Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Curr Top Med Chem ; 18(27): 2367-2378, 2018.
Article in English | MEDLINE | ID: mdl-30582479

ABSTRACT

BACKGROUND: Inflammation and oxidative stress are very closely related to pathophysiological processes and linked to multiple chronic diseases. Traditionally, the coconut fruits were used in Guatemala for treatment of dermatitis and inflammation. Isolation of the anti-inflammatory agent from the hard shell of the coconut fruit was targeted in the current study. METHODS: Fractionation of ethanolic extract of the coconut hard shell was done by using column chromatography, solvent treatments and TLC that led to the isolation of a molecule. RESULTS AND DISCUSSION: Spectral characterization of the molecule by LC-MS/MS QTOF, FTIR, 1HNMR, 13C-NMR, HMQC and HMBC indicated that it is a novel keto fatty acid, which is named as nuciferoic acid. Hyaluronidase inhibitory potential of the nuciferoic acid was found to be moderate. It was further docked in all the ten cavities of hyaluronidase and was compared with the substrate hyaluronic acid. Cavity 1 and cavity 4 could be the probable sites of action on hyaluronidase for nuciferoic acid. ADME and toxicological characterization suggested that the key sites of metabolism on nuciferoic acid are C1, C2, C14 and C17. Toxicity prediction against 55 toxicological endpoints revealed that nuciferoic acid does not have any indication of existing toxicological features. CONCLUSION: A novel keto fatty acid, nuciferoic acid, from C. nucifera hard shell has been isolated and characterized. It was found to inhibit hyaluronidase activity, which indicated its potential application as an anti-inflammatory drug or as an adjuvant.


Subject(s)
Cocos/chemistry , Fatty Acids/pharmacology , Glycoside Hydrolase Inhibitors/pharmacology , Hyaluronoglucosaminidase/antagonists & inhibitors , Keto Acids/pharmacology , Dose-Response Relationship, Drug , Fatty Acids/chemistry , Fatty Acids/isolation & purification , Glycoside Hydrolase Inhibitors/chemistry , Glycoside Hydrolase Inhibitors/isolation & purification , Humans , Hyaluronoglucosaminidase/metabolism , Keto Acids/chemistry , Keto Acids/isolation & purification , Molecular Structure , Structure-Activity Relationship
2.
Br J Nutr ; 119(1): 22-29, 2018 01.
Article in English | MEDLINE | ID: mdl-29208058

ABSTRACT

Diabetic nephropathy (DN) is a major cause of chronic kidney disease. We aimed to investigate the effect of the low-protein diets (LPD) supplemented with ketoacids (LPD+KA) in KKAy mice, an early type 2 DN model. KKAy mice were treated with normal protein diet (NPD), LPD or LPD+KA from 12 to 24 weeks of age. A period of 12-week treatment with LPD significantly reduced albuminuria as compared with that observed after NPD treatment. Treatment with LPD+KA further reduced albuminuria as compared with that observed with LPD treatment alone. Moreover, LPD treatment reduced mesangial expansion, thickness of glomerular basement membrane and the severity of the podocyte foot process effacement in KKAy mice; these effects were more pronounced in KKAy mice treated with LPD+KA. Both LPD and LPD+KA treatments slightly reduced total body weight, but had no significant effect on kidney weight and blood glucose concentrations when compared with NPD-treated KKAy mice. LPD treatment slightly attenuated oxidative stress in kidneys as compared with that observed in NPD-treated KKAy mice; however, LPD+KA treatment remarkably ameliorated oxidative stress in diabetic kidneys as shown by decreased malondialdehyde concentrations, protein carbonylation, nitrotyrosine expression and increased superoxide dismutase expression. Nutritional therapy using LPD+KA confers additional renal benefits as compared with those of LPD treatment alone in early type 2 DN through inhibition of oxidative stress.


Subject(s)
Diabetic Nephropathies/diet therapy , Diet, Protein-Restricted , Keto Acids/chemistry , Oxidative Stress , Renal Insufficiency, Chronic/diet therapy , Albuminuria/therapy , Animals , Diabetes Mellitus, Experimental/diet therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/diet therapy , Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/metabolism , Dietary Supplements , Disease Models, Animal , Glomerular Basement Membrane/metabolism , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Organ Size , Renal Insufficiency, Chronic/metabolism
3.
Chembiochem ; 16(1): 110-8, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25393087

ABSTRACT

Isobutanol is deemed to be a next-generation biofuel and a renewable platform chemical.1 Non-natural biosynthetic pathways for isobutanol production have been implemented in cell-based and in vitro systems with Bacillus subtilis acetolactate synthase (AlsS) as key biocatalyst.2-6 AlsS catalyzes the condensation of two pyruvate molecules to acetolactate with thiamine diphosphate and Mg(2+) as cofactors. AlsS also catalyzes the conversion of 2-ketoisovalerate into isobutyraldehyde, the immediate precursor of isobutanol. Our phylogenetic analysis suggests that the ALS enzyme family forms a distinct subgroup of ThDP-dependent enzymes. To unravel catalytically relevant structure-function relationships, we solved the AlsS crystal structure at 2.3 Å in the presence of ThDP, Mg(2+) and in a transition state with a 2-lactyl moiety bound to ThDP. We supplemented our structural data by point mutations in the active site to identify catalytically important residues.


Subject(s)
Acetolactate Synthase/chemistry , Bacillus subtilis/chemistry , Bacterial Proteins/chemistry , Butanols/chemistry , Acetolactate Synthase/genetics , Acetolactate Synthase/metabolism , Aldehydes/chemistry , Aldehydes/metabolism , Bacillus subtilis/classification , Bacillus subtilis/enzymology , Bacillus subtilis/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biocatalysis , Biofuels , Butanols/metabolism , Catalytic Domain , Cations, Divalent , Coenzymes/chemistry , Coenzymes/metabolism , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Hemiterpenes , Keto Acids/chemistry , Keto Acids/metabolism , Lactates/chemistry , Lactates/metabolism , Magnesium/chemistry , Magnesium/metabolism , Models, Molecular , Phylogeny , Point Mutation , Pyruvic Acid/chemistry , Pyruvic Acid/metabolism , Structure-Activity Relationship , Thiamine Pyrophosphate/chemistry , Thiamine Pyrophosphate/metabolism
4.
ChemSusChem ; 6(11): 2149-56, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23934656

ABSTRACT

Polymers based on renewable resources have become increasingly important. The natural functionalization of fats and oils enables an easy access to interesting monomeric building blocks, which in turn transform the derivative biopolymers into high-performance materials. Unfortunately, interesting building blocks of medium-chain length are difficult to obtain by traditional chemical means. Herein, a biotechnological pathway is established that could provide an environmentally suitable and sustainable alternative. A multiple enzyme two-step one-pot process efficiently catalyzed by a coupled 9S-lipoxygenase (St-LOX1, Solanum tuberosum) and 9/13-hydroperoxide lyase (Cm-9/13HPL, Cucumis melo) cascade reaction is proposed as a potential route for the conversion of linoleic acid into 9-oxononanoic acid, which is a precursor for biopolymers. Lipoxygenase catalyzes the insertion of oxygen into linoleic acid through a radical mechanism to give 9S-hydroperoxy-octadecadienoic acid (9S-HPODE) as a cascade intermediate, which is subsequently cleaved by the action of Cm-9/13HPL. This one-pot process afforded a yield of 73 % combined with high selectivity. The best reaction performance was achieved when lipoxygenase and hydroperoxide lyase were applied in a successive rather than a simultaneous manner. Green leaf volatiles, which are desired flavor and fragrance products, are formed as by-products in this reaction cascade. Furthermore, we have investigated the enantioselectivity of 9/13-HPLs, which exhibited a strong preference for 9S-HPODE over 9R-HPODE.


Subject(s)
Biopolymers/chemistry , Fatty Acids/chemical synthesis , Keto Acids/chemical synthesis , Biocatalysis , Chemistry Techniques, Synthetic , Cucumis melo/enzymology , Fatty Acids/chemistry , Hydro-Lyases/metabolism , Hydrogen-Ion Concentration , Keto Acids/chemistry , Linoleic Acid/chemistry , Lipoxygenase/metabolism , Solanum tuberosum/enzymology , Stereoisomerism , Substrate Specificity
5.
Anal Biochem ; 439(2): 116-22, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23684523

ABSTRACT

Branched-chain keto acids (BCKAs) are associated with increased susceptibility to several degenerative diseases. However, BCKA concentrations in tissues or the amounts of tissue available are frequently at the limit of detection for standard plasma methods. To accurately and quickly determine tissue BCKAs, we have developed a sensitive ultra fast liquid chromatography-mass spectrometry (UFLC-MS) method. BCKAs from deproteinized tissue extractions were o-phenylenediamine (OPD) derivatized, ethyl acetate extracted, lyophilized in a vacuum centrifuge, and reconstituted in 200 mM ammonium acetate. Samples were injected onto a Shimadzu UFLC system coupled to an AB-Sciex 5600 Triple TOF mass spectrometer instrument that detected masses of the OPD BCKA products using a multiple reaction monitoring method. An OPD-derivatized (13)C-labeled keto acid was used as an internal standard. Application of the method for C57BL/6J (wild-type) and PP2Cm knockout mouse tissues, including kidney, adipose tissue, liver, gastrocnemius, and hypothalamus, is shown. The lowest tissue concentration measured by this method was 20 nM, with the standard curve covering a wide range (7.8-32,000 nM). Liquid chromatography-mass spectrometry run times for this assay were less than 5 min, facilitating high throughput, and the OPD derivatives were found to be stable over several days.


Subject(s)
Chromatography, Liquid/methods , Keto Acids/chemistry , Mass Spectrometry/methods , Tissue Distribution/physiology , Adipose Tissue/chemistry , Animals , Hypothalamus/chemistry , Keto Acids/metabolism , Kidney/chemistry , Liver/chemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/chemistry , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Protein Phosphatase 2C
6.
Br J Nutr ; 104(10): 1438-42, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20594391

ABSTRACT

During exercise, ammonia levels are related to the appearance of both central and peripheral fatigue. Therefore, controlling the increase in ammonia levels is an important strategy in ameliorating the metabolic response to exercise and in improving athletic performance. Free amino acids can be used as substrates for ATP synthesis that produces ammonia as a side product. Keto analogues act in an opposite way, being used to synthesise amino acids whilst decreasing free ammonia in the blood. Adult male rats were divided into four groups based on receiving either keto analogues associated with amino acids (KAAA) or a placebo and resistance exercise or no exercise. There was an approximately 40% increase in ammonaemia due to KAAA supplementation in resting animals. Exercise increased ammonia levels twofold with respect to the control, with a smaller increase (about 20%) in ammonia levels due to exercise. Exercise itself causes a significant increase in blood urea levels (17%). However, KAAA reduced blood urea levels to 75% of the pre-exercise values. Blood urate levels increased 28% in the KAAA group, independent of exercise. Supplementation increased glucose levels by 10% compared with control animals. Exercise did not change glucose levels in either the control or supplemented groups. Exercise promoted a 57% increase in lactate levels in the control group. Supplementation promoted a twofold exercise-induced increase in blood lactate levels. The present results suggest that an acute supplementation of KAAA can decrease hyperammonaemia induced by exercise.


Subject(s)
Amino Acids/pharmacology , Keto Acids/chemistry , Keto Acids/pharmacology , Motor Activity/physiology , Physical Conditioning, Animal/physiology , Amino Acids/administration & dosage , Animal Feed , Animal Nutritional Physiological Phenomena , Animals , Diet , Dietary Supplements , Hyperammonemia , Keto Acids/administration & dosage , Lactic Acid/blood , Male , Rats , Rats, Wistar , Uric Acid/blood
7.
Cancer Prev Res (Phila) ; 2(7): 683-93, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19584079

ABSTRACT

Histone deacetylase (HDAC) inhibitors are gaining interest as cancer therapeutic agents. We tested the hypothesis that natural organoselenium compounds might be metabolized to HDAC inhibitors in human prostate cancer cells. Se-Methyl-L-selenocysteine (MSC) and selenomethionine are amino acid components of selenium-enriched yeast. In a cell-free system, glutamine transaminase K (GTK) and L-amino acid oxidase convert MSC to the corresponding alpha-keto acid, beta-methylselenopyruvate (MSP), and L-amino acid oxidase converts selenomethionine to its corresponding alpha-keto acid, alpha-keto-gamma-methylselenobutyrate (KMSB). Although methionine (sulfur analogue of selenomethionine) is an excellent substrate for GTK, selenomethionine is poorly metabolized. Structurally, MSP and KMSB resemble the known HDAC inhibitor butyrate. We examined androgen-responsive LNCaP cells and androgen-independent LNCaP C4-2, PC-3, and DU145 cells and found that these human prostate cancer cells exhibit endogenous GTK activities. In the corresponding cytosolic extracts, the metabolism of MSC was accompanied by the concomitant formation of MSP. In MSP-treated and KMSB-treated prostate cancer cell lines, acetylated histone 3 levels increased within 5 hours, and returned to essentially baseline levels by 24 hours, suggesting a rapid, transient induction of histone acetylation. In an in vitro HDAC activity assay, the selenoamino acids, MSC and selenomethionine, had no effect at concentrations up to 2.5 mmol/L, whereas MSP and KMSB both inhibited HDAC activity. We conclude that, in addition to targeting redox-sensitive signaling proteins and transcription factors, alpha-keto acid metabolites of MSC and selenomethionine can alter HDAC activity and histone acetylation status. These findings provide a potential new paradigm by which naturally occurring organoselenium might prevent the progression of human prostate cancer.


Subject(s)
Histone Deacetylase Inhibitors , Keto Acids/chemistry , Prostatic Neoplasms/drug therapy , Cell Line, Tumor , Chromatography, High Pressure Liquid , Histones/chemistry , Humans , Inhibitory Concentration 50 , L-Amino Acid Oxidase/chemistry , Lyases/chemistry , Male , Mass Spectrometry/methods , Methionine/chemistry , Prostatic Neoplasms/prevention & control , Protein Processing, Post-Translational , Selenium/chemistry , Transaminases/chemistry
8.
J Biomol NMR ; 43(2): 111-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19115043

ABSTRACT

A strategy for the introduction of ((1)H,(13)C-methyl)-alanine into perdeuterated proteins is described. Specific protonation of alanine methyl groups to a level of 95% can be achieved by overexpressing proteins in M9/D(2)O based bacterial growth medium supplemented with 800 mg/l of 2-[(2)H], 3-[(13)C] L: -alanine. However, though simple, this approach results in undesired, non-specific background labeling due to isotope scrambling via different amino acid metabolic pathways. Following a careful analysis of known metabolic pathways we found that co-addition of perdeuterated forms of alpha-ketoisovalerate-d(7), succinate-d(4) and L: -isoleucine-d(10) with labeled L: -alanine, reduces undesired background labeling to <1%. When combined with recently developed methyl TROSY experiments, this methyl-specific labeling protocol permits the acquisition of excellent quality correlation spectra of alanine methyl groups in high molecular weight proteins. Our cost effective strategy offers a significant enhancement in the level of incorporation of methyl-labeled alanine in overexpressed proteins over previously reported methods.


Subject(s)
Isotope Labeling/methods , Nuclear Magnetic Resonance, Biomolecular , Proteins/chemistry , Proteins/metabolism , Alanine/chemistry , Alanine/metabolism , Carbon Isotopes/chemistry , Carbon Isotopes/metabolism , Culture Media , Deuterium/chemistry , Deuterium/metabolism , Escherichia coli/genetics , Hemiterpenes , Humans , Isoleucine/chemistry , Isoleucine/metabolism , Keto Acids/chemistry , Keto Acids/metabolism , Malate Synthase/chemistry , Malate Synthase/metabolism , Metabolic Networks and Pathways , Proteins/genetics , Succinic Acid/chemistry , Succinic Acid/metabolism , Ubiquitin/chemistry , Ubiquitin/genetics , Ubiquitin/metabolism
9.
J Am Chem Soc ; 130(50): 17075-84, 2008 Dec 17.
Article in English | MEDLINE | ID: mdl-19012396

ABSTRACT

There has been considerable interest in protein tyrosine phosphatase 1B (PTP1B) as a therapeutic target for diabetes, obesity, as well as cancer. Identifying inhibitory compounds with good bioavailability is a major challenge of drug discovery programs targeted toward PTPs. Most current PTP active site-directed pharmacophores are negatively charged pTyr mimetics which cannot readily enter the cell. This lack of cell permeability limits the utility of such compounds in signaling studies and further therapeutic development. We identify aryl diketoacids as novel pTyr surrogates and show that neutral amide-linked aryl diketoacid dimers also exhibit excellent PTP inhibitory activity. Kinetic studies establish that these aryl diketoacid derivatives act as noncompetitive inhibitors of PTP1B. Crystal structures of ligand-bound PTP1B reveal that both the aryl diketoacid and its dimeric derivative bind PTP1B at the active site, albeit with distinct modes of interaction, in the catalytically inactive, WPD loop open conformation. Furthermore, dimeric aryl diketoacids are cell permeable and enhance insulin signaling in hepatoma cells, suggesting that targeting the inactive conformation may provide a unique opportunity for creating active site-directed PTP1B inhibitors with improved pharmacological properties.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Keto Acids/chemical synthesis , Keto Acids/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Amides/chemistry , Cell Line, Tumor , Crystallography, X-Ray , Dimerization , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Humans , Keto Acids/chemistry , Models, Molecular , Molecular Structure , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 1/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Structure-Activity Relationship
10.
J Biol Chem ; 280(9): 7588-96, 2005 Mar 04.
Article in English | MEDLINE | ID: mdl-15611050

ABSTRACT

A complex mixture of fatty acid-derived aldehydes, ketones, and alcohols is released upon wounding of the moss Physcomitrella patens. To investigate the formation of these oxylipins at the molecular level we isolated a lipoxygenase from P. patens, which was identified in an EST library by sequence homology to lipoxygenases from plants. Sequence analysis of the cDNA showed that it exhibits a domain structure similar to that of type2 lipoxygenases from plants, harboring an N-terminal import signal for chloroplasts. The recombinant protein was identified as arachidonate 12-lipoxygenase and linoleate 13-lipoxygenase with a preference for arachidonic acid and eicosapentaenoic acid. In contrast to any other lipoxygenase cloned so far, this enzyme exhibited in addition an unusual high hydroperoxidase and also a fatty acid chain-cleaving lyase activity. Because of these unique features the pronounced formation of (2Z)-octen-1-ol, 1-octen-3-ol, the dienal (5Z,8Z,10E)-12-oxo-dodecatrienoic acid and 12-keto eicosatetraenoic acid was observed when arachidonic acid was administered as substrate. 12-Hydroperoxy eicosatetraenoic acid was found to be only a minor product. Moreover, the P. patens LOX has a relaxed substrate tolerance accepting C(18)-C(22) fatty acids giving rise to even more LOX-derived products. In contrast to other lipoxygenases a highly diverse product spectrum is formed by a single enzyme accounting for most of the observed oxylipins produced by the moss. This single enzyme might, in a fast and effective way, be involved in the formation of signal and/or defense molecules thus contributing to the broad resistance of mosses against pathogens.


Subject(s)
Bryopsida/chemistry , Bryopsida/enzymology , Fatty Acids/metabolism , Hydrogen Peroxide/chemistry , Lipoxygenase/physiology , Amino Acid Sequence , Arachidonate 12-Lipoxygenase/chemistry , Catalytic Domain , Chloroplasts/metabolism , Chromatography, High Pressure Liquid , Cloning, Molecular , DNA, Complementary/metabolism , Eicosapentaenoic Acid/chemistry , Expressed Sequence Tags , Gas Chromatography-Mass Spectrometry , Gene Library , Genes, Plant , Keto Acids/chemistry , Lipoxygenase/chemistry , Lyases/chemistry , Models, Chemical , Molecular Sequence Data , Phylogeny , Plant Proteins/chemistry , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Time Factors , Ultraviolet Rays
11.
J Biol Chem ; 277(9): 6985-93, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11744710

ABSTRACT

Glycine oxidase (GO) is a homotetrameric flavoenzyme that contains one molecule of non-covalently bound flavin adenine dinucleotide per 47 kDa protein monomer. GO is active on various amines (sarcosine, N-ethylglycine, glycine) and d-amino acids (d-alanine, d-proline). The products of GO reaction with various substrates have been determined, and it has been clearly shown that GO catalyzes the oxidative deamination of primary and secondary amines, a reaction similar to that of d-amino acid oxidase, although its sequence homology is higher with enzymes such as sarcosine oxidase and N-methyltryptophane oxidase. GO shows properties that are characteristic of the oxidase class of flavoproteins: it stabilizes the anionic flavin semiquinone and forms a reversible covalent flavin-sulfite complex. The approximately 300 mV separation between the two FAD redox potentials is in accordance with the high amount of the anionic semiquinone formed on photoreduction. GO can be distinguished from d-amino acid oxidase by its low catalytic efficiency and high apparent K(m) value for d-alanine. A number of active site ligands have been identified; the tightest binding is observed with glycolate, which acts as a competitive inhibitor with respect to sarcosine. The presence of a carboxylic group and an amino group on the substrate molecule is not mandatory for binding and catalysis.


Subject(s)
Amino Acid Oxidoreductases/chemistry , Amino Acid Oxidoreductases/physiology , Bacillus subtilis/enzymology , Flavoproteins/chemistry , Aldehyde Oxidoreductases/metabolism , Binding Sites , Butyrates/chemistry , Catalysis , DNA, Complementary/metabolism , Escherichia coli/metabolism , Glutamate Dehydrogenase/metabolism , Hydrogen-Ion Concentration , Keto Acids/chemistry , Kinetics , Ligands , Models, Chemical , Oxidation-Reduction , Oxygen/metabolism , Protein Binding , Protein Structure, Tertiary , Substrate Specificity , Sulfites/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL