Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Int J Mol Sci ; 24(19)2023 Oct 06.
Article in English | MEDLINE | ID: mdl-37834405

ABSTRACT

Thioesters of coenzyme A (CoA) carrying different acyl chains (acyl-CoAs) are central intermediates of many metabolic pathways and donor molecules for protein lysine acylation. Acyl-CoA species largely differ in terms of cellular concentrations and physico-chemical properties, rendering their analysis challenging. Here, we compare several approaches to quantify cellular acyl-CoA concentrations in normal and ischemic rat liver, using HPLC and LC-MS/MS for multi-acyl-CoA analysis, as well as NMR, fluorimetric and spectrophotometric techniques for the quantification of acetyl-CoAs. In particular, we describe a simple LC-MS/MS protocol that is suitable for the relative quantification of short and medium-chain acyl-CoA species. We show that ischemia induces specific changes in the short-chain acyl-CoA relative concentrations, while mild ischemia (1-2 min), although reducing succinyl-CoA, has little effects on acetyl-CoA, and even increases some acyl-CoA species upstream of the tricarboxylic acid cycle. In contrast, advanced ischemia (5-6 min) also reduces acetyl-CoA levels. Our approach provides the keys to accessing the acyl-CoA metabolome for a more in-depth analysis of metabolism, protein acylation and epigenetics.


Subject(s)
Acyl Coenzyme A , Tandem Mass Spectrometry , Rats , Animals , Acetyl Coenzyme A/analysis , Chromatography, Liquid/methods , Acyl Coenzyme A/metabolism , Coenzyme A/analysis , Ischemia , Liver/metabolism
2.
Anal Bioanal Chem ; 413(30): 7333-7340, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34705077

ABSTRACT

Stable isotope labelling in combination with high-resolution mass spectrometry approaches are increasingly used to analyze both metabolite and protein modification dynamics. To enable correct estimation of the resulting dynamics, it is critical to correct the measured values for naturally occurring stable isotopes, a process commonly called isotopologue correction or deconvolution. While the importance of isotopologue correction is well recognized in metabolomics, it has received far less attention in proteomics approaches. Although several tools exist that enable isotopologue correction of mass spectrometry data, the majority is tailored for the analysis of low molecular weight metabolites. We here present PICor which has been developed for isotopologue correction of complex isotope labelling experiments in proteomics or metabolomics and demonstrate the importance of appropriate correction for accurate determination of protein modifications dynamics, using histone acetylation as an example.


Subject(s)
Isotope Labeling/methods , Proteins/chemistry , Acetyl Coenzyme A/analysis , Acetylation , Animals , Chromatography, Liquid/methods , HEK293 Cells , Humans , Mice , Molecular Weight , Protein Processing, Post-Translational , Proteomics , RAW 264.7 Cells , Tandem Mass Spectrometry/methods
3.
Metabolomics ; 15(12): 154, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31773381

ABSTRACT

INTRODUCTION: Relative oxidation of different metabolic substrates in the heart varies both physiologically and pathologically, in order to meet metabolic demands under different circumstances. 13C labelled substrates have become a key tool for studying substrate use-yet an accurate model is required to analyse the complex data produced as these substrates become incorporated into the Krebs cycle. OBJECTIVES: We aimed to generate a network model for the quantitative analysis of Krebs cycle intermediate isotopologue distributions measured by mass spectrometry, to determine the 13C labelled proportion of acetyl-CoA entering the Krebs cycle. METHODS: A model was generated, and validated ex vivo using isotopic distributions measured from isolated hearts perfused with buffer containing 11 mM glucose in total, with varying fractions of universally labelled with 13C. The model was then employed to determine the relative oxidation of glucose and triacylglycerol by hearts perfused with 11 mM glucose and 0.4 mM equivalent Intralipid (a triacylglycerol mixture). RESULTS: The contribution of glucose to Krebs cycle oxidation was measured to be 79.1 ± 0.9%, independent of the fraction of buffer glucose which was U-13C labelled, or of which Krebs cycle intermediate was assessed. In the presence of Intralipid, glucose and triglyceride were determined to contribute 58 ± 3.6% and 35.6 ± 0.8% of acetyl-CoA entering the Krebs cycle, respectively. CONCLUSION: These results demonstrate the accuracy of a functional model of Krebs cycle metabolism, which can allow quantitative determination of the effects of therapeutics and pathology on cardiac substrate metabolism.


Subject(s)
Mitochondria/metabolism , Myocardium/metabolism , Acetyl Coenzyme A/analysis , Animals , Carbon Isotopes , Citric Acid Cycle/physiology , Glucose/metabolism , Heart/physiology , Male , Mass Spectrometry/methods , Models, Biological , Oxidation-Reduction , Rats , Rats, Wistar
4.
Int J Mol Sci ; 20(22)2019 Nov 16.
Article in English | MEDLINE | ID: mdl-31744143

ABSTRACT

Using a closed-head impact acceleration model of mild or severe traumatic brain injury (mTBI or sTBI, respectively) in rats, we evaluated the effects of graded head impacts on the gene and protein expressions of pyruvate dehydrogenase (PDH), as well as major enzymes of mitochondrial tricarboxylic acid cycle (TCA). TBI was induced in anaesthetized rats by dropping 450 g from 1 (mTBI) or 2 m height (sTBI). After 6 h, 12 h, 24 h, 48 h, and 120 h gene expressions of enzymes and subunits of PDH. PDH kinases and phosphatases (PDK1-4 and PDP1-2, respectively), citrate synthase (CS), isocitrate dehydrogenase (IDH), oxoglutarate dehydrogenase (OGDH), succinate dehydrogenase (SDH), succinyl-CoA synthase (SUCLG), and malate dehydrogenase (MDH) were determined in whole brain extracts (n = 6 rats at each time for both TBI levels). In the same samples, the high performance liquid chromatographic (HPLC) determination of acetyl-coenzyme A (acetyl-CoA) and free coenzyme A (CoA-SH) was performed. Sham-operated animals (n = 6) were used as controls. After mTBI, the results indicated a general transient decrease, followed by significant increases, in PDH and TCA gene expressions. Conversely, permanent PDH and TCA downregulation occurred following sTBI. The inhibitory conditions of PDH (caused by PDP1-2 downregulations and PDK1-4 overexpression) and SDH appeared to operate only after sTBI. This produced almost no change in acetyl-CoA and free CoA-SH following mTBI and a remarkable depletion of both compounds after sTBI. These results again demonstrated temporary or steady mitochondrial malfunctioning, causing minimal or profound modifications to energy-related metabolites, following mTBI or sTBI, respectively. Additionally, PDH and SDH appeared to be highly sensitive to traumatic insults and are deeply involved in mitochondrial-related energy metabolism imbalance.


Subject(s)
Brain Injuries, Traumatic/pathology , Citric Acid Cycle/genetics , Pyruvate Dehydrogenase Complex/metabolism , Acetyl Coenzyme A/analysis , Animals , Brain Injuries, Traumatic/metabolism , Chromatography, High Pressure Liquid , Coenzyme A/analysis , Down-Regulation , Energy Metabolism , Male , Mitochondria/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Pyruvate Dehydrogenase Complex/genetics , Rats , Rats, Wistar , Severity of Illness Index , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism , Up-Regulation
5.
Methods Enzymol ; 626: 1-21, 2019.
Article in English | MEDLINE | ID: mdl-31606071

ABSTRACT

Dynamic interplay between cellular metabolism and histone acetylation is a key mechanism underlying metabolic control of epigenetics. In particular, the central metabolite acetyl-coenzyme A (acetyl-CoA) acts as the acetyl-donor for histone acetylation in both an enzymatic and non-enzymatic manner. Since members of the family of histone acetyl transferases (HATs) that catalyze the acetylation of histone tails possess a Michaelis constant (Km) within the range of physiological cellular acetyl-CoA concentrations, changing concentrations of acetyl-CoA can restrict or promote enzymatic histone acetylation. Likewise, non-enzymatic histone acetylation occurs at physiological concentrations. These concepts implicate acetyl-CoA as a rheostat for nutrient availability acting, in part, by controlling histone acetylation. Histone acetylation is an important epigenetic modification that controls gene expression and acetyl-CoA dependent changes in both histone acetylation and gene expression have been shown in yeast and mammalian systems. However, quantifying the metabolic conditions required to achieve specific changes in histone acetylation is a major challenge. The relationship between acetyl-CoA and histone acetylation may be influenced by a variety of factors including sub-cellular location of metabolites and enzymes, relative quantities of metabolites, and substrate availability/preference. A diversity of substrates can contribute the two-carbon acyl-chain to acetyl-CoA, a number of pathways can create or degrade acetyl-CoA, and only a handful of potential mechanisms for the crosstalk between metabolism and histone acetylation have been explored. The centrality of acetyl-CoA in intermediary metabolism means that acetyl-CoA levels may change, or be resistant to change, in unexpected ways. Thus, quantification of relevant metabolites is critical evidence in understanding how the nutrient rheostat is set in normal and pathological contexts. Coupling metabolite quantitation with isotope tracing to examine fate of specific metabolites is critical to the crosstalk between metabolism and histone acetylation, including but not limited to acetyl-CoA provides necessary context. This chapter provides guidance on experimental design of quantification with isotope dilution and/or tracing of acetyl-CoA within a targeted or highly multiplexed multi-analyte workflow.


Subject(s)
Acetyl Coenzyme A/metabolism , Histones/metabolism , Acetyl Coenzyme A/analysis , Acetylation , Animals , Chromatography, High Pressure Liquid/methods , Histones/chemistry , Humans , Mass Spectrometry/methods , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism
6.
Anal Chem ; 91(3): 2464-2471, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30608643

ABSTRACT

Coenzyme A (CoA) and acetyl-coenzyme A (acetyl-CoA) are ubiquitous cellular molecules, which mediate hundreds of anabolic and catabolic reactions including energy metabolism. Highly sensitive methods including absorption spectroscopy and mass spectrometry enable their analysis, albeit with many limitations. To date, however, NMR spectroscopy has not been used to analyze these important molecules. Building on our recent efforts, which enabled simultaneous analysis of a large number of metabolites in tissue and blood including many coenzymes and antioxidants ( Anal. Chem. 2016, 88, 4817-24; ibid 2017, 89, 4620-4627), we describe here a new method for identification and quantitation of CoA and acetyl-CoA ex vivo in tissue. Using mouse heart, kidney, liver, brain, and skeletal tissue, we show that a simple 1H NMR experiment can simultaneously measure these molecules. Identification of the two species involved a comprehensive analysis of the different tissue types using 1D and 2D NMR, in combination with spectral databases for standards, as well as spiking with authentic compounds. Time dependent studies showed that while the acetyl-CoA levels remain unaltered, CoA levels diminish by more than 50% within 24 h, which indicates that CoA is labile in solution; however, degassing the sample with helium gas halted its oxidation. Further, interestingly, we also identified endogenous coenzyme A glutathione disulfide (CoA-S-S-G) in tissue for the first time by NMR and show that CoA, when oxidized in tissue extract, also forms the same disulfide metabolite. The ability to simultaneously visualize absolute concentrations of CoA, acetyl-CoA, and endogenous CoA-S-S-G along with redox coenzymes (NAD+, NADH, NADP+, NADPH), energy coenzymes (ATP, ADP, AMP), antioxidants (GSH, GSSG), and a vast pool of other metabolites using a single 1D NMR spectrum offers a new avenue in the metabolomics field for investigation of cellular function in health and disease.


Subject(s)
Acetyl Coenzyme A/analysis , Adenine Nucleotides/analysis , Animals , Coenzyme A/analysis , Coenzymes/analysis , Glutathione/analysis , Male , Metabolomics/methods , Mice , Proton Magnetic Resonance Spectroscopy
7.
J Biosci Bioeng ; 120(5): 510-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26055446

ABSTRACT

Genetic engineering and metabolite profiling for the overproduction of polyhydroxybutyrate (PHB), which is a carbon material in biodegradable plastics, were examined in the unicellular cyanobacterium Synechocystis sp. PCC 6803. Transconjugants harboring cyanobacterial expression vectors that carried the pha genes for PHB biosynthesis were constructed. The overproduction of PHB by the engineering cells was confirmed through microscopic observations using Nile red, transmission electron microscopy (TEM), or nuclear magnetic resonance (NMR). We successfully recovered PHB from transconjugants prepared from nitrogen-depleted medium without sugar supplementation in which PHB reached approximately 7% (w/w) of the dry cell weight, showing a value of 12-fold higher productivity in the transconjugant than that in the control strain. We also measured the intracellular levels of acetyl-CoA, acetoacetyl-CoA, and 3-hydroxybutyryl-CoA (3HB-CoA), which are intermediate products for PHB. The results obtained indicated that these products were absent or at markedly low levels when cells were subjected to the steady-state growth phase of cultivation under nitrogen depletion for the overproduction of bioplastics. Based on these results, efficient factors were discussed for the overproduction of PHB in recombinant cyanobacteria.


Subject(s)
Genetic Engineering , Hydroxybutyrates/metabolism , Metabolome , Synechocystis/genetics , Synechocystis/metabolism , Acetyl Coenzyme A/analysis , Acetyl Coenzyme A/metabolism , Acyl Coenzyme A/analysis , Acyl Coenzyme A/metabolism , Conjugation, Genetic , Magnetic Resonance Spectroscopy , Microscopy, Electron, Transmission , Nitrogen/deficiency , Oxazines , Synechocystis/ultrastructure
8.
Biochem Soc Trans ; 42(4): 1107-11, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25110010

ABSTRACT

CoA (coenzyme A) is a ubiquitous and essential cofactor that acts as an acyl group carrier in biochemical reactions. Apart from participating in numerous metabolic pathways as substrates and intermediates, CoA and a number of its thioester derivatives, such as acetyl-CoA, can also directly regulate the activity of proteins by allosteric mechanisms and by affecting protein acetylation reactions. Cellular levels of CoA and CoA thioesters change under various physiological and pathological conditions. Defective CoA biosynthesis is implicated in NBIA (neurodegeneration with brain iron accumulation). However, the exact role of CoA in the pathogenesis of NBIA is not well understood. Accurate and reliable assays for measuring CoA species in biological samples are essential for studying the roles of CoA and CoA derivatives in health and disease. The present mini-review discusses methods that are commonly used to measure CoA species in biological samples.


Subject(s)
Coenzyme A/analysis , Enzyme Assays/methods , Acetyl Coenzyme A/analysis , Animals , Chromatography, High Pressure Liquid , Humans
9.
Br J Clin Pharmacol ; 78(6): 1366-77, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25041869

ABSTRACT

AIM: Further to its pivotal role in haemostasis, factor Xa (FXa) promotes effects on the vascular wall. The purpose of the study was to evaluate if FXa modifies the expression level of energy metabolism and oxidative stress-related proteins in femoral arteries obtained from type 2 diabetic patients with end-stage vasculopathy. METHODS: Femoral arteries were obtained from 12 type 2 diabetic patients who underwent leg amputation. Segments from the femoral arteries were incubated in vitro alone and in the presence of 25 nmol l(-1) FXa and 25 nmol l(-1) FXa + 50 nmol l(-1) rivaroxaban. RESULTS: In the femoral arteries, FXa increased triosephosphate isomerase and glyceraldehyde-3-phosphate dehydrogenase isotype 1 expression but decreased pyruvate dehydrogenase expression. These facts were accompanied by an increased content of acetyl-CoA. Aconitase activity was reduced in FXa-incubated femoral arteries as compared with control. Moreover, FXa increased the protein expression level of oxidative stress-related proteins which was accompanied by an increased malonyldialdehyde arterial content. The FXa inhibitor, rivaroxaban, failed to prevent the reduced expression of pyruvate dehydrogenase induced by FXa but reduced acetyl-CoA content and reverted the decreased aconitase activity observed with FXa alone. Rivaroxaban + FXa but not FXa alone increased the expression level of carnitine palmitoyltransferase I and II, two mitochondrial long chain fatty acid transporters. Rivaroxaban also prevented the increased expression of oxidative stress-related proteins induced by FXa alone. CONCLUSIONS: In femoral isolated arteries from type 2 diabetic patients with end-stage vasculopathy, FXa promoted disruption of the aerobic mitochondrial metabolism. Rivaroxaban prevented such effects and even seemed to favour long chain fatty acid transport into mitochondria.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Factor Xa/pharmacology , Femoral Artery/metabolism , Acetyl Coenzyme A/analysis , Aged , Carnitine O-Palmitoyltransferase/genetics , Diabetic Angiopathies/metabolism , Energy Metabolism , Female , Glycolysis , Humans , Male , Mitochondria/metabolism , Morpholines/pharmacology , Oxidative Stress , Rivaroxaban , Thiophenes/pharmacology
10.
J Sep Sci ; 36(23): 3807-12, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24123860

ABSTRACT

A new CZE method was developed for the determination of 12 purine and pyrimidine nucleotides, two adenine coenzymes and their reduced forms, and acetyl coenzyme A in various cell extracts. As the concentration levels of these metabolites in living cells are low; CZE was combined with field-enhanced sample stacking. As a result, the separation conditions were optimised to achieve a suitable resolution at the relatively high sample volume provided by this on-line pre-concentration technique. The optimum BGE was 150 mM glycine buffer (pH 9.5). Samples were introduced hydrodynamically using a pressure of 35 mbar (3.5 kPa) for 25 s, and data were collected at a detection wavelength of 260 nm. An applied voltage of 30 kV (positive polarity) and capillary temperature of 25°C gave the best separation of these compounds. The optimised method was validated by determining the linearity, sensitivity and repeatability and it was successfully applied for the analysis of extracts from Paracoccus denitrificans bacteria and from stem cells.


Subject(s)
Chemistry Techniques, Analytical/methods , Acetyl Coenzyme A/analysis , Adenosine Triphosphate/analysis , Chemistry Techniques, Analytical/standards , Cytidine Triphosphate/analysis , Embryonic Stem Cells/chemistry , Guanosine Triphosphate/analysis , Humans , Limit of Detection , Paracoccus denitrificans/chemistry , Reproducibility of Results
11.
J Nutr Sci Vitaminol (Tokyo) ; 59(2): 93-9, 2013.
Article in English | MEDLINE | ID: mdl-23727638

ABSTRACT

D-Pantethine is a compound in which two molecules of D-pantetheine bind through an S-S linkage. D-Pantethine is available from commercial sources as well as from D-pantothenic acid. We investigated if D-pantethine has the same vitamin activity as D-pantothenic acid by comparing the recovery from a deficiency of D-pantothenic acid in rats. D-Pantothenic acid-deficient rats were developed by weaning rats on a diet lacking D-pantothenic acid for 47 d. At that time, the urinary excretion of D-pantothenic acid was almost zero, and the body weight extremely low, compared with the control (p<0.05); the contents of free D-pantothenic acid were also significantly reduced in comparison with those of controls (p<0.05). D-Pantothenic acid-deficient rats were administered a diet containing D-pantothenic acid or D-pantethine for 7 d. D-Pantethine and D-pantothenic acid contents of the diets were equimolar in forms of D-pantothenic acid. We compared various parameters concerning nutritional status between rats fed D-pantothenic acid- and D-pantethine-containing diets. The recoveries of body weight, tissue weights, and tissue concentrations of free D-pantothenic acid, dephospho-CoA, CoA, and acetyl-CoA were identical between rats fed diets containing D-pantothenic acid and D-pantethine. Thus, the biological efficiency for recovering from a deficiency of D-pantothenic acid in rats was equivalent between D-pantothenic acid and D-pantethine.


Subject(s)
Pantetheine/analogs & derivatives , Pantothenic Acid/deficiency , Vitamins/pharmacology , Acetyl Coenzyme A/analysis , Animals , Body Weight/drug effects , Coenzyme A/analysis , Diet , Male , Organ Size/drug effects , Pantetheine/blood , Pantetheine/pharmacology , Pantothenic Acid/blood , Pantothenic Acid/pharmacology , Rats , Rats, Wistar , Vitamins/blood , Weaning
12.
J Dairy Sci ; 96(7): 4615-23, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23684018

ABSTRACT

We previously showed that propionic acid was more hypophagic than acetic acid when infused intraruminally in cows in the postpartum period and that the degree of hypophagia from short-term propionic acid infusion (18 h) was related to the acetyl coenzyme A (CoA) concentration in the liver. The objective of this experiment was to evaluate adaptation over time with longer-term infusions over 3 d. Twelve multiparous cows (2-13 d postpartum) were blocked by calving date and assigned randomly to treatment sequence in a crossover design experiment. The experiment was 12 d long with covariate periods preceding each 3-d infusion period. Treatments were 1.0 M propionic acid or 1.0 M acetic acid, infused intraruminally at 0.5 mol of volatile fatty acids/h beginning 6 h before feeding and continuing for 78 h with 3 d between infusions. Propionic acid decreased dry matter intake (DMI) relative to acetic acid (15.9 vs. 17.0 kg/d). However, a period-by- treatment interaction was detected for DMI. During period 1, propionic acid decreased DMI relative to acetic acid (14.3 vs. 17.5 kg/d) because of a reduction in meal size (1.30 vs. 1.65 kg), with no effect on intermeal interval. Propionic acid decreased DMI over the first 4 h following feeding (5.86 vs. 8.23 kg) but did not affect DMI 4 to 24 h after feeding. The depression in DMI in period 1 was positively related to hepatic acetyl-CoA concentration during the covariate period. Propionic acid was increasingly more hypophagic than acetic acid as hepatic acetyl-CoA concentration was elevated. No treatment-by-day interaction for DMI was observed, suggesting little or no measurable adaptation to treatment over the 3-d infusion period. These results suggest that hypophagia from propionic acid is enhanced when hepatic acetyl-CoA concentrations are elevated, such as when cows are in a lipolytic state.


Subject(s)
Cattle/physiology , Eating/drug effects , Lactation/physiology , Postpartum Period/physiology , Propionates/administration & dosage , Rumen/drug effects , Acetyl Coenzyme A/analysis , Animals , Fatty Acids, Volatile/analysis , Feeding Behavior/drug effects , Female , Fermentation , Hydrogen-Ion Concentration , Lipolysis , Liver/chemistry , Propionates/metabolism , Rumen/chemistry , Rumen/metabolism , Starch/metabolism
13.
Se Pu ; 31(1): 33-7, 2013 Jan.
Article in Chinese | MEDLINE | ID: mdl-23667987

ABSTRACT

A high performance liquid chromatography-tandem mass spectrometry (HPLC-MS/ MS) method was developed for the determination of acetyl coenzyme A in grape berries using n-propionyl coenzyme A as an internal standard (IS). The sample was extracted with water and then centrifuged for 5 min at 10 000 r/min on a centrifuge at 4 degrees C, and cleaned-up with a C18 solid phase extraction cartridge. The identification and quantification were carried out by using electrospray ionization in positive ion mode (ESI +) with multiple reaction monitoring (MRM). The total run time was 1 min and the elution of both acetyl coenzyme A and n-propionyl coenzyme A occurred at about 0.45 min. This was achieved with a mobile phase consisting of 5 mmol/L ammonium formate-acetonitrile (20: 80, v/v) at a flow rate of 0.4 mL/min on a C18 column. A linear response function was established for the concentration range of 1 - 2 000 microg/L for acetyl coenzyme A and the correlation coefficient was more than 0.99. The limit of detection of acetyl coenzyme A was 0.1 microg/L. The spiked recoveries at 50, 500, 1 000 microg/L ranged from 82.87% - 89.67% with the relative standard deviations less than 10%. This method is simple, rapid, sensitive and can significantly reduce the loss of acetyl coenzyme A during the experiment. It is suitable for the determination of acetyl coenzyme A in grape berries.


Subject(s)
Acetyl Coenzyme A/analysis , Chromatography, High Pressure Liquid/methods , Tandem Mass Spectrometry/methods , Vitis/enzymology , Food Analysis/methods , Fruit/enzymology
14.
Chem Commun (Camb) ; 49(28): 2876-8, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23443073

ABSTRACT

The application of a new rhodamine-based fluorescent probe, RH-NH2 3 and an acyl transfer promoter, PBu3, to Hela cells induced a time-dependent increase in fluorescence in the mitochondria, which was most likely due to acetylation of RH-NH2 3 with activated acetyl-CoA by the artificial reaction promoter in living cells.


Subject(s)
Acetyl Coenzyme A/analysis , Fluorescent Dyes/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Acetyl Coenzyme A/metabolism , Fluorescent Dyes/analysis , HeLa Cells , Humans , Molecular Imaging , Rhodamines/chemistry
15.
Anal Biochem ; 430(2): 151-5, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22922385

ABSTRACT

We describe here a simultaneous high-performance liquid chromatography method for practical and rapid determination of coenzyme A (CoA), dephospho-CoA, and acetyl-CoA in tissues. These coenzymes are biosynthesized from the vitamin pantothenic acid (PaA), which is involved in the metabolism of fatty acids, amino acid catabolism, and several other nutrients. The method employed a Tosoh TSK-GEL ODS-100 V column (250×4.6mm i.d., particle size 5µm) eluted with 100mmol/L NaH(2)PO(4) and 75mmol/L CH(3)COONa (pH was adjusted to 4.6 by the addition of concentrated H(3)PO(4))-acetonitrile (94:6, v/v) at a flow rate of 1.0ml/min. The ultraviolet detector was set at 259nm. The limits of detection for CoA, dephospho-CoA, and acetyl-CoA all were 10pmol. The method was applied to the analysis of several tissues of rats fed normal and PaA-free diets. The results clearly showed that the method was suitable for the simultaneous determination of CoA, dephospho-CoA, and acetyl-CoA in the liver, heart, kidney, spleen, testis, large colon, and muscle, but not for the small intestine, of rats.


Subject(s)
Acetyl Coenzyme A/analysis , Acyl Coenzyme A/analysis , Chromatography, High Pressure Liquid , Coenzyme A/analysis , Animals , Diet , Male , Pantothenic Acid/deficiency , Pantothenic Acid/metabolism , Rats , Rats, Wistar , Spectrophotometry, Ultraviolet
16.
J Dairy Sci ; 95(6): 3259-68, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22612960

ABSTRACT

Thirty multiparous lactating dairy cows were used in a randomized block design experiment to evaluate factors related to the degree of hypophagia from intraruminal infusion of propionate. Cows between 3 and 40 d postpartum at the start of the experiment were blocked by calving date and randomly assigned to treatment. Treatments were 1.0 mol/L propionic acid or 1.0 mol/L acetic acid adjusted to pH 6 with sodium hydroxide and infused at 0.5 mol of volatile fatty acid/h from 6h before feeding until 12h after feeding. Propionate infusion decreased dry matter intake by 20.0%, total metabolizable energy intake by 22.5%, and plasma ß-hydroxybutyrate concentration by 54.3% compared with acetate infusion. Effects of treatment on dry matter intake were related to concentration of acetyl coenzyme A (CoA) in the liver; hypophagic effects of propionate compared with acetate increased as liver acetyl CoA concentration increased. Hypophagic effects of propionate are greater for cows with elevated concentrations of acetyl CoA in the liver.


Subject(s)
Acetyl Coenzyme A/analysis , Eating/drug effects , Liver/chemistry , Postpartum Period/drug effects , Propionates/pharmacology , 3-Hydroxybutyric Acid/blood , Animals , Cattle , Diet/veterinary , Energy Intake/drug effects , Female , Postpartum Period/physiology , Pregnancy , Propionates/administration & dosage
17.
Magn Reson Med ; 66(6): 1526-30, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21608032

ABSTRACT

Deuterated water is widely used for measuring de novo lipogenesis on the basis of quantifying lipid (2)H-enrichment relative to that of body water. However, incorporation of (2)H-enrichment from body water into newly synthesized lipid molecules is incomplete therefore the true lipid precursor enrichment differs from that of body water. We describe a novel measurement of de novo lipogenesis that is based on a true precursor-product analysis of hepatic acetyl-CoA and triglyceride methyl enrichments from deuterated water. After deuterated water administration to seven in situ and seven perfused livers, acetyl-CoA methyl enrichment was inferred from (2)H nuclear magnetic resonance analysis of hepatic glutamate/glutamine (Glx) enrichment and triglyceride methyl enrichment was directly determined by (2)H nuclear magnetic resonance of triglycerides. Acetyl-CoA (2) H-enrichment was 71% ± 1% that of body water for in situ livers and 53% ± 2% of perfusate water for perfused livers. From the ratio of triglyceride-methyl/acetyl-CoA enrichments, fractional de novo lipogenesis rates of 0.97% ± 0.09%/2 hr and 7.92% ± 1.47%/48 hr were obtained for perfused and in situ liver triglycerides, respectively. Our method reveals that acetyl-CoA enrichment is significantly less than body water both for in situ and perfused livers. Furthermore, the difference between acetyl-CoA and body water enrichments is sensitive to the experimental setting.


Subject(s)
Acetyl Coenzyme A/analysis , Acetyl Coenzyme A/chemistry , Deuterium Exchange Measurement/methods , Glutamic Acid/metabolism , Glutamine/metabolism , Lipogenesis/physiology , Liver/metabolism , Animals , Citric Acid Cycle/physiology , Hydrogen/metabolism , Magnetic Resonance Spectroscopy/methods , Male , Rats , Rats, Wistar
18.
Biomed Chromatogr ; 25(12): 1352-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21381064

ABSTRACT

A highly sensitive and specific LC-MS/MS method was developed for simultaneous estimation of acetyl co-enzyme A (ACoA) and malonyl co-enzyme A (MCoA) in surrogate matrix using n-propionyl co-enzyme A as an internal standard (IS). LC-MS/MS was operated under the multiple reaction-monitoring mode using the electrospray ionization technique. Simple acidification followed by dilution using an assay buffer process was used to extract ACoA, MCoA and IS from surrogate matrix and tissue samples. The total run time was 3 min and the elution of both analytes (ACoA, MCoA) and IS occurred at 1.28 min; this was achieved with a mobile phase consisting of 5 mM ammonium formate (pH 7.5)-acetonitrile (30:70, v/v) delivered at a flow rate of 1 mL/min on a monolithic RP-18e column. A linear response function was established for the range of concentrations 1.09-2187 and 1.09-2193 ng/mL for ACoA and MCoA, respectively. The intra- and inter-day precision values for ACoA and MCoA met the acceptance as per FDA guidelines. ACoA and MCoA were stable in a battery of stability studies viz. bench-top, auto-sampler and long-term. The developed assay was used to quantitate ACoA and MCoA levels in various tissues of rat.


Subject(s)
Acetyl Coenzyme A/analysis , Chromatography, Liquid/methods , Malonyl Coenzyme A/analysis , Tandem Mass Spectrometry/methods , Acetonitriles , Animals , Calibration , Chromatography, Liquid/standards , Drug Stability , Formates , High-Throughput Screening Assays , Linear Models , Liver/chemistry , Male , Muscle, Skeletal/chemistry , Myocardium/chemistry , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity , Tandem Mass Spectrometry/standards
19.
J Dent Res ; 89(12): 1383-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20924070

ABSTRACT

Dental caries is initiated by demineralization of the tooth surface through acid production by sugar metabolism of supragingival plaque microflora. To elucidate the sugar metabolic system, we used CE-MS to perform metabolomics of the central carbon metabolism, the EMP pathway, the pentose-phosphate pathway, and the TCA cycle in supra- gingival plaque and representative oral bacteria, Streptococcus and Actinomyces. Supragingival plaque contained all the targeted metabolites in the central carbon metabolism, except erythrose 4-phosphate in the pentose-phosphate pathway. After glucose rinse, glucose 6-phosphate, fructose 6-phosphate, fructose 1,6-bisphosphate, dihydroxyacetone phosphate, and pyruvate in the EMP pathway and 6-phosphogluconate, ribulose 5-phosphate, and sedoheptulose 7-phosphate in the pentose-phosphate pathway, and acetyl CoA were increased. Meanwhile, 3-phosphoglycerate and phosphoenolpyruvate in the EMP pathway and succinate, fumarate, and malate in the TCA cycle were decreased. These pathways and changes in metabolites observed in supragingival plaque were similar to the integration of metabolite profiles in Streptococcus and Actinomyces.


Subject(s)
Actinomyces/metabolism , Dental Plaque/microbiology , Metabolomics , Streptococcus/metabolism , Acetyl Coenzyme A/analysis , Actinomyces/classification , Adult , Bacteriological Techniques , Carbon/metabolism , Citric Acid Cycle/physiology , Dihydroxyacetone Phosphate/analysis , Female , Fructosediphosphates/analysis , Fructosephosphates/analysis , Fumarates/analysis , Gluconates/analysis , Glucose/metabolism , Glucose-6-Phosphate/analysis , Glyceric Acids/analysis , Glycolysis/physiology , Humans , Malates/analysis , Male , Pentose Phosphate Pathway/physiology , Phosphoenolpyruvate/analysis , Pyruvic Acid/analysis , Ribulosephosphates/analysis , Streptococcus/classification , Streptococcus mutans/metabolism , Succinic Acid/analysis , Sugar Phosphates/analysis
20.
Biochem Biophys Res Commun ; 402(1): 158-62, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20933504

ABSTRACT

Levels of three coenzyme A (CoA) molecular species, i.e., nonesterified CoA (CoASH), acetyl-CoA, and malonyl-CoA, in fasted and fed rat tissues were analyzed by the acyl-CoA cycling method which makes detection possible at the pmol level. Malonyl-CoA in brain tissues readily increased with feeding, and inversely, acetyl-CoA decreased. This phenomenon occurred in the cerebral cortex, hippocampus, cerebellum, and medulla oblongata, as well as in the hypothalamus which controls energy balance by monitoring malonyl-CoA. In the non-brain tissues, the sizes of the acetyl-CoA, malonyl-CoA, and CoASH pools depended on the tissues. The total CoA pools consisting of the above three CoA species in the liver, heart, and brown adipose tissue were larger and those of the perirenal, epididymal, and ovarian adipose tissues were much smaller, compared with those of other tissues including brain tissues. In addition, the response of each CoA pool to feeding was not uniform, suggesting that the tissue-specific metabolism individually functions in the non-brain tissues. Thus, a comprehensive analysis of thirteen types of rat tissue revealed that CoA pools have different sizes and showed a different response to fasting and feeding depending on the tissue.


Subject(s)
Acetyl Coenzyme A/metabolism , Coenzyme A/metabolism , Eating , Fasting/metabolism , Malonyl Coenzyme A/metabolism , Acetyl Coenzyme A/analysis , Animals , Brain/enzymology , Coenzyme A/analysis , Female , Male , Malonyl Coenzyme A/analysis , Rats , Rats, Wistar , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...