Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
Biochim Biophys Acta Bioenerg ; 1865(3): 149046, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38642871

ABSTRACT

The respiratory chain alternative enzymes (AEs) NDX and AOX from the tunicate Ciona intestinalis (Ascidiacea) have been xenotopically expressed and characterized in human cells in culture and in the model organisms Drosophila melanogaster and mouse, with the purpose of developing bypass therapies to combat mitochondrial diseases in human patients with defective complexes I and III/IV, respectively. The fact that the genes coding for NDX and AOX have been lost from genomes of evolutionarily successful animal groups, such as vertebrates and insects, led us to investigate if the composition of the respiratory chain of Ciona and other tunicates differs significantly from that of humans and Drosophila, to accommodate the natural presence of AEs. We have failed to identify in tunicate genomes fifteen orthologous genes that code for subunits of the respiratory chain complexes; all of these putatively missing subunits are peripheral to complexes I, III and IV in mammals, and many are important for complex-complex interaction in supercomplexes (SCs), such as NDUFA11, UQCR11 and COX7A. Modeling of all respiratory chain subunit polypeptides of Ciona indicates significant structural divergence that is consistent with the lack of these fifteen clear orthologous subunits. We also provide evidence using Ciona AOX expressed in Drosophila that this AE cannot access the coenzyme Q pool reduced by complex I, but it is readily available to oxidize coenzyme Q molecules reduced by glycerophosphate oxidase, a mitochondrial inner membrane-bound dehydrogenase that is not involved in SCs. Altogether, our results suggest that Ciona AEs might have evolved in a mitochondrial inner membrane environment much different from that of mammals and insects, possibly without SCs; this correlates with the preferential functional interaction between these AEs and non-SC dehydrogenases in heterologous mammalian and insect systems. We discuss the implications of these findings for the applicability of Ciona AEs in human bypass therapies and for our understanding of the evolution of animal respiratory chain.


Subject(s)
Ciona intestinalis , Mitochondrial Proteins , Oxidative Phosphorylation , Animals , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Ciona intestinalis/genetics , Ciona intestinalis/enzymology , Humans , Oxidoreductases/genetics , Oxidoreductases/metabolism , Protein Subunits/metabolism , Protein Subunits/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/enzymology , Urochordata/genetics , Urochordata/enzymology , Electron Transport , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Phylogeny , Plant Proteins
2.
Elife ; 102021 11 15.
Article in English | MEDLINE | ID: mdl-34779404

ABSTRACT

Positively charged amino acids respond to membrane potential changes to drive voltage sensor movement in voltage-gated ion channels, but determining the displacements of voltage sensor gating charges has proven difficult. We optically tracked the movement of the two most extracellular charged residues (R1 and R2) in the Shaker potassium channel voltage sensor using a fluorescent positively charged bimane derivative (qBBr) that is strongly quenched by tryptophan. By individually mutating residues to tryptophan within the putative pathway of gating charges, we observed that the charge motion during activation is a rotation and a tilted translation that differs between R1 and R2. Tryptophan-induced quenching of qBBr also indicates that a crucial residue of the hydrophobic plug is linked to the Cole-Moore shift through its interaction with R1. Finally, we show that this approach extends to additional voltage-sensing membrane proteins using the Ciona intestinalis voltage-sensitive phosphatase (CiVSP).


Subject(s)
Ion Channel Gating/physiology , Potassium Channels, Voltage-Gated/physiology , Potassium Channels/physiology , Animals , Biophysical Phenomena , Bridged Bicyclo Compounds, Heterocyclic , Ciona intestinalis/enzymology , Membrane Potentials , Shaker Superfamily of Potassium Channels , Tryptophan/chemistry , Xenopus laevis
3.
Nature ; 585(7824): 288-292, 2020 09.
Article in English | MEDLINE | ID: mdl-32641834

ABSTRACT

The mitochondrial electron transport chain (ETC) is necessary for tumour growth1-6 and its inhibition has demonstrated anti-tumour efficacy in combination with targeted therapies7-9. Furthermore, human brain and lung tumours display robust glucose oxidation by mitochondria10,11. However, it is unclear why a functional ETC is necessary for tumour growth in vivo. ETC function is coupled to the generation of ATP-that is, oxidative phosphorylation and the production of metabolites by the tricarboxylic acid (TCA) cycle. Mitochondrial complexes I and II donate electrons to ubiquinone, resulting in the generation of ubiquinol and the regeneration of the NAD+ and FAD cofactors, and complex III oxidizes ubiquinol back to ubiquinone, which also serves as an electron acceptor for dihydroorotate dehydrogenase (DHODH)-an enzyme necessary for de novo pyrimidine synthesis. Here we show impaired tumour growth in cancer cells that lack mitochondrial complex III. This phenotype was rescued by ectopic expression of Ciona intestinalis alternative oxidase (AOX)12, which also oxidizes ubiquinol to ubiquinone. Loss of mitochondrial complex I, II or DHODH diminished the tumour growth of AOX-expressing cancer cells deficient in mitochondrial complex III, which highlights the necessity of ubiquinone as an electron acceptor for tumour growth. Cancer cells that lack mitochondrial complex III but can regenerate NAD+ by expression of the NADH oxidase from Lactobacillus brevis (LbNOX)13 targeted to the mitochondria or cytosol were still unable to grow tumours. This suggests that regeneration of NAD+ is not sufficient to drive tumour growth in vivo. Collectively, our findings indicate that tumour growth requires the ETC to oxidize ubiquinol, which is essential to drive the oxidative TCA cycle and DHODH activity.


Subject(s)
Mitochondria/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Ubiquinone/analogs & derivatives , Animals , Cell Line, Tumor , Cell Proliferation , Ciona intestinalis/enzymology , Citric Acid Cycle , Cytosol/metabolism , Dihydroorotate Dehydrogenase , Electron Transport , Electron Transport Complex I/metabolism , Electron Transport Complex II/metabolism , Electron Transport Complex III/deficiency , Electron Transport Complex III/metabolism , Humans , Levilactobacillus brevis/enzymology , Male , Mice , Mitochondria/enzymology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , NAD/metabolism , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , Neoplasms/enzymology , Oxidative Phosphorylation , Oxidoreductases/genetics , Oxidoreductases/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Ubiquinone/metabolism
4.
Biogerontology ; 21(2): 155-171, 2020 04.
Article in English | MEDLINE | ID: mdl-31749111

ABSTRACT

Mitochondrial alternative NADH dehydrogenase (aNDH) was found to extend lifespan when expressed in the fruit fly. We have found that fruit flies expressing aNDH from Ciona intestinalis (NDX) had 17-71% lifespan prolongation on media with different protein-tocarbohydrate ratios except NDX-expressing males that had 19% shorter lifespan than controls on a high protein diet. NDX-expressing flies were more resistant to organic xenobiotics, 2,4-dichlorophenoxyacetic acid and alloxan, and inorganic toxicant potassium iodate, and partially to sodium molybdate treatments. On the other hand, NDX-expressing flies were more sensitive to catechol and sodium chromate. Enzymatic analysis showed that NDX-expressing males had higher glucose 6-phosphate dehydrogenase activity, whilst both sexes showed increased glutathione S-transferase activity.


Subject(s)
Ciona intestinalis/enzymology , Drosophila melanogaster/drug effects , Drosophila melanogaster/enzymology , Drug Resistance , Energy Metabolism , Longevity , NADH Dehydrogenase/metabolism , Xenobiotics/pharmacology , Animals , Animals, Genetically Modified , Ciona intestinalis/genetics , Drosophila melanogaster/genetics , Drug Resistance/genetics , Energy Metabolism/genetics , Female , Gene Expression Regulation , Longevity/genetics , Male , NADH Dehydrogenase/genetics , Sex Factors
5.
Physiol Rep ; 7(13): e14159, 2019 08.
Article in English | MEDLINE | ID: mdl-31267687

ABSTRACT

The alternative oxidase (AOX) from Ciona intestinalis was previously shown to be expressible in mice and to cause no physiological disturbance under unstressed conditions. Because AOX is known to become activated under some metabolic stress conditions, resulting in altered energy balance, we studied its effects in mice subjected to dietary stress. Wild-type mice (Mus musculus, strain C57BL/6JOlaHsd) fed a high-fat or ketogenic (high-fat, low-carbohydrate) diet show weight gain with increased fat mass, as well as loss of performance, compared with chow-fed animals. Unexpectedly, AOX-expressing mice fed on these metabolically stressful, fat-rich diets showed almost indistinguishable patterns of weight gain and altered body composition as control animals. Cardiac performance was impaired to a similar extent by ketogenic diet in AOX mice as in nontransgenic littermates. AOX and control animals fed on ketogenic diet both showed wide variance in weight gain. Analysis of the gut microbiome in stool revealed a strong correlation with diet, rather than with genotype. The microbiome of the most and least obese outliers reared on the ketogenic diet showed no consistent trends compared with animals of normal body weight. We conclude that AOX expression in mice does not modify physiological responses to extreme diets.


Subject(s)
Diet, Ketogenic/adverse effects , Oxidoreductases/genetics , Phenotype , Stress, Physiological , Animals , Body Composition , Ciona intestinalis/enzymology , Ciona intestinalis/genetics , Gastrointestinal Microbiome , Genotype , Heart/physiology , Male , Mice , Mice, Inbred C57BL , Oxidoreductases/metabolism , Transgenes , Weight Gain
6.
J Exp Zool A Ecol Integr Physiol ; 331(6): 341-356, 2019 07.
Article in English | MEDLINE | ID: mdl-31218852

ABSTRACT

The mitochondrial alternative oxidase, AOX, present in most eukaryotes apart from vertebrates and insects, catalyzes the direct oxidation of ubiquinol by oxygen, by-passing the terminal proton-motive steps of the respiratory chain. Its physiological role is not fully understood, but it is proposed to buffer stresses in the respiratory chain similar to those encountered in mitochondrial diseases in humans. Previously, we found that the ubiquitous expression of AOX from Ciona intestinalis in Drosophila perturbs the development of flies cultured under low-nutrient conditions (media containing only glucose and yeast). Here we tested the effects of a wide range of nutritional supplements on Drosophila development, to gain insight into the physiological mechanism underlying this developmental failure. On low-nutrient medium, larvae contained decreased amounts of triglycerides, lactate, and pyruvate, irrespective of AOX expression. Complex food supplements, including treacle (molasses), restored normal development to AOX-expressing flies, but many individual additives did not. Inhibition of AOX by treacle extract was excluded as a mechanism, since the supplement did not alter the enzymatic activity of AOX in vitro. Furthermore, antibiotics did not influence the organismal phenotype, indicating that commensal microbes were not involved. Fractionation of treacle identified a water-soluble fraction with low solubility in ethanol, rich in lactate and tricarboxylic acid cycle intermediates, which contained the critical activity. We propose that the partial activation of AOX during metamorphosis impairs the efficient use of stored metabolites, resulting in developmental failure.


Subject(s)
Animal Nutritional Physiological Phenomena , Drosophila/enzymology , Mitochondria/genetics , Mitochondrial Proteins/metabolism , Oxidoreductases/metabolism , Plant Proteins/metabolism , Animals , Animals, Genetically Modified/growth & development , Animals, Genetically Modified/metabolism , Ciona intestinalis/enzymology , Diet , Drosophila/growth & development , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , HEK293 Cells , Humans , Larva/enzymology , Larva/growth & development , Metamorphosis, Biological , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Molasses/analysis , Oxidoreductases/genetics , Plant Proteins/genetics
7.
Genes Genet Syst ; 94(2): 81-93, 2019 Apr 27.
Article in English | MEDLINE | ID: mdl-30930342

ABSTRACT

Apurinic/apyrimidinic (AP) sites are the most common form of cytotoxic DNA damage. Since AP sites inhibit DNA replication and transcription, repairing them is critical for cell growth. However, the significance of repairing AP sites during early embryonic development has not yet been clearly determined. Here, we focused on APEX1 from the ascidian Ciona intestinalis (CiApex1), a homolog of human AP endonuclease 1 (APEX1), and examined its role in early embryonic development. Recombinant CiApex1 protein complemented the drug sensitivities of an AP endonuclease-deficient Escherichia coli mutant, and exhibited Mg2+-dependent AP endonuclease activity, like human APEX1, in vitro. Next, the effects of abnormal AP site repair on embryonic development were investigated. Treatment with methyl methanesulfonate, which alkylates DNA bases and generates AP sites, induced abnormal embryonic development. This abnormal phenotype was also caused by treatment with methoxyamine, which inhibits AP endonuclease activity. Furthermore, we constructed dominant-negative CiApex1, which inhibits CiApex1 action, and found that its expression impaired embryonic growth. These results suggested that AP site repair is essential for embryonic development and CiApex1 plays an important role in AP site repair during early embryonic development in C. intestinalis.


Subject(s)
Ciona intestinalis/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Embryonic Development/genetics , Animals , Ciona intestinalis/embryology , Ciona intestinalis/enzymology , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Mutation
8.
Fish Shellfish Immunol ; 86: 213-222, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30453047

ABSTRACT

Matrix metalloproteinases (MMPs) are a family of endopeptidases collectively able to degrade the components of the extracellular matrix (ECM), with important roles in many biological processes, such as embryogenesis, normal tissue remodelling, angiogenesis and wound healing. New views on the function of MMPs reveal that they regulate inflammatory response and therefore might represent an early step in the evolution of the immune system. MMPs can affect the activity of cytokines involved in inflammation including TGF-ß and TNF-α. MMPs are widely distributed in all kingdoms of life and have likely evolved from a single-domain protein which underwent successive rounds of duplications. In this study, we focused on the Ciona robusta (formerly known as Ciona intestinalis) MMP gelatinase homologue. Gene organization, phylogenetic analysis and 3D modeling supported the closest correlation of C. robusta gelatinase with the human MMP-9. Real-time PCR analysis and zymographic assay showed a prompt expression induced by LPS inoculation and an upregulation of enzymatic activity. Furthermore, we showed that before of the well-known increase of TGF-ß and TNF-α levels, a MMP-9like boost occurred, suggesting a possible involvement of MMP-9like in regulating inflammatory response in C. robusta.


Subject(s)
Ciona intestinalis/enzymology , Inflammation/enzymology , Matrix Metalloproteinases/genetics , Animals , Ciona intestinalis/genetics , Gelatinases/chemistry , Gelatinases/genetics , Inflammation/chemically induced , Lipopolysaccharides/toxicity , Matrix Metalloproteinases/chemistry , Models, Molecular , Phylogeny , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism
9.
EMBO Mol Med ; 11(1)2019 01.
Article in English | MEDLINE | ID: mdl-30530468

ABSTRACT

Alternative oxidase (AOX) is a non-mammalian enzyme that can bypass blockade of the complex III-IV segment of the respiratory chain (RC). We crossed a Ciona intestinalis AOX transgene into RC complex III (cIII)-deficient Bcs1lp.S78G knock-in mice, displaying multiple visceral manifestations and premature death. The homozygotes expressing AOX were viable, and their median survival was extended from 210 to 590 days due to permanent prevention of lethal cardiomyopathy. AOX also prevented renal tubular atrophy and cerebral astrogliosis, but not liver disease, growth restriction, or lipodystrophy, suggesting distinct tissue-specific pathogenetic mechanisms. Assessment of reactive oxygen species (ROS) production and damage suggested that ROS were not instrumental in the rescue. Cardiac mitochondrial ultrastructure, mitochondrial respiration, and pathological transcriptome and metabolome alterations were essentially normalized by AOX, showing that the restored electron flow upstream of cIII was sufficient to prevent cardiac energetic crisis and detrimental decompensation. These findings demonstrate the value of AOX, both as a mechanistic tool and a potential therapeutic strategy, for cIII deficiencies.


Subject(s)
Cardiomyopathies/prevention & control , Cell Respiration , Electron Transport Complex III/deficiency , Mitochondrial Proteins/metabolism , Oxidoreductases/metabolism , Plant Proteins/metabolism , Recombinant Proteins/metabolism , Animals , Ciona intestinalis/enzymology , Ciona intestinalis/genetics , Gene Knock-In Techniques , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Oxidoreductases/genetics , Plant Proteins/genetics , Recombinant Proteins/genetics , Survival Analysis
10.
Cell Tissue Res ; 370(1): 113-128, 2017 10.
Article in English | MEDLINE | ID: mdl-28547657

ABSTRACT

Many heterotrophic animals have a one-way alimentary canal that is essential for their nutrition and sequential steps of the digestive system, namely ingestion, digestion, absorption and elimination, are widely shared among bilaterians. Morphological, functional and molecular knowledge of the alimentary canal has been obtained in particular from mammalian research but the shared features and evolution of these aspects of the highly diverged alimentary canal in the animal kingdom are still unclear. We therefore investigate spatial gene expression patterns of pancreatic- and gastric-related molecules of ascidians (a sister group of vertebrates) with special reference to the functional regionality of the gastrointestinal tract. Genome-wide surveys of ascidian homologs to mammalian exocrine digestive enzyme genes revealed that pancreatic enzymes, namely alpha-amylase, lipase, phospholipase A2, trypsin, chymotrypsin and carboxypeptidase, exist in the ascidian genome. However, an ascidian homolog of the mammalian gastric enzyme pepsin has not been identified, although molecules resembling cathepsin D, a pepsin relative, are indeed present. Spatial expression analyses in the ascidian Ciona intestinalis, by means of whole-mount in situ hybridization, have elucidated that the expression of Ciona homologs of pancreatic- and gastric-related exocrine enzyme genes and of their transcriptional regulator genes is restricted to the Ciona stomach. Furthermore, the expression of these genes is localized to specific regions of the stomach epithelium according to their regionality in the vertebrate digestive system. The compartmentalized expression patterns of Ciona homologs imply primitive and/or ancestral aspects of molecular, functional and morphological bases among Olfactores.


Subject(s)
Ciona intestinalis/enzymology , Ciona intestinalis/genetics , Animals , Aspartic Acid Proteases/analysis , Aspartic Acid Proteases/genetics , Ciona intestinalis/anatomy & histology , Ciona intestinalis/physiology , Digestion , Gastrointestinal Tract/anatomy & histology , Gastrointestinal Tract/enzymology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/physiology , Gene Expression Regulation, Enzymologic , Transcription Factors/analysis , Transcription Factors/genetics
11.
Dis Model Mech ; 10(2): 163-171, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28067626

ABSTRACT

Plants and many lower organisms, but not mammals, express alternative oxidases (AOXs) that branch the mitochondrial respiratory chain, transferring electrons directly from ubiquinol to oxygen without proton pumping. Thus, they maintain electron flow under conditions when the classical respiratory chain is impaired, limiting excess production of oxygen radicals and supporting redox and metabolic homeostasis. AOX from Ciona intestinalis has been used to study and mitigate mitochondrial impairments in mammalian cell lines, Drosophila disease models and, most recently, in the mouse, where multiple lentivector-AOX transgenes conferred substantial expression in specific tissues. Here, we describe a genetically tractable mouse model in which Ciona AOX has been targeted to the Rosa26 locus for ubiquitous expression. The AOXRosa26 mouse exhibited only subtle phenotypic effects on respiratory complex formation, oxygen consumption or the global metabolome, and showed an essentially normal physiology. AOX conferred robust resistance to inhibitors of the respiratory chain in organello; moreover, animals exposed to a systemically applied LD50 dose of cyanide did not succumb. The AOXRosa26 mouse is a useful tool to investigate respiratory control mechanisms and to decipher mitochondrial disease aetiology in vivo.


Subject(s)
Mitochondrial Proteins/metabolism , Oxidoreductases/metabolism , Physiological Phenomena , Plant Proteins/metabolism , Animals , Ciona intestinalis/enzymology , Cyanides/administration & dosage , Cyanides/toxicity , Mice, Transgenic , Mitochondria/metabolism , Protective Agents/metabolism , RNA, Untranslated/genetics
12.
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
13.
Nat Chem Biol ; 12(4): 261-7, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26878552

ABSTRACT

Allostery provides a critical control over enzyme activity, biasing the catalytic site between inactive and active states. We found that the Ciona intestinalis voltage-sensing phosphatase (Ci-VSP), which modifies phosphoinositide signaling lipids (PIPs), has not one but two sequential active states with distinct substrate specificities, whose occupancy is allosterically controlled by sequential conformations of the voltage-sensing domain (VSD). Using fast fluorescence resonance energy transfer (FRET) reporters of PIPs to monitor enzyme activity and voltage-clamp fluorometry to monitor conformational changes in the VSD, we found that Ci-VSP switches from inactive to a PIP3-preferring active state when the VSD undergoes an initial voltage-sensing motion and then into a second PIP2-preferring active state when the VSD activates fully. This two-step allosteric control over a dual-specificity enzyme enables voltage to shape PIP concentrations in time, and provides a mechanism for the complex modulation of PIP-regulated ion channels, transporters, cell motility, endocytosis and exocytosis.


Subject(s)
Phosphatidylinositols/metabolism , Phosphoric Monoester Hydrolases/metabolism , Allosteric Regulation , Allosteric Site , Animals , Catalytic Domain , Ciona intestinalis/enzymology , Fluorescence Resonance Energy Transfer , Models, Molecular , Mutagenesis, Site-Directed , Oocytes , Patch-Clamp Techniques , Phosphoric Monoester Hydrolases/genetics , Substrate Specificity , Xenopus laevis
14.
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
15.
PLoS One ; 10(8): e0135924, 2015.
Article in English | MEDLINE | ID: mdl-26288188

ABSTRACT

Meprins are astacin metalloproteases with a characteristic, easily recognizable structure, given that they are the only proteases with both MAM and MATH domains plus a transmembrane region. So far assumed to be vertebrate-specific, it is shown here, using a combination of evolutionary and genomic analyses, that meprins originated before the urochordates/vertebrates split. In particular, three genes encoding structurally typical meprin proteins are arranged in tandem in the genome of the urochordate Ciona intestinalis. Phylogenetic analyses showed that the protease and MATH domains present in the meprin-like proteins encoded by the Ciona genes are very similar in sequence to the domains found in vertebrate meprins, which supports them having a common origin. While many vertebrates have the two canonical meprin-encoding genes orthologous to human MEP1A and MEP1B (which respectively encode for the proteins known as meprin α and meprin ß), a single gene has been found so far in the genome of the chondrichthyan fish Callorhinchus milii, and additional meprin-encoding genes are present in some species. Particularly, a group of bony fish species have genes encoding highly divergent meprins, here named meprin-F. Genes encoding meprin-F proteins, derived from MEP1B genes, are abundant in some species, as the Amazon molly, Poecilia formosa, which has 7 of them. Finally, it is confirmed that the MATH domains of meprins are very similar to the ones in TRAF ubiquitin ligases, which suggests that meprins originated when protease and TRAF E3-encoding sequences were combined.


Subject(s)
Biological Evolution , Metalloendopeptidases/genetics , Animals , Ciona intestinalis/enzymology , Ciona intestinalis/genetics , Humans , Poecilia/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics
16.
J Exp Zool B Mol Dev Evol ; 324(2): 141-51, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25786783

ABSTRACT

Two distinct Ciona intestinalis phenoloxidases (CinPO1, 2) had previously been cloned and sequenced. The CinPO2 is involved in innate immunity and is expressed by inflammatory hemocytes that populate the tunic and pharynx vessels as a response to LPS inoculation. In situ hybridization and immunohistochemistry assays on histological section, showed that the expression of this gene and the produced protein are shared with oogenesis, embryogenesis and larval morphogenesis. Intriguingly, upregulation of gene transcription was found in the test cell layer that envelopes the ovary follicle, ovulated egg, and gastrula, as well as it was modulated in the zygotic nucleus of outer balstomers of 32-cell embryo, neurula presumptive epidermis tissue and larval mesenchyme. The anti-CinPO2 antibodies, specific for adult inflammatory cells, recognize epitopes in the cytoplasm of ovarian oocytes, ovulated eggs, development stages and larval mesenchyme. The overall findings disclose the precocious activation of the CinPO2 immunity-related gene, and show a developmentally programmed expression of this phenoloxidase. Furthermore, these findings support the multifunctional roles of immunity-related genes and allows us to explore new perspectives on ascidian development and immunity.


Subject(s)
Ciona intestinalis/genetics , Gene Expression Regulation, Developmental , Monophenol Monooxygenase/genetics , Animals , Cell Differentiation , Ciona intestinalis/embryology , Ciona intestinalis/enzymology , Ciona intestinalis/growth & development , Embryo, Nonmammalian/enzymology , Female , Immunohistochemistry , In Situ Hybridization , Larva/enzymology , Monophenol Monooxygenase/metabolism , Ovary/enzymology , Sequence Analysis, Protein , Zygote/enzymology
17.
J Invertebr Pathol ; 126: 6-11, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25659264

ABSTRACT

We investigated the role of phenoloxidases (POs) in ascidians inflammatory reaction, a components of a copper-containing protein family involved in invertebrate immune system. In Ciona intestinalis two phenoloxidases (CinPO-1, CinPO-2) have been sequenced. In the present study, real time PCR analysis showed that both CinPO-1 and CinPO-2 genes were modulated by LPS inoculation suggesting that they are inducible and highly expressed in the inflamed pharynx. In situ hybridization disclosed CinPO-1 and CinPO-2 transcripts in pharynx hemocytes (granulocytes) and, mainly, in unilocular refractile granulocytes (URG) which mainly populated the inflamed tunic matrix. Interestingly, the genes are also upregulated by LPS in the endostyle (zones 7, 8 and 9) that is considered homolog to the vertebrate thyroid.


Subject(s)
Ciona intestinalis/enzymology , Lipopolysaccharides/pharmacology , Monophenol Monooxygenase/metabolism , Animals , Ciona intestinalis/drug effects , Ciona intestinalis/immunology , Hemocytes/drug effects , Hemocytes/enzymology , Hemocytes/immunology , In Situ Hybridization , Monophenol Monooxygenase/genetics , Real-Time Polymerase Chain Reaction , Up-Regulation/drug effects
18.
G3 (Bethesda) ; 4(10): 2013-21, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25147191

ABSTRACT

A point mutation [technical knockout(25t) (tko(25t))] in the Drosophila gene coding for mitoribosomal protein S12 generates a phenotype of developmental delay and bang sensitivity. tko(25t) has been intensively studied as an animal model for human mitochondrial diseases associated with deficiency of mitochondrial protein synthesis and consequent multiple respiratory chain defects. Transgenic expression in Drosophila of the alternative oxidase (AOX) derived from Ciona intestinalis has previously been shown to mitigate the toxicity of respiratory chain inhibitors and to rescue mutant and knockdown phenotypes associated with cytochrome oxidase deficiency. We therefore tested whether AOX expression could compensate the mutant phenotype of tko(25t) using the GeneSwitch system to activate expression at different times in development. The developmental delay of tko(25t) was not mitigated by expression of AOX throughout development. AOX expression for 1 d after eclosion, or continuously throughout development, had no effect on the bang sensitivity of tko(25t) adults, and continued expression in adults older than 30 d also produced no amelioration of the phenotype. In contrast, transgenic expression of the yeast alternative NADH dehydrogenase Ndi1 was synthetically semi-lethal with tko(25t) and was lethal when combined with both AOX and tko(25t). We conclude that AOX does not rescue tko(25t) and that the mutant phenotype is not solely due to limitations on electron flow in the respiratory chain, but rather to a more complex metabolic defect. The future therapeutic use of AOX in disorders of mitochondrial translation may thus be of limited value.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/enzymology , Drosophila/genetics , Mitochondrial Proteins/metabolism , Oxidoreductases/metabolism , Plant Proteins/metabolism , Ribosomal Proteins/metabolism , Adenosine Triphosphate/pharmacology , Animals , Ciona intestinalis/enzymology , Drosophila/growth & development , Drosophila/metabolism , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Embryonic Development/drug effects , Female , Genotype , Hormone Antagonists/pharmacology , Male , Mifepristone/pharmacology , Mitochondria/metabolism , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , NADH Dehydrogenase/genetics , NADH Dehydrogenase/metabolism , Oxidoreductases/genetics , Phenotype , Plant Proteins/genetics , Reactive Oxygen Species/metabolism , Ribosomal Proteins/deficiency , Ribosomal Proteins/genetics , beta Carotene/analogs & derivatives
19.
Int J Mol Sci ; 15(8): 13192-208, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-25073090

ABSTRACT

Adenylyl cyclase (AC) is a key enzyme that synthesizes cyclic AMP (cAMP) at the onset of the signaling pathway to activate sperm motility. Here, we showed that both transmembrane AC (tmAC) and soluble AC (sAC) are distinctly involved in the regulation of sperm motility in the ascidian Ciona intestinalis. A tmAC inhibitor blocked both cAMP synthesis and the activation of sperm motility induced by the egg factor sperm activating and attracting factor (SAAF), as well as those induced by theophylline, an inhibitor of phoshodiesterase. It also significantly inhibited cAMP-dependent phosphorylation of a set of proteins at motility activation. On the other hand, a sAC inhibitor does not affect on SAAF-induced transient increase of cAMP, motility activation or protein phosphorylation, but it reduced swimming velocity to half in theophylline-induced sperm. A sAC inhibitor KH-7 induced circular swimming trajectory with smaller diameter and significantly suppressed chemotaxis of sperm to SAAF. These results suggest that tmAC is involved in the basic mechanism for motility activation through cAMP-dependent protein phosphorylation, whereas sAC plays distinct roles in increase of flagellar beat frequency and in the Ca2+-dependent chemotactic movement of sperm.


Subject(s)
Adenylyl Cyclases/metabolism , Spermatozoa/physiology , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/classification , Animals , Bicarbonates/pharmacology , Calcium/metabolism , Ciona intestinalis/enzymology , Ciona intestinalis/metabolism , Cyclic AMP/metabolism , Enzyme Inhibitors/pharmacology , Male , Phosphorylation/drug effects , Phylogeny , Sperm Motility/drug effects , Spermatozoa/enzymology , Testis/enzymology , Theophylline/pharmacology , Valinomycin/pharmacology
20.
Aquat Toxicol ; 152: 47-56, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24727215

ABSTRACT

The major thiol-containing molecules involved in controlling the level of intracellular ROS in eukaryotes, acting as a nonenzymatic detoxification system, are metallothioneins (MTs), glutathione (GSH) and phytochelatins (PCs). Both MTs and GSH are well-known in the animal kingdom. PC was considered a prerogative of the plant kingdom but, in 2001, a phytochelatin synthase (PCS) gene was described in the nematode Caenorhabditis elegans; additional genes encoding this enzyme were later described in the earthworm Eisenia fetida and in the parasitic nematode Schistosoma mansoni but scanty data are available, up to now, for Deuterostomes. Here, we describe the molecular characteristics and transcription pattern, in the presence of Cd, of a PCS gene from the invertebrate chordate Ciona intestinalis, a ubiquitous solitary tunicate and demonstrate the presence of PCs in tissue extracts. We also studied mRNA localization by in situ hybridization. In addition, we analyzed the behavior of hemocytes and tunic cells consequent to Cd exposure as well as the transcription pattern of the Ciona orthologous for proliferating cell nuclear antigen (PCNA), usually considered a proliferation marker, and observed that cell proliferation occurs after 96h of Cd treatment. This matches the hypothesis of Cd-induced cell proliferation, as already suggested by previous data on the expression of a metallothionein gene in the same animal.


Subject(s)
Aminoacyltransferases/genetics , Cadmium/toxicity , Ciona intestinalis/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Water Pollutants, Chemical/toxicity , Amino Acid Sequence , Aminoacyltransferases/chemistry , Aminoacyltransferases/metabolism , Animals , Cadmium/analysis , Ciona intestinalis/chemistry , Ciona intestinalis/classification , Ciona intestinalis/enzymology , Ciona intestinalis/genetics , Gene Expression Profiling , Gene Order , Molecular Sequence Data , Phylogeny , Sequence Alignment , Water Pollutants, Chemical/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...