Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Microbiologyopen ; 10(5): e1238, 2021 10.
Article En | MEDLINE | ID: mdl-34713605

Om45 is a major protein of the yeast's outer mitochondrial membrane under respiratory conditions. However, the cellular role of the protein has remained obscure. Previously, deletion mutant phenotypes have not been found, and clear amino acid sequence similarities that would allow inferring its functional role are not available. In this work, we describe synthetic petite mutants of GEM1 and UGO1 that depend on the presence of OM45 for respiratory growth, as well as the identification of several multicopy suppressors of the synthetic petite phenotypes. In the analysis of our mutants, we demonstrate that Om45p and Gem1p have a collaborative role in the maintenance of mitochondrial morphology, cristae structure, and mitochondrial DNA maintenance. A group of multicopy suppressors rescuing the synthetic lethal phenotypes of the mutants on non-fermentable carbon sources additionally supports this result. Our results imply that the synthetic petite phenotypes we observed are due to the disturbance of the inner mitochondrial membrane and point to this mitochondrial sub-compartment as the main target of action of Om45p, Ugo1p, and the yeast Miro GTPase Gem1p.


Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Precursor Protein Import Complex Proteins/metabolism , Mitochondrial Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , DNA, Fungal , DNA, Mitochondrial/metabolism , GTP Phosphohydrolases/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Precursor Protein Import Complex Proteins/genetics , Mutation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
2.
Sci Rep ; 9(1): 12038, 2019 08 19.
Article En | MEDLINE | ID: mdl-31427678

A large number of studies have demonstrated significance of polyunsaturated fatty acids (PUFAs) for human health. However, many aspects on signals translating PUFA-sensing into body homeostasis have remained enigmatic. To shed light on PUFA physiology, we have generated a mouse line defective in mitochondrial dienoyl-CoA reductase (Decr), which is a key enzyme required for ß-oxidation of PUFAs. Previously, we have shown that these mice, whose oxidation of saturated fatty acid is intact but break-down of unsaturated fatty acids is blunted, develop severe hypoglycemia during metabolic stresses and fatal hypothermia upon acute cold challenge. In the current work, indirect calorimetry and thermography suggested that cold intolerance of Decr-/- mice is due to failure in maintaining appropriate heat production at least partly due to failure of brown adipose tissue (BAT) thermogenesis. Magnetic resonance imaging, electron microscopy, mass spectrometry and biochemical analysis showed attenuation in activation of lipolysis despite of functional NE-signaling and inappropriate expression of genes contributing to thermogenesis in iBAT when the Decr-/- mice were exposed to cold. We hypothesize that the failure in turning on BAT thermogenesis occurs due to accumulation of unsaturated long-chain fatty acids or their metabolites in Decr-/- mice BAT suppressing down-stream propagation of NE-signaling.


Adipose Tissue, Brown/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Oxidoreductases Acting on CH-CH Group Donors/deficiency , Thermogenesis/genetics , Adipose Tissue, White/metabolism , Animals , Fatty Acids/metabolism , Gene Expression , Humans , Lipolysis , Metabolic Networks and Pathways , Mice , Mice, Knockout , Oxidation-Reduction , Stress, Physiological , Thermography
3.
J Neurosci ; 38(45): 9781-9800, 2018 11 07.
Article En | MEDLINE | ID: mdl-30266742

There has been a growing interest toward mitochondrial fatty acid synthesis (mtFAS) since the recent discovery of a neurodegenerative human disorder termed MEPAN (mitochondrial enoyl reductase protein associated neurodegeneration), which is caused by mutations in the mitochondrial enoyl-CoA/ACP (acyl carrier protein) reductase (MECR) carrying out the last step of mtFAS. We show here that MECR protein is highly expressed in mouse Purkinje cells (PCs). To elucidate mtFAS function in neural tissue, here, we generated a mouse line with a PC-specific knock-out (KO) of Mecr, leading to inactivation of mtFAS confined to this cell type. Both sexes were studied. The mitochondria in KO PCs displayed abnormal morphology, loss of protein lipoylation, and reduced respiratory chain enzymatic activities by the time these mice were 6 months of age, followed by nearly complete loss of PCs by 9 months of age. These animals exhibited balancing difficulties ∼7 months of age and ataxic symptoms were evident from 8-9 months of age on. Our data show that impairment of mtFAS results in functional and ultrastructural changes in mitochondria followed by death of PCs, mimicking aspects of the clinical phenotype. This KO mouse represents a new model for impaired mitochondrial lipid metabolism and cerebellar ataxia with a distinct and well trackable cellular phenotype. This mouse model will allow the future investigation of the feasibility of metabolite supplementation approaches toward the prevention of neurodegeneration due to dysfunctional mtFAS.SIGNIFICANCE STATEMENT We have recently reported a novel neurodegenerative disorder in humans termed MEPAN (mitochondrial enoyl reductase protein associated neurodegeneration) (Heimer et al., 2016). The cause of neuron degeneration in MEPAN patients is the dysfunction of the highly conserved mitochondrial fatty acid synthesis (mtFAS) pathway due to mutations in MECR, encoding mitochondrial 2-enoyl-CoA/ACP reductase. The report presented here describes the analysis of the first mouse model suffering from mtFAS-defect-induced neurodegenerative changes due to specific disruption of the Mecr gene in Purkinje cells. Our work sheds a light on the mechanisms of neurodegeneration caused by mtFAS deficiency and provides a test bed for future treatment approaches.


Cerebellum/metabolism , Fatty Acids/biosynthesis , Mitochondria/metabolism , Nerve Degeneration/metabolism , Animals , Animals, Newborn , Cerebellum/pathology , Fatty Acids/genetics , Female , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/genetics , Mitochondria/pathology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Oxidoreductases Acting on CH-CH Group Donors/biosynthesis , Oxidoreductases Acting on CH-CH Group Donors/genetics
4.
Hum Mol Genet ; 26(11): 2104-2117, 2017 06 01.
Article En | MEDLINE | ID: mdl-28369354

Mitochondrial fatty acid synthesis (mtFAS) is an underappreciated but highly conserved metabolic process, indispensable for mitochondrial respiration. It was recently reported that dysfunction of mtFAS causes childhood onset of dystonia and optic atrophy in humans (MEPAN). To study the role of mtFAS in mammals, we generated three different mouse lines with modifications of the Mecr gene, encoding mitochondrial enoyl-CoA/ACP reductase (Mecr). A knock-out-first type mutation, relying on insertion of a strong transcriptional terminator between the first two exons of Mecr, displayed embryonic lethality over a broad window of time and due to a variety of causes. Complete removal of exon 2 or replacing endogenous Mecr by its functional prokaryotic analogue fabI (Mecrtm(fabI)) led to more consistent lethality phenotypes and revealed a hypoplastic placenta. Analyses of several mitochondrial parameters indicate that mitochondrial capacity for aerobic metabolism is reduced upon disrupting mtFAS function. Further analysis of the synthetic Mecrtm(fabI) models disclosed defects in development of placental trophoblasts consistent with disturbed peroxisome proliferator-activated receptor signalling. We conclude that disrupted mtFAS leads to deficiency in mitochondrial respiration, which lies at the root of the observed pantropic effects on embryonic and placental development in these mouse models.


Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/genetics , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Animals , Fatty Acid Desaturases/metabolism , Fatty Acids/metabolism , Female , Mice , Mice, Knockout , Mitochondria/metabolism , Oxidoreductases/metabolism , Placenta , Placentation/genetics , Placentation/physiology , Pregnancy
5.
Biochem J ; 461(1): 125-35, 2014 Jul 01.
Article En | MEDLINE | ID: mdl-24735479

Cholesterol is catabolized to bile acids by peroxisomal ß-oxidation in which the side chain of C27-bile acid intermediates is shortened by three carbon atoms to form mature C24-bile acids. Knockout mouse models deficient in AMACR (α-methylacyl-CoA racemase) or MFE-2 (peroxisomal multifunctional enzyme type 2), in which this ß-oxidation pathway is prevented, display a residual C24-bile acid pool which, although greatly reduced, implies the existence of alternative pathways of bile acid synthesis. One alternative pathway could involve Mfe-1 (peroxisomal multifunctional enzyme type 1) either with or without Amacr. To test this hypothesis, we generated a double knockout mouse model lacking both Amacr and Mfe-1 activities and studied the bile acid profiles in wild-type, Mfe-1 and Amacr single knockout mouse line and Mfe-1 and Amacr double knockout mouse lines. The total bile acid pool was decreased in Mfe-1-/- mice compared with wild-type and the levels of mature C24-bile acids were reduced in the double knockout mice when compared with Amacr-deficient mice. These results indicate that Mfe-1 can contribute to the synthesis of mature bile acids in both Amacr-dependent and Amacr-independent pathways.


Bile Acids and Salts/biosynthesis , Multienzyme Complexes/physiology , Racemases and Epimerases/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Multienzyme Complexes/deficiency , Multienzyme Complexes/genetics , Racemases and Epimerases/deficiency , Racemases and Epimerases/genetics , Signal Transduction/genetics , Signal Transduction/physiology
6.
PLoS Genet ; 10(3): e1004193, 2014 Mar.
Article En | MEDLINE | ID: mdl-24603431

Although the growth factor (GF) signaling guiding renal branching is well characterized, the intracellular cascades mediating GF functions are poorly understood. We studied mitogen-activated protein kinase (MAPK) pathway specifically in the branching epithelia of developing kidney by genetically abrogating the pathway activity in mice lacking simultaneously dual-specificity protein kinases Mek1 and Mek2. Our data show that MAPK pathway is heterogeneously activated in the subset of G1- and S-phase epithelial cells, and its tissue-specific deletion results in severe renal hypodysplasia. Consequently to the deletion of Mek1/2, the activation of ERK1/2 in the epithelium is lost and normal branching pattern in mutant kidneys is substituted with elongation-only phenotype, in which the epithelium is largely unable to form novel branches and complex three-dimensional patterns, but able to grow without primary defects in mitosis. Cellular characterization of double mutant epithelium showed increased E-cadherin at the cell surfaces with its particular accumulation at baso-lateral locations. This indicates changes in cellular adhesion, which were revealed by electron microscopic analysis demonstrating intercellular gaps and increased extracellular space in double mutant epithelium. When challenged to form monolayer cultures, the mutant epithelial cells were impaired in spreading and displayed strong focal adhesions in addition to spiky E-cadherin. Inhibition of MAPK activity reduced paxillin phosphorylation on serine 83 while remnants of phospho-paxillin, together with another focal adhesion (FA) protein vinculin, were augmented at cell surface contacts. We show that MAPK activity is required for branching morphogenesis, and propose that it promotes cell cycle progression and higher cellular motility through remodeling of cellular adhesions.


Focal Adhesions/genetics , Kidney/growth & development , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/genetics , Animals , Epithelial Cells/metabolism , Kidney/metabolism , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , MAP Kinase Signaling System/genetics , Mice , Mitogen-Activated Protein Kinases/genetics , Morphogenesis/genetics , Phosphorylation , Signal Transduction/genetics , Vinculin/metabolism
7.
PLoS One ; 7(4): e34530, 2012.
Article En | MEDLINE | ID: mdl-22506025

BACKGROUND: Glycosomes are a specialized form of peroxisomes (microbodies) present in unicellular eukaryotes that belong to the Kinetoplastea order, such as Trypanosoma and Leishmania species, parasitic protists causing severe diseases of livestock and humans in subtropical and tropical countries. The organelles harbour most enzymes of the glycolytic pathway that is responsible for substrate-level ATP production in the cell. Glycolysis is essential for bloodstream-form Trypanosoma brucei and enzymes comprising this pathway have been validated as drug targets. Glycosomes are surrounded by a single membrane. How glycolytic metabolites are transported across the glycosomal membrane is unclear. METHODS/PRINCIPAL FINDINGS: We hypothesized that glycosomal membrane, similarly to membranes of yeast and mammalian peroxisomes, contains channel-forming proteins involved in the selective transfer of metabolites. To verify this prediction, we isolated a glycosomal fraction from bloodstream-form T. brucei and reconstituted solubilized membrane proteins into planar lipid bilayers. The electrophysiological characteristics of the channels were studied using multiple channel recording and single channel analysis. Three main channel-forming activities were detected with current amplitudes 70-80 pA, 20-25 pA, and 8-11 pA, respectively (holding potential +10 mV and 3.0 M KCl as an electrolyte). All channels were in fully open state in a range of voltages ±150 mV and showed no sub-conductance transitions. The channel with current amplitude 20-25 pA is anion-selective (P(K+)/P(Cl-)∼0.31), while the other two types of channels are slightly selective for cations (P(K+)/P(Cl-) ratios ∼1.15 and ∼1.27 for the high- and low-conductance channels, respectively). The anion-selective channel showed an intrinsic current rectification that may suggest a functional asymmetry of the channel's pore. CONCLUSIONS/SIGNIFICANCE: These results indicate that the membrane of glycosomes apparently contains several types of pore-forming channels connecting the glycosomal lumen and the cytosol.


Intracellular Membranes/metabolism , Ion Channels/metabolism , Membrane Proteins/metabolism , Microbodies/metabolism , Trypanosoma brucei brucei/metabolism , Animals , Biological Transport , Cytosol/metabolism , Glycolysis , Lipid Bilayers/metabolism , Organelles/metabolism , Rats , Rats, Wistar
8.
FASEB J ; 23(11): 3682-91, 2009 Nov.
Article En | MEDLINE | ID: mdl-19571038

Mitochondrial fatty acid synthesis (FAS) generates the octanoyl-group that is required for the synthesis of lipoic acid and is linked to mitochondrial RNA metabolism. All of the human enzymes involved in mitochondrial FAS have been characterized except for beta-ketoacyl thioester reductase (HsKAR), which catalyzes the second step in the pathway. We report here the unexpected finding that a heterotetramer composed of human 17beta-hydroxysteroid dehydrogenase type 8 (Hs17beta-HSD8) and human carbonyl reductase type 4 (HsCBR4) forms the long-sought HsKAR. Both proteins share sequence similarities to the yeast 3-oxoacyl-(acyl carrier protein) reductase (Oar1p) and the bacterial FabG, although HsKAR is NADH dependent, whereas FabG and Oar1p are NADPH dependent. Hs17beta-HSD8 and HsCBR4 show a strong genetic interaction in vivo in yeast, where, only if they are expressed together, they rescue the respiratory deficiency and restore the lipoic acid content of oar1Delta cells. Moreover, these two proteins display a stable physical interaction and form an active heterotetramer. Both Hs17beta-HSD8 and HsCBR4 are targeted to mitochondria in vivo in cultured HeLa cells. Notably, 17beta-HSD8 was previously classified as a steroid-metabolizing enzyme, but our data suggest that 17beta-HSD8 is primarily involved in mitochondrial FAS.


17-Hydroxysteroid Dehydrogenases/metabolism , Alcohol Oxidoreductases/biosynthesis , Fatty Acid Synthases/biosynthesis , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Animals , Gene Knockout Techniques , HeLa Cells , Humans , Kinetics , Mice , Oxidoreductases , Protein Multimerization , Proteins , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Thioctic Acid/metabolism
9.
PLoS Genet ; 5(7): e1000543, 2009 Jul.
Article En | MEDLINE | ID: mdl-19578400

The mitochondrial beta-oxidation system is one of the central metabolic pathways of energy metabolism in mammals. Enzyme defects in this pathway cause fatty acid oxidation disorders. To elucidate the role of 2,4-dienoyl-CoA reductase (DECR) as an auxiliary enzyme in the mitochondrial beta-oxidation of unsaturated fatty acids, we created a DECR-deficient mouse line. In Decr(-/-) mice, the mitochondrial beta-oxidation of unsaturated fatty acids with double bonds is expected to halt at the level of trans-2, cis/trans-4-dienoyl-CoA intermediates. In line with this expectation, fasted Decr(-/-) mice displayed increased serum acylcarnitines, especially decadienoylcarnitine, a product of the incomplete oxidation of linoleic acid (C(18:2)), urinary excretion of unsaturated dicarboxylic acids, and hepatic steatosis, wherein unsaturated fatty acids accumulate in liver triacylglycerols. Metabolically challenged Decr(-/-) mice turned on ketogenesis, but unexpectedly developed hypoglycemia. Induced expression of peroxisomal beta-oxidation and microsomal omega-oxidation enzymes reflect the increased lipid load, whereas reduced mRNA levels of PGC-1alpha and CREB, as well as enzymes in the gluconeogenetic pathway, can contribute to stress-induced hypoglycemia. Furthermore, the thermogenic response was perturbed, as demonstrated by intolerance to acute cold exposure. This study highlights the necessity of DECR and the breakdown of unsaturated fatty acids in the transition of intermediary metabolism from the fed to the fasted state.


Hypoglycemia/physiopathology , Ketone Bodies/biosynthesis , Mitochondria/enzymology , Oxidoreductases Acting on CH-CH Group Donors/deficiency , Stress, Physiological , Animals , Fatty Acids, Unsaturated/metabolism , Female , Glucose/metabolism , Hypoglycemia/enzymology , Hypoglycemia/genetics , Hypoglycemia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Oxidation-Reduction , Oxidoreductases Acting on CH-CH Group Donors/genetics , Triglycerides/metabolism
10.
PLoS One ; 4(5): e5589, 2009.
Article En | MEDLINE | ID: mdl-19440339

It has been recently recognized that mammalian mitochondria contain most, if not all, of the components of fatty acid synthesis type II (FAS II). Among the components identified is 2-enoyl thioester reductase/mitochondrial enoyl-CoA reductase (Etr1/Mecr), which catalyzes the NADPH-dependent reduction of trans-2-enoyl thioesters, generating saturated acyl-groups. Although the FAS type II pathway is highly conserved, its physiological role in fatty acid synthesis, which apparently occurs simultaneously with breakdown of fatty acids in the same subcellular compartment in mammals, has remained an enigma. To study the in vivo function of the mitochondrial FAS in mammals, with special reference to Mecr, we generated mice overexpressing Mecr under control of the mouse metallothionein-1 promoter. These Mecr transgenic mice developed cardiac abnormalities as demonstrated by echocardiography in vivo, heart perfusion ex vivo, and electron microscopy in situ. Moreover, the Mecr transgenic mice showed decreased performance in endurance exercise testing. Our results showed a ventricular dilatation behind impaired heart function upon Mecr overexpression, concurrent with appearance of dysmorphic mitochondria. Furthermore, the data suggested that inappropriate expression of genes of FAS II can result in the development of hereditary cardiomyopathy.


Gene Expression , Heart Diseases/physiopathology , Mitochondrial Proteins/physiology , Myocardium/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Animals , Blotting, Northern , Blotting, Southern , Echocardiography , Exercise Test , Heart Diseases/genetics , In Situ Nick-End Labeling , Male , Mice , Mice, Transgenic , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxygen Consumption/genetics , Oxygen Consumption/physiology , Reverse Transcriptase Polymerase Chain Reaction , fas Receptor/genetics
11.
FASEB J ; 22(2): 569-78, 2008 Feb.
Article En | MEDLINE | ID: mdl-17898086

In bacteria, functionally related gene products are often encoded by a common transcript. Such polycistronic transcripts are rare in eukaryotes. Here we isolated several clones from human cDNA libraries, which rescued the respiratory-deficient phenotype of a yeast mitochondrial 3-hydroxyacyl thioester dehydratase 2 (htd2) mutant strain. All complementing cDNAs were derived from the RPP14 transcript previously described to encode the RPP14 subunit of the human ribonuclease P (RNase P) complex. We identified a second, 3' open reading frame (ORF) on the RPP14 transcript encoding a protein showing similarity to known dehydratases and hydratase 2 enzymes. The protein was localized in mitochondria, and the recombinant enzyme exhibited (3R)-specific hydratase 2 activity. Based on our results, we named the protein human 3-hydroxyacyl-thioester dehydratase 2 (HsHTD2), which is involved in mitochondrial fatty acid synthesis. The bicistronic arrangement of RPP14 and HsHTD2, as well as the general exon structure of the gene, is conserved in vertebrates from fish to humans, indicating a genetic link conserved for 400 million years between RNA processing and mitochondrial fatty acid synthesis.


Fatty Acids/biosynthesis , Mitochondria/genetics , Mitochondria/metabolism , RNA/genetics , Vertebrates/genetics , Vertebrates/metabolism , Amino Acid Sequence , Animals , Conserved Sequence , DNA, Complementary/genetics , Gene Expression Regulation, Enzymologic , Genome/genetics , Humans , Hydro-Lyases/chemistry , Hydro-Lyases/genetics , Hydro-Lyases/isolation & purification , Hydro-Lyases/metabolism , Mitochondrial Proteins , Molecular Sequence Data , Mutation/genetics , Open Reading Frames/genetics , Phylogeny , Ribonuclease P/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sequence Alignment , Transcription, Genetic/genetics
12.
J Biol Chem ; 278(22): 20154-61, 2003 May 30.
Article En | MEDLINE | ID: mdl-12654921

A data base search with YBR026c/MRF1', which encodes trans-2-enoyl thioester reductase of the intramitochondrial fatty acid synthesis (FAS) type II in yeast (Torkko, J. M., Koivuranta, K. T., Miinalainen, I. J., Yagi, A. I., Schmitz, W., Kastaniotis, A. J., Airenne, T. T., Gurvitz, A., and Hiltunen, K. J. (2001) Mol. Cell. Biol. 21, 6243-6253), revealed the clone AA393871 (HsNrbf-1, nuclear receptor binding factor 1) in human EST data bank. Expression of HsNrbf-1, tagged C-terminally with green fluorescent protein, in HeLa cells, resulted in a punctated fluorescence signal, superimposable with the MitoTracker Red dye. Wild-type polypeptide was immunoisolated from the extract of bovine heart mitochondria. Recombinant HsNrbf-1p reduces trans-2-enoyl-CoA to acyl-CoA with chain length from C6 to C16 in an NADPH-dependent manner with preference to medium chain length substrate. Furthermore, expression of HsNRBF-1 in the ybr026cDelta yeast strain restored mitochondrial respiratory function allowing growth on glycerol. These findings provide evidence that Nrbf-1ps act as a mitochondrial 2-enoyl thioester reductase, and mammalian cells may possess bacterial type fatty acid synthetase (FAS type II) in mitochondria, in addition to FAS type I in the cytoplasm.


Fatty Acid Desaturases/chemistry , NADH, NADPH Oxidoreductases , Saccharomyces cerevisiae/enzymology , Amino Acid Sequence , Animals , Base Sequence , Cattle , DNA Primers , Fatty Acid Desaturases/genetics , Fatty Acid Desaturases/isolation & purification , Fatty Acid Desaturases/metabolism , HeLa Cells , Humans , Microscopy, Fluorescence , Mitochondria, Heart/enzymology , Molecular Sequence Data , Oxidoreductases Acting on CH-CH Group Donors , Sequence Homology, Amino Acid
...