Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Mol Nutr Food Res ; 67(23): e2300398, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37867207

ABSTRACT

SCOPE: Olive oil, rapeseed oil, and lard are dietary fats rich in monounsaturated fatty acids, but the effects of dietary oils enriched in monounsaturated fatty acids on hepatic lipid deposition have seldom been compared. METHODS AND RESULTS: Ninety 8-week-old C57BL/6J male mice are randomly divided into six groups and fed diets containing lard, rapeseed oil, or olive oil with a 10% or 45% fat energy supply for 16 weeks. Under high-fat conditions, serum total cholesterol levels in the lard and olive oil groups are significantly higher than those in the rapeseed oil group. Hepatic lipid content in the olive oil group is higher than that in the other two groups. Compared with rapeseed oil, lard increases the liver levels of arachidonic, palmitic, and myristic acids and decreases the levels of eicosapentaenoic linolenic acid and linoleic acid. Olive oil increases the liver levels of docosatrienoic, arachidonic, oleic, and myristic acids; maltose; and fructose and decreases the levels of eicosapentaenoic, linolenic, and linoleic acids. CONCLUSION: Olive oil probably causes hepatic lipid deposition in mice, which may enhance hepatic lipid synthesis by activating the starch and sucrose metabolic pathways. By contrast, rapeseed oil shows a significant anti-lipid deposition effect on the liver.


Subject(s)
Cholesterol , Glucose , Male , Animals , Mice , Olive Oil/pharmacology , Rapeseed Oil , Glucose/metabolism , Lipid Metabolism , Transcriptome , Mice, Inbred C57BL , Dietary Fats , Liver/metabolism , Fatty Acids, Monounsaturated/pharmacology , Myristic Acids/metabolism , Plant Oils/pharmacology , Fatty Acids/metabolism
2.
Biotechnol Bioeng ; 119(9): 2482-2493, 2022 09.
Article in English | MEDLINE | ID: mdl-35680651

ABSTRACT

High value unsaturated fatty acids can be produced by de novo synthesis in microalgal cells, especially via heterotrophic cultivation. Unfortunately, the lipid accumulation of heterotrophic microalgae cannot be improved efficiently in conventional ways. Here we reported heterotrophic Tribonema minus, a promising resource for the production of palmitoleic acid which has increasing demands in health service for patients with metabolic syndrome, as whole-cell biocatalyst to develop a novel way of shifting low value exogenous saturated fatty acids to high value ones. Results showed that myristic acid is the best precursor for whole-cell catalysis; it elevated the lipid content of T. minus to 42.2%, the highest among the tried precursors. The influences of cultivation condition on the utilization of extrinsic myristic acid and lipid accumulation were also determined. Under the optimized condition, the lipid content reached as high as 48.9%. In addition, our findings showed that ~13.0% of C16:1 in T. minus is derived from extrinsic myristic acid, and 30.1% of metabolized precursor is converted into heterologous fatty acids. Thus, a feasible approach for both increasing the value of low value saturated fatty acid by bioconversion and enhancing the lipid accumulation in microalgae is proposed by supplementing extrinsic myristic acid.


Subject(s)
Microalgae , Stramenopiles , Biofuels , Biomass , Catalysis , Fatty Acids/metabolism , Humans , Microalgae/metabolism , Myristic Acids/metabolism
3.
Mol Plant Microbe Interact ; 35(6): 464-476, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35285673

ABSTRACT

Arbuscular mycorrhizal fungi (AMF) colonize roots, where they provide nutrients in exchange for sugars and lipids. Because AMF lack genes for cytosolic fatty acid de novo synthase (FAS), they depend on host-derived fatty acids. AMF colonization is accompanied by expression of specific lipid genes and synthesis of sn-2 monoacylglycerols (MAGs). It is unknown how host-derived fatty acids are taken up by AMF. We describe the characterization of two AMP-binding domain protein genes from Rhizophagus irregularis, RiFAT1 and RiFAT2, with sequence similarity to Saccharomyces cerevisiae fatty acid transporter 1 (FAT1). Uptake of 13C-myristic acid (14:0) and, to a lesser extent, 13C-palmitic acid (16:0) was enhanced after expression of RiFAT1 or RiFAT2 in S. cerevisiae Δfat1 cells. The uptake of 2H-labeled fatty acids from 2H-myristoylglycerol or 2H-palmitoylglycerol was also increased after RiFAT1 and RiFAT2 expression in Δfat, but intact 2H-MAGs were not detected. RiFAT1 and RiFAT2 expression was induced in colonized roots compared with extraradical mycelium. 13C-label in the AMF-specific palmitvaccenic acid (16:1Δ11) and eicosatrienoic acid (20:3) were detected in colonized roots only when 13C2-acetate was supplemented but not 13C-fatty acids, demonstrating that de novo synthesized, host-derived fatty acids are rapidly taken up by R. irregularis from the roots. The results show that RiFAT1 and RiFAT2 are involved in the uptake of myristic acid (14:0) and palmitic acid (16:0), while fatty acids from MAGs are only taken up after hydrolysis. Therefore, the two proteins might be involved in fatty acid import into the fungal arbuscules in colonized roots.[Formula: see text] Copyright © 2022 The Author(s). This is an open access article distributed under the CC BY-NC-ND 4.0 International license.


Subject(s)
Glomeromycota , Mycorrhizae , Saccharomyces cerevisiae Proteins , Adenosine Monophosphate/metabolism , Carrier Proteins/metabolism , Fatty Acid Transport Proteins/metabolism , Fatty Acids/metabolism , Fungi , Glomeromycota/genetics , Glomeromycota/metabolism , Myristic Acids/metabolism , Palmitic Acids/metabolism , Plant Roots/microbiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
4.
J Biomol Screen ; 18(3): 341-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23015018

ABSTRACT

LpxA, the first enzyme in the biosynthetic pathway for the Lipid A component of the outer membrane lipopolysaccharide in Gram-negative bacteria, is a potential target for novel antibacterial drug discovery. A fluorescence polarization assay was developed to facilitate high-throughput screening for competitive inhibitors of LpxA. The assay detects displacement of a fluorescently labeled peptide inhibitor, based on the previously reported inhibitor peptide 920, by active site ligands. The affinity of the fluorescent ligand was increased ~10-fold by acyl carrier protein (ACP). Competition with peptide binding was observed with UDP-N-acetylglucosamine (IC(50) ~6 mM), UDP-3-O-(R-3-hydroxymyristoyl)-N-acetylglucosamine (IC(50) ~200 nM), and DL-3-hydroxymyristic acid (IC(50) ~50 µM) and peptide 920 (IC(50) ~600 nM). The IC(50)s were not significantly affected by the presence of ACP.


Subject(s)
Acyltransferases/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Escherichia coli/enzymology , Fluorescence Polarization/methods , High-Throughput Screening Assays/methods , Acyl Carrier Protein/metabolism , Acyltransferases/chemistry , Binding, Competitive , Catalytic Domain , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Escherichia coli/drug effects , Escherichia coli/metabolism , Inhibitory Concentration 50 , Ligands , Lipid A/metabolism , Myristic Acids/chemistry , Myristic Acids/metabolism , Peptides/chemistry , Peptides/metabolism , Uridine Diphosphate N-Acetylglucosamine/analogs & derivatives , Uridine Diphosphate N-Acetylglucosamine/chemistry , Uridine Diphosphate N-Acetylglucosamine/metabolism
5.
J Biol Chem ; 279(21): 21849-56, 2004 May 21.
Article in English | MEDLINE | ID: mdl-15010473

ABSTRACT

Human ClC-2 Cl(-) (hClC-2) channels are activated by protein kinase A (PKA) and low extracellular pH(o). Both of these effects are prevented by the PKA inhibitor, myristoylated PKI. The aims of the present study were to identify the PKA phosphorylation site(s) important for PKA activation of hClC-2 at neutral and low pH(o) and to examine the relationship between PKA and low pH(o) activation. Recombinant hClC-2 with point mutations of consensus phosphorylation sites was prepared and stably expressed in HEK-293 cells. The responses to forskolin plus isobutylmethylxanthine at neutral and acidic pH(o) were studied by whole cell patch clamp in the presence and absence of phosphatase inhibitors. The double phosphorylation site (RRAT655(A) plus RGET691(A)) mutant hClC-2 lost PKA activation and low pH(o) activation. Either RRAT or RGET was sufficient for PKA activation of hClC-2 at pH(o) 7.4, as long as phosphatase inhibitors (cyclosporin A or endothal) were present. At pH(o) 6 only RGET was needed for PKA activation of hClC-2. Low pH(o) activation of hClC-2 Cl(-) channel activity was PKA-dependent, retained in RGET(A) mutant hClC-2, but lost in RRAT(A) mutant hClC-2. RRAT655(D) mutant hClC-2 was constitutively active and was further activated by PKA at pH(o) 7.4 and 6.0, consistent with the above findings. These results show that activation of hClC-2 is differentially regulated by PKA at two sites, RRAT655 and RGET691. Either RRAT655 or RGET691 was sufficient for activation at pH(o) 7.4. RGET, but not RRAT, was sufficient for activation at pH(o) 6.0. However, in the RGET691(D) mutant, there was PKA activation at pH(o) 6.0.


Subject(s)
Chloride Channels/chemistry , Cyclic AMP-Dependent Protein Kinases/chemistry , Arachidonic Acid/pharmacology , Binding Sites , CLC-2 Chloride Channels , Cell Line , Chlorides/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclosporine/pharmacology , DNA, Complementary/metabolism , Dicarboxylic Acids/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Hydrogen-Ion Concentration , Mutagenesis, Site-Directed , Mutation , Myristic Acids/metabolism , Patch-Clamp Techniques , Phosphorylation , Point Mutation , Recombinant Proteins/chemistry , Transfection
6.
Eur J Biochem ; 271(4): 863-74, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14764103

ABSTRACT

In order to determine the amino-terminal sequence requirements for protein N-myristoylation, site-directed mutagenesis of the N-terminal region was performed using tumor necrosis factor (TNF) mutants as model substrate proteins. Subsequently, the susceptibility of these mutants to protein N-myristoylation was evaluated by metabolic labeling in an in vitro translation system using rabbit reticulocyte lysate. A TNF mutant having the sequence MGAAAAAAAA at its N-terminus was used as the starting sequence to identify elements critical for protein N-myristoylation. Sequential vertical-scanning mutagenesis of amino acids at a distinct position in this model N-terminal sequence revealed the major sequence requirements for protein N-myristoylation: the combination of amino acids at position 3 and 6 constitutes a major determinant for the susceptibility to protein N-myristoylation. When Ser was located at position 6, 11 amino acids (Gly, Ala, Ser, Cys, Thr, Val, Asn, Leu, Ile, Gln, His) were permitted at position 3 to direct efficient protein N-myristoylation. In this case, the presence of Lys at position 7 was found to affect the amino acid requirement at position 3 and Lys became permitted at this position. When Ser was not located at position 6, only 3 amino acids (Ala, Asn, Gln) were permitted at position 3 to direct efficient protein N-myristoylation. The amino acid requirements found in this study were fully consistent with the N-terminal sequence of 78 N-myristoylated proteins in which N-myristoylation was experimentally verified. These observations strongly indicate that the combination of amino acids at position 3, 6 and 7 is a major determinant for protein N-myristoylation.


Subject(s)
Amino Acids/chemistry , Amino Acids/genetics , Myristic Acids/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , COS Cells , DNA Primers/genetics , DNA, Complementary/metabolism , Mutagenesis, Site-Directed , Precipitin Tests , Protein Biosynthesis , RNA, Messenger/genetics , Rabbits , Reticulocytes/metabolism , Transfection , Tritium , Tumor Necrosis Factor-alpha/chemistry
7.
Mol Cell Biochem ; 254(1-2): 9-19, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14674678

ABSTRACT

Rod outer segment membrane guanylate cyclase (ROS-GC) is a critical component of the vertebrate phototransduction machinery. In response to photoillumination, it senses a decline in free Ca(2+) levels from 500 to below 100 nM, becomes activated, and replenishes the depleted cyclic GMP pool to restore the dark state of the photoreceptor cell. It exists in two forms, ROS-GC1 and ROS-GC2. In outer segments, ROS-GCs sense fluctuations in Ca(2+) via two Ca(2+)-binding proteins, which have been termed GCAP1 and GCAP2. In the present study we report on the cloning of two ROS-GCs from the frog retinal cDNA library. These cyclases are the structural and functional counterparts of the mammalian ROS-GC1 and ROS-GC2. There is, however, an important difference between the regulation of mammalian and frog ROS-GC1: In contrast to the mammalian, the frog form does not require the myristoylated form of GCAP1 for its Ca(2+)-dependent modulation. This feature is not dependent upon the ability of frog GCAP1 to bind Ca(2+) because unmyristoylated GCAP1 mutants which do not bind Ca(2+), activate frog ROS-GC1. The findings establish frog as a suitable phototransduction model and show a facet of frog ROS-GC signaling, which is not shared by the mammalian form.


Subject(s)
Calcium/metabolism , Gene Expression Regulation , Guanylate Cyclase/biosynthesis , Guanylate Cyclase/physiology , Receptors, Cell Surface , Amino Acid Sequence , Animals , Cattle , Cell Line , Cloning, Molecular , Cyclic GMP/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Gene Library , Guanylate Cyclase/metabolism , Insecta , Molecular Sequence Data , Mutation , Myristic Acids/metabolism , Protein Binding , Ranidae , Rod Cell Outer Segment/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Transfection , Vision, Ocular
8.
Mol Genet Metab ; 73(3): 276-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11461195

ABSTRACT

Malonyl-CoA decarboxylase deficiency is a rare inborn error of metabolism. It has been suggested but never demonstrated that many of the clinical features arise due to inhibition of mitochondrial fatty acid oxidation by accumulated malonyl-CoA. We studied the oxidation of fatty acids in cultured skin fibroblasts from a recently described patient with malonyl-CoA decarboxylase deficiency. There was a marked reduction in the oxidation of palmitic and myristic acids both under baseline conditions and when the cells were cultured in the presence of high concentrations of acetate, a malonyl-CoA precursor. These results suggest that there is inhibition of fatty acid oxidation in malonyl-CoA decarboxylase deficiency and that this inhibition may be related to some of the clinical phenotypes.


Subject(s)
Carboxy-Lyases/deficiency , Fatty Acids/metabolism , Fibroblasts/metabolism , Mitochondria/metabolism , Oxygen/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Cells, Cultured , Child , DNA, Complementary/metabolism , Exons , Humans , Male , Myristic Acids/metabolism , Palmitic Acid/metabolism , Phenotype , Skin/cytology
9.
J Biol Chem ; 276(13): 10505-13, 2001 Mar 30.
Article in English | MEDLINE | ID: mdl-11124252

ABSTRACT

To examine the amino-terminal sequence requirements for cotranslational protein N-myristoylation, a series of site-directed mutagenesis of N-terminal region were performed using tumor necrosis factor as a nonmyristoylated model protein. Subsequently, the susceptibility of these mutants to protein N-myristoylation was evaluated by metabolic labeling in an in vitro translation system or in transfected cells. It was found that the amino acid residue at position 3 in an N-myristoylation consensus motif, Met-Gly-X-X-X-Ser-X-X-X, strongly affected the susceptibility of the protein to two different cotranslational protein modifications, N-myristoylation and N-acetylation; 10 amino acids (Ala, Ser, Cys, Thr, Val, Asn, Leu, Ile, Gln, and His) with a radius of gyration smaller than 1.80 A directed N-myristoylation, two negatively charged residues (Asp and Glu) directed N-acetylation, and two amino acids (Gly and Met) directed heterogeneous modification with both N-myristoylation and N-acetylation. The amino acid requirements at this position for the two modifications were dramatically changed when Ser at position 6 in the consensus motif was replaced with Ala. Thus, the amino acid residue penultimate to the N-terminal Gly residue strongly affected two cotranslational protein modifications, N-myristoylation and N-acetylation, and the amino acid requirements at this position for these two modifications were significantly affected by downstream residues.


Subject(s)
Amino Acids/chemistry , Glycine/chemistry , Myristic Acids/metabolism , Protein Biosynthesis , Acetylation , Amino Acid Motifs , Animals , Blotting, Western , COS Cells , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , Humans , Models, Genetic , Mutagenesis, Site-Directed , Mutation , Ovalbumin/metabolism , Plasmids/metabolism , Precipitin Tests , Rabbits , Reticulocytes/metabolism , Transfection , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
10.
J Biol Chem ; 276(13): 10320-9, 2001 Mar 30.
Article in English | MEDLINE | ID: mdl-11104758

ABSTRACT

Since little is known of how the primitive protozoan parasite, Giardia lamblia, senses and responds to its changing environment, we characterized a giardial protein kinase A (gPKA) catalytic subunit with unusual subcellular localization. Sequence analysis of the 1080-base pair open reading frame shows 48% amino acid identity with the cyclic AMP-dependent kinase from Euglena gracilis. Northern analysis indicated a 1.28- kilobase pair transcript at relatively constant concentrations during growth and encystation. gPKA is autophosphorylated, although amino acid residues corresponding to Thr-197 and Ser-338 of human protein kinase A (PKA) that are important for autophosphorylation are absent. Kinetic analysis of the recombinant PKA showed that ATP and magnesium are preferred over GTP and manganese. Kinase activity of the native PKA has also been detected in crude extracts using kemptide as a substrate. A myristoylated PKA inhibitor, amide 14-22, inhibited excystation with an IC(50) of 3 microm, suggesting an important role of gPKA during differentiation from the dormant cyst form into the active trophozoite. gPKA localizes independently of cell density to the eight flagellar basal bodies between the two nuclei together with centrin, a basal body/centrosome-specific protein. However, localization of gPKA to marginal plates along the intracellular portions of the anterior and caudal pairs of flagella was evident only at low cell density and higher endogenous cAMP concentrations or after refeeding with fresh medium. These data suggest an important role of PKA in trophozoite motility during vegetative growth and the cellular activation of excystation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/physiology , Giardia lamblia/enzymology , Movement/physiology , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Amino Acids/metabolism , Animals , Blotting, Northern , Blotting, Southern , Blotting, Western , Catalysis , Cell Differentiation , Centrosome/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA, Complementary/metabolism , Flagella/metabolism , Gene Deletion , Gene Library , Guanosine Triphosphate/metabolism , Inhibitory Concentration 50 , Kinetics , Microscopy, Fluorescence , Molecular Sequence Data , Myristic Acids/metabolism , Oligopeptides/pharmacology , Open Reading Frames , Phosphorylation , Protein Isoforms , RNA, Messenger/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Signal Transduction , Time Factors , Transcription, Genetic
11.
Biochem Biophys Res Commun ; 277(2): 361-7, 2000 Oct 22.
Article in English | MEDLINE | ID: mdl-11032731

ABSTRACT

We have reported that both dehydroepiandrosterone (DHEA) and dexamethasone (Dexa) directly activate PKC. In this study, we investigated the effects of these hormones on conventional PKC (cPKC) and atypical PKC (aPKC). DHEA and Dexa directly activated PKCbeta and PKCzeta to the same degree. In rat adipocytes, DHEA and Dexa activated endogenous immunoprecitable PKCzeta to 246 and 164%, respectively, from basal level (100%). In adipocytes, 5 min treatment with DHEA increased phosphatidylinositol 3-kinase (PI 3-kinase) activity in immunoprecipitate with anti-phosphotyrotyrosine antibody to 235%. Preincubation with wortmannin, myristoylated PKCzeta pseudosubstrate, but not with Go6976, abolished DHEA-induced 2-deoxyglucose (DOG) uptake. cPKC inhibitors prevented Dexa-induced insulin resistance. Moreover, DHEA and Dexa increased DOG uptake to 330 and 220%, respectively, in adipocytes overexpressed with wild-type PKCzeta, but not in those overexpressed with dominant negative. These results indicate that DHEA and Dexa activate both cPKC and aPKC, and Dexa-induced cPKC activation may lead to insulin resistance. In contrast, DHEA may mimic or enhance insulin action via PI 3-kinase and aPKC.


Subject(s)
Dehydroepiandrosterone/pharmacology , Dexamethasone/pharmacology , Glucose/pharmacokinetics , Insulin Resistance/physiology , Protein Kinase C/physiology , Adipocytes/metabolism , Adjuvants, Immunologic/pharmacology , Androstadienes/pharmacology , Animals , Antimetabolites/pharmacokinetics , Carbazoles/pharmacology , Cells, Cultured , Deoxyglucose/pharmacokinetics , Enzyme Activation , Enzyme Inhibitors/pharmacology , Genes, Dominant , Glucocorticoids/pharmacology , Indoles/pharmacology , Insulin Antagonists/pharmacology , Male , Myristic Acids/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Precipitin Tests , Protein Kinase C/genetics , Protein Kinase C/metabolism , Rats , Time Factors , Transfection , Wortmannin
12.
Food Chem Toxicol ; 36(9-10): 771-9, 1998.
Article in English | MEDLINE | ID: mdl-9737424

ABSTRACT

Two experiments with Sprague Dawley rats tested their ability to hydrolyse myristoyl-methionine (M-M) into myristic acid and L-methionine (M). In the first experiment, lasting for 3 days. male rats were orally administered [9,10-3H]myristoyl-L-[35S]methionine. The recovery of radioactivity was approximately 90% for both isotopes; 19% of the administered 3H was recovered in the urine and 16% in the faeces, while the recovered 35S activity was 13 and 12%, respectively. The balance of the radioactivity was found among the tissues, organs and blood. In the second experiment, male and female rats received soybean-based diets which were supplemented with either 0.305% M-M or 0.2% M (both diets contained equal amounts of M) for periods up to 4 weeks. The growth rate of the rats receiving the 0.305% M-M diets was slightly slower than that for the rats on the 0.2% M diet, but the difference was not statistically significant (P > 0.05). The M-M rats had a transitory decrease in feed consumption, suggesting that palatability may have contributed to the growth difference and that a somewhat greater amount of M-M was necessary for the rat to attain the same growth rate as that produced by 0.2% M. When the amount of dietary M-M was increased to 3.05% M-M, a greater reduction in feed consumption and body weight gain was observed. This latter diet was an initial attempt to study the potential toxicity of M-M. None of the haematological, clinical chemistry or organ weight data suggested that M-M was overtly toxic per se, but longer-term feeding studies are needed to evaluate the potential toxicity of M-M more fully.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Methionine/analogs & derivatives , Myristic Acids/metabolism , Administration, Oral , Animal Feed , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Biomarkers/blood , Biomarkers/urine , Body Weight/drug effects , Diet , Dietary Supplements , Eating/drug effects , Feces/chemistry , Female , Male , Methionine/metabolism , Methionine/pharmacokinetics , Methionine/toxicity , Myristic Acids/pharmacokinetics , Myristic Acids/toxicity , Rats , Rats, Sprague-Dawley , Sex Factors , Sulfur Radioisotopes , Tissue Distribution , Tritium
13.
Biochemistry ; 37(1): 193-200, 1998 Jan 06.
Article in English | MEDLINE | ID: mdl-9425039

ABSTRACT

N-Terminal myristoylation and thiopalmitoylation of the endothelial isoform of nitric oxide synthase (eNOS) are required for targeting the enzyme to specialized signal-transducing microdomains of plasma membrane termed caveolae. We have previously documented that the subcellular localization of eNOS is dynamically regulated by agonists such as bradykinin, which promotes enzyme depalmitoylation and translocation from caveolae. More recently, we have shown that association of eNOS with caveolin, the principal structural protein in caveolae, leads to enzyme inhibition, in a reversible process modulated by Ca2+-calmodulin (CaM). We now report studies of the respective roles of acylation and caveolin interaction for regulating eNOS activity. Using eNOS truncation and deletion mutants expressed in COS-7 cells, we have identified an obligatory role for the N-terminal half of eNOS in stabilizing its association with caveolin. By exploring the differential effects of detergents (CHAPS vs octyl glucoside), we have shown that this direct interaction between both proteins is facilitated by, but does not require, eNOS acylation, and, importantly, that treatment of intact aortic endothelial cells with the calcium ionophore A23187 leads to the rapid disruption of the eNOS-caveolin complexes. Finally, using transiently transfected COS-7 cells, we have observed that the myristoylation-deficient cytosol-restricted eNOS mutant (myr-) as well as the cytosolic fraction of the palmitoylation-deficient eNOS mutant (palm-) may both interact with caveolin; this association also leads to a marked inhibition of enzyme activity, which is completely reversed by addition of calmodulin. We conclude that the regulatory eNOS-caveolin association is independent of the state of eNOS acylation, indicating that agonist-evoked Ca2+/CaM-dependent disruption of the caveolin-eNOS complex, rather than agonist-promoted depalmitoylation of eNOS, relieves caveolin's tonic inhibition of enzyme activity. We therefore propose that caveolin may serve as an eNOS chaperone regulating NO production independently of the enzyme's residence within caveolae or its state of acylation.


Subject(s)
Caveolins , Endothelium, Vascular/enzymology , Membrane Proteins/metabolism , Nitric Oxide Synthase/metabolism , Acylation/drug effects , Amino Acid Sequence , Animals , Aorta , COS Cells , Calcimycin/pharmacology , Cattle , Caveolin 1 , Cells, Cultured , Endothelium, Vascular/cytology , Membrane Proteins/drug effects , Membrane Proteins/physiology , Molecular Sequence Data , Mutagenesis, Site-Directed , Myristic Acids/metabolism , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/genetics , Palmitic Acids/metabolism , Polymers/metabolism , Sequence Deletion
14.
Neuroreport ; 8(4): 937-40, 1997 Mar 03.
Article in English | MEDLINE | ID: mdl-9141068

ABSTRACT

The neuronal-specific G protein Gz is known to interact with a large variety of receptors for neurotransmitters and hormones. Fatty acylations on the N-terminus of the alpha subunit of Gz (alpha z) provide anchorage to the plasma membrane. Fatty acylation-deficient mutants of alpha z have previously been shown to exhibit altered signaling properties. Since the N-terminus of alpha z is likely to play a critical role in beta gamma binding, we examined the ability of these mutants to interact with beta gamma subunits by means of receptor-mediated stimulation of beta gamma-sensitive type II adenylyl cyclase. Our results indicate that lack of myristoylation, but not lack of palmitoylation, impaired the ability of alpha z to mediate receptor-induced release of beta gamma subunits.


Subject(s)
GTP-Binding Proteins/metabolism , Mutagenesis, Site-Directed , Receptors, Opioid, delta/physiology , Acylation , Adenylate Cyclase Toxin , Adenylyl Cyclases/metabolism , Animals , COS Cells , Cyclic AMP/metabolism , DNA, Complementary , Enkephalin, D-Penicillamine (2,5)- , Enkephalins/pharmacology , GTP-Binding Proteins/biosynthesis , GTP-Binding Proteins/genetics , Macromolecular Substances , Myristic Acid , Myristic Acids/metabolism , Neurons/metabolism , Palmitic Acid/metabolism , Point Mutation , Receptors, Opioid, delta/biosynthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Signal Transduction , Transfection , Virulence Factors, Bordetella/pharmacology
15.
Eur J Clin Invest ; 26(9): 755-62, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8889437

ABSTRACT

To assess the influence of dietary fat composition on the contribution of dietary myristic and palmitic acid to total fat oxidation and energy production, eight healthy men consumed diets containing 40% of total energy as fat, largely as either butter, tallow or corn oil, for 11 days. On days 8 and 11 of each diet, [1-13C]-myristic or [1-13C]-palmitic acid (20 mg kg-1 body weight) was ingested mixed with the test breakfast meal. Respiratory gas exchange was measured before, and for 9h after, consumption of the meal. Breath 13CO2 enrichments were determined hourly by isotope ratio mass spectrometry. Cumulative 9-h percentage oxidation of dietary myristic acid exceeded that of palmitic acid (P < 0.01), but neither was influenced by fat treatment [n = 8, 7.1% (1.0) (SEM), 8.6% (0.9) and 8.9% (0.6) of dietary myristic acid and 3.3% (0.7), 3.0% (0.9), and 2.5% (0.6) of dietary palmitic acid from butter, tallow and corn oil meals respectively]. Net dietary myristic acid oxidation was greater (P < 0.05) after consumption of the meal high in butter than after consumption of other fats. Net dietary palmitic acid oxidation was similar after consumption of all test meals. Precedent fat treatment had no measurable effect on net fat or carbohydrate oxidation or energy expenditure. The overall contribution of dietary myristic or palmitic acid to total fat oxidation did not exceed 1% over 9 h for any dietary fat. These results suggest that, although dietary fatty acid content is the principal determinant of net dietary fatty acid oxidation, dietary fat sources with moderate differences in fat composition do not measurably alter total energy or substrate utilization after a meal.


Subject(s)
Dietary Fats/metabolism , Myristic Acids/metabolism , Palmitic Acid/metabolism , Adult , Butter , Corn Oil/chemistry , Corn Oil/metabolism , Eating , Fats/chemistry , Fats/metabolism , Fatty Acids/analysis , Humans , Male , Myristic Acid , Oxidation-Reduction , Postprandial Period
16.
Plant Physiol ; 110(1): 203-10, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8587983

ABSTRACT

The seed oil of Cuphea palustris has an unusual fatty-acyl composition, whereby the principal fatty-acyl groups, myristate (64%) and caprylate (20%), differ by more than two methylenes. We have isolated two thioesterase (TE) cDNAs from C. palustris, encoding proteins designated Cp FatB1 and Cp FatB2, which, when expressed in Escherichia coli, have TE activities specific for 8:0/10:0- and 14:0/16:0-acyl carrier protein substrates, respectively. The specific activities of the recombinant affinity-purified enzymes indicate that Cp FatB2 is kinetically superior to Cp FatB1. This result is consistent with the predominance of 14:0 in the seed oil, despite apparently equal mRNA abundance of the two transcripts in the seed. In C. palustris the expression of both sequences is confined to the seed tissues. Based on these findings we propose that these two enzymes are major factors determining the bimodal chain-length composition of C. palustris oil. Analysis of the immature and mature seed oil by reverse-phase high-performance liquid chromatography confirmed that the principal triglycerides contain both 8:0 and 14:0. This result indicates that both fatty acids are synthesized at the same time and in the same cells at all developmental stages during oil deposition, suggesting that the two TEs act together in the same fatty acid synthesis system.


Subject(s)
Fatty Acids/biosynthesis , Plant Oils/chemistry , Plant Proteins , Plants/genetics , Seeds/chemistry , Thiolester Hydrolases/genetics , Amino Acid Sequence , Base Sequence , Caprylates/metabolism , DNA, Complementary/genetics , Escherichia coli/genetics , Gene Library , Molecular Sequence Data , Myristic Acid , Myristic Acids/metabolism , Plants/chemistry , Plants/enzymology , Protein Structure, Secondary , RNA, Messenger/analysis , RNA, Plant/analysis , Recombinant Proteins/metabolism , Sequence Analysis, DNA , Substrate Specificity , Thiolester Hydrolases/metabolism , Tissue Distribution , Triglycerides/chemistry
17.
J Dairy Sci ; 78(4): 872-9, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7790578

ABSTRACT

The objective of this work was to predict changes in milk fat composition caused by differences in dietary fat. Twenty-two references describing 35 experiments and 108 treatments were used in the analysis. For lauric, myristic, and palmitic acids in milk, proportions in the dietary fat and the total dietary fat concentration were important predictors for their concentrations in milk as well as for stearic and oleic acids in milk. Using a model that included these four parameters, the residual standard deviation around the observed versus predicted line within experiments was approximately 10% of the mean for short-chain fatty acids (< C12); for lauric, myristic, palmitic, and oleic acid; and for total C18 fatty acids in milk. The model also effectively predicted milk fatty acid profile with respect to lauric, myristic, palmitic, and oleic acid and total C18 fatty acids across experiments despite differences in breed, basal diet, and milk yield among experiments. The content of short-chain acids, stearic acid, and poly-unsaturated fatty acids were less effectively predicted across experiments. Possible explanations for the differing predictabilities for different milk fatty acids are discussed.


Subject(s)
Cattle/metabolism , Dietary Fats/administration & dosage , Fatty Acids/metabolism , Milk/metabolism , Animals , Dietary Fats/analysis , Fatty Acids/analysis , Female , Lactation , Lauric Acids/metabolism , Myristic Acid , Myristic Acids/metabolism , Palmitic Acid , Palmitic Acids/metabolism , Regression Analysis
18.
Am J Clin Nutr ; 60(6 Suppl): 986S-990S, 1994 12.
Article in English | MEDLINE | ID: mdl-7977157

ABSTRACT

Stearic acid is a long-chain saturated fatty acid. However, in contrast with other saturated fatty acids, stearic acid apparently does not raise serum cholesterol concentrations. Studies carried out three decades ago provided strong suggestive evidence that this was the case. More recent investigations that specifically compared stearic acid with other fatty acids in human studies have confirmed that stearic acid is not hypercholesterolemic. Stearic acid was shown not to raise low-density-lipoprotein cholesterol relative to oleic acid, which is known to be neutral in its effects on cholesterol concentrations. In contrast, palmitic acid, another long-chain saturated fatty acid, definitely raises cholesterol concentrations. For this reason, fats rich in stearic acid might be used in place of those high in palmitic acid in cholesterol-lowering diets.


Subject(s)
Cholesterol/blood , Dietary Fats/metabolism , Fatty Acids/metabolism , Stearic Acids/metabolism , Animals , Dietary Carbohydrates/metabolism , Fatty Acids, Monounsaturated/metabolism , Fatty Acids, Unsaturated/metabolism , Humans , Lauric Acids/metabolism , Myristic Acid , Myristic Acids/metabolism , Palmitic Acid , Palmitic Acids/metabolism
19.
Biochem J ; 303 ( Pt 3): 697-700, 1994 Nov 01.
Article in English | MEDLINE | ID: mdl-7980434

ABSTRACT

The alpha-subunit of the G-protein Gi1 carries two fatty acyl moieties covalently bound to its N-terminal region: myristic acid is linked to glycine-2 and palmitic acid is linked to cysteine-3. Using site-directed mutagenesis on a cDNA construct of alpha i1 we have generated an alpha i1-G2A mutant, carrying alanine instead of glycine at position 2, and alpha i1-C3S mutant, in which serine replaced cysteine-3 and a double mutant with both substitutions (alpha i1-G2A/C3S). These constructs were individually expressed by transfection in Cos-7 cells, and incorporation of fatty acids into the various mutants was compared with wild-type alpha i1 monitoring metabolic labelling with [3H]palmitate or [3H]myristate. The disruption of the palmitoylation site in alpha i1-C3S did not influence myristoylation, whereas prevention of myristoylation in alpha i1-G2A also abolished palmitoylation. Co-translational myristoylation is thus an absolute requirement for alpha i1 to be post-translationally palmitoylated. The non-palmitoylated alpha i1-C3S showed reduced membrane binding to the same extent as the non-myristoylated/non-palmitoylated alpha i1-G2A and alpha i1-G2A/C3S mutants, indicating that the attachment of palmitic acid is necessary for proper interaction with the membrane.


Subject(s)
Fatty Acids/metabolism , GTP-Binding Proteins/metabolism , Myristic Acids/metabolism , Palmitic Acids/metabolism , Acylation , Animals , Base Sequence , Binding Sites , Cell Line , DNA, Complementary , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Myristic Acid , Palmitic Acid , Rats
20.
J Cell Biol ; 127(3): 751-62, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7962057

ABSTRACT

Saccharomyces cerevisiae has been used as a model for studying the regulation of protein N-myristoylation. MyristoylCoA:protein N-myristoyl-transferase (Nmt1p), is essential for vegetative growth and uses myristoylCoA as its substrate. MyristoylCoA is produced by the fatty acid synthetase (Fas) complex and by cellular acylCoA synthetases. We have recently isolated three unlinked Fatty Acid Activation (FAA) genes encoding long chain acylCoA synthetases and have now recovered a fourth by genetic complementation. When Fas is active and NMT1 cells are grown on media containing a fermentable carbon source, none of the FAA genes is required for vegetative growth. When Fas is inactivated by a specific inhibitor (cerulenin), NMT1 cells are not viable unless the media is supplemented with long chain fatty acids. Supplementation of cellular myristoylCoA pools through activation of imported myristate (C14:0) is predominantly a function of Faa1p, although Faa4p contributes to this process. Cells with nmt181p need larger pools of myristoylCoA because of the mutant enzyme's reduced affinity for this substrate. Faa1p and Faa4p are required for maintaining the viability of nmt1-181 strains even when Fas is active. Overexpression of Faa2p can rescue nmt1-181 cells due to activation of an endogenous pool of C14:0. This pool appears to be derived in part from membrane phospholipids since overexpression of Plb1p, a nonessential lysophospholipase/phospholipase B, suppresses the temperature-sensitive growth arrest and C14:0 auxotrophy produced by nmt1-181. None of the four known FAAs is exclusively responsible for targeting imported fatty acids to peroxisomal beta-oxidation pathways. Introduction of a peroxisomal assembly mutation, pas1 delta, into isogenic NMT1 and nmt1-181 strains with wild type FAA alleles revealed that when Fas is inhibited, peroxisomes contribute to myristoylCoA pools used by Nmt1p. When Fas is active, a fraction of cellular myristoylCoA is targeted to peroxisomes. A NMT1 strain with deletions of all four FAAs is still viable at 30 degrees C on media containing myristate, palmitate, or oleate as the sole carbon source--indicating that S. cerevisiae contains at least one other FAA which directs fatty acids to beta-oxidation pathways.


Subject(s)
Fatty Acids/metabolism , Fungal Proteins/genetics , Genes, Fungal , Myristic Acids/metabolism , Protein Processing, Post-Translational , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Acyl Coenzyme A/metabolism , Amino Acid Sequence , Animals , Bacteria/genetics , Base Sequence , Fatty Acid Synthases/metabolism , Fungal Proteins/metabolism , Humans , Lipid Metabolism , Mammals , Molecular Sequence Data , Myristic Acid , Oligodeoxyribonucleotides , Phylogeny , Saccharomyces cerevisiae/growth & development , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL