Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Crit Care Explor ; 2(7): e0150, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32766551

RESUMEN

Arginine is a conditionally essential amino acid, the precursor for nitric oxide and a key factor in cell proliferation, protein synthesis, and energy metabolism. When there is increased demand in the setting of inflammation, ischemia-reperfusion injury, and organ dysfunction, endogenous arginine production falls short, and external supplementation may be necessary. The goal of this study was to assess changes in concentrations of plasma arginine, citrulline, ornithine, glutamine, and plasma arginase in infants and children undergoing surgery for congenital heart disease with cardiopulmonary bypass. DESIGN: Prospective observational study. SETTING: The study was conducted in the Heart Center at Texas Children's Hospital. SUBJECTS: Children undergoing surgery for congenital heart disease with cardiopulmonary bypass. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: Serial perioperative blood samples were collected for quantification of amino acids, arginase, nitric oxide metabolites, and markers of organ function (lactate, Pao2/Fio2 ratio, and creatinine clearance). Thirty children (18 males) were included in the study; median (interquartile range) age 0.5 years (0.3-0.9 yr). The mean ± sd for plasma amino acid concentrations before cardiopulmonary bypass: arginine 62 ± 20 µmol/L, citrulline 24 ± 6 µmol/L, ornithine 53 ± 32 µmol/L, and glutamine 591 ± 126 µmol/L. Arginine concentration was decreased within the first 24 hours (43 ± 15 µmol/L; p = 0.004), citrulline and glutamine concentrations decreased over the first 48 hours (11 ± 4 µmol/L; p < 0.001 and 493 ± 131 µmol/L; p = 0.019, respectively) and were associated with an increase in arginase (3.8 ± 3 µg/mL; p < 0.05). There was an increase in Vasoactive-Inotropic Score (5.9 ± 19 vs 0.5 ± 2; p < 0.001), decrease in creatinine clearance (76 ± 24 vs 93 ± 31; p = 0.002), and Pao2/Fio2 ratio (243 ± 138 vs 374 ± 200; p = 0.007) comparing to baseline. CONCLUSIONS: A widely variable degree of arginine, citrulline, and glutamine depletion occurs in children after surgery for congenital heart disease. These findings were associated with increased arginase and coincide with some of the markers of organ perfusion.

2.
Am J Physiol Renal Physiol ; 318(1): F175-F182, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31760772

RESUMEN

Developmental changes in the renal expression and activity of argininosuccinate synthase (ASS1) and argininosuccinate lyase (ASL), enzymes that use citrulline for the production of arginine, have been reported. Thus, the ability of neonates, and especially premature neonates, to produce arginine may be compromised. To determine the utilization of citrulline in vivo, we measured renal expression of ASS1 and ASL and conducted citrulline compartmental and noncompartmental kinetics using [15N]citrulline in pigs of five different ages (from 10 days preterm to 5 wk of age). The tracer was given in substrate amounts to also test the ability of neonatal pigs to use exogenous citrulline. Preterm and term pigs at birth had lower ASS1 and ASL expression than older animals, which was reflected in the longer half-life of citrulline in the neonatal groups. The production and utilization of citrulline by 1-wk-old pigs was greater than in pigs of other ages, including 5-wk-old animals. Plasma citrulline concentration was not able to capture these differences in citrulline production and utilization. In conclusion, the developmental changes in renal ASS1 and ASL gene expression are reflected in the ability of the pigs to use citrulline. However, it seems that there is an excess capacity to use citrulline at all ages, including during prematurity, since the bolus dose of tracer did not result in an increase in endogenous citrulline. Our results support the idea that citrulline supplementation in neonatal, including premature, pigs is a viable option to increase arginine availability.


Asunto(s)
Envejecimiento/metabolismo , Arginina/metabolismo , Citrulina/metabolismo , Animales , Animales Recién Nacidos , Femenino , Riñón/metabolismo , Masculino , Porcinos
3.
Am J Physiol Gastrointest Liver Physiol ; 315(4): G638-G649, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30048597

RESUMEN

Necrotizing enterocolitis (NEC) is associated with low plasma arginine and vascular dysfunction. It is not clear whether low intestinal citrulline production, the precursor for arginine synthesis, occurs before and thus predisposes to NEC or if it results from tissue damage. This study was designed to test the hypothesis that whole body rates of citrulline, arginine, and nitric oxide synthesis are low in premature pigs and that they precede NEC. Piglets delivered by cesarean section at 103 days [preterm (PT)], 110 days [near-term (NT)], or 114 days [full-term (FT)] of gestation were given total parenteral nutrition and after 2 days orogastrically fed infant formula for 42 h to induce NEC. Citrulline and arginine fluxes were determined before and during the feeding protocol. Gross macroscopic and histological NEC scores and plasma fatty acid binding protein (iFABP) concentration were determined as indicators of NEC. Intestinal gene expression for enzymes of the arginine pathway were quantitated. A lower ( P < 0.05) survival rate was observed for PT (8/27) than for NT (9/9) and FT pigs (11/11). PT pigs had higher macroscopic gross ( P < 0.05) and histological NEC ( P < 0.05) scores and iFABP concentration ( P < 0.05) than pigs of more advanced gestational age. PT pigs had lower citrulline production and arginine fluxes ( P < 0.05) throughout and a reduced gene expression in genes of the citrulline-arginine pathway. In summary, intestinal enzyme expression and whole body citrulline and arginine fluxes were reduced in PT pigs compared with animals of more advance gestational age and preceded the development of NEC. NEW & NOTEWORTHY Arginine supplementation prevents necrotizing enterocolitis (NEC), the most common gastrointestinal emergency of prematurity. Citrulline (precursor for arginine) production is reduced during NEC, and this is believed to be a consequence of intestinal damage. In a swine model of NEC, we show that intestinal gene expression of the enzymes for citrulline production and whole body citrulline and arginine fluxes are reduced and precede the onset of NEC in premature pigs. Reduced citrulline production during prematurity may be a predisposition to NEC.


Asunto(s)
Arginina/metabolismo , Citrulina/metabolismo , Enterocolitis Necrotizante/etiología , Desarrollo Fetal , Mucosa Intestinal/metabolismo , Óxido Nítrico/metabolismo , Animales , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/patología , Mucosa Intestinal/crecimiento & desarrollo , Mucosa Intestinal/patología , Porcinos
4.
J Nutr ; 147(4): 596-602, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28179487

RESUMEN

Background: Arginine is considered to be an essential amino acid in various (patho)physiologic conditions of high demand. However, dietary arginine supplementation suffers from various drawbacks, including extensive first-pass extraction. Citrulline supplementation may be a better alternative than arginine, because its only fate in vivo is conversion into arginine.Objective: The goal of the present research was to determine the relative efficiency of arginine and citrulline supplementation to improve arginine availability.Methods: Six-week-old C57BL/6J male mice fitted with gastric catheters were adapted to 1 of 7 experimental diets for 2 wk. The basal diet contained 2.5 g l-arginine/kg, whereas the supplemented diets contained an additional 2.5, 7.5, and 12.5 g/kg diet of either l-arginine or l-citrulline. On the final day, after a 3-h food deprivation, mice were continuously infused intragastrically with an elemental diet similar to the dietary treatment, along with l-[13C6]arginine, to determine the splanchnic first-pass metabolism (FPM) of arginine. In addition, tracers were continuously infused intravenously to determine the fluxes and interconversions between citrulline and arginine. Linear regression slopes were compared to determine the relative efficiency of each supplement.Results: Whereas all the supplemented citrulline (105% ± 7% SEM) appeared in plasma and resulted in a marginal increase of 86% in arginine flux, supplemental arginine underwent an ∼70% FPM, indicating that only 30% of the supplemental arginine entered the peripheral circulation. However, supplemental arginine did not increase arginine flux. Both supplements linearly increased (P < 0.01) plasma arginine concentration from 109 µmol/L for the basal diet to 159 and 214 µmol/L for the highest arginine and citrulline supplementation levels, respectively. However, supplemental citrulline increased arginine concentrations to a greater extent (35%, P < 0.01).Conclusions: Citrulline supplementation is more efficient at increasing arginine availability than is arginine supplementation itself in mice.


Asunto(s)
Arginina/farmacocinética , Citrulina/farmacología , Alimentación Animal/análisis , Animales , Arginasa/genética , Arginasa/metabolismo , Arginina/administración & dosificación , Disponibilidad Biológica , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Citrulina/administración & dosificación , Citrulina/farmacocinética , Dieta/veterinaria , Suplementos Dietéticos , Regulación de la Expresión Génica/efectos de los fármacos , Hígado/enzimología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Curr Opin Clin Nutr Metab Care ; 19(1): 62-6, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26560519

RESUMEN

PURPOSE OF REVIEW: This article analyzes the contribution of glutamine to the synthesis of citrulline and reviews the evidence that glutamine supplementation increases citrulline production. RECENT FINDINGS: Glutamine supplementation has been proposed in the treatment of critically ill patients; however, a recent large multicenter randomized controlled trial resulted in increased mortality in the glutamine-supplemented group. Within this context, defining the contribution of glutamine to the production of citrulline, and thus to de-novo arginine synthesis, has become a pressing issue. SUMMARY: The beneficial effects of glutamine supplementation may be partially mediated by the effects of glutamine on citrulline synthesis by the gut and the de-novo synthesis of arginine by the kidney and other tissues. Although there is no strong evidence to support that glutamine is a major precursor for citrulline synthesis in humans, glutamine has the potential to increase overall gut function and in this way increase citrulline production.


Asunto(s)
Citrulina/biosíntesis , Suplementos Dietéticos , Tracto Gastrointestinal/efectos de los fármacos , Glutamina/farmacología , Animales , Arginina/biosíntesis , Tracto Gastrointestinal/metabolismo , Glutamina/metabolismo , Humanos , Riñón/metabolismo
7.
Mol Genet Metab ; 107(3): 315-21, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23040521

RESUMEN

OBJECTIVE: To compare the effects of combinatorial therapy with low-dose arginine and a nitrogen scavenging agent (sodium phenylbutyrate) vs. monotherapy with high-dose arginine on liver function tests in patients with argininosuccinic aciduria (ASA). STUDY DESIGN: Twelve patients with ASA were enrolled in a double-blind, placebo-controlled, cross-over study design. Subjects were randomized to receive either a low-dose of arginine therapy (100 mg · kg(-1) · d(-1)) combined with sodium phenylbutyrate (500 mg · kg(-1) · d(-1)) (LDA arm) or a high-dose of arginine alone (500 mg · kg(-1) · d(-1)) (HDA arm) for one week. At the end of one week of therapy, liver function tests were assessed and metabolite fluxes were measured using a multi-tracer stable isotope protocol. RESULTS: Plasma aspartate aminotransferase (AST), alanine aminotransferase (ALT), and measures of synthetic functions of the liver were the primary outcomes. Subjects had significantly increased levels of argininosuccinate (P<0.03) and AST levels (P<0.01) after treatment with high-dose arginine. In the subset of subjects with elevated AST or ALT, treatment with high-dose of arginine was associated with further increases in plasma levels of both aminotransferases. Whereas subjects had increased arginine and citrulline flux with high-dose arginine therapy, the glutamine flux was not different between the two treatment arms. The synthetic liver functions as assessed by prothrombin time, INR, and coagulation factor levels were not different between the HDA and LDA arms. CONCLUSIONS: Administering higher doses of arginine in subjects with ASA results in increases in AST and ALT levels, especially in the subset of patients with elevated baseline aminotransferases. Hence, low-dose arginine sufficient to normalize arginine levels in plasma combined with nitrogen scavenging therapy should be considered as a therapeutic option for treatment of ASA in patients with elevations of hepatic aminotransferases.


Asunto(s)
Arginina/uso terapéutico , Aciduria Argininosuccínica/tratamiento farmacológico , Fenilbutiratos/uso terapéutico , Adolescente , Alanina Transaminasa/sangre , Arginina/sangre , Ácido Argininosuccínico/sangre , Aciduria Argininosuccínica/sangre , Aspartato Aminotransferasas/sangre , Niño , Preescolar , Estudios Cruzados , Método Doble Ciego , Esquema de Medicación , Quimioterapia Combinada , Femenino , Humanos , Pruebas de Función Hepática , Masculino , Fenilbutiratos/sangre , Placebos , Adulto Joven
8.
Am J Hum Genet ; 90(5): 836-46, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22541557

RESUMEN

Argininosuccinate lyase (ASL) is required for the synthesis and channeling of L-arginine to nitric oxide synthase (NOS) for nitric oxide (NO) production. Congenital ASL deficiency causes argininosuccinic aciduria (ASA), the second most common urea-cycle disorder, and leads to deficiency of both ureagenesis and NO production. Subjects with ASA have been reported to develop long-term complications such as hypertension and neurocognitive deficits despite early initiation of therapy and the absence of documented hyperammonemia. In order to distinguish the relative contributions of the hepatic urea-cycle defect from those of the NO deficiency to the phenotype, we performed liver-directed gene therapy in a mouse model of ASA. Whereas the gene therapy corrected the ureagenesis defect, the systemic hypertension in mice could be corrected by treatment with an exogenous NO source. In an ASA subject with severe hypertension refractory to antihypertensive medications, monotherapy with NO supplements resulted in the long-term control of hypertension and a decrease in cardiac hypertrophy. In addition, the NO therapy was associated with an improvement in some neuropsychological parameters pertaining to verbal memory and nonverbal problem solving. Our data show that ASA, in addition to being a classical urea-cycle disorder, is also a model of congenital human NO deficiency and that ASA subjects could potentially benefit from NO supplementation. Hence, NO supplementation should be investigated for the long-term treatment of this condition.


Asunto(s)
Aciduria Argininosuccínica/tratamiento farmacológico , Aciduria Argininosuccínica/fisiopatología , Terapia Genética , Óxido Nítrico/deficiencia , Óxido Nítrico/farmacología , Adolescente , Animales , Arginina/sangre , Argininosuccinatoliasa/genética , Aciduria Argininosuccínica/complicaciones , Aciduria Argininosuccínica/genética , Preescolar , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Humanos , Hipertensión/complicaciones , Hipertensión/tratamiento farmacológico , Hígado/enzimología , Masculino , Ratones , Óxido Nítrico/biosíntesis
9.
Am J Clin Nutr ; 93(6): 1248-54, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21490144

RESUMEN

BACKGROUND: Phenylbutyrate is a drug used in patients with urea cycle disorder to elicit alternative pathways for nitrogen disposal. However, phenylbutyrate administration decreases plasma branched-chain amino acid (BCAA) concentrations, and previous research suggests that phenylbutyrate administration may increase leucine oxidation, which would indicate increased protein degradation and net protein loss. OBJECTIVE: We investigated the effects of phenylbutyrate administration on whole-body protein metabolism, glutamine, leucine, and urea kinetics in healthy and ornithine transcarbamylase-deficient (OTCD) subjects and the possible benefits of BCAA supplementation during phenylbutyrate therapy. DESIGN: Seven healthy control and 7 partial-OTCD subjects received either phenylbutyrate or no treatment in a crossover design. In addition, the partial-OTCD and 3 null-OTCD subjects received phenylbutyrate and phenylbutyrate plus BCAA supplementation. A multitracer protocol was used to determine the whole-body fluxes of urea and amino acids of interest. RESULTS: Phenylbutyrate administration reduced ureagenesis by ≈15% without affecting the fluxes of leucine, tyrosine, phenylalanine, or glutamine and the oxidation of leucine or phenylalanine. The transfer of (15)N from glutamine to urea was reduced by 35%. However, a reduction in plasma concentrations of BCAAs due to phenylbutyrate treatment was observed. BCAA supplementation did not alter the respective baseline fluxes. CONCLUSIONS: Prolonged phenylbutyrate administration reduced ureagenesis and the transfer of (15)N from glutamine to urea without parallel reductions in glutamine flux and concentration. There were no changes in total-body protein breakdown and amino acid catabolism, which suggests that phenylbutyrate can be used to dispose of nitrogen effectively without adverse effects on body protein economy.


Asunto(s)
Aminoácidos de Cadena Ramificada/sangre , Glutamina/metabolismo , Nitrógeno/metabolismo , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/metabolismo , Fenilbutiratos/farmacología , Proteínas/metabolismo , Urea/metabolismo , Adolescente , Adulto , Anciano , Aminoácidos/metabolismo , Aminoácidos/farmacología , Aminoácidos de Cadena Ramificada/farmacología , Niño , Femenino , Humanos , Masculino , Redes y Vías Metabólicas/efectos de los fármacos , Persona de Mediana Edad , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/tratamiento farmacológico , Fenilbutiratos/uso terapéutico , Adulto Joven
10.
Am J Physiol Endocrinol Metab ; 300(1): E188-94, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20978229

RESUMEN

The synthesis of citrulline from arginine in the small intestine depends on the provision of ornithine. To test the hypothesis that arginase II plays a central role in the supply of ornithine for citrulline synthesis, the contribution of dietary arginine, glutamine, and proline was determined by utilizing multitracer stable isotope protocols in arginase II knockout (AII(-/-)) and wild-type (WT) mice. The lack of arginase II resulted in a lower citrulline rate of appearance (121 vs. 137 µmol·kg(-1)·h(-1)) due to a reduced availability of ornithine; ornithine supplementation was able to restore the rate of citrulline production in AII(-/-) to levels comparable with WT mice. There were significant differences in the utilization of dietary citrulline precursors. The contribution of dietary arginine to the synthesis of citrulline was reduced from 45 to 10 µmol·kg(-1)·h(-1) due to the lack of arginase II. No enteral utilization of arginine was observed in AII(-/-) mice (WT = 25 µmol·kg(-1)·h(-1)), and the contribution of dietary arginine through plasma ornithine was reduced in the transgenic mice (20 vs. 13 µmol·kg(-1)·h(-1)). Dietary glutamine and proline utilization were greater in AII(-/-) than in WT mice (20 vs. 13 and 1.4 vs. 3.7 µmol·kg(-1)·h(-1), respectively). Most of the contribution of glutamine and proline was enteral rather than through plasma ornithine. The arginase isoform present in the small intestinal mucosa has the role of providing ornithine for citrulline synthesis. The lack of arginase II results in a greater contribution of plasma ornithine and dietary glutamine and proline to the synthesis of citrulline.


Asunto(s)
Arginasa/metabolismo , Citrulina/metabolismo , Mucosa Intestinal/enzimología , Intestino Delgado/enzimología , Ornitina/metabolismo , Animales , Arginasa/genética , Arginina/administración & dosificación , Arginina/sangre , Arginina/química , Arginina/metabolismo , Isótopos de Carbono , Citrulina/sangre , Citrulina/química , Glutamina/administración & dosificación , Glutamina/metabolismo , Riñón/enzimología , Hígado/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Isótopos de Nitrógeno , Ornitina/administración & dosificación , Ornitina/sangre , Ornitina/química , Fenilalanina/sangre , Fenilalanina/química , Prolina/administración & dosificación , Prolina/sangre , Prolina/química , Prolina/metabolismo , Distribución Aleatoria
11.
Am J Physiol Endocrinol Metab ; 293(6): E1764-71, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17925451

RESUMEN

The spf-ash mutation in mice results in reduced hepatic and intestinal ornithine transcarbamylase. However, a reduction in enzyme activity only translates in reduced ureagenesis and hyperammonemia when an unbalanced nitrogen load is imposed. Six-week-old wild-type control and spf-ash mutant male mice from different genetic backgrounds (B6 and ICR) were infused intravenously with [(13)C(18)O]urea, l-[(15)N(2)]arginine, l-[5,5 D(2)]ornithine, l-[6-(13)C, 4,4,5,5, D(4)]citrulline, and l-[ring-D(5)]phenylalanine to investigate the interaction between genetic background and spf-ash mutation on ureagenesis, arginine metabolism, and nitric oxide production. ICR(spf-ash) mice maintained ureagenesis (5.5 +/- 0.3 mmol.kg(-1).h(-1)) and developed mild hyperammonemia (145 +/- 19 micromol/l) when an unbalanced nitrogen load was imposed; however, B6(spf-ash) mice became hyperammonemic (671 +/- 15 micromol/l) due to compromised ureagenesis (3.4 +/- 0.1 mmol.kg(-1).h(-1)). Ornithine supplementation restored ureagenesis and mitigated hyperammonemia. A reduction in citrulline entry rate was observed due to the mutation in both genetic backgrounds (wild-type: 128, spf-ash: 60; SE 4.0 micromol.kg(-1).h(-1)). Arginine entry rate was only reduced in B6(spf-ash) mice (B6(spf-ash): 332, ICR(spf-ash): 453; SE 20.6 micromol.kg(-1).h(-1)). Genetic background and mutation had an effect on nitric oxide production (B6: 3.4, B6(spf-ash): 2.8, ICR: 9.0, ICR(spf-ash): 4.6, SE 0.7 micromol.kg(-1).h(-1)). Protein breakdown was the main source of arginine during the postabsorptive state and was higher in ICR(spf-ash) than in B6(spf-ash) mice (phenylalanine entry rate 479 and 327, respectively; SE 18 micromol.kg(-1).h(-1)). Our results highlight the importance of the interaction between mutation and genetic background on ureagenesis, arginine metabolism, and nitric oxide production. These observations help explain the wide phenotypic variation of ornithine transcarbamylase deficiency in the human population.


Asunto(s)
Mutación , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/metabolismo , Ornitina Carbamoiltransferasa/metabolismo , Alanina/administración & dosificación , Alanina/metabolismo , Alanina/farmacocinética , Amoníaco/sangre , Amoníaco/metabolismo , Animales , Arginina/administración & dosificación , Arginina/metabolismo , Arginina/farmacocinética , Citrulina/sangre , Citrulina/metabolismo , Modelos Animales de Enfermedad , Femenino , Glicina/administración & dosificación , Glicina/metabolismo , Glicina/farmacocinética , Inyecciones Intravenosas , Mucosa Intestinal/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Endogámicos , Ratones Mutantes , Modelos Biológicos , Óxido Nítrico/metabolismo , Ornitina/administración & dosificación , Ornitina/metabolismo , Ornitina/farmacocinética , Ornitina Carbamoiltransferasa/genética , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/genética , Fenotipo , Fenilalanina/administración & dosificación , Fenilalanina/metabolismo , Fenilalanina/farmacocinética , Urea/sangre , Urea/metabolismo
12.
J Nutr ; 136(7): 1834-8, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16772445

RESUMEN

We showed that Otc(spf-ash) mice, a model of ornithine transcarbamylase deficiency, were able to sustain ureagenesis at the same rate as control mice, despite reduced enzyme activity, when a complete mixture of amino acids was provided. An unbalanced amino acid mixture, however, resulted in reduced ureagenesis and hyperammonemia. To study the effect of ornithine supplementation [316 micromol/(kg.h)] on urea and glutamine kinetics in conscious Otc(spf-ash) mice under a glycine-alanine load [6.06 mmol/(kg.h)], a multiple tracer infusion protocol ([(13)C(18)O]urea, [5-(15)N]glutamine, [2,3,3,4,4 D(5)]glutamine and [ring-D(5)] phenylalanine) was conducted. Ornithine supplementation increased ureagenesis [3.18 +/- 0.88 vs. 4.56 +/- 0.51 mmol/(kg.h), P < 0.001], reduced plasma ammonia concentration (1125 +/- 621 vs. 193 +/- 94 micromol/L, P < 0.001), and prevented acute hepatic enlargement (P < 0.006) in Otc(spf-ash) mice. Ornithine supplementation also increased [96 +/- 20 vs. 120 +/- 16 micromol/(kg.h), P < 0.001] the transfer of (15)N from glutamine to urea, to values observed in the control mice [123 +/- 17 micromol/(kg.h)]. De novo amido-N glutamine flux was higher [1.57 +/- 0.37 vs. 3.04 +/- 0.86 mmol/(kg.h); P < 0.001] in Otc(spf-ash) mice, but ornithine supplementation had no effect (P < 0.56). The flux of glutamine carbon skeleton was affected by both genotype (P < 0.0001) and by ornithine (P 0. 036). In conclusion, ornithine supplementation restored ureagenesis, mitigated hyperammonemia, prevented liver enlargement, and normalized the transfer of (15)N from glutamine to urea. These data strongly suggest that ornithine has the potential for the biochemical correction of OTCD in Otc(spf-ash) mice.


Asunto(s)
Aminoácidos/metabolismo , Glutamina/metabolismo , Hiperamonemia/tratamiento farmacológico , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa , Ornitina/uso terapéutico , Urea/metabolismo , Aminoácidos/sangre , Animales , Ratones , Ornitina/administración & dosificación , Ornitina Carbamoiltransferasa/metabolismo , Urea/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA