Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Sci Rep ; 14(1): 11729, 2024 05 22.
Article in English | MEDLINE | ID: mdl-38778216

ABSTRACT

Filamentous fungi are eukaryotic microorganisms that differentiate into diverse cellular forms. Recent research demonstrated that phospholipid homeostasis is crucial for the morphogenesis of filamentous fungi. However, phospholipids involved in the morphological regulation are yet to be systematically analyzed. In this study, we artificially controlled the amount of phosphatidylcholine (PC), a primary membrane lipid in many eukaryotes, in a filamentous fungus Aspergillus oryzae, by deleting the genes involved in PC synthesis or by repressing their expression. Under the condition where only a small amount of PC was synthesized, A. oryzae hardly formed aerial hyphae, the basic structures for asexual development. In contrast, hyphae were formed on the surface or in the interior of agar media (we collectively called substrate hyphae) under the same conditions. Furthermore, we demonstrated that supplying sufficient choline to the media led to the formation of aerial hyphae from the substrate hyphae. We suggested that acyl chains in PC were shorter in the substrate hyphae than in the aerial hyphae by utilizing the strain in which intracellular PC levels were controlled. Our findings suggested that the PC levels regulate hyphal elongation and differentiation processes in A. oryzae and that phospholipid composition varied depending on the hyphal types.


Subject(s)
Aspergillus oryzae , Hyphae , Phosphatidylcholines , Hyphae/growth & development , Hyphae/metabolism , Phosphatidylcholines/metabolism , Aspergillus oryzae/metabolism , Aspergillus oryzae/genetics , Aspergillus oryzae/growth & development , Choline/metabolism , Gene Expression Regulation, Fungal , Fungal Proteins/metabolism , Fungal Proteins/genetics
2.
Sci Rep ; 13(1): 4285, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36922566

ABSTRACT

Conidiation is an important reproductive process in Aspergillus. We previously reported, in A. nidulans, that the deletion of a putative glycosyltransferase gene, rseA/cpsA, causes an increase in the production of extracellular hydrolases and a severe reduction in conidiation. The aim of this study was to obtain novel genetic factors involved in the repression of conidiation in the rseA deletion mutant. We isolated mutants in which the rseA deletion mutant conidiation defect is suppressed and performed a comparative genomic analysis of these mutants. A gene encoding a putative transcription factor was identified as the associated candidate causative gene. The candidate gene was designated as srdA (suppressor gene for the conidiation defect of the rseA deletion mutant). The conidiation efficiency of the rseAsrdA double-deletion mutant was increased. Introduction of wild-type srdA into the suppressor mutants caused a conidiation defect similar to that of the rseA deletion mutant. Notably, the conidiation efficiencies of the rseAsrdA double-deletion and srdA single-deletion mutants were higher than that of the wild-type strain. These results indicate that srdA is a novel genetic factor that strongly represses conidiation of the rseA deletion mutant, and a putative transcriptional regulator, SrdA is a negative regulator of conidiation in A. nidulans.


Subject(s)
Aspergillus nidulans , Aspergillus nidulans/genetics , Aspergillus nidulans/metabolism , Gene Expression Regulation, Fungal , Fungal Proteins/genetics , Fungal Proteins/metabolism , Mutation , Transcription Factors/metabolism , Spores, Fungal/genetics , Spores, Fungal/metabolism , Gene Deletion
3.
World J Microbiol Biotechnol ; 39(4): 97, 2023 Feb 14.
Article in English | MEDLINE | ID: mdl-36781616

ABSTRACT

Yarrowia lipolytica, a dimorphic yeast belonging to the Ascomycota, has potent abilities to utilize hydrophobic compounds, such as n-alkanes and fatty acids, as carbon and energy sources. Yarrowia lipolytica can synthesize and accumulate large amounts of lipids, making it a promising host to produce various lipids and convert n-alkanes to useful compounds. For advanced use of Y. lipolytica in these applications, it is necessary to understand the metabolism of these hydrophobic compounds in this yeast and the underlying molecular mechanisms. In this review, current knowledge on the n-alkane metabolism and how this is regulated in Y. lipolytica is summarized. Furthermore, recent studies revealed that lipid transfer proteins are involved in the utilization of n-alkanes and the regulation of cell morphology in response to n-alkanes. This review discusses the roles of membrane lipids in these processes in Y. lipolytica.


Subject(s)
Yarrowia , Yarrowia/metabolism , Alkanes/metabolism , Fatty Acids/metabolism
4.
FEMS Yeast Res ; 22(1)2022 02 15.
Article in English | MEDLINE | ID: mdl-35134922

ABSTRACT

The dimorphic yeast Yarrowia lipolytica has an ability to assimilate n-alkanes as carbon and energy sources. In this study, the roles of orthologs of Saccharomyces cerevisiae SEC14 family gene SFH2, which we named SFH21, SFH22, SFH23 and SFH24, of Y. lipolytica were investigated. The transcript levels of SFH21, SFH22 and SFH23, determined by RNA-seq analysis, qRT-PCR analysis and northern blot analysis, were found to increase in the presence of n-alkanes. The deletion mutant of SFH21, but not that of SFH22, SFH23 or SFH24, showed defects in growth in the media containing n-alkanes and in filamentous growth on the solid media containing n-alkanes. Additional deletions of SFH22 and SFH23 significantly exaggerated the defect in filamentous growth of the deletion mutant of SFH21, and expression of SFH22 or SFH24 using the SFH21 promoter partially suppressed the growth defect of the deletion mutant of SFH21 on n-alkanes. These results suggest that SFH2 orthologs are involved in the utilization of n-alkanes and filamentous growth in response to n-alkanes in Y. lipolytica.


Subject(s)
Saccharomyces cerevisiae Proteins , Yarrowia , Alkanes , Fungal Proteins/genetics , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Promoter Regions, Genetic , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Yarrowia/metabolism
5.
J Biosci Bioeng ; 131(6): 589-598, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33827772

ABSTRACT

Koji molds, such as Aspergillus oryzae and Aspergillus sojae, are used in the food industry in East Asia and have been explored for the large-scale production of extracellular hydrolases. We previously found that the deletion of a gene encoding a putative GT2 glycosyltransferase increased production of extracellular hydrolases in A. sojae. The gene was named rseA (regulator of the secretory enzyme A). We predicted that intracellular signaling pathways were involved in the increased production of hydrolases in the ΔrseA mutant of A. sojae. However, little has been reported on molecular biological knowledge about A. sojae. Hence, Aspergillus nidulans, a typical model organism used in molecular biology, was employed for the functional characterization of rseA in this study. Deletion of the rseA ortholog in A. nidulans induced increased extracellular production of hydrolases under the solid-state cultivation condition, similar to that in A. sojae. The involvement of the cell wall integrity pathway and the high osmolarity glycerol pathway in ΔrseA was further investigated. The results indicated that the HOG pathway played an important role in the increased extracellular production of hydrolases caused by the deletion of the rseA gene. rseA ortholog in A. nidulans was identical to cpsA, which was reported to function as a regulator of mycotoxin production, morphogenesis, and cell wall biosynthesis. However, this is the first study reporting that rseA/cpsA regulates extracellular hydrolase production in A. nidulans.


Subject(s)
Aspergillus nidulans/genetics , Glycerol/metabolism , Glycosyltransferases/genetics , Hydrolases/metabolism , Aspergillus/enzymology , Aspergillus/genetics , Aspergillus nidulans/metabolism , Cell Wall/metabolism , Culture Media/chemistry , Extracellular Space/enzymology , Extracellular Space/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Deletion , Glycosyltransferases/metabolism , Hydrolases/genetics , Metabolic Engineering/methods , Metabolic Networks and Pathways/genetics , Microbiological Techniques , Organisms, Genetically Modified , Osmolar Concentration , Secretory Pathway/genetics
6.
J Gen Appl Microbiol ; 67(1): 9-14, 2021 Apr 16.
Article in English | MEDLINE | ID: mdl-33100277

ABSTRACT

The yeast Yarrowia lipolytica assimilates hydrophobic compounds, such as n-alkanes and fatty acids, as sole carbon and energy sources. It has been shown that the acyl-CoA synthetase (ACS) genes, FAT1 and FAA1, are involved in the activation of fatty acids produced during the metabolism of n-alkanes, but the ACS genes that are involved in the metabolism of fatty acids from the culture medium remains to be identified. In this paper, we have identified the ACS genes involved in the utilization of exogenous fatty acids. RNA-seq analysis and qRT-PCR revealed that the transcript levels of the peroxisomal ACS-like protein-encoding genes AAL4 and AAL7 were increased in the presence of oleic acid. The single deletion mutant of AAL4 or AAL7 and double deletion mutant of AAL4 and AAL7 did not show any defects in the growth on the medium containing glucose, glycerol, n-alkanes, or fatty acids. In contrast, the mutant with deletion of seven genes, FAA1, FAT1-FAT4, AAL4, and AAL7, showed severe growth defects on the medium containing dodecanoic acid or oleic acid. These results suggest that Aal4p and Aal7p play important roles in the metabolism of exogenous fatty acids in collaboration with Faa1p and Fat1p-Fat4p.


Subject(s)
Coenzyme A Ligases/metabolism , Fatty Acids/metabolism , Yarrowia/genetics , Yarrowia/metabolism , Alkanes/metabolism , Coenzyme A Ligases/genetics , Fatty Acid Transport Proteins/genetics , Fatty Acid Transport Proteins/metabolism , Gene Deletion , Gene Expression Regulation, Fungal , Glycerol/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
7.
J Biosci Bioeng ; 131(2): 139-146, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33109479

ABSTRACT

Phosphatidylserine decarboxylases (PSDs) catalyze the production of phosphatidylethanolamine (PE) from phosphatidylserine (PS) and are crucial for the maintenance of PE levels in fungi. The PSDs are classified into two types; the type I PSDs are conserved from bacteria to humans, while the type II PSDs exist only in fungi and plants. In yeasts, the deletion of type I PSD-encoding genes causes severe growth retardation. In contrast, the deletion of type II PSD-encoding genes has little or no effect. In this study, we found four genes encoding type II PSD orthologs in the filamentous fungus Aspergillus nidulans; these included psdB, psdC, psdD, and psdE. Deletion of psdB caused severe growth defects on minimal medium and these defects were partially restored by the addition of ethanolamine, choline, PE, or phosphatidylcholine into the medium. The conidiation efficiency of the psdB deletion mutant was dramatically decreased and its conidiophore structures were aberrant. In the psdB deletion mutant, the PE content decreased while the PS content increased. We further showed that PsdB had a major PSD activity. Our findings suggest that the type II PSDs exert important roles in the phospholipid homeostasis, and in the growth and morphogenesis of filamentous fungi.


Subject(s)
Aspergillus nidulans/enzymology , Aspergillus nidulans/growth & development , Carboxy-Lyases/metabolism , Aspergillus nidulans/genetics , Carboxy-Lyases/deficiency , Carboxy-Lyases/genetics , Gene Deletion , Homeostasis , Humans , Morphogenesis
8.
PLoS One ; 14(4): e0215009, 2019.
Article in English | MEDLINE | ID: mdl-30958856

ABSTRACT

Interorganelle phospholipid transfer is critical for eukaryotic membrane biogenesis. In the yeast Saccharomyces cerevisiae, phosphatidylserine (PS) synthesized by PS synthase, Pss1, in the endoplasmic reticulum (ER) is decarboxylated to phosphatidylethanolamine (PE) by PS decarboxylase, Psd1, in the ER and mitochondria or by Psd2 in the endosome, Golgi, and/or vacuole, but the mechanism of interorganelle PS transport remains to be elucidated. Here we report that Sfh1, a member of Sec14 family proteins of S. cerevisiae, possesses the ability to enhance PE production by Psd2. Overexpression of SFH1 in the strain defective in Psd1 restored its growth on non-fermentable carbon sources and increased the intracellular and mitochondrial PE levels. Sfh1 was found to bind various phospholipids, including PS, in vivo. Bacterially expressed and purified Sfh1 was suggested to have the ability to transport fluorescently labeled PS between liposomes by fluorescence dequenching assay in vitro. Biochemical subcellular fractionation suggested that a fraction of Sfh1 localizes to the endosome, Golgi, and/or vacuole. We propose a model that Sfh1 promotes PE production by Psd2 by transferring phospholipids between the ER and endosome.


Subject(s)
Carboxy-Lyases/deficiency , Cell Cycle Proteins/biosynthesis , Chromosomal Proteins, Non-Histone/biosynthesis , Mitochondria/metabolism , Models, Biological , Oxygen Consumption , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae/metabolism , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endosomes/genetics , Endosomes/metabolism , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Mitochondria/genetics , Phosphatidylethanolamines/metabolism , Phosphatidylserines/genetics , Phosphatidylserines/metabolism , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Vacuoles/genetics , Vacuoles/metabolism
9.
FEBS Lett ; 592(11): 1829-1836, 2018 06.
Article in English | MEDLINE | ID: mdl-29782033

ABSTRACT

To elucidate the mechanism of acyl chain remodeling at the sn-1 position of phosphatidylcholine (PC), we investigated acyl chain introduction using a newly synthesized 1-hydroxy-2-hexadecyl-sn-glycero-3-phosphocholine (HHPC) in Saccharomyces cerevisiae. HHPC is incorporated into yeast cells and converted to a PC species containing acyl residues of 16 or 18 carbons. The efficiency of palmitoleic acid introduction to HHPCin vitro is lower in the reaction with the extract from the deletion mutant of ALE1, which encodes a membrane-bound O-acyltransferase, than in that with extracts from the wild-type strain. In addition, deletion of ALE1 causes reductions in the molecular species containing acyl residues in HHPC. These results reveal that ALE1 is involved in acyl chain transfer to the sn-1 position of HHPC in yeast.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Phosphatidylcholines/biosynthesis , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Gene Deletion , Phosphatidylcholines/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
10.
Biosci Biotechnol Biochem ; 82(9): 1633-1639, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29804512

ABSTRACT

DDL1 encodes a mitochondrial phospholipase A1 involved in acyl chain remodeling of mitochondrial phospholipids and degradation of cardiolipin in Saccharomyces cerevisiae. The deletion of DDL1 leads to respiratory growth defects. To elucidate the physiological role of DDL1, we screened for genes that, when overexpressed, suppress the respiratory growth defect of the DDL1 deletion mutant. Introduction of COQ8, COQ9, or COQ5, which are involved in coenzyme Q (CoQ) synthesis, using a multicopy vector suppressed the respiratory growth defect of the DDL1 deletion mutant. In contrast, introduction of COQ8 using a multicopy vector did not accelerate the growth of the deletion mutants of TAZ1 or CLD1, which encode an acyltransferase or phospholipase A2, respectively, involved in the remodeling of cardiolipin. These results suggest genetic interactions between the mitochondrial phospholipase A1 gene and the genes involved in CoQ synthesis.


Subject(s)
Gene Expression Regulation, Fungal , Genes, Fungal , Mitochondria/enzymology , Mutation , Phospholipases A1/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/metabolism , Ubiquinone/biosynthesis , Electron Transport , Gene Deletion , Genetic Vectors , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism
11.
Biochem Biophys Res Commun ; 499(4): 836-842, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29621549

ABSTRACT

In this study, we investigated the role of OSH6, which encodes a homolog of the oxysterol-binding protein, in the assimilation of n-alkanes in the yeast Yarrowia lipolytica. The deletion mutant of OSH6 showed growth defects on n-alkanes of 10-16 carbons. In the deletion mutant, production of the functional cytochrome P450 was not observed. However, transcription of ALK1, encoding a major P450 belonging to the CYP52 family that plays a critical role in n-alkane hydroxylation, and further translation of its transcript were noted in the deletion mutant as well as in the wild-type strain. The phospholipid composition was altered and, the ratio of phosphatidylserine (PS) was reduced by the deletion of OSH6. Residues involved in the transport of PS and phosphatidylinositol-4-phosphate in Osh6 of Saccharomyces cerevisiae are conserved in Y. lipolytica Osh6p and substitutions of these residues resulted in a defect in the n-alkane assimilation by Y. lipolytica. From these results, we propose a hypothesis that Osh6p provides an ideal endoplasmic reticulum membrane environment for Alk proteins to have a functional conformation via lipid transport activity in Y. lipolytica.


Subject(s)
Alkanes/metabolism , Cytochrome P-450 Enzyme System/metabolism , Receptors, Steroid/chemistry , Receptors, Steroid/metabolism , Sequence Homology, Amino Acid , Yarrowia/metabolism , Amino Acid Sequence , Biological Transport , Fungal Proteins/metabolism , Gene Deletion , Phosphatidylinositol Phosphates/metabolism , Phosphatidylserines/metabolism , Saccharomyces cerevisiae/metabolism , Yarrowia/growth & development
12.
J Biol Chem ; 293(15): 5636-5648, 2018 04 13.
Article in English | MEDLINE | ID: mdl-29487131

ABSTRACT

Sterols are present in eukaryotic membranes and significantly affect membrane fluidity, permeability, and microdomain formation. They are synthesized in the endoplasmic reticulum (ER) and transported to other organelles and the plasma membrane. Sterols play important roles in the biogenesis and maintenance of mitochondrial membranes. However, the mechanisms underlying ER-to-mitochondrion sterol transport remain to be identified. Here, using purified yeast membrane fractions enriched in ER and mitochondria, we show that the oxysterol-binding protein homologs encoded by the OSH genes in the yeast Saccharomyces cerevisiae mediate sterol transport from the ER to mitochondria. Combined depletion of all seven Osh proteins impaired sterol transport from the ER to mitochondria in vitro; however, sterol transport was recovered at different levels upon adding one of the Osh proteins. Of note, the sterol content in the mitochondrial fraction was significantly decreased in vivo after Osh4 inactivation in a genetic background in which all the other OSH genes were deleted. We also found that Osh5-Osh7 bind cholesterol in vitro We propose a model in which Osh proteins share a common function to transport sterols between membranes, with varying contributions by these proteins, depending on the target membranes. In summary, we have developed an in vitro system to examine intracellular sterol transport and provide evidence for involvement of Osh proteins in sterol transport from the ER to mitochondria in yeast.


Subject(s)
Carrier Proteins/metabolism , Cholesterol/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Receptors, Steroid/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Biological Transport, Active/physiology , Carrier Proteins/genetics , Cholesterol/genetics , Endoplasmic Reticulum/genetics , Fatty Acid-Binding Proteins , Gene Deletion , Membrane Proteins/genetics , Mitochondria/genetics , Receptors, Steroid/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
13.
Biochem Biophys Res Commun ; 488(1): 165-170, 2017 06 17.
Article in English | MEDLINE | ID: mdl-28483524

ABSTRACT

We investigated the role of FAD2, which was predicted to encode a fatty acid desaturase of the n-alkane-assimilating yeast Yarrowia lipolytica. Northern blot analysis suggested that FAD2 transcription was upregulated at low temperature or in the presence of n-alkanes or oleic acid. The FAD2 deletion mutant grew as well as the wild-type strain on glucose, n-alkanes, or oleic acid at 30 °C, but grew at a slower rate at 12 °C, when compared to the wild-type strain. The growth of the FAD2 deletion mutant at 12 °C was restored by the addition of 18:2, but not 18:1, fatty acids. The amount of 18:2 fatty acid in the wild-type strain was increased by the incubation at 12 °C and in the presence of n-octadecane. In contrast, 18:2 fatty acid was not detected in the deletion mutant of FAD2, confirming that FAD2 encodes the Δ12-fatty acid desaturase. These results suggest that Δ12-fatty acid desaturase is involved in the growth of Y. lipolytica at low temperature.


Subject(s)
Fatty Acid Desaturases/metabolism , Temperature , Yarrowia/enzymology , Yarrowia/growth & development , Fatty Acid Desaturases/deficiency , Fatty Acid Desaturases/genetics , Yarrowia/genetics , Yarrowia/metabolism
14.
Fungal Genet Biol ; 91: 43-54, 2016 06.
Article in English | MEDLINE | ID: mdl-27039152

ABSTRACT

Yarrowia lipolytica possesses twelve ALK genes, which encode cytochromes P450 in the CYP52 family. In this study, using a Y. lipolytica strain from which all twelve ALK genes had been deleted, strains individually expressing each of the ALK genes were constructed and their roles and substrate specificities were determined by observing their growth on n-alkanes and analyzing fatty acid metabolism. The results suggested that the twelve Alk proteins can be categorized into four groups based on their substrate specificity: Alk1p, Alk2p, Alk9p, and Alk10p, which have significant activities to hydroxylate n-alkanes; Alk4p, Alk5p, and Alk7p, which have significant activities to hydroxylate the ω-terminal end of dodecanoic acid; Alk3p and Alk6p, which have significant activities to hydroxylate both n-alkanes and dodecanoic acid; and Alk8p, Alk11p, and Alk12p, which showed faint or no activities to oxidize these substrates. The involvement of Alk proteins in the oxidation of fatty alcohols and fatty aldehydes was also analyzed by measuring viability of the mutant deleted for twelve ALK genes in medium containing dodecanol and by observing growth on dodecanal of a mutant strain, in which twelve ALK genes were deleted along with four fatty aldehyde dehydrogenase genes. It was suggested that ALK gene(s) is/are involved in the detoxification of dodecanol and the assimilation of dodecanal. These results imply that genes encoding CYP52-family P450s have undergone multiplication and diversification in Y. lipolytica for assimilation of various hydrophobic compounds.


Subject(s)
Aldehyde Dehydrogenase/genetics , Alkanes/metabolism , Cytochrome P-450 Enzyme System/genetics , Yarrowia/enzymology , Aldehydes/metabolism , Cytochrome P-450 Enzyme System/classification , Cytochrome P-450 Enzyme System/metabolism , Fatty Acids/metabolism , Fatty Alcohols/metabolism , Gene Deletion , Oxidation-Reduction , Substrate Specificity
15.
Yeast ; 32(12): 691-701, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26284565

ABSTRACT

In the n-alkane-assimilating yeast Yarrowia lipolytica, the transcription of ALK1, encoding cytochrome P450, that catalyses n-alkane hydroxylation is activated by a complex composed of Yas1p and Yas2p via a promoter element, ARE1, in response to n-alkanes. An Opi1-family transcription factor, Yas3p, represses the transcription by binding to Yas2p in the nucleus when cultured in glucose-containing medium, but it is localized to the ER, presumably through interaction with acidic phospholipids, phosphatidic acid and/or phospho inositides, when cultured in n-alkane-containing medium. Here, to elucidate the mechanisms regulating the localization of Yas3p, point and deletion mutants of Yas3p were constructed and analysed. The substitution of Trp(360) and Cys(361) by Arg abrogated the localization of Yas3p to the ER and decreased ARE1-mediated transcriptional activation by n-alkane. A Yas3p truncation mutant consisting of residues 259-422 did not bind to acidic phospholipids, but it was localized to the ER in the presence of n-alkane, implying the acidic-phospholipid-independent recruitment of this mutant to the ER in response to n-alkane. The W360R and C361R substitutions in this truncation mutant abolished its localization to the ER. The results suggest that these residues are implicated in the acidic phospholipid-independent interaction of Yas3p to the ER.


Subject(s)
Endoplasmic Reticulum/metabolism , Fungal Proteins/metabolism , Yarrowia/genetics , Yarrowia/metabolism , Alkanes/metabolism , Alkanes/pharmacology , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/genetics , Fungal Proteins/genetics , Gene Deletion , Liposomes/metabolism , Mutation , Phosphatidic Acids/metabolism , Phospholipids/metabolism , Phospholipids/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation , Yarrowia/enzymology , beta-Galactosidase/metabolism
16.
Biosci Biotechnol Biochem ; 79(10): 1608-14, 2015.
Article in English | MEDLINE | ID: mdl-26106800

ABSTRACT

To elucidate the mechanism of interorganelle sterol transport, a system to evaluate sterol transport from the endoplasmic reticulum (ER) to the mitochondria was constructed. A bacterial glycerophospholipid: cholesterol acyltransferase fused with a mitochondria-targeting sequence and a membrane-spanning domain of the mitochondrial inner membrane protein Pet100 and enhanced green fluorescent protein was expressed in a Saccharomyces cerevisiae mutant deleted for ARE1 and ARE2 encoding acyl-CoA:sterol acyltransferases. Microscopic observation and subcellular fractionation suggested that this fusion protein, which was named mito-SatA-EGFP, was localized in the mitochondria. Steryl esters were synthesized in the mutant expressing mito-SatA-EGFP. This system will be applicable for evaluations of sterol transport from the ER to the mitochondria in yeast by examining sterol esterification in the mitochondria.


Subject(s)
Endoplasmic Reticulum/metabolism , Gene Expression Regulation, Fungal , Mitochondria/metabolism , Saccharomyces cerevisiae/metabolism , Sterols/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Transport , Cholesterol Esters/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sterol O-Acyltransferase/deficiency , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism
17.
FEMS Yeast Res ; 15(4): fov031, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26019148

ABSTRACT

Here, we investigated the roles of YAL1 (FAA1) and FAT1 encoding acyl-CoA synthetases (ACSs) and three additional orthologs of ACS genes FAT2-FAT4 of the yeast Yarrowia lipolytica in the assimilation or utilization of n-alkanes and fatty acids. ACS deletion mutants were generated to characterize their function. The FAT1 deletion mutant exhibited decreased growth on n-alkanes of 10-18 carbons, whereas the FAA1 mutant showed growth reduction on n-alkane of 16 carbons. However, FAT2-FAT4 deletion mutants did not show any growth defects, suggesting that FAT1 and FAA1 are involved in the activation of fatty acids produced during the metabolism of n-alkanes. In contrast, deletions of FAA1 and FAT1-FAT4 conferred no defect in growth on fatty acids. The wild-type strain grew in the presence of cerulenin, an inhibitor of fatty acid synthesis, by utilizing exogenously added fatty acid or fatty acid derived from n-alkane when oleic acid or n-alkane of 18 carbons was supplemented. However, the FAA1 deletion mutant did not grow, indicating a critical role for FAA1 in the utilization of fatty acids. Fluorescent microscopic observation and biochemical analyses suggested that Fat1p is present in the peroxisome and Faa1p is localized in the cytosol and to membranes.


Subject(s)
Alkanes/metabolism , Coenzyme A Ligases/metabolism , Fatty Acids/metabolism , Yarrowia/enzymology , Yarrowia/metabolism , Coenzyme A Ligases/genetics , Culture Media/chemistry , Gene Deletion , Metabolic Networks and Pathways/genetics , Yarrowia/genetics , Yarrowia/growth & development
18.
FEMS Yeast Res ; 15(3)2015 May.
Article in English | MEDLINE | ID: mdl-25805841

ABSTRACT

The yeast Yarrowia lipolytica can assimilate hydrophobic substrates, including n-alkanes and fatty alcohols. Here, eight alcohol dehydrogenase genes, ADH1-ADH7 and FADH, and a fatty alcohol oxidase gene, FAO1, were analyzed to determine their roles in the metabolism of hydrophobic substrates. A mutant deleted for all of these genes (ALCY02 strain) showed severely defective growth on fatty alcohols, and enhanced sensitivity to fatty alcohols in glucose-containing media. The ALCY02 strain grew normally on n-tetradecane or n-hexadecane, but exhibited slightly defective growth on n-decane or n-dodecane. It accumulated more 1-dodecanol and less dodecanoic acid than the wild-type strain when n-dodecane was fed. Expression of ADH1, ADH3 or FAO1, but not that of other ADH genes or FADH, in the ALCY02 strain restored its growth on fatty alcohols. In addition, a triple deletion mutant of ADH1, ADH3 and FAO1 showed similarly defective growth on fatty alcohols and on n-dodecane to the ALCY02 strain. Microscopic observation suggests that Adh1p and Adh3p are localized in the cytosol and Fao1p is in the peroxisome. These results suggest that Adh1p, Adh3p and Fao1p are responsible for the oxidation of exogenous fatty alcohols but play less prominent roles in the oxidation of fatty alcohols derived from n-alkanes.


Subject(s)
Alcohol Dehydrogenase/metabolism , Alcohol Oxidoreductases/metabolism , Fatty Alcohols/metabolism , Yarrowia/enzymology , Yarrowia/metabolism , Alcohol Dehydrogenase/genetics , Alcohol Oxidoreductases/genetics , Culture Media/chemistry , Cytosol/chemistry , Gene Deletion , Microscopy, Confocal , Microscopy, Fluorescence , Peroxisomes/chemistry , Yarrowia/genetics , Yarrowia/growth & development
19.
J Biol Chem ; 289(48): 33275-86, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25315778

ABSTRACT

In the n-alkane assimilating yeast Yarrowia lipolytica, n-alkanes are oxidized to fatty acids via fatty alcohols and fatty aldehydes, after which they are utilized as carbon sources. Here, we show that four genes (HFD1-HFD4) encoding fatty aldehyde dehydrogenases (FALDHs) are involved in the metabolism of n-alkanes in Y. lipolytica. A mutant, in which all of four HFD genes are deleted (Δhfd1-4 strain), could not grow on n-alkanes of 12-18 carbons; however, the expression of one of those HFD genes restored its growth on n-alkanes. Production of Hfd2Ap or Hfd2Bp, translation products of transcript variants generated from HFD2 by the absence or presence of splicing, also supported the growth of the Δhfd1-4 strain on n-alkanes. The FALDH activity in the extract of the wild-type strain was increased when cells were incubated in the presence of n-decane, whereas this elevation in FALDH activity by n-decane was not observed in Δhfd1-4 strain extract. Substantial FALDH activities were detected in the extracts of Escherichia coli cells expressing the HFD genes. Fluorescent microscopic observation suggests that Hfd3p and Hfd2Bp are localized predominantly in the peroxisome, whereas Hfd1p and Hfd2Ap are localized in both the endoplasmic reticulum and the peroxisome. These results suggest that the HFD multigene family is responsible for the oxidation of fatty aldehydes to fatty acids in the metabolism of n-alkanes, and raise the possibility that Hfd proteins have diversified by gene multiplication and RNA splicing to efficiently assimilate or detoxify fatty aldehydes in Y. lipolytica.


Subject(s)
Aldehyde Oxidoreductases , Alkanes/metabolism , Fungal Proteins , Multigene Family/physiology , Yarrowia , Aldehyde Oxidoreductases/genetics , Aldehyde Oxidoreductases/metabolism , Aldehydes/metabolism , Base Sequence , Fatty Acids/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Molecular Sequence Data , Oxidation-Reduction , Yarrowia/enzymology , Yarrowia/genetics
20.
Biochem Biophys Res Commun ; 449(1): 26-31, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24802409

ABSTRACT

CTP:phosphoethanolamine cytidylyltransferase (ECT) is a key enzyme in the CDP-ethanolamine branch of the Kennedy pathway, which is the primary pathway of phosphatidylethanolamine (PE) synthesis in mammalian cells. Here, the enzymatic properties of recombinant human ECT (hECT) were characterized. The catalytic reaction of hECT obeyed Michaelis-Menten kinetics with respect to both CTP and phosphoethanolamine. hECT is composed of two tandem cytidylyltransferase (CT) domains as ECTs of other organisms. The histidines, especially the first histidine, in the CTP-binding motif HxGH in the N-terminal CT domain were critical for its catalytic activity in vitro, while those in the C-terminal CT domain were not. Overexpression of the wild-type hECT and hECT mutants containing amino acid substitutions in the HxGH motif in the C-terminal CT domain suppressed the growth defect of the Saccharomyces cerevisiae mutant of ECT1 encoding ECT in the absence of a PE supply via the decarboxylation of phosphatidylserine, but overexpression of hECT mutants of the N-terminal CT domain did not. These results suggest that the N-terminal CT domain of hECT contributes to its catalytic reaction, but C-terminal CT domain does not.


Subject(s)
Phosphatidylethanolamines/chemistry , RNA Nucleotidyltransferases/chemistry , RNA Nucleotidyltransferases/metabolism , Amino Acid Motifs , Binding Sites , Catalysis , Enzyme Activation , Humans , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...