Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Cell Mol Life Sci ; 71(5): 933-48, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23864032

ABSTRACT

Acyl-CoA thioesterase (ACOT) activities are found in prokaryotes and in several compartments of eukaryotes where they hydrolyze a wide range of acyl-CoA substrates and thereby regulate intracellular acyl-CoA/CoA/fatty acid levels. ACOT9 is a mitochondrial ACOT with homologous genes found from bacteria to humans and in this study we have carried out an in-depth kinetic characterization of ACOT9 to determine its possible physiological function. ACOT9 showed unusual kinetic properties with activity peaks for short-, medium-, and saturated long-chain acyl-CoAs with highest V max with propionyl-CoA and (iso) butyryl-CoA while K cat/K m was highest with saturated long-chain acyl-CoAs. Further characterization of the short-chain acyl-CoA activity revealed that ACOT9 also hydrolyzes a number of short-chain acyl-CoAs and short-chain methyl-branched CoA esters that suggest a role for ACOT9 in regulation also of amino acid metabolism. In spite of markedly different K ms, ACOT9 can hydrolyze both short- and long-chain acyl-CoAs simultaneously, indicating that ACOT9 may provide a novel regulatory link between fatty acid and amino acid metabolism in mitochondria. Based on similar acyl-CoA chain-length specificities of recombinant ACOT9 and ACOT activity in mouse brown adipose tissue and kidney mitochondria, we conclude that ACOT9 is the major mitochondrial ACOT hydrolyzing saturated C2-C20-CoA in these tissues. Finally, ACOT9 activity is strongly regulated by NADH and CoA, suggesting that mitochondrial metabolic state regulates the function of ACOT9.


Subject(s)
Amino Acids/metabolism , Fatty Acids/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Thiolester Hydrolases/genetics , Thiolester Hydrolases/metabolism , Animals , Base Sequence , Blotting, Western , Chromatography, High Pressure Liquid , Chromosome Mapping , Cloning, Molecular , Cluster Analysis , Computational Biology , DNA Primers/genetics , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Molecular Sequence Data , Phylogeny , Sequence Analysis, DNA , Spectrophotometry
2.
J Biol Chem ; 287(20): 16158-67, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22408254

ABSTRACT

Lysine acetylation is a major post-translational modification of proteins and regulates many physiological processes such as metabolism, cell migration, aging, and inflammation. Proteomic studies have identified numerous lysine-acetylated proteins in human and mouse models (Kim, S. C., Sprung, R., Chen, Y., Xu, Y., Ball, H., Pei, J., Cheng, T., Kho, Y., Xiao, H., Xiao, L., Grishin, N. V., White, M., Yang, X. J., and Zhao, Y. (2006) Mol. Cell 23, 607-618). One family of proteins identified in this study was the murine glycine N-acyltransferase (GLYAT) enzymes, which are acetylated on lysine 19. Lysine 19 is a conserved residue in human glycine N-acyltransferase-like 2 (hGLYATL2) and in several other species, showing that this residue may be important for enzyme function. Mutation of lysine 19 in recombinant hGLYATL2 to glutamine (K19Q) and arginine (K19R) resulted in a 50-80% lower production of N-oleoyl glycine and N-arachidonoylglycine, indicating that lysine 19 is important for enzyme function. LC/MS/MS confirmed that Lys-19 is not acetylated in wild-type hGLYATL2, indicating that Lys-19 requires to be deacetylated for full activity. The hGLYATL2 enzyme conjugates medium- and long-chain saturated and unsaturated acyl-CoA esters to glycine, resulting in the production of N-oleoyl glycine and also N-arachidonoyl glycine. N-Oleoyl glycine and N-arachidonoyl glycine are structurally and functionally related to endocannabinoids and have been identified as signaling molecules that regulate functions like the perception of pain and body temperature and also have anti-inflammatory properties. In conclusion, acetylation of lysine(s) in hGLYATL2 regulates the enzyme activity, thus linking post-translational modification of proteins with the production of biological signaling molecules, the N-acyl glycines.


Subject(s)
Acyltransferases/metabolism , Arachidonic Acids/biosynthesis , Glycine/analogs & derivatives , Oleic Acids/biosynthesis , Protein Processing, Post-Translational/physiology , Acetylation , Acyltransferases/genetics , Amino Acid Substitution , Animals , Arachidonic Acids/genetics , Glycine/biosynthesis , Glycine/genetics , HEK293 Cells , HeLa Cells , Humans , Mice , Mutation, Missense , Oleic Acids/genetics
3.
Biochim Biophys Acta ; 1822(9): 1397-410, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22465940

ABSTRACT

The importance of peroxisomes in lipid metabolism is now well established and peroxisomes contain approximately 60 enzymes involved in these lipid metabolic pathways. Several acyl-CoA thioesterase enzymes (ACOTs) have been identified in peroxisomes that catalyze the hydrolysis of acyl-CoAs (short-, medium-, long- and very long-chain), bile acid-CoAs, and methyl branched-CoAs, to the free fatty acid and coenzyme A. A number of acyltransferase enzymes, which are structurally and functionally related to ACOTs, have also been identified in peroxisomes, which conjugate (or amidate) bile acid-CoAs and acyl-CoAs to amino acids, resulting in the production of amidated bile acids and fatty acids. The function of ACOTs is to act as auxiliary enzymes in the α- and ß-oxidation of various lipids in peroxisomes. Human peroxisomes contain at least two ACOTs (ACOT4 and ACOT8) whereas mouse peroxisomes contain six ACOTs (ACOT3, 4, 5, 6, 8 and 12). Similarly, human peroxisomes contain one bile acid-CoA:amino acid N-acyltransferase (BAAT), whereas mouse peroxisomes contain three acyltransferases (BAAT and acyl-CoA:amino acid N-acyltransferases 1 and 2: ACNAT1 and ACNAT2). This review will focus on the human and mouse peroxisomal ACOT and acyltransferase enzymes identified to date and discuss their cellular localizations, emerging structural information and functions as auxiliary enzymes in peroxisomal metabolic pathways.


Subject(s)
Acyltransferases/physiology , Lipid Metabolism , Palmitoyl-CoA Hydrolase/physiology , Peroxisomes/enzymology , Acyl Coenzyme A/metabolism , Acyltransferases/genetics , Acyltransferases/metabolism , Animals , Bile Acids and Salts/metabolism , Cholic Acids/blood , Cholic Acids/genetics , Humans , Hydrolysis , Models, Molecular , Palmitoyl-CoA Hydrolase/chemistry , Palmitoyl-CoA Hydrolase/metabolism , Peroxisomes/metabolism , Protein Conformation , Steroid Metabolism, Inborn Errors/enzymology , Steroid Metabolism, Inborn Errors/genetics
4.
Biochem Biophys Res Commun ; 430(1): 54-9, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23159632

ABSTRACT

Pancreatic ß-cells secrete insulin in response to various stimuli to control blood glucose levels. This insulin release is the result of a complex interplay between signaling, membrane potential and intracellular calcium levels. Various nutritional and hormonal factors are involved in regulating this process. N-Acyl taurines are a group of fatty acids which are amidated (or conjugated) to taurine and little is known about their physiological functions. In this study, treatment of pancreatic ß-cell lines (HIT-T15) and rat islet cell lines (INS-1) with N-acyl taurines (N-arachidonoyl taurine and N-oleoyl taurine), induced a high frequency of calcium oscillations in these cells. Treatment with N-arachidonoyl taurine and N-oleoyl taurine also resulted in a significant increase in insulin secretion from pancreatic ß-cell lines as determined by insulin release assay and immunofluorescence (p<0.05). Our data also show that the transient receptor potential vanilloid 1 (TRPV1) channel is involved in insulin secretion in response to N-arachidonoyl taurine and N-oleoyl taurine treatment. However our data also suggest that receptors other than TRPV1 are involved in the insulin secretion response to treatment with N-oleoyl taurine.


Subject(s)
Arachidonic Acids/pharmacology , Calcium/metabolism , Insulin-Secreting Cells/drug effects , Insulin/metabolism , Oleic Acids/pharmacology , Taurine/analogs & derivatives , Animals , Cell Line , Cricetinae , Cytoplasm/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Rats , TRPV Cation Channels/metabolism , Taurine/pharmacology
5.
FASEB J ; 24(8): 2795-803, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20305126

ABSTRACT

The discovery of glycine conjugates of long-chain fatty acids (N-acyl glycines) in the brain and other non-neuronal tissues has led to the identification of an emerging class of bioactive lipids. The biological activities of N-acyl glycines include antinociceptive, anti-inflammatory and antiproliferative effects, and activation of G-protein-coupled receptors. However, despite the fact that N-acyl glycines are emerging as a distinct lipid signaling family, pathways for their production are not fully elucidated. Here we report on the characterization of human glycine N-acyltransferase-like 2 (hGLYATL2), a member of a gene family of 4 putative glycine conjugating enzymes, and show that it synthesizes various N-acyl glycines. Recombinantly expressed hGLYATL2 efficiently conjugated oleoyl-CoA, arachidonoyl-CoA, and other medium- and long-chain acyl-CoAs to glycine. The enzyme was specific for glycine as an acceptor molecule, and preferentially produced N-oleoyl glycine. The hGLYATL2 enzyme is localized to the endoplasmic reticulum, and the mRNA shows highest expression in salivary gland and trachea, but is also detected in spinal cord and skin fibroblasts. The expression pattern and the identification of high levels of N-acyl glycines in skin and lung may indicate a role for N-acyl glycines in barrier function/immune response and the potential role of hGLYATL2 in this regard is discussed.


Subject(s)
Acyltransferases/metabolism , Acyl Coenzyme A/metabolism , Acyltransferases/genetics , Glycine/analogs & derivatives , Glycine/biosynthesis , Humans , Lung/chemistry , RNA, Messenger/analysis , Skin/chemistry , Tissue Distribution
6.
Biochem Biophys Res Commun ; 388(2): 355-9, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19666004

ABSTRACT

The distribution of some enzymes between peroxisomes and cytosol, or a dual localization in both these compartments, can be difficult to reconcile. We have used photobleaching in live cells expressing green fluorescent protein (GFP)-fusion proteins to show that imported bona fide peroxisomal matrix proteins are retained in the peroxisome. The high mobility of the GFP-fusion proteins in the cytosol and absence of peroxisomal escape makes it possible to eliminate the cytosolic fluorescence by photobleaching, to distinguish between exclusively cytosolic proteins and proteins that are also present at low levels in peroxisomes. Using this technique we found that GFP tagged bile acid-CoA:amino acid N-acyltransferase (BAAT) was exclusively localized in the cytosol in HeLa cells. We conclude that the cytosolic localization was due to its carboxyterminal non-consensus peroxisomal targeting signal (-SQL) since mutation of the -SQL to -SKL resulted in BAAT being efficiently imported into peroxisomes.


Subject(s)
Acyltransferases/analysis , Peroxisomes/enzymology , Acyltransferases/genetics , Acyltransferases/metabolism , Amino Acid Sequence , Amino Acid Substitution , Cytosol/enzymology , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Photobleaching , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
7.
Front Microbiol ; 10: 1701, 2019.
Article in English | MEDLINE | ID: mdl-31428062

ABSTRACT

Elastase B (lasB) is a multifunctional metalloenzyme secreted by the gram-negative pathogen Pseudomonas aeruginosa, and this enzyme orchestrates several physiopathological events during bacteria-host interplays. LasB is considered to be a potential target for the development of an innovative chemotherapeutic approach, especially against multidrug-resistant strains. Recently, our group showed that 1,10-phenanthroline-5,6-dione (phendione), [Ag(phendione)2]ClO4 (Ag-phendione) and [Cu(phendione)3](ClO4)2.4H2O (Cu-phendione) had anti-P. aeruginosa action against both planktonic- and biofilm-growing cells. In the present work, we have evaluated the effects of these compounds on the (i) interaction with the lasB active site using in silico approaches, (ii) lasB proteolytic activity by using a specific fluorogenic peptide substrate, (iii) lasB gene expression by real time-polymerase chain reaction, (iv) lasB protein secretion by immunoblotting, (v) ability to block the damages induced by lasB on a monolayer of lung epithelial cells, and (vi) survivability of Galleria mellonella larvae after being challenged with purified lasB and lasB-rich bacterial secretions. Molecular docking analyses revealed that phendione and its Ag+ and Cu2+ complexes were able to interact with the amino acids forming the active site of lasB, particularly Cu-phendione which exhibited the most favorable interaction energy parameters. Additionally, the test compounds were effective inhibitors of lasB activity, blocking the in vitro cleavage of the peptide substrate, aminobenzyl-Ala-Gly-Leu-Ala-p-nitrobenzylamide, with Cu-phendione having the best inhibitory action (K i = 90 nM). Treating living bacteria with a sub-inhibitory concentration (½ × MIC value) of the test compounds caused a significant reduction in the expression of the lasB gene as well as its mature protein production/secretion. Further, Ag-phendione and Cu-phendione offered protective action for lung epithelial cells, reducing the A549 monolayer damage by approximately 32 and 42%, respectively. Interestingly, Cu-phendione mitigated the toxic effect of both purified lasB molecules and lasB-containing bacterial secretions in the in vivo model, increasing the survival time of G. mellonella larvae. Collectively, these data reinforce the concept of lasB being a veritable therapeutic target and phendione-based compounds (mainly Cu-phendione) being prospective anti-virulence drugs against P. aeruginosa.

8.
J Biochem ; 144(5): 655-63, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18799520

ABSTRACT

Coenzyme A (CoASH) is an obligate cofactor for lipids undergoing beta-oxidation in peroxisomes. Although the peroxisomal membrane appears to be impermeable to CoASH, peroxisomes contain their own pool of CoASH. It is believed that CoASH enters peroxisomes as acyl-CoAs, but it is not known how this pool is regulated. The mouse nudix hydrolase 7 (NUDT7alpha) was previously identified in peroxisomes as a CoA-diphosphatase, and therefore suggested to be involved in regulation of peroxisomal CoASH levels. Here we show that mouse NUDT7alpha mainly acts as an acyl-CoA diphosphatase, with highest activity towards medium-chain acyl-CoAs, and much lower activity with CoASH. Nudt7alpha mRNA is highly expressed in liver, brown adipose tissue and heart, similar to enzymes involved in peroxisomal lipid degradation. Nudt7alpha mRNA is down-regulated by Wy-14,643, a peroxisome proliferator-activated receptor alpha (PPARalpha) ligand, in a PPARalpha-dependent manner in mouse liver. In highly purified peroxisomes, nudix hydrolase activity is highest with C(6)-CoA and is decreased by fibrate treatment. Under certain conditions, such as treatment with peroxisome proliferators or fasting, an increase in peroxisomal CoASH levels has been reported, which is in line with a decreased expression/activity of NUDT7alpha. Taken together these data suggest that NUDT7alpha function is tightly linked to peroxisomal CoASH/acyl-CoA homeostasis.


Subject(s)
Coenzyme A/metabolism , Homeostasis , Isoenzymes/metabolism , Peroxisomes/metabolism , Pyrophosphatases/metabolism , Acyl Coenzyme A/chemistry , Acyl Coenzyme A/metabolism , Adipose Tissue, Brown/enzymology , Alternative Splicing , Amino Acid Sequence , Animals , Coenzyme A/chemistry , Isoenzymes/genetics , Liver/enzymology , Mice , Molecular Sequence Data , PPAR gamma/metabolism , Pyrophosphatases/genetics , Sequence Alignment , Tissue Distribution , Nudix Hydrolases
9.
FASEB J ; 21(1): 99-107, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17116739

ABSTRACT

A wide variety of endogenous carboxylic acids and xenobiotics are conjugated with amino acids, before excretion in urine or bile. The conjugation of carboxylic acids and bile acids with taurine and glycine has been widely characterized, and de novo synthesized bile acids are conjugated to either glycine or taurine in peroxisomes. Peroxisomes are also involved in the oxidation of several other lipid molecules, such as very long chain acyl-CoAs, branched chain acyl-CoAs, and prostaglandins. In this study, we have now identified a novel peroxisomal enzyme called acyl-coenzyme A:amino acid N-acyltransferase (ACNAT1). Recombinantly expressed ACNAT1 acts as an acyltransferase that efficiently conjugates very long-chain and long-chain fatty acids to taurine. The enzyme shows no conjugating activity with glycine, showing that it is a specific taurine conjugator. Acnat1 is mainly expressed in liver and kidney, and the gene is localized in a gene cluster, together with two further acyltransferases, one of which conjugates bile acids to glycine and taurine. In conclusion, these data describe ACNAT1 as a new acyltransferase, involved in taurine conjugation of fatty acids in peroxisomes, identifying a novel pathway for production of N-acyltaurines as signaling molecules or for excretion of fatty acids.


Subject(s)
Acyltransferases/metabolism , Fatty Acids/metabolism , Peroxisomes/enzymology , Taurine/metabolism , Acyltransferases/chemistry , Acyltransferases/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Male , Mice , Molecular Sequence Data , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Spectrometry, Mass, Electrospray Ionization
10.
FASEB J ; 20(11): 1855-64, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16940157

ABSTRACT

The maintenance of cellular levels of free fatty acids and acyl-CoAs, the activated form of free fatty acids, is extremely important, as imbalances in lipid metabolism have serious consequences for human health. Acyl-coenzyme A (CoA) thioesterases (ACOTs) hydrolyze acyl-CoAs to the free fatty acid and CoASH, and thereby have the potential to regulate intracellular levels of these compounds. We previously identified and characterized a mouse ACOT gene cluster comprised of six genes that apparently arose by gene duplications encoding acyl-CoA thioesterases with localizations in cytosol (ACOT1), mitochondria (ACOT2), and peroxisomes (ACOT3-6). However, the corresponding human gene cluster contains only three genes (ACOT1, ACOT2, and ACOT4) coding for full-length thioesterase proteins, of which only one is peroxisomal (ACOT4). We therefore set out to characterize the human genes, and we show here that the human ACOT4 protein catalyzes the activities of three mouse peroxisomal ACOTs (ACOT3, 4, and 5), being active on succinyl-CoA and medium to long chain acyl-CoAs, while ACOT1 and ACOT2 carry out similar functions to the corresponding mouse genes. These data strongly suggest that the human ACOT4 gene has acquired the functions of three mouse genes by a functional convergent evolution that also provides an explanation for the unexpectedly low number of human genes.


Subject(s)
Evolution, Molecular , Multigene Family , Peroxisomes/enzymology , Thiolester Hydrolases/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA Primers , Humans , Mice , Molecular Sequence Data , Open Reading Frames , Palmitoyl-CoA Hydrolase/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Transcription, Genetic
11.
Prog Lipid Res ; 41(2): 99-130, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11755680

ABSTRACT

Acyl-CoA thioesterases are a group of enzymes that catalyze the hydrolysis of acyl-CoAs to the free fatty acid and coenzyme A (CoASH), providing the potential to regulate intracellular levels of acyl-CoAs, free fatty acids and CoASH. These enzymes are localized in almost all cellular compartments such as endoplasmic reticulum, cytosol, mitochondria and peroxisomes. Acyl-CoA thioesterases are highly regulated by peroxisome proliferator-activated receptors (PPARs), and other nutritional factors, which has led to the conclusion that they are involved in lipid metabolism. Although the physiological functions for these enzymes are not yet fully understood, recent cloning and more in-depth characterization of acyl-CoA thioesterases has assisted in discussion of putative functions for specific enzymes. Here we review the acyl-CoA thioesterases characterized to date and also address the diverse putative functions for these enzymes, such as in ligand supply for nuclear receptors, and regulation and termination of fatty acid oxidation in mitochondria and peroxisomes.


Subject(s)
Lipid Metabolism , Thiolester Hydrolases/physiology , Amino Acid Sequence , Animals , Brain/metabolism , Cloning, Molecular , Cytosol/enzymology , Humans , Liver/enzymology , Mitochondria/enzymology , Molecular Sequence Data , Rats , Terminology as Topic , Thiolester Hydrolases/isolation & purification
12.
Biochim Biophys Acta ; 1593(2-3): 283-9, 2003 Feb 17.
Article in English | MEDLINE | ID: mdl-12581873

ABSTRACT

Sterol 27-hydroxylase has been suggested to be involved in an alternative pathway for the elimination of cholesterol from macrophages and early atherosclerotic lesions. We have previously shown that human lung macrophages as well as monocyte-derived macrophages have a relatively high activity of sterol 27-hydroxylase (CYP27). This enzyme converts intracellular cholesterol into 27-hydroxycholesterol and cholestenoic acid that flux from cultured cells into the medium. It is shown here that human monocytes have very low CYP27 activity and CYP27 mRNA levels. During differentiation into macrophages, both CYP27 activity and CYP27 mRNA levels increase markedly after 4 days of culture in serum-free medium. Addition of macrophage-colony stimulating factor had no significant effect on the induction and addition of fetal calf serum had an inhibitory effect. Cholesterol synthesis was found to be a critical factor for the production of 27-oxygenated products by the macrophages cultured in serum-free medium. The increased capacity of the differentiated cells to eliminate intracellular cholesterol is of interest and supports the contention that CYP27 is an antiatherogenic factor.


Subject(s)
Cholesterol/analogs & derivatives , Macrophages/metabolism , Monocytes/cytology , RNA, Messenger , Steroid Hydroxylases/biosynthesis , Cell Differentiation , Cells, Cultured , Cholestanetriol 26-Monooxygenase , Cholesterol/analysis , Cholesterol/metabolism , Culture Media, Serum-Free , Humans , Hydroxycholesterols/analysis , Hydroxycholesterols/metabolism , Macrophage Colony-Stimulating Factor , Macrophages/enzymology , Monocytes/metabolism , RNA, Messenger/biosynthesis , Simvastatin/pharmacology , Time Factors
13.
Biochimie ; 98: 45-55, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24389458

ABSTRACT

Peroxisomes are nearly ubiquitous organelles involved in a number of metabolic pathways that vary between organisms and tissues. A common metabolic function in mammals is the partial degradation of various (di)carboxylic acids via α- and ß-oxidation. While only a small number of enzymes catalyze the reactions of ß-oxidation, numerous auxiliary enzymes have been identified to be involved in uptake of fatty acids and cofactors required for ß-oxidation, regulation of ß-oxidation and transport of metabolites across the membrane. These proteins include membrane transporters/channels, acyl-CoA thioesterases, acyl-CoA:amino acid N-acyltransferases, carnitine acyltransferases and nudix hydrolases. Here we review the current view of the role of these auxiliary enzymes in peroxisomal lipid metabolism and propose that they function in concert to provide a means to regulate fatty acid metabolism and transport of products across the peroxisomal membrane.


Subject(s)
Acyl Coenzyme A/metabolism , Coenzyme A/metabolism , Peroxisomes/metabolism , Acyltransferases/metabolism , Animals , Biological Transport/physiology , Coenzyme A-Transferases/metabolism , Fatty Acids/metabolism , Humans , Lipid Metabolism , Pyrophosphatases/metabolism , Thiolester Hydrolases/metabolism , Nudix Hydrolases
14.
Lipids ; 47(4): 355-61, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22160494

ABSTRACT

Endocannabinoids have been implicated in cancer development and cause heterogenous effects in tumor cells, by inducing apoptosis, reducing migration, causing anti-angiogenic activity and alterations in the cell cycle resulting in growth arrest. Recently, several novel amides of fatty acids that are structurally related to endocannabinoids have been isolated from mammalian sources, although the functions of these fatty amides are not well studied. One group of these novel fatty acid amides are the N-acyl taurines (fatty acids conjugated to the amino acid taurine). This study examined if N-acyl taurines, specifically N-arachidonoyl taurine and N-oleoyl taurine could function in a similar way to endocannabinoids and result in cell cycle alterations or growth arrest in the human prostate adenocarcinoma cell line PC-3. PC-3 cells were treated with various concentrations of N-arachidonoyl taurine and N-oleoyl taurine and cell proliferation and viability was measured using resazurin and colony formation assays. Effects of N-acyl taurines on the cell cycle was measured using FACS analysis. Treatment with N-arachidonoyl taurine and N-oleoyl taurine resulted in a significant reduction in proliferation of PC-3 cells, even at concentrations as low as 1 µM. Treatment with N-oleoyl taurine resulted in an increased number of cells in the subG1 population, suggesting apoptosis, and a lower number of cells in S-phase of the cell cycle. In summary, our results show that novel biologically active lipids, the N-acyl taurines, result in reduced proliferation in PC-3 cells.


Subject(s)
Adenocarcinoma/drug therapy , Arachidonic Acids/pharmacology , Prostate/drug effects , Prostatic Neoplasms/drug therapy , Taurine/analogs & derivatives , Adenocarcinoma/pathology , Apoptosis/drug effects , Arachidonic Acids/chemistry , Cannabinoid Receptor Modulators/chemistry , Cannabinoid Receptor Modulators/pharmacology , Cell Count , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Male , Oxazines , Prostate/pathology , Prostatic Neoplasms/pathology , Taurine/chemistry , Taurine/pharmacology , Xanthenes
15.
Prog Lipid Res ; 47(6): 405-21, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18538142

ABSTRACT

Peroxisomes are single membrane bound organelles present in almost all eukaryotic cells, and to date have been shown to contain approximately 60 identified enzymes involved in various metabolic pathways, including the oxidation of a variety of lipids. These lipids include very long-chain fatty acids, methyl branched fatty acids, prostaglandins, bile-acid precursors and xenobiotics that are either beta-oxidized or alpha-oxidized in peroxisomes. The recent identification of several acyl-CoA thioesterases and acyltransferases in peroxisomes has revealed their various functions in acting as auxiliary enzymes in alpha- and beta-oxidation in this organelle. To date, 9 functional acyl-CoA thioesterases and acyltransferases have been identified in mouse and 4 functional acyl-CoA thioesterases and acyltransferases in human, thus these enzymes make up a substantial portion of peroxisomal proteins. This review will therefore focus on new and emerging roles for these enzymes in assisting with the oxidation of various lipids, amidation of lipids for excretion from peroxisomes, and in controlling coenzyme A levels in peroxisomes.


Subject(s)
Acyltransferases/physiology , Lipid Metabolism/physiology , Peroxisomes/metabolism , Thiolester Hydrolases/physiology , Animals , Bile Acids and Salts/metabolism , Coenzyme A/metabolism , Dicarboxylic Acids/metabolism , Fatty Acid-Binding Proteins/physiology , Fatty Acids/metabolism , Humans , Mice , Oxidation-Reduction
16.
J Lipid Res ; 48(8): 1781-91, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17485727

ABSTRACT

The cytosolic acyl-coenzyme A thioesterase I (Acot1) is an enzyme that hydrolyzes long-chain acyl-CoAs of C(12)-C(20)-CoA in chain length to the free fatty acid and CoA. Acot1 was shown previously to be strongly upregulated at the mRNA and protein level in rodents by fibrates. In this study, we show that Acot1 mRNA levels were increased by 90-fold in liver by treatment with Wy-14,643 and that Acot1 mRNA was also increased by 15-fold in the liver of hepatocyte nuclear factor 4alpha (HNF4alpha) knockout animals. Our study identified a direct repeat 1 (DR1) located in the Acot1 gene promoter in mouse, which binds the peroxisome proliferator-activated receptor alpha (PPARalpha) and HNF4alpha. Chromatin immunoprecipitation (ChIP) assay showed that the identified DR1 bound PPARalpha/retinoid X receptor alpha (RXRalpha) and HNF4alpha, whereas the binding in ChIP was abrogated in the PPARalpha and HNF4alpha knockout mouse models. Reporter gene assays showed activation of the Acot1 promoter in cells by the PPARalpha agonist Wy-14,643 after cotransfection with PPARalpha/RXRalpha. However, transfection with a plasmid containing HNF4alpha also resulted in an increase in promoter activity. Together, these data show that Acot1 is under regulation by an interplay between HNF4alpha and PPARalpha.


Subject(s)
Hepatocyte Nuclear Factor 4/metabolism , PPAR alpha/metabolism , Palmitoyl-CoA Hydrolase/genetics , Promoter Regions, Genetic , Response Elements , Thiolester Hydrolases/genetics , Animals , Binding Sites , Clofibric Acid/pharmacology , Gene Expression Regulation, Enzymologic , Liver/drug effects , Liver/enzymology , Liver/metabolism , Male , Mice , Mice, Knockout , Palmitoyl-CoA Hydrolase/metabolism , Promoter Regions, Genetic/drug effects , Pyrimidines/pharmacology , Retinoid X Receptor alpha/metabolism , Thiolester Hydrolases/metabolism , Transcription Initiation Site
17.
J Biol Chem ; 282(37): 26707-26716, 2007 Sep 14.
Article in English | MEDLINE | ID: mdl-17613526

ABSTRACT

Phytanic acid and pristanic acid are derived from phytol, which enter the body via the diet. Phytanic acid contains a methyl group in position three and, therefore, cannot undergo beta-oxidation directly but instead must first undergo alpha-oxidation to pristanic acid, which then enters beta-oxidation. Both these pathways occur in peroxisomes, and in this study we have identified a novel peroxisomal acyl-CoA thioesterase named ACOT6, which we show is specifically involved in phytanic acid and pristanic acid metabolism. Sequence analysis of ACOT6 revealed a putative peroxisomal targeting signal at the C-terminal end, and cellular localization experiments verified it as a peroxisomal enzyme. Subcellular fractionation experiments showed that peroxisomes contain by far the highest phytanoyl-CoA/pristanoyl-CoA thioesterase activity in the cell, which could be almost completely immunoprecipitated using an ACOT6 antibody. Acot6 mRNA was mainly expressed in white adipose tissue and was co-expressed in tissues with Acox3 (the pristanoyl-CoA oxidase). Furthermore, Acot6 was identified as a target gene of the peroxisome proliferator-activated receptor alpha (PPARalpha) and is up-regulated in mouse liver in a PPARalpha-dependent manner.


Subject(s)
Coenzyme A/metabolism , Fatty Acids/metabolism , Peroxisomes/enzymology , Phytanic Acid/analogs & derivatives , Thiolester Hydrolases/physiology , Animals , Base Sequence , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Oxidation-Reduction , PPAR alpha/physiology , Phytanic Acid/metabolism
18.
Biochem Biophys Res Commun ; 360(2): 437-40, 2007 Aug 24.
Article in English | MEDLINE | ID: mdl-17601491

ABSTRACT

The metabolic regulator fibroblast growth factor 21 (FGF21) has antidiabetic properties in animal models of diabetes and obesity. Using quantitative RT-PCR, we here show that the hepatic gene expression of FGF21 is regulated by the peroxisome proliferator-activated receptor alpha (PPARalpha). Fasting or treatment of mice with the PPARalpha agonist Wy-14,643 induced FGF21 mRNA by 10-fold and 8-fold, respectively. In contrast, FGF21 mRNA was low in PPARalpha deficient mice, and fasting or treatment with Wy-14,643 did not induce FGF21. Obese ob/ob mice, known to have increased PPARalpha levels, displayed 12-fold increased hepatic FGF21 mRNA levels. The potential importance of PPARalpha for FGF21 expression also in human liver was shown by Wy-14,643 induction of FGF21 mRNA in human primary hepatocytes, and PPARalpha response elements were identified in both the human and mouse FGF21 promoters. Further studies on the mechanisms of regulation of FGF21 by PPARalpha in humans will be of great interest.


Subject(s)
Fibroblast Growth Factors/metabolism , Hepatocytes/metabolism , Liver Neoplasms/metabolism , Liver/metabolism , PPAR alpha/metabolism , Animals , Humans , Male , Mice , Mice, Knockout , Tumor Cells, Cultured
19.
J Biol Chem ; 280(46): 38125-32, 2005 Nov 18.
Article in English | MEDLINE | ID: mdl-16141203

ABSTRACT

Dicarboxylic acids are formed by omega-oxidation of fatty acids in the endoplasmic reticulum and degraded as the CoA ester via beta-oxidation in peroxisomes. Both synthesis and degradation of dicarboxylic acids occur mainly in kidney and liver, and the chain-shortened dicarboxylic acids are excreted in the urine as the free acids, implying that acyl-CoA thioesterases (ACOTs), which hydrolyze CoA esters to the free acid and CoASH, are needed for the release of the free acids. Recent studies show that peroxisomes contain several acyl-CoA thioesterases with different functions. We have now expressed a peroxisomal acyl-CoA thioesterase with a previously unknown function, ACOT4, which we show is active on dicarboxylyl-CoA esters. We also expressed ACOT8, another peroxisomal acyl-CoA thioesterase that was previously shown to hydrolyze a large variety of CoA esters. Acot4 and Acot8 are both strongly expressed in kidney and liver and are also target genes for the peroxisome proliferator-activated receptor alpha. Enzyme activity measurements with expressed ACOT4 and ACOT8 show that both enzymes hydrolyze CoA esters of dicarboxylic acids with high activity but with strikingly different specificities. Whereas ACOT4 mainly hydrolyzes succinyl-CoA, ACOT8 preferentially hydrolyzes longer dicarboxylyl-CoA esters (glutaryl-CoA, adipyl-CoA, suberyl-CoA, sebacyl-CoA, and dodecanedioyl-CoA). The identification of a highly specific succinyl-CoA thioesterase in peroxisomes strongly suggests that peroxisomal beta-oxidation of dicarboxylic acids leads to formation of succinate, at least under certain conditions, and that ACOT4 and ACOT8 are responsible for the termination of beta-oxidation of dicarboxylic acids of medium-chain length with the concomitant release of the corresponding free acids.


Subject(s)
Palmitoyl-CoA Hydrolase/physiology , Peroxisomes/metabolism , Succinic Acid/chemistry , Thiolester Hydrolases/physiology , Animals , Blotting, Western , Cloning, Molecular , Dicarboxylic Acids/chemistry , Dicarboxylic Acids/metabolism , Electrophoresis, Polyacrylamide Gel , Endoplasmic Reticulum/metabolism , Fatty Acids/metabolism , Fibroblasts/metabolism , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , Humans , Hydrolysis , Kidney/metabolism , Kinetics , Liver/metabolism , Male , Mice , Mice, Transgenic , Models, Biological , PPAR alpha/metabolism , Palmitoyl-CoA Hydrolase/chemistry , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Recombinant Proteins/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Substrate Specificity , Thiolester Hydrolases/chemistry , Thiolester Hydrolases/metabolism , Time Factors , Tissue Distribution , Up-Regulation
20.
J Lipid Res ; 46(9): 2029-32, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16103133

ABSTRACT

Acyl-CoA thioesterases, also known as acyl-CoA hydrolases, are a group of enzymes that hydrolyze CoA esters such as acyl-CoAs (saturated, unsaturated, branched-chain), bile acid-CoAs, CoA esters of prostaglandins, etc., to the corresponding free acid and CoA. However, there is significant confusion regarding the nomenclature of these genes. In agreement with the HUGO Gene Nomenclature Committee and the Mouse Genomic Nomenclature Committee, a revised nomenclature for mammalian acyl-CoA thioesterases/hydrolases has been suggested for the 12 member family. The family root symbol is ACOT, with human genes named ACOT1-ACOT12, and rat and mouse genes named Acot1-Acot12. Several of the ACOT genes are the result of splicing events, and these splice variants are cataloged.


Subject(s)
Palmitoyl-CoA Hydrolase , Terminology as Topic , Alternative Splicing , Animals , Humans , Mice , Multigene Family , Palmitoyl-CoA Hydrolase/genetics , Rats
SELECTION OF CITATIONS
SEARCH DETAIL