Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biosens Bioelectron ; 264: 116647, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-39173338

RESUMEN

ß-Hydroxybutyrate (BHB) is a substantial physiological ketone body. Its elevated concentration causes ketoacidosis, which is a disorder with a high mortality rate. Therefore, there is an urgent need to develop a simple method for the in-situ monitoring of BHB in urine. In this study, a photonic crystal hydrogel (PCH) sensing material for the detection of urinary ketones was prepared by embedding a two-dimensional polystyrene photonic crystal array (PCA) in a hydrogel functionalized with ß-hydroxybutyrate dehydrogenase (BHBDH). BHBDH catalyzes the interconversion between ß-hydroxybutyrate and acetoacetic acid and relies on the cofactor nicotinamide adenine dinucleotide (NAD+) to participate in the reaction process. The catalytic cycle of converting ß-hydroxybutyrate to acetoacetate generates H+, which reduces the electrostatic repulsion between the carboxyl groups in the hydrogel network, ultimately leading to the shrinkage of the hydrogel volume. The hydrogel volume change was detected by measuring the diameter of the Debye diffraction ring, thus reflecting the concentration of BHB. When the concentration of BHB was increased from 0 to 10 mM, the reflection spectrum of PCH shifted for 117 nm within 60 min, consequently, the structural color of PCH changed from red to green and finally to blue. The material was used for quantitative detection of BHB with a detection limit of 48.94 µM. Then it was used for detection in artificial urine samples. While, this smart and reusable sensing material could provide a more convenient and efficient strategy for the ketone body detection in clinical diagnosis and point-of-care monitoring.


Asunto(s)
Ácido 3-Hidroxibutírico , Técnicas Biosensibles , Hidroxibutirato Deshidrogenasa , Hidroxibutirato Deshidrogenasa/química , Ácido 3-Hidroxibutírico/química , Ácido 3-Hidroxibutírico/orina , Ácido 3-Hidroxibutírico/análisis , Humanos , Cuerpos Cetónicos/orina , Cuerpos Cetónicos/química , Hidrogeles/química , Enzimas Inmovilizadas/química , Límite de Detección , Fotones
2.
Protein Sci ; 33(7): e5025, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38864689

RESUMEN

Polyhydroxyalkanoates are a class of biodegradable, thermoplastic polymers which represent a major carbon source for various bacteria. Proteins which mediate the translocation of polyhydroxyalkanoate breakdown products, such as ß-hydroxybutyrate (BHB)-a ketone body which in humans serves as an important biomarker, have not been well characterized. In our investigation to screen a solute-binding protein (SBP) which can act as a suitable recognition element for BHB, we uncovered insights at the intersection of bacterial metabolism and diagnostics. Herein, we identify SBPs associated with putative ATP-binding cassette transporters that specifically recognize BHB, with the potential to serve as recognition elements for continuous quantification of this analyte. Through bioinformatic analysis, we identified candidate SBPs from known metabolizers of polyhydroxybutyrate-including proteins from Cupriavidus necator, Ensifer meliloti, Paucimonas lemoignei, and Thermus thermophilus. After recombinant expression in Escherichia coli, we demonstrated with intrinsic tryptophan fluorescence spectroscopy that four candidate proteins interacted with BHB, ranging from nanomolar to micromolar affinity. Tt.2, an intrinsically thermostable protein from Thermus thermophilus, was observed to have the tightest binding and specificity for BHB, which was confirmed by isothermal calorimetry. Structural analyses facilitated by AlphaFold2, along with molecular docking and dynamics simulations, were used to hypothesize key residues in the binding pocket and to model the conformational dynamics of substrate unbinding. Overall, this study provides strong evidence identifying the cognate ligands of SBPs which we hypothesize to be involved in prokaryotic cellular translocation of polyhydroxyalkanoate breakdown products, while highlighting these proteins' promising biotechnological application.


Asunto(s)
Ácido 3-Hidroxibutírico , Ácido 3-Hidroxibutírico/metabolismo , Ácido 3-Hidroxibutírico/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas de Unión Periplasmáticas/metabolismo , Proteínas de Unión Periplasmáticas/química , Proteínas de Unión Periplasmáticas/genética , Escherichia coli/metabolismo , Escherichia coli/genética , Cuerpos Cetónicos/metabolismo , Cuerpos Cetónicos/química
3.
Bioessays ; 43(6): e2000312, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33857328

RESUMEN

Biocidal agents such as formaldehyde and glutaraldehyde are able to inactivate several coronaviruses including SARS-CoV-2. In this article, an insight into one mechanism for the inactivation of these viruses by those two agents is presented, based on analysis of previous observations during electron microscopic examination of several members of the orthocoronavirinae subfamily, including the new virus SARS-CoV-2. This inactivation is proposed to occur through Schiff base reaction-induced conformational changes in the spike glycoprotein leading to its disruption or breakage, which can prevent binding of the virus to cellular receptors. Also, a new prophylactic and therapeutic measure against SARS-CoV-2 using acetoacetate is proposed, suggesting that it could similarly break the viral spike through Schiff base reaction with lysines of the spike protein. This measure needs to be confirmed experimentally before consideration. In addition, a new line of research is proposed to help find a broad-spectrum antivirus against several members of this subfamily.


Asunto(s)
Desinfectantes/farmacología , Cuerpos Cetónicos/farmacología , SARS-CoV-2/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/metabolismo , Animales , Antivirales/química , Antivirales/farmacología , Desinfectantes/química , Formaldehído/química , Formaldehído/farmacología , Glutaral/química , Glutaral/farmacología , Humanos , Cuerpos Cetónicos/química , Cuerpos Cetónicos/metabolismo , Cetosis/etiología , Cetosis/virología , SARS-CoV-2/patogenicidad , Virión/efectos de los fármacos , Virión/patogenicidad
4.
Biochimie ; 183: 55-62, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33596448

RESUMEN

Succinyl-CoA:3-oxoacid coenzyme A transferase deficiency (SCOTD) is a rare autosomal recessive disorder of ketone body utilization caused by mutations in OXCT1. We performed a systematic literature search and evaluated clinical, biochemical and genetic data on 34 previously published and 10 novel patients with SCOTD. Structural mapping and in silico analysis of protein variants is also presented. All patients presented with severe ketoacidotic episodes. Age at first symptoms ranged from 36 h to 3 years (median 7 months). About 70% of patients manifested in the first year of life, approximately one quarter already within the neonatal period. Two patients died, while the remainder (95%) were alive at the time of the report. Almost all the surviving patients (92%) showed normal psychomotor development and no neurologic abnormalities. A total of 29 missense mutations are reported. Analysis of the published crystal structure of the human SCOT enzyme, paired with both sequence-based and structure-based methods to predict variant pathogenicity, provides insight into the biochemical consequences of the reported variants. Pathogenic variants cluster in SCOT protein regions that affect certain structures of the protein. The described pathogenic variants can be viewed in an interactive map of the SCOT protein at https://michelanglo.sgc.ox.ac.uk/r/oxct. This comprehensive data analysis provides a systematic overview of all cases of SCOTD published to date. Although SCOTD is a rather benign disorder with often favourable outcome, metabolic crises can be life-threatening or even fatal. As the diagnosis can only be made by enzyme studies or mutation analyses, SCOTD may be underdiagnosed.


Asunto(s)
Acidosis , Encefalopatías Metabólicas Innatas , Coenzima A Transferasas/deficiencia , Mutación Missense , Trastornos del Neurodesarrollo , Acidosis/enzimología , Acidosis/genética , Encefalopatías Metabólicas Innatas/enzimología , Encefalopatías Metabólicas Innatas/genética , Coenzima A Transferasas/química , Coenzima A Transferasas/genética , Coenzima A Transferasas/metabolismo , Cristalografía por Rayos X , Humanos , Cuerpos Cetónicos/química , Cuerpos Cetónicos/genética , Cuerpos Cetónicos/metabolismo , Trastornos del Neurodesarrollo/enzimología , Trastornos del Neurodesarrollo/genética , Dominios Proteicos
5.
J Clin Endocrinol Metab ; 105(9)2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32525972

RESUMEN

Ketone bodies - 3-hydroxybutyrate (3-OHB), acetoacetate, and acetone - are ancient, evolutionarily preserved, small fuel substrates, which uniquely can substitute and alternate with glucose under conditions of fuel and food deficiency. Once canonized as a noxious, toxic pathogen leading to ketoacidosis in patients with diabetes, it is now becoming increasingly clear that 3-OHB possesses a large number of beneficial, life-preserving effects in the fields of clinical science and medicine. 3-OHB, the most prominent ketone body, binds to specific hydroxyl-carboxylic acid receptors and inhibits histone deacetylase enzymes, free fatty acid receptors, and the NOD-like receptor protein 3 inflammasome, tentatively inhibiting lipolysis, inflammation, oxidative stress, cancer growth, angiogenesis, and atherosclerosis, and perhaps contributing to the increased longevity associated with exercise and caloric restriction. Clinically ketone bodies/ketogenic diets have for a long time been used to reduce the incidence of seizures in epilepsy and may have a role in the treatment of other neurological diseases such as dementia. 3-OHB also acts to preserve muscle protein during systemic inflammation and is an important component of the metabolic defense against insulin-induced hypoglycemia. Most recently, a number of studies have reported that 3-OHB dramatically increases myocardial blood flow and cardiac output in control subjects and patients with heart failure. At the moment, scientific interest in ketone bodies, in particular 3-OHB, is in a hectic transit and, hopefully, future, much needed, controlled clinical studies will reveal and determine to which extent the diverse biological manifestations of 3-OHB should be introduced medically.


Asunto(s)
Ácido 3-Hidroxibutírico/fisiología , Cuerpos Cetónicos/fisiología , Ácido 3-Hidroxibutírico/química , Ácido 3-Hidroxibutírico/metabolismo , Animales , Sistema Cardiovascular/metabolismo , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/fisiología , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Ejercicio Físico/fisiología , Ayuno/fisiología , Humanos , Inflamación/metabolismo , Inflamación/patología , Cuerpos Cetónicos/química , Cuerpos Cetónicos/metabolismo , Longevidad/fisiología , Redes y Vías Metabólicas/fisiología , Síndrome Metabólico/metabolismo , Síndrome Metabólico/patología
6.
Curr Pharm Des ; 26(12): 1377-1387, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31957603

RESUMEN

BACKGROUND: The ketone bodies (KB), ß-hydroxybutyrate (BHB) and acetoacetate, have been proposed for the treatment of acute and chronic neurological disorders, however, the molecular mechanisms involved in KB protection are not well understood. KB can substitute for glucose and support mitochondrial metabolism increasing cell survival. We have reported that the D-isomer of BHB (D-BHB) stimulates autophagic degradation during glucose deprivation in cultured neurons increasing cell viability. Autophagy is a lysosomal degradation process of damaged proteins and organelles activated during nutrient deprivation to obtain building blocks and energy. However, impaired or excessive autophagy can contribute to neuronal death. OBJECTIVE: The aim of the present study was to test whether D-BHB can preserve autophagic function in an in vivo model of excitotoxic damage induced by the administration of the glutamate receptor agonist, N-methyl-Daspartate (NMDA), in the rat striatum. METHODS: D-BHB was administered through an intravenous injection followed by either an intraperitoneal injection (i.v+i.p) or a continuous epidural infusion (i.v+pump), or through a continuous infusion of D-BHB alone. Changes in the autophagy proteins ATG7, ATG5, BECLIN 1 (BECN1), LC3, Sequestrosome1/p62 (SQSTM1/ p62) and the lysosomal membrane protein LAMP2, were evaluated by immunoblot. The lesion volume was measured in cresyl violet-stained brain sections. RESULTS: Autophagy is activated early after NMDA injection but autophagic degradation is impaired due to the cleavage of LAMP2. Twenty-four h after NMDA intrastriatal injection, the autophagic flux is re-established, but LAMP2 cleavage is still observed. The administration of D-BHB through the i.v+pump protocol reduced the content of autophagic proteins and the cleavage of LAMP2, suggesting decreased autophagosome formation and lysosomal membrane preservation, improving autophagic degradation. D-BHB also reduced brain injury. The i.v+i.p administration protocol and the infusion of D-BHB alone showed no effect on autophagy activation or degradation.


Asunto(s)
Autofagia , N-Metilaspartato , Ácido 3-Hidroxibutírico , Animales , Cuerpos Cetónicos/química , Neuronas/química , Neuronas/fisiología , Ratas
7.
J Photochem Photobiol B ; 185: 223-234, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29966989

RESUMEN

Raman spectroscopy (RS) is a vibrational technique that is suitable for performing biochemical analyses in human tissues and fluids. This work has investigated the identification of biochemical markers due to physical performance in the urine of swimming athletes. This was achieved by means of the Raman features that were found before and after the swimming training compared to the sedentary control subjects. These particular biochemical marker identifications refer to and infer the physiological status of individuals. The urine samples (single stream) were collected before and after the training (velocity, middle distance and distance) of professional swimmers, as well as from sedentary subjects (control). The urine samples were submitted to RS (830 nm excitation, 350 mW, 400-1800 cm-1 spectral range, 4 cm-1 resolution) and the spectra after the training were compared to the spectra before training, and subsequently, to the control subjects. The principal component analysis (PCA) was employed in order to identify the biochemicals that were responsible for the spectral differences. The Raman features of the urine samples after training showed peaks that were related to common urine metabolites, such as urea and creatinine. PCA analysis also revealed Raman features that were attributed to other metabolites, such as creatine, ketone bodies, phosphate and nitrogenous compounds in the swimmers after training. RS was a rapid and reliable technique for the evaluation of urine metabolites that were related to the physical performance of high-level swimmers, which then allowed for an accurate assessment and a control of their physiological efficiencies.


Asunto(s)
Biomarcadores/orina , Natación , Adolescente , Atletas , Creatinina/orina , Femenino , Humanos , Cuerpos Cetónicos/química , Masculino , Análisis de Componente Principal , Espectrometría Raman , Urea/orina , Adulto Joven
8.
Artículo en Inglés | MEDLINE | ID: mdl-29547803

RESUMEN

Short chain fatty acids (SCFA) and ketone bodies recently emerged as important physiological relevant metabolites because of their association with microbiota, immunology, obesity and other metabolic states. They were commonly analyzed by GC-MS with long run time and laborious sample preparation. In this study we developed a novel LC-MS/MS method using fast derivatization coupled with liquid-liquid extraction to detect SCFA and ketone bodies in plasma and feces. Several different derivatization reagents were evaluated to compare the efficiency, the sensitivity and chromatographic separation of structural isomers. O­benzylhydroxylamine was selected for its superior overall performance in reaction time and isomeric separation that allowed the measurement of each SCFAs and ketone bodies free from interferences. The derivatization procedure is facile and reproducible in aqueous-organic medium, which abolished the evaporation procedure hampering the analysis of volatile short chain acids. Enhancement in sensitivity remarkably improved the detection limit of SCFA and ketone bodies to sub-fmol level. This novel method was applied to quantify these metabolites in fecal and plasma samples from lean and DIO mouse.


Asunto(s)
Cromatografía Liquida/métodos , Ácidos Grasos Volátiles/análisis , Cuerpos Cetónicos/análisis , Extracción Líquido-Líquido/métodos , Espectrometría de Masas en Tándem/métodos , Animales , Ácidos Grasos Volátiles/sangre , Ácidos Grasos Volátiles/química , Ácidos Grasos Volátiles/aislamiento & purificación , Heces/química , Cromatografía de Gases y Espectrometría de Masas , Cuerpos Cetónicos/sangre , Cuerpos Cetónicos/química , Cuerpos Cetónicos/aislamiento & purificación , Límite de Detección , Modelos Lineales , Ratones , Ratones Obesos , Reproducibilidad de los Resultados
9.
Cell Chem Biol ; 24(8): 935-943.e7, 2017 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-28820963

RESUMEN

The α-oxoaldehyde methylglyoxal is a ubiquitous and highly reactive metabolite known to be involved in aging- and diabetes-related diseases. If not detoxified by the endogenous glyoxalase system, it exerts its detrimental effects primarily by reacting with biopolymers such as DNA and proteins. We now demonstrate that during ketosis, another metabolic route is operative via direct non-enzymatic aldol reaction between methylglyoxal and the ketone body acetoacetate, leading to 3-hydroxyhexane-2,5-dione. This novel metabolite is present at a concentration of 10%-20% of the methylglyoxal level in the blood of insulin-starved patients. By employing a metabolite-alkyne-tagging strategy it is clarified that 3-hydroxyhexane-2,5-dione is further metabolized to non-glycating species in human blood. The discovery represents a new direction within non-enzymatic metabolism and within the use of alkyne-tagging for metabolism studies and it revitalizes acetoacetate as a competent endogenous carbon nucleophile.


Asunto(s)
Acetoacetatos/química , Cuerpos Cetónicos/química , Piruvaldehído/sangre , Acetoacetatos/metabolismo , Alquinos/química , Secuencia de Aminoácidos , Cromatografía Líquida de Alta Presión , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Hexanonas/análisis , Hexanonas/sangre , Hexanonas/metabolismo , Humanos , Cuerpos Cetónicos/metabolismo , Espectrometría de Masas , Piruvaldehído/análisis , Piruvaldehído/metabolismo , Albúmina Sérica/química , Albúmina Sérica/metabolismo
10.
J Biol Chem ; 291(5): 2181-95, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26645687

RESUMEN

Acetoacetate (AA) is a ketone body and acts as a fuel to supply energy for cellular activity of various tissues. Here, we uncovered a novel function of AA in promoting muscle cell proliferation. Notably, the functional role of AA in regulating muscle cell function is further evidenced by its capability to accelerate muscle regeneration in normal mice, and it ameliorates muscular dystrophy in mdx mice. Mechanistically, our data from multiparameter analyses consistently support the notion that AA plays a non-metabolic role in regulating muscle cell function. Finally, we show that AA exerts its function through activation of the MEK1-ERK1/2-cyclin D1 pathway, revealing a novel mechanism in which AA serves as a signaling metabolite in mediating muscle cell function. Our findings highlight the profound functions of a small metabolite as signaling molecule in mammalian cells.


Asunto(s)
Acetoacetatos/farmacología , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Distrofia Muscular Animal/tratamiento farmacológico , Regeneración/efectos de los fármacos , Animales , Proliferación Celular , Ciclina D1/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Cuerpos Cetónicos/química , MAP Quinasa Quinasa 1/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/efectos de los fármacos , Células Satélite del Músculo Esquelético/citología , Transducción de Señal
11.
J Biomol Struct Dyn ; 34(12): 2658-2666, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26621475

RESUMEN

Acetoacetate (AA) is an important ketone body, which produces reactive oxygen species (ROS). Advanced glycation end products (AGEs) are defined as final products of glycation process whose production is influenced by the levels of ROS. The accumulation of AGEs in the body contributes to pathogenesis of many diseases including complications of diabetes, and Alzheimer's and Parkinson's disease. Here, we evaluated the impact of AA on production of AGEs upon incubation of human serum albumin (HSA) with glucose. The effect of AA on the AGEs formation of HSA was studied under physiological conditions after incubation with glucose for 35 days. The physical techniques including circular dichroism (CD) and fluorescence spectroscopy were used to assess the impact of AA on formation and structural changes of glycated HSA (GHSA). Our results indicated that the secondary and tertiary structural changes of GHSA were increased in the presence of AA. The fluorescence intensity measurements of AGEs also showed an increase in AGEs formation. Acetoacetate has an activator effect in formation of AGEs through ROS production. The presence of AA may result in enhanced glycation in the presence of glucose and severity of complications associated with accumulation of AGEs.


Asunto(s)
Acetoacetatos/química , Productos Finales de Glicación Avanzada/química , Dicroismo Circular , Fluorescencia , Cuerpos Cetónicos/química , Lisina/química , Especies Reactivas de Oxígeno/química , Espectrometría de Fluorescencia , Compuestos de Sulfhidrilo/química
12.
Med Hypotheses ; 85(6): 1041-3, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26541314

RESUMEN

The hypothesis is that poly-(R)-3-hydroxybutyrates (PHB), linear polymers of the ketone body, R-3-hydroxybutyrate (R-3HB), are atherogenic components of lipoprotein Lp(a). PHB are universal constituents of biological cells and are thus components of all foods. Medium chain-length PHB (<200 residues) (mPHB) are located in membranes and organelles, and short-chain PHB (<15 residues) are covalently attached to certain proteins (cPHB). PHB are highly insoluble in water, but soluble in lipids in which they exhibit a high intrinsic viscosity. They have a higher density than other cellular lipids and they are very adhesive, i.e. they engage in multiple noncovalent interactions with other molecules and salts via hydrogen, hydrophobic and coordinate bonds, thus producing insoluble deposits. Following digestive processes, PHB enter the circulation in chylomicrons and very low density lipoproteins (VLDL). The majority of the PHB (>70%) are absorbed by albumin, which transports them to the liver for disposal. When the amount of PHB in the diet exceed the capacity of albumin to safely remove them from the circulation, the excess PHB remain in the lipid core of LDL particles that become constituents of lipoprotein Lp(a), and contribute to the formation of arterial deposits. In summary, the presence of PHB ­ water-insoluble, dense, viscous, adhesive polymers ­ in the lipid cores of the LDL moieties of Lp(a) particles supports the hypothesis that PHB are atherogenic components of Lp(a).


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Hidroxibutiratos/sangre , Hidroxibutiratos/química , Lipoproteína(a)/química , Poliésteres/química , Albúminas/química , Transporte Biológico , Quilomicrones/química , Humanos , Cuerpos Cetónicos/química , Lípidos/química , Lipoproteínas VLDL/química , Polímeros/química , Prohibitinas , Factores de Riesgo , Solubilidad , Viscosidad
13.
Mol Endocrinol ; 29(8): 1134-43, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26087172

RESUMEN

Acute liver failure (ALF) is a severe and rapid liver injury, often occurring without any preexisting liver disease, which may precipitate multiorgan failure and death. ALF is often associated with impaired ß-oxidation and increased oxidative stress (OS), characterized by elevated levels of hepatic reactive oxygen species (ROS) and lipid peroxidation (LPO) products. Peroxisome proliferator-activated receptor (PPAR)α has been shown to confer hepatoprotection in acute and chronic liver injury, at least in part, related to its ability to control peroxisomal and mitochondrial ß-oxidation. To study the pathophysiological role of PPARα in hepatic response to high OS, we induced a pronounced LPO by treating wild-type and Pparα-deficient mice with high doses of fish oil (FO), containing n-3 polyunsaturated fatty acids. FO feeding of Pparα-deficient mice, in contrast to control sunflower oil, surprisingly induced coma and death due to ALF as indicated by elevated serum alanine aminotransferase, aspartate aminotransferase, ammonia, and a liver-specific increase of ROS and LPO-derived malondialdehyde. Reconstitution of PPARα specifically in the liver using adeno-associated serotype 8 virus-PPARα in Pparα-deficient mice restored ß-oxidation and ketogenesis and protected mice from FO-induced lipotoxicity and death. Interestingly, administration of the ketone body ß-hydroxybutyrate prevented FO-induced ALF in Pparα-deficient mice, and normalized liver ROS and malondialdehyde levels. Therefore, PPARα protects the liver from FO-induced OS through its regulatory actions on ketone body levels. ß-Hydroxybutyrate treatment could thus be an option to prevent LPO-induced liver damage.


Asunto(s)
Hígado Graso/metabolismo , Cuerpos Cetónicos/química , Fallo Hepático Agudo/metabolismo , PPAR alfa/deficiencia , PPAR alfa/metabolismo , Ácido 3-Hidroxibutírico/química , Animales , Ácidos Grasos/metabolismo , Hígado Graso/prevención & control , Femenino , Peroxidación de Lípido , Hígado/metabolismo , Fallo Hepático Agudo/prevención & control , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Peroxisomas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Triglicéridos/metabolismo
14.
Biomed Res Int ; 2014: 869186, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24900994

RESUMEN

BACKGROUND: Acetone, ß -hydroxybutyric acid, and acetoacetic acid are three types of ketone body that may be found in the breath, blood, and urine. Detecting altered concentrations of ketones in the breath, blood, and urine is crucial for the diagnosis and treatment of diabetic ketosis. The aim of this study was to evaluate the advantages of different detection methods for ketones, and to establish whether detection of the concentration of ketones in the breath is an effective and practical technique. METHODS: We measured the concentrations of acetone in the breath using gas chromatography-mass spectrometry and ß -hydroxybutyrate in fingertip blood collected from 99 patients with diabetes assigned to groups 1 (-), 2 (±), 3 (+), 4 (++), or 5 (+++) according to urinary ketone concentrations. RESULTS: There were strong relationships between fasting blood glucose, age, and diabetic ketosis. Exhaled acetone concentration significantly correlated with concentrations of fasting blood glucose, ketones in the blood and urine, LDL-C, creatinine, and blood urea nitrogen. CONCLUSIONS: Breath testing for ketones has a high sensitivity and specificity and appears to be a noninvasive, convenient, and repeatable method for the diagnosis and therapeutic monitoring of diabetic ketosis.


Asunto(s)
Biomarcadores/química , Cetoacidosis Diabética/diagnóstico , Cetonas/química , Ácido 3-Hidroxibutírico/sangre , Ácido 3-Hidroxibutírico/química , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Biomarcadores/orina , Glucemia/metabolismo , Nitrógeno de la Urea Sanguínea , Pruebas Respiratorias/métodos , Niño , Creatinina/metabolismo , Diabetes Mellitus/sangre , Diabetes Mellitus/metabolismo , Cetoacidosis Diabética/sangre , Cetoacidosis Diabética/metabolismo , Cetoacidosis Diabética/orina , Femenino , Cromatografía de Gases y Espectrometría de Masas/métodos , Humanos , Cuerpos Cetónicos/química , Cetonas/sangre , Cetonas/orina , Masculino , Persona de Mediana Edad , Adulto Joven
15.
Adv Exp Med Biol ; 765: 365-370, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22879057

RESUMEN

UNLABELLED: Ketone bodies are an alternative energy substrate to glucose in brain. Under conditions of oxidative stress, we hypothesize that ketosis stabilizes glucose metabolism by partitioning glucose away from oxidative metabolism towards ketone body oxidation. In this study we assessed oxidative metabolism in ketotic rat brain using stable isotope mass spectrometry analysis. The contribution of glucose and ketone bodies to oxidative metabolism was studied in cortical brain homogenates isolated from anesthetized ketotic rats. To induce chronic ketosis, rats were fed either a ketogenic (high-fat, carbohydrate restricted) or standard rodent chow for 3 weeks and then infused intravenously with tracers of [U-(13)C] glucose or [U-(13)C] acetoacetate for 60 min. The measured percent contribution of glucose or ketone bodies to oxidative metabolism was analyzed by measuring the (13)C-label incorporation into acetyl-CoA. Using mass spectrometry (gas-chromatography; GC-MS, and liquid-chromatography; LCMS) and isotopomer analysis, the fractional amount of substrate oxidation was measured as the M + 2 enrichment (%) of acetyl-CoA relative to the achieved enrichment of the infused precursors, [U-(13)C]glucose or [U-(13)C] acetoacetate. RESULTS: the percent contribution of glucose oxidation in cortical brain in rats fed the ketogenic diet was 71.2 ± 16.8 (mean% ± SD) compared to the standard chow, 89.0 ± 14.6. Acetoacetate oxidation was significantly higher with ketosis compared to standard chow, 41.7 ± 9.4 vs. 21.9 ± 10.6. These data confer the high oxidative capacity for glucose irrespective of ketotic or non-ketotic states. With ketosis induced by 3 weeks of diet, cortical brain utilizes twice as much acetoacetate compared to non-ketosis.


Asunto(s)
Acetoacetatos/metabolismo , Acetilcoenzima A/metabolismo , Encéfalo/metabolismo , Glucosa/química , Glucosa/metabolismo , Cuerpos Cetónicos/química , Cuerpos Cetónicos/metabolismo , Animales , Radioisótopos de Carbono , Cromatografía Liquida , Dieta Cetogénica , Cromatografía de Gases y Espectrometría de Masas , Masculino , Oxidación-Reducción , Ratas , Ratas Wistar
16.
J Biol Chem ; 287(40): 33227-36, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22865860

RESUMEN

3-Hydroxy-3-methylglutaryl-CoA lyase-like protein (HMGCLL1) has been annotated in the Mammalian Genome Collection as a previously unidentified human HMG-CoA lyase (HMGCL). To test the validity of this annotation and evaluate the physiological role of the protein, plasmids were constructed for protein expression in Escherichia coli and Pichia pastoris. Protein expression in E. coli produced insoluble material. In contrast, active HMGCLL1 could be recovered upon expression in P. pastoris. Antibodies were prepared against a unique peptide sequence found in the N terminus of the protein. In immunodetection experiments, the antibodies discriminated between HMGCLL1 and mitochondrial HMGCL. Purified enzyme was characterized and demonstrated to cleave HMG-CoA to acetoacetate and acetyl-CoA with catalytic and affinity properties comparable with human mitochondrial HMGCL. The deduced HMGCLL1 sequence contains an N-terminal myristoylation motif; the putative modification site was eliminated by construction of a G2A HMGCLL1. Modification of both proteins was attempted using human N-myristoyltransferase and [(3)H]myristoyl-CoA. Wild-type protein was clearly modified, whereas G2A protein was not labeled. Myristoylation of HMGCLL1 affects its cellular localization. Upon transfection of appropriate expression plasmids into COS1 cells, immunofluorescence detection indicates that G2A HMGCLL1 exhibits a diffuse pattern, suggesting a cytosolic location. In contrast, wild-type HMGCLL1 exhibits a punctate as well as a perinuclear immunostaining pattern, indicating myristoylation dependent association with nonmitochondrial membrane compartments. In control experiments with the HMGCL expression plasmid, protein is localized in the mitochondria, as anticipated. The available results for COS1 cell expression, as well as endogenous expression in U87 cells, indicate that HMGCLL1 is an extramitochondrial hydroxymethylglutaryl-CoA lyase.


Asunto(s)
Oxo-Ácido-Liasas/química , Acilcoenzima A/química , Animales , Células COS , Catálisis , Línea Celular Tumoral , Chlorocebus aethiops , Metabolismo Energético , Escherichia coli/metabolismo , Femenino , Humanos , Cuerpos Cetónicos/química , Cetonas , Lípidos/química , Lipogénesis , Masculino , Mitocondrias/metabolismo , Modelos Químicos , Mutagénesis , Neoplasias/metabolismo , Oxo-Ácido-Liasas/genética , Péptidos/química , Plásmidos/metabolismo , Ratas
17.
J Biol Chem ; 285(32): 24447-56, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-20529848

RESUMEN

Heart muscle is metabolically versatile, converting energy stored in fatty acids, glucose, lactate, amino acids, and ketone bodies. Here, we use mouse models in ketotic nutritional states (24 h of fasting and a very low carbohydrate ketogenic diet) to demonstrate that heart muscle engages a metabolic response that limits ketone body utilization. Pathway reconstruction from microarray data sets, gene expression analysis, protein immunoblotting, and immunohistochemical analysis of myocardial tissue from nutritionally modified mouse models reveal that ketotic states promote transcriptional suppression of the key ketolytic enzyme, succinyl-CoA:3-oxoacid CoA transferase (SCOT; encoded by Oxct1), as well as peroxisome proliferator-activated receptor alpha-dependent induction of the key ketogenic enzyme HMGCS2. Consistent with reduction of SCOT, NMR profiling demonstrates that maintenance on a ketogenic diet causes a 25% reduction of myocardial (13)C enrichment of glutamate when (13)C-labeled ketone bodies are delivered in vivo or ex vivo, indicating reduced procession of ketones through oxidative metabolism. Accordingly, unmetabolized substrate concentrations are higher within the hearts of ketogenic diet-fed mice challenged with ketones compared with those of chow-fed controls. Furthermore, reduced ketone body oxidation correlates with failure of ketone bodies to inhibit fatty acid oxidation. These results indicate that ketotic nutrient environments engage mechanisms that curtail ketolytic capacity, controlling the utilization of ketone bodies in ketotic states.


Asunto(s)
Miocardio/metabolismo , Animales , Isótopos de Carbono/química , Coenzima A Transferasas/metabolismo , Inmunohistoquímica/métodos , Cuerpos Cetónicos/química , Cetonas/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Miocitos Cardíacos/citología , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Ratas
18.
J Biol Chem ; 284(41): 27799-27807, 2009 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-19666922

RESUMEN

We investigated the interrelations between C(4) ketogenesis (production of beta-hydroxybutyrate + acetoacetate), C(5) ketogenesis (production of beta-hydroxypentanoate + beta-ketopentanoate), and anaplerosis in isolated rat livers perfused with (13)C-labeled octanoate, heptanoate, or propionate. Mass isotopomer analysis of C(4) and C(5) ketone bodies and of related acyl-CoA esters reveal that C(4) and C(5) ketogenesis share the same pool of acetyl-CoA. Although the uptake of octanoate and heptanoate by the liver are similar, the rate of C(5) ketogenesis from heptanoate is much lower than the rate of C(4) ketogenesis from octanoate. This results from the channeling of the propionyl moiety of heptanoate into anaplerosis of the citric acid cycle. C(5) ketogenesis from propionate is virtually nil because acetoacyl-CoA thiolase does not favor the formation of beta-ketopentanoyl-CoA from propionyl-CoA and acetyl-CoA. Anaplerosis and gluconeogenesis from heptanoate are inhibited by octanoate. The data have implications for the design of diets for the treatment of long chain fatty acid oxidation disorders, such as the triheptanoin-based diet.


Asunto(s)
Ácido 3-Hidroxibutírico/metabolismo , Acetoacetatos/metabolismo , Cuerpos Cetónicos/biosíntesis , Hígado/metabolismo , Ácidos Pentanoicos/metabolismo , Ácido 3-Hidroxibutírico/química , Acetoacetatos/química , Animales , Caprilatos/química , Caprilatos/metabolismo , Ácidos Grasos/química , Ácidos Grasos/metabolismo , Cromatografía de Gases y Espectrometría de Masas , Glucosa/metabolismo , Heptanoatos/química , Heptanoatos/metabolismo , Cuerpos Cetónicos/química , Metabolismo de los Lípidos , Masculino , Oxidación-Reducción , Ácidos Pentanoicos/química , Propionatos/química , Propionatos/metabolismo , Ratas , Ratas Sprague-Dawley
19.
Biomed Chromatogr ; 22(5): 450-3, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18254151

RESUMEN

Simultaneous determination of lactic acid, pyruvic acid, 3-hydroxybutyric acid and acetoacetic acid for clinical monitoring of lactic acidosis and ketone body formation in human plasma (20 microL) was performed by gas chromatography-mass spectrometry in selected ion monitoring (SIM) mode after generating methoxime/tert-butyldimethylsilyl derivatives. All of the targeted carboxylic acids were detected by characteristic fragment ions, which permitted sensitive and selective identification in the presence of co-extracted free fatty acids and other acidic metabolites at much higher levels. The method was linear (r>or=0.9991), reproducible (% relative standard deviation=1.2-5.8), and accurate (% relative error=-7.2-7.6), with detection limits of 0.05-1.7 ng/mL. This rapid, accurate and selective method using minimal plasma samples (20 microL) is useful in the clinical monitoring of lactic acidosis and ketone body formation in plasma.


Asunto(s)
Cromatografía de Gases y Espectrometría de Masas/métodos , Cuerpos Cetónicos/sangre , Ácido Láctico/sangre , Compuestos de Organosilicio/química , Oximas/química , Ácido Pirúvico/sangre , Humanos , Cuerpos Cetónicos/química , Ácido Láctico/química , Masculino , Ácido Pirúvico/química , Reproducibilidad de los Resultados
20.
CNS Drug Rev ; 11(2): 113-40, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16007235

RESUMEN

KTX 0101 is the sodium salt of the physiological ketone, D-beta-hydroxybutyrate (betaOHB). This neuroprotectant, which has recently successfully completed clinical Phase IA evaluation, is being developed as an intravenous infusion fluid to prevent the cognitive deficits caused by ischemic foci in the brain during cardiopulmonary bypass (CPB) surgery. KTX 0101 maintains cellular viability under conditions of physiological stress by acting as a "superfuel" for efficient ATP production in the brain and peripheral tissues. Unlike glucose, this ketone does not require phosphorylation before entering the TCA cycle, thereby sparing vital ATP stores. Although no reliable models of CPB-induced ischemia exist, KTX 0101 is powerfully cytoprotectant under the more severe ischemic conditions of global and focal cerebral ischemia, cardiac ischemia and lung hemorrhage. Neuroprotection has been demonstrated by reductions in infarct volume, edema, markers of apoptosis and functional impairment. One significant difference between KTX 0101 and other potential neuroprotectants in development is that betaOHB is a component of human metabolic physiology which exploits the body's own neuroprotective mechanisms. KTX 0101 also protects hippocampal organotypic cultures against early and delayed cell death in an in vitro model of status epilepticus, indicating that acute KTX 0101 intervention in this condition could help prevent the development of epileptiform foci, a key mechanism in the etiology of intractable epilepsy. In models of chronic neurodegenerative disorders, KTX 0101 protects neurons against damage caused by dopaminergic neurotoxins and by the fragment of beta-amyloid, Abeta(1-42), implying possible therapeutic applications for ketogenic strategies in treating Parkinson's and Alzheimer's diseases. Major obstacles to the use of KTX 0101 for long term therapy in chronic disorders, e.g., Parkinson's and Alzheimer's diseases, are the sodium loading problem and the need to administer it in relatively large amounts because of its rapid mitochondrial metabolism. These issues are being addressed by designing and synthesizing orally bioavailable multimers of betaOHB with improved pharmacokinetics.


Asunto(s)
Cuerpos Cetónicos/uso terapéutico , Enfermedades del Sistema Nervioso/prevención & control , Fármacos Neuroprotectores/uso terapéutico , Complicaciones Posoperatorias/prevención & control , Ácido 3-Hidroxibutírico/química , Ácido 3-Hidroxibutírico/metabolismo , Ácido 3-Hidroxibutírico/farmacología , Ácido 3-Hidroxibutírico/uso terapéutico , Adenosina Trifosfato/metabolismo , Animales , Infarto Encefálico/etiología , Infarto Encefálico/prevención & control , Muerte Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación de Medicamentos/métodos , Humanos , Hidroxibutiratos , Técnicas In Vitro , Isquemia/etiología , Isquemia/prevención & control , Cuerpos Cetónicos/química , Cuerpos Cetónicos/metabolismo , Cuerpos Cetónicos/farmacología , Modelos Biológicos , Enfermedades del Sistema Nervioso/clasificación , Estado Epiléptico/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA