate wound healing, we propose increased mitochondrial heat production as the most likely primary mechanism of action of AOX in promoting cell migration in these various contexts.


Subject(s)
Cell Movement/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Mitochondrial Proteins/metabolism , Oxidoreductases/metabolism , Plant Proteins/metabolism , Signal Transduction/physiology , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Ciona intestinalis/metabolism , Ciona intestinalis/physiology , Down-Regulation/physiology , Drosophila/metabolism , Drosophila/physiology , Drosophila Proteins/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/physiology , Male , Mice , Mitochondria/metabolism , Mitochondria/physiology , Phenotype , Thorax/metabolism , Thorax/physiology , Transcription Factor AP-1/metabolism , Transcription, Genetic/physiology , Wound Healing/physiology
6.
Sci Rep ; 8(1): 10882, 2018 Jul 18.
Article in English | MEDLINE | ID: mdl-30022066

ABSTRACT

The xenotopic expression of the alternative oxidase AOX from the tunicate Ciona intestinalis in diverse models of human disease partially alleviates the phenotypic effects of mitochondrial respiratory chain defects. AOX is a non-proton pumping, mitochondrial inner membrane-bound, single-subunit enzyme that can bypass electron transport through the cytochrome segment, providing an additional site for ubiquinone reoxidation and oxygen reduction upon respiratory chain overload. We set out to investigate whether AOX expression in Drosophila could counteract the effects of mitochondrial DNA (mtDNA) replication defects caused by disturbances in the mtDNA helicase or DNA polymerase γ. We observed that the developmental arrest imposed by either the expression of mutant forms of these enzymes or their knockdown was not rescued by AOX. Considering also the inability of AOX to ameliorate the phenotype of tko25t, a fly mutant with mitochondrial translation deficiency, we infer that this alternative enzyme may not be applicable to cases of mitochondrial gene expression defects. Finding the limitations of AOX applicability will help establish the parameters for the future putative use of this enzyme in gene therapies for human mitochondrial diseases.


Subject(s)
DNA Replication , DNA, Mitochondrial/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Mitochondria/pathology , Mitochondrial Diseases/physiopathology , Mitochondrial Proteins/metabolism , Oxidoreductases/metabolism , Plant Proteins/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Electron Transport , Female , Genes, Lethal , Male , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mutation , Oxidoreductases/genetics , Phenotype , Plant Proteins/genetics
7.
BMC Dev Biol ; 17(1): 9, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28673232

ABSTRACT

BACKGROUND: Mitochondrial alternative respiratory-chain enzymes are phylogenetically widespread, and buffer stresses affecting oxidative phosphorylation in species that possess them. However, they have been lost in the evolutionary lineages leading to vertebrates and arthropods, raising the question as to what survival or reproductive disadvantages they confer. Recent interest in using them in therapy lends a biomedical dimension to this question. METHODS: Here, we examined the impact of the expression of Ciona intestinalis alternative oxidase, AOX, on the reproductive success of Drosophila melanogaster males. Sperm-competition assays were performed between flies carrying three copies of a ubiquitously expressed AOX construct, driven by the α-tubulin promoter, and wild-type males of the same genetic background. RESULTS: In sperm-competition assays, AOX conferred a substantial disadvantage, associated with decreased production of mature sperm. Sperm differentiation appeared to proceed until the last stages, but was spatially deranged, with spermatozoids retained in the testis instead of being released to the seminal vesicle. High AOX expression was detected in the outermost cell-layer of the testis sheath, which we hypothesize may disrupt a signal required for sperm maturation. CONCLUSIONS: AOX expression in Drosophila thus has effects that are deleterious to male reproductive function. Our results imply that AOX therapy must be developed with caution.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Spermatogenesis/genetics , Animals , Ciona intestinalis/genetics , Drosophila melanogaster/enzymology , Gene Expression , Male , Testis/embryology , Testis/enzymology
8.
G3 (Bethesda) ; 6(9): 2839-46, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27412986

ABSTRACT

Culture of Drosophila expressing the steroid-dependent GeneSwitch transcriptional activator under the control of the ubiquitous α-tubulin promoter was found to produce extensive pupal lethality, as well as a range of dysmorphic adult phenotypes, in the presence of high concentrations of the inducing drug RU486. Prominent among these was cleft thorax, seen previously in flies bearing mutant alleles of the nuclear receptor Ultraspiracle and many other mutants, as well as notched wings, leg malformations, and bristle abnormalities. Neither the α-tubulin-GeneSwitch driver nor the inducing drug on their own produced any of these effects. A second GeneSwitch driver, under the control of the daughterless promoter, which gave much lower and more tissue-restricted transgene expression, exhibited only mild bristle abnormalities in the presence of high levels of RU486. Coexpression of the alternative oxidase (AOX) from Ciona intestinalis produced a substantial shift in the developmental outcome toward a wild-type phenotype, which was dependent on the AOX expression level. Neither an enzymatically inactivated variant of AOX, nor GFP, or the alternative NADH dehydrogenase Ndi1 from yeast gave any such rescue. Users of the GeneSwitch system should be aware of the potential confounding effects of its application in developmental studies.


Subject(s)
Ciona intestinalis/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Embryonic Development/genetics , Mitochondrial Proteins/genetics , Oxidoreductases/genetics , Plant Proteins/genetics , Transcription Factors/genetics , Wings, Animal/abnormalities , Animals , Ciona intestinalis/enzymology , Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , Electron Transport Complex I/genetics , Genotype , Ligands , Mifepristone/pharmacology , Mutation , Phenotype , Pupa/drug effects , Pupa/genetics , Saccharomyces cerevisiae Proteins/genetics , Thorax/abnormalities , Thorax/drug effects , Transgenes/genetics , Wings, Animal/drug effects
9.
Sci Rep ; 5: 18295, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26672986

ABSTRACT

The mitochondrial alternative oxidase, AOX, carries out the non proton-motive re-oxidation of ubiquinol by oxygen in lower eukaryotes, plants and some animals. Here we created a modified version of AOX from Ciona instestinalis, carrying mutations at conserved residues predicted to be required for chelation of the diiron prosthetic group. The modified protein was stably expressed in mammalian cells or flies, but lacked enzymatic activity and was unable to rescue the phenotypes of flies knocked down for a subunit of cytochrome oxidase. The mutated AOX transgene is thus a potentially useful tool in studies of the physiological effects of AOX expression.


Subject(s)
Ciona intestinalis/enzymology , Drosophila melanogaster/enzymology , Electron Transport Complex IV/metabolism , Iron/metabolism , Mitochondrial Proteins/metabolism , Mutation , Oxidoreductases/metabolism , Plant Proteins/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Cell Line , Ciona intestinalis/genetics , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Electron Transport Complex IV/genetics , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Iron/chemistry , Male , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Models, Molecular , Molecular Sequence Data , Oxidoreductases/chemistry , Oxidoreductases/genetics , Oxygen Consumption , Plant Proteins/chemistry , Plant Proteins/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid
10.
Acta Chim Slov ; 61(4): 858-65, 2014.
Article in English | MEDLINE | ID: mdl-25551727

ABSTRACT

The aim of this research was to optimize the extraction condition of ultrasound-assisted extraction (UAE) of phenols from the red grape of Vranac variety (Vitis vinifera L.) pomace seeds. The minimum experiments needed for optimization of UAE by response surface methodology (RSM) were obtained by spectrophotometric and HPLC analyses of seed extracts. UAE greatly depends on three independent variables: extraction temperature, time and liquid/solid ratio. The RSM can be used for optimization of UAE conditions to obtain maximum responses such as extraction yield, TPC, (+)-catechin, (-)-epicatechin and proanthocyanidin content. The predicted values of the model were in accordance with experimental data under the same conditions (RSD was 0.74%). Experimental data also confirmed that UAE gives a better yield of phenolics than conventional solvent extraction (23.76% increase). The UAE under optimal extraction conditions is suitable for obtaining extracts that are rich in phenolic content, and have strong antioxidant activity which could be used as additives in food and medicaments.


Subject(s)
Phenols/chemistry , Seeds/chemistry , Ultrasonics/methods , Vitis/chemistry , Antioxidants/chemistry , Chemistry Techniques, Analytical , Plant Extracts/chemistry , Solvents
SELECTION OF CITATIONS
SEARCH DETAIL