Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros

Métodos Terapéuticos y Terapias MTCI
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Genet Metab ; 131(3): 316-324, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33127324

RESUMEN

Propionic acidemia (PA) is caused by inherited deficiency of mitochondrial propionyl-CoA carboxylase (PCC) and results in significant neurodevelopmental and cardiac morbidity. However, relationships among therapeutic intervention, biochemical markers, and disease progression are poorly understood. Sixteen individuals homozygous for PCCB c.1606A > G (p.Asn536Asp) variant PA participated in a two-week suspension of therapy. Standard metabolic markers (plasma amino acids, blood spot methylcitrate, plasma/urine acylcarnitines, urine organic acids) were obtained before and after stopping treatment. These same markers were obtained in sixteen unaffected siblings. Echocardiography and electrocardiography were obtained from all subjects. We characterized the baseline biochemical phenotype of untreated PCCB c.1606A > G homozygotes and impact of treatment on PCC deficiency biomarkers. Therapeutic regimens varied widely. Suspension of therapy did not significantly alter branched chain amino acid levels, their alpha-ketoacid derivatives, or urine ketones. Carnitine supplementation significantly increased urine propionylcarnitine and its ratio to total carnitine. Methylcitrate blood spot and urine levels did not correlate with other biochemical measures or cardiac outcomes. Treatment of PCCB c.1606A > G homozygotes with protein restriction, prescription formula, and/or various dietary supplements has a limited effect on core biomarkers of PCC deficiency. These patients require further longitudinal study with standardized approaches to better understand the relationship between biomarkers and disease burden.


Asunto(s)
Ligasas de Carbono-Carbono/genética , Corazón/fisiopatología , Trastornos del Neurodesarrollo/genética , Acidemia Propiónica/genética , Ácidos/sangre , Ácidos/orina , Adolescente , Adulto , Aminoácidos/sangre , Aminoácidos/orina , Biomarcadores/sangre , Biomarcadores/orina , Ligasas de Carbono-Carbono/sangre , Ligasas de Carbono-Carbono/orina , Carnitina/sangre , Carnitina/orina , Niño , Preescolar , Ecocardiografía , Femenino , Corazón/diagnóstico por imagen , Humanos , Masculino , Mitocondrias/genética , Mitocondrias/metabolismo , Mutación/genética , Trastornos del Neurodesarrollo/sangre , Trastornos del Neurodesarrollo/diagnóstico por imagen , Trastornos del Neurodesarrollo/orina , Compuestos Orgánicos/sangre , Compuestos Orgánicos/orina , Fenotipo , Acidemia Propiónica/sangre , Acidemia Propiónica/diagnóstico por imagen , Acidemia Propiónica/orina , Adulto Joven
2.
Nutrients ; 12(8)2020 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-32727157

RESUMEN

Fortification of human milk (HM) for preterm and very low-birth weight (VLBW) infants is a standard practice in most neonatal intensive care units. The optimal fortification strategy and the most suitable protein source for achieving better tolerance and growth rates for fortified infants are still being investigated. In a previous clinical trial, preterm and VLBW infants receiving supplementation of HM with experimental donkey milk-based fortifiers (D-HMF) showed decreased signs of feeding intolerance, including feeding interruptions, bilious gastric residuals and vomiting, with respect to infants receiving bovine milk-based fortifiers (B-HMF). In the present ancillary study, the urinary metabolome of infants fed B-HMF (n = 27) and D-HMF (n = 27) for 21 days was analyzed by 1H NMR spectroscopy at the beginning (T0) and at the end (T1) of the observation period. Results showed that most temporal changes in the metabolic responses were common in the two groups, providing indications of postnatal adaptation. The significantly higher excretion of galactose in D-HMF and of carnitine, choline, lysine and leucine in B-HMF at T1 were likely due to different formulations. In conclusion, isocaloric and isoproteic HM fortification may result in different metabolic patterns, as a consequence of the different quality of the nutrients provided by the fortifiers.


Asunto(s)
Nutrición Enteral/métodos , Alimentos Fortificados , Recien Nacido Prematuro/orina , Leche Humana/metabolismo , Estado Nutricional , Animales , Carnitina/orina , Bovinos , Colina/orina , Equidae , Femenino , Galactosa/orina , Humanos , Recién Nacido , Leucina/orina , Lisina/orina , Masculino , Metaboloma , Leche Humana/química
3.
Nutr Res ; 78: 72-81, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32544852

RESUMEN

Diets including red meat and other animal-sourced foods may increase proteolytic fermentation and microbial-generated trimethylamine (TMA) and, subsequently, trimethylamine-N-oxide (TMAO), a metabolite associated with increased risk of cardiovascular disease and dementia. It was hypothesized that compared to usual dietary intake, a maintenance-energy high-protein diet (HPD) would increase products of proteolytic fermentation, whereas adjunctive prebiotic, probiotic, and synbiotic supplementation may mitigate these effects. An exploratory aim was to determine the association of the relative abundance of the TMA-generating taxon, Emergencia timonensis, with serum and urinary TMAO. At 5 time points (usual dietary intake, HPD diet, HPD + prebiotic, HPD + probiotic, and HPD + synbiotic), urinary (24-hour) and serum metabolites and fecal microbiota profile of healthy older women (n = 20) were measured by liquid chromatography-tandem mass spectrometry and 16S rRNA gene amplicon sequencing analyses, respectively. The HPD induced increases in serum levels of l-carnitine, indoxyl sulfate, and phenylacetylglutamine but not TMAO or p-cresyl sulfate. Urinary excretion of l-carnitine, indoxyl sulfate, phenylacetylglutamine, and TMA increased with the HPD but not TMAO or p-cresyl sulfate. Most participants had undetectable levels of E.timonensis at baseline and only 50% during the HPD interventions, suggesting other taxa are responsible for the microbial generation of TMA in these individuals. An HPD diet with or without a prebiotic, probiotic, or synbiotic elicited an increase in products of proteolytic fermentation. The urinary l-carnitine response suggests that the additional dietary l-carnitine provided was primarily bioavailable, providing little substrate for microbial conversion to TMA and subsequent TMAO formation.


Asunto(s)
Dieta Rica en Proteínas , Carne , Metilaminas/sangre , Metilaminas/orina , Anciano , Carnitina/sangre , Carnitina/orina , Clostridiales/aislamiento & purificación , Cresoles/sangre , Cresoles/orina , Estudios Cruzados , Heces/microbiología , Femenino , Microbioma Gastrointestinal , Glutamina/análogos & derivados , Glutamina/orina , Humanos , Indicán/sangre , Indicán/orina , Prebióticos , Probióticos , Ésteres del Ácido Sulfúrico/sangre , Ésteres del Ácido Sulfúrico/orina , Simbióticos
4.
PLoS One ; 15(3): e0229772, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32126131

RESUMEN

BACKGROUND: Fatigue is a common adverse event during lenvatinib treatment in patients with hepatocellular carcinoma. One mechanism contributing to development of fatigue might involve abnormal adenosine triphosphate synthesis that is caused by carnitine deficiency. To address this possibility, we examined the relationship between carnitine levels and fatigue during lenvatinib treatment. METHODS: This prospective study evaluated 20 patients with hepatocellular carcinoma who underwent lenvatinib treatment. Both blood and urine samples were collected from the patients before starting lenvatinib therapy (day 0), and on days 3, 7, 14, and 28 thereafter. Plasma and urine concentrations of free and acyl carnitine (AC) were assessed at each time point. The changes in daily fatigue were evaluated using the Brief Fatigue Inventory (BFI). RESULTS: Plasma levels of free carnitine (FC) at days 3 and 7 were significantly higher compared with baseline (p = 0.005, p = 0.005, respectively). The urine FC level at day 3 was significantly higher compared with baseline (p = 0.030) and that of day 7 tended to be higher compared with baseline (p = 0.057). The plasma AC concentration at days 14 and 28 was significantly higher compared with that of baseline (p = 0.002, p = 0.005, respectively). The plasma AC-to-FC (AC/FC) ratio on days 14 and 28 was significantly higher compared with baseline (p = 0.001, p = 0.003, respectively). There were significant correlations between the plasma AC/FC ratio and the change in the BFI score at days 14 and 28 (r = 0.461, p = 0.041; r = 0.770, p = 0.002, respectively). CONCLUSIONS: Longitudinal assessments of carnitine and fatigue in patients with hepatocellular carcinoma suggest that lenvatinib affects the carnitine system in patients undergoing lenvatinib therapy and that carnitine insufficiency increases fatigue. The occurrence of carnitine insufficiency may be a common cause of fatigue during the treatment.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Cardiomiopatías/inducido químicamente , Carnitina/deficiencia , Fatiga/etiología , Hiperamonemia/inducido químicamente , Neoplasias Hepáticas/tratamiento farmacológico , Enfermedades Musculares/inducido químicamente , Compuestos de Fenilurea/efectos adversos , Quinolinas/efectos adversos , Anciano , Anciano de 80 o más Años , Carcinoma Hepatocelular/sangre , Carcinoma Hepatocelular/orina , Cardiomiopatías/sangre , Cardiomiopatías/complicaciones , Cardiomiopatías/dietoterapia , Carnitina/administración & dosificación , Carnitina/sangre , Carnitina/orina , Suplementos Dietéticos , Fatiga/sangre , Fatiga/diagnóstico , Fatiga/prevención & control , Femenino , Humanos , Hiperamonemia/sangre , Hiperamonemia/complicaciones , Hiperamonemia/dietoterapia , Neoplasias Hepáticas/sangre , Neoplasias Hepáticas/orina , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Enfermedades Musculares/sangre , Enfermedades Musculares/complicaciones , Enfermedades Musculares/dietoterapia , Estudios Prospectivos , Resultado del Tratamiento
5.
J Pediatr Hematol Oncol ; 42(5): e321-e327, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32032238

RESUMEN

Carnitine is an essential cofactor for mitochondrial import and oxidation of fatty acids. High-dose chemotherapy and radiation, often required for hematopoietic stem cell transplant (HSCT), leads to tissue damage, mitochondrial dysfunction, and alterations in carnitine metabolism. The aim of this pilot cohort study was to describe plasma and urinary carnitine profiles during pediatric HSCT and their relationships with clinical outcomes. Plasma and urinary carnitine samples were collected from 22 pediatric patients before and through day 180 post-HSCT. Associations were observed between graft-versus-host disease and an elevated plasma total carnitine (P=0.019), and also increased plasma acyl:free carnitine ratio with veno-occlusive disease (P=0.016). Mortality was observed in those with their highest urinary total carnitine losses on day 0 (P=0.005), and in those with an abnormal day 28 plasma ratio either above or below the reference range (P=0.007). Changes in carnitine profiles were more reflective of metabolic stress and negative outcomes than of inadequate dietary intake. Associations observed direct larger studies to assess the validity of carnitine profiles as a prognostic indicator and also to assess whether prophylactic carnitine supplementation pre-HSCT could reduce mitochondrial injury and urinary losses and help mitigate inflammatory and metabolic comorbidities of HSCT.


Asunto(s)
Biomarcadores/análisis , Carnitina/sangre , Carnitina/orina , Enfermedad Injerto contra Huésped/diagnóstico , Neoplasias Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Enfermedades Vasculares/diagnóstico , Adolescente , Niño , Preescolar , Estudios de Cohortes , Femenino , Estudios de Seguimiento , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/metabolismo , Neoplasias Hematológicas/patología , Humanos , Lactante , Masculino , Proyectos Piloto , Pronóstico , Enfermedades Vasculares/etiología , Enfermedades Vasculares/metabolismo
6.
BMC Pediatr ; 19(1): 344, 2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31597564

RESUMEN

BACKGROUND: Mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase (mHS) deficiency is an autosomal recessive inborn error of metabolism, which will give rise to failure of ketogenesis in liver during illness or fasting. It is a very rare disease with only a few patients reported worldwide, most of which had a good prognosis after proper therapies. CASE PRESENTATION: We report a 9-month-old boy with mHS deficiency presenting with unusually severe and persistent acidosis after diarrhea and reduced oral food intake. The metabolic acidosis persisted even after supplementation with sugar and alkaline solution. Blood purification and assisted respiration alleviated symptoms, but a second onset induced by respiratory infection several days later led to multiple organ failure and death. Urine organic acid analysis during the acute episode revealed a complex pattern of ketogenic dicarboxylic and 3-hydroxydicarboxylic aciduria with prominent elevation of glutaric acid and adipic acid, which seem to be specific to mHS deficiency. Plasma acylcarnitine analysis revealed elevated 3-hydroxybutyrylcarnitine and acetylcarnitine. This is the first report of elevated 3-hydroxybutyrylcarnitine in mHS deficiency. Whole exome sequencing revealed a novel compound heterozygous mutation in HMGCS2 (c.100C > T and c.1465delA). CONCLUSION: This severe case suggests the need for patients with mHS deficiency to avoid recurrent illness because it can induce severe metabolic crisis, possibly leading to death. Such patients may also require special treatment, such as blood purification. Urine organic acid profile during the acute episode may give a hint to the disease.


Asunto(s)
Acidosis/genética , Acilcoenzima A/deficiencia , Hidroximetilglutaril-CoA Sintasa/genética , Mitocondrias/enzimología , Mutación/genética , Acidosis/terapia , Acidosis/orina , Adipatos/orina , Carnitina/análogos & derivados , Carnitina/sangre , Carnitina/orina , Diarrea/complicaciones , Ácidos Dicarboxílicos/orina , Resultado Fatal , Mutación del Sistema de Lectura/genética , Glutaratos/orina , Humanos , Lactante , Masculino , Insuficiencia Multiorgánica/complicaciones , Infecciones del Sistema Respiratorio/complicaciones , Secuenciación del Exoma
7.
Eur J Nutr ; 55(1): 207-17, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25612929

RESUMEN

PURPOSE: More than 95% of the body carnitine is located in skeletal muscle, where it is essential for energy metabolism. Vegetarians ingest less carnitine and carnitine precursors and have lower plasma carnitine concentrations than omnivores. Principle aims of the current study were to assess the plasma and skeletal muscle carnitine content and physical performance of male vegetarians and matched omnivores under basal conditions and after L-carnitine supplementation. RESULTS: Sixteen vegetarians and eight omnivores participated in this interventional study with oral supplementation of 2 g L-carnitine for 12 weeks. Before carnitine supplementation, vegetarians had a 10% lower plasma carnitine concentration, but maintained skeletal muscle carnitine stores compared to omnivores. Skeletal muscle phosphocreatine, ATP, glycogen and lactate contents were also not different from omnivores. Maximal oxygen uptake (VO2max) and workload (P max) per bodyweight (bicycle spiroergometry) were not significantly different between vegetarians and omnivores. Sub-maximal exercise (75% VO2max for 1 h) revealed no significant differences between vegetarians and omnivores (respiratory exchange ratio, blood lactate and muscle metabolites). Supplementation with L-carnitine significantly increased the total plasma carnitine concentration (24% in omnivores, 31% in vegetarians) and the muscle carnitine content in vegetarians (13%). Despite this increase, P max and VO2max as well as muscle phosphocreatine, lactate and glycogen were not significantly affected by carnitine administration. CONCLUSIONS: Vegetarians have lower plasma carnitine concentrations, but maintained muscle carnitine stores compared to omnivores. Oral L-carnitine supplementation normalizes the plasma carnitine stores and slightly increases the skeletal muscle carnitine content in vegetarians, but without affecting muscle function and energy metabolism.


Asunto(s)
Carnitina/administración & dosificación , Suplementos Dietéticos , Metabolismo Energético/efectos de los fármacos , Ejercicio Físico/fisiología , Músculo Esquelético/efectos de los fármacos , Administración Oral , Adolescente , Adulto , Índice de Masa Corporal , Peso Corporal , Carnitina/sangre , Carnitina/orina , Carbohidratos de la Dieta/administración & dosificación , Grasas de la Dieta/administración & dosificación , Proteínas en la Dieta/administración & dosificación , Ingestión de Energía , Glucógeno/metabolismo , Humanos , Masculino , Músculo Esquelético/metabolismo , Vegetarianos , Adulto Joven
8.
Nutr. hosp ; 32(6): 2613-2617, dic. 2015. tab, graf
Artículo en Español | IBECS | ID: ibc-146124

RESUMEN

Introducción: la cistinosis nefropática infantil (CNI) es una enfermedad genética debida a un defecto del transporte de la cistina, con la subsecuente acumulación de este aminoácido predominantemente en el riñón. Existen pocos estudios sobre la evaluación del estado nutricional en pacientes con esta patología, pero se sabe que tienen una excreción de carnitina urinaria aumentada, lo que puede dar como resultado una deficiencia plasmática y muscular de este compuesto; sin embargo, la suplementación de carnitina en CNI es controversial. Objetivo: comparar la concentración sanguínea de carnitina libre (C0) con el estado nutricional de una cohorte de pacientes con CNI. Material y métodos: evaluación antropométrica mediante la medición de peso, talla, perímetro braquial (PB) y pliegue cutáneo tricipital (PCT). La C0 se cuantificó mediante espectrometría de masas en tándem en muestras de sangre en ayuno. Resultados: se analizaron 10 pacientes con CNI, 5 con y 5 sin trasplante renal. De acuerdo con el IMC, 3/10 presentaron desnutrición. La reserva de masa magra se encontró baja en 8/10 pacientes (3 no trasplantados y todos los trasplantados). El PB mostró correlación con las concentraciones sanguíneas de C0 (r2=0,353); Los pacientes no trasplantados tuvieron niveles de C0 significativamente más bajos que los trasplantados (Chi2=0,0027). Conclusión: en esta población de pacientes con CNI se encontró un 70% de sujetos con C0 baja, que se correlaciona con la masa magra disminuida. Es recomendable hacer una evaluación nutricional de rutina que incluya los tres parámetros antropométricos como parte del seguimiento médico-nutricional integral de estos pacientes (AU)


Introduction: infantile nephropathic cystinosis (INC) is an autosomal recessive disorder that causes defects in cystine transport with subsequent accumulation in almost all body tissues, especially kidneys. There are few studies regarding the nutritional status assessment of patients with INC. It has been reported that patients with INC showed increased urinary losses of carnitine, resulting in plasma and muscle carnitine deficiency also increased metabolic requirements of carnitine in this patients have also been proposed, but to date carnitine supplementation is controversial. Objective: the aim of this study was to compare carnitine blood concentrations with nutritional status assessed by three anthropometric parameters: body mass index, mid-upper arm circumference and tricipital skin fold in patients with INC. Material and methods: anthropometric assessment of 10 patients with INC which included measurement of weight, height, mid-upper arm circumference and tricipital skin fold thickness. Free carnitine (C0) was measured by tandem mass spectrometry in fasting blood samples. Results: a total of 10 patients with INC were analyzed, 5 with and 5 without renal graft. According to the body mass index, 3/10 presented malnutrition. Muscular mass was found low in 8/10 patients (3 without renal graft and all the transplanted) the mid-upper arm circumference showed correlation with C0 blood concentrations (r2=0.353); non transplanted patients had C0 levels significantly lower than the transplanted ones (Chi2=0.0027). Conclusion: in this study we found that 70% of patients had low C0 blood levels that had a correlation with depleted lean body mass. It is recommendable to evaluate the nutritional status of these patients as part of their routine medical evaluation (AU)


Asunto(s)
Humanos , Cistinosis/fisiopatología , Carnitina/orina , Estado Nutricional/fisiología , Síndrome de Fanconi/fisiopatología , Antropometría/métodos , Pesos y Medidas Corporales/estadística & datos numéricos , Biomarcadores/análisis , Trasplante de Riñón/estadística & datos numéricos
9.
Appl Physiol Nutr Metab ; 40(3): 280-91, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25723909

RESUMEN

The aim of this study was to estimate the effect of carnitine supplementation on lipid disorders and peripheral tissue insulin sensitivity in a non-obese animal model of insulin resistance, the hereditary hypertriglyceridemic (HHTg) rat. Male HHTg rats were fed a standard diet, and half of them received daily doses of carnitine (500 mg·kg(-1) body weight) for 8 weeks. Rats of the original Wistar strain were used for comparison. HHTg rats exhibited increased urinary excretion of free carnitine and reduced carnitine content in the liver and blood. Carnitine supplementation compensated for this shortage and promoted urinary excretion of acetylcarnitine without any signs of (acyl)carnitine accumulation in skeletal muscle. Compared with their untreated littermates, carnitine-treated HHTg rats exhibited lower weight gain, reduced liver steatosis, lower fasting triglyceridemia, and greater reduction of serum free fatty acid content after glucose load. Carnitine treatment was associated with increased mitochondrial biogenesis and oxidative capacity for fatty acids, amelioration of oxidative stress, and restored substrate switching in the liver. In skeletal muscle (diaphragm), carnitine supplementation was associated with significantly higher palmitate oxidation and a more favorable complete to incomplete oxidation products ratio. Carnitine supplementation further enhanced insulin sensitivity ex vivo. No effects on whole-body glucose tolerance were observed. Our data suggest that some metabolic syndrome-related disorders, particularly fatty acid oxidation, steatosis, and oxidative stress in the liver, could be attenuated by carnitine supplementation. The effect of carnitine could be explained, at least partly, by enhanced substrate oxidation and increased fatty acid transport from tissues in the form of short-chain acylcarnitines.


Asunto(s)
Carnitina/farmacología , Hipertrigliceridemia/genética , Metabolismo de los Lípidos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Animales , Carnitina/administración & dosificación , Carnitina/análogos & derivados , Carnitina/sangre , Carnitina/metabolismo , Carnitina/orina , ADN Mitocondrial/genética , Suplementos Dietéticos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Predisposición Genética a la Enfermedad , Homeostasis , Hipertrigliceridemia/metabolismo , Resistencia a la Insulina , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/metabolismo , Masculino , Músculo Esquelético/metabolismo , Ratas
10.
Life Sci ; 117(2): 84-92, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25301199

RESUMEN

AIMS: Trimethylamine-N-oxide (TMAO) is produced in host liver from trimethylamine (TMA). TMAO and TMA share common dietary quaternary amine precursors, carnitine and choline, which are metabolized by the intestinal microbiota. TMAO recently has been linked to the pathogenesis of atherosclerosis and severity of cardiovascular diseases. We examined the effects of anti-atherosclerotic compound meldonium, an aza-analogue of carnitine bioprecursor gamma-butyrobetaine (GBB), on the availability of TMA and TMAO. MAIN METHODS: Wistar rats received L-carnitine, GBB or choline alone or in combination with meldonium. Plasma, urine and rat small intestine perfusate samples were assayed for L-carnitine, GBB, choline and TMAO using UPLC-MS/MS. Meldonium effects on TMA production by intestinal bacteria from L-carnitine and choline were tested. KEY FINDINGS: Treatment with meldonium significantly decreased intestinal microbiota-dependent production of TMA/TMAO from L-carnitine, but not from choline. 24hours after the administration of meldonium, the urinary excretion of TMAO was 3.6 times lower in the combination group than in the L-carnitine-alone group. In addition, the administration of meldonium together with L-carnitine significantly increased GBB concentration in blood plasma and in isolated rat small intestine perfusate. Meldonium did not influence bacterial growth and bacterial uptake of L-carnitine, but TMA production by the intestinal microbiota bacteria K. pneumoniae was significantly decreased. SIGNIFICANCE: We have shown for the first time that TMA/TMAO production from quaternary amines could be decreased by targeting bacterial TMA-production. In addition, the production of pro-atherogenic TMAO can be suppressed by shifting the microbial degradation pattern of supplemental/dietary quaternary amines.


Asunto(s)
Vías Biosintéticas/efectos de los fármacos , Carnitina/metabolismo , Tracto Gastrointestinal/microbiología , Metilaminas/metabolismo , Metilhidrazinas/farmacología , Microbiota/fisiología , Animales , Betaína/administración & dosificación , Betaína/análogos & derivados , Betaína/sangre , Vías Biosintéticas/fisiología , Isótopos de Carbono/metabolismo , Carnitina/administración & dosificación , Carnitina/sangre , Carnitina/orina , Colina/metabolismo , Cromatografía Líquida de Alta Presión , Metilaminas/orina , Ratas , Ratas Wistar , Estadísticas no Paramétricas , Espectrometría de Masas en Tándem
11.
J Nutr ; 144(12): 1977-84, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25122647

RESUMEN

BACKGROUND: Biotin functions as a cofactor for several carboxylase enzymes with key roles in metabolism. At present, the dietary requirement for biotin is unknown and intake recommendations are provided as Adequate Intakes (AIs). The biotin AI for adults and pregnant women is 30 µg/d, whereas 35 µg/d is recommended for lactating women. However, pregnant and lactating women may require more biotin to meet the demands of these reproductive states. OBJECTIVE: The current study sought to quantify the impact of reproductive state on biotin status response to a known dietary intake of biotin. METHODS: To achieve this aim, we measured a panel of biotin biomarkers among pregnant (gestational week 27 at study entry; n = 26), lactating (postnatal week 5 at study entry; n = 28), and control (n = 21) women who participated in a 10- to 12-wk feeding study providing 57 µg of dietary biotin/d as part of a mixed diet. RESULTS: Over the course of the study, pregnant women excreted 69% more (vs. control; P < 0.001) 3-hydroxyisovaleric acid (3-HIA), a metabolite that accumulates during the catabolism of leucine when the activity of biotin-dependent methylcrotonyl-coenzyme A carboxylase is impaired. Interestingly, urinary excretion of 3-hydroxyisovaleryl-carnitine (3-HIA-carnitine), a downstream metabolite of 3-HIA, was 27% lower (P = 0.05) among pregnant (vs. control) women, a finding that may arise from carnitine inadequacy during gestation. No differences (P > 0.05) were detected in plasma biotin, urinary biotin, or urinary bisnorbiotin between pregnant and control women. Lactating women excreted 76% more (vs. control; P = 0.001) of the biotin catabolite bisnorbiotin, indicating that lactation accelerates biotin turnover and loss. Notably, with respect to control women, lactating women excreted 23% less (P = 0.04) urinary 3-HIA and 26% less (P = 0.05) urinary 3-HIA-carnitine, suggesting that lactation reduces leucine catabolism and that these metabolites may not be useful indicators of biotin status during lactation. CONCLUSIONS: Overall, these data demonstrate significant alterations in markers of biotin metabolism during pregnancy and lactation and suggest that biotin intakes exceeding current recommendations are needed to meet the demands of these reproductive states. This trial was registered at clinicaltrials.gov as NCT01127022.


Asunto(s)
Biotina/análogos & derivados , Biotina/metabolismo , Dieta , Lactancia/sangre , Embarazo , Adulto , Biomarcadores/sangre , Biotina/sangre , Biotina/orina , Ligasas de Carbono-Carbono/metabolismo , Carnitina/análogos & derivados , Carnitina/orina , Colina/administración & dosificación , Cromatografía Liquida , Suplementos Dietéticos , Femenino , Humanos , Leucina/metabolismo , Leche Humana/química , New York , Cooperación del Paciente , Espectrometría de Masas en Tándem , Valeratos/orina , Adulto Joven
12.
Vet Q ; 34(2): 85-91, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24893614

RESUMEN

BACKGROUND: Insulin resistance in horses is an emerging field of interest as it is thought to be a contributing factor in the pathogenesis of many equine conditions. OBJECTIVES: The objectives of the present study were to determine the effects of long-term oral administration of L-carnitine on insulin sensitivity, glucose disposal, plasma leptin concentrations and acylcarnitine spectrum both in plasma and urine. ANIMALS AND METHODS: Six 3-year-old healthy warmblood geldings were used. In a double blind 2 × 2 Latin square design at a dosage of 100 mg/kg body weight (BW)/day for 28 days the effects of oral supplementation of L-carnitine (as fumarate) were assessed. Glucose disposal and insulin sensitivity were measured by means of the euglycemic-hyperinsulinemic clamp technique. Radioimmunoassays were used to determine plasma leptin and insulin concentrations. Electrospray tandem mass spectrometry was used to assess acylcarnitines both in plasma and urine. Statistical analysis was performed using a linear mixed-effects model and P values <0.05 were considered significant. RESULTS: Long-term L-carnitine administration did not affect insulin sensitivity. Plasma leptin and free carnitine concentrations in plasma and urine increased significantly (P = 0.047 and 0.000, respectively) following L-carnitine administration as well as short-chain acylcarnitines in plasma and urinary excretion of short- and medium-chain acylcarnitines. CONCLUSION AND CLINICAL RELEVANCE: Given the effects of oral administration of L-carnitine further clinical study is necessary in order to assess the potential beneficial effects in equine patients suffering from metabolic myopathies such as acquired multiple acyl-CoA dehydrogenase deficiency. IMPACT FOR HUMAN MEDICINE: The current study supports the treatment rationale of short-chain acyl-CoA dehydrogenase deficiency in humans with L-carnitine at an oral dosage of 100 mg/kg BW/day.


Asunto(s)
Carnitina/análogos & derivados , Carnitina/administración & dosificación , Resistencia a la Insulina , Leptina/sangre , Complejo Vitamínico B/administración & dosificación , Animales , Carnitina/sangre , Carnitina/orina , Técnica de Clampeo de la Glucosa/veterinaria , Caballos/sangre , Caballos/orina , Masculino , Plasma , Radioinmunoensayo/veterinaria
13.
Eur J Nutr ; 53(6): 1313-25, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24337254

RESUMEN

PURPOSE: Pharmacokinetics and effects on skeletal muscle and physical performance of oral acetylcarnitine and propionylcarnitine are not well characterized. We therefore investigated the influence of oral acetylcarnitine, propionylcarnitine, and carnitine on body carnitine homeostasis, energy metabolism, and physical performance in mice and compared the findings to non-supplemented control animals. METHODS: Mice were supplemented orally with 2 mmol/kg/day carnitine, acetylcarnitine, or propionylcarnitine for 4 weeks and studied either at rest or after exhaustive exercise. RESULTS: In the supplemented groups, total plasma and urine carnitine concentrations were significantly higher than in the control group receiving no carnitine, whereas the skeletal muscle carnitine content remained unchanged. The supplemented acylcarnitines were hydrolyzed in intestine and liver and reached the systemic circulation as carnitine. Bioavailability of carnitine and acylcarnitines, determined as the urinary excretion of total carnitine, was in the range of 19 %. Skeletal muscle morphology, including fiber-type composition, was not affected, and oxygen consumption by soleus or gastrocnemius fibers was not different between the groups. Supplementation with carnitine or acylcarnitines had no significant impact on the running capacity, but was associated with lower plasma lactate levels and a higher glycogen content in white skeletal muscle after exhaustive exercise. CONCLUSIONS: Oral supplementation of carnitine, acetylcarnitine, or propionylcarnitine in mice is associated with increased plasma and urine total carnitine concentrations, but does not affect the skeletal muscle carnitine content. Despite better preservation of skeletal muscle glycogen and lower plasma lactate levels, physical performance was not improved by carnitine or acylcarnitine supplementation.


Asunto(s)
Acetilcarnitina/administración & dosificación , Carnitina/análogos & derivados , Suplementos Dietéticos , Músculo Esquelético/efectos de los fármacos , Condicionamiento Físico Animal , Acetilcarnitina/sangre , Acetilcarnitina/farmacocinética , Acetilcarnitina/orina , Administración Oral , Animales , Disponibilidad Biológica , Biomarcadores/sangre , Biomarcadores/orina , Carnitina/administración & dosificación , Carnitina/sangre , Carnitina/farmacocinética , Carnitina/orina , Metabolismo Energético , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Estrés Oxidativo/efectos de los fármacos , Consumo de Oxígeno
14.
Talanta ; 88: 193-200, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22265487

RESUMEN

Urinary organic acids, plasma amino acids and acylcarnitine profile analyses are the main tools used to diagnose inborn errors of metabolisms (IEMs). However, without metabolic decompensation, these parameters are often not helpful. On the other hand, in cases of IEM, acylglycines are consistently raised even when patients appear to be in remission. This study aims to set-up a simple liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS) method for the determination of urine acylglycines, complementary to organic acid and acylcarnitine profiles, for the diagnosis of IEM. In addition, local reference intervals for various acylglycines are established by using this method. Acylglycines were isolated by solid-phase extraction, derivatized with n-butanol, separated by HPLC, and detected by ESI-MS/MS. Acylglycines were quantified with deuterated internal standards. Mean recoveries of acylglycines ranged from 90.2 to 109.3%. Within- and between-run imprecisions for all acylglycines have CVs less than 10%. Linear regression coefficients were greater than 0.99. Reference intervals were established according to CLSI guidelines by analyzing 204 samples from apparently healthy individuals less than 18 years of age. The distributions of AG in the "normal" urine were skewed towards the right. After log transformation, all the results were normally distributed. Partitioning into age group reference intervals was not indicated, according to the Harris and Boyd approach. In this context, a single reference interval for each acylglycine could be used. This method of urine acylglycines analysis is a powerful diagnostic tool, complementary to urine organic acids and plasma acylcarnitine profiling, for detecting certain inborn errors of metabolism.


Asunto(s)
Glicina/análogos & derivados , Glicina/orina , Errores Innatos del Metabolismo/orina , 1-Butanol/química , Acilación , Adolescente , Pueblo Asiatico , Calibración , Carnitina/análogos & derivados , Carnitina/orina , Niño , Preescolar , Cromatografía Líquida de Alta Presión , Deuterio , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Errores Innatos del Metabolismo/diagnóstico , Valores de Referencia , Sensibilidad y Especificidad , Extracción en Fase Sólida , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem
15.
Pediatr Transplant ; 16(3): 230-4, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22151065

RESUMEN

Propionic acidemia is a rare autosomal recessive disorder affecting the catabolism of branched-chain amino acids because of a genetic defect in PCC. Despite the improvements in medical treatment with protein restriction, sufficient caloric intake, supplementation of l-carnitine, and metronidazole, patients with the severe form of propionic acidemia have life-threatening metabolic acidosis, hyperammonemia, and cardiomyopathy, which results in serious neurologic sequelae and sometimes death. This study retrospectively reviewed three children with neonatal-onset propionic acidemia who received LDLT. Between November 2005 and December 2010, 148 children underwent LDLT, with an overall patient survival of 90.5%, in our center. Three patients were indicated for transplantation because of propionic acidemia. All recipients achieved a resolution of metabolic derangement and better quality of life with protein restriction and medication, although urine methylcitrate and serum propionylcarnitine levels did not decrease markedly. LT can reduce the magnitude of progressive cardiac/neurologic disability as a result of poor metabolic control. Further evaluation is therefore required to determine the long-term suitability of this treatment modality.


Asunto(s)
Trasplante de Hígado/métodos , Acidemia Propiónica/terapia , Carnitina/análogos & derivados , Carnitina/orina , Preescolar , Citratos/orina , Femenino , Humanos , Lactante , Donadores Vivos , Complicaciones Posoperatorias/terapia , Calidad de Vida , Estudios Retrospectivos , Resultado del Tratamiento
16.
Cell Mol Neurobiol ; 32(1): 77-82, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21833551

RESUMEN

Propionic (PA) and methylmalonic (MMA) acidurias are inherited disorders caused by deficiency of propionyl-CoA carboxylase and methylmalonyl-CoA mutase, respectively. Affected patients present acute metabolic crises in the neonatal period and long-term neurological deficits. Treatments of these diseases include a protein restricted diet and L: -carnitine supplementation. L: -Carnitine is widely used in the therapy of these diseases to prevent secondary L: -carnitine deficiency and promote detoxification, and several recent in vitro and in vivo studies have reported antioxidant and antiperoxidative effects of this compound. In this study, we evaluated the oxidative stress parameters, isoprostane and di-tyrosine levels, and the antioxidant capacity, in urine from patients with PA and MMA at the diagnosis, and during treatment with L: -carnitine and protein-restricted diet. We verified a significant increase of isoprostanes and di-tyrosine, as well as a significant reduction of the antioxidant capacity in urine from these patients at diagnosis, as compared to controls. Furthermore, treated patients presented a marked reduction of isoprostanes and di-tyrosine levels in relation to untreated patients. In addition, patients with higher levels of protein and lipid oxidative damage, determined by di-tyrosine and isoprostanes levels, also presented lower urinary concentrations of total and free L: -carnitine. In conclusion, the present results indicate that treatment with low protein diet and L: -carnitine significantly reduces urinary biomarkers of protein and lipid oxidative damage in patients with disorders of propionate metabolism and that L: -carnitine supplementation may be specially involved in this protection.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/dietoterapia , Errores Innatos del Metabolismo de los Aminoácidos/orina , Carnitina/uso terapéutico , Estrés Oxidativo/fisiología , Propionatos/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Antioxidantes/análisis , Antioxidantes/metabolismo , Carnitina/administración & dosificación , Carnitina/análisis , Carnitina/orina , Niño , Preescolar , Dieta con Restricción de Proteínas , Suplementos Dietéticos , Humanos , Lactante , Recién Nacido , Análisis por Apareamiento , Ácido Metilmalónico/metabolismo , Ácido Metilmalónico/orina , Estrés Oxidativo/efectos de los fármacos , Propionatos/orina , Resultado del Tratamiento , Tirosina/análisis , Tirosina/orina
17.
J Nutr ; 141(3): 353-8, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21248194

RESUMEN

Mounting evidence indicates that marginal biotin deficiency is not rare, contrary to previous assumptions. Accordingly, robust indicators of biotin status would be useful. In a study of 10 healthy adults, we recently provided evidence that abnormally increased plasma concentration of 3-hydroxyisovaleryl carnitine (3HIA-carnitine) is a sensitive indicator of marginal biotin deficiency. We sought to determine whether urinary excretion of 3HIA-carnitine (expressed as the ratio to urinary creatinine) significantly increases in marginal biotin deficiency. Marginal, asymptomatic biotin deficiency was induced experimentally in the same 10 healthy adults (8 women) by feeding undenatured egg white with meals for 28 d. Biotin status was repleted by a mixed general diet plus biotin supplementation. Urinary excretion of 3HIA-carnitine was determined by liquid chromatography-tandem MS on d 0, 14, and 28 (depletion) and on d 35 and 50 (repletion). Mean urinary 3HIA-carnitine concentration increased with depletion (P < 0.0001; d 0 vs. 28) and decreased with repletion (P = 0.0002; d 28 vs. 50). Urinary 3HIA-carnitine excretion was greater than the upper limit of normal in 9 of 10 participants by d 14 and decreased to within normal limits by d 50 in all participants. This study provides evidence that urinary excretion of 3HIA-carnitine is an early and sensitive indicator of marginal biotin deficiency. The ease of collection of untimed urine samples and application of a new analytical method with simplified sample preparation suggest that urinary 3HIA-carnitine is likely to be a useful indicator for large population studies.


Asunto(s)
Biotina/deficiencia , Carnitina/análogos & derivados , Estado Nutricional , Deficiencia de Vitamina B/diagnóstico , Deficiencia de Vitamina B/orina , Adulto , Biomarcadores/sangre , Biomarcadores/orina , Biotina/uso terapéutico , Carnitina/orina , Clara de Huevo , Femenino , Humanos , Linfocitos/enzimología , Masculino , Metilmalonil-CoA Descarboxilasa/sangre , Valores de Referencia , Factores de Tiempo , Deficiencia de Vitamina B/sangre , Deficiencia de Vitamina B/tratamiento farmacológico
18.
Tohoku J Exp Med ; 221(3): 191-5, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20543534

RESUMEN

Carnitine palmitoyltransferase 2 (CPT2) deficiency is one of the most common mitochondrial beta-oxidation defects. A female patient with an infantile form of CPT2 deficiency first presented as having a Reye-like syndrome with hypoglycemic convulsions. Oral L-carnitine supplementation was administered since serum free carnitine level was very low (less than 10 micromol/L), indicating secondary carnitine deficiency. Her serum and urinary acylcarnitine profiles were analyzed successively to evaluate time-course effects of L-carnitine supplementation. After the first two days of L-carnitine supplementation, the serum level of free carnitine was elevated; however, the serum levels of acylcarnitines and the urinary excretion of both free carnitine and acylcarnitines remained low. A peak of the serum free carnitine level was detected on day 5, followed by a peak of acetylcarnitine on day 7, and peaks of long-chain acylcarnitines, such as C16, C18, C18:1 and C18:2 carnitines, on day 9. Thereafter free carnitine became predominant again. These peaks of the serum levels corresponded to urinary excretion peaks of free carnitine, acetylcarnitine, and medium-chain dicarboxylic carnitines, respectively. It took several days for oral L-carnitine administration to increase the serum carnitine levels, probably because the intracellular stores were depleted. Thereafter, the administration increased the excretion of abnormal acylcarnitines, some of which had accumulated within the tissues. The excretion of medium-chain dicarboxylic carnitines dramatically decreased on day 13, suggesting improvement of tissue acylcarnitine accumulation. These time-course changes in blood and urinary acylcarnitine levels after L-carnitine supplementation support the effectiveness of L-carnitine supplementation to CPT2-deficient patients.


Asunto(s)
Carnitina O-Palmitoiltransferasa/deficiencia , Carnitina/deficiencia , Carnitina/orina , Acetilcarnitina/sangre , Acetilcarnitina/deficiencia , Acetilcarnitina/orina , Errores Innatos del Metabolismo de los Aminoácidos/sangre , Errores Innatos del Metabolismo de los Aminoácidos/orina , Aminoácidos/sangre , Aminoácidos/deficiencia , Aminoácidos/orina , Análisis Químico de la Sangre , Carnitina/análogos & derivados , Carnitina/sangre , Carnitina O-Palmitoiltransferasa/sangre , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Lactante , Errores Innatos del Metabolismo Lipídico/sangre , Errores Innatos del Metabolismo Lipídico/orina , Síndrome de Reye/sangre , Síndrome de Reye/orina , Factores de Tiempo , Resultado del Tratamiento , Complejo Vitamínico B/sangre , Complejo Vitamínico B/orina
19.
Int J Toxicol ; 29(3): 313-7, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20448264

RESUMEN

This investigation was aimed to study the effect of 2,3-dimercapto-1-propanesulfonic acid (DMPS) on mercuric chloride (HgCl(2))-induced alterations in urinary excretion of various carnitine fractions including free carnitine (FC), acylcarnitine (AC), and total carnitine (TC). Different groups of Wistar male rats were treated with HgCl(2) at the doses of 0.1, 0.5, 1.0, 2.0, and 3.0 mg/kg body weight, and the animals were sacrificed at 24 hours following HgCl(2) injection. A separate batch of animals received HgCl(2) (2 mg/kg) with or without DMPS (100 mg/kg) and sacrificed at 24 or 48 hours after dosing. Administration of HgCl(2) resulted in statistically significant and dose-dependent increase in the urinary excretion of FC, AC, and TC in rats. However, the ratio of urinary AC:FC was significantly decreased by HgCl(2). Pretreatment with DMPS offered statistically significant protection against HgCl(2)-induced alterations in various urinary carnitine fractions in rats.


Asunto(s)
Antídotos/farmacología , Carnitina/análogos & derivados , Carnitina/orina , Quelantes/farmacología , Sustancias Peligrosas/toxicidad , Cloruro de Mercurio/toxicidad , Unitiol/farmacología , Algoritmos , Animales , Antídotos/uso terapéutico , Biomarcadores/orina , Quelantes/uso terapéutico , Terapia por Quelación , Relación Dosis-Respuesta a Droga , Enfermedades Renales/inducido químicamente , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/orina , Masculino , Ratas , Ratas Wistar , Factores de Tiempo , Unitiol/uso terapéutico
20.
Nephrol Dial Transplant ; 25(1): 69-76, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19737869

RESUMEN

BACKGROUND: This study examined whether carnitine deficiency is a risk factor and should be viewed as a mechanism during the development of gentamicin (GM)-induced ARF as well as exploring if carnitine supplementation could offer protection against this toxicity. METHODS: Adult male Wistar albino rats were assigned to one of six treatment groups: group 1 (control) rats were given daily intraperitoneal (I.P.) injections of normal saline for 8 consecutive days; groups 2, 3 and 4 rats were given GM (80 mg/kg/day, I.P.), l-carnitine (200 mg/kg/day, I.P.) and d-carnitine (250 mg/kg/day, I.P.), respectively, for 8 consecutive days. Rats of group 5 (GM plus d-carnitine) received a daily I.P. injection of d-carnitine (250 mg/kg/day) 1 h before GM (80 mg/kg/day) for 8 consecutive days. Rats of group 6 (GM plus l-carnitine) received a daily I.P. injection of l-carnitine (200 mg/kg/day) 1 h before GM (80 mg/kg/day) for 8 consecutive days. RESULTS: GM significantly increased serum creatinine, blood urea nitrogen (BUN), urinary carnitine excretion, intramitochondrial acetyl-CoA and total nitrate/nitrite (NOx) and thiobarbituric acid reactive substances (TBARS) in kidney tissues and significantly decreased total carnitine, intramitochondrial CoA-SH, ATP, ATP/ADP and reduced glutathione (GSH) in kidney tissues. In carnitine-depleted rats, GM caused a progressive increase in serum creatinine, BUN and urinary carnitine excretion and a progressive decrease in total carnitine, intamitochondrial CoA-SH and ATP. Interestingly, l-carnitine supplementation resulted in a complete reversal of the increase in serum creatinine, BUN, urinary carnitine excretion and the decrease in total carnitine, intramitochondrial CoA-SH and ATP, induced by GM, to the control values. Moreover, the histopathological examination of kidney tissues confirmed the biochemical data, where l-carnitine prevents and d-carnitine aggravates GM-induced ARF. CONCLUSIONS: (i) GM-induced nephrotoxicity leads to increased urinary losses of carnitine; (ii) carnitine deficiency is a risk factor and should be viewed as a mechanism during the development of GM-induced ARF; and (iii) carnitine supplementation ameliorates the severity of GM-induced kidney dysfunction by increasing the intramitochondrial CoA-SH/acetyl-CoA ratio and ATP production.


Asunto(s)
Lesión Renal Aguda/metabolismo , Carnitina/orina , Coenzima A/metabolismo , Mitocondrias/metabolismo , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/prevención & control , Adenosina Trifosfato/metabolismo , Animales , Nitrógeno de la Urea Sanguínea , Carnitina/farmacología , Creatinina/sangre , Modelos Animales de Enfermedad , Gentamicinas/efectos adversos , Riñón/efectos de los fármacos , Riñón/metabolismo , Masculino , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA