Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Commun Biol ; 2: 194, 2019.
Article in English | MEDLINE | ID: mdl-31123718

ABSTRACT

Hydrogen sulfide (H2S) plays important roles in metabolism and health. Its enzymatic generation from sulfur-containing amino acids (SAAs) is well characterized. However, the existence of non-enzymatic H2S production from SAAs, the chemical mechanism, and its biological implications remain unclear. Here we present non-enzymatic H2S production in vitro and in blood via a reaction specific for the SAA cysteine serving as substrate and requires coordinated catalysis by Vitamin B6, pyridoxal(phosphate), and iron under physiological conditions. An initial cysteine-aldimine is formed by nucleophilic attack of the cysteine amino group to the pyridoxal(phosphate) aldehyde group. Free or heme-bound iron drives the formation of a cysteine-quinonoid, thiol group elimination, and hydrolysis of the desulfurated aldimine back to pyridoxal(phosphate). The reaction ultimately produces pyruvate, NH3, and H2S. This work highlights enzymatic production is inducible and robust in select tissues, whereas iron-catalyzed production contributes underappreciated basal H2S systemically with pathophysiological implications in hemolytic, iron overload, and hemorrhagic disorders.


Subject(s)
Cysteine/metabolism , Hydrogen Sulfide/metabolism , Iron/metabolism , Pyridoxal Phosphate/metabolism , Vitamin B 6/metabolism , Animals , Catalysis , Erythrocytes/cytology , Erythrocytes/metabolism , Hemolysis , Hydrolysis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Spectrophotometry, Ultraviolet
2.
Am J Physiol Endocrinol Metab ; 313(6): E690-E698, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28830869

ABSTRACT

The period around bariatric surgery offers a unique opportunity to characterize metabolism responses to dynamic shifts in energy, gut function, and anesthesia. We analyzed plasma acylcarnitines in obese women (n = 17) sampled in the overnight fasted/postabsorptive state approximately 1-2 wk before surgery (condition A), the morning of surgery (prior restriction to a 48-h clear liquid diet coupled in some cases a standard polyethylene glycol gut evacuation: condition B), and following induction of anesthesia (condition C). Comparisons tested if 1) plasma acylcarnitine derivatives reflective of fatty acid oxidation (FAO) and xenometabolism would be significantly increased and decreased, respectively, by preoperative gut preparation/negative energy balance (condition A vs. B), and 2) anesthesia would acutely depress markers of FAO. Acylcarnitines associated with fat mobilization and FAO were significantly increased in condition B: long-chain acylcarnitines (i.e., C18:1, ~70%), metabolites from active but incomplete FAO [i.e., C14:1 (161%) and C14:2 (102%)] and medium- to short-chain acylcarnitines [i.e., C2 (91%), R-3-hydroxybutyryl-(245%), C6 (45%), and cis-3,4-methylene-heptanoyl-(17%), etc.]. Branched-chain amino acid markers displayed disparate patterns [i.e., isobutyryl-(40% decreased) vs. isovaleryl carnitine (51% increased)]. Anesthesia reduced virtually every acylcarnitine. These results are consistent with a fasting-type metabolic phenotype coincident with the presurgical "gut preparation" phase of bariatric surgery, and a major and rapid alteration of both fat and amino acid metabolism with onset of anesthesia. Whether presurgical or anesthesia-associated metabolic shifts in carnitine and fuel metabolism impact patient outcomes or surgical risks remains to be evaluated experimentally.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Anesthesia , Bariatric Surgery , Carnitine/analogs & derivatives , Cathartics/adverse effects , Lipid Metabolism/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Adult , Anesthesia/adverse effects , Anesthesia/methods , Bariatric Surgery/adverse effects , Bariatric Surgery/methods , Carnitine/blood , Cathartics/pharmacology , Fasting/metabolism , Fatty Acids/metabolism , Female , Humans , Middle Aged , Obesity, Morbid/metabolism , Obesity, Morbid/surgery , Oxidation-Reduction/drug effects , Preoperative Care/adverse effects , Preoperative Care/methods , Young Adult
3.
J Chromatogr B Analyt Technol Biomed Life Sci ; 1061-1062: 128-133, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28734160

ABSTRACT

Tandem MS acylcarnitine "profiles" are extremely valuable. Although used appropriately in newborn screening programs to identify patients with possible diseases, their inadequate quantitative accuracy and lack of selectivity is problematic for confirmatory testing. In this report, we show the application of our validated, selective, accurate, precise, and robust UHPLC-MS/MS method for quantitation of acylcarnitines, specifically to C5 acylcarnitines: pivaloyl-, 2-methylbutyryl-, isovaleryl-, and valerylcarnitine. Standardized calibrants were used to generate 13-point, 200-fold concentration range calibration curves. Samples were isolated by solid-phase extraction and derivatized with pentafluorophenacyl trifluoromethanesulfonate. Acylcarnitine pentafluorophenacyl esters were eluted in 14min chromatograms. Data demonstrating quantitative stability and method robustness over a five year time period are shown and these results validate the method's accuracy and robustness. Urine from patients with isovaleric acidemia (with the disease marker isovalerylcarnitine) and with pivaloylcarnitine present are shown. These results demonstrate the method's ability to distinguish true isovaleric acidemia from pivalate derived interference. Our method for acylcarnitine quantitation is shown to be accurate, precise, and robust for selective quantitation of isovalerylcarnitine, and thus is recommended for confirmatory testing of suspected isovaleric acidemia patients.


Subject(s)
Amino Acid Metabolism, Inborn Errors/urine , Carnitine/analogs & derivatives , Chromatography, High Pressure Liquid/methods , Isovaleryl-CoA Dehydrogenase/deficiency , Tandem Mass Spectrometry/methods , Carnitine/urine , Humans , Isovaleryl-CoA Dehydrogenase/urine , Limit of Detection , Reproducibility of Results
4.
Mol Genet Metab ; 120(4): 363-369, 2017 04.
Article in English | MEDLINE | ID: mdl-28190699

ABSTRACT

While selectively quantifying acylcarnitines in thousands of patient samples using UHPLC-MS/MS, we have occasionally observed unidentified branched-chain C8 acylcarnitines. Such observations are not possible using tandem MS methods, which generate pseudo-quantitative acylcarnitine "profiles". Since these "profiles" select for mass alone, they cannot distinguish authentic signal from isobaric and isomeric interferences. For example, some of the samples containing branched-chain C8 acylcarnitines were, in fact, expanded newborn screening false positive "profiles" for medium-chain acyl-CoA dehydrogenase deficiency (MCADD). Using our fast, highly selective, and quantitatively accurate UHPLC-MS/MS acylcarnitine determination method, we corrected the false positive tandem MS results and reported the sample results as normal for octanoylcarnitine (the marker for MCADD). From instances such as these, we decided to further investigate the presence of branched-chain C8 acylcarnitines in patient samples. To accomplish this, we synthesized and chromatographically characterized several branched-chain C8 acylcarnitines (in addition to valproylcarnitine): 2-methylheptanoylcarnitine, 6-methylheptanoylcarnitine, 2,2-dimethylhexanoylcarnitine, 3,3-dimethylhexanoylcarnitine, 3,5-dimethylhexanoylcarnitine, 2-ethylhexanoylcarnitine, and 2,4,4-trimethylpentanoylcarnitine. We then compared their behavior with branched-chain C8 acylcarnitines observed in patient samples and demonstrated our ability to chromographically resolve, and thus distinguish, octanoylcarnitine from branched-chain C8 acylcarnitines, correcting false positive MCADD results from expanded newborn screening.


Subject(s)
Acyl-CoA Dehydrogenase/deficiency , Carnitine/analogs & derivatives , Carnitine/metabolism , Lipid Metabolism, Inborn Errors/diagnosis , Neonatal Screening/standards , Carnitine/chemical synthesis , Carnitine/isolation & purification , Chromatography, High Pressure Liquid/methods , False Positive Reactions , Humans , Infant, Newborn , Sensitivity and Specificity , Tandem Mass Spectrometry
5.
J Appl Lab Med ; 2(3): 335-344, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-33636854

ABSTRACT

BACKGROUND: Although correctly used in expanded newborn screening programs to identify patients with possible diseases, flow-injection tandem mass spectrometry (MS/MS) acylcarnitine "profiles" are inadequate for standard clinical uses owing to their limited quantitative accuracy and lack of selectivity. We report the application of our selective, accurate, and precise method for quantification of acylcarnitines, applied to urine glutarylcarnitine from a patient with glutaric acidemia type I (GAI). METHODS: A previously validated acylcarnitine ultra-HPLC-MS/MS method was used, with a focus on analysis of glutarylcarnitine. Calibrants and samples were isolated by solid-phase extraction and derivatized with pentafluorophenacyl trifluoromethanesulfonate. Acylcarnitine pentafluorophenacyl esters were eluted in 14-min chromatograms. Standardized calibrants and a 13-point, 200-fold concentration range calibration curve were used for accurate quantification of glutarylcarnitine. Quality control samples validated method accuracy and long-term analytic stability. RESULTS: Quantification of glutarylcarnitine in urine from a patient with GAI is reported. Long-term analytical stability of the method over a 5-year period is shown. CONCLUSIONS: Our method for acylcarnitine quantification is shown to be selective, accurate, and precise; thus, we recommend it for confirmatory testing and monitoring of plasma and urine samples from patients with GAI.

6.
Mol Genet Metab ; 116(4): 231-41, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26458767

ABSTRACT

Tandem MS "profiling" of acylcarnitines and amino acids was conceived as a first-tier screening method, and its application to expanded newborn screening has been enormously successful. However, unlike amino acid screening (which uses amino acid analysis as its second-tier validation of screening results), acylcarnitine "profiling" also assumed the role of second-tier validation, due to the lack of a generally accepted second-tier acylcarnitine determination method. In this report, we present results from the application of our validated UHPLC-MS/MS second-tier method for the quantification of total carnitine, free carnitine, butyrobetaine, and acylcarnitines to patient samples with known diagnoses: malonic acidemia, short-chain acyl-CoA dehydrogenase deficiency (SCADD) or isobutyryl-CoA dehydrogenase deficiency (IBD), 3-methyl-crotonyl carboxylase deficiency (3-MCC) or ß-ketothiolase deficiency (BKT), and methylmalonic acidemia (MMA). We demonstrate the assay's ability to separate constitutional isomers and diastereomeric acylcarnitines and generate values with a high level of accuracy and precision. These capabilities are unavailable when using tandem MS "profiles". We also show examples of research interest, where separation of acylcarnitine species and accurate and precise acylcarnitine quantification is necessary.


Subject(s)
Acetyl-CoA C-Acyltransferase/deficiency , Acyl-CoA Dehydrogenase/deficiency , Amino Acid Metabolism, Inborn Errors/diagnosis , Carbon-Carbon Ligases/deficiency , Carnitine/analogs & derivatives , Lipid Metabolism, Inborn Errors/diagnosis , Urea Cycle Disorders, Inborn/diagnosis , Acetyl-CoA C-Acyltransferase/blood , Acetyl-CoA C-Acyltransferase/cerebrospinal fluid , Acetyl-CoA C-Acyltransferase/urine , Acyl-CoA Dehydrogenase/blood , Acyl-CoA Dehydrogenase/cerebrospinal fluid , Acyl-CoA Dehydrogenase/urine , Amino Acid Metabolism, Inborn Errors/blood , Amino Acid Metabolism, Inborn Errors/cerebrospinal fluid , Amino Acid Metabolism, Inborn Errors/urine , Betaine/analogs & derivatives , Betaine/blood , Betaine/cerebrospinal fluid , Betaine/urine , Carbon-Carbon Ligases/blood , Carbon-Carbon Ligases/cerebrospinal fluid , Carbon-Carbon Ligases/urine , Carnitine/blood , Carnitine/cerebrospinal fluid , Carnitine/urine , Chromatography, High Pressure Liquid/methods , Chromatography, High Pressure Liquid/standards , Female , Humans , Infant, Newborn , Isomerism , Lipid Metabolism, Inborn Errors/blood , Lipid Metabolism, Inborn Errors/cerebrospinal fluid , Lipid Metabolism, Inborn Errors/urine , Male , Neonatal Screening , Reproducibility of Results , Sensitivity and Specificity , Tandem Mass Spectrometry/standards , Urea Cycle Disorders, Inborn/blood , Urea Cycle Disorders, Inborn/cerebrospinal fluid , Urea Cycle Disorders, Inborn/urine
7.
Anal Chem ; 87(17): 8994-9001, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26270397

ABSTRACT

A validated quantitative method for the determination of free and total carnitine, butyrobetaine, and acylcarnitines is presented. The versatile method has four components: (1) isolation using strong cation-exchange solid-phase extraction, (2) derivatization with pentafluorophenacyl trifluoromethanesulfonate, (3) sequential ion-exchange/reversed-phase (ultra) high-performance liquid chromatography [(U)HPLC] using a strong cation-exchange trap in series with a fused-core HPLC column, and (4) detection with electrospray ionization multiple reaction monitoring (MRM) mass spectrometry (MS). Standardized carnitine along with 65 synthesized, standardized acylcarnitines (including short-chain, medium-chain, long-chain, dicarboxylic, hydroxylated, and unsaturated acyl moieties) were used to construct multiple-point calibration curves, resulting in accurate and precise quantification. Separation of the 65 acylcarnitines was accomplished in a single chromatogram in as little as 14 min. Validation studies were performed showing a high level of accuracy, precision, and reproducibility. The method provides capabilities unavailable by tandem MS procedures, making it an ideal approach for confirmation of newborn screening results and for clinical and basic research projects, including treatment protocol studies, acylcarnitine biomarker studies, and metabolite studies using plasma, urine, tissue, or other sample matrixes.


Subject(s)
Betaine/analogs & derivatives , Carnitine/analogs & derivatives , Carnitine/analysis , Muscle, Skeletal/chemistry , Animals , Betaine/analysis , Betaine/blood , Betaine/urine , Carnitine/blood , Carnitine/urine , Chromatography, High Pressure Liquid , Diabetes Mellitus, Experimental/diagnosis , Humans , Mesylates/chemistry , Rats , Solid Phase Extraction , Spectrometry, Mass, Electrospray Ionization , Trimethylsilyl Compounds/chemistry
8.
J Biol Chem ; 289(14): 10223-34, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24558043

ABSTRACT

Rat hearts were perfused with [1,2,3,4-(13)C4]palmitic acid (M+4), and the isotopic patterns of myocardial acylcarnitines and acyl-CoAs were analyzed using ultra-HPLC-MS/MS. The 91.2% (13)C enrichment in palmitoylcarnitine shows that little endogenous (M+0) palmitate contributed to its formation. The presence of M+2 myristoylcarnitine (95.7%) and M+2 acetylcarnitine (19.4%) is evidence for ß-oxidation of perfused M+4 palmitic acid. Identical enrichment data were obtained in the respective acyl-CoAs. The relative (13)C enrichment in M+4 (84.7%, 69.9%) and M+6 (16.2%, 17.8%) stearoyl- and arachidylcarnitine, respectively, clearly shows that the perfused palmitate is chain-elongated. The observed enrichment of (13)C in acetylcarnitine (19%), M+6 stearoylcarnitine (16.2%), and M+6 arachidylcarnitine (17.8%) suggests that the majority of two-carbon units for chain elongation are derived from ß-oxidation of [1,2,3,4-(13)C4]palmitic acid. These data are explained by conversion of the M+2 acetyl-CoA to M+2 malonyl-CoA, which serves as the acceptor for M+4 palmitoyl-CoA in chain elongation. Indeed, the (13)C enrichment in mitochondrial acetyl-CoA (18.9%) and malonyl-CoA (19.9%) are identical. No (13)C enrichment was found in acylcarnitine species with carbon chain lengths between 4 and 12, arguing against the simple reversal of fatty acid ß-oxidation. Furthermore, isolated, intact rat heart mitochondria 1) synthesize malonyl-CoA with simultaneous inhibition of carnitine palmitoyltransferase 1b and 2) catalyze the palmitoyl-CoA-dependent incorporation of (14)C from [2-(14)C]malonyl-CoA into lipid-soluble products. In conclusion, rat heart has the capability to chain-elongate fatty acids using mitochondria-derived two-carbon chain extenders. The data suggest that the chain elongation process is localized on the outer surface of the mitochondrial outer membrane.


Subject(s)
Acetyl Coenzyme A/metabolism , Enzyme Inhibitors/pharmacology , Mitochondria, Heart/metabolism , Myocardium/metabolism , Palmitic Acid/metabolism , Palmitic Acid/pharmacology , Animals , Carnitine O-Palmitoyltransferase/metabolism , Enzyme Inhibitors/metabolism , Malonyl Coenzyme A/metabolism , Muscle Proteins/metabolism , Oxidation-Reduction , Palmitoyl Coenzyme A/metabolism , Perfusion , Rats , Rats, Inbred F344
9.
FASEB J ; 26(10): 4316-26, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22782973

ABSTRACT

Mitochondrial enoyl-CoA isomerase (ECI1) is an auxiliary enzyme involved in unsaturated fatty acid oxidation. In contrast to most of the other enzymes involved in fatty acid oxidation, a deficiency of ECI1 has yet to be identified in humans. We used wild-type (WT) and Eci1-deficient knockout (KO) mice to explore a potential presentation of human ECI1 deficiency. Upon food withdrawal, Eci1-deficient mice displayed normal blood ß-hydroxybutyrate levels (WT 1.09 mM vs. KO 1.10 mM), a trend to lower blood glucose levels (WT 4.58 mM vs. KO 3.87 mM, P=0.09) and elevated blood levels of unsaturated acylcarnitines, in particular C12:1 acylcarnitine (WT 0.03 µM vs. KO 0.09 µM, P<0.01). Feeding an olive oil-rich diet induced an even greater increase in C12:1 acylcarnitine levels (WT 0.01 µM vs. KO 0.04 µM, P<0.01). Overall, the phenotypic presentation of Eci1-deficient mice is mild, possibly caused by the presence of a second enoyl-CoA isomerase (Eci2) in mitochondria. Knockdown of Eci2 in Eci1-deficient fibroblasts caused a more pronounced accumulation of C12:1 acylcarnitine on incubation with unsaturated fatty acids (12-fold, P<0.05). We conclude that Eci2 compensates for Eci1 deficiency explaining the mild phenotype of Eci1-deficient mice. Hypoglycemia and accumulation of C12:1 acylcarnitine might be diagnostic markers to identify ECI1 deficiency in humans.


Subject(s)
Carbon-Carbon Double Bond Isomerases/metabolism , Fatty Acids, Unsaturated/metabolism , Mitochondria/enzymology , Animals , Blood Glucose/metabolism , Carbon-Carbon Double Bond Isomerases/genetics , Carnitine/analogs & derivatives , Carnitine/blood , Cells, Cultured , Chromatography, High Pressure Liquid , Dodecenoyl-CoA Isomerase , Immunoblotting , Mass Spectrometry , Mice , Mice, Knockout , Oxidation-Reduction , Real-Time Polymerase Chain Reaction
10.
Metabolism ; 61(7): 966-73, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22209095

ABSTRACT

Hydroxybutyrylcarnitine (HB-carnitine) is a metabolite that has been associated with insulin resistance and type 2 diabetes mellitus. It is currently unknown whether HB-carnitine can be produced from D-3-hydroxybutyrate (D-3HB), a ketone body; but its formation from L-3-HB-CoA, a fatty acid ß-oxidation intermediate, is well established. We aimed to assess which stereoisomers of 3-HB-carnitine are present in vivo. Ketosis and increased fatty acid oxidation were induced in 12 lean healthy men by a 38-hour fasting period. The D-3HB kinetics (stable isotope technique) and stereoisomers of muscle 3-HB-carnitine (high-performance liquid chromatography/ultra-performance liquid chromatography-tandem mass spectrometry) were measured. Muscle D-3HB-carnitine content was much higher compared with L-3HB-carnitine. In addition, muscle D-3HB-carnitine correlated significantly with D-3-HB production. Following the finding that a ketone body can be converted into a carnitine ester in vivo, we show in vitro that D-3-HB can be converted into HB-carnitine (ketocarnitine) via an acyl-CoA synthetase reaction in several tissues including human muscle. During fasting, HB-carnitine in muscle is derived mainly from the ketone body D-3HB. The role of D-3HB-carnitine synthesis in metabolism remains to be elucidated.


Subject(s)
Carnitine/analogs & derivatives , Carnitine/metabolism , Ketosis/metabolism , Vitamin B Complex/metabolism , Adolescent , Adult , Carnitine/analysis , Coenzyme A Ligases/metabolism , Fasting/metabolism , Fatty Acids/metabolism , Humans , Ketone Bodies/metabolism , Male , Middle Aged , Muscle, Skeletal/chemistry , Vitamin B Complex/analysis , Young Adult
11.
Biochim Biophys Acta ; 1807(11): 1373-82, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21320465

ABSTRACT

Mitochondrial dysfunction is a major contributor in heart failure (HF). We investigated whether the decrease in respirasome organization reported by us previously in cardiac mitochondria in HF is due to changes in the phospholipids of the mitochondrial inner membrane or modifications of the subunits of the electron transport chain (ETC) complexes. The contents of the main phospholipid species, including cardiolipin, as well as the molecular species of cardiolipin were unchanged in cardiac mitochondria in HF. Oxidized cardiolipin molecular species were not observed. In heart mitochondria isolated from HF, complex IV not incorporated into respirasomes exhibits increased threonine phosphorylation. Since HF is associated with increased adrenergic drive to cardiomyocytes, this increased protein phosphorylation might be explained by the involvement of cAMP-activated protein kinase. Does the preservation of cAMP-induced phosphorylation changes of mitochondrial proteins or the addition of exogenous cAMP have similar effects on oxidative phosphorylation? The usage of phosphatase inhibitors revealed a specific decrease in complex I-supported respiration with glutamate. In saponin-permeabilized cardiac fibers, pre-incubation with cAMP decreases oxidative phosphorylation due to a defect localized at complex IV of the ETC inter alia. We propose that phosphorylation of specific complex IV subunits decreases oxidative phosphorylation either by limiting the incorporation of complex IV in supercomplexes or by decreasing supercomplex stability.


Subject(s)
Cardiolipins/metabolism , Electron Transport Complex IV/metabolism , Heart Failure/physiopathology , Mitochondria, Heart/chemistry , Mitochondria, Heart/metabolism , Threonine/metabolism , Animals , Cardiolipins/chemistry , Cell Respiration/physiology , Cyclic AMP/metabolism , Dogs , Humans , Male , Myocardium/cytology , Myocardium/metabolism , Oxidative Phosphorylation , Oxygen/metabolism , Oxygen Consumption , Phospholipids/chemistry , Phospholipids/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley
12.
J Lipid Res ; 52(1): 125-35, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20858593

ABSTRACT

Cardiolipin (CL) is a phospholipid predominantly found in the mitochondrial inner membrane and is associated structurally with individual complexes of the electron transport chain (ETC). Because the ETC is the major mitochondrial reactive oxygen species (ROS)-generating site, the proximity to the ETC and bisallylic methylenes of the PUFA chains of CL make it a likely target of ROS in the mitochondrial inner membrane. Oxidized cellular CL products, uniquely derived from ROS-induced autoxidation, could serve as biomarkers for the presence of the ROS and could help in the understanding of the mechanism of oxidative stress. Because major CL species have four unsaturated acyl chains, whereas other phospholipids usually have only one in the sn-2 position, characterization of oxidized CL is highly challenging. In the current study, we exposed CL, under aerobic conditions, to singlet oxygen (¹O2), the radical initiator 2,2'-azobis(2-methylpropionamidine) dihydrochloride, or room air, and the oxidized CL species were characterized by HPLC-tandem mass spectrometry (MS/MS). Our reverse-phase ion-pair HPLC-MS/MS method can characterize the major and minor oxidized CL species by detecting distinctive fragment ions associated with specific oxidized species. The HPLC-MS/MS results show that monohydroperoxides and bis monohydroperoxides were generated under all three conditions. However, significant amounts of CL dihydroperoxides were produced only by ¹O2-mediated oxidation. These products were barely detectable from radical oxidation either in a liposome bilayer or in thin film. These observations are only possible due to the chromatographic separation of the different oxidized species.


Subject(s)
Cardiolipins/chemistry , Amidines/chemistry , Amidines/metabolism , Cardiolipins/metabolism , Chromatography, High Pressure Liquid/methods , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/metabolism , Singlet Oxygen/metabolism , Tandem Mass Spectrometry
13.
Anal Biochem ; 401(1): 114-24, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20184857

ABSTRACT

The measurement of acyl-CoA dehydrogenase activities is an essential part of the investigation of patients with suspected defects in fatty acid oxidation. Multiple methods are available for the synthesis of the substrates used for measuring acyl-CoA dehydrogenase activities; however, the yields are low and the products are used without purification. In addition, the reported characterization of acyl-CoAs focuses on the CoA moiety, not on the acyl group. Here we describe the synthesis of three medium-chain acyl-CoAs from mixed anhydrides of the fatty acids using an aqueous-organic solvent mixture optimized to obtain the highest yield. First, cis-4-decenoic acid and 2,6-dimethylheptanoic acid were prepared (3-phenylpropionic acid is commercially available). These were characterized by gas chromatography/mass spectrometry (GC/MS), (1)H nuclear magnetic resonance (NMR), and (13)C NMR. Then cis-4-decenoyl-CoA, 3-phenylpropionyl-CoA, and 2,6-dimethylheptanoyl-CoA were synthesized. These were then purified by ion exchange solid-phase extraction using 2-(2-pyridyl)ethyl-functionalized silica gel, followed by reversed-phase semipreparative high-performance liquid chromatography with ultraviolet detection (HPLC-UV). The purified acyl-CoAs were characterized by analytical HPLC-UV followed by data-dependent tandem mass spectrometry (MS/MS) analysis on the largest responding MS mass (HPLC-UV-MS-MS/MS) and (13)C NMR. The yields of the purified acyl-CoAs were between 75% and 78% based on coenzyme A trilithium salt (CoASH). Acyl-CoA dehydrogenase activities were measured in rat skeletal muscle mitochondria using, as substrates, the synthesized cis-4-decenoyl-CoA, 3-phenylpropionyl-CoA, and 2,6-dimethylheptanoyl-CoA. These results were compared with the results using our standard substrates butyryl-CoA, octanoyl-CoA, and palmitoyl-CoA.


Subject(s)
Acyl Coenzyme A/chemical synthesis , Coenzyme A/chemical synthesis , Acyl Coenzyme A/chemistry , Acyl Coenzyme A/isolation & purification , Coenzyme A/chemistry , Coenzyme A/isolation & purification , Gas Chromatography-Mass Spectrometry , Magnetic Resonance Spectroscopy , Solid Phase Extraction
14.
J Lipid Res ; 51(4): 856-65, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19965604

ABSTRACT

An improved high-performance liquid chromatography-mass spectrometry method for the separation and characterization of cardiolipin molecular species is presented. Reverse-phase ion pair chromatography with acidified triethylamine resulted in increased chromatographic retention and resolution when compared with chromatography without acidified triethylamine. Using a hybrid triple quadrupole linear ion trap mass spectrometer to generate MS/MS spectra revealed three regions within each spectrum that could be used to deduce the structure of the cardiolipin molecular species: the diacylglycerol phosphate region, the monoacylglycerol phosphate region, and the fatty acid region. Cardiolipin standards of known composition were analyzed and exhibited expected chromatographic and mass spectral results. Two minor components in commercial bovine heart cardiolipin, (with the same molecular weight but different chromatographic retention times), were shown to differ by fatty acid composition: (C18:2)(2)(C18:1)(2) versus (C18:2)(3)(C18:0)(1). These compounds were then analyzed by HPLC-MS(3) to examine specific diacylglycerol phosphate generated fatty acid fragmentation. Also, two commercial sources of bovine heart cardiolipin were shown to have minor differences in cardiolipin species content. Cardiolipin isolated from rat liver, mouse heart, and dog heart mitochondria were then characterized and the relative distributions of the major cardiolipin species were determined.


Subject(s)
Cardiolipins/chemistry , Cardiolipins/isolation & purification , Chromatography, High Pressure Liquid/methods , Tandem Mass Spectrometry/methods , Animals , Chromatography, Reverse-Phase , Dogs , Ethylamines , Fatty Acids/analysis , Hydrogen-Ion Concentration , Male , Mice , Mice, Inbred C57BL , Mitochondria, Heart/chemistry , Mitochondria, Liver , Molecular Structure , Organ Specificity , Rats , Rats, Sprague-Dawley , Species Specificity , Tandem Mass Spectrometry/instrumentation
15.
Biochim Biophys Acta ; 1791(8): 806-15, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19465148

ABSTRACT

Several mouse models for mitochondrial fatty acid beta-oxidation (FAO) defects have been developed. So far, these models have contributed little to our current understanding of the pathophysiology. The objective of this study was to explore differences between murine and human FAO. Using a combination of analytical, biochemical and molecular methods, we compared fibroblasts of long chain acyl-CoA dehydrogenase knockout (LCAD(-/-)), very long chain acyl-CoA dehydrogenase knockout (VLCAD(-/-)) and wild type mice with fibroblasts of VLCAD-deficient patients and human controls. We show that in mice, LCAD and VLCAD have overlapping and distinct roles in FAO. The absence of VLCAD is apparently fully compensated, whereas LCAD deficiency is not. LCAD plays an essential role in the oxidation of unsaturated fatty acids such as oleic acid, but seems redundant in the oxidation of saturated fatty acids. In strong contrast, LCAD is neither detectable at the mRNA level nor at the protein level in men, making VLCAD indispensable in FAO. Our findings open new avenues to employ the existing mouse models to study the pathophysiology of human FAO defects.


Subject(s)
Fatty Acids/metabolism , Mitochondria/metabolism , Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Animals , Carnitine/analogs & derivatives , Carnitine/chemistry , Carnitine/metabolism , Cell Line , Chromatography, High Pressure Liquid , Fibroblasts/enzymology , Humans , Mice , Oxidation-Reduction , Tandem Mass Spectrometry
16.
J Nutr ; 139(6): 1073-81, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19369366

ABSTRACT

Inefficient muscle long-chain fatty acid (LCFA) combustion is associated with insulin resistance, but molecular links between mitochondrial fat catabolism and insulin action remain controversial. We hypothesized that plasma acylcarnitine profiling would identify distinct metabolite patterns reflective of muscle fat catabolism when comparing individuals bearing a missense G304A uncoupling protein 3 (UCP3 g/a) polymorphism to controls, because UCP3 is predominantly expressed in skeletal muscle and g/a individuals have reduced whole-body fat oxidation. MS analyses of 42 carnitine moieties in plasma samples from fasting type 2 diabetics (n = 44) and nondiabetics (n = 12) with or without the UCP3 g/a polymorphism (n = 28/genotype: 22 diabetic, 6 nondiabetic/genotype) were conducted. Contrary to our hypothesis, genotype had a negligible impact on plasma metabolite patterns. However, a comparison of nondiabetics vs. type 2 diabetics revealed a striking increase in the concentrations of fatty acylcarnitines reflective of incomplete LCFA beta-oxidation in the latter (i.e. summed C10- to C14-carnitine concentrations were approximately 300% of controls; P = 0.004). Across all volunteers (n = 56), acetylcarnitine rose and propionylcarnitine decreased with increasing hemoglobin A1c (r = 0.544, P < 0.0001; and r = -0.308, P < 0.05, respectively) and with increasing total plasma acylcarnitine concentration. In proof-of-concept studies, we made the novel observation that C12-C14 acylcarnitines significantly stimulated nuclear factor kappa-B activity (up to 200% of controls) in RAW264.7 cells. These results are consistent with the working hypothesis that inefficient tissue LCFA beta-oxidation, due in part to a relatively low tricarboxylic acid cycle capacity, increases tissue accumulation of acetyl-CoA and generates chain-shortened acylcarnitine molecules that activate proinflammatory pathways implicated in insulin resistance.


Subject(s)
Black or African American , Carnitine/analogs & derivatives , Citric Acid Cycle/physiology , Diabetes Mellitus, Type 2/blood , Fatty Acids/metabolism , Adult , Aged , Aged, 80 and over , Carnitine/blood , Diabetes Mellitus, Type 2/ethnology , Diabetes Mellitus, Type 2/genetics , Fatty Acids/chemistry , Female , Humans , Ion Channels/genetics , Middle Aged , Mitochondrial Proteins/genetics , Mutation, Missense , NF-kappa B/metabolism , Obesity/complications , Oxidation-Reduction , Polymorphism, Genetic , Uncoupling Protein 3 , Young Adult
17.
J Mol Cell Cardiol ; 46(6): 1008-15, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19303420

ABSTRACT

Aging enhances cardiac injury during ischemia and reperfusion compared to the adult heart, including in the Fischer 344 rat model of aging (F344). In interfibrillar cardiac mitochondria obtained from the elderly F344 rat, the rate of oxidative phosphorylation and the activity of electron transport complex III is decreased, concomitant with an increase in the production of reactive oxygen species. In the isolated, perfused heart, 25 min of global ischemia results in additional damage to complex III. We proposed that ischemic damage superimposed upon the aging defect augments production of reactive oxygen species leading to greater oxidative damage in the aged heart. Cardiolipin is an oxidatively sensitive phospholipid located in the inner mitochondrial membrane. Oxidative damage to cardiolipin was assessed by characterization of the individual molecular species of cardiolipin via reverse phase HPLC and electrospray mass spectrometry (MS). The predominant molecular species of cardiolipin (95%) contains four linoleic acid residues (C18:2). Ischemia and reperfusion did not alter the content or composition of cardiolipin in the adult heart. Following ischemia and reperfusion in the aged heart, a new molecular species of cardiolipin was present with mass increased by 48 Da, suggesting the addition of three oxygen atoms. MS fragmentation localized the added mass to the C18:2 residues. Ischemia alone was sufficient to modify cardiolipin in the aged heart whereas cardiolipin in the adult heart remained unaltered. Thus, age-enhanced oxidative damage occurs within mitochondria in the heart during ischemia and reperfusion, especially during ischemia.


Subject(s)
Aging/physiology , Cardiolipins/chemistry , Cardiolipins/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Animals , Chromatography, High Pressure Liquid , Electron Transport Complex III/metabolism , In Vitro Techniques , Lipid Peroxidation , Male , Mitochondria, Heart/metabolism , Molecular Structure , Myocardium/pathology , Oxidation-Reduction , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Spectrometry, Mass, Electrospray Ionization
18.
Clin Chem ; 54(9): 1451-62, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18678604

ABSTRACT

BACKGROUND: Analysis of carnitine and acylcarnitines by tandem mass spectrometry (MS/MS) has limitations. First, preparation of butyl esters partially hydrolyzes acylcarnitines. Second, isobaric nonacylcarnitine compounds yield false-positive results in acylcarnitine tests. Third, acylcarnitine constitutional isomers cannot be distinguished. METHODS: Carnitine and acylcarnitines were isolated by ion-exchange solid-phase extraction, derivatized with pentafluorophenacyl trifluoromethanesulfonate, separated by HPLC, and detected with an ion trap mass spectrometer. Carnitine was quantified with d(3)-carnitine as the internal standard. Acylcarnitines were quantified with 42 synthesized calibrators. The internal standards used were d(6)-acetyl-, d(3)-propionyl-, undecanoyl-, undecanedioyl-, and heptadecanoylcarnitine. RESULTS: Example recoveries [mean (SD)] were 69.4% (3.9%) for total carnitine, 83.1% (5.9%) for free carnitine, 102.2% (9.8%) for acetylcarnitine, and 107.2% (8.9%) for palmitoylcarnitine. Example imprecision results [mean (SD)] within runs (n = 6) and between runs (n = 18) were, respectively: total carnitine, 58.0 (0.9) and 57.4 (1.7) micromol/L; free carnitine, 44.6 (1.5) and 44.3 (1.2) micromol/L; acetylcarnitine, 7.74 (0.51) and 7.85 (0.69) micromol/L; and palmitoylcarnitine, 0.12 (0.01) and 0.11 (0.02) micromol/L. Standard-addition slopes and linear regression coefficients were 1.00 and 0.9998, respectively, for total carnitine added to plasma, 0.99 and 0.9997 for free carnitine added to plasma, 1.04 and 0.9972 for octanoylcarnitine added to skeletal muscle, and 1.05 and 0.9913 for palmitoylcarnitine added to skeletal muscle. Reference intervals for plasma, urine, and skeletal muscle are provided. CONCLUSIONS: This method for analysis of carnitine and acylcarnitines overcomes the observed limitations of MS/MS methods.


Subject(s)
Carnitine/analysis , Carnitine/chemistry , Chromatography, High Pressure Liquid/methods , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Acetylation , Calibration , Carnitine/metabolism , Humans , Molecular Structure
19.
Arch Physiol Biochem ; 114(3): 161-70, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18629681

ABSTRACT

Hepatic mitochondrial fatty acid oxidation and ketogenesis increase during starvation. Carnitine palmitoyltransferase I (CPT-I) catalyses the rate-controlling step in the overall pathway and retains its control over beta-oxidation under fed, starved and diabetic conditions. To determine the factors contributing to the reported several-fold increase in fatty acid oxidation in perfused livers, we measured the V(max) and K(m) values for palmitoyl-CoA and carnitine, the K(i) (and IC(50)) values for malonyl-CoA in isolated liver mitochondria as well as the hepatic malonyl-CoA and carnitine contents in control and 48 h starved rats. Since CPT-I is localized in the mitochondrial outer membrane and in contact sites, the kinetic properties of CPT-I also was determined in these submitochondrial structures. After 48 h starvation, there is: (a) a significant increase in K(i) and decrease in hepatic malonyl-CoA content; (b) a decreased K(m) for palmitoyl-CoA; and (c) increased catalytic activity (V(max)) and CPT-I protein abundance that is significantly greater in contact sites compared with outer membranes. Based on these changes the estimated increase in mitochondrial fatty acid oxidation is significantly less than that observed in perfused liver. This suggests that CPT-I is regulated in vivo by additional mechanism(s) lost during mitochondrial isolation or/and that mitochondrial oxidation of peroxisomal beta-oxidation products contribute to the increased ketogenesis by bypassing CPT-I. Furthermore, the greater increase in CPT-I protein in contact sites as compared to outer membranes emphasizes the significance of contact sites in hepatic fatty acid oxidation.


Subject(s)
Carnitine O-Palmitoyltransferase/metabolism , Carnitine/metabolism , Liver/metabolism , Malonyl Coenzyme A/metabolism , Mitochondria, Liver/enzymology , Starvation/metabolism , Animals , Blotting, Western , Body Weight , Electrophoresis, Polyacrylamide Gel , Male , Organ Size , Rats , Rats, Sprague-Dawley
20.
Anal Biochem ; 376(2): 275-6, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18355435

ABSTRACT

A novel procedure for the quantitative isolation and purification of acyl-coenzyme A esters is presented. The procedure involves two steps: (1) tissue extraction using acetonitrile/2-propanol (3+1, v+v) followed by 0.1M potassium phosphate, pH 6.7, and (2) purification using 2-(2-pyridyl)ethyl-functionalized silica gel. Recoveries determined by adding radiolabeled acetyl-, malonyl-, octanoyl-, oleoyl-, palmitoyl-, or arachidonyl-coenzyme A to powdered rat liver varied 93-104% for tissue extraction and 83-90% for solid-phase extraction. The procedure described allows for isolation and purification, with high recoveries, of acyl-coenzyme A esters differing widely in chain length and saturation.


Subject(s)
Acyl Coenzyme A/isolation & purification , Liver/chemistry , Solid Phase Extraction/methods , 2-Propanol/chemistry , Acetonitriles/chemistry , Acyl Coenzyme A/chemistry , Animals , Esters , Malonyl Coenzyme A/chemistry , Malonyl Coenzyme A/isolation & purification , Palmitoyl Coenzyme A/chemistry , Palmitoyl Coenzyme A/isolation & purification , Phosphates/chemistry , Potassium Compounds/chemistry , Protons , Rats , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...